1
|
Ruvira S, Rodríguez-Rodríguez P, Abderrahim F, Morales D, Cañas S, Valdivieso A, Ramiro-Cortijo D, Arribas SM. Resistance artery vasodilator pathways involved in the antihypertensive effects of cocoa shell extract in rats exposed to fetal undernutrition. J Physiol 2024; 602:6065-6085. [PMID: 39388282 DOI: 10.1113/jp287097] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2024] [Accepted: 08/20/2024] [Indexed: 10/12/2024] Open
Abstract
Fetal undernutrition establishes the foundations for hypertension development, with oxidative stress being a key hallmark. A growing interest in nutraceuticals for treating hypertension and environmental waste concerns prompted the present study aiming to evaluate whether supplementation with a polyphenol enriched extract from cocoa shell (CSE), a by-product from the chocolate industry with antioxidant properties, reduces hypertension of developmental origin, thus improving mesenteric resistance artery (MRA) vasodilatation. Adult male and female offspring from rats exposed to 50% food restriction from mid-gestation (maternal undernutrition, MUN) and controls were used. Supplementation was given through a gelatine (vehicle, VEH) or containing CSE (250 mg kg-1 day-1) 5 days week-1 for 3 weeks. Systolic blood pressure (SBP) was assessed by tail-cuff plethysmography. MRA function was studied by wire myography, and superoxide anion and nitric oxide were investigated by fluorescent indicators and confocal microscopy. Compared to control-VEH, MUN-VEH males showed significantly higher SBP, reduced MRA as well as relaxation to ACh, sodium nitroprusside and the AMPK agonist 5-aminoimidazole-4-carboxamide riboside, but not to isoproterenol. In MUN males, endothelial endothelium-derived hyperpolarizing factor and nitric oxide were unaltered, but MRA released a vasoconstrictor prostanoid and produced higher levels of superoxide anion. CSE normalized blood pressure and improved all above-mentioned MRA alterations in MUN males without an effect on control counterparts, except the reduction of superoxide anion. MUN-VEH females were normotensive and only showed a tendency towards larger superoxide anion production, which was abolished by CSE. CSE supplementation reduces SBP improving endothelium-dependent and independent MRA vasodilatation, related to local superoxide anion reduction, being a potential nutraceutical ingredient to counteract hypertension, in addition to contributing to the circular economy. KEY POINTS: Fetal undernutrition induces hypertension in males associated with deficient resistance artery vasodilatation, being normalized by cocoa shell extract (CSE). Release of a cyclooxygenase-derived contractile factor is the main endothelial alteration, which is abolished by CSE. AMPK and soluble guanylyl cyclase-mediated relaxation are also reduced in smooth muscle cells from maternal undernutrition resistance arteries, being improved by CSE. Vascular oxidative damage caused by excess superoxide anion generation can account for impaired vasodilatation, which is improved by CSE. The capacity of CSE to improve relaxation is probably related to its antioxidant bioactive factors, and thus cocoa shell is a potential food by-product to treat hypertension.
Collapse
Affiliation(s)
- Santiago Ruvira
- Department of Physiology, Faculty of Medicine, Universidad Autónoma de Madrid, Madrid, Spain
- Food, Oxidative Stress and Cardiovascular Health (FOSCH) Research Group, Universidad Autónoma de Madrid, Madrid, Spain
| | - Pilar Rodríguez-Rodríguez
- Department of Physiology, Faculty of Medicine, Universidad Autónoma de Madrid, Madrid, Spain
- Food, Oxidative Stress and Cardiovascular Health (FOSCH) Research Group, Universidad Autónoma de Madrid, Madrid, Spain
| | - Fatima Abderrahim
- Department of Physiology, Faculty of Medicine, Universidad Autónoma de Madrid, Madrid, Spain
| | - Dolores Morales
- Confocal Microscopy Service (SiDI), Faculty of Medicine, Universidad Autonoma de Madrid, Madrid, Spain
| | - Silvia Cañas
- Food, Oxidative Stress and Cardiovascular Health (FOSCH) Research Group, Universidad Autónoma de Madrid, Madrid, Spain
- Institute of Food Science Research (CIAL), Universidad Autónoma de Madrid (UAM-CSIC), Madrid, Spain
- Department of Agricultural Chemistry and Food Science, Faculty of Science, Universidad Autónoma de Madrid, Madrid, Spain
| | - Alberto Valdivieso
- Department of Physiology, Faculty of Medicine, Universidad Autónoma de Madrid, Madrid, Spain
| | - David Ramiro-Cortijo
- Department of Physiology, Faculty of Medicine, Universidad Autónoma de Madrid, Madrid, Spain
- Food, Oxidative Stress and Cardiovascular Health (FOSCH) Research Group, Universidad Autónoma de Madrid, Madrid, Spain
| | - Silvia M Arribas
- Department of Physiology, Faculty of Medicine, Universidad Autónoma de Madrid, Madrid, Spain
- Food, Oxidative Stress and Cardiovascular Health (FOSCH) Research Group, Universidad Autónoma de Madrid, Madrid, Spain
| |
Collapse
|
2
|
Vidović A, Dolinar K, Chibalin AV, Pirkmajer S. AMPK and glucose deprivation exert an isoform-specific effect on the expression of Na +,K +-ATPase subunits in cultured myotubes. J Muscle Res Cell Motil 2024; 45:139-154. [PMID: 38709429 DOI: 10.1007/s10974-024-09673-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Accepted: 04/24/2024] [Indexed: 05/07/2024]
Abstract
In skeletal muscle, Na+,K+-ATPase (NKA), a heterodimeric (α/β) P-type ATPase, has an essential role in maintenance of Na+ and K+ homeostasis, excitability, and contractility. AMP-activated protein kinase (AMPK), an energy sensor, increases the membrane abundance and activity of NKA in L6 myotubes, but its potential role in regulation of NKA content in skeletal muscle, which determines maximum capacity for Na+ and K+ transport, has not been clearly delineated. We examined whether energy stress and/or AMPK affect expression of NKA subunits in rat L6 and primary human myotubes. Energy stress, induced by glucose deprivation, increased protein content of NKAα1 and NKAα2 in L6 myotubes, while decreasing the content of NKAα1 in human myotubes. Pharmacological AMPK activators (AICAR, A-769662, and diflunisal) modulated expression of NKA subunits, but their effects only partially mimicked those that occurred in response to glucose deprivation, indicating that AMPK does not mediate all effects of energy stress on NKA expression. Gene silencing of AMPKα1/α2 increased protein levels of NKAα1 in L6 myotubes and NKAα1 mRNA levels in human myotubes, while decreasing NKAα2 protein levels in L6 myotubes. Collectively, our results suggest a role for energy stress and AMPK in modulation of NKA expression in skeletal muscle. However, their modulatory effects were not conserved between L6 myotubes and primary human myotubes, which suggests that coupling between energy stress, AMPK, and regulation of NKA expression in vitro depends on skeletal muscle cell model.
Collapse
Affiliation(s)
- Anja Vidović
- Faculty of Medicine, Institute of Pathophysiology, University of Ljubljana, Ljubljana, Slovenia
| | - Klemen Dolinar
- Faculty of Medicine, Institute of Pathophysiology, University of Ljubljana, Ljubljana, Slovenia
| | - Alexander V Chibalin
- Department of Molecular Medicine and Surgery, Integrative Physiology, Karolinska Institutet, Stockholm, Sweden
- National Research Tomsk State University, Tomsk, Russia
| | - Sergej Pirkmajer
- Faculty of Medicine, Institute of Pathophysiology, University of Ljubljana, Ljubljana, Slovenia.
| |
Collapse
|
3
|
MacMillan S, Burns DP, O'Halloran KD, Evans AM. SubSol-HIe is an AMPK-dependent hypoxia-responsive subnucleus of the nucleus tractus solitarius that coordinates the hypoxic ventilatory response and protects against apnoea in mice. Pflugers Arch 2024; 476:1087-1107. [PMID: 38635058 PMCID: PMC11166843 DOI: 10.1007/s00424-024-02957-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Revised: 03/24/2024] [Accepted: 03/31/2024] [Indexed: 04/19/2024]
Abstract
Functional magnetic resonance imaging (fMRI) suggests that the hypoxic ventilatory response is facilitated by the AMP-activated protein kinase (AMPK), not at the carotid bodies, but within a subnucleus (Bregma -7.5 to -7.1 mm) of the nucleus tractus solitarius that exhibits right-sided bilateral asymmetry. Here, we map this subnucleus using cFos expression as a surrogate for neuronal activation and mice in which the genes encoding the AMPK-α1 (Prkaa1) and AMPK-α2 (Prkaa2) catalytic subunits were deleted in catecholaminergic cells by Cre expression via the tyrosine hydroxylase promoter. Comparative analysis of brainstem sections, relative to controls, revealed that AMPK-α1/α2 deletion inhibited, with right-sided bilateral asymmetry, cFos expression in and thus activation of a neuronal cluster that partially spanned three interconnected anatomical nuclei adjacent to the area postrema: SolDL (Bregma -7.44 mm to -7.48 mm), SolDM (Bregma -7.44 mm to -7.48 mm) and SubP (Bregma -7.48 mm to -7.56 mm). This approximates the volume identified by fMRI. Moreover, these nuclei are known to be in receipt of carotid body afferent inputs, and catecholaminergic neurons of SubP and SolDL innervate aspects of the ventrolateral medulla responsible for respiratory rhythmogenesis. Accordingly, AMPK-α1/α2 deletion attenuated hypoxia-evoked increases in minute ventilation (normalised to metabolism), reductions in expiration time, and increases sigh frequency, but increased apnoea frequency during hypoxia. The metabolic response to hypoxia in AMPK-α1/α2 knockout mice and the brainstem and spinal cord catecholamine levels were equivalent to controls. We conclude that within the brainstem an AMPK-dependent, hypoxia-responsive subnucleus partially spans SubP, SolDM and SolDL, namely SubSol-HIe, and is critical to coordination of active expiration, the hypoxic ventilatory response and defence against apnoea.
Collapse
Affiliation(s)
- Sandy MacMillan
- Centre for Discovery Brain Sciences, College of Medicine and Veterinary Medicine, Hugh Robson Building, University of Edinburgh, Edinburgh, EH8 9XD, UK
| | - David P Burns
- Department of Physiology, School of Medicine, College of Medicine & Health, University College Cork, Cork, Ireland
| | - Ken D O'Halloran
- Department of Physiology, School of Medicine, College of Medicine & Health, University College Cork, Cork, Ireland
| | - A Mark Evans
- Centre for Discovery Brain Sciences, College of Medicine and Veterinary Medicine, Hugh Robson Building, University of Edinburgh, Edinburgh, EH8 9XD, UK.
| |
Collapse
|
4
|
Liu Y, Cheng Y, Xiang N, Wang Z, Li S, Gong L, Wang X. Aerobic exercise improves BK Ca channel-mediated vasodilation in diabetic vascular smooth muscle via AMPK/Nrf2/HO-1 pathway. Acta Diabetol 2024; 61:425-434. [PMID: 38041787 DOI: 10.1007/s00592-023-02210-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/01/2023] [Accepted: 11/07/2023] [Indexed: 12/03/2023]
Abstract
AIMS This study aims to investigate the effect of aerobic exercise training on BKCa channel in diabetic vascular smooth muscle and explore the underlying mechanism. METHODS Control m/m mice and diabetic db/db mice were randomly assigned to sedentary groups (W and D) and exercise training groups (WE and DE). Mice in exercise groups underwent training sessions lasting for 12 weeks, with a speed of 12 m/min for 60 min, five times per week. The thoracic aorta was extracted isolated and examined for measurement of vascular structure, global levels of reactive oxygen species (ROS), vasodilation, and protein expression. Rat thoracic aorta vascular smooth muscle cells (USMCs) were cultured, and siRNA transfection was conducted to detect whether AMPK contributed to the regulation. ROS level and protein expression were measured. RESULTS Compared with control mice, diabetic mice had a larger vascular medium thickness, impaired BKCa-mediated vasodilation, a higher level of ROS, and a lower expression of BKCa α, BKCa β1, Nrf2, p-Nrf2, p-Nrf2/Nrf2, HO-1, and p-AMPK/AMPK. Exercise training increased the expression of BKCa α, Nrf2, p-Nrf2, p-Nrf2/Nrf2, HO-1, and p-AMPK/AMPK. AMPK deletion led to lower ROS levels and expression of BKCa α, β1, Nrf2, and HO-1 in USMCs cultured in high glucose conditions. CONCLUSIONS BKCa channel protein expression reduction in VSMCs contributes to vasodilation and vascular remodeling dysfunction in diabetes mellitus. Aerobic exercise can promote the expression of BKCa channel and improve BKCa-mediated vascular dysfunction in diabetic VSMCs through AMPK/Nrf2/HO-1 signaling pathway.
Collapse
Affiliation(s)
- Yujia Liu
- Department of Physical Education, Jiangsu Normal University, Xuzhou, China
| | - Yue Cheng
- Department of Physical Education, Jiangsu Normal University, Xuzhou, China
| | - Na Xiang
- Caoxian People's Hospital, Heze, China
| | - Zhiyuan Wang
- China Institute of Sport and Health Science, Beijing Sport University, Beijing, China
| | - Siyu Li
- China Institute of Sport and Health Science, Beijing Sport University, Beijing, China
| | - Lijing Gong
- China Institute of Sport and Health Science, Beijing Sport University, Beijing, China
| | - Xingqi Wang
- Biomedical R&d Center, School of Life Science, Jiangsu Normal University, Tongshan District, No. 101, Shanghai Road, Xuzhou, Jiangsu, China.
| |
Collapse
|
5
|
Zhuang W, Mun SY, Park M, Jeong J, Kim HR, Na S, Lee SJ, Park H, Park WS. Inhibition of voltage-dependent K + channels in rabbit coronary arterial smooth muscle cells by the atypical antipsychotic agent sertindole. J Appl Toxicol 2024; 44:391-399. [PMID: 37786982 DOI: 10.1002/jat.4549] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2023] [Revised: 09/13/2023] [Accepted: 09/18/2023] [Indexed: 10/04/2023]
Abstract
The regulation of membrane potential and the contractility of vascular smooth muscle cells (VSMCs) by voltage-dependent K+ (Kv) potassium channels are well-established. In this study, native VSMCs from rabbit coronary arteries were used to investigate the inhibitory effect of sertindole, an atypical antipsychotic agent, on Kv channels. Sertindole induced dose-dependent inhibition of Kv channels, with an IC50 of 3.13 ± 0.72 μM. Although sertindole did not cause a change in the steady-state activation curve, it did lead to a negative shift in the steady-state inactivation curve. The application of 1- or 2-Hz train pulses failed to alter the sertindole-induced inhibition of Kv channels, suggesting use-independent effects of the drug. The inhibitory response to sertindole was significantly diminished by pretreatment with a Kv1.5 inhibitor but not by Kv2.1 and Kv7 subtype inhibitors. These findings demonstrate the sertindole dose-dependent and use-independent inhibition of vascular Kv channels (mainly the Kv1.5 subtype) through a mechanism that involves altering steady-state inactivation curves. Therefore, the use of sertindole as an antipsychotic drug may have adverse effects on the cardiovascular system.
