1
|
Li T, Xu L, Shen P, Qiu J, Wang Y, Hu J, Guan P, Lin H, Jiang Z, Chen K, Wang J. The role of miRNAs in the associations between particulate matter and ischemic stroke: A nested case-control study. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2025; 368:125750. [PMID: 39870131 DOI: 10.1016/j.envpol.2025.125750] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/25/2024] [Revised: 01/03/2025] [Accepted: 01/24/2025] [Indexed: 01/29/2025]
Abstract
Epidemiological studies have reported that atmospheric particulate matter (PM) contributes to ischemic stroke (IS). Biological studies also indicated that the pathway where PM induces IS involves several pathological processes. Moreover, exposure to PM can alter the expression of specific microRNAs (miRNAs) and ultimately accelerate the onset of IS by regulating related pathways. However, evidence on the role of miRNAs between PM and IS still needs to be fully elucidated. We used the miRNA sequencing datasets from the GEO (Gene Expression Omnibus) to screen miRNAs associated with IS. A nested case-control study was performed, including all incident ischemic stroke cases during the follow-up period and controls matched by age, sex, and entry seasons. Land use regression (LUR) models were constructed to estimate the levels of PM2.5 and PM10. The real-time quantitative PCR (RT-qPCR) assay was applied to detect the expression of candidate miRNAs in plasma samples collected at baseline to verify whether candidate miRNAs differentially expressed between cases and controls. Mediation analyses were applied to evaluate whether PM could induce IS by affecting the expression of miRNAs. We screened 23 miRNAs expressed differentially between cases and controls from the GEO database. A total of 605 incident ischemic stroke patients were finally included in the case group, and 605 healthy controls were matched. The RT-qPCR assay detected 15 differentially expressed miRNAs. Mediating effects of hsa-miR-107, hsa-miR-320b, hsa-miR-423-5p, hsa-miR-483-5p, and hsa-miR-935 were observed for the associations between PM and IS, indicating that PM could promote IS by altering the expression of those miRNAs. In this nested case-control study, PM might induce IS by affecting the expression of hsa-miR-107, hsa-miR-320b, hsa-miR-423-5p, hsa-miR-483-5p and hsa-miR-935.
Collapse
Affiliation(s)
- Tiezheng Li
- Department of Public Health, and Department of Endocrinology of the Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Children's Health, Hangzhou, China
| | - Lisha Xu
- Department of Public Health, and Department of Endocrinology of the Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Children's Health, Hangzhou, China
| | - Peng Shen
- Department of Chronic Disease and Health Promotion, Yinzhou District Center for Disease Control and Prevention, Ningbo, China
| | - Jie Qiu
- Department of Public Health, and Department of Endocrinology of the Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Children's Health, Hangzhou, China
| | - Yixing Wang
- Department of Public Health, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Jingjing Hu
- Department of Public Health, and Department of Endocrinology of the Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Children's Health, Hangzhou, China
| | - Peng Guan
- Department of Epidemiology, School of Public Health, China Medical University, Shenyang, China
| | - Hongbo Lin
- Department of Chronic Disease and Health Promotion, Yinzhou District Center for Disease Control and Prevention, Ningbo, China
| | - Zhiqin Jiang
- Department of Chronic Disease and Health Promotion, Yinzhou District Center for Disease Control and Prevention, Ningbo, China
| | - Kun Chen
- Department of Public Health, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.
| | - Jianbing Wang
- Department of Public Health, and Department of Endocrinology of the Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Children's Health, Hangzhou, China.
| |
Collapse
|
2
|
Li Y, Hsu CT, Yang TT, Cheng KC. Syringaldehyde Alleviates Cardiac Hypertrophy Induced by Hyperglycemia in H9c2 Cells Through GLP-1 Receptor Signals. Pharmaceuticals (Basel) 2025; 18:110. [PMID: 39861172 PMCID: PMC11768131 DOI: 10.3390/ph18010110] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2024] [Revised: 01/07/2025] [Accepted: 01/13/2025] [Indexed: 01/27/2025] Open
Abstract
Background: Cardiac hypertrophy is a significant complication of diabetes, often triggered by hyperglycemia. Glucagon-like peptide-1 (GLP-1) receptor agonists alleviate cardiac hypertrophy, but their efficacy diminishes under GLP-1 resistance. Syringaldehyde (SA), a natural phenolic compound, may activate GLP-1 receptors and mitigate hypertrophy. This study explores SA's therapeutic potential in hyperglycemia-induced cardiac hypertrophy in H9c2 cardiomyocytes. Methods: H9c2 cells were exposed to high glucose to induce hypertrophy. Cells were treated with varying SA concentrations, and hypertrophic biomarkers were analyzed using ELISA, qPCR, and Western blot. Results: SA reduced cell size and hypertrophic biomarkers in a dose-dependent manner while increasing GLP-1 receptor expression and cAMP levels. These effects were attenuated in GLP-1-resistant cells, highlighting the role of GLP-1 receptor activation. AMPK activation was essential, as its inhibition abolished SA's effects. SA also decreased O-linked N-acetylglucosamine transferase (OGT) expression via AMPK activation, contributing to reduced hypertrophy. Conclusions: SA alleviates hyperglycemia-induced cardiac hypertrophy in H9c2 cells by activating the GLP-1 receptor and AMPK signaling pathway.
Collapse
Affiliation(s)
- Yingxiao Li
- Department of Anatomy, College of Medicine, I-Shou University, Kaohsiung 824005, Taiwan;
| | - Chao-Tien Hsu
- Department of Pathology, E-Da Hospital, I-Shou University, Kaohsiung 824005, Taiwan;
| | - Ting-Ting Yang
- School of Chinese Medicine for Post Baccalaureate, College of Medicine, I-Shou University, Kaohsiung 824005, Taiwan;
| | - Kai-Chun Cheng
- Department of Pharmacy, College of Pharmacy and Health Care, Tajen University, Pingtung 90741, Taiwan
| |
Collapse
|
3
|
Abel ED, Gloyn AL, Evans-Molina C, Joseph JJ, Misra S, Pajvani UB, Simcox J, Susztak K, Drucker DJ. Diabetes mellitus-Progress and opportunities in the evolving epidemic. Cell 2024; 187:3789-3820. [PMID: 39059357 PMCID: PMC11299851 DOI: 10.1016/j.cell.2024.06.029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2024] [Revised: 06/21/2024] [Accepted: 06/24/2024] [Indexed: 07/28/2024]
Abstract
Diabetes, a complex multisystem metabolic disorder characterized by hyperglycemia, leads to complications that reduce quality of life and increase mortality. Diabetes pathophysiology includes dysfunction of beta cells, adipose tissue, skeletal muscle, and liver. Type 1 diabetes (T1D) results from immune-mediated beta cell destruction. The more prevalent type 2 diabetes (T2D) is a heterogeneous disorder characterized by varying degrees of beta cell dysfunction in concert with insulin resistance. The strong association between obesity and T2D involves pathways regulated by the central nervous system governing food intake and energy expenditure, integrating inputs from peripheral organs and the environment. The risk of developing diabetes or its complications represents interactions between genetic susceptibility and environmental factors, including the availability of nutritious food and other social determinants of health. This perspective reviews recent advances in understanding the pathophysiology and treatment of diabetes and its complications, which could alter the course of this prevalent disorder.
Collapse
Affiliation(s)
- E Dale Abel
- Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, CA, USA.
| | - Anna L Gloyn
- Department of Pediatrics, Division of Endocrinology & Diabetes, Department of Genetics, Stanford Diabetes Research Center, Stanford University School of Medicine, Stanford, CA, USA
| | - Carmella Evans-Molina
- Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Joshua J Joseph
- Division of Endocrinology, Diabetes and Metabolism, The Ohio State University College of Medicine, Columbus, OH, USA
| | - Shivani Misra
- Department of Metabolism, Digestion and Reproduction, Imperial College London, and Imperial College NHS Trust, London, UK
| | - Utpal B Pajvani
- Department of Medicine, Columbia University Irving Medical Center, New York, NY, USA
| | - Judith Simcox
- Howard Hughes Medical Institute, Department of Biochemistry, University of Wisconsin-Madison, Madison, WI, USA
| | - Katalin Susztak
- Renal, Electrolyte, and Hypertension Division, Department of Medicine, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA, USA
| | - Daniel J Drucker
- Lunenfeld-Tanenbaum Research Institute, Sinai Health System, Toronto, ON, Canada; Department of Medicine, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
4
|
Marino Y, Arangia A, D'Amico R, Cordaro M, Siracusa R, Impellizzeri D, Gugliandolo E, Fusco R, Cuzzocrea S, Di Paola R. Aggravation of TGFβ1-Smad Pathway and Autoimmune Myocarditis by Fungicide (Tebuconazole) Exposure. Int J Mol Sci 2023; 24:11510. [PMID: 37511266 PMCID: PMC10380223 DOI: 10.3390/ijms241411510] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Revised: 06/29/2023] [Accepted: 07/14/2023] [Indexed: 07/30/2023] Open
Abstract
Myocarditis is an inflammatory cardiac disorder and the primary cause of heart failure in young adults. Its origins can be attributed to various factors, including bacterial or viral infections, exposure to toxins or drugs, endocrine disruptors (EDs), and autoimmune processes. Tebuconazole (TEB), which is a member of the triazole fungicide family, is utilized to safeguard agricultural crop plants against fungal pathogens. Although TEB poses serious threats to mammal health, the information about how it induces toxic effects through various pathways, particularly in autoimmune diseases, are still limited. Thus, the aim of this paper was to evaluate the effect of TEB exposure in autoimmune myocarditis (AM). To induce AM, rats were immunized with porcine cardiac myosin and exposed to TEB for 21 days. Thereafter, animals were sacrificed, and histological, biochemical, and molecular analyses were performed. TEB exposure increased heart weight, systolic blood pressure and heart rate already augmented by AM. Additionally, it significantly increased creatine phosphokinase heart (CK-MB), creatine phosphokinase (CPK), cardiac troponin T (cTnT), and cardiac troponin I (cTnI), as compared to the control. From the histological perspective, TEB exacerbates the histological damage induced by AM (necrosis, inflammation and cell infiltration) and increased fibrosis and collagen deposition. TEB exposure strongly increased pro-inflammatory cytokines and prooxidant levels (O2-, H2O2, NO2-, lipid peroxidation) and reduced antioxidant enzyme levels, which were already dysregulated by AM. Additionally, TEB increased NOX-4 expression and the TGFβ1-Smads pathway already activated by AM. Overall, our results showed that TEB exposure strongly aggravated the cardiotoxicity induced by AM.
Collapse
Affiliation(s)
- Ylenia Marino
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, 98166 Messina, Italy
| | - Alessia Arangia
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, 98166 Messina, Italy
| | - Ramona D'Amico
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, 98166 Messina, Italy
| | - Marika Cordaro
- Department of Biomedical, Dental and Morphological and Functional Imaging, University of Messina, Consolare Valeria, 98100 Messina, Italy
| | - Rosalba Siracusa
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, 98166 Messina, Italy
| | - Daniela Impellizzeri
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, 98166 Messina, Italy
| | - Enrico Gugliandolo
- Department of Veterinary Sciences, University of Messina, 98168 Messina, Italy
| | - Roberta Fusco
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, 98166 Messina, Italy
| | - Salvatore Cuzzocrea
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, 98166 Messina, Italy
| | - Rosanna Di Paola
- Department of Veterinary Sciences, University of Messina, 98168 Messina, Italy
| |
Collapse
|
5
|
Bae HR, Chandy M, Aguilera J, Smith EM, Nadeau KC, Wu JC, Paik DT. Adverse effects of air pollution-derived fine particulate matter on cardiovascular homeostasis and disease. Trends Cardiovasc Med 2022; 32:487-498. [PMID: 34619335 PMCID: PMC9063923 DOI: 10.1016/j.tcm.2021.09.010] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/18/2021] [Revised: 09/15/2021] [Accepted: 09/28/2021] [Indexed: 12/11/2022]
Abstract
Air pollution is a rapidly growing major health concern around the world. Atmospheric particulate matter that has a diameter of less than 2.5 µm (PM2.5) refers to an air pollutant composed of particles and chemical compounds that originate from various sources. While epidemiological studies have established the association between PM2.5 exposure and cardiovascular diseases, the precise cellular and molecular mechanisms by which PM2.5 promotes cardiovascular complications are yet to be fully elucidated. In this review, we summarize the various sources of PM2.5, its components, and the concentrations of ambient PM2.5 in various settings. We discuss the experimental findings to date that evaluate the potential adverse effects of PM2.5 on cardiovascular homeostasis and function, and the possible therapeutic options that may alleviate PM2.5-driven cardiovascular damage.
Collapse
Affiliation(s)
- Hye Ryeong Bae
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA, USA
| | - Mark Chandy
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA, USA; Division of Cardiovascular Medicine, Department of Medicine, Stanford University, Stanford, CA, USA
| | - Juan Aguilera
- Sean N. Parker Center for Allergy and Asthma Research and the Division of Pulmonary, Allergy & Critical Care Medicine, Department of Medicine, Stanford University, Stanford, CA, USA
| | - Eric M Smith
- Sean N. Parker Center for Allergy and Asthma Research and the Division of Pulmonary, Allergy & Critical Care Medicine, Department of Medicine, Stanford University, Stanford, CA, USA
| | - Kari C Nadeau
- Sean N. Parker Center for Allergy and Asthma Research and the Division of Pulmonary, Allergy & Critical Care Medicine, Department of Medicine, Stanford University, Stanford, CA, USA
| | - Joseph C Wu
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA, USA; Division of Cardiovascular Medicine, Department of Medicine, Stanford University, Stanford, CA, USA
| | - David T Paik
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA, USA; Division of Cardiovascular Medicine, Department of Medicine, Stanford University, Stanford, CA, USA.
| |
Collapse
|
6
|
Li T, Yu Z, Xu L, Wu Y, Yu L, Yang Z, Shen P, Lin H, Shui L, Tang M, Jin M, Chen K, Wang J. Residential greenness, air pollution, and incident ischemic heart disease: A prospective cohort study in China. THE SCIENCE OF THE TOTAL ENVIRONMENT 2022; 838:155881. [PMID: 35569653 DOI: 10.1016/j.scitotenv.2022.155881] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/07/2022] [Revised: 04/23/2022] [Accepted: 05/08/2022] [Indexed: 06/15/2023]
Abstract
Greener residential surroundings are associated with beneficial health outcomes, whereas higher air pollution exposure is linked with elevated risks of chronic diseases. To date, limited studies have explored the interaction between residential greenness and air pollution on the risk of ischemic heart disease (IHD). We performed a prospective cohort study that included 29,141 adult participants recruited from Yinzhou District, Ningbo, China. Normalized Difference Vegetation Index (NDVI) around each participant's residence was calculated to measure residential greenness exposure. Land-use regression models were conducted to estimate long-term individual exposure to air pollutants, including nitrogen dioxide (NO2) and particulate matter with aerodynamic diameters ≤ 2.5 μm (PM2.5) and ≤10 μm (PM10). Cox proportional hazard models were used to calculate the hazard ratios (HRs) and 95% confidence intervals (95% CIs) for the associations of residential greenness and air pollutants with the risk of incident IHD. During 101,172.5 person-years of follow-up, 1392 incident IHD cases were reported in the study population. Residential greenness, expressed as an interquartile range (IQR) increase in NDVI within 250 m, was inversely associated with incident IHD (HR = 0.89, 95%CI: 0.81,0.98). However, long-term exposures to air pollution were associated with higher IHD incidence (HR = 1.21, 95%CI:1.10,1.33 per IQR increase for PM2.5; HR = 1.12, 95%CI:1.03,1.22 per IQR increase for PM10; HR = 1.09, 95%CI:1.02,1.16 per IQR increase for NO2). Mediation analyses suggested that the beneficial effect of residential greenness on incident IHD could be partly mediated by reducing the exposure to PM2.5. These findings suggested that higher greenness was associated with decreased risk of IHD, while air pollutants were positively associated with incident IHD. Meanwhile, residential greenness may decrease the risk of IHD by reducing exposure to PM2.5.
