1
|
Tain YL, Hsu CN. Kidney Programming and Hypertension: Linking Prenatal Development to Adulthood. Int J Mol Sci 2024; 25:13610. [PMID: 39769369 PMCID: PMC11677590 DOI: 10.3390/ijms252413610] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2024] [Revised: 12/12/2024] [Accepted: 12/18/2024] [Indexed: 01/11/2025] Open
Abstract
The complex relationship between kidney disease and hypertension represents a critical area of research, yet less attention has been devoted to exploring how this connection develops early in life. Various environmental factors during pregnancy and lactation can significantly impact kidney development, potentially leading to kidney programming that results in alterations in both structure and function. This early programming can contribute to adverse long-term kidney outcomes, such as hypertension. In the context of kidney programming, the molecular pathways involved in hypertension are intricate and include epigenetic modifications, oxidative stress, impaired nitric oxide pathway, inappropriate renin-angiotensin system (RAS) activation, disrupted nutrient sensing, gut microbiota dysbiosis, and altered sodium transport. This review examines each of these mechanisms and highlights reprogramming interventions proposed in preclinical studies to prevent hypertension related to kidney programming. Given that reprogramming strategies differ considerably from conventional treatments for hypertension in kidney disease, it is essential to shift focus toward understanding the processes of kidney programming and its role in the development of programmed hypertension.
Collapse
Affiliation(s)
- You-Lin Tain
- Division of Pediatric Nephrology, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung 833, Taiwan;
- College of Medicine, Chang Gung University, Taoyuan 333, Taiwan
| | - Chien-Ning Hsu
- Department of Pharmacy, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung 833, Taiwan
- School of Pharmacy, Kaohsiung Medical University, Kaohsiung 807, Taiwan
| |
Collapse
|
2
|
Liu H, Li Y, Li M, Xie L, Li F, Pan R, Pei F. Follistatin-like 1 protects endothelial function in the spontaneously hypertensive rat by inhibition of endoplasmic reticulum stress through AMPK-dependent mechanism. Clin Exp Hypertens 2023; 45:2277654. [PMID: 37963199 DOI: 10.1080/10641963.2023.2277654] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2023] [Accepted: 10/26/2023] [Indexed: 11/16/2023]
Abstract
OBJECTIVE Endothelial dysfunction is a critical initiating factor in the development of hypertension and related complications. Follistatin-like 1 (FSTL1) can promote endothelial cell function and stimulates revascularization in response to ischemic insult. However, it is unclear whether FSTL1 has an effect on ameliorating endothelial dysfunction in spontaneously hypertensive rats (SHRs). METHODS Wistar Kyoto (WKY) and SHRs were treated with a tail vein injection of vehicle (1 mL/day) or recombinant FSTL1 (100 μg/kg body weight/day) for 4 weeks. Blood pressure was measured by tail-cuff plethysmograph, and vascular reactivity in mesenteric arteries was measured using wire myography. RESULTS We found that treatment with FSTL1 reversed impaired endothelium-dependent relaxation (EDR) in mesenteric arteries and lowered blood pressure of SHRs. Decreased AMP-activated protein kinase (AMPK) phosphorylation, elevated endoplasmic reticulum (ER) stress markers, increased reactive oxygen species (ROS), and reduction of nitric oxide (NO) production in mesenteric arteries of SHRs were also reversed by FSTL1 treatment. Ex vivo treatment with FSTL1 improved the impaired EDR in mesenteric arteries from SHRs and reversed tunicamycin (ER stress inducer)-induced ER stress and the impairment of EDR in mesenteric arteries from WKY rats. The effects of FSTL1 were abolished by cotreatment of compound C (AMPK inhibitor). CONCLUSIONS These results suggest that FSTL1 prevents endothelial dysfunction in mesenteric arteries of SHRs through inhibiting ER stress and ROS and increasing NO production via activation of AMPK signaling.
Collapse
Affiliation(s)
- Hanwen Liu
- Department of Neurosurgery, Ganzhou Hospital of Guangdong Provincial People's Hospital, Ganzhou Municipal Hospital, Ganzhou, Jiangxi, China
| | - Yanwen Li
- Department of Neurosurgery, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Maogang Li
- Department of Neurosurgery, Ganzhou Hospital of Guangdong Provincial People's Hospital, Ganzhou Municipal Hospital, Ganzhou, Jiangxi, China
| | - Linghai Xie
- Department of Neurosurgery, Ganzhou Hospital of Guangdong Provincial People's Hospital, Ganzhou Municipal Hospital, Ganzhou, Jiangxi, China
| | - Feng Li
- Department of Neurosurgery, Ganzhou Hospital of Guangdong Provincial People's Hospital, Ganzhou Municipal Hospital, Ganzhou, Jiangxi, China
| | - Runmei Pan
- Operating room, Ganzhou Hospital of Guangdong Provincial People's Hospital, Ganzhou Municipal Hospital, Ganzhou, China
| | - Fang Pei
- Department of Cardiology, Ganzhou Hospital of Guangdong Provincial People's Hospital, Ganzhou Municipal Hospital, Ganzhou, Jiangxi, China
| |
Collapse
|
3
|
Pyla R, Hartney TJ, Segar L. AICAR promotes endothelium-independent vasorelaxation by activating AMP-activated protein kinase via increased ZMP and decreased ATP/ADP ratio in aortic smooth muscle. J Basic Clin Physiol Pharmacol 2022; 33:759-768. [PMID: 35503763 DOI: 10.1515/jbcpp-2021-0308] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2021] [Accepted: 04/05/2022] [Indexed: 11/15/2022]
Abstract
OBJECTIVES AICAR, an adenosine analog, has been shown to exhibit vascular protective effects through activation of AMP-activated protein kinase (AMPK). However, it remains unclear as to whether adenosine kinase-mediated ZMP formation or adenosine receptor activation contributes to AICAR-mediated AMPK activation and/or vasorelaxant response in vascular smooth muscle. METHODS AND RESULTS In the present study using endothelium-denuded rat aortic ring preparations, isometric tension measurements revealed that exposure to 1 mM AICAR for 30 min resulted in inhibition of phenylephrine (1 μM)-induced smooth muscle contractility by ∼35%. Importantly, this vasorelaxant response by AICAR was prevented after pretreatment of aortic rings with an AMPK inhibitor (compound C, 40 µM) and adenosine kinase inhibitor (5-iodotubercidin, 1 µM), but not with an adenosine receptor blocker (8-sulfophenyltheophylline, 100 µM). Immunoblot analysis of respective aortic tissues showed that AMPK activation seen during vasorelaxant response by AICAR was abolished by compound C and 5-iodotubercidin, but not by 8-sulfophenyltheophylline, suggesting ZMP involvement in AMPK activation. Furthermore, LC-MS/MS MRM analysis revealed that exposure of aortic smooth muscle cells to 1 mM AICAR for 30 min enhanced ZMP level to 2014.9 ± 179.4 picomoles/mg protein (vs. control value of 8.5 ± 0.6; p<0.01), which was accompanied by a significant decrease in ATP/ADP ratio (1.08 ± 0.02 vs. 2.08 ± 0.06; p<0.01). CONCLUSIONS Together, the present findings demonstrate that AICAR-mediated ZMP elevation and the resultant AMPK activation in vascular smooth muscle contribute to vasorelaxation.
Collapse
Affiliation(s)
- Rajkumar Pyla
- Charlie Norwood VA Medical Center, Augusta, GA, USA.,Center for Pharmacy and Experimental Therapeutics, University of Georgia College of Pharmacy, Augusta, GA, USA
| | | | - Lakshman Segar
- Charlie Norwood VA Medical Center, Augusta, GA, USA.,Center for Pharmacy and Experimental Therapeutics, University of Georgia College of Pharmacy, Augusta, GA, USA.,Vascular Biology Center, Department of Pharmacology and Toxicology, Augusta University, Augusta, GA, USA.,Department of Medicine, Pennsylvania State University College of Medicine, Hershey, PA, USA
| |
Collapse
|
4
|
Lorca RA, Houck JA, Laurent LC, Matarazzo CJ, Baker K, Horii M, Nelson KK, Bales ES, Euser AG, Parast MM, Moore LG, Julian CG. High altitude regulates the expression of AMPK pathways in human placenta. Placenta 2021; 104:267-276. [PMID: 33472134 DOI: 10.1016/j.placenta.2021.01.010] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/16/2020] [Revised: 12/29/2020] [Accepted: 01/08/2021] [Indexed: 12/24/2022]
Abstract
INTRODUCTION High-altitude (>2500 m) residence augments the risk of intrauterine growth restriction (IUGR) and preeclampsia likely due, in part, to uteroplacental hypoperfusion. Previous genomic and transcriptomic studies in humans and functional studies in mice and humans suggest a role for AMP-activated protein kinase (AMPK) pathway in protecting against hypoxia-associated IUGR. AMPK is a metabolic sensor activated by hypoxia that is ubiquitously expressed in vascular beds and placenta. METHODS We measured gene expression and protein levels of AMPK and its upstream regulators and downstream targets in human placentas from high (>2500 m) vs. moderate (~1700 m) and low (~100 m) altitude. RESULTS We found that phosphorylated AMPK protein levels and its downstream target TSC2 were increased in placentas from high and moderate vs. low altitude, whereas the phosphorylated form of the downstream target translation repressor protein 4E-BP1 was increased in high compared to moderate as well as low altitude placentas. Mean birth weights progressively fell with increasing altitude but no infants, by study design, were clinically growth-restricted. Gene expression analysis showed moderate increases in PRKAG2, encoding the AMPK γ2 subunit, and mechanistic target of rapamycin, MTOR, expression. DISCUSSION These results highlight a differential regulation of placental AMPK pathway activation in women residing at low, moderate or high altitude during pregnancy, suggesting AMPK may be serving as a metabolic regulator for integrating hypoxic stimuli with placental function.
Collapse
Affiliation(s)
- Ramón A Lorca
- Division of Reproductive Sciences, Department of Obstetrics and Gynecology, University of Colorado Denver-Anschutz Medical Campus, Aurora, CO, 80045, USA.
| | - Julie A Houck
- Division of Biomedical Informatics and Personalized Medicine, Department of Medicine, University of Colorado Denver-Anschutz Medical Campus, Aurora, CO, 80045, USA
| | - Louise C Laurent
- Department of Reproductive Medicine, University of California San Diego, La Jolla, 92093, USA
| | - Christopher J Matarazzo
- Division of Biomedical Informatics and Personalized Medicine, Department of Medicine, University of Colorado Denver-Anschutz Medical Campus, Aurora, CO, 80045, USA
| | - Kori Baker
- Division of Reproductive Sciences, Department of Obstetrics and Gynecology, University of Colorado Denver-Anschutz Medical Campus, Aurora, CO, 80045, USA
| | - Mariko Horii
- Department of Pathology, University of California San Diego, La Jolla, 92093, USA
| | - Katharine K Nelson
- Department of Pathology, University of California San Diego, La Jolla, 92093, USA
| | - Elise S Bales
- Division of Reproductive Sciences, Department of Obstetrics and Gynecology, University of Colorado Denver-Anschutz Medical Campus, Aurora, CO, 80045, USA
| | - Anna G Euser
- Division of Maternal-Fetal Medicine, Department of Obstetrics and Gynecology, University of Colorado Denver-Anschutz Medical Campus, Aurora, CO, 80045, USA
| | - Mana M Parast
- Department of Pathology, University of California San Diego, La Jolla, 92093, USA
| | - Lorna G Moore
- Division of Reproductive Sciences, Department of Obstetrics and Gynecology, University of Colorado Denver-Anschutz Medical Campus, Aurora, CO, 80045, USA
| | - Colleen G Julian
- Division of Biomedical Informatics and Personalized Medicine, Department of Medicine, University of Colorado Denver-Anschutz Medical Campus, Aurora, CO, 80045, USA
| |
Collapse
|
5
|
Lane SL, Houck JA, Doyle AS, Bales ES, Lorca RA, Julian CG, Moore LG. AMP-activated protein kinase activator AICAR attenuates hypoxia-induced murine fetal growth restriction in part by improving uterine artery blood flow. J Physiol 2020; 598:4093-4105. [PMID: 32592403 DOI: 10.1113/jp279341] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2019] [Accepted: 06/24/2020] [Indexed: 12/20/2022] Open
Abstract
KEY POINTS Pregnancy at high altitude is associated with a greater incidence of fetal growth restriction due, in part, to lesser uterine artery blood flow. AMP-activated protein kinase (AMPK) activation vasodilates arteries and may increase uterine artery blood flow. In this study, pharmacological activation of AMPK by the drug AICAR improved fetal growth and elevated uterine artery blood flow. These results suggest that AMPK activation is a potential strategy for improving fetal growth and raising uterine artery blood flow in pregnancy, which may be important in pregnancy disorders characterized by uteroplacental ischaemia and/or fetal hypoxia. ABSTRACT Uteroplacental hypoxia is associated with pregnancy disorders such as intrauterine growth restriction and preeclampsia, which are characterized by uteroplacental ischaemia and/or fetal hypoxia. Activation of AMP-activated protein kinase (AMPK) results in vasodilatation and is therefore a potential therapeutic strategy for restoring uteroplacental perfusion in pregnancy disorders. In this study, C57Bl/6 mice were treated with subcutaneous pellets containing vehicle, the AMPK activator AICAR (200 mg kg-1 day-1 ), or the AMPK inhibitor Compound C (20 mg kg-1 day-1 ) beginning on gestational day 13.5, and were exposed to hypoxia starting on gestational day 14.5 that induced intrauterine growth restriction. Pharmacological AMPK activation by AICAR partially prevented hypoxia-induced fetal growth restriction (P < 0.01), due in part to increased uterine artery blood flow (P < 0.0001). The proportion of total cardiac output flowing through the uterine artery was increased with AICAR in hypoxic mice (P < 0.001), suggesting that the vasodilator effect of AICAR was selective for the uterine circulation. Further, pharmacological inhibition of AMPK with Compound C reduced uterine artery diameter and increased uterine artery contractility in normoxic mice, providing evidence that physiological levels of AMPK activation are necessary for vasodilatation in healthy pregnancy. Two-way ANOVA analyses indicated that hypoxia reduced AMPK activation in the uterine artery and placenta, and AICAR increased AMPK activation in these tissues compared to vehicle. These findings provide support for further investigation into the utility of pharmacological AMPK activation for treatment of fetal growth restriction.