Collapse
Affiliation(s)
- Wenwen Zhuang
- Institute of Medical Sciences, Department of Physiology, Kangwon National University School of Medicine, Chuncheon, South Korea
| | - Seo-Yeong Mun
- Institute of Medical Sciences, Department of Physiology, Kangwon National University School of Medicine, Chuncheon, South Korea
| | - Minju Park
- Institute of Medical Sciences, Department of Physiology, Kangwon National University School of Medicine, Chuncheon, South Korea
| | - Junsu Jeong
- Institute of Medical Sciences, Department of Physiology, Kangwon National University School of Medicine, Chuncheon, South Korea
| | - Hye Ryung Kim
- Institute of Medical Sciences, Department of Physiology, Kangwon National University School of Medicine, Chuncheon, South Korea
| | - Sunghun Na
- Institute of Medical Sciences, Department of Obstetrics and Gynecology, Kangwon National University Hospital, Kangwon National University School of Medicine, Chuncheon, South Korea
| | - Se Jin Lee
- Institute of Medical Sciences, Department of Obstetrics and Gynecology, Kangwon National University Hospital, Kangwon National University School of Medicine, Chuncheon, South Korea
| | - Hongzoo Park
- Institute of Medical Sciences, Department of Urology, Kangwon National University School of Medicine, Chuncheon, South Korea
| | - Won Sun Park
- Institute of Medical Sciences, Department of Physiology, Kangwon National University School of Medicine, Chuncheon, South Korea
| |
Collapse
|
6
|
Sahinturk S. Cilostazol induces vasorelaxation through the activation of the eNOS/NO/cGMP pathway, prostanoids, AMPK, PKC, potassium channels, and calcium channels. Prostaglandins Other Lipid Mediat 2023; 169:106782. [PMID: 37741358 DOI: 10.1016/j.prostaglandins.2023.106782] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2023] [Revised: 08/02/2023] [Accepted: 09/15/2023] [Indexed: 09/25/2023]
Abstract
OBJECTIVE This study aimed to investigate vasoactive effect mechanisms of cilostazol in rat thoracic aorta. MATERIALS AND METHODS The vessel rings prepared from the thoracic aortas of the male rats were placed in the chambers of the isolated tissue bath system. The resting tone was adjusted to 1 g. Following the equilibration phase, potassium chloride or phenylephrine was used to contract the vessel rings. When achieving a steady contraction, cilostazol was applied cumulatively (10-8-10-4 M). In the presence of potassium channel blockers or signaling pathway inhibitors, the same experimental procedure was performed. RESULTS Cilostazol exhibited a significant vasorelaxant effect in a concentration-dependent manner (pD2: 5.94 ± 0.94) (p < .001). The vasorelaxant effect level of cilostazol was significantly reduced by the endothelial nitric oxide synthase inhibitor L-NAME (10-4 M), soluble guanylate cyclase inhibitor methylene blue (10 µM), cyclooxygenase 1/2 inhibitor indomethacin (5 µM), adenosine monophosphate-activated protein kinase inhibitor compound C (10 µM), non-selective potassium channel blocker tetraethylammonium chloride (10 mM), large-conductance calcium-activated potassium channel blocker iberiotoxin (20 nM), voltage-gated potassium channel blocker 4-Aminopyridine (1 mM), and inward-rectifier potassium channel blocker BaCl2 (30 µM) (p < .001). Moreover, incubation of cilostazol (10-4 M) significantly reduced caffeine (10 mM), cyclopiazonic acid (10 µM), and phorbol 12-myristate 13-acetate-induced (100 µM) vascular contractions (p < .001). CONCLUSIONS In the rat thoracic aorta, the vasodilator action level of cilostazol is quite noticeable. The vasorelaxant effects of cilostazol are mediated by the eNOS/NO/cGMP pathway, prostanoids, AMPK pathway, PKC, potassium channels, and calcium channels.
Collapse
Affiliation(s)
- Serdar Sahinturk
- Bursa Uludag University Medicine School, Physiology Department, 16059, Bursa, Turkey.
| |
Collapse
|
7
|
Higashikuni Y, Liu W, Sata M. Not a small frog in a big pond: targeting bradykinin receptor B2 signaling in vascular smooth muscle cells for treatment of hypertension. Hypertens Res 2023; 46:2415-2418. [PMID: 37507534 DOI: 10.1038/s41440-023-01385-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2023] [Accepted: 07/01/2023] [Indexed: 07/30/2023]
Affiliation(s)
- Yasutomi Higashikuni
- Department of Cardiovascular Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8655, Japan.
| | - Wenhao Liu
- Department of Cardiovascular Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8655, Japan
| | - Masataka Sata
- Department of Cardiovascular Medicine, Tokushima University Graduate School, 3-18-15 Kuramoto-cho, Tokushima-shi, Tokushima, 770-8503, Japan
| |
Collapse
|
8
|
Li XY, Qian LL, Wu Y, Zhang YM, Dang SP, Liu XY, Tang X, Lu CY, Wang RX. Advanced glycation end products impair coronary artery BK channels via AMPK/Akt/FBXO32 signaling pathway. Diab Vasc Dis Res 2023; 20:14791641231197107. [PMID: 37592725 PMCID: PMC10439763 DOI: 10.1177/14791641231197107] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 08/19/2023] Open
Abstract
Background: Advanced glycation end products (AGEs) impair vascular physiology in Diabetes mellitus (DM). However, the underlying mechanisms remain unclear. Vascular large conductance calcium-activated potassium (BK) channels play important roles in coronary arterial function.Purpose: Our study aimed to investigate the regulatory role of AGEs in BK channels.Research Design: Using gavage of vehicle (V, normal saline) or aminoguanidine (A) for 8 weeks, normal and diabetic rats were divided into four groups: C+V group, DM+V group, C+A group, and DM+A group.Study Sample: Coronary arteries from different groups of rats and human coronary smooth muscle cells were used in this study.Data Collection and Analysis: Data were presented as mean ± SEM (standard error of mean). Student's t-test was used to compare data between two groups. One-way ANOVA with post-hoc LSD analysis was used to compare data between multiple groups.Results: Compared to the C+V group, vascular contraction induced by iberiotoxin (IBTX), a BK channel inhibitor, was impaired, and BK channel densities decreased in the DM+V group. However, aminoguanidine administration reduced the impairment. Protein expression of BK-β1, phosphorylation of adenosine 5'-monophosphate-activated protein kinase (AMPK), and protein kinase B (PKB or Akt) were down-regulated, while F-box protein 32 (FBXO32) expression increased in the DM+V group and in high glucose (HG) cultured human coronary smooth muscle cells. Treatment with aminoguanidine in vitro and in vivo could reverse the above protein expression. The effect of aminoguanidine on the improvement of BK channel function by inhibiting the generation of AGEs was reversed by adding MK2206 (Akt inhibitor) or Compound C (AMPK inhibitor) in HG conditions in vitro.Conclusions: AGEs aggravate BK channel dysfunction via the AMPK/Akt/FBXO32 signaling pathway.
Collapse
Affiliation(s)
- Xiao-Yan Li
- Department of Cardiology, The Affiliated Wuxi People’s Hospital of Nanjing Medical University, Wuxi, China
| | - Ling-Ling Qian
- Department of Cardiology, The Affiliated Wuxi People’s Hospital of Nanjing Medical University, Wuxi, China
| | - Ying Wu
- Department of Cardiology, The Affiliated Wuxi People’s Hospital of Nanjing Medical University, Wuxi, China
| | - Yu-Min Zhang
- Department of Cardiology, The Affiliated Wuxi People’s Hospital of Nanjing Medical University, Wuxi, China
| | - Shi-Peng Dang
- Department of Cardiology, The Affiliated Wuxi People’s Hospital of Nanjing Medical University, Wuxi, China
| | - Xiao-Yu Liu
- Department of Cardiology, The Affiliated Wuxi People’s Hospital of Nanjing Medical University, Wuxi, China
| | - Xu Tang
- Department of Cardiology, The Affiliated Wuxi People’s Hospital of Nanjing Medical University, Wuxi, China
| | - Cun-yu Lu
- Department of Cardiology, The Affiliated Wuxi People’s Hospital of Nanjing Medical University, Wuxi, China
| | - Ru-Xing Wang
- Department of Cardiology, The Affiliated Wuxi People’s Hospital of Nanjing Medical University, Wuxi, China
| |
Collapse
|
9
|
Abdelazeem H, Tu L, Thuillet R, Ottaviani M, Boulfrad A, Beck T, Senbel A, Mani S, Castier Y, Guyard A, Tran-Dinh A, El-Benna J, Longrois D, Silverstein AM, Guignabert C, Norel X. AMPK activation by metformin protects against pulmonary hypertension in rats and relaxes isolated human pulmonary artery. Eur J Pharmacol 2023; 946:175579. [PMID: 36914083 DOI: 10.1016/j.ejphar.2023.175579] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2022] [Revised: 02/03/2023] [Accepted: 02/05/2023] [Indexed: 03/13/2023]
Abstract
Pulmonary hypertension (PH) is associated with pulmonary vasoconstriction and endothelial dysfunction leading to impaired nitric oxide (NO) and prostacyclin (PGI2) pathways. Metformin, the first line treatment for type 2 diabetes and AMP-activated protein kinase (AMPK) activator, has been recently highlighted as a potential PH treatment. AMPK activation has been reported to improve endothelial function by enhancing endothelial NO synthase (eNOS) activity and to have relaxant effects in blood vessels. In this study, we examined the effect of metformin treatment on PH as well as on NO and PGI2 pathways in monocrotaline (MCT)-injected rats with established PH. Moreover, we investigated the anti-contractile effects of AMPK activators on endothelium-denuded human pulmonary arteries (HPA) from Non-PH and Group 3 PH patients (due to lung diseases and/or hypoxia). Furthermore, we explored the interaction between treprostinil and the AMPK/eNOS pathway. Our results showed that metformin protected against PH progression in MCT rats where it reduced the mean pulmonary artery pressure, pulmonary vascular remodeling and right ventricular hypertrophy and fibrosis compared to vehicle-treated MCT rats. The protective effects on rat lungs were mediated in part by increasing eNOS activity and protein kinase G-1 expression but not through the PGI2 pathway. In addition, incubation with AMPK activators reduced the phenylephrine-induced contraction of endothelium-denuded HPA from Non-PH and PH patients. Finally, treprostinil also augmented eNOS activity in HPA smooth muscle cells. In conclusion, we found that AMPK activation can enhance the NO pathway, attenuate vasoconstriction by direct effects on smooth muscles, and reverse established MCT-induced PH in rats.
Collapse
Affiliation(s)
- Heba Abdelazeem
- Université Paris Cité and Université Sorbonne Paris Nord, INSERM, LVTS, F-75018, Paris, France; Department of Pharmacology and Toxicology, Faculty of Pharmacy, Alexandria University, Egypt
| | - Ly Tu
- INSERM UMR_S 999 «Pulmonary Hypertension: Pathophysiology and Novel Therapies», Hôpital Marie Lannelongue, 92350, Le Plessis-Robinson, France; Université Paris-Saclay, Faculté de Médecine, 94270, Le Kremlin-Bicêtre, France
| | - Raphaël Thuillet
- INSERM UMR_S 999 «Pulmonary Hypertension: Pathophysiology and Novel Therapies», Hôpital Marie Lannelongue, 92350, Le Plessis-Robinson, France; Université Paris-Saclay, Faculté de Médecine, 94270, Le Kremlin-Bicêtre, France
| | - Mina Ottaviani
- INSERM UMR_S 999 «Pulmonary Hypertension: Pathophysiology and Novel Therapies», Hôpital Marie Lannelongue, 92350, Le Plessis-Robinson, France; Université Paris-Saclay, Faculté de Médecine, 94270, Le Kremlin-Bicêtre, France
| | - Achraf Boulfrad
- Université Paris Cité and Université Sorbonne Paris Nord, INSERM, LVTS, F-75018, Paris, France
| | - Thomas Beck
- Université Paris Cité and Université Sorbonne Paris Nord, INSERM, LVTS, F-75018, Paris, France
| | - Amira Senbel
- Arab Academy for Science, Technology & Maritime Transport, College of Pharmacy, Alexandria, Egypt
| | - Salma Mani
- Université Paris Cité and Université Sorbonne Paris Nord, INSERM, LVTS, F-75018, Paris, France; Université de Monastir-Tunisia, Institut Supérieur de Biotechnologie de Monastir (ISBM), Tunisia
| | - Yves Castier
- Hôpital Bichat-Claude Bernard, Assistance Publique-Hôpitaux de Paris, Université Paris Cité, Paris, France
| | - Alice Guyard
- Hôpital Bichat-Claude Bernard, Assistance Publique-Hôpitaux de Paris, Université Paris Cité, Paris, France
| | - Alexy Tran-Dinh
- Université Paris Cité and Université Sorbonne Paris Nord, INSERM, LVTS, F-75018, Paris, France; Hôpital Bichat-Claude Bernard, Assistance Publique-Hôpitaux de Paris, Université Paris Cité, Paris, France
| | - Jamel El-Benna
- Université Paris Cité, INSERM-U1149, CNRS-ERL8252, Centre de Recherche sur l'Inflammation, Laboratoire d'Excellence Inflamex, Faculté de Médecine, Site Xavier Bichat, Paris, F-75018, France
| | - Dan Longrois
- Université Paris Cité and Université Sorbonne Paris Nord, INSERM, LVTS, F-75018, Paris, France; Hôpital Bichat-Claude Bernard, Assistance Publique-Hôpitaux de Paris, Université Paris Cité, Paris, France
| | | | - Christophe Guignabert
- INSERM UMR_S 999 «Pulmonary Hypertension: Pathophysiology and Novel Therapies», Hôpital Marie Lannelongue, 92350, Le Plessis-Robinson, France; Université Paris-Saclay, Faculté de Médecine, 94270, Le Kremlin-Bicêtre, France
| | - Xavier Norel
- Université Paris Cité and Université Sorbonne Paris Nord, INSERM, LVTS, F-75018, Paris, France.
| |
Collapse
|
10
|
Feng D, Guo YY, Wang W, Yan LF, Sun T, Liu QQ, Cui GB, Nan HY. α-Subunit Tyrosine Phosphorylation Is Required for Activation of the Large Conductance Ca 2+-Activated Potassium Channel in the Rabbit Sphincter of Oddi. THE AMERICAN JOURNAL OF PATHOLOGY 2022; 192:1725-1744. [PMID: 36150507 DOI: 10.1016/j.ajpath.2022.08.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Revised: 08/06/2022] [Accepted: 08/31/2022] [Indexed: 06/16/2023]
Abstract
Large conductance Ca2+-activated potassium (BKCa) channels are regulated by intracellular free Ca2+ concentrations ([Ca2+]i) and channel protein phosphorylation. In hypercholesterolemia (HC), motility impairment of the sphincter of Oddi (SO) is associated with abnormal [Ca2+]i accumulation in smooth muscle cells of the rabbit SO (RSOSMCs), which is closely related to BKCa channel activity. However, the underlying mechanisms regulating channel activity remain unclear. In this study, an HC rabbit model was generated and used to investigate BKCa channel activity of RSOSMCs via SO muscle tone measurement in vitro and manometry in vivo, electrophysiological recording, intracellular calcium measurement, and Western blot analyses. BKCa channel activity was decreased, which correlated with [Ca2+]i overload and reduced tyrosine phosphorylation of the BKCa α-subunit in the HC group. The abnormal [Ca2+]i accumulation and decreased BKCa channel activity were partially restored by Na3VO4 pretreatment but worsened by genistein in RSOSMCs in the HC group. This study suggests that α-subunit tyrosine phosphorylation is required for [Ca2+]i to activate BKCa channels, and there is a negative feedback between the BKCa channel and the L-type voltage-dependent Ca2+ channel that regulates [Ca2+]i. This study provides direct evidence that tyrosine phosphorylation of BKCa α-subunits is required for [Ca2+]i to activate BKCa channels in RSOSMCs, which may be the underlying physiological and pathologic mechanism regulating the activity of BKCa channels in SO cells.
