1
|
Zhuang T, Lei Y, Chang JJ, Zhou YP, Li Y, Li YX, Yang YF, Chen MH, Meng T, Fu SM, Huang LH, Cheang WS, Cooke JP, Dong ZH, Bai YN, Ruan CC. A2AR-mediated lymphangiogenesis via VEGFR2 signaling prevents salt-sensitive hypertension. Eur Heart J 2023; 44:2730-2742. [PMID: 37377160 PMCID: PMC10393074 DOI: 10.1093/eurheartj/ehad377] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/28/2022] [Revised: 04/17/2023] [Accepted: 05/24/2023] [Indexed: 06/29/2023] Open
Abstract
AIMS Excess dietary sodium intake and retention lead to hypertension. Impaired dermal lymphangiogenesis and lymphatic dysfunction-mediated sodium and fluid imbalance are pathological mechanisms. The adenosine A2A receptor (A2AR) is expressed in lymphatic endothelial cells (LECs), while the roles and mechanisms of LEC-A2AR in skin lymphangiogenesis during salt-induced hypertension are not clear. METHODS AND RESULTS The expression of LEC-A2AR correlated with lymphatic vessel density in both high-salt diet (HSD)-induced hypertensive mice and hypertensive patients. Lymphatic endothelial cell-specific A2AR knockout mice fed HSD exhibited 17 ± 2% increase in blood pressure and 17 ± 3% increase in Na+ content associated with decreased lymphatic density (-19 ± 2%) compared with HSD-WT mice. A2AR activation by agonist CGS21680 increased lymphatic capillary density and decreased blood pressure in HSD-WT mice. Furthermore, this A2AR agonist activated MSK1 directly to promote VEGFR2 activation and endocytosis independently of VEGF as assessed by phosphoprotein profiling and immunoprecipitation assays in LECs. VEGFR2 kinase activity inhibitor fruquintinib or VEGFR2 knockout in LECs but not VEGF-neutralizing antibody bevacizumab suppressed A2AR activation-mediated decrease in blood pressure. Immunostaining revealed phosphorylated VEGFR2 and MSK1 expression in the LECs were positively correlated with skin lymphatic vessel density and A2AR level in hypertensive patients. CONCLUSION The study highlights a novel A2AR-mediated VEGF-independent activation of VEGFR2 signaling in dermal lymphangiogenesis and sodium balance, which might be a potential therapeutic target in salt-sensitive hypertension.
Collapse
Affiliation(s)
- Tao Zhuang
- Department of Physiology and Pathophysiology, Shanghai Key Laboratory of Bioactive Small Molecules, State Key Laboratory of Medical Neurobiology, School of Basic Medical Sciences, and Jinshan Hospital, Fudan University, 138 Yi-Xue-Yuan Road, Shanghai 200032, China
| | - Yu Lei
- Department of Physiology and Pathophysiology, Shanghai Key Laboratory of Bioactive Small Molecules, State Key Laboratory of Medical Neurobiology, School of Basic Medical Sciences, and Jinshan Hospital, Fudan University, 138 Yi-Xue-Yuan Road, Shanghai 200032, China
| | - Jin-Jia Chang
- Department of Gastrointestinal Medical Oncology, Fudan University Shanghai Cancer Center, 270 Dong-An Road, Shanghai 200032, China
| | - Yan-Ping Zhou
- Department of Radiology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, 160 Pu-Jian Road, Shanghai 200032, China
| | - Yan Li
- Department of Cardiology, RuiJin Hospital/LuWan Branch, Shanghai Jiao Tong University School of Medicine, 149 Chong-Qing-Nan Road, Shanghai 200032, China
| | - Yan-Xiu Li
- Department of Critical Care Medicine, The First Affiliated Hospital of Nanjing Medical University, 300 Guang-Zhou Road, Nanjing 210000, China
| | - Yong-Feng Yang
- Department of Physiology and Pathophysiology, Shanghai Key Laboratory of Bioactive Small Molecules, State Key Laboratory of Medical Neurobiology, School of Basic Medical Sciences, and Jinshan Hospital, Fudan University, 138 Yi-Xue-Yuan Road, Shanghai 200032, China
| | - Mei-Hua Chen
- Department of Physiology and Pathophysiology, Shanghai Key Laboratory of Bioactive Small Molecules, State Key Laboratory of Medical Neurobiology, School of Basic Medical Sciences, and Jinshan Hospital, Fudan University, 138 Yi-Xue-Yuan Road, Shanghai 200032, China
| | - Ting Meng
- Department of Physiology and Pathophysiology, Shanghai Key Laboratory of Bioactive Small Molecules, State Key Laboratory of Medical Neurobiology, School of Basic Medical Sciences, and Jinshan Hospital, Fudan University, 138 Yi-Xue-Yuan Road, Shanghai 200032, China
| | - Shi-Man Fu
- Department of Physiology and Pathophysiology, Shanghai Key Laboratory of Bioactive Small Molecules, State Key Laboratory of Medical Neurobiology, School of Basic Medical Sciences, and Jinshan Hospital, Fudan University, 138 Yi-Xue-Yuan Road, Shanghai 200032, China
| | - Li-Hao Huang
- Department of Chemistry and Institute of Metabolism and Integrative Biology, Shanghai Key Laboratory of Metabolic Remodeling and Health, Fudan University, 38 Yi-Xue-Yuan Road, Shanghai 200032, China
| | - Wai-San Cheang
- Institute of Chinese Medical Sciences, State Key Laboratory of Quality Research in Chinese Medicine, University of Macau, Long-Ma Road, Macau 999078, China
| | - John P Cooke
- Department of Cardiovascular Sciences, Center for Cardiovascular Regeneration, Houston Methodist Research Institute, Houston, TX 77030, USA
| | - Zhi-Hui Dong
- Department of Vascular Surgery, Zhongshan Hospital, and Center for Vascular Surgery and Wound Care, Jinshan Hospital, Fudan University, 180 Feng-Lin Road, Shanghai 200032, China
| | - Ying-Nan Bai
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, 180 Feng-Lin Road, Shanghai 200032, China
| | - Cheng-Chao Ruan
- Department of Physiology and Pathophysiology, Shanghai Key Laboratory of Bioactive Small Molecules, State Key Laboratory of Medical Neurobiology, School of Basic Medical Sciences, and Jinshan Hospital, Fudan University, 138 Yi-Xue-Yuan Road, Shanghai 200032, China
| |
Collapse
|
2
|
Raph SM, Dwenger MM, Hu X, Nystoriak MA. Basal NAD(H) redox state permits hydrogen peroxide-induced mesenteric artery dilatation. J Physiol 2023; 601:2621-2634. [PMID: 37114864 PMCID: PMC11714382 DOI: 10.1113/jp284195] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2022] [Accepted: 04/26/2023] [Indexed: 04/29/2023] Open
Abstract
Smooth muscle voltage-gated K+ (Kv) channels in resistance arteries control vascular tone and contribute to the coupling of blood flow with local metabolic activity. Members of the Kv1 family are expressed in vascular smooth muscle and are modulated upon physiological elevation of local metabolites, including the glycolytic end-product l-lactate and superoxide-derived hydrogen peroxide (H2 O2 ). Here, we show that l-lactate elicits vasodilatation of small-diameter mesenteric arteries in a mechanism that requires lactate dehydrogenase (LDH). Using the inside-out configuration of the patch clamp technique, we show that increases in NADH that reflect LDH-mediated conversion of l-lactate to pyruvate directly stimulate the activity of single Kv1 channels and significantly enhance the sensitivity of Kv1 activity to H2 O2 . Consistent with these findings, H2 O2 -evoked vasodilatation was significantly greater in the presence of 10 mM l-lactate relative to lactate-free conditions, yet was abolished in the presence of 10 mM pyruvate, which shifts the LDH reaction towards the generation of NAD+ . Moreover, the enhancement of H2 O2 -induced vasodilatation was abolished in arteries from double transgenic mice with selective overexpression of the intracellular Kvβ1.1 subunit in smooth muscle cells. Together, our results indicate that the Kvβ complex of native vascular Kv1 channels serves as a nodal effector for multiple redox signals to precisely control channel activity and vascular tone in the face of dynamic tissue-derived metabolic cues. KEY POINTS: Vasodilatation of mesenteric arteries by elevated external l-lactate requires its conversion by lactate dehydrogenase. Application of either NADH or H2 O2 potentiates single Kv channel currents in excised membrane patches from mesenteric artery smooth muscle cells. The binding of NADH enhances the stimulatory effects of H2 O2 on single Kv channel activity. The vasodilatory response to H2 O2 is differentially modified upon elevation of external l-lactate or pyruvate. The presence of l-lactate enhances the vasodilatory response to H2 O2 via the Kvβ subunit complex in smooth muscle.