Collapse
Affiliation(s)
- Tiezheng Li
- Department of Epidemiology and Biostatistics at School of Public Health and National Clinical Research Center for Child Health of the Children's Hospital, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Zhebin Yu
- Department of Epidemiology and Biostatistics, Zhejiang University School of Public Health, Zhejiang University 866 Yuhangtang Road, Hangzhou 310058, China
| | - Lisha Xu
- Department of Epidemiology and Biostatistics at School of Public Health and National Clinical Research Center for Child Health of the Children's Hospital, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Yonghao Wu
- Department of Epidemiology and Biostatistics at School of Public Health and National Clinical Research Center for Child Health of the Children's Hospital, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Luhua Yu
- Department of Epidemiology and Biostatistics at School of Public Health and National Clinical Research Center for Child Health of the Children's Hospital, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Zongming Yang
- Department of Epidemiology and Biostatistics at School of Public Health and National Clinical Research Center for Child Health of the Children's Hospital, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Peng Shen
- Department of Chronic Disease and Health Promotion, Yinzhou District Center for Disease Control and Prevention, 1221 Xueshi Road, Ningbo, Zhejiang 315100, China
| | - Hongbo Lin
- Department of Chronic Disease and Health Promotion, Yinzhou District Center for Disease Control and Prevention, 1221 Xueshi Road, Ningbo, Zhejiang 315100, China
| | - Liming Shui
- Yinzhou District Health Bureau of Ningbo, 1221 Xueshi Road, Ningbo, Zhejiang 315100, China
| | - Mengling Tang
- Department of Epidemiology and Biostatistics at School Public Health and the Fourth Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Mingjuan Jin
- Department of Epidemiology and Biostatistics, Cancer Institute of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Kun Chen
- Department of Epidemiology and Biostatistics, Cancer Institute of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310058, China..
| | - Jianbing Wang
- Department of Epidemiology and Biostatistics at School of Public Health and National Clinical Research Center for Child Health of the Children's Hospital, Zhejiang University School of Medicine, Hangzhou 310058, China.
| |
Collapse
|
7
|
Wu T, Tong M, Chu A, Wu K, Niu X, Zhang Z. PM2.5-Induced Programmed Myocardial Cell Death via mPTP Opening Results in Deteriorated Cardiac Function in HFpEF Mice. Cardiovasc Toxicol 2022; 22:746-762. [PMID: 35593990 DOI: 10.1007/s12012-022-09753-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Accepted: 05/06/2022] [Indexed: 11/03/2022]
Abstract
PM2.5 exposure can induce or exacerbate heart failure and is associated with an increased risk of heart failure hospitalization and mortality; however, the underlying mechanisms remain unclear. This study focuses on the potential mechanisms underlying PM2.5 induction of cardiomyocyte programmed necrosis as well as its promotion of cardiac function impairment in a mouse model of heart failure with preserved ejection fraction (HFpEF). HFpEF mice were exposed to concentrated ambient PM2.5 (CAP) (CAP group) or filtered air (FA) (FA group) for 6 weeks. Changes in myocardial pathology and cardiac function were documented for comparisons between the two groups. In vitro experiments were performed to measure oxidative stress and mitochondrial permeability transition pore (mPTP) dynamics in H9C2 cells following 24 h exposure to PM2.5. Additionally, co-immunoprecipitation was conducted to detect p53 and cyclophilin D (CypD) interactions. The results showed exposure to CAP promoted cardiac function impairment in HFpEF mice. Myocardial pathology analysis and in vitro experiments demonstrated that PM2.5 led to mitochondrial damage in cardiomyocytes and, eventually, their necrosis. Moreover, our experiments also suggested that PM2.5 increases mitochondrial reactive oxygen species (ROS), induces DNA oxidative damage, and decreases the inner mitochondrial membrane potential (ΔΨm). This indicates the presence of mPTP opening. Co-immunoprecipitation results showed a p53/CypD interaction in the myocardial tissue of HFpEF mice in the CAP group. Inhibition of CypD by cyclosporin A was found to reverse PM2.5-induced mPTP opening and H9C2 cell death. In conclusion, PM2.5 induces mPTP opening to stimulate mitochondria-mediated programmed necrosis of cardiomyocytes, and it might exacerbate cardiac function impairment in HFpEF mice.
Collapse
Affiliation(s)
- Tingting Wu
- The First Clinical Medical College of Lanzhou University, Lanzhou, 730000, China
- The Second Hospital of Lanzhou University, Lanzhou, 730030, China
| | - Minghui Tong
- The Second Hospital of Lanzhou University, Lanzhou, 730030, China
| | - Aiai Chu
- Department of Cardiology, Gansu Provincial Hospital, Lanzhou, 730000, China
| | - Kaiyue Wu
- Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, 200030, China
| | - Xiaowei Niu
- Heart Center, The First Hospital of Lanzhou University, Lanzhou, 730000, China
| | - Zheng Zhang
- Heart Center, The First Hospital of Lanzhou University, Lanzhou, 730000, China.
| |
Collapse
|
8
|
Molecular Correlates of Early Onset of Diabetic Cardiomyopathy: Possible Therapeutic Targets. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:9014155. [PMID: 35464763 PMCID: PMC9023181 DOI: 10.1155/2022/9014155] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/06/2022] [Accepted: 03/24/2022] [Indexed: 11/18/2022]
Abstract
Diabetes mellitus (DM) is associated with mitochondrial dysfunction and oxidative stress that can lead to diabetic cardiomyopathy (DCM), which can often remain undetected until late stages of the disease. However, myocardial injury occurs before the onset of measurable cardiac dysfunction, although its molecular correlates are poorly understood. In this study, we made a DM rat induced by a high-fat diet combined with low and high doses of streptozotocin (STZ) to emulate pre and early DCM. RNA-sequencing analysis of ventricular tissue revealed a differential transcriptome profile and abnormal activation of pathways involved in fatty acid metabolism, oxidative phosphorylation, cardiac structure and function, insulin resistance, calcium signalling, apoptosis, and TNF signalling. Moreover, using high glucose-treated human induced pluripotent stem cell-derived cardiomyocytes (iPSC-CM), we recapitulated the cardiac cellular phenotype of DM and identified several molecular correlates that may promote the development of DCM. In conclusion, we have developed an experimental framework to target pathways underlying the progression of DCM.
Collapse
|
9
|
Kobos L, Shannahan J. Particulate matter inhalation and the exacerbation of cardiopulmonary toxicity due to metabolic disease. Exp Biol Med (Maywood) 2021; 246:822-834. [PMID: 33467887 DOI: 10.1177/1535370220983275] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Particulate matter is a significant public health issue in the United States and globally. Inhalation of particulate matter is associated with a number of systemic and organ-specific adverse health outcomes, with the pulmonary and cardiovascular systems being particularly vulnerable. Certain subpopulations are well-recognized as being more susceptible to inhalation exposures, such as the elderly and those with pre-existing respiratory disease. Metabolic syndrome is becoming increasingly prevalent in our society and has known adverse effects on the heart, lungs, and vascular systems. The limited evaluations of individuals with metabolic syndromehave demonstrated that theymay compose a sensitive subpopulation to particulate exposures. However, the toxicological mechanisms responsible for this increased vulnerability are not fully understood. This review evaluates the currently available literature regarding how the response of an individual's pulmonary and cardiovascular systems is influenced by metabolic syndrome and metabolic syndrome-associated conditions such as hypertension, dyslipidemia, and diabetes. Further, we will discuss potential therapeutic agents and targets for the alleviation and treatment of particulate-matter induced metabolic illness. The information reviewed here may contribute to the understanding of metabolic illness as a risk factor for particulate matter exposure and further the development of therapeutic approaches to treat vulnerable subpopulations, such as those with metabolic diseases.
Collapse
Affiliation(s)
- Lisa Kobos
- School of Health Sciences, College of Human and Health Sciences, Purdue University, West Lafayette, IN 47907, USA
| | - Jonathan Shannahan
- School of Health Sciences, College of Human and Health Sciences, Purdue University, West Lafayette, IN 47907, USA
| |
Collapse
|
10
|
Gratz D, Hund TJ, Falvo MJ, Wold LE. Reverse Translation: Using Computational Modeling to Enhance Translational Research. Circ Res 2019; 122:1496-1498. [PMID: 29798899 DOI: 10.1161/circresaha.118.313003] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Affiliation(s)
- Daniel Gratz
- From the Dorothy M. Davis Heart and Lung Research Institute (D.G., T.J.H., L.E.W.).,The Ohio State University Wexner Medical Center, Columbus; Department of Biomedical Engineering, College of Engineering (D.G., T.J.H.)
| | - Thomas J Hund
- From the Dorothy M. Davis Heart and Lung Research Institute (D.G., T.J.H., L.E.W.).,Department of Internal Medicine (T.J.H.).,The Ohio State University Wexner Medical Center, Columbus; Department of Biomedical Engineering, College of Engineering (D.G., T.J.H.)
| | - Michael J Falvo
- The Ohio State University, Columbus; War Related Illness and Injury Study Center, Department of Veterans Affairs, New Jersey Health Care System, East Orange (M.J.F.).,Rutgers Biomedical and Health Sciences, New Jersey Medical School, Newark (M.J.F.)
| | - Loren E Wold
- From the Dorothy M. Davis Heart and Lung Research Institute (D.G., T.J.H., L.E.W.) .,College of Nursing (L.E.W.).,Department of Physiology and Cell Biology (L.E.W.)
| |
Collapse
|
11
|
TGR5 activation ameliorates hyperglycemia-induced cardiac hypertrophy in H9c2 cells. Sci Rep 2019; 9:3633. [PMID: 30842472 PMCID: PMC6403401 DOI: 10.1038/s41598-019-40002-0] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2018] [Accepted: 02/06/2019] [Indexed: 12/20/2022] Open
Abstract
Left ventricular hypertrophy is an independent risk factor in diabetic patients. TGR5 is shown to express in hearts, but its functional role in diabetes-induced cardiac hypertrophy remained unclear. The current study investigated the role of TGR5 on high glucose-induced hypertrophy of H9C2 cells. After incubation with a high level of glucose, H9C2 cells showed hypertrophic responses. Activation of TGR5 by lithocholic acid (LCA) ameliorated cell hypertrophy and enhanced SERCA2a and phosphorylated phospholamban (PLN) expression in H9C2 cells. Triamterene inhibited these effects at an effective dose to block TGR5. However, LCA failed to modify the free radical elevation induced by high-glucose in the H9c2 cells. Moreover, PKA inhibitors, but not an Epac blocker, markedly improved hyperglycemia-induced hypertrophy and attenuated the increased SERCA2a expression by LCA; it also attenuated the phosphorylated PLN and SERCA2a protein expression levels in high glucose-treated H9C2 cells. In conclusion, TGR5 activation stimulated protein kinase A (PKA) to enhance PLN phosphorylation, which activated SERCA2a to remove Ca2+ from cytosol to sarcoplasmic reticulum in addition to the reduction of calcineurin/NFAT pathway signaling to ameliorate the hyperglycemia-induced cardiac hypertrophy shown in cardiomyocytes. TGR5 may service as a new target in the control of diabetic cardiomyopathy.
Collapse
|
12
|
Qu Y, Pan Y, Niu H, He Y, Li M, Li L, Liu J, Li B. Short-term effects of fine particulate matter on non-accidental and circulatory diseases mortality: A time series study among the elder in Changchun. PLoS One 2018; 13:e0209793. [PMID: 30596713 PMCID: PMC6312390 DOI: 10.1371/journal.pone.0209793] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2018] [Accepted: 12/11/2018] [Indexed: 12/26/2022] Open
Abstract
Background and objectives Fine particulate matter (PM2.5, particulate matter with an aerodynamic diameter less than or equal to 2.5 μm) has multiple adverse effects on human health, especially on the respiratory and circulatory system. The purpose of this study was to evaluate the short-term effect of PM2.5 on the mortality risk of non-accidental and circulatory diseases, and to explore the potential effect modification by sex, education and death location. Methods We collected daily mortality counts of Changchun (China) residents, daily meteorology and air pollution data, from January 1, 2014, to January 1, 2017. We focused on the elderly (≥65 years old) population who died from non-accidental causes and circulatory diseases, and stratified them by sex, education, and death location. A generalized additive Poisson regression model (GAM) was used to analyse the impact of air pollutants on mortality. We fit single pollutant models to examine PM2.5 effects with different lag structures of single-day (distributed lag:lag0-lag3) and multi-day (moving average lag: lag01-lag03). To test the sensitivity of the model, a multi-pollutant model was established when the PM2.5 effect was strongest. Results In the single pollutant models, an increment of PM2.5 by 10 μg/m3 at lag0-3 was associated with a 0.385% (95% CI: 0.069% to 0.702%) increase in daily non-accidental mortality and a 0.442% (95% CI: 0.038% to 0.848%) increase in daily circulatory disease mortality. NO2 (lag1) and O3 (lag0, lag1, lag2, lag01,lag02, lag03) were associated with daily non-accidental death and NO2 (lag1, lag3, lag03) and O3 (lag0, lag1, lag01,lag02, lag03) were associated with daily circulatory disease mortality. In the co-pollutant models, the risk estimates for PM2.5 changed slightly. The excess mortality risk of non-accidental and circulatory diseases was higher for women, people with low education, and died outside hospital. Conclusions We found that short-term exposure to PM2.5 increased the mortality risk of non-accidental and circulatory diseases among the elderly in Changchun. Women, people with low education and died outside hospital are more susceptible to PM2.5. NO2 and O3 were also associated with an increase in mortality from non-accidental and circulatory diseases and the O3 is a high effect.
