1
|
Alvarado-Jiménez D, Donzelli G, Morales-Suárez-Varela M. A systematic review on the association between exposure to air particulate matter during pregnancy and the development of hypertensive disorders of pregnancy and gestational diabetes mellitus. REVIEWS ON ENVIRONMENTAL HEALTH 2024; 39:619-641. [PMID: 37141623 DOI: 10.1515/reveh-2022-0258] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Accepted: 04/17/2023] [Indexed: 05/06/2023]
Abstract
Particulate matter (PM) is considered an intrauterine toxin that can cross the blood-placental barrier and circulate in fetal blood, affecting fetal development, and implicating placental and intrauterine inflammation, and oxidative damage. However, the relationship between PM exposure and adverse pregnancy outcomes is still unclear and our aim was to systematically review toxicological evidence on the link between PM exposure during pregnancy and the development of gestational diabetes mellitus or hypertensive disorders of pregnancy, including gestational hypertension and pre-eclampsia. PubMed and Science Direct were searched until January 2022. Of the 204 studies identified, 168 were excluded. The remaining articles were assessed in full-text, and after evaluation, 27 were included in the review. Most of the studies showed an association between PM exposure and gestational hypertension, systolic and diastolic blood pressure, pre-eclampsia, and gestational diabetes mellitus. These results should be interpreted with caution due to the heterogeneity of baseline concentrations, which ranged from 3.3 μg/m3 to 85.9 μg/m3 and from 21.8 μg/m3 to 92.2 μg/m3, respectively for PM2.5 and PM10. Moreover, critical exposure periods were not consistent among studies, with five out of ten observational studies reporting the second trimester as the critical period for hypertensive disorders of pregnancy, and ten out of twelve observational studies reporting the first or second trimester as the critical period for gestational diabetes mellitus. Overall, the findings support an association between PM exposure during pregnancy and adverse pregnancy outcomes, highlighting the need for further research to identify the critical exposure periods and underlying mechanisms.
Collapse
Affiliation(s)
| | - Gabriele Donzelli
- Department of Health Sciences, University of Florence, 50134 Florence, Italy
| | - María Morales-Suárez-Varela
- Department of Preventive Medicine and Public Health, Food Sciences, Toxicology, and Legal Medicine, School of Pharmacy, University of Valencia, Burjassot, Valencia, Spain
- CIBER of Epidemiology and Public Health (CIBERESP), Madrid, Spain
| |
Collapse
|
2
|
Zhang F, Armando I, Jose PA, Zeng C, Yang J. G protein-coupled receptor kinases in hypertension: physiology, pathogenesis, and therapeutic targets. Hypertens Res 2024; 47:2317-2336. [PMID: 38961282 PMCID: PMC11374685 DOI: 10.1038/s41440-024-01763-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2023] [Revised: 05/10/2024] [Accepted: 06/06/2024] [Indexed: 07/05/2024]
Abstract
G protein-coupled receptors (GPCRs) mediate cellular responses to a myriad of hormones and neurotransmitters that play vital roles in the regulation of physiological processes such as blood pressure. In organs such as the artery and kidney, hormones or neurotransmitters, such as angiotensin II (Ang II), dopamine, epinephrine, and norepinephrine exert their functions via their receptors, with the ultimate effect of keeping normal vascular reactivity, normal body sodium, and normal blood pressure. GPCR kinases (GRKs) exert their biological functions, by mediating the regulation of agonist-occupied GPCRs, non-GPCRs, or non-receptor substrates. In particular, increasing number of studies show that aberrant expression and activity of GRKs in the cardiovascular system and kidney inhibit or stimulate GPCRs (e.g., dopamine receptors, Ang II receptors, and α- and β-adrenergic receptors), resulting in hypertension. Current studies focus on the effect of selective GRK inhibitors in cardiovascular diseases, including hypertension. Moreover, genetic studies show that GRK gene variants are associated with essential hypertension, blood pressure response to antihypertensive medicines, and adverse cardiovascular outcomes of antihypertensive treatment. In this review, we present a comprehensive overview of GRK-mediated regulation of blood pressure, role of GRKs in the pathogenesis of hypertension, and highlight potential strategies for the treatment of hypertension. Schematic representation of GPCR desensitization process. Activation of GPCRs begins with the binding of an agonist to its corresponding receptor. Then G proteins activate downstream effectors that are mediated by various signaling pathways. GPCR signaling is halted by GRK-mediated receptor phosphorylation, which causes receptor internalization through β-arrestin.
Collapse
Affiliation(s)
- Fuwei Zhang
- Research Center for Metabolic and Cardiovascular Diseases, The Third Affiliated Hospital of Chongqing Medical University, Chongqing, PR China
- Department of Nutrition, The Third Affiliated Hospital of Chongqing Medical University, Chongqing, PR China
- Department of Cardiology, The Third Affiliated Hospital of Chongqing Medical University, Chongqing, PR China
| | - Ines Armando
- Division of Renal Diseases & Hypertension, Department of Medicine and Department of Physiology/Pharmacology, The George Washington University School of Medicine & Health Sciences, Washington, DC, USA
| | - Pedro A Jose
- Division of Renal Diseases & Hypertension, Department of Medicine and Department of Physiology/Pharmacology, The George Washington University School of Medicine & Health Sciences, Washington, DC, USA
| | - Chunyu Zeng
- Department of Cardiology, Daping Hospital, The Third Military Medical University (Army Medical University), Chongqing, PR China
- Chongqing Key Laboratory for Hypertension Research, Chongqing Cardiovascular Clinical Research Center, Chongqing Institute of Cardiology, Chongqing, PR China
| | - Jian Yang
- Research Center for Metabolic and Cardiovascular Diseases, The Third Affiliated Hospital of Chongqing Medical University, Chongqing, PR China.
- Department of Nutrition, The Third Affiliated Hospital of Chongqing Medical University, Chongqing, PR China.
| |
Collapse
|
3
|
Tain YL, Hsu CN. The Impact of the Aryl Hydrocarbon Receptor on Antenatal Chemical Exposure-Induced Cardiovascular-Kidney-Metabolic Programming. Int J Mol Sci 2024; 25:4599. [PMID: 38731818 PMCID: PMC11083012 DOI: 10.3390/ijms25094599] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2024] [Revised: 04/18/2024] [Accepted: 04/20/2024] [Indexed: 05/13/2024] Open
Abstract
Early life exposure lays the groundwork for the risk of developing cardiovascular-kidney-metabolic (CKM) syndrome in adulthood. Various environmental chemicals to which pregnant mothers are commonly exposed can disrupt fetal programming, leading to a wide range of CKM phenotypes. The aryl hydrocarbon receptor (AHR) has a key role as a ligand-activated transcription factor in sensing these environmental chemicals. Activating AHR through exposure to environmental chemicals has been documented for its adverse impacts on cardiovascular diseases, hypertension, diabetes, obesity, kidney disease, and non-alcoholic fatty liver disease, as evidenced by both epidemiological and animal studies. In this review, we compile current human evidence and findings from animal models that support the connection between antenatal chemical exposures and CKM programming, focusing particularly on AHR signaling. Additionally, we explore potential AHR modulators aimed at preventing CKM syndrome. As the pioneering review to present evidence advocating for the avoidance of toxic chemical exposure during pregnancy and deepening our understanding of AHR signaling, this has the potential to mitigate the global burden of CKM syndrome in the future.
