1
|
Yan W, Dai YL, Han JX. A-kinase anchoring protein 1: an independent predictor of coronary artery disease. BMC Cardiovasc Disord 2025; 25:156. [PMID: 40055597 PMCID: PMC11887123 DOI: 10.1186/s12872-025-04613-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Accepted: 02/27/2025] [Indexed: 05/13/2025] Open
Abstract
INTRODUCTION Coronary artery disease (CAD) is the leading cause of death worldwide. A-kinase anchoring protein 1 (AKAP1), thought to regulate the function and structure of mitochondria, is enriched in the heart, where it plays a protective role. However, data on the serum AKAP1 concentration levels in patients with CAD are currently lacking. To address this, the serum levels of AKAP1 in patients with CAD were quantified and their predictive ability for CAD was evaluated in this study. METHODS A total of 255 patients referred for coronary angiography were included in this study and classified into two groups (CAD and non-CAD group). A comparative analysis of clinical data and serum AKAP1 concentration levels was performed between the two groups. The patients were then divided into quartiles according to AKAP1 levels. A multivariate logistic regression model was used to assess the independent association of AKAP1 with CAD. RESULTS The CAD group showed a lower AKAP1 concentration than the non-CAD group (P < 0.01). The AKAP1 level was correlated with a history of CAD (P < 0.001). The receiver operator characteristic (ROC) curve analysis showed a low ability of AKAP1 in predicting CAD (area under the ROC curve = 0.649). Finally, in the multivariate logistic regression model with the highest quartile as the reference, the lowest quartile of AKAP1 remained significantly associated with an increased risk for CAD (odds ratio (OR) = 5.677, 95% confidence interval [CI] 1.704 to 18.912, P = 0.005). CONCLUSIONS Our results confirmed that serum AKAP1 levels are inversely associated with CAD and may therefore be used as a marker for CAD prediction. But additional studies are needed to confirm and further elucidate our results.
Collapse
Affiliation(s)
- Wei Yan
- Department of Geriatric Medicine, the First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Yun-Lang Dai
- Department of Cardiology, the First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Jun-Xia Han
- Department of Endocrinology, the First Affiliated Hospital of Soochow University, 188 Shizijie Road, Suzhou City, 215006, Jiangsu Province, People's Republic of China.
| |
Collapse
|
2
|
Chen G, Zhang L, Van Schepdael A, Wang X. Recent Advances in Activation of Endothelial Nitric Oxide Synthase by Natural Products: An Effects and Mechanisms Review. FOOD REVIEWS INTERNATIONAL 2023. [DOI: 10.1080/87559129.2023.2166061] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Affiliation(s)
- Guangxuan Chen
- State Key Laboratory of Food Nutrition and Safety, College of Food Science and Engineering, Tianjin University of Science and Technology, Tianjin, China
| | - Le Zhang
- Department of Chemistry, University of Missouri, Columbia, Missouri, USA
| | - Ann Van Schepdael
- Department of Pharmaceutical and Pharmacological Sciences, Pharmaceutical Analysis, KU Leuven - University of Leuven, Leuven, Belgium
| | - Xu Wang
- State Key Laboratory of Food Nutrition and Safety, College of Food Science and Engineering, Tianjin University of Science and Technology, Tianjin, China
| |
Collapse
|
3
|
Tracy EP, Hughes W, Beare JE, Rowe G, Beyer A, LeBlanc AJ. Aging-Induced Impairment of Vascular Function: Mitochondrial Redox Contributions and Physiological/Clinical Implications. Antioxid Redox Signal 2021; 35:974-1015. [PMID: 34314229 PMCID: PMC8905248 DOI: 10.1089/ars.2021.0031] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Significance: The vasculature responds to the respiratory needs of tissue by modulating luminal diameter through smooth muscle constriction or relaxation. Coronary perfusion, diastolic function, and coronary flow reserve are drastically reduced with aging. This loss of blood flow contributes to and exacerbates pathological processes such as angina pectoris, atherosclerosis, and coronary artery and microvascular disease. Recent Advances: Increased attention has recently been given to defining mechanisms behind aging-mediated loss of vascular function and development of therapeutic strategies to restore youthful vascular responsiveness. The ultimate goal aims at providing new avenues for symptom management, reversal of tissue damage, and preventing or delaying of aging-induced vascular damage and dysfunction in the first place. Critical Issues: Our major objective is to describe how aging-associated mitochondrial dysfunction contributes to endothelial and smooth muscle dysfunction via dysregulated reactive oxygen species production, the clinical impact of this phenomenon, and to discuss emerging therapeutic strategies. Pathological changes in regulation of mitochondrial oxidative and nitrosative balance (Section 1) and mitochondrial dynamics of fission/fusion (Section 2) have widespread effects on the mechanisms underlying the ability of the vasculature to relax, leading to hyperconstriction with aging. We will focus on flow-mediated dilation, endothelial hyperpolarizing factors (Sections 3 and 4), and adrenergic receptors (Section 5), as outlined in Figure 1. The clinical implications of these changes on major adverse cardiac events and mortality are described (Section 6). Future Directions: We discuss antioxidative therapeutic strategies currently in development to restore mitochondrial redox homeostasis and subsequently vascular function and evaluate their potential clinical impact (Section 7). Antioxid. Redox Signal. 35, 974-1015.
Collapse
Affiliation(s)
- Evan Paul Tracy
- Department of Physiology, University of Louisville, Louisville, Kentucky, USA
| | - William Hughes
- Department of Medicine and Physiology, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| | - Jason E Beare
- Cardiovascular Innovation Institute, University of Louisville, Louisville, Kentucky, USA.,Kentucky Spinal Cord Injury Research Center, University of Louisville, Louisville, Kentucky, USA
| | - Gabrielle Rowe
- Department of Physiology, University of Louisville, Louisville, Kentucky, USA
| | - Andreas Beyer
- Department of Medicine and Physiology, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| | - Amanda Jo LeBlanc
- Department of Physiology, University of Louisville, Louisville, Kentucky, USA.,Cardiovascular Innovation Institute, University of Louisville, Louisville, Kentucky, USA
| |
Collapse
|
4
|
Brown OI, Bridge KI, Kearney MT. Nicotinamide Adenine Dinucleotide Phosphate Oxidases in Glucose Homeostasis and Diabetes-Related Endothelial Cell Dysfunction. Cells 2021; 10:cells10092315. [PMID: 34571964 PMCID: PMC8469180 DOI: 10.3390/cells10092315] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Revised: 08/30/2021] [Accepted: 09/01/2021] [Indexed: 12/15/2022] Open
Abstract
Oxidative stress within the vascular endothelium, due to excess generation of reactive oxygen species (ROS), is thought to be fundamental to the initiation and progression of the cardiovascular complications of type 2 diabetes mellitus. The term ROS encompasses a variety of chemical species including superoxide anion (O2•-), hydroxyl radical (OH-) and hydrogen peroxide (H2O2). While constitutive generation of low concentrations of ROS are indispensable for normal cellular function, excess O2•- can result in irreversible tissue damage. Excess ROS generation is catalysed by xanthine oxidase, uncoupled nitric oxide synthases, the mitochondrial electron transport chain and the nicotinamide adenine dinucleotide phosphate (NADPH) oxidases. Amongst enzymatic sources of O2•- the Nox2 isoform of NADPH oxidase is thought to be critical to the oxidative stress found in type 2 diabetes mellitus. In contrast, the transcriptionally regulated Nox4 isoform, which generates H2O2, may fulfil a protective role and contribute to normal glucose homeostasis. This review describes the key roles of Nox2 and Nox4, as well as Nox1 and Nox5, in glucose homeostasis, endothelial function and oxidative stress, with a key focus on how they are regulated in health, and dysregulated in type 2 diabetes mellitus.
Collapse
|
5
|
Azad BJ, Heshmati J, Daneshzad E, Palmowski A. Effects of coffee consumption on arterial stiffness and endothelial function: a systematic review and meta-analysis of randomized clinical trials. Crit Rev Food Sci Nutr 2021; 61:1013-1026. [PMID: 32292049 DOI: 10.1080/10408398.2020.1750343] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
BACKGROUND Endothelial function (EF) and arterial stiffness (AS) are predictors of cardiovascular disease. As previous research concerning the effect of coffee intake on EF and AS was controversial, we conducted a systematic review and meta-analysis to synthesize research. METHODS We performed a systematic search in PubMed, Scopus and Web of Science to find clinical trials investigating the effect of coffee intake on EF or AS up to March 2020.Random-effects models were used to estimate the pooled weighted mean difference (WMD) between intervention and control groups for randomized controlled trials (RCTs). Between study heterogeneity was estimated using Cochran's Q and the I 2-inconsistency index. Internal validity of included randomized trials was determined with the Cochrane Collaboration's tool for assessing the risk of bias. RESULTS Twenty-three articles were included for qualitative and 11 articles for quantitative synthesis. Meta-analysis of 14 RCTs (nine articles) indicated a positive short-term (postprandial) effect of coffee intake on flow-mediated dilation (FMD) as a measure of EF (WMD: 1.93%[95% CI: 1.10-2.75]; I 2= 97.9%). Meta-analysis of three long-term RCTs(two articles) found no such effect on FMD (WMD: -0.08% [-3.82 to 3.66]; I 2= 61.4%).Most short-term information was from studies at low or unclear risk of bias, while the proportion of long-term information from studies at high risk of bias was considerable. CONCLUSION The results from this meta-analysis suggest a beneficial short-term effect of coffee
intake on EF as measured by FMD. However, there might be unfavorable effects on AS. Our findings must be interpreted cautiously as the number of studies were low and included studies had a considerable risk of bias.
Collapse
Affiliation(s)
- Banafsheh Jafari Azad
- Department of Cellular and Molecular Nutrition, School of Nutritional Sciences and Dietetics, Tehran University of Medical Sciences, Tehran, Iran
| | - Javad Heshmati
- Department of Nutritional Science, School of Nutritional Science and Food Technology, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Elnaz Daneshzad
- Department of Community Nutrition, School of Nutritional Sciences and Dietetics, Tehran University of Medical Sciences, Tehran, Iran
- Students' Scientific Research Center (SSRC), Tehran University of Medical Sciences (TUMS), Tehran, Iran
| | - Andriko Palmowski
- Department of Rheumatology and Clinical Immunology, Charité - Universitätsmedizin Berlin, Berlin, Germany
| |
Collapse
|
6
|
He A, Zuo D, Liang X, Guo Y, Suxin L, Xia Y. Hypoglycemia increases endothelial-dependent vasodilation through suppressing phosphorylation at Threonine 495/497 site of endothelial nitric oxide synthase. Microvasc Res 2021; 133:104075. [PMID: 32950484 DOI: 10.1016/j.mvr.2020.104075] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2020] [Revised: 08/06/2020] [Accepted: 09/14/2020] [Indexed: 12/20/2022]
Abstract
OBJECTIVE Phosphorylation plays an essential role in the regulation of endothelial nitric oxide synthase (eNOS) activity. However, the phosphorylation of eNOS under hypoglycemia and whether hypoglycemia changes eNOS activity is unknown. This paper aims to clarify the regulation of eNOS phosphorylation and its activity change under hypoglycemia. METHODS Bovine aortic endothelial cells (BAECs) and Sprague-Dawley rats were treated with hypoglycemia, and the phosphorylation of eNOS was subjected to western blot. Blood nitric oxide (NO) concentration was determined by NO kit and endothelial-dependent vasodilation was detected by multi-wire myograph. RESULTS In both BAECs and rats' thoracic aorta, hypoglycemia induced eNOS phosphorylation decrease specifically on Threonine (Thr) 497. Inhibition of ubiquitination of protein kinase C α subunit (PKCα) reverses the decrease of eNOS phosphorylation in hypoglycemia. Ubiquitinated PKCα can be reversed by AMPK knockdown. In rats, insulin induced hypoglycemia increased the concentration of NO in arterial blood, and progressively enhanced the endothelium-dependent vasodilation of the thoracic and mesenteric aorta. CONCLUSIONS In vitro, the activation of AMPK may lead to the expression of PKCα by regulating ubiquitination, resulting in a decrease in the level of P-eNOS Thr497 phosphorylation under hypoglycemia. In vivo, insulin-induced hypoglycemia produces a beneficial cardiovascular effect on rats.
Collapse
Affiliation(s)
- An He
- Division of Cardiology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Deyu Zuo
- Division of Cardiology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Xiaoxue Liang
- Division of Cardiology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Yongzheng Guo
- Division of Cardiology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Luo Suxin
- Division of Cardiology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China.
| | - Yong Xia
- Division of Cardiology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China; Institute of Life Science, Chongqing Medical University, Chongqing 400016, China.
| |
Collapse
|
7
|
Lin X, Han T, Fan Y, Wu S, Wang F, Wang C. Quercetin improves vascular endothelial function through promotion of autophagy in hypertensive rats. Life Sci 2020; 258:118106. [PMID: 32682916 DOI: 10.1016/j.lfs.2020.118106] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2020] [Revised: 07/01/2020] [Accepted: 07/13/2020] [Indexed: 12/18/2022]
Abstract
AIMS Endothelial dysfunction is a hallmark of hypertension. Herein, we assessed the effect of quercetin, a common dietary antioxidant, on endothelial function of spontaneously hypertensive rats (SHRs), and investigated the underlying molecular mechanisms. MAIN METHODS The Wistar-Kyoto (WKY) and SHR rats were administered vehicle (1% w/v methyl cellulose) or quercetin (10 mg/kg body weight) by oral gavage once a day for 6 weeks. Systolic blood pressure (SBP) and diastolic blood pressure (DBP) were measured with a tail-cuff system. Functional of rat mesenteric arterioles was assessed by the temperature-controlled myograph. A dose-response curve was generated by the cumulative addition of acetylcholine (ACh) or sodium nitroprusside (SNP). NO production in the culture medium was assessed by measuring the concentration of nitrite, a stable metabolite of NO, using a modified Griess reagent. KEY FINDINGS Quercetin improved endothelial function and decreased blood pressure in SHRs. Endothelial autophagy, an important cellular homeostatic process, was increased in the early phase of treatment, and decreased in the late phase of treatment. Quercetin promoted autophagy in cultured endothelial cells under both normal and oxidative stress conditions. Pharmacological inhibition of autophagy aggravated endothelial dysfunction in quercetin-treated endothelial cells under oxidative stress, and attenuated the antihypertensive and endothelial protective effects of quercetin in SHRs. SIGNIFICANCE Quercetin protects endothelial function in hypertensive rats through promotion of autophagy. Thus, autophagy could serve as a potential therapeutic target for hypertension.