Collapse
Affiliation(s)
- Yangming Qu
- Key Laboratory of Zoonosis Research, Ministry of Education, School of Public Health, Jilin University, Changchun, Jilin, China
| | - Yang Pan
- Jilin Provincial Center for Disease Control and Prevention, Changchun, Jilin, China
| | - Huikun Niu
- Key Laboratory of Zoonosis Research, Ministry of Education, School of Public Health, Jilin University, Changchun, Jilin, China
| | - Yinghua He
- Jilin Provincial Center for Disease Control and Prevention, Changchun, Jilin, China
| | - Meiqi Li
- Key Laboratory of Zoonosis Research, Ministry of Education, School of Public Health, Jilin University, Changchun, Jilin, China
| | - Lu Li
- Key Laboratory of Zoonosis Research, Ministry of Education, School of Public Health, Jilin University, Changchun, Jilin, China
| | - Jianwei Liu
- Jilin Provincial Center for Disease Control and Prevention, Changchun, Jilin, China
| | - Bo Li
- Key Laboratory of Zoonosis Research, Ministry of Education, School of Public Health, Jilin University, Changchun, Jilin, China
- * E-mail:
| |
Collapse
|
13
|
Chen W, Sun Q, Ju J, Chen W, Zhao X, Zhang Y, Yang Y. Effect of Astragalus Polysaccharides on Cardiac Dysfunction in db/db Mice with Respect to Oxidant Stress. BIOMED RESEARCH INTERNATIONAL 2018; 2018:8359013. [PMID: 30581869 PMCID: PMC6276493 DOI: 10.1155/2018/8359013] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/10/2018] [Revised: 08/25/2018] [Accepted: 09/06/2018] [Indexed: 01/03/2023]
Abstract
OBJECTIVE Oxidant stress plays an important role in the development of diabetic cardiomyopathy. Previously we reported that Astragalus polysaccharides (APS) rescued heart dysfunction and cardinal pathological abnormalities in diabetic mice. In the current study, we determined whether the effect of APS on diabetic cardiomyopathy was associated with its impact on oxidant stress. METHODS Db/db diabetic mice were employed and administered with APS. The hematodynamics, cardiac ultra-structure, apoptosis, and ROS formation of myocardium were assessed. The cardiac protein expression of apoptosis target genes (Bax, Bcl-2, and caspase-3) and oxidation target genes (Gpx, SOD2, t/p-JNK, catalase, t/p-p38 MAPK, and t/p-ERK) were evaluated, respectively. RESULTS APS therapy improved hematodynamics and cardinal ultra-structure with reduced apoptosis and ROS formation in db/db hearts. In addition, APS therapy inhibited the protein expression of apoptosis target genes (Bax, Bcl-2, and caspase-3) and regulated the protein expression of oxidation target genes (enhancing Gpx, SOD2, and catalase, while reducing t/p-JNK, t/p-ERK, and t/p-p38 MAPK) in db/db hearts. CONCLUSION Our findings suggest that APS has benefits in diabetic cardiomyopathy, which may be partly associated with its impact on cardiac oxidant stress.
Collapse
Affiliation(s)
- Wei Chen
- Department of Geriatrics, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - Qilin Sun
- Department of Geriatrics, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - Jing Ju
- Department of Geriatrics, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - Wenjie Chen
- Department of Geriatrics, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - Xuelan Zhao
- Department of Geriatrics, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - Yu Zhang
- Department of Geriatrics, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - Yehong Yang
- Department of Endocrinology, Huashan Hospital, Fudan University, Shanghai 200040, China
| |
Collapse
|
14
|
Kuo SC, Li Y, Cheng YZ, Lee WJ, Cheng JT, Cheng KC. Molecular mechanisms regarding potassium bromate‑induced cardiac hypertrophy without apoptosis in H9c2 cells. Mol Med Rep 2018; 18:4700-4708. [PMID: 30221729 DOI: 10.3892/mmr.2018.9470] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2018] [Accepted: 08/16/2018] [Indexed: 11/05/2022] Open
Abstract
Cardiac hypertrophy is commonly involved in cardiac injury. Oxidative stress can induce cardiac hypertrophy with apoptosis. Potassium bromate (KBrO3) has been widely used as a food additive due to its oxidizing properties. In the present study, the rat‑derived heart cell line H9c2 was used to investigate the effect of KBrO3 on cell size. KBrO3 increased cell size at concentrations <250 µM, in a dose‑dependent manner. Additionally, KBrO3 also promoted the gene expression of two biomarkers of cardiac hypertrophy, brain/B‑type natriuretic peptides (BNP) and β‑Myosin Heavy Chain (β‑MHC). However, apoptosis remained unobserved in these cells. Moreover, mediation of free radicals was investigated using a fluorescence assay, and it was observed that superoxide and reactive oxygen species (ROS) levels increased with KBrO3. Effects of KBrO3 were significantly reduced by tiron at concentrations sufficient to produce antioxidant‑like action. Additionally, signals involved in cardiac hypertrophy such as calcineurin and nuclear factor of activated T‑cells (NFAT) were also determined using western blot analysis. KBrO3 increased the protein levels of both these molecules which were decreased by tiron in a dose‑dependent manner. Additionally, cyclosporine A attenuated the cardiac hypertrophy induced by KBrO3 in H9c2 cells at concentrations effective to inhibit calcineurin, in addition to reducing mRNA levels of BNP or β‑MHC. Finally, apoptosis was also identified in H9c2 cells incubated with KBrO3 at concentrations >300 µM. Collectively, these results provided a novel perspective that KBrO3 induces cardiac hypertrophy without apoptosis at a low dose through the generation of ROS, activating the calcineurin/NFAT signaling pathway in H9c2 cells. Therefore, at a dose <250 µM, KBrO3 can be applied as an inducer of cardiac hypertrophy without apoptosis in H9c2 cells. KBrO3 can also be developed as a tool to induce cardiac hypertrophy in animals.
Collapse
Affiliation(s)
- Shu-Chun Kuo
- Department of Optometry, Chung Hwa University of Medical Technology, Tainan 7170, Taiwan R.O.C
| | - Yingxiao Li
- Department of Medical Research, Chi‑Mei Medical Center, Tainan 71003, Taiwan R.O.C
| | - Yung-Ze Cheng
- Department of Emergency Medicine, Chi‑Mei Medical Center, Tainan 71003, Taiwan, R.O.C
| | - Wei-Jing Lee
- Department of Emergency Medicine, Chi‑Mei Medical Center, Tainan 71003, Taiwan, R.O.C
| | - Juei-Tang Cheng
- Department of Medical Research, Chi‑Mei Medical Center, Tainan 71003, Taiwan R.O.C
| | - Kai-Chun Cheng
- Department of Psychosomatic Internal Medicine, Kagoshima University Graduate School of Medical and Dental Sciences, Kagoshima 890, Japan
| |
Collapse
|
15
|
Cheng KC, Chang WT, Li Y, Cheng YZ, Cheng JT, Chen ZC. GW0742 activates peroxisome proliferator-activated receptor δ to reduce free radicals and alleviate cardiac hypertrophy induced by hyperglycemia in cultured H9c2 cells. J Cell Biochem 2018; 119:9532-9542. [PMID: 30129179 DOI: 10.1002/jcb.27270] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2018] [Accepted: 06/26/2018] [Indexed: 01/24/2023]
Abstract
Peroxisome proliferator-activated receptor δ (PPARδ), the predominant PPAR subtype in the heart, is known to regulate cardiac function. PPARδ activation may inhibit cardiac hypertrophy in H9c2 cells while the potential mechanism has not been elucidated. Then, H9c2 cells incubated with high glucose to induce hypertrophy were used to investigate using GW0742 to activate PPARδ. The fluorescence assays were applied to determine the changes in cell size, cellular calcium levels, and free radicals. Western blot analyses for hypertrophic signals and assays of messenger RNA (mRNA) levels for hypertrophic biomarkers were performed. In H9c2 cells, GW0742 inhibited cardiac hypertrophy. In addition, increases in cellular calcium and hypertrophic signals, including calcineurin and nuclear factor of activated T-cells, were reduced by GW0742. This reduction was parallel to the decrease in the mRNA levels of biomarkers, such as brain/B-type natriuretic peptides and β-myosin heavy chain. These effects of GW0742 were dose-dependently inhibited by GSK0660 indicating an activation of PPARδ by GW0742 to alleviate cardiac hypertrophy. Moreover, free radicals produced by hyperglycemia were also markedly inhibited by GW0742 and were later reversed by GSK0660. GW0742 promoted the expression of thioredoxin, an antioxidant enzyme. Direct inhibition of reactive oxygen species by GW0742 was also identified in the oxidant potassium bromate stimulated H9c2 cells. Taken together, these findings suggest that PPARδ agonists can inhibit free radicals, resulting in lower cellular calcium for reduction of hypertrophic signaling to alleviate cardiac hypertrophy in H9c2 cells. Therefore, PPARδ activation can be used to develop agent(s) for treating cardiac hypertrophy.
Collapse
Affiliation(s)
- Kai-Chun Cheng
- Department of Psychosomatic Internal Medicine, Kagoshima University Graduate School of Medical and Dental Sciences, Kagoshima, Japan
| | - Wei-Ting Chang
- Department of Cardiology, Chi-Mei Medical Center, Tainan, Taiwan
| | - Yingxiao Li
- Department of Psychosomatic Internal Medicine, Kagoshima University Graduate School of Medical and Dental Sciences, Kagoshima, Japan.,Department of Medical Research, Chi-Mei Medical Center, Tainan, Taiwan
| | - Yung-Ze Cheng
- Department of Emergency Medicine, Chi-Mei Medical Center, Tainan, Taiwan
| | - Juei-Tang Cheng
- Department of Medical Research, Chi-Mei Medical Center, Tainan, Taiwan.,Graduate Institute of Medical Science, Chang Jung Christian University, Gueiren, Tainan, Taiwan
| | - Zhih-Cherng Chen
- Department of Cardiology, Chi-Mei Medical Center, Tainan, Taiwan.,Department of Pharmacy, Chia Nan University of Pharmacy & Science, Tainan, Taiwan
| |
Collapse
|
16
|
Qin G, Xia J, Zhang Y, Guo L, Chen R, Sang N. Ambient fine particulate matter exposure induces reversible cardiac dysfunction and fibrosis in juvenile and older female mice. Part Fibre Toxicol 2018; 15:27. [PMID: 29941001 PMCID: PMC6019275 DOI: 10.1186/s12989-018-0264-2] [Citation(s) in RCA: 59] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2017] [Accepted: 06/14/2018] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND Cardiovascular disease is the leading cause of mortality in the advanced world, and age is an important determinant of cardiac function. The purpose of the study is to determine whether the PM2.5-induced cardiac dysfunction is age-dependent and whether the adverse effects can be restored after PM2.5 exposure withdrawal. METHODS Female C57BL/6 mice at different ages (4-week-old, 4-month-old, and 10-month-old) received oropharyngeal aspiration of 3 mg/kg b.w. PM2.5 every other day for 4 weeks. Then, 10-month-old and 4-week-old mice were exposed to PM2.5 for 4 weeks and withdrawal PM2.5 1 or 2 weeks. Heart rate and systolic blood pressure were measured using a tail-cuff system. Cardiac function was assessed by echocardiography. Left ventricles were processed for histology to assess myocardial fibrosis. ROS generation was detected by photocatalysis using 2',7'-dichlorodihydrofluorescein diacetate (DCFHDA). The expression of cardiac fibrosis markers (Col1a1, Col3a1) and possible signaling molecules, including NADPH oxidase 4 (NOX-4), transforming growth factor β1 (TGFβ1), and Smad3, were detected by qPCR and/ or Western blot. RESULTS PM2.5 exposure induced cardiac diastolic dysfunction of mice, elevated the heart rate and blood pressure, developed cardiac systolic dysfunction of 10-month-old mice, and caused fibrosis in both 4-week-old and 10-month-old mice. PM2.5 exposure increased the expression of Col1a1, Col3a1, NOX-4, and TGFβ1, activated Smad3, and generated more reactive oxygen species in the myocardium of 4-week-old and 10-month-old mice. The withdrawal from PM2.5 exposure restored blood pressure, heart rate, cardiac function, expression of collagens, and malonaldehyde (MDA) levels in hearts of both 10-month-old and 4-week-old mice. CONCLUSION Juvenile and older mice are more sensitive to PM2.5 than adults and suffer from cardiac dysfunction. PM2.5 exposure reversibly elevated heart rate and blood pressure, induced cardiac systolic dysfunction of older mice, and reversibly induced fibrosis in juvenile and older mice. The mechanism by which PM2.5 exposure resulted in cardiac lesions might involve oxidative stress, NADPH oxidase, TGFβ1, and Smad-dependent pathways.
