51
|
Zhang Y, Qian H, Wu B, You S, Wu S, Lu S, Wang P, Cao L, Zhang N, Sun Y. E3 Ubiquitin ligase NEDD4 family‑regulatory network in cardiovascular disease. Int J Biol Sci 2020; 16:2727-2740. [PMID: 33110392 PMCID: PMC7586430 DOI: 10.7150/ijbs.48437] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2020] [Accepted: 08/06/2020] [Indexed: 12/17/2022] Open
Abstract
Protein ubiquitination represents a critical modification occurring after translation. E3 ligase catalyzes the covalent binding of ubiquitin to the protein substrate, which could be degraded. Ubiquitination as an important protein post-translational modification is closely related to cardiovascular disease. The NEDD4 family, belonging to HECT class of E3 ubiquitin ligases can recognize different substrate proteins, including PTEN, ENaC, Nav1.5, SMAD2, PARP1, Septin4, ALK1, SERCA2a, TGFβR3 and so on, via the WW domain to catalyze ubiquitination, thus participating in multiple cardiovascular-related disease such as hypertension, arrhythmia, myocardial infarction, heart failure, cardiotoxicity, cardiac hypertrophy, myocardial fibrosis, cardiac remodeling, atherosclerosis, pulmonary hypertension and heart valve disease. However, there is currently no review comprehensively clarifying the important role of NEDD4 family proteins in the cardiovascular system. Therefore, the present review summarized recent studies about NEDD4 family members in cardiovascular disease, providing novel insights into the prevention and treatment of cardiovascular disease. In addition, assessing transgenic animals and performing gene silencing would further identify the ubiquitination targets of NEDD4. NEDD4 quantification in clinical samples would also constitute an important method for determining NEDD4 significance in cardiovascular disease.
Collapse
Affiliation(s)
- Ying Zhang
- Department of Cardiology, the First Hospital of China Medical University, Shenyang, Liaoning, P.R. China
| | - Hao Qian
- Department of Cardiology, the First Hospital of China Medical University, Shenyang, Liaoning, P.R. China
| | - Boquan Wu
- Department of Cardiology, the First Hospital of China Medical University, Shenyang, Liaoning, P.R. China
| | - Shilong You
- Department of Cardiology, the First Hospital of China Medical University, Shenyang, Liaoning, P.R. China
| | - Shaojun Wu
- Department of Cardiology, the First Hospital of China Medical University, Shenyang, Liaoning, P.R. China
| | - Saien Lu
- Department of Cardiology, the First Hospital of China Medical University, Shenyang, Liaoning, P.R. China
| | - Pingyuan Wang
- Staff scientist, Center for Molecular Medicine National Heart Lung and Blood Institute, National Institutes of Health, the United States
| | - Liu Cao
- Key Laboratory of Medical Cell Biology, Ministry of Education; Institute of Translational Medicine, China Medical University; Liaoning Province Collaborative Innovation Center of Aging Related Disease Diagnosis and Treatment and Prevention, Shenyang, Liaoning, China
| | - Naijin Zhang
- Department of Cardiology, the First Hospital of China Medical University, Shenyang, Liaoning, P.R. China
| | - Yingxian Sun
- Department of Cardiology, the First Hospital of China Medical University, Shenyang, Liaoning, P.R. China
| |
Collapse
|
52
|
Zhuge Y, Zhang J, Qian F, Wen Z, Niu C, Xu K, Ji H, Rong X, Chu M, Jia C. Role of smooth muscle cells in Cardiovascular Disease. Int J Biol Sci 2020; 16:2741-2751. [PMID: 33110393 PMCID: PMC7586427 DOI: 10.7150/ijbs.49871] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2020] [Accepted: 08/06/2020] [Indexed: 12/13/2022] Open
Abstract
Normally, smooth muscle cells (SMCs) are localized in the tunica media of the vasculature, where they take responsibility for vascular contraction and extracellular matrix (ECM) generation. SMCs also play a significant role in obedience and elastic rebound of the artery in response to the haemodynamic condition. However, under pathological or stressed conditions, phenotype switching from contractile to synthetic state or other cell types will occur in SMCs to positively or negatively contribute to disease progression. Various studies demonstrated that functional changes of SMCs are implicated in several cardiovascular diseases. In this review, we present the function of vascular SMCs (VSMCs) and the involved molecular mechanisms about phenotype switching, and summarize the roles of SMCs in atherosclerosis, hypertension, arterial aneurysms and myocardial infarction, hoping to obtain potential therapeutic targets against cardiovascular disease in the clinical practices.
Collapse
Affiliation(s)
- Yingzhi Zhuge
- Pediatric Research Institute, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325027, China.,Children's Heart Center, Institute of Cardiovascular Development and Translational Medicine, The Second Affiliated Hospital and Yuying children's Hospital of Wenzhou Medical University, Wenzhou 325027, China
| | - Jian Zhang
- Pediatric Research Institute, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325027, China.,Children's Heart Center, Institute of Cardiovascular Development and Translational Medicine, The Second Affiliated Hospital and Yuying children's Hospital of Wenzhou Medical University, Wenzhou 325027, China
| | - Fanyu Qian
- Pediatric Research Institute, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325027, China.,Children's Heart Center, Institute of Cardiovascular Development and Translational Medicine, The Second Affiliated Hospital and Yuying children's Hospital of Wenzhou Medical University, Wenzhou 325027, China
| | - Zhengwang Wen
- Children's Heart Center, Institute of Cardiovascular Development and Translational Medicine, The Second Affiliated Hospital and Yuying children's Hospital of Wenzhou Medical University, Wenzhou 325027, China
| | - Chao Niu
- Pediatric Research Institute, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325027, China.,Children's Heart Center, Institute of Cardiovascular Development and Translational Medicine, The Second Affiliated Hospital and Yuying children's Hospital of Wenzhou Medical University, Wenzhou 325027, China
| | - Ke Xu
- The Institute of Life Sciences, Wenzhou University, Wenzhou, Zhejiang, China
| | - Hao Ji
- The Institute of Life Sciences, Wenzhou University, Wenzhou, Zhejiang, China
| | - Xing Rong
- Children's Heart Center, Institute of Cardiovascular Development and Translational Medicine, The Second Affiliated Hospital and Yuying children's Hospital of Wenzhou Medical University, Wenzhou 325027, China
| | - Maoping Chu
- Pediatric Research Institute, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325027, China.,Children's Heart Center, Institute of Cardiovascular Development and Translational Medicine, The Second Affiliated Hospital and Yuying children's Hospital of Wenzhou Medical University, Wenzhou 325027, China
| | - Chang Jia
- Pediatric Research Institute, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325027, China.,Children's Heart Center, Institute of Cardiovascular Development and Translational Medicine, The Second Affiliated Hospital and Yuying children's Hospital of Wenzhou Medical University, Wenzhou 325027, China
| |
Collapse
|
53
|
Rao KNS, Shen X, Pardue S, Krzywanski DM. Nicotinamide nucleotide transhydrogenase (NNT) regulates mitochondrial ROS and endothelial dysfunction in response to angiotensin II. Redox Biol 2020; 36:101650. [PMID: 32763515 PMCID: PMC7408723 DOI: 10.1016/j.redox.2020.101650] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2020] [Revised: 07/16/2020] [Accepted: 07/18/2020] [Indexed: 12/22/2022] Open
Abstract
Endothelial dysfunction is a critical, initiating step in the development of hypertension (HTN) and mitochondrial reactive oxygen species (ROS) are important contributors to endothelial dysfunction. Genome-wide association studies (GWAS) have identified single nucleotide polymorphisms (SNPs) in the nicotinamide nucleotide transhydrogenase (Nnt) gene that are associated with endothelial dysfunction and increased risk for HTN. NNT is emerging as an important enzyme that regulates mitochondrial NADPH levels and mitochondrial redox balance by supporting the thiol dependent peroxidase systems in the mitochondria. We have previously shown that the absence of NNT in C57Bl/6J animals promotes a more severe hypertensive phenotype through reductions in •NO and endothelial dependent vessel dilation. However, the impact of NNT on human endothelial cell function remains unclear. We utilized NNT directed shRNA in human aortic endothelial cells to test the hypothesis that NNT critically regulates mitochondrial redox balance and endothelial function in response to angiotensin II (Ang II). We demonstrate that NNT expression and activity are elevated in response to the mitochondrial dysfunction and oxidative stress associated with Ang II treatment. Knockdown of NNT led to a significant elevation of mitochondrial ROS production and impaired glutathione peroxidase and glutathione reductase activities associated with a reduction in the NADPH/NADP+ ratio. Loss of NNT also promoted mitochondrial dysfunction, disruption of the mitochondrial membrane potential, and impaired ATP production in response to Ang II. Finally, we observed that, while the loss of NNT augmented eNOS phosphorylation at Ser1177, neither eNOS activity nor nitric oxide production were similarly increased. The results from these studies clearly demonstrate that NNT is critical for the maintenance of mitochondrial redox balance and mitochondrial function. Loss of NNT and disruption of redox balance leads to oxidative stress that compromises eNOS activity that could have a profound effect on the endothelium dependent regulation of vascular tone.
Collapse
Affiliation(s)
- K N Shashanka Rao
- Department of Cellular Biology and Anatomy, School of Medicine, Center for Cardiovascular Diseases and Sciences, Louisiana State University Health Sciences Center - Shreveport, Shreveport, LA, 71103, United States
| | - Xinggui Shen
- Department of Cellular Biology and Anatomy, School of Medicine, Center for Cardiovascular Diseases and Sciences, Louisiana State University Health Sciences Center - Shreveport, Shreveport, LA, 71103, United States
| | - Sibile Pardue
- Department of Cellular Biology and Anatomy, School of Medicine, Center for Cardiovascular Diseases and Sciences, Louisiana State University Health Sciences Center - Shreveport, Shreveport, LA, 71103, United States
| | - David M Krzywanski
- Department of Cellular Biology and Anatomy, School of Medicine, Center for Cardiovascular Diseases and Sciences, Louisiana State University Health Sciences Center - Shreveport, Shreveport, LA, 71103, United States.
| |
Collapse
|
54
|
Forrester SJ, Preston KJ, Cooper HA, Boyer MJ, Escoto KM, Poltronetti AJ, Elliott KJ, Kuroda R, Miyao M, Sesaki H, Akiyama T, Kimura Y, Rizzo V, Scalia R, Eguchi S. Mitochondrial Fission Mediates Endothelial Inflammation. Hypertension 2020; 76:267-276. [PMID: 32389075 PMCID: PMC7289685 DOI: 10.1161/hypertensionaha.120.14686] [Citation(s) in RCA: 67] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2020] [Accepted: 03/31/2020] [Indexed: 01/04/2023]
Abstract
Endothelial inflammation and mitochondrial dysfunction have been implicated in cardiovascular diseases, yet, a unifying mechanism tying them together remains limited. Mitochondrial dysfunction is frequently associated with mitochondrial fission/fragmentation mediated by the GTPase Drp1 (dynamin-related protein 1). Nuclear factor (NF)-κB, a master regulator of inflammation, is implicated in endothelial dysfunction and resultant complications. Here, we explore a causal relationship between mitochondrial fission and NF-κB activation in endothelial inflammatory responses. In cultured endothelial cells, TNF-α (tumor necrosis factor-α) or lipopolysaccharide induces mitochondrial fragmentation. Inhibition of Drp1 activity or expression suppresses mitochondrial fission, NF-κB activation, vascular cell adhesion molecule-1 induction, and leukocyte adhesion induced by these proinflammatory factors. Moreover, attenuations of inflammatory leukocyte adhesion were observed in Drp1 heterodeficient mice as well as endothelial Drp1 silenced mice. Intriguingly, inhibition of the canonical NF-κB signaling suppresses endothelial mitochondrial fission. Mechanistically, NF-κB p65/RelA seems to mediate inflammatory mitochondrial fission in endothelial cells. In addition, the classical anti-inflammatory drug, salicylate, seems to maintain mitochondrial fission/fusion balance against TNF-α via inhibition of NF-κB. In conclusion, our results suggest a previously unknown mechanism whereby the canonical NF-κB cascade and a mitochondrial fission pathway interdependently regulate endothelial inflammation.
Collapse
Affiliation(s)
- Steven J. Forrester
- Cardiovascular Research Center, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, U.S.A
| | - Kyle J. Preston
- Cardiovascular Research Center, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, U.S.A
| | - Hannah A. Cooper
- Cardiovascular Research Center, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, U.S.A
| | - Michael J. Boyer
- Cardiovascular Research Center, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, U.S.A
| | - Kathleen M. Escoto
- Cardiovascular Research Center, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, U.S.A
| | - Anthony J. Poltronetti
- Cardiovascular Research Center, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, U.S.A
| | - Katherine J. Elliott
- Cardiovascular Research Center, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, U.S.A
| | - Ryohei Kuroda
- Cardiovascular Research Center, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, U.S.A
| | - Masashi Miyao
- Cardiovascular Research Center, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, U.S.A
- Department of Forensic Medicine, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Hiromi Sesaki
- Department of Cell Biology, Johns Hopkins School of Medicine, Baltimore, MD, U.S.A
| | - Tomoko Akiyama
- Advanced Medical Research Center, Yokohama City University, Yokohama, Japan
| | - Yayoi Kimura
- Advanced Medical Research Center, Yokohama City University, Yokohama, Japan
| | - Victor Rizzo
- Cardiovascular Research Center, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, U.S.A
| | - Rosario Scalia
- Cardiovascular Research Center, Lewis Katz School of Medicine, Temple University, 3500 N. Broad Street, Philadelphia, PA19140
| | - Satoru Eguchi
- Cardiovascular Research Center, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, U.S.A
| |
Collapse
|
55
|
Pauls AD, Sandhu V, Young D, Nevay DL, Yeung DF, Sirrs S, Tsang MY, Tsang TSM, Lehman A, Mezei MM, Poburko D. High rate of hypertension in patients with m.3243A>G MELAS mutations and POLG variants. Mitochondrion 2020; 53:194-202. [PMID: 32502631 DOI: 10.1016/j.mito.2020.05.011] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2019] [Revised: 05/22/2020] [Accepted: 05/26/2020] [Indexed: 12/14/2022]
Abstract
Animal studies suggest that decreased vascular mitochondrial DNA copy number can promote hypertension. We conducted a chart review of blood pressure and hemodynamics in patients with either mitochondrial encephalopathy, lactic acidosis, and stroke-like episodes (MELAS, n = 36) or individuals with variants in the mitochondrial DNA polymerase gamma (POLG, n = 26). The latter included both pathogenic variants and variants of unknown significance (VUS). Hypertension rates (MELAS 50%, POLG 50%) were elevated relative to Canadian norms in 20-39 (MELAS) and 40-59 (MELAS and POLG) years of age groups. Peripheral resistance was high in the hypertensive versus normotensive patients, potentially indicative of microvascular disease. Despite antihypertensive treatment, systolic blood pressure remained elevated in the POLG versus MELAS group. The risk of hypertension was not associated with MELAS heteroplasmy. Hypertension rates were not different between individuals with known pathogenic POLG variants and those with VUS, including common variants. Hypertension (HT) also did not differ between patients with POLG variants with (n = 17) and without chronic progressive external opthalmoplegia (n = 9) (CPEO). HT was associated with variants in all three functional domains of POLG. These findings suggest that both pathogenic variants and several VUS in the POLG gene may promote human hypertension and extend our past reports that increased risk of HT is associated with MELAS.
Collapse
Affiliation(s)
- Andrew D Pauls
- Biomedical Physiology & Kinesiology, Simon Fraser University, Burnaby, Canada
| | - Vikrant Sandhu
- Biomedical Physiology & Kinesiology, Simon Fraser University, Burnaby, Canada
| | - Dana Young
- Adult Metabolic Diseases Unit, Vancouver General Hospital, Vancouver, BC, Canada
| | - Dayna-Lynn Nevay
- Adult Metabolic Diseases Unit, Vancouver General Hospital, Vancouver, BC, Canada
| | - Darwin F Yeung
- Division of Cardiology, University of British Columbia, Vancouver, BC, Canada
| | - Sandra Sirrs
- Adult Metabolic Diseases Unit, Vancouver General Hospital, Vancouver, BC, Canada
| | - Michael Y Tsang
- Division of Cardiology, University of British Columbia, Vancouver, BC, Canada
| | - Teresa S M Tsang
- Division of Cardiology, University of British Columbia, Vancouver, BC, Canada
| | - Anna Lehman
- Department of Medical Genetics, University of British Columbia, Vancouver, BC, Canada
| | - Michelle M Mezei
- Adult Metabolic Diseases Unit, Vancouver General Hospital, Vancouver, BC, Canada; Division of Neurology, University of British Columbia, Vancouver, BC, Canada
| | - Damon Poburko
- Biomedical Physiology & Kinesiology, Simon Fraser University, Burnaby, Canada; Centre for Cell Biology, Development, and Disease, Simon Fraser University, Burnaby, BC, Canada.
| |
Collapse
|
56
|
El-Mahdy MA, Abdelghany TM, Hemann C, Ewees MG, Mahgoup EM, Eid MS, Shalaan MT, Alzarie YA, Zweier JL. Chronic cigarette smoke exposure triggers a vicious cycle of leukocyte and endothelial-mediated oxidant stress that results in vascular dysfunction. Am J Physiol Heart Circ Physiol 2020; 319:H51-H65. [PMID: 32412791 DOI: 10.1152/ajpheart.00657.2019] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Although there is a strong association between cigarette smoking exposure (CSE) and vascular endothelial dysfunction (VED), the underlying mechanisms by which CSE triggers VED remain unclear. Therefore, studies were performed to define these mechanisms using a chronic mouse model of cigarette smoking (CS)-induced cardiovascular disease mirroring that in humans. C57BL/6 male mice were subjected to CSE for up to 48 wk. CSE impaired acetylcholine (ACh)-induced relaxation of aortic and mesenteric segments and triggered hypertension, with mean arterial blood pressure at 32 and 48 wk of exposure of 122 ± 6 and 135 ± 5 mmHg compared with 99 ± 4 and 102 ± 6 mmHg, respectively, in air-exposed mice. CSE led to monocyte activation with superoxide generation in blood exiting the pulmonary circulation. Macrophage infiltration with concomitant increase in NADPH oxidase subunits p22phox and gp91phox was seen in aortas of CS-exposed mice at 16 wk, with further increase out to 48 wk. Associated with this, increased superoxide production was detected that decreased with Nox inhibition. Tetrahydrobiopterin was progressively depleted in CS-exposed mice but not in air-exposed controls, resulting in endothelial nitric oxide synthase (eNOS) uncoupling and secondary superoxide generation. CSE led to a time-dependent decrease in eNOS and Akt expression and phosphorylation. Overall, CSE induces vascular monocyte infiltration with increased NADPH oxidase-mediated reactive oxygen species generation and depletes the eNOS cofactor tetrahydrobiopterin, uncoupling eNOS and triggering a vicious cycle of oxidative stress with VED and hypertension. Our study provides important insights toward understanding the process by which smoking contributes to the genesis of cardiovascular disease and identifies biomarkers predictive of disease.NEW & NOTEWORTHY In a chronic model of smoking-induced cardiovascular disease, we define underlying mechanisms of smoking-induced vascular endothelial dysfunction (VED). Smoking exposure triggered VED and hypertension and led to vascular macrophage infiltration with concomitant increase in superoxide and NADPH oxidase levels as early as 16 wk of exposure. This oxidative stress was accompanied by tetrahydrobiopterin depletion, resulting in endothelial nitric oxide synthase uncoupling with further superoxide generation triggering a vicious cycle of oxidative stress and VED.