Collapse
Affiliation(s)
- Sean M. Raph
- Department of Pharmacology and Toxicology, University of Louisville, Louisville, KY 40202
| | - Marc M. Dwenger
- Department of Pharmacology and Toxicology, University of Louisville, Louisville, KY 40202
| | - Xuemei Hu
- Department of Pharmacology and Toxicology, University of Louisville, Louisville, KY 40202
| | - Matthew A. Nystoriak
- Department of Medicine, Division of Environmental Medicine, University of Louisville, Louisville, KY 40202
| |
Collapse
|
3
|
Nayeem MA, Hanif A, Geldenhuys WJ, Agba S. Crosstalk between adenosine receptors and CYP450-derived oxylipins in the modulation of cardiovascular, including coronary reactive hyperemic response. Pharmacol Ther 2022; 240:108213. [PMID: 35597366 DOI: 10.1016/j.pharmthera.2022.108213] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Revised: 05/11/2022] [Accepted: 05/12/2022] [Indexed: 12/14/2022]
Abstract
Adenosine is a ubiquitous endogenous nucleoside or autacoid that affects the cardiovascular system through the activation of four G-protein coupled receptors: adenosine A1 receptor (A1AR), adenosine A2A receptor (A2AAR), adenosine A2B receptor (A2BAR), and adenosine A3 receptor (A3AR). With the rapid generation of this nucleoside from cellular metabolism and the widespread distribution of its four G-protein coupled receptors in almost all organs and tissues of the body, this autacoid induces multiple physiological as well as pathological effects, not only regulating the cardiovascular system but also the central nervous system, peripheral vascular system, and immune system. Mounting evidence shows the role of CYP450-enzymes in cardiovascular physiology and pathology, and the genetic polymorphisms in CYP450s can increase susceptibility to cardiovascular diseases (CVDs). One of the most important physiological roles of CYP450-epoxygenases (CYP450-2C & CYP2J2) is the metabolism of arachidonic acid (AA) and linoleic acid (LA) into epoxyeicosatrienoic acids (EETs) and epoxyoctadecaenoic acid (EpOMEs) which generally involve in vasodilation. Like an increase in coronary reactive hyperemia (CRH), an increase in anti-inflammation, and cardioprotective effects. Moreover, the genetic polymorphisms in CYP450-epoxygenases will change the beneficial cardiovascular effects of metabolites or oxylipins into detrimental effects. The soluble epoxide hydrolase (sEH) is another crucial enzyme ubiquitously expressed in all living organisms and almost all organs and tissues. However, in contrast to CYP450-epoxygenases, sEH converts EETs into dihydroxyeicosatrienoic acid (DHETs), EpOMEs into dihydroxyoctadecaenoic acid (DiHOMEs), and others and reverses the beneficial effects of epoxy-fatty acids leading to vasoconstriction, reducing CRH, increase in pro-inflammation, increase in pro-thrombotic and become less cardioprotective. Therefore, polymorphisms in the sEH gene (Ephx2) cause the enzyme to become overactive, making it more vulnerable to CVDs, including hypertension. Besides the sEH, ω-hydroxylases (CYP450-4A11 & CYP450-4F2) derived metabolites from AA, ω terminal-hydroxyeicosatetraenoic acids (19-, 20-HETE), lipoxygenase-derived mid-chain hydroxyeicosatetraenoic acids (5-, 11-, 12-, 15-HETEs), and the cyclooxygenase-derived prostanoids (prostaglandins: PGD2, PGF2α; thromboxane: Txs, oxylipins) are involved in vasoconstriction, hypertension, reduction in CRH, pro-inflammation and cardiac toxicity. Interestingly, the interactions of adenosine receptors (A2AAR, A1AR) with CYP450-epoxygenases, ω-hydroxylases, sEH, and their derived metabolites or oxygenated polyunsaturated fatty acids (PUFAs or oxylipins) is shown in the regulation of the cardiovascular functions. In addition, much evidence demonstrates polymorphisms in CYP450-epoxygenases, ω-hydroxylases, and sEH genes (Ephx2) and adenosine receptor genes (ADORA1 & ADORA2) in the human population with the susceptibility to CVDs, including hypertension. CVDs are the number one cause of death globally, coronary artery disease (CAD) was the leading cause of death in the US in 2019, and hypertension is one of the most potent causes of CVDs. This review summarizes the articles related to the crosstalk between adenosine receptors and CYP450-derived oxylipins in vascular, including the CRH response in regular salt-diet fed and high salt-diet fed mice with the correlation of heart perfusate/plasma oxylipins. By using A2AAR-/-, A1AR-/-, eNOS-/-, sEH-/- or Ephx2-/-, vascular sEH-overexpressed (Tie2-sEH Tr), vascular CYP2J2-overexpressed (Tie2-CYP2J2 Tr), and wild-type (WT) mice. This review article also summarizes the role of pro-and anti-inflammatory oxylipins in cardiovascular function/dysfunction in mice and humans. Therefore, more studies are needed better to understand the crosstalk between the adenosine receptors and eicosanoids to develop diagnostic and therapeutic tools by using plasma oxylipins profiles in CVDs, including hypertensive cases in the future.
Collapse
Affiliation(s)
- Mohammed A Nayeem
- Faculties of the Department of Pharmaceutical Sciences, School of Pharmacy, West Virginia University, Morgantown, WV, USA.
| | - Ahmad Hanif
- Faculties of the Department of Pharmaceutical Sciences, School of Pharmacy, West Virginia University, Morgantown, WV, USA
| | - Werner J Geldenhuys
- Faculties of the Department of Pharmaceutical Sciences, School of Pharmacy, West Virginia University, Morgantown, WV, USA
| | - Stephanie Agba
- Graduate student, Department of Pharmaceutical Sciences, School of Pharmacy, West Virginia University, Morgantown, WV, USA
| |
Collapse
|
4
|
Mustapha S, Azemi AK, Wan Ahmad WAN, Rasool AHG, Mustafa MR, Mokhtar SS. Inhibition of Endoplasmic Reticulum Stress Improves Acetylcholine-Mediated Relaxation in the Aorta of Type-2 Diabetic Rats. Molecules 2022; 27:5107. [PMID: 36014347 PMCID: PMC9413505 DOI: 10.3390/molecules27165107] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2022] [Revised: 08/05/2022] [Accepted: 08/08/2022] [Indexed: 12/02/2022] Open
Abstract
Endoplasmic reticulum (ER) stress contributes to insulin resistance and macro- and microvascular complications associated with diabetes. This study aimed to evaluate the effect of ER stress inhibition on endothelial function in the aorta of type-2 diabetic rats. Type-2 diabetes was developed in male Sprague-Dawley rats using a high-fat diet and low-dose streptozotocin. Rat aortic tissues were harvested to study endothelial-dependent relaxation. The mechanisms for acetylcholine-mediated relaxation were investigated using pharmacological blockers, Western blotting, oxidative stress, and inflammatory markers. Acetylcholine-mediated relaxation was diminished in the aorta of diabetic rats compared to control rats; supplementation with TUDCA improved relaxation. In the aortas of control and diabetic rats receiving TUDCA, the relaxation was mediated via eNOS/PI3K/Akt, NAD(P)H, and the KATP channel. In diabetic rats, acetylcholine-mediated relaxation involved eNOS/PI3K/Akt and NAD(P)H, but not the KATP channel. The expression of ER stress markers was upregulated in the aorta of diabetic rats and reduced with TUDCA supplementation. The expression of eNOS and Akt were lower in diabetic rats but were upregulated after supplementation with TUDCA. The levels of MDA, IL-6, and SOD activity were higher in the aorta of the diabetic rats compared to control rats. This study demonstrated that endothelial function was impaired in diabetes, however, supplementation with TUDCA improved the function via eNOS/Akt/PI3K, NAD(P)H, and the KATP channel. The improvement of endothelial function was associated with increased expressions of eNOS and Akt. Thus, ER stress plays a crucial role in the impairment of endothelial-dependent relaxation. Mitigating ER stress could be a potential strategy for improving endothelial dysfunction in type-2 diabetes.
Collapse
Affiliation(s)
- Sagir Mustapha
- Department of Pharmacology, School of Medical Sciences, Universiti Sains Malaysia, Kota Bharu 16150, Kelantan, Malaysia
- Department of Pharmacology and Therapeutics, Ahmadu Bello University, Zaria 810107, Kaduna, Nigeria
| | - Ahmad Khusairi Azemi
- Institute of Marine Biotechnology, Universiti Malaysia Terengganu, Kuala Terengganu 21030, Terengganu, Malaysia
| | - Wan Amir Nizam Wan Ahmad
- Biomedicine Programme, School of Health Sciences, Universiti Sains Malaysia, Kota Bharu 16150, Kelantan, Malaysia
| | - Aida Hanum Ghulam Rasool
- Department of Pharmacology, School of Medical Sciences, Universiti Sains Malaysia, Kota Bharu 16150, Kelantan, Malaysia
| | - Mohd Rais Mustafa
- Department of Pharmacology, Faculty of Medicine, University of Malaya, Kuala Lumpur 50603, Selangor, Malaysia
| | - Siti Safiah Mokhtar
- Department of Pharmacology, School of Medical Sciences, Universiti Sains Malaysia, Kota Bharu 16150, Kelantan, Malaysia
| |
Collapse
|
5
|
Zhang Y, Wernly B, Cao X, Mustafa SJ, Tang Y, Zhou Z. Adenosine and adenosine receptor-mediated action in coronary microcirculation. Basic Res Cardiol 2021; 116:22. [PMID: 33755785 PMCID: PMC7987637 DOI: 10.1007/s00395-021-00859-7] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/17/2020] [Accepted: 03/08/2021] [Indexed: 12/20/2022]
Abstract
Adenosine is an ubiquitous extracellular signaling molecule and plays a fundamental role in the regulation of coronary microcirculation through activation of adenosine receptors (ARs). Adenosine is regulated by various enzymes and nucleoside transporters for its balance between intra- and extracellular compartments. Adenosine-mediated coronary microvascular tone and reactive hyperemia are through receptors mainly involving A2AR activation on both endothelial and smooth muscle cells, but also involving interaction among other ARs. Activation of ARs further stimulates downstream targets of H2O2, KATP, KV and KCa2+ channels leading to coronary vasodilation. An altered adenosine-ARs signaling in coronary microcirculation has been observed in several cardiovascular diseases including hypertension, diabetes, atherosclerosis and ischemic heart disease. Adenosine as a metabolite and its receptors have been studied for its both therapeutic and diagnostic abilities. The present review summarizes important aspects of adenosine metabolism and AR-mediated actions in the coronary microcirculation.
