1
|
Buraczynska M, Boczkowska S, Zaluska W. Association of KCNJ11 E23K/rs5219 Gene Polymorphism with Type 2 Diabetes and Diabetes-Related Cardiovascular Disease. Diabetes Metab Syndr Obes 2025; 18:653-661. [PMID: 40034481 PMCID: PMC11874986 DOI: 10.2147/dmso.s506639] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/27/2024] [Accepted: 02/03/2025] [Indexed: 03/05/2025] Open
Abstract
Objective A potassium voltage-gated channel subfamily J member 11 (KCNJ11) is a candidate gene for diabetes and cardiovascular disease. We investigated the relationship of KCNJ11 E23K gene polymorphism with type 2 diabetes (T2DM) and diabetes-related cardiovascular disease (CVD). Methods In this case-control study, the KCNJ11 E23K (rs5219) single nucleotide polymorphism was evaluated using the PCR-RFLP method in 780 patients with T2DM and 425 healthy controls. The genotype distribution was compared between subgroups of patients with CVD (524) and without CVD (256). Results The genotyping results showed that the T allele and TT genotype were associated with the risk of T2DM (OR 1.26, p = 0.008 and OR 1.55, p = 0.0019, respectively). The T2DM group was analyzed according to the presence or absence of CVD. The T allele frequency was significantly higher in CVD+ than CVD- patients (49% vs 28%, p = 0.0001). The frequency of TT genotype in CVD+ subgroup was 20% compared to 8.5% in CVD-. This shows the significant correlation of the T allele with CVD in T2DM patients in all genetic association models. The OR for T allele was 2.44, p < 0.0001 representing 2.5-fold higher odds of CVD. For TT genotype, the OR 5.61, p < 0.0001 represents almost 6-fold higher risk of CVD development. The multiple logistic regression analysis showed that KCNJ11 E23K polymorphism was a significant risk predictor for CVD development (p < 0.0001). Conclusion This is the first study of the relationship between KCNJ11 gene polymorphism and cardiovascular risk in T2DM patients in Polish population. The E23K (rs5219) polymorphism is associated with T2DM. It also increases the risk of cardiovascular disease in T2DM patients. If confirmed in other studies, it can be considered a potential marker for predicting the risk of CVD in T2DM patients.
Collapse
Affiliation(s)
| | - Sylwia Boczkowska
- Department of Nephrology, Medical University of Lublin, Lublin, Poland
| | - Wojciech Zaluska
- Department of Nephrology, Medical University of Lublin, Lublin, Poland
| |
Collapse
|
2
|
Rajkovic J, Peric M, Stanisic J, Gostimirovic M, Novakovic R, Djokic V, Tepavcevic S, Rakocevic J, Labudovic-Borovic M, Gojkovic-Bukarica L. Effect of Type-2 Diabetes Mellitus on the Expression and Function of Smooth Muscle ATP-Sensitive Potassium Channels in Human Internal Mammary Artery Grafts. Pharmaceuticals (Basel) 2024; 17:857. [PMID: 39065708 PMCID: PMC11280115 DOI: 10.3390/ph17070857] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2024] [Revised: 06/10/2024] [Accepted: 06/27/2024] [Indexed: 07/28/2024] Open
Abstract
Here we have shown for the first time altered expression of the vascular smooth muscle (VSM) KATP channel subunits in segments of the human internal mammary artery (HIMA) in patients with type-2 diabetes mellitus (T2DM). Functional properties of vascular KATP channels in the presence of T2DM, and the interaction between its subunits and endogenous ligands known to relax this vessel, were tested using the potassium (K) channels opener, pinacidil. HIMA is the most commonly used vascular graft in cardiac surgery. Previously it was shown that pinacidil relaxes HIMA segments through interaction with KATP (SUR2B/Kir6.1) vascular channels, but it is unknown whether pinacidil sensitivity is changed in the presence of T2DM, considering diabetes-induced vascular complications commonly seen in patients undergoing coronary artery bypass graft surgery (CABG). KATP subunits were detected in HIMA segments using Western blot and immunohistochemistry analyses. An organ bath system was used to interrogate endothelium-independent vasorelaxation caused by pinacidil. In pharmacological experiments, pinacidil was able to relax HIMA from patients with T2DM, with sensitivity comparable to our previous results. All three KATP subunits (SUR2B, Kir6.1 and Kir6.2) were observed in HIMA from patients with and without T2DM. There were no differences in the expression of the SUR2B subunit. The expression of the Kir6.1 subunit was lower in HIMA from T2DM patients. In the same group, the expression of the Kir6.2 subunit was higher. Therefore, KATP channels might not be the only method of pinacidil-induced dilatation of T2DM HIMA. T2DM may decrease the level of Kir6.1, a dominant subunit in VSM of HIMA, altering the interaction between pinacidil and those channels.
Collapse
Affiliation(s)
- Jovana Rajkovic
- Institute of Pharmacology, Clinical Pharmacology and Toxicology, Faculty of Medicine, University of Belgrade, 11000 Belgrade, Serbia; (M.G.); (L.G.-B.)
| | - Miodrag Peric
- Dedinje Cardiovascular Institute, 11000 Belgrade, Serbia;
| | - Jelena Stanisic
- Laboratory for Molecular Biology and Endocrinology, Vinca Institute of Nuclear Sciences, University of Belgrade, 11000 Belgrade, Serbia; (J.S.); (S.T.)
| | - Milos Gostimirovic
- Institute of Pharmacology, Clinical Pharmacology and Toxicology, Faculty of Medicine, University of Belgrade, 11000 Belgrade, Serbia; (M.G.); (L.G.-B.)
| | - Radmila Novakovic
- Center for Genome Sequencing and Bioinformatics, Institute of Molecular Genetics and Genetic Engineering, University of Belgrade, 11000 Belgrade, Serbia;
| | - Vladimir Djokic
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, MN 55905, USA;
| | - Snezana Tepavcevic
- Laboratory for Molecular Biology and Endocrinology, Vinca Institute of Nuclear Sciences, University of Belgrade, 11000 Belgrade, Serbia; (J.S.); (S.T.)
| | - Jelena Rakocevic
- Institute of Histology and Embryology, Faculty of Medicine, University of Belgrade, 11000 Belgrade, Serbia; (J.R.); (M.L.-B.)
| | - Milica Labudovic-Borovic
- Institute of Histology and Embryology, Faculty of Medicine, University of Belgrade, 11000 Belgrade, Serbia; (J.R.); (M.L.-B.)
| | - Ljiljana Gojkovic-Bukarica
- Institute of Pharmacology, Clinical Pharmacology and Toxicology, Faculty of Medicine, University of Belgrade, 11000 Belgrade, Serbia; (M.G.); (L.G.-B.)
- Dedinje Cardiovascular Institute, 11000 Belgrade, Serbia;
| |
Collapse
|
3
|
Sedghi S, Khadra WZ, Pourafkari L, Knight PR, Alderson FA, Nader ND. Sevoflurane-mediated modulation of oxidative myocardial injury. J Cardiovasc Thorac Res 2023; 15:138-144. [PMID: 38028722 PMCID: PMC10590461 DOI: 10.34172/jcvtr.2023.31724] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2023] [Accepted: 08/19/2023] [Indexed: 12/01/2023] Open
Abstract
Introduction Volatile anesthetics offer protection when administered throughout an ischemic injury. We examined how volatile anesthetics modulate the cardiac myocytic injury associated with hydrogen peroxide. Methods Forty-eight Long-Evans rats were divided into four groups depending on the treatment: none (CONT), Glibenclamide (GLB); Sevoflurane (SEV); or GLB+SEV. Each group was further divided into two, one of which was exposed to hydrogen peroxide (H2O2). Oral GLB was administered 48 hours before myocardial isolation. All rats were anesthetized by intraperitoneal injection of Ketamine, and the hearts were harvested after heparinization. Cardiomyocytes were isolated using a combination of mechanical mincing and enzymatic digestion. After isolation, the aliquots of cells were exposed to H2O2 and FeSO4 for 30 minutes. The cell suspensions were then bubbled for 10 minutes with 100% oxygen and 1.5% SEV if appropriate. Apoptosis was detected by fluorescein-bound annexin-V (ANX-V), necrosis by propidium iodide, and ELISA assessed caspase-3 activity in all groups. Results There was an increase in apoptosis, necrosis, and caspase-3 activity in the cells following exposure to hydrogen peroxide. SEV reduced the rate of cell necrosis and apoptosis. Pretreatment with GLB did not alter the effects of SEV. Similarly, caspase-3 activity did not change with GLB, although SEV administration reduced this enzymatic activity in response to hydrogen peroxide. Conclusion In this oxidant injury model, we demonstrated that incubating isolated cardiomyocytes with SEV profoundly diminished H2O2-induced apoptotic and necrotic cells compared to their CONTs. These results support the hypothesis that KATP channels are not the sole mediators associated with anesthetic preconditioning.
Collapse
Affiliation(s)
- Siavash Sedghi
- Department of Anesthesiology, University at Buffalo, Jacobs School of Medicine and Biomedical Sciences, Buffalo, New York, USA
- VA Western New York Healthcare System, Buffalo, New York, USA
| | - Wiam Z. Khadra
- Department of Anesthesiology, University at Buffalo, Jacobs School of Medicine and Biomedical Sciences, Buffalo, New York, USA
- VA Western New York Healthcare System, Buffalo, New York, USA
| | - Leili Pourafkari
- Department of Anesthesiology, University at Buffalo, Jacobs School of Medicine and Biomedical Sciences, Buffalo, New York, USA
- Cardiac Imaging, The Lundquist Institute, Harbor-University of California at Los Angles Medical Center, Los Angles, USA
| | - Paul R. Knight
- Department of Anesthesiology, University at Buffalo, Jacobs School of Medicine and Biomedical Sciences, Buffalo, New York, USA
| | | | - Nader D. Nader
- Department of Anesthesiology, University at Buffalo, Jacobs School of Medicine and Biomedical Sciences, Buffalo, New York, USA
- VA Western New York Healthcare System, Buffalo, New York, USA
| |
Collapse
|
4
|
Deng J, Jiang Y, Chen ZB, Rhee JW, Deng Y, Wang ZV. Mitochondrial Dysfunction in Cardiac Arrhythmias. Cells 2023; 12:679. [PMID: 36899814 PMCID: PMC10001005 DOI: 10.3390/cells12050679] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2022] [Revised: 02/14/2023] [Accepted: 02/17/2023] [Indexed: 02/24/2023] Open
Abstract
Electrophysiological and structural disruptions in cardiac arrhythmias are closely related to mitochondrial dysfunction. Mitochondria are an organelle generating ATP, thereby satisfying the energy demand of the incessant electrical activity in the heart. In arrhythmias, the homeostatic supply-demand relationship is impaired, which is often accompanied by progressive mitochondrial dysfunction leading to reduced ATP production and elevated reactive oxidative species generation. Furthermore, ion homeostasis, membrane excitability, and cardiac structure can be disrupted through pathological changes in gap junctions and inflammatory signaling, which results in impaired cardiac electrical homeostasis. Herein, we review the electrical and molecular mechanisms of cardiac arrhythmias, with a particular focus on mitochondrial dysfunction in ionic regulation and gap junction action. We provide an update on inherited and acquired mitochondrial dysfunction to explore the pathophysiology of different types of arrhythmias. In addition, we highlight the role of mitochondria in bradyarrhythmia, including sinus node dysfunction and atrioventricular node dysfunction. Finally, we discuss how confounding factors, such as aging, gut microbiome, cardiac reperfusion injury, and electrical stimulation, modulate mitochondrial function and cause tachyarrhythmia.
Collapse
Affiliation(s)
- Jielin Deng
- Department of Diabetes and Cancer Metabolism, Beckman Research Institute, City of Hope National Medical Center, Duarte, CA 91010, USA
| | - Yunqiu Jiang
- Department of Diabetes and Cancer Metabolism, Beckman Research Institute, City of Hope National Medical Center, Duarte, CA 91010, USA
- Irell and Manella Graduate School of Biological Sciences, City of Hope National Medical Center, Duarte, CA 91010, USA
| | - Zhen Bouman Chen
- Irell and Manella Graduate School of Biological Sciences, City of Hope National Medical Center, Duarte, CA 91010, USA
- Department of Diabetes Complications and Metabolism, Arthur Riggs Diabetes and Metabolism Research Institute, Beckman Research Institute, City of Hope National Medical Center, Duarte, CA 91010, USA
- City of Hope Comprehensive Cancer Center, Duarte, CA 91010, USA
| | - June-Wha Rhee
- Irell and Manella Graduate School of Biological Sciences, City of Hope National Medical Center, Duarte, CA 91010, USA
- City of Hope Comprehensive Cancer Center, Duarte, CA 91010, USA
- Department of Medicine, Beckman Research Institute, City of Hope National Medical Center, Duarte, CA 91010, USA
| | - Yingfeng Deng
- Department of Diabetes and Cancer Metabolism, Beckman Research Institute, City of Hope National Medical Center, Duarte, CA 91010, USA
- Irell and Manella Graduate School of Biological Sciences, City of Hope National Medical Center, Duarte, CA 91010, USA
| | - Zhao V. Wang
- Department of Diabetes and Cancer Metabolism, Beckman Research Institute, City of Hope National Medical Center, Duarte, CA 91010, USA
- Irell and Manella Graduate School of Biological Sciences, City of Hope National Medical Center, Duarte, CA 91010, USA
- City of Hope Comprehensive Cancer Center, Duarte, CA 91010, USA
| |
Collapse
|
5
|
Premont A, Saadeh K, Edling C, Lewis R, Marr CM, Jeevaratnam K. Cardiac ion channel expression in the equine model - In-silico prediction utilising RNA sequencing data from mixed tissue samples. Physiol Rep 2022; 10:e15273. [PMID: 35880716 PMCID: PMC9316921 DOI: 10.14814/phy2.15273] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2021] [Revised: 03/19/2022] [Accepted: 04/03/2022] [Indexed: 06/15/2023] Open
Abstract
Understanding cardiomyocyte ion channel expression is crucial to understanding normal cardiac electrophysiology and underlying mechanisms of cardiac pathologies particularly arrhythmias. Hitherto, equine cardiac ion channel expression has rarely been investigated. Therefore, we aim to predict equine cardiac ion channel gene expression. Raw RNAseq data from normal horses from 9 datasets was retrieved from ArrayExpress and European Nucleotide Archive and reanalysed. The normalised (FPKM) read counts for a gene in a mix of tissue were hypothesised to be the average of the expected expression in each tissue weighted by the proportion of the tissue in the mix. The cardiac-specific expression was predicted by estimating the mean expression in each other tissues. To evaluate the performance of the model, predicted gene expression values were compared to the human cardiac gene expression. Cardiac-specific expression could be predicted for 91 ion channels including most expressed Na+ channels, K+ channels and Ca2+ -handling proteins. These revealed interesting differences from what would be expected based on human studies. These differences included predominance of NaV 1.4 rather than NaV 1.5 channel, and RYR1, SERCA1 and CASQ1 rather than RYR2, SERCA2, CASQ2 Ca2+ -handling proteins. Differences in channel expression not only implicate potentially different regulatory mechanisms but also pathological mechanisms of arrhythmogenesis.