Collapse
Affiliation(s)
- You-Lin Tain
- Division of Pediatric Nephrology, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung 833, Taiwan;
- Institute for Translational Research in Biomedicine, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung 833, Taiwan
- College of Medicine, Chang Gung University, Taoyuan 333, Taiwan
| | - Chien-Ning Hsu
- Department of Pharmacy, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung 833, Taiwan
- School of Pharmacy, Kaohsiung Medical University, Kaohsiung 807, Taiwan
| |
Collapse
|
4
|
Youn EK, Cho HM, Jung JK, Yoon GE, Eto M, Kim JI. Pathologic HDAC1/c-Myc signaling axis is responsible for angiotensinogen transcription and hypertension induced by high-fat diet. Biomed Pharmacother 2023; 164:114926. [PMID: 37244179 DOI: 10.1016/j.biopha.2023.114926] [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: 03/20/2023] [Revised: 05/03/2023] [Accepted: 05/22/2023] [Indexed: 05/29/2023] Open
Abstract
High-fat diet (HFD)-induced obesity is a cause of resistant hypertension. We have shown a possible link between histone deacetylases (HDACs) and renal angiotensinogen (Agt) upregulation in the HFD-induced hypertension, whereas the underlying mechanisms remain to be elucidated. Here, using a HDAC1/2 inhibitor romidepsin (FK228) and siRNAs, we determined roles of HDAC1 and HDAC2 in HFD-induced hypertension and found the pathologic signaling axis between HDAC1 and Agt transcription. Treatment with FK228 canceled the increased blood pressure of male C57BL/6 mice induced by HFD. FK228 also blocked upregulation of renal Agt mRNA, protein, angiotensin II (Ang II) or serum Ang II. Activation and nuclear accumulation of both HDAC1 and HDAC2 occurred in the HFD group. The HFD-induced HDAC activation was associated with an increase in deacetylated c-Myc transcription factor. Silencing of HDAC1, HDAC2 or c-Myc in HRPTEpi cells decreased Agt expression. However, only HDAC1 knockdown, but not HDAC2, increased c-Myc acetylation, suggesting selective roles in two enzymes. Chromatin immunoprecipitation assay revealed that HFD induced the binding of HDAC1 and deacetylated c-Myc at the Agt gene promoter. A putative c-Myc binding sequence in the promotor region was necessary for Agt transcription. Inhibition of c-Myc downregulated Agt and Ang II levels in kidney and serum, ameliorating HFD-induced hypertension. Thus, the abnormal HDAC1/2 in the kidney may be responsible for the upregulation of the Agt gene expression and hypertension. The results expose the pathologic HDAC1/c-myc signaling axis in kidney as a promising therapeutic target for obesity-associated resistant hypertension.
Collapse
Affiliation(s)
- Eui Kyung Youn
- Department of Molecular Medicine, Keimyung University School of Medicine, Daegu 42601, Republic of Korea
| | - Hyun Min Cho
- Department of Molecular Medicine, Keimyung University School of Medicine, Daegu 42601, Republic of Korea
| | - Jin Ki Jung
- Department of Molecular Medicine, Keimyung University School of Medicine, Daegu 42601, Republic of Korea
| | - Ga-Eun Yoon
- Department of Molecular Medicine, Keimyung University School of Medicine, Daegu 42601, Republic of Korea
| | - Masumi Eto
- Department of Veterinary Medicine, Okayama University of Science, Ehime 794-8555, Japan
| | - Jee In Kim
- Department of Molecular Medicine, Keimyung University School of Medicine, Daegu 42601, Republic of Korea.
| |
Collapse
|
5
|
Garcia A, Santa-Helena E, De Falco A, de Paula Ribeiro J, Gioda A, Gioda CR. Toxicological Effects of Fine Particulate Matter (PM 2.5): Health Risks and Associated Systemic Injuries-Systematic Review. WATER, AIR, AND SOIL POLLUTION 2023; 234:346. [PMID: 37250231 PMCID: PMC10208206 DOI: 10.1007/s11270-023-06278-9] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/09/2022] [Accepted: 03/29/2023] [Indexed: 05/31/2023]
Abstract
Previous studies focused on investigating particulate matter with aerodynamic diameter ≤ 2.5 µm (PM2.5) have shown the risk of disease development, and association with increased morbidity and mortality rates. The current review investigate epidemiological and experimental findings from 2016 to 2021, which enabled the systemic overview of PM2.5's toxic impacts on human health. The Web of Science database search used descriptive terms to investigate the interaction among PM2.5 exposure, systemic effects, and COVID-19 disease. Analyzed studies have indicated that cardiovascular and respiratory systems have been extensively investigated and indicated as the main air pollution targets. Nevertheless, PM2.5 reaches other organic systems and harms the renal, neurological, gastrointestinal, and reproductive systems. Pathologies onset and/or get worse due to toxicological effects associated with the exposure to this particle type, since it can trigger several reactions, such as inflammatory responses, oxidative stress generation and genotoxicity. These cellular dysfunctions lead to organ malfunctions, as shown in the current review. In addition, the correlation between COVID-19/Sars-CoV-2 and PM2.5 exposure was also assessed to help better understand the role of atmospheric pollution in the pathophysiology of this disease. Despite the significant number of studies about PM2.5's effects on organic functions, available in the literature, there are still gaps in knowledge about how this particulate matter can hinder human health. The current review aimed to approach the main findings about the effect of PM2.5 exposure on different systems, and demonstrate the likely interaction of COVID-19/Sars-CoV-2 and PM2.5.
Collapse
Affiliation(s)
- Amanda Garcia
- Instituto de Ciências Biológicas, Universidade Federal do Rio Grande - FURG, Rio Grande, RS Brazil
- Programa de Pós Graduação Em Ciências Fisiológicas, Universidade Federal do Rio Grande - FURG, Av. Itália Km 8, Campus Carreiros, Rio Grande, RS 96203-900 Brazil
| | - Eduarda Santa-Helena
- Instituto de Ciências Biológicas, Universidade Federal do Rio Grande - FURG, Rio Grande, RS Brazil
- Programa de Pós Graduação Em Ciências Fisiológicas, Universidade Federal do Rio Grande - FURG, Av. Itália Km 8, Campus Carreiros, Rio Grande, RS 96203-900 Brazil
- Pontifícia Universidade Católica do Rio de Janeiro (PUC-Rio), Departmento de Química, Rio de Janeiro, Brazil
| | - Anna De Falco
- Pontifícia Universidade Católica do Rio de Janeiro (PUC-Rio), Departmento de Química, Rio de Janeiro, Brazil
| | - Joaquim de Paula Ribeiro
- Instituto de Ciências Biológicas, Universidade Federal do Rio Grande - FURG, Rio Grande, RS Brazil
- Programa de Pós Graduação Em Ciências Fisiológicas, Universidade Federal do Rio Grande - FURG, Av. Itália Km 8, Campus Carreiros, Rio Grande, RS 96203-900 Brazil
| | - Adriana Gioda
- Pontifícia Universidade Católica do Rio de Janeiro (PUC-Rio), Departmento de Química, Rio de Janeiro, Brazil
| | - Carolina Rosa Gioda
- Instituto de Ciências Biológicas, Universidade Federal do Rio Grande - FURG, Rio Grande, RS Brazil
- Programa de Pós Graduação Em Ciências Fisiológicas, Universidade Federal do Rio Grande - FURG, Av. Itália Km 8, Campus Carreiros, Rio Grande, RS 96203-900 Brazil
| |
Collapse
|
6
|
Liu Y, Li Y, Xu H, Zhao X, Zhu Y, Zhao B, Yao Q, Duan H, Guo C, Li Y. Pre- and postnatal particulate matter exposure and blood pressure in children and adolescents: A systematic review and meta-analysis. ENVIRONMENTAL RESEARCH 2023; 223:115373. [PMID: 36731599 DOI: 10.1016/j.envres.2023.115373] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/12/2022] [Revised: 01/10/2023] [Accepted: 01/25/2023] [Indexed: 06/18/2023]
Abstract
BACKGROUND Early life is a susceptible period of air pollution-related adverse health effects. Hypertension in children might be life-threatening without prevention or treatment. Nevertheless, the causative association between environmental factors and childhood hypertension was limited. In the light of particulate matter (PM) as an environmental risk factor for cardiovascular diseases, this study investigated the association of pre- and postnatal PM exposure with blood pressure (BP) and hypertension among children and adolescents. METHOD Four electronic databases were searched for related epidemiological studies published up to September 13, 2022. Stata 14.0 was applied to examine the heterogeneity among the studies and evaluate the combined effect sizes per 10 μg/m3 increase of PM by selecting the corresponding models. Besides, subgroup analysis, sensitivity analysis, and publication bias test were also conducted. RESULTS Prenatal PM2.5 exposure was correlated with increased diastolic blood pressure (DBP) in offspring [1.14 mmHg (95% CI: 0.12, 2.17)]. For short-term postnatal exposure effects, PM2.5 (7-day average) was significantly associated with systolic blood pressure (SBP) [0.20 mmHg (95% CI: 0.16, 0.23)] and DBP [0.49 mmHg (95% CI: 0.45, 0.53)]; and also, PM10 (7-day average) was significantly associated with SBP [0.14 mmHg (95% CI: 0.12, 0.16)]. For long-term postnatal exposure effects, positive associations were manifested in SBP with PM2.5 [β = 0.44, 95% CI: 0.40, 0.48] and PM10 [β = 0.35, 95% CI: 0.19, 0.51]; DBP with PM1 [β = 0.45, 95% CI: 0.42, 0.49], PM2.5 [β = 0.31, 95% CI: 0.27, 0.35] and PM10 [β = 0.32, 95% CI: 0.19, 0.45]; and hypertension with PM1 [OR = 1.43, 95% CI: 1.40, 1.46], PM2.5 [OR = 1.65, 95% CI: 1.29, 2.11] and PM10 [OR = 1.26, 95% CI: 1.09, 1.45]. CONCLUSION Both prenatal and postnatal exposure to PM can increase BP, contributing to a higher prevalence of hypertension in children and adolescents.