Collapse
Affiliation(s)
- Ying Zhang
- The International Collaborative Centre On Big Science Plan for Purinergic Signalling, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Bernhard Wernly
- Department of Anaesthesiology, Perioperative Medicine and Intensive Care Medicine, Paracelsus Medical University of Salzburg, Salzburg, Austria
| | - Xin Cao
- The International Collaborative Centre On Big Science Plan for Purinergic Signalling, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - S Jamal Mustafa
- Department of Physiology and Pharmacology, West Virginia University, Morgantown, USA
| | - Yong Tang
- The International Collaborative Centre On Big Science Plan for Purinergic Signalling, Chengdu University of Traditional Chinese Medicine, Chengdu, China.,Acupuncture and Chronobiology Key Laboratory of Sichuan Province, Chengdu, China
| | - Zhichao Zhou
- Division of Cardiology, Department of Medicine, Karolinska Institutet, Karolinska University Hospital, 17176, Stockholm, Sweden.
| |
Collapse
|
6
|
Zhou R, Dang X, Sprague RS, Mustafa SJ, Zhou Z. Alteration of purinergic signaling in diabetes: Focus on vascular function. J Mol Cell Cardiol 2020; 140:1-9. [PMID: 32057736 DOI: 10.1016/j.yjmcc.2020.02.004] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/02/2019] [Revised: 01/02/2020] [Accepted: 02/09/2020] [Indexed: 12/11/2022]
Abstract
Diabetes is an important risk factor for the development of cardiovascular disease including atherosclerosis and ischemic heart disease. Vascular complications including macro- and micro-vascular dysfunction are the leading causes of morbidity and mortality in diabetes. Disease mechanisms at present are unclear and no ideal therapies are available, which urgently calls for the identification of novel therapeutic targets/agents. An altered nucleotide- and nucleoside-mediated purinergic signaling has been implicated to cause diabetes-associated vascular dysfunction in major organs. Alteration of both purinergic P1 and P2 receptor sensitivity rather than the changes in receptor expression accounts for vascular dysfunction in diabetes. Activation of P2X7 receptors plays a crucial role in diabetes-induced retinal microvascular dysfunction. Recent findings have revealed that both ecto-nucleotidase CD39, a key enzyme hydrolyzing ATP, and CD73, an enzyme regulating adenosine turnover, are involved in the renal vascular injury in diabetes. Interestingly, erythrocyte dysfunction in diabetes by decreasing ATP release in response to physiological stimuli may serve as an important trigger to induce vascular dysfunction. Nucleot(s)ide-mediated purinergic activation also exerts long-term actions including inflammatory and atherogenic effects in hyperglycemic and diabetic conditions. This review highlights the current knowledge regarding the altered nucleot(s)ide-mediated purinergic signaling as an important disease mechanism for the diabetes-associated vascular complications. Better understanding the role of key receptor-mediated signaling in diabetes will provide more insights into their potential as targets for the treatment.
Collapse
Affiliation(s)
- Rui Zhou
- Institute of Cardiovascular Research, The Key Laboratory of Medical Electrophysiology of Ministry of Education, Southwest Medical University, Luzhou, PR China
| | - Xitong Dang
- Institute of Cardiovascular Research, The Key Laboratory of Medical Electrophysiology of Ministry of Education, Southwest Medical University, Luzhou, PR China
| | - Randy S Sprague
- Department of Pharmacology and Physiology, Saint Louis University School of Medicine, St. Louis, MO, USA
| | - S Jamal Mustafa
- Department of Physiology and Pharmacology, West Virginia University, Morgantown, WV, USA
| | - Zhichao Zhou
- Division of Cardiology, Department of Medicine, Karolinska University Hospital, Karolinska Institutet, Stockholm, Sweden.
| |
Collapse
|
7
|
Sun C, Jiao T, Merkus D, Duncker DJ, Mustafa SJ, Zhou Z. Activation of adenosine A 2A but not A 2B receptors is involved in uridine adenosine tetraphosphate-induced porcine coronary smooth muscle relaxation. J Pharmacol Sci 2019; 141:64-69. [PMID: 31640919 PMCID: PMC7418061 DOI: 10.1016/j.jphs.2019.09.006] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2019] [Revised: 08/05/2019] [Accepted: 08/07/2019] [Indexed: 12/18/2022] Open
Abstract
Activation of both adenosine A2A and A2B receptors (A2BR) contributes to coronary vasodilation. We previously demonstrated that uridine adenosine tetraphosphate (Up4A) is a novel vasodilator in the porcine coronary microcirculation, acting mainly on A2AR in smooth muscle cells (SMC). We further investigated whether activation of A2BR is involved in Up4A-mediated coronary SMC relaxation. Both A2AR and A2BR may stimulate H2O2 production leading to activation of KATP channels in SMCs, we also studied the involvement of H2O2 and KATP channels in Up4A-mediated effect. Coronary small arteries dissected from the apex of porcine hearts were mounted on wire myograph for Up4A concentration responses. Up4A-induced coronary SMC relaxation was attenuated by A2AR but not A2BR antagonism or non-selective P2R antagonism, despite greater endogenous A2BR expression vs. A2AR in both coronary small arteries and primary cultured coronary SMCs. Moreover, Up4A-induced coronary SMC relaxation was blunted by H2O2 catabolism. This effect was not altered by KATP channel blockade. Combination of H2O2 catabolism and A2AR antagonism attenuated Up4A-induced coronary SMC relaxation to the similar extent as A2AR antagonism alone. Collectively, Up4A-induced porcine coronary SMC relaxation is mediated by activation of A2AR-H2O2 pathway. This process does not involve A2BR, P2R or KATP channels.
Collapse
Affiliation(s)
- Changyan Sun
- Department of Physiology and Pharmacology, West Virginia University, Morgantown, WV, USA
| | - Tong Jiao
- Division of Cardiology, Department of Medicine, Karolinska University Hospital, Karolinska Institutet, Stockholm, Sweden
| | - Daphne Merkus
- Division of Experimental Cardiology, Department of Cardiology, Erasmus University Medical Center, Rotterdam, the Netherlands; Walter-Brendel-Centre of Experimental Medicine, University Hostpital, LMU Munich, Munich, Germany
| | - Dirk J Duncker
- Division of Experimental Cardiology, Department of Cardiology, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - S Jamal Mustafa
- Department of Physiology and Pharmacology, West Virginia University, Morgantown, WV, USA
| | - Zhichao Zhou
- Department of Physiology and Pharmacology, West Virginia University, Morgantown, WV, USA; Division of Cardiology, Department of Medicine, Karolinska University Hospital, Karolinska Institutet, Stockholm, Sweden.
| |
Collapse
|
8
|
Yadav VR, Teng B, Mustafa SJ. Enhanced A 1 adenosine receptor-induced vascular contractions in mesenteric artery and aorta of in L-NAME mouse model of hypertension. Eur J Pharmacol 2018; 842:111-117. [PMID: 30347181 DOI: 10.1016/j.ejphar.2018.10.024] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2018] [Revised: 10/13/2018] [Accepted: 10/17/2018] [Indexed: 01/28/2023]
Abstract
L-NAME-induced hypertension is commonly used to study endothelial dysfunction and related vascular effects. It has been reported that genetic deletion of A1 adenosine receptor (AR) reduces blood pressure (BP) increases in mice and thus, suggesting the involvement of A1AR. Thus, we sought to determine whether A1AR-induced vascular responses were altered in this mouse model of hypertension. L-NAME (1 mg/ml) was given in the drinking water for 28 days to mice. The BP was monitored using non-invasive tail-cuff system. Muscle tension studies were performed using DMT for mesenteric arteries (MAs) and organ bath for aorta. Protein expression was analyzed by western blot. Significantly, higher systolic and mean arterial blood pressure was noted in L-NAME mice. In MAs, higher 2-Chloro-N6-cyclopentyladenosine (CCPA, selective A1AR agonist) induced contractions in hypertensive mice were observed. This enhanced contraction was inhibited by HET0016 (Cytochrome 450 4A inhibitor, 10 µM, 15 min). Contrary, 5'-(N-Ethylcarboxamido) adenosine (NECA, non-selective AR agonist) induced vascular responses were comparable in both groups. Pinacidil (KATP channel opener) induced relaxation was significantly increased in hypertensive mice. In aorta, CCPA-induced contractions were enhanced and inhibited by HET0016 in hypertensive mice. Notably, NECA-induced contractions in aorta were enhanced in hypertensive mice. Higher expressions of A1AR and Cyp4A were noted in MAs of hypertensive mice. In addition, in aorta, higher A1AR and comparable Cyp4A levels were observed in hypertensive mice. A1AR-induced vascular contractions were enhanced in hypertensive mice aorta and MAs. Cyp4A plays a role in altered vascular responses in MAs.