Collapse
Affiliation(s)
- Xuemei Lin
- Department of Cardiology, The Second Affiliated Hospital of Xi'an Jiaotong University, No. 157, Xiwu Road, Xi'an 710004, Shaanxi, China; Department of Neurology, The First Hospital of Xi'an, No. 30, Fengxiang Road, South Street, Xi'an 710002, Shaanxi, China
| | - Tuo Han
- Department of Cardiology, The Second Affiliated Hospital of Xi'an Jiaotong University, No. 157, Xiwu Road, Xi'an 710004, Shaanxi, China
| | - Yajie Fan
- Department of Cardiology, The Second Affiliated Hospital of Xi'an Jiaotong University, No. 157, Xiwu Road, Xi'an 710004, Shaanxi, China
| | - Songdi Wu
- Department of Neurology, The First Hospital of Xi'an, No. 30, Fengxiang Road, South Street, Xi'an 710002, Shaanxi, China
| | - Fang Wang
- Department of Neurology, The First Hospital of Xi'an, No. 30, Fengxiang Road, South Street, Xi'an 710002, Shaanxi, China
| | - Congxia Wang
- Department of Cardiology, The Second Affiliated Hospital of Xi'an Jiaotong University, No. 157, Xiwu Road, Xi'an 710004, Shaanxi, China.
| |
Collapse
|
8
|
Downregulation of thioredoxin-1-dependent CD95 S-nitrosation by Sorafenib reduces liver cancer. Redox Biol 2020; 34:101528. [PMID: 32388267 PMCID: PMC7210585 DOI: 10.1016/j.redox.2020.101528] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2020] [Accepted: 03/27/2020] [Indexed: 12/30/2022] Open
Abstract
Hepatocellular carcinoma (HCC) represents 80% of the primary hepatic neoplasms. It is the sixth most frequent neoplasm, the fourth cause of cancer-related death, and 7% of registered malignancies. Sorafenib is the first line molecular targeted therapy for patients in advanced stage of HCC. The present study shows that Sorafenib exerts free radical scavenging properties associated with the downregulation of nuclear factor E2-related factor 2 (Nrf2)-regulated thioredoxin 1 (Trx1) expression in liver cancer cells. The experimental downregulation and/or overexpression strategies showed that Trx1 induced activation of nitric oxide synthase (NOS) type 3 (NOS3) and S-nitrosation (SNO) of CD95 receptor leading to an increase of caspase-8 activity and cell proliferation, as well as reduction of caspase-3 activity in liver cancer cells. In addition, Sorafenib transiently increased mRNA expression and activity of S-nitrosoglutathione reductase (GSNOR) in HepG2 cells. Different experimental models of hepatocarcinogenesis based on the subcutaneous implantation of HepG2 cells in nude mice, as well as the induction of HCC by diethylnitrosamine (DEN) confirmed the relevance of Trx1 downregulation during the proapoptotic and antiproliferative properties induced by Sorafenib. In conclusion, the induction of apoptosis and antiproliferative properties by Sorafenib were related to Trx1 downregulation that appeared to play a relevant role on SNO of NOS3 and CD95 in HepG2 cells. The transient increase of GSNOR might also participate in the deactivation of CD95-dependent proliferative signaling in liver cancer cells. Sorafenib induces mitochondrial ROS generation, but also acts as nucleophilic scavenger. Sorafenib reduces Nrf2-depenent Trx1 expression, and SNO–NOS3 and SNO-CD95 ratios. Sorafenib-related antitumoral in vivo activity involves diminution of Trx1 and SNO-CD95.
Collapse
|
9
|
Chen C, Huang J, Shen J, Bai Q. Quercetin improves endothelial insulin sensitivity in obese mice by inhibiting Drp1 phosphorylation at serine 616 and mitochondrial fragmentation. Acta Biochim Biophys Sin (Shanghai) 2019; 51:1250-1257. [PMID: 31781748 DOI: 10.1093/abbs/gmz127] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2019] [Revised: 09/30/2019] [Accepted: 09/30/2019] [Indexed: 01/20/2023] Open
Abstract
Studies have shown that endothelial insulin resistance induced by oxidative stress contributes to vascular dysfunction in metabolic disorders. Quercetin, a natural antioxidant, has been recently shown to exert protective effects on endothelial function. However, the effects of quercetin on endothelial insulin resistance and its underlying mechanism are unclear. Here, we found that chronic oral treatment of obese mice with quercetin increased vascular endothelial insulin sensitivity, accompanied by alleviated mitochondrial fragmentation as revealed by confocal imaging. In addition, western blot analysis showed that quercetin treatment suppressed the levels of dynamin-related protein 1 (Drp1) and phosphorylation at serine 616 in endothelial cells of obese mice. Mechanistically, quercetin specifically suppressed Drp1 phosphorylation at serine 616, whereas it showed little effects on the Drp1 level and its phosphorylation at serine 637 in cultured endothelial cells under oxidative stress. Furthermore, our results also showed that quercetin suppressed Drp1 phosphorylation at serine 616 by inhibiting PKCδ as revealed by western blot analysis. Knockdown of PKCδ with siRNA alleviated the protective effects of quercetin on endothelial-mitochondrial dynamics and insulin sensitivity. These results suggest that chronic oral treatment with quercetin exerts endothelial protective effects through inhibition of PKCδ and the resultant mitochondrial fragmentation.
Collapse
Affiliation(s)
- Cuirong Chen
- Department of Neurology, Renmin Hospital of Pudong New District, Shanghai 201200, China
| | - Jing Huang
- The Central Hospital of Xuhui District, Shanghai 201231, China
| | - Jian Shen
- Department of Neurology, Renmin Hospital of Pudong New District, Shanghai 201200, China
| | - Qingke Bai
- Department of Neurology, Renmin Hospital of Pudong New District, Shanghai 201200, China
| |
Collapse
|
10
|
Marin W. A-kinase anchoring protein 1 (AKAP1) and its role in some cardiovascular diseases. J Mol Cell Cardiol 2019; 138:99-109. [PMID: 31783032 DOI: 10.1016/j.yjmcc.2019.11.154] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/11/2019] [Revised: 11/08/2019] [Accepted: 11/22/2019] [Indexed: 01/09/2023]
Abstract
A-kinase anchoring proteins (AKAPs) play crucial roles in regulating compartmentalized multi-protein signaling networks related to PKA-mediated phosphorylation. The mitochondrial AKAP - AKAP1 proteins are enriched in heart and play cardiac protective roles. This review aims to thoroughly summarize AKAP1 variants from their sequence features to the structure-function relationships between AKAP1 and its binding partners, as well as the molecular mechanisms of AKAP1 in cardiac hypertrophy, hypoxia-induced myocardial infarction and endothelial cells dysfunction, suggesting AKAP1 as a candidate for cardiovascular therapy.
Collapse
Affiliation(s)
- Wenwen Marin
- Institute for Translational Medicine, Medical Faculty of Qingdao University, Qingdao 266021, China.
| |
Collapse
|
11
|
Higashi Y. Coffee and Endothelial Function: A Coffee Paradox? Nutrients 2019; 11:nu11092104. [PMID: 31487926 PMCID: PMC6770186 DOI: 10.3390/nu11092104] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2019] [Revised: 08/28/2019] [Accepted: 09/01/2019] [Indexed: 02/07/2023] Open
Abstract
Coffee is a popular beverage throughout the world. Coffee contains various chemical compounds (e.g., caffeine, chlorogenic acids, hydroxyhydroquinone, kahweol, cafestol, and complex chemical mixtures). Caffeine is also the most widely consumed pharmacological substance in the world and is included in various beverages (e.g., coffee, tea, soft drinks, and energy drinks), products containing chocolate, and drugs. The effects of coffee and caffeine on cardiovascular diseases remain controversial. It is well known that there are J-curve-type or U-curve-type associations of coffee consumption with cardiovascular events including myocardial infarction and stroke. However, there is little information on the direct and indirect effects of coffee consumption on endothelial function in humans. It is likely that the coffee paradox or caffeine paradox exists the association of coffee intake with cardiovascular diseases, cardiovascular outcomes, and endothelial function. This review focusses on the effects of coffee and caffeine on endothelial function from molecular mechanisms to clinical perspectives.
Collapse
Affiliation(s)
- Yukihito Higashi
- Department of Cardiovascular Regeneration and Medicine, Research Institute for Radiation Biology and Medicine, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima 734-8551, Japan.
- Division of Regeneration and Medicine, Medical Center for Translational and Clinical Research, Hiroshima University Hospital, Hiroshima 734-8551, Japan.
| |
Collapse
|
12
|
Mazrouei S, Sharifpanah F, Caldwell RW, Franz M, Shatanawi A, Muessig J, Fritzenwanger M, Schulze PC, Jung C. Regulation of MAP kinase-mediated endothelial dysfunction in hyperglycemia via arginase I and eNOS dysregulation. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2019; 1866:1398-1411. [DOI: 10.1016/j.bbamcr.2019.05.004] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/18/2018] [Revised: 12/14/2018] [Accepted: 05/20/2019] [Indexed: 12/24/2022]
|
13
|
Effect of eNOS on Ischemic Postconditioning-Induced Autophagy against Ischemia/Reperfusion Injury in Mice. BIOMED RESEARCH INTERNATIONAL 2019; 2019:5201014. [PMID: 30881990 PMCID: PMC6387714 DOI: 10.1155/2019/5201014] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/27/2018] [Accepted: 01/15/2019] [Indexed: 12/21/2022]
Abstract
Autophagy is involved in the development of numerous illnesses, including ischemia/reperfusion (I/R). Endothelial nitric oxide synthase (eNOS) participates in the protective effects of ischemic postconditioning (IPostC). However, it remains unclear whether eNOS-mediated autophagy serves as a critical role in IPostC in the hearts of mice, in protecting against I/R injury. In the present study, the hearts of mice with left anterior descending coronary artery ligation were studied as I/R models. H9c2 cells underwent exposure to hypoxia/reoxygenation (H/R) and were examined as in vitro model. IPostC reduced mice myocardial infarct size and improved the structure of the heart. IPostC increased the formation of autophagosomes and increased the phosphorylation of eNOS and adenosine monophosphate-activated protein kinase (AMPK). Autophagy and eNOS inhibition suppressed the cardioprotective effects of IPostC. AMPK or eNOS inhibition abolished the improvement effect of IPostC on autophagy. AMPK inhibition decreased eNOS phosphorylation in the heart. Additionally, H9c2 cells suffering hypoxia were used as in vitro model. Autophagy or eNOS inhibition abolished the protective effects of hypoxic postconditioning (HPostC) against H/R injury. AMPK and eNOS inhibition/knockout decreased autophagic activity in the HPostC group. These results indicated that IPostC protects the heart against I/R injury, partially via promoting AMPK/eNOS-mediated autophagy.