Collapse
Affiliation(s)
- Guohua Qin
- College of Environment and Resource, Research Center of Environment and Health, Shanxi University, Taiyuan, Shanxi 030006 People’s Republic of China
| | - Jin Xia
- College of Environment and Resource, Research Center of Environment and Health, Shanxi University, Taiyuan, Shanxi 030006 People’s Republic of China
| | - Yingying Zhang
- College of Environment and Resource, Research Center of Environment and Health, Shanxi University, Taiyuan, Shanxi 030006 People’s Republic of China
| | - Lianghong Guo
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing, 100085 People’s Republic of China
| | - Rui Chen
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety& CAS Center for Excellence in Nanoscience, Beijing Key Laboratory of Ambient Particles Health Effects and PreventionTechniques, National Center for Nanoscience & Technology of China, Beijing, 100190 People’s Republic of China
| | - Nan Sang
- College of Environment and Resource, Research Center of Environment and Health, Shanxi University, Taiyuan, Shanxi 030006 People’s Republic of China
| |
Collapse
|
17
|
Hou J, Zheng D, Xiao W, Li D, Ma J, Hu Y. Mangiferin Enhanced Autophagy via Inhibiting mTORC1 Pathway to Prevent High Glucose-Induced Cardiomyocyte Injury. Front Pharmacol 2018; 9:383. [PMID: 29719509 PMCID: PMC5913280 DOI: 10.3389/fphar.2018.00383] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2018] [Accepted: 04/04/2018] [Indexed: 12/11/2022] Open
Abstract
Mangiferin functions as a perfect anti-oxidative compound in the diabetic heart, however, the exact mechanism remains to be elucidated. Here, we show the cardioprotective effect of mangiferin under high glucose-induced cardiotoxic condition mainly contributed to enhanced autophagy via suppressing mTORC1 downstream signal transduction. Primary neonatal rat cardiomyocytes were cultured to detect myocytes injury, autophagy, and related signal transduction under different doses of glucose and mangiferin treatment. High glucose (30 mM) reduced autophagic flux, and increased myocyte apoptosis and death compared with normal glucose (5.5 mM) as determined by variation of autophagy markers LC3-II, p62, parkin, GFP-LC3, or mRFP-LC3 fluorescence puncta, cell viability, cleaved caspase 3, cleaved PARP apoptosis indices, reactive oxygen species (ROS), MAO, and PI death indices. Conversely, mangiferin inhibited hyperglycemia associated oxidative stress by reducing ROS, MAO, cleaved caspase 3, and cleaved PARP generation, reestablishing cell viability, mitochondrial membrane potential, and enhancing autophagic flux, thereby preventing myocytes from high glucose-induced toxicity. Furthermore, cardioprotection with mangiferin was potentially related to the decreased mTOR phosphorylation and suppression of mTORC1 downstream signaling pathway. These data indicated the valuable effects of mangiferin on regulation of cardiac autophagy and pointed to the promising utilization for hyperglycemia control.
Collapse
Affiliation(s)
- Jun Hou
- Department of Pharmacy, Chengdu Military General Hospital, Chengdu, China
| | - Dezhi Zheng
- Department of Cardiovascular Surgery, Jinan Military General Hospital, Jinan, China
| | - Wenjing Xiao
- Department of Pharmacy, Chengdu Military General Hospital, Chengdu, China
| | - Dandan Li
- Base for Drug Clinical Trial, Xinqiao Hospital, Army Medical University, Chongqing, China
| | - Jie Ma
- Department of Pharmacy, Chengdu Military General Hospital, Chengdu, China
| | - Yonghe Hu
- Department of Pharmacy, Chengdu Military General Hospital, Chengdu, China
| |
Collapse
|
18
|
Fiordelisi A, Piscitelli P, Trimarco B, Coscioni E, Iaccarino G, Sorriento D. The mechanisms of air pollution and particulate matter in cardiovascular diseases. Heart Fail Rev 2018; 22:337-347. [PMID: 28303426 DOI: 10.1007/s10741-017-9606-7] [Citation(s) in RCA: 228] [Impact Index Per Article: 32.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Clinical and epidemiological studies demonstrate that short- and long-term exposure to air pollution increases mortality due to respiratory and cardiovascular diseases. Given the increased industrialization and the increased sources of pollutants (i.e., cars exhaust emissions, cigarette smoke, industry emissions, burning of fossil fuels, incineration of garbage), air pollution has become a key public health issue to solve. Among pollutants, the particulate matter (PM) is a mixture of solid and liquid particles which differently affects human health depending on their size (i.e., PM10 with a diameter <10 μm reach the lung and PM2.5 with a diameter <2.5 μm penetrate deeper into the lung). In particular, the acute exposure to PM10 and PM2.5 increases the rate of cardiovascular deaths. Thus, appropriate interventions to reduce air pollution may promote great benefits to public health by reducing the risk of cardiovascular diseases. Several biological mechanisms have been identified to date which could be responsible for PM-dependent adverse cardiovascular outcomes. Indeed, the exposure to PM10 and PM2.5 induces sustained oxidative stress and inflammation. PM2.5 is also able to increase autonomic nervous system activation. Some potential therapeutic approaches have been tested both in pre-clinical and clinical studies, based on the intake of antioxidants from dietary or by pharmacological administration. Studies are still in progress to increase the knowledge of PM activation of intracellular pathways and propose new strategies of intervention.
Collapse
Affiliation(s)
- Antonella Fiordelisi
- I.O.S, Southern Italy Hospital Institute, Medicina Futura Research, 80100, Naples, Italy
| | - Prisco Piscitelli
- I.O.S, Southern Italy Hospital Institute, Medicina Futura Research, 80100, Naples, Italy
| | - Bruno Trimarco
- Department of Advanced Biomedical Sciences, University Federico II of Naples, Via Pansini 5, 80131, Naples, Italy
| | - Enrico Coscioni
- Division of Cardiac Surgery, AOU San Giovanni di Dio e Ruggi d'Aragona, Via Largo d'Ippocrate, 84131, Salerno, Italy
| | - Guido Iaccarino
- Division of Cardiac Surgery, AOU San Giovanni di Dio e Ruggi d'Aragona, Via Largo d'Ippocrate, 84131, Salerno, Italy
- Department of Medicine and Surgery, University of Salerno, Via Salvator Allende, 84081, Baronissi, SA, Italy
| | - Daniela Sorriento
- Department of Advanced Biomedical Sciences, University Federico II of Naples, Via Pansini 5, 80131, Naples, Italy.
| |
Collapse
|
19
|
Liang H, Qiu H, Tian L. Short-term effects of fine particulate matter on acute myocardial infraction mortality and years of life lost: A time series study in Hong Kong. THE SCIENCE OF THE TOTAL ENVIRONMENT 2018; 615:558-563. [PMID: 28988091 DOI: 10.1016/j.scitotenv.2017.09.266] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/31/2017] [Revised: 09/15/2017] [Accepted: 09/25/2017] [Indexed: 06/07/2023]
Abstract
Previous studies have applied years of life lost (YLL) as a complementary indicator to assess the short-term effect of the air pollution on the health burden from all-cause mortality, but sparsely focused on individual diseases such as acute myocardial infraction (AMI). In this study, we aimed to conduct a time-series analysis to evaluate short-term effects of fine particulate matter (PM2.5) on mortality and YLL from AMI in Hong Kong from 2011 to 2015, and explore the potential effect modifiers including sex and age by subgroup analysis. We applied generalized additive Poisson and Gaussian regression model for daily death count and YLL, respectively. We found that 10μg/m3 increment in concentration of PM2.5 lasting for two days (lag01) was associated with a 2.35% (95% CI 0.38% to 4.36%) increase in daily mortality count and a 1.69 (95% CI 0.01 to 3.37) years increase in YLL from AMI. The association between PM2.5 and AMI mortality count was stronger among women and older people than men and young people, respectively. We concluded that acute exposure to PM2.5 may increase the risk of mortality and YLL from AMI in Hong Kong and this effect can be modified by age and gender. These findings add to the evidence base for public health policy formulation and resource allocation.
Collapse
Affiliation(s)
- Haiqing Liang
- School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong Special Administrative Region
| | - Hong Qiu
- School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong Special Administrative Region
| | - Linwei Tian
- School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong Special Administrative Region.
| |
Collapse
|
20
|
Zhu H, Gao Y, Zhu S, Cui Q, Du J. Klotho Improves Cardiac Function by Suppressing Reactive Oxygen Species (ROS) Mediated Apoptosis by Modulating Mapks/Nrf2 Signaling in Doxorubicin-Induced Cardiotoxicity. Med Sci Monit 2017; 23:5283-5293. [PMID: 29107939 PMCID: PMC5687120 DOI: 10.12659/msm.907449] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Background Anthracyclines-induced cardiotoxicity has become one of the major restrictions of their clinical applications. Klotho showed cardioprotective effects. This study aimed to investigate the effects and possible mechanisms of klotho on doxorubicin (DOX)-induced cardiotoxicity. Material/Methods Rats and isolated myocytes were exposed to DOX and treated with exogenous klotho. Specific inhibitors and siRNAs silencing MAPKs were also used to treat the animals and/or myocytes. An invasive hemodynamic method was used to determine cardiac functions. Intracellular ROS generation was evaluated by DHE staining. Western blotting was used to determine the phosphorylation levels of JNK, ERK, and p38 MAPKs in plasma extracts and Nrf2 in nuclear extracts. Nuclear translocation of Nrf2 in myocytes was evaluated by immunohistochemistry. Cell apoptosis was evaluated by TUNEL assay and flow cytometry. Results Klotho treatment improved DOX-induced cardiac dysfunction in rats. The DOX-induced ROS accumulation and cardiac apoptosis were attenuated by klotho. Impaired phosphorylations of MAPKs, Nrf2 translocation and expression levels of HO1 and Prx1 were also attenuated by klotho treatment. However, the anti-oxidant and anti-apoptotic effects of klotho on DOX-exposed myocardium and myocytes were impaired by both specific inhibitors and siRNAs against MAPKs. Moreover, the recovery effects of klotho on phosphorylations of MAPKs, Nrf2 translocation and expression levels of HO1 and Prx1 were also impaired by specific inhibitors and siRNAs against MAPKs. Conclusions By recovering the activation of MAPKs signaling, klotho improved cardiac function loss which was triggered by DOX-induced ROS mediated cardiac apoptosis.
Collapse
Affiliation(s)
- Huolan Zhu
- Department of Cardiology, Shaanxi Provincial People's Hospital, Xi'an, Shaanxi, China (mainland)
| | - Yan Gao
- ECG Exam Room, Function Testing Lab, Shaanxi Provincial People's Hospital, Xi'an, Shaanxi, China (mainland)
| | - Shunming Zhu
- Department of Cardiology, Shaanxi Provincial People's Hospital, Xi'an, Shaanxi, China (mainland)
| | - Qianwei Cui
- Department of Cardiology, Shaanxi Provincial People's Hospital, Xi'an, Shaanxi, China (mainland)
| | - Jie Du
- Health Examination Center, Shaanxi Provincial People's Hospital, Xi'an, Shaanxi, China (mainland)
| |
Collapse
|
21
|
Gorr MW, Falvo MJ, Wold LE. Air Pollution and Other Environmental Modulators of Cardiac Function. Compr Physiol 2017; 7:1479-1495. [PMID: 28915333 PMCID: PMC7249238 DOI: 10.1002/cphy.c170017] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Cardiovascular disease (CVD) is the leading cause of death in developed regions and a worldwide health concern. Multiple external causes of CVD are well known, including obesity, diabetes, hyperlipidemia, age, and sedentary behavior. Air pollution has been linked with the development of CVD for decades, though the mechanistic characterization remains unknown. In this comprehensive review, we detail the background and epidemiology of the effects of air pollution and other environmental modulators on the heart, including both short- and long-term consequences. Then, we provide the experimental data and current hypotheses of how pollution is able to cause the CVD, and how exposure to pollutants is exacerbated in sensitive states. Published 2017. Compr Physiol 7:1479-1495, 2017.
Collapse
Affiliation(s)
- Matthew W. Gorr
- Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University Wexner College of Medicine, Columbus, Ohio, USA
- College of Nursing, The Ohio State University, Columbus, Ohio, USA
| | - Michael J. Falvo
- War Related Illness and Injury Study Center, Department of Veterans Affairs, New Jersey Health Care System, East Orange, New Jersey, USA
- New Jersey Medical School, Rutgers Biomedical and Health Sciences, Newark, New Jersey, USA
| | - Loren E. Wold
- Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University Wexner College of Medicine, Columbus, Ohio, USA
- College of Nursing, The Ohio State University, Columbus, Ohio, USA
- Department of Physiology and Cell Biology, The Ohio State University, Columbus, Ohio, USA
| |
Collapse
|
22
|
Goodson JM, Weldy CS, MacDonald JW, Liu Y, Bammler TK, Chien WM, Chin MT. In utero exposure to diesel exhaust particulates is associated with an altered cardiac transcriptional response to transverse aortic constriction and altered DNA methylation. FASEB J 2017; 31:4935-4945. [PMID: 28751527 DOI: 10.1096/fj.201700032r] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2017] [Accepted: 07/10/2017] [Indexed: 12/13/2022]
Abstract
In utero exposure to diesel exhaust air pollution has been associated with increased adult susceptibility to heart failure in mice, but the mechanisms by which this exposure promotes susceptibility to heart failure are poorly understood. To identify the potential transcriptional effects that mediate this susceptibility, we have performed RNA sequencing analysis on adult hearts from mice that were exposed to diesel exhaust in utero and that have subsequently undergone transverse aortic constriction. We identified 3 target genes, Mir133a-2, Ptprf, and Pamr1, which demonstrate dysregulation after exposure and aortic constriction. Examination of expression patterns in human heart tissues indicates a correlation between expression and heart failure. We subsequently assessed DNA methylation modifications at these candidate loci in neonatal cultured cardiac myocytes after in utero exposure to diesel exhaust and found that the promoter for Mir133a-2 is differentially methylated. These target genes in the heart are the first genes to be identified that likely play an important role in mediating adult sensitivity to heart failure. We have also shown a change in DNA methylation within cardiomyocytes as a result of in utero exposure to diesel exhaust.-Goodson, J. M., Weldy, C. S., MacDonald, J. W., Liu, Y., Bammler, T. K., Chien, W.-M., Chin, M. T. In utero exposure to diesel exhaust particulates is associated with an altered cardiac transcriptional response to transverse aortic constriction and altered DNA methylation.