Collapse
Affiliation(s)
- Sydney L Lane
- Integrated Physiology PhD Program, University of Colorado Graduate School, Aurora, CO, USA.,Division of Reproductive Sciences, Department of Obstetrics and Gynecology, University of Colorado School of Medicine, Aurora, CO, USA
| | - Julie A Houck
- Division of Bioinformatics and Personalized Medicine, Department of Medicine, University of Colorado School of Medicine, Aurora, CO, USA
| | - Alexandrea S Doyle
- Division of Bioinformatics and Personalized Medicine, Department of Medicine, University of Colorado School of Medicine, Aurora, CO, USA
| | - Elise S Bales
- Division of Reproductive Sciences, Department of Obstetrics and Gynecology, University of Colorado School of Medicine, Aurora, CO, USA
| | - Ramón A Lorca
- Division of Reproductive Sciences, Department of Obstetrics and Gynecology, University of Colorado School of Medicine, Aurora, CO, USA
| | - Colleen G Julian
- Division of Bioinformatics and Personalized Medicine, Department of Medicine, University of Colorado School of Medicine, Aurora, CO, USA
| | - Lorna G Moore
- Division of Reproductive Sciences, Department of Obstetrics and Gynecology, University of Colorado School of Medicine, Aurora, CO, USA
| |
Collapse
|
6
|
Oduro PK, Fang J, Niu L, Li Y, Li L, Zhao X, Wang Q. Pharmacological management of vascular endothelial dysfunction in diabetes: TCM and western medicine compared based on biomarkers and biochemical parameters. Pharmacol Res 2020; 158:104893. [PMID: 32434053 DOI: 10.1016/j.phrs.2020.104893] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/12/2020] [Revised: 04/18/2020] [Accepted: 05/03/2020] [Indexed: 12/20/2022]
Abstract
Diabetes, a worldwide health concern while burdening significant populace of countries with time due to a hefty increase in both incidence and prevalence rates. Hyperglycemia has been buttressed both in clinical and experimental studies to modulate widespread molecular actions that effect macro and microvascular dysfunctions. Endothelial dysfunction, activation, inflammation, and endothelial barrier leakage are key factors contributing to vascular complications in diabetes, plus the development of diabetes-induced cardiovascular diseases. The recent increase in molecular, transcriptional, and clinical studies has brought a new scope to the understanding of molecular mechanisms and the therapeutic targets for endothelial dysfunction in diabetes. In this review, an attempt made to discuss up to date critical and emerging molecular signaling pathways involved in the pathophysiology of endothelial dysfunction and viable pharmacological management targets. Importantly, we exploit some Traditional Chinese Medicines (TCM)/TCM isolated bioactive compounds modulating effects on endothelial dysfunction in diabetes. Finally, clinical studies data on biomarkers and biochemical parameters involved in the assessment of the efficacy of treatment in vascular endothelial dysfunction in diabetes was compared between clinically used western hypoglycemic drugs and TCM formulas.
Collapse
Affiliation(s)
- Patrick Kwabena Oduro
- Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, PR China; Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Ministry of Education, Tianjin 301617, PR China
| | - Jingmei Fang
- Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, PR China; Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Ministry of Education, Tianjin 301617, PR China
| | - Lu Niu
- Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, PR China; Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Ministry of Education, Tianjin 301617, PR China
| | - Yuhong Li
- Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, PR China; Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Ministry of Education, Tianjin 301617, PR China; Tianjin Key Laboratory of Chinese medicine Pharmacology, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, PR China
| | - Lin Li
- Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, PR China; Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Ministry of Education, Tianjin 301617, PR China; Tianjin Key Laboratory of Chinese medicine Pharmacology, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, PR China
| | - Xin Zhao
- Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, PR China; Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Ministry of Education, Tianjin 301617, PR China; Tianjin Key Laboratory of Chinese medicine Pharmacology, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, PR China
| | - Qilong Wang
- Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, PR China; Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Ministry of Education, Tianjin 301617, PR China; Tianjin Key Laboratory of Chinese medicine Pharmacology, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, PR China.
| |
Collapse
|
7
|
Chen H, Vanhoutte PM, Leung SWS. Vascular adenosine monophosphate-activated protein kinase: Enhancer, brake or both? Basic Clin Pharmacol Toxicol 2019; 127:81-91. [PMID: 31671245 DOI: 10.1111/bcpt.13357] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2019] [Accepted: 10/24/2019] [Indexed: 12/25/2022]
Abstract
Adenosine monophosphate-activated protein kinase (AMPK), expressed/present ubiquitously in the body, contributes to metabolic regulation. In the vasculature, activation of AMPK is associated with several beneficial biological effects including enhancement of vasodilatation, reduction of oxidative stress and inhibition of inflammatory reactions. The vascular protective effects of certain anti-diabetic (metformin and sitagliptin) or lipid-lowering (simvastatin and fenofibrate) therapeutic agents, of active components of Chinese medicinal herbs (resveratrol and berberine) and of pharmacological agents (AICAR, A769662 and PT1) have been attributed to the activation of AMPK (in endothelial cells, vascular smooth muscle cells and/or perivascular adipocytes), independently of changes in the metabolic profile (eg glucose tolerance and/or plasma lipoprotein levels), leading to improved endothelium-derived nitric oxide-mediated vasodilatation and attenuated endothelium-derived cyclooxygenase-dependent vasoconstriction. By contrast, endothelial AMPK activation with pharmacological agents or by genetic modification is associated with reduced endothelium-dependent relaxations in small blood vessels and elevated systolic blood pressure. Indeed, AMPK activators inhibit endothelium-dependent hyperpolarization (EDH)-type relaxations in superior mesenteric arteries, partly by inhibiting endothelial calcium-activated potassium channel signalling. Therefore, AMPK activation is not necessarily beneficial in terms of endothelial function. The contribution of endothelial AMPK in the regulation of vascular tone, in particular in the microvasculature where EDH plays a more important role, remains to be characterized.
Collapse
Affiliation(s)
- Hui Chen
- Department of Pharmacology and Pharmacy, The University of Hong Kong, Hong Kong, China
| | - Paul Michel Vanhoutte
- Department of Pharmacology and Pharmacy, The University of Hong Kong, Hong Kong, China.,State Key Laboratory of Pharmaceutical Biotechnology, The University of Hong Kong, Hong Kong, China.,Department of Cardiovascular and Renal Research, Institute for Molecular Medicine, University of Southern Denmark, Odense, Denmark
| | - Susan Wai Sum Leung
- Department of Pharmacology and Pharmacy, The University of Hong Kong, Hong Kong, China
| |
Collapse
|
8
|
Ying L, Li N, He Z, Zeng X, Nan Y, Chen J, Miao P, Ying Y, Lin W, Zhao X, Lu L, Chen M, Cen W, Guo T, Li X, Huang Z, Wang Y. Fibroblast growth factor 21 Ameliorates diabetes-induced endothelial dysfunction in mouse aorta via activation of the CaMKK2/AMPKα signaling pathway. Cell Death Dis 2019; 10:665. [PMID: 31511499 PMCID: PMC6739326 DOI: 10.1038/s41419-019-1893-6] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2019] [Revised: 08/06/2019] [Accepted: 08/20/2019] [Indexed: 12/16/2022]
Abstract
Endothelial dysfunction initiates and exacerbates hypertension, atherosclerosis and other cardiovascular complications in diabetic mellitus. FGF21 is a hormone that mediates a number of beneficial effects relevant to metabolic disorders and their associated complications. Nevertheless, it remains unclear as to whether FGF21 ameliorates endothelial dysfunction. Therefore, we investigated the effect of FGF21 on endothelial function in both type 1 and type 2 diabetes. We found that FGF21 reduced hyperglycemia and ameliorated insulin resistance in type 2 diabetic mice, an effect that was totally lost in type 1 diabetic mice. However, FGF21 activated AMPKα, suppressing oxidative stress and enhancing endothelium-dependent vasorelaxation of aorta in both types, suggesting a mechanism that is independent of its glucose-lowering and insulin-sensitizing effects. In vitro, we identified a direct action of FGF21 on endothelial cells of the aorta, in which it bounds to FGF receptors to alleviate impaired endothelial function challenged with high glucose. Furthermore, the CaMKK2-AMPKα signaling pathway was activated to suppress oxidative stress. Apart from its anti-oxidative capacity, FGF21 activated eNOS to dilate the aorta via CaMKK2/AMPKα activation. Our data suggest expanded potential uses of FGF21 for the treatment of vascular diseases in diabetes.
Collapse
Affiliation(s)
- Lei Ying
- Department of Pathophysiology, School of Basic Medical Sciences, Wenzhou Medical University, 325035, Wenzhou, Zhejiang, China
| | - Na Li
- Department of Pathophysiology, School of Basic Medical Sciences, Wenzhou Medical University, 325035, Wenzhou, Zhejiang, China.,Department of Pathology, Wenzhou Central Hospital, 325035, Wenzhou, Zhejiang, China
| | - Zhengyue He
- Department of Pathophysiology, School of Basic Medical Sciences, Wenzhou Medical University, 325035, Wenzhou, Zhejiang, China.,Department of Pathology, Suining Central Hospital, 629000, Suining, Sichuan, China
| | - Xueqin Zeng
- Department of Pathophysiology, School of Basic Medical Sciences, Wenzhou Medical University, 325035, Wenzhou, Zhejiang, China
| | - Yan Nan
- The Second Affiliated Hospital and Yuying Children's Hospital, Wenzhou Medical University, 325035, Wenzhou, Zhejiang, China
| | - Jiantong Chen
- Department of Pathophysiology, School of Basic Medical Sciences, Wenzhou Medical University, 325035, Wenzhou, Zhejiang, China
| | - Peipei Miao
- Department of Pathophysiology, School of Basic Medical Sciences, Wenzhou Medical University, 325035, Wenzhou, Zhejiang, China.,Department of Pharmacy, the Second People's Hospital of Pingyang, 325035, Wenzhou, Zhejiang, China
| | - Yunyun Ying
- Department of Pathophysiology, School of Basic Medical Sciences, Wenzhou Medical University, 325035, Wenzhou, Zhejiang, China.,The First Affiliated Hospital & School of the First Clinical Medical Sciences, Wenzhou Medical University, 325035, Wenzhou, Zhejiang, China
| | - Wei Lin
- School of Pharmaceutical Sciences, Wenzhou Medical University, 325035, Wenzhou, Zhejiang, China
| | - Xinyu Zhao
- School of Pharmaceutical Sciences, Wenzhou Medical University, 325035, Wenzhou, Zhejiang, China
| | - Lu Lu
- The Second Affiliated Hospital and Yuying Children's Hospital, Wenzhou Medical University, 325035, Wenzhou, Zhejiang, China
| | - Mengke Chen
- The First Affiliated Hospital & School of the First Clinical Medical Sciences, Wenzhou Medical University, 325035, Wenzhou, Zhejiang, China
| | - Wei Cen
- The First Affiliated Hospital & School of the First Clinical Medical Sciences, Wenzhou Medical University, 325035, Wenzhou, Zhejiang, China
| | - Tonglin Guo
- School of Ophthalmology and Optometry, Wenzhou Medical University, 325035, Wenzhou, Zhejiang, China
| | - Xiaokun Li
- School of Pharmaceutical Sciences, Wenzhou Medical University, 325035, Wenzhou, Zhejiang, China.
| | - Zhifeng Huang
- School of Pharmaceutical Sciences, Wenzhou Medical University, 325035, Wenzhou, Zhejiang, China.
| | - Yang Wang
- Department of Pathophysiology, School of Basic Medical Sciences, Wenzhou Medical University, 325035, Wenzhou, Zhejiang, China.
| |
Collapse
|
9
|
Chen H, Vanhoutte PM, Leung SWS. Acute activation of endothelial AMPK surprisingly inhibits endothelium-dependent hyperpolarization-like relaxations in rat mesenteric arteries. Br J Pharmacol 2019; 176:2905-2921. [PMID: 31116877 DOI: 10.1111/bph.14716] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2018] [Revised: 05/04/2019] [Accepted: 05/08/2019] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND AND PURPOSE Endothelium-dependent hyperpolarizations (EDHs) contribute to the regulation of peripheral resistance. They are initiated through opening of endothelial calcium-activated potassium channels (KCa ); the potassium ions released then diffuse to the underlying smooth muscle cells, causing hyperpolarization and thus relaxation. The present study aimed to examine whether or not AMPK modulates EDH-like relaxations in rat mesenteric arteries. EXPERIMENTAL APPROACH Arterial rings were isolated for isometric tension recording. AMPK activity and protein level were measured by ELISA and western blotting respectively. KEY RESULTS The AMPK activator, AICAR, reduced ACh-induced EDH-like relaxations and increased AMPK activity in preparations with endothelium; these responses were prevented by compound C, an AMPK inhibitor. AICAR inhibited relaxations induced by SKA-31 (opener of endothelial KCa ) but did not affect potassium-induced, hyperpolarization-attributable relaxations or increase AMPK activity in preparations without endothelium. A769662, another AMPK activator, not only caused a similar inhibition of relaxations to ACh and SKA-31 in preparations with endothelium but also inhibited hyperpolarization-attributable relaxations and augmented AMPK activity in rings without endothelium. Protein levels of total AMPKα, AMPKα1, or AMPKβ1/2 were comparable between preparations with and without endothelium. CONCLUSIONS AND IMPLICATIONS Activation of endothelial AMPK, by either AICAR or A769662, acutely inhibits EDH-like relaxations of rat mesenteric arteries. Furthermore, A769662 inhibits signalling downstream of smooth muscle hyperpolarization. In view of the major blunting effect of AMPK activation on EDH-like relaxations, caution should be applied when administering therapeutic agents that activate AMPK in patients with endothelial dysfunction characterized by reduced production and/or bioavailability of NO.
Collapse
Affiliation(s)
- Hui Chen
- Department of Pharmacology and Pharmacy, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, SAR, China
| | - Paul M Vanhoutte
- Department of Pharmacology and Pharmacy, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, SAR, China
| | - Susan W S Leung
- Department of Pharmacology and Pharmacy, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, SAR, China
| |
Collapse
|
10
|
Balcilar C, Ozakca-Gunduz I, Altan VM. Contributions of Rho-kinase and AMP-related kinase signaling pathways to responses mediated by endothelium-derived contracting factors in diabetic rat aorta. Can J Physiol Pharmacol 2019; 97:600-610. [DOI: 10.1139/cjpp-2018-0698] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Diabetes-induced endothelial damage leads to vascular dysfunction. The current study investigated the effects of short-term (4-week) streptozotocin (STZ)-induced diabetes on responses mediated by endothelium-derived contracting factors (EDCFs) as well as possible contributions of Rho-kinase and AMP-activated kinase (AMPK) signaling pathways. The effects of STZ-diabetes on vascular function were examined in isolated thoracic aorta preparations of 30-week-old rats (n = 27). The diabetes-associated changes in vascular function were studied with calcium ionophore A23187, acetylcholine, Rho-kinase inhibitor Y27632 ((R)-(+)-trans-4-(1-aminoethyl)-N-(4-pyridyl)cyclohexanecarboxamide dihydrochloride), and AMPK activator AICAR (5-aminoimidazole-4-carboxamide-1-β-d-ribofuranoside). The phosphorylation of acetyl-CoA carboxylase, AMPK, and phospholamban and the protein levels of sarcoplasmic/endoplasmic Ca2+-ATPase 2 (SERCA2) and Rho-associated protein kinase (ROCKII) were measured in aortic preparations. Although the acetylcholine-mediated relaxation responses were preserved in 4-week STZ-induced diabetes, the increased activation of the Rho-kinase pathway was demonstrated via twofold enhancement in A23187-mediated contractile responses and significantly augmented protein levels of ROCKII. The AICAR-activated AMPK-mediated relaxation response was also augmented ∼4-fold in diabetic rats, without any alteration in phospholamban phosphorylation; further, this relaxation response suppressed A23187-mediated contraction in both groups. Diabetic rats showed an increase in AICAR-induced AMPK-mediated vasorelaxation and a 2.5-fold elevation of phosphorylated AMPK levels. These results indicate a possible compensation between hyperglycemia-induced endothelium-dependent hypercontractility and AMPK-mediated vasorelaxation in diabetes.