Collapse
Affiliation(s)
- Antoine Premont
- Faculty of Health and Medical SciencesUniversity of SurreyGuildfordSurreyUK
| | - Khalil Saadeh
- Faculty of Health and Medical SciencesUniversity of SurreyGuildfordSurreyUK
- School of Clinical MedicineUniversity of CambridgeCambridgeUK
| | - Charlotte Edling
- Faculty of Health and Medical SciencesUniversity of SurreyGuildfordSurreyUK
| | - Rebecca Lewis
- Faculty of Health and Medical SciencesUniversity of SurreyGuildfordSurreyUK
| | - Celia M. Marr
- Faculty of Health and Medical SciencesUniversity of SurreyGuildfordSurreyUK
- School of Clinical MedicineUniversity of CambridgeCambridgeUK
- Rossdales Equine Hospital and Diagnostic CentreExningSuffolkUK
| | | |
Collapse
|
6
|
Lv J, Xiao X, Bi M, Tang T, Kong D, Diao M, Jiao Q, Chen X, Yan C, Du X, Jiang H. ATP-sensitive potassium channels: A double-edged sword in neurodegenerative diseases. Ageing Res Rev 2022; 80:101676. [PMID: 35724860 DOI: 10.1016/j.arr.2022.101676] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 04/15/2022] [Accepted: 06/14/2022] [Indexed: 11/25/2022]
Abstract
ATP-sensitive potassium channels (KATP channels), a group of vital channels that link the electrical activity of the cell membrane with cell metabolism, were discovered on the ventricular myocytes of guinea pigs by Noma using the patch-clamp technique in 1983. Subsequently, KATP channels have been found to be expressed in pancreatic β cells, cardiomyocytes, skeletal muscle cells, and nerve cells in the substantia nigra (SN), hippocampus, cortex, and basal ganglia. KATP channel openers (KCOs) diazoxide, nicorandil, minoxidil, and the KATP channel inhibitor glibenclamide have been shown to have anti-hypertensive, anti-myocardial ischemia, and insulin-releasing regulatory effects. Increasing evidence has suggested that KATP channels also play roles in Alzheimer's disease (AD), Parkinson's disease (PD), vascular dementia (VD), Huntington's disease (HD) and other neurodegenerative diseases. KCOs and KATP channel inhibitors protect neurons from injury by regulating neuronal excitability and neurotransmitter release, inhibiting abnormal protein aggregation and Ca2+ overload, reducing reactive oxygen species (ROS) production and microglia activation. However, KATP channels have dual effects in some cases. In this review, we focus on the roles of KATP channels and their related openers and inhibitors in neurodegenerative diseases. This will enable us to precisely take advantage of the KATP channels and provide new ideas for the treatment of neurodegenerative diseases.
Collapse
Affiliation(s)
- Jirong Lv
- Department of Physiology, Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders and State Key Disciplines: Physiology, School of Basic Medicine, Medical College, Qingdao University, Qingdao, China
| | - Xue Xiao
- Department of Physiology, Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders and State Key Disciplines: Physiology, School of Basic Medicine, Medical College, Qingdao University, Qingdao, China
| | - Mingxia Bi
- Department of Physiology, Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders and State Key Disciplines: Physiology, School of Basic Medicine, Medical College, Qingdao University, Qingdao, China
| | - Tingting Tang
- Department of Physiology, Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders and State Key Disciplines: Physiology, School of Basic Medicine, Medical College, Qingdao University, Qingdao, China
| | - Deao Kong
- Department of Physiology, Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders and State Key Disciplines: Physiology, School of Basic Medicine, Medical College, Qingdao University, Qingdao, China
| | - Meining Diao
- Department of Physiology, Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders and State Key Disciplines: Physiology, School of Basic Medicine, Medical College, Qingdao University, Qingdao, China
| | - Qian Jiao
- Department of Physiology, Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders and State Key Disciplines: Physiology, School of Basic Medicine, Medical College, Qingdao University, Qingdao, China
| | - Xi Chen
- Department of Physiology, Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders and State Key Disciplines: Physiology, School of Basic Medicine, Medical College, Qingdao University, Qingdao, China
| | - Chunling Yan
- Department of Physiology, Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders and State Key Disciplines: Physiology, School of Basic Medicine, Medical College, Qingdao University, Qingdao, China
| | - Xixun Du
- Department of Physiology, Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders and State Key Disciplines: Physiology, School of Basic Medicine, Medical College, Qingdao University, Qingdao, China.
| | - Hong Jiang
- Department of Physiology, Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders and State Key Disciplines: Physiology, School of Basic Medicine, Medical College, Qingdao University, Qingdao, China.
| |
Collapse
|
7
|
Caruba T, Charles-Nelson A, Alexander JH, Mehta RH, Heringlake M, Toller W, Yavchitz A, Sabatier B, Chatellier G, Cholley B. Prophylactic levosimendan in patients with low ejection fraction undergoing coronary artery bypass grafting: A pooled analysis of two multicentre randomised controlled trials. Anaesth Crit Care Pain Med 2022; 41:101107. [PMID: 35643391 DOI: 10.1016/j.accpm.2022.101107] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Revised: 04/01/2022] [Accepted: 04/01/2022] [Indexed: 11/18/2022]
Abstract
OBJECTIVES To assess the effect of preoperative levosimendan on mortality at Day-90 in patients with left ventricular ejection fraction (LVEF) ≤ 40%, and to investigate a possible differential effect between patients undergoing isolated coronary artery bypass grafting (CABG) versus CABG combined with valve replacement surgery. DESIGN Pooled analysis of two multicentre randomised controlled trials (RCT) investigating prophylactic levosimendan versus placebo prior to CABG surgery on mortality at Day-90 in patients with LVEF ≤ 40%. A meta-analysis of all RCT investigating the same issue was also conducted. RESULTS A cohort of 1084 patients (809 isolated CABG, and 275 combined surgery) resulted from the merging of LEVO-CTS and LICORN databases. Seventy-two patients were dead at day 90. The mortality at day 90 was not different between levosimendan and placebo (Hazard Ratio (HR): 0.73, 95% CI: 0.41-1.28, p = 0.27). However, there was a significant interaction between the type of surgery and the study drug (p = 0.004). We observed a decrease in mortality at day 90 in the isolated CABG subgroup (HR: 0.39, 95% CI: 0.19-0.82, p = 0.013), but not in the combined surgery subgroup (HR: 1.73, 95% CI: 0.77-3.92, p = 0.19). The meta-analysis of 6 RCT involving 1441 patients confirmed the differential effect on mortality at day 30 between the 2 subgroups. CONCLUSIONS Preoperative levosimendan did not reduce mortality in a mixed surgical population with LV dysfunction. However, the subgroup of patients undergoing isolated CABG had a reduction in mortality at day 90, whereas there was no significant effect in combined surgery patients. This finding requires confirmation with a specific prospective trial.
Collapse
Affiliation(s)
- Thibaut Caruba
- AP-HP, Hôpital Européen Georges Pompidou, F-75015 Paris, France
| | | | - John H Alexander
- Duke Clinical Research Institute, Duke University School of Medicine, Durham, NC, USA
| | - Rajendra H Mehta
- Duke Clinical Research Institute, Duke University School of Medicine, Durham, NC, USA
| | - Matthias Heringlake
- Dept. of Anaesthesiology and Intensive Care Medicine, Heart- and Diabetes Center, Karlsburg, Germany
| | | | - Amelie Yavchitz
- AP-HP, Hôpital Européen Georges Pompidou, F-75015 Paris, France
| | - Brigitte Sabatier
- AP-HP, Hôpital Européen Georges Pompidou, F-75015 Paris, France; Inserm UMR_1138, Centre de Recherche des Cordeliers, Sorbonne Université, Université de Paris Cité, F-75006 Paris, France and Inria, HeKA, PariSantéCampus, Paris, France
| | - Gilles Chatellier
- AP-HP, Hôpital Européen Georges Pompidou, F-75015 Paris, France; Université de Paris, F-75006 Paris, France
| | - Bernard Cholley
- AP-HP, Hôpital Européen Georges Pompidou, F-75015 Paris, France; Université de Paris, INSERM UMR_S 1140 "Innovations Thérapeutiques en Hémostase", F-75006 Paris, France.
| |
Collapse
|
8
|
Shiels HA, White E, Couturier CS, Hall D, Royal S, Galli GL, Stecyk JA. The air-breathing Alaska blackfish (Dallia pectoralis) remodels ventricular Ca2+ cycling with chronic hypoxic submergence to maintain ventricular contractility. Curr Res Physiol 2022; 5:25-35. [PMID: 35072107 PMCID: PMC8763628 DOI: 10.1016/j.crphys.2022.01.001] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Revised: 12/22/2021] [Accepted: 01/06/2022] [Indexed: 11/24/2022] Open
Abstract
The Alaska blackfish (Dallia pectoralis) is a facultative air-breather endemic to northern latitudes where it remains active in winter under ice cover in cold hypoxic waters. To understand the changes in cellular Ca2+ cycling that allow the heart to function in cold hypoxic water, we acclimated Alaska blackfish to cold (5 °C) normoxia or cold hypoxia (2.1–4.2 kPa; no air access) for 5–8 weeks. We then assessed the impact of the acclimation conditions on intracellular Ca2+ transients (Δ[Ca2+]i) of isolated ventricular myocytes and contractile performance of isometrically-contracting ventricular strips. Measurements were obtained at various contractile frequencies (0.2–0.6 Hz) in normoxia, during acute exposure to hypoxia, and reoxygenation at 5 °C. The results show that hypoxia-acclimated Alaska blackfish compensate against the depressive effects of hypoxia on excitation-contraction coupling by remodelling cellular Δ[Ca2+]i to maintain ventricular contractility. When measured at 0.2 Hz in normoxia, hypoxia-acclimated ventricular myocytes had a 3.8-fold larger Δ[Ca2+]i peak amplitude with a 4.1-fold faster rate of rise, compared to normoxia-acclimated ventricular myocytes. At the tissue level, maximal developed force was 2.1-fold greater in preparations from hypoxia-acclimated animals. However, maximal attainable contraction frequencies in hypoxia were lower in hypoxia-acclimated myocytes and strips than preparations from normoxic animals. Moreover, the inability of hypoxia-acclimated ventricular myocytes and strips to contract at high frequency persisted upon reoxygenation. Overall, the findings indicate that hypoxia alters aspects of Alaska blackfish cardiac myocyte Ca2+ cycling, and that there may be consequences for heart rate elevation during hypoxia, which may impact cardiac output in vivo. The air-breathing Alaska blackfish remains active under ice cover in hypoxic waters. Maintained activity is supported by compensation of intracellular Ca2+ transients. The compensation permits greater ventricular maximal developed force. However, maximal attainable contraction frequencies are limited by hypoxia exposure.
Collapse
|
9
|
Balci CN, Firat T, Acar N, Kukner A. Carvacrol treatment opens Kir6.2 ATP-dependent potassium channels and prevents apoptosis on rat testis following ischemia-reperfusion injury model. ROMANIAN JOURNAL OF MORPHOLOGY AND EMBRYOLOGY 2021; 62:179-190. [PMID: 34609420 PMCID: PMC8597367 DOI: 10.47162/rjme.62.1.17] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Testicular torsion is a urological problem that causes subfertility and testicular damage in males. Testis torsion and detorsion lead to ischemia–reperfusion (IR) injury in the testis. Testicular IR injury causes the increase of reactive oxygen species (ROS), oxidative stress (OS) and germ cell-specific apoptosis. In this study, we aimed to investigate whether Carvacrol has a protective effect on testicular IR injury and its effects on Kir6.2 channels, which is a member of adenosine triphosphate (ATP)-dependent potassium channels. In the study, 2–4 months old 36 albino Wistar rats were used. For experimental testicular IR model, the left testis was rotated counterclockwise at 720° for two hours, and after two hours following torsion, detorsion was performed. Carvacrol was dissolved in 5% Dimethyl Sulfoxide (DMSO) at a dose of 73 mg/kg and half an hour before detorsion, 0.2 mL was administered intraperitoneally. In testicular tissues, caspase 3 and Kir6.2 immunoexpressions were examined. Serum malondialdehyde (MDA) and testosterone levels were measured. Apoptotic cells and serum MDA levels were significantly decreased and Kir6.2 activation was significantly increased in Carvacrol-administrated IR group. As a result of our study, Carvacrol may activates Kir6.2 channels and inhibits apoptosis and may have a protective effect on testicular IR injury.