Collapse
Affiliation(s)
- Vishal R Yadav
- Department of Physiology and Pharmacology, West Virginia University, Morgantown, WV, USA
| | - Bunyen Teng
- Coagulation and Blood Research Task Area, US Army Institute of Surgical Research, San Antonio, TX, USA
| | - S Jamal Mustafa
- Department of Physiology and Pharmacology, West Virginia University, Morgantown, WV, USA; Center for Translational Science Institute, West Virginia University, Morgantown, WV, USA.
| |
Collapse
|
9
|
Ohanyan V, Yin L, Bardakjian R, Kolz C, Enrick M, Hakobyan T, Luli J, Graham K, Khayata M, Logan S, Kmetz J, Chilian WM. Kv1.3 channels facilitate the connection between metabolism and blood flow in the heart. Microcirculation 2017; 24:10.1111/micc.12334. [PMID: 28504408 PMCID: PMC5433265 DOI: 10.1111/micc.12334] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2016] [Revised: 10/23/2016] [Accepted: 11/01/2016] [Indexed: 12/17/2022]
Abstract
The connection between metabolism and flow in the heart, metabolic dilation, is essential for cardiac function. We recently found redox-sensitive Kv1.5 channels play a role in coronary metabolic dilation; however, more than one ion channel likely plays a role in this process as animals null for these channels still showed limited coronary metabolic dilation. Accordingly, we examined the role of another Kv1 family channel, the energetically linked Kv1.3 channel, in coronary metabolic dilation. We measured myocardial blood flow (contrast echocardiography) during norepinephrine-induced increases in cardiac work (heart rate x mean arterial pressure) in WT, WT mice given correolide (preferential Kv1.3 antagonist), and Kv1.3-null mice (Kv1.3-/- ). We also measured relaxation of isolated small arteries mounted in a myograph. During increased cardiac work, myocardial blood flow was attenuated in Kv1.3-/- and in correolide-treated mice. In isolated vessels from Kv1.3-/- mice, relaxation to H2 O2 was impaired (vs WT), but responses to adenosine and acetylcholine were equivalent to WT. Correolide reduced dilation to adenosine and acetylcholine in WT and Kv1.3-/- , but had no effect on H2 O2 -dependent dilation in vessels from Kv1.3-/- mice. We conclude that Kv1.3 channels participate in the connection between myocardial blood flow and cardiac metabolism.
Collapse
Affiliation(s)
- Vahagn Ohanyan
- Department of Integrative Medical Sciences, Northeast Ohio Medical University, Rootstown, OH, USA
| | - Liya Yin
- Department of Integrative Medical Sciences, Northeast Ohio Medical University, Rootstown, OH, USA
| | | | - Christopher Kolz
- Department of Integrative Medical Sciences, Northeast Ohio Medical University, Rootstown, OH, USA
| | - Molly Enrick
- Department of Integrative Medical Sciences, Northeast Ohio Medical University, Rootstown, OH, USA
| | - Tatevik Hakobyan
- Department of Integrative Medical Sciences, Northeast Ohio Medical University, Rootstown, OH, USA
| | - Jordan Luli
- Department of Integrative Medical Sciences, Northeast Ohio Medical University, Rootstown, OH, USA
| | - Kathleen Graham
- Department of Integrative Medical Sciences, Northeast Ohio Medical University, Rootstown, OH, USA
| | | | - Suzanna Logan
- Department of Integrative Medical Sciences, Northeast Ohio Medical University, Rootstown, OH, USA
| | - John Kmetz
- Department of Integrative Medical Sciences, Northeast Ohio Medical University, Rootstown, OH, USA
| | - William M Chilian
- Department of Integrative Medical Sciences, Northeast Ohio Medical University, Rootstown, OH, USA
| |
Collapse
|
10
|
Tykocki NR, Boerman EM, Jackson WF. Smooth Muscle Ion Channels and Regulation of Vascular Tone in Resistance Arteries and Arterioles. Compr Physiol 2017; 7:485-581. [PMID: 28333380 DOI: 10.1002/cphy.c160011] [Citation(s) in RCA: 236] [Impact Index Per Article: 29.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Vascular tone of resistance arteries and arterioles determines peripheral vascular resistance, contributing to the regulation of blood pressure and blood flow to, and within the body's tissues and organs. Ion channels in the plasma membrane and endoplasmic reticulum of vascular smooth muscle cells (SMCs) in these blood vessels importantly contribute to the regulation of intracellular Ca2+ concentration, the primary determinant of SMC contractile activity and vascular tone. Ion channels provide the main source of activator Ca2+ that determines vascular tone, and strongly contribute to setting and regulating membrane potential, which, in turn, regulates the open-state-probability of voltage gated Ca2+ channels (VGCCs), the primary source of Ca2+ in resistance artery and arteriolar SMCs. Ion channel function is also modulated by vasoconstrictors and vasodilators, contributing to all aspects of the regulation of vascular tone. This review will focus on the physiology of VGCCs, voltage-gated K+ (KV) channels, large-conductance Ca2+-activated K+ (BKCa) channels, strong-inward-rectifier K+ (KIR) channels, ATP-sensitive K+ (KATP) channels, ryanodine receptors (RyRs), inositol 1,4,5-trisphosphate receptors (IP3Rs), and a variety of transient receptor potential (TRP) channels that contribute to pressure-induced myogenic tone in resistance arteries and arterioles, the modulation of the function of these ion channels by vasoconstrictors and vasodilators, their role in the functional regulation of tissue blood flow and their dysfunction in diseases such as hypertension, obesity, and diabetes. © 2017 American Physiological Society. Compr Physiol 7:485-581, 2017.
Collapse
Affiliation(s)
- Nathan R Tykocki
- Department of Pharmacology, University of Vermont, Burlington, Vermont, USA
| | - Erika M Boerman
- Department of Medical Pharmacology and Physiology, University of Missouri, Columbia, Missouri, USA
| | - William F Jackson
- Department of Pharmacology and Toxicology, Michigan State University, East Lansing, Michigan, USA
| |
Collapse
|
11
|
Inosine attenuates spontaneous activity in the rat neurogenic bladder through an A 2B pathway. Sci Rep 2017; 7:44416. [PMID: 28294142 PMCID: PMC5353659 DOI: 10.1038/srep44416] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2016] [Accepted: 02/07/2017] [Indexed: 01/16/2023] Open
Abstract
Neurogenic detrusor overactivity (NDO) is among the most challenging complications of spinal cord injury (SCI). A recent report by us demonstrated an improvement in NDO in SCI rats following chronic systemic treatment with the purine nucleoside inosine. The objective of this study was to investigate the mechanism of action of inosine underlying improvement of NDO. Male Sprague-Dawley rats underwent complete spinal cord transection at T8. Inosine (1 mM) delivered intravesically to SCI rats during conscious cystometry significantly decreased the frequency of spontaneous non-voiding contractions. In isolated tissue assays, inosine (1 mM) significantly decreased the amplitude of spontaneous activity (SA) in SCI bladder muscle strips. This effect was prevented by a pan-adenosine receptor antagonist CGS15943, but not by A1 or A3 receptor antagonists. The A2A antagonist ZM241385 and A2B antagonist PSB603 prevented the effect of inosine. The effect of inosine was mimicked by the adenosine receptor agonist NECA and the A2B receptor agonist BAY60-6583. The inhibition of SA by inosine was not observed in the presence of the BK antagonist, iberiotoxin, but persisted in the presence of KATP and SK antagonists. These findings demonstrate that inosine acts via an A2B receptor-mediated pathway that impinges on specific potassium channel effectors.
Collapse
|
12
|
Effect of Soluble Epoxide Hydrolase on the Modulation of Coronary Reactive Hyperemia: Role of Oxylipins and PPARγ. PLoS One 2016; 11:e0162147. [PMID: 27583776 PMCID: PMC5008628 DOI: 10.1371/journal.pone.0162147] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2016] [Accepted: 08/17/2016] [Indexed: 11/19/2022] Open
Abstract
Coronary reactive hyperemia (CRH) is a physiological response to ischemic insult that prevents the potential harm associated with an interruption of blood supply. The relationship between the pharmacologic inhibition of soluble epoxide hydrolase (sEH) and CRH response to a brief ischemia is not known. sEH is involved in the main catabolic pathway of epoxyeicosatrienoic acids (EETs), which are converted into dihydroxyeicosatrienoic acids (DHETs). EETs protect against ischemia/reperfusion injury and have numerous beneficial physiological effects. We hypothesized that inhibition of sEH by t-AUCB enhances CRH in isolated mouse hearts through changing the oxylipin profiles, including an increase in EETs/DHETs ratio. Compared to controls, t-AUCB-treated mice had increased CRH, including repayment volume (RV), repayment duration, and repayment/debt ratio (p < 0.05). Treatment with t-AUCB significantly changed oxylipin profiles, including an increase in EET/DHET ratio, increase in EpOME/DiHOME ratio, increase in the levels of HODEs, decrease in the levels of mid-chain HETEs, and decrease in prostanoids (p < 0.05). Treatment with MS-PPOH (CYP epoxygenase inhibitor) reduced CRH, including RV (p < 0.05). Involvement of PPARγ in the modulation of CRH was demonstrated using a PPARγ-antagonist (T0070907) and a PPARγ-agonist (rosiglitazone). T0070907 reduced CRH (p < 0.05), whereas rosiglitazone enhanced CRH (p < 0.05) in isolated mouse hearts compared to the non-treated. These data demonstrate that sEH inhibition enhances, whereas CYP epoxygenases-inhibition attenuates CRH, PPARγ mediate CRH downstream of the CYP epoxygenases-EET pathway, and the changes in oxylipin profiles associated with sEH-inhibition collectively contributed to the enhanced CRH.
Collapse
|
13
|
Potassium Channels in Regulation of Vascular Smooth Muscle Contraction and Growth. ADVANCES IN PHARMACOLOGY (SAN DIEGO, CALIF.) 2016; 78:89-144. [PMID: 28212804 DOI: 10.1016/bs.apha.2016.07.001] [Citation(s) in RCA: 88] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Potassium channels importantly contribute to the regulation of vascular smooth muscle (VSM) contraction and growth. They are the dominant ion conductance of the VSM cell membrane and importantly determine and regulate membrane potential. Membrane potential, in turn, regulates the open-state probability of voltage-gated Ca2+ channels (VGCC), Ca2+ influx through VGCC, intracellular Ca2+, and VSM contraction. Membrane potential also affects release of Ca2+ from internal stores and the Ca2+ sensitivity of the contractile machinery such that K+ channels participate in all aspects of regulation of VSM contraction. Potassium channels also regulate proliferation of VSM cells through membrane potential-dependent and membrane potential-independent mechanisms. VSM cells express multiple isoforms of at least five classes of K+ channels that contribute to the regulation of contraction and cell proliferation (growth). This review will examine the structure, expression, and function of large conductance, Ca2+-activated K+ (BKCa) channels, intermediate-conductance Ca2+-activated K+ (KCa3.1) channels, multiple isoforms of voltage-gated K+ (KV) channels, ATP-sensitive K+ (KATP) channels, and inward-rectifier K+ (KIR) channels in both contractile and proliferating VSM cells.