Collapse
Affiliation(s)
- Cennet Balcilar
- Department of Pharmacology, Faculty of Pharmacy, Ankara University, Ankara, Turkey
| | - Isil Ozakca-Gunduz
- Department of Pharmacology, Faculty of Pharmacy, Ankara University, Ankara, Turkey
| | - V. Melih Altan
- Department of Pharmacology, Faculty of Pharmacy, Bezmialem Vakif University, Istanbul, Turkey
| |
Collapse
|
11
|
Chen C, Kassan A, Castañeda D, Gabani M, Choi SK, Kassan M. Metformin prevents vascular damage in hypertension through the AMPK/ER stress pathway. Hypertens Res 2019; 42:960-969. [PMID: 30664704 DOI: 10.1038/s41440-019-0212-z] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2018] [Revised: 11/23/2018] [Accepted: 12/20/2018] [Indexed: 11/09/2022]
Abstract
Metformin is an antidiabetic drug. However, the pleiotropic beneficial effects of metformin in nondiabetic models still need to be defined. The objective of this study is to investigate the effect of metformin on angiotensin II (Ang II)-induced hypertension and cardiovascular diseases. Mice were infused with Ang II (400 ng/kg per min) with or without metformin for 2 weeks. Mice infused with angiotensin II displayed an increase in blood pressure associated with enhanced vascular endoplasmic reticulum (ER) stress markers, which were blunted after metformin treatment. Moreover, hypertension-induced reduction in phosphorylated AMPK, endothelial nitric oxide synthase (eNOs) phosphorylation, and endothelium-dependent relaxation (EDR) in mesenteric resistance arteries (MRA) were rescued after metformin treatment. Infusion of ER stress inducer (tunicamycin, Tun) in control mice induced ER stress in MRA and reduced phosphorylation of AMPK, eNOS synthase phosphorylation, and EDR in MRA without affecting systolic blood pressure (SBP). All these factors were reversed subsequently with metformin treatment. ER stress inhibition by metformin improves vascular function in hypertension. Therefore, metformin could be a potential therapy for cardiovascular diseases in hypertension independent of its effects on diabetes.
Collapse
Affiliation(s)
- Cheng Chen
- Department of Emergency and Critical Care, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Adam Kassan
- West Coast University, School of Pharmacy, Los Angeles, CA, 90004, USA
| | - Diana Castañeda
- California State University, Los Angeles, Los Angeles, CA, 90032, USA
| | - Mohanad Gabani
- Cardiovascular Division, Department of Medicine, and Abboud Cardiovascular Research Center, University of Iowa Carver College of Medicine, IA City, IA, 52242, USA
| | - Soo-Kyoung Choi
- Department of Physiology, College of Medicine, Brain Korea 21 PLUS Project for Medical Science, Yonsei University, Seoul, 03722, Korea
| | - Modar Kassan
- Cardiovascular Division, Department of Medicine, and Abboud Cardiovascular Research Center, University of Iowa Carver College of Medicine, IA City, IA, 52242, USA.
| |
Collapse
|
12
|
Tain YL, Hsu CN. AMP-Activated Protein Kinase as a Reprogramming Strategy for Hypertension and Kidney Disease of Developmental Origin. Int J Mol Sci 2018; 19:ijms19061744. [PMID: 29895790 PMCID: PMC6032132 DOI: 10.3390/ijms19061744] [Citation(s) in RCA: 57] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2018] [Revised: 06/10/2018] [Accepted: 06/10/2018] [Indexed: 12/18/2022] Open
Abstract
Suboptimal early-life conditions affect the developing kidney, resulting in long-term programming effects, namely renal programming. Adverse renal programming increases the risk for developing hypertension and kidney disease in adulthood. Conversely, reprogramming is a strategy aimed at reversing the programming processes in early life. AMP-activated protein kinase (AMPK) plays a key role in normal renal physiology and the pathogenesis of hypertension and kidney disease. This review discusses the regulation of AMPK in the kidney and provides hypothetical mechanisms linking AMPK to renal programming. This will be followed by studies targeting AMPK activators like metformin, resveratrol, thiazolidinediones, and polyphenols as reprogramming strategies to prevent hypertension and kidney disease. Further studies that broaden our understanding of AMPK isoform- and tissue-specific effects on renal programming are needed to ultimately develop reprogramming strategies. Despite the fact that animal models have provided interesting results with regard to reprogramming strategies targeting AMPK signaling to protect against hypertension and kidney disease with developmental origins, these results await further clinical translation.
Collapse
Affiliation(s)
- You-Lin Tain
- Departments of Pediatrics, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung 833, Taiwan.
- Institute for Translational Research in Biomedicine, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung 833, Taiwan.
| | - Chien-Ning Hsu
- Department of Pharmacy, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung 833, Taiwan.
| |
Collapse
|
13
|
Moore LG. Human Genetic Adaptation to High Altitudes: Current Status and Future Prospects. QUATERNARY INTERNATIONAL : THE JOURNAL OF THE INTERNATIONAL UNION FOR QUATERNARY RESEARCH 2017; 461:4-13. [PMID: 29375239 PMCID: PMC5784843 DOI: 10.1016/j.quaint.2016.09.045] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
Abstract
The question of whether human populations have adapted genetically to high altitude has been of interest since studies began there in the early 1900s. Initially there was debate as to whether genetic adaptation to high altitude has taken place based, in part, on disciplinary orientation and the sources of evidence being considered. Studies centered on short-term responses, termed acclimatization, and the developmental changes occurring across lifetimes. A paradigm shift occurred with the advent of single nucleotide polymorphism (SNP) technologies and statistical methods for detecting evidence of natural selection, resulting in an exponential rise in the number of publications reporting genetic adaptation. Reviewed here are the various kinds of evidence by which adaptation to high altitude has been assessed and which have led to widespread acceptance of the idea that genetic adaptation to high altitude has occurred. While methodological and other challenges remain for determining the specific gene or genes involved and the physiological mechanisms by which they are exerting their effects, considerable progress has been realized as shown by recent studies in Tibetans, Andeans and Ethiopians. Further advances are anticipated with the advent of new statistical methods, whole-genome sequencing and other molecular techniques for finer-scale genetic mapping, and greater intradisciplinary and interdisciplinary collaboration to identify the functional consequences of the genes or gene regions implicated and the time scales involved.
Collapse
Affiliation(s)
- Lorna G Moore
- Department of Obstetrics & Gynecology, University of Colorado Denver, Aurora CO (formerly of the Department of Anthropology, University of Colorado Denver, Denver CO)
| |
Collapse
|
14
|
Huang Y, Smith CA, Chen G, Sharma B, Miner AS, Barbee RW, Ratz PH. The AMP-Dependent Protein Kinase (AMPK) Activator A-769662 Causes Arterial Relaxation by Reducing Cytosolic Free Calcium Independently of an Increase in AMPK Phosphorylation. Front Pharmacol 2017; 8:756. [PMID: 29093683 PMCID: PMC5651270 DOI: 10.3389/fphar.2017.00756] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2017] [Accepted: 10/05/2017] [Indexed: 12/15/2022] Open
Abstract
Although recent studies reveal that activation of the metabolic and Ca2+ sensor AMPK strongly inhibits smooth muscle contraction, there is a paucity of information about the potential linkage between pharmacological AMPK activation and vascular smooth muscle (VSM) contraction regulation. Our aim was to test the general hypothesis that the allosteric AMPK activator A-769662 causes VSM relaxation via inhibition of contractile protein activation, and to specifically determine which activation mechanism(s) is(are) affected. The ability of A-769662 to cause endothelium-independent relaxation of contractions induced by several contractile stimuli was examined in large and small musculocutaneous and visceral rabbit arteries. For comparison, the structurally dissimilar AMPK activators MET, SIM, and BBR were assessed. A-769662 displayed artery- and agonist-dependent differential inhibitory activities that depended on artery size and location. A-769662 did not increase AMPK-pT172 levels, but did increase phosphorylation of the downstream AMPK substrate, acetyl-CoA carboxylase (ACC). A-769662 did not inhibit basal phosphorylation levels of several contractile protein regulatory proteins, and did not alter the activation state of rhoA. A-769662 did not inhibit Ca2+- and GTPγS-induced contractions in β-escin-permeabilized muscle, suggesting that A-769662 must act by inhibiting Ca2+ signaling. In intact artery, A-769662 immediately reduced basal intracellular free calcium ([Ca2+]i), inhibited a stimulus-induced increase in [Ca2+]i, and inhibited a cyclopiazonic acid (CPA)-induced contraction. MET increased AMPK-pT172, and caused neither inhibition of contraction nor inhibition of [Ca2+]i. Together, these data support the hypothesis that the differential inhibition of stimulus-induced arterial contractions by A-769662 was due to selective inhibition of a Ca2+ mobilization pathway, possibly involving CPA-dependent Ca2+ entry via an AMPK-independent pathway. That MET activated AMPK without causing arterial relaxation suggests that AMPK activation does not necessarily cause VSM relaxation.
Collapse
Affiliation(s)
- Yi Huang
- Department of Emergency Medicine and Physiology, Virginia Commonwealth University, Richmond, VA, United States
| | - Corey A Smith
- Department of Biochemistry and Molecular Biology, School of Medicine, Virginia Commonwealth University, Richmond, VA, United States
| | - Grace Chen
- Department of Biochemistry and Molecular Biology, School of Medicine, Virginia Commonwealth University, Richmond, VA, United States
| | - Bharti Sharma
- Department of Biochemistry and Molecular Biology, School of Medicine, Virginia Commonwealth University, Richmond, VA, United States
| | - Amy S Miner
- Department of Biochemistry and Molecular Biology, School of Medicine, Virginia Commonwealth University, Richmond, VA, United States
| | - Robert W Barbee
- Department of Emergency Medicine and Physiology, Virginia Commonwealth University, Richmond, VA, United States
| | - Paul H Ratz
- Department of Biochemistry and Molecular Biology, School of Medicine, Virginia Commonwealth University, Richmond, VA, United States
| |
Collapse
|
15
|
Balcilar C, Özakça I, Altan VM. Contribution of Rho-kinase and Adenosine Monophosphate-Activated Protein Kinase Signaling Pathways to Endothelium-Derived Contracting Factors Responses. Turk J Pharm Sci 2017; 14:207-212. [PMID: 32454615 DOI: 10.4274/tjps.26349] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2016] [Accepted: 02/10/2017] [Indexed: 12/01/2022]
Abstract
Vascular tonus is controlled by endothelium-derived relaxing factor (EDRF), endothelium-derived hyperpolarizing factor (EDHF) and endothelium-derived contracting factor (EDCF) under physiological circumstances. In pathological conditions, impairment of endothelium-derived relaxation can be caused by both decrease in EDRF release and increase in EDCF release. The increase in EDCF is observed with diseases such as hypertension and diabetes. The contribution of Rho-kinase and activated protein kinase (AMPK), which have opposite effects, to the increased EDCF responses was investigated. Rho-kinases are the effectors of Rho which is one of the small guanosine triphosphate-binding proteins. They increase cytosolic Ca+2 concentration and cause vascular smooth muscle to contract, keeping myosin light chain (MLC) in phosphorylated state by affecting myosin phosphatase target subunit which dephosphorylates the MLC. The activities of Rho-kinases increase with the increase of EDCF function. AMPK is the energy sensor of the cell. It provides a vasculoprotective effect by causing endothelium-dependent and endothelium-independent relaxation in smooth muscle. In contrast to Rho-kinase pathway activity, AMPK pathway activity decreases with diseases in which the EDCF function increases. In cases such as diabetes and hypertension that endothelial function impairs toward vasocontraction, it is considered that evaluating Rho-kinase and AMPK pathways which mediate contraction and relaxation in vascular smooth muscle respectively, would provide clues on choosing therapeutic target for pathologies in which endothelial dysfunction is observed.
Collapse
Affiliation(s)
- Cennet Balcilar
- Ankara University, Faculty Of Pharmacy, Department Of Pharmacology, Ankara, Turkey
| | - Işıl Özakça
- Ankara University, Faculty Of Pharmacy, Department Of Pharmacology, Ankara, Turkey
| | - Vecdi Melih Altan
- Ankara University, Faculty Of Pharmacy, Department Of Pharmacology, Ankara, Turkey
| |
Collapse
|
16
|
Schubert KM, Qiu J, Blodow S, Wiedenmann M, Lubomirov LT, Pfitzer G, Pohl U, Schneider H. The AMP-Related Kinase (AMPK) Induces Ca
2+
-Independent Dilation of Resistance Arteries by Interfering With Actin Filament Formation. Circ Res 2017; 121:149-161. [DOI: 10.1161/circresaha.116.309962] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/12/2016] [Revised: 05/23/2017] [Accepted: 06/06/2017] [Indexed: 12/13/2022]
Abstract
Rationale:
Decreasing Ca
2+
sensitivity of vascular smooth muscle (VSM) allows for vasodilation without lowering of cytosolic Ca
2+
. This may be particularly important in states requiring maintained dilation, such as hypoxia. AMP-related kinase (AMPK) is an important cellular energy sensor in VSM. Regulation of Ca
2+
sensitivity usually is attributed to myosin light chain phosphatase activity, but findings in non-VSM identified changes in the actin cytoskeleton. The potential role of AMPK in this setting is widely unknown.
Objective:
To assess the influence of AMPK on the actin cytoskeleton in VSM of resistance arteries with regard to potential Ca
2+
desensitization of VSM contractile apparatus.
Methods and Results:
AMPK induced a slowly developing dilation at unchanged cytosolic Ca
2+
levels in potassium chloride–constricted intact arteries isolated from mouse mesenteric tissue. This dilation was not associated with changes in phosphorylation of myosin light chain or of myosin light chain phosphatase regulatory subunit. Using ultracentrifugation and confocal microscopy, we found that AMPK induced depolymerization of F-actin (filamentous actin). Imaging of arteries from LifeAct mice showed F-actin rarefaction in the midcellular portion of VSM. Immunoblotting revealed that this was associated with activation of the actin severing factor cofilin. Coimmunoprecipitation experiments indicated that AMPK leads to the liberation of cofilin from 14-3-3 protein.
Conclusions:
AMPK induces actin depolymerization, which reduces vascular tone and the response to vasoconstrictors. Our findings demonstrate a new role of AMPK in the control of actin cytoskeletal dynamics, potentially allowing for long-term dilation of microvessels without substantial changes in cytosolic Ca
2+
.