Collapse
|
14
|
Schiattarella GG, Cattaneo F, Carrizzo A, Paolillo R, Boccella N, Ambrosio M, Damato A, Pironti G, Franzone A, Russo G, Magliulo F, Pirozzi M, Storto M, Madonna M, Gargiulo G, Trimarco V, Rinaldi L, De Lucia M, Garbi C, Feliciello A, Esposito G, Vecchione C, Perrino C. Akap1
Regulates Vascular Function and Endothelial Cells Behavior. Hypertension 2018; 71:507-517. [DOI: 10.1161/hypertensionaha.117.10185] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2017] [Revised: 08/29/2017] [Accepted: 12/14/2017] [Indexed: 11/16/2022]
Affiliation(s)
- Gabriele Giacomo Schiattarella
- From the Department of Advanced Biomedical Sciences (G.G.S., F.C., R.P., N.B., A.F., F.M., G.G., G.E., C.P.), Department of Molecular Medicine and Medical Biotechnologies (G.R., L.R., C.G., A.F.), and Department of Neuroscience, Reproductive Science and Odontostomatology (V.T.), University of Naples “Federico II”, Italy; IRCCS Neuromed, Pozzilli, Italy (A.C., M.A., A.D., M.S., M.M., M.D.L., C.V.); Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden (G.P.); Department
| | - Fabio Cattaneo
- From the Department of Advanced Biomedical Sciences (G.G.S., F.C., R.P., N.B., A.F., F.M., G.G., G.E., C.P.), Department of Molecular Medicine and Medical Biotechnologies (G.R., L.R., C.G., A.F.), and Department of Neuroscience, Reproductive Science and Odontostomatology (V.T.), University of Naples “Federico II”, Italy; IRCCS Neuromed, Pozzilli, Italy (A.C., M.A., A.D., M.S., M.M., M.D.L., C.V.); Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden (G.P.); Department
| | - Albino Carrizzo
- From the Department of Advanced Biomedical Sciences (G.G.S., F.C., R.P., N.B., A.F., F.M., G.G., G.E., C.P.), Department of Molecular Medicine and Medical Biotechnologies (G.R., L.R., C.G., A.F.), and Department of Neuroscience, Reproductive Science and Odontostomatology (V.T.), University of Naples “Federico II”, Italy; IRCCS Neuromed, Pozzilli, Italy (A.C., M.A., A.D., M.S., M.M., M.D.L., C.V.); Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden (G.P.); Department
| | - Roberta Paolillo
- From the Department of Advanced Biomedical Sciences (G.G.S., F.C., R.P., N.B., A.F., F.M., G.G., G.E., C.P.), Department of Molecular Medicine and Medical Biotechnologies (G.R., L.R., C.G., A.F.), and Department of Neuroscience, Reproductive Science and Odontostomatology (V.T.), University of Naples “Federico II”, Italy; IRCCS Neuromed, Pozzilli, Italy (A.C., M.A., A.D., M.S., M.M., M.D.L., C.V.); Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden (G.P.); Department
| | - Nicola Boccella
- From the Department of Advanced Biomedical Sciences (G.G.S., F.C., R.P., N.B., A.F., F.M., G.G., G.E., C.P.), Department of Molecular Medicine and Medical Biotechnologies (G.R., L.R., C.G., A.F.), and Department of Neuroscience, Reproductive Science and Odontostomatology (V.T.), University of Naples “Federico II”, Italy; IRCCS Neuromed, Pozzilli, Italy (A.C., M.A., A.D., M.S., M.M., M.D.L., C.V.); Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden (G.P.); Department
| | - Mariateresa Ambrosio
- From the Department of Advanced Biomedical Sciences (G.G.S., F.C., R.P., N.B., A.F., F.M., G.G., G.E., C.P.), Department of Molecular Medicine and Medical Biotechnologies (G.R., L.R., C.G., A.F.), and Department of Neuroscience, Reproductive Science and Odontostomatology (V.T.), University of Naples “Federico II”, Italy; IRCCS Neuromed, Pozzilli, Italy (A.C., M.A., A.D., M.S., M.M., M.D.L., C.V.); Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden (G.P.); Department
| | - Antonio Damato
- From the Department of Advanced Biomedical Sciences (G.G.S., F.C., R.P., N.B., A.F., F.M., G.G., G.E., C.P.), Department of Molecular Medicine and Medical Biotechnologies (G.R., L.R., C.G., A.F.), and Department of Neuroscience, Reproductive Science and Odontostomatology (V.T.), University of Naples “Federico II”, Italy; IRCCS Neuromed, Pozzilli, Italy (A.C., M.A., A.D., M.S., M.M., M.D.L., C.V.); Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden (G.P.); Department
| | - Gianluigi Pironti
- From the Department of Advanced Biomedical Sciences (G.G.S., F.C., R.P., N.B., A.F., F.M., G.G., G.E., C.P.), Department of Molecular Medicine and Medical Biotechnologies (G.R., L.R., C.G., A.F.), and Department of Neuroscience, Reproductive Science and Odontostomatology (V.T.), University of Naples “Federico II”, Italy; IRCCS Neuromed, Pozzilli, Italy (A.C., M.A., A.D., M.S., M.M., M.D.L., C.V.); Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden (G.P.); Department
| | - Anna Franzone
- From the Department of Advanced Biomedical Sciences (G.G.S., F.C., R.P., N.B., A.F., F.M., G.G., G.E., C.P.), Department of Molecular Medicine and Medical Biotechnologies (G.R., L.R., C.G., A.F.), and Department of Neuroscience, Reproductive Science and Odontostomatology (V.T.), University of Naples “Federico II”, Italy; IRCCS Neuromed, Pozzilli, Italy (A.C., M.A., A.D., M.S., M.M., M.D.L., C.V.); Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden (G.P.); Department
| | - Giusi Russo
- From the Department of Advanced Biomedical Sciences (G.G.S., F.C., R.P., N.B., A.F., F.M., G.G., G.E., C.P.), Department of Molecular Medicine and Medical Biotechnologies (G.R., L.R., C.G., A.F.), and Department of Neuroscience, Reproductive Science and Odontostomatology (V.T.), University of Naples “Federico II”, Italy; IRCCS Neuromed, Pozzilli, Italy (A.C., M.A., A.D., M.S., M.M., M.D.L., C.V.); Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden (G.P.); Department
| | - Fabio Magliulo
- From the Department of Advanced Biomedical Sciences (G.G.S., F.C., R.P., N.B., A.F., F.M., G.G., G.E., C.P.), Department of Molecular Medicine and Medical Biotechnologies (G.R., L.R., C.G., A.F.), and Department of Neuroscience, Reproductive Science and Odontostomatology (V.T.), University of Naples “Federico II”, Italy; IRCCS Neuromed, Pozzilli, Italy (A.C., M.A., A.D., M.S., M.M., M.D.L., C.V.); Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden (G.P.); Department
| | - Marinella Pirozzi
- From the Department of Advanced Biomedical Sciences (G.G.S., F.C., R.P., N.B., A.F., F.M., G.G., G.E., C.P.), Department of Molecular Medicine and Medical Biotechnologies (G.R., L.R., C.G., A.F.), and Department of Neuroscience, Reproductive Science and Odontostomatology (V.T.), University of Naples “Federico II”, Italy; IRCCS Neuromed, Pozzilli, Italy (A.C., M.A., A.D., M.S., M.M., M.D.L., C.V.); Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden (G.P.); Department
| | - Marianna Storto
- From the Department of Advanced Biomedical Sciences (G.G.S., F.C., R.P., N.B., A.F., F.M., G.G., G.E., C.P.), Department of Molecular Medicine and Medical Biotechnologies (G.R., L.R., C.G., A.F.), and Department of Neuroscience, Reproductive Science and Odontostomatology (V.T.), University of Naples “Federico II”, Italy; IRCCS Neuromed, Pozzilli, Italy (A.C., M.A., A.D., M.S., M.M., M.D.L., C.V.); Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden (G.P.); Department
| | - Michele Madonna
- From the Department of Advanced Biomedical Sciences (G.G.S., F.C., R.P., N.B., A.F., F.M., G.G., G.E., C.P.), Department of Molecular Medicine and Medical Biotechnologies (G.R., L.R., C.G., A.F.), and Department of Neuroscience, Reproductive Science and Odontostomatology (V.T.), University of Naples “Federico II”, Italy; IRCCS Neuromed, Pozzilli, Italy (A.C., M.A., A.D., M.S., M.M., M.D.L., C.V.); Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden (G.P.); Department
| | - Giuseppe Gargiulo
- From the Department of Advanced Biomedical Sciences (G.G.S., F.C., R.P., N.B., A.F., F.M., G.G., G.E., C.P.), Department of Molecular Medicine and Medical Biotechnologies (G.R., L.R., C.G., A.F.), and Department of Neuroscience, Reproductive Science and Odontostomatology (V.T.), University of Naples “Federico II”, Italy; IRCCS Neuromed, Pozzilli, Italy (A.C., M.A., A.D., M.S., M.M., M.D.L., C.V.); Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden (G.P.); Department
| | - Valentina Trimarco
- From the Department of Advanced Biomedical Sciences (G.G.S., F.C., R.P., N.B., A.F., F.M., G.G., G.E., C.P.), Department of Molecular Medicine and Medical Biotechnologies (G.R., L.R., C.G., A.F.), and Department of Neuroscience, Reproductive Science and Odontostomatology (V.T.), University of Naples “Federico II”, Italy; IRCCS Neuromed, Pozzilli, Italy (A.C., M.A., A.D., M.S., M.M., M.D.L., C.V.); Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden (G.P.); Department
| | - Laura Rinaldi
- From the Department of Advanced Biomedical Sciences (G.G.S., F.C., R.P., N.B., A.F., F.M., G.G., G.E., C.P.), Department of Molecular Medicine and Medical Biotechnologies (G.R., L.R., C.G., A.F.), and Department of Neuroscience, Reproductive Science and Odontostomatology (V.T.), University of Naples “Federico II”, Italy; IRCCS Neuromed, Pozzilli, Italy (A.C., M.A., A.D., M.S., M.M., M.D.L., C.V.); Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden (G.P.); Department
| | - Massimiliano De Lucia
- From the Department of Advanced Biomedical Sciences (G.G.S., F.C., R.P., N.B., A.F., F.M., G.G., G.E., C.P.), Department of Molecular Medicine and Medical Biotechnologies (G.R., L.R., C.G., A.F.), and Department of Neuroscience, Reproductive Science and Odontostomatology (V.T.), University of Naples “Federico II”, Italy; IRCCS Neuromed, Pozzilli, Italy (A.C., M.A., A.D., M.S., M.M., M.D.L., C.V.); Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden (G.P.); Department
| | - Corrado Garbi
- From the Department of Advanced Biomedical Sciences (G.G.S., F.C., R.P., N.B., A.F., F.M., G.G., G.E., C.P.), Department of Molecular Medicine and Medical Biotechnologies (G.R., L.R., C.G., A.F.), and Department of Neuroscience, Reproductive Science and Odontostomatology (V.T.), University of Naples “Federico II”, Italy; IRCCS Neuromed, Pozzilli, Italy (A.C., M.A., A.D., M.S., M.M., M.D.L., C.V.); Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden (G.P.); Department
| | - Antonio Feliciello
- From the Department of Advanced Biomedical Sciences (G.G.S., F.C., R.P., N.B., A.F., F.M., G.G., G.E., C.P.), Department of Molecular Medicine and Medical Biotechnologies (G.R., L.R., C.G., A.F.), and Department of Neuroscience, Reproductive Science and Odontostomatology (V.T.), University of Naples “Federico II”, Italy; IRCCS Neuromed, Pozzilli, Italy (A.C., M.A., A.D., M.S., M.M., M.D.L., C.V.); Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden (G.P.); Department
| | - Giovanni Esposito
- From the Department of Advanced Biomedical Sciences (G.G.S., F.C., R.P., N.B., A.F., F.M., G.G., G.E., C.P.), Department of Molecular Medicine and Medical Biotechnologies (G.R., L.R., C.G., A.F.), and Department of Neuroscience, Reproductive Science and Odontostomatology (V.T.), University of Naples “Federico II”, Italy; IRCCS Neuromed, Pozzilli, Italy (A.C., M.A., A.D., M.S., M.M., M.D.L., C.V.); Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden (G.P.); Department
| | - Carmine Vecchione
- From the Department of Advanced Biomedical Sciences (G.G.S., F.C., R.P., N.B., A.F., F.M., G.G., G.E., C.P.), Department of Molecular Medicine and Medical Biotechnologies (G.R., L.R., C.G., A.F.), and Department of Neuroscience, Reproductive Science and Odontostomatology (V.T.), University of Naples “Federico II”, Italy; IRCCS Neuromed, Pozzilli, Italy (A.C., M.A., A.D., M.S., M.M., M.D.L., C.V.); Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden (G.P.); Department
| | - Cinzia Perrino
- From the Department of Advanced Biomedical Sciences (G.G.S., F.C., R.P., N.B., A.F., F.M., G.G., G.E., C.P.), Department of Molecular Medicine and Medical Biotechnologies (G.R., L.R., C.G., A.F.), and Department of Neuroscience, Reproductive Science and Odontostomatology (V.T.), University of Naples “Federico II”, Italy; IRCCS Neuromed, Pozzilli, Italy (A.C., M.A., A.D., M.S., M.M., M.D.L., C.V.); Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden (G.P.); Department
| |
Collapse
|
15
|
Zhou H, Fu B, Xu B, Mi X, Li G, Ma C, Xie J, Li J, Wang Z. Rosmarinic Acid Alleviates the Endothelial Dysfunction Induced by Hydrogen Peroxide in Rat Aortic Rings via Activation of AMPK. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2017; 2017:7091904. [PMID: 28883905 PMCID: PMC5572610 DOI: 10.1155/2017/7091904] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/07/2017] [Revised: 06/09/2017] [Accepted: 06/20/2017] [Indexed: 12/21/2022]
Abstract
Endothelial dysfunction is the key player in the development and progression of vascular events. Oxidative stress is involved in endothelial injury. Rosmarinic acid (RA) is a natural polyphenol with antioxidative, antiapoptotic, and anti-inflammatory properties. The present study investigates the protective effect of RA on endothelial dysfunction induced by hydrogen peroxide (H2O2). Compared with endothelium-denuded aortic rings, the endothelium significantly alleviated the decrease of vasoconstrictive reactivity to PE and KCl induced by H2O2. H2O2 pretreatment significantly injured the vasodilative reactivity to ACh in endothelium-intact aortic rings in a concentration-dependent manner. RA individual pretreatment had no obvious effect on the vasoconstrictive reaction to PE and KCl, while its cotreatment obviously mitigated the endothelium-dependent relaxation impairments and the oxidative stress induced by H2O2. The RA cotreatment reversed the downregulation of AMPK and eNOS phosphorylation induced by H2O2 in HAEC cells. The pretreatment with the inhibitors of AMPK (compound C) and eNOS (L-NAME) wiped off RA's beneficial effects. All these results demonstrated that RA attenuated the endothelial dysfunction induced by oxidative stress by activating the AMPK/eNOS pathway.
Collapse
Affiliation(s)
- Hui Zhou
- Center for Mitochondria and Healthy Aging, College of Life Sciences, Yantai University, Yantai 264005, China
| | - Baocai Fu
- Intensive Care Unit, Yantaishan Hospital, Yantai 264001, China
| | - Bo Xu
- Center for Mitochondria and Healthy Aging, College of Life Sciences, Yantai University, Yantai 264005, China
| | - Xiangquan Mi
- School of Medicine, Shihezi University, Shihezi 832002, China
| | - Gang Li
- Center for Mitochondria and Healthy Aging, College of Life Sciences, Yantai University, Yantai 264005, China
| | - Chengjun Ma
- Center for Mitochondria and Healthy Aging, College of Life Sciences, Yantai University, Yantai 264005, China
| | - Jianxin Xie
- School of Medicine, Shihezi University, Shihezi 832002, China
| | - Ji Li
- Center for Mitochondria and Healthy Aging, College of Life Sciences, Yantai University, Yantai 264005, China
| | - Zhenhua Wang
- Center for Mitochondria and Healthy Aging, College of Life Sciences, Yantai University, Yantai 264005, China
| |
Collapse
|
16
|
Pereira PC, Pernomian L, Côco H, Gomes MS, Franco JJ, Marchi KC, Hipólito UV, Uyemura SA, Tirapelli CR, de Oliveira AM. Auto-inhibitory regulation of angiotensin II functionality in hamster aorta during the early phases of dyslipidemia. Eur J Pharmacol 2016; 781:1-9. [DOI: 10.1016/j.ejphar.2016.04.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2015] [Revised: 03/31/2016] [Accepted: 04/07/2016] [Indexed: 10/22/2022]
|
17
|
Li J, Zhang Y, Luan H, Chen X, Han Y, Wang C. l-carnitine protects human hepatocytes from oxidative stress-induced toxicity through Akt-mediated activation of Nrf2 signaling pathway. Can J Physiol Pharmacol 2016; 94:517-25. [DOI: 10.1139/cjpp-2015-0305] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
In our previous study, l-carnitine was shown to have cytoprotective effect against hydrogen peroxide (H2O2)-induced injury in human normal HL7702 hepatocytes. The aim of this study was to investigate whether the protective effect of l-carnitine was associated with the nuclear factor erythroid 2 (NFE2)-related factor 2 (Nrf2) pathway. Our results showed that pretreatment with l-carnitine augmented Nrf2 nuclear translocation, DNA binding activity and heme oxygenase-1 (HO-1) expression in H2O2-treated HL7702 cells, although l-carnitine treatment alone had no effect on them. Analysis using Nrf2 siRNA demonstrated that Nrf2 activation was involved in l-carnitine-induced HO-1 expression. In addition, l-carnitine-mediated protection against H2O2 toxicity was abrogated by Nrf2 siRNA, indicating the important role of Nrf2 in l-carnitine-induced cytoprotection. Further experiments revealed that l-carnitine pretreatment enhanced the phosphorylation of Akt in H2O2-treated cells. Blocking Akt pathway with inhibitor partly abrogated the protective effect of l-carnitine. Moreover, our finding demonstrated that the induction of Nrf2 translocation and HO-1 expression by l-carnitine directly correlated with the Akt pathway because Akt inhibitor showed inhibitory effects on the Nrf2 translocation and HO-1 expression. Altogether, these results demonstrate that l-carnitine protects HL7702 cells against H2O2-induced cell damage through Akt-mediated activation of Nrf2 signaling pathway.
