1
|
Specterman MJ, Aziz Q, Li Y, Anderson NA, Ojake L, Ng KE, Thomas AM, Finlay MC, Schilling RJ, Lambiase PD, Tinker A. Hypoxia Promotes Atrial Tachyarrhythmias via Opening of ATP-Sensitive Potassium Channels. Circ Arrhythm Electrophysiol 2023; 16:e011870. [PMID: 37646176 PMCID: PMC10510820 DOI: 10.1161/circep.123.011870] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/14/2022] [Accepted: 08/15/2023] [Indexed: 09/01/2023]
Abstract
BACKGROUND Hypoxia-ischemia predisposes to atrial arrhythmia. Atrial ATP-sensitive potassium channel (KATP) modulation during hypoxia has not been explored. We investigated the effects of hypoxia on atrial electrophysiology in mice with global deletion of KATP pore-forming subunits. METHODS Whole heart KATP RNA expression was probed. Whole-cell KATP current and action potentials were recorded in isolated wild-type (WT), Kir6.1 global knockout (6.1-gKO), and Kir6.2 global knockout (6.2-gKO) murine atrial myocytes. Langendorff-perfused hearts were assessed for atrial effective refractory period (ERP), conduction velocity, wavefront path length (WFPL), and arrhymogenicity under normoxia/hypoxia using a microelectrode array and programmed electrical stimulation. Heart histology was assessed. RESULTS Expression patterns were essentially identical for all KATP subunit RNA across human heart, whereas in mouse, Kir6.1 and SUR2 (sulphonylurea receptor subunit) were higher in ventricle than atrium, and Kir6.2 and SUR1 were higher in atrium. Compared with WT, 6.2-gKO atrial myocytes had reduced tolbutamide-sensitive current and action potentials were more depolarized with slower upstroke and reduced peak amplitude. Action potential duration was prolonged in 6.1-gKO atrial myocytes, absent of changes in other ion channel gene expression or atrial myocyte hypertrophy. In Langendorff-perfused hearts, baseline atrial ERP was prolonged and conduction velocity reduced in both KATP knockout mice compared with WT, without histological fibrosis. Compared with baseline, hypoxia led to conduction velocity slowing, stable ERP, and WFPL shortening in WT and 6.1-gKO hearts, whereas WFPL was stable in 6.2-gKO hearts due to ERP prolongation with conduction velocity slowing. Tolbutamide reversed hypoxia-induced WFPL shortening in WT and 6.1-gKO hearts through ERP prolongation. Atrial tachyarrhythmias inducible with programmed electrical stimulation during hypoxia in WT and 6.1-gKO mice correlated with WFPL shortening. Spontaneous arrhythmia was not seen. CONCLUSIONS KATP block/absence leads to cellular and tissue level atrial electrophysiological modification. Kir6.2 global knockout prevents hypoxia-induced atrial WFPL shortening and atrial arrhythmogenicity to programmed electrical stimulation. This mechanism could be explored translationally to treat ischemically driven atrial arrhythmia.
Collapse
Affiliation(s)
- Mark J. Specterman
- William Harvey Research Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, United Kingdom (M.J.S., Q.A., Y.L., N.A.A., L.O., K.-E.N., A.M.T., M.C.F., R.J.S., A.T.)
| | - Qadeer Aziz
- William Harvey Research Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, United Kingdom (M.J.S., Q.A., Y.L., N.A.A., L.O., K.-E.N., A.M.T., M.C.F., R.J.S., A.T.)
| | - Yiwen Li
- William Harvey Research Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, United Kingdom (M.J.S., Q.A., Y.L., N.A.A., L.O., K.-E.N., A.M.T., M.C.F., R.J.S., A.T.)
| | - Naomi A. Anderson
- William Harvey Research Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, United Kingdom (M.J.S., Q.A., Y.L., N.A.A., L.O., K.-E.N., A.M.T., M.C.F., R.J.S., A.T.)
| | - Leona Ojake
- William Harvey Research Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, United Kingdom (M.J.S., Q.A., Y.L., N.A.A., L.O., K.-E.N., A.M.T., M.C.F., R.J.S., A.T.)
| | - Keat-Eng Ng
- William Harvey Research Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, United Kingdom (M.J.S., Q.A., Y.L., N.A.A., L.O., K.-E.N., A.M.T., M.C.F., R.J.S., A.T.)
| | - Alison M. Thomas
- William Harvey Research Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, United Kingdom (M.J.S., Q.A., Y.L., N.A.A., L.O., K.-E.N., A.M.T., M.C.F., R.J.S., A.T.)
| | - Malcolm C. Finlay
- William Harvey Research Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, United Kingdom (M.J.S., Q.A., Y.L., N.A.A., L.O., K.-E.N., A.M.T., M.C.F., R.J.S., A.T.)
| | - Richard J. Schilling
- William Harvey Research Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, United Kingdom (M.J.S., Q.A., Y.L., N.A.A., L.O., K.-E.N., A.M.T., M.C.F., R.J.S., A.T.)
| | - Pier D. Lambiase
- Institute of Cardiovascular Science, University College London, United Kingdom (P.D.L.)
| | - Andrew Tinker
- William Harvey Research Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, United Kingdom (M.J.S., Q.A., Y.L., N.A.A., L.O., K.-E.N., A.M.T., M.C.F., R.J.S., A.T.)
| |
Collapse
|
2
|
Davis MJ, Kim HJ, Nichols CG. K ATP channels in lymphatic function. Am J Physiol Cell Physiol 2022; 323:C1018-C1035. [PMID: 35785984 PMCID: PMC9550566 DOI: 10.1152/ajpcell.00137.2022] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Revised: 06/22/2022] [Accepted: 06/23/2022] [Indexed: 11/22/2022]
Abstract
KATP channels function as negative regulators of active lymphatic pumping and lymph transport. This review summarizes and critiques the evidence for the expression of specific KATP channel subunits in lymphatic smooth muscle and endothelium, the roles that they play in normal lymphatic function, and their possible involvement in multiple diseases, including metabolic syndrome, lymphedema, and Cantú syndrome. For each of these topics, suggestions are made for directions for future research.
Collapse
Affiliation(s)
- Michael J Davis
- Department of Medical Pharmacology and Physiology, University of Missouri School of Medicine, Columbia, Missouri
| | - Hae Jin Kim
- Department of Medical Pharmacology and Physiology, University of Missouri School of Medicine, Columbia, Missouri
| | - Colin G Nichols
- Department of Cell Biology and Physiology, Washington University School of Medicine, St. Louis, Missouri
| |
Collapse
|
3
|
Garner BR, Stolarz AJ, Stuckey D, Sarimollaoglu M, Liu Y, Palade PT, Rusch NJ, Mu S. K ATP Channel Openers Inhibit Lymphatic Contractions and Lymph Flow as a Possible Mechanism of Peripheral Edema. J Pharmacol Exp Ther 2021; 376:40-50. [PMID: 33100270 PMCID: PMC7745085 DOI: 10.1124/jpet.120.000121] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2020] [Accepted: 10/08/2020] [Indexed: 11/22/2022] Open
Abstract
Pharmacological openers of ATP-sensitive potassium (KATP) channels are effective antihypertensive agents, but off-target effects, including severe peripheral edema, limit their clinical usefulness. It is presumed that the arterial dilation induced by KATP channel openers (KCOs) increases capillary pressure to promote filtration edema. However, KATP channels also are expressed by lymphatic muscle cells (LMCs), raising the possibility that KCOs also attenuate lymph flow to increase interstitial fluid. The present study explored the effect of KCOs on lymphatic contractile function and lymph flow. In isolated rat mesenteric lymph vessels (LVs), the prototypic KATP channel opener cromakalim (0.01-3 µmol/l) progressively inhibited rhythmic contractions and calculated intraluminal flow. Minoxidil sulfate and diazoxide (0.01-100 µmol/l) had similar effects at clinically relevant plasma concentrations. High-speed in vivo imaging of the rat mesenteric lymphatic circulation revealed that superfusion of LVs with cromakalim and minoxidil sulfate (0.01-10 µmol/l) maximally decreased lymph flow in vivo by 38.4% and 27.4%, respectively. Real-time polymerase chain reaction and flow cytometry identified the abundant KATP channel subunits in LMCs as the pore-forming Kir6.1/6.2 and regulatory sulfonylurea receptor 2 subunits. Patch-clamp studies detected cromakalim-elicited unitary K+ currents in cell-attached patches of LMCs with a single-channel conductance of 46.4 pS, which is a property consistent with Kir6.1/6.2 tetrameric channels. Addition of minoxidil sulfate and diazoxide elicited unitary currents of similar amplitude. Collectively, our findings indicate that KCOs attenuate lymph flow at clinically relevant plasma concentrations as a potential contributing mechanism to peripheral edema. SIGNIFICANCE STATEMENT: ATP-sensitive potassium (KATP) channel openers (KCOs) are potent antihypertensive medications, but