Collapse
Affiliation(s)
- Kai Michael Schubert
- From the Walter Brendel Centre of Experimental Medicine, Biomedical Center of LMU, Ludwig Maximilian University of Munich, Germany (K.M.S., J.Q., S.B., M.W., U.P., H.S.); Munich Cluster for Systems Neurology (SyNergy), Germany (K.M.S., S.B., U.P., H.S.); Deutsches Zentrum für Herz- Kreislauf-Forschung (German Center for Cardiovascular Research), Partner Site Munich Heart Alliance, Munich, Germany (K.M.S., S.B., U.P., H.S.); and Institute of Vegetative Physiology, University of Cologne, Germany (L.T
| | - Jiehua Qiu
- From the Walter Brendel Centre of Experimental Medicine, Biomedical Center of LMU, Ludwig Maximilian University of Munich, Germany (K.M.S., J.Q., S.B., M.W., U.P., H.S.); Munich Cluster for Systems Neurology (SyNergy), Germany (K.M.S., S.B., U.P., H.S.); Deutsches Zentrum für Herz- Kreislauf-Forschung (German Center for Cardiovascular Research), Partner Site Munich Heart Alliance, Munich, Germany (K.M.S., S.B., U.P., H.S.); and Institute of Vegetative Physiology, University of Cologne, Germany (L.T
| | - Stephanie Blodow
- From the Walter Brendel Centre of Experimental Medicine, Biomedical Center of LMU, Ludwig Maximilian University of Munich, Germany (K.M.S., J.Q., S.B., M.W., U.P., H.S.); Munich Cluster for Systems Neurology (SyNergy), Germany (K.M.S., S.B., U.P., H.S.); Deutsches Zentrum für Herz- Kreislauf-Forschung (German Center for Cardiovascular Research), Partner Site Munich Heart Alliance, Munich, Germany (K.M.S., S.B., U.P., H.S.); and Institute of Vegetative Physiology, University of Cologne, Germany (L.T
| | - Margarethe Wiedenmann
- From the Walter Brendel Centre of Experimental Medicine, Biomedical Center of LMU, Ludwig Maximilian University of Munich, Germany (K.M.S., J.Q., S.B., M.W., U.P., H.S.); Munich Cluster for Systems Neurology (SyNergy), Germany (K.M.S., S.B., U.P., H.S.); Deutsches Zentrum für Herz- Kreislauf-Forschung (German Center for Cardiovascular Research), Partner Site Munich Heart Alliance, Munich, Germany (K.M.S., S.B., U.P., H.S.); and Institute of Vegetative Physiology, University of Cologne, Germany (L.T
| | - Lubomir T. Lubomirov
- From the Walter Brendel Centre of Experimental Medicine, Biomedical Center of LMU, Ludwig Maximilian University of Munich, Germany (K.M.S., J.Q., S.B., M.W., U.P., H.S.); Munich Cluster for Systems Neurology (SyNergy), Germany (K.M.S., S.B., U.P., H.S.); Deutsches Zentrum für Herz- Kreislauf-Forschung (German Center for Cardiovascular Research), Partner Site Munich Heart Alliance, Munich, Germany (K.M.S., S.B., U.P., H.S.); and Institute of Vegetative Physiology, University of Cologne, Germany (L.T
| | - Gabriele Pfitzer
- From the Walter Brendel Centre of Experimental Medicine, Biomedical Center of LMU, Ludwig Maximilian University of Munich, Germany (K.M.S., J.Q., S.B., M.W., U.P., H.S.); Munich Cluster for Systems Neurology (SyNergy), Germany (K.M.S., S.B., U.P., H.S.); Deutsches Zentrum für Herz- Kreislauf-Forschung (German Center for Cardiovascular Research), Partner Site Munich Heart Alliance, Munich, Germany (K.M.S., S.B., U.P., H.S.); and Institute of Vegetative Physiology, University of Cologne, Germany (L.T
| | - Ulrich Pohl
- From the Walter Brendel Centre of Experimental Medicine, Biomedical Center of LMU, Ludwig Maximilian University of Munich, Germany (K.M.S., J.Q., S.B., M.W., U.P., H.S.); Munich Cluster for Systems Neurology (SyNergy), Germany (K.M.S., S.B., U.P., H.S.); Deutsches Zentrum für Herz- Kreislauf-Forschung (German Center for Cardiovascular Research), Partner Site Munich Heart Alliance, Munich, Germany (K.M.S., S.B., U.P., H.S.); and Institute of Vegetative Physiology, University of Cologne, Germany (L.T
| | - Holger Schneider
- From the Walter Brendel Centre of Experimental Medicine, Biomedical Center of LMU, Ludwig Maximilian University of Munich, Germany (K.M.S., J.Q., S.B., M.W., U.P., H.S.); Munich Cluster for Systems Neurology (SyNergy), Germany (K.M.S., S.B., U.P., H.S.); Deutsches Zentrum für Herz- Kreislauf-Forschung (German Center for Cardiovascular Research), Partner Site Munich Heart Alliance, Munich, Germany (K.M.S., S.B., U.P., H.S.); and Institute of Vegetative Physiology, University of Cologne, Germany (L.T
| |
Collapse
|
17
|
Abstract
The AMP-activated protein kinase (AMPK) is a key regulator of cellular and whole-body energy homeostasis, which acts to restore energy homoeostasis whenever cellular energy charge is depleted. Over the last 2 decades, it has become apparent that AMPK regulates several other cellular functions and has specific roles in cardiovascular tissues, acting to regulate cardiac metabolism and contractile function, as well as promoting anticontractile, anti-inflammatory, and antiatherogenic actions in blood vessels. In this review, we discuss the role of AMPK in the cardiovascular system, including the molecular basis of mutations in AMPK that alter cardiac physiology and the proposed mechanisms by which AMPK regulates vascular function under physiological and pathophysiological conditions.
Collapse
Affiliation(s)
- Ian P Salt
- From the Institute of Cardiovascular & Medical Sciences, College of Medical, Veterinary & Life Sciences, University of Glasgow, Scotland, United Kingdom (I.P.S.); and Division of Cell Signalling & Immunology, School of Life Sciences, University of Dundee, Scotland, United Kingdom (D.G.H.).
| | - D Grahame Hardie
- From the Institute of Cardiovascular & Medical Sciences, College of Medical, Veterinary & Life Sciences, University of Glasgow, Scotland, United Kingdom (I.P.S.); and Division of Cell Signalling & Immunology, School of Life Sciences, University of Dundee, Scotland, United Kingdom (D.G.H.)
| |
Collapse
|
18
|
Mori A, Ishikawa E, Amano T, Sakamoto K, Nakahara T. Anti-diabetic drug metformin dilates retinal blood vessels through activation of AMP-activated protein kinase in rats. Eur J Pharmacol 2017; 798:66-71. [PMID: 28087254 DOI: 10.1016/j.ejphar.2017.01.003] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2016] [Revised: 12/20/2016] [Accepted: 01/09/2017] [Indexed: 12/29/2022]
Abstract
The aim of this study was to examine whether metformin, a biguanide anti-hyperglycemic drug, dilates retinal blood vessels in rats. Ocular fundus images were captured with an original high-resolution digital fundus camera in vivo and diameters of retinal blood vessels were measured. Both systemic blood pressure and heart rate were continuously recorded. Metformin (0.01-0.3mg/kg/min) increased diameters of retinal blood vessels in a dose-dependent manner. This retinal vasodilator effect of metformin was abolished by compound C, an inhibitor of AMP-activated protein kinase (AMPK), and NG-nitro-L-arginine methyl ester, an inhibitor of nitric oxide (NO) synthase. Similar results were obtained with the AMPK activator 5-aminoimidazole-4-carboxamide-1-β-D-ribonucleoside (AICAR, 0.01-1mg/kg/min). Neither metformin nor AICAR exerted significant effect on mean blood pressure and heart rate. However, a significant pressor response to AICAR was observed upon inhibition of NO synthase. These results suggest that metformin dilates retinal blood vessels through activation of AMPK, and NO plays an important role in the retinal vasodilator response following AMPK activation.
Collapse
Affiliation(s)
- Asami Mori
- Department of Molecular Pharmacology, Kitasato University School of Pharmaceutical Sciences, 5-9-1 Shirokane, Minato-ku, Tokyo 108-8641, Japan
| | - Eriko Ishikawa
- Department of Molecular Pharmacology, Kitasato University School of Pharmaceutical Sciences, 5-9-1 Shirokane, Minato-ku, Tokyo 108-8641, Japan
| | - Tomoyo Amano
- Department of Molecular Pharmacology, Kitasato University School of Pharmaceutical Sciences, 5-9-1 Shirokane, Minato-ku, Tokyo 108-8641, Japan
| | - Kenji Sakamoto
- Department of Molecular Pharmacology, Kitasato University School of Pharmaceutical Sciences, 5-9-1 Shirokane, Minato-ku, Tokyo 108-8641, Japan
| | - Tsutomu Nakahara
- Department of Molecular Pharmacology, Kitasato University School of Pharmaceutical Sciences, 5-9-1 Shirokane, Minato-ku, Tokyo 108-8641, Japan.
| |
Collapse
|
19
|
Li X, Dai Y, Yan S, Shi Y, Li J, Liu J, Cha L, Mu J. Resveratrol lowers blood pressure in spontaneously hypertensive rats via calcium-dependent endothelial NO production. Clin Exp Hypertens 2016; 38:287-93. [PMID: 27018796 DOI: 10.3109/10641963.2015.1089882] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
OBJECTIVE Resveratrol, a polyphenol of natural compounds, has beneficial cardiovascular effects, many of which are mediated by nitric oxide (NO). Resveratrol increases intracellular calcium and activates AMP-activated protein kinase (AMPK), all of which could increase NO production. We hypothesized that resveratrol via a calcium-dependent NO production lowers blood pressure (BP) in spontaneously hypertensive rats (SHR). METHODS Acetylcholine (Ach)-induced endothelium-dependent relaxations in rat aortas were examined by organ chamber. Blood pressures were determined by radiotelemetry methods. RESULTS Incubation of isolated aortas from SHR with resveratrol dramatically improved vasorelaxation induced by Ach. Preincubation of aortas with endothelial NO synthase (eNOS) inhibitor or calcium chelant blunted the effects of resveratrol on Ach-induced relaxation, as wells as NO production and eNOS phosphorylation. In animal studies, administration of resveratrol significantly lowered systemic BP in SHR. CONCLUSION Resveratrol increases endothelial NO production to improve endothelial dysfunction and lowers BP in hypertensive rats, which depends on calcium-eNOS activation.
Collapse
Affiliation(s)
- Xin Li
- a Department of Cardiology , The First Affiliated Hospital of Medical College, Xi'an Jiao Tong University , Xi'an , China.,b Department of Cardiology , The Fourth Affiliated Hospital of Harbin Medical University , Harbin , China
| | - Yingnan Dai
- b Department of Cardiology , The Fourth Affiliated Hospital of Harbin Medical University , Harbin , China
| | - Shujun Yan
- b Department of Cardiology , The Fourth Affiliated Hospital of Harbin Medical University , Harbin , China
| | - Yanli Shi
- b Department of Cardiology , The Fourth Affiliated Hospital of Harbin Medical University , Harbin , China
| | - Jingxiu Li
- b Department of Cardiology , The Fourth Affiliated Hospital of Harbin Medical University , Harbin , China
| | - Jinglu Liu
- b Department of Cardiology , The Fourth Affiliated Hospital of Harbin Medical University , Harbin , China
| | - Li Cha
- b Department of Cardiology , The Fourth Affiliated Hospital of Harbin Medical University , Harbin , China
| | - Jianjun Mu
- a Department of Cardiology , The First Affiliated Hospital of Medical College, Xi'an Jiao Tong University , Xi'an , China
| |
Collapse
|
20
|
Denniss SG, Ford RJ, Smith CS, Jeffery AJ, Rush JWE. Chronic in vivo or acute in vitro resveratrol attenuates endothelium-dependent cyclooxygenase-mediated contractile signaling in hypertensive rat carotid artery. J Appl Physiol (1985) 2016; 120:1141-50. [PMID: 26917696 DOI: 10.1152/japplphysiol.00675.2015] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2015] [Accepted: 02/22/2016] [Indexed: 01/13/2023] Open
Abstract
Exaggerated cyclooxygenase (COX) and thromboxane-prostanoid (TP) receptor-mediated endothelium-dependent contraction can contribute to endothelial dysfunction. This study examined the effect of resveratrol (RSV) on endothelium-dependent contraction and cell signaling in the common carotid artery (CCA) from spontaneously hypertensive rats (SHR) and Wistar Kyoto rats (WKY). Acetylcholine (Ach)-stimulated endothelium-dependent nitric oxide synthase (NOS)-mediated relaxation in precontracted SHR CCA was impaired (maximum 73 ± 6% vs. 87 ± 5% in WKY) (P < 0.05) by competitive COX-mediated contraction. Chronic (28-day) treatment in vivo (drinking water) with a ∼0.075 mg·kg(-1)·day(-1) RSV dose affected neither endothelium-dependent relaxation nor endothelium-dependent contraction and associated prostaglandin (PG) production evaluated in non-precontracted NOS-blocked CCA. In contrast, a chronic ∼7.5 mg·kg(-1)·day(-1) RSV dose improved endothelium-dependent relaxation (94 ± 6%) and attenuated endothelium-dependent contraction (58 ± 4% vs. 73 ± 5% in No-RSV) and PG production (183 ± 43 vs. 519 ± 93 pg/ml) in SHR CCA, while U46619-stimulated TP receptor-mediated contraction was unaffected. In separate acute in vitro experiments, 20-μM RSV preincubation attenuated endothelium-dependent contraction (6 ± 4% vs. 62 ± 2% in No Drug) and PG production (121 ± 15 vs. 491 ± 93 pg/ml) and attenuated U46619-stimulated contraction (134 ± 5% vs. 171 ± 4%) in non-precontracted NOS-blocked SHR CCA. Compound C, a known AMP-activated protein kinase (AMPK) inhibitor, did not prevent the RSV attenuating effect on Ach- and U46619-stimulated contraction but did prevent the RSV attenuating effect on PG production (414 ± 58 pg/ml). These data demonstrate that RSV can attenuate endothelium-dependent contraction both by suppressing arterial wall PG production, which may be partially mediated by AMPK, and by TP receptor hyporesponsiveness, which does not appear to be mediated by AMPK.
Collapse
Affiliation(s)
- Steven G Denniss
- Integrative Vascular Biology Laboratory, Department of Kinesiology, Faculty of Applied Health Sciences, University of Waterloo, Waterloo, Ontario, Canada
| | - Rebecca J Ford
- Integrative Vascular Biology Laboratory, Department of Kinesiology, Faculty of Applied Health Sciences, University of Waterloo, Waterloo, Ontario, Canada
| | - Christopher S Smith
- Integrative Vascular Biology Laboratory, Department of Kinesiology, Faculty of Applied Health Sciences, University of Waterloo, Waterloo, Ontario, Canada
| | - Andrew J Jeffery
- Integrative Vascular Biology Laboratory, Department of Kinesiology, Faculty of Applied Health Sciences, University of Waterloo, Waterloo, Ontario, Canada
| | - James W E Rush
- Integrative Vascular Biology Laboratory, Department of Kinesiology, Faculty of Applied Health Sciences, University of Waterloo, Waterloo, Ontario, Canada
| |
Collapse
|
21
|
Zhao Y, Zhang X, Li J, Bian Y, Sheng M, Liu B, Fu Z, Zhang Y, Yang B. Jujuboside B Reduces Vascular Tension by Increasing Ca2+ Influx and Activating Endothelial Nitric Oxide Synthase. PLoS One 2016; 11:e0149386. [PMID: 26901291 PMCID: PMC4762982 DOI: 10.1371/journal.pone.0149386] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2015] [Accepted: 02/01/2016] [Indexed: 11/24/2022] Open
Abstract
Jujuboside B has been reported to have protective effect on many cardiovascular diseases. However, the effects of Jujuboside B on vascular tension and endothelial function are unknown. The present study investigated the effects of Jujuboside B on reducing vascular tension, protecting endothelial function and the potential mechanisms. The tension of isolated rat thoracic aorta ring was measured by Wire myograph system. The concentration of nitric oxide (NO) and the activity of endothelial nitric oxide synthase (eNOS) in human aortic endothelial cells (HAECs) were determined by Griess reagent method and enzyme-linked immune sorbent assay. The protein levels of eNOS and p-eNOS at Serine-1177 were determined by western blot analysis. Intracellular Ca2+ concentration in HAECs was measured by laser confocal imaging microscopy. Results showed that Jujuboside B reduced the tension of rat thoracic aorta rings with intact endothelium in a dose-dependent manner. L-NAME, KN93, EGTA, SKF96365, iberiotoxin and glibenclamide significantly attenuated Jujuboside B-induced vasodilation in endothelium-intact tissues. In contrast, indometacin and 4-DAMP had no such effects. Jujuboside B also promoted NO generation and increased eNOS activity, which were attenuated by L-NAME, EGTA and SKF96365. Moreover, Jujuboside B increased intracellular Ca2+ concentration dose-dependently, which was inhibited by EGTA and SKF96365. Besides, Jujuboside B induced a rapid Ca2+ influx instantaneously after depleting intracellular Ca2+ store, which was significantly inhibited by SKF96365. In conclusion, this study preliminarily confirmed that Jujuboside B reduced vascular tension endothelium-dependently. The underlying mechanisms involved that Jujuboside B increased extracellular Ca2+ influx through endothelial transient receptor potential cation (TRPC) channels, phosphorylated eNOS and promoted NO generation in vascular endothelial cells. In addition, Jujuboside B-induced vasodilation involved endothelium-dependent hyperpolarizaiton through endothelial potassium channels. Jujuboside B is a natural compound with new pharmacological effects on improving endothelial dysfunction and treating vascular diseases.