Collapse
|
14
|
Hanif A, Edin ML, Zeldin DC, Morisseau C, Nayeem MA. Deletion of soluble epoxide hydrolase enhances coronary reactive hyperemia in isolated mouse heart: role of oxylipins and PPARγ. Am J Physiol Regul Integr Comp Physiol 2016; 311:R676-R688. [PMID: 27488890 DOI: 10.1152/ajpregu.00237.2016] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2016] [Accepted: 07/28/2016] [Indexed: 11/22/2022]
Abstract
The relationship between soluble epoxide hydrolase (sEH) and coronary reactive hyperemia (CRH) response to a brief ischemic insult is not known. Epoxyeicosatrienoic acids (EETs) exert cardioprotective effects in ischemia/reperfusion injury. sEH converts EETs into dihydroxyeicosatrienoic-acids (DHETs). Therefore, we hypothesized that knocking out sEH enhances CRH through modulation of oxylipin profiles, including an increase in EET/DHET ratio. Compared with sEH+/+, sEH-/- mice showed enhanced CRH, including greater repayment volume (RV; 28% higher, P < 0.001) and repayment/debt ratio (32% higher, P < 0.001). Oxylipins from the heart perfusates were analyzed by LC-MS/MS. The 14,15-EET/14,15-DHET ratio was 3.7-fold higher at baseline (P < 0.001) and 5.6-fold higher post-ischemia (P < 0.001) in sEH-/- compared with sEH+/+ mice. Likewise, the baseline 9,10- and 12,13-EpOME/DiHOME ratios were 3.2-fold (P < 0.01) and 3.7-fold (P < 0.001) higher, respectively in sEH-/- compared with sEH+/+ mice. 13-HODE was also significantly increased at baseline by 71% (P < 0.01) in sEH-/- vs. sEH+/+ mice. Levels of 5-, 11-, 12-, and 15-hydroxyeicosatetraenoic acids were not significantly different between the two strains (P > 0.05), but were decreased postischemia in both groups (P = 0.02, P = 0.04, P = 0.05, P = 0.03, respectively). Modulation of CRH by peroxisome proliferator-activated receptor gamma (PPARγ) was demonstrated using a PPARγ-antagonist (T0070907), which reduced repayment volume by 25% in sEH+/+ (P < 0.001) and 33% in sEH-/- mice (P < 0.01), and a PPARγ-agonist (rosiglitazone), which increased repayment volume by 37% in both sEH+/+ (P = 0.04) and sEH-/- mice (P = 0.04). l-NAME attenuated CRH in both sEH-/- and sEH+/+ These data demonstrate that genetic deletion of sEH resulted in an altered oxylipin profile, which may have led to an enhanced CRH response.
Collapse
Affiliation(s)
- Ahmad Hanif
- Basic Pharmaceutical Sciences, School of Pharmacy, Center for Basic and Translational Stroke Research, West Virginia University, Morgantown, West Virginia
| | - Matthew L Edin
- Division of Intramural Research, National Institute of Environmental Health Sciences/National Institutes of Health, Research Triangle Park, North Carolina; and
| | - Darryl C Zeldin
- Division of Intramural Research, National Institute of Environmental Health Sciences/National Institutes of Health, Research Triangle Park, North Carolina; and
| | | | - Mohammed A Nayeem
- Basic Pharmaceutical Sciences, School of Pharmacy, Center for Basic and Translational Stroke Research, West Virginia University, Morgantown, West Virginia;
| |
Collapse
|
15
|
Zhou X, Teng B, Mustafa SJ. Sex Difference in Coronary Endothelial Dysfunction in Apolipoprotein E Knockout Mouse: Role of NO and A2A Adenosine Receptor. Microcirculation 2016. [PMID: 26201383 DOI: 10.1111/micc.12222] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
OBJECTIVE Sex plays an important role in the pathophysiology of cardiovascular diseases. This study aims to investigate how sex impacts on the coronary flow regulation during atherosclerosis. METHODS ApoE KO mouse fed with western diet were used for atherosclerosis model. Coronary RH and flow response were measured using Langendorff-perfused isolated hearts. RESULTS Coronary RH and A23187-induced NO-dependent flow increases were significantly reduced in female (by ~28% and 48%, respectively), but not in male atherosclerotic mice. However, SNP-induced coronary vasodilation remains intact in both sexes of ApoE KO mice. L-NAME (NOS inhibitor) reduced baseline flow and RH to a lesser extent in ApoE KO (by ~19% and 31%) vs. WT (~30% and 59%, respectively), and abolished the sex difference in RH. In contrast, SCH58261 (a selective A2A AR antagonist) reduced the baseline flow and RH to a greater extent in atherosclerotic mice, but did not affect the sex difference. Immunofluorescent staining of coronary arteries showed a similar A2A AR upregulation in both sexes of ApoE KO mice. CONCLUSIONS Our results suggest that during atherosclerosis, female mice are more susceptible to NO-dependent endothelial dysfunction and the upregulation of A2A AR may serve as a compensatory mechanism to counteract the compromised endothelial function.
Collapse
Affiliation(s)
- Xueping Zhou
- Department of Physiology and Pharmacology, West Virginia University, Morgantown, West Virginia, USA.,Center for Cardiovascular and Respiratory Sciences and West Virginia Clinical & Translational Science Institute, Morgantown, West Virginia, USA
| | - Bunyen Teng
- Department of Physiology and Pharmacology, West Virginia University, Morgantown, West Virginia, USA.,Center for Cardiovascular and Respiratory Sciences and West Virginia Clinical & Translational Science Institute, Morgantown, West Virginia, USA
| | - S J Mustafa
- Department of Physiology and Pharmacology, West Virginia University, Morgantown, West Virginia, USA.,Center for Cardiovascular and Respiratory Sciences and West Virginia Clinical & Translational Science Institute, Morgantown, West Virginia, USA
| |
Collapse
|
16
|
Arsyad A, Dobson GP. Adenosine relaxation in isolated rat aortic rings and possible roles of smooth muscle Kv channels, KATP channels and A2a receptors. BMC Pharmacol Toxicol 2016; 17:23. [PMID: 27211886 PMCID: PMC4876563 DOI: 10.1186/s40360-016-0067-8] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2016] [Accepted: 04/29/2016] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND An area of ongoing controversy is the role adenosine to regulate vascular tone in conduit vessels that regulate compliance, and the role of nitric oxide (NO), potassium channels and receptor subtypes involved. The aim of our study was to investigate adenosine relaxation in rat thoracic aortic rings, and the effect of inhibitors of NO, prostanoids, Kv, KATP channels, and A2a and A2b receptors. METHODS Aortic rings were freshly harvested from adult male Sprague Dawley rats and equilibrated in an organ bath containing oxygenated, modified Krebs-Henseleit solution, 11 mM glucose, pH 7.4, 37 °C. Isolated rings were pre-contracted sub-maximally with 0.3 μM norepinephrine (NE), and the effect of increasing concentrations of adenosine (1 to 1000 μM) were examined. The drugs L-NAME, indomethacin, 4-aminopyridine (4-AP), glibenclamide, 5-hydroxydecanoate, ouabain, 8-(3-chlorostyryl) caffeine and PSB-0788 were examined in intact and denuded rings. Rings were tested for viability after each experiment. RESULTS Adenosine induced a dose-dependent, triphasic relaxation response, and the mechanical removal of the endothelium significantly deceased adenosine relaxation above 10 μM. Interestingly, endothelial removal significantly decreased the responsiveness (defined as % relaxation per μM adenosine) by two-thirds between 10 and 100 μM, but not in the lower (1-10 μM) or higher (>100 μM) ranges. In intact rings, L-NAME significantly reduced relaxation, but not indomethacin. Antagonists of voltage-dependent Kv (4-AP), sarcolemma KATP (glibenclamide) and mitochondrial KATP channels (5-HD) led to significant reductions in relaxation in both intact and denuded rings, with ouabain having little or no effect. Adenosine-induced relaxation appeared to involve the A2a receptor, but not the A2b subtype. CONCLUSIONS It was concluded that adenosine relaxation in NE-precontracted rat aortic rings was triphasic and endothelium-dependent above 10 μM, and relaxation involved endothelial nitric oxide (not prostanoids) and a complex interplay between smooth muscle A2a subtype and voltage-dependent Kv, SarcKATP and MitoKATP channels. The possible in vivo significance of the regulation of arterial compliance to left ventricular function coupling is discussed.