Collapse
Affiliation(s)
- Jinlian Li
- Medical College, Qingdao University, 308 Ningxia Road, Qingdao 266071, China
- Binzhou Medical University, 346 Guanhai Road, Yantai 264003, China
| | - Yanli Zhang
- Binzhou Medical University, 346 Guanhai Road, Yantai 264003, China
| | - Haiyun Luan
- Binzhou Medical University, 346 Guanhai Road, Yantai 264003, China
| | - Xuehong Chen
- Department of Pharmacology, Medical College, Qingdao University, 308 Ningxia Road, Qingdao 266071, China
| | - Yantao Han
- Department of Pharmacology, Medical College, Qingdao University, 308 Ningxia Road, Qingdao 266071, China
| | - Chunbo Wang
- Department of Pharmacology, Medical College, Qingdao University, 308 Ningxia Road, Qingdao 266071, China
| |
Collapse
|
18
|
Onyango IG, Dennis J, Khan SM. Mitochondrial Dysfunction in Alzheimer's Disease and the Rationale for Bioenergetics Based Therapies. Aging Dis 2016; 7:201-14. [PMID: 27114851 PMCID: PMC4809610 DOI: 10.14336/ad.2015.1007] [Citation(s) in RCA: 184] [Impact Index Per Article: 20.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2015] [Accepted: 10/07/2015] [Indexed: 12/19/2022] Open
Abstract
Alzheimer’s disease (AD) is a debilitating neurodegenerative disorder characterized by the progressive loss of cholinergic neurons, leading to the onset of severe behavioral, motor and cognitive impairments. It is a pressing public health problem with no effective treatment. Existing therapies only provide symptomatic relief without being able to prevent, stop or reverse the pathologic process. While the molecular basis underlying this multifactorial neurodegenerative disorder remains a significant challenge, mitochondrial dysfunction appears to be a critical factor in the pathogenesis of this disease. It is therefore important to target mitochondrial dysfunction in the prodromal phase of AD to slow or prevent the neurodegenerative process and restore neuronal function. In this review, we discuss mechanisms of action and translational potential of current mitochondrial and bioenergetic therapeutics for AD including: mitochondrial enhancers to potentiate energy production; antioxidants to scavenge reactive oxygen species and reduce oxidative damage; glucose metabolism and substrate supply; and candidates that target apoptotic and mitophagy pathways to remove damaged mitochondria. While mitochondrial therapeutic strategies have shown promise at the preclinical stage, there has been little progress in clinical trials thus far.
Collapse
Affiliation(s)
- Isaac G Onyango
- Gencia Biotechnology, 706 B Forest St, Charlottesville, VA 22903, USA
| | - Jameel Dennis
- Gencia Biotechnology, 706 B Forest St, Charlottesville, VA 22903, USA
| | - Shaharyah M Khan
- Gencia Biotechnology, 706 B Forest St, Charlottesville, VA 22903, USA
| |
Collapse
|
19
|
Sharma A, Yuen D, Huet O, Pickering R, Stefanovic N, Bernatchez P, de Haan JB. Lack of glutathione peroxidase-1 facilitates a pro-inflammatory and activated vascular endothelium. Vascul Pharmacol 2015; 79:32-42. [PMID: 26569096 DOI: 10.1016/j.vph.2015.11.001] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2015] [Revised: 10/26/2015] [Accepted: 11/08/2015] [Indexed: 11/30/2022]
Abstract
A critical early event in the pathogenesis of atherosclerosis is vascular inflammation leading to endothelial dysfunction (ED). Reactive oxygen species and inflammation are inextricably linked and declining antioxidant defense is implicated in ED. We have previously shown that Glutathione peroxidase-1 (GPx1) is a crucial antioxidant enzyme in the protection against diabetes-associated atherosclerosis. In this study we aimed to investigate mechanisms by which lack of GPx1 affects pro-inflammatory mediators in primary aortic endothelial cells (PAECs) isolated from GPx1 knockout (GPx1 KO) mice. Herein, we demonstrate that lack of GPx1 prolonged TNF-α induced phosphorylation of P38, ERK and JNK, all of which was reversed upon treatment with the GPx1 mimetic, ebselen. In addition, Akt phosphorylation was reduced in GPx1 KO PAECs, which correlated with decreased nitric oxide (NO) bioavailability as compared to WT PAECs. Furthermore, IκB degradation was prolonged in GPx1 KO PAECS suggesting an augmentation of NF-κB activity. In addition, the expression of vascular cell adhesion molecule (VCAM-1) was significantly increased in GPx1 KO PAECs and aortas. Static and dynamic flow adhesion assays showed significantly increased adhesion of fluorescently labeled leukocytes to GPx1 KO PAECS and aortas respectively, which were significantly reduced by ebselen treatment. Our results suggest that GPx1 plays a critical role in regulating pro-inflammatory pathways, including MAPK and NF-κB, and down-stream mediators such as VCAM-1, in vascular endothelial cells. Lack of GPx1, via effects on p-AKT also affects signaling to eNOS-derived NO. We speculate based on these results that declining antioxidant defenses as seen in cardiovascular diseases, by failing to regulate these pro-inflammatory pathways, facilitates an inflammatory and activated endothelium leading to ED and atherogenesis.
Collapse
Affiliation(s)
- Arpeeta Sharma
- Baker IDI Heart and Diabetes Institute, Diabetic Complications Division, Melbourne, Victoria, Australia
| | - Derek Yuen
- Baker IDI Heart and Diabetes Institute, Diabetic Complications Division, Melbourne, Victoria, Australia
| | - Olivier Huet
- Baker IDI Heart and Diabetes Institute, Diabetic Complications Division, Melbourne, Victoria, Australia; Department of Anaesthesia and Intensive Care, CHRU La Cavale Blanche, Université de Bretagne Ouest, Brest, France
| | - Raelene Pickering
- Baker IDI Heart and Diabetes Institute, Diabetic Complications Division, Melbourne, Victoria, Australia
| | - Nada Stefanovic
- Baker IDI Heart and Diabetes Institute, Diabetic Complications Division, Melbourne, Victoria, Australia
| | - Pascal Bernatchez
- Department of Anesthesiology, Pharmacology and Therapeutics, University of British Columbia, Canada
| | - Judy B de Haan
- Baker IDI Heart and Diabetes Institute, Diabetic Complications Division, Melbourne, Victoria, Australia.
| |
Collapse
|
20
|
Wang L, Chen Q, Li G, Ke D. Ghrelin ameliorates impaired angiogenesis of ischemic myocardium through GHSR1a-mediated AMPK/eNOS signal pathway in diabetic rats. Peptides 2015; 73:77-87. [PMID: 26364514 DOI: 10.1016/j.peptides.2015.09.004] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/28/2015] [Revised: 08/21/2015] [Accepted: 09/08/2015] [Indexed: 11/24/2022]
Abstract
OBJECTIVES Ghrelin, an endogenous ligand of the growth hormone secretagogue receptor (GHSR), has been found to stimulate angiogenesis in vivo and in vitro. However, the effect and the corresponding mechanisms of ghrelin on impaired myocardial angiogenesis in diabetic and myocardial infarction (MI) rat model are still unknown. METHODS In the present study, adult SD rats were randomly divided into 4 groups: control, DM, DM+ghrelin, DM+ghrelin+[D-Lys3]-GHRP-6 groups. DM was induced by streptozotocin (STZ) 60 mg/kg body weight. 12 weeks post STZ injection all groups were subjected to MI, which was induced by ligation left anterior descending artery (LAD). Ghrelin and [D-Lys3]-GHRP-6 were administered via intraperitoneal injection at the doses 200 μg/kg and 50mg/kg for 4 weeks, respectively. Left ventricular function, microvascular density (MVD), myocardial infarct size, the expression of hypoxia-inducible factor (HIF1α), vascular endothelial growth factor (VEGF), fetal liver kinase-1 (Flk-1) and fms-like tyrosine kinase-1 (Flt-1), AMPK and endothelial nitric oxide synthase (eNOS) phosphorylation were examined. RESULTS Compared with the DM group, left ventricular ejection fraction (LVEF), fractional shortening (FS), and MVD were increased, whereas myocardial infarct size decreased remarkably in DM+ghrelin group. For the mechanism study, we found that ghrelin promoted the HIF1α, VEGF, Flk-1 and Flt-1 expression, AMPK and eNOS phosphorylation in diabetic rats. However, the above biochemical events in ghrelin treated diabetic rats were completely inhibited by GHSR-1a blocker [D-Lys3]-GHRP-6. CONCLUSIONS These results suggest that administration of ghrelin ameliorates impaired angiogenesis in diabetic MI rats. And these beneficial effects derive from regulating GHSR1a-mediated AMPK/eNOS signal pathway by upregulating of HIF1α, VEGF and its receptors Flk-1, Flt-1 expressions.
Collapse
Affiliation(s)
- Li Wang
- Department of Geriatrics, the Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, China
| | - Qingwei Chen
- Department of Geriatrics, the Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, China.
| | - Guiqiong Li
- Department of Geriatrics, the Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, China
| | - Dazhi Ke
- Department of Geriatrics, the Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, China
| |
Collapse
|
21
|
Meyer G, André L, Kleindienst A, Singh F, Tanguy S, Richard S, Obert P, Boucher F, Jover B, Cazorla O, Reboul C. Carbon monoxide increases inducible NOS expression that mediates CO-induced myocardial damage during ischemia-reperfusion. Am J Physiol Heart Circ Physiol 2015; 308:H759-67. [PMID: 25595132 DOI: 10.1152/ajpheart.00702.2014] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/30/2014] [Accepted: 01/13/2015] [Indexed: 11/22/2022]
Abstract
We investigated the role of inducible nitric oxide (NO) synthase (iNOS) on ischemic myocardial damage in rats exposed to daily low nontoxic levels of carbon monoxide (CO). CO is a ubiquitous environmental pollutant that impacts on mortality and morbidity from cardiovascular diseases. We have previously shown that CO exposure aggravates myocardial ischemia-reperfusion (I/R) injury partly because of increased oxidative stress. Nevertheless, cellular mechanisms underlying cardiac CO toxicity remain hypothetical. Wistar rats were exposed to simulated urban CO pollution for 4 wk. First, the effects of CO exposure on NO production and NO synthase (NOS) expression were evaluated. Myocardial I/R was performed on isolated perfused hearts in the presence or absence of S-methyl-isothiourea (1 μM), a NOS inhibitor highly specific for iNOS. Finally, Ca(2+) handling was evaluated in isolated myocytes before and after an anoxia-reoxygenation performed with or without S-methyl-isothiourea or N-acetylcystein (20 μM), a nonspecific antioxidant. Our main results revealed that 1) CO exposure altered the pattern of NOS expression, which is characterized by increased neuronal NOS and iNOS expression; 2) cardiac NO production increased in CO rats because of its overexpression of iNOS; and 3) the use of a specific inhibitor of iNOS reduced myocardial hypersensitivity to I/R (infarct size, 29 vs. 51% of risk zone) in CO rat hearts. These last results are explained by the deleterious effects of NO and reactive oxygen species overproduction by iNOS on diastolic Ca(2+) overload and myofilaments Ca(2+) sensitivity. In conclusion, this study highlights the involvement of iNOS overexpression in the pathogenesis of simulated urban CO air pollution exposure.
Collapse
Affiliation(s)
| | - Lucas André
- Institut national de la santé et de la recherche médicale, Université Montpellier1, Université Montpellier2, Montpellier, France
| | | | - François Singh
- Fédération de Médecine Translationelle, Faculty of Medicine, Université de Strasbourg, Strasbourg France
| | - Stéphane Tanguy
- Université d'Avignon, Avignon, France; Laboratoire Techniques for biomedical engineering and complexity management-informatics, mathematics, and applications-Grenoble, Bâtiment Jean Roget-Domaine de la Merci, Université Joseph Fourier, La Tronche Cedex, France
| | - Sylvain Richard
- Institut national de la santé et de la recherche médicale, Université Montpellier1, Université Montpellier2, Montpellier, France
| | | | - François Boucher
- Laboratoire Techniques for biomedical engineering and complexity management-informatics, mathematics, and applications-Grenoble, Bâtiment Jean Roget-Domaine de la Merci, Université Joseph Fourier, La Tronche Cedex, France
| | - Bernard Jover
- Centre de Pharmacologie et Innovation dans le Diabète, Faculty of Pharmacy, Université Montpellier1, Montpellier, France; and
| | - Olivier Cazorla
- Institut national de la santé et de la recherche médicale, Université Montpellier1, Université Montpellier2, Montpellier, France
| | | |
Collapse
|
22
|
Xu L, Wang S, Li B, Sun A, Zou Y, Ge J. A protective role of ciglitazone in ox-LDL-induced rat microvascular endothelial cells via modulating PPARγ-dependent AMPK/eNOS pathway. J Cell Mol Med 2014; 19:92-102. [PMID: 25388834 PMCID: PMC4288353 DOI: 10.1111/jcmm.12463] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2014] [Accepted: 09/19/2014] [Indexed: 01/10/2023] Open
Abstract
Thiazolidinediones, the antidiabetic agents such as ciglitazone, has been proved to be effective in limiting atherosclerotic events. However, the underlying mechanism remains elucidative. Ox-LDL receptor-1 (LOX-1) plays a central role in ox-LDL-mediated atherosclerosis via endothelial nitric oxide synthase (eNOS) uncoupling and nitric oxide reduction. Therefore, we tested the hypothesis that ciglitazone, the PPARγ agonist, protected endothelial cells against ox-LDL through regulating eNOS activity and LOX-1 signalling. In the present study, rat microvascular endothelial cells (RMVECs) were stimulated by ox-LDL. The impact of ciglitazone on cell apoptosis and angiogenesis, eNOS expression and phosphorylation, nitric oxide synthesis and related AMPK, Akt and VEGF signalling pathway were observed. Our data showed that both eNOS and Akt phosphorylation, VEGF expression and nitric oxide production were significantly decreased, RMVECs ageing and apoptosis increased after ox-LDL induction for 24 hrs, all of which were effectively reversed by ciglitazone pre-treatment. Meanwhile, phosphorylation of AMP-activated protein kinase (AMPK) was suppressed by ox-LDL, which was also prevented by ciglitazone. Of interest, AMPK inhibition abolished ciglitazone-mediated eNOS function, nitric oxide synthesis and angiogenesis, and increased RMVECs ageing and apoptosis. Further experiments showed that inhibition of PPARγ significantly suppressed AMPK phosphorylation, eNOS expression and nitric oxide production. Ciglitazone-mediated angiogenesis and reduced cell ageing and apoptosis were reversed. Furthermore, LOX-1 protein expression in RMVECs was suppressed by ciglitazone, but re-enhanced by blocking PPARγ or AMPK. Ox-LDL-induced suppression of eNOS and nitric oxide synthesis were largely prevented by silencing LOX-1. Collectively, these data demonstrate that ciglitazone-mediated PPARγ activation suppresses LOX-1 and moderates AMPK/eNOS pathway, which contributes to endothelial cell survival and function preservation.