Collapse
Affiliation(s)
- Yixiu Zhao
- Department of Pharmacology, Harbin Medical University, Harbin, Heilongjiang, PR China
- Key Laboratory of Cardiovascular Medicine Research (Harbin Medical University), Ministry of Education, Harbin, Heilongjiang, PR China
| | - Xin Zhang
- Department of Pharmacology, Harbin Medical University, Harbin, Heilongjiang, PR China
- Key Laboratory of Cardiovascular Medicine Research (Harbin Medical University), Ministry of Education, Harbin, Heilongjiang, PR China
| | - Jiannan Li
- Department of Pharmacology, Harbin Medical University, Harbin, Heilongjiang, PR China
- Key Laboratory of Cardiovascular Medicine Research (Harbin Medical University), Ministry of Education, Harbin, Heilongjiang, PR China
| | - Yu Bian
- Department of Pharmacology, Harbin Medical University, Harbin, Heilongjiang, PR China
- Key Laboratory of Cardiovascular Medicine Research (Harbin Medical University), Ministry of Education, Harbin, Heilongjiang, PR China
| | - Miaomiao Sheng
- Department of Pharmacology, Harbin Medical University, Harbin, Heilongjiang, PR China
- Key Laboratory of Cardiovascular Medicine Research (Harbin Medical University), Ministry of Education, Harbin, Heilongjiang, PR China
| | - Bin Liu
- Department of Pharmacology, Harbin Medical University, Harbin, Heilongjiang, PR China
- Key Laboratory of Cardiovascular Medicine Research (Harbin Medical University), Ministry of Education, Harbin, Heilongjiang, PR China
| | - Zidong Fu
- Department of Pharmacology, Harbin Medical University, Harbin, Heilongjiang, PR China
- Key Laboratory of Cardiovascular Medicine Research (Harbin Medical University), Ministry of Education, Harbin, Heilongjiang, PR China
| | - Yan Zhang
- Department of Pharmacology, Harbin Medical University, Harbin, Heilongjiang, PR China
- Key Laboratory of Cardiovascular Medicine Research (Harbin Medical University), Ministry of Education, Harbin, Heilongjiang, PR China
- * E-mail: (YZ); (BFY)
| | - Baofeng Yang
- Department of Pharmacology, Harbin Medical University, Harbin, Heilongjiang, PR China
- Key Laboratory of Cardiovascular Medicine Research (Harbin Medical University), Ministry of Education, Harbin, Heilongjiang, PR China
- * E-mail: (YZ); (BFY)
| |
Collapse
|
22
|
Skeffington KL, Higgins JS, Mahmoud AD, Evans AM, Sferruzzi-Perri AN, Fowden AL, Yung HW, Burton GJ, Giussani DA, Moore LG. Hypoxia, AMPK activation and uterine artery vasoreactivity. J Physiol 2015; 594:1357-69. [PMID: 26110512 DOI: 10.1113/jp270995] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2015] [Accepted: 06/21/2015] [Indexed: 01/12/2023] Open
Abstract
Genes near adenosine monophosphate-activated protein kinase-α1 (PRKAA1) have been implicated in the greater uterine artery (UtA) blood flow and relative protection from fetal growth restriction seen in altitude-adapted Andean populations. Adenosine monophosphate-activated protein kinase (AMPK) activation vasodilates multiple vessels but whether AMPK is present in UtA or placental tissue and influences UtA vasoreactivity during normal or hypoxic pregnancy remains unknown. We studied isolated UtA and placenta from near-term C57BL/6J mice housed in normoxia (n = 8) or hypoxia (10% oxygen, n = 7-9) from day 14 to day 19, and placentas from non-labouring sea level (n = 3) or 3100 m (n = 3) women. Hypoxia increased AMPK immunostaining in near-term murine UtA and placental tissue. RT-PCR products for AMPK-α1 and -α2 isoforms and liver kinase B1 (LKB1; the upstream kinase activating AMPK) were present in murine and human placenta, and hypoxia increased LKB1 and AMPK-α1 and -α2 expression in the high- compared with low-altitude human placentas. Pharmacological AMPK activation by A769662 caused phenylephrine pre-constricted UtA from normoxic or hypoxic pregnant mice to dilate and this dilatation was partially reversed by the NOS inhibitor l-NAME. Hypoxic pregnancy sufficient to restrict fetal growth markedly augmented the UtA vasodilator effect of AMPK activation in opposition to PE constriction as the result of both NO-dependent and NO-independent mechanisms. We conclude that AMPK is activated during hypoxic pregnancy and that AMPK activation vasodilates the UtA, especially in hypoxic pregnancy. AMPK activation may be playing an adaptive role by limiting cellular energy depletion and helping to maintain utero-placental blood flow in hypoxic pregnancy.
Collapse
Affiliation(s)
- K L Skeffington
- Centre for Trophoblast Research, Department of Physiology Development & Neuroscience, University of Cambridge, Cambridge, UK
| | - J S Higgins
- Centre for Trophoblast Research, Department of Physiology Development & Neuroscience, University of Cambridge, Cambridge, UK
| | - A D Mahmoud
- Centre for Integrative Physiology, College of Medicine and Veterinary Medicine, University of Edinburgh, Edinburgh, UK
| | - A M Evans
- Centre for Integrative Physiology, College of Medicine and Veterinary Medicine, University of Edinburgh, Edinburgh, UK
| | - A N Sferruzzi-Perri
- Centre for Trophoblast Research, Department of Physiology Development & Neuroscience, University of Cambridge, Cambridge, UK
| | - A L Fowden
- Centre for Trophoblast Research, Department of Physiology Development & Neuroscience, University of Cambridge, Cambridge, UK
| | - H W Yung
- Centre for Trophoblast Research, Department of Physiology Development & Neuroscience, University of Cambridge, Cambridge, UK
| | - G J Burton
- Centre for Trophoblast Research, Department of Physiology Development & Neuroscience, University of Cambridge, Cambridge, UK
| | - D A Giussani
- Centre for Trophoblast Research, Department of Physiology Development & Neuroscience, University of Cambridge, Cambridge, UK
| | - L G Moore
- Division of Basic Reproductive Sciences, Department of Obstetrics & Gynaecology, University of Colorado Denver, Aurora, CO, USA
| |
Collapse
|
23
|
Pyla R, Pichavaram P, Fairaq A, Park MA, Kozak M, Kamath V, Patel VS, Segar L. Altered energy state reversibly controls smooth muscle contractile function in human saphenous vein during acute hypoxia-reoxygenation: Role of glycogen, AMP-activated protein kinase, and insulin-independent glucose uptake. Biochem Pharmacol 2015. [PMID: 26212549 DOI: 10.1016/j.bcp.2015.06.036] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Hypoxia is known to promote vasodilation of coronary vessels through several mediators including cardiac-derived adenosine and endothelium-derived prostanoids and nitric oxide. To date, the impact of endogenous glycogen depletion in vascular smooth muscle and the resultant alterations in cellular energy state (e.g., AMP-activated protein kinase, AMPK) on the contractile response to G protein-coupled receptor agonists (e.g., serotonin, 5-HT) has not yet been studied. In the present study, ex vivo exposure of endothelium-denuded human saphenous vein rings to hypoxic and glucose-deprived conditions during KCl-induced contractions for 30 min resulted in a marked depletion of endogenous glycogen by ∼80% (from ∼1.78 μmol/g under normoxia to ∼0.36 μmol/g under hypoxia). Importantly, glycogen-depleted HSV rings, which were maintained under hypoxia/reoxygenation and glucose-deprived conditions, exhibited significant increases in basal AMPK phosphorylation (∼6-fold ↑) and 5-HT-induced AMPK phosphorylation (∼19-fold ↑) with an accompanying suppression of 5-HT-induced maximal contractile response (∼68% ↓), compared with respective controls. Exposure of glycogen-depleted HSV rings to exogenous D-glucose, but not the inactive glucose analogs, prevented the exaggerated increase in 5-HT-induced AMPK phosphorylation and restored 5-HT-induced maximal contractile response. In addition, the ability of exogenous D-glucose to rescue cellular stress and impaired contractile function occurred through GLUT1-mediated but insulin/GLUT4-independent mechanisms. Together, the present findings from clinically-relevant human saphenous vein suggest that the loss of endogenous glycogen in vascular smooth muscle and the resultant accentuation of AMPK phosphorylation by GPCR agonists may constitute a yet another mechanism of metabolic vasodilation of coronary vessels in ischemic heart disease.
Collapse
Affiliation(s)
- Rajkumar Pyla
- Center for Pharmacy and Experimental Therapeutics, University of Georgia College of Pharmacy, Augusta, GA, USA; Charlie Norwood VA Medical Center, Augusta, GA, USA
| | - Prahalathan Pichavaram
- Center for Pharmacy and Experimental Therapeutics, University of Georgia College of Pharmacy, Augusta, GA, USA; Charlie Norwood VA Medical Center, Augusta, GA, USA
| | - Arwa Fairaq
- Center for Pharmacy and Experimental Therapeutics, University of Georgia College of Pharmacy, Augusta, GA, USA; Charlie Norwood VA Medical Center, Augusta, GA, USA
| | - Mary Anne Park
- Division of Clinical and Translational Sciences, Clinical Trials Office, Georgia Regents University, Augusta, GA, USA
| | - Mark Kozak
- Heart and Vascular Institute, Department of Medicine, Pennsylvania State University College of Medicine, Hershey, PA, USA
| | - Vinayak Kamath
- Heart and Cardiovascular Health Services, Department of Surgery, Georgia Regents University, Augusta, GA, USA
| | - Vijay S Patel
- Heart and Cardiovascular Health Services, Department of Surgery, Georgia Regents University, Augusta, GA, USA; Vascular Biology Center, Department of Pharmacology and Toxicology, Georgia Regents University, Augusta, GA, USA
| | - Lakshman Segar
- Center for Pharmacy and Experimental Therapeutics, University of Georgia College of Pharmacy, Augusta, GA, USA; Charlie Norwood VA Medical Center, Augusta, GA, USA; Heart and Vascular Institute, Department of Medicine, Pennsylvania State University College of Medicine, Hershey, PA, USA; Vascular Biology Center, Department of Pharmacology and Toxicology, Georgia Regents University, Augusta, GA, USA.
| |
Collapse
|
24
|
Matsumoto T, Goulopoulou S, Taguchi K, Tostes RC, Kobayashi T. Constrictor prostanoids and uridine adenosine tetraphosphate: vascular mediators and therapeutic targets in hypertension and diabetes. Br J Pharmacol 2015; 172:3980-4001. [PMID: 26031319 DOI: 10.1111/bph.13205] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2015] [Revised: 04/16/2015] [Accepted: 05/19/2015] [Indexed: 12/22/2022] Open
Abstract
Vascular dysfunction plays a pivotal role in the development of systemic complications associated with arterial hypertension and diabetes. The endothelium, or more specifically, various factors derived from endothelial cells tightly regulate vascular function, including vascular tone. In physiological conditions, there is a balance between endothelium-derived factors, that is, relaxing factors (endothelium-derived relaxing factors; EDRFs) and contracting factors (endothelium-derived contracting factors; EDCFs), which mediate vascular homeostasis. However, in disease states, such as diabetes and arterial hypertension, there is an imbalance between EDRF and EDCF, with a reduction of EDRF signalling and an increase of EDCF signalling. Among EDCFs, COX-derived vasoconstrictor prostanoids play an important role in the development of vascular dysfunction associated with hypertension and diabetes. Moreover, uridine adenosine tetraphosphate (Up4 A), identified as an EDCF in 2005, also modulates vascular function. However, the role of Up4 A in hypertension- and diabetes-associated vascular dysfunction is unclear. In the present review, we focused on experimental and clinical evidence that implicate these two EDCFs (vasoconstrictor prostanoids and Up4 A) in vascular dysfunction associated with hypertension and diabetes.
Collapse
Affiliation(s)
- Takayuki Matsumoto
- Department of Physiology and Morphology, Institute of Medicinal Chemistry, Hoshi University, Shinagawa-ku, Tokyo, Japan
| | - Styliani Goulopoulou
- Department of Integrative Physiology and Anatomy, Obstetrics and Gynecology, University of North Texas Health Science Center, Fort Worth, TX, USA
| | - Kumiko Taguchi
- Department of Physiology and Morphology, Institute of Medicinal Chemistry, Hoshi University, Shinagawa-ku, Tokyo, Japan
| | - Rita C Tostes
- Department of Pharmacology, Ribeirao Preto Medical School University of Sao Paulo, Ribeirao Preto, SP, Brazil
| | - Tsuneo Kobayashi
- Department of Physiology and Morphology, Institute of Medicinal Chemistry, Hoshi University, Shinagawa-ku, Tokyo, Japan
| |
Collapse
|
25
|
Schneider H, Schubert KM, Blodow S, Kreutz CP, Erdogmus S, Wiedenmann M, Qiu J, Fey T, Ruth P, Lubomirov LT, Pfitzer G, Mederos y Schnitzler M, Hardie DG, Gudermann T, Pohl U. AMPK Dilates Resistance Arteries via Activation of SERCA and BK
Ca
Channels in Smooth Muscle. Hypertension 2015; 66:108-16. [DOI: 10.1161/hypertensionaha.115.05514] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2015] [Accepted: 04/30/2015] [Indexed: 01/12/2023]
Affiliation(s)
- Holger Schneider
- From the Walter-Brendel Centre of Experimental Medicine and Biomedical Center (H.S., K.M.S., S.B., C.-P.K., M.W., J.Q., T.F., U.P.) and Walther Straub Institute, Pharmacology (S.E., M.M.y.S., T.G.), Ludwig-Maximilians Universität München, Munich, Germany; Munich Cluster for Systems Neurology (SyNergy), Munich, Germany (H.S., K.M.S., S.B., U.P.); DZHK (German Center for Cardiovascular Research), partner site Munich Heart Alliance, Munich, Germany (H.S., K.M.S., S.B., T.F., M.M.y.S., T.G., U.P.)
| | - Kai Michael Schubert
- From the Walter-Brendel Centre of Experimental Medicine and Biomedical Center (H.S., K.M.S., S.B., C.-P.K., M.W., J.Q., T.F., U.P.) and Walther Straub Institute, Pharmacology (S.E., M.M.y.S., T.G.), Ludwig-Maximilians Universität München, Munich, Germany; Munich Cluster for Systems Neurology (SyNergy), Munich, Germany (H.S., K.M.S., S.B., U.P.); DZHK (German Center for Cardiovascular Research), partner site Munich Heart Alliance, Munich, Germany (H.S., K.M.S., S.B., T.F., M.M.y.S., T.G., U.P.)