Collapse
Affiliation(s)
- Dan Feng
- Department of Radiology and Functional and Molecular Imaging Key Lab of Shaanxi Province, Tangdu Hospital, Fourth Military Medical University, Xi'an, China
| | - Yan-Yan Guo
- Department of Pharmacy, Tangdu Hospital, Fourth Military Medical University, Xi'an, China
| | - Wen Wang
- Department of Radiology and Functional and Molecular Imaging Key Lab of Shaanxi Province, Tangdu Hospital, Fourth Military Medical University, Xi'an, China
| | - Lin-Feng Yan
- Department of Radiology and Functional and Molecular Imaging Key Lab of Shaanxi Province, Tangdu Hospital, Fourth Military Medical University, Xi'an, China
| | - Ting Sun
- Department of Pharmacy, Tangdu Hospital, Fourth Military Medical University, Xi'an, China
| | - Qing-Qing Liu
- Department of Pharmacy, Tangdu Hospital, Fourth Military Medical University, Xi'an, China
| | - Guang-Bin Cui
- Department of Radiology and Functional and Molecular Imaging Key Lab of Shaanxi Province, Tangdu Hospital, Fourth Military Medical University, Xi'an, China.
| | - Hai-Yan Nan
- Department of Radiology and Functional and Molecular Imaging Key Lab of Shaanxi Province, Tangdu Hospital, Fourth Military Medical University, Xi'an, China.
| |
Collapse
|
11
|
Zaabalawi A, Renshall L, Beards F, Lightfoot AP, Degens H, Alexander Y, Hasan R, Bilal H, Graf BA, Harris LK, Azzawi M. Internal Mammary Arteries as a Model to Demonstrate Restoration of the Impaired Vasodilation in Hypertension, Using Liposomal Delivery of the CYP1B1 Inhibitor, 2,3',4,5'-Tetramethoxystilbene. Pharmaceutics 2022; 14:2046. [PMID: 36297480 PMCID: PMC9611804 DOI: 10.3390/pharmaceutics14102046] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2022] [Revised: 09/20/2022] [Accepted: 09/22/2022] [Indexed: 11/23/2022] Open
Abstract
A significant number of patients with severe cardiovascular disease, undergoing coronary artery bypass grafting (CABG), present with hypertension. While internal mammary arteries (IMAs) may be a better alternative to vein grafts, their impaired vasodilator function affects their patency. Our objectives were to (1) determine if inhibition of the cytochrome P450 enzyme CYP1B1, using liposome-encapsulated 2,3′,4,5′-tetramethoxystilbene (TMS), can potentiate vasodilation of IMAs from CABG patients, and (2) assess mechanisms involved using coronary arteries from normal rats, in an ex vivo model of hypertension. PEGylated liposomes were synthesized and loaded with TMS (mean diameter 141 ± 0.9 nm). Liposomal delivery of TMS improved its bioavailability Compared to TMS solution (0.129 ± 0.02 ng/mL vs. 0.086 ± 0.01 ng/mL at 4 h; p < 0.05). TMS-loaded liposomes alleviated attenuated endothelial-dependent acetylcholine (ACh)-induced dilation in diseased IMAs (@ACh 10−4 M: 56.9 ± 5.1%; n = 8 vs. 12.7 ± 7.8%; n = 6; p < 0.01) for TMS-loaded liposomes vs. blank liposomes, respectively. The alleviation in dilation may be due to the potent inhibition of CYP1B1 by TMS, and subsequent reduction in reactive oxygen species (ROS) moieties and stimulation of nitric oxide synthesis. In isolated rat coronary arteries exposed to a hypertensive environment, TMS-loaded liposomes potentiated nitric oxide and endothelium-derived hyperpolarization pathways via AMPK. Our findings are promising for the future development of TMS-loaded liposomes as a promising therapeutic strategy to enhance TMS bioavailability and potentiate vasodilator function in hypertension, with relevance for early and long-term treatment of CABG patients, via the sustained and localized TMS release within IMAs.
Collapse
Affiliation(s)
- Azziza Zaabalawi
- Department of Life Sciences, Manchester Metropolitan University, Manchester M1 5GD, UK
| | - Lewis Renshall
- Division of Pharmacy and Optometry, University of Manchester, Manchester M13 9PL, UK
- Maternal & Fetal Health Research Centre, University of Manchester, Manchester M13 9WL, UK
- Manchester Academic Health Science Centre, Central Manchester University Hospitals NHS Foundation Trust, Manchester M13 9WL, UK
| | - Frances Beards
- Division of Pharmacy and Optometry, University of Manchester, Manchester M13 9PL, UK
- Maternal & Fetal Health Research Centre, University of Manchester, Manchester M13 9WL, UK
- Manchester Academic Health Science Centre, Central Manchester University Hospitals NHS Foundation Trust, Manchester M13 9WL, UK
| | - Adam P. Lightfoot
- Department of Life Sciences, Manchester Metropolitan University, Manchester M1 5GD, UK
- School of Pharmacy and Biomolecular Sciences, Liverpool John Moores University, Liverpool L3 3AP, UK
| | - Hans Degens
- Department of Life Sciences, Manchester Metropolitan University, Manchester M1 5GD, UK
- Institute of Sport Science and Innovations, Lithuanian Sports University, 44221 Kaunas, Lithuania
| | - Yvonne Alexander
- Department of Life Sciences, Manchester Metropolitan University, Manchester M1 5GD, UK
| | - Ragheb Hasan
- Department of Cardiothoracic Surgery, Manchester Foundation Trust, Manchester M13 9WL, UK
| | - Haris Bilal
- Department of Cardiothoracic Surgery, Manchester Foundation Trust, Manchester M13 9WL, UK
| | - Brigitte A. Graf
- Faculty of Health and Education, Manchester Metropolitan University, Manchester M15 6BG, UK
| | - Lynda K. Harris
- Division of Pharmacy and Optometry, University of Manchester, Manchester M13 9PL, UK
- Maternal & Fetal Health Research Centre, University of Manchester, Manchester M13 9WL, UK
- Manchester Academic Health Science Centre, Central Manchester University Hospitals NHS Foundation Trust, Manchester M13 9WL, UK
| | - May Azzawi
- Department of Life Sciences, Manchester Metropolitan University, Manchester M1 5GD, UK
| |
Collapse
|
12
|
Sahinturk S, Demirel S, Ozyener F, Isbil N. Vascular Functional Effect Mechanisms of Elabela in Rat Thoracic Aorta. Ann Vasc Surg 2022; 84:381-397. [PMID: 35472496 DOI: 10.1016/j.avsg.2022.04.033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2022] [Revised: 04/14/2022] [Accepted: 04/19/2022] [Indexed: 11/20/2022]
Abstract
BACKGROUND Elabela is a recently discovered peptide hormone. The present study aims to investigate the vasorelaxant effect mechanisms of elabela in the rat thoracic aorta. METHODS The vascular rings obtained from the thoracic aortas of the male Wistar albino rats were placed in the isolated tissue bath system. Resting tension was set to 1 gram. After the equilibration period, the vessel rings were contracted with phenylephrine or potassium chloride. Once a stable contraction was achieved, elabela-32 was applied cumulatively (10-9-10-6 molar) to the vascular rings. The experimental protocol was repeated in the presence of specific signaling pathway inhibitors or potassium channel blockers to determine the effect mechanisms of elabela. RESULTS Elabela showed a significant vasorelaxant effect in a concentration-dependent manner (P < 0.001). The vasorelaxant effect level of elabela was significantly reduced by the apelin receptor antagonist F13A, cyclooxygenase inhibitor indomethacin, adenosine monophosphate-activated protein kinase inhibitor dorsomorphin, protein kinase C inhibitor bisindolmaleimide, large-conductance calcium-activated potassium channel blocker iberiotoxin, and intermediate-conductance calcium-activated potassium channel blocker TRAM-34 (P < 0.001). However, the vasorelaxant effect level of elabela was not significantly affected by the endothelial nitric oxide synthase inhibitor nitro-L-arginine methyl ester and mitogen-activated protein kinase inhibitor U0126. CONCLUSIONS Elabela exhibits a prominent vasodilator effect in rat thoracic aorta. Apelin receptor, prostanoids, adenosine monophosphate-activated protein kinase, protein kinase C, and calcium-activated potassium channels are involved in the vasorelaxant effect mechanisms of elabela.
Collapse
Affiliation(s)
- Serdar Sahinturk
- Physiology Department, Bursa Uludag University Medicine School, Bursa, Turkey.
| | - Sadettin Demirel
- Physiology Department, Bursa Uludag University Medicine School, Bursa, Turkey
| | - Fadil Ozyener
- Physiology Department, Bursa Uludag University Medicine School, Bursa, Turkey
| | - Naciye Isbil
- Physiology Department, Bursa Uludag University Medicine School, Bursa, Turkey
| |
Collapse
|
13
|
Nemirovskaya TL, Sharlo KA. Roles of ATP and SERCA in the Regulation of Calcium Turnover in Unloaded Skeletal Muscles: Current View and Future Directions. Int J Mol Sci 2022; 23:ijms23136937. [PMID: 35805949 PMCID: PMC9267070 DOI: 10.3390/ijms23136937] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Accepted: 06/20/2022] [Indexed: 11/16/2022] Open
Abstract
A decrease in skeletal muscle contractile activity or its complete cessation (muscle unloading or disuse) leads to muscle fibers’ atrophy and to alterations in muscle performance. These changes negatively affect the quality of life of people who, for one reason or another, are forced to face a limitation of physical activity. One of the key regulatory events leading to the muscle disuse-induced changes is an impairment of calcium homeostasis, which leads to the excessive accumulation of calcium ions in the sarcoplasm. This review aimed to analyze the triggering mechanisms of calcium homeostasis impairment (including those associated with the accumulation of high-energy phosphates) under various types of muscle unloading. Here we proposed a hypothesis about the regulatory mechanisms of SERCA and IP3 receptors activity during muscle unloading, and about the contribution of these mechanisms to the excessive calcium ion myoplasmic accumulation and gene transcription regulation via excitation–transcription coupling.
Collapse
|
14
|
Mohd Nor NA, Budin SB, Zainalabidin S, Jalil J, Sapian S, Jubaidi FF, Mohamad Anuar NN. The Role of Polyphenol in Modulating Associated Genes in Diabetes-Induced Vascular Disorders. Int J Mol Sci 2022; 23:6396. [PMID: 35742837 PMCID: PMC9223817 DOI: 10.3390/ijms23126396] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2022] [Revised: 06/02/2022] [Accepted: 06/04/2022] [Indexed: 01/05/2023] Open
Abstract
Diabetes-induced vascular disorder is considered one of the deadly risk factors among diabetic patients that are caused by persistent hyperglycemia that eventually leads to cardiovascular diseases. Elevated reactive oxygen species (ROS) due to high blood glucose levels activate signaling pathways such as AGE/RAGE, PKC, polyol, and hexosamine pathways. The activated signaling pathway triggers oxidative stress, inflammation, and apoptosis which later lead to vascular dysfunction induced by diabetes. Polyphenol is a bioactive compound that can be found abundantly in plants such as vegetables, fruits, whole grains, and nuts. This compound exerts therapeutic effects in alleviating diabetes-induced vascular disorder, mainly due to its potential as an anti-oxidative, anti-inflammatory, and anti-apoptotic agent. In this review, we sought to summarize the recent discovery of polyphenol treatments in modulating associated genes involved in the progression of diabetes-induced vascular disorder.
Collapse
Affiliation(s)
- Nor Anizah Mohd Nor
- Centre for Diagnostic, Therapeutic and Investigative Studies, Faculty of Health Sciences, Universiti Kebangsaan Malaysia, Kuala Lumpur 50300, Malaysia; (N.A.M.N.); (S.B.B.); (S.S.); (F.F.J.)
- PICOMS International University College, Taman Batu Muda, Batu Caves, Kuala Lumpur 68100, Malaysia
| | - Siti Balkis Budin
- Centre for Diagnostic, Therapeutic and Investigative Studies, Faculty of Health Sciences, Universiti Kebangsaan Malaysia, Kuala Lumpur 50300, Malaysia; (N.A.M.N.); (S.B.B.); (S.S.); (F.F.J.)
| | - Satirah Zainalabidin
- Programme of Biomedical Science, Centre for Toxicology and Health Risk Research, Faculty of Health Sciences, Universiti Kebangsaan Malaysia, Kuala Lumpur 50300, Malaysia;
| | - Juriyati Jalil
- Center for Drug and Herbal Development, Faculty of Pharmacy, Universiti Kebangsaan Malaysia, Kuala Lumpur 50300, Malaysia;
| | - Syaifuzah Sapian
- Centre for Diagnostic, Therapeutic and Investigative Studies, Faculty of Health Sciences, Universiti Kebangsaan Malaysia, Kuala Lumpur 50300, Malaysia; (N.A.M.N.); (S.B.B.); (S.S.); (F.F.J.)
| | - Fatin Farhana Jubaidi
- Centre for Diagnostic, Therapeutic and Investigative Studies, Faculty of Health Sciences, Universiti Kebangsaan Malaysia, Kuala Lumpur 50300, Malaysia; (N.A.M.N.); (S.B.B.); (S.S.); (F.F.J.)
| | - Nur Najmi Mohamad Anuar
- Programme of Biomedical Science, Centre for Toxicology and Health Risk Research, Faculty of Health Sciences, Universiti Kebangsaan Malaysia, Kuala Lumpur 50300, Malaysia;
| |
Collapse
|
15
|
Virginia DM, Patramurti C, Fenty, Setiawan CH, Julianus J, Hendra P, Susanto NAP. Single Nucleotide Polymorphism in the 3' Untranslated Region of PRKAA2 on Cardiometabolic Parameters in Type 2 Diabetes Mellitus Patients Who Received Metformin. Ther Clin Risk Manag 2022; 18:349-357. [PMID: 35414746 PMCID: PMC8995000 DOI: 10.2147/tcrm.s349900] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2021] [Accepted: 03/14/2022] [Indexed: 11/29/2022] Open
Abstract
Purpose This study aimed to explore the association of rs857148 A>C as 3'UTR variants with blood pressure, HbA1c profile, and lipid profiles as cardiometabolic parameters among patients with T2DM receiving metformin. Patients and Methods This cross-sectional analytic research was conducted with 114 consecutively selected patients with T2DM. Polymerase chain reaction-restriction fragment length polymorphism was conducted to determine rs857148. A total of 108 patients fulfilled inclusion and exclusion criteria. Results Genotype distribution agreed with the Hardy Weinberg Equation for Equilibrium (p>0.05) but wildtype allele was found as the minor allele. Subjects with CC genotype and C allele had enhanced HbA1c levels (OR=7.12; 95% CI=1.05-48.26; p=0.04; OR=1.66; 95% CI=1.06-2.60; p=0.03, respectively). It was confirmed by dominant model whereas subjects with AA tended to have reduced HbA1c compared to AC+CC genotype (OR=0.15; 95% CI=0.02-0.97; p=0.047). AC genotype had significant correlation to total cholesterol (OR=1.05; 95% CI=1.01-1.10; p=0.03) compared to AA genotype. Conclusion We conclude that polymorphism of rs87148, specifically CC genotype and C allele, has a significant association with HbA1c and total cholesterol after considering oral hypoglycemia agent dose, age, gender, and combination therapy, compared to AA genotype. Future studies that involve a larger sample population and more rigorous selection criteria are required.
Collapse
Affiliation(s)
- Dita Maria Virginia
- Department of Pharmacology and Clinical Pharmacy, Faculty of Pharmacy, Sanata Dharma University, Yogyakarta, Indonesia
| | - Christine Patramurti
- Department of Pharmaceutical Analysis and Medicinal Chemistry, Faculty of Pharmacy, Sanata Dharma University, Yogyakarta, Indonesia
| | - Fenty
- Department of Pharmacology and Clinical Pharmacy, Faculty of Pharmacy, Sanata Dharma University, Yogyakarta, Indonesia
- Bethesda Lempuyangwangi Hospital, Yogyakarta, Indonesia
| | - Christianus Heru Setiawan
- Department of Pharmacology and Clinical Pharmacy, Faculty of Pharmacy, Sanata Dharma University, Yogyakarta, Indonesia
| | - Jeffry Julianus
- Department of Pharmaceutical Analysis and Medicinal Chemistry, Faculty of Pharmacy, Sanata Dharma University, Yogyakarta, Indonesia
| | - Phebe Hendra
- Department of Pharmacology and Clinical Pharmacy, Faculty of Pharmacy, Sanata Dharma University, Yogyakarta, Indonesia
| | | |
Collapse
|
16
|
Heidary Moghaddam R, Samimi Z, Asgary S, Mohammadi P, Hozeifi S, Hoseinzadeh-Chahkandak F, Xu S, Farzaei MH. Natural AMPK Activators in Cardiovascular Disease Prevention. Front Pharmacol 2022; 12:738420. [PMID: 35046800 PMCID: PMC8762275 DOI: 10.3389/fphar.2021.738420] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2021] [Accepted: 11/03/2021] [Indexed: 12/11/2022] Open
Abstract
Cardiovascular diseases (CVD), as a life-threatening global disease, is receiving worldwide attention. Seeking novel therapeutic strategies and agents is of utmost importance to curb CVD. AMP-activated protein kinase (AMPK) activators derived from natural products are promising agents for cardiovascular drug development owning to regulatory effects on physiological processes and diverse cardiometabolic disorders. In the past decade, different therapeutic agents from natural products and herbal medicines have been explored as good templates of AMPK activators. Hereby, we overviewed the role of AMPK signaling in the cardiovascular system, as well as evidence implicating AMPK activators as potential therapeutic tools. In the present review, efforts have been made to compile and update relevant information from both preclinical and clinical studies, which investigated the role of natural products as AMPK activators in cardiovascular therapeutics.