Collapse
Affiliation(s)
- Jamie M Goodson
- Department of Pathology, University of Washington School of Medicine, University of Washington, Seattle, Washington, USA
| | - Chad S Weldy
- Department of Pathology, University of Washington School of Medicine, University of Washington, Seattle, Washington, USA.,Division of Cardiology, Department of Medicine, University of Washington School of Medicine, University of Washington, Seattle, Washington, USA
| | - James W MacDonald
- Department of Environmental and Occupational Health Sciences, University of Washington School of Public Health, University of Washington, Seattle, Washington, USA
| | - Yonggang Liu
- Division of Cardiology, Department of Medicine, University of Washington School of Medicine, University of Washington, Seattle, Washington, USA
| | - Theo K Bammler
- Department of Environmental and Occupational Health Sciences, University of Washington School of Public Health, University of Washington, Seattle, Washington, USA
| | - Wei-Ming Chien
- Division of Cardiology, Department of Medicine, University of Washington School of Medicine, University of Washington, Seattle, Washington, USA
| | - Michael T Chin
- Department of Pathology, University of Washington School of Medicine, University of Washington, Seattle, Washington, USA .,Division of Cardiology, Department of Medicine, University of Washington School of Medicine, University of Washington, Seattle, Washington, USA
| |
Collapse
|
23
|
Jiang P, Zhang D, Qiu H, Yi X, Zhang Y, Cao Y, Zhao B, Xia Z, Wang C. Tiron ameliorates high glucose-induced cardiac myocyte apoptosis by PKCδ-dependent inhibition of osteopontin. Clin Exp Pharmacol Physiol 2017; 44:760-770. [PMID: 28394420 DOI: 10.1111/1440-1681.12762] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2016] [Revised: 03/17/2017] [Accepted: 03/31/2017] [Indexed: 12/25/2022]
Affiliation(s)
- Ping Jiang
- Department of Cardiovascular Medicine; The People's Hospital of Gongan County; Gongan China
- Department of Pathology & Pathophysiology; Wuhan University School of Basic Medical Sciences; Wuhan China
| | - Deling Zhang
- Department of Pathology & Pathophysiology; Wuhan University School of Basic Medical Sciences; Wuhan China
| | - Hong Qiu
- Department of Laboratory; Dongfeng General Hospital of Hubei Medical University; Shiyan China
| | - Xianqi Yi
- Department of Cardiovascular Medicine; The People's Hospital of Gongan County; Gongan China
- Department of Pathology & Pathophysiology; Wuhan University School of Basic Medical Sciences; Wuhan China
| | - Yemin Zhang
- Department of Pathology & Pathophysiology; Wuhan University School of Basic Medical Sciences; Wuhan China
| | - Yingkang Cao
- Department of Pathology & Pathophysiology; Wuhan University School of Basic Medical Sciences; Wuhan China
| | - Bo Zhao
- Department of Anesthesiology; Wuhan University Renmin Hospital; Wuhan China
| | - Zhongyuan Xia
- Department of Anesthesiology; Wuhan University Renmin Hospital; Wuhan China
| | - Changhua Wang
- Department of Pathology & Pathophysiology; Wuhan University School of Basic Medical Sciences; Wuhan China
| |
Collapse
|
24
|
Simon F, Tapia P, Armisen R, Echeverria C, Gatica S, Vallejos A, Pacheco A, Sanhueza ME, Alvo M, Segovia E, Torres R. Human Peritoneal Mesothelial Cell Death Induced by High-Glucose Hypertonic Solution Involves Ca 2+ and Na + Ions and Oxidative Stress with the Participation of PKC/NOX2 and PI3K/Akt Pathways. Front Physiol 2017; 8:379. [PMID: 28659813 PMCID: PMC5468383 DOI: 10.3389/fphys.2017.00379] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2017] [Accepted: 05/22/2017] [Indexed: 01/07/2023] Open
Abstract
Chronic peritoneal dialysis (PD) therapy is equally efficient as hemodialysis while providing greater patient comfort and mobility. Therefore, PD is the treatment of choice for several types of renal patients. During PD, a high-glucose hyperosmotic (HGH) solution is administered into the peritoneal cavity to generate an osmotic gradient that promotes water and solutes transport from peritoneal blood to the dialysis solution. Unfortunately, PD has been associated with a loss of peritoneal viability and function through the generation of a severe inflammatory state that induces human peritoneal mesothelial cell (HPMC) death. Despite this deleterious effect, the precise molecular mechanism of HPMC death as induced by HGH solutions is far from being understood. Therefore, the aim of this study was to explore the pathways involved in HGH solution-induced HPMC death. HGH-induced HPMC death included influxes of intracellular Ca2+ and Na+. Furthermore, HGH-induced HPMC death was inhibited by antioxidant and reducing agents. In line with this, HPMC death was induced solely by increased oxidative stress. In addition to this, the cPKC/NOX2 and PI3K/Akt intracellular signaling pathways also participated in HGH-induced HPMC death. The participation of PI3K/Akt intracellular is in agreement with previously shown in rat PMC apoptosis. These findings contribute toward fully elucidating the underlying molecular mechanism mediating peritoneal mesothelial cell death induced by high-glucose solutions during peritoneal dialysis.
Collapse
Affiliation(s)
- Felipe Simon
- Departamento de Ciencias Biologicas, Facultad de Ciencias Biologicas and Facultad de Medicina, Universidad Andres BelloSantiago, Chile.,Millennium Institute on Immunology and ImmunotherapySantiago, Chile
| | - Pablo Tapia
- Unidad de Paciente Critico, Hospital Clínico Metropolitano de La FloridaSantiago, Chile
| | - Ricardo Armisen
- Centro de Investigación y Tratamiento del Cancer, Facultad de Medicina, Universidad de ChileSantiago, Chile.,Center for Excellence in Precision Medicine Pfizer, Pfizer ChileSantiago, Chile
| | - Cesar Echeverria
- Centro Integrativo de Biología y Química Aplicada, Universidad Bernardo OHigginsSantiago, Chile
| | - Sebastian Gatica
- Departamento de Ciencias Biologicas, Facultad de Ciencias Biologicas and Facultad de Medicina, Universidad Andres BelloSantiago, Chile
| | - Alejandro Vallejos
- Departamento de Ciencias Biologicas, Facultad de Ciencias Biologicas and Facultad de Medicina, Universidad Andres BelloSantiago, Chile
| | - Alejandro Pacheco
- Sección de Nefrología, Departamento de Medicina, Hospital Clínico Universidad de ChileSantiago, Chile
| | - Maria E Sanhueza
- Sección de Nefrología, Departamento de Medicina, Hospital Clínico Universidad de ChileSantiago, Chile
| | - Miriam Alvo
- Sección de Nefrología, Departamento de Medicina, Hospital Clínico Universidad de ChileSantiago, Chile
| | - Erico Segovia
- Centro Integrativo de Biología y Química Aplicada, Universidad Bernardo OHigginsSantiago, Chile
| | - Rubén Torres
- Sección de Nefrología, Departamento de Medicina, Hospital Clínico Universidad de ChileSantiago, Chile.,Facultad de Medicina, Instituto de Ciencias Biomédicas, Universidad de ChileSantiago, Chile
| |
Collapse
|
25
|
Yang L, Wang WC, Lung SCC, Sun Z, Chen C, Chen JK, Zou Q, Lin YH, Lin CH. Polycyclic aromatic hydrocarbons are associated with increased risk of chronic obstructive pulmonary disease during haze events in China. THE SCIENCE OF THE TOTAL ENVIRONMENT 2017; 574:1649-1658. [PMID: 27614859 DOI: 10.1016/j.scitotenv.2016.08.211] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/04/2016] [Revised: 07/28/2016] [Accepted: 08/31/2016] [Indexed: 06/06/2023]
Abstract
Although exposure to particulate matter with a diameter of <2.5μm (PM2.5) is associated with chronic obstructive pulmonary disease (COPD), the major components of PM2.5 in COPD pathogenesis are controversial. Here we employed the human lung epithelial cell line BEAS-2B to elucidate the association between COPD and the organic and water-soluble components of PM2.5. We found that the PM2.5 organic extract was a potential major risk factor for pulmonary epithelial barrier dysfunction through the depletion of proteins from the zonula occludens. This extract induced severe oxidative stress that increased DNA damage and the production of proinflammatory cytokines by BEAS-2B cells as well as decreased α1-antitrypsin expression, suggesting a mechanism that increases the risk of COPD. These effects were mainly mediated by polycyclic aromatic hydrocarbons (PAHs) through the aryl hydrocarbon receptor pathway. PAHs with high benzo(a)pyrene (BaP)-equivalent concentrations, but not major PAH components, have an increased risk of causing COPD, suggesting that BaP-equivalent concentrations represent a PM2.5-induced COPD risk metric, which may contribute to provide a rationale for the remediation of air pollution.
Collapse
Affiliation(s)
- Lingyan Yang
- Key Laboratory of Nano-Bio Interface, Division of Nanobiomedicine, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou 215123, China
| | - Wen-Cheng Wang
- Research Center for Environmental Changes, Academia Sinica, Taipei 11529, Taiwan
| | | | - Zhelin Sun
- Key Laboratory of Nano-Bio Interface, Division of Nanobiomedicine, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou 215123, China
| | - Chongjun Chen
- School of Environmental Science and Engineering, Suzhou University of Science and Technology, Suzhou 215009, China
| | - Jen-Kun Chen
- Institute of Biomedical Engineering & Nanomedicine, National Health Research Institutes, Miaoli 35053, Taiwan
| | - Qiang Zou
- Suzhou Environmental Monitor Center, Suzhou 215004, China
| | - Yu-Hsin Lin
- Department of Food and Beverage Management, Taipei College of Maritime Technology, Taipei 11174, Taiwan
| | - Chia-Hua Lin
- Department of Biotechnology, National Formosa University, Yunlin 63208, Taiwan.
| |
Collapse
|
26
|
He F, Li J, Liu Z, Chuang CC, Yang W, Zuo L. Redox Mechanism of Reactive Oxygen Species in Exercise. Front Physiol 2016; 7:486. [PMID: 27872595 PMCID: PMC5097959 DOI: 10.3389/fphys.2016.00486] [Citation(s) in RCA: 224] [Impact Index Per Article: 24.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2016] [Accepted: 10/10/2016] [Indexed: 01/04/2023] Open
Abstract
It is well known that regular exercise can benefit health by enhancing antioxidant defenses in the body. However, unaccustomed and/or exhaustive exercise can generate excessive reactive oxygen species (ROS), leading to oxidative stress-related tissue damages and impaired muscle contractility. ROS are produced in both aerobic and anaerobic exercise. Mitochondria, NADPH oxidases and xanthine oxidases have all been identified as potential contributors to ROS production, yet the exact redox mechanisms underlying exercise-induced oxidative stress remain elusive. Interestingly, moderate exposure to ROS is necessary to induce body's adaptive responses such as the activation of antioxidant defense mechanisms. Dietary antioxidant manipulation can also reduce ROS levels and muscle fatigue, as well as enhance exercise recovery. To elucidate the complex role of ROS in exercise, this review updates on new findings of ROS origins within skeletal muscles associated with various types of exercises such as endurance, sprint and mountain climbing. In addition, we will examine the corresponding antioxidant defense systems as well as dietary manipulation against damages caused by ROS.
Collapse
Affiliation(s)
- Feng He
- Department of Kinesiology, California State University-Chico Chico, CA, USA
| | - Juan Li
- Department of Physical Education, Anhui University Anhui, China
| | - Zewen Liu
- Affiliated Ezhou Central Hospital at Medical School of Wuhan UniversityHubei, China; Radiologic Sciences and Respiratory Therapy Division, School of Health and Rehabilitation Sciences, The Ohio State University College of MedicineColumbus, OH, USA
| | - Chia-Chen Chuang
- Radiologic Sciences and Respiratory Therapy Division, School of Health and Rehabilitation Sciences, The Ohio State University College of MedicineColumbus, OH, USA; Interdisciplinary Biophysics Graduate Program, The Ohio State UniversityColumbus, OH, USA
| | - Wenge Yang
- Department of Physical Education, China University of Geosciences Beijing, China
| | - Li Zuo
- Radiologic Sciences and Respiratory Therapy Division, School of Health and Rehabilitation Sciences, The Ohio State University College of MedicineColumbus, OH, USA; Interdisciplinary Biophysics Graduate Program, The Ohio State UniversityColumbus, OH, USA
| |
Collapse
|
27
|
Zuo L, He F, Tinsley GM, Pannell BK, Ward E, Arciero PJ. Comparison of High-Protein, Intermittent Fasting Low-Calorie Diet and Heart Healthy Diet for Vascular Health of the Obese. Front Physiol 2016; 7:350. [PMID: 27621707 PMCID: PMC5002412 DOI: 10.3389/fphys.2016.00350] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2016] [Accepted: 08/02/2016] [Indexed: 12/14/2022] Open
Abstract
AIM It has been debated whether different diets are more or less effective in long-term weight loss success and cardiovascular disease prevention among men and women. To further explore these questions, the present study evaluated the combined effects of a high-protein, intermittent fasting, low-calorie diet plan compared with a heart healthy diet plan during weight loss, and weight loss maintenance on blood lipids and vascular compliance of obese individuals. METHODS The experiment involved 40 obese adults (men, n = 21; women, n = 19) and was divided into two phases: (a) 12-week high-protein, intermittent fasting, low-calorie weight loss diet comparing men and women (Phase 1) and (b) a 1-year weight maintenance phase comparing high-protein, intermittent fasting with a heart healthy diet (Phase 2). Body weight, body mass index (BMI), blood lipids, and arterial compliance outcomes were assessed at weeks 1 (baseline control), 12 (weight loss), and 64 (12 + 52 week; weight loss maintenance). RESULTS At the end of weight loss intervention, concomitant reductions in body weight, BMI and blood lipids were observed, as well as enhanced arterial compliance. No sex-specific differences in responses were observed. During phase 2, the high-protein, intermittent fasting group demonstrated a trend for less regain in BMI, low-density lipoprotein (LDL), and aortic pulse wave velocity than the heart healthy group. CONCLUSION Our results suggest that a high-protein, intermittent fasting and low-calorie diet is associated with similar reductions in BMI and blood lipids in obese men and women. This diet also demonstrated an advantage in minimizing weight regain as well as enhancing arterial compliance as compared to a heart healthy diet after 1 year.
Collapse
Affiliation(s)
- Li Zuo
- Radiologic Sciences and Respiratory Therapy Division, School of Health and Rehabilitation Sciences, The Ohio State University College of Medicine, The Ohio State University Wexner Medical CenterColumbus, OH, USA
| | - Feng He
- Department of Kinesiology, California State University, ChicoChico, CA, USA
- Human Nutrition and Metabolism Laboratory, Health and Exercise Sciences Department, Skidmore CollegeSaratoga Springs, NY, USA
| | - Grant M. Tinsley
- Department of Kinesiology and Sport Management, Texas Tech UniversityLubbock, TX, USA
| | - Benjamin K. Pannell
- Radiologic Sciences and Respiratory Therapy Division, School of Health and Rehabilitation Sciences, The Ohio State University College of Medicine, The Ohio State University Wexner Medical CenterColumbus, OH, USA
| | - Emery Ward
- Human Nutrition and Metabolism Laboratory, Health and Exercise Sciences Department, Skidmore CollegeSaratoga Springs, NY, USA
| | - Paul J. Arciero
- Human Nutrition and Metabolism Laboratory, Health and Exercise Sciences Department, Skidmore CollegeSaratoga Springs, NY, USA
| |
Collapse
|
28
|
Ma L, Chuang CC, Weng W, Zhao L, Zheng Y, Zhang J, Zuo L. Paeonol Protects Rat Heart by Improving Regional Blood Perfusion during No-Reflow. Front Physiol 2016; 7:298. [PMID: 27493631 PMCID: PMC4954854 DOI: 10.3389/fphys.2016.00298] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2016] [Accepted: 06/28/2016] [Indexed: 01/22/2023] Open
Abstract
No-reflow phenomenon, defined as inadequate perfusion of myocardium without evident artery obstruction, occurs at a high incidence after coronary revascularization. The mechanisms underlying no-reflow is only partially understood. It is commonly caused by the swelling of endothelial cells, neutrophil accumulation, and vasoconstriction, which are all related to acute inflammation. Persistent no-reflow can lead to hospitalization and mortality. However, an effective preventive intervention has not yet been established. We have previously found that paeonol, an active extraction from the root of Paeonia suffruticosa, can benefit the heart function by inhibiting tissue damage after ischemia, reducing inflammation, and inducing vasodilatation. To further investigate the potential cardioprotective action of paeonol on no-reflow, healthy male Wistar rats were randomly divided into four groups: sham, ischemia-reperfusion (I/R) injury (left anterior descending coronary artery was ligated for 4 h followed by reperfusion for 8 h), and I/R injury pretreated with paeonol at two different doses. Real-time myocardial contrast echocardiography was used to monitor regional blood perfusion and cardiac functions. Our data indicated that paeonol treatment significantly reduces myocardial infarct area and no-reflow area (n = 8; p < 0.05). Regional myocardial perfusion (A·β) and cardiac functions such as ejection fraction, stroke volume, and fractional shortening were elevated by paeonol (n = 8; p < 0.05). Paeonol also lowered the serum levels of lactate dehydrogenase, creatine kinase, cardiac troponin T, and C-reactive protein, as indices of myocardial injury. Paeonol exerts beneficial effects on attenuating I/R-associated no-reflow injuries, and may be considered as a potential preventive treatment for cardiac diseases or post-coronary revascularization in which no-reflow often occurs.