Collapse
Affiliation(s)
- Mohamed A El-Mahdy
- Division of Cardiovascular Medicine, Department of Internal Medicine, Davis Heart and Lung Research Institute, College of Medicine, The Ohio State University, Columbus, Ohio
| | - Tamer M Abdelghany
- Division of Cardiovascular Medicine, Department of Internal Medicine, Davis Heart and Lung Research Institute, College of Medicine, The Ohio State University, Columbus, Ohio.,Department of Pharmacology and Toxicology, College of Pharmacy, Al-Azhar University, Cairo, Egypt
| | - Craig Hemann
- Division of Cardiovascular Medicine, Department of Internal Medicine, Davis Heart and Lung Research Institute, College of Medicine, The Ohio State University, Columbus, Ohio
| | - Mohamed G Ewees
- Division of Cardiovascular Medicine, Department of Internal Medicine, Davis Heart and Lung Research Institute, College of Medicine, The Ohio State University, Columbus, Ohio.,Department of Pharmacology and Toxicology, College of Pharmacy, Al-Azhar University, Cairo, Egypt
| | - Elsayed M Mahgoup
- Division of Cardiovascular Medicine, Department of Internal Medicine, Davis Heart and Lung Research Institute, College of Medicine, The Ohio State University, Columbus, Ohio.,Department of Pharmacology and Toxicology, College of Pharmacy, Al-Azhar University, Cairo, Egypt
| | - Mahmoud S Eid
- Division of Cardiovascular Medicine, Department of Internal Medicine, Davis Heart and Lung Research Institute, College of Medicine, The Ohio State University, Columbus, Ohio.,Department of Pharmacology and Toxicology, College of Pharmacy, Al-Azhar University, Cairo, Egypt
| | - Mahmoud T Shalaan
- Division of Cardiovascular Medicine, Department of Internal Medicine, Davis Heart and Lung Research Institute, College of Medicine, The Ohio State University, Columbus, Ohio.,Department of Pharmacology and Toxicology, College of Pharmacy, Al-Azhar University, Cairo, Egypt
| | - Yasmin A Alzarie
- Division of Cardiovascular Medicine, Department of Internal Medicine, Davis Heart and Lung Research Institute, College of Medicine, The Ohio State University, Columbus, Ohio.,Department of Pharmacology and Toxicology, College of Pharmacy, Helwan University, National Organization of Drug Control and Research, Cairo, Egypt
| | - Jay L Zweier
- Division of Cardiovascular Medicine, Department of Internal Medicine, Davis Heart and Lung Research Institute, College of Medicine, The Ohio State University, Columbus, Ohio
| |
Collapse
|
57
|
Touyz RM, Rios FJ, Alves-Lopes R, Neves KB, Camargo LL, Montezano AC. Oxidative Stress: A Unifying Paradigm in Hypertension. Can J Cardiol 2020; 36:659-670. [PMID: 32389339 PMCID: PMC7225748 DOI: 10.1016/j.cjca.2020.02.081] [Citation(s) in RCA: 143] [Impact Index Per Article: 28.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2020] [Revised: 02/19/2020] [Accepted: 02/19/2020] [Indexed: 02/07/2023] Open
Abstract
The etiology of hypertension involves complex interactions among genetic, environmental, and pathophysiologic factors that influence many regulatory systems. Hypertension is characteristically associated with vascular dysfunction, cardiovascular remodelling, renal dysfunction, and stimulation of the sympathetic nervous system. Emerging evidence indicates that the immune system is also important and that activated immune cells migrate and accumulate in tissues promoting inflammation, fibrosis, and target-organ damage. Common to these processes is oxidative stress, defined as an imbalance between oxidants and antioxidants in favour of the oxidants that leads to a disruption of oxidation-reduction (redox) signalling and control and molecular damage. Physiologically, reactive oxygen species (ROS) act as signalling molecules and influence cell function through highly regulated redox-sensitive signal transduction. In hypertension, oxidative stress promotes posttranslational modification (oxidation and phosphorylation) of proteins and aberrant signalling with consequent cell and tissue damage. Many enzymatic systems generate ROS, but NADPH oxidases (Nox) are the major sources in cells of the heart, vessels, kidneys, and immune system. Expression and activity of Nox are increased in hypertension and are the major systems responsible for oxidative stress in cardiovascular disease. Here we provide a unifying concept where oxidative stress is a common mediator underlying pathophysiologic processes in hypertension. We focus on some novel concepts whereby ROS influence vascular function, aldosterone/mineralocorticoid actions, and immunoinflammation, all important processes contributing to the development of hypertension.
Collapse
Affiliation(s)
- Rhian M Touyz
- Institute of Cardiovascular and Medical Sciences, British Heart Foundation Glasgow Cardiovascular Research Centre, University of Glasgow, Glasgow, Scotland, United Kingdom.
| | - Francisco J Rios
- Institute of Cardiovascular and Medical Sciences, British Heart Foundation Glasgow Cardiovascular Research Centre, University of Glasgow, Glasgow, Scotland, United Kingdom
| | - Rhéure Alves-Lopes
- Institute of Cardiovascular and Medical Sciences, British Heart Foundation Glasgow Cardiovascular Research Centre, University of Glasgow, Glasgow, Scotland, United Kingdom
| | - Karla B Neves
- Institute of Cardiovascular and Medical Sciences, British Heart Foundation Glasgow Cardiovascular Research Centre, University of Glasgow, Glasgow, Scotland, United Kingdom
| | - Livia L Camargo
- Institute of Cardiovascular and Medical Sciences, British Heart Foundation Glasgow Cardiovascular Research Centre, University of Glasgow, Glasgow, Scotland, United Kingdom
| | - Augusto C Montezano
- Institute of Cardiovascular and Medical Sciences, British Heart Foundation Glasgow Cardiovascular Research Centre, University of Glasgow, Glasgow, Scotland, United Kingdom
| |
Collapse
|
58
|
Noh MR, Kong MJ, Han SJ, Kim JI, Park KM. Isocitrate dehydrogenase 2 deficiency aggravates prolonged high-fat diet intake-induced hypertension. Redox Biol 2020; 34:101548. [PMID: 32388270 PMCID: PMC7210593 DOI: 10.1016/j.redox.2020.101548] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2020] [Revised: 04/16/2020] [Accepted: 04/20/2020] [Indexed: 02/07/2023] Open
Abstract
The development of hypertension is associated with mitochondrial redox balance disruptions. NADP+-dependent isocitrate dehydrogenase 2 (IDH2) plays an important role in the maintenance of mitochondrial redox balance by producing mitochondrial NADPH, which is an essential cofactor in the reduction of glutathione (from GSSG to GSH) to reduced form of glutathione (GSH). We investigated the association of IDH2 between the development of prolonged high-fat diet (HFD)-induced hypertension. Idh2 gene-deleted (Idh2-/-) male mice and wild-type (Idh2+/+) littermates were fed either HFD or low-fat diet (LFD). Some mice were administrated with Mito-TEMPO, a mitochondria-specific antioxidant. HFD feeding increased blood pressure (BP) in both Idh2-/- mice and Idh2+/+ mice. HFD-induced BP increase was greater in Idh2-/- than Idh2+/+ mice. HFD intake decreased IDH2 activity, NADPH levels, and the GSH/(GSH + GSSG) ratio in the renal mitochondria. However, HFD intake increased mitochondrial ROS levels, along with the accompanying oxidative stress and damage. HFD intake increased angiotensin II receptor 1 type 1 mRNA levels in the kidneys and plasma renin and angiotensin II concentrations. These HFD-induced changes were more prominent in Idh2-/- mice than Idh2+/+ mice. Mito-TEMPO mitigated the HFD-induced changes in both Idh2-/- and Idh2+/+ mice, with greater effects in Idh2-/- mice than Idh2+/+ mice. These results indicate that prolonged HFD intake disrupts the IDH2-NADPH-GSH-associated antioxidant system and activates the renin-angiotensin system in the kidney, leading to increased BP, suggesting that IDH2 is a critical enzyme in the development of hypertension and that the IDH2-associated antioxidant system could serve as a potential hypertension treatment target.
Collapse
Affiliation(s)
- Mi Ra Noh
- Department of Anatomy, Cardiovascular Research Center and BK21 Plus, School of Medicine, Kyungpook National University, 680 Gukchaebosang-ro, Junggu, Daegu, 41944, Republic of Korea
| | - Min Jung Kong
- Department of Anatomy, Cardiovascular Research Center and BK21 Plus, School of Medicine, Kyungpook National University, 680 Gukchaebosang-ro, Junggu, Daegu, 41944, Republic of Korea
| | - Sang Jun Han
- Department of Anatomy, Cardiovascular Research Center and BK21 Plus, School of Medicine, Kyungpook National University, 680 Gukchaebosang-ro, Junggu, Daegu, 41944, Republic of Korea
| | - Jee In Kim
- Department of Molecular Medicine, Keimyung University School of Medicine, 1095 Dalgubeol-daero, Dalseogu, Daegu, 42601, Republic of Korea
| | - Kwon Moo Park
- Department of Anatomy, Cardiovascular Research Center and BK21 Plus, School of Medicine, Kyungpook National University, 680 Gukchaebosang-ro, Junggu, Daegu, 41944, Republic of Korea.
| |
Collapse
|
59
|
Qian H, Zhang N, Wu B, Wu S, You S, Zhang Y, Sun Y. The E3 ubiquitin ligase Smurf2 regulates PARP1 stability to alleviate oxidative stress-induced injury in human umbilical vein endothelial cells. J Cell Mol Med 2020; 24:4600-4611. [PMID: 32167680 PMCID: PMC7176845 DOI: 10.1111/jcmm.15121] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2019] [Revised: 02/02/2020] [Accepted: 02/07/2020] [Indexed: 12/15/2022] Open
Abstract
Oxidative stress injury is involved in many cardiovascular diseases, like hypertension and myocardial infarction. Ubiquitination is a ubiquitous protein post-translational modification that controls a wide range of biological functions and plays a crucial role in maintaining the homeostasis of cells in physiology and disease. Many studies have shown that oxidative stress damage is inextricably linked to ubiquitination. We demonstrate that Smurf2, an E3 ubiquitinated ligase, is involved in HUVEC apoptosis induced by oxidative stress to alleviate H2 O2 -induced reactive oxygen species (ROS) production and the apoptosis of human umbilical vein endothelial cells (HUVECs). At the same time, we found that Smurf2 can bind the poly(ADP-ribose) polymerase-1(PARP1), and the interaction is enhanced under the stimulation of oxidative stress. We further study and prove that Smurf2 can promote PARP1 ubiquitination and degradation. Collectively, we demonstrate Smurf2 degradation of overactivated PARP1 by ubiquitin-proteasome pathway to protect HUVEC and alleviate oxidative stress injury.
Collapse
Affiliation(s)
- Hao Qian
- Department of Cardiology, the First Hospital of China Medical University, Shenyang, China
| | - Naijin Zhang
- Department of Cardiology, the First Hospital of China Medical University, Shenyang, China
| | - Boquan Wu
- Department of Cardiology, the First Hospital of China Medical University, Shenyang, China
| | - Shaojun Wu
- Department of Cardiology, the First Hospital of China Medical University, Shenyang, China
| | - Shilong You
- Department of Cardiology, the First Hospital of China Medical University, Shenyang, China
| | - Ying Zhang
- Department of Cardiology, the First Hospital of China Medical University, Shenyang, China
| | - Yingxian Sun
- Department of Cardiology, the First Hospital of China Medical University, Shenyang, China
| |
Collapse
|
60
|
Wang H, Li X, Li RJ, Yan J, Lan Z, Chen J, Zhang L. Associations of exposure to metals with the risk of hypertension among an older population aged 40-75 years in rural southwest China. J Appl Toxicol 2020; 40:1076-1086. [PMID: 32163192 DOI: 10.1002/jat.3968] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2020] [Revised: 02/24/2020] [Accepted: 02/25/2020] [Indexed: 12/13/2022]
Abstract
Metal exposure has recently been related to the risk of hypertension. However, the association remains unclear and relevant epidemiologic studies are limited. The present study aimed to assess the associations between exposure to metals and the odds of hypertension, as well as blood pressure (BP) levels. A total of 816 participants were enrolled in southwestern China. Hypertension was defined as a systolic BP (SBP) of ≥140 mmHg or diastolic BP (DBP) of ≥90 mmHg, a self-reported physician diagnosis, or current use of antihypertensive medication. Blood samples were used to detect the levels of exposure to metals, ie, magnesium (Mg), calcium (Ca), iron (Fe), zinc (Zn), arsenic (As), cadmium (Cd), copper (Cu) and lead (Pb). Logistic and linear regression models were used to assess the potential associations. The results show that positive trends for elevated odds of hypertension with increasing quartiles of Fe in a polluted area; and of Mg, Ca and Cu in an unpolluted area. Compared with those in the lowest quartiles, participants in the highest quartiles of Fe, Mg and Ca had 2.7-, 9.0- and 5.1-fold increased odds of hypertension, respectively. High blood Fe and Pb levels in the Cd-polluted area, and Mg and Fe in the unpolluted area were found to be related to increasing SBP and DBP levels. Our findings suggest that exposure to Fe and/or Pb in the polluted area; and Mg, Ca and Fe in the unpolluted area might increase the risk of hypertension or elevate BP levels.
Collapse
Affiliation(s)
- He Wang
- West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, China.,Food Safety Monitoring and Risk Assessment Key Laboratory of Sichuan Province, Sichuan University, Chengdu, China
| | - Xiaomeng Li
- West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, China.,Food Safety Monitoring and Risk Assessment Key Laboratory of Sichuan Province, Sichuan University, Chengdu, China
| | - Ren Jia Li
- West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, China.,Food Safety Monitoring and Risk Assessment Key Laboratory of Sichuan Province, Sichuan University, Chengdu, China
| | - Jiuming Yan
- West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, China.,Food Safety Monitoring and Risk Assessment Key Laboratory of Sichuan Province, Sichuan University, Chengdu, China
| | - Zhen Lan
- Department of Nutrition and Food Safety, Sichuan Center for Disease Control and P-revention, Chengdu, China
| | - Jinyao Chen
- West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, China.,Food Safety Monitoring and Risk Assessment Key Laboratory of Sichuan Province, Sichuan University, Chengdu, China
| | - Lishi Zhang
- West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, China.,Food Safety Monitoring and Risk Assessment Key Laboratory of Sichuan Province, Sichuan University, Chengdu, China
| |
Collapse
|
61
|
Jusic A, Devaux Y. Mitochondrial noncoding RNA-regulatory network in cardiovascular disease. Basic Res Cardiol 2020; 115:23. [PMID: 32140778 DOI: 10.1007/s00395-020-0783-5] [Citation(s) in RCA: 65] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/04/2019] [Accepted: 02/17/2020] [Indexed: 12/22/2022]
Abstract
Mitochondrial function and integrity are vital for the maintenance of cellular homeostasis, particularly in high-energy demanding cells. Cardiomyocytes have a large number of mitochondria, which provide a continuous and bulk supply of the ATP necessary for cardiac mechanical function. More than 90% of the ATP consumed by the heart is derived from the mitochondrial oxidative metabolism. Decreased energy supply as the main consequence of mitochondrial dysfunction is closely linked to cardiovascular disease (CVD). The discovery of noncoding RNA (ncRNAs) in the mitochondrial compartment has changed the traditional view of molecular pathways involved in the regulatory network of CVD. Mitochondrial ncRNAs participate in controlling cardiovascular pathogenesis by regulating glycolysis, mitochondrial energy status, and the expression of genes involved in mitochondrial metabolism. Understanding the underlying mechanisms of the association between impaired mitochondrial function resulting from fluctuation in expression levels of ncRNAs and specific disease phenotype can aid in preventing and treating CVD. This review presents an overview of the role of mitochondrial ncRNAs in the complex regulatory network of the cardiovascular pathology. We will summarize and discuss (1) mitochondrial microRNAs (mitomiRs) and long noncoding RNAs (lncRNAs) encoded either by nuclear or mitochondrial genome which are involved in the regulation of mitochondrial metabolism; (2) the role of mitomiRs and lncRNAs in the pathogenesis of several CVD such as hypertension, cardiac hypertrophy, acute myocardial infarction and heart failure; (3) the biomarker and therapeutic potential of mitochondrial ncRNAs in CVD; (4) and the challenges inherent to their translation into clinical application.
Collapse
Affiliation(s)
- Amela Jusic
- Department of Biology, Faculty of Natural Sciences and Mathematics, University of Tuzla, Tuzla, Bosnia and Herzegovina
| | - Yvan Devaux
- Cardiovascular Research Unit, Luxembourg Institute of Health, 1A-B rue Edison, 1445, Strassen, Luxembourg.
| | | |
Collapse
|
62
|
Dera AA, Rajagopalan P, Alfhili MA, Ahmed I, Chandramoorthy HC. Thymoquinone attenuates oxidative stress of kidney mitochondria and exerts nephroprotective effects in oxonic acid-induced hyperuricemia rats. Biofactors 2020; 46:292-300. [PMID: 31758843 DOI: 10.1002/biof.1590] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/06/2019] [Accepted: 10/30/2019] [Indexed: 12/16/2022]
Abstract
BACKGROUND Recent studies indicate hyperuricemia as an aggravating factor for kidney diseases progression. Basic research for novel agents to reduce hyperuricemia and kidney abnormalities will be highly rewarding. Herein, we report Thymoquinone (Tq) as an active constituent of Nigella sativa to have renal protective effective against oxonic acid (OA)-induced hyperuricemia, hypertension, and renal oxidative stress in rat models. METHODS OA 750 mg/kg BW for 12 weeks was used to induce uricemia in Sprague dawley rats. Tq at 10 and 20 mg/kg BW were administered along with OA for treatment groups. Plasma uric acid concentration and systolic blood pressure were measured. Oxidative stress markers, total ATP content, and membrane bound ATPases were measured in renal mitochondria. Anti-oxidant enzymes were analyzed in the renal tissues. Apoptosis in renal tissue was detected. Key signaling proteins for apoptosis, oxidative stress, and lipid oxidation pathways were determined. RESULTS OA induced both circulating uric acid levels and hypertension in the control group which was brought down on Tq treatments. Tq effectively prevented accumulation of uric acid and oxidative stress in the renal tissues. Tq also proved to increase the total ATP content of the renal mitochondria and prevented the apoptosis induced by OA. Tq increased the expressions of phosphorylated Akt, Nrf2, and HO-1 proteins while decreasing the levels of cleaved caspase-3 in renal cells. CONCLUSION In summary, Tq exhibited protective effects on hyperuricemia-mediated renal oxidative stress and mitochondrial abnormalities which could be mediated by Nrf2/HO-1, Akt signaling pathways.
Collapse
Affiliation(s)
- Ayed A Dera
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, King Khalid University, Abha, Saudi Arabia
- Central Research Laboratory, College of Applied Medical Sciences, King Khalid University, Abha, Saudi Arabia
| | - Prasanna Rajagopalan
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, King Khalid University, Abha, Saudi Arabia
- Central Research Laboratory, College of Applied Medical Sciences, King Khalid University, Abha, Saudi Arabia
| | - Mohammad A Alfhili
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, King Saud University, Riyadh, Saudi Arabia
| | - Irfan Ahmed
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, King Khalid University, Abha, Saudi Arabia
- Central Research Laboratory, College of Applied Medical Sciences, King Khalid University, Abha, Saudi Arabia
| | - Harish C Chandramoorthy
- Department of Microbiology & Clinical Parasitology and Centre for Stem Cell Research, College of Medicine, King Khalid University, Abha, Saudi Arabia
| |
Collapse
|
63
|
Nigro E, Perrotta F, Polito R, D'Agnano V, Scialò F, Bianco A, Daniele A. Metabolic Perturbations and Severe COVID-19 Disease: Implication of Molecular Pathways. Int J Endocrinol 2020; 2020:8896536. [PMID: 33312199 PMCID: PMC7703458 DOI: 10.1155/2020/8896536] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/16/2020] [Revised: 11/04/2020] [Accepted: 11/17/2020] [Indexed: 01/08/2023] Open
Abstract
Coronavirus disease (COVID-19) is caused by SARS-CoV-2 virus, which can result in serious respiratory illnesses such as pneumonia leading to respiratory failure. It was first reported in Wuhan, Hubei, China, in December 2019 and rapidly spread globally, becoming a pandemic in March 2020. Among comorbidities observed in SARS-CoV-2 positive patients, hypertension (68.3%) and type 2-diabetes (30.1%) are the most frequent conditions. Although symptoms are highly heterogeneous (ranging from absence of symptoms to severe acute respiratory failure), patients with metabolic-associated diseases often experience worse COVID-19 outcomes. This review investigates the association between metabolic disorders and COVID-19 severity, exploring the molecular mechanisms potentially underlying this relationship and those that are responsible for more severe COVID-19 outcomes. In addition, the role of the main biological processes that may connect metabolic alterations to SARS-CoV-2 infection such as hyperglycemia, immune system deregulation, ACE-2 receptor modulation, and inflammatory response is described. The impact of metabolic disorders on the prognosis of COVID-19 has major implications in public health especially for countries affected by a high incidence of metabolic diseases.