Collapse
Affiliation(s)
- Lei Xu
- Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, Shanghai, China; Institutes of Biomedical Science, Fudan University, Shanghai, China
| | | | | | | | | | | |
Collapse
|
23
|
AMPK-α1 functions downstream of oxidative stress to mediate neuronal atrophy in Huntington's disease. Biochim Biophys Acta Mol Basis Dis 2014; 1842:1668-80. [DOI: 10.1016/j.bbadis.2014.06.012] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2013] [Revised: 05/27/2014] [Accepted: 06/09/2014] [Indexed: 01/29/2023]
|
24
|
Awad H, Nolette N, Hinton M, Dakshinamurti S. AMPK and FoxO1 regulate catalase expression in hypoxic pulmonary arterial smooth muscle. Pediatr Pulmonol 2014; 49:885-97. [PMID: 24167160 DOI: 10.1002/ppul.22919] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/06/2013] [Accepted: 08/13/2013] [Indexed: 11/10/2022]
Abstract
BACKGROUND Hypoxia and reactive oxygen species (ROS) including H(2)O(2) play major roles in triggering and progression of pulmonary vascular remodeling in persistent pulmonary hypertension. Catalase (CAT), the major endogenous enzyme scavenging H(2)O(2), is regulated in a tissue- and context-specific manner. OBJECTIVE To investigate mechanisms by which hypoxia and H(2)O(2) regulate catalase expression, and the role of AMPK-FoxO pathway, in neonatal porcine pulmonary artery smooth muscle (PASMC). DESIGN/METHODS PASMC were grown in hypoxia (10% O(2)) or normoxia (21% O(2)) for 72 hr. We measured catalase activity and lipid peroxidation; CAT, FoxO1, and FoxO3a expression by qPCR; protein contents of CAT, FoxOs, p-AMPK, p-AKT, p-JNK, p-ERK1/2 in whole lysates, and FoxOs in nuclear extracts, by immunoblot; and FoxO-1 nuclear localization by immunocytochemistry, quantified by laser scanning cytometry. RESULTS Hypoxia upregulated CAT transcription, content and activity, by increasing CAT transcription factors FoxO1 and FoxO3a mRNA, and promoting nuclear translocation of FoxO1. However, lipid peroxidation increased in hypoxic PASMC. Among candidate FoxO regulatory kinases, hypoxia activated AMPK, and decreased p-Akt and ERK1/2. AMPK activation increased FoxO1 (total and nuclear) and CAT, while AMPK inhibition inhibited FoxO1 and CAT, but not FoxO3a. Exogenous H(2)O(2) decreased p-AMPK and increased p-AKT in hypoxic PASMC. This decreased active FoxO1, and reduced mRNA and protein content of CAT. Hypoxic induction of CAT, AKT inhibition (LY294002), or addition of PEG-catalase partly ameliorated the H(2)O(2) -mediated loss of nuclear FoxO1. CONCLUSIONS Hypoxia induces catalase expression, though this adaptation is insufficient to protect PASMC from hypoxia-induced lipid peroxidation. This occurs via hypoxic activation of AMPK, which promotes nuclear FoxO1 and thus catalase expression. Exogenous ROS may downregulate cellular antioxidant defenses; H(2)O(2) activates survival factor Akt, decreasing nuclear FoxO1 and thus catalase.
Collapse
Affiliation(s)
- Hanan Awad
- Biology of Breathing Group, Manitoba Institute of Child Health, Winnipeg, Canada
| | | | | | | |
Collapse
|
25
|
d'Uscio LV, He T, Santhanam AVR, Tai LJ, Evans RM, Katusic ZS. Mechanisms of vascular dysfunction in mice with endothelium-specific deletion of the PPAR-δ gene. Am J Physiol Heart Circ Physiol 2014; 306:H1001-10. [PMID: 24486511 PMCID: PMC3962632 DOI: 10.1152/ajpheart.00761.2013] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/01/2013] [Accepted: 01/27/2014] [Indexed: 02/06/2023]
Abstract
Peroxisome proliferator-activated receptor (PPAR)-δ is a nuclear hormone receptor that is mainly involved in lipid metabolism. Recent studies have suggested that PPAR-δ agonists exert vascular protective effects. The present study was designed to characterize vascular function in mice with genetic inactivation of PPAR-δ in the endothelium. Mice with vascular endothelial cell-specific deletion of the PPAR-δ gene (ePPARδ(-/-) mice) were generated using loxP/Cre technology. ePPARδ(-/-) mice were normotensive and did not display any sign of metabolic syndrome. Endothelium-dependent relaxations to ACh and endothelium-independent relaxations to the nitric oxide (NO) donor diethylammonium (Z)-1-(N,N-diethylamino)diazen-1-ium-1,2-diolate were both significantly impaired in the aorta and carotid arteries of ePPARδ(-/-) mice (P < 0.05). In ePPARδ(-/-) mouse aortas, phosphorylation of endothelial NO synthase at Ser(1177) was significantly decreased (P < 0.05). However, basal levels of cGMP were unexpectedly increased (P < 0.05). Enzymatic activity of GTP-cyclohydrolase I and tetrahydrobiopterin levels were also enhanced in ePPARδ(-/-) mice (P < 0.05). Most notably, endothelium-specific deletion of the PPAR-δ gene significantly decreased protein expressions of catalase and glutathione peroxidase 1 and resulted in increased levels of H2O2 in the aorta (P < 0.05). In contrast, superoxide anion production was unaltered. Moreover, treatment with catalase prevented the endothelial dysfunction and elevation of cGMP detected in aortas of ePPARδ(-/-) mice. The findings suggest that increased levels of cGMP caused by H2O2 impair vasodilator reactivity to endogenous and exogenous NO. We speculate that chronic elevation of H2O2 predisposes PPAR-δ-deficient arteries to oxidative stress and vascular dysfunction.
Collapse
Affiliation(s)
- Livius V d'Uscio
- Department of Anesthesiology and Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic College of Medicine, Rochester, Minnesota; and
| | | | | | | | | | | |
Collapse
|
26
|
Guterbaum TJ, Braunstein TH, Fossum A, Holstein-Rathlou NH, Torp-Pedersen CT, Domínguez H. Endothelial nitric oxide synthase phosphorylation at Threonine 495 and mitochondrial reactive oxygen species formation in response to a high H₂O₂ concentration. J Vasc Res 2013; 50:410-20. [PMID: 24008236 DOI: 10.1159/000354225] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2011] [Accepted: 07/05/2013] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND Hydrogen peroxide (H₂O₂) is produced in vessels during ischemia/reperfusion and during inflammation, both leading to vascular dysfunction. We investigated cellular pathways involved in endothelial nitric oxide synthase (eNOS) phosphorylation at Threonine 495 (Thr(495)) in human umbilical vein endothelial cells (HUVECs) exposed to H₂O₂. METHODS HUVECs were exposed to 400 μM H₂O₂ for 30 min. Phosphorylation at Thr(495) was assessed by Western blotting and reactive oxygen species (ROS) monitored by flow cytometry. Protein kinase C (PKC) pathways were investigated by pretreatment with PKC-β inhibitor ruboxistaurin or pan-PKC inhibitor GF109203X. In addition, we investigated ROCK and ERK pathways by MEKK1/2 inhibitor U0126 and ROCK inhibitor Y27632. RESULTS H₂O₂ increased eNOS phosphorylation at Thr(495) (to 176% vs. control (100%), p < 0.001) along with increased mitochondrial ROS formation (from 19.7 to 45.3%, p < 0.01). This rise in phosphorylation could be prevented by U0126 and Y27632 in a dose-dependent manner, but did not result in lowered mitochondrial ROS formation. Conversely, addition of the antioxidant N-acetyl-L-cysteine only prevented mitochondrial ROS formation but did not prevent phosphorylation of eNOS Thr(495). CONCLUSION H₂O₂-mediated phosphorylation of eNOS Thr(495) is mediated by ROCK and ERK activity, but not by PKC, and is uncoupled from mitochondrial ROS signaling. Furthermore, ERK inhibition increased mitochondrial ROS formation.
Collapse
Affiliation(s)
- T J Guterbaum
- The Danish National Research Foundation Center for Cardiac Arrhythmia, University of Copenhagen, Copenhagen, Denmark
| | | | | | | | | | | |
Collapse
|
27
|
Brunt KR, Wu J, Chen Z, Poeckel D, Dercho RA, Melo LG, Funk CD, Ward CA, Li RK. Ex vivo Akt/HO-1 gene therapy to human endothelial progenitor cells enhances myocardial infarction recovery. Cell Transplant 2013; 21:1443-61. [PMID: 22776314 DOI: 10.3727/096368912x653002] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
The aim of this study was to evaluate the overexpression of genes central to cell survival and angiogenesis to enhance the function of human late outgrowth endothelial progenitor cells (EPCs) and their utility for infarct recovery. Ischemic myocardial injury creates a hostile microenvironment, which is characterized by hypoxia, oxidative stress, and inflammation. The infarct microenvironment prevents adhesion, survival, and integration of cell transplants that promote neovascularization. EPCs are dysfunctional as a result of risk factors in cardiovascular patients. Protein kinase B (Akt) and heme-oxygenase-1 (HO-1) are intracellular proteins that play an important role in angiogenesis and cell survival. Late outgrowth EPCs transduced ex vivo with Akt and HO-1 demonstrate improved adhesion to extracellular matrix, improved migration toward human cardiomyocytes, and an improved paracrine profile under stress. Enhanced late outgrowth EPCs reduce the tumor necrosis factor-α (TNF-α) burden both in vitro and in vivo, attenuating nuclear factor-κB (NF-κB) activity and promoting cell survival. Akt and HO-1 enhance late outgrowth EPC neovascularization, resulting in improved cardiac performance and reduced negative remodeling after myocardial infarction in nude mice. Alteration of the infarct microenvironment through gene modification of human late outgrowth EPCs enhances the function and integration of transplanted cells for restoration of cardiac function.
Collapse
Affiliation(s)
- Keith R Brunt
- Department of Physiology, Queen's University, Kingston, ON, Canada
| | | | | | | | | | | | | | | | | |
Collapse
|
28
|
AMPK-regulated and Akt-dependent enhancement of glucose uptake is essential in ischemic preconditioning-alleviated reperfusion injury. PLoS One 2013; 8:e69910. [PMID: 23922853 PMCID: PMC3724784 DOI: 10.1371/journal.pone.0069910] [Citation(s) in RCA: 64] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2013] [Accepted: 06/13/2013] [Indexed: 01/04/2023] Open
Abstract
AIMS Ischemic preconditioning (IPC) is a potent form of endogenous protection. However, IPC-induced cardioprotective effect is significantly blunted in insulin resistance-related diseases and the underlying mechanism is unclear. This study aimed to determine the role of glucose metabolism in IPC-reduced reperfusion injury. METHODS Normal or streptozotocin (STZ)-treated diabetic rats subjected to 2 cycles of 5 min ischemia/5 min reperfusion prior to myocardial ischemia (30 min)/reperfusion (3 h). Myocardial glucose uptake was determined by (18)F-fluorodeoxyglucose-positron emission tomography (PET) scan and gamma-counter biodistribution assay. RESULTS IPC exerted significant cardioprotection and markedly improved myocardial glucose uptake 1 h after reperfusion (P<0.01) as evidenced by PET images and gamma-counter biodistribution assay in ischemia/reperfused rats. Meanwhile, myocardial translocation of glucose transporter 4 (GLUT4) to plasma membrane together with myocardial Akt and AMPK phosphorylation were significantly enhanced in preconditioned hearts. Intramyocardial injection of GLUT4 siRNA markedly decreased GLUT4 expression and blocked the cardioprotection of IPC as evidence by increased myocardial infarct size. Moreover, the PI3K inhibitor wortmannin significantly inhibited activation of Akt and AMPK, reduced GLUT4 translocation, glucose uptake and ultimately, depressed IPC-induced cardioprotection. Furthermore, IPC-afforded antiapoptotic effect was markedly blunted in STZ-treated diabetic rats. Exogenous insulin supplementation significantly improved glucose uptake via co-activation of myocardial AMPK and Akt and alleviated ischemia/reperfusion injury as evidenced by reduced myocardial apoptosis and infarction size in STZ-treated rats (P<0.05). CONCLUSIONS The present study firstly examined the role of myocardial glucose metabolism during reperfusion in IPC using direct genetic modulation in vivo. Augmented glucose uptake via co-activation of myocardial AMPK and Akt in reperfused myocardium is essential to IPC-alleviated reperfusion injury. This intrinsic metabolic modulation and cardioprotective capacity are present in STZ-treated hearts and can be triggered by insulin.