Collapse
Affiliation(s)
- Lina Ma
- Graduate School, Beijing University of Chinese MedicineBeijing, China; Institute of Basic Medical Sciences, Xiyuan Hospital, China Academy of Chinese Medical SciencesBeijing, China
| | - Chia-Chen Chuang
- Radiologic Sciences and Respiratory Therapy Division, School of Health and Rehabilitation Sciences, The Ohio State University College of MedicineColumbus, OH, USA; Interdisciplinary Biophysics Graduate Program, The Ohio State UniversityColumbus, OH, USA
| | - Weiliang Weng
- Institute of Basic Medical Sciences, Xiyuan Hospital, China Academy of Chinese Medical Sciences Beijing, China
| | - Le Zhao
- Institute of Basic Medical Sciences, Xiyuan Hospital, China Academy of Chinese Medical Sciences Beijing, China
| | - Yongqiu Zheng
- Institute of Basic Medical Sciences, Xiyuan Hospital, China Academy of Chinese Medical Sciences Beijing, China
| | - Jinyan Zhang
- Institute of Basic Medical Sciences, Xiyuan Hospital, China Academy of Chinese Medical Sciences Beijing, China
| | - Li Zuo
- Radiologic Sciences and Respiratory Therapy Division, School of Health and Rehabilitation Sciences, The Ohio State University College of MedicineColumbus, OH, USA; Interdisciplinary Biophysics Graduate Program, The Ohio State UniversityColumbus, OH, USA
| |
Collapse
|
29
|
da Cunha AA, Nuñez NK, de Souza RG, Vargas MHM, Silveira JS, Antunes GL, Schmitz F, de Souza Wyse AT, Jones MH, Pitrez PM. Recombinant human deoxyribonuclease attenuates oxidative stress in a model of eosinophilic pulmonary response in mice. Mol Cell Biochem 2016; 413:47-55. [PMID: 26738487 DOI: 10.1007/s11010-015-2638-1] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2015] [Accepted: 12/23/2015] [Indexed: 12/14/2022]
Abstract
The inflammatory cells infiltrating the airways produce several mediators, such as reactive oxygen species (ROS). ROS and the oxidant-antioxidant imbalance might play an important role in the modulation of airways inflammation. In order to avoid the undesirable effects of ROS, various endogenous antioxidant strategies have evolved, incorporating both enzymatic and non-enzymatic mechanisms. Recombinant human deoxyribonuclease (rhDNase) in clinical studies demonstrated a reduction in sputum viscosity, cleaving extracellular DNA in the airways, and facilitating mucus clearance, but an antioxidant effect was not studied so far. Therefore, we evaluated whether the administration of rhDNase improves oxidative stress in a murine model of asthma. Mice were sensitized by two subcutaneous injections of ovalbumin (OVA), on days 0 and 7, followed by three lung challenges with OVA on days 14, 15, and 16. On days 15 and 16, after 2 h of the challenge with OVA, mice received 1 mg/mL of rhDNase in the lungs. Bronchoalveolar lavage fluid and lung tissue were obtained on day 17, for inflammatory and oxidative stress analysis. We showed that rhDNase did not alter the population of inflammatory cells, such as eosinophil cells, in OVA-treated rhDNase group but significantly improved oxidative stress in lung tissue, by decreasing oxygen reactive species and increasing superoxide dismutase/catalase ratio, glutathione peroxidase activity, and thiol content. Our data provide the first evidence that rhDNase decreases some measures of oxidative stress and antioxidant status in a murine model of asthma, with a potential antioxidant effect to be further studied in human asthma.
Collapse
Affiliation(s)
- Aline Andrea da Cunha
- Laboratory of Pediatric Respirology, Infant Center, Institute of Biomedical Research, Pontifícia Universidade Católica do Rio Grande do Sul, 6690 Ipiranga Avenue, 2nd Floor, Room 13, Porto Alegre, RS, 90610-000, Brazil.
| | - Nailê Karine Nuñez
- Laboratory of Pediatric Respirology, Infant Center, Institute of Biomedical Research, Pontifícia Universidade Católica do Rio Grande do Sul, 6690 Ipiranga Avenue, 2nd Floor, Room 13, Porto Alegre, RS, 90610-000, Brazil
| | - Rodrigo Godinho de Souza
- Laboratory of Pediatric Respirology, Infant Center, Institute of Biomedical Research, Pontifícia Universidade Católica do Rio Grande do Sul, 6690 Ipiranga Avenue, 2nd Floor, Room 13, Porto Alegre, RS, 90610-000, Brazil
| | - Mauro Henrique Moraes Vargas
- Laboratory of Pediatric Respirology, Infant Center, Institute of Biomedical Research, Pontifícia Universidade Católica do Rio Grande do Sul, 6690 Ipiranga Avenue, 2nd Floor, Room 13, Porto Alegre, RS, 90610-000, Brazil
| | - Josiane Silva Silveira
- Laboratory of Pediatric Respirology, Infant Center, Institute of Biomedical Research, Pontifícia Universidade Católica do Rio Grande do Sul, 6690 Ipiranga Avenue, 2nd Floor, Room 13, Porto Alegre, RS, 90610-000, Brazil
| | - Géssica Luana Antunes
- Laboratory of Pediatric Respirology, Infant Center, Institute of Biomedical Research, Pontifícia Universidade Católica do Rio Grande do Sul, 6690 Ipiranga Avenue, 2nd Floor, Room 13, Porto Alegre, RS, 90610-000, Brazil
| | - Felipe Schmitz
- Laboratory of Neuroprotection and Neurometabolic Disease, Department of Biochemistry, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Angela Terezinha de Souza Wyse
- Laboratory of Neuroprotection and Neurometabolic Disease, Department of Biochemistry, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Marcus Herbert Jones
- Laboratory of Respiratory Physiology, Infant Center, Institute of Biomedical Research, Pontifícia Universidade Católica do Rio Grande do Sul, Porto Alegre, Brazil
| | - Paulo Márcio Pitrez
- Laboratory of Pediatric Respirology, Infant Center, Institute of Biomedical Research, Pontifícia Universidade Católica do Rio Grande do Sul, 6690 Ipiranga Avenue, 2nd Floor, Room 13, Porto Alegre, RS, 90610-000, Brazil
| |
Collapse
|
30
|
He F, Zuo L. Redox Roles of Reactive Oxygen Species in Cardiovascular Diseases. Int J Mol Sci 2015; 16:27770-80. [PMID: 26610475 PMCID: PMC4661917 DOI: 10.3390/ijms161126059] [Citation(s) in RCA: 159] [Impact Index Per Article: 15.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2015] [Revised: 11/06/2015] [Accepted: 11/11/2015] [Indexed: 02/07/2023] Open
Abstract
Cardiovascular disease (CVD), a major cause of mortality in the world, has been extensively studied over the past decade. However, the exact mechanism underlying its pathogenesis has not been fully elucidated. Reactive oxygen species (ROS) play a pivotal role in the progression of CVD. Particularly, ROS are commonly engaged in developing typical characteristics of atherosclerosis, one of the dominant CVDs. This review will discuss the involvement of ROS in atherosclerosis, specifically their effect on inflammation, disturbed blood flow and arterial wall remodeling. Pharmacological interventions target ROS in order to alleviate oxidative stress and CVD symptoms, yet results are varied due to the paradoxical role of ROS in CVD. Lack of effectiveness in clinical trials suggests that understanding the exact role of ROS in the pathophysiology of CVD and developing novel treatments, such as antioxidant gene therapy and nanotechnology-related antioxidant delivery, could provide a therapeutic advance in treating CVDs. While genetic therapies focusing on specific antioxidant expression seem promising in CVD treatments, multiple technological challenges exist precluding its immediate clinical applications.
Collapse
Affiliation(s)
- Feng He
- Department of Kinesiology, California State University-Chico, Chico, CA 95929, USA.
| | - Li Zuo
- Molecular Physiology and Rehabilitation Research Lab, Radiologic Sciences and Respiratory Therapy Division, School of Health and Rehabilitation Sciences, the Ohio State University College of Medicine, Columbus, OH 43210, USA.
- Interdisciplinary Biophysics Graduate Program, the Ohio State University, Columbus, OH 43210, USA.
| |
Collapse
|
31
|
Zuo L, Pannell BK. Redox Characterization of Functioning Skeletal Muscle. Front Physiol 2015; 6:338. [PMID: 26635624 PMCID: PMC4649055 DOI: 10.3389/fphys.2015.00338] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2015] [Accepted: 11/02/2015] [Indexed: 12/17/2022] Open
Abstract
Skeletal muscle physiology is influenced by the presence of chemically reactive molecules such as reactive oxygen species (ROS). These molecules regulate multiple redox-sensitive signaling pathways that play a critical role in cellular processes including gene expression and protein modification. While ROS have gained much attention for their harmful effects in muscle fatigue and dysfunction, research has also shown ROS to facilitate muscle adaptation after stressors such as physical exercise. This manuscript aims to provide a comprehensive review of the current understanding of redox signaling in skeletal muscle. ROS-induced oxidative stress and its role in the aging process are discussed. Mitochondria have been shown to generate large amounts of ROS during muscular contractions, and thus are susceptible to oxidative stress. ROS can modify proteins located in the mitochondrial membrane leading to cell death and osmotic swelling. ROS also contribute to the necrosis and inflammation of muscle fibers that is associated with muscular diseases including Duchenne muscular dystrophy. It is imperative that future research continues to investigate the exact role of ROS in normal skeletal muscle function as well as muscular dysfunction and disease.
Collapse
Affiliation(s)
- Li Zuo
- Radiologic Sciences and Respiratory Therapy Division, School of Health and Rehabilitation Sciences, The Ohio State University College of Medicine Columbus, OH, USA ; Interdisciplinary Biophysics Graduate Program, The Ohio State University Columbus, OH, USA
| | - Benjamin K Pannell
- Radiologic Sciences and Respiratory Therapy Division, School of Health and Rehabilitation Sciences, The Ohio State University College of Medicine Columbus, OH, USA
| |
Collapse
|
32
|
Zuo L, Lucas K, Fortuna CA, Chuang CC, Best TM. Molecular Regulation of Toll-like Receptors in Asthma and COPD. Front Physiol 2015; 6:312. [PMID: 26617525 PMCID: PMC4637409 DOI: 10.3389/fphys.2015.00312] [Citation(s) in RCA: 77] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2015] [Accepted: 10/19/2015] [Indexed: 11/13/2022] Open
Abstract
Asthma and chronic obstructive pulmonary disease (COPD) have both been historically associated with significant morbidity and financial burden. These diseases can be induced by several exogenous factors, such as pathogen-associated molecular patterns (PAMPs) (e.g., allergens and microbes). Endogenous factors, including reactive oxygen species, and damage-associated molecular patterns (DAMPs) recognized by toll-like receptors (TLRs), can also result in airway inflammation. Asthma is characterized by the dominant presence of eosinophils, mast cells, and clusters of differentiation (CD)4+ T cells in the airways, while COPD typically results in the excessive formation of neutrophils, macrophages, and CD8+ T cells in the airways. In both asthma and COPD, in the respiratory tract, TLRs are the primary proteins of interest associated with the innate and adaptive immune responses; hence, multiple treatment options targeting TLRs are being explored in an effort to reduce the severity of the symptoms of these disorders. TLR-mediated pathways for both COPD and asthma have their similarities and differences with regards to cell types and the pro-inflammatory cytotoxins present in the airway. Because of the complex TLR cascade, a variety of treatments have been used to minimize airway hypersensitivity and promote bronchodilation. Although unsuccessful at completely alleviating COPD and severe asthmatic symptoms, new studies are focused on possible targets within the TLR cascade to ameliorate airway inflammation.
Collapse
Affiliation(s)
- Li Zuo
- Radiologic Sciences and Respiratory Therapy Division, The Ohio State University Wexner Medical Center, School of Health and Rehabilitation Sciences, The Ohio State University College of Medicine, Columbus, Ohio State University Columbus, OH, USA ; Interdisciplinary Biophysics Graduate Program, The Ohio State University Columbus, OH, USA
| | - Kurt Lucas
- Multiphase Chemistry Department, Max Planck Institute for Chemistry Mainz, Germany
| | - Christopher A Fortuna
- Radiologic Sciences and Respiratory Therapy Division, The Ohio State University Wexner Medical Center, School of Health and Rehabilitation Sciences, The Ohio State University College of Medicine, Columbus, Ohio State University Columbus, OH, USA
| | - Chia-Chen Chuang
- Radiologic Sciences and Respiratory Therapy Division, The Ohio State University Wexner Medical Center, School of Health and Rehabilitation Sciences, The Ohio State University College of Medicine, Columbus, Ohio State University Columbus, OH, USA ; Interdisciplinary Biophysics Graduate Program, The Ohio State University Columbus, OH, USA
| | - Thomas M Best
- Division of Sports Medicine, Department of Family Medicine, Sports Health and Performance Institute, The Ohio State University Wexner Medical Center Columbus, OH, USA
| |
Collapse
|
33
|
Ni L, Chuang CC, Zuo L. Fine particulate matter in acute exacerbation of COPD. Front Physiol 2015; 6:294. [PMID: 26557095 PMCID: PMC4617054 DOI: 10.3389/fphys.2015.00294] [Citation(s) in RCA: 115] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2015] [Accepted: 10/05/2015] [Indexed: 12/17/2022] Open
Abstract
Chronic obstructive pulmonary disease (COPD) is a common airway disorder. In particular, acute exacerbations of COPD (AECOPD) can significantly reduce pulmonary function. The majority of AECOPD episodes are attributed to infections, although environmental stress also plays a role. Increasing urbanization and associated air pollution, especially in developing countries, have been shown to contribute to COPD pathogenesis. Elevated levels of particulate matter (PM) in polluted air are strongly correlated with the onset and development of various respiratory diseases. In this review, we have conducted an extensive literature search of recent studies of the role of PM2.5 (fine PM) in AECOPD. PM2.5 leads to AECOPD via inflammation, oxidative stress (OS), immune dysfunction, and altered airway epithelial structure and microbiome. Reducing PM2.5 levels is a viable approach to lower AECOPD incidence, attenuate COPD progression and decrease the associated healthcare burden.