Collapse
Affiliation(s)
- Reza Heidary Moghaddam
- Clinical Research Development Center, Imam Ali and Taleghani Hospital, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Zeinab Samimi
- Pharmaceutical Sciences Research Center, Health Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Sedigheh Asgary
- Isfahan Cardiovascular Research Center, Cardiovascular Research Institute,.Isfahan University of Medical Sciences, Isfahan, Iran
| | - Pantea Mohammadi
- Medical Biology Research Center, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Soroush Hozeifi
- School of Medicine, Birjand University of Medical Sciences, Birjand, Iran
| | | | - Suowen Xu
- Department of Endocrinology and Metabolism, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Mohammad Hosein Farzaei
- Pharmaceutical Sciences Research Center, Health Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran.,Medical Technology Research Center, Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran
| |
Collapse
|
17
|
Virginia DM, Wahyuningsih MSH, Nugrahaningsih DAA. PRKAA2 variation and the clinical characteristics of patients newly diagnosed with type 2 diabetes mellitus in Yogyakarta, Indonesia. ASIAN BIOMED 2021; 15:161-170. [PMID: 37551330 PMCID: PMC10388783 DOI: 10.2478/abm-2021-0021] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Background Adenosine monophosphate (AMP)-activated protein kinase (AMPK; EC 2.7.11.31) enzymes play a pivotal role in cell metabolism. They are involved in type 2 diabetes mellitus (T2DM) pathogenesis. Genetic variation of PRKAA2 coding for the AMPK α2 catalytic subunit (AMPKα2) is reported to be associated with susceptibility for T2DM. Objectives To determine the association between PRKAA2 genetic variations (rs2796498, rs9803799, and rs2746342) with clinical characteristics in patients newly diagnosed with T2DM. Methods We performed a cross-sectional study including 166 T2DM patients from 10 primary health care centers in Yogyakarta, Indonesia. We measured fasting plasma glucose, hemoglobin A1c, serum creatinine, glomerular filtration rate, blood pressure, and body mass index as clinical characteristics. PRKAA2 genetic variations were determined by TaqMan SNP genotyping assay. Hardy-Weinberg equilibrium was calculated using χ2 tests. Results There was no difference in clinical characteristics for genotypes rs2796498, rs9803799, or rs2746342 (P > 0.05). No significant association was found between PRKAA2 genetic variations and any clinical feature observed. Further subgroup analysis adjusting for age, sex, and waist circumference did not detect any significant association of PRKAA2 genetic variations with clinical characteristics (P > 0.05). Conclusion PRKAA2 genetic variation is not associated with the clinical characteristics of Indonesian patients with newly diagnosed T2DM.
Collapse
Affiliation(s)
- Dita Maria Virginia
- Department of Pharmacology and Therapy, Faculty of Medicine, Public Health and Nursing, Universitas Gadjah Mada, Yogyakarta55281, Indonesia
- Faculty of Pharmacy, Universitas Sanata Dharma, Yogyakarta552181, Indonesia
| | - Mae Sri Hartati Wahyuningsih
- Department of Pharmacology and Therapy, Faculty of Medicine, Public Health and Nursing, Universitas Gadjah Mada, Yogyakarta55281, Indonesia
| | - Dwi Aris Agung Nugrahaningsih
- Department of Pharmacology and Therapy, Faculty of Medicine, Public Health and Nursing, Universitas Gadjah Mada, Yogyakarta55281, Indonesia
- Center of Genetic Study, Faculty of Medicine, Public Health and Nursing, Universitas Gadjah Mada, Yogyakarta55281, Indonesia
| |
Collapse
|
18
|
Rodríguez C, Muñoz M, Contreras C, Prieto D. AMPK, metabolism, and vascular function. FEBS J 2021; 288:3746-3771. [PMID: 33825330 DOI: 10.1111/febs.15863] [Citation(s) in RCA: 64] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Revised: 03/04/2021] [Accepted: 04/04/2021] [Indexed: 12/12/2022]
Abstract
Adenosine monophosphate-activated protein kinase (AMPK) is a cellular energy sensor activated during energy stress that plays a key role in maintaining energy homeostasis. This ubiquitous signaling pathway has been implicated in multiple functions including mitochondrial biogenesis, redox regulation, cell growth and proliferation, cell autophagy and inflammation. The protective role of AMPK in cardiovascular function and the involvement of dysfunctional AMPK in the pathogenesis of cardiovascular disease have been highlighted in recent years. In this review, we summarize and discuss the role of AMPK in the regulation of blood flow in response to metabolic demand and the basis of the AMPK physiological anticontractile, antioxidant, anti-inflammatory, and antiatherogenic actions in the vascular system. Investigations by others and us have demonstrated the key role of vascular AMPK in the regulation of endothelial function, redox homeostasis, and inflammation, in addition to its protective role in the hypoxia and ischemia/reperfusion injury. The pathophysiological implications of AMPK involvement in vascular function with regard to the vascular complications of metabolic disease and the therapeutic potential of AMPK activators are also discussed.
Collapse
Affiliation(s)
- Claudia Rodríguez
- Departamento de Fisiología, Facultad de Farmacia, Universidad Complutense, Madrid, Spain
| | - Mercedes Muñoz
- Departamento de Fisiología, Facultad de Farmacia, Universidad Complutense, Madrid, Spain
| | - Cristina Contreras
- Departamento de Fisiología, Facultad de Farmacia, Universidad Complutense, Madrid, Spain
| | - Dolores Prieto
- Departamento de Fisiología, Facultad de Farmacia, Universidad Complutense, Madrid, Spain
| |
Collapse
|
19
|
Ottolini M, Sonkusare SK. The Calcium Signaling Mechanisms in Arterial Smooth Muscle and Endothelial Cells. Compr Physiol 2021; 11:1831-1869. [PMID: 33792900 PMCID: PMC10388069 DOI: 10.1002/cphy.c200030] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
The contractile state of resistance arteries and arterioles is a crucial determinant of blood pressure and blood flow. Physiological regulation of arterial contractility requires constant communication between endothelial and smooth muscle cells. Various Ca2+ signals and Ca2+ -sensitive targets ensure dynamic control of intercellular communications in the vascular wall. The functional effect of a Ca2+ signal on arterial contractility depends on the type of Ca2+ -sensitive target engaged by that signal. Recent studies using advanced imaging methods have identified the spatiotemporal signatures of individual Ca2+ signals that control arterial and arteriolar contractility. Broadly speaking, intracellular Ca2+ is increased by ion channels and transporters on the plasma membrane and endoplasmic reticular membrane. Physiological roles for many vascular Ca2+ signals have already been confirmed, while further investigation is needed for other Ca2+ signals. This article focuses on endothelial and smooth muscle Ca2+ signaling mechanisms in resistance arteries and arterioles. We discuss the Ca2+ entry pathways at the plasma membrane, Ca2+ release signals from the intracellular stores, the functional and physiological relevance of Ca2+ signals, and their regulatory mechanisms. Finally, we describe the contribution of abnormal endothelial and smooth muscle Ca2+ signals to the pathogenesis of vascular disorders. © 2021 American Physiological Society. Compr Physiol 11:1831-1869, 2021.
Collapse
Affiliation(s)
- Matteo Ottolini
- Department of Pharmacology, University of Virginia, Charlottesville, Virginia, USA
| | - Swapnil K Sonkusare
- Department of Pharmacology, University of Virginia, Charlottesville, Virginia, USA.,Department of Molecular Physiology & Biological Physics, University of Virginia, Charlottesville, Virginia, USA.,Robert M. Berne Cardiovascular Research Center, University of Virginia, Charlottesville, Virginia, USA
| |
Collapse
|
20
|
The role of AMPK in regulation of Na +,K +-ATPase in skeletal muscle: does the gauge always plug the sink? J Muscle Res Cell Motil 2021; 42:77-97. [PMID: 33398789 DOI: 10.1007/s10974-020-09594-3] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2020] [Accepted: 12/14/2020] [Indexed: 12/14/2022]
Abstract
AMP-activated protein kinase (AMPK) is a cellular energy gauge and a major regulator of cellular energy homeostasis. Once activated, AMPK stimulates nutrient uptake and the ATP-producing catabolic pathways, while it suppresses the ATP-consuming anabolic pathways, thus helping to maintain the cellular energy balance under energy-deprived conditions. As much as ~ 20-25% of the whole-body ATP consumption occurs due to a reaction catalysed by Na+,K+-ATPase (NKA). Being the single most important sink of energy, NKA might seem to be an essential target of the AMPK-mediated energy saving measures, yet NKA is vital for maintenance of transmembrane Na+ and K+ gradients, water homeostasis, cellular excitability, and the Na+-coupled transport of nutrients and ions. Consistent with the model that AMPK regulates ATP consumption by NKA, activation of AMPK in the lung alveolar cells stimulates endocytosis of NKA, thus suppressing the transepithelial ion transport and the absorption of the alveolar fluid. In skeletal muscles, contractions activate NKA, which opposes a rundown of transmembrane ion gradients, as well as AMPK, which plays an important role in adaptations to exercise. Inhibition of NKA in contracting skeletal muscle accentuates perturbations in ion concentrations and accelerates development of fatigue. However, different models suggest that AMPK does not inhibit or even stimulates NKA in skeletal muscle, which appears to contradict the idea that AMPK maintains the cellular energy balance by always suppressing ATP-consuming processes. In this short review, we examine the role of AMPK in regulation of NKA in skeletal muscle and discuss the apparent paradox of AMPK-stimulated ATP consumption.
Collapse
|
21
|
Haye A, Ansari MA, Rahman SO, Shamsi Y, Ahmed D, Sharma M. Role of AMP-activated protein kinase on cardio-metabolic abnormalities in the development of diabetic cardiomyopathy: A molecular landscape. Eur J Pharmacol 2020; 888:173376. [PMID: 32810493 DOI: 10.1016/j.ejphar.2020.173376] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2020] [Revised: 07/10/2020] [Accepted: 07/13/2020] [Indexed: 12/13/2022]
Abstract
Cardiovascular complications associated with diabetes mellitus remains a leading cause of morbidity and mortality across the world. Diabetic cardiomyopathy is a descriptive pathology that in absence of co-morbidities such as hypertension, dyslipidemia initially characterized by cardiac stiffness, myocardial fibrosis, ventricular hypertrophy, and remodeling. These abnormalities further contribute to diastolic dysfunctions followed by systolic dysfunctions and eventually results in clinical heart failure (HF). The clinical outcomes associated with HF are considerably worse in patients with diabetes. The complexity of the pathogenesis and clinical features of diabetic cardiomyopathy raises serious questions in developing a therapeutic strategy to manage cardio-metabolic abnormalities. Despite extensive research in the past decade the compelling approaches to manage and treat diabetic cardiomyopathy are limited. AMP-Activated Protein Kinase (AMPK), a serine-threonine kinase, often referred to as cellular "metabolic master switch". During the development and progression of diabetic cardiomyopathy, a plethora of evidence demonstrate the beneficial role of AMPK on cardio-metabolic abnormalities including altered substrate utilization, impaired cardiac insulin metabolic signaling, mitochondrial dysfunction and oxidative stress, myocardial inflammation, increased accumulation of advanced glycation end-products, impaired cardiac calcium handling, maladaptive activation of the renin-angiotensin-aldosterone system, endoplasmic reticulum stress, myocardial fibrosis, ventricular hypertrophy, cardiac apoptosis, and impaired autophagy. Therefore, in this review, we have summarized the findings from pre-clinical and clinical studies and provided a collective overview of the pathophysiological mechanism and the regulatory role of AMPK on cardio-metabolic abnormalities during the development of diabetic cardiomyopathy.
Collapse
Affiliation(s)
- Abdul Haye
- Pharmaceutical Medicine, Department of Pharmacology, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, 110062, India
| | - Mohd Asif Ansari
- Department of Pharmacology, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, 110062, India
| | - Syed Obaidur Rahman
- Pharmaceutical Medicine, Department of Pharmacology, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, 110062, India
| | - Yasmeen Shamsi
- Department of Moalejat, School of Unani Medical Education and Research, Jamia Hamdard, New Delhi, 110062, India
| | - Danish Ahmed
- Department of Pharmaceutical Sciences, Faculty of Health Sciences, Sam Higginbottom University of Agriculture Technology and Sciences, Allahabad, Uttar Pradesh, India
| | - Manju Sharma
- Department of Pharmacology, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, 110062, India.
| |
Collapse
|
22
|
Rogacka D, Audzeyenka I, Piwkowska A. Regulation of podocytes function by AMP-activated protein kinase. Arch Biochem Biophys 2020; 692:108541. [PMID: 32781053 DOI: 10.1016/j.abb.2020.108541] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2020] [Revised: 07/23/2020] [Accepted: 08/06/2020] [Indexed: 01/08/2023]
Abstract
Podocytes are unique, highly specialized, terminally differentiated cells that form an essential, integral part of the glomerular filter. These cells limit the outside border of the glomerular basement membrane, forming a tight barrier that prevents significant protein loss from the capillary space. The slit diaphragm formed by podocytes is crucial for maintaining glomerular integrity and function. They are the target of injury in many glomerular diseases, including hypertension and diabetes mellitus. Accumulating studies have revealed that AMP-activated protein kinase (AMPK), an essential cellular energy sensor, might play a fundamental role in regulating podocyte metabolism and function. AMPK participates in insulin signaling, therefore controls glucose uptake and podocytes insulin sensitivity. It is also involved in insulin-dependent cytoskeleton reorganization in podocytes, mediating glomerular albumin permeability. AMPK plays an important role in the regulation of autophagy/apoptosis processes, which influence podocytes viability. The present review aimed to highlight the molecular mechanisms associated with AMPK that are involved in the regulation of podocyte function in health and disease states.