Collapse
|
29
|
Takai J, Santu A, Zheng H, Koh SD, Ohta M, Filimban LM, Lemaître V, Teraoka R, Jo H, Miura H. Laminar shear stress upregulates endothelial Ca²⁺-activated K⁺ channels KCa2.3 and KCa3.1 via a Ca²⁺/calmodulin-dependent protein kinase kinase/Akt/p300 cascade. Am J Physiol Heart Circ Physiol 2013; 305:H484-93. [PMID: 23792675 DOI: 10.1152/ajpheart.00642.2012] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
In endothelial cells (ECs), Ca²⁺-activated K⁺ channels KCa2.3 and KCa3.1 play a crucial role in the regulation of arterial tone via producing NO and endothelium-derived hyperpolarizing factors. Since a rise in intracellular Ca²⁺ levels and activation of p300 histone acetyltransferase are early EC responses to laminar shear stress (LS) for the transcriptional activation of genes, we examined the role of Ca²⁺/calmodulin-dependent kinase kinase (CaMKK), the most upstream element of a Ca²⁺/calmodulin-kinase cascade, and p300 in LS-dependent regulation of KCa2.3 and KCa3.1 in ECs. Exposure to LS (15 dyn/cm²) for 24 h markedly increased KCa2.3 and KCa3.1 mRNA expression in cultured human coronary artery ECs (3.2 ± 0.4 and 45 ± 10 fold increase, respectively; P < 0.05 vs. static condition; n = 8-30), whereas oscillatory shear (OS; ± 5 dyn/cm² × 1 Hz) moderately increased KCa3.1 but did not affect KCa2.3. Expression of KCa2.1 and KCa2.2 was suppressed under both LS and OS conditions, whereas KCa1.1 was slightly elevated in LS and unchanged in OS. Inhibition of CaMKK attenuated LS-induced increases in the expression and channel activity of KCa2.3 and KCa3.1, and in phosphorylation of Akt (Ser473) and p300 (Ser1834). Inhibition of Akt abolished the upregulation of these channels by diminishing p300 phosphorylation. Consistently, disruption of the interaction of p300 with transcription factors eliminated the induction of these channels. Thus a CaMKK/Akt/p300 cascade plays an important role in LS-dependent induction of KCa2.3 and KCa3.1 expression, thereby regulating EC function and adaptation to hemodynamic changes.
Collapse
Affiliation(s)
- Jun Takai
- Department of Physiology and Cell Biology, University of Nevada School of Medicine, Reno, Nevada
| | | | | | | | | | | | | | | | | | | |
Collapse
|
30
|
González R, Ferrín G, Aguilar-Melero P, Ranchal I, Linares CI, Bello RI, De la Mata M, Gogvadze V, Bárcena JA, Alamo JM, Orrenius S, Padillo FJ, Zhivotovsky B, Muntané J. Targeting hepatoma using nitric oxide donor strategies. Antioxid Redox Signal 2013; 18:491-506. [PMID: 22861189 DOI: 10.1089/ars.2011.4476] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
AIMS The study evaluated the role of increased intracellular nitric oxide (NO) concentration using NO donors or stably NO synthase-3 (NOS-3) overexpression during CD95-dependent cell death in hepatoma cells. The expression of cell death receptors and caspase activation, RhoA kinase activity, NOS-3 expression/activity, oxidative/nitrosative stress, and p53 expression were analyzed. The antitumoral activity of NO was also evaluated in the subcutaneous implantation of NOS-3-overexpressing hepatoma cells, as well NO donor injection into wild-type hepatoma-derived tumors implanted in xenograft mouse models. RESULTS NO donor increased CD95 expression and activation of caspase-8 and 3 in HepG2, Huh7, and Hep3B cells. NOS-3 overexpression increased oxidative/nitrosative stress, p53 and CD95 expression, cellular Fas-associated death domain (FADD)-like IL-1beta converting enzyme (FLICE) inhibitory protein long (cFLIP(L)) and its short isoform (cFLIP(S)) shift, and cell death in HepG2 (4TO-NOS) cells. The inhibition of RhoA kinase and p53 knockdown using RNA interference reduced cell death in 4TO-NOS cells. The supplementation with hydrogen peroxide (H(2)O(2)) increased NOS-3 activity and cell death in 4TO-NOS cells. NOS-3 overexpression or NO donor injection into hepatoma-derived tumors reduced the size and increased p53 and cell death receptor expression in nude mice. INNOVATION AND CONCLUSIONS The increase of intracellular NO concentration promoted oxidative and nitrosative stress, Rho kinase activity, p53 and CD95 expression, and cell death in cultured hepatoma cells. NOS-3-overexpressed HepG2 cells or intratumoral NO donor administration reduced tumor cell growth and increased the expression of p53 and cell death receptors in tumors developed in a xenograft mouse model.
Collapse
Affiliation(s)
- Raúl González
- Department of Biochemistry and Molecular Biology, University of Córdoba, Córdoba, Spain
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Yan J, Tie G, Wang S, Messina KE, DiDato S, Guo S, Messina LM. Type 2 diabetes restricts multipotency of mesenchymal stem cells and impairs their capacity to augment postischemic neovascularization in db/db mice. J Am Heart Assoc 2012; 1:e002238. [PMID: 23316315 PMCID: PMC3540677 DOI: 10.1161/jaha.112.002238] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/23/2012] [Accepted: 09/12/2012] [Indexed: 02/07/2023]
Abstract
Background This study tested the hypothesis that type 2 diabetes restricts multipotency of db/db mesenchymal stem cells (MSCs), promotes their terminal differentiation into adipocytes rather than endothelial cells, thereby promotes adipocytic infiltration into ischemic muscles, and reduces their capacity to participate in postischemic neovascularization. Methods and Results To test this hypothesis, we transplanted MSCs from db/db or wild-type (WT) mice into WT recipients after induction of hind limb ischemia. WT recipients of db/db MSCs demonstrated adipocyte infiltration of ischemic muscle and impaired neovascularization; WT recipients of WT MSCs showed no intramuscular adipocyte infiltration and had significantly enhanced neovascularization (P<0.05; n=6). Confocal microscopy showed that the percentage of MSCs that differentiated into an adipocyte phenotype was greater and into an endothelial cell was less in WT recipients transplanted with db/db MSCs than those transplanted with WT MSCs (P<0.05; n=6). In vitro, db/db MSCs exhibited greater oxidant stress, greater adipocyte differentiation, and less endothelial differentiation than WT MSCs, and these differences were reversed by treatment with N-acetylcysteine or Nox4 siRNA (P<0.05; n=6). Insulin increased Nox4 expression, oxidant stress, and adipocyte differentiation in WT MSCs, and these insulin-induced effects were reversed by Nox4 siRNA (P<0.05; n=6). Reversal of db/db MSC oxidant stress by in vivo pretreatment with Nox4 siRNA before transplantation reversed their impaired capacity to augment postischemic neovascularization. Conclusions Type 2 diabetes–induced oxidant stress restricts the multipotency of MSCs and impairs their capacity to increase blood flow recovery after the induction of hind-limb ischemia. Reversal of MSC oxidant stress might permit greater leverage of the therapeutic potential of MSC transplantation in the setting of diabetes.
Collapse
Affiliation(s)
- Jinglian Yan
- Division of Vascular and Endovascular Surgery, University of Massachusetts Medical School, Worcester, MA 01655, USA
| | | | | | | | | | | | | |
Collapse
|
32
|
Silva BR, Pernomian L, Bendhack LM. Contribution of oxidative stress to endothelial dysfunction in hypertension. Front Physiol 2012; 3:441. [PMID: 23227009 PMCID: PMC3514688 DOI: 10.3389/fphys.2012.00441] [Citation(s) in RCA: 88] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2012] [Accepted: 11/05/2012] [Indexed: 12/12/2022] Open
Abstract
Endothelial dysfunction is the hallmark of hypertension, which is a multifactorial disorder. In the cardiovascular system reactive oxygen species play a pivotal role in controlling the endothelial function and vascular tone. Physiologically, the endothelium-derived relaxing factors (EDRFs) and endothelium-derived contractile factors (EDCFs) that have functions on the vascular smooth muscle cells. The relaxation induced by the EDRFs nitric oxide (NO), prostacyclin, and the endothelium-derived hyperpolarization factor (EDHF) could be impaired in hypertension. The impaired ability of endothelial cells to release NO along with enhanced EDCFs production has been described to contribute to the endothelium dysfunction, which appears to lead to several cardiovascular diseases. The present review discusses the role of oxidative stress, vascular endothelium, and vascular tone control by EDRFs, mainly NO, and EDCFs in different models of experimental hypertension.
Collapse
Affiliation(s)
- Bruno R Silva
- Department of Pharmacology, School of Medicine of Ribeirão Preto, University of São Paulo, Ribeirão Preto São Paulo, Brazil
| | | | | |
Collapse
|
33
|
Ou HC, Hsieh YL, Yang NC, Tsai KL, Chen KL, Tsai CS, Chen IJ, Wu BT, Lee SD. Ginkgo biloba extract attenuates oxLDL-induced endothelial dysfunction via an AMPK-dependent mechanism. J Appl Physiol (1985) 2012. [PMID: 23195633 DOI: 10.1152/japplphysiol.00367.2012] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Atherosclerosis is a complex inflammatory arterial disease, and oxidized low-density lipoprotein (oxLDL) is directly associated with chronic vascular inflammation. Previous studies have shown that Ginkgo biloba extract (GbE) acts as a therapeutic agent for neurological and cardiovascular disorders. However, the mechanisms mediating the actions of GbE are still largely unknown. In the present study, we tested the hypothesis that GbE protects against oxLDL-induced endothelial dysfunction via an AMP-activated protein kinase (AMPK)-dependent mechanism. Human umbilical vein endothelial cells were treated with GbE, followed by oxLDL, for indicated time periods. Results from Western blot showed that GbE inhibited the membrane translocation of the NADPH oxidase subunits p47(phox) and Rac-1 and attenuated the increase in protein expression of membrane subunits gp91 and p22(phox) caused by oxLDL-induced AMPK dephosphorylation and subsequent PKC activation. AMPK-α(1)-specific small interfering RNA-transfected cells that had been exposed to GbE followed by oxLDL revealed elevated levels of PKC and p47(phox). In addition, exposure to oxLDL resulted in reduced AMPK-mediated Akt/endothelial nitric oxide (NO) synthase signaling and the induction of phosphorylation of p38 mitogen-activated protein kinase, which, in turn, activated NF-κB-mediated inflammatory responses, such as the release of interleukin-8, the expression of the adhesion molecule, and the adherence of monocytic cells to human umbilical vein endothelial cells. Furthermore, oxLDL upregulated the expression of inducible NO synthase, thereby augmenting the formation of NO and protein nitrosylation. Pretreatment with GbE, however, exerted significant cytoprotective effects in a dose-dependent manner. Results from this study may provide insight into a possible molecular mechanism by which GbE protects against oxLDL-induced endothelial dysfunction.
Collapse
Affiliation(s)
- Hsiu-Chung Ou
- Department of Physical Therapy and Graduate Institute of Rehabilitation Science, China Medical University, Taichung, Taiwan
| | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Zhou X, Yuan D, Wang M, He P. H2O2-induced endothelial NO production contributes to vascular cell apoptosis and increased permeability in rat venules. Am J Physiol Heart Circ Physiol 2012; 304:H82-93. [PMID: 23086988 DOI: 10.1152/ajpheart.00300.2012] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Although elevated levels of H(2)O(2) have been implicated to play important roles in the pathogenesis of various cardiovascular diseases, the underlying mechanisms remain unclear. This study aims to examine the effect of H(2)O(2) on endothelial nitric oxide (NO) production in intact venules, and elucidate the role and mechanisms of NO in H(2)O(2)-induced increases in microvessel permeability. Experiments were conducted on individually perfused rat mesenteric venules. Microvessel permeability was determined by measuring hydraulic conductivity (Lp), and endothelial [Ca(2+)](i) was measured on fura-2-loaded vessels. Perfusion of H(2)O(2) (10 μM) caused a delayed and progressively increased endothelial [Ca(2+)](i) and Lp, a pattern different from inflammatory mediator-induced immediate and transient response. Under the same experimental conditions, measuring endothelial NO via DAF-2 and the spatial detection of cell apoptosis by fluorescent markers revealed that H(2)O(2) induced two phases of NO production followed by caspase activation, intracellular Ca(2+) accumulation, and vascular cell apoptosis. The initial NO production was correlated with increased endothelial NO synthase (eNOS) Ser(1177) phosphorylation in the absence of elevated endothelial [Ca(2+)](i), whereas the second phase of NO depended on increased [Ca(2+)](i) and was associated with Thr(495) dephosphorylation without increased Ser(1177) phosphorylation. Inhibition of NOS prevented H(2)O(2)-induced caspase activation, cell apoptosis, and increases in endothelial [Ca(2+)](i) and Lp. Our results indicate that H(2)O(2) at micromolar concentration is able to induce a large magnitude of NO in intact venules, causing caspase activation-mediated endothelial Ca(2+) accumulation, cell apoptosis, and increases in permeability. The mechanisms revealed from intact microvessels may contribute to the pathogenesis of oxidant-related cardiovascular diseases.
Collapse
Affiliation(s)
- Xueping Zhou
- Department of Physiology and Pharmacology, School of Medicine, West Virginia University, Morgantown, WV 26505, USA
| | | | | | | |
Collapse
|
35
|
Acute exercise induce endothelial nitric oxide synthase phosphorylation via Akt and AMP-activated protein kinase in aorta of rats: Role of reactive oxygen species. Int J Cardiol 2012; 167:2983-8. [PMID: 22989604 DOI: 10.1016/j.ijcard.2012.08.050] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/19/2012] [Accepted: 08/31/2012] [Indexed: 02/02/2023]
Abstract
BACKGROUND Acute exercise increases reactive oxygen species (ROS) levels, including hydrogen peroxide (H2O2). H2O2 promotes endothelial nitric oxide synthase (eNOS) activation and phosphorylation in endothelial cells. With this in mind, the present study was designed to evaluate ex vivo eNOS phosphorylation in rat aortas incubated with H2O2 and to test this hypothesis in vivo in the aortas of rats submitted to acute exercise. METHODS For ex vivo studies, six groups of aortic tissue were formed: control, H2O2, N-acetylcysteine (NAC), LY294002, compound C, and LY294002 plus compound C. While incubation with H2O2 increased Akt, AMPK and eNOS phosphorylation, pre-incubation with NAC strongly reduced the phosphorylation of these enzymes. For in vivo studies, male Wistar rats were divided into four groups: control, cont+NAC, exercise, and exer+NAC. After a 3h swimming session, animals were decapitated and aortas were excised for biochemical and immunoblotting analysis. RESULTS Acute exercise increased superoxide levels and dichlorofluorescein (DCF) concentrations, and this increase was related to phosphorylation of Akt, AMPK and eNOS. On the other hand, use of NAC reduced superoxide levels and DCF concentration. Reduced superoxide levels and DCF in the exer+NAC group were associated with decreased Akt, AMPK and eNOS phosphorylation. These results appear to be connected with vascular function because VASP phosphorylation increased in acute exercise and decreased in exer+NAC. CONCLUSION Our results indicate that ROS induced by acute exercise play the important role of activating eNOS, a process apparently mediated by Akt and AMPK.