Collapse
Affiliation(s)
- Aryadi Arsyad
- Physiology Department, Medical Faculty, Hasanuddin University, Jl. Perintis Kemerdekaan, Km. 10, Tamalanrea, Makassar, 90213, Indonesia
| | - Geoffrey P Dobson
- Heart, Trauma and Sepsis Research Laboratory, Australian Institute of Tropical Health and Medicine, College of Medicine and Dentistry, James Cook University, 1 James Cook Drive, Queensland, 4811, Australia.
| |
Collapse
|
17
|
Labazi H, Teng B, Zhou Z, Mustafa SJ. Enhanced A2A adenosine receptor-mediated increase in coronary flow in type I diabetic mice. J Mol Cell Cardiol 2016; 90:30-7. [PMID: 26654777 PMCID: PMC4729563 DOI: 10.1016/j.yjmcc.2015.11.033] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/17/2015] [Revised: 11/09/2015] [Accepted: 11/30/2015] [Indexed: 02/08/2023]
Abstract
Adenosine A2A receptor (A2AAR) activation plays a major role in the regulation of coronary flow (CF). Recent studies from our laboratory and others have suggested that A2AAR expression and/or signaling is altered in disease conditions. However, the coronary response to AR activation, in particular A2AAR, in diabetes is not fully understood. In this study, we use an STZ mouse model of type 1 diabetes (T1D) to look at CF responses to the nonspecific AR agonist NECA and the A2AAR specific agonist CGS 21680 in-vivo and ex-vivo. Using immunofluorescence, we also explored the effect of diabetes on A2AAR expression in coronary arteries. NECA mediated increase in CF was significantly increased in hearts isolated from STZ-induced diabetic mice. In addition, both in in-vivo and ex-vivo responses to A2AAR activation using CGS 21680 were significantly higher in diabetic mice when compared to their controls. Immunohistochemistry showed an upregulation of A2AAR in both coronary smooth muscle and endothelial cells (~160% and ~140%, respectively). Our data suggest that diabetes resulted in an increased A2AAR expression in coronary arteries which resulted in enhanced A2AAR-mediated increase in CF observed in diabetic hearts. This is the first report implying that A2AAR has a role in the regulation of CF in diabetes, supporting recent studies suggesting that the use of adenosine and its A2A selective agonist (regadenoson, Lexiscan®) may not be appropriate for the detection of coronary artery diseases in T1D and the estimation of coronary reserve.
Collapse
MESH Headings
- Adenosine/analogs & derivatives
- Adenosine/pharmacology
- Adenosine A2 Receptor Agonists/pharmacology
- Adenosine-5'-(N-ethylcarboxamide)/pharmacology
- Animals
- Coronary Circulation/drug effects
- Coronary Vessels/drug effects
- Coronary Vessels/metabolism
- Coronary Vessels/pathology
- Diabetes Mellitus, Experimental/genetics
- Diabetes Mellitus, Experimental/metabolism
- Diabetes Mellitus, Experimental/pathology
- Diabetes Mellitus, Type 1/genetics
- Diabetes Mellitus, Type 1/metabolism
- Diabetes Mellitus, Type 1/pathology
- Endothelial Cells/drug effects
- Endothelial Cells/metabolism
- Endothelial Cells/pathology
- Gene Expression Regulation
- Heart/drug effects
- Heart/physiopathology
- Humans
- Male
- Mice
- Muscle, Smooth, Vascular/drug effects
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/pathology
- Organ Culture Techniques
- Phenethylamines/pharmacology
- Receptor, Adenosine A2A/genetics
- Receptor, Adenosine A2A/metabolism
- Receptor, Adenosine A2B/genetics
- Receptor, Adenosine A2B/metabolism
- Signal Transduction
- Vasodilation/drug effects
- Vasodilator Agents/pharmacology
Collapse
Affiliation(s)
- Hicham Labazi
- Department of Physiology and Pharmacology, Center for Cardiovascular and Respiratory Sciences and Clinical Translational Science Institute, West Virginia University, Morgantown, WV, United States
| | - Bunyen Teng
- Department of Physiology and Pharmacology, Center for Cardiovascular and Respiratory Sciences and Clinical Translational Science Institute, West Virginia University, Morgantown, WV, United States
| | - Zhichao Zhou
- Department of Physiology and Pharmacology, Center for Cardiovascular and Respiratory Sciences and Clinical Translational Science Institute, West Virginia University, Morgantown, WV, United States
| | - S Jamal Mustafa
- Department of Physiology and Pharmacology, Center for Cardiovascular and Respiratory Sciences and Clinical Translational Science Institute, West Virginia University, Morgantown, WV, United States.
| |
Collapse
|
18
|
Yadav VR, Nayeem MA, Tilley SL, Mustafa SJ. Angiotensin II stimulation alters vasomotor response to adenosine in mouse mesenteric artery: role for A1 and A2B adenosine receptors. Br J Pharmacol 2015; 172:4959-69. [PMID: 26227882 DOI: 10.1111/bph.13265] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2015] [Revised: 07/16/2015] [Accepted: 07/26/2015] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND AND PURPOSE Stimulation of the A1 adenosine receptor and angiotensin II receptor type-1 (AT1 receptor) causes vasoconstriction through activation of cytochrome P450 4A (CYP4A) and ERK1/2. Thus, we hypothesized that acute angiotensin II activation alters the vasomotor response induced by the non-selective adenosine receptor agonist, NECA, in mouse mesenteric arteries (MAs). EXPERIMENTAL APPROACH We used a Danish Myo Technology wire myograph to measure muscle tension in isolated MAs from wild type (WT), A1 receptor and A2B receptor knockout (KO) mice. Western blots were performed to determine the expression of AT1 receptors and CYP4A. KEY RESULTS Acute exposure (15 min) to angiotensin II attenuated the NECA-dependent vasodilatation and enhanced vasoconstriction. This vasoconstrictor effect of angiotensin II in NECA-treated MAs was abolished in A1 receptor KO mice and in WT mice treated with the A1 receptor antagonist DPCPX, CYP4A inhibitor HET0016 and ERK1/2 inhibitor PD98059. In MAs from A2B receptor KO mice, the vasoconstrictor effect of angiotensin II on the NECA-induced response was shown to be dependent on A1 receptors. Furthermore, in A2B receptor KO mice, the expression of AT1 receptors and CYP4A was increased and the angiotensin II-induced vasoconstriction enhanced. In addition, inhibition of KATP channels with glibenclamide significantly reduced NECA-induced vasodilatation in WT mice. CONCLUSIONS AND IMPLICATIONS Acute angiotensin II stimulation enhanced A1 receptor-dependent vasoconstriction and inhibited A2B receptor-dependent vasodilatation, leading to a net vasoconstriction and altered vasomotor response to NECA in MAs. This interaction may be important in the regulation of BP.
Collapse
Affiliation(s)
- Vishal R Yadav
- Department of Physiology and Pharmacology, School of Medicine, Morgantown, WV, USA
| | - Mohammed A Nayeem
- Department of Pharmaceutical Sciences, School of Pharmacy, West Virginia University, Morgantown, WV, USA
| | - Stephen L Tilley
- Department of Medicine, University of North Carolina, Chapel Hill, NC, USA
| | - S Jamal Mustafa
- Department of Physiology and Pharmacology, School of Medicine, Morgantown, WV, USA.,West Virginia Center for Translational Science Institute, Morgantown, WV, USA
| |
Collapse
|
19
|
Zhou Z, Rajamani U, Labazi H, Tilley SL, Ledent C, Teng B, Mustafa SJ. Involvement of NADPH oxidase in A2A adenosine receptor-mediated increase in coronary flow in isolated mouse hearts. Purinergic Signal 2015; 11:263-73. [DOI: 10.1007/s11302-015-9451-x] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2015] [Accepted: 04/16/2015] [Indexed: 12/22/2022] Open
|
20
|
Zhou X, Teng B, Tilley S, Ledent C, Mustafa SJ. Metabolic hyperemia requires ATP-sensitive K+ channels and H2O2 but not adenosine in isolated mouse hearts. Am J Physiol Heart Circ Physiol 2014; 307:H1046-55. [PMID: 25108010 DOI: 10.1152/ajpheart.00421.2014] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
We have previously demonstrated that adenosine-mediated H2O2 production and opening of ATP-sensitive K(+) (KATP) channels contributes to coronary reactive hyperemia. The present study aimed to investigate the roles of adenosine, H2O2, and KATP channels in coronary metabolic hyperemia (MH). Experiments were conducted on isolated Langendorff-perfused mouse hearts using combined pharmacological approaches with adenosine receptor (AR) knockout mice. MH was induced by electrical pacing at graded frequencies. Coronary flow increased linearly from 14.4 ± 1.2 to 20.6 ± 1.2 ml·min(-1)·g(-1) with an increase in heart rate from 400 to 650 beats/min in wild-type mice. Neither non-selective blockade of ARs by 8-(p-sulfophenyl)theophylline (8-SPT; 50 μM) nor selective A2AAR blockade by SCH-58261 (1 μM) or deletion affected MH, although resting flow and left ventricular developed pressure were reduced. Combined A2AAR and A2BAR blockade or deletion showed similar effects as 8-SPT. Inhibition of nitric oxide synthesis by N-nitro-l-arginine methyl ester (100 μM) or combined 8-SPT administration failed to reduce MH, although resting flows were reduced (by ∼20%). However, glibenclamide (KATP channel blocker, 5 μM) decreased not only resting flow (by ∼45%) and left ventricular developed pressure (by ∼36%) but also markedly reduced MH by ∼94%, resulting in cardiac contractile dysfunction. Scavenging of H2O2 by catalase (2,500 U/min) also decreased resting flow (by ∼16%) and MH (by ∼24%) but to a lesser extent than glibenclamide. Our results suggest that while adenosine modulates coronary flow under both resting and ischemic conditions, it is not required for MH. However, H2O2 and KATP channels are important local control mechanisms responsible for both coronary ischemic and metabolic vasodilation.
Collapse
Affiliation(s)
- Xueping Zhou
- Department of Physiology and Pharmacology, West Virginia University, Morgantown, West Virginia; Center for Cardiovascular and Respiratory Sciences, West Virginia University, Morgantown, West Virginia
| | - Bunyen Teng
- Department of Physiology and Pharmacology, West Virginia University, Morgantown, West Virginia; Center for Cardiovascular and Respiratory Sciences, West Virginia University, Morgantown, West Virginia
| | - Stephen Tilley
- Department of Medicine, University of North Carolina, Chapel Hill, North Carolina; and
| | | | - S Jamal Mustafa
- Department of Physiology and Pharmacology, West Virginia University, Morgantown, West Virginia; Center for Cardiovascular and Respiratory Sciences, West Virginia University, Morgantown, West Virginia;
| |
Collapse
|
21
|
Matsumoto N, Nagao K, Hirayama A, Kasama S. Adenosine-induced coronary vasospasm following drug-eluting stent implantation. BMJ Case Rep 2014; 2014:bcr-2013-202996. [PMID: 24518394 DOI: 10.1136/bcr-2013-202996] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
Abstract
We present the case of coronary vasospasm during adenosine stress in a patient with a prior drug-eluting stent implantation. The patient had a stent implantation in the left anterior descending coronary artery 3 years ago. Recently, he developed a chest pain and underwent adenosine stress myocardial perfusion single photon emission CT (SPECT). During the adenosine stress, he felt severe chest pain and ST elevation on electrocardiogram. An invasive coronary angiography showed no in-stent restenosis. This phenomenon deemed to be adenosine-induced coronary vasospasm after stent implantation.