Collapse
Affiliation(s)
- Dorota Rogacka
- Mossakowski Medical Research Centre Polish Academy of Sciences, Laboratory of Molecular and Cellular Nephrology, Wita Stwosza 63, 80-308, Gdansk, Poland; University of Gdansk, Faculty of Chemistry, Department of Molecular Biotechnology, Wita Stwosza 63, 80-308, Gdansk, Poland.
| | - Irena Audzeyenka
- Mossakowski Medical Research Centre Polish Academy of Sciences, Laboratory of Molecular and Cellular Nephrology, Wita Stwosza 63, 80-308, Gdansk, Poland; University of Gdansk, Faculty of Chemistry, Department of Molecular Biotechnology, Wita Stwosza 63, 80-308, Gdansk, Poland.
| | - Agnieszka Piwkowska
- Mossakowski Medical Research Centre Polish Academy of Sciences, Laboratory of Molecular and Cellular Nephrology, Wita Stwosza 63, 80-308, Gdansk, Poland; University of Gdansk, Faculty of Chemistry, Department of Molecular Biotechnology, Wita Stwosza 63, 80-308, Gdansk, Poland.
| |
Collapse
|
23
|
Lorca RA, Matarazzo CJ, Bales ES, Houck JA, Orlicky DJ, Euser AG, Julian CG, Moore LG. AMPK activation in pregnant human myometrial arteries from high-altitude and intrauterine growth-restricted pregnancies. Am J Physiol Heart Circ Physiol 2020; 319:H203-H212. [PMID: 32502374 DOI: 10.1152/ajpheart.00644.2019] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
High-altitude (>2,500 m) residence increases the incidence of intrauterine growth restriction (IUGR) due, in part, to reduced uterine artery blood flow and impaired myometrial artery (MA) vasodilator response. A role for the AMP-activated protein kinase (AMPK) pathway in protecting against hypoxia-associated IUGR is suggested by genomic and transcriptomic studies in humans and functional studies in mice. AMPK is a hypoxia-sensitive metabolic sensor with vasodilatory properties. Here we hypothesized that AMPK-dependent vasodilation was increased in MAs from high versus low-altitude (<1,700 m) Colorado women with appropriate for gestational age (AGA) pregnancies and reduced in IUGR pregnancies regardless of altitude. Vasoreactivity studies showed that, in AGA pregnancies, MAs from high-altitude women were more sensitive to vasodilation by activation of AMPK with A769662 due chiefly to increased endothelial nitric oxide production, whereas MA responses to AMPK activation in the low-altitude women were endothelium independent. MAs from IUGR compared with AGA pregnancies had blunted vasodilator responses to acetylcholine at high altitude. We concluded that 1) blunted vasodilator responses in IUGR pregnancies confirm the importance of MA vasodilation for normal fetal growth and 2) the increased sensitivity to AMPK activation in AGA pregnancies at high altitude suggests that AMPK activation helped maintain MA vasodilation and fetal growth. These results highlight a novel mechanism for vasodilation of MAs under conditions of chronic hypoxia and suggest that AMPK activation could provide a therapy for increasing uteroplacental blood flow and improving fetal growth in IUGR pregnancies.NEW & NOTEWORTHY Intrauterine growth restriction (IUGR) impairs infant well- being and increases susceptibility to later-in-life diseases for mother and child. Our study reveals a novel role for AMPK in vasodilating the myometrial artery (MA) from women residing at high altitude (>2,500 m) with appropriate for gestational age pregnancies but not in IUGR pregnancies at any altitude.
Collapse
Affiliation(s)
- Ramón A Lorca
- Division of Reproductive Sciences, Department of Obstetrics and Gynecology, University of Colorado Denver-Anschutz Medical Campus, Aurora, Colorado
| | - Christopher J Matarazzo
- Division of Biomedical Informatics and Personalized Medicine, Department of Medicine, University of Colorado Denver-Anschutz Medical Campus, Aurora, Colorado
| | - Elise S Bales
- Division of Reproductive Sciences, Department of Obstetrics and Gynecology, University of Colorado Denver-Anschutz Medical Campus, Aurora, Colorado
| | - Julie A Houck
- Division of Biomedical Informatics and Personalized Medicine, Department of Medicine, University of Colorado Denver-Anschutz Medical Campus, Aurora, Colorado
| | - David J Orlicky
- Department of Pathology, University of Colorado Denver-Anschutz Medical Campus, Aurora, Colorado
| | - Anna G Euser
- Division of Maternal-Fetal Medicine, Department of Obstetrics and Gynecology, University of Colorado Denver-Anschutz Medical Campus, Aurora, Colorado
| | - Colleen G Julian
- Division of Biomedical Informatics and Personalized Medicine, Department of Medicine, University of Colorado Denver-Anschutz Medical Campus, Aurora, Colorado
| | - Lorna G Moore
- Division of Reproductive Sciences, Department of Obstetrics and Gynecology, University of Colorado Denver-Anschutz Medical Campus, Aurora, Colorado
| |
Collapse
|
24
|
Rodríguez C, Contreras C, Sáenz-Medina J, Muñoz M, Corbacho C, Carballido J, García-Sacristán A, Hernandez M, López M, Rivera L, Prieto D. Activation of the AMP-related kinase (AMPK) induces renal vasodilatation and downregulates Nox-derived reactive oxygen species (ROS) generation. Redox Biol 2020. [PMID: 32470915 DOI: 10.1016/j.redox.2020.101575.] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
AMP-activated protein kinase (AMPK) is a cellular energy sensor activated during energy stress to stimulate ATP production pathways and restore homeostasis. AMPK is widely expressed in the kidney and involved in mitochondrial protection and biogenesis upon acute renal ischemia, AMPK activity being blunted in metabolic disease-associated kidney disease. Since little is known about AMPK in the regulation of renal blood flow, the present study aimed to assess the role of AMPK in renal vascular function. Functional responses to the selective AMPK activator A769662 were assessed in intrarenal small arteries isolated from the kidney of renal tumour patients and Wistar rats and mounted in microvascular myographs to perform simultaneous measurements of intracellular calcium [Ca2+]i and tension. Superoxide (O2.-) and hydrogen peroxide (H2O2) production were measured by chemiluminescence and fluorescence and protein expression by Western blot. Activation of AMPK with A769662 increased AMPKα phosphorylation at Thr-172 and induced potent relaxations compared to AICAR in isolated human and rat intrarenal arteries, through both endothelium-dependent mechanisms involving nitric oxide (NO) and intermediate-conductance calcium-activated potassium (IKCa) channels, as well as activation of ATP-sensitive (KATP) channels and sarcoplasmic reticulum Ca2+-ATPase (SERCA) in vascular smooth muscle (VSM). Furthermore, AMPK activator reduced NADPH oxidase 4 (Nox4) and Nox2-derived reactive oxygen species (ROS) production. These results demonstrate that A769662 has potent vasodilator and antioxidant effects in intrarenal arteries. The benefits of AMPK activation in rat kidney are reproduced in human arteries and therefore vascular AMPK activation might be a therapeutic target in the treatment of metabolic disease-associated kidney injury.
Collapse
Affiliation(s)
- Claudia Rodríguez
- Departamento de Fisiología, Facultad de Farmacia, Universidad Complutense, Madrid, Spain
| | - Cristina Contreras
- Departamento de Fisiología, Facultad de Farmacia, Universidad Complutense, Madrid, Spain
| | - Javier Sáenz-Medina
- Departamento de Urología, Hospital Universitario Puerta de Hierro-Majadahonda, Madrid, Spain
| | - Mercedes Muñoz
- Departamento de Fisiología, Facultad de Farmacia, Universidad Complutense, Madrid, Spain
| | - César Corbacho
- Departamento de Anatomía Patológica, Hospital Universitario Puerta de Hierro-Majadahonda, Madrid, Spain
| | - Joaquín Carballido
- Departamento de Urología, Hospital Universitario Puerta de Hierro-Majadahonda, Madrid, Spain
| | | | - Medardo Hernandez
- Departamento de Fisiología, Facultad de Farmacia, Universidad Complutense, Madrid, Spain
| | - Miguel López
- NeurObesity Group, Department of Physiology, CIMUS, Universidad de Santiago de Compostela-Instituto de Investigación Sanitaria, Santiago de Compostela, Spain
| | - Luis Rivera
- Departamento de Fisiología, Facultad de Farmacia, Universidad Complutense, Madrid, Spain
| | - Dolores Prieto
- Departamento de Fisiología, Facultad de Farmacia, Universidad Complutense, Madrid, Spain.
| |
Collapse
|
25
|
Activation of the AMP-related kinase (AMPK) induces renal vasodilatation and downregulates Nox-derived reactive oxygen species (ROS) generation. Redox Biol 2020; 34:101575. [PMID: 32470915 PMCID: PMC7256643 DOI: 10.1016/j.redox.2020.101575] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2020] [Accepted: 05/10/2020] [Indexed: 12/19/2022] Open
Abstract
AMP-activated protein kinase (AMPK) is a cellular energy sensor activated during energy stress to stimulate ATP production pathways and restore homeostasis. AMPK is widely expressed in the kidney and involved in mitochondrial protection and biogenesis upon acute renal ischemia, AMPK activity being blunted in metabolic disease-associated kidney disease. Since little is known about AMPK in the regulation of renal blood flow, the present study aimed to assess the role of AMPK in renal vascular function. Functional responses to the selective AMPK activator A769662 were assessed in intrarenal small arteries isolated from the kidney of renal tumour patients and Wistar rats and mounted in microvascular myographs to perform simultaneous measurements of intracellular calcium [Ca2+]i and tension. Superoxide (O2.-) and hydrogen peroxide (H2O2) production were measured by chemiluminescence and fluorescence and protein expression by Western blot. Activation of AMPK with A769662 increased AMPKα phosphorylation at Thr-172 and induced potent relaxations compared to AICAR in isolated human and rat intrarenal arteries, through both endothelium-dependent mechanisms involving nitric oxide (NO) and intermediate-conductance calcium-activated potassium (IKCa) channels, as well as activation of ATP-sensitive (KATP) channels and sarcoplasmic reticulum Ca2+-ATPase (SERCA) in vascular smooth muscle (VSM). Furthermore, AMPK activator reduced NADPH oxidase 4 (Nox4) and Nox2-derived reactive oxygen species (ROS) production. These results demonstrate that A769662 has potent vasodilator and antioxidant effects in intrarenal arteries. The benefits of AMPK activation in rat kidney are reproduced in human arteries and therefore vascular AMPK activation might be a therapeutic target in the treatment of metabolic disease-associated kidney injury.
Collapse
|
26
|
Evans AM, Hardie DG. AMPK and the Need to Breathe and Feed: What's the Matter with Oxygen? Int J Mol Sci 2020; 21:ijms21103518. [PMID: 32429235 PMCID: PMC7279029 DOI: 10.3390/ijms21103518] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2020] [Revised: 05/11/2020] [Accepted: 05/12/2020] [Indexed: 12/12/2022] Open
Abstract
We live and to do so we must breathe and eat, so are we a combination of what we eat and breathe? Here, we will consider this question, and the role in this respect of the AMP-activated protein kinase (AMPK). Emerging evidence suggests that AMPK facilitates central and peripheral reflexes that coordinate breathing and oxygen supply, and contributes to the central regulation of feeding and food choice. We propose, therefore, that oxygen supply to the body is aligned with not only the quantity we eat, but also nutrient-based diet selection, and that the cell-specific expression pattern of AMPK subunit isoforms is critical to appropriate system alignment in this respect. Currently available information on how oxygen supply may be aligned with feeding and food choice, or vice versa, through our motivation to breathe and select particular nutrients is sparse, fragmented and lacks any integrated understanding. By addressing this, we aim to provide the foundations for a clinical perspective that reveals untapped potential, by highlighting how aberrant cell-specific changes in the expression of AMPK subunit isoforms could give rise, in part, to known associations between metabolic disease, such as obesity and type 2 diabetes, sleep-disordered breathing, pulmonary hypertension and acute respiratory distress syndrome.
Collapse
Affiliation(s)
- A. Mark Evans
- Centre for Discovery Brain Sciences and Cardiovascular Science, Edinburgh Medical School, Hugh Robson Building, University of Edinburgh, Edinburgh EH8 9XD, UK
- Correspondence:
| | - D. Grahame Hardie
- Division of Cell Signalling and Immunology, School of Life Sciences, University of Dundee, Dow Street, Dundee DD1 5EH, UK;
| |
Collapse
|
27
|
Steinberg GR, Carling D. AMP-activated protein kinase: the current landscape for drug development. Nat Rev Drug Discov 2020; 18:527-551. [PMID: 30867601 DOI: 10.1038/s41573-019-0019-2] [Citation(s) in RCA: 409] [Impact Index Per Article: 81.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Since the discovery of AMP-activated protein kinase (AMPK) as a central regulator of energy homeostasis, many exciting insights into its structure, regulation and physiological roles have been revealed. While exercise, caloric restriction, metformin and many natural products increase AMPK activity and exert a multitude of health benefits, developing direct activators of AMPK to elicit beneficial effects has been challenging. However, in recent years, direct AMPK activators have been identified and tested in preclinical models, and a small number have entered clinical trials. Despite these advances, which disease(s) represent the best indications for therapeutic AMPK activation and the long-term safety of such approaches remain to be established.
Collapse
Affiliation(s)
- Gregory R Steinberg
- Centre for Metabolism, Obesity and Diabetes Research, Department of Medicine and Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Ontario, Canada.
| | - David Carling
- Cellular Stress Group, Medical Research Council London Institute of Medical Sciences, Hammersmith Hospital, Imperial College, London, UK
| |
Collapse
|
28
|
Liu G, Wu F, Jiang X, Que Y, Qin Z, Hu P, Lee KSS, Yang J, Zeng C, Hammock BD, Tong X. Inactivation of Cys 674 in SERCA2 increases BP by inducing endoplasmic reticulum stress and soluble epoxide hydrolase. Br J Pharmacol 2020; 177:1793-1805. [PMID: 31758704 DOI: 10.1111/bph.14937] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2019] [Revised: 10/29/2019] [Accepted: 10/30/2019] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND AND PURPOSE The kidney is essential in regulating sodium homeostasis and BP. The irreversible oxidation of Cys674 (C674) in the sarcoplasmic/endoplasmic reticulum calcium ATPase 2 (SERCA2) is increased in the renal cortex of hypertensive mice. Whether inactivation of C674 promotes hypertension is unclear. Here we have investigated the effects on BP of the inactivation of C674, and its role in the kidney. EXPERIMENTAL APPROACH We used heterozygous SERCA2 C674S knock-in (SKI) mice, where half of C674 was substituted by serine, to represent partial irreversible oxidation of C674. The BP, urine volume, and urine composition of SKI mice and their littermate wild-type (WT) mice were measured. The kidneys were collected for cell culture, Na+ /K+ -ATPase activity, protein expression, and immunohistological analysis. KEY RESULTS Compared with WT mice, SKI mice had higher BP, lower urine volume and sodium excretion, up-regulated endoplasmic reticulum (ER) stress markers and soluble epoxide hydrolase (sEH), and down-regulated dopamine D1 receptors in renal cortex and cells from renal proximal tubule. ER stress and sEH were mutually regulated, and both upstream of D1 receptors. Inhibition of ER stress or sEH up-regulated expression of D1 receptors, decreased the activity of Na+ /K+ -ATPase, increased sodium excretion, and lowered BP in SKI mice. CONCLUSIONS AND IMPLICATIONS The inactivation of SERCA2 C674 promotes the development of hypertension by inducing ER stress and sEH. Our study highlights the importance of C674 redox status in BP control and the contribution of SERCA2 to sodium homeostasis and BP in the kidney.
Collapse
Affiliation(s)
- Gang Liu
- School of Pharmaceutical Sciences, Chongqing University, Chongqing, China
| | - Fuhua Wu
- School of Pharmaceutical Sciences, Chongqing University, Chongqing, China
| | - Xiaoli Jiang
- School of Pharmaceutical Sciences, Chongqing University, Chongqing, China
| | - Yumei Que
- School of Pharmaceutical Sciences, Chongqing University, Chongqing, China
| | - Zhexue Qin
- Department of Cardiovascular Diseases, Xinqiao Hospital, Third Military Medical University, Chongqing, China
| | - Pingping Hu
- School of Pharmaceutical Sciences, Chongqing University, Chongqing, China
| | - Kin Sing Stephen Lee
- Department of Entomology & UCD Comprehensive Cancer Center, University of California-Davis, Davis, California.,Department of Pharmacology & Toxicology, Michigan State University, East Lansing, Michigan
| | - Jian Yang
- Department of Clinical Nutrition, The Third Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Chunyu Zeng
- Department of Cardiology, Daping Hospital, Third Military Medical University, Chongqing, China
| | - Bruce D Hammock
- Department of Entomology & UCD Comprehensive Cancer Center, University of California-Davis, Davis, California
| | - Xiaoyong Tong
- School of Pharmaceutical Sciences, Chongqing University, Chongqing, China
| |
Collapse
|
29
|
Vyas V, Guerra DD, Bok R, Powell T, Jansson T, Hurt KJ. Adiponectin links maternal metabolism to uterine contractility. FASEB J 2019; 33:14588-14601. [PMID: 31665924 PMCID: PMC6894045 DOI: 10.1096/fj.201901646r] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2019] [Accepted: 09/23/2019] [Indexed: 12/31/2022]
Abstract
Adiponectin is secreted by adipose tissue and promotes insulin sensitivity. Low circulating adiponectin is associated with increased risk for preterm labor, but the influence of adiponectin on uterine myometrial physiology is unknown. We hypothesized that adiponectin receptors (AdipoRs) decrease myometrial contractility via AMPK to promote uterine quiescence in pregnancy. Using quantitative RT-PCR, we found that nonpregnant or pregnant human and mouse myometrium express AdipoR1 and AdipoR2 mRNAs. We confirmed AdipoR2 protein expression in human and mouse myometrium, with increased abundance in late mouse pregnancy. Both recombinant adiponectin and a pharmacologic AdipoR agonist, AdipoRon, potently inhibited uterine myometrial strip contractions in physiologic organ bath. The relaxation was independent of contractile stimulus (oxytocin, KCl, U46619). AdipoR agonists increased AMPK phosphorylation in pregnant mouse myometrium, and the direct AMPK activator A769662 also relaxed myometrial strips. However, the AMPK inhibitor dorsomorphin (compound C) blocked AMPK phosphorylation but did not abolish relaxation with either AdipoRon or A769662. In summary, adiponectin inhibits myometrial contractility consistent with the possibility that it is a previously unrecognized link between maternal metabolism and pregnancy maintenance. We also identify a separate role for AMPK regulating myometrial contractions that may influence labor onset.-Vyas, V., Guerra, D. D., Bok, R., Powell, T., Jansson, T., Hurt, K. J. Adiponectin links maternal metabolism to uterine contractility.