Collapse
Affiliation(s)
- Lei Ni
- Radiologic Sciences and Respiratory Therapy Division, School of Health and Rehabilitation Sciences, Davis Heart and Lung Research Institute, The Ohio State University College of Medicine, The Ohio State University Wexner Medical Center Columbus, OH, USA ; Department of Pulmonary Medicine, Ruijin Hospital, School of Medicine, Shanghai Jiao Tong University Shanghai, China ; Shanghai Key Laboratory of Meteorology and Health, Pudong Meteorological Service Shanghai, China
| | - Chia-Chen Chuang
- Radiologic Sciences and Respiratory Therapy Division, School of Health and Rehabilitation Sciences, Davis Heart and Lung Research Institute, The Ohio State University College of Medicine, The Ohio State University Wexner Medical Center Columbus, OH, USA ; Interdisciplinary Biophysics Program, The Ohio State University Columbus, OH, USA
| | - Li Zuo
- Radiologic Sciences and Respiratory Therapy Division, School of Health and Rehabilitation Sciences, Davis Heart and Lung Research Institute, The Ohio State University College of Medicine, The Ohio State University Wexner Medical Center Columbus, OH, USA ; Interdisciplinary Biophysics Program, The Ohio State University Columbus, OH, USA
| |
Collapse
|
34
|
Zuo L, Pannell BK, Re AT, Best TM, Wagner PD. Po2 cycling protects diaphragm function during reoxygenation via ROS, Akt, ERK, and mitochondrial channels. Am J Physiol Cell Physiol 2015; 309:C759-66. [PMID: 26423578 DOI: 10.1152/ajpcell.00174.2015] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2015] [Accepted: 09/25/2015] [Indexed: 11/22/2022]
Abstract
Po2 cycling, often referred to as intermittent hypoxia, involves exposing tissues to brief cycles of low oxygen environments immediately followed by hyperoxic conditions. After experiencing long-term hypoxia, muscle can be damaged during the subsequent reintroduction of oxygen, which leads to muscle dysfunction via reperfusion injury. The protective effect and mechanism behind Po2 cycling in skeletal muscle during reoxygenation have yet to be fully elucidated. We hypothesize that Po2 cycling effectively increases muscle fatigue resistance through reactive oxygen species (ROS), protein kinase B (Akt), extracellular signal-regulated kinase (ERK), and certain mitochondrial channels during reoxygenation. Using a dihydrofluorescein fluorescent probe, we detected the production of ROS in mouse diaphragmatic skeletal muscle in real time under confocal microscopy. Muscles treated with Po2 cycling displayed significantly attenuated ROS levels (n = 5; P < 0.001) as well as enhanced force generation compared with controls during reperfusion (n = 7; P < 0.05). We also used inhibitors for signaling molecules or membrane channels such as ROS, Akt, ERK, as well as chemical stimulators to close mitochondrial ATP-sensitive potassium channel (KATP) or open mitochondrial permeability transition pore (mPTP). All these blockers or stimulators abolished improved muscle function with Po2 cycling treatment. This current investigation has discovered a correlation between KATP and mPTP and the Po2 cycling pathway in diaphragmatic skeletal muscle. Thus we have identified a unique signaling pathway that may involve ROS, Akt, ERK, and mitochondrial channels responsible for Po2 cycling protection during reoxygenation conditions in the diaphragm.
Collapse
Affiliation(s)
- Li Zuo
- Radiologic Sciences and Respiratory Therapy Division, School of Health and Rehabilitation Sciences, The Ohio State University College of Medicine, The Ohio State University Wexner Medical Center, Columbus, Ohio; Interdisciplinary Biophysics Graduate Program, The Ohio State University, Columbus, Ohio
| | - Benjamin K Pannell
- Radiologic Sciences and Respiratory Therapy Division, School of Health and Rehabilitation Sciences, The Ohio State University College of Medicine, The Ohio State University Wexner Medical Center, Columbus, Ohio
| | - Anthony T Re
- Radiologic Sciences and Respiratory Therapy Division, School of Health and Rehabilitation Sciences, The Ohio State University College of Medicine, The Ohio State University Wexner Medical Center, Columbus, Ohio
| | - Thomas M Best
- Division of Sports Medicine, Department of Family Medicine, Sports Health and Performance Institute, The Ohio State University, Columbus, Ohio; and
| | - Peter D Wagner
- Department of Medicine, University of California, San Diego, La Jolla, California
| |
Collapse
|
35
|
Gorr MW, Youtz DJ, Eichenseer CM, Smith KE, Nelin TD, Cormet-Boyaka E, Wold LE. In vitro particulate matter exposure causes direct and lung-mediated indirect effects on cardiomyocyte function. Am J Physiol Heart Circ Physiol 2015; 309:H53-62. [PMID: 25957217 DOI: 10.1152/ajpheart.00162.2015] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/06/2015] [Accepted: 05/04/2015] [Indexed: 01/16/2023]
Abstract
Particulate matter (PM) exposure induces a pathological response from both the lungs and the cardiovascular system. PM is capable of both manifestation into the lung epithelium and entrance into the bloodstream. Therefore, PM has the capacity for both direct and lung-mediated indirect effects on the heart. In the present studies, we exposed isolated rat cardiomyocytes to ultrafine particulate matter (diesel exhaust particles, DEP) and examined their contractile function and calcium handling ability. In another set of experiments, lung epithelial cells (16HBE14o- or Calu-3) were cultured on permeable supports that allowed access to both the basal (serosal) and apical (mucosal) media; the basal media was used to culture cardiomyocytes to model the indirect, lung-mediated effects of PM on the heart. Both the direct and indirect treatments caused a reduction in contractility as evidenced by reduced percent sarcomere shortening and reduced calcium handling ability measured in field-stimulated cardiomyocytes. Treatment of cardiomyocytes with various anti-oxidants before culture with DEP was able to partially prevent the contractile dysfunction. The basal media from lung epithelial cells treated with PM contained several inflammatory cytokines, and we found that monocyte chemotactic protein-1 was a key trigger for cardiomyocyte dysfunction. These results indicate the presence of both direct and indirect effects of PM on cardiomyocyte function in vitro. Future work will focus on elucidating the mechanisms involved in these separate pathways using in vivo models of air pollution exposure.
Collapse
Affiliation(s)
- Matthew W Gorr
- Dorothy M. Davis Heart and Lung Research Institute, College of Medicine, The Ohio State University, Columbus, Ohio
| | - Dane J Youtz
- College of Nursing, The Ohio State University, Columbus, Ohio; and
| | | | - Korbin E Smith
- College of Nursing, The Ohio State University, Columbus, Ohio; and
| | - Timothy D Nelin
- Dorothy M. Davis Heart and Lung Research Institute, College of Medicine, The Ohio State University, Columbus, Ohio; College of Nursing, The Ohio State University, Columbus, Ohio; and
| | - Estelle Cormet-Boyaka
- Department of Veterinary Biosciences, College of Veterinary Medicine, The Ohio State University, Columbus, Ohio
| | - Loren E Wold
- Dorothy M. Davis Heart and Lung Research Institute, College of Medicine, The Ohio State University, Columbus, Ohio; College of Nursing, The Ohio State University, Columbus, Ohio; and
| |
Collapse
|
36
|
Zuo L, Best TM, Roberts WJ, Diaz PT, Wagner PD. Characterization of reactive oxygen species in diaphragm. Acta Physiol (Oxf) 2015; 213:700-10. [PMID: 25330121 DOI: 10.1111/apha.12410] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2014] [Revised: 07/19/2014] [Accepted: 10/16/2014] [Indexed: 12/17/2022]
Abstract
Reactive oxygen species (ROS) exist as natural mediators of metabolism to maintain cellular homeostasis. However, ROS production may significantly increase in response to environmental stressors, resulting in extensive cellular damage. Although several potential sources of increased ROS have been proposed, exact mechanisms of their generation have not been completely elucidated. This is particularly true for diaphragmatic skeletal muscle, the key muscle used for respiration. Several experimental models have focused on detection of ROS generation in rodent diaphragm tissue under stressful conditions, including hypoxia, exercise, and heat, as well as ROS formation in single myofibres. Identification methods include direct detection of ROS with confocal or fluorescent microscopy and indirect detection of ROS through end product analysis. This article explores implications of ROS generation and oxidative stress, and also evaluates potential mechanisms of cellular ROS formation in diaphragmatic skeletal muscle.
Collapse
Affiliation(s)
- L. Zuo
- Radiologic Sciences and Respiratory Therapy Division; School of Health and Rehabilitation Sciences; The Ohio State University College of Medicine; The Ohio State University Wexner Medical Center; Columbus OH USA
| | - T. M. Best
- Division of Sports Medicine; Department of Family Medicine Sports Health and Performance Institute; The Ohio State University; Columbus OH USA
| | - W. J. Roberts
- Radiologic Sciences and Respiratory Therapy Division; School of Health and Rehabilitation Sciences; The Ohio State University College of Medicine; The Ohio State University Wexner Medical Center; Columbus OH USA
| | - P. T. Diaz
- Division of Pulmonary, Allergy, Critical Care & Sleep Medicine; The Ohio State University Wexner Medical Center; Columbus OH USA
| | - P. D. Wagner
- Department of Medicine; University of California, San Diego; La Jolla CA USA
| |
Collapse
|
37
|
Liu Z, Cai H, Zhu H, Toque H, Zhao N, Qiu C, Guan G, Dang Y, Wang J. Protein kinase RNA-like endoplasmic reticulum kinase (PERK)/calcineurin signaling is a novel pathway regulating intracellular calcium accumulation which might be involved in ventricular arrhythmias in diabetic cardiomyopathy. Cell Signal 2014; 26:2591-600. [PMID: 25152364 DOI: 10.1016/j.cellsig.2014.08.015] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2014] [Accepted: 08/17/2014] [Indexed: 11/18/2022]
Abstract
We previously found that endoplasmic reticulum (ER) stress was involved in ventricular arrhythmias in diabetic cardiomyopathy. The present study was aimed to investigate the possible mechanism. In the in vivo study, diabetes cardiomyopathy (DCM) was induced by streptozotocin (STZ) injection. Hemodynamic and plasma brain natriuretic peptide (BNP) detections were used to evaluate cardiac functions; ECG was used to assess the vulnerability to arrhythmias by recording ventricular arrhythmia events (VAEs). In the in vitro study, high-glucose incubation was employed to mimic the diabetic environment of myocytes. Immunofluorescent staining was used to investigate the nuclear factor of activated T cells (NFAT) nuclear translocation and (FK506-binding protein 12.6) FKBP12.6 disassociation. [(3)H]-ryanodine binding assay was implemented to assess the channel activity of ryanodine receptor. In both in vivo and in vitro studies, activity of calcineurin was determined by colorimetric method, and western blotting was used to detect protein expression levels. In the in vivo study, we found that inhibition of both of ER stress and PERK activation decreased the VAEs in DCM rats, accompanied by reduced activity of calcineurin in myocardial tissue. In the in vitro study, in high-glucose incubated myocytes, the depletion of PERK reduced activity of calcineurin, decreased NFAT translocation and FKBP12.6 disassociation from ryanodine receptor 2 (RyR2). Furthermore, PERK deletion also reduced RyR2 channel activity and consequently impaired intracellular calcium accumulation. We concluded that PERK/calcineurin-pathway was involved in intracellular calcium regulation in myocytes in diabetic heart, which might be the mechanism inducing arrhythmias in DCM.
Collapse
Affiliation(s)
- Zhongwei Liu
- Department of Cardiology, Shaanxi Provincial People's Hospital, China
| | - Hui Cai
- Department of Anesthesiology, First Affiliated Hospital of Xi'an Jiaotong University, China
| | - Haitao Zhu
- School of Medicine, Xi'an Jiaotong University, China
| | - Haroldo Toque
- Department of Pharmacology and Toxicology, GA Regents University, USA
| | - Na Zhao
- Department of Cardiology, Shaanxi Provincial People's Hospital, China
| | - Chuan Qiu
- School of Public Health & Tropical Medicine, Tulane University, USA
| | - Gongchang Guan
- Department of Cardiology, Shaanxi Provincial People's Hospital, China
| | - Yonghui Dang
- Department of Forensic Science, Xi'an Jiaotong University School of Medicine, China.
| | - Junkui Wang
- Department of Cardiology, Shaanxi Provincial People's Hospital, China.
| |
Collapse
|
38
|
Pan Q, Xie X, Guo Y, Wang H. Simvastatin promotes cardiac microvascular endothelial cells proliferation, migration and survival by phosphorylation of p70 S6K and FoxO3a. Cell Biol Int 2014; 38:599-609. [PMID: 24375611 DOI: 10.1002/cbin.10236] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2013] [Accepted: 12/10/2013] [Indexed: 01/19/2023]
Affiliation(s)
- Qiao Pan
- Department of Cardiology; Xijing Hospital; Fourth Military Medical University; Xi'an Shaanxi 710032 China
| | - Xiaobo Xie
- Department of Disease Surveillance and Control; Centers for Diseases Control and Prevention of Guangzhou Military District; Guangzhou 510507 China
| | - Yan Guo
- Department of Oncology; State Key Discipline of Cell Biology; Xijing Hospital, Fourth Military Medical University; Xi'an Shaanxi 710032 China
| | - Haichang Wang
- Department of Cardiology; Xijing Hospital; Fourth Military Medical University; Xi'an Shaanxi 710032 China
| |
Collapse
|
39
|
Liu ZW, Zhu HT, Chen KL, Qiu C, Tang KF, Niu XL. Selenium attenuates high glucose-induced ROS/TLR-4 involved apoptosis of rat cardiomyocyte. Biol Trace Elem Res 2013; 156:262-70. [PMID: 24214856 DOI: 10.1007/s12011-013-9857-7] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/30/2013] [Accepted: 10/30/2013] [Indexed: 01/04/2023]
Abstract
The potential mechanism of high glucose-induced cardiomyocyte apoptosis and selenium's protective effects were investigated in this study. Myocytes isolated from neonate rats were cultured in high-glucose medium (25.5 mmol/L glucose) to mimic sustained hyperglycemia. Before high-glucose incubation, myocytes were pretreated by sodium selenite solution. Cell apoptosis was evaluated by annexin V/propidium iodide (PI) staining and caspase activation. Expression of Toll-like receptor 4 (TLR-4) and myeloid differentiation factor 88 (MyD-88) was examined at both mRNA and protein levels. The intracellular reactive oxygen species (ROS) production and glutathione peroxidase (GPx) activity in myocytes were also detected. We found high glucose-induced cell apoptosis and activation of TLR-4/MyD-88/caspase-8/caspase-3 signaling, accompanied by increased production of ROS. Selenium pretreatment attenuated apoptosis in high glucose-incubated myocytes, and mechanically, this protective effect was found to be associated with attenuating oxidative status by increasing activity of GPx, decreasing the generation of ROS, as well as inhibition of the activation of TLR-4/MyD-88/caspase-8/caspase-3 signaling in myocytes. These results suggest that activation of TLR-4/MyD-88 signaling pathway plays an important role in high glucose-induced cardiomyocyte apoptosis. Additionally, by modulating TLR-4/MyD-88 signaling pathway, which is linked to ROS formation, selenium exerts its antioxidative and antiapoptotic effects in high glucose-incubated myocytes.