Collapse
|
36
|
Maron BA, Zhang YY, White K, Chan SY, Handy DE, Mahoney CE, Loscalzo J, Leopold JA. Aldosterone inactivates the endothelin-B receptor via a cysteinyl thiol redox switch to decrease pulmonary endothelial nitric oxide levels and modulate pulmonary arterial hypertension. Circulation 2012; 126:963-74. [PMID: 22787113 DOI: 10.1161/circulationaha.112.094722] [Citation(s) in RCA: 150] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
BACKGROUND Pulmonary arterial hypertension (PAH) is characterized, in part, by decreased endothelial nitric oxide (NO(·)) production and elevated levels of endothelin-1. Endothelin-1 is known to stimulate endothelial nitric oxide synthase (eNOS) via the endothelin-B receptor (ET(B)), suggesting that this signaling pathway is perturbed in PAH. Endothelin-1 also stimulates adrenal aldosterone synthesis; in systemic blood vessels, hyperaldosteronism induces vascular dysfunction by increasing endothelial reactive oxygen species generation and decreasing NO(·) levels. We hypothesized that aldosterone modulates PAH by disrupting ET(B)-eNOS signaling through a mechanism involving increased pulmonary endothelial oxidant stress. METHODS AND RESULTS In rats with PAH, elevated endothelin-1 levels were associated with elevated aldosterone levels in plasma and lung tissue and decreased lung NO(·) metabolites in the absence of left-sided heart failure. In human pulmonary artery endothelial cells, endothelin-1 increased aldosterone levels via peroxisome proliferator-activated receptor gamma coactivator-1α/steroidogenesis factor-1-dependent upregulation of aldosterone synthase. Aldosterone also increased reactive oxygen species production, which oxidatively modified cysteinyl thiols in the eNOS-activating region of ET(B) to decrease endothelin-1-stimulated eNOS activity. Substitution of ET(B)-Cys405 with alanine improved ET(B)-dependent NO(·) synthesis under conditions of oxidant stress, confirming that Cys405 is a redox-sensitive thiol that is necessary for ET(B)-eNOS signaling. In human pulmonary artery endothelial cells, mineralocorticoid receptor antagonism with spironolactone decreased aldosterone-mediated reactive oxygen species generation and restored ET(B)-dependent NO(·) production. Spironolactone or eplerenone prevented or reversed pulmonary vascular remodeling and improved cardiopulmonary hemodynamics in 2 animal models of PAH in vivo. CONCLUSIONS Our findings demonstrate that aldosterone modulates an ET(B) cysteinyl thiol redox switch to decrease pulmonary endothelium-derived NO(·) and promote PAH.
Collapse
Affiliation(s)
- Bradley A Maron
- Cardiovascular Division, Department of Medicine, Brigham and Women's Hospital & Harvard Medical School, 75 Francis St, PBB-1, Boston, MA 02115, USA.
| | | | | | | | | | | | | | | |
Collapse
|
37
|
Chandra S, Romero MJ, Shatanawi A, Alkilany AM, Caldwell RB, Caldwell RW. Oxidative species increase arginase activity in endothelial cells through the RhoA/Rho kinase pathway. Br J Pharmacol 2012; 165:506-19. [PMID: 21740411 DOI: 10.1111/j.1476-5381.2011.01584.x] [Citation(s) in RCA: 122] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
BACKGROUND AND PURPOSE NO produced by endothelial NOS is needed for normal vascular function. During diabetes, aging and hypertension, elevated levels of arginase can compete with NOS for available l-arginine, reducing NO and increasing superoxide (O(2) (.-)) production via NOS uncoupling. Elevated O(2) (.-) combines with NO to form peroxynitrite (ONOO(-)), further reducing NO. Oxidative species increase arginase activity, but the mechanism(s) involved are not known. Our study determined the mechanism involved in peroxynitrite and hydrogen peroxide-induced enhancement in endothelial arginase activity. We hypothesized that oxidative species increase arginase activity through PKC-activated RhoA/Rho kinase (ROCK) pathway. EXPERIMENTAL APPROACH Arginase activity/expression was analysed in bovine aortic endothelial cells (BAEC) treated with an ONOO(-) generator (SIN-1) or H(2) O(2). Pretreatment with inhibitors of Rho kinase (Y-27632) or PKC (Gö6976) was used to investigate the mechanism involved in arginase activation. KEY RESULTS Exposure to SIN-1 (25 µM, 24 h) or H(2) O(2) (25 µM, 8 h) increased arginase I expression and arginase activity (35% and 50%, respectively), which was prevented by ROCK inhibitor, Y-27632, PKC inhibitor, Gö6976 or siRNA to p115-Rho GEF. There was an early activation of p115-Rho GEF (SIN-1, 2 h; H(2) O(2), 1 h) and Rho A (SIN-1, 4 h; H(2) O(2), 1 h) that was prevented by using the PKC inhibitor. Exposure to SIN-1 and H(2) O(2 ) also reduced NOS activity, which was blocked by pretreatment with p115-RhoGEF siRNA. CONCLUSIONS AND IMPLICATIONS Our data indicate that the oxidative species ONOO(-) and H(2) O(2) increase arginase activity/expression through PKC-mediated activation of RhoA/Rho kinase pathway.
Collapse
Affiliation(s)
- S Chandra
- Department of Pharmacology and Toxicology, Medical College of Georgia, Georgia Health Sciences University, Augusta, Georgia, USA
| | | | | | | | | | | |
Collapse
|
38
|
Bretón-Romero R, González de Orduña C, Romero N, Sánchez-Gómez FJ, de Álvaro C, Porras A, Rodríguez-Pascual F, Laranjinha J, Radi R, Lamas S. Critical role of hydrogen peroxide signaling in the sequential activation of p38 MAPK and eNOS in laminar shear stress. Free Radic Biol Med 2012; 52:1093-100. [PMID: 22281399 DOI: 10.1016/j.freeradbiomed.2011.12.026] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/22/2011] [Revised: 12/23/2011] [Accepted: 12/28/2011] [Indexed: 01/02/2023]
Abstract
Laminar shear stress (LSS) is a protective hemodynamic regulator of endothelial function and limits the development of atherosclerosis and other vascular wall diseases related to pathophysiological generation of reactive oxygen species. LSS activates several endothelial signaling responses, including the activation of MAPKs and eNOS. Here, we explored the mechanisms of activation of these key endothelial signaling pathways. Using the cone/plate model we found that LSS (12 dyn/cm(2)) rapidly promotes endothelial intracellular generation of superoxide and hydrogen peroxide (H(2)O(2)). Physiological concentrations of H(2)O(2) (flux of 0.1 nM/min and 15 μM added extracellularly) significantly activated both eNOS and p38 MAPK. Pharmacological inhibition of NADPH oxidases (NOXs) and specific knockdown of NOX4 decreased LSS-induced p38 MAPK activation. Whereas the absence of eNOS did not alter LSS-induced p38 MAPK activation, pharmacological inhibition and knockdown of p38α MAPK blocked H(2)O(2)- and LSS-induced eNOS phosphorylation and reduced (•)NO levels. We propose a model in which LSS promotes the formation of signaling levels of H(2)O(2), which in turn activate p38α MAPK and then stimulate eNOS, leading to increased (•)NO generation and protection of endothelial function.
Collapse
Affiliation(s)
- Rosa Bretón-Romero
- Laboratorio Mixto CSIC-FRIAT de Fisiopatología Vascular y Renal, Centro de Biología Molecular Severo Ochoa, Campus Universidad Autónoma, 28049 Madrid, Spain
| | | | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Carreira RS, Lee P, Gottlieb RA. Mitochondrial therapeutics for cardioprotection. Curr Pharm Des 2012; 17:2017-35. [PMID: 21718247 DOI: 10.2174/138161211796904777] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2011] [Accepted: 06/27/2011] [Indexed: 12/22/2022]
Abstract
Mitochondria represent approximately one-third of the mass of the heart and play a critical role in maintaining cellular function-however, they are also a potent source of free radicals and pro-apoptotic factors. As such, maintaining mitochondrial homeostasis is essential to cell survival. As the dominant source of ATP, continuous quality control is mandatory to ensure their ongoing optimal function. Mitochondrial quality control is accomplished by the dynamic interplay of fusion, fission, autophagy, and mitochondrial biogenesis. This review examines these processes in the heart and considers their role in the context of ischemia-reperfusion injury. Interventions that modulate mitochondrial turnover, including pharmacologic agents, exercise, and caloric restriction are discussed as a means to improve mitochondrial quality control, ameliorate cardiovascular dysfunction, and enhance longevity.
Collapse
Affiliation(s)
- Raquel S Carreira
- BioScience Center, San Diego State University, 5500 Campanile Drive, San Diego, CA 92182-4650, USA
| | | | | |
Collapse
|
40
|
Tovilovic G, Zogovic N, Harhaji-Trajkovic L, Misirkic-Marjanovic M, Janjetovic K, Vucicevic L, Kostic-Rajacic S, Schrattenholz A, Isakovic A, Soskic V, Trajkovic V. Arylpiperazine Dopamineric Ligands Protect Neuroblastoma Cells from Nitric Oxide (NO)-Induced Mitochondrial Damage and Apoptosis. ChemMedChem 2012; 7:495-508. [DOI: 10.1002/cmdc.201100537] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2011] [Indexed: 01/26/2023]
|
41
|
Overexpression of hepatitis B x-interacting protein in HepG2 cells enhances tumor-induced angiogenesis. Mol Cell Biochem 2011; 364:165-71. [DOI: 10.1007/s11010-011-1215-5] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2011] [Accepted: 12/21/2011] [Indexed: 11/26/2022]
|
42
|
Wang J, Alexanian A, Ying R, Kizhakekuttu TJ, Dharmashankar K, Vasquez-Vivar J, Gutterman DD, Widlansky ME. Acute exposure to low glucose rapidly induces endothelial dysfunction and mitochondrial oxidative stress: role for AMP kinase. Arterioscler Thromb Vasc Biol 2011; 32:712-20. [PMID: 22207730 DOI: 10.1161/atvbaha.111.227389] [Citation(s) in RCA: 103] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
OBJECTIVE Hypoglycemia is associated with increased mortality. The reasons for this remain unclear, and the effects of low glucose exposure on vascular endothelial function remain largely unknown. We endeavored to determine the effects of low glucose on endothelial cells and intact human arterioles. METHODS AND RESULTS We exposed human umbilical vein endothelial cells to low glucose conditions in a clinically relevant range (40-70 mg/dL) and found rapid and marked reductions in nitric oxide (NO) bioavailability (P<0.001). This was associated with concomitantly increased mitochondrial superoxide production (P<0.001) and NO-dependent mitochondrial hyperpolarization (P<0.001). Reduced NO bioavailability was rapid and attributable to reduced endothelial nitric oxide synthase activity and destruction of NO. Low glucose rapidly activated AMP kinase, but physiological activation failed to restore NO bioavailability. Pharmacological AMP kinase activation led to phosphorylation of endothelial nitric oxide synthase's Ser633 activation site, reversing the adverse effects of low glucose. This protective effect was prevented by L-NG-Nitroarginine methyl ester. Intact human arterioles exposed to low glucose demonstrated marked endothelial dysfunction, which was prevented by either metformin or TEMPOL. CONCLUSION Our data suggest that moderate low glucose exposure rapidly impairs NO bioavailability and endothelial function in the human endothelium and that pharmacological AMP kinase activation inhibit this effect in an NO-dependent manner.
Collapse
Affiliation(s)
- Jingli Wang
- Department of Medicine, Division of Cardiovascular Medicine, Graduate School of Bioscience, Medical College of Wisconsin, 9200 W Wisconsin Ave, FEC Ste E5100, Milwaukee, WI 53226, USA
| | | | | | | | | | | | | | | |
Collapse
|
43
|
Xu JF, Qu JM, Li HP. N-Acetylcysteine modulates acute lung injury induced by Pseudomonas aeruginosa in rats. Clin Exp Pharmacol Physiol 2011; 38:345-51. [PMID: 21401696 DOI: 10.1111/j.1440-1681.2011.05515.x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
1. In critically ill patients, Pseudomonas aeruginosa-induced pneumonia and the lung injury associated with infection are major causes of mortality. The aim of the present study was to evaluate the protective properties of N-acetylcysteine (NAC) in rats infected with P. aeruginosa and the role of nitric oxide synthases (NOS) protein in this process. 2. Pneumonia was induced in rats by infecting them with P. aeruginosa intratracheally. One group of rats was treated with NAC (150 mg/kg per day, i.p., for 7 days). An untreated group served as the control. Samples were collected both before (0 h) and after infection (24 h). Bacterial loads in lung tissue, the lung wet : dry (W/D) ratio and pulmonary vascular permeability were assessed. Total cell and polymorphonuclear leucocyte cell counts in bronchoalveolar lavage fluid were determined. The expression of inducible (i) NOS and endothelial (e) NOS protein was analysed and correlated with indices of lung injury using Pearson's correlation analysis. 3. Bacterial load, lung injury indices and NOS expression increased after infection. Pretreatment with NAC mitigated lung injury although it did not significantly change bacterial loads. Furthermore, NAC treatment increased eNOS protein expression, but decreased iNOS expression, in lung tissues after infection. The expression of iNOS protein was positively correlated with indices of lung injury, whereas there was a negative correlation between eNOS expression and lung injury indices. 4. N-Acetylcysteine modulated P. aeruginosa-induced lung injury in rats. The results suggest that this effect maybe due to regulation of iNOS and eNOS protein expression by NAC.