| | - Stephanie Blodow
- From the Walter-Brendel Centre of Experimental Medicine and Biomedical Center (H.S., K.M.S., S.B., C.-P.K., M.W., J.Q., T.F., U.P.) and Walther Straub Institute, Pharmacology (S.E., M.M.y.S., T.G.), Ludwig-Maximilians Universität München, Munich, Germany; Munich Cluster for Systems Neurology (SyNergy), Munich, Germany (H.S., K.M.S., S.B., U.P.); DZHK (German Center for Cardiovascular Research), partner site Munich Heart Alliance, Munich, Germany (H.S., K.M.S., S.B., T.F., M.M.y.S., T.G., U.P.)
| | - Claus-Peter Kreutz
- From the Walter-Brendel Centre of Experimental Medicine and Biomedical Center (H.S., K.M.S., S.B., C.-P.K., M.W., J.Q., T.F., U.P.) and Walther Straub Institute, Pharmacology (S.E., M.M.y.S., T.G.), Ludwig-Maximilians Universität München, Munich, Germany; Munich Cluster for Systems Neurology (SyNergy), Munich, Germany (H.S., K.M.S., S.B., U.P.); DZHK (German Center for Cardiovascular Research), partner site Munich Heart Alliance, Munich, Germany (H.S., K.M.S., S.B., T.F., M.M.y.S., T.G., U.P.)
| | - Serap Erdogmus
- From the Walter-Brendel Centre of Experimental Medicine and Biomedical Center (H.S., K.M.S., S.B., C.-P.K., M.W., J.Q., T.F., U.P.) and Walther Straub Institute, Pharmacology (S.E., M.M.y.S., T.G.), Ludwig-Maximilians Universität München, Munich, Germany; Munich Cluster for Systems Neurology (SyNergy), Munich, Germany (H.S., K.M.S., S.B., U.P.); DZHK (German Center for Cardiovascular Research), partner site Munich Heart Alliance, Munich, Germany (H.S., K.M.S., S.B., T.F., M.M.y.S., T.G., U.P.)
| | - Margarethe Wiedenmann
- From the Walter-Brendel Centre of Experimental Medicine and Biomedical Center (H.S., K.M.S., S.B., C.-P.K., M.W., J.Q., T.F., U.P.) and Walther Straub Institute, Pharmacology (S.E., M.M.y.S., T.G.), Ludwig-Maximilians Universität München, Munich, Germany; Munich Cluster for Systems Neurology (SyNergy), Munich, Germany (H.S., K.M.S., S.B., U.P.); DZHK (German Center for Cardiovascular Research), partner site Munich Heart Alliance, Munich, Germany (H.S., K.M.S., S.B., T.F., M.M.y.S., T.G., U.P.)
| | - Jiehua Qiu
- From the Walter-Brendel Centre of Experimental Medicine and Biomedical Center (H.S., K.M.S., S.B., C.-P.K., M.W., J.Q., T.F., U.P.) and Walther Straub Institute, Pharmacology (S.E., M.M.y.S., T.G.), Ludwig-Maximilians Universität München, Munich, Germany; Munich Cluster for Systems Neurology (SyNergy), Munich, Germany (H.S., K.M.S., S.B., U.P.); DZHK (German Center for Cardiovascular Research), partner site Munich Heart Alliance, Munich, Germany (H.S., K.M.S., S.B., T.F., M.M.y.S., T.G., U.P.)
| | - Theres Fey
- From the Walter-Brendel Centre of Experimental Medicine and Biomedical Center (H.S., K.M.S., S.B., C.-P.K., M.W., J.Q., T.F., U.P.) and Walther Straub Institute, Pharmacology (S.E., M.M.y.S., T.G.), Ludwig-Maximilians Universität München, Munich, Germany; Munich Cluster for Systems Neurology (SyNergy), Munich, Germany (H.S., K.M.S., S.B., U.P.); DZHK (German Center for Cardiovascular Research), partner site Munich Heart Alliance, Munich, Germany (H.S., K.M.S., S.B., T.F., M.M.y.S., T.G., U.P.)
| | - Peter Ruth
- From the Walter-Brendel Centre of Experimental Medicine and Biomedical Center (H.S., K.M.S., S.B., C.-P.K., M.W., J.Q., T.F., U.P.) and Walther Straub Institute, Pharmacology (S.E., M.M.y.S., T.G.), Ludwig-Maximilians Universität München, Munich, Germany; Munich Cluster for Systems Neurology (SyNergy), Munich, Germany (H.S., K.M.S., S.B., U.P.); DZHK (German Center for Cardiovascular Research), partner site Munich Heart Alliance, Munich, Germany (H.S., K.M.S., S.B., T.F., M.M.y.S., T.G., U.P.)
| | - Lubomir T. Lubomirov
- From the Walter-Brendel Centre of Experimental Medicine and Biomedical Center (H.S., K.M.S., S.B., C.-P.K., M.W., J.Q., T.F., U.P.) and Walther Straub Institute, Pharmacology (S.E., M.M.y.S., T.G.), Ludwig-Maximilians Universität München, Munich, Germany; Munich Cluster for Systems Neurology (SyNergy), Munich, Germany (H.S., K.M.S., S.B., U.P.); DZHK (German Center for Cardiovascular Research), partner site Munich Heart Alliance, Munich, Germany (H.S., K.M.S., S.B., T.F., M.M.y.S., T.G., U.P.)
| | - Gabriele Pfitzer
- From the Walter-Brendel Centre of Experimental Medicine and Biomedical Center (H.S., K.M.S., S.B., C.-P.K., M.W., J.Q., T.F., U.P.) and Walther Straub Institute, Pharmacology (S.E., M.M.y.S., T.G.), Ludwig-Maximilians Universität München, Munich, Germany; Munich Cluster for Systems Neurology (SyNergy), Munich, Germany (H.S., K.M.S., S.B., U.P.); DZHK (German Center for Cardiovascular Research), partner site Munich Heart Alliance, Munich, Germany (H.S., K.M.S., S.B., T.F., M.M.y.S., T.G., U.P.)
| | - Michael Mederos y Schnitzler
- From the Walter-Brendel Centre of Experimental Medicine and Biomedical Center (H.S., K.M.S., S.B., C.-P.K., M.W., J.Q., T.F., U.P.) and Walther Straub Institute, Pharmacology (S.E., M.M.y.S., T.G.), Ludwig-Maximilians Universität München, Munich, Germany; Munich Cluster for Systems Neurology (SyNergy), Munich, Germany (H.S., K.M.S., S.B., U.P.); DZHK (German Center for Cardiovascular Research), partner site Munich Heart Alliance, Munich, Germany (H.S., K.M.S., S.B., T.F., M.M.y.S., T.G., U.P.)
| | - D. Grahame Hardie
- From the Walter-Brendel Centre of Experimental Medicine and Biomedical Center (H.S., K.M.S., S.B., C.-P.K., M.W., J.Q., T.F., U.P.) and Walther Straub Institute, Pharmacology (S.E., M.M.y.S., T.G.), Ludwig-Maximilians Universität München, Munich, Germany; Munich Cluster for Systems Neurology (SyNergy), Munich, Germany (H.S., K.M.S., S.B., U.P.); DZHK (German Center for Cardiovascular Research), partner site Munich Heart Alliance, Munich, Germany (H.S., K.M.S., S.B., T.F., M.M.y.S., T.G., U.P.)
| | - Thomas Gudermann
- From the Walter-Brendel Centre of Experimental Medicine and Biomedical Center (H.S., K.M.S., S.B., C.-P.K., M.W., J.Q., T.F., U.P.) and Walther Straub Institute, Pharmacology (S.E., M.M.y.S., T.G.), Ludwig-Maximilians Universität München, Munich, Germany; Munich Cluster for Systems Neurology (SyNergy), Munich, Germany (H.S., K.M.S., S.B., U.P.); DZHK (German Center for Cardiovascular Research), partner site Munich Heart Alliance, Munich, Germany (H.S., K.M.S., S.B., T.F., M.M.y.S., T.G., U.P.)
| | - Ulrich Pohl
- From the Walter-Brendel Centre of Experimental Medicine and Biomedical Center (H.S., K.M.S., S.B., C.-P.K., M.W., J.Q., T.F., U.P.) and Walther Straub Institute, Pharmacology (S.E., M.M.y.S., T.G.), Ludwig-Maximilians Universität München, Munich, Germany; Munich Cluster for Systems Neurology (SyNergy), Munich, Germany (H.S., K.M.S., S.B., U.P.); DZHK (German Center for Cardiovascular Research), partner site Munich Heart Alliance, Munich, Germany (H.S., K.M.S., S.B., T.F., M.M.y.S., T.G., U.P.)
| |
Collapse
|
26
|
García-Prieto CF, Hernández-Nuño F, Rio DD, Ruiz-Hurtado G, Aránguez I, Ruiz-Gayo M, Somoza B, Fernández-Alfonso MS. High-fat diet induces endothelial dysfunction through a down-regulation of the endothelial AMPK-PI3K-Akt-eNOS pathway. Mol Nutr Food Res 2014; 59:520-32. [PMID: 25421217 DOI: 10.1002/mnfr.201400539] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2014] [Revised: 11/08/2014] [Accepted: 11/13/2014] [Indexed: 11/11/2022]
Abstract
SCOPE Activation of endothelial adenosine monophosphate-activated protein kinase (AMPK) contributes to increase nitric oxide (NO) availability. The aim of this study was to assess if high-fat diet (HFD)-induced endothelial dysfunction is linked to AMPK deregulation. METHODS AND RESULTS Twelve-week-old Sprague Dawley male rats were assigned either to control (10 kcal % from fat) or to HFD (45 kcal % from fat) for 8 wk. HFD rats segregated in obesity-prone (OP) or obesity-resistant (OR) rats according to body weight. HFD triggered an impaired glucose management together with impaired endothelium-dependent relaxation, reduced endothelial AMPK activity and lower NO availability in aortic rings of OP and OR cohorts. Relaxation evoked by AMPK activator, 5-aminoimidazole-4-carboxamide-1-β-d-ribofuranoside (AICAR) was reduced in both OP and OR rings, which exhibited lower p-AMPKα-Thr(172) /AMPKα ratios that negatively correlated with plasma non-esterified fatty acids (NEFA) and triglycerides (TG). Inhibition of PI3K (wortmannin, 10(-7) M) or Akt (triciribine, 10(-5) M) reduced relaxation to AICAR only in the control group (p < 0.001). Akt (p-Akt-Ser(473) ) and eNOS phosphorylation (p-eNOS-Ser(1177) ) were significantly reduced in OP and OR (p < 0.01). CONCLUSION Endothelial dysfunction caused by HFD is related to a dysfunctional endothelial AMPK-PI3K-Akt-eNOS pathway correlating with the increase of plasma NEFA, TG, and an impaired glucose management.
Collapse
Affiliation(s)
- Concha F García-Prieto
- Departamento de Ciencias Farmacéuticas y de la Salud, Facultad de Farmacia, Universidad CEU-San Pablo, Madrid, Spain
| | | | | | | | | | | | | | | |
Collapse
|
27
|
Gonzalez-Vicente A, Cabral PD, Garvin JL. Resveratrol increases nitric oxide production in the rat thick ascending limb via Ca2+/calmodulin. PLoS One 2014; 9:e110487. [PMID: 25314136 PMCID: PMC4196991 DOI: 10.1371/journal.pone.0110487] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2014] [Accepted: 09/19/2014] [Indexed: 12/12/2022] Open
Abstract
The thick ascending limb of the loop of Henle reabsorbs 30% of the NaCl filtered through the glomerulus. Nitric oxide (NO) produced by NO synthase 3 (NOS3) inhibits NaCl absorption by this segment. Resveratrol, a polyphenol, has beneficial cardiovascular and renal effects, many of which are mediated by NO. Resveratrol increases intracellular Ca2+ (Cai) and AMP kinase (AMPK) and NAD-dependent deacetylase sirtuin1 (SIRT1) activities, all of which could activate NO production. We hypothesized that resveratrol stimulates NO production by thick ascending limbs via a Ca2+/calmodulin-dependent mechanism. To test this, the effect of resveratrol on NO bioavailability was measured in thick ascending limb suspensions. Cai was measured in single perfused thick ascending limbs. SIRT1 activity and expression were measured in thick ascending limb lysates. Resveratrol (100 µM) increased NO bioavailability in thick ascending limb suspensions by 1.3±0.2 AFU/mg/min (p<0.03). The NOS inhibitor L-NAME blunted resveratrol-stimulated NO bioavailability by 96±11% (p<0.03). The superoxide scavenger tempol had no effect. Resveratrol elevated Cai from 48±7 to 135±24 nM (p<0.01) in single tubules. In Ca2+-free media, the resveratrol-induced increase in NO was blunted by 60±20% (p<0.05) and the rise in Cai reduced by 80%. Calmodulin inhibition prevented the resveratrol-induced increase in NO (p<0.002). AMPK inhibition had no effect. Resveratrol did not increase SIRT1 activity. We conclude that resveratrol increases NO production in thick ascending limbs via a Ca2+/calmodulin dependent mechanism, and SIRT1 and AMPK do not participate. Resveratrol-stimulated NO production in thick ascending limbs may account for part of its beneficial effects.
Collapse
Affiliation(s)
- Agustin Gonzalez-Vicente
- Department of Physiology and Biophysics, Case Western Reserve University, School of Medicine, Cleveland, Ohio, United States of America
- Universidad de Buenos Aires, Facultad de Farmacia y Bioquímica, Ciudad Autónoma de Buenos Aires, Buenos Aires, Argentina
| | - Pablo D. Cabral
- Department of Physiology and Biophysics, Case Western Reserve University, School of Medicine, Cleveland, Ohio, United States of America
- Universidad de Buenos Aires, Facultad de Medicina, Ciudad Autónoma de Buenos Aires, Buenos Aires, Argentina
| | - Jeffrey L. Garvin
- Department of Physiology and Biophysics, Case Western Reserve University, School of Medicine, Cleveland, Ohio, United States of America
- Hypertension and Vascular Research, Department of Internal Medicine, Henry Ford Hospital, Detroit, Michigan, United States of America
- * E-mail:
| |
Collapse
|
28
|
Kim YH, Hwang JH, Kim KS, Noh JR, Gang GT, Oh WK, Jeong KH, Kwak TH, Choi HS, Lee IK, Lee CH. Enhanced activation of NAD(P)H: quinone oxidoreductase 1 attenuates spontaneous hypertension by improvement of endothelial nitric oxide synthase coupling via tumor suppressor kinase liver kinase B1/adenosine 5'-monophosphate-activated protein kinase-mediated guanosine 5'-triphosphate cyclohydrolase 1 preservation. J Hypertens 2014; 32:306-17. [PMID: 24241058 DOI: 10.1097/hjh.0000000000000018] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
AIMS Guanosine 5'-triphosphate cyclohydrolase-1 (GTPCH-1) is a rate-limiting enzyme in de-novo synthesis of tetrahydrobiopterin (BH4), an essential cofactor for endothelial nitric oxide synthase (eNOS) coupling. Adenosine 5'-monophosphate-activated protein kinase (AMPK) is crucial for GTPCH-1 preservation, and tumor suppressor kinase liver kinase B1 (LKB1), an upstream kinase of AMPK, is activated by NAD-dependent class III histone deacetylase sirtuin 1 (SIRT1)-mediated deacetylation. β-Lapachone has been shown to increase cellular NAD/NADH ratio via NAD(P)H quinone oxidoreductase 1 (NQO1) activation. In this study, we have evaluated whether β-lapachone-induced NQO1 activation modulates blood pressure (BP) through preservation of GTPCH-1 in a hypertensive animal model. METHODS AND RESULTS Spontaneously hypertensive rats (SHRs), primary aortic endothelial cells, and endothelial cell line were used to investigate the hypotensive effect of β-lapachone and its action mechanism. β-Lapachone treatment dramatically lowered BP and vascular tension in SHRs and induced eNOS activation in endothelial cells. Consistent with these effects, β-lapachone treatment also elevated levels of both aortic cGMP and plasma nitric oxide in SHRs. Meanwhile, β-lapachone-treated SHRs showed significantly increased levels of aortic NAD, LKB1 deacetylation, and AMPK Thr phosphorylation followed by increased GTPCH-1 and tetrahydrobiopterin/dihydrobiopterin ratio. In-vitro study revealed that AMPK inhibition by overexpression of dominant-negative AMPK nearly abolished GTPCH-1 protein conservation. Enhanced LKB1 deacetylation and AMPK activation were also elicited by β-lapachone in endothelial cells. However, inhibition of LKB1 deacetylation by blocking of NQO1 or SIRT1 blunted AMPK activation by β-lapachone. CONCLUSION This is the first study demonstrating that eNOS coupling can be regulated by NQO1 activation via LKB1/AMPK/GTPCH-1 modulation, which is possibly correlated with relieving hypertension. These findings provide strong evidence to suggest that NQO1 might be a new therapeutic target for hypertension.