Collapse
Affiliation(s)
- Yufan Liu
- Department of Toxicology and Sanitary Chemistry, School of Public Health, Capital Medical University, Beijing 100069, China; Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing 100069, China
| | - Yan Li
- Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing 100069, China; Department of Occupational Health and Environmental Health, School of Public Health, Capital Medical University, Beijing, 100069, China
| | - Hailin Xu
- Department of Toxicology and Sanitary Chemistry, School of Public Health, Capital Medical University, Beijing 100069, China; Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing 100069, China
| | - Xinying Zhao
- Department of Toxicology and Sanitary Chemistry, School of Public Health, Capital Medical University, Beijing 100069, China; Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing 100069, China
| | - Yawen Zhu
- Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing 100069, China; Department of Occupational Health and Environmental Health, School of Public Health, Capital Medical University, Beijing, 100069, China
| | - Bosen Zhao
- Department of Toxicology and Sanitary Chemistry, School of Public Health, Capital Medical University, Beijing 100069, China; Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing 100069, China
| | - Qing Yao
- Department of Toxicology and Sanitary Chemistry, School of Public Health, Capital Medical University, Beijing 100069, China; Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing 100069, China
| | - Huawei Duan
- Key Laboratory of Chemical Safety and Health, National Institute for Occupational Health and Poison Control, Chinese Center for Disease Control and Prevention, Beijing 100050, China
| | - Caixia Guo
- Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing 100069, China; Department of Occupational Health and Environmental Health, School of Public Health, Capital Medical University, Beijing, 100069, China.
| | - Yanbo Li
- Department of Toxicology and Sanitary Chemistry, School of Public Health, Capital Medical University, Beijing 100069, China; Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing 100069, China.
| |
Collapse
|
7
|
Predisposed obesity and long-term metabolic diseases from maternal exposure to fine particulate matter (PM2.5) — A review of its effect and potential mechanisms. Life Sci 2022; 310:121054. [DOI: 10.1016/j.lfs.2022.121054] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Revised: 09/30/2022] [Accepted: 10/05/2022] [Indexed: 11/05/2022]
|
8
|
Yang J, Hall JE, Jose PA, Chen K, Zeng C. Comprehensive insights in GRK4 and hypertension: From mechanisms to potential therapeutics. Pharmacol Ther 2022; 239:108194. [PMID: 35487286 PMCID: PMC9728143 DOI: 10.1016/j.pharmthera.2022.108194] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2021] [Revised: 03/30/2022] [Accepted: 04/21/2022] [Indexed: 11/24/2022]
Abstract
G protein-coupled receptors (GPCRs) mediate cellular responses to diverse extracellular stimuli that play vital roles in the regulation of biology, including behavior. Abnormal G protein-coupled receptor kinase (GRK)-mediated regulation of GPCR function is involved in the pathogenesis of hypertension. Among the seven GRK subtypes, GRK4 has attracted attention because of its constitutive activity and tissue-specific expression. Increasing number of studies show that GRK4 affects blood pressure by GPCR-mediated regulation of renal and arterial function. The target receptor of GRK4 is confined not only to GPCRs, but also to other blood pressure-regulating receptors, such as the adiponectin receptor. Genetic studies in humans show that in several ethnic groups, GRK4 gene variants (R65L, A142V, and A486V) are associated with salt-sensitive or salt-resistant essential hypertension and blood pressure responses to antihypertensive medicines. In this article, we present a comprehensive overview of GRK-mediated regulation of blood pressure, focusing on the latest research progress on GRK4 and hypertension and highlighting potential and novel strategies for the prevention and treatment of hypertension.
Collapse
Affiliation(s)
- Jian Yang
- Department of Clinical Nutrition, The Third Affiliated Hospital of Chongqing Medical University, Chongqing, PR China; Research Center for Metabolic and Cardiovascular Diseases, The Third Affiliated Hospital of Chongqing Medical University, Chongqing, China.
| | - John E Hall
- Department of Physiology and Biophysics, Mississippi Center for Obesity Research, University of Mississippi Medical Center, Jackson, MS, USA
| | - Pedro A Jose
- Division of Renal Diseases & Hypertension, The George Washington University School of Medicine & Health Sciences, Washington, DC, USA
| | - Ken Chen
- Department of Cardiology, Daping Hospital, The Third Military Medical University, Chongqing, PR China; Cardiovascular Research Center of Chongqing College, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Chongqing, PR China.
| | - Chunyu Zeng
- Department of Cardiology, Daping Hospital, The Third Military Medical University, Chongqing, PR China; Heart Center of Fujian Province, Union Hospital, Fujian Medical University, Fuzhou, PR China; Department of Cardiology, Chongqing General Hospital, Chongqing, PR China; Cardiovascular Research Center of Chongqing College, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Chongqing, PR China.
| |
Collapse
|
9
|
Pryor JT, Cowley LO, Simonds SE. The Physiological Effects of Air Pollution: Particulate Matter, Physiology and Disease. Front Public Health 2022; 10:882569. [PMID: 35910891 PMCID: PMC9329703 DOI: 10.3389/fpubh.2022.882569] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Accepted: 06/15/2022] [Indexed: 01/19/2023] Open
Abstract
Nine out of 10 people breathe air that does not meet World Health Organization pollution limits. Air pollutants include gasses and particulate matter and collectively are responsible for ~8 million annual deaths. Particulate matter is the most dangerous form of air pollution, causing inflammatory and oxidative tissue damage. A deeper understanding of the physiological effects of particulate matter is needed for effective disease prevention and treatment. This review will summarize the impact of particulate matter on physiological systems, and where possible will refer to apposite epidemiological and toxicological studies. By discussing a broad cross-section of available data, we hope this review appeals to a wide readership and provides some insight on the impacts of particulate matter on human health.