Collapse
Affiliation(s)
- Ersilia Nigro
- Dipartimento di Scienze e Tecnologie Ambientali Biologiche Farmaceutiche, Università Degli Studi Della Campania “Luigi Vanvitelli”, Via G. Vivaldi 42, Caserta 81100, Italy
- CEINGE-Biotecnologie Avanzate Scarl, Via G. Salvatore 486, Napoli 80145, Italy
| | - Fabio Perrotta
- Dipartimento di Medicina e Scienze Della Salute “V. Tiberio”, Università Del Molise, Campobasso 86100, Italy
| | - Rita Polito
- CEINGE-Biotecnologie Avanzate Scarl, Via G. Salvatore 486, Napoli 80145, Italy
| | - Vito D'Agnano
- Dipartimento di Scienze Mediche Traslazionali e Chirurgiche, Università Della Campania “L. Vanvitelli”, Napoli 80131, Italy
| | - Filippo Scialò
- Dipartimento di Scienze Mediche Traslazionali e Chirurgiche, Università Della Campania “L. Vanvitelli”, Napoli 80131, Italy
| | - Andrea Bianco
- Dipartimento di Scienze Mediche Traslazionali e Chirurgiche, Università Della Campania “L. Vanvitelli”, Napoli 80131, Italy
| | - Aurora Daniele
- Dipartimento di Scienze e Tecnologie Ambientali Biologiche Farmaceutiche, Università Degli Studi Della Campania “Luigi Vanvitelli”, Via G. Vivaldi 42, Caserta 81100, Italy
- CEINGE-Biotecnologie Avanzate Scarl, Via G. Salvatore 486, Napoli 80145, Italy
| |
Collapse
|
64
|
Abstract
The microcirculation maintains tissue homeostasis through local regulation of blood flow and oxygen delivery. Perturbations in microvascular function are characteristic of several diseases and may be early indicators of pathological changes in the cardiovascular system and in parenchymal tissue function. These changes are often mediated by various reactive oxygen species and linked to disruptions in pathways such as vasodilation or angiogenesis. This overview compiles recent advances relating to redox regulation of the microcirculation by adopting both cellular and functional perspectives. Findings from a variety of vascular beds and models are integrated to describe common effects of different reactive species on microvascular function. Gaps in understanding and areas for further research are outlined. © 2020 American Physiological Society. Compr Physiol 10:229-260, 2020.
Collapse
Affiliation(s)
- Andrew O Kadlec
- Department of Physiology, Medical College of Wisconsin, Milwaukee, Wisconsin, USA.,Medical Scientist Training Program, Medical College of Wisconsin, Milwaukee, Wisconsin, USA.,Cardiovascular Center, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| | - David D Gutterman
- Department of Physiology, Medical College of Wisconsin, Milwaukee, Wisconsin, USA.,Department of Medicine-Division of Cardiology, Medical College of Wisconsin, Milwaukee, Wisconsin, USA.,Cardiovascular Center, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| |
Collapse
|
65
|
Lipecz A, Miller L, Kovacs I, Czakó C, Csipo T, Baffi J, Csiszar A, Tarantini S, Ungvari Z, Yabluchanskiy A, Conley S. Microvascular contributions to age-related macular degeneration (AMD): from mechanisms of choriocapillaris aging to novel interventions. GeroScience 2019; 41:813-845. [PMID: 31797238 PMCID: PMC6925092 DOI: 10.1007/s11357-019-00138-3] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2019] [Accepted: 11/12/2019] [Indexed: 12/13/2022] Open
Abstract
Aging of the microcirculatory network plays a central role in the pathogenesis of a wide range of age-related diseases, from heart failure to Alzheimer's disease. In the eye, changes in the choroid and choroidal microcirculation (choriocapillaris) also occur with age, and these changes can play a critical role in the pathogenesis of age-related macular degeneration (AMD). In order to develop novel treatments for amelioration of choriocapillaris aging and prevention of AMD, it is essential to understand the cellular and functional changes that occur in the choroid and choriocapillaris during aging. In this review, recent advances in in vivo analysis of choroidal structure and function in AMD patients and patients at risk for AMD are discussed. The pathophysiological roles of fundamental cellular and molecular mechanisms of aging including oxidative stress, mitochondrial dysfunction, and impaired resistance to molecular stressors in the choriocapillaris are also considered in terms of their contribution to the pathogenesis of AMD. The pathogenic roles of cardiovascular risk factors that exacerbate microvascular aging processes, such as smoking, hypertension, and obesity as they relate to AMD and choroid and choriocapillaris changes in patients with these cardiovascular risk factors, are also discussed. Finally, future directions and opportunities to develop novel interventions to prevent/delay AMD by targeting fundamental cellular and molecular aging processes are presented.
Collapse
Affiliation(s)
- Agnes Lipecz
- Translational Geroscience Laboratory, Center for Geroscience and Healthy Brain Aging/Reynolds Oklahoma Center on Aging, Department of Biochemistry and Molecular Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Vascular Cognitive Impairment and Neurodegeneration Program, Center for Geroscience and Healthy Brain Aging/Reynolds Oklahoma Center on Aging, Department of Biochemistry and Molecular Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Department of Ophthalmology, Josa Andras Hospital, Nyiregyhaza, Hungary
- International Training Program in Geroscience, Doctoral School of Basic and Translational Medicine/Department of Public Health, Semmelweis University, Budapest, Hungary
- Department of Ophthalmology, Semmelweis University, Budapest, Hungary
| | - Lauren Miller
- Vascular Cognitive Impairment and Neurodegeneration Program, Center for Geroscience and Healthy Brain Aging/Reynolds Oklahoma Center on Aging, Department of Biochemistry and Molecular Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Department of Cell Biology, University of Oklahoma Health Sciences Center, 940 Stanton L. Young Blvd. BMSB553, Oklahoma City, OK, 73104, USA
| | - Illes Kovacs
- Vascular Cognitive Impairment and Neurodegeneration Program, Center for Geroscience and Healthy Brain Aging/Reynolds Oklahoma Center on Aging, Department of Biochemistry and Molecular Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Department of Ophthalmology, Semmelweis University, Budapest, Hungary
- Department of Ophthalmology, Weill Cornell Medical College, New York City, NY, USA
| | - Cecília Czakó
- Department of Ophthalmology, Semmelweis University, Budapest, Hungary
| | - Tamas Csipo
- Translational Geroscience Laboratory, Center for Geroscience and Healthy Brain Aging/Reynolds Oklahoma Center on Aging, Department of Biochemistry and Molecular Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Vascular Cognitive Impairment and Neurodegeneration Program, Center for Geroscience and Healthy Brain Aging/Reynolds Oklahoma Center on Aging, Department of Biochemistry and Molecular Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- International Training Program in Geroscience, Doctoral School of Basic and Translational Medicine/Department of Public Health, Semmelweis University, Budapest, Hungary
- International Training Program in Geroscience, Division of Clinical Physiology, Department of Cardiology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Judit Baffi
- Vascular Cognitive Impairment and Neurodegeneration Program, Center for Geroscience and Healthy Brain Aging/Reynolds Oklahoma Center on Aging, Department of Biochemistry and Molecular Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Anna Csiszar
- Translational Geroscience Laboratory, Center for Geroscience and Healthy Brain Aging/Reynolds Oklahoma Center on Aging, Department of Biochemistry and Molecular Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Vascular Cognitive Impairment and Neurodegeneration Program, Center for Geroscience and Healthy Brain Aging/Reynolds Oklahoma Center on Aging, Department of Biochemistry and Molecular Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- International Training Program in Geroscience, Doctoral School of Basic and Translational Medicine/Department of Public Health, Semmelweis University, Budapest, Hungary
- International Training Program in Geroscience, Theoretical Medicine Doctoral School, University of Szeged, Szeged, Hungary
| | - Stefano Tarantini
- Translational Geroscience Laboratory, Center for Geroscience and Healthy Brain Aging/Reynolds Oklahoma Center on Aging, Department of Biochemistry and Molecular Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Vascular Cognitive Impairment and Neurodegeneration Program, Center for Geroscience and Healthy Brain Aging/Reynolds Oklahoma Center on Aging, Department of Biochemistry and Molecular Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- International Training Program in Geroscience, Doctoral School of Basic and Translational Medicine/Department of Public Health, Semmelweis University, Budapest, Hungary
- International Training Program in Geroscience, Theoretical Medicine Doctoral School, University of Szeged, Szeged, Hungary
| | - Zoltan Ungvari
- Translational Geroscience Laboratory, Center for Geroscience and Healthy Brain Aging/Reynolds Oklahoma Center on Aging, Department of Biochemistry and Molecular Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Vascular Cognitive Impairment and Neurodegeneration Program, Center for Geroscience and Healthy Brain Aging/Reynolds Oklahoma Center on Aging, Department of Biochemistry and Molecular Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- International Training Program in Geroscience, Doctoral School of Basic and Translational Medicine/Department of Public Health, Semmelweis University, Budapest, Hungary
- International Training Program in Geroscience, Theoretical Medicine Doctoral School, University of Szeged, Szeged, Hungary
- Department of Health Promotion Sciences, College of Public Health, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Andriy Yabluchanskiy
- Translational Geroscience Laboratory, Center for Geroscience and Healthy Brain Aging/Reynolds Oklahoma Center on Aging, Department of Biochemistry and Molecular Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Vascular Cognitive Impairment and Neurodegeneration Program, Center for Geroscience and Healthy Brain Aging/Reynolds Oklahoma Center on Aging, Department of Biochemistry and Molecular Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Shannon Conley
- Vascular Cognitive Impairment and Neurodegeneration Program, Center for Geroscience and Healthy Brain Aging/Reynolds Oklahoma Center on Aging, Department of Biochemistry and Molecular Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA.
- Department of Cell Biology, University of Oklahoma Health Sciences Center, 940 Stanton L. Young Blvd. BMSB553, Oklahoma City, OK, 73104, USA.
| |
Collapse
|
66
|
Sun D, Wei Y, Zheng HX, Jin L, Wang J. Contribution of Mitochondrial DNA Variation to Chronic Disease in East Asian Populations. Front Mol Biosci 2019; 6:128. [PMID: 31803756 PMCID: PMC6873657 DOI: 10.3389/fmolb.2019.00128] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2019] [Accepted: 10/29/2019] [Indexed: 12/17/2022] Open
Abstract
Mitochondria are the main producers of energy in eukaryotic cells. Mitochondrial dysfunction is associated with specific mitochondrial DNA (mtDNA) variations (haplogroups), and these variations can contribute to human disease. East Asian populations show enrichment of many mitochondrial haplogroups, including A, B, D, G, M7, M8, M9, N9, R9, and exhibit half of the known haplogroups of worldwide. In this review, we summarize the current research in the field of mtDNA variation and associated disease in East Asian populations and discuss the physiological and pathological relevance of mitochondrial biology. mtDNA haplogroups are associated with various metabolic disorders ascribed to altered oxidative phosphorylation. The same mitochondrial haplogroup can show either a negative or positive association with different diseases. Mitochondrial dynamics, mitophagy, and mitochondrial oxidative stress, ultimately influence susceptibility to various diseases. In addition, mitochondrial retrograde signaling pathways may have profound effects on nuclear-mitochondrial interactions, affecting cellular morphology, and function. Other complex networks including proteostasis, mitochondrial unfolded protein response and reactive oxygen species signaling may also play pivotal roles in metabolic performance.
Collapse
Affiliation(s)
- Dayan Sun
- State Key Laboratory of Genetic Engineering and Collaborative Innovation Center for Genetics and Development, School of Life Sciences, Fudan University, Shanghai, China
- Human Phenome Institute, Fudan University, Shanghai, China
| | - Yang Wei
- State Key Laboratory of Genetic Engineering and Collaborative Innovation Center for Genetics and Development, School of Life Sciences, Fudan University, Shanghai, China
- Human Phenome Institute, Fudan University, Shanghai, China
| | - Hong-Xiang Zheng
- Ministry of Education Key Laboratory of Contemporary Anthropology, Department of Anthropology and Human Genetics, School of Life Sciences, Fudan University, Shanghai, China
| | - Li Jin
- State Key Laboratory of Genetic Engineering and Collaborative Innovation Center for Genetics and Development, School of Life Sciences, Fudan University, Shanghai, China
- Human Phenome Institute, Fudan University, Shanghai, China
| | - Jiucun Wang
- State Key Laboratory of Genetic Engineering and Collaborative Innovation Center for Genetics and Development, School of Life Sciences, Fudan University, Shanghai, China
- Human Phenome Institute, Fudan University, Shanghai, China
| |
Collapse
|
67
|
Zhong Q, Jiang CX, Zhang C, Zhang Q, Qin QR, Wang XD, Huang F. Urinary Metal Concentrations and the Incidence of Hypertension Among Adult Residents Along the Yangtze River, China. ARCHIVES OF ENVIRONMENTAL CONTAMINATION AND TOXICOLOGY 2019; 77:490-500. [PMID: 31363802 DOI: 10.1007/s00244-019-00655-4] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/02/2019] [Accepted: 07/15/2019] [Indexed: 06/10/2023]
Abstract
Metals from the natural environment have potential hypertension effects. However, relevant studies on this topic are few. A total of 1358 adults aged 18-74 years from Chizhou, Maanshan, and Tongling of Anhui Province participated in the baseline study from 2014 to 2015. The follow-up study was performed from 2016 to 2017. Inductively coupled plasma optical emission spectrometry (7000 DV) was used to measure urinary Cr, Mn, Fe, Cu, and Zn of residents. Urinary concentrations of Cd determined via TAS-900 atomic absorption spectrophotometry at 228.8 nm wavelength. A total of 275 hypertension cases were identified. After adjusting for sociodemographic factors and risk factors for hypertension, four metals (Cd, Cr, Cu, and Mn) were significantly associated with hypertension in the single-metal model. Upon including all metals in the same model, the hazard ratios of the highest quartiles Cd and Cu compared with the reference group were 1.42 (95% confidence interval [CI] 1.09-2.02) and 1.56 (95% CI 1.16-2.09) for cases of hypertension. Our findings suggested that high levels of Cd and Cu might increase the incidence of hypertension. Further studies involving larger population should be conducted to confirm these findings.
Collapse
Affiliation(s)
- Qi Zhong
- Department of Epidemiology and Biostatistics Statistics, School of Public Health, Anhui Medical University, No. 81 Meishan Road, Shushan Districts, Hefei, 230032, Anhui, People's Republic of China
| | - Chun-Xiao Jiang
- Department of Epidemiology and Biostatistics Statistics, School of Public Health, Anhui Medical University, No. 81 Meishan Road, Shushan Districts, Hefei, 230032, Anhui, People's Republic of China
| | - Chi Zhang
- Department of Epidemiology and Biostatistics Statistics, School of Public Health, Anhui Medical University, No. 81 Meishan Road, Shushan Districts, Hefei, 230032, Anhui, People's Republic of China
| | - Qian Zhang
- Department of Epidemiology and Biostatistics Statistics, School of Public Health, Anhui Medical University, No. 81 Meishan Road, Shushan Districts, Hefei, 230032, Anhui, People's Republic of China
| | - Qi-Rong Qin
- Ma'anshan Center for Disease Control and Provention, Ma'anshan, Anhui, People's Republic of China
| | - Xiao-Dong Wang
- Yian Center for Disease Control and Provention, Tongling, Anhui, People's Republic of China
| | - Fen Huang
- Department of Epidemiology and Biostatistics Statistics, School of Public Health, Anhui Medical University, No. 81 Meishan Road, Shushan Districts, Hefei, 230032, Anhui, People's Republic of China.
- Central Laboratory of Preventive Medicine, School of Public Health, Anhui Medical University, Hefei, 230032, Anhui, People's Republic of China.
| |
Collapse
|
68
|
Dikalov SI, Dikalova AE. Crosstalk Between Mitochondrial Hyperacetylation and Oxidative Stress in Vascular Dysfunction and Hypertension. Antioxid Redox Signal 2019; 31:710-721. [PMID: 30618267 PMCID: PMC6708267 DOI: 10.1089/ars.2018.7632] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Significance: Vascular dysfunction plays a key role in the development of arteriosclerosis, heart disease, and hypertension, which causes one-third of deaths worldwide. Vascular oxidative stress and metabolic disorders contribute to vascular dysfunction, leading to impaired vasorelaxation, vascular hypertrophy, fibrosis, and aortic stiffening. Mitochondria are critical in the regulation of metabolic and antioxidant functions; therefore, mitochondria-targeted treatments could be beneficial. Recent Advances: Vascular dysfunction is crucial in hypertension pathophysiology and exhibits bidirectional relationship. Metabolic disorders and oxidative stress contribute to the pathogenesis of vascular dysfunction and hypertension, which are associated with mitochondrial impairment and hyperacetylation. Mitochondrial deacetylase Sirtuin 3 (Sirt3) is critical in the regulation of metabolic and antioxidant functions. Clinical studies show that cardiovascular disease risk factors reduce Sirt3 level and Sirt3 declines with age, paralleling the increased incidence of cardiovascular disease and hypertension. An imbalance between mitochondrial acetylation and reduced Sirt3 activity contributes to mitochondrial dysfunction and oxidative stress. We propose that mitochondrial hyperacetylation drives a vicious cycle between metabolic disorders and mitochondrial oxidative stress, promoting vascular dysfunction and hypertension. Critical Issues: The mechanisms of mitochondrial dysfunction are still obscure in human hypertension. Mitochondrial hyperacetylation and oxidative stress contribute to mitochondrial dysfunction; however, regulation of mitochondrial acetylation, the role of GCN5L1 (acetyl-CoA-binding protein promoting acetyltransferase protein acetylation) acetyltransferase, Sirt3 deacetylase, and acetylation of specific proteins require further investigations. Future Directions: There is an urgent need to define molecular mechanisms and the pathophysiological role of mitochondrial hyperacetylation, identify novel pharmacological targets, and develop therapeutic approaches to reduce this phenomenon.
Collapse
Affiliation(s)
- Sergey I Dikalov
- Department of Medicine, Division of Clinical Pharmacology, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Anna E Dikalova
- Department of Medicine, Division of Clinical Pharmacology, Vanderbilt University Medical Center, Nashville, Tennessee
| |
Collapse
|
69
|
Czigler A, Toth L, Szarka N, Berta G, Amrein K, Czeiter E, Lendvai-Emmert D, Bodo K, Tarantini S, Koller A, Ungvari Z, Buki A, Toth P. Hypertension Exacerbates Cerebrovascular Oxidative Stress Induced by Mild Traumatic Brain Injury: Protective Effects of the Mitochondria-Targeted Antioxidative Peptide SS-31. J Neurotrauma 2019; 36:3309-3315. [PMID: 31266393 DOI: 10.1089/neu.2019.6439] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Traumatic brain injury (TBI) induces cerebrovascular oxidative stress, which is associated with neurovascular uncoupling, autoregulatory dysfunction, and persisting cognitive decline in both pre-clinical models and patients. However, single mild TBI (mTBI), the most frequent form of brain trauma, increases cerebral generation of reactive oxygen species (ROS) only transiently. We hypothesized that comorbid conditions might exacerbate long-term ROS generation in cerebral arteries after mTBI. Because hypertension is the most important cerebrovascular risk factor in populations prone to mild brain trauma, we induced mTBI in normotensive and spontaneously hypertensive rats (SHR) and assessed changes in cytoplasmic and mitochondrial superoxide (O2-) production by confocal microscopy in isolated middle cerebral arteries (MCA) 2 weeks after mTBI using dihydroethidine (DHE) and the mitochondria-targeted redox-sensitive fluorescent indicator dye MitoSox. We found that mTBI induced a significant increase in long-term cytoplasmic and mitochondrial O2- production in MCAs of SHRs and increased expression of the nicotinamide adenine dinucleotide phosphate (NADPH) oxidase subunit Nox4, which were reversed to the normal level by treating the animals with the cell-permeable, mitochondria-targeted antioxidant peptide SS-31 (5.7 mg kg-1 day-1, i.p.). Persistent mTBI-induced oxidative stress in MCAs of SHRs was significantly decreased by inhibiting vascular NADPH oxidase (apocyinin). We propose that hypertension- and mTBI-induced cerebrovascular oxidative stress likely lead to persistent dysregulation of cerebral blood flow (CBF) and cognitive dysfunction, which might be reversed by SS-31 treatment.