Collapse
Affiliation(s)
- Yong-Hoon Kim
- aLaboratory Animal Center, Korea Research Institute of Bioscience and Biotechnology, Yuseong-gu, Daejeon bKorea Bioactive Natural Material Bank, College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul cDiabetes and Metabolic Disease Research Center, Lee Gil Ya Cancer & Diabetes Institute, Gachon University of Medicine, Incheon dKT&G Life Sciences Corporation/R&D Center, Suwon eHormone Research Center, School of Biological Sciences and Technology, Chonnam National University, Gwangju fDepartment of Internal Medicine, Kyungpook National University School of Medicine, Daegu, South Korea
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
29
|
Pyla R, Osman I, Pichavaram P, Hansen P, Segar L. Metformin exaggerates phenylephrine-induced AMPK phosphorylation independent of CaMKKβ and attenuates contractile response in endothelium-denuded rat aorta. Biochem Pharmacol 2014; 92:266-79. [PMID: 25179145 DOI: 10.1016/j.bcp.2014.08.024] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2014] [Accepted: 08/21/2014] [Indexed: 02/08/2023]
Abstract
Metformin, a widely prescribed antidiabetic drug, has been shown to reduce the risk of cardiovascular disease, including hypertension. Its beneficial effect toward improved vasodilation results from its ability to activate AMPK and enhance nitric oxide formation in the endothelium. To date, metformin regulation of AMPK has not been fully studied in intact arterial smooth muscle, especially during contraction evoked by G protein-coupled receptor (GPCR) agonists. In the present study, ex vivo incubation of endothelium-denuded rat aortic rings with 3mM metformin for 2h resulted in significant accumulation of metformin (∼ 600 pmoles/mg tissue), as revealed by LC-MS/MS MRM analysis. However, metformin did not show significant increase in AMPK phosphorylation under these conditions. Exposure of aortic rings to a GPCR agonist (e.g., phenylephrine) resulted in enhanced AMPK phosphorylation by ∼ 2.5-fold. Importantly, in metformin-treated aortic rings, phenylephrine challenge showed an exaggerated increase in AMPK phosphorylation by ∼ 9.7-fold, which was associated with an increase in AMP/ATP ratio. Pretreatment with compound C (AMPK inhibitor) prevented AMPK phosphorylation induced by phenylephrine alone and also that induced by phenylephrine after metformin treatment. However, pretreatment with STO-609 (CaMKKβ inhibitor) diminished AMPK phosphorylation induced by phenylephrine alone but not that induced by phenylephrine after metformin treatment. Furthermore, attenuation of phenylephrine-induced contraction (observed after metformin treatment) was prevented by AMPK inhibition but not by CaMKKβ inhibition. Together, these findings suggest that, upon endothelial damage in the vessel wall, metformin uptake by the underlying vascular smooth muscle would accentuate AMPK phosphorylation by GPCR agonists independent of CaMKKβ to promote vasorelaxation.
Collapse
Affiliation(s)
- Rajkumar Pyla
- Center for Pharmacy and Experimental Therapeutics, University of Georgia College of Pharmacy, 1120 15th Street, HM-1200 Georgia Regents University Campus, Augusta, GA 30912-2450, USA; Charlie Norwood VA Medical Center, Augusta, GA, USA
| | - Islam Osman
- Center for Pharmacy and Experimental Therapeutics, University of Georgia College of Pharmacy, 1120 15th Street, HM-1200 Georgia Regents University Campus, Augusta, GA 30912-2450, USA; Charlie Norwood VA Medical Center, Augusta, GA, USA
| | - Prahalathan Pichavaram
- Center for Pharmacy and Experimental Therapeutics, University of Georgia College of Pharmacy, 1120 15th Street, HM-1200 Georgia Regents University Campus, Augusta, GA 30912-2450, USA; Charlie Norwood VA Medical Center, Augusta, GA, USA
| | - Paul Hansen
- Center for Pharmacy and Experimental Therapeutics, University of Georgia College of Pharmacy, 1120 15th Street, HM-1200 Georgia Regents University Campus, Augusta, GA 30912-2450, USA; Charlie Norwood VA Medical Center, Augusta, GA, USA
| | - Lakshman Segar
- Center for Pharmacy and Experimental Therapeutics, University of Georgia College of Pharmacy, 1120 15th Street, HM-1200 Georgia Regents University Campus, Augusta, GA 30912-2450, USA; Charlie Norwood VA Medical Center, Augusta, GA, USA; Vascular Biology Center, Department of Pharmacology and Toxicology, Georgia Regents University, Augusta, GA, USA; Department of Medicine, Pennsylvania State University College of Medicine, Hershey, PA, USA.
| |
Collapse
|
30
|
Wang Z, Zhang J, Li B, Gao X, Liu Y, Mao W, Chen SL. Resveratrol ameliorates low shear stress‑induced oxidative stress by suppressing ERK/eNOS‑Thr495 in endothelial cells. Mol Med Rep 2014; 10:1964-72. [PMID: 25198200 DOI: 10.3892/mmr.2014.2390] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2013] [Accepted: 05/09/2014] [Indexed: 11/06/2022] Open
Abstract
Fluid shear stress has been revealed to differentially regulate endothelial nitric oxide synthase (eNOS) distribution in vessels. eNOS, a key enzyme in controlling nitric oxide (NO) release, has a crucial role in mediating oxidative stress, and resveratrol (RSV)‑mediated eNOS also attenuates oxidative damage and suppresses endothelial dysfunction. To observe the protective effect of RSV on low shear stress (LSS)‑induced oxidative damage and the potential mechanisms involved, a parallel‑plate flow chamber, which imposed a low level of stress of 2 dynes/cm2 to cells, was employed. Reactive oxygen species (ROS), NO and apoptotic cells were examined in LSS‑treated endothelial cells (ECs) with or without RSV. Western blot analysis was used to examine LSS‑regulated eNOS‑Ser1177, Thr495 and Ser633, which were tightly associated with NO release. To further determine the underlying signaling pathways involved, extracellular signal‑regulated kinase (ERK), a possible upstream target of eNOS‑Thr495, was investigated, followed by examination of eNOS‑Thr495 in ERK‑inhibited cells. Additionally, eNOS mRNA expression levels were analyzed in cells challenged with LSS. The results revealed that RSV markedly decreased LSS‑induced oxidative damage in ECs. Furthermore, eNOS‑Ser1177 and Thr495 as well as phospho‑ERK were time‑dependently activated by LSS. The ERK inhibitor deactivated eNOS‑Thr495, which was accompanied by increased intracellular superoxide dismutase (SOD) levels. Of note, the activation effect of LSS on ERK/eNOS was markedly eliminated by RSV. In conclusion, RSV exerts antioxidant effects by suppressing LSS-activated ERK/eNOS and may provide a potential therapeutic target for atherosclerosis.
Collapse
Affiliation(s)
- Zhimei Wang
- Department of Cardiology, Nanjing First Hospital, Nanjing Medical University, Nanjing, Jiangsu 210006, P.R. China
| | - Junxia Zhang
- Department of Cardiology, Nanjing First Hospital, Nanjing Medical University, Nanjing, Jiangsu 210006, P.R. China
| | - Bing Li
- Department of Cardiology, Nanjing First Hospital, Nanjing Medical University, Nanjing, Jiangsu 210006, P.R. China
| | - Xiaofei Gao
- Department of Cardiology, Nanjing First Hospital, Nanjing Medical University, Nanjing, Jiangsu 210006, P.R. China
| | - Yanrong Liu
- Department of Cardiology, Nanjing First Hospital, Nanjing Medical University, Nanjing, Jiangsu 210006, P.R. China
| | - Wenxing Mao
- Department of Cardiology, Nanjing First Hospital, Nanjing Medical University, Nanjing, Jiangsu 210006, P.R. China
| | - Shao-Liang Chen
- Department of Cardiology, Nanjing First Hospital, Nanjing Medical University, Nanjing, Jiangsu 210006, P.R. China
| |
Collapse
|
31
|
Turkseven S, Ertuna E. Prolonged AMP-activated protein kinase induction impairs vascular functions. Can J Physiol Pharmacol 2013; 91:1025-30. [DOI: 10.1139/cjpp-2013-0160] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
AMP-activated protein kinase (AMPK) is a regulator of cellular metabolism and is involved in the pathogenesis of several diseases, including type 2 diabetes and cardiovascular diseases. Data showing the effects of AMPK on vasculature are controversial. Therefore, the aim of this study was to determine the impact of prolonged AMPK activation on vascular functions. For this purpose we have examined the role of AMPK in endothelium-dependent and -independent relaxation and vascular contractions. For this, we incubated thoracic aortic rings, from rats, with AMPK activator 5-aminoimidazole-4-carboxamide-1-4-ribofuranoside (AICAR, 500 μmol/L or 2 mmol/L) in the presence or absence of AMPK inhibitor compound C (10 μmol/L). Next, cumulative dose–response curves to acetylcholine (ACh) (10−9−10−4 mol/L), nitroglycerine (NG) (10−9–3 × 10−5 mol/L), and noradrenaline (NA) (10−9−10−4 mol/L) were obtained. Endothelial nitric oxide synthase (eNOS) protein expression was determined. Our results show that endothelium-dependent relaxation was inhibited after AICAR treatment, and that this effect was reversed by AMPK inhibition. Moreover, AICAR enhanced the contractile response to NA and caused a decrease in eNOS protein expression. In conclusion, prolonged AMPK induction causes endothelial impairment, possibly via increased degradation and (or) reduced expression of eNOS.
Collapse
Affiliation(s)
- Saadet Turkseven
- Ege University, Faculty of Pharmacy, Department of Pharmacology, Bornova-Izmir 35100, Turkey
| | - Elif Ertuna
- Ege University, Faculty of Pharmacy, Department of Pharmacology, Bornova-Izmir 35100, Turkey
| |
Collapse
|
32
|
Carrizzo A, Puca A, Damato A, Marino M, Franco E, Pompeo F, Traficante A, Civitillo F, Santini L, Trimarco V, Vecchione C. Resveratrol improves vascular function in patients with hypertension and dyslipidemia by modulating NO metabolism. Hypertension 2013; 62:359-66. [PMID: 23753407 DOI: 10.1161/hypertensionaha.111.01009] [Citation(s) in RCA: 111] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Epidemiological studies have demonstrated that the Mediterranean diet, which is rich in resveratrol, is associated with a significantly reduced risk of cardiovascular disease. However, the molecular mechanisms that underlie the beneficial effects of resveratrol on cardiovascular function remain incompletely understood. Therefore, we set out to identify the molecular target(s) mediating the protective action of resveratrol on vascular function. To this end, we performed vascular reactivity studies to evaluate the effects of resveratrol on superior thyroid artery obtained from 59 patients with hypertension and dyslipidemia. We found that resveratrol evoked vasorelaxation and reduced endothelial dysfunction through the modulation of NO metabolism via (1) an 5' adenosine monophosphate-activated protein kinase-mediated increase in endothelial NO synthase activity; (2) a rise in tetrahydrobiopterin levels, which also increases endothelial NO synthase activity; and (3) attenuation of vascular oxidative stress, brought about by overexpression of manganese superoxide dismutase via an nuclear factor erythroid-derived 2-like 2-dependent mechanism. The effects of resveratrol on acetylcholine vasorelaxation were also tested in vessels from patients with nonhypertensive nondyslipidemia undergoing thyroid surgery. In this setting, resveratrol failed to exert any effect. Thus, our finding that resveratrol reduces endothelial dysfunction, an early pathophysiological feature and independent predictor of poor prognosis in most forms of cardiovascular disease, supports the concept that the risk of vascular events could be further reduced by adherence to a set of dietary and behavioral guidelines.
Collapse
Affiliation(s)
- Albino Carrizzo
- Vascular Physiopathology Unit, University of Naples Federico II, Naples, Italy
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Lee KY, Choi HC. Acetylcholine-induced AMP-activated protein kinase activation attenuates vasoconstriction through an LKB1-dependent mechanism in rat aorta. Vascul Pharmacol 2013; 59:96-102. [PMID: 23916920 DOI: 10.1016/j.vph.2013.07.007] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2013] [Revised: 06/07/2013] [Accepted: 07/24/2013] [Indexed: 01/27/2023]
Abstract
Numerous studies of acetylcholine (ACh)-induced endothelium-dependent relaxation in arteries have been reported since the original description by Furchgott and Zawadzki (1980). ACh also produces endothelium-independent relaxation. However, it is still unknown whether ACh-induced AMP-activated protein kinase (AMPK) activation can attenuate vasoconstriction in endothelium-denuded rat aorta. Here, we investigated whether ACh may exert a regulatory effect for vascular tone via AMPK activation and its underlying mechanism in vascular smooth muscle cells (VSMCs). Western blotting showed that ACh dose- and time-dependently increased LKB1 and AMPK phosphorylation in VSMCs. The ACh-induced activation of AMPK required muscarinic receptors in VSMCs. LKB1 and AMPK activation by ACh inhibited myosin light-chain kinase (MLCK) and phosphorylated myosin light chain (p-MLC) expression in VSMCs. In addition, a tension study showed the inhibitory effect of ACh-induced AMPK activation on phenylephrine-mediated contraction in endothelium-denuded rat aorta. These data suggest that the ACh-induced activation of AMPK may attenuate vasoconstriction via LKB1-AMPK-dependent mechanism in endothelium-denuded rat aorta.