Collapse
Affiliation(s)
- Cemre Nur Balci
- Department of Histology and Embryology, Faculty of Medicine, Akdeniz University, Antalya, Turkey;
| | | | | | | |
Collapse
|
10
|
Raza ST, Singh SP, Rizvi S, Zaidi A, Srivastava S, Hussain A, Mahdi F. Association of eNOS (G894T, rs1799983) and KCNJ11 (E23K, rs5219) gene polymorphism with coronary artery disease in North Indian population. Afr Health Sci 2021; 21:1163-1171. [PMID: 35222579 PMCID: PMC8843271 DOI: 10.4314/ahs.v21i3.25] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Background Endothelial nitric oxide synthase (eNOS) and potassium voltage-gated channel subfamily J member 11 (KCNJ11) could be the candidate genes for coronary artery disease (CAD). This study investigated the relationship of the eNOS (rs1799983) and KCNJ11 (rs5219) polymorphisms with the presence and severity of CAD in the North Indian population. Methods This study included 300 subjects, 150 CAD cases and 150 healthy controls. Single nucleotide polymorphism was evaluated by Polymerase chain reaction and Restriction fragment length polymorphism (PCR-RFLP). Analysis was performed by SPSS (version 21.0). Results We observed that KK genotype of KCNJ11E23K (rs5219) polymorphism (P=0.0001) genotypes and K allele (P=0.0001) was found to be a positive risk factor and strongly associated with CAD. In the case of eNOSG894T (rs1799983) there was no association found with CAD. Conclusion These results illustrate the probability of associations between SNPs and CAD although specific genetic polymorphisms affecting ion channel function and expression have still to be clarified by further investigations involving larger cohorts.
Collapse
Affiliation(s)
- Syed Tasleem Raza
- Department of Biochemistry, Era's Lucknow Medical College and Hospital, Lucknow, India
| | - Sachendra P Singh
- Department of Biochemistry, Era's Lucknow Medical College and Hospital, Lucknow, India
| | - Saliha Rizvi
- Department of Biochemistry, Era's Lucknow Medical College and Hospital, Lucknow, India
| | - Alina Zaidi
- Department of Biochemistry, Era's Lucknow Medical College and Hospital, Lucknow, India
| | - Sanchita Srivastava
- Department of Biochemistry, Era's Lucknow Medical College and Hospital, Lucknow, India
| | - Arif Hussain
- Department of Molecular biology, Manipal Academy of Higher Education
| | - Farzana Mahdi
- Department of Biochemistry, Era's Lucknow Medical College and Hospital, Lucknow, India
| |
Collapse
|
11
|
Mutated CCDC51 Coding for a Mitochondrial Protein, MITOK Is a Candidate Gene Defect for Autosomal Recessive Rod-Cone Dystrophy. Int J Mol Sci 2021; 22:ijms22157875. [PMID: 34360642 PMCID: PMC8346125 DOI: 10.3390/ijms22157875] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2021] [Revised: 07/05/2021] [Accepted: 07/15/2021] [Indexed: 11/17/2022] Open
Abstract
The purpose of this work was to identify the gene defect underlying a relatively mild rod-cone dystrophy (RCD), lacking disease-causing variants in known genes implicated in inherited retinal disorders (IRD), and provide transcriptomic and immunolocalization data to highlight the best candidate. The DNA of the female patient originating from a consanguineous family revealed no large duplication or deletion, but several large homozygous regions. In one of these, a homozygous frameshift variant, c.244_246delins17 p.(Trp82Valfs*4); predicted to lead to a nonfunctional protein, was identified in CCDC51. CCDC51 encodes the mitochondrial coiled-coil domain containing 51 protein, also called MITOK. MITOK ablation causes mitochondrial dysfunction. Here we show for the first time that CCDC51/MITOK localizes in the retina and more specifically in the inner segments of the photoreceptors, well known to contain mitochondria. Mitochondrial proteins have previously been implicated in IRD, although usually in association with syndromic disease, unlike our present case. Together, our findings add another ultra-rare mutation implicated in non-syndromic IRD, whose pathogenic mechanism in the retina needs to be further elucidated.
Collapse
|
12
|
Mondéjar-Parreño G, Cogolludo A, Perez-Vizcaino F. Potassium (K +) channels in the pulmonary vasculature: Implications in pulmonary hypertension Physiological, pathophysiological and pharmacological regulation. Pharmacol Ther 2021; 225:107835. [PMID: 33744261 DOI: 10.1016/j.pharmthera.2021.107835] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2021] [Revised: 02/26/2021] [Accepted: 03/02/2021] [Indexed: 02/06/2023]
Abstract
The large K+ channel functional diversity in the pulmonary vasculature results from the multitude of genes expressed encoding K+ channels, alternative RNA splicing, the post-transcriptional modifications, the presence of homomeric or heteromeric assemblies of the pore-forming α-subunits and the existence of accessory β-subunits modulating the functional properties of the channel. K+ channels can also be regulated at multiple levels by different factors controlling channel activity, trafficking, recycling and degradation. The activity of these channels is the primary determinant of membrane potential (Em) in pulmonary artery smooth muscle cells (PASMC), providing an essential regulatory mechanism to dilate or contract pulmonary arteries (PA). K+ channels are also expressed in pulmonary artery endothelial cells (PAEC) where they control resting Em, Ca2+ entry and the production of different vasoactive factors. The activity of K+ channels is also important in regulating the population and phenotype of PASMC in the pulmonary vasculature, since they are involved in cell apoptosis, survival and proliferation. Notably, K+ channels play a major role in the development of pulmonary hypertension (PH). Impaired K+ channel activity in PH results from: 1) loss of function mutations, 2) downregulation of its expression, which involves transcription factors and microRNAs, or 3) decreased channel current as a result of increased vasoactive factors (e.g., hypoxia, 5-HT, endothelin-1 or thromboxane), exposure to drugs with channel-blocking properties, or by a reduction in factors that positively regulate K+ channel activity (e.g., NO and prostacyclin). Restoring K+ channel expression, its intracellular trafficking and the channel activity is an attractive therapeutic strategy in PH.
Collapse
Affiliation(s)
- Gema Mondéjar-Parreño
- Department of Pharmacology and Toxicology, School of Medicine, University Complutense of Madrid, Spain; Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Madrid, Spain; Ciber Enfermedades Respiratorias (CIBERES), Spain
| | - Angel Cogolludo
- Department of Pharmacology and Toxicology, School of Medicine, University Complutense of Madrid, Spain; Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Madrid, Spain; Ciber Enfermedades Respiratorias (CIBERES), Spain
| | - Francisco Perez-Vizcaino
- Department of Pharmacology and Toxicology, School of Medicine, University Complutense of Madrid, Spain; Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Madrid, Spain; Ciber Enfermedades Respiratorias (CIBERES), Spain.
| |
Collapse
|
13
|
Saadeh K, Fazmin IT. Mitochondrial Dysfunction Increases Arrhythmic Triggers and Substrates; Potential Anti-arrhythmic Pharmacological Targets. Front Cardiovasc Med 2021; 8:646932. [PMID: 33659284 PMCID: PMC7917191 DOI: 10.3389/fcvm.2021.646932] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2020] [Accepted: 01/26/2021] [Indexed: 12/31/2022] Open
Abstract
Incidence of cardiac arrhythmias increases significantly with age. In order to effectively stratify arrhythmic risk in the aging population it is crucial to elucidate the relevant underlying molecular mechanisms. The changes underlying age-related electrophysiological disruption appear to be closely associated with mitochondrial dysfunction. Thus, the present review examines the mechanisms by which age-related mitochondrial dysfunction promotes arrhythmic triggers and substrate. Namely, via alterations in plasmalemmal ionic currents (both sodium and potassium), gap junctions, cellular Ca2+ homeostasis, and cardiac fibrosis. Stratification of patients' mitochondrial function status permits application of appropriate anti-arrhythmic therapies. Here, we discuss novel potential anti-arrhythmic pharmacological interventions that specifically target upstream mitochondrial function and hence ameliorates the need for therapies targeting downstream changes which have constituted traditional antiarrhythmic therapy.
Collapse
Affiliation(s)
- Khalil Saadeh
- School of Clinical Medicine, University of Cambridge, Cambridge, United Kingdom.,Faculty of Health and Medical Sciences, University of Surrey, Guildford, United Kingdom
| | - Ibrahim Talal Fazmin
- School of Clinical Medicine, University of Cambridge, Cambridge, United Kingdom.,Faculty of Health and Medical Sciences, University of Surrey, Guildford, United Kingdom.,Royal Papworth Hospital NHS Foundation Trust, Cambridge, United Kingdom
| |
Collapse
|
14
|
Stecyk JAW, Couturier CS, Abramochkin DV, Hall D, Arrant-Howell A, Kubly KL, Lockmann S, Logue K, Trueblood L, Swalling C, Pinard J, Vogt A. Cardiophysiological responses of the air-breathing Alaska blackfish to cold acclimation and chronic hypoxic submergence at 5°C. J Exp Biol 2020; 223:jeb225730. [PMID: 33020178 PMCID: PMC7687868 DOI: 10.1242/jeb.225730] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2020] [Accepted: 09/28/2020] [Indexed: 01/29/2023]
Abstract
The Alaska blackfish (Dallia pectoralis) remains active at cold temperatures when experiencing aquatic hypoxia without air access. To discern the cardiophysiological adjustments that permit this behaviour, we quantified the effect of acclimation from 15°C to 5°C in normoxia (15N and 5N fish), as well as chronic hypoxic submergence (6-8 weeks; ∼6.3-8.4 kPa; no air access) at 5°C (5H fish), on in vivo and spontaneous heart rate (fH), electrocardiogram, ventricular action potential (AP) shape and duration (APD), the background inward rectifier (IK1) and rapid delayed rectifier (IKr) K+ currents and ventricular gene expression of proteins involved in excitation-contraction coupling. In vivo fH was ∼50% slower in 5N than in 15N fish, but 5H fish did not display hypoxic bradycardia. Atypically, cold acclimation in normoxia did not induce shortening of APD or alter resting membrane potential. Rather, QT interval and APD were ∼2.6-fold longer in 5N than in 15N fish because outward IK1 and IKr were not upregulated in 5N fish. By contrast, chronic hypoxic submergence elicited a shortening of QT interval and APD, driven by an upregulation of IKr The altered electrophysiology of 5H fish was accompanied by increased gene expression of kcnh6 (3.5-fold; Kv11.2 of IKr), kcnj12 (7.4-fold; Kir2.2 of IK1) and kcnj14 (2.9-fold; Kir2.4 of IK1). 5H fish also exhibited a unique gene expression pattern that suggests modification of ventricular Ca2+ cycling. Overall, the findings reveal that Alaska blackfish exposed to chronic hypoxic submergence prioritize the continuation of cardiac performance to support an active lifestyle over reducing cardiac ATP demand.
Collapse
Affiliation(s)
- Jonathan A W Stecyk
- Department of Biological Sciences, University of Alaska Anchorage, Anchorage, AK 99508, USA
| | - Christine S Couturier
- Department of Biological Sciences, University of Alaska Anchorage, Anchorage, AK 99508, USA
| | - Denis V Abramochkin
- Department of Human and Animal Physiology, Lomonosov Moscow State University, 1-12 Leninskiye Gory, 119991 Moscow, Russia
- Ural Federal University, 19 Mira Street, 620002 Ekaterinburg, Russia
- Laboratory of Cardiac Physiology, Institute of Physiology of Kоmi Science Centre of the Ural Branch of the Russian Academy of Sciences, FRC Komi SC UB RAS, 50 Pervomayskaya Str., 167982 Syktyvkar, Komi Republic, Russia
| | - Diarmid Hall
- Department of Biological Sciences, University of Alaska Anchorage, Anchorage, AK 99508, USA
| | - Asia Arrant-Howell
- Department of Biological Sciences, University of Alaska Anchorage, Anchorage, AK 99508, USA
| | - Kerry L Kubly
- Department of Biological Sciences, University of Alaska Anchorage, Anchorage, AK 99508, USA
| | - Shyanne Lockmann
- Department of Biological Sciences, University of Alaska Anchorage, Anchorage, AK 99508, USA
| | - Kyle Logue
- Department of Biological Sciences, University of Alaska Anchorage, Anchorage, AK 99508, USA
| | - Lenett Trueblood
- Department of Biological Sciences, University of Alaska Anchorage, Anchorage, AK 99508, USA
| | - Connor Swalling
- Department of Biological Sciences, University of Alaska Anchorage, Anchorage, AK 99508, USA
| | - Jessica Pinard
- Department of Biological Sciences, University of Alaska Anchorage, Anchorage, AK 99508, USA
| | - Angela Vogt
- Department of Biological Sciences, University of Alaska Anchorage, Anchorage, AK 99508, USA
| |
Collapse
|
15
|
Djokic V, Jankovic-Raznatovic S, Novakovic R, Kostic M, Rajkovic J, Labudovic-Borovic M, Rakocevic J, Stanisic J, Djuric M, Gojkovic-Bukarica L. Effect of gestational diabetes mellitus and pregnancy-induced hypertension on human umbilical vein smooth muscle K ATP channels. Exp Mol Pathol 2019; 111:104323. [PMID: 31669131 DOI: 10.1016/j.yexmp.2019.104323] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2019] [Revised: 09/03/2019] [Accepted: 10/21/2019] [Indexed: 01/25/2023]
Abstract
Gestational diabetes mellitus (GDM) and pregnancy-induced hypertension (PIH) can jeopardize mother and/or fetus. Vascular ATP-sensitive potassium (KATP) channels most likely participate in the processes of diabetes and hypertension. The aim of this research was to examine whether GDM and PIH cause changes in the expression and function of KATP channels in vascular smooth muscle of human umbilical vein (HUV). Western blot and immunohistochemistry detected significantly decreased expression of Kir6.1 subunit of KATP channels in GDM and PIH, while the expression of SUR2B was unchanged. In GDM, a K+ channel opener, pinacidil caused reduced relaxation of the endothelium-denuded HUVs compared to normal pregnancy. However, its effects in HUVs from PIH subjects were similar to normal pregnancy. In all groups KATP channel blocker glibenclamide antagonized the relaxation of HUV induced by pinacidil without change in the maximal relaxations indicating additional KATP channel-independent mechanisms of pinacidil action. Iberiotoxin, a selective antagonist of large-conductance calcium-activated potassium channels, inhibited the relaxant effect of pinacidil in PIH, but not in normal pregnancy and GDM. Experiments performed in K+-rich solution confirmed the existence of K+-independent effects of pinacidil, which also appear to be impaired in GDM and PIH. Thus, the expression of KATP channels is decreased in GDM and PIH. In GDM, vasorelaxant response of HUV to pinacidil is reduced, while in PIH it remains unchanged. It is very likely that KATP channels modulation and more detailed insight in KATP channel-independent actions of pinacidil may be precious in the therapy of pathological pregnancies.