Collapse
Affiliation(s)
- Andras Czigler
- Department of Neurosurgery and Szentagothai Research Center, University of Pecs, Medical School, Pecs, Hungary.,Institute for Translational Medicine, Departments of University of Pecs, Medical School, Pecs, Hungary
| | - Luca Toth
- Department of Neurosurgery and Szentagothai Research Center, University of Pecs, Medical School, Pecs, Hungary.,Institute for Translational Medicine, Departments of University of Pecs, Medical School, Pecs, Hungary
| | - Nikolett Szarka
- Institute for Translational Medicine, Departments of University of Pecs, Medical School, Pecs, Hungary
| | - Gergely Berta
- Medical Biology and University of Pecs, Medical School, Pecs, Hungary
| | - Kriszitina Amrein
- Department of Neurosurgery and Szentagothai Research Center, University of Pecs, Medical School, Pecs, Hungary
| | - Endre Czeiter
- Department of Neurosurgery and Szentagothai Research Center, University of Pecs, Medical School, Pecs, Hungary.,Immunology and Biotechnology, University of Pecs, Medical School, Pecs, Hungary
| | - Dominika Lendvai-Emmert
- Department of Neurosurgery and Szentagothai Research Center, University of Pecs, Medical School, Pecs, Hungary
| | - Kornelia Bodo
- Immunology and Biotechnology, University of Pecs, Medical School, Pecs, Hungary
| | - Stefano Tarantini
- Reynolds Oklahoma Center on Aging, Donald W. Reynolds Department of Geriatric Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma
| | - Akos Koller
- Department of Neurosurgery and Szentagothai Research Center, University of Pecs, Medical School, Pecs, Hungary.,Department of Morphology and Physiology, Semmelweis University, Budapest, Hungary.,Sport-Physiology Research Center, University of Physical Education, Budapest, Hungary.,Department of Physiology, New York Medical College, Valhalla, New York
| | - Zoltan Ungvari
- Reynolds Oklahoma Center on Aging, Donald W. Reynolds Department of Geriatric Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma
| | - Andras Buki
- Department of Neurosurgery and Szentagothai Research Center, University of Pecs, Medical School, Pecs, Hungary
| | - Peter Toth
- Department of Neurosurgery and Szentagothai Research Center, University of Pecs, Medical School, Pecs, Hungary.,Institute for Translational Medicine, Departments of University of Pecs, Medical School, Pecs, Hungary.,MTA-PTE Clinical Neuroscience MR Research Group, Pecs, Hungary
| |
Collapse
|
70
|
He J, Liu X, Su C, Wu F, Sun J, Zhang J, Yang X, Zhang C, Zhou Z, Zhang X, Lin X, Tao J. Inhibition of Mitochondrial Oxidative Damage Improves Reendothelialization Capacity of Endothelial Progenitor Cells via SIRT3 (Sirtuin 3)-Enhanced SOD2 (Superoxide Dismutase 2) Deacetylation in Hypertension. Arterioscler Thromb Vasc Biol 2019; 39:1682-1698. [PMID: 31189433 DOI: 10.1161/atvbaha.119.312613] [Citation(s) in RCA: 57] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
OBJECTIVE Dysfunction of endothelial progenitor cells (EPCs) leads to impaired endothelial repair capacity in patients with hypertension, but the mechanisms remain incompletely understood. Mitochondrial oxidative stress is involved in endothelial injury in hypertension. In this study, we aim to investigate the role of mitochondrial oxidative stress in the deficient endothelial reparative capacity of EPCs and identify enhanced SIRT3 (sirtuin 3)-mediated SOD2 (superoxide dismutase 2) deacetylation as a novel endothelial protective mechanism in hypertension. Approach and Results: Hypertension-EPCs displayed increased mitochondrial reactive oxygen species and mitochondrial damage, including loss of mitochondrial membrane potential, abnormal mitochondrial ultrastructure, and mtDNA oxidative injury, which was coincided with impaired in vitro function and in vivo reendothelialization capacity. The harmful effects of hypertension on mitochondrial function of EPCs were in vitro mimicked by angiotensin II coincubation. Scavenging of mitochondrial reactive oxygen species with mitoTEMPO attenuated mitochondrial oxidative damage and rescued reendothelialization capacity. Enzymatic activity and deacetylation level of SOD2 were significantly reduced in hypertension-EPCs, which was accompanied with decreased SIRT3 expression. Knockdown of SIRT3 in EPCs resulted in mitochondrial oxidative damage, hyperacetylation of SOD2, and suppression of reendothelialization capacity. SIRT3 physically interacted with SOD2 and eliminated excess mitochondrial reactive oxygen species, restored mitochondrial function through enhancing SOD2 activity by deacetylation of K68. Upregulation of SIRT3/SOD2 signaling improved reendothelialization capability of EPCs. CONCLUSIONS The present study demonstrated for the first time that mitochondrial oxidative damage because of deficient SIRT3/SOD2 signaling contributes to the decline in reendothelialization capacity of EPCs in hypertension. Maintenance of mitochondrial redox homeostasis in EPCs may be a novel therapeutic target for endothelial injury.
Collapse
Affiliation(s)
- Jiang He
- From the Department of Hypertension and Vascular Disease, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China (J.H., X.L., C.S., F.W., J.S., J.Z., X.Y., C.Z., Z.Z., X.Z., J.T.)
| | - Xing Liu
- From the Department of Hypertension and Vascular Disease, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China (J.H., X.L., C.S., F.W., J.S., J.Z., X.Y., C.Z., Z.Z., X.Z., J.T.)
| | - Chen Su
- From the Department of Hypertension and Vascular Disease, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China (J.H., X.L., C.S., F.W., J.S., J.Z., X.Y., C.Z., Z.Z., X.Z., J.T.)
| | - Fang Wu
- From the Department of Hypertension and Vascular Disease, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China (J.H., X.L., C.S., F.W., J.S., J.Z., X.Y., C.Z., Z.Z., X.Z., J.T.)
| | - Jiapan Sun
- From the Department of Hypertension and Vascular Disease, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China (J.H., X.L., C.S., F.W., J.S., J.Z., X.Y., C.Z., Z.Z., X.Z., J.T.)
| | - Jianning Zhang
- From the Department of Hypertension and Vascular Disease, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China (J.H., X.L., C.S., F.W., J.S., J.Z., X.Y., C.Z., Z.Z., X.Z., J.T.)
| | - Xulong Yang
- From the Department of Hypertension and Vascular Disease, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China (J.H., X.L., C.S., F.W., J.S., J.Z., X.Y., C.Z., Z.Z., X.Z., J.T.)
| | - Chanjuan Zhang
- From the Department of Hypertension and Vascular Disease, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China (J.H., X.L., C.S., F.W., J.S., J.Z., X.Y., C.Z., Z.Z., X.Z., J.T.)
| | - Ziting Zhou
- From the Department of Hypertension and Vascular Disease, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China (J.H., X.L., C.S., F.W., J.S., J.Z., X.Y., C.Z., Z.Z., X.Z., J.T.)
| | - Xiaoyu Zhang
- From the Department of Hypertension and Vascular Disease, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China (J.H., X.L., C.S., F.W., J.S., J.Z., X.Y., C.Z., Z.Z., X.Z., J.T.)
| | - Xiufang Lin
- Department of Cardiology, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, Guangdong, China (X.L.)
| | - Jun Tao
- From the Department of Hypertension and Vascular Disease, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China (J.H., X.L., C.S., F.W., J.S., J.Z., X.Y., C.Z., Z.Z., X.Z., J.T.)
| |
Collapse
|
71
|
Chen S, Wang Y, Zhang H, Chen R, Lv F, Li Z, Jiang T, Lin D, Zhang H, Yang L, Kong X. The Antioxidant MitoQ Protects Against CSE-Induced Endothelial Barrier Injury and Inflammation by Inhibiting ROS and Autophagy in Human Umbilical Vein Endothelial Cells. Int J Biol Sci 2019; 15:1440-1451. [PMID: 31337974 PMCID: PMC6643142 DOI: 10.7150/ijbs.30193] [Citation(s) in RCA: 50] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2018] [Accepted: 01/12/2019] [Indexed: 12/15/2022] Open
Abstract
Chronic obstructive pulmonary disease (COPD) is a common disease characterized by persistent airflow limitation. Pulmonary vascular endothelial barrier injury and inflammation are increasingly considered to be important pathophysiological processes in cigarette smoke extract (CSE)-induced COPD, but the mechanism remains unclear. To identify the cellular mechanism of endothelial barrier injury and inflammation in CSE-treated human umbilical vein endothelial cells (HUVECs), we investigated the effect of the mitochondrion-targeting antioxidant mitoquinone (MitoQ) on endothelial barrier injury and inflammation. We demonstrated that MitoQ restored endothelial barrier integrity by preventing VE-cadherin disassembly and actin cytoskeleton remodeling, as well as decreased inflammation by the NF-κB and NLRP3 inflammasome pathways in endothelial cells. In addition, MitoQ also maintained mitochondrial function by reducing the production of ROS and excess autophagy. Inhibition of autophagy by 3-MA protected against cytotoxicity that was induced by CSE in HUVECs. Overall, our study indicated that mitochondrial damage is a key promoter in the induction of endothelial barrier dysfunction and inflammation by CSE. The protective effect of MitoQ is related to the inhibition of ROS and excess autophagy in CSE-induced HUVEC injury.
Collapse
Affiliation(s)
- Sha Chen
- School of Basic Medical Sciences, Institute of Hypoxia Medicine, Wenzhou Medical University, Wenzhou, Zhejiang 325035, PR China
| | - Yu Wang
- School of Basic Medical Sciences, Institute of Hypoxia Medicine, Wenzhou Medical University, Wenzhou, Zhejiang 325035, PR China
| | - Hailin Zhang
- Department of Children's Respiration, The Second Affiliated Hospital & Yuying Children's Hospital, Wenzhou Medical University, Wenzhou, Zhejiang 325027, PR China
| | - Ran Chen
- School of Basic Medical Sciences, Institute of Hypoxia Medicine, Wenzhou Medical University, Wenzhou, Zhejiang 325035, PR China
| | - Fangfang Lv
- Department of Children's Respiration, The Second Affiliated Hospital & Yuying Children's Hospital, Wenzhou Medical University, Wenzhou, Zhejiang 325027, PR China
| | - Zhengmao Li
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, PR China
| | - Ting Jiang
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, PR China
| | - Daopeng Lin
- Department of Children's Respiration, The Second Affiliated Hospital & Yuying Children's Hospital, Wenzhou Medical University, Wenzhou, Zhejiang 325027, PR China
| | - Hongyu Zhang
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, PR China
| | - Li Yang
- Department of Respiratory Medicine, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325035, PR China
| | - Xiaoxia Kong
- School of Basic Medical Sciences, Institute of Hypoxia Medicine, Wenzhou Medical University, Wenzhou, Zhejiang 325035, PR China
| |
Collapse
|
72
|
Jayeola OC, Oyagbemi AA, Okunlola OI, Olubamiwa O, Omobowale TO, Ajibade TO, Bolaji-Alabi FB, Ogunpolu BS, Falayi OO, Saba AB, Adedapo AA, Yakubu MA, Oluwadun A, Oguntibeju OO. Effect of cocoa powder on hypertension and antioxidant status in uninephrectomized hypertensive rats. Vet World 2019; 13:695-705. [PMID: 32546914 PMCID: PMC7245715 DOI: 10.14202/vetworld.2020.695-705] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2019] [Accepted: 02/12/2020] [Indexed: 11/16/2022] Open
Abstract
BACKGROUND AND AIM High salt diet and uninephrectomy are associated with high blood pressure with attendant cardiovascular disease conditions such as hypertension, renal damage, myocardial infarction, and stroke. The aim of this study was to investigate the beneficial effects of consumption of cocoa and cocoa-containing products in the management of high blood pressure in uninephrectomized hypertensive rats. MATERIALS AND METHODS The effect of cocoa powder on blood pressure, markers of inflammation, oxidative stress, and histopathology were investigated in uninephrectomized animals fed with cocoa feed alone or in combination with a high salt diet. Male rats were randomly divided into five groups: Group A was the control group and fed with normal feed alone, Group B was fed with cocoa feed alone, Group C was fed with high salt diet (8% salt), GroupD was fed with cocoa-feed compounded with 8% salt for 4weeks after uninephrectomy, and GroupE was uninephrectomized rats on a normal diet. The left kidneys of animals in GroupsC, D, and E were removed by surgery. After 4weeks of treatment, the systolic, diastolic, and mean arterial blood pressure was measured. The serum markers of renal damage and oxidative stress were determined. Histological examination was also performed on renal and cardiac tissues. RESULTS Results showed significant increases in biomarkers of oxidative stress, inflammation, and renal damage with a concomitant decrease in antioxidant status in hypertensive uninephrectomized rats. Cocoa feed, however, significantly improved blood pressure and nitric oxide bioavailability, antioxidant status and reduced markers of inflammation and oxidative stress. CONCLUSION These findings show that cocoa powder could be used to maintain blood pressure levels in hypertensive rats through its antioxidant capacity.
Collapse
Affiliation(s)
| | - Ademola Adetokunbo Oyagbemi
- Department of Veterinary Physiology and Biochemistry, Faculty of Veterinary Medicine, University of Ibadan, Ibadan, Nigeria
| | | | | | | | - Temitayo Olabisi Ajibade
- Department of Veterinary Physiology and Biochemistry, Faculty of Veterinary Medicine, University of Ibadan, Ibadan, Nigeria
| | | | - Blessing Seun Ogunpolu
- Department of Veterinary Medicine, Faculty of Veterinary Medicine, University of Ibadan, Nigeria
| | - Olufunke Olubunmi Falayi
- Department of Veterinary Pharmacology and Toxicology, Faculty of Veterinary Medicine, University of Ibadan, Ibadan, Nigeria
| | - Adebowale Benard Saba
- Department of Veterinary Pharmacology and Toxicology, Faculty of Veterinary Medicine, University of Ibadan, Ibadan, Nigeria
| | - Adeolu Alex Adedapo
- Department of Veterinary Pharmacology and Toxicology, Faculty of Veterinary Medicine, University of Ibadan, Ibadan, Nigeria
| | - Momoh Audu Yakubu
- Department of Environmental and Interdisciplinary Sciences, College of Science, Engineering and Technology, Vascular Biology Unit, Center for Cardiovascular Diseases, College of Pharmacy and Health Sciences (COPHS), Texas Southern University, Houston, Texas, USA
| | - Afolabi Oluwadun
- Department of Medical Microbiology, Olabisi Onabanjo University, Sagamu Campus, Sagamu, Nigeria
| | - Oluwafemi Omoniyi Oguntibeju
- Phytomedicine and Phytochemistry Group, Department of Biomedical Sciences, Faculty of Health and Wellness Sciences, Cape Peninsula University of Technology, Bellville 7535, South Africa
| |
Collapse
|
73
|
Knockout of dihydrofolate reductase in mice induces hypertension and abdominal aortic aneurysm via mitochondrial dysfunction. Redox Biol 2019; 24:101185. [PMID: 30954686 PMCID: PMC6451172 DOI: 10.1016/j.redox.2019.101185] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2019] [Revised: 03/15/2019] [Accepted: 03/28/2019] [Indexed: 12/14/2022] Open
Abstract
Hypertension and abdominal aortic aneurysm (AAA) are severe cardiovascular diseases with incompletely defined molecular mechanisms. In the current study we generated dihydrofolate reductase (DHFR) knockout mice for the first time to examine its potential contribution to the development of hypertension and AAA, as well as the underlying molecular mechanisms. Whereas the homozygote knockout mice were embryonically lethal, the heterozygote knockout mice had global reduction in DHFR protein expression and activity. Angiotensin II infusion into these animals resulted in substantially exaggerated elevation in blood pressure and development of AAA, which was accompanied by excessive eNOS uncoupling activity (featured by significantly impaired tetrahydrobiopterin and nitric oxide bioavailability), vascular remodeling (MMP2 activation, medial elastin breakdown and adventitial fibrosis) and inflammation (macrophage infiltration). Importantly, scavenging of mitochondrial reactive oxygen species with Mito-Tempo in vivo completely abrogated development of hypertension and AAA in DHFR knockout mice, indicating a novel role of mitochondria in mediating hypertension and AAA downstream of DHFR deficiency-dependent eNOS uncoupling. These data for the first time demonstrate that targeting DHFR-deficiency driven mitochondrial dysfunction may represent an innovative therapeutic option for the treatment of AAA and hypertension.
Collapse
|
74
|
Pharmacological strategies to lower crosstalk between nicotinamide adenine dinucleotide phosphate (NADPH) oxidase and mitochondria. Biomed Pharmacother 2019; 111:1478-1498. [DOI: 10.1016/j.biopha.2018.11.128] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2018] [Revised: 11/23/2018] [Accepted: 11/27/2018] [Indexed: 02/07/2023] Open
|
75
|
Fiorenza M, Gunnarsson TP, Ehlers TS, Bangsbo J. High-intensity exercise training ameliorates aberrant expression of markers of mitochondrial turnover but not oxidative damage in skeletal muscle of men with essential hypertension. Acta Physiol (Oxf) 2019; 225:e13208. [PMID: 30339318 DOI: 10.1111/apha.13208] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2018] [Revised: 09/18/2018] [Accepted: 10/14/2018] [Indexed: 12/21/2022]
Abstract
AIM To examine whether hypertensive individuals exhibit altered muscle mitochondrial turnover and redox homeostasis compared with healthy normotensive counterparts, and whether the antihypertensive effect of high-intensity exercise training is associated with improved mitochondrial quality and enhanced anti-oxidant defence. METHODS In a cross-sectional and longitudinal parallel design, 24 essential hypertensive (HYP) and 13 healthy normotensive (NORM) men completed 6 weeks of high-intensity interval training (HIIT). Twenty four-hour ambulatory blood pressure, body composition, cardiorespiratory fitness, exercise capacity and skeletal muscle characteristics were examined before and after HIIT. Expression of markers of mitochondrial turnover, anti-oxidant protection and oxidative damage was determined in vastus lateralis muscle biopsies. Muscle protein levels of eNOS and VEGF, and muscle capillarity were also evaluated. RESULTS At baseline, HYP exhibited lower expression of markers of mitochondrial volume/biogenesis, mitochondrial fusion/fission and autophagy along with depressed eNOS expression compared with NORM. Expression of markers of anti-oxidant protection was similar in HYP and NORM, whereas oxidative damage was higher in HYP than in NORM. In HYP, HIIT lowered blood pressure, improved body composition, cardiorespiratory fitness and exercise capacity, up-regulated markers of mitochondrial volume/biogenesis and autophagy and increased eNOS and VEGF protein content. Furthermore, in HYP, HIIT induced divergent responses in markers of mitochondrial fusion and anti-oxidant protection, did not affect markers of mitochondrial fission, and increased apoptotic susceptibility and oxidative damage. CONCLUSION The present results indicate aberrant muscle mitochondrial turnover and augmented oxidative damage in hypertensive individuals. High-intensity exercise training can partly reverse hypertension-related impairments in muscle mitochondrial turnover, but not redox imbalance.