Collapse
Affiliation(s)
- Kyung Young Lee
- Department of Pharmacology, Aging-associated Vascular Disease Research Center, College of Medicine, Yeungnam University, Daegu, 705-717, Korea
| | | |
Collapse
|
34
|
Seto SW, Au ALS, Poon CCW, Zhang Q, Li RWS, Yeung JHK, Kong SK, Ngai SM, Wan S, Ho HP, Lee SMY, Hoi MPM, Chan SW, Leung GPH, Kwan YW. Acute simvastatin inhibits K ATP channels of porcine coronary artery myocytes. PLoS One 2013; 8:e66404. [PMID: 23799098 PMCID: PMC3684588 DOI: 10.1371/journal.pone.0066404] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2013] [Accepted: 05/06/2013] [Indexed: 01/19/2023] Open
Abstract
Background Statins (3-hydroxy-3-methyl-glutaryl coenzyme A (HMG-CoA) reductase inhibitors) consumption provides beneficial effects on cardiovascular systems. However, effects of statins on vascular KATP channel gatings are unknown. Methods Pig left anterior descending coronary artery and human left internal mammary artery were isolated and endothelium-denuded for tension measurements and Western immunoblots. Enzymatically-dissociated/cultured arterial myocytes were used for patch-clamp electrophysiological studies and for [Ca2+]i, [ATP]i and [glucose]o uptake measurements. Results The cromakalim (10 nM to 10 µM)- and pinacidil (10 nM to 10 µM)-induced concentration-dependent relaxation of porcine coronary artery was inhibited by simvastatin (3 and 10 µM). Simvastatin (1, 3 and 10 µM) suppressed (in okadaic acid (10 nM)-sensitive manner) cromakalim (10 µM)- and pinacidil (10 µM)-mediated opening of whole-cell KATP channels of arterial myocytes. Simvastatin (10 µM) and AICAR (1 mM) elicited a time-dependent, compound C (1 µM)-sensitive [3H]-2-deoxy-glucose uptake and an increase in [ATP]i levels. A time (2–30 min)- and concentration (0.1–10 µM)-dependent increase by simvastatin of p-AMPKα-Thr172 and p-PP2A-Tyr307 expression was observed. The enhanced p-AMPKα-Thr172 expression was inhibited by compound C, ryanodine (100 µM) and KN93 (10 µM). Simvastatin-induced p-PP2A-Tyr307 expression was suppressed by okadaic acid, compound C, ryanodine, KN93, phloridzin (1 mM), ouabain (10 µM), and in [glucose]o-free or [Na+]o-free conditions. Conclusions Simvastatin causes ryanodine-sensitive Ca2+ release which is important for AMPKα-Thr172 phosphorylation via Ca2+/CaMK II. AMPKα-Thr172 phosphorylation causes [glucose]o uptake (and an [ATP]i increase), closure of KATP channels, and phosphorylation of AMPKα-Thr172 and PP2A-Tyr307 resulted. Phosphorylation of PP2A-Tyr307 occurs at a site downstream of AMPKα-Thr172 phosphorylation.
Collapse
Affiliation(s)
- Sai Wang Seto
- The Vascular Biology Unit, Queensland Research Centre for Peripheral Vascular Disease, School of Medicine and Dentistry, James Cook University, Townsville, Queensland, Australia
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong, PR of China
| | - Alice Lai Shan Au
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong, PR of China
| | - Christina Chui Wa Poon
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong, PR of China
| | - Qian Zhang
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong, PR of China
| | - Rachel Wai Sum Li
- Department of Pharmacology and Pharmacy, Faculty of Medicine, The University of Hong Kong, Hong Kong, PR of China
| | - John Hok Keung Yeung
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong, PR of China
| | - Siu Kai Kong
- School of Life Sciences, Faculty of Science, The Chinese University of Hong Kong, Shatin, Hong Kong, PR of China
| | - Sai Ming Ngai
- School of Life Sciences, Faculty of Science, The Chinese University of Hong Kong, Shatin, Hong Kong, PR of China
| | - Song Wan
- Department of Surgery, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong, PR of China
| | - Ho Pui Ho
- Department of Electronic Engineering, Faculty of Engineering, The Chinese University of Hong Kong, Shatin, Hong Kong, PR of China
| | - Simon Ming Yuen Lee
- Institute of Chinese Medical Sciences, the University of Macau, Macau, PR of China
| | - Maggie Pui Man Hoi
- Institute of Chinese Medical Sciences, the University of Macau, Macau, PR of China
| | - Shun Wan Chan
- State Key Laboratory of Chinese Medicine and Molecular Pharmacology, Department of Applied Biology and Chemical Technology, The Hong Kong Polytechnic University, Kowloon, Hong Kong, PR of China
- * E-mail: (YWK); (GPHL); (SWC)
| | - George Pak Heng Leung
- Department of Pharmacology and Pharmacy, Faculty of Medicine, The University of Hong Kong, Hong Kong, PR of China
- * E-mail: (YWK); (GPHL); (SWC)
| | - Yiu Wa Kwan
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong, PR of China
- * E-mail: (YWK); (GPHL); (SWC)
| |
Collapse
|
35
|
Banek CT, Bauer AJ, Needham KM, Dreyer HC, Gilbert JS. AICAR administration ameliorates hypertension and angiogenic imbalance in a model of preeclampsia in the rat. Am J Physiol Heart Circ Physiol 2013; 304:H1159-65. [PMID: 23417865 PMCID: PMC3625906 DOI: 10.1152/ajpheart.00903.2012] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/05/2012] [Accepted: 02/12/2013] [Indexed: 01/15/2023]
Abstract
Previous studies suggest restoration of angiogenic balance can lower blood pressure and improve vascular endothelium function in models of preeclampsia. Our laboratory has recently reported exercise training mitigates hypertension in an animal model of preeclampsia, but the mechanisms are unknown. AMP-activated protein kinase (AMPK) is stimulated during exercise and has been shown to increase expression of VEGF. Therefore, the purpose of this study was to determine whether AICAR (5-aminoimidazole-4-carboxamide-3-ribonucleoside), a potent AMPK stimulator, would increase circulating VEGF, improve angiogenic potential, decrease oxidative stress, and abrogate placental ischemia-induced hypertension. In rats, reduced uteroplacental perfusion pressure (RUPP) was induced on day 14 of gestation by introducing silver clips on the inferior abdominal aorta and ovarian arteries. AICAR was administered intraperitoneally (50 mg/kg b.i.d.) days 14-18, and blood pressure and tissues were collected on day 19. RUPP-induced hypertension was ameliorated (P < 0.05) with AICAR versus RUPP. AICAR increased (P < 0.05) plasma VEGF and decreased (P < 0.05) plasma soluble VEGF receptor-1 in the RUPP + AICAR versus RUPP. Antioxidant capacity was restored (P < 0.05) by AICAR in RUPP placenta. Renal and placental catalase activity was decreased (P < 0.05) in RUPP + AICAR versus RUPP. Angiogenic potential was increased (P < 0.05) in RUPP + AICAR versus RUPP. Fetal and placental weights were unaffected by AICAR. Placental AMPK phosphorylation was increased (P < 0.05) in RUPP + AICAR versus normal pregnant and RUPP. These findings suggest AICAR may be useful to mitigate angiogenic imbalance, renal, and placental oxidative stress and increase in blood pressure associated with RUPP hypertension. Furthermore, placental AMPK phosphorylation was observed only in the setting of ischemia.
Collapse
Affiliation(s)
- Christopher T Banek
- Department of Human Physiology, University of Oregon, Eugene, Oregon 97402-1240, USA
| | | | | | | | | |
Collapse
|
36
|
Liang B, Wang S, Wang Q, Zhang W, Viollet B, Zhu Y, Zou MH. Aberrant endoplasmic reticulum stress in vascular smooth muscle increases vascular contractility and blood pressure in mice deficient of AMP-activated protein kinase-α2 in vivo. Arterioscler Thromb Vasc Biol 2013; 33:595-604. [PMID: 23288166 DOI: 10.1161/atvbaha.112.300606] [Citation(s) in RCA: 67] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
OBJECTIVE The endoplasmic reticulum (ER) plays a critical role in ensuring proper folding of newly synthesized proteins. Aberrant ER stress is reported to play a causal role in cardiovascular diseases. However, the effects of ER stress on vascular smooth muscle contractility and blood pressure remain unknown. The aim of this study was to investigate whether aberrant ER stress causes abnormal vasoconstriction and consequent high blood pressure in mice. METHODS AND RESULTS ER stress markers, vascular smooth muscle contractility, and blood pressure were monitored in mice. Incubation of isolated aortic rings with tunicamycin or MG132, 2 structurally unrelated ER stress inducers, significantly increased both phenylephrine-induced vasoconstriction and the phosphorylation of myosin light chain (Thr18/Ser19), both of which were abrogated by pretreatment with chemical chaperones or 5-Aminoimidazole-4-carboxamide ribonucleotide and metformin, 2 potent activators for the AMP-activated protein kinase. Consistently, administration of tauroursodeoxycholic acid or 4-phenyl butyric acid, 2 structurally unrelated chemical chaperones, in AMP-activated protein kinase-α2 knockout mice lowered blood pressure and abolished abnormal vasoconstrictor response of AMP-activated protein kinase-α2 knockout mice to phenylephrine. Consistently, tunicamycin (0.01 μg/g per day) infusion markedly increased both systolic and diastolic blood pressure, both of which were ablated by coadministration of 4-phenyl butyric acid. Furthermore, 4-phenyl butyric acid or tauroursodeoxycholic acid, which suppressed angiotensin II infusion-induced ER stress markers in vivo, markedly lowered blood pressure in angiotensin II-infused mice in vivo. CONCLUSIONS We conclude that ER stress increases vascular smooth muscle contractility resulting in high blood pressure, and AMP-activated protein kinase activation mitigates high blood pressure through the suppression of ER stress in vivo.
Collapse
Affiliation(s)
- Bin Liang
- Section of Molecular Medicine, Department of Medicine, University of Oklahoma Health Science Center, Oklahoma City, OK 73104, USA
| | | | | | | | | | | | | |
Collapse
|
37
|
Davis B, Rahman A, Arner A. AMP-activated kinase relaxes agonist induced contractions in the mouse aorta via effects on PKC signaling and inhibits NO-induced relaxation. Eur J Pharmacol 2012; 695:88-95. [PMID: 22921370 DOI: 10.1016/j.ejphar.2012.07.025] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2012] [Revised: 06/26/2012] [Accepted: 07/02/2012] [Indexed: 10/28/2022]
Abstract
Adenosine monophosphate activated kinase (AMPK), a regulator of cellular metabolism, has been shown to relax arterial smooth muscle via endothelium-dependent and independent mechanisms. We have examined the role of AMPK in different smooth muscles using the activating compound, 5-amino-4-imidazolecarboxamide riboside-1-β-d-ribofuranoside (AICAR). Isolated preparations of mouse aorta, saphenous artery, ileum and urinary bladder were compared. AICAR produced a reversible dose-dependent relaxation in aortic rings pre-incubated with AICAR and activated with phenylephrine. Less prominent relaxation was noted in the other tissues. This difference in sensitivity to AICAR was not due to differences in the expression levels of AMPK α1 mRNA. In the aorta, AICAR had a greater effect on contractions induced by phenylephrine, compared to high-K(+) induced contractions. Contractions of the aorta in response to the protein kinase C activator PDBu were prominently inhibited by AICAR. The AICAR relaxation observed in the aorta was not prevented by the NOS inhibitor L-NAME, Indomethacin or endothelium removal. Nitric oxide (NO) mediated relaxations in aortic preparations induced by acetylcholine or sodium nitroprusside (SNP) were attenuated by AICAR. In conclusion, AMPK induced relaxation of smooth muscle is tissue-dependent and most prominent in large elastic arteries. The smooth muscle relaxation is NO-independent and occurs downstream of PKC activation and is associated with attenuated relaxant responses to NO.
Collapse
Affiliation(s)
- Benjamin Davis
- Department of Physiology & Pharmacology, Karolinska Institutet, Von Eulers väg 8, Stockholm, Sweden
| | | | | |
Collapse
|
38
|
AMP-activated protein kinase activator AICAR acutely lowers blood pressure and relaxes isolated resistance arteries of hypertensive rats. J Hypertens 2012; 30:725-33. [PMID: 22306847 DOI: 10.1097/hjh.0b013e32835050ca] [Citation(s) in RCA: 57] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
OBJECTIVES AMP-activated protein kinase (AMPK) activity may alter blood pressure by directly influencing vascular tone. The purpose of this study is to examine if these effects occur acutely in a model of hypertension. METHODS AND RESULTS Using distinct groups of Wistar-Kyoto rats (WKY) and spontaneously hypertensive rats (SHR) we compare baseline aortic and mesenteric artery AMPK activation (by immunoblotting), hemodynamic (blood pressure and heart rate via carotid catheter) and biochemical responses to an acute injection of AMPK activator 5-aminoimidazole-4-carboxyamide-1-β-D-ribofuranoside (AICAR) in vivo and vasomotor responses of isolated mesenteric vessels to AICAR exposure in vitro using myography. Mean arterial pressure (MAP) decreased from 196 ± 3 to 122 ± 15 mmHg (P < 0.001) during the 30 min following AICAR injection in SHR (an effect partially prevented by NOS inhibitor L-NAME), but in WKY MAP was unaffected by AICAR. Basal AMPK activation (phosphorylation of AMPK activation site threonine 172) was reduced by approximately 50% in aorta of SHR vs. WKY (0.49 ± 0.1 vs. 1.0 ± 0.1 arbitrary units, P < 0.001), and was improved approximately 1.6-fold in SHR but not in WKY aorta 30 min following AICAR injection. In isolated vessel experiments, dose-dependent vasorelaxation to AICAR was similar in mesenteric arteries of SHR and WKY, although responses were more reliant on nitric oxide in SHR vs. WKY. CONCLUSIONS The ability of AICAR to improve vascular AMPK activation, and to generate parallel reductions in blood pressure and relaxation of SHR resistance vasculature, highlights the potential importance of AMPK in the regulation of blood pressure and vascular tone.
Collapse
|
39
|
Sung JY, Choi HC. Metformin-induced AMP-activated protein kinase activation regulates phenylephrine-mediated contraction of rat aorta. Biochem Biophys Res Commun 2012; 421:599-604. [PMID: 22525678 DOI: 10.1016/j.bbrc.2012.04.052] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2012] [Accepted: 04/10/2012] [Indexed: 12/01/2022]
Abstract
The aim of the present study is to determine the effects and molecular mechanisms by which activation of LKB1-AMP-activated protein kinase (AMPK) by metformin regulates vascular smooth muscle contraction. The essential ability of vascular smooth muscle cells (VSMCs) to contract and relax in response to an elevation and reduction in intravascular pressure is necessary for appropriate blood flow regulation. Thus, vessel contraction is a critical mechanism for systemic blood flow regulation. In cultured rat VSMCs, AMPK activation through LKB1 by metformin-inhibited phenylephrine-mediated myosin light chain kinase (MLCK) and myosin light chain phosphorylation (p-MLC). Conversely, inhibition of AMPK and LKB1 reversed phenylephrine-induced MLCK and p-MLC phosphorylation. Measurement of the tension trace in rat aortic rings also showed that the effect of AMPK activation by metformin decreased phenylephrine-induced contraction. Metformin inhibited PE-induced p-MLC and α-smooth muscle actin co-localization. Our results suggest that activation of AMPK by LKB1 decreases VSMC contraction by inhibiting MLCK and p-MLC, indicating that induction by the AMPK-LKB1 pathway may be a new therapeutic target to lower high blood pressure.
Collapse
Affiliation(s)
- Jin Young Sung
- Department of Pharmacology, Aging-associated Vascular Disease Research Center, Yeungnam University, Daegu 705-717, Republic of Korea
| | | |
Collapse
|
40
|
Nagai A, Sado T, Naruse K, Noguchi T, Haruta S, Yoshida S, Tanase Y, Tsunemi T, Kobayashi H. Antiangiogenic-Induced Hypertension: The Molecular Basis of Signaling Network. Gynecol Obstet Invest 2012; 73:89-98. [DOI: 10.1159/000334458] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2011] [Accepted: 10/16/2011] [Indexed: 01/09/2023]
|
41
|
Aspirin-induced AMP-activated protein kinase activation regulates the proliferation of vascular smooth muscle cells from spontaneously hypertensive rats. Biochem Biophys Res Commun 2011; 408:312-7. [DOI: 10.1016/j.bbrc.2011.04.027] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2011] [Accepted: 04/06/2011] [Indexed: 11/18/2022]
|