Collapse
Affiliation(s)
- Vibhuti Vyas
- Division of Reproductive Sciences, Department of Obstetrics and Gynecology, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Damian D. Guerra
- Division of Reproductive Sciences, Department of Obstetrics and Gynecology, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Rachael Bok
- Division of Reproductive Sciences, Department of Obstetrics and Gynecology, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Theresa Powell
- Division of Reproductive Sciences, Department of Obstetrics and Gynecology, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
- Division of Neonatology, Department of Pediatrics, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA; and
| | - Thomas Jansson
- Division of Reproductive Sciences, Department of Obstetrics and Gynecology, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - K. Joseph Hurt
- Division of Reproductive Sciences, Department of Obstetrics and Gynecology, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
- Division of Maternal Fetal Medicine, Department of Obstetrics and Gynecology, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| |
Collapse
|
30
|
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
|
31
|
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
|
32
|
AMPK breathing and oxygen supply. Respir Physiol Neurobiol 2019; 265:112-120. [DOI: 10.1016/j.resp.2018.08.011] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2018] [Revised: 08/06/2018] [Accepted: 08/31/2018] [Indexed: 01/28/2023]
|
33
|
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
|
34
|
Endothelial AMP-Activated Kinase α1 Phosphorylates eNOS on Thr495 and Decreases Endothelial NO Formation. Int J Mol Sci 2018; 19:ijms19092753. [PMID: 30217073 PMCID: PMC6165563 DOI: 10.3390/ijms19092753] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2018] [Revised: 09/07/2018] [Accepted: 09/11/2018] [Indexed: 02/08/2023] Open
Abstract
AMP-activated protein kinase (AMPK) is frequently reported to phosphorylate Ser1177 of the endothelial nitric-oxide synthase (eNOS), and therefore, is linked with a relaxing effect. However, previous studies failed to consistently demonstrate a major role for AMPK on eNOS-dependent relaxation. As AMPK also phosphorylates eNOS on the inhibitory Thr495 site, this study aimed to determine the role of AMPKα1 and α2 subunits in the regulation of NO-mediated vascular relaxation. Vascular reactivity to phenylephrine and acetylcholine was assessed in aortic and carotid artery segments from mice with global (AMPKα-/-) or endothelial-specific deletion (AMPKαΔEC) of the AMPKα subunits. In control and AMPKα1-depleted human umbilical vein endothelial cells, eNOS phosphorylation on Ser1177 and Thr495 was assessed after AMPK activation with thiopental or ionomycin. Global deletion of the AMPKα1 or α2 subunit in mice did not affect vascular reactivity. The endothelial-specific deletion of the AMPKα1 subunit attenuated phenylephrine-mediated contraction in an eNOS- and endothelium-dependent manner. In in vitro studies, activation of AMPK did not alter the phosphorylation of eNOS on Ser1177, but increased its phosphorylation on Thr495. Depletion of AMPKα1 in cultured human endothelial cells decreased Thr495 phosphorylation without affecting Ser1177 phosphorylation. The results of this study indicate that AMPKα1 targets the inhibitory phosphorylation Thr495 site in the calmodulin-binding domain of eNOS to attenuate basal NO production and phenylephrine-induced vasoconstriction.
Collapse
|
35
|
MacMillan S, Evans AM. AMPK-α1 or AMPK-α2 Deletion in Smooth Muscles Does Not Affect the Hypoxic Ventilatory Response or Systemic Arterial Blood Pressure Regulation During Hypoxia. Front Physiol 2018; 9:655. [PMID: 29928235 PMCID: PMC5997817 DOI: 10.3389/fphys.2018.00655] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2018] [Accepted: 05/14/2018] [Indexed: 12/31/2022] Open
Abstract
The hypoxic ventilatory response (HVR) is markedly attenuated by AMPK-α1 deletion conditional on the expression of Cre-recombinase in tyrosine hydroxylase (TH) expressing cells, precipitating marked increases in apnea frequency and duration. It was concluded that ventilatory dysfunction caused by AMPK deficiency was driven by neurogenic mechanisms. However, TH is transiently expressed in other cell types during development, and it is evident that central respiratory depression can also be triggered by myogenic mechanisms that impact blood supply to the brain. We therefore assessed the effect on the HVR and systemic arterial blood pressure of AMPK deletion in vascular smooth muscles. There was no difference in minute ventilation during normoxia. However, increases in minute ventilation during severe hypoxia (8% O2) were, if affected at all, augmented by AMPK-α1 and AMPK-α2 deletion in smooth muscles; despite the fact that hypoxia (8% O2) evoked falls in arterial SpO2 comparable with controls. Surprisingly, these mice exhibited no difference in systolic, diastolic or mean arterial blood pressure during normoxia or hypoxia. We conclude that neither AMPK-α1 nor AMPK-α2 are required in smooth muscle for the regulation of systemic arterial blood pressure during hypoxia, and that AMPK-α1 deficiency does not impact the HVR by myogenic mechanisms.
Collapse
Affiliation(s)
- Sandy MacMillan
- Centre for Discovery Brain Sciences and Centre for Cardiovascular Science, College of Medicine and Veterinary Medicine, University of Edinburgh, Edinburgh, United Kingdom
| | - A Mark Evans
- Centre for Discovery Brain Sciences and Centre for Cardiovascular Science, College of Medicine and Veterinary Medicine, University of Edinburgh, Edinburgh, United Kingdom
| |
Collapse
|
36
|
Keeley TP, Siow RCM, Jacob R, Mann GE. Reduced SERCA activity underlies dysregulation of Ca 2+ homeostasis under atmospheric O 2 levels. FASEB J 2017; 32:2531-2538. [PMID: 29273673 PMCID: PMC5901376 DOI: 10.1096/fj.201700685rrr] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Unregulated increases in cellular Ca2+ homeostasis are a hallmark of pathophysiological conditions and a key trigger of cell death. Endothelial cells cultured under physiologic O2 conditions (5% O2) exhibit a reduced cytosolic Ca2+ response to stimulation. The mechanism for reduced plateau [Ca2+]i upon stimulation was due to increased sarco/endoplasmic reticulum Ca2+ ATPase (SERCA)-mediated reuptake rather than changes in Ca2+ influx capacity. Agonist-stimulated phosphorylation of the SERCA regulatory protein phospholamban was increased in cells cultured under 5% O2. Elevation of cytosolic and mitochondrial [Ca2+] and cell death after prolonged ionomycin treatment, as a model of Ca2+ overload, were lower when cells were cultured long-term under 5% compared with 18% O2. This protection was abolished by cotreatment with the SERCA inhibitor cyclopiazonic acid. Taken together, these results demonstrate that culturing cells under hyperoxic conditions reduces their ability to efficiently regulate [Ca2+]i, resulting in greater sensitivity to cytotoxic stimuli.-Keeley, T. P., Siow, R. C. M., Jacob, R., Mann, G. E. Reduced SERCA activity underlies dysregulation of Ca2+ homeostasis under atmospheric O2 levels.
Collapse
Affiliation(s)
- Thomas P Keeley
- King's British Heart Foundation Centre for Research Excellence, School of Cardiovascular Medicine and Sciences, Faculty of Life Sciences and Medicine, King's College London, London, United Kingdom
| | - Richard C M Siow
- King's British Heart Foundation Centre for Research Excellence, School of Cardiovascular Medicine and Sciences, Faculty of Life Sciences and Medicine, King's College London, London, United Kingdom
| | - Ron Jacob
- King's British Heart Foundation Centre for Research Excellence, School of Cardiovascular Medicine and Sciences, Faculty of Life Sciences and Medicine, King's College London, London, United Kingdom
| | - Giovanni E Mann
- King's British Heart Foundation Centre for Research Excellence, School of Cardiovascular Medicine and Sciences, Faculty of Life Sciences and Medicine, King's College London, London, United Kingdom
| |
Collapse
|
37
|
Pogoda K, Mannell H, Blodow S, Schneider H, Schubert KM, Qiu J, Schmidt A, Imhof A, Beck H, Tanase LI, Pfeifer A, Pohl U, Kameritsch P. NO Augments Endothelial Reactivity by Reducing Myoendothelial Calcium Signal Spreading: A Novel Role for Cx37 (Connexin 37) and the Protein Tyrosine Phosphatase SHP-2. Arterioscler Thromb Vasc Biol 2017; 37:2280-2290. [PMID: 29025706 DOI: 10.1161/atvbaha.117.309913] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2016] [Accepted: 09/26/2017] [Indexed: 01/31/2023]
Abstract
OBJECTIVE Because of its strategic position between endothelial and smooth muscle cells in microvessels, Cx37 (Connexin 37) plays an important role in myoendothelial gap junctional intercellular communication. We have shown before that NO inhibits gap junctional intercellular communication through gap junctions containing Cx37. However, the underlying mechanism is not yet identified. APPROACH AND RESULTS Using channel-forming Cx37 mutants exhibiting partial deletions or amino acid exchanges in their C-terminal loops, we now show that the phosphorylation state of a tyrosine residue at position 332 (Y332) in the C-terminus of Cx37 controls the gap junction-dependent spread of calcium signals. Mass spectra revealed that NO protects Cx37 from dephosphorylation at Y332 by inhibition of the protein tyrosine phosphatase SHP-2. Functionally, the inhibition of gap junctional intercellular communication by NO decreased the spread of the calcium signal (induced by mechanical stimulation of individual endothelial cells) from endothelial to smooth muscle cells in intact vessels, while, at the same time, augmenting the calcium signal spreading within the endothelium. Consequently, preincubation of small resistance arteries with exogenous NO enhanced the endothelium-dependent dilator response to acetylcholine in spite of a pharmacological blockade of NO-dependent cGMP formation by the soluable guanylyl cyclase inhibitor ODQ (1H-[1,2,4]oxadiazolo[4,3-a]quinoxalin-1-one). CONCLUSIONS Our results identify a novel mechanism by which NO can increase the efficacy of calcium, rising vasoactive agonists in the microvascular endothelium.
Collapse
Affiliation(s)
- Kristin Pogoda
- From the Walter Brendel Centre of Experimental Medicine, University Hospital, Biomedical Center, Munich, Germany (K.P., H.M., S.B., H.S., K.M.S., J.Q., H.B., L.I.T., U.P., P.K.); Protein Analysis Unit, Biomedical Center, Munich, Germany (A.S., A.I.); DZHK (German Center for Cardiovascular Research), partner site Munich Heart Alliance, Germany (K.P., H.M., S.B., H.S., K.M.S., J.Q., H.B., U.P., P.K.); Munich Cluster for Systems Neurology (SyNergY), Germany (A.I., U.P.); and Institute of Pharmacology and Toxicology, Rheinische Friedrich-Wilhelms-Universität Bonn, Germany (A.P.)
| | - Hanna Mannell
- From the Walter Brendel Centre of Experimental Medicine, University Hospital, Biomedical Center, Munich, Germany (K.P., H.M., S.B., H.S., K.M.S., J.Q., H.B., L.I.T., U.P., P.K.); Protein Analysis Unit, Biomedical Center, Munich, Germany (A.S., A.I.); DZHK (German Center for Cardiovascular Research), partner site Munich Heart Alliance, Germany (K.P., H.M., S.B., H.S., K.M.S., J.Q., H.B., U.P., P.K.); Munich Cluster for Systems Neurology (SyNergY), Germany (A.I., U.P.); and Institute of Pharmacology and Toxicology, Rheinische Friedrich-Wilhelms-Universität Bonn, Germany (A.P.)
| | - Stephanie Blodow
- From the Walter Brendel Centre of Experimental Medicine, University Hospital, Biomedical Center, Munich, Germany (K.P., H.M., S.B., H.S., K.M.S., J.Q., H.B., L.I.T., U.P., P.K.); Protein Analysis Unit, Biomedical Center, Munich, Germany (A.S., A.I.); DZHK (German Center for Cardiovascular Research), partner site Munich Heart Alliance, Germany (K.P., H.M., S.B., H.S., K.M.S., J.Q., H.B., U.P., P.K.); Munich Cluster for Systems Neurology (SyNergY), Germany (A.I., U.P.); and Institute of Pharmacology and Toxicology, Rheinische Friedrich-Wilhelms-Universität Bonn, Germany (A.P.)
| | - Holger Schneider
- From the Walter Brendel Centre of Experimental Medicine, University Hospital, Biomedical Center, Munich, Germany (K.P., H.M., S.B., H.S., K.M.S., J.Q., H.B., L.I.T., U.P., P.K.); Protein Analysis Unit, Biomedical Center, Munich, Germany (A.S., A.I.); DZHK (German Center for Cardiovascular Research), partner site Munich Heart Alliance, Germany (K.P., H.M., S.B., H.S., K.M.S., J.Q., H.B., U.P., P.K.); Munich Cluster for Systems Neurology (SyNergY), Germany (A.I., U.P.); and Institute of Pharmacology and Toxicology, Rheinische Friedrich-Wilhelms-Universität Bonn, Germany (A.P.)
| | - Kai Michael Schubert
- From the Walter Brendel Centre of Experimental Medicine, University Hospital, Biomedical Center, Munich, Germany (K.P., H.M., S.B., H.S., K.M.S., J.Q., H.B., L.I.T., U.P., P.K.); Protein Analysis Unit, Biomedical Center, Munich, Germany (A.S., A.I.); DZHK (German Center for Cardiovascular Research), partner site Munich Heart Alliance, Germany (K.P., H.M., S.B., H.S., K.M.S., J.Q., H.B., U.P., P.K.); Munich Cluster for Systems Neurology (SyNergY), Germany (A.I., U.P.); and Institute of Pharmacology and Toxicology, Rheinische Friedrich-Wilhelms-Universität Bonn, Germany (A.P.)
| | - Jiehua Qiu
- From the Walter Brendel Centre of Experimental Medicine, University Hospital, Biomedical Center, Munich, Germany (K.P., H.M., S.B., H.S., K.M.S., J.Q., H.B., L.I.T., U.P., P.K.); Protein Analysis Unit, Biomedical Center, Munich, Germany (A.S., A.I.); DZHK (German Center for Cardiovascular Research), partner site Munich Heart Alliance, Germany (K.P., H.M., S.B., H.S., K.M.S., J.Q., H.B., U.P., P.K.); Munich Cluster for Systems Neurology (SyNergY), Germany (A.I., U.P.); and Institute of Pharmacology and Toxicology, Rheinische Friedrich-Wilhelms-Universität Bonn, Germany (A.P.)