Collapse
Affiliation(s)
- Naoya Matsumoto
- Department of Cardiology, Nihon University Surugadai Hospital, Tokyo, Japan
| | | | | | | |
Collapse
|
22
|
Echavarría-Pinto M, Gonzalo N, Ibañez B, Petraco R, Jimenez-Quevedo P, Sen S, Nijjer S, Tarkin J, Alfonso F, Núñez-Gil IJ, Bañuelos C, Quirós A, Fernández-Ortiz A, Macaya C, Koo BK, Davies J, Escaned J. Low Coronary Microcirculatory Resistance Associated With Profound Hypotension During Intravenous Adenosine Infusion. Circ Cardiovasc Interv 2014; 7:35-42. [DOI: 10.1161/circinterventions.113.000659] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Affiliation(s)
- Mauro Echavarría-Pinto
- From the Cardiovascular Institute, Hospital Clínico San Carlos and Centro Nacional de Investigaciones Cardiovasculares Carlos III, Madrid, Spain (M.E.-P., B.I., A.Q., A.F.-O., J.E.); Cardiovascular Institute, Hospital Clínico San Carlos, Madrid, Spain (N.G., P.J.-Q., F.A., I.J.N.-G., C.B., C.M.); International Centre for Circulatory Health, National Heart and Lung Institute, Imperial College London and Imperial College Healthcare NHS Trust, London, United Kingdom (R.P., S.S., S.N., J.T., J.D.); and
| | - Nieves Gonzalo
- From the Cardiovascular Institute, Hospital Clínico San Carlos and Centro Nacional de Investigaciones Cardiovasculares Carlos III, Madrid, Spain (M.E.-P., B.I., A.Q., A.F.-O., J.E.); Cardiovascular Institute, Hospital Clínico San Carlos, Madrid, Spain (N.G., P.J.-Q., F.A., I.J.N.-G., C.B., C.M.); International Centre for Circulatory Health, National Heart and Lung Institute, Imperial College London and Imperial College Healthcare NHS Trust, London, United Kingdom (R.P., S.S., S.N., J.T., J.D.); and
| | - Borja Ibañez
- From the Cardiovascular Institute, Hospital Clínico San Carlos and Centro Nacional de Investigaciones Cardiovasculares Carlos III, Madrid, Spain (M.E.-P., B.I., A.Q., A.F.-O., J.E.); Cardiovascular Institute, Hospital Clínico San Carlos, Madrid, Spain (N.G., P.J.-Q., F.A., I.J.N.-G., C.B., C.M.); International Centre for Circulatory Health, National Heart and Lung Institute, Imperial College London and Imperial College Healthcare NHS Trust, London, United Kingdom (R.P., S.S., S.N., J.T., J.D.); and
| | - Ricardo Petraco
- From the Cardiovascular Institute, Hospital Clínico San Carlos and Centro Nacional de Investigaciones Cardiovasculares Carlos III, Madrid, Spain (M.E.-P., B.I., A.Q., A.F.-O., J.E.); Cardiovascular Institute, Hospital Clínico San Carlos, Madrid, Spain (N.G., P.J.-Q., F.A., I.J.N.-G., C.B., C.M.); International Centre for Circulatory Health, National Heart and Lung Institute, Imperial College London and Imperial College Healthcare NHS Trust, London, United Kingdom (R.P., S.S., S.N., J.T., J.D.); and
| | - Pilar Jimenez-Quevedo
- From the Cardiovascular Institute, Hospital Clínico San Carlos and Centro Nacional de Investigaciones Cardiovasculares Carlos III, Madrid, Spain (M.E.-P., B.I., A.Q., A.F.-O., J.E.); Cardiovascular Institute, Hospital Clínico San Carlos, Madrid, Spain (N.G., P.J.-Q., F.A., I.J.N.-G., C.B., C.M.); International Centre for Circulatory Health, National Heart and Lung Institute, Imperial College London and Imperial College Healthcare NHS Trust, London, United Kingdom (R.P., S.S., S.N., J.T., J.D.); and
| | - Sayan Sen
- From the Cardiovascular Institute, Hospital Clínico San Carlos and Centro Nacional de Investigaciones Cardiovasculares Carlos III, Madrid, Spain (M.E.-P., B.I., A.Q., A.F.-O., J.E.); Cardiovascular Institute, Hospital Clínico San Carlos, Madrid, Spain (N.G., P.J.-Q., F.A., I.J.N.-G., C.B., C.M.); International Centre for Circulatory Health, National Heart and Lung Institute, Imperial College London and Imperial College Healthcare NHS Trust, London, United Kingdom (R.P., S.S., S.N., J.T., J.D.); and
| | - Sukkinder Nijjer
- From the Cardiovascular Institute, Hospital Clínico San Carlos and Centro Nacional de Investigaciones Cardiovasculares Carlos III, Madrid, Spain (M.E.-P., B.I., A.Q., A.F.-O., J.E.); Cardiovascular Institute, Hospital Clínico San Carlos, Madrid, Spain (N.G., P.J.-Q., F.A., I.J.N.-G., C.B., C.M.); International Centre for Circulatory Health, National Heart and Lung Institute, Imperial College London and Imperial College Healthcare NHS Trust, London, United Kingdom (R.P., S.S., S.N., J.T., J.D.); and
| | - Jason Tarkin
- From the Cardiovascular Institute, Hospital Clínico San Carlos and Centro Nacional de Investigaciones Cardiovasculares Carlos III, Madrid, Spain (M.E.-P., B.I., A.Q., A.F.-O., J.E.); Cardiovascular Institute, Hospital Clínico San Carlos, Madrid, Spain (N.G., P.J.-Q., F.A., I.J.N.-G., C.B., C.M.); International Centre for Circulatory Health, National Heart and Lung Institute, Imperial College London and Imperial College Healthcare NHS Trust, London, United Kingdom (R.P., S.S., S.N., J.T., J.D.); and
| | - Fernando Alfonso
- From the Cardiovascular Institute, Hospital Clínico San Carlos and Centro Nacional de Investigaciones Cardiovasculares Carlos III, Madrid, Spain (M.E.-P., B.I., A.Q., A.F.-O., J.E.); Cardiovascular Institute, Hospital Clínico San Carlos, Madrid, Spain (N.G., P.J.-Q., F.A., I.J.N.-G., C.B., C.M.); International Centre for Circulatory Health, National Heart and Lung Institute, Imperial College London and Imperial College Healthcare NHS Trust, London, United Kingdom (R.P., S.S., S.N., J.T., J.D.); and
| | - Ivan J. Núñez-Gil
- From the Cardiovascular Institute, Hospital Clínico San Carlos and Centro Nacional de Investigaciones Cardiovasculares Carlos III, Madrid, Spain (M.E.-P., B.I., A.Q., A.F.-O., J.E.); Cardiovascular Institute, Hospital Clínico San Carlos, Madrid, Spain (N.G., P.J.-Q., F.A., I.J.N.-G., C.B., C.M.); International Centre for Circulatory Health, National Heart and Lung Institute, Imperial College London and Imperial College Healthcare NHS Trust, London, United Kingdom (R.P., S.S., S.N., J.T., J.D.); and
| | - Camino Bañuelos
- From the Cardiovascular Institute, Hospital Clínico San Carlos and Centro Nacional de Investigaciones Cardiovasculares Carlos III, Madrid, Spain (M.E.-P., B.I., A.Q., A.F.-O., J.E.); Cardiovascular Institute, Hospital Clínico San Carlos, Madrid, Spain (N.G., P.J.-Q., F.A., I.J.N.-G., C.B., C.M.); International Centre for Circulatory Health, National Heart and Lung Institute, Imperial College London and Imperial College Healthcare NHS Trust, London, United Kingdom (R.P., S.S., S.N., J.T., J.D.); and
| | - Alicia Quirós
- From the Cardiovascular Institute, Hospital Clínico San Carlos and Centro Nacional de Investigaciones Cardiovasculares Carlos III, Madrid, Spain (M.E.-P., B.I., A.Q., A.F.-O., J.E.); Cardiovascular Institute, Hospital Clínico San Carlos, Madrid, Spain (N.G., P.J.-Q., F.A., I.J.N.-G., C.B., C.M.); International Centre for Circulatory Health, National Heart and Lung Institute, Imperial College London and Imperial College Healthcare NHS Trust, London, United Kingdom (R.P., S.S., S.N., J.T., J.D.); and
| | - Antonio Fernández-Ortiz
- From the Cardiovascular Institute, Hospital Clínico San Carlos and Centro Nacional de Investigaciones Cardiovasculares Carlos III, Madrid, Spain (M.E.-P., B.I., A.Q., A.F.-O., J.E.); Cardiovascular Institute, Hospital Clínico San Carlos, Madrid, Spain (N.G., P.J.-Q., F.A., I.J.N.-G., C.B., C.M.); International Centre for Circulatory Health, National Heart and Lung Institute, Imperial College London and Imperial College Healthcare NHS Trust, London, United Kingdom (R.P., S.S., S.N., J.T., J.D.); and
| | - Carlos Macaya
- From the Cardiovascular Institute, Hospital Clínico San Carlos and Centro Nacional de Investigaciones Cardiovasculares Carlos III, Madrid, Spain (M.E.-P., B.I., A.Q., A.F.-O., J.E.); Cardiovascular Institute, Hospital Clínico San Carlos, Madrid, Spain (N.G., P.J.-Q., F.A., I.J.N.-G., C.B., C.M.); International Centre for Circulatory Health, National Heart and Lung Institute, Imperial College London and Imperial College Healthcare NHS Trust, London, United Kingdom (R.P., S.S., S.N., J.T., J.D.); and
| | - Bon-Kwon Koo