Collapse
Affiliation(s)
- Vladimir Djokic
- Department of Pharmacology, Clinical Pharmacology and Toxicology, Faculty of Medicine, University of Belgrade, 11000 Belgrade, Serbia.
| | - Svetlana Jankovic-Raznatovic
- Department of Obstetrics and Gynecology "Narodni front", Faculty of Medicine, University of Belgrade, 11000 Belgrade, Serbia
| | - Radmila Novakovic
- Department of Pharmacology, Clinical Pharmacology and Toxicology, Faculty of Medicine, University of Belgrade, 11000 Belgrade, Serbia
| | - Milan Kostic
- Vinca Institute of Nuclear Sciences, 11000 Belgrade, Serbia
| | - Jovana Rajkovic
- Department of Pharmacology, Clinical Pharmacology and Toxicology, Faculty of Medicine, University of Belgrade, 11000 Belgrade, Serbia
| | - Milica Labudovic-Borovic
- Institute of Histology and Embryology "Aleksandar Dj. Kostic", Faculty of Medicine, University of Belgrade, 11000 Belgrade, Serbia
| | - Jelena Rakocevic
- Institute of Histology and Embryology "Aleksandar Dj. Kostic", Faculty of Medicine, University of Belgrade, 11000 Belgrade, Serbia
| | | | - Milos Djuric
- Department of Pharmacology, Clinical Pharmacology and Toxicology, Faculty of Medicine, University of Belgrade, 11000 Belgrade, Serbia
| | - Ljiljana Gojkovic-Bukarica
- Department of Pharmacology, Clinical Pharmacology and Toxicology, Faculty of Medicine, University of Belgrade, 11000 Belgrade, Serbia
| |
Collapse
|
16
|
Djuric M, Nikolic Turnic T, Kostic S, Radonjic K, Jeremic J, Petkovic A, Bradic J, Milosavljevic I, Srejovic I, Zivkovic V, Djuric D, Jakovljevic V, Stevanovic P. Inhibition of gasotransmitters production and calcium influx affect cardiodynamic variables and cardiac oxidative stress in propofol-anesthetized male Wistar rats. Can J Physiol Pharmacol 2019; 97:850-856. [DOI: 10.1139/cjpp-2018-0719] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
It has been assumed that the cardioprotective effects of propofol are due to its non-anesthetic pleiotropic cardiac and vasodilator effects, in which gasotransmitters (NO, H2S, and CO) as well as calcium influx could be involved. The study on isolated rat heart was performed using 4 experimental groups (n = 7 in each): (1) bolus injection of propofol (100 mg/kg body mass, i.p.); (2) L-NAME (NO synthase inhibitor, 60 mg/kg body mass, i.p.) + propofol; (3) DL-PAG (H2S synthase inhibitor, 50 mg/kg body mass, i.p.) + propofol; (4) ZnPPIX (CO synthase inhibitor, 50 μmol/kg body mass, i.p.) + propofol. Before and after the verapamil (3 μmol/L) administration, cardiodynamic parameters were recorded (dp/dtmax, dp/dtmin, systolic left ventricular pressure, diastolic left ventricular pressure, heart rate, coronary flow), as well as coronary and cardiac oxidative stress parameters. The results showed significant increases of diastolic left ventricular pressure following NO and CO inhibition, but also increases of coronary flow following H2S and CO inhibition. Following verapamil administration, significant decreases of dp/dtmax were noted after NO and CO inhibition, then increase of diastolic left ventricular pressure following CO inhibition, and increase of coronary flow following NO, H2S, or CO inhibition. Oxidative stress markers were increased but catalase activity was significantly decreased in cardiac tissue. Gasotransmitters and calcium influx are involved in pleiotropic cardiovascular effects of propofol in male Wistar rats.
Collapse
Affiliation(s)
- M. Djuric
- Department of Anesthesiology, Reanimatology and Intensive Care Medicine, University Clinical Hospital Center “Dr. Dragisa Misovic - Dedinje”, Belgrade, Serbia
| | - T. Nikolic Turnic
- Department of Pharmacy, Faculty of Medical Sciences, University of Kragujevac, Kragujevac, Serbia
| | - S. Kostic
- Faculty of Medicine, University of Belgrade, Belgrade, Serbia
| | - K. Radonjic
- Department of Pharmacy, Faculty of Medical Sciences, University of Kragujevac, Kragujevac, Serbia
| | - J. Jeremic
- Department of Pharmacy, Faculty of Medical Sciences, University of Kragujevac, Kragujevac, Serbia
| | - A. Petkovic
- Department of Pharmacy, Faculty of Medical Sciences, University of Kragujevac, Kragujevac, Serbia
| | - J. Bradic
- Department of Pharmacy, Faculty of Medical Sciences, University of Kragujevac, Kragujevac, Serbia
| | - I. Milosavljevic
- Department of Pharmacy, Faculty of Medical Sciences, University of Kragujevac, Kragujevac, Serbia
| | - I. Srejovic
- Department of Physiology, Faculty of Medical Sciences, University of Kragujevac, Kragujevac, Serbia
| | - V. Zivkovic
- Department of Physiology, Faculty of Medical Sciences, University of Kragujevac, Kragujevac, Serbia
| | - D. Djuric
- Institute of Medical Physiology “Richard Burian”, Faculty of Medicine, University of Belgrade, Belgrade, Serbia
| | - V. Jakovljevic
- Department of Physiology, Faculty of Medical Sciences, University of Kragujevac, Kragujevac, Serbia
- Department of Human Pathology, 1st Moscow State Medical, University IM Sechenov, Moscow, Russian Federation
| | - P. Stevanovic
- Department of Anesthesiology, Reanimatology and Intensive Care Medicine, University Clinical Hospital Center “Dr. Dragisa Misovic - Dedinje”, Belgrade, Serbia
| |
Collapse
|
17
|
Chuang CC, Zhou T, Olfert IM, Zuo L. Hypoxic Preconditioning Attenuates Reoxygenation-Induced Skeletal Muscle Dysfunction in Aged Pulmonary TNF-α Overexpressing Mice. Front Physiol 2019; 9:1720. [PMID: 30622474 PMCID: PMC6308319 DOI: 10.3389/fphys.2018.01720] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2017] [Accepted: 11/15/2018] [Indexed: 11/26/2022] Open
Abstract
Aim: Skeletal muscle subjected to hypoxia followed by reoxygenation is susceptible to injury and subsequent muscle function decline. This phenomenon can be observed in the diaphragm during strenuous exercise or in pulmonary diseases such as chronic obstructive pulmonary diseases (COPD). Previous studies have shown that PO2 cycling or hypoxic preconditioning (HPC), as it can also be referred to as, protects muscle function via mechanisms involving reactive oxygen species (ROS). However, this HPC protection has not been fully elucidated in aged pulmonary TNF-α overexpressing (Tg+) mice (a COPD-like model). We hypothesize that HPC can exert protection on the diaphragms of Tg+ mice during reoxygenation through pathways involving ROS/phosphoinositide 3-kinase (PI3K)/protein kinase B (Akt)/extracellular signal regulated kinase (ERK), as well as the downstream activation of mitochondrial ATP-sensitive potassium channel (mitoKATP) and inhibition of mitochondrial permeability transition pore (mPTP). Methods: Isolated Tg+ diaphragm muscle strips were pre-treated with inhibitors for ROS, PI3K, Akt, ERK, or a combination of mitoKATP inhibitor and mPTP opener, respectively, prior to HPC. Another two groups of muscles were treated with either mitoKATP activator or mPTP inhibitor without HPC. Muscles were treated with 30-min hypoxia, followed by 15-min reoxygenation. Data were analyzed by multi-way ANOVA and expressed as means ± SE. Results: Muscle treated with HPC showed improved muscle function during reoxygenation (n = 5, p < 0.01). Inhibition of ROS, PI3K, Akt, or ERK abolished the protective effect of HPC. Simultaneous inhibition of mitoKATP and activation of mPTP also diminished HPC effects. By contrast, either the opening of mitoKATP channel or the closure of mPTP provided a similar protective effect to HPC by alleviating muscle function decline, suggesting that mitochondria play a role in HPC initiation (n = 5; p < 0.05). Conclusion: Hypoxic preconditioning may protect respiratory skeletal muscle function in Tg+ mice during reoxygenation through redox-sensitive signaling cascades and regulations of mitochondrial channels.
Collapse
Affiliation(s)
- Chia-Chen Chuang
- Radiologic Sciences and Respiratory Therapy Division, School of Health and Rehabilitation Sciences, The Ohio State University College of Medicine, The Ohio State University Wexner Medical Center, Columbus, OH, United States.,Interdisciplinary Biophysics Graduate Program, The Ohio State University, Columbus, OH, United States
| | - Tingyang Zhou
- Radiologic Sciences and Respiratory Therapy Division, School of Health and Rehabilitation Sciences, The Ohio State University College of Medicine, The Ohio State University Wexner Medical Center, Columbus, OH, United States.,Interdisciplinary Biophysics Graduate Program, The Ohio State University, Columbus, OH, United States
| | - I Mark Olfert
- Division of Exercise Physiology, West Virginia University School of Medicine, Morgantown, WV, United States
| | - Li Zuo
- Radiologic Sciences and Respiratory Therapy Division, School of Health and Rehabilitation Sciences, The Ohio State University College of Medicine, The Ohio State University Wexner Medical Center, Columbus, OH, United States.,Interdisciplinary Biophysics Graduate Program, The Ohio State University, Columbus, OH, United States.,Department of Biology, The University of Maine, Presque Isle, ME, United States
| |
Collapse
|
18
|
Kon N, Abe N, Miyazaki M, Mushiake H, Kazama I. Partial exposure of frog heart to high-potassium solution: an easily reproducible model mimicking ST segment changes. J Vet Med Sci 2018; 80:578-582. [PMID: 29503350 PMCID: PMC5938182 DOI: 10.1292/jvms.18-0010] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
By simply inducing burn injuries on the bullfrog heart, we previously reported a simple
model of abnormal ST segment changes observed in human ischemic heart disease. In the
present study, instead of inducing burn injuries, we partially exposed the surface of the
frog heart to high-potassium (K+) solution to create a concentration gradient
of the extracellular K+ within the myocardium. Dual recordings of ECG and the
cardiac action potential demonstrated significant elevation of the ST segment and the
resting membrane potential, indicating its usefulness as a simple model of heart injury.
Additionally, from our results, Na+/K+-ATPase activity was thought
to be primarily responsible for generating the K+ concentration gradient and
inducing the ST segment changes in ECG.
Collapse
Affiliation(s)
- Nobuaki Kon
- Department of Physiology, Tohoku University Graduate School of Medicine, Seiryo-cho, Aoba-ku, Sendai, Miyagi 980-8575, Japan
| | - Nozomu Abe
- Department of Physiology, Tohoku University Graduate School of Medicine, Seiryo-cho, Aoba-ku, Sendai, Miyagi 980-8575, Japan.,Department of Anesthesiology, Tohoku University Hospital, Seiryo-cho, Aoba-ku, Sendai, Miyagi 980-8575, Japan
| | - Masahiro Miyazaki
- Department of Physiology, Tohoku University Graduate School of Medicine, Seiryo-cho, Aoba-ku, Sendai, Miyagi 980-8575, Japan
| | - Hajime Mushiake
- Department of Physiology, Tohoku University Graduate School of Medicine, Seiryo-cho, Aoba-ku, Sendai, Miyagi 980-8575, Japan
| | - Itsuro Kazama
- School of Nursing, Miyagi University, Gakuen, Taiwa-cho, Kurokawa-gun, Miyagi 981-3298, Japan.,Department of Physiology, Tohoku University Graduate School of Medicine, Seiryo-cho, Aoba-ku, Sendai, Miyagi 980-8575, Japan
| |
Collapse
|
19
|
A Protective Role of Glibenclamide in Inflammation-Associated Injury. Mediators Inflamm 2017; 2017:3578702. [PMID: 28740332 PMCID: PMC5504948 DOI: 10.1155/2017/3578702] [Citation(s) in RCA: 62] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2016] [Revised: 03/26/2017] [Accepted: 04/06/2017] [Indexed: 12/13/2022] Open
Abstract
Glibenclamide is the most widely used sulfonylurea drug for the treatment of type 2 diabetes mellitus (DM). Recent studies have suggested that glibenclamide reduced adverse neuroinflammation and improved behavioral outcomes following central nervous system (CNS) injury. We reviewed glibenclamide's anti-inflammatory effects: abundant evidences have shown that glibenclamide exerted an anti-inflammatory effect in respiratory, digestive, urological, cardiological, and CNS diseases, as well as in ischemia-reperfusion injury. Glibenclamide might block KATP channel, Sur1-Trpm4 channel, and NOD-like receptor pyrin domain containing 3 (NLRP3) inflammasome activation, decrease the production of proinflammatory mediators (TNF-α, IL-1β, and reactive oxygen species), and suppress the accumulation of inflammatory cells. Glibenclamide's anti-inflammation warrants further investigation.