| | - Andreas Schmidt
- From the Walter Brendel Centre of Experimental Medicine, University Hospital, Biomedical Center, Munich, Germany (K.P., H.M., S.B., H.S., K.M.S., J.Q., H.B., L.I.T., U.P., P.K.); Protein Analysis Unit, Biomedical Center, Munich, Germany (A.S., A.I.); DZHK (German Center for Cardiovascular Research), partner site Munich Heart Alliance, Germany (K.P., H.M., S.B., H.S., K.M.S., J.Q., H.B., U.P., P.K.); Munich Cluster for Systems Neurology (SyNergY), Germany (A.I., U.P.); and Institute of Pharmacology and Toxicology, Rheinische Friedrich-Wilhelms-Universität Bonn, Germany (A.P.)
| | - Axel Imhof
- From the Walter Brendel Centre of Experimental Medicine, University Hospital, Biomedical Center, Munich, Germany (K.P., H.M., S.B., H.S., K.M.S., J.Q., H.B., L.I.T., U.P., P.K.); Protein Analysis Unit, Biomedical Center, Munich, Germany (A.S., A.I.); DZHK (German Center for Cardiovascular Research), partner site Munich Heart Alliance, Germany (K.P., H.M., S.B., H.S., K.M.S., J.Q., H.B., U.P., P.K.); Munich Cluster for Systems Neurology (SyNergY), Germany (A.I., U.P.); and Institute of Pharmacology and Toxicology, Rheinische Friedrich-Wilhelms-Universität Bonn, Germany (A.P.)
| | - Heike Beck
- From the Walter Brendel Centre of Experimental Medicine, University Hospital, Biomedical Center, Munich, Germany (K.P., H.M., S.B., H.S., K.M.S., J.Q., H.B., L.I.T., U.P., P.K.); Protein Analysis Unit, Biomedical Center, Munich, Germany (A.S., A.I.); DZHK (German Center for Cardiovascular Research), partner site Munich Heart Alliance, Germany (K.P., H.M., S.B., H.S., K.M.S., J.Q., H.B., U.P., P.K.); Munich Cluster for Systems Neurology (SyNergY), Germany (A.I., U.P.); and Institute of Pharmacology and Toxicology, Rheinische Friedrich-Wilhelms-Universität Bonn, Germany (A.P.)
| | - Laurentia Irina Tanase
- From the Walter Brendel Centre of Experimental Medicine, University Hospital, Biomedical Center, Munich, Germany (K.P., H.M., S.B., H.S., K.M.S., J.Q., H.B., L.I.T., U.P., P.K.); Protein Analysis Unit, Biomedical Center, Munich, Germany (A.S., A.I.); DZHK (German Center for Cardiovascular Research), partner site Munich Heart Alliance, Germany (K.P., H.M., S.B., H.S., K.M.S., J.Q., H.B., U.P., P.K.); Munich Cluster for Systems Neurology (SyNergY), Germany (A.I., U.P.); and Institute of Pharmacology and Toxicology, Rheinische Friedrich-Wilhelms-Universität Bonn, Germany (A.P.)
| | - Alexander Pfeifer
- From the Walter Brendel Centre of Experimental Medicine, University Hospital, Biomedical Center, Munich, Germany (K.P., H.M., S.B., H.S., K.M.S., J.Q., H.B., L.I.T., U.P., P.K.); Protein Analysis Unit, Biomedical Center, Munich, Germany (A.S., A.I.); DZHK (German Center for Cardiovascular Research), partner site Munich Heart Alliance, Germany (K.P., H.M., S.B., H.S., K.M.S., J.Q., H.B., U.P., P.K.); Munich Cluster for Systems Neurology (SyNergY), Germany (A.I., U.P.); and Institute of Pharmacology and Toxicology, Rheinische Friedrich-Wilhelms-Universität Bonn, Germany (A.P.)
| | - Ulrich Pohl
- From the Walter Brendel Centre of Experimental Medicine, University Hospital, Biomedical Center, Munich, Germany (K.P., H.M., S.B., H.S., K.M.S., J.Q., H.B., L.I.T., U.P., P.K.); Protein Analysis Unit, Biomedical Center, Munich, Germany (A.S., A.I.); DZHK (German Center for Cardiovascular Research), partner site Munich Heart Alliance, Germany (K.P., H.M., S.B., H.S., K.M.S., J.Q., H.B., U.P., P.K.); Munich Cluster for Systems Neurology (SyNergY), Germany (A.I., U.P.); and Institute of Pharmacology and Toxicology, Rheinische Friedrich-Wilhelms-Universität Bonn, Germany (A.P.).
| | - Petra Kameritsch
- From the Walter Brendel Centre of Experimental Medicine, University Hospital, Biomedical Center, Munich, Germany (K.P., H.M., S.B., H.S., K.M.S., J.Q., H.B., L.I.T., U.P., P.K.); Protein Analysis Unit, Biomedical Center, Munich, Germany (A.S., A.I.); DZHK (German Center for Cardiovascular Research), partner site Munich Heart Alliance, Germany (K.P., H.M., S.B., H.S., K.M.S., J.Q., H.B., U.P., P.K.); Munich Cluster for Systems Neurology (SyNergY), Germany (A.I., U.P.); and Institute of Pharmacology and Toxicology, Rheinische Friedrich-Wilhelms-Universität Bonn, Germany (A.P.)
| |
Collapse
|
38
|
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
|
39
|
Smith CA, Miner AS, Barbee RW, Ratz PH. Metabolic Stress-Induced Activation of AMPK and Inhibition of Constitutive Phosphoproteins Controlling Smooth Muscle Contraction: Evidence for Smooth Muscle Fatigue? Front Physiol 2017; 8:681. [PMID: 28943852 PMCID: PMC5596101 DOI: 10.3389/fphys.2017.00681] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2017] [Accepted: 08/24/2017] [Indexed: 11/23/2022] Open
Abstract
Metabolic stress diminishes smooth muscle contractile strength by a poorly defined mechanism. To test the hypothesis that metabolic stress activates a compensatory cell signaling program to reversibly downregulate contraction, arterial rings and bladder muscle strips in vitro were deprived of O2 and glucose for 30 and 60 min (“starvation”) to induce metabolic stress, and the phosphorylation status of proteins involved in regulation of contraction and metabolic stress were assessed in tissues under basal and stimulated conditions. A 15–30 min recovery period (O2 and glucose repletion) tested whether changes induced by starvation were reversible. Starvation decreased basal phosphorylation of myosin regulatory light chain (MLC-pS19) and of the rho kinase (ROCK) downstream substrates cofilin (cofilin-pS3) and myosin phosphatase targeting subunit MYPT1 (MYPT1-pT696 and MYPT1-pT853), and abolished the ability of contractile stimuli to cause a strong, sustained contraction. Starvation increased basal phosphorylation of AMPK (AMPK-pT172) and 3 downstream AMPK substrates, acetyl-CoA carboxylase (ACC-pS79), rhoA (rhoA-pS188), and phospholamban (PLB-pS16). Increases in rhoA-pS188 and PLB-pS16 would be expected to inhibit contraction. Recovery restored basal AMPK-pT172 and MLC-pS19 to control levels, and restored contraction. In AMPKα2 deficient mice (AMPKα2-/-), the basal level of AMPK-pT172 was reduced by 50%, and MLC-pS19 was elevated by 50%, but AMPKα2-/- did not prevent starvation-induced contraction inhibition nor enhance recovery from starvation. These results indicate that constitutive AMPK activity participates in constitutive regulation of contractile proteins, and suggest that AMPK activation is necessary, but may not be sufficient, to cause smooth muscle contraction inhibition during metabolic stress.
Collapse
Affiliation(s)
- Corey A Smith
- Department of Biochemistry and Molecular Biology, Virginia Commonwealth UniversityRichmond, VA, United States
| | - Amy S Miner
- Department of Biochemistry and Molecular Biology, Virginia Commonwealth UniversityRichmond, VA, United States
| | - Robert W Barbee
- Departments of Emergency Medicine and Physiology, Virginia Commonwealth UniversityRichmond, VA, United States
| | - Paul H Ratz
- Department of Biochemistry and Molecular Biology, Virginia Commonwealth UniversityRichmond, VA, United States
| |
Collapse
|
40
|
Woodman RJ, Baghdadi LR, Shanahan ME, Mangoni AA. The Temporal Relationship between Arterial Stiffening and Blood Pressure Is Modified by Methotrexate Treatment in Patients with Rheumatoid Arthritis. Front Physiol 2017; 8:593. [PMID: 28861004 PMCID: PMC5559508 DOI: 10.3389/fphys.2017.00593] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2017] [Accepted: 07/31/2017] [Indexed: 01/17/2023] Open
Abstract
Background: The temporal relationship between arterial stiffness and blood pressure (BP) may vary depending on age and other clinical and demographic factors. Since both BP and arterial stiffness are also affected by inflammatory processes, we examined the temporal arterial stiffness-BP relationship in patients with rheumatoid arthritis (RA) treated with either methotrexate (MTX), an anti-rheumatic agent shown to reduce cardiovascular risk in meta-analyses, or other disease-modifying anti-rheumatic drugs (DMARDs). Methods: Measurements of clinic and 24-h peripheral and central systolic and diastolic BP (SBP and DBP), and pulse wave velocity (PWV) were assessed in RA patients on stable treatment with either MTX ± other DMARDs (MTX group, n = 41, age 61 ± 14 years, 73% females) or other DMARDs (non-MTX group, n = 18, age 65 ± 13 years, 89% females). Measurements were performed at baseline and after 8 months. The temporal relationships were examined using cross-lagged path analysis with models that included age, sex, body mass index, prednisolone, and folic acid use and 28-joint disease activity score. Results: There were significant differences in the temporal arterial stiffness-BP relationships between those in the MTX and DMARD groups. A higher PWV at baseline caused a significant increase in 6 out of 8 different measures of SBP at 8 months amongst those treated with DMARDs (standardized β, range = 0.54–0.66, p < 0.003 for each) and 3 out of 8 different measures of DBP (standardized β, range = 0.52–0.61, p < 0.003 for each) but was not associated with either SBP or DBP at 8 months amongst those treated with MTX. The difference in the effect of baseline PWV on 8-month BP between the 2 groups was also significant (p < 0.003) for 4 measures including clinic peripheral SBP (β = 7.0, 95% CI = 2.8–11.1 mmHg per 1 m/s higher baseline PWV; p < 0.001). Conclusions: Higher arterial stiffness preceded increases in BP in subjects with RA treated with DMARDs, but these effects did not occur amongst those treated with MTX. The different effects were seen mostly in measures of SBP but were also present in some measures of DBP. Our findings suggest MTX may confer a protective effect against stiffness mediated increases in BP in patients with RA.
Collapse
Affiliation(s)
- Richard J Woodman
- Centre for Epidemiology and Biostatistics, School of Medicine, Flinders UniversityAdelaide, SA, Australia
| | - Leena R Baghdadi
- Centre for Epidemiology and Biostatistics, School of Medicine, Flinders UniversityAdelaide, SA, Australia.,Department of Clinical Pharmacology, School of Medicine, Flinders University and Flinders Medical CentreAdelaide, SA, Australia.,Department of Family and Community Medicine, King Saud UniversityRiyadh, Saudi Arabia
| | - Michael E Shanahan
- Department of Rheumatology, Flinders University and Southern Adelaide Local Health NetworkAdelaide, SA, Australia
| | - Arduino A Mangoni
- Department of Clinical Pharmacology, School of Medicine, Flinders University and Flinders Medical CentreAdelaide, SA, Australia
| |
Collapse
|
41
|
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
|
42
|
Duan Q, Song P, Ding Y, Zou MH. Activation of AMP-activated protein kinase by metformin ablates angiotensin II-induced endoplasmic reticulum stress and hypertension in mice in vivo. Br J Pharmacol 2017; 174:2140-2151. [PMID: 28436023 DOI: 10.1111/bph.13833] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2017] [Revised: 03/29/2017] [Accepted: 04/16/2017] [Indexed: 12/29/2022] Open
Abstract
BACKGROUND AND PURPOSE Metformin, one of the most frequently prescribed medications for type 2 diabetes, reportedly exerts BP-lowering effects in patients with diabetes. However, the effects and underlying mechanisms of metformin on BP in non-diabetic conditions remain to be determined. The aim of the present study was to determine the effects of metformin on angiotensin II (Ang II) infusion-induced hypertension in vivo. EXPERIMENTAL APPROACH The effects of metformin on BP were investigated in wild-type (WT) C57BL/6J mice and in mice lacking AMP-activated protein kinase α2 (AMPKα2) mice with or without Ang II infusion. Also, the effect of metformin on Ang II-induced endoplasmic reticulum (ER) stress was explored in cultured human vascular smooth muscle cells (hVSMCs). KEY RESULTS Metformin markedly reduced BP in Ang II-infused WT mice but not in AMPKα2-deficient mice. In cultured hVSMCs, Ang II treatment resulted in inactivation of AMPK, as well as the subsequent induction of spliced X-box binding protein-1, phosphorylation of eukaryotic translation initiation factor 2α and expression of glucose-regulated protein 78 kDa, representing three well-characterized ER stress biomarkers. Moreover, AMPK activation by metformin ablated Ang II-induced ER stress in hVSMCs. Mechanistically, metformin-activated AMPKα2 suppressed ER stress by increasing phospholamban phosphorylation. CONCLUSION AND IMPLICATIONS Metformin alleviates Ang II-triggered hypertension in mice by activating AMPKα2, which mediates phospholamban phosphorylation and inhibits Ang II-induced ER stress in vascular smooth muscle cells.
Collapse
Affiliation(s)
- Quanlu Duan
- Center for Molecular and Translational Medicine, Georgia State University, Atlanta, GA, USA.,Division of Cardiology, Department Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ping Song
- Center for Molecular and Translational Medicine, Georgia State University, Atlanta, GA, USA
| | - Ye Ding
- Center for Molecular and Translational Medicine, Georgia State University, Atlanta, GA, USA
| | - Ming-Hui Zou
- Center for Molecular and Translational Medicine, Georgia State University, Atlanta, GA, USA
| |
Collapse
|
43
|
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
|
44
|
Laskowski M, Andersson C, Eliasson E, Golubinskaya V, Nilsson H. Potassium-Channel-Independent Relaxing Influence of Adipose Tissue on Mouse Carotid Artery. J Vasc Res 2017; 54:51-57. [PMID: 28334715 DOI: 10.1159/000458421] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2016] [Accepted: 01/27/2017] [Indexed: 11/19/2022] Open
Abstract
Since the cardiovascular consequences of obesity reportedly vary in different types of obesity, we investigated the influence of adipose tissue from different locales on the phenylephrine-induced tone of the mouse carotid artery. Vessels were mounted in a Mulvany-Halpern-type wire myograph, and adipose tissue, from the back (brown) or mesenteric or inguinal subcutaneous (white), was placed around the artery. Contractile responses to phenylephrine were not affected by brown adipose tissue but were reduced (p < 0.001) by either type of white adipose tissue, with no difference between the 2 locales. The relaxing effect persisted in the presence of the Kv7 channel inhibitor XE991 (10,10-bis(4-pyridinylmethyl)-9(10H)-anthracenone), the KATP channel inhibitor glibenclamide (1 µM), or the KV channel inhibitor 4-amino pyridine (1 mM), as well as after elevation of the extracellular potassium concentration to 30 mM. Contractions of rat carotid artery were equally reduced by mouse and rat subcutaneous adipose tissue. Thus, white, but not brown, adipose tissue reduces the adrenergic contractions of the carotid artery with no differences between the locales of origin, and the effect appears largely independent of potassium channels.