Collapse
Affiliation(s)
- Jin-Fu Xu
- Department of Respiratory Medicine, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
| | | | | |
Collapse
|
44
|
Abstract
Cardiovascular diseases remain the leading cause of mortality worldwide. Recent studies of AMP-activated protein kinase (AMPK), a highly conserved sensor of cellular energy status, suggest that there might be therapeutic value in targeting the AMPK signaling pathway. AMPK is found in most mammalian tissues, including those of the cardiovascular system. As cardiovascular diseases are typically associated with blood flow occlusion and blood occlusion may induce rapid energy deficit, AMPK activation may occur during the early phase upon nutrient deprivation in cardiovascular organs. Therefore, investigation of AMPK in cardiovascular organs may help us to understand the pathophysiology of defence mechanisms in these organs. Recent studies have provided proof of concept for the idea that AMPK is protective in heart as well as in vascular endothelial and smooth muscle cells. Moreover, dysfunction of the AMPK signalling pathway is involved in the genesis and development of various cardiovascular diseases, including atherosclerosis, hypertension and stroke. The roles of AMPK in the cardiovascular system, as they are currently understood, will be presented in this review. The interaction between AMPK and other cardiovascular signalling pathways such as nitric oxide signalling is also discussed.
Collapse
Affiliation(s)
- Qiang Xu
- Department of Geriatrics, Southwest Hospital, Third Military Medical University, Chongqing, China
| | | |
Collapse
|
45
|
Mild mitochondrial uncoupling and calorie restriction increase fasting eNOS, akt and mitochondrial biogenesis. PLoS One 2011; 6:e18433. [PMID: 21483800 PMCID: PMC3069103 DOI: 10.1371/journal.pone.0018433] [Citation(s) in RCA: 57] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2010] [Accepted: 03/04/2011] [Indexed: 01/28/2023] Open
Abstract
Enhanced mitochondrial biogenesis promoted by eNOS activation is believed to play a central role in the beneficial effects of calorie restriction (CR). Since treatment of mice with dinitrophenol (DNP) promotes health and lifespan benefits similar to those observed in CR, we hypothesized that it could also impact biogenesis. We found that DNP and CR increase citrate synthase activity, PGC-1α, cytochrome c oxidase and mitofusin-2 expression, as well as fasting plasma levels of NO• products. In addition, eNOS and Akt phosphorylation in skeletal muscle and visceral adipose tissue was activated in fasting CR and DNP animals. Overall, our results indicate that systemic mild uncoupling activates eNOS and Akt-dependent pathways leading to mitochondrial biogenesis.
Collapse
|
46
|
Hirai DM, Copp SW, Schwagerl PJ, Musch TI, Poole DC. Acute effects of hydrogen peroxide on skeletal muscle microvascular oxygenation from rest to contractions. J Appl Physiol (1985) 2011; 110:1290-8. [PMID: 21372096 DOI: 10.1152/japplphysiol.01489.2010] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Reactive oxygen species, such as hydrogen peroxide (H(2)O(2)), exert a critical regulatory role on skeletal muscle function. Whether acute increases in H(2)O(2) modulate muscle microvascular O(2) delivery-utilization (Qo(2)/Vo(2)) matching [i.e., microvascular partial pressure of O(2) (Pmv(O(2)))] at rest and following the onset of contractions is unknown. The hypothesis was tested that H(2)O(2) treatment (exogenous H(2)O(2)) would enhance Pmv(O(2)) and slow Pmv(O(2)) kinetics during contractions compared with control. Anesthetized, healthy young Sprague-Dawley rats had their spinotrapezius muscles either exposed for measurement of blood flow (and therefore QO(2)), VO(2), and Pmv(O(2)), or exteriorized for measurement of force production. Electrically stimulated twitch contractions (1 Hz, ~7 V, 2-ms pulse duration, 3 min) were evoked following acute superfusion with Krebs-Henseleit (control) and H(2)O(2) (100 μM). Relative to control, H(2)O(2) treatment elicited disproportionate increases in QO(2) and VO(2) that elevated Pmv(O(2)) at rest and throughout contractions and slowed overall Pmv(O(2)) kinetics (i.e., ~85% slower mean response time; P < 0.05). Accordingly, H(2)O(2) resulted in ~33% greater overall Pmv(O(2)), as assessed by the area under the Pmv(O(2)) curve (P < 0.05). Muscle force production was not altered with H(2)O(2) treatment (P > 0.05), evidencing reduced economy during contractions (~40% decrease in the force/VO(2) relationship; P < 0.05). These findings indicate that, although increasing the driving force for blood-myocyte O(2) flux (i.e., Pmv(O(2))), transient elevations in H(2)O(2) impair skeletal muscle function (i.e., reduced economy during contractions), which mechanistically may underlie, in part, the reduced exercise tolerance in conditions associated with oxidative stress.
Collapse
Affiliation(s)
- Daniel M Hirai
- Department of Anatomy and Physiology, Kansas State University, Manhattan, KS 66506-5802, USA
| | | | | | | | | |
Collapse
|
47
|
Naito Y, Hirotani S, Sawada H, Akahori H, Tsujino T, Masuyama T. Dietary Iron Restriction Prevents Hypertensive Cardiovascular Remodeling in Dahl Salt-Sensitive Rats. Hypertension 2011; 57:497-504. [DOI: 10.1161/hypertensionaha.110.159681] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Iron accumulation is associated with the pathogenesis of several cardiovascular diseases. However, the preventive effects of iron restriction (IR) against cardiovascular disease remain obscure. We investigated the effects of dietary IR on cardiovascular pathophysiology and the involved mechanism in Dahl salt-sensitive rats. Dahl salt-sensitive rats were provided either a normal or high-salt (HS) diet. Another subset of Dahl salt-sensitive rats were fed an HS with iron-restricted (HS+IR) diet for 11 weeks. Dahl salt-sensitive rats given an HS diet developed hypertension, heart failure, and decreased a survival rate after 11 weeks on the diet. In contrast, IR attenuated the development of hypertension and heart failure, thereby improving survival rate. Dietary IR suppressed cardiovascular hypertrophy, fibrosis, and inflammation in HS rats. The phosphorylation of Akt, AMP-activated protein kinase, and endothelial nitric oxide synthase was decreased in the aorta of HS rats, whereas they were ameliorated by the IR diet. Aortic expression of the cellular iron import protein transferrin receptor 1, and the iron storage protein ferritin H-subunit, was upregulated in HS rats. IR also attenuated proteinuria and increased oxidative stress in the HS group.
N
G
-nitro-
l
-arginine methyl ester abolished the beneficial effects of IR and decreased survival rate in HS+IR rats. Dietary IR had protective effects on salt-induced hypertension, cardiovascular remodeling, and proteinuria through the inhibition of oxidative stress, and maintenance of Akt, AMP-activated protein kinase, and endothelial nitric oxide synthase in the aorta. IR could be an effective strategy for prevention of HS-induced organ damage in salt-sensitive hypertensive patients.
Collapse
Affiliation(s)
- Yoshiro Naito
- From the Cardiovascular Division, Department of Internal Medicine (Y.N., S.H., H.S., H.A., T.M.), Hyogo College of Medicine, Nishinomiya, Japan; Department of Pharmacy (T.T.), Hyogo University of Health Sciences, Kobe, Japan
| | - Shinichi Hirotani
- From the Cardiovascular Division, Department of Internal Medicine (Y.N., S.H., H.S., H.A., T.M.), Hyogo College of Medicine, Nishinomiya, Japan; Department of Pharmacy (T.T.), Hyogo University of Health Sciences, Kobe, Japan
| | - Hisashi Sawada
- From the Cardiovascular Division, Department of Internal Medicine (Y.N., S.H., H.S., H.A., T.M.), Hyogo College of Medicine, Nishinomiya, Japan; Department of Pharmacy (T.T.), Hyogo University of Health Sciences, Kobe, Japan
| | - Hirokuni Akahori
- From the Cardiovascular Division, Department of Internal Medicine (Y.N., S.H., H.S., H.A., T.M.), Hyogo College of Medicine, Nishinomiya, Japan; Department of Pharmacy (T.T.), Hyogo University of Health Sciences, Kobe, Japan
| | - Takeshi Tsujino
- From the Cardiovascular Division, Department of Internal Medicine (Y.N., S.H., H.S., H.A., T.M.), Hyogo College of Medicine, Nishinomiya, Japan; Department of Pharmacy (T.T.), Hyogo University of Health Sciences, Kobe, Japan
| | - Tohru Masuyama
- From the Cardiovascular Division, Department of Internal Medicine (Y.N., S.H., H.S., H.A., T.M.), Hyogo College of Medicine, Nishinomiya, Japan; Department of Pharmacy (T.T.), Hyogo University of Health Sciences, Kobe, Japan
| |
Collapse
|
48
|
Nediani C, Raimondi L, Borchi E, Cerbai E. Nitric oxide/reactive oxygen species generation and nitroso/redox imbalance in heart failure: from molecular mechanisms to therapeutic implications. Antioxid Redox Signal 2011; 14:289-331. [PMID: 20624031 DOI: 10.1089/ars.2010.3198] [Citation(s) in RCA: 61] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Adaptation of the heart to intrinsic and external stress involves complex modifications at the molecular and cellular levels that lead to tissue remodeling, functional and metabolic alterations, and finally to failure depending upon the nature, intensity, and chronicity of the stress. Reactive oxygen species (ROS) have long been considered as merely harmful entities, but their role as second messengers has gradually emerged. At the same time, our comprehension of the multifaceted role of nitric oxide (NO) and the related reactive nitrogen species (RNS) has been upgraded. The tight interlay between ROS and RNS suggests that their imbalance may implicate the impairment in physiological NO/redox-based signaling that contributes to the failing of the cardiovascular system. This review initially provides basic concepts on the role of nitroso/oxidative stress in the pathophysiology of heart failure with a particular focus on sources of ROS/RNS, their downstream targets, and endogenous modulators. Then, the role of NO/redox regulation of cardiomyocyte function, including calcium homeostasis, electrogenesis, and insulin signaling pathways, is described. Finally, an overview of old and emerging therapeutic opportunities in heart failure is presented, focusing on modulation of NO/redox mechanisms and discussing benefits and limitations.
Collapse
Affiliation(s)
- Chiara Nediani
- Department of Biochemical Sciences, University of Florence, Florence, Italy.
| | | | | | | |
Collapse
|
49
|
Wong WT, Tian XY, Xu A, Ng CF, Lee HK, Chen ZY, Au CL, Yao X, Huang Y. Angiotensin II type 1 receptor-dependent oxidative stress mediates endothelial dysfunction in type 2 diabetic mice. Antioxid Redox Signal 2010; 13:757-68. [PMID: 20136508 DOI: 10.1089/ars.2009.2831] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
The mechanisms underlying the effect of the renin-angiotensin-aldosterone system (RAAS) inhibition on endothelial dysfunction in type 2 diabetes are incompletely understood. This study explored a causal relationship between RAAS activation and oxidative stress involved in diabetes-associated endothelial dysfunction. Daily oral administration of valsartan or enalapril at 10 mg/kg/day to db/db mice for 6 weeks reversed the blunted acetylcholine-induced endothelium-dependent dilatations, suppressed the upregulated expression of angiotensin II type 1 receptor (AT(1)R) and NAD(P)H oxidase subunits (p22(phox) and p47(phox)), and reduced reactive oxygen species (ROS) production. Acute exposure to AT(1)R blocker losartan restored the impaired endothelium-dependent dilatations in aortas of db/db mice and also in renal arteries of diabetic patients (fasting plasma glucose level > or =7.0 mmol/l). Similar observations were also made with apocynin, diphenyliodonium, or tempol treatment in db/db mouse aortas. DHE fluorescence revealed an overproduction of ROS in db/db aortas which was sensitive to inhibition by losartan or ROS scavengers. Losartan also prevented the impairment of endothelium-dependent dilatations under hyperglycemic conditions that were accompanied by high ROS production. The present study has identified an initiative role of AT(1)R activation in mediating endothelial dysfunction of arteries from db/db mice and diabetic patients.
Collapse
Affiliation(s)
- Wing Tak Wong
- Institute of Vascular Medicine and Li Ka Shing Institute of Health Sciences, Chinese University of Hong Kong, China
| | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Hien TT, Kim ND, Pokharel YR, Oh SJ, Lee MY, Kang KW. Ginsenoside Rg3 increases nitric oxide production via increases in phosphorylation and expression of endothelial nitric oxide synthase: essential roles of estrogen receptor-dependent PI3-kinase and AMP-activated protein kinase. Toxicol Appl Pharmacol 2010; 246:171-83. [PMID: 20546771 DOI: 10.1016/j.taap.2010.05.008] [Citation(s) in RCA: 65] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2010] [Revised: 05/11/2010] [Accepted: 05/14/2010] [Indexed: 11/29/2022]
Abstract
We previously showed that ginsenosides increase nitric oxide (NO) production in vascular endothelium and that ginsenoside Rg3 (Rg3) is the most active one among ginseng saponins. However, the mechanism for Rg3-mediated nitric oxide production is still uncertain. In this study, we determined whether Rg3 affects phosphorylation and expression of endothelial nitric oxide synthase (eNOS) in ECV 304 human endothelial cells. Rg3 increased both the phosphorylation and the expression of eNOS in a concentration-dependent manner and a maximal effect was found at 10μg/ml of Rg3. The enzyme activities of phosphatidylinositol 3-kinase (PI3-kinase), c-Jun N-terminal kinase (JNK), and p38 kinase were enhanced as were estrogen receptor (ER)- and glucocorticoid receptor (GR)-dependent reporter gene transcriptions in Rg3-treated endothelial cells. Rg3-induced eNOS phosphorylation required the ER-mediated PI3-kinase/Akt pathway. Moreover, Rg3 activates AMP-activated protein kinase (AMPK) through up-regulation of CaM kinase II and Rg3-stimulated eNOS phosphorylation was reversed by AMPK inhibition. The present results provide a mechanism for Rg3-stimulated endothelial NO production.
Collapse
Affiliation(s)
- Tran Thi Hien
- BK21 Project Team, College of Pharmacy, Chosun University, 375 Seosuk-dong, Dong-gu, Gwangju 501-759, South Korea
| | - Nak Doo Kim
- Seoul National University, Seoul 151-745, South Korea
| | - Yuba Raj Pokharel
- BK21 Project Team, College of Pharmacy, Chosun University, 375 Seosuk-dong, Dong-gu, Gwangju 501-759, South Korea
| | - Seok Jeong Oh
- College of Pharmacy, Chonnam National University, Gwangju 500-757, South Korea
| | - Moo Yeol Lee
- College of Pharmacy, Chonnam National University, Gwangju 500-757, South Korea
| | - Keon Wook Kang
- BK21 Project Team, College of Pharmacy, Chosun University, 375 Seosuk-dong, Dong-gu, Gwangju 501-759, South Korea
| |
Collapse
|