Collapse
Affiliation(s)
- Zhong-Wei Liu
- Department of Cardiology, Second Affiliated Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi, 710004, China
| | | | | | | | | | | |
Collapse
|
40
|
Liu ZW, Zhu HT, Chen KL, Dong X, Wei J, Qiu C, Xue JH. Protein kinase RNA-like endoplasmic reticulum kinase (PERK) signaling pathway plays a major role in reactive oxygen species (ROS)-mediated endoplasmic reticulum stress-induced apoptosis in diabetic cardiomyopathy. Cardiovasc Diabetol 2013; 12:158. [PMID: 24180212 PMCID: PMC4176998 DOI: 10.1186/1475-2840-12-158] [Citation(s) in RCA: 175] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/15/2013] [Accepted: 10/29/2013] [Indexed: 12/12/2022] Open
Abstract
Background Endoplasmic reticulum (ER) stress is considered one of the mechanisms contributing to reactive oxygen species (ROS)- mediated cell apoptosis. In diabetic cardiomyopathy (DCM), cell apoptosis is generally accepted as the etiological factor and closely related to cardiac ROS generation. ER stress is proposed the link between ROS and cell apoptosis; however, the signaling pathways and their roles in participating ER stress- induced apoptosis in DCM are still unclear. Methods In this study, we investigated the signaling transductions in ROS- dependent ER stress- induced cardiomocyte apoptosis in animal model of DCM. Moreover, in order to clarify the roles of IRE1 (inositol - requiring enzyme-1), PERK (protein kinase RNA (PKR)- like ER kinase) and ATF6 (activating transcription factor-6) in conducting apoptotic signal in ROS- dependent ER stress- induced cardiomocyte apoptosis, we further investigated apoptosis in high- glucose incubated cardiomyocytes with IRE1, ATF6 and PERK- knocked down respectively. Results we demonstrated that the ER stress sensors, referred as PERK, IRE1 and ATF6, were activated in ROS- mediated ER stress- induced cell apoptosis in rat model of DCM which was characterized by cardiac pump and electrical dysfunctions. The deletion of PERK in myocytes exhibited stronger protective effect against apoptosis induced by high- glucose incubation than deletion of ATF6 or IRE in the same myocytes. By subcellular fractionation, rather than ATF6 and IRE1, in primary cardiomyocytes, PERK was found a component of MAMs (mitochondria-associated endoplasmic reticulum membranes) which was the functional and physical contact site between ER and mitochondria. Conclusions ROS- stimulated activation of PERK signaling pathway takes the major responsibility rather than IRE1 or ATF6 signaling pathways in ROS- medicated ER stress- induced myocyte apoptosis in DCM.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Jia-Hong Xue
- Department of Cardiology, Second Affiliated Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi, China.
| |
Collapse
|
41
|
Zhu X, Zuo L. Characterization of oxygen radical formation mechanism at early cardiac ischemia. Cell Death Dis 2013; 4:e787. [PMID: 24008731 PMCID: PMC3789172 DOI: 10.1038/cddis.2013.313] [Citation(s) in RCA: 55] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2013] [Revised: 06/12/2013] [Accepted: 07/03/2013] [Indexed: 12/31/2022]
Abstract
Myocardial ischemia–reperfusion (I/R) causes severe cardiac damage. Although the primary function of oxymyoglobin (Mb) has been considered to be cellular O2 storage and supply, previous research has suggested that Mb is a potentially protective element against I/R injury. However, the mechanism of its protective action is still largely unknown. With a real-time fluorescent technique, we observed that at the onset of ischemia, there was a small burst of superoxide (O2•–) release, as visualized in an isolated rat heart. Thus, we hypothesize that the formation of O2•– correlates to Mb due to a decrease in oxygen tension in the myocardium. Measurement of O2•– production in a Langendorff apparatus was performed using surface fluorometry. An increase in fluorescence was observed during the onset of ischemia in hearts perfused with a solution of hydroethidine, a fluorescent dye sensitive to intracellular O2•–. The increase of fluorescence in the ischemic heart was abolished by a superoxide dismutase mimic, carbon monoxide, or by Mb-knockout gene technology. Furthermore, we identified that O2•– was not generated from the intracellular endothelium but from the myocytes, which are a rich source of Mb. These results suggest that during the onset of ischemia, Mb is responsible for generating O2•–. This novel mechanism may shed light on the protective role of Mb in I/R injury.
Collapse
Affiliation(s)
- X Zhu
- Department of Pulmonary Medicine, Ruijin Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | | |
Collapse
|
42
|
Molecular mechanisms of reactive oxygen species-related pulmonary inflammation and asthma. Mol Immunol 2013; 56:57-63. [PMID: 23665383 DOI: 10.1016/j.molimm.2013.04.002] [Citation(s) in RCA: 123] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2013] [Accepted: 04/07/2013] [Indexed: 02/07/2023]
Abstract
Asthma is a highly relevant disorder that can be induced by many environmental factors such as allergens and pollutants. One of the most critical pathological symptoms of asthma is airway inflammation. In order to identify a cause of respiratory inflammation, we thoroughly examine the unique role of reactive oxygen species (ROS). Evidence supports that the inhalation of aggravating compounds such as allergens can promote the increased generation of ROS. Accordingly, ROS have a proven role in the cellular signaling cascades of many respiratory diseases that cause respiratory inflammation, including asthma. Although there is no known cure for asthma, current treatments effectively lessen the inflammation symptom. Based on the investigations of asthma pathogenesis and the mechanism of ROS formation, we have identified several novel anti-inflammatory therapeutic treatments, shedding light on a fundamental understanding for the cure of this disorder. In this review, we will outline the pathogenesis of asthma and its relationship to ROS, oxidative stress, and pulmonary inflammation.
Collapse
|
43
|
Wang D, Luo P, Wang Y, Li W, Wang C, Sun D, Zhang R, Su T, Ma X, Zeng C, Wang H, Ren J, Cao F. Glucagon-like peptide-1 protects against cardiac microvascular injury in diabetes via a cAMP/PKA/Rho-dependent mechanism. Diabetes 2013; 62:1697-708. [PMID: 23364453 PMCID: PMC3636622 DOI: 10.2337/db12-1025] [Citation(s) in RCA: 138] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Impaired cardiac microvascular function contributes to cardiovascular complications in diabetes. Glucagon-like peptide-1 (GLP-1) exhibits potential cardioprotective properties in addition to its glucose-lowering effect. This study was designed to evaluate the impact of GLP-1 on cardiac microvascular injury in diabetes and the underlying mechanism involved. Experimental diabetes was induced using streptozotocin in rats. Cohorts of diabetic rats received a 12-week treatment of vildagliptin (dipeptidyl peptidase-4 inhibitor) or exenatide (GLP-1 analog). Experimental diabetes attenuated cardiac function, glucose uptake, and microvascular barrier function, which were significantly improved by vildagliptin or exenatide treatment. Cardiac microvascular endothelial cells (CMECs) were isolated and cultured in normal or high glucose medium with or without GLP-1. GLP-1 decreased high-glucose-induced reactive oxygen species production and apoptotic index, as well as the levels of NADPH oxidase such as p47(phox) and gp91(phox). Furthermore, cAMP/PKA (cAMP-dependent protein kinase activity) was increased and Rho-expression was decreased in high-glucose-induced CMECs after GLP-1 treatment. In conclusion, GLP-1 could protect the cardiac microvessels against oxidative stress, apoptosis, and the resultant microvascular barrier dysfunction in diabetes, which may contribute to the improvement of cardiac function and cardiac glucose metabolism in diabetes. The protective effects of GLP-1 are dependent on downstream inhibition of Rho through a cAMP/PKA-mediated pathway.
Collapse
Affiliation(s)
- Dongjuan Wang
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Peng Luo
- Department of Cardiovascular Surgery, Xijing Hospital, Fourth Military Medical University, Xi’an, Shaanxi, China
| | - Yabin Wang
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Weijie Li
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Chen Wang
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Dongdong Sun
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Rongqing Zhang
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Tao Su
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Xiaowei Ma
- Department of Nuclear Medicine, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Chao Zeng
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Haichang Wang
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Jun Ren
- Center for Cardiovascular Research and Alternative Medicine, University of Wyoming College of Health Sciences, Laramie, Wyoming
| | - Feng Cao
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, China
- Corresponding author: Feng Cao, , or Jun Ren, , or Haichang Wang,
| |
Collapse
|
44
|
The influence of Nrf2 on cardiac responses to environmental stressors. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2013; 2013:901239. [PMID: 23738044 PMCID: PMC3655674 DOI: 10.1155/2013/901239] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/11/2013] [Accepted: 03/26/2013] [Indexed: 01/01/2023]
Abstract
Nrf2 protects the lung from adverse responses to oxidants, including 100% oxygen (hyperoxia) and airborne pollutants like particulate matter (PM) exposure, but the role of Nrf2 on heart rate (HR) and heart rate variability (HRV) responses is not known. We hypothesized that genetic disruption of Nrf2 would exacerbate murine HR and HRV responses to severe hyperoxia or moderate PM exposures. Nrf2−/− and Nrf2+/+ mice were instrumented for continuous ECG recording to calculate HR and HRV (low frequency (LF), high frequency (HF), and total power (TP)). Mice were then either exposed to hyperoxia for up to 72 hrs or aspirated with ultrafine PM (UF-PM). Compared to respective controls, UF-PM induced significantly greater effects on HR (P < 0.001) and HF HRV (P < 0.001) in Nrf2−/− mice compared to Nrf2+/+ mice. Nrf2−/− mice tolerated hyperoxia significantly less than Nrf2+/+ mice (~22 hrs; P < 0.001). Reductions in HR, LF, HF, and TP HRV were also significantly greater in Nrf2−/− compared to Nrf2+/+ mice (P < 0.01). Results demonstrate that Nrf2 deletion increases susceptibility to change in HR and HRV responses to environmental stressors and suggest potential therapeutic strategies to prevent cardiovascular alterations.
Collapse
|
45
|
Zuo L, Shiah A, Roberts WJ, Chien MT, Wagner PD, Hogan MC. Low Po₂ conditions induce reactive oxygen species formation during contractions in single skeletal muscle fibers. Am J Physiol Regul Integr Comp Physiol 2013; 304:R1009-16. [PMID: 23576612 DOI: 10.1152/ajpregu.00563.2012] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Contractions in whole skeletal muscle during hypoxia are known to generate reactive oxygen species (ROS); however, identification of real-time ROS formation within isolated single skeletal muscle fibers has been challenging. Consequently, there is no convincing evidence showing increased ROS production in intact contracting fibers under low Po₂ conditions. Therefore, we hypothesized that intracellular ROS generation in single contracting skeletal myofibers increases during low Po₂ compared with a value approximating normal resting Po₂. Dihydrofluorescein was loaded into single frog (Xenopus) fibers, and fluorescence was used to monitor ROS using confocal microscopy. Myofibers were exposed to two maximal tetanic contractile periods (1 contraction/3 s for 2 min, separated by a 60-min rest period), each consisting of one of the following treatments: high Po₂ (30 Torr), low Po₂ (3-5 Torr), high Po₂ with ebselen (antioxidant), or low Po₂ with ebselen. Ebselen (10 μM) was administered before the designated contractile period. ROS formation during low Po₂ treatment was greater than during high Po₂ treatment, and ebselen decreased ROS generation in both low- and high-Po₂ conditions (P < 0.05). ROS accumulated at a faster rate in low vs. high Po₂. Force was reduced >30% for each condition except low Po₂ with ebselen, which only decreased ~15%. We concluded that single myofibers under low Po₂ conditions develop accelerated and more oxidative stress than at Po₂ = 30 Torr (normal human resting Po₂). Ebselen decreases ROS formation in both low and high Po₂, but only mitigates skeletal muscle fatigue during reduced Po₂ conditions.
Collapse
Affiliation(s)
- Li Zuo
- Department of Medicine, University of California, San Diego, La Jolla, CA, USA.
| | | | | | | | | | | |
Collapse
|
46
|
Direct and indirect effects of particulate matter on the cardiovascular system. Toxicol Lett 2011; 208:293-9. [PMID: 22119171 DOI: 10.1016/j.toxlet.2011.11.008] [Citation(s) in RCA: 147] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2011] [Revised: 11/08/2011] [Accepted: 11/09/2011] [Indexed: 01/17/2023]
Abstract
Human exposure to particulate matter (PM) elicits a variety of responses on the cardiovascular system through both direct and indirect pathways. Indirect effects of PM on the cardiovascular system are mediated through the autonomic nervous system, which controls heart rate variability, and inflammatory responses, which augment acute cardiovascular events and atherosclerosis. Recent research demonstrates that PM also affects the cardiovascular system directly by entry into the systemic circulation. This process causes myocardial dysfunction through mechanisms of reactive oxygen species production, calcium ion interference, and vascular dysfunction. In this review, we will present key evidence in both the direct and indirect pathways, suggest clinical applications of the current literature, and recommend directions for future research.
Collapse
|