Collapse
|
20
|
Isakova ZT, Talaibekova ET, Asambaeva DA, Kerimkulova AS, Lunegova OS, Aldashev AA. Association of the polymorphic marker Glu23Lys in the KCNJ11 gene with hypertension in Kyrgyz patients. TERAPEVT ARKH 2017; 89:14-17. [DOI: 10.17116/terarkh201789114-17] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Aim. To study the association of the polymorphic marker Glu23Lys in the KCNJ11 with the development of hypertension in Kyrgyz patients. Subjects and methods. This case-control study enrolled 214 unrelated ethnic Kyrgyzes, in which a study group included 152 hypertensive patients (82 men and 70 women) and a control group consisted of 109 apparently healthy individuals (61 men and 48 women). The examinees’ mean age was 55.2±10.1 years. Hypertension was verified when blood pressure (BP) was above 140/90 mm Hg. Polymerase chain reaction-restriction fragment length polymorphism analysis was used to identify the polymorphic marker Glu23Lys in the KCNJ11 gene. Results. In the hypertension and control groups, the prevalence of 3 genotypes (Glu23Glu, Glu23Lys, and Lys23Lys) of the Glu23Lys polymorphism in the KCNJ11 gene differed significantly (χ2=8.04; p=0.018). The Lys23Lys and Glu23Lys genotypes were statistically more frequently recorded in the hypertension group and the homozygous Glu23Glu genotype was, on the contrary, more common in the control group than in the study one. In the hypertension group, the 23Lys allele frequency was statistically significantly higher than that in the control one (χ2=7.36; p=0.0067). The carriage of the 23Lys allele increased the risk of hypertension by 1.68 times (odds ratio (OR), 1.68; 95% confidence interval (CI), 1.17—2.41), that of the Glu23 allele had, on the contrary, a protective effect (OR, 0.60; 95% CI, 0.41—0.86). Conclusion. The polymorphic marker Glu23Lys in the KCNJ11 gene is associated with hypertension in the Kyrgyzes. The 23Lys allele is a marker for the higher risk of hypertension.
Collapse
|
21
|
Goharbari MH, Shadboores A, Abdollahi M. Inhibitory Effects of Thyroid Hormones on Mitochondrial
Oxidative Stress: A Systematic Review. INT J PHARMACOL 2016. [DOI: 10.3923/ijp.2016.249.261] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
|
22
|
Li SS, Cui N, Yang Y, Trower TC, Wei YM, Wu Y, Zhang S, Jin X, Jiang C. Impairment of the Vascular KATP Channel Imposes Fatal Susceptibility to Experimental Diabetes Due to Multi-Organ Injuries. J Cell Physiol 2015; 230:2915-26. [PMID: 25825210 DOI: 10.1002/jcp.25003] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2014] [Accepted: 03/25/2015] [Indexed: 12/19/2022]
Abstract
The vascular isoform of ATP-sensitive K(+) (KATP ) channels regulates blood flow to all organs. The KATP channel is strongly inhibited by reactive oxygen and carbonyl species produced in diabetic tissue inflammation. To address how such channel inhibition impacts vascular regulation as well as tissue viability, we performed studies in experimental diabetic mice. Strikingly, we found that knockout of the Kcnj8 encoding Kir6.1 subunit (Kcnj8-KO) caused mice to be fatally susceptible to diabetes. Organ perfusion studies suggested that the lack of this vascular K(+) channel handicapped activity-dependent vasodilation, leading to hypoperfusion, tissue hypoxia, and multi-organ failure. Morphologically, Kcnj8-KO mice showed greater inflammatory cell infiltration, higher levels of expression of inflammation indicator proteins, more severe cell apoptosis, and worse tissue disruptions. These were observed in the kidney, liver, and heart under diabetic condition in parallel comparison to tissues from WT mice. Patch clamping and molecular studies showed that the KATP channel was S-glutathionylated in experimental diabetes contributing to the inhibition of channel activity as well as the reduced arterial responses to vasodilators. These results suggest that the vascular KATP channel is organ protective in diabetic condition, and since the channel is suppressed by diabetic oxidative stress, therapeutical interventions to the maintenance of functional KATP channels may help to lower or prevent diabetic organ dysfunction.
Collapse
Affiliation(s)
- Shan-Shan Li
- Department of Biology, Georgia State University, 50 Decatur Street, Atlanta, Georgia.,Harbin Medical University School of Pharmacy, Harbin, Heilongjiang, China
| | - Ningren Cui
- Department of Biology, Georgia State University, 50 Decatur Street, Atlanta, Georgia
| | - Yang Yang
- Department of Biology, Georgia State University, 50 Decatur Street, Atlanta, Georgia
| | - Timothy C Trower
- Department of Biology, Georgia State University, 50 Decatur Street, Atlanta, Georgia
| | - Yu-Min Wei
- Department of Biology, Georgia State University, 50 Decatur Street, Atlanta, Georgia
| | - Yang Wu
- Department of Biology, Georgia State University, 50 Decatur Street, Atlanta, Georgia
| | - Shuang Zhang
- Department of Biology, Georgia State University, 50 Decatur Street, Atlanta, Georgia
| | - Xin Jin
- Department of Biology, Georgia State University, 50 Decatur Street, Atlanta, Georgia
| | - Chun Jiang
- Department of Biology, Georgia State University, 50 Decatur Street, Atlanta, Georgia
| |
Collapse
|
23
|
Yang KC, Kyle JW, Makielski JC, Dudley SC. Mechanisms of sudden cardiac death: oxidants and metabolism. Circ Res 2015; 116:1937-55. [PMID: 26044249 PMCID: PMC4458707 DOI: 10.1161/circresaha.116.304691] [Citation(s) in RCA: 94] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/19/2014] [Accepted: 02/09/2015] [Indexed: 02/07/2023]
Abstract
Ventricular arrhythmia is the leading cause of sudden cardiac death (SCD). Deranged cardiac metabolism and abnormal redox state during cardiac diseases foment arrhythmogenic substrates through direct or indirect modulation of cardiac ion channel/transporter function. This review presents current evidence on the mechanisms linking metabolic derangement and excessive oxidative stress to ion channel/transporter dysfunction that predisposes to ventricular arrhythmias and SCD. Because conventional antiarrhythmic agents aiming at ion channels have proven challenging to use, targeting arrhythmogenic metabolic changes and redox imbalance may provide novel therapeutics to treat or prevent life-threatening arrhythmias and SCD.
Collapse
Affiliation(s)
- Kai-Chien Yang
- From the Department of Pharmacology (K.-C.Y.) and Division of Cardiology, Department of Internal Medicine (K.-C.Y.), National Taiwan University Hospital, Taipei, Taiwan; Division of Cardiovascular Medicine, Department of Medicine, University of Wisconsin, Madison (J.W.K., J.C.M.); and Lifespan Cardiovascular Institute, the Providence VA Medical Center, and Brown University, RI (S.C.D.)
| | - John W Kyle
- From the Department of Pharmacology (K.-C.Y.) and Division of Cardiology, Department of Internal Medicine (K.-C.Y.), National Taiwan University Hospital, Taipei, Taiwan; Division of Cardiovascular Medicine, Department of Medicine, University of Wisconsin, Madison (J.W.K., J.C.M.); and Lifespan Cardiovascular Institute, the Providence VA Medical Center, and Brown University, RI (S.C.D.)
| | - Jonathan C Makielski
- From the Department of Pharmacology (K.-C.Y.) and Division of Cardiology, Department of Internal Medicine (K.-C.Y.), National Taiwan University Hospital, Taipei, Taiwan; Division of Cardiovascular Medicine, Department of Medicine, University of Wisconsin, Madison (J.W.K., J.C.M.); and Lifespan Cardiovascular Institute, the Providence VA Medical Center, and Brown University, RI (S.C.D.).
| | - Samuel C Dudley
- From the Department of Pharmacology (K.-C.Y.) and Division of Cardiology, Department of Internal Medicine (K.-C.Y.), National Taiwan University Hospital, Taipei, Taiwan; Division of Cardiovascular Medicine, Department of Medicine, University of Wisconsin, Madison (J.W.K., J.C.M.); and Lifespan Cardiovascular Institute, the Providence VA Medical Center, and Brown University, RI (S.C.D.).
| |
Collapse
|
24
|
Geiseler SJ, Ludvigsen S, Folkow LP. KATP-channels play a minor role in the protective hypoxic shut-down of cerebellar activity in eider ducks (Somateria mollissima). Neuroscience 2014; 284:751-758. [PMID: 25451290 DOI: 10.1016/j.neuroscience.2014.10.046] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2014] [Revised: 10/02/2014] [Accepted: 10/15/2014] [Indexed: 10/24/2022]
Abstract
Eider duck (Somateria mollissima) cerebellar neurons are highly tolerant toward hypoxia in vitro, which in part is due to a hypoxia-induced depression of their spontaneous activity. We have studied whether this response involves ATP-sensitive potassium (KATP) channels, which are known to be involved in the hypoxic/ischemic defense of mammalian neural and muscular tissues, by causing hyperpolarization and reduced ATP demand. Extracellular recordings in the Purkinje layer of isolated normoxic eider duck cerebellar slices showed that their spontaneous neuronal activity decreased significantly compared to in control slices when the KATP channel opener diazoxide (600 μM) was added (F1,70=92.781, p<0.001). Adding the KATP channel blocker tolbutamide (400 μM) 5 min prior to diazoxide completely abolished its effect (F1,55=39.639, p<0.001), strongly suggesting that these drugs have a similar mode of action in this avian species as in mammals. The spontaneous activity of slices treated with tolbutamide in combined hypoxia/chemical anoxia (95% N2-5% CO2 and 2 mM NaCN) was not significantly different from that of control slices (F1,203=0.071, p=0.791). Recovery from hypoxia/anoxia was, however, slightly but significantly weaker in tolbutamide-treated slices than in control slices (F1,137=15.539, p<0.001). We conclude that KATP channels are present in eider duck cerebellar neurons and are activated in hypoxia/anoxia, but that they do not play a key role in the protective shut-down response to hypoxia/anoxia.
Collapse
Affiliation(s)
- S J Geiseler
- Department of Arctic and Marine Biology, University of Tromsø - the Arctic University of Norway, Breivika, NO-9037 Tromsø, Norway.
| | - S Ludvigsen
- Department of Arctic and Marine Biology, University of Tromsø - the Arctic University of Norway, Breivika, NO-9037 Tromsø, Norway
| | - L P Folkow
- Department of Arctic and Marine Biology, University of Tromsø - the Arctic University of Norway, Breivika, NO-9037 Tromsø, Norway
| |
Collapse
|
25
|
Nnorom CC, Davis C, Fedinec AL, Howell K, Jaggar JH, Parfenova H, Pourcyrous M, Leffler CW. Contributions of KATP and KCa channels to cerebral arteriolar dilation to hypercapnia in neonatal brain. Physiol Rep 2014; 2:2/8/e12127. [PMID: 25168876 PMCID: PMC4246596 DOI: 10.14814/phy2.12127] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Mechanisms by which Pco2 controls cerebral vascular tone remain uncertain. We hypothesize that potassium channel activation contributes to the neonatal cerebrovascular dilation in response to increases in Paco2. To test this hypothesis, experiments were performed on newborn pigs with surgically implanted, closed cranial windows. Hypercapnia was induced by ventilation with elevated Pco2 gas in the absence and presence of the KATP channel inhibitor, glibenclamide and/or the KCa channel inhibitor, paxillin. Dilations to pinacidil, a selective KATP channel activator, without and with glibenclamide, were used to evaluate the efficacy of KATP channel inhibition. Dilations to NS1619, a selective KCa channel activator, without and with paxillin, were used to evaluate the efficacy of KCa channel inhibition. Cerebrovascular responses to the KATP and KCa channel activators, pinacidil and NS1619, respectively, cAMP‐dependent dilator, isoproterenol, and cGMP‐dependent dilator, sodium nitroprusside (SNP), were used to evaluate the selectivity of glibenclamide and paxillin. Glibenclamide blocked dilation to pinacidil, but did not inhibit dilations to NS1619, isoproterenol, or SNP. Glibenclamide prior to hypercapnia decreased mean pial arteriole dilation ~60%. Glibenclamide treatment during hypercapnia constricted arterioles ~35%. The level of hypercapnia, Paco2 between 50 and 75 mmHg, did not appear to be involved in efficacy of glibenclamide in blocking dilation to Paco2. Similarly to glibenclamide and KATP channel inhibition, paxillin blocked dilation to the KCa channel agonist, NS1619, and attenuated, but did not block, arteriolar dilation to hypercapnia. Treatment with both glibenclamide and paxillin abolished dilation to hypercapnia. Therefore, either glibenclamide or paxillin that block dilation to their channel agonists, pinacidil or NS1619, respectively, only partially inhibit dilation to hypercapnia. Block of both KATP and KCa channels completely prevent dilation hypercapnia. These data suggest hypercapnia activates both KATP and KCa channels leading to cerebral arteriolar dilation in newborn pigs. Mechanisms by which Pco2 controls vascular tone remain uncertain. We hypothesize KATP and KCa channel activation contributes to the neonatal cerebrovascular dilation in response to increases in Paco2. Presented data support this hypothesis.
Collapse
Affiliation(s)
- Chukwuma C Nnorom
- Departments of Physiology and Pediatrics, University of Tennessee Health Science Center, Memphis, Tennessee
| | - Corinne Davis
- Departments of Physiology and Pediatrics, University of Tennessee Health Science Center, Memphis, Tennessee
| | - Alexander L Fedinec
- Departments of Physiology and Pediatrics, University of Tennessee Health Science Center, Memphis, Tennessee
| | - Khadesia Howell
- Departments of Physiology and Pediatrics, University of Tennessee Health Science Center, Memphis, Tennessee
| | - Jonathan H Jaggar
- Departments of Physiology and Pediatrics, University of Tennessee Health Science Center, Memphis, Tennessee
| | - Helena Parfenova
- Departments of Physiology and Pediatrics, University of Tennessee Health Science Center, Memphis, Tennessee
| | - Massroor Pourcyrous
- Departments of Physiology and Pediatrics, University of Tennessee Health Science Center, Memphis, Tennessee
| | - Charles W Leffler
- Departments of Physiology and Pediatrics, University of Tennessee Health Science Center, Memphis, Tennessee
| |
Collapse
|
26
|
Yang KC, Bonini MG, Dudley SC. Mitochondria and arrhythmias. Free Radic Biol Med 2014; 71:351-361. [PMID: 24713422 PMCID: PMC4096785 DOI: 10.1016/j.freeradbiomed.2014.03.033] [Citation(s) in RCA: 106] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/31/2013] [Revised: 03/21/2014] [Accepted: 03/24/2014] [Indexed: 12/31/2022]
Abstract
Mitochondria are essential to providing ATP, thereby satisfying the energy demand of the incessant electrical activity and contractile action of cardiac muscle. Emerging evidence indicates that mitochondrial dysfunction can adversely affect cardiac electrical functioning by impairing the intracellular ion homeostasis and membrane excitability through reduced ATP production and excessive reactive oxygen species (ROS) generation, resulting in increased propensity to cardiac arrhythmias. In this review, the molecular mechanisms linking mitochondrial dysfunction to cardiac arrhythmias are discussed with an emphasis on the impact of increased mitochondrial ROS on the cardiac ion channels and transporters that are critical to maintaining normal electromechanical functioning of the cardiomyocytes. The potential of using mitochondria-targeted antioxidants as a novel antiarrhythmia therapy is highlighted.
Collapse
Affiliation(s)
- Kai-Chien Yang
- Lifespan Cardiovascular Institute, Providence VA Medical Center, and Brown University, Providence, RI 02903, USA
| | - Marcelo G Bonini
- Department of Medicine/Cardiology, University of Illinois at Chicago, Chicago, IL 60612, USA; Department of Pathology, and University of Illinois at Chicago, Chicago, IL 60612, USA; Department of Pharmacology, University of Illinois at Chicago, Chicago, IL 60612, USA
| | - Samuel C Dudley
- Lifespan Cardiovascular Institute, Providence VA Medical Center, and Brown University, Providence, RI 02903, USA.
| |
Collapse
|
27
|
Protić D, Beleslin-Čokić B, Spremović-Rađenović S, Radunović N, Heinle H, Šćepanović R, Gojković Bukarica L. The Different Effects of Resveratrol and Naringenin on Isolated Human Umbilical Vein: The Role of ATP-Sensitive K+
Channels. Phytother Res 2014; 28:1412-8. [DOI: 10.1002/ptr.5145] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2013] [Revised: 02/19/2014] [Accepted: 02/21/2014] [Indexed: 01/06/2023]
Affiliation(s)
- Dragana Protić
- Department of Pharmacology; Clinical Pharmacology and Toxicology; Faculty of Medicine; University of Belgrade; 11000 Belgrade Serbia
| | - Bojana Beleslin-Čokić
- Clinic for Endocrinology, Diabetes and Metabolic Diseases, Clinical Center of Serbia; Dr. Subotica 13 11000 Belgrade Serbia
| | - Svetlana Spremović-Rađenović
- Faculty of Medicine; University of Belgrade, Clinic for Gynecology and Obstetrics, Clinical Center of Serbia; 11000 Belgrade Serbia
| | - Nebojša Radunović
- Faculty of Medicine; University of Belgrade, Clinic for Gynecology and Obstetrics, Clinical Center of Serbia; 11000 Belgrade Serbia
| | - Helmut Heinle
- Institute of Physiology; University of Tüebingen; Germany
| | - Radisav Šćepanović
- Faculty of Medicine; University of Belgrade, Clinical Center Dr. Dragisa Misovic; 11000 Belgrade Serbia
| | - Ljiljana Gojković Bukarica
- Department of Pharmacology; Clinical Pharmacology and Toxicology; Faculty of Medicine; University of Belgrade; 11000 Belgrade Serbia
| |
Collapse
|
28
|
Shimizu S, Oikawa R, Tsounapi P, Inoue K, Shimizu T, Tanaka K, Martin DT, Honda M, Sejima T, Tomita S, Saito M. Blocking of the ATP sensitive potassium channel ameliorates the ischaemia-reperfusion injury in the rat testis. Andrology 2014; 2:458-65. [PMID: 24604784 DOI: 10.1111/j.2047-2927.2014.00199.x] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2013] [Revised: 01/21/2014] [Accepted: 02/01/2014] [Indexed: 12/17/2022]
Abstract
There is increasing evidence that the effects of administered ATP sensitive potassium (KATP ) channel openers or blockers during ischaemia are still controversial in many organs/tissues. Testicular torsion detorsion which causes ischaemia-reperfusion (IR) injury, cannot be predicted, thus an effective drug should be administered during or after the ischaemia. The aim of this study was to examine whether the administration of KATP channel openers or blockers during ischaemia ameliorates IR injury in the testis. Eight-week-old male Sprague-Dawley rats were subjected to 2 h right testicular ischaemia followed by 24 h reperfusion. The selective mitochondrial (mito) KATP channel blocker, 5-hydroxydecanoate (5-HD) (40 mg/kg), the non-selective KATP channel blocker glibenclamide (5 mg/kg), the selective mito KATP channel opener diazoxide (10 mg/kg) and the non-selective KATP channel opener cromakalim (300 μg/kg) were administered intraperitoneally 15 min prior to the ischaemia or 75 min after the induction of ischaemia. Tissue damage was evaluated by malondialdehyde concentration, myeloperoxidase activity, histological evaluation and TdT-mediated dUTP nick end labelling assay in the testis. There was a significant increase in oxidative stress, neutrophil infiltration, histological damage and apoptosis in the testicular IR model. A significant reduction in the testicular IR injury was observed with the administration of glibenclamide, but not 5-HD, diazoxide or cromakalim during ischaemia. The administration of non-selective KATP channel blocker glibenclamide ameliorated the testicular IR injury. On the other hand, the selective mito KATP channel blocker, 5-HD and KATP channel openers did not reduce the testicular IR injury. These data suggest that blocking of the membrane KATP channel may have a protective effect during the testicular ischaemia. Glibenclamide could be an effective drug to manage the post-ischaemic injury caused by the testicular torsion-detorsion.
Collapse
Affiliation(s)
- S Shimizu
- Division of Molecular Pharmacology, Tottori University School of Medicine, Yonago, Japan
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
29
|
Van Allen NR, Krafft PR, Leitzke AS, Applegate RL, Tang J, Zhang JH. The role of Volatile Anesthetics in Cardioprotection: a systematic review. Med Gas Res 2012; 2:22. [PMID: 22929111 PMCID: PMC3598931 DOI: 10.1186/2045-9912-2-22] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2012] [Accepted: 08/10/2012] [Indexed: 02/06/2023] Open
Abstract
This review evaluates the mechanism of volatile anesthetics as cardioprotective agents in both clinical and laboratory research and furthermore assesses possible cardiac side effects upon usage. Cardiac as well as non-cardiac surgery may evoke perioperative adverse events including: ischemia, diverse arrhythmias and reperfusion injury. As volatile anesthetics have cardiovascular effects that can lead to hypotension, clinicians may choose to administer alternative anesthetics to patients with coronary artery disease, particularly if the patient has severe preoperative ischemia or cardiovascular instability. Increasing preclinical evidence demonstrated that administration of inhaled anesthetics - before and during surgery - reduces the degree of ischemia and reperfusion injury to the heart. Recently, this preclinical data has been implemented clinically, and beneficial effects have been found in some studies of patients undergoing coronary artery bypass graft surgery. Administration of volatile anesthetic gases was protective for patients undergoing cardiac surgery through manipulation of the potassium ATP (KATP) channel, mitochondrial permeability transition pore (mPTP), reactive oxygen species (ROS) production, as well as through cytoprotective Akt and extracellular-signal kinases (ERK) pathways. However, as not all studies have demonstrated improved outcomes, the risks for undesirable hemodynamic effects must be weighed against the possible benefits of using volatile anesthetics as a means to provide cardiac protection in patients with coronary artery disease who are undergoing surgery.
Collapse
Affiliation(s)
- Nicole R Van Allen
- Department of Physiology, Loma Linda University School of Medicine, Risley Hall, Room 223, Loma Linda, CA 92354, USA.
| | | | | | | | | | | |
Collapse
|
30
|
Liu YH, Lu M, Hu LF, Wong PTH, Webb GD, Bian JS. Hydrogen sulfide in the mammalian cardiovascular system. Antioxid Redox Signal 2012; 17:141-85. [PMID: 22304473 DOI: 10.1089/ars.2011.4005] [Citation(s) in RCA: 205] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
For more than a century, hydrogen sulfide (H(2)S) has been regarded as a toxic gas. This review surveys the growing recognition of the role of H(2)S as an endogenous signaling molecule in mammals, with emphasis on its physiological and pathological pathways in the cardiovascular system. In biological fluids, H(2)S gas is a weak acid that exists as about 15% H(2)S, 85% HS(-), and a trace of S(2-). Here, we use "H(2)S" to refer to this mixture. H(2)S has been found to influence heart contractile functions and may serve as a cardioprotectant for treating ischemic heart diseases and heart failure. Alterations of the endogenous H(2)S level have been found in animal models with various pathological conditions such as myocardial ischemia, spontaneous hypertension, and hypoxic pulmonary hypertension. In the vascular system, H(2)S exerts biphasic regulation of a vascular tone with varying effects based on its concentration and in the presence of nitric oxide. Over the past decade, several H(2)S-releasing compounds (NaHS, Na(2)S, GYY4137, etc.) have been utilized to test the effect of exogenous H(2)S under different physiological and pathological situations in vivo and in vitro. H(2)S has been found to promote angiogenesis and to protect against atherosclerosis and hypertension, while excess H(2)S may promote inflammation in septic or hemorrhagic shock. H(2)S-releasing compounds and inhibitors of H(2)S synthesis hold promise in alleviating specific disease conditions. This comprehensive review covers in detail the effects of H(2)S on the cardiovascular system, especially in disease situations, and also the various underlying mechanisms.
Collapse
Affiliation(s)
- Yi-Hong Liu
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | | | | | | | | | | |
Collapse
|
31
|
Tsounapi P, Saito M, Dimitriadis F, Kitatani K, Kinoshita Y, Shomori K, Takenaka A, Satoh K. The role of KATP channels on ischemia-reperfusion injury in the rat testis. Life Sci 2012; 90:649-56. [PMID: 22480512 DOI: 10.1016/j.lfs.2012.03.006] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2011] [Revised: 02/16/2012] [Accepted: 02/28/2012] [Indexed: 12/30/2022]
|
32
|
Abstract
Hydrogen sulfide (H2S) is an endogenous gasotransmitter produced in mammalian cells. It is responsible for physiological functions in many organs and systems, with attention focused mainly on the cardiovascular and nervous systems. In the vascular system, H2S produces biphasic effects in regulation of vascular tone. At lower concentrations, it induces vasoconstriction predominantly via decreasing cyclic adenosine monophosphate in smooth muscle cell and inhibiting the production and bioavailability of nitric oxide. At higher concentrations, it produces vasorelaxation mainly through opening of KATP channels and induction of intracellular acidification. Scavenging reactive oxygen species and elevation of cyclic guanosine monophosphate are also implicated in the vasorelaxant response. This review presents an overview of the current knowledge of H2S in the vascular system, with special emphasis and discussion on the involvement of various signaling pathways and ion channels based on current understanding and reported literature till date.
Collapse
|
33
|
Barajas-Martínez H, Hu D, Ferrer T, Onetti CG, Wu Y, Burashnikov E, Boyle M, Surman T, Urrutia J, Veltmann C, Schimpf R, Borggrefe M, Wolpert C, Ibrahim BB, Sánchez-Chapula JA, Winters S, Haïssaguerre M, Antzelevitch C. Molecular genetic and functional association of Brugada and early repolarization syndromes with S422L missense mutation in KCNJ8. Heart Rhythm 2011; 9:548-55. [PMID: 22056721 DOI: 10.1016/j.hrthm.2011.10.035] [Citation(s) in RCA: 115] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/25/2011] [Accepted: 10/30/2011] [Indexed: 12/18/2022]
Abstract
BACKGROUND Adenosine triphosphate (ATP)-sensitive potassium cardiac channels consist of inward-rectifying channel subunits Kir6.1 or Kir6.2 (encoded by KCNJ8 or KCNJ11) and the sulfonylurea receptor subunits SUR2A (encoded by ABCC9). OBJECTIVE To examine the association of mutations in KCNJ8 with Brugada syndrome (BrS) and early repolarization syndrome (ERS) and to elucidate the mechanism underlying the gain of function of ATP-sensitive potassium channel current. METHODS Direct sequencing of KCNJ8 and other candidate genes was performed on 204 BrS and ERS probands and family members. Whole-cell and inside-out patch-clamp methods were used to study mutated channels expressed in TSA201 cells. RESULTS The same missense mutation, p.Ser422Leu (c.1265C>T) in KCNJ8, was identified in 3 BrS and 1 ERS probands but was absent in 430 alleles from ethnically matched healthy controls. Additional genetic variants included CACNB2b-D601E. Whole-cell patch-clamp studies showed a 2-fold gain of function of glibenclamide-sensitive ATP-sensitive potassium channel current when KCNJ8-S422L was coexpressed with SUR2A-wild type. Inside-out patch-clamp evaluation yielded a significantly greater half maximal inhibitory concentration for ATP in the mutant channels (785.5 ± 2 vs 38.4 ± 3 μM; n = 5; P <.01), pointing to incomplete closing of the ATP-sensitive potassium channels under normoxic conditions. Patients with a CACNB2b-D601E polymorphism displayed longer QT/corrected QT intervals, likely owing to their effect to induce an increase in L-type calcium channel current (I(Ca-L)). CONCLUSIONS Our results support the hypothesis that KCNJ8 is a susceptibility gene for BrS and ERS and point to S422L as a possible hotspot mutation. Our findings suggest that the S422L-induced gain of function in ATP-sensitive potassium channel current is due to reduced sensitivity to intracellular ATP.
Collapse
Affiliation(s)
- Hector Barajas-Martínez
- Molecular Genetics Department, Masonic Medical Research Laboratory, Utica, New York 13501, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Wu XF, Liu WT, Liu YP, Huang ZJ, Zhang YK, Song XJ. Reopening of ATP-sensitive potassium channels reduces neuropathic pain and regulates astroglial gap junctions in the rat spinal cord. Pain 2011; 152:2605-2615. [PMID: 21907492 DOI: 10.1016/j.pain.2011.08.003] [Citation(s) in RCA: 57] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2011] [Revised: 07/26/2011] [Accepted: 08/02/2011] [Indexed: 01/27/2023]
Abstract
Adenosine triphosphate-sensitive potassium (K(ATP)) channels are suggested to be involved in pathogenesis of neuropathic pain, but remain underinvestigated in primary afferents and in the spinal cord. We examined alterations of K(ATP) channels in rat spinal cord and tested whether and how they could contribute to neuropathic pain. The results showed that protein expression for K(ATP) channel subunits SUR1, SUR2, and Kir6.1, but not Kir6.2, were significantly downregulated and associated with thermal hyperalgesia and mechanical allodynia after sciatic nerve injury. Spinal administration of a K(ATP) channel opener cromakalim (CRO, 5, 10, and 20 μg, respectively) prevented or suppressed, in a dose-dependent manner, the hyperalgesia and allodynia. Nerve injury also significantly increased expression and phosphorylation of connexin 43, an astroglial gap junction protein. Such an increase of phosphorylation of connexin 43 was inhibited by CRO treatment. Furthermore, preadministration of an astroglial gap junction decoupler carbenoxolone (10 μg) completely reversed the inhibitory effects of CRO treatment on the hyperalgesia and allodynia and phosphorylation of NR1 and NR2B receptors and the subsequent activation of Ca(2+)-dependent signals Ca(2+)/calmodulin-dependent kinase II and cyclic adenosine monophosphate (cAMP) response element binding protein. These findings suggest that nerve injury-induced downregulation of the K(ATP) channels in the spinal cord may interrupt the astroglial gap junctional function and contribute to neuropathic pain, thus the K(ATP) channels opener can reduce neuropathic pain probably partly via regulating the astroglial gap junctions. This study may provide a new strategy for treating neuropathic pain using K(ATP) channel openers in the clinic.
Collapse
Affiliation(s)
- Xue-Feng Wu
- Department of Neurobiology, Parker University Research Institute, Dallas, TX, USA State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University, Nanjing, Jiangsu, China
| | | | | | | | | | | |
Collapse
|
35
|
Shimizu S, Saito M, Kinoshita Y, Ohmasa F, Dimitriadis F, Shomori K, Hayashi A, Satoh K. Nicorandil ameliorates ischaemia-reperfusion injury in the rat kidney. Br J Pharmacol 2011; 163:272-82. [PMID: 21250976 PMCID: PMC3087131 DOI: 10.1111/j.1476-5381.2011.01231.x] [Citation(s) in RCA: 59] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2010] [Revised: 09/21/2010] [Accepted: 10/28/2010] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND AND PURPOSE Nicorandil, an ATP-sensitive potassium (K(ATP) ) channel opener and nitric oxide donor, is used in the treatment of angina and acute heart failure. Here we investigated the effects of two K(ATP) channel openers, nicorandil and cromakalim on ischaemia reperfusion (I-R) injury in the kidney. EXPERIMENTAL APPROACH Right nephrectomy was performed in 8-week-old male Sprague-Dawley rats and they were then divided into six groups: control group; I-R, including 30 min of left renal ischaemia followed by 24 h of reperfusion; I-R groups plus nicorandil 3 or 10 mg·kg⁻¹ i.p.; and I-R groups plus cromakalim 100 or 300 µg·kg⁻¹ i.p. After reperfusion, renal function was estimated by serum creatinine (SCr), urinary albumin:creatinine ratio (ACR) and urinary β2-microglobulin (β2-MG). Levels of K(ATP) channel subtypes were investigated by Western blot. Kidney sections were stained for 4-hydroxy-2-nonenal and 8-hydroxy-2'-deoxyguanosine. KEY RESULTS Renal I-R induced significant increases in SCr, ACR and β2-MG levels compared with the control animals. Treatment with K(ATP) channel openers reduced urinary β2-MG levels, raised by I-R. Both K(IR) 6.1 and K(IR) 6.2 channels were expressed. Expression of K(IR) 6.2 channels in the I-R group was lower than in the control group, which was restored to normal by treatment with K(ATP) channel openers. Histologically, severe acute tubular damage was observed in the I-R kidney and this damage was ameliorated by K(ATP) channel openers, dose-dependently. CONCLUSIONS AND IMPLICATIONS ATP-sensitive potassium channel openers protected against proximal tubule damage after I-R injury. Nicorandil could represent a powerful additional component in the treatment of patients undergoing partial nephrectomy or renal transplantation.
Collapse
Affiliation(s)
- Shogo Shimizu
- Division of Molecular Pharmacology, Tottori University School of MedicineYonago, Japan
| | - Motoaki Saito
- Division of Molecular Pharmacology, Tottori University School of MedicineYonago, Japan
| | - Yukako Kinoshita
- Division of Molecular Pharmacology, Tottori University School of MedicineYonago, Japan
| | - Fumiya Ohmasa
- Division of Molecular Pharmacology, Tottori University School of MedicineYonago, Japan
| | - Fotios Dimitriadis
- Division of Molecular Pharmacology, Tottori University School of MedicineYonago, Japan
| | - Kohei Shomori
- Division of Organ Pathology, Tottori University School of MedicineYonago, Japan
| | - Atsushi Hayashi
- Department of Pediatrics, Yonago Medical CenterYonago, Japan
| | - Keisuke Satoh
- Division of Molecular Pharmacology, Tottori University School of MedicineYonago, Japan
| |
Collapse
|
36
|
Antzelevitch C, Barajas-Martinez H. A gain-of-function I(K-ATP) mutation and its role in sudden cardiac death associated with J-wave syndromes. Heart Rhythm 2010; 7:1472-4. [PMID: 20736095 PMCID: PMC2946491 DOI: 10.1016/j.hrthm.2010.07.027] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/23/2010] [Indexed: 02/07/2023]
|
37
|
Targeting Hypertension With a New Adenosine Triphosphate–sensitive Potassium Channel Opener Iptakalim. J Cardiovasc Pharmacol 2010; 56:215-28. [DOI: 10.1097/fjc.0b013e3181e23e2b] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
|
38
|
Ohmasa F, Saito M, Shimizu S, Taniguchi S, Dimitriadis F, Satoh I, Kinoshita Y, Satoh K. The role of ATP-sensitive potassium channel on acute urinary retention and subsequent catheterization in the rat. Eur J Pharmacol 2010; 635:194-7. [PMID: 20303940 DOI: 10.1016/j.ejphar.2010.02.052] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2009] [Revised: 02/08/2010] [Accepted: 02/24/2010] [Indexed: 10/19/2022]
|
39
|
Raeis V, Philip-Couderc P, Roatti A, Habre W, Sierra J, Kalangos A, Beghetti M, Baertschi AJ. Central Venous Hypoxemia Is a Determinant of Human Atrial ATP-Sensitive Potassium Channel Expression. Hypertension 2010; 55:1186-92. [DOI: 10.1161/hypertensionaha.109.148767] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Affiliation(s)
- Véronique Raeis
- From the Departments of Neuroscience (V.R., P.P.-C., A.R., A.J.B.), Anaesthesia (W.H.), Surgery (J.S., A.K.), and Child and Adolescent (M.B.), University of Geneva, Geneva, Switzerland
| | - Pierre Philip-Couderc
- From the Departments of Neuroscience (V.R., P.P.-C., A.R., A.J.B.), Anaesthesia (W.H.), Surgery (J.S., A.K.), and Child and Adolescent (M.B.), University of Geneva, Geneva, Switzerland
| | - Angela Roatti
- From the Departments of Neuroscience (V.R., P.P.-C., A.R., A.J.B.), Anaesthesia (W.H.), Surgery (J.S., A.K.), and Child and Adolescent (M.B.), University of Geneva, Geneva, Switzerland
| | - Walid Habre
- From the Departments of Neuroscience (V.R., P.P.-C., A.R., A.J.B.), Anaesthesia (W.H.), Surgery (J.S., A.K.), and Child and Adolescent (M.B.), University of Geneva, Geneva, Switzerland
| | - Jorge Sierra
- From the Departments of Neuroscience (V.R., P.P.-C., A.R., A.J.B.), Anaesthesia (W.H.), Surgery (J.S., A.K.), and Child and Adolescent (M.B.), University of Geneva, Geneva, Switzerland
| | - Afksendyios Kalangos
- From the Departments of Neuroscience (V.R., P.P.-C., A.R., A.J.B.), Anaesthesia (W.H.), Surgery (J.S., A.K.), and Child and Adolescent (M.B.), University of Geneva, Geneva, Switzerland
| | - Maurice Beghetti
- From the Departments of Neuroscience (V.R., P.P.-C., A.R., A.J.B.), Anaesthesia (W.H.), Surgery (J.S., A.K.), and Child and Adolescent (M.B.), University of Geneva, Geneva, Switzerland
| | - Alex J. Baertschi
- From the Departments of Neuroscience (V.R., P.P.-C., A.R., A.J.B.), Anaesthesia (W.H.), Surgery (J.S., A.K.), and Child and Adolescent (M.B.), University of Geneva, Geneva, Switzerland
| |
Collapse
|
40
|
Energetic myocardial metabolism and oxidative stress: let's make them our friends in the fight against heart failure. Biomed Pharmacother 2009; 64:203-7. [PMID: 19954925 DOI: 10.1016/j.biopha.2009.10.002] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2009] [Accepted: 10/19/2009] [Indexed: 01/31/2023] Open
Abstract
Heart failure (HF) is a syndrome causing a huge burden in morbidity and mortality worldwide. Current medical therapies for HF are aimed at suppressing the neurohormonal activation. However, novel therapies are needed for HF, independent of the neurohormonal axis, that can improve cardiac performance and prevent the progression of heart dysfunction. The modulation of cardiac metabolism may represent a new approach to the treatment of HF. The healthy heart converts chemical energy stored in fatty acids (FA) and glucose. Utilization of FA costs more oxygen per unit of ATP generated than glucose, and the heart gets 60-90% of its energy for oxidative phosphorylation from FA oxidation. The failing heart has been demonstrated to be metabolically abnormal, in both animal models and in patients, showing a shift toward an increased glucose uptake and utilization. The manipulation of myocardial substrate oxidation toward greater carbohydrate oxidation and less FA oxidation may improve ventricular performance and slow the progression of heart dysfunction. Impaired mitochondrial function and oxidative phosphorylation can reduce cardiac function by providing an insufficient supply of ATP to cardiomyocytes and by increasing myocardial oxidative stress. Although there are no effective stimulators of oxidative phosphorylation, several classes of drugs have been shown to open mitochondrial K(ATP) channels and, indirectly, to improve cardiac protection against oxidative stress. This article focuses on the energetic myocardial metabolism and oxidative status in the normal and failing heart, and briefly, it overviews the therapeutic potential strategies to improve cardiac energy and oxidative status in HF patients.
Collapse
|
41
|
The role of KATP channels on propofol preconditioning in a cellular model of renal ischemia-reperfusion. Anesth Analg 2009; 109:1486-92. [PMID: 19843786 DOI: 10.1213/ane.0b013e3181b76396] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
BACKGROUND Propofol (2,6-diisopropylphenol) has been shown to protect several organs, including the kidneys, from ischemia-reperfusion (I-R)-induced injury. Although propofol affects adenosine triphosphate-sensitive potassium (K(ATP)) channels in nonrenal tissues, it is still not clear by which mechanisms propofol protects renal cells from such damage. In this study, we investigated whether propofol induces renal preconditioning through renal K(ATP) channels. METHODS A reversible ATP depletion (antimycin A) followed by restoration of substrate supply in LLC-PK1 cells was used as an in vitro model of renal I-R. Cell viability was assessed by dimethylthiazol-diphenyltetrazol bromide and trypan blue dye exclusion test assays. Apoptosis was evaluated by annexin V-fluorescein isothiocyanate staining by flow cytometry and immunofluorescence. Propofol treatments were initiated at various time intervals: 1 or 24 h before ischemia, only during ischemia, or only during reperfusion. To evaluate the mechanisms of propofol protection, specific K(ATP) channel inhibitors or activators were used in some experiments during propofol pretreatment. RESULTS Propofol attenuated I-R injury on LLC-PK1 cells when present either 1 or 24 h before initiated I-R, and also during the recovery period, but not when added only during ischemia. Propofol pretreatment significantly protected LLC-PK1 from I-R-induced apoptosis. The protective effect of propofol was prevented by glibenclamide (a sarcolemmal ATP-dependent K(+) channel blocker) and decreased by 5-hydroxidecanoic acid (a mitochondrial ATP-dependent K(+) channel blocker), but it was not modified by diazoxide (a selective opener of ATP-sensitive K(+) channel). CONCLUSION Propofol protected cells against apoptosis induced by I-R. This protection was probably due to a preconditioning effect of propofol and was, at least in part, mediated by K(ATP) channels.
Collapse
|
42
|
Shears SB. Diphosphoinositol polyphosphates: metabolic messengers? Mol Pharmacol 2009; 76:236-52. [PMID: 19439500 PMCID: PMC2713120 DOI: 10.1124/mol.109.055897] [Citation(s) in RCA: 125] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2009] [Accepted: 05/12/2009] [Indexed: 12/18/2022] Open
Abstract
The diphosphoinositol polyphosphates ("inositol pyrophosphates") are a specialized subgroup of the inositol phosphate signaling family. This review proposes that many of the current data concerning the metabolic turnover and biological effects of the diphosphoinositol polyphosphates are linked by a common theme: these polyphosphates act as metabolic messengers. This review will also discuss the latest proposals concerning possible molecular mechanisms of action of this intriguing class of molecules.
Collapse
Affiliation(s)
- Stephen B Shears
- Inositide Signaling Group, National Institute of Environmental Health Sciences, NIH, DHHS, Research Triangle Park, NC 27709, USA.
| |
Collapse
|
43
|
Vasileiou I, Xanthos T, Koudouna E, Perrea D, Klonaris C, Katsargyris A, Papadimitriou L. Propofol: A review of its non-anaesthetic effects. Eur J Pharmacol 2009; 605:1-8. [DOI: 10.1016/j.ejphar.2009.01.007] [Citation(s) in RCA: 173] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
|
44
|
Activation of kappa-opioid receptor as a method for prevention of ischemic and reperfusion arrhythmias: role of protein kinase C and K(ATP) channels. Bull Exp Biol Med 2008; 143:187-90. [PMID: 17970197 DOI: 10.1007/s10517-007-0046-6] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
Intravenous pretreatment with kappa-opioid receptor antagonist (-)-U-50,488 (1 mg/kg) improved heart resistance to the arrhythmogenic effect of coronary occlusion and reperfusion. Selective kappa1-opioid receptor antagonist norbinaltorphimine and nonselective blocker of peripheral opioid receptors methylnaloxone abolished this antiarrhythmic effect. Preliminary blockade of protein kinase C with chelerythrine or inhibition of ATP-dependent K+ channels (K(ATP) channels) with glybenclamide abolished the antiarrhythmic effect of kappa-opioid receptor activation. Selective inhibitor of sarcolemmal K(ATP) channels did not modulate the kappa-opioid receptor-mediated increase in cardiac electrical stability. Our results suggest that protein kinase C and mitochondrial K(ATP) channels play an important role in the antiarrhythmic effect associated with activation of peripheral kappa-opioid receptors.
Collapse
|
45
|
Protective role of pinacidil against adrenaline-induced myocardium injury in guinea pig liver mitochondria. Open Life Sci 2007. [DOI: 10.2478/s11535-007-0039-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
AbstractWe investigated the role of the ATP-sensitive potassium channel opener pinacidil and blocker glibenclamide on guinea pig liver mitochondrial function, and a possible significance of pinacidil in the pharmacological treatment during myocardium dystrophy. First, a series of experiments was performed to determine the effect of pinacidil and glibenclamide on mitochondrial oxygen consumption. We found that pinacidil increased the rate of mitochondrial respiration for FAD-generated substrate (succinate oxidation), but was most effective for α-ketoglutarate oxidation with enhancement of respiratory control ratio. Oxidation of FAD-generated substrate inhibited efficiency of phosphorylation for α-ketoglutarate oxidation in pinacidil-treated animals. Glibenclamide decreased the rate of respiration with the lowest value of efficiency of phosphorylation, especially for α-ketoglutarate oxidation. A second series of experiments was performed to determine the effects of pinacidil and glibenclamide on oxidative phosphorylation during adrenaline-induced myocardium dystrophy. The increase in respiratory control ratio and efficiency of phosphorylation for α-ketoglutarate oxidation was greater than for succinate oxidation in mitochondria of pinacidil-pretreated animals during myocardium dystrophy. Inhibitory analysis with malonate suggested that endogenous succinate increased oxidation of NADH-generated substrates in mitochondria. Pinacidil is mainly involved in the adrenaline-induced alterations of mitochondrial function due to elevation of phosphorylation efficiency for α-ketoglutarate oxidation and a decreased level of lipid peroxidation.
Collapse
|
46
|
Effects of intracellular MgADP and acidification on the inhibition of cardiac sarcolemmal ATP-sensitive potassium channels by propofol. J Anesth 2007; 21:472-9. [PMID: 18008114 DOI: 10.1007/s00540-007-0551-9] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2006] [Accepted: 06/29/2007] [Indexed: 10/22/2022]
Abstract
PURPOSE Propofol inhibits adenosine triphosphate-sensitive potassium (K(ATP)) channels, which may result in the blocking of ischemic preconditioning in the heart. During cardiac ischemia, sarcolemmal K(ATP) channel activity is regulated by the increased levels of cytosolic metabolites, such as adenosine diphosphate (ADP) and protons. However, it remains unclear whether these cytosolic metabolites modulate the inhibitory action of propofol. The aim of this study was to investigate the effects of intracellular MgADP and acidification on K(ATP) channel inhibition by propofol. METHODS We used inside-out patch-clamp configurations to investigate the effects of propofol on the activities of recombinant cardiac sarcolemmal K(ATP) channels, which are reassociated by expressed subunits, sulfonylurea receptor (SUR) 2A, and inwardly rectifying potassium channels (Kir6.2). RESULTS In the absence of MgADP, propofol inhibited the SUR2A/Kir6.2 channel currents in a concentration-dependent manner, and an IC(50) of 78 microM. Increasing the intracellular MgADP concentrations to 0.1 and 0.3 mM markedly attenuated the inhibitory potency of propofol, and shifted the IC(50) to 183 and 265 microM, respectively. Moreover, decreasing the intracellular pH from 7.4 to 6.5 attenuated the inhibitory potency of propofol, and shifted the IC(50) to 277 microM. In addition, propofol-induced inhibition of truncated Kir6.2DeltaC36 currents, which form a functional channel without SUR2A, was not affected by an increase in intracellular MgADP. However, intracellular acidification (pH 6.5) significantly reduced the propofol sensitivity of Kir6.2DeltaC36 channels. CONCLUSION Our results demonstrated that the existence of intracellular MgADP and protons attenuated the direct inhibitory potency of propofol on recombinant cardiac sarcolemmal K(ATP) channels, via SUR2A and Kir6.2 subunits, respectively.
Collapse
|
47
|
Misaki N, Mao X, Lin YF, Suga S, Li GH, Liu Q, Chang Y, Wang H, Wakui M, Wu J. Iptakalim, a vascular ATP-sensitive potassium (KATP) channel opener, closes rat pancreatic beta-cell KATP channels and increases insulin release. J Pharmacol Exp Ther 2007; 322:871-8. [PMID: 17522344 DOI: 10.1124/jpet.107.121129] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Sulfonylureas have been the leading oral antihyperglycemic agents, and they presently continue to be the most popular antidiabetic drugs prescribed for treatment of type 2 diabetes. However, concern has arisen over the side effects of sulfonylureas on the cardiovascular system. Here, we tested the hypothesis that iptakalim, a novel vascular ATP-sensitive potassium (K(ATP)) channel opener, closes rat pancreatic beta-cell K(ATP) channels and increases insulin release. Rat pancreatic beta-cell K(ATP) channels and heterologously expressed K(ATP) channels in both human embryonic kidney (HEK) 293 cells and Xenopus oocytes were used to test the pharmacological effects of iptakalim. Patch-clamp recordings, Ca(2+) imaging, and measurements of insulin release were applied. Patch-clamp whole-cell recordings revealed that iptakalim depolarized beta-cells, induced action potential firing, and reduced K(ATP) channel-mediated currents. Single-channel recordings revealed that iptakalim reduced the open probability of K(ATP) channels without changing channel sensitivity to ATP. By closing beta-cell K(ATP) channels, iptakalim elevated intracellular Ca(2+) concentrations and increased insulin release. In addition, iptakalim decreased the open probability of recombinant Kir6.2FL4A (a trafficking mutant of the Kir6.2) K(ATP) channels heterologously expressed in HEK 293 cells, suggesting that iptakalim suppressed the function of beta-cell K(ATP) channels by directly inhibiting the Kir6.2 subunit. Finally, iptakalim inhibited Kir6.2/SUR1, but it activated Kir6.1/SUR2B (vascular-type), K(ATP) channels heterologously expressed in Xenopus oocytes. Iptakalim bidirectionally regulated pancreatic-type and vascular-type K(ATP) channels, and this unique pharmacological property suggests the potential use of iptakalim as a new therapeutic strategy for treating type 2 diabetes with the additional benefit of alleviating vascular disorders.
Collapse
Affiliation(s)
- Naoko Misaki
- Department of Physiology, Hirosaki University School of Medicine, Zaifucho, Japan
| | | | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Wan J, Xiong C, Zheng F, Zhou X, Huang C, Jiang H. Study of Kir6.2/KCNJ11 gene in a sudden cardiac death pedigree. Mol Biol Rep 2007; 35:119-23. [PMID: 17431820 DOI: 10.1007/s11033-007-9060-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2006] [Accepted: 01/17/2007] [Indexed: 11/28/2022]
Abstract
In clinic, the patients with acute myocardial infarction (AMI) are at high risk to develop ischemia-induced ventricular arrhythmias leading to sudden cardiac death (SCD). Some studies suggest that individual susceptibility to ischemia-induced arrhythmia may be related to the genes encoding ion channels. One of them is the cardiac ATP-sensitive potassium channel (K(ATP)), which is an octamer composed of four pore-forming inwardly rectifying potassium-channel subunits (Kir6.2) and four regulatory sulfonylurea-receptor subunits (SUR2A). They play important roles in the physiology and pathophysiology of cardiovascular system by coupling the metabolic state of the cells to cellular electrical activity. So far, some mutations and polymorphisms of Kir6.2/KCNJ11 gene showed significant correlation with type 2 diabetes. But it was not sure whether it was associated with acute myocardial diseases. Hence a complete mutational analysis of Kir6.2/KCNJ11 gene was performed in a pedigree of sudden cardiac death. The complete coding region and the intron-exon boundaries of KCNJ11 were amplified from genomic DNA using polymerase chain reaction (PCR). Direct sequencing was done to identify any mutations and then further confirmed by restriction site polymorphism (RSP) approach. No mutation was detected in the samples analyzed, a common polymorphism K23E (A>G) was noticed in this pedigree and the proband showed a homozygote genotype (G/G). The result suggests that the Kir6.2/KCNJ11 gene is not related to sudden cardiac death in this family.
Collapse
Affiliation(s)
- Jun Wan
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, 430064, China
| | | | | | | | | | | |
Collapse
|
49
|
Abstract
Sarcolemmal ATP-sensitive K(+) (K(ATP)) channels are abundant in cardiac myocytes where they couple the cellular metabolic state with membrane excitability. Structurally, these channels are composed of Kir6.2, a pore-forming subunit, SUR2A, a regulatory subunit, and at least four accessory proteins. The activation of K(ATP) channels occurs during ischaemia to promote cardiac viability under this adverse condition. Age-dependent changes in the myocardial susceptibility to ischaemia have been reported in experimental animals as well as in humans. Recent research has demonstrated that ageing is associated with a decrease in the number of cardiac sarcolemmal K(ATP) channels in hearts from females, but not males. This alteration is likely to be due to an age-dependent decrease in the concentration of circulating estrogens. In the heart, SUR2A is the least expressed protein of all K(ATP) channel-forming proteins. The consequence of this phenomenon is that the level of SUR2A is the main factor controlling the number of sarcolemmal K(ATP) channels. Estrogens specifically up-regulate SUR2A and govern the number of sarcolemmal K(ATP) channels, and this may explain the effect of decreasing estrogen levels on the heart. An age-dependent decrease in the number of sarcolemmal K(ATP) channels generates a cardiac phenotype more sensitive to ischaemia, which seems to be responsible for the ageing-associated decrease in myocardial tolerance to stress that occurs in elderly women.
Collapse
Affiliation(s)
- Aleksandar Jovanović
- Maternal and Child Health Sciences, Tayside Institute of Child Health, Ninewells Hospital and Medical School, University of Dundee, Dundee, UK.
| |
Collapse
|
50
|
Budas GR, Sukhodub A, Alessi DR, Jovanović A. 3'Phosphoinositide-dependent kinase-1 is essential for ischemic preconditioning of the myocardium. FASEB J 2006; 20:2556-8. [PMID: 17077284 PMCID: PMC2128707 DOI: 10.1096/fj.06-6252fje] [Citation(s) in RCA: 36] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
Brief periods of ischemia and reperfusion that precede sustained ischemia lead to a reduction in myocardial infarct size. This phenomenon, known as ischemic preconditioning, is mediated by signaling pathway(s) that are yet to be fully defined. 3'-Phosphoinositide-dependent kinase-1 (PDK1) has been implicated in numerous cellular processes. However, the involvement of PDK1 in preconditioning has yet to be elucidated. Studying PDK1 is not as straightforward as it is for the majority of kinases, due to the lack of a specific inhibitor of PDK1. Therefore, we have taken advantage of PDK1 hypomorphic mutant mice with reduced expression of PDK1 to study the role of PDK1 in preconditioning. Whole heart and single cell models of preconditioning demonstrated that the hearts and cardiac cells from PDK1 hypomorphic mice could not be preconditioned. The cardioprotective effect of PDK1 was not related to the effect that preconditioning has on sarcolemmal membrane action potential as revealed by di-8-ANEPPS, a sarcolemmal-potential sensitive dye, and laser confocal microscopy. In contrast, experiments with JC-1, a mitochondrial membrane potential-sensitive dye, has demonstrated that intact PDK1 levels were required for preconditioning-mediated regulation of mitochondrial membrane potential. Western blotting combined with functional experiments have shown that intact PDK1 levels were required for preconditioning-induced phosphorylation of protein kinase B (PKB), glycogen synthase kinase-3beta (GSK-3beta), and cardioprotection. We conclude that PDK1 mediates preconditioning in the heart by regulating activating PKB-GSK-3beta to regulate mitochondrial but not sarcolemmal membrane potential. 3'Phosphoinositide-dependent kinase-1 (PDK1) is essential for ischemic preconditioning of the myocardium.
Collapse
Affiliation(s)
- Grant R. Budas
- Maternal and Child Health Sciences, Ninewells Hospital and Medical School, University of Dundee, Dundee, Scotland, UK
| | - Andrey Sukhodub
- Maternal and Child Health Sciences, Ninewells Hospital and Medical School, University of Dundee, Dundee, Scotland, UK
| | - Dario R. Alessi
- MRC Protein Phosphorylation Unit, School of Life Sciences, University of Dundee, Dundee, Scotland, UK
| | - Aleksandar Jovanović
- Maternal and Child Health Sciences, Ninewells Hospital and Medical School, University of Dundee, Dundee, Scotland, UK
| |
Collapse
|