Collapse
Affiliation(s)
- Matteo Fiorenza
- Department of Nutrition, Exercise and Sports University of Copenhagen Copenhagen Denmark
- Department of Neurosciences, Biomedicine and Movement Sciences University of Verona Verona Italy
| | - Thomas P. Gunnarsson
- Department of Nutrition, Exercise and Sports University of Copenhagen Copenhagen Denmark
| | - Thomas S. Ehlers
- Department of Nutrition, Exercise and Sports University of Copenhagen Copenhagen Denmark
| | - Jens Bangsbo
- Department of Nutrition, Exercise and Sports University of Copenhagen Copenhagen Denmark
| |
Collapse
|
76
|
Liu L, He X, Zhao M, Yang S, Wang S, Yu X, Liu J, Zang W. Regulation of DNA methylation and 2-OG/TET signaling by choline alleviated cardiac hypertrophy in spontaneously hypertensive rats. J Mol Cell Cardiol 2019; 128:26-37. [PMID: 30660679 DOI: 10.1016/j.yjmcc.2019.01.011] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/23/2018] [Revised: 12/24/2018] [Accepted: 01/14/2019] [Indexed: 12/17/2022]
Abstract
DNA methylation is a well-defined epigenetic modification that regulates gene transcription. However, the role of DNA methylation in the cardiac hypertrophy seen in hypertension is unclear. This study was performed to investigate genome-wide DNA methylation profiles in spontaneously hypertensive rats (SHRs) and Wistar-Kyoto rats (WKY), and the cardioprotective effect of choline. Eight-week-old male SHRs received intraperitoneal injections of choline (8 mg/kg/day) for 8 weeks. SHRs showed aberrant methylation distribution on chromosomes and genome regions, with decreased methylation levels at CHG and CHH sites. A total of 91,559 differentially methylated regions (DMRs) were detected between SHRs and WKY rats, of which 28,197 were demethylated and 63,362 were methylated. Choline treatment partly restored the DMRs in SHRs, which were related to 131 genes. Gene ontology analysis and Kyoto Encyclopedia of Genes and Genomes analysis of DMRs suggested that choline partly reversed the dysfunctions of biological processes, cellular components and molecular functions in SHRs. Moreover, the inhibition of 2-oxoglutarate accumulation by choline, thereby inhibiting excessive activation of ten-eleven translocation methylcytosine dioxygenase enzymes, may correlate with the beneficial effects of choline on methylation levels, cardiac hypertrophy and cardiac function of SHRs, as indicated by decreased heart rate and blood pressure, and increased ejection fraction and fractional shortening. This study provides the first genome-wide DNA methylation profile of the hypertrophic myocardium of SHRs and suggests a novel role for this epigenetic modification in hypertension. Choline treatment may represent a promising approach for modification of DNA methylation and optimization of the epigenetic profile for antihypertensive therapy.
Collapse
Affiliation(s)
- Longzhu Liu
- Department of Pharmacology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, PR China
| | - Xi He
- Department of Pharmacology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, PR China
| | - Ming Zhao
- Department of Pharmacology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, PR China
| | - Si Yang
- Department of Pharmacology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, PR China
| | - Shengpeng Wang
- Cardiovascular Research Center, School of Basic Medical Sciences, Xi'an Jiaotong University, Xi'an, PR China
| | - Xiaojiang Yu
- Department of Pharmacology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, PR China
| | - Jiankang Liu
- Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, Shaanxi, PR China
| | - Weijin Zang
- Department of Pharmacology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, PR China.
| |
Collapse
|
77
|
Dikalov S, Itani H, Richmond B, Vergeade A, Rahman SMJ, Boutaud O, Blackwell T, Massion PP, Harrison DG, Dikalova A. Tobacco smoking induces cardiovascular mitochondrial oxidative stress, promotes endothelial dysfunction, and enhances hypertension. Am J Physiol Heart Circ Physiol 2019; 316:H639-H646. [PMID: 30608177 DOI: 10.1152/ajpheart.00595.2018] [Citation(s) in RCA: 124] [Impact Index Per Article: 20.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Tobacco smoking is a major risk factor for cardiovascular disease and hypertension. It is associated with the oxidative stress and induces metabolic reprogramming, altering mitochondrial function. We hypothesized that cigarette smoke induces cardiovascular mitochondrial oxidative stress, which contributes to endothelial dysfunction and hypertension. To test this hypothesis, we studied whether the scavenging of mitochondrial H2O2 in transgenic mice expressing mitochondria-targeted catalase (mCAT) attenuates the development of cigarette smoke/angiotensin II-induced mitochondrial oxidative stress and hypertension compared with wild-type mice. Two weeks of exposure of wild-type mice with cigarette smoke increased systolic blood pressure by 17 mmHg, which was similar to the effect of a subpresssor dose of angiotensin II (0.2 mg·kg-1·day-1), leading to a moderate increase to the prehypertensive level. Cigarette smoke exposure and a low dose of angiotensin II cooperatively induced severe hypertension in wild-type mice, but the scavenging of mitochondrial H2O2 in mCAT mice completely prevented the development of hypertension. Cigarette smoke and angiotensin II cooperatively induced oxidation of cardiolipin (a specific biomarker of mitochondrial oxidative stress) in wild-type mice, which was abolished in mCAT mice. Cigarette smoke and angiotensin II impaired endothelium-dependent relaxation and induced superoxide overproduction, which was diminished in mCAT mice. To mimic the tobacco smoke exposure, we used cigarette smoke condensate, which induced mitochondrial superoxide overproduction and reduced endothelial nitric oxide (a hallmark of endothelial dysfunction in hypertension). Western blot experiments indicated that tobacco smoke and angiotensin II reduce the mitochondrial deacetylase sirtuin-3 level and cause hyperacetylation of a key mitochondrial antioxidant, SOD2, which promotes mitochondrial oxidative stress. NEW & NOTEWORTHY This work demonstrates tobacco smoking-induced mitochondrial oxidative stress, which contributes to endothelial dysfunction and development of hypertension. We suggest that the targeting of mitochondrial oxidative stress can be beneficial for treatment of pathological conditions associated with tobacco smoking, such as endothelial dysfunction, hypertension, and cardiovascular diseases.
Collapse
Affiliation(s)
- Sergey Dikalov
- Division of Clinical Pharmacology, Vanderbilt University Medical Center , Nashville, Tennessee
| | - Hana Itani
- Division of Clinical Pharmacology, Vanderbilt University Medical Center , Nashville, Tennessee.,Pharmacology and Toxicology, Faculty of Medicine, American University of Beirut , Beirut , Lebanon
| | - Bradley Richmond
- Division of Allergy, Pulmonary and Critical Care Medicine, Vanderbilt University Medical Center , Nashville, Tennessee
| | - Aurelia Vergeade
- Division of Clinical Pharmacology, Vanderbilt University Medical Center , Nashville, Tennessee
| | - S M Jamshedur Rahman
- Division of Allergy, Pulmonary and Critical Care Medicine, Vanderbilt University Medical Center , Nashville, Tennessee
| | - Olivier Boutaud
- Department of Pharmacology, Vanderbilt University , Nashville, Tennessee
| | - Timothy Blackwell
- Division of Allergy, Pulmonary and Critical Care Medicine, Vanderbilt University Medical Center , Nashville, Tennessee.,Veterans Affairs, Tennessee Valley Healthcare System, Nashville, Tennessee
| | - Pierre P Massion
- Division of Allergy, Pulmonary and Critical Care Medicine, Vanderbilt University Medical Center , Nashville, Tennessee.,Veterans Affairs, Tennessee Valley Healthcare System, Nashville, Tennessee
| | - David G Harrison
- Division of Clinical Pharmacology, Vanderbilt University Medical Center , Nashville, Tennessee
| | - Anna Dikalova
- Division of Clinical Pharmacology, Vanderbilt University Medical Center , Nashville, Tennessee
| |
Collapse
|
78
|
Carlstrom M, Montenegro MF. Therapeutic value of stimulating the nitrate-nitrite-nitric oxide pathway to attenuate oxidative stress and restore nitric oxide bioavailability in cardiorenal disease. J Intern Med 2019; 285:2-18. [PMID: 30039620 DOI: 10.1111/joim.12818] [Citation(s) in RCA: 66] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Cardiovascular disorders including hypertension and associated renal disease are major health problems affecting more than 1.5 billion people worldwide. Apart from nonmodifiable factors such as ageing, family history and gender, both sedentary lifestyle and unhealthy dietary habits are considered as major risk factors. The disorders are interrelated suggesting common pathological pathways. Mechanistically, oxidative stress and compromised function of the nitric oxide synthase (NOS) system leading to endothelial dysfunction and reduction in nitric oxide (NO) bioavailability have been widely implicated and associated with development and progression of disease. New strategies that correct this redox imbalance and increase NO bioactivity may have major clinical implications. The inorganic anions, nitrate and nitrite, are endogenously formed by oxidization of NOS-derived NO, but there are also high amounts of nitrate in our daily diet. In this regard, accumulated evidence over the past two decades demonstrates that these anions can be recycled back to NO and other bioactive nitrogen oxides, thus offering an attractive alternative strategy for therapeutic exploitation. In this review, we describe how dietary stimulation of the nitrate-nitrite-NO pathway affects cardiovascular and renal functions in health and disease via modulation of oxidative stress and NO bioavailability. Clinical studies addressing potential effects on the renal system are still limited, but blood pressure-lowering effects of nitrate supplementation have been demonstrated in healthy and hypertensive subjects as well as in patients with chronic kidney disease. However, larger clinical studies are warranted to reveal whether chronic nitrate treatment can slow-down the progression of cardiorenal disease and associated complications.
Collapse
Affiliation(s)
- M Carlstrom
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - M F Montenegro
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
79
|
Abstract
Advancing age promotes cardiovascular disease (CVD), the leading cause of death in the United States and many developed nations. Two major age-related arterial phenotypes, large elastic artery stiffening and endothelial dysfunction, are independent predictors of future CVD diagnosis and likely are responsible for the development of CVD in older adults. Not limited to traditional CVD, these age-related changes in the vasculature also contribute to other age-related diseases that influence mammalian health span and potential life span. This review explores mechanisms that influence age-related large elastic artery stiffening and endothelial dysfunction at the tissue level via inflammation and oxidative stress and at the cellular level via Klotho and energy-sensing pathways (AMPK [AMP-activated protein kinase], SIRT [sirtuins], and mTOR [mammalian target of rapamycin]). We also discuss how long-term calorie restriction-a health span- and life span-extending intervention-can prevent many of these age-related vascular phenotypes through the prevention of deleterious alterations in these mechanisms. Lastly, we discuss emerging novel mechanisms of vascular aging, including senescence and genomic instability within cells of the vasculature. As the population of older adults steadily expands, elucidating the cellular and molecular mechanisms of vascular dysfunction with age is critical to better direct appropriate and measured strategies that use pharmacological and lifestyle interventions to reduce risk of CVD within this population.
Collapse
Affiliation(s)
- Anthony J. Donato
- University of Utah, Department of Internal Medicine, Division of Geriatrics, Salt Lake City, Utah
- Veterans Affairs Medical Center-Salt Lake City, Geriatrics Research Education and Clinical Center, Salt Lake City, Utah
| | - Daniel R. Machin
- University of Utah, Department of Internal Medicine, Division of Geriatrics, Salt Lake City, Utah
- Veterans Affairs Medical Center-Salt Lake City, Geriatrics Research Education and Clinical Center, Salt Lake City, Utah
| | - Lisa A. Lesniewski
- University of Utah, Department of Internal Medicine, Division of Geriatrics, Salt Lake City, Utah
- Veterans Affairs Medical Center-Salt Lake City, Geriatrics Research Education and Clinical Center, Salt Lake City, Utah
| |
Collapse
|
80
|
Sodium acetate and androgen receptor blockade improve gestational androgen excess-induced deteriorated glucose homeostasis and antioxidant defenses in rats: roles of adenosine deaminase and xanthine oxidase activities. J Nutr Biochem 2018; 62:65-75. [PMID: 30267975 DOI: 10.1016/j.jnutbio.2018.08.018] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2018] [Revised: 08/28/2018] [Accepted: 08/29/2018] [Indexed: 12/26/2022]
Abstract
Nutritional challenges and androgen excess have been implicated in the development of gestational diabetes and poor fetal outcome, but the mechanisms are not well delineated. The effects of short chain fatty acid (SCFA) on glucose dysmetabolism and poor fetal outcome induced by gestational androgen excess is also not known. We tested the hypothesis that blockade of androgen receptor (AR) and suppression of late gestational androgen excess prevents glucose dysmetabolism and poor fetal outcome through suppression of adenosine deaminase (ADA)/xanthine oxidase (XO) pathway. Twenty-four pregnant Wistar rats were treated (sc) with olive oil, testosterone propionate (0.5 mg/kg) singly or in combination with SCFA (sodium acetate; 200 mg/kg; p.o.) or AR blocker (flutamide; 7.5 mg/kg; p.o.) between gestational days 14 and 19. The results showed that late gestational androgen excess led to glucose deregulation, poor fetal outcome, increased plasma and hepatic free fatty acid and lactate dehydrogenase, liver function marker enzymes, malondialdehyde, uric acid, ADA and XO activities. Conversely, gestational androgen excess resulted in reduced body weight gain, visceral adiposity, plasma and hepatic anti-oxidant defenses (glutathione peroxidase, reduced glutathione/glutathione disulphide ratio, glucose-6-phosphate dehydrogenase, adenosine and nitric oxide). However, all these effects were ameliorated by either sodium acetate or flutamide treatment. The study demonstrates that suppression of testosterone by SCFA or AR blockade protects against glucose deregulation and poor fetal outcome by improvement of anti-oxidant defenses and replenishment of hepatic oxidative capacity through suppression of ADA/XO pathway. Hence, utility of SCFA should be encouraged for prevention of glucose dysmetabolism and poor fetal outcome.
Collapse
|
81
|
Carlström M, Lundberg JO, Weitzberg E. Mechanisms underlying blood pressure reduction by dietary inorganic nitrate. Acta Physiol (Oxf) 2018; 224:e13080. [PMID: 29694703 DOI: 10.1111/apha.13080] [Citation(s) in RCA: 57] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2018] [Revised: 03/28/2018] [Accepted: 04/18/2018] [Indexed: 12/20/2022]
Abstract
Nitric oxide (NO) importantly contributes to cardiovascular homeostasis by regulating blood flow and maintaining endothelial integrity. Conversely, reduced NO bioavailability is a central feature during natural ageing and in many cardiovascular disorders, including hypertension. The inorganic anions nitrate and nitrite are endogenously formed after oxidation of NO synthase (NOS)-derived NO and are also present in our daily diet. Knowledge accumulated over the past two decades has demonstrated that these anions can be recycled back to NO and other bioactive nitrogen oxides via serial reductions that involve oral commensal bacteria and various enzymatic systems. Intake of inorganic nitrate, which is predominantly found in green leafy vegetables and beets, has a variety of favourable cardiovascular effects. As hypertension is a major risk factor of morbidity and mortality worldwide, much attention has been paid to the blood pressure reducing effect of inorganic nitrate. Here, we describe how dietary nitrate, via stimulation of the nitrate-nitrite-NO pathway, affects various organ systems and discuss underlying mechanisms that may contribute to the observed blood pressure-lowering effect.
Collapse
Affiliation(s)
- M. Carlström
- Department of Physiology and Pharmacology; Karolinska Institutet; Stockholm Sweden
| | - J. O. Lundberg
- Department of Physiology and Pharmacology; Karolinska Institutet; Stockholm Sweden
| | - E. Weitzberg
- Department of Physiology and Pharmacology; Karolinska Institutet; Stockholm Sweden
| |
Collapse
|
82
|
Dang AK, Chaplin NL, Murtazina DA, Boehm U, Clay CM, Amberg GC. Subplasmalemmal hydrogen peroxide triggers calcium influx in gonadotropes. J Biol Chem 2018; 293:16028-16042. [PMID: 30154243 DOI: 10.1074/jbc.ra118.001830] [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: 01/09/2018] [Revised: 08/14/2018] [Indexed: 01/01/2023] Open
Abstract
Gonadotropin-releasing hormone (GnRH) stimulation of its eponymous receptor on the surface of endocrine anterior pituitary gonadotrope cells (gonadotropes) initiates multiple signaling cascades that culminate in the secretion of luteinizing and follicle-stimulating hormones, which have critical roles in fertility and reproduction. Enhanced luteinizing hormone biosynthesis, a necessary event for ovulation, requires a signaling pathway characterized by calcium influx through L-type calcium channels and subsequent activation of the mitogen-activated protein kinase extracellular signal-regulated kinase (ERK). We previously reported that highly localized subplasmalemmal calcium microdomains produced by L-type calcium channels (calcium sparklets) play an essential part in GnRH-dependent ERK activation. Similar to calcium, reactive oxygen species (ROS) are ubiquitous intracellular signaling molecules whose subcellular localization determines their specificity. To investigate the potential influence of oxidant signaling in gonadotropes, here we examined the impact of ROS generation on L-type calcium channel function. Total internal reflection fluorescence (TIRF) microscopy revealed that GnRH induces spatially restricted sites of ROS generation in gonadotrope-derived αT3-1 cells. Furthermore, GnRH-dependent stimulation of L-type calcium channels required intracellular hydrogen peroxide signaling in these cells and in primary mouse gonadotropes. NADPH oxidase and mitochondrial ROS generation were each necessary for GnRH-mediated stimulation of L-type calcium channels. Congruently, GnRH increased oxidation within subplasmalemmal mitochondria, and L-type calcium channel activity correlated strongly with the presence of adjacent mitochondria. Collectively, our results provide compelling evidence that NADPH oxidase activity and mitochondria-derived hydrogen peroxide signaling play a fundamental role in GnRH-dependent stimulation of L-type calcium channels in anterior pituitary gonadotropes.
Collapse
Affiliation(s)
- An K Dang
- From the Department of Biomedical Sciences, Colorado State University, Fort Collins, Colorado 80523 and
| | - Nathan L Chaplin
- From the Department of Biomedical Sciences, Colorado State University, Fort Collins, Colorado 80523 and
| | - Dilyara A Murtazina
- From the Department of Biomedical Sciences, Colorado State University, Fort Collins, Colorado 80523 and
| | - Ulrich Boehm
- Department of Pharmacology and Toxicology, Center for Molecular Signaling (PZMS), Saarland University School of Medicine, 66421 Homburg, Germany
| | - Colin M Clay
- From the Department of Biomedical Sciences, Colorado State University, Fort Collins, Colorado 80523 and
| | - Gregory C Amberg
- From the Department of Biomedical Sciences, Colorado State University, Fort Collins, Colorado 80523 and
| |
Collapse
|
83
|
Fang G, Hong L, Liu C, Yang Q, Zhang Q, Li Y, Li B, Wu D, Wu W, Shi H. Oxidative status of cardinal ligament in pelvic organ prolapse. Exp Ther Med 2018; 16:3293-3302. [PMID: 30250520 PMCID: PMC6143997 DOI: 10.3892/etm.2018.6633] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2017] [Accepted: 09/22/2017] [Indexed: 12/15/2022] Open
Abstract
Pelvic organ prolapse (POP) is a common and distressing health problem in adult women, but the pathophysiological mechanism is yet to be fully elucidated. Previous studies have indicated that oxidative stress may be associated with POP. Thus, the aim of the present study was to investigate the oxidative status of pelvic supportive tissue in POP and further demonstrate that oxidative stress is associated with the pathogenesis of POP. A total of 60 samples were collected from females undergoing hysterectomy for POP or cervical intraepithelial neoplasia (CIN). This included 16 females with POP II, 24 females with POP III–IV (according to the POP-Q system) and 20 females with CIN II–III as the control group. Immunohistochemistry was utilized to measure the expression of oxidative biomarkers, 8-hydroxydeoxyguanosine (8-OHdG) and 4-hydroxynonenal (4-HNE). Major antioxidative enzymes, mitochondrial superoxide dismutase (MnSOD) and glutathione peroxidase 1 (GPx1) were measured through reverse transcription-quantitative polymerase chain reaction, western blotting and enzyme activity assays. The results demonstrated that in the cardinal ligament, the expression of 8-OHdG and 4-HNE was higher in the POP III–IV group compared with the POP II group and control group. The MnSOD and GPx1 protein level and enzyme activity were lower in the POP III–IV group compared with the POP II or the control group, while the mRNA expression level of MnSOD and GPx1 was increased. In conclusion, oxidative damage is increased in the pelvic supportive ligament of female patients with POP and the antioxidative defense capacity is decreased. These results support previous findings that oxidative stress is involved in the pathogenesis of POP.
Collapse
Affiliation(s)
- Gui Fang
- Department of Obstetrics and Gynecology Ultrasound, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Li Hong
- Department of Obstetrics and Gynecology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Cheng Liu
- Department of Obstetrics and Gynecology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Qing Yang
- Department of Obstetrics and Gynecology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Qifan Zhang
- Department of Obstetrics and Gynecology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Yang Li
- Department of Obstetrics and Gynecology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Bingshu Li
- Department of Obstetrics and Gynecology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Debin Wu
- Department of Obstetrics and Gynecology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Wenying Wu
- Department of Obstetrics and Gynecology Ultrasound, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Hua Shi
- Department of Obstetrics and Gynecology Ultrasound, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| |
Collapse
|
84
|
Li X, Yu Y, Gorshkov B, Haigh S, Bordan Z, Weintraub D, Rudic RD, Chakraborty T, Barman SA, Verin AD, Su Y, Lucas R, Stepp DW, Chen F, Fulton DJR. Hsp70 Suppresses Mitochondrial Reactive Oxygen Species and Preserves Pulmonary Microvascular Barrier Integrity Following Exposure to Bacterial Toxins. Front Immunol 2018; 9:1309. [PMID: 29951058 PMCID: PMC6008539 DOI: 10.3389/fimmu.2018.01309] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2018] [Accepted: 05/25/2018] [Indexed: 01/22/2023] Open
Abstract
Pneumonia is a leading cause of death in children and the elderly worldwide, accounting for 15% of all deaths of children under 5 years old. Streptococcus pneumoniae is a common and aggressive cause of pneumonia and can also contribute to meningitis and sepsis. Despite the widespread use of antibiotics, mortality rates for pneumonia remain unacceptably high in part due to the release of bacterial toxins. Pneumolysin (PLY) is a cholesterol-dependent toxin that is produced by Streptococcus, and it is both necessary and sufficient for the development of the extensive pulmonary permeability edema that underlies acute lung injury. The mechanisms by which PLY disrupts the pulmonary endothelial barrier are not fully understood. Previously, we found that reactive oxygen species (ROS) contribute to the barrier destructive effects of PLY and identified an unexpected but potent role of Hsp70 in suppressing ROS production. The ability of Hsp70 to influence PLY-induced barrier dysfunction is not yet described, and the goal of the current study was to identify whether Hsp70 upregulation is an effective strategy to protect the lung microvascular endothelial barrier from G+ bacterial toxins. Overexpression of Hsp70 via adenovirus-mediated gene transfer attenuated PLY-induced increases in permeability in human lung microvascular endothelial cells (HLMVEC) with no evidence of cytotoxicity. To adopt a more translational approach, we employed a pharmacological approach using geranylgeranylacetone (GGA) to acutely upregulate endogenous Hsp70 expression. Following acute treatment (6 h) with GGA, HLMVECs exposed to PLY displayed improved cell viability and enhanced endothelial barrier function as measured by both Electric Cell-substrate Impedance Sensing (ECIS) and transwell permeability assays compared to control treated cells. PLY promoted increased mitochondrial ROS, decreased mitochondrial oxygen consumption, and increased caspase 3 cleavage and cell death, which were collectively improved in cells pretreated with GGA. In mice, IP pretreatment with GGA 24 h prior to IT administration of PLY resulted in significantly less Evans Blue Dye extravasation compared to vehicle, indicating preserved endothelial barrier integrity and suggesting that the acute upregulation of Hsp70 may be an effective therapeutic approach in the treatment of lung injury associated with pneumonia.
Collapse
Affiliation(s)
- Xueyi Li
- Vascular Biology Center, Medical College of Georgia at Augusta University, Augusta, Georgia
| | - Yanfang Yu
- Vascular Biology Center, Medical College of Georgia at Augusta University, Augusta, Georgia.,Department of Forensic Medicine, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Boris Gorshkov
- Vascular Biology Center, Medical College of Georgia at Augusta University, Augusta, Georgia
| | - Stephen Haigh
- Vascular Biology Center, Medical College of Georgia at Augusta University, Augusta, Georgia
| | - Zsuzsanna Bordan
- Vascular Biology Center, Medical College of Georgia at Augusta University, Augusta, Georgia
| | - Daniel Weintraub
- Vascular Biology Center, Medical College of Georgia at Augusta University, Augusta, Georgia
| | - Radu Daniel Rudic
- Department of Pharmacology and Toxicology, Medical College of Georgia at Augusta University, Augusta, GA, United States
| | - Trinad Chakraborty
- Institute for Medical Microbiology, Justus-Liebig University Giessen, Giessen, Germany
| | - Scott A Barman
- Department of Pharmacology and Toxicology, Medical College of Georgia at Augusta University, Augusta, GA, United States
| | - Alexander D Verin
- Vascular Biology Center, Medical College of Georgia at Augusta University, Augusta, Georgia
| | - Yunchao Su
- Department of Pharmacology and Toxicology, Medical College of Georgia at Augusta University, Augusta, GA, United States
| | - Rudolf Lucas
- Vascular Biology Center, Medical College of Georgia at Augusta University, Augusta, Georgia.,Department of Pharmacology and Toxicology, Medical College of Georgia at Augusta University, Augusta, GA, United States
| | - David W Stepp
- Department of Pharmacology and Toxicology, Medical College of Georgia at Augusta University, Augusta, GA, United States
| | - Feng Chen
- Vascular Biology Center, Medical College of Georgia at Augusta University, Augusta, Georgia.,Department of Forensic Medicine, Nanjing Medical University, Nanjing, Jiangsu, China
| | - David J R Fulton
- Vascular Biology Center, Medical College of Georgia at Augusta University, Augusta, Georgia.,Department of Pharmacology and Toxicology, Medical College of Georgia at Augusta University, Augusta, GA, United States
| |
Collapse
|
85
|
Associations of Annual Ambient Fine Particulate Matter Mass and Components with Mitochondrial DNA Abundance. Epidemiology 2018; 28:763-770. [PMID: 28953603 DOI: 10.1097/ede.0000000000000717] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
BACKGROUND Fine particulate matter (PM2.5) represents a mixture of components with potentially different toxicities. However, little is known about the relative effects of PM2.5 mass and PM2.5 components on mitochondrial DNA (mtDNA) abundance, which may lie on the pathway of PM2.5-associated disease. METHODS We studied 646 elderly male participants in the Normative Aging Study from Greater Boston to investigate associations of long-term exposure to PM2.5 mass and PM2.5 components with mtDNA abundance. We estimated concentrations of pollutants for the 365-day preceding examination at each participant's address using spatial- and temporal-resolved chemical transport models. We measured blood mtDNA abundance using RT-PCR. We applied a shrinkage and selection method (adaptive LASSO) to identify components most predictive of mtDNA abundance, and fit multipollutant linear mixed-effects models with subject-specific intercept to estimate the relative effects of individual PM component. RESULTS MtDNA abundance was negatively associated with PM2.5 mass in the previous year and-after adjusting for PM2.5 mass-several PM2.5 components, including organic carbon, sulfate (marginally), and nitrate. In multipollutant models including as independent variables PM2.5 mass and PM2.5 components selected by LASSO, nitrate was associated with mtDNA abundance. An SD increase in annual PM2.5-associated nitrate was associated with a 0.12 SD (95% confidence intervals [CI] = -0.18, -0.07) decrease in mtDNA abundance. Analyses restricted to PM2.5 annual concentration below the current 1-year U.S. Environmental Protection Agency standard produced similar results. CONCLUSIONS Long-term exposures to PM2.5-associated nitrate were related to decreased mtDNA abundance independent of PM2.5 mass. Mass alone may not fully capture the potential of PM2.5 to oxidize the mitochondrial genome.See video abstract at, http://links.lww.com/EDE/B274.
Collapse
|
86
|
Siasos G, Tsigkou V, Kosmopoulos M, Theodosiadis D, Simantiris S, Tagkou NM, Tsimpiktsioglou A, Stampouloglou PK, Oikonomou E, Mourouzis K, Philippou A, Vavuranakis M, Stefanadis C, Tousoulis D, Papavassiliou AG. Mitochondria and cardiovascular diseases-from pathophysiology to treatment. ANNALS OF TRANSLATIONAL MEDICINE 2018; 6:256. [PMID: 30069458 DOI: 10.21037/atm.2018.06.21] [Citation(s) in RCA: 175] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Mitochondria are the source of cellular energy production and are present in different types of cells. However, their function is especially important for the heart due to the high demands in energy which is achieved through oxidative phosphorylation. Mitochondria form large networks which regulate metabolism and the optimal function is achieved through the balance between mitochondrial fusion and mitochondrial fission. Moreover, mitochondrial function is upon quality control via the process of mitophagy which removes the damaged organelles. Mitochondrial dysfunction is associated with the development of numerous cardiac diseases such as atherosclerosis, ischemia-reperfusion (I/R) injury, hypertension, diabetes, cardiac hypertrophy and heart failure (HF), due to the uncontrolled production of reactive oxygen species (ROS). Therefore, early control of mitochondrial dysfunction is a crucial step in the therapy of cardiac diseases. A number of anti-oxidant molecules and medications have been used but the results are inconsistent among the studies. Eventually, the aim of future research is to design molecules which selectively target mitochondrial dysfunction and restore the capacity of cellular anti-oxidant enzymes.
Collapse
Affiliation(s)
- Gerasimos Siasos
- Department of Cardiology, "Hippokration" General Hospital, National and Kapodistrian University of Athens, School of Medicine, Athens, Greece.,Division of Cardiovascular, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Vasiliki Tsigkou
- Department of Cardiology, "Hippokration" General Hospital, National and Kapodistrian University of Athens, School of Medicine, Athens, Greece
| | - Marinos Kosmopoulos
- Department of Cardiology, "Hippokration" General Hospital, National and Kapodistrian University of Athens, School of Medicine, Athens, Greece
| | - Dimosthenis Theodosiadis
- Department of Cardiology, "Hippokration" General Hospital, National and Kapodistrian University of Athens, School of Medicine, Athens, Greece
| | - Spyridon Simantiris
- Department of Cardiology, "Hippokration" General Hospital, National and Kapodistrian University of Athens, School of Medicine, Athens, Greece
| | - Nikoletta Maria Tagkou
- Department of Cardiology, "Hippokration" General Hospital, National and Kapodistrian University of Athens, School of Medicine, Athens, Greece
| | - Athina Tsimpiktsioglou
- Department of Cardiology, "Hippokration" General Hospital, National and Kapodistrian University of Athens, School of Medicine, Athens, Greece
| | - Panagiota K Stampouloglou
- Department of Cardiology, "Hippokration" General Hospital, National and Kapodistrian University of Athens, School of Medicine, Athens, Greece
| | - Evangelos Oikonomou
- Department of Cardiology, "Hippokration" General Hospital, National and Kapodistrian University of Athens, School of Medicine, Athens, Greece
| | - Konstantinos Mourouzis
- Department of Cardiology, "Hippokration" General Hospital, National and Kapodistrian University of Athens, School of Medicine, Athens, Greece
| | - Anastasios Philippou
- Department of Experimental Physiology, Medical School, National and Kapodistrian University of Athens, Greece
| | - Manolis Vavuranakis
- Department of Cardiology, "Hippokration" General Hospital, National and Kapodistrian University of Athens, School of Medicine, Athens, Greece
| | | | - Dimitris Tousoulis
- Department of Cardiology, "Hippokration" General Hospital, National and Kapodistrian University of Athens, School of Medicine, Athens, Greece
| | - Athanasios G Papavassiliou
- Department of Biological Chemistry, National and Kapodistrian University of Athens, School of Medicine, Athens, Greece
| |
Collapse
|
87
|
Wu W, Jiang S, Zhao Q, Zhang K, Wei X, Zhou T, Liu D, Zhou H, Zhong R, Zeng Q, Cheng L, Miao X, Lu Q. Associations of environmental exposure to metals with the risk of hypertension in China. THE SCIENCE OF THE TOTAL ENVIRONMENT 2018; 622-623:184-191. [PMID: 29216461 DOI: 10.1016/j.scitotenv.2017.11.343] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/11/2017] [Revised: 11/29/2017] [Accepted: 11/29/2017] [Indexed: 06/07/2023]
Abstract
Hypertension contributes largely to the global burden of disease and mortality. Environmental exposure to metals might be a causative factor for hypertension, but the association remains unclear. The present case-control study of 502 hypertension patients and 502 healthy participants aimed to evaluate the potential relationships between the concentrations of 20 metal in urine and the risk of hypertension in a Chinese population. Multivariate logistic analyses adjusted for potential confounders were performed separately considering the effects of single and multi-metal. We found the increasing trends of urinary Fe, Co, Ni, Cu, Zn and Sr quartiles and the decreasing trends of urinary V and Rb quartiles with the ORs for hypertension. These dose-response associations were confirmed in the RCS models and remained robust in the multi-metal model. Urinary Hg quartiles were positively associated with the risk of hypertension in the models of single-metal and multi-metal. Urinary Cd quartiles were inversely associated with the risk of hypertension in the multi-metal model. Besides, modification effects of gender, BMI and smoking status on the associations of the exposure to various metals with the risk of hypertension were also suggested in the subgroup analysis. Our findings suggest that environmental exposure to V, Fe, Co, Ni, Cu, Zn, Rb, Sr, Cd and Hg might be related with the prevalence of hypertension. Further studies with prospective design should be conducted to confirm these findings.
Collapse
Affiliation(s)
- Weixiang Wu
- State Key Laboratory of Environment Health (Incubation), Key Laboratory of Environment and Health, Ministry of Education, Key Laboratory of Environment and Health (Wuhan), Ministry of Environmental Protection, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, #13 Hangkong Road, Wuhan, Hubei 430030, China
| | - Shunli Jiang
- State Key Laboratory of Environment Health (Incubation), Key Laboratory of Environment and Health, Ministry of Education, Key Laboratory of Environment and Health (Wuhan), Ministry of Environmental Protection, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, #13 Hangkong Road, Wuhan, Hubei 430030, China
| | - Qiang Zhao
- State Key Laboratory of Environment Health (Incubation), Key Laboratory of Environment and Health, Ministry of Education, Key Laboratory of Environment and Health (Wuhan), Ministry of Environmental Protection, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, #13 Hangkong Road, Wuhan, Hubei 430030, China
| | - Ke Zhang
- Department of Clinical Laboratory, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, #1277 Jiefang Road, Wuhan, Hubei 430022, China
| | - Xiaoyun Wei
- State Key Laboratory of Environment Health (Incubation), Key Laboratory of Environment and Health, Ministry of Education, Key Laboratory of Environment and Health (Wuhan), Ministry of Environmental Protection, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, #13 Hangkong Road, Wuhan, Hubei 430030, China
| | - Tong Zhou
- State Key Laboratory of Environment Health (Incubation), Key Laboratory of Environment and Health, Ministry of Education, Key Laboratory of Environment and Health (Wuhan), Ministry of Environmental Protection, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, #13 Hangkong Road, Wuhan, Hubei 430030, China
| | - Dayang Liu
- State Key Laboratory of Environment Health (Incubation), Key Laboratory of Environment and Health, Ministry of Education, Key Laboratory of Environment and Health (Wuhan), Ministry of Environmental Protection, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, #13 Hangkong Road, Wuhan, Hubei 430030, China
| | - Hao Zhou
- State Key Laboratory of Environment Health (Incubation), Key Laboratory of Environment and Health, Ministry of Education, Key Laboratory of Environment and Health (Wuhan), Ministry of Environmental Protection, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, #13 Hangkong Road, Wuhan, Hubei 430030, China
| | - Rong Zhong
- State Key Laboratory of Environment Health (Incubation), Key Laboratory of Environment and Health, Ministry of Education, Key Laboratory of Environment and Health (Wuhan), Ministry of Environmental Protection, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, #13 Hangkong Road, Wuhan, Hubei 430030, China
| | - Qiang Zeng
- State Key Laboratory of Environment Health (Incubation), Key Laboratory of Environment and Health, Ministry of Education, Key Laboratory of Environment and Health (Wuhan), Ministry of Environmental Protection, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, #13 Hangkong Road, Wuhan, Hubei 430030, China
| | - Liming Cheng
- Department of Laboratory Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiaoping Miao
- State Key Laboratory of Environment Health (Incubation), Key Laboratory of Environment and Health, Ministry of Education, Key Laboratory of Environment and Health (Wuhan), Ministry of Environmental Protection, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, #13 Hangkong Road, Wuhan, Hubei 430030, China
| | - Qing Lu
- State Key Laboratory of Environment Health (Incubation), Key Laboratory of Environment and Health, Ministry of Education, Key Laboratory of Environment and Health (Wuhan), Ministry of Environmental Protection, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, #13 Hangkong Road, Wuhan, Hubei 430030, China.
| |
Collapse
|
88
|
Evans LC, Dayton A, Yang C, Liu P, Kurth T, Ahn KW, Komas S, Stingo FC, Laud PW, Vannucci M, Liang M, Cowley AW. Transcriptomic analysis reveals inflammatory and metabolic pathways that are regulated by renal perfusion pressure in the outer medulla of Dahl-S rats. Physiol Genomics 2018; 50:440-447. [PMID: 29602296 DOI: 10.1152/physiolgenomics.00034.2018] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Studies exploring the development of hypertension have traditionally been unable to distinguish which of the observed changes are underlying causes from those that are a consequence of elevated blood pressure. In this study, a custom-designed servo-control system was utilized to precisely control renal perfusion pressure to the left kidney continuously during the development of hypertension in Dahl salt-sensitive rats. In this way, we maintained the left kidney at control blood pressure while the right kidney was exposed to hypertensive pressures. As each kidney was exposed to the same circulating factors, differences between them represent changes induced by pressure alone. RNA sequencing analysis identified 1,613 differently expressed genes affected by renal perfusion pressure. Three pathway analysis methods were applied, one a novel approach incorporating arterial pressure as an input variable allowing a more direct connection between the expression of genes and pressure. The statistical analysis proposed several novel pathways by which pressure affects renal physiology. We confirmed the effects of pressure on p-Jnk regulation, in which the hypertensive medullas show increased p-Jnk/Jnk ratios relative to the left (0.79 ± 0.11 vs. 0.53 ± 0.10, P < 0.01, n = 8). We also confirmed pathway predictions of mitochondrial function, in which the respiratory control ratio of hypertensive vs. control mitochondria are significantly reduced (7.9 ± 1.2 vs. 10.4 ± 1.8, P < 0.01, n = 6) and metabolomic profile, in which 14 metabolites differed significantly between hypertensive and control medullas ( P < 0.05, n = 5). These findings demonstrate that subtle differences in the transcriptome can be used to predict functional changes of the kidney as a consequence of pressure elevation.
Collapse
Affiliation(s)
- Louise C Evans
- Department of Physiology, Medical College of Wisconsin , Milwaukee, Wisconsin
| | - Alex Dayton
- Department of Physiology, Medical College of Wisconsin , Milwaukee, Wisconsin
| | - Chun Yang
- Department of Physiology, Medical College of Wisconsin , Milwaukee, Wisconsin
| | - Pengyuan Liu
- Department of Physiology, Medical College of Wisconsin , Milwaukee, Wisconsin.,Center of Systems Molecular Medicine, Medical College of Wisconsin , Milwaukee, Wisconsin
| | - Theresa Kurth
- Department of Physiology, Medical College of Wisconsin , Milwaukee, Wisconsin
| | - Kwang Woo Ahn
- Division of Biostatistics, Medical College of Wisconsin , Milwaukee, Wisconsin
| | - Steve Komas
- Cancer Center, Redox and Bioenergetics Shared Resource, Medical College of Wisconsin , Milwaukee, Wisconsin
| | | | - Purushottam W Laud
- Center for Patient Care and Outcomes Research, Medical College of Wisconsin , Milwaukee, Wisconsin
| | | | - Mingyu Liang
- Department of Physiology, Medical College of Wisconsin , Milwaukee, Wisconsin.,Center of Systems Molecular Medicine, Medical College of Wisconsin , Milwaukee, Wisconsin
| | - Allen W Cowley
- Department of Physiology, Medical College of Wisconsin , Milwaukee, Wisconsin
| |
Collapse
|
89
|
Nguyen EK, Koval OM, Noble P, Broadhurst K, Allamargot C, Wu M, Strack S, Thiel WH, Grumbach IM. CaMKII (Ca 2+/Calmodulin-Dependent Kinase II) in Mitochondria of Smooth Muscle Cells Controls Mitochondrial Mobility, Migration, and Neointima Formation. Arterioscler Thromb Vasc Biol 2018; 38:1333-1345. [PMID: 29599132 DOI: 10.1161/atvbaha.118.310951] [Citation(s) in RCA: 49] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2017] [Accepted: 03/12/2018] [Indexed: 12/19/2022]
Abstract
OBJECTIVE The main objective of this study is to define the mechanisms by which mitochondria control vascular smooth muscle cell (VSMC) migration and impact neointimal hyperplasia. APPROACH AND RESULTS The multifunctional CaMKII (Ca2+/calmodulin-dependent kinase II) in the mitochondrial matrix of VSMC drove a feed-forward circuit with the mitochondrial Ca2+ uniporter (MCU) to promote matrix Ca2+ influx. MCU was necessary for the activation of mitochondrial CaMKII (mtCaMKII), whereas mtCaMKII phosphorylated MCU at the regulatory site S92 that promotes Ca2+ entry. mtCaMKII was necessary and sufficient for platelet-derived growth factor-induced mitochondrial Ca2+ uptake. This effect was dependent on MCU. mtCaMKII and MCU inhibition abrogated VSMC migration and mitochondrial translocation to the leading edge. Overexpression of wild-type MCU, but not MCU S92A, mutant in MCU-/- VSMC rescued migration and mitochondrial mobility. Inhibition of microtubule, but not of actin assembly, blocked mitochondrial mobility. The outer mitochondrial membrane GTPase Miro-1 promotes mitochondrial mobility via microtubule transport but arrests it in subcellular domains of high Ca2+ concentrations. In Miro-1-/- VSMC, mitochondrial mobility and VSMC migration were abolished, and overexpression of mtCaMKII or a CaMKII inhibitory peptide in mitochondria (mtCaMKIIN) had no effect. Consistently, inhibition of mtCaMKII increased and prolonged cytosolic Ca2+ transients. mtCaMKII inhibition diminished phosphorylation of focal adhesion kinase and myosin light chain, leading to reduced focal adhesion turnover and cytoskeletal remodeling. In a transgenic model of selective mitochondrial CaMKII inhibition in VSMC, neointimal hyperplasia was significantly reduced after vascular injury. CONCLUSIONS These findings identify mitochondrial CaMKII as a key regulator of mitochondrial Ca2+ uptake via MCU, thereby controlling mitochondrial translocation and VSMC migration after vascular injury.
Collapse
Affiliation(s)
- Emily K Nguyen
- From the Department of Internal Medicine, Carver College of Medicine (E.K.N., O.M.K., P.N., K.B., W.H.T., I.M.G.).,Interdisciplinary Program in Molecular and Cellular Biology (E.K.N.)
| | - Olha M Koval
- From the Department of Internal Medicine, Carver College of Medicine (E.K.N., O.M.K., P.N., K.B., W.H.T., I.M.G.)
| | - Paige Noble
- From the Department of Internal Medicine, Carver College of Medicine (E.K.N., O.M.K., P.N., K.B., W.H.T., I.M.G.)
| | - Kim Broadhurst
- From the Department of Internal Medicine, Carver College of Medicine (E.K.N., O.M.K., P.N., K.B., W.H.T., I.M.G.)
| | | | - Meng Wu
- Division of Medicinal and Natural Products Chemistry, Department of Pharmaceutical Sciences and Experimental Therapeutics, College of Pharmacy (M.W.).,High Throughput Screening Facility (M.W.).,Department of Biochemistry, Carver College of Medicine (M.W.)
| | - Stefan Strack
- Department of Pharmacology, Carver College of Medicine (S.S.)
| | - William H Thiel
- From the Department of Internal Medicine, Carver College of Medicine (E.K.N., O.M.K., P.N., K.B., W.H.T., I.M.G.).,François Abboud Cardiovascular Research Center (W.H.T., I.M.G.)
| | - Isabella M Grumbach
- From the Department of Internal Medicine, Carver College of Medicine (E.K.N., O.M.K., P.N., K.B., W.H.T., I.M.G.) .,François Abboud Cardiovascular Research Center (W.H.T., I.M.G.).,Iowa City Veterans Affairs Healthcare System (I.M.G.), University of Iowa, Iowa City
| |
Collapse
|
90
|
Abd Rahman N, Ibrahim F, Aeinehvand MM, Yusof R, Madou M. A Microfluidic Lab-on-a-Disc (LOD) for Antioxidant Activities of Plant Extracts. MICROMACHINES 2018; 9:E140. [PMID: 30424074 PMCID: PMC6187507 DOI: 10.3390/mi9040140] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/13/2018] [Revised: 03/15/2018] [Accepted: 03/19/2018] [Indexed: 11/25/2022]
Abstract
Antioxidants are an important substance that can fight the deterioration of free radicals and can easily oxidize when exposed to light. There are many methods to measure the antioxidant activity in a biological sample, for example 2,2-diphenyl-1-picrylhydrazyl (DPPH) antioxidant activity test, which is one of the simplest methods used. Despite its simplicity, the organic solvent that has been used to dilute DPPH is easily evaporated and degraded with respect to light exposure and time. Thus, it needs to be used at the earliest convenient time prior to the experiment. To overcome this issue, a rapid and close system for antioxidant activity is required. In this paper, we introduced the Lab-on-a-Disc (LoD) method that integrates the DPPH antioxidant activity test on a microfluidic compact disc (CD). We used ascorbic acid, quercetin, Areca catechu, Polygonum minus, and Syzygium polyanthum plant extracts to compare the results of our proposed LoD method with the conventional method. Contrasted to the arduous laborious conventional method, our proposed method offer rapid analysis and simple determination of antioxidant. This proposed LoD method for antioxidant activity in plants would be a platform for the further development of antioxidant assay.
Collapse
Affiliation(s)
- Nurhaslina Abd Rahman
- Department of Biomedical Engineering, Faculty of Engineering, University of Malaya, 50603 Kuala Lumpur, Malaysia.
- Centre for Innovation in Medical Engineering, Faculty of Engineering, University of Malaya, 50603 Kuala Lumpur, Malaysia.
| | - Fatimah Ibrahim
- Department of Biomedical Engineering, Faculty of Engineering, University of Malaya, 50603 Kuala Lumpur, Malaysia.
- Centre for Innovation in Medical Engineering, Faculty of Engineering, University of Malaya, 50603 Kuala Lumpur, Malaysia.
| | - Mohammad M Aeinehvand
- Centre for Innovation in Medical Engineering, Faculty of Engineering, University of Malaya, 50603 Kuala Lumpur, Malaysia.
- School of Engineering and Sciences, Tecnologico de Monterrey, Av. Eugenio Garza Sada 2501 Sur, 64849 Monterrey, NL, Mexico.
| | - Rohana Yusof
- Department of Molecular Medicine, Faculty of Medicine, University of Malaya, 50603 Kuala Lumpur, Malaysia.
| | - Marc Madou
- Centre for Innovation in Medical Engineering, Faculty of Engineering, University of Malaya, 50603 Kuala Lumpur, Malaysia.
- Department of Biomedical Engineering, University of California, Irvine, CA 92697, USA.
| |
Collapse
|
91
|
Lai CL, Xing JP, Liu XH, Qi J, Zhao JQ, Ji YR, Yang WX, Yan PJ, Luo CY, Ruan LF. Relationships of Inflammatory Factors and Risk Factors with Different Target Organ Damage in Essential Hypertension Patients. Chin Med J (Engl) 2018; 130:1296-1302. [PMID: 28524828 PMCID: PMC5455038 DOI: 10.4103/0366-6999.206343] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023] Open
Abstract
BACKGROUND Atherosclerosis (AS) is an inflammatory disease. Inflammation was considered to play a role in the whole process of AS. This study aimed to analyze the relationships of inflammatory factors and risk factors with different target organ damages (TOD) in essential hypertension (EH) patients and to explore its clinical significance. METHODS A total of 294 EH patients were selected and divided into four groups according to their conditions of TOD. Forty-eight healthy subjects were selected as control. The clinical biochemical parameters, serum amyloid A, serum tryptase, and lipoprotein-associated phospholipase A2 (Lp-PLA2) in each group were detected, and the related risk factors were also statistically analyzed. RESULTS Fibrinogen (Fbg) was the most significant independent risk factor in acute coronary syndrome (ACS) group (odds ratio [OR]: 22.242, 95% confidence interval [CI]: 6.458-76.609, P< 0.001) with the largest absolute value of the standardized partial regression coefficient B' (b': 1.079). Lp-PLA2 was the most significant independent risk factor in stroke group (OR: 13.699, 95% CI: 5.236-35.837, P< 0.001) with b' = 0.708. Uric acid (UA) was the most significant independent risk factor in renal damage group (OR: 15.307, 95% CI: 4.022-58.250, P< 0.001) with b' = 1.026. CONCLUSIONS Fbg, Lp-PLA2, and UA are the strongest independent risk factors toward the occurrence of ACS, ischemic stroke, and renal damage in EH patients, thus exhibiting the greatest impacts on the occurrence of ACS, ischemic stroke, and renal damage in EH patients, respectively.
Collapse
Affiliation(s)
- Chun-Lin Lai
- Department of Cardiology, Shanxi Provincial People's Hospital, Taiyuan, Shanxi 030012, China
| | - Jin-Ping Xing
- Department of Cardiology, Shanxi Provincial People's Hospital, Taiyuan, Shanxi 030012, China
| | - Xiao-Hong Liu
- Department of Cardiology, Shanxi Provincial People's Hospital, Taiyuan, Shanxi 030012, China
| | - Jie Qi
- Department of Cardiology, Shanxi Provincial People's Hospital, Taiyuan, Shanxi 030012, China
| | - Jian-Qiang Zhao
- Department of Cardiology, Shanxi Provincial People's Hospital, Taiyuan, Shanxi 030012, China
| | - You-Rui Ji
- Department of Cardiology, Shanxi Provincial People's Hospital, Taiyuan, Shanxi 030012, China
| | - Wu-Xiao Yang
- Department of Cardiology, Shanxi Provincial People's Hospital, Taiyuan, Shanxi 030012, China
| | - Pu-Juan Yan
- Postgraduate School of Shanxi Medical University, Taiyuan, Shanxi 030012, China
| | - Chun-Yan Luo
- Postgraduate School of Shanxi Medical University, Taiyuan, Shanxi 030012, China
| | - Lu-Fang Ruan
- Postgraduate School of Shanxi Medical University, Taiyuan, Shanxi 030012, China
| |
Collapse
|
92
|
Valli H, Ahmad S, Jiang AY, Smyth R, Jeevaratnam K, Matthews HR, Huang CLH. Cardiomyocyte ionic currents in intact young and aged murine Pgc-1β -/- atrial preparations. Mech Ageing Dev 2017; 169:1-9. [PMID: 29197478 PMCID: PMC5846848 DOI: 10.1016/j.mad.2017.11.016] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2017] [Revised: 11/11/2017] [Accepted: 11/28/2017] [Indexed: 12/19/2022]
Abstract
Energetically-deficient Pgc-1β−/− murine atria show age-dependent arrhythmia. Voltage clamp studies investigated their underlying membrane current changes. Pgc-1β−/− atria showed reduced inward Na+ currents with normal voltage-dependences. Outward repolarising K+ currents retained normal activation and rectification. A resulting slowed action potential conduction explains the arrhythmic phenotype.
Introduction Recent studies reported that energetically deficient murine Pgc-1β−/− hearts replicate age-dependent atrial arrhythmic phenotypes associated with their corresponding clinical conditions, implicating action potential (AP) conduction slowing consequent upon reduced AP upstroke rates. Materials and methods We tested a hypothesis implicating Na+ current alterations as a mechanism underlying these electrophysiological phenotypes. We applied loose patch-clamp techniques to intact young and aged, WT and Pgc-1β−/−, atrial cardiomyocyte preparations preserving their in vivo extracellular and intracellular conditions. Results and discussion Depolarising steps activated typical voltage-dependent activating and inactivating inward (Na+) currents whose amplitude increased or decreased with the amplitudes of the activating, or preceding inactivating, steps. Maximum values of peak Na+ current were independently influenced by genotype but not age or interacting effects of genotype and age on two-way ANOVA. Neither genotype, nor age, whether independently or interactively, influenced voltages at half-maximal current, or steepness factors, for current activation and inactivation, or time constants for recovery from inactivation following repolarisation. In contrast, delayed outward (K+) currents showed similar activation and rectification properties through all experimental groups. These findings directly demonstrate and implicate reduced Na+ in contrast to unchanged K+ current, as a mechanism for slowed conduction causing atrial arrhythmogenicity in Pgc-1β−/− hearts.
Collapse
Affiliation(s)
- Haseeb Valli
- Physiological Laboratory, University of Cambridge, Downing Street, Cambridge CB2 3EG, United Kingdom
| | - Shiraz Ahmad
- Physiological Laboratory, University of Cambridge, Downing Street, Cambridge CB2 3EG, United Kingdom
| | - Anita Y Jiang
- Physiological Laboratory, University of Cambridge, Downing Street, Cambridge CB2 3EG, United Kingdom
| | - Robert Smyth
- Physiological Laboratory, University of Cambridge, Downing Street, Cambridge CB2 3EG, United Kingdom
| | - Kamalan Jeevaratnam
- Physiological Laboratory, University of Cambridge, Downing Street, Cambridge CB2 3EG, United Kingdom; Faculty of Health and Medical Sciences, University of Surrey, Guildford GU2 7AL, Surrey, United Kingdom; PU-RCSI School of Medicine, Perdana University, 43400 Serdang, Selangor Darul Ehsan, Malaysia
| | - Hugh R Matthews
- Physiological Laboratory, University of Cambridge, Downing Street, Cambridge CB2 3EG, United Kingdom
| | - Christopher L-H Huang
- Physiological Laboratory, University of Cambridge, Downing Street, Cambridge CB2 3EG, United Kingdom; Department of Biochemistry, University of Cambridge, Tennis Court Road, Cambridge CB2 1QW, United Kingdom.
| |
Collapse
|
93
|
Kirkman DL, Muth BJ, Ramick MG, Townsend RR, Edwards DG. Role of mitochondria-derived reactive oxygen species in microvascular dysfunction in chronic kidney disease. Am J Physiol Renal Physiol 2017; 314:F423-F429. [PMID: 29117995 DOI: 10.1152/ajprenal.00321.2017] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
Cardiovascular disease is the leading cause of mortality in chronic kidney disease (CKD). Mitochondrial dysfunction secondary to CKD is a potential source of oxidative stress that may impair vascular function. This study sought to determine if mitochondria-derived reactive oxygen species contribute to microvascular dysfunction in stage 3-5 CKD. Cutaneous vasodilation in response to local heating was assessed in 20 CKD patients [60 ± 13 yr; estimated glomerular filtration rate (eGFR) 46 ± 13 ml·kg-1·1.73 m-2] and 11 matched healthy participants (58 ± 2 yr; eGFR >90 ml·kg-1·1.73 m-2). Participants were instrumented with two microdialysis fibers for the delivery of 1) Ringer solution, and 2) the mitochondria- specific superoxide scavenger MitoTempo. Skin blood flow was measured via laser Doppler flowmetry during standardized local heating (42°C). Cutaneous vascular conductance (CVC) was calculated as a percentage of the maximum conductance achieved with sodium nitroprusside infusion at 43°C. Urinary isofuran/F2-isoprostane ratios were assessed by gas-chromatography mass spectroscopy. Isofuran-to-F2-isoprostane ratios were increased in CKD patients (3.08 ± 0.32 vs. 1.69 ± 0.12 arbitrary units; P < 0.01) indicative of mitochondria-derived oxidative stress. Cutaneous vasodilation was impaired in CKD compared with healthy controls (87 ± 1 vs. 92 ± 1%CVCmax; P < 0.01). Infusion of MitoTempo significantly increased the plateau phase CVC in CKD patients (CKD Ringer vs. CKD MitoTempo: 87 ± 1 vs. 93 ± 1%CVCmax; P < 0.01) to similar levels observed in healthy controls ( P = 0.9). These data provide in vivo evidence that mitochondria-derived reactive oxygen species contribute to microvascular dysfunction in CKD and suggest that mitochondrial dysfunction may be a potential therapeutic target to improve CKD-related vascular dysfunction.
Collapse
Affiliation(s)
- Danielle L Kirkman
- Department of Kinesiology and Applied Physiology, University of Delaware , Newark, Delaware
| | - Bryce J Muth
- Department of Kinesiology and Applied Physiology, University of Delaware , Newark, Delaware
| | - Meghan G Ramick
- Department of Kinesiology and Applied Physiology, University of Delaware , Newark, Delaware
| | - Raymond R Townsend
- Clinical and Translational Research Center, University of Pennsylvania , Philadelphia, Pennsylvania
| | - David G Edwards
- Department of Kinesiology and Applied Physiology, University of Delaware , Newark, Delaware
| |
Collapse
|
94
|
Prestes PR, Charchar FJ. Is there a link between mitochondrial DNA and blood pressure? J Hum Hypertens 2017; 31:761-762. [DOI: 10.1038/jhh.2017.73] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
95
|
Valli H, Ahmad S, Chadda KR, Al-Hadithi ABAK, Grace AA, Jeevaratnam K, Huang CLH. Age-dependent atrial arrhythmic phenotype secondary to mitochondrial dysfunction in Pgc-1β deficient murine hearts. Mech Ageing Dev 2017; 167:30-45. [PMID: 28919427 PMCID: PMC5652526 DOI: 10.1016/j.mad.2017.09.002] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2017] [Revised: 08/24/2017] [Accepted: 09/13/2017] [Indexed: 12/19/2022]
Abstract
INTRODUCTION Ageing and several age-related chronic conditions including obesity, insulin resistance and hypertension are associated with mitochondrial dysfunction and represent independent risk factors for atrial fibrillation (AF). MATERIALS AND METHODS Atrial arrhythmogenesis was investigated in Langendorff-perfused young (3-4 month) and aged (>12 month), wild type (WT) and peroxisome proliferator activated receptor-γ coactivator-1β deficient (Pgc-1β-/-) murine hearts modeling age-dependent chronic mitochondrial dysfunction during regular pacing and programmed electrical stimulation (PES). RESULTS AND DISCUSSION The Pgc-1β-/- genotype was associated with a pro-arrhythmic phenotype progressing with age. Young and aged Pgc-1β-/- hearts showed compromised maximum action potential (AP) depolarization rates, (dV/dt)max, prolonged AP latencies reflecting slowed action potential (AP) conduction, similar effective refractory periods and baseline action potential durations (APD90) but shortened APD90 in APs in response to extrasystolic stimuli at short stimulation intervals. Electrical properties of APs triggering arrhythmia were similar in WT and Pgc-1β-/- hearts. Pgc-1β-/- hearts showed accelerated age-dependent fibrotic change relative to WT, with young Pgc-1β-/- hearts displaying similar fibrotic change as aged WT, and aged Pgc-1β-/- hearts the greatest fibrotic change. Mitochondrial deficits thus result in an arrhythmic substrate, through slowed AP conduction and altered repolarisation characteristics, arising from alterations in electrophysiological properties and accelerated structural change.
Collapse
Affiliation(s)
- Haseeb Valli
- Physiological Laboratory, University of Cambridge, Downing Street, Cambridge, CB2 3EG, United Kingdom
| | - Shiraz Ahmad
- Physiological Laboratory, University of Cambridge, Downing Street, Cambridge, CB2 3EG, United Kingdom
| | - Karan R Chadda
- Physiological Laboratory, University of Cambridge, Downing Street, Cambridge, CB2 3EG, United Kingdom; Faculty of Health and Medical Sciences, University of Surrey, GU2 7AL, Guildford, Surrey, United Kingdom
| | - Ali B A K Al-Hadithi
- Physiological Laboratory, University of Cambridge, Downing Street, Cambridge, CB2 3EG, United Kingdom
| | - Andrew A Grace
- Department of Biochemistry, University of Cambridge, Tennis Court Road, Cambridge, CB2 1QW, United Kingdom
| | - Kamalan Jeevaratnam
- Physiological Laboratory, University of Cambridge, Downing Street, Cambridge, CB2 3EG, United Kingdom; Faculty of Health and Medical Sciences, University of Surrey, GU2 7AL, Guildford, Surrey, United Kingdom; PU-RCSI School of Medicine, Perdana University, 43400, Serdang, Selangor Darul Ehsan, Malaysia
| | - Christopher L-H Huang
- Physiological Laboratory, University of Cambridge, Downing Street, Cambridge, CB2 3EG, United Kingdom; Department of Biochemistry, University of Cambridge, Tennis Court Road, Cambridge, CB2 1QW, United Kingdom.
| |
Collapse
|
96
|
da Costa CA, Ognibene DT, Cordeiro VSC, de Bem GF, Santos IB, Soares RA, de Melo Cunha LL, Carvalho LCR, de Moura RS, Resende AC. Effect of Euterpe oleracea Mart. Seeds Extract on Chronic Ischemic Renal Injury in Renovascular Hypertensive Rats. J Med Food 2017. [DOI: 10.1089/jmf.2017.0011] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Affiliation(s)
- Cristiane Aguiar da Costa
- Department of Pharmacology, Institute of Biology, Rio de Janeiro State University, Rio de Janeiro, Brazil
| | - Dayane Teixeira Ognibene
- Department of Pharmacology, Institute of Biology, Rio de Janeiro State University, Rio de Janeiro, Brazil
| | | | - Graziele Freitas de Bem
- Department of Pharmacology, Institute of Biology, Rio de Janeiro State University, Rio de Janeiro, Brazil
| | - Izabelle Barcellos Santos
- Department of Pharmacology, Institute of Biology, Rio de Janeiro State University, Rio de Janeiro, Brazil
| | - Ricardo Andrade Soares
- Department of Pharmacology, Institute of Biology, Rio de Janeiro State University, Rio de Janeiro, Brazil
| | - Letícia L. de Melo Cunha
- Department of Pharmacology, Institute of Biology, Rio de Janeiro State University, Rio de Janeiro, Brazil
| | - Lenize C. R.M. Carvalho
- Department of Pharmacology, Institute of Biology, Rio de Janeiro State University, Rio de Janeiro, Brazil
| | - Roberto Soares de Moura
- Department of Pharmacology, Institute of Biology, Rio de Janeiro State University, Rio de Janeiro, Brazil
| | - Angela Castro Resende
- Department of Pharmacology, Institute of Biology, Rio de Janeiro State University, Rio de Janeiro, Brazil
| |
Collapse
|
97
|
Priya LB, Baskaran R, Huang CY, Padma VV. Neferine ameliorates cardiomyoblast apoptosis induced by doxorubicin: possible role in modulating NADPH oxidase/ROS-mediated NFκB redox signaling cascade. Sci Rep 2017; 7:12283. [PMID: 28947826 PMCID: PMC5612945 DOI: 10.1038/s41598-017-12060-9] [Citation(s) in RCA: 79] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2017] [Accepted: 08/29/2017] [Indexed: 12/04/2022] Open
Abstract
Doxorubicin (DOX) mediated cardiomyopathy is a major challenge in cancer chemotherapy. Redox-cycling of doxorubicin by flavoenzymes makes the heart more vulnerable to oxidative stress leading to cardiac dysfunction. The present study evaluates the role of neferine, a bisbenzylisoquinoline alkaloid, in curbing the molecular consequences of DOX-exposure in H9c2 cardiomyoblasts. Neferine pre-treatment increased cell viability upon DOX-exposure. DOX activates NADPH oxidase subunits, (p22phox, p47phox, gp91phox) as the primary event followed by peak in [Ca2+]i accumulation by 2 h, ROS by 3 h and activated ERK1/2 and p38 MAPKinases, time dependently along with the activation and translocation of NFκB and up-regulated COX2 and TNF-α expressions. Neferine pre-treatment modulated NADPH oxidase/ROS system, inhibited MAPKinases and NFκB activation, reduced sub G1 cell population and concomitantly increased cyclin D1 expression reducing DOX-mediated apoptosis. The study demonstrates for the first time, the molecular sequential events behind DOX toxicity and the mechanism of protection offered by neferine with specific relevance to NADPH oxidase system, MAPKinases, inflammation and apoptosis in H9c2 cells. Our data suggests the use of neferine as a new approach in pharmacological interventions against cardiovascular disorders as secondary complications.
Collapse
Affiliation(s)
- Lohanathan Bharathi Priya
- Translational Research Laboratory, Department of Biotechnology, School of Biotechnology and Genetic Engineering, Bharathiar University, Coimbatore, 641046, Tamil Nadu, India
| | - Rathinasamy Baskaran
- National Institute of Cancer Research, National Health Research Institutes, Zhunan, Miaoli County, Taiwan
| | - Chih-Yang Huang
- Graduate Institute of Basic Medical Science, China Medical University, Taichung, Taiwan
- Graduate Institute of Chinese Medical Science, China Medical University, Taichung, Taiwan
- Department of Health and Nutrition Biotechnology, Asia University, Taichung, Taiwan
| | - Viswanadha Vijaya Padma
- Translational Research Laboratory, Department of Biotechnology, School of Biotechnology and Genetic Engineering, Bharathiar University, Coimbatore, 641046, Tamil Nadu, India.
- Graduate Institute of Basic Medical Science, China Medical University, Taichung, Taiwan.
- Department of Health and Nutrition Biotechnology, Asia University, Taichung, Taiwan.
| |
Collapse
|
98
|
Togliatto G, Lombardo G, Brizzi MF. The Future Challenge of Reactive Oxygen Species (ROS) in Hypertension: From Bench to Bed Side. Int J Mol Sci 2017; 18:ijms18091988. [PMID: 28914782 PMCID: PMC5618637 DOI: 10.3390/ijms18091988] [Citation(s) in RCA: 66] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2017] [Revised: 09/07/2017] [Accepted: 09/13/2017] [Indexed: 02/07/2023] Open
Abstract
Reactive oxygen species (ROS) act as signaling molecules that control physiological processes, including cell adaptation to stress. Redox signaling via ROS has quite recently become the focus of much attention in numerous pathological contexts, including neurodegenerative diseases, kidney and cardiovascular disease. Imbalance in ROS formation and degradation has also been implicated in essential hypertension. Essential hypertension is characterized by multiple genetic and environmental factors which do not completely explain its associated risk factors. Thereby, even if advances in therapy have led to a significant reduction in hypertension-associated complications, to interfere with the unbalance of redox signals might represent an additional therapeutic challenge. The decrease of nitric oxide (NO) levels, the antioxidant activity commonly found in preclinical models of hypertension and the ability of antioxidant approaches to reduce ROS levels have spurred clinicians to investigate the contribution of ROS in humans. Indeed, particular effort has recently been devoted to understanding how redox signaling may contribute to vascular pathobiology in human hypertension. However, although biomarkers of oxidative stress have been found to positively correlate with blood pressure in preclinical model of hypertension, human data are less convincing. We herein provide an overview of the most relevant mechanisms via which oxidative stress might contribute to the pathophysiology of essential hypertension. Moreover, alternative approaches, which are directed towards improving antioxidant machinery and/or interfering with ROS production, are also discussed.
Collapse
Affiliation(s)
- Gabriele Togliatto
- Department of Medical Sciences, University of Torino, 10126 Torino, Italy.
| | - Giusy Lombardo
- Department of Medical Sciences, University of Torino, 10126 Torino, Italy.
| | | |
Collapse
|
99
|
Martínez-Revelles S, García-Redondo AB, Avendaño MS, Varona S, Palao T, Orriols M, Roque FR, Fortuño A, Touyz RM, Martínez-González J, Salaices M, Rodríguez C, Briones AM. Lysyl Oxidase Induces Vascular Oxidative Stress and Contributes to Arterial Stiffness and Abnormal Elastin Structure in Hypertension: Role of p38MAPK. Antioxid Redox Signal 2017; 27:379-397. [PMID: 28010122 PMCID: PMC5563924 DOI: 10.1089/ars.2016.6642] [Citation(s) in RCA: 86] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/29/2016] [Revised: 12/22/2016] [Accepted: 12/22/2016] [Indexed: 12/19/2022]
Abstract
AIMS Vascular stiffness, structural elastin abnormalities, and increased oxidative stress are hallmarks of hypertension. Lysyl oxidase (LOX) is an elastin crosslinking enzyme that produces H2O2 as a by-product. We addressed the interplay between LOX, oxidative stress, vessel stiffness, and elastin. RESULTS Angiotensin II (Ang II)-infused hypertensive mice and spontaneously hypertensive rats (SHR) showed increased vascular LOX expression and stiffness and an abnormal elastin structure. Mice over-expressing LOX in vascular smooth muscle cells (TgLOX) exhibited similar mechanical and elastin alterations to those of hypertensive models. LOX inhibition with β-aminopropionitrile (BAPN) attenuated mechanical and elastin alterations in TgLOX mice, Ang II-infused mice, and SHR. Arteries from TgLOX mice, Ang II-infused mice, and/or SHR exhibited increased vascular H2O2 and O2.- levels, NADPH oxidase activity, and/or mitochondrial dysfunction. BAPN prevented the higher oxidative stress in hypertensive models. Treatment of TgLOX and Ang II-infused mice and SHR with the mitochondrial-targeted superoxide dismutase mimetic mito-TEMPO, the antioxidant apocynin, or the H2O2 scavenger polyethylene glycol-conjugated catalase (PEG-catalase) reduced oxidative stress, vascular stiffness, and elastin alterations. Vascular p38 mitogen-activated protein kinase (p38MAPK) activation was increased in Ang II-infused and TgLOX mice and this effect was prevented by BAPN, mito-TEMPO, or PEG-catalase. SB203580, the p38MAPK inhibitor, normalized vessel stiffness and elastin structure in TgLOX mice. INNOVATION We identify LOX as a novel source of vascular reactive oxygen species and a new pathway involved in vascular stiffness and elastin remodeling in hypertension. CONCLUSION LOX up-regulation is associated with enhanced oxidative stress that promotes p38MAPK activation, elastin structural alterations, and vascular stiffness. This pathway contributes to vascular abnormalities in hypertension. Antioxid. Redox Signal. 27, 379-397.
Collapse
Affiliation(s)
- Sonia Martínez-Revelles
- Departamento de Farmacología, Universidad Autónoma de Madrid (UAM), Instituto de Investigación Hospital Universitario La Paz (IdiPAZ), Madrid, Spain
- CIBER de Enfermedades Cardiovasculares, Spain
| | - Ana B. García-Redondo
- Departamento de Farmacología, Universidad Autónoma de Madrid (UAM), Instituto de Investigación Hospital Universitario La Paz (IdiPAZ), Madrid, Spain
- CIBER de Enfermedades Cardiovasculares, Spain
| | - María S. Avendaño
- Departamento de Farmacología, Universidad Autónoma de Madrid (UAM), Instituto de Investigación Hospital Universitario La Paz (IdiPAZ), Madrid, Spain
| | - Saray Varona
- CIBER de Enfermedades Cardiovasculares, Spain
- Centro de Investigación Cardiovascular (CSIC-ICCC), IIB-Sant Pau, Barcelona, Spain
| | - Teresa Palao
- Departamento de Farmacología, Universidad Autónoma de Madrid (UAM), Instituto de Investigación Hospital Universitario La Paz (IdiPAZ), Madrid, Spain
| | - Mar Orriols
- CIBER de Enfermedades Cardiovasculares, Spain
- Centro de Investigación Cardiovascular (CSIC-ICCC), IIB-Sant Pau, Barcelona, Spain
| | - Fernanda R. Roque
- Departamento de Farmacología, Universidad Autónoma de Madrid (UAM), Instituto de Investigación Hospital Universitario La Paz (IdiPAZ), Madrid, Spain
| | - Ana Fortuño
- Program of Cardiovascular Diseases, Centre for Applied Medical Research, University of Navarra, Pamplona, Spain
| | - Rhian M. Touyz
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, United Kingdom
| | - Jose Martínez-González
- CIBER de Enfermedades Cardiovasculares, Spain
- Centro de Investigación Cardiovascular (CSIC-ICCC), IIB-Sant Pau, Barcelona, Spain
| | - Mercedes Salaices
- Departamento de Farmacología, Universidad Autónoma de Madrid (UAM), Instituto de Investigación Hospital Universitario La Paz (IdiPAZ), Madrid, Spain
- CIBER de Enfermedades Cardiovasculares, Spain
| | - Cristina Rodríguez
- CIBER de Enfermedades Cardiovasculares, Spain
- Centro de Investigación Cardiovascular (CSIC-ICCC), IIB-Sant Pau, Barcelona, Spain
| | - Ana M. Briones
- Departamento de Farmacología, Universidad Autónoma de Madrid (UAM), Instituto de Investigación Hospital Universitario La Paz (IdiPAZ), Madrid, Spain
- CIBER de Enfermedades Cardiovasculares, Spain
| |
Collapse
|
100
|
Wang B, Xiong S, Lin S, Xia W, Li Q, Zhao Z, Wei X, Lu Z, Wei X, Gao P, Liu D, Zhu Z. Enhanced Mitochondrial Transient Receptor Potential Channel, Canonical Type 3-Mediated Calcium Handling in the Vasculature From Hypertensive Rats. J Am Heart Assoc 2017; 6:e005812. [PMID: 28711865 PMCID: PMC5586301 DOI: 10.1161/jaha.117.005812] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/09/2017] [Accepted: 05/11/2017] [Indexed: 12/18/2022]
Abstract
BACKGROUND Mitochondrial Ca2+ homeostasis is fundamental to the regulation of mitochondrial reactive oxygen species (ROS) generation and adenosine triphosphate production. Recently, transient receptor potential channel, canonical type 3 (TRPC3), has been shown to localize to the mitochondria and to play a role in maintaining mitochondrial calcium homeostasis. Inhibition of TRPC3 attenuates vascular calcium influx in spontaneously hypertensive rats (SHRs). However, it remains elusive whether mitochondrial TRPC3 participates in hypertension by increasing mitochondrial calcium handling and ROS production. METHODS AND RESULTS In this study we demonstrated increased TRPC3 expression in purified mitochondria in the vasculature from SHRs, which facilitates enhanced mitochondrial calcium uptake and ROS generation compared with Wistar-Kyoto rats. Furthermore, inhibition of TRPC3 by its specific inhibitor, Pyr3, significantly decreased the vascular mitochondrial ROS production and H2O2 synthesis and increased adenosine triphosphate content. Administration of telmisartan can improve these abnormalities. This beneficial effect was associated with improvement of the mitochondrial respiratory function through recovering the activity of pyruvate dehydrogenase in the vasculature of SHRs. In vivo, chronic administration of telmisartan suppressed TRPC3-mediated excessive mitochondrial ROS generation and vasoconstriction in the vasculature of SHRs. More importantly, TRPC3 knockout mice exhibited significantly ameliorated hypertension through reduction of angiotensin II-induced mitochondrial ROS generation. CONCLUSIONS Together, we give experimental evidence for a potential mechanism by which enhanced TRPC3 activity at the cytoplasmic and mitochondrial levels contributes to redox signaling and calcium dysregulation in the vasculature from SHRs. Angiotensin II or telmisartan can regulate [Ca2+]mito, ROS production, and mitochondrial energy metabolism through targeting TRPC3.
Collapse
MESH Headings
- Adenosine Triphosphate/metabolism
- Angiotensin II Type 1 Receptor Blockers/pharmacology
- Animals
- Antihypertensive Agents/pharmacology
- Benzimidazoles/pharmacology
- Benzoates/pharmacology
- Blood Pressure
- Calcium/metabolism
- Calcium Signaling/drug effects
- Cells, Cultured
- Disease Models, Animal
- Energy Metabolism
- Hypertension/drug therapy
- Hypertension/genetics
- Hypertension/metabolism
- Hypertension/physiopathology
- Male
- Mice, Knockout
- Mitochondria/drug effects
- Mitochondria/metabolism
- Muscle, Smooth, Vascular/drug effects
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/physiopathology
- Myocytes, Smooth Muscle/drug effects
- Myocytes, Smooth Muscle/metabolism
- Oxidation-Reduction
- Rats, Inbred SHR
- Rats, Inbred WKY
- Reactive Oxygen Species/metabolism
- TRPC Cation Channels/genetics
- TRPC Cation Channels/metabolism
- Telmisartan
- Time Factors
- Up-Regulation
- Vasoconstriction
Collapse
Affiliation(s)
- Bin Wang
- Center for Hypertension and Metabolic Diseases, Department of Hypertension and Endocrinology, Daping Hospital, Third Military Medical University Chongqing Institute of Hypertension, Chongqing, China
| | - Shiqiang Xiong
- Center for Hypertension and Metabolic Diseases, Department of Hypertension and Endocrinology, Daping Hospital, Third Military Medical University Chongqing Institute of Hypertension, Chongqing, China
| | - Shaoyang Lin
- Center for Hypertension and Metabolic Diseases, Department of Hypertension and Endocrinology, Daping Hospital, Third Military Medical University Chongqing Institute of Hypertension, Chongqing, China
| | - Weijie Xia
- Center for Hypertension and Metabolic Diseases, Department of Hypertension and Endocrinology, Daping Hospital, Third Military Medical University Chongqing Institute of Hypertension, Chongqing, China
| | - Qiang Li
- Center for Hypertension and Metabolic Diseases, Department of Hypertension and Endocrinology, Daping Hospital, Third Military Medical University Chongqing Institute of Hypertension, Chongqing, China
| | - Zhigang Zhao
- Center for Hypertension and Metabolic Diseases, Department of Hypertension and Endocrinology, Daping Hospital, Third Military Medical University Chongqing Institute of Hypertension, Chongqing, China
| | - Xing Wei
- Center for Hypertension and Metabolic Diseases, Department of Hypertension and Endocrinology, Daping Hospital, Third Military Medical University Chongqing Institute of Hypertension, Chongqing, China
| | - Zongshi Lu
- Center for Hypertension and Metabolic Diseases, Department of Hypertension and Endocrinology, Daping Hospital, Third Military Medical University Chongqing Institute of Hypertension, Chongqing, China
| | - Xiao Wei
- Center for Hypertension and Metabolic Diseases, Department of Hypertension and Endocrinology, Daping Hospital, Third Military Medical University Chongqing Institute of Hypertension, Chongqing, China
| | - Peng Gao
- Center for Hypertension and Metabolic Diseases, Department of Hypertension and Endocrinology, Daping Hospital, Third Military Medical University Chongqing Institute of Hypertension, Chongqing, China
| | - Daoyan Liu
- Center for Hypertension and Metabolic Diseases, Department of Hypertension and Endocrinology, Daping Hospital, Third Military Medical University Chongqing Institute of Hypertension, Chongqing, China
| | - Zhiming Zhu
- Center for Hypertension and Metabolic Diseases, Department of Hypertension and Endocrinology, Daping Hospital, Third Military Medical University Chongqing Institute of Hypertension, Chongqing, China
| |
Collapse
|