- From the Cardiovascular Institute, Hospital Clínico San Carlos and Centro Nacional de Investigaciones Cardiovasculares Carlos III, Madrid, Spain (M.E.-P., B.I., A.Q., A.F.-O., J.E.); Cardiovascular Institute, Hospital Clínico San Carlos, Madrid, Spain (N.G., P.J.-Q., F.A., I.J.N.-G., C.B., C.M.); International Centre for Circulatory Health, National Heart and Lung Institute, Imperial College London and Imperial College Healthcare NHS Trust, London, United Kingdom (R.P., S.S., S.N., J.T., J.D.); and
| | - Justin Davies
- From the Cardiovascular Institute, Hospital Clínico San Carlos and Centro Nacional de Investigaciones Cardiovasculares Carlos III, Madrid, Spain (M.E.-P., B.I., A.Q., A.F.-O., J.E.); Cardiovascular Institute, Hospital Clínico San Carlos, Madrid, Spain (N.G., P.J.-Q., F.A., I.J.N.-G., C.B., C.M.); International Centre for Circulatory Health, National Heart and Lung Institute, Imperial College London and Imperial College Healthcare NHS Trust, London, United Kingdom (R.P., S.S., S.N., J.T., J.D.); and
| | - Javier Escaned
- From the Cardiovascular Institute, Hospital Clínico San Carlos and Centro Nacional de Investigaciones Cardiovasculares Carlos III, Madrid, Spain (M.E.-P., B.I., A.Q., A.F.-O., J.E.); Cardiovascular Institute, Hospital Clínico San Carlos, Madrid, Spain (N.G., P.J.-Q., F.A., I.J.N.-G., C.B., C.M.); International Centre for Circulatory Health, National Heart and Lung Institute, Imperial College London and Imperial College Healthcare NHS Trust, London, United Kingdom (R.P., S.S., S.N., J.T., J.D.); and
| |
Collapse
|
23
|
Zhou X, Teng B, Tilley S, Mustafa SJ. A1 adenosine receptor negatively modulates coronary reactive hyperemia via counteracting A2A-mediated H2O2 production and KATP opening in isolated mouse hearts. Am J Physiol Heart Circ Physiol 2013; 305:H1668-79. [PMID: 24043252 DOI: 10.1152/ajpheart.00495.2013] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
We previously demonstrated that A2A, but not A2B, adenosine receptors (ARs) mediate coronary reactive hyperemia (RH), possibly by producing H2O2 and, subsequently, opening ATP-dependent K(+) (KATP) channels in coronary smooth muscle cells. In this study, A1 AR knockout (KO), A3 AR KO, and A1 and A3 AR double-KO (A1/A3 DKO) mice were used to investigate the roles and mechanisms of A1 and A3 ARs in modulation of coronary RH. Coronary flow of isolated hearts was measured using the Langendorff system. A1 KO and A1/A3 DKO, but not A3 KO, mice showed a higher flow debt repayment [~30% more than wild-type (WT) mice, P < 0.05] following a 15-s occlusion. SCH-58261 (a selective A2A AR antagonist, 1 μM) eliminated the augmented RH, suggesting the involvement of enhanced A2A AR-mediated signaling in A1 KO mice. In isolated coronary arteries, immunohistochemistry showed an upregulation of A2A AR (1.6 ± 0.2 times that of WT mice, P < 0.05) and a higher magnitude of adenosine-induced H2O2 production in A1 KO mice (1.8 ± 0.3 times that of WT mice, P < 0.05), which was blocked by SCH-58261. Catalase (2,500 U/ml) and glibenclamide (a KATP channel blocker, 5 μM), but not N(G)-nitro-l-arginine methyl ester, also abolished the enhanced RH in A1 KO mice. Our data suggest that A1, but not A3, AR counteracts the A2A AR-mediated CF increase and that deletion of A1 AR results in upregulation of A2A AR and/or removal of the negative modulatory effect of A1 AR, thus leading to an enhanced A2A AR-mediated H2O2 production, KATP channel opening, and coronary vasodilation during RH. This is the first report implying that A1 AR has a role in coronary RH.
Collapse
Affiliation(s)
- Xueping Zhou
- Department of Physiology and Pharmacology, West Virginia University, Morgantown, West Virginia
| | | | | | | |
Collapse
|
24
|
El-Awady MS, Rajamani U, Teng B, Tilley SL, Mustafa SJ. Evidence for the involvement of NADPH oxidase in adenosine receptors-mediated control of coronary flow using A 1 and A 3 knockout mice. Physiol Rep 2013; 1:e00070. [PMID: 24159377 PMCID: PMC3804374 DOI: 10.1002/phy2.70] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
The NADPH oxidase (Nox) subunits 1, 2 (gp91 phox) and 4 are the major sources for reactive oxygen species (ROS) in cardiovascular system. In conditions such as ischemia-reperfusion injury and hypoxia, both ROS and adenosine are released suggesting a possible interaction. We hypothesized that ROS generated through Nox is involved in adenosine-induced coronary flow (CF) responses. Adenosine (10-8-10-5.5 M) increased CF in isolated hearts from wild type (WT; C57/BL6), A1 adenosine receptor (AR) knockout (A1KO), A3AR KO (A3KO) and A1 and A3AR double KO (A1/A3DKO) mice. The Nox inhibitors apocynin (10-5 M) and gp91 ds-tat (10-6 M) or the SOD and catalase-mimicking agent EUK134 (50 μM) decreased the adenosine-enhanced CF in the WT and all the KOs. Additionally, adenosine increased phosphorylation of p47-phox subunit and ERK 1/2 without changing protein expression of Nox isoforms in WT. Moreover, intracellular superoxide production was increased by adenosine and CGS-21680 (a selective A2A agonist), but not BAY 60-6583 (a selective A2B agonist), in mouse coronary artery smooth muscle cells (CASMCs) and endothelial cells (CAECs). This superoxide increase was inhibited by the gp91 ds-tat and ERK 1/2 inhibitor (PD98059). In conclusion, adenosine-induced increase in CF in isolated heart involves Nox2-generated superoxide, possibly through ERK 1/2 phosphorylation with subsequent p47-phox subunit phosphorylation. This adenosine/Nox/ROS interaction occurs in both CASMCs and CAECs, and involves neither A1 nor A3 ARs, but possibly A2A ARs in mouse.
Collapse
Affiliation(s)
- Mohammed S El-Awady
- Department of Physiology and Pharmacology,Center for Cardiovascular and Respiratory Sciences and Clinical & Translational Science Institute, West Virginia University, Morgantown, WV 26505, USA ; Department of Pharmacology and Toxicology, Faculty of Pharmacy, Mansoura University, Mansoura 35516, Egypt
| | | | | | | | | |
Collapse
|
25
|
Kunduri S, Dick G, Nayeem M, Mustafa S. Adenosine A 1 receptor signaling inhibits BK channels through a PKCα-dependent mechanism in mouse aortic smooth muscle. Physiol Rep 2013; 1. [PMID: 23977428 PMCID: PMC3747964 DOI: 10.1002/phy2.37] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Adenosine receptors (AR; A1, A2A, A2B, and A3) contract and relax smooth muscle through different signaling mechanisms. Deciphering these complex responses remains difficult because relationships between AR subtypes and various end-effectors (e.g., enzymes and ion channels) remain to be identified. A1AR stimulation is associated with the production of 20–hydroxyeicosatetraenoic acid (20–HETE) and activation of protein kinase C (PKC). 20–HETE and PKC can inhibit large conductance Ca2+/voltage-sensitive K+ (BK) channels that regulate smooth muscle contraction. We tested the hypothesis that activation of A1AR inhibits BK channels via a PKC-dependent mechanism. Patch clamp recordings and Western blots were performed using aortae of wild type (WT) and A1AR knockout (A1KO) mice. There were no differences in whole-cell K+ current or α and β1 subunits expression between WT and A1KO. 20–HETE (100 nmol/L) inhibited BK current similarly in WT and A1KO mice. NECA (5′–N–ethylcarboxamidoadenosine; 10 μmol/L), a nonselective AR agonist, increased BK current in myocytes from both WT and A1KO mice, but the increase was greater in A1KO (52 ± 15 vs. 17 ± 3%; P < 0.05). This suggests that A1AR signaling negatively regulates BK channel activity. Accordingly, CCPA (2–chloro–N(6)-cyclopentyladenosine; 100 nmol/L), an A1AR-selective agonist, inhibited BK current in myocytes from WT but not A1KO mice (81 ± 4 vs. 100 ± 7% of control; P < 0.05). Gö6976 (100 nmol/L), a PKCα inhibitor, abolished the effect of CCPA to inhibit BK current (99 ± 3% of control). These data lead us to conclude that, in aortic smooth muscle, A1AR inhibits BK channel activity and that this occurs via a mechanism involving PKCα.
Collapse
Affiliation(s)
- Ss Kunduri
- Department of Physiology & Pharmacology, West Virginia University School of Medicine ; Center for Cardiovascular and Respiratory Sciences, West Virginia University School of Medicine
| | | | | | | |
Collapse
|