Collapse
Affiliation(s)
- Jack T. Pryor
- Metabolism, Diabetes and Obesity Programme, Monash Biomedicine Discovery Institute, Monash University, Clayton, VIC, Australia
- Woodrudge LTD, London, United Kingdom
| | - Lachlan O. Cowley
- Metabolism, Diabetes and Obesity Programme, Monash Biomedicine Discovery Institute, Monash University, Clayton, VIC, Australia
| | - Stephanie E. Simonds
- Metabolism, Diabetes and Obesity Programme, Monash Biomedicine Discovery Institute, Monash University, Clayton, VIC, Australia
- *Correspondence: Stephanie E. Simonds
| |
Collapse
|
10
|
Yang L, Zhang Y, Qi W, Zhao T, Zhang L, Zhou L, Ye L. Adverse effects of PM 2.5 on cardiovascular diseases. REVIEWS ON ENVIRONMENTAL HEALTH 2022; 37:71-80. [PMID: 33793141 DOI: 10.1515/reveh-2020-0155] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/24/2020] [Accepted: 03/14/2021] [Indexed: 06/12/2023]
Abstract
As an air pollutant, fine particulate matter with a diameter ≤ 2.5 μm (PM2.5) can enter the body through the respiratory tract and cause adverse cardiovascular effects. Here, the effects of PM2.5 on atherosclerosis, hypertension, arrhythmia, myocardial infarction are summarized from the perspective researches of human epidemiology, animal, cell and molecule. The results of this review should be proved useful as a scientific basis for the prevention and treatment of cardiovascular disease caused by PM2.5.
Collapse
Affiliation(s)
- Liwei Yang
- Department of Occupational and Environmental Health, School of Public Health, Jilin University, Changchun, China
| | - Yuezhu Zhang
- Department of Occupational and Environmental Health, School of Public Health, Jilin University, Changchun, China
| | - Wen Qi
- Department of Occupational and Environmental Health, School of Public Health, Jilin University, Changchun, China
| | - Tianyang Zhao
- Department of Occupational and Environmental Health, School of Public Health, Jilin University, Changchun, China
| | - Lele Zhang
- Department of Occupational and Environmental Health, School of Public Health, Jilin University, Changchun, China
| | - Liting Zhou
- Department of Occupational and Environmental Health, School of Public Health, Jilin University, Changchun, China
| | - Lin Ye
- Department of Occupational and Environmental Health, School of Public Health, Jilin University, Changchun, China
| |
Collapse
|
11
|
Hu C, Tao Y, Deng Y, Cai Q, Ren H, Yu C, Zheng S, Yang J, Zeng C. Paternal long-term PM2.5 exposure causes hypertension via increased renal AT1R expression and function in male offspring. Clin Sci (Lond) 2021; 135:2575-2588. [PMID: 34779863 PMCID: PMC8628185 DOI: 10.1042/cs20210802] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2021] [Revised: 11/10/2021] [Accepted: 11/15/2021] [Indexed: 01/15/2023]
Abstract
Maternal exposure to fine particulate matter (PM2.5) causes hypertension in offspring. However, paternal contribution of PM2.5 exposure to hypertension in offspring remains unknown. In the present study, male Sprague-Dawley rats were treated with PM2.5 suspension (10 mg/ml) for 12 weeks and/or fed with tap water containing an antioxidant tempol (1 mM/L) for 16 weeks. The blood pressure, 24 h-urine volume and sodium excretion were determined in male offspring. The offspring were also administrated with losartan (20 mg/kg/d) for 4 weeks. The expressions of angiotensin II type 1 receptor (AT1R) and G-protein-coupled receptor kinase type 4 (GRK4) were determined by qRT-PCR and immunoblotting. We found that long-term PM2.5 exposure to paternal rats caused hypertension and impaired urine volume and sodium excretion in male offspring. Both the mRNA and protein expression of GRK4 and its downstream target AT1R were increased in offspring of PM2.5-exposed paternal rats, which was reflected in its function because treatment with losartan, an AT1R antagonist, decreased the blood pressure and increased urine volume and sodium excretion. In addition, the oxidative stress level was increased in PM2.5-treated paternal rats. Administration with tempol in paternal rats restored the increased blood pressure and decreased urine volume and sodium excretion in the offspring of PM2.5-exposed paternal rats. Treatment with tempol in paternal rats also reversed the increased expressions of AT1R and GRK4 in the kidney of their offspring. We suggest that paternal PM2.5 exposure causes hypertension in offspring. The mechanism may be involved that paternal PM2.5 exposure-associated oxidative stress induces the elevated renal GRK4 level, leading to the enhanced AT1R expression and its-mediated sodium retention, consequently causes hypertension in male offspring.
Collapse
Affiliation(s)
- Cuimei Hu
- Department of Cardiology, Daping Hospital, The Third Military Medical University, Chongqing, China
- Chongqing Key Laboratory for Hypertension Research, Chongqing Cardiovascular Clinical Research Center, Chongqing Institute of Cardiology, Chongqing, P. R. China
| | - Yu Tao
- Department of Cardiology, Daping Hospital, The Third Military Medical University, Chongqing, China
- Chongqing Key Laboratory for Hypertension Research, Chongqing Cardiovascular Clinical Research Center, Chongqing Institute of Cardiology, Chongqing, P. R. China
| | - Yi Deng
- Department of General Practice Medicine, The General Hospital of Western Theater Command, Chengdu, Sichuan, China
| | - Qi Cai
- Department of Cardiology, Fujian Heart Center, Provincial Institute of Coronary Disease, Fujian Medical University Union Hospital, Fuzhou, Fujian, China
| | - Hongmei Ren
- Department of Cardiology, Daping Hospital, The Third Military Medical University, Chongqing, China
- Chongqing Key Laboratory for Hypertension Research, Chongqing Cardiovascular Clinical Research Center, Chongqing Institute of Cardiology, Chongqing, P. R. China
| | - Cheng Yu
- Department of Cardiology, Fujian Heart Center, Provincial Institute of Coronary Disease, Fujian Medical University Union Hospital, Fuzhou, Fujian, China
| | - Shuo Zheng
- Department of Cardiology, Daping Hospital, The Third Military Medical University, Chongqing, China
- Chongqing Key Laboratory for Hypertension Research, Chongqing Cardiovascular Clinical Research Center, Chongqing Institute of Cardiology, Chongqing, P. R. China
| | - Jian Yang
- Department of Clinical Nutrition, The Third Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Chunyu Zeng
- Department of Cardiology, Daping Hospital, The Third Military Medical University, Chongqing, China
- Chongqing Key Laboratory for Hypertension Research, Chongqing Cardiovascular Clinical Research Center, Chongqing Institute of Cardiology, Chongqing, P. R. China
- State Key Laboratory of Trauma, Burns and Combined Injury, Daping Hospital, The Third Military Medical University, Chongqing, China
- Cardiovascular Research Center of Chongqing College, Department of Cardiology of Chongqing General Hospital, University of Chinese Academy of Sciences, Chongqing, China
| |
Collapse
|
12
|
Li R, Peng X, Wu Y, Lv W, Xie H, Ishii Y, Zhang C. Exposure to PM 2.5 during pregnancy causes lung inflammation in the offspring: Mechanism of action of mogrosides. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2021; 228:112955. [PMID: 34781127 DOI: 10.1016/j.ecoenv.2021.112955] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/10/2021] [Revised: 10/16/2021] [Accepted: 10/28/2021] [Indexed: 06/13/2023]
Abstract
Epidemiological and toxicological studies have demonstrated that exposure to fine particulate matter (PM2.5) during pregnancy is harmful to the tissues of the offspring. However, the mechanism by which PM2.5 exposure causes lung damage in the offspring or potential dietary therapy for this condition remains unclear. Mogrosides (MGs) are derived from the traditional plant Siraitia grosvenorii and are used medicinally, where they can moisten the lungs and relieve coughing. In this study, pregnant rats were exposed to PM2.5 by intratracheal instillation and treated with MGs by gavage to model the effect of PM2.5 in the offspring and the interventional effect of MGs on lung tissue. We then used transcriptomics, metabolomics, and RT-qPCR as tools to look for metabolite and genetic changes in the offspring. We found that when compared to the control group, the mRNA levels of the inflammatory mediator Pla2g2d and the metabolites lysophosphatidylcholines (LysoPCs) and arachidonic acid (AA) were up-regulated in the lung tissues of PM2.5 group. In contrast, these inflammatory changes were restored after treatment with MGs during pregnancy. In addition, the levels of AA, LPC 15:0 and LPC 18:0 were elevated in the PM2.5 group compared with control group. This increase was inhibited by co-administration of MGs. The change of PGA1 was adverse. In conclusion, even a relatively low exposure to PM2.5 in rats during pregnancy produces inflammation in the lungs of the male offspring, and an intervention with MGs could significantly alleviate this effect. Furthermore, Pla2g2d may represent a potential target for MGs resulting in the improvement of PM2.5-induced lung injury.
Collapse
Affiliation(s)
- Renshi Li
- State Key Laboratory of Natural Medicines, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 210009, China; Sino-Jan Joint Lab of Natural Health Products Research, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Xuewei Peng
- State Key Laboratory of Natural Medicines, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 210009, China; Sino-Jan Joint Lab of Natural Health Products Research, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Yanliang Wu
- State Key Laboratory of Natural Medicines, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Weichao Lv
- State Key Laboratory of Natural Medicines, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Haifeng Xie
- Research and Development Department, Chengdu Biopurify Phytochemicals Ltd., Chengdu 611130, China
| | - Yuji Ishii
- Laboratory of Molecular Life Sciences, Graduate School of Pharmaceutical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan
| | - Chaofeng Zhang
- State Key Laboratory of Natural Medicines, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 210009, China; Sino-Jan Joint Lab of Natural Health Products Research, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 210009, China.
| |
Collapse
|
13
|
Hsu CN, Tain YL. Adverse Impact of Environmental Chemicals on Developmental Origins of Kidney Disease and Hypertension. Front Endocrinol (Lausanne) 2021; 12:745716. [PMID: 34721300 PMCID: PMC8551449 DOI: 10.3389/fendo.2021.745716] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/22/2021] [Accepted: 09/27/2021] [Indexed: 01/09/2023] Open
Abstract
Chronic kidney disease (CKD) and hypertension are becoming a global health challenge, despite developments in pharmacotherapy. Both diseases can begin in early life by so-called "developmental origins of health and disease" (DOHaD). Environmental chemical exposure during pregnancy can affect kidney development, resulting in renal programming. Here, we focus on environmental chemicals that pregnant mothers are likely to be exposed, including dioxins, bisphenol A (BPA), phthalates, per- and polyfluoroalkyl substances (PFAS), polycyclic aromatic hydrocarbons (PAH), heavy metals, and air pollution. We summarize current human evidence and animal models that supports the link between prenatal exposure to environmental chemicals and developmental origins of kidney disease and hypertension, with an emphasis on common mechanisms. These include oxidative stress, renin-angiotensin system, reduced nephron numbers, and aryl hydrocarbon receptor signaling pathway. Urgent action is required to identify toxic chemicals in the environment, avoid harmful chemicals exposure during pregnancy and lactation, and continue to discover other potentially harmful chemicals. Innovation is also needed to identify kidney disease and hypertension in the earliest stage, as well as translating effective reprogramming interventions from animal studies into clinical practice. Toward DOHaD approach, prohibiting toxic chemical exposure and better understanding of underlying mechanisms, we have the potential to reduce global burden of kidney disease and hypertension.
Collapse
Affiliation(s)
- Chien-Ning Hsu
- Department of Pharmacy, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung, Taiwan
- School of Pharmacy, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - You-Lin Tain
- Department of Pediatrics, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan
- Institute for Translational Research in Biomedicine, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung, Taiwan
| |
Collapse
|
14
|
Johnson NM, Hoffmann AR, Behlen JC, Lau C, Pendleton D, Harvey N, Shore R, Li Y, Chen J, Tian Y, Zhang R. Air pollution and children's health-a review of adverse effects associated with prenatal exposure from fine to ultrafine particulate matter. Environ Health Prev Med 2021; 26:72. [PMID: 34253165 PMCID: PMC8274666 DOI: 10.1186/s12199-021-00995-5] [Citation(s) in RCA: 119] [Impact Index Per Article: 29.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2021] [Accepted: 07/01/2021] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Particulate matter (PM), a major component of ambient air pollution, accounts for a substantial burden of diseases and fatality worldwide. Maternal exposure to PM during pregnancy is particularly harmful to children's health since this is a phase of rapid human growth and development. METHOD In this review, we synthesize the scientific evidence on adverse health outcomes in children following prenatal exposure to the smallest toxic components, fine (PM2.5) and ultrafine (PM0.1) PM. We highlight the established and emerging findings from epidemiologic studies and experimental models. RESULTS Maternal exposure to fine and ultrafine PM directly and indirectly yields numerous adverse birth outcomes and impacts on children's respiratory systems, immune status, brain development, and cardiometabolic health. The biological mechanisms underlying adverse effects include direct placental translocation of ultrafine particles, placental and systemic maternal oxidative stress and inflammation elicited by both fine and ultrafine PM, epigenetic changes, and potential endocrine effects that influence long-term health. CONCLUSION Policies to reduce maternal exposure and health consequences in children should be a high priority. PM2.5 levels are regulated, yet it is recognized that minority and low socioeconomic status groups experience disproportionate exposures. Moreover, PM0.1 levels are not routinely measured or currently regulated. Consequently, preventive strategies that inform neighborhood/regional planning and clinical/nutritional recommendations are needed to mitigate maternal exposure and ultimately protect children's health.
Collapse
Affiliation(s)
- Natalie M Johnson
- Department of Environmental and Occupational Health, Texas A&M University, College Station, TX, 77843, USA.
| | | | - Jonathan C Behlen
- Department of Environmental and Occupational Health, Texas A&M University, College Station, TX, 77843, USA
| | - Carmen Lau
- Department of Veterinary Pathobiology, Texas A&M University, College Station, TX, 77843, USA
| | - Drew Pendleton
- Department of Environmental and Occupational Health, Texas A&M University, College Station, TX, 77843, USA
| | - Navada Harvey
- Department of Environmental and Occupational Health, Texas A&M University, College Station, TX, 77843, USA
| | - Ross Shore
- Department of Environmental and Occupational Health, Texas A&M University, College Station, TX, 77843, USA
| | - Yixin Li
- Department of Chemistry, Texas A&M University, College Station, TX, 77843, USA
| | - Jingshu Chen
- Department of Veterinary Physiology and Pharmacology, Texas A&M University, College Station, TX, 77843, USA
| | - Yanan Tian
- Department of Veterinary Physiology and Pharmacology, Texas A&M University, College Station, TX, 77843, USA
| | - Renyi Zhang
- Department of Chemistry, Texas A&M University, College Station, TX, 77843, USA
| |
Collapse
|
15
|
Chen H, Oliver BG, Pant A, Olivera A, Poronnik P, Pollock CA, Saad S. Particulate Matter, an Intrauterine Toxin Affecting Foetal Development and Beyond. Antioxidants (Basel) 2021; 10:antiox10050732. [PMID: 34066412 PMCID: PMC8148178 DOI: 10.3390/antiox10050732] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Revised: 04/30/2021] [Accepted: 05/02/2021] [Indexed: 02/07/2023] Open
Abstract
Air pollution is the 9th cause of the overall disease burden globally. The solid component in the polluted air, particulate matters (PMs) with a diameter of 2.5 μm or smaller (PM2.5) possess a significant health risk to several organ systems. PM2.5 has also been shown to cross the blood–placental barrier and circulate in foetal blood. Therefore, it is considered an intrauterine environmental toxin. Exposure to PM2.5 during the perinatal period, when the foetus is particularly susceptible to developmental defects, has been shown to reduce birth weight and cause preterm birth, with an increase in adult disease susceptibility in the offspring. However, few studies have thoroughly studied the health outcome of foetuses due to intrauterine exposure and the underlying mechanisms. This perspective summarises currently available evidence, which suggests that intrauterine exposure to PM2.5 promotes oxidative stress and inflammation in a similar manner as occurs in response to direct PM exposure. Oxidative stress and inflammation are likely to be the common mechanisms underlying the dysfunction of multiple systems, offering potential targets for preventative strategies in pregnant mothers for an optimal foetal outcome.
Collapse
Affiliation(s)
- Hui Chen
- School of Life Sciences, Faculty of Science, University of Technology Sydney, Sydney, NSW 2007, Australia; (B.G.O.); (A.O.)
- Correspondence: (H.C.); (S.S.)
| | - Brian G. Oliver
- School of Life Sciences, Faculty of Science, University of Technology Sydney, Sydney, NSW 2007, Australia; (B.G.O.); (A.O.)
| | - Anushriya Pant
- School of Medical Sciences, Faculty of Medicine and Health, University of Sydney, Sydney, NSW 2006, Australia; (A.P.); (P.P.)
| | - Annabel Olivera
- School of Life Sciences, Faculty of Science, University of Technology Sydney, Sydney, NSW 2007, Australia; (B.G.O.); (A.O.)
| | - Philip Poronnik
- School of Medical Sciences, Faculty of Medicine and Health, University of Sydney, Sydney, NSW 2006, Australia; (A.P.); (P.P.)
| | - Carol A. Pollock
- Renal Research Laboratory, Kolling Institute of Medical Research, Sydney, NSW 2065, Australia;
| | - Sonia Saad
- Renal Research Laboratory, Kolling Institute of Medical Research, Sydney, NSW 2065, Australia;
- Correspondence: (H.C.); (S.S.)
| |
Collapse
|
16
|
Yang J, Villar VAM, Jose PA, Zeng C. Renal Dopamine Receptors and Oxidative Stress: Role in Hypertension. Antioxid Redox Signal 2021; 34:716-735. [PMID: 32349533 PMCID: PMC7910420 DOI: 10.1089/ars.2020.8106] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Significance: The kidney plays an important role in the long-term control of blood pressure. Oxidative stress is one of the fundamental mechanisms responsible for the development of hypertension. Dopamine, via five subtypes of receptors, plays an important role in the control of blood pressure by various mechanisms, including the inhibition of oxidative stress. Recent Advances: Dopamine receptors exert their regulatory function to decrease the oxidative stress in the kidney and ultimately maintain normal sodium balance and blood pressure homeostasis. An aberration of this regulation may be involved in the pathogenesis of hypertension. Critical Issues: Our present article reviews the important role of oxidative stress and intrarenal dopaminergic system in the regulation of blood pressure, summarizes the current knowledge on renal dopamine receptor-mediated antioxidation, including decreasing reactive oxygen species production, inhibiting pro-oxidant enzyme nicotinamide-adenine dinucleotide phosphate (NADPH) oxidase, and stimulating antioxidative enzymes, and also discusses its underlying mechanisms, including the increased activity of G protein-coupled receptor kinase 4 (GRK4) and abnormal trafficking of renal dopamine receptors in hypertensive status. Future Directions: Identifying the mechanisms of renal dopamine receptors in the regulation of oxidative stress and their contribution to the pathogenesis of hypertension remains an important research focus. Increased understanding of the role of reciprocal regulation between renal dopamine receptors and oxidative stress in the regulation of blood pressure may give us novel insights into the pathogenesis of hypertension and provide a new treatment strategy for hypertension.
Collapse
Affiliation(s)
- Jian Yang
- Department of Clinical Nutrition, The Third Affiliated Hospital of Chongqing Medical University, Chongqing, People's Republic of China
| | - Van Anthony M Villar
- Division of Renal Diseases & Hypertension, Department of Medicine, The George Washington University School of Medicine and Health Sciences, Washington, District of Columbia, USA
| | - Pedro A Jose
- Division of Renal Diseases & Hypertension, Department of Medicine, The George Washington University School of Medicine and Health Sciences, Washington, District of Columbia, USA
| | - Chunyu Zeng
- Department of Cardiology, Fujian Heart Medical Center, Fujian Medical University Union Hospital, Fuzhou, People's Republic of China.,Department of Cardiology, Daping Hospital, The Third Military Medical University, Chongqing, People's Republic of China
| |
Collapse
|
17
|
The Role of the Renal Dopaminergic System and Oxidative Stress in the Pathogenesis of Hypertension. Biomedicines 2021; 9:biomedicines9020139. [PMID: 33535566 PMCID: PMC7912729 DOI: 10.3390/biomedicines9020139] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Revised: 01/26/2021] [Accepted: 01/28/2021] [Indexed: 01/11/2023] Open
Abstract
The kidney is critical in the long-term regulation of blood pressure. Oxidative stress is one of the many factors that is accountable for the development of hypertension. The five dopamine receptor subtypes (D1R–D5R) have important roles in the regulation of blood pressure through several mechanisms, such as inhibition of oxidative stress. Dopamine receptors, including those expressed in the kidney, reduce oxidative stress by inhibiting the expression or action of receptors that increase oxidative stress. In addition, dopamine receptors stimulate the expression or action of receptors that decrease oxidative stress. This article examines the importance and relationship between the renal dopaminergic system and oxidative stress in the regulation of renal sodium handling and blood pressure. It discusses the current information on renal dopamine receptor-mediated antioxidative network, which includes the production of reactive oxygen species and abnormalities of renal dopamine receptors. Recognizing the mechanisms by which renal dopamine receptors regulate oxidative stress and their degree of influence on the pathogenesis of hypertension would further advance the understanding of the pathophysiology of hypertension.
Collapse
|
18
|
Yu C, Chen S, Wang X, Wu G, Zhang Y, Fu C, Hu C, Liu Z, Luo X, Wang J, Chen L. Exposure to maternal diabetes induces endothelial dysfunction and hypertension in adult male rat offspring. Microvasc Res 2021; 133:104076. [PMID: 32956647 DOI: 10.1016/j.mvr.2020.104076] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2020] [Revised: 09/15/2020] [Accepted: 09/16/2020] [Indexed: 01/10/2023]
Abstract
The adverse environment in early life can modulate adult phenotype, including blood pressure. Our previous study shows, in a rat streptozotocin (STZ)-induced maternal diabetes model, fetal exposure to maternal diabetes is characterized by established hypertension in the offspring. However, the exact mechanisms are not known. Our present study found, as compared with male control mother offspring (CMO), male diabetic mother offspring (DMO) had higher blood pressure with arterial dysfunction, i.e., decreased acetylcholine (Ach)-induced vasodilation. But there is no difference in blood pressure between female CMO and DMO. The decreased Ach-induced vasodilation was related to decreased nitric oxide (NO) production in the endothelium, not NO sensitivity in vascular smooth muscle because sodium nitroprusside (SNP)-mediated vasodilation was preserved; there was decreased NO production and lower eNOS phosphorylation in male DMO. The reactive oxygen species (ROS) level was increased in male DMO than CMO; normalized ROS levels with tempol increased NO production, normalized Ach-mediated vasodilation, and lowered blood pressure in male DMO rats. It indicates that diabetic programming hypertension is related to arterial dysfunction; normalizing ROS might be a potential strategy for the prevention of hypertension in the offspring.
Collapse
MESH Headings
- Age Factors
- Animals
- Arterial Pressure
- Blood Glucose/metabolism
- Cyclic GMP/metabolism
- Diabetes Mellitus, Experimental/blood
- Diabetes Mellitus, Experimental/complications
- Diabetes Mellitus, Experimental/physiopathology
- Diabetes, Gestational/blood
- Diabetes, Gestational/physiopathology
- Endothelium, Vascular/metabolism
- Endothelium, Vascular/physiopathology
- Female
- Hypertension/etiology
- Hypertension/metabolism
- Hypertension/physiopathology
- Male
- Mesenteric Artery, Superior/metabolism
- Mesenteric Artery, Superior/physiopathology
- Nitric Oxide/metabolism
- Oxidative Stress
- Pregnancy
- Prenatal Exposure Delayed Effects
- Rats, Sprague-Dawley
- Reactive Oxygen Species/metabolism
- Sex Factors
- Vasodilation
- Rats
Collapse
Affiliation(s)
- Cheng Yu
- Department of Cardiology, Fujian Heart Center, Provincial Institute of Coronary Disease, Fujian Medical University Union Hospital, Fuzhou, Fujian, China; Department of Cardiology, Daping Hospital, Third Military Medical University; Chongqing Institute of Cardiology, Chongqing, China
| | - Shuo Chen
- Department of Cardiology, Daping Hospital, Third Military Medical University; Chongqing Institute of Cardiology, Chongqing, China
| | - Xinquan Wang
- Department of Cardiology, Daping Hospital, Third Military Medical University; Chongqing Institute of Cardiology, Chongqing, China
| | - Gengze Wu
- Department of Cardiology, Daping Hospital, Third Military Medical University; Chongqing Institute of Cardiology, Chongqing, China
| | - Ye Zhang
- Department of Cardiology, Daping Hospital, Third Military Medical University; Chongqing Institute of Cardiology, Chongqing, China
| | - Chunjiang Fu
- Department of Cardiology, Daping Hospital, Third Military Medical University; Chongqing Institute of Cardiology, Chongqing, China
| | - Cuimei Hu
- Department of Cardiology, Daping Hospital, Third Military Medical University; Chongqing Institute of Cardiology, Chongqing, China
| | - Zhengbi Liu
- Center of Laboratory Animal, Daping Hospital, Third Military Medical University, Chongqing, China
| | - Xiaoli Luo
- Department of Cardiology, Daping Hospital, Third Military Medical University; Chongqing Institute of Cardiology, Chongqing, China
| | - Jialiang Wang
- Department of Cardiology, Daping Hospital, Third Military Medical University; Chongqing Institute of Cardiology, Chongqing, China.
| | - Lianglong Chen
- Department of Cardiology, Fujian Heart Center, Provincial Institute of Coronary Disease, Fujian Medical University Union Hospital, Fuzhou, Fujian, China.
| |
Collapse
|
19
|
Cowell WJ, Brunst KJ, Malin AJ, Coull BA, Gennings C, Kloog I, Lipton L, Wright RO, Enlow MB, Wright RJ. Prenatal Exposure to PM2.5 and Cardiac Vagal Tone during Infancy: Findings from a Multiethnic Birth Cohort. ENVIRONMENTAL HEALTH PERSPECTIVES 2019; 127:107007. [PMID: 31663780 PMCID: PMC6867319 DOI: 10.1289/ehp4434] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/05/2018] [Revised: 09/19/2019] [Accepted: 09/19/2019] [Indexed: 05/20/2023]
Abstract
BACKGROUND The autonomic nervous system plays a key role in maintaining homeostasis and responding to external stimuli. In adults, exposure to fine particulate matter (PM2.5) has been associated with reduced heart rate variability (HRV), an indicator of cardiac autonomic control. OBJECTIVES Our goal was to investigate the associations of exposure to fine particulate matter (PM2.5) with HRV as an indicator of cardiac autonomic control during early development. METHODS We studied 237 maternal-infant pairs in a Boston-based birth cohort. We estimated daily residential PM2.5 using satellite data in combination with land-use regression predictors. In infants at 6 months of age, we measured parasympathetic nervous system (PNS) activity using continuous electrocardiogram monitoring during the Repeated Still-Face Paradigm, an experimental protocol designed to elicit autonomic reactivity in response to maternal interaction and disengagement. We used multivariable linear regression to examine average PM2.5 exposure across pregnancy in relation to PNS withdrawal and activation, indexed by changes in respiration-corrected respiratory sinus arrhythmia (RSAc)-an established metric of HRV that reflects cardiac vagal tone. We examined interactions with infant sex using cross-product terms. RESULTS In adjusted models we found that a 1-unit increase in PM2.5 (in micrograms per cubic meter) was associated with a 3.53% decrease in baseline RSAc (95% CI: -6.96, 0.02). In models examining RSAc change between episodes, higher PM2.5 was generally associated with reduced PNS withdrawal during stress and reduced PNS activation during recovery; however, these associations were not statistically significant. We did not observe a significant interaction between PM2.5 and sex. DISCUSSION Prenatal exposure to PM2.5 may disrupt cardiac vagal tone during infancy. Future research is needed to replicate these preliminary findings. https://doi.org/10.1289/EHP4434.
Collapse
Affiliation(s)
- Whitney J. Cowell
- Department of Environmental Medicine and Public Health, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Kelly J. Brunst
- Department of Environmental Health, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| | - Ashley J. Malin
- Department of Environmental Medicine and Public Health, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Brent A. Coull
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, USA
- Department of Environmental Health, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, USA
| | - Chris Gennings
- Department of Environmental Medicine and Public Health, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Itai Kloog
- Department of Geography and Environmental Development, Faculty of Humanities and Social Sciences, Ben-Gurion University of the Negev, Beer Sheva, Israel
| | - Lianna Lipton
- Department of Pediatrics, Kravis Children’s Hospital, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Robert O. Wright
- Department of Environmental Medicine and Public Health, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Michelle Bosquet Enlow
- Department of Psychiatry, Boston Children’s Hospital, Boston, Massachusetts, USA
- Department of Psychiatry, Harvard Medical School, Boston, Massachusetts, USA
| | - Rosalind J. Wright
- Department of Environmental Medicine and Public Health, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Department of Pediatrics, Kravis Children’s Hospital, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| |
Collapse
|
20
|
Prenatal cold exposure causes hypertension in offspring by hyperactivity of the sympathetic nervous system. Clin Sci (Lond) 2019; 133:1097-1113. [PMID: 31015358 PMCID: PMC6833955 DOI: 10.1042/cs20190254] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2019] [Revised: 04/15/2019] [Accepted: 04/23/2019] [Indexed: 02/07/2023]
Abstract
Environmental temperature plays a role in the variation of blood pressure. Maternal cold stress could affect the physiological phenotype of the offspring, including blood pressure elevation. In the present study, we found that adult offspring of dams exposed to cold have increased systolic and diastolic blood pressure, and decreased urine volume and sodium excretion, accompanied by increased heart rate and heart rate variability, secondary to increased activity of the sympathetic nervous system. Renal denervation or adrenergic receptor blockade decreased blood pressure and increased sodium excretion. The increase in peripheral sympathetic nerve activity can be ascribed to the central nervous system because administration of clonidine, a centrally acting α2 adrenergic receptor agonist, lowered blood pressure to a greater degree in the prenatal cold-exposed than control offspring. Moreover, these prenatal cold-exposed offspring had hypothalamic paraventricular nucleus (PVN) disorder because magnetic resonance spectroscopy showed decreased N-acetylaspartate and increased choline and creatine ratios in the PVN. Additional studies found that prenatal cold exposure impaired the balance between inhibitory and excitatory neurons. This led to PVN overactivation that was related to enhanced PVN-angiotensin II type 1 (AT1) receptor expression and function. Microinjection of the AT1 receptor antagonist losartan in the PVN lowered blood pressure to a greater extent in prenatal cold-exposed that control offspring. The present study provides evidence for overactive peripheral and central sympathetic nervous systems in the pathogenesis of prenatal cold-induced hypertension. Central AT1 receptor blockade in the PVN may be a key step for treatment of this type hypertension.
Collapse
|
21
|
Wang S, Tan X, Chen P, Zheng S, Ren H, Cai J, Zhou L, Jose PA, Yang J, Zeng C. Role of Thioredoxin 1 in Impaired Renal Sodium Excretion of hD 5 R F173L Transgenic Mice. J Am Heart Assoc 2019; 8:e012192. [PMID: 30957627 PMCID: PMC6507211 DOI: 10.1161/jaha.119.012192] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/28/2019] [Accepted: 03/12/2019] [Indexed: 12/12/2022]
Abstract
Background Dopamine D5 receptor (D5R) plays an important role in the maintenance of blood pressure by regulating renal sodium transport. Our previous study found that human D5R mutant F173L transgenic ( hD 5 R F173L-TG) mice are hypertensive. In the present study, we aimed to investigate the mechanisms causing this renal D5R dysfunction in hD 5 R F173L-TG mice. Methods and Results Compared with wild-type D5R-TG ( hD 5 R WT-TG) mice, hD 5 R F173L-TG mice have higher blood pressure, lower basal urine flow and sodium excretion, and impaired agonist-mediated natriuresis and diuresis. Enhanced reactive oxygen species production in hD 5 R F173L-TG mice is caused, in part, by decreased expression of antioxidant enzymes, including thioredoxin 1 (Trx1). Na+-K+-ATPase activity is increased in mouse renal proximal tubule cells transfected with hD 5 R F173L, but is normalized by treatment with exogenous recombinant human Trx1 protein. Regulation of Trx1 by D5R occurs by the phospholipase C/ protein kinase C (PKC) pathway because upregulation of Trx1 expression by D5R does not occur in renal proximal tubule cells from D1R knockout mice in the presence of a phospholipase C or PKC inhibitor. Fenoldopam, a D1R and D5R agonist, stimulates PKC activity in primary renal proximal tubule cells of hD5R WT -TG mice, but not in those of hD 5 R F173L-TG mice. Hyperphosphorylation of hD5RF173L and its dissociation from Gαs and Gαq are associated with impairment of D5R-mediated inhibition of Na+-K+-ATPase activity in hD 5 R F173L-TG mice. Conclusions These suggest that hD 5 R F173L increases blood pressure, in part, by decreasing renal Trx1 expression and increasing reactive oxygen species production. Hyperphosphorylation of hD5RF173L, with its dissociation from Gαs and Gαq, is the key factor in impaired D5R function of hD 5 R F173L-TG mice.
Collapse
Affiliation(s)
- Shaoxiong Wang
- Department of CardiologyDaping HospitalArmy Medical University of PLAChongqingP.R. China
| | - Xiaorong Tan
- Department of CardiologyDaping HospitalArmy Medical University of PLAChongqingP.R. China
| | - Peng Chen
- Department of CardiologyDaping HospitalArmy Medical University of PLAChongqingP.R. China
| | - Shuo Zheng
- Department of CardiologyDaping HospitalArmy Medical University of PLAChongqingP.R. China
| | - Hongmei Ren
- Department of CardiologyDaping HospitalArmy Medical University of PLAChongqingP.R. China
| | - Jin Cai
- Department of CardiologyDaping HospitalArmy Medical University of PLAChongqingP.R. China
| | - Lin Zhou
- Department of CardiologyDaping HospitalArmy Medical University of PLAChongqingP.R. China
| | - Pedro A. Jose
- Division of Renal Disease & HypertensionDepartments of Medicine and Pharmacology/PhysiologyThe George Washington University School of Medicine and Health SciencesWashingtonDC
| | - Jian Yang
- Department of Clinical NutritionThe Third Affiliated Hospital of Chongqing Medical UniversityChongqingP.R. China
| | - Chunyu Zeng
- Department of CardiologyDaping HospitalArmy Medical University of PLAChongqingP.R. China
| |
Collapse
|
22
|
Sun D, Chen K, Wang J, Zhou L, Zeng C. In-utero cold stress causes elevation of blood pressure via impaired vascular dopamine D1 receptor in offspring. Clin Exp Hypertens 2019; 42:99-104. [PMID: 30698033 DOI: 10.1080/10641963.2019.1571603] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Affiliation(s)
- Dongdong Sun
- Department of Cardiology, Daping Hospital, The Third Military Medical University, Chongqing, P.R. China
- Chongqing Institute of Cardiology, Chongqing, P.R. China
| | - Ken Chen
- Department of Cardiology, Daping Hospital, The Third Military Medical University, Chongqing, P.R. China
- Chongqing Institute of Cardiology, Chongqing, P.R. China
- Department of Cardiology, Chengdu Military General Hospital, Chengdu, Sichuan, P.R. China
| | - Jialiang Wang
- Department of Cardiology, Daping Hospital, The Third Military Medical University, Chongqing, P.R. China
- Chongqing Institute of Cardiology, Chongqing, P.R. China
| | - Lin Zhou
- Department of Cardiology, Daping Hospital, The Third Military Medical University, Chongqing, P.R. China
- Chongqing Institute of Cardiology, Chongqing, P.R. China
| | - Chunyu Zeng
- Department of Cardiology, Daping Hospital, The Third Military Medical University, Chongqing, P.R. China
- Chongqing Institute of Cardiology, Chongqing, P.R. China
| |
Collapse
|
23
|
Wang J, Deng Y, Zou X, Luo H, Jose PA, Fu C, Yang J, Zeng C. Long-term low salt diet increases blood pressure by activation of the renin-angiotensin and sympathetic nervous systems. Clin Exp Hypertens 2018; 41:739-746. [PMID: 30451012 DOI: 10.1080/10641963.2018.1545850] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Background The aim of this study was to investigate the effect of long-term low salt diet on blood pressure and its underlying mechanisms.Methods Male Sprague-Dawley (SD) rats were divided into normal salt diet group (0.4%) and low salt diet group (0.04%). Blood pressure was measured with the non-invasive tail-cuff method. The contractile response of isolated mesenteric arteries was measured using a small vessel myograph. The effects on renal function of the intrarenal arterial infusion of candesartan (10 μg/kg/min), an angiotensin II receptor type 1 (AT1R) antagonist, were also measured. The expressions of renal AT1R and mesenteric arterial α1A, α1B, and α1D adrenergic receptors were quantified by immunoblotting. Plasma levels of angiotensin II were also measured.Results Systolic blood pressure was significantly increased after 8 weeks of low salt diet. There were no obvious differences in the renal structure between the low and normal salt diet groups. However, the plasma angiotensin II levels and renal AT1R expression were higher in low than normal salt diet group. The intrarenal arterial infusion of candesartan increased urine flow and sodium excretion to a greater extent in the low than normal salt diet group. The expressions of α1A and α1D, but not α1B, adrenergic receptors, and phenylephrine-induced contraction were increased in mesenteric arteries from the low salt, relative to the normal salt diet group.Conclusion Activation of the renin-angiotensin and sympathetic nervous systems may be involved in the pathogenesis of long-term low salt diet-induced hypertension.
Collapse
Affiliation(s)
- Jialiang Wang
- Department of Cardiology, Daping Hospital, The Third Military Medical University, Chongqing, P.R. China
| | - Yi Deng
- Department of Cardiology, Daping Hospital, The Third Military Medical University, Chongqing, P.R. China
| | - Xue Zou
- Department of Cardiology, Daping Hospital, The Third Military Medical University, Chongqing, P.R. China
| | - Hao Luo
- Department of Cardiology, Daping Hospital, The Third Military Medical University, Chongqing, P.R. China
| | - Pedro A Jose
- Division of Renal Diseases & Hypertension, Departments of Medicine and Pharmacology/Physiology, The George Washington University School of Medicine and Health Sciences, Washington, DC, USA
| | - Chunjiang Fu
- Department of Cardiology, Daping Hospital, The Third Military Medical University, Chongqing, P.R. China
| | - Jian Yang
- Department of Clinical Nutrition, The Third Affiliated Hospital of Chongqing Medical University, Chongqing, P.R. China
| | - Chunyu Zeng
- Department of Cardiology, Daping Hospital, The Third Military Medical University, Chongqing, P.R. China
| |
Collapse
|