Collapse
Affiliation(s)
- Marta Laskowski
- Department of Physiology, Institute of Neuroscience and Physiology, Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
| | | | | | | | | |
Collapse
|
45
|
Niclosamide ethanolamine inhibits artery constriction. Pharmacol Res 2017; 115:78-86. [DOI: 10.1016/j.phrs.2016.11.008] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/22/2016] [Revised: 11/09/2016] [Accepted: 11/09/2016] [Indexed: 01/25/2023]
|
46
|
Fu J, Han Y, Wang J, Liu Y, Zheng S, Zhou L, Jose PA, Zeng C. Irisin Lowers Blood Pressure by Improvement of Endothelial Dysfunction via AMPK-Akt-eNOS-NO Pathway in the Spontaneously Hypertensive Rat. J Am Heart Assoc 2016; 5:e003433. [PMID: 27912206 PMCID: PMC5210324 DOI: 10.1161/jaha.116.003433] [Citation(s) in RCA: 101] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/19/2016] [Accepted: 09/06/2016] [Indexed: 12/13/2022]
Abstract
BACKGROUND Exercise is a major nonpharmacological treatment for hypertension, but its underlying mechanisms are still not completely elucidated. Irisin, a polypeptide containing 112 amino acids, which is secreted mainly by skeletal muscle cells during exercise, exerts a protective role in metabolic diseases, such as diabetes mellitus and obesity. Because of the close relationship between irisin and metabolic diseases, we hypothesized that irisin may play a role in the regulation of blood pressure. METHODS AND RESULTS Blood pressures of male Wistar-Kyoto (WKY) rats and spontaneously hypertensive rats (SHRs) were monitored through the carotid artery. Our study found that acute intravenous injection of irisin reduced blood pressure in SHRs, but not WKY rats. Irisin, by itself, had no direct vasorelaxing effect in phenylephrine-preconstricted mesenteric arteries from SHRs. However, irisin augmented the acetylcholine-induced vasorelaxation in mesenteric arteries from SHRs that could be reversed by Nω-nitro-l-arginine-methyl ester (L-NAME; 100 μmol/L), indicating a role of nitric oxide (NO) in this action. Indeed, irisin increased NO production and phosphorylation of endothelial nirtic oxide synthase (eNOS) in endothelial cells. 5'-AMP-activated protein kinase (AMPK) was involved in the vasorelaxing effect of irisin because compound C (20 μmol/L), an AMPK inhibitor, blocked the irisin-mediated increase in phosphorylation of eNOS and protein kinase B (Akt) in endothelial cells and vasodilation in mesenteric arteries. CONCLUSIONS We conclude that acute administration of irisin lowers blood pressure of SHRs by amelioration of endothelial dysfunction of the mesenteric artery through the AMPK-Akt-eNOS-NO signaling pathway.
Collapse
Affiliation(s)
- Jinjuan Fu
- Department of Cardiology, Daping Hospital, Chongqing Institute of Cardiology, The Third Military Medical University, Chongqing, China
| | - Yu Han
- Department of Cardiology, Daping Hospital, Chongqing Institute of Cardiology, The Third Military Medical University, Chongqing, China
| | - Jialiang Wang
- Department of Cardiology, Daping Hospital, Chongqing Institute of Cardiology, The Third Military Medical University, Chongqing, China
| | - Yukai Liu
- Department of Cardiology, Daping Hospital, Chongqing Institute of Cardiology, The Third Military Medical University, Chongqing, China
| | - Shuo Zheng
- Department of Cardiology, Daping Hospital, Chongqing Institute of Cardiology, The Third Military Medical University, Chongqing, China
| | - Lin Zhou
- Department of Cardiology, Daping Hospital, Chongqing Institute of Cardiology, The Third Military Medical University, Chongqing, China
| | - Pedro A Jose
- Division of Renal Diseases & Hypertension, Department of Medicine, The George Washington University School of Medicine & Health Sciences, Washington, DC
| | - Chunyu Zeng
- Department of Cardiology, Daping Hospital, Chongqing Institute of Cardiology, The Third Military Medical University, Chongqing, China
| |
Collapse
|
47
|
Zhang YQ, Shen X, Xiao XL, Liu MY, Li SL, Yan J, Jin J, Gao JL, Zhen CL, Hu N, Zhang XZ, Tai Y, Zhang LS, Bai YL, Dong DL. Mitochondrial uncoupler carbonyl cyanide m-chlorophenylhydrazone induces vasorelaxation without involving K ATP channel activation in smooth muscle cells of arteries. Br J Pharmacol 2016; 173:3145-3158. [PMID: 27534899 DOI: 10.1111/bph.13578] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2016] [Revised: 08/16/2016] [Accepted: 08/16/2016] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND AND PURPOSE The effects and mechanisms of chemical mitochondrial uncouplers on vascular function have never been identified. Here, we characterized the effects of the typical mitochondrial uncoupler carbonyl cyanide m-chlorophenylhydrazone (CCCP) on vascular function in rat mesenteric arteries and aorta and elucidated the potential mechanisms. EXPERIMENTAL APPROACH Isometric tension of mesenteric artery and thoracic aorta was recorded by using a multiwire myograph system. Protein levels were measured by western blot analyses. Cytosolic [Ca2+ ]i , mitochondrial ROS (mitoROS) and mitochondrial membrane potential of smooth muscle cells (A10) were measured by laser scanning confocal microscopy. KEY RESULTS Acute treatment with CCCP relaxed phenylephrine (PE)- and high K+ (KPSS)-induced constriction of rat mesenteric arteries with intact and denuded endothelium. Pretreatment with CCCP prevented PE- and KPSS-induced constriction of rat mesenteric arteries with intact and denuded endothelium. Similarly, CCCP prevented PE- and KPSS-induced constriction of rat thoracic aorta. CCCP increased the cellular ADP/ATP ratio in vascular smooth muscle cells (A10) and activated AMPK in A10 cells and rat thoracic aorta tissues. CCCP-induced aorta relaxation was attenuated in AMPK α1 knockout (-/-) mice. SERCA inhibitors thapsigargin and cyclopiazonic acid (CPA) but not the KATP channel blocker glibenclamide partially inhibited CCCP-induced vasorelaxation in endothelium-denuded rat mesenteric arteries. CCCP increased cytosolic [Ca2+ ]i , mitoROS production and depolarized mitochondrial membrane potential in A10 cells. FCCP, the analogue of CCCP, had similar vasoactivity as CCCP in rat mesenteric arteries. CONCLUSIONS AND IMPLICATIONS CCCP induces vasorelaxation by a mechanism that does not involve KATP channel activation in smooth muscle cells of arteries.
Collapse
Affiliation(s)
- Yan-Qiu Zhang
- Department of Pharmacology (the State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy; Translational Medicine Research and Cooperation Center of Northern China, Heilongjiang Academy of Medical Sciences, Harbin Medical University, Harbin, PR China
| | - Xin Shen
- Department of Pharmacology (the State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy; Translational Medicine Research and Cooperation Center of Northern China, Heilongjiang Academy of Medical Sciences, Harbin Medical University, Harbin, PR China
| | - Xiao-Lin Xiao
- Department of Pharmacology (the State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy; Translational Medicine Research and Cooperation Center of Northern China, Heilongjiang Academy of Medical Sciences, Harbin Medical University, Harbin, PR China
| | - Ming-Yu Liu
- Department of Pharmacology (the State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy; Translational Medicine Research and Cooperation Center of Northern China, Heilongjiang Academy of Medical Sciences, Harbin Medical University, Harbin, PR China
| | - Shan-Liang Li
- Department of Pharmacology (the State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy; Translational Medicine Research and Cooperation Center of Northern China, Heilongjiang Academy of Medical Sciences, Harbin Medical University, Harbin, PR China
| | - Jie Yan
- Department of Pharmacology (the State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy; Translational Medicine Research and Cooperation Center of Northern China, Heilongjiang Academy of Medical Sciences, Harbin Medical University, Harbin, PR China
| | - Jing Jin
- Department of Pharmacology (the State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy; Translational Medicine Research and Cooperation Center of Northern China, Heilongjiang Academy of Medical Sciences, Harbin Medical University, Harbin, PR China
| | - Jin-Lai Gao
- Department of Pharmacology (the State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy; Translational Medicine Research and Cooperation Center of Northern China, Heilongjiang Academy of Medical Sciences, Harbin Medical University, Harbin, PR China
| | - Chang-Lin Zhen
- Department of Pharmacology (the State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy; Translational Medicine Research and Cooperation Center of Northern China, Heilongjiang Academy of Medical Sciences, Harbin Medical University, Harbin, PR China
| | - Nan Hu
- Department of Pharmacology (the State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy; Translational Medicine Research and Cooperation Center of Northern China, Heilongjiang Academy of Medical Sciences, Harbin Medical University, Harbin, PR China
| | - Xin-Zi Zhang
- Department of Pharmacology (the State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy; Translational Medicine Research and Cooperation Center of Northern China, Heilongjiang Academy of Medical Sciences, Harbin Medical University, Harbin, PR China
| | - Yu Tai
- Department of Pharmacology (the State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy; Translational Medicine Research and Cooperation Center of Northern China, Heilongjiang Academy of Medical Sciences, Harbin Medical University, Harbin, PR China
| | - Liang-Shuan Zhang
- Department of Pharmacology (the State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy; Translational Medicine Research and Cooperation Center of Northern China, Heilongjiang Academy of Medical Sciences, Harbin Medical University, Harbin, PR China
| | - Yun-Long Bai
- Department of Pharmacology (the State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy; Translational Medicine Research and Cooperation Center of Northern China, Heilongjiang Academy of Medical Sciences, Harbin Medical University, Harbin, PR China
| | - De-Li Dong
- Department of Pharmacology (the State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy; Translational Medicine Research and Cooperation Center of Northern China, Heilongjiang Academy of Medical Sciences, Harbin Medical University, Harbin, PR China.
| |
Collapse
|
48
|
Evans AM, Mahmoud AD, Moral-Sanz J, Hartmann S. The emerging role of AMPK in the regulation of breathing and oxygen supply. Biochem J 2016; 473:2561-72. [PMID: 27574022 PMCID: PMC5003690 DOI: 10.1042/bcj20160002] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2016] [Revised: 04/20/2016] [Accepted: 05/03/2016] [Indexed: 01/25/2023]
Abstract
Regulation of breathing is critical to our capacity to accommodate deficits in oxygen availability and demand during, for example, sleep and ascent to altitude. It is generally accepted that a fall in arterial oxygen increases afferent discharge from the carotid bodies to the brainstem and thus delivers increased ventilatory drive, which restores oxygen supply and protects against hypoventilation and apnoea. However, the precise molecular mechanisms involved remain unclear. We recently identified as critical to this process the AMP-activated protein kinase (AMPK), which is key to the cell-autonomous regulation of metabolic homoeostasis. This observation is significant for many reasons, not least because recent studies suggest that the gene for the AMPK-α1 catalytic subunit has been subjected to natural selection in high-altitude populations. It would appear, therefore, that evolutionary pressures have led to AMPK being utilized to regulate oxygen delivery and thus energy supply to the body in the short, medium and longer term. Contrary to current consensus, however, our findings suggest that AMPK regulates ventilation at the level of the caudal brainstem, even when afferent input responses from the carotid body are normal. We therefore hypothesize that AMPK integrates local hypoxic stress at defined loci within the brainstem respiratory network with an index of peripheral hypoxic status, namely afferent chemosensory inputs. Allied to this, AMPK is critical to the control of hypoxic pulmonary vasoconstriction and thus ventilation-perfusion matching at the lungs and may also determine oxygen supply to the foetus by, for example, modulating utero-placental blood flow.
Collapse
Affiliation(s)
- A Mark Evans
- Centre for Integrative Physiology, College of Medicine and Veterinary Medicine, Hugh Robson Building, University of Edinburgh, Edinburgh EH8 9XD, U.K.
| | - Amira D Mahmoud
- Centre for Integrative Physiology, College of Medicine and Veterinary Medicine, Hugh Robson Building, University of Edinburgh, Edinburgh EH8 9XD, U.K
| | - Javier Moral-Sanz
- Centre for Integrative Physiology, College of Medicine and Veterinary Medicine, Hugh Robson Building, University of Edinburgh, Edinburgh EH8 9XD, U.K
| | - Sandy Hartmann
- Centre for Integrative Physiology, College of Medicine and Veterinary Medicine, Hugh Robson Building, University of Edinburgh, Edinburgh EH8 9XD, U.K
| |
Collapse
|
49
|
Pirkmajer S, Chibalin AV. Na,K-ATPase regulation in skeletal muscle. Am J Physiol Endocrinol Metab 2016; 311:E1-E31. [PMID: 27166285 DOI: 10.1152/ajpendo.00539.2015] [Citation(s) in RCA: 74] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/31/2015] [Accepted: 05/02/2016] [Indexed: 12/17/2022]
Abstract
Skeletal muscle contains one of the largest and the most dynamic pools of Na,K-ATPase (NKA) in the body. Under resting conditions, NKA in skeletal muscle operates at only a fraction of maximal pumping capacity, but it can be markedly activated when demands for ion transport increase, such as during exercise or following food intake. Given the size, capacity, and dynamic range of the NKA pool in skeletal muscle, its tight regulation is essential to maintain whole body homeostasis as well as muscle function. To reconcile functional needs of systemic homeostasis with those of skeletal muscle, NKA is regulated in a coordinated manner by extrinsic stimuli, such as hormones and nerve-derived factors, as well as by local stimuli arising in skeletal muscle fibers, such as contractions and muscle energy status. These stimuli regulate NKA acutely by controlling its enzymatic activity and/or its distribution between the plasma membrane and the intracellular storage compartment. They also regulate NKA chronically by controlling NKA gene expression, thus determining total NKA content in skeletal muscle and its maximal pumping capacity. This review focuses on molecular mechanisms that underlie regulation of NKA in skeletal muscle by major extrinsic and local stimuli. Special emphasis is given to stimuli and mechanisms linking regulation of NKA and energy metabolism in skeletal muscle, such as insulin and the energy-sensing AMP-activated protein kinase. Finally, the recently uncovered roles for glutathionylation, nitric oxide, and extracellular K(+) in the regulation of NKA in skeletal muscle are highlighted.
Collapse
Affiliation(s)
- Sergej Pirkmajer
- Institute of Pathophysiology, Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia; and
| | - Alexander V Chibalin
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
50
|
Loss of anti-contractile effect of perivascular adipose tissue in offspring of obese rats. Int J Obes (Lond) 2016; 40:1205-14. [PMID: 27102050 PMCID: PMC4973217 DOI: 10.1038/ijo.2016.62] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/18/2015] [Revised: 02/26/2016] [Accepted: 03/22/2016] [Indexed: 12/21/2022]
Abstract
Rationale: Maternal obesity pre-programmes offspring to develop obesity and associated cardiovascular disease. Perivascular adipose tissue (PVAT) exerts an anti-contractile effect on the vasculature, which is reduced in hypertension and obesity. Objective: The objective of this study was to determine whether maternal obesity pre-programmes offspring to develop PVAT dysfunction in later life. Methods: Female Sprague–Dawley rats were fed a diet containing 10% (control) or 45% fat (high fat diet, HFD) for 12 weeks prior to mating and during pregnancy and lactation. Male offspring were killed at 12 or 24 weeks of age and tension in PVAT-intact or -denuded mesenteric artery segments was measured isometrically. Concentration–response curves were constructed to U46619 and norepinephrine. Results: Only 24-week-old HFD offspring were hypertensive (P<0.0001), although the anti-contractile effect of PVAT was lost in vessels from HFD offspring of each age. Inhibition of nitric oxide (NO) synthase with 100 μMl-NMMA attenuated the anti-contractile effect of PVAT and increased contractility of PVAT-denuded arteries (P<0.05, P<0.0001). The increase in contraction was smaller in PVAT-intact than PVAT-denuded vessels from 12-week-old HFD offspring, suggesting decreased PVAT-derived NO and release of a contractile factor (P<0.07). An additional, NO-independent effect of PVAT was evident only in norepinephrine-contracted vessels. Activation of AMP-activated kinase (with 10 μM A769662) was anti-contractile in PVAT-denuded (P<0.0001) and -intact (P<0.01) vessels and was due solely to NO in controls; the AMPK effect was similar in HFD offspring vessels (P<0.001 and P<0.01, respectively) but was partially NO-independent. Conclusions: The diminished anti-contractile effects of PVAT in offspring of HFD dams are primarily due to release of a PVAT-derived contractile factor and reduced NO bioavailability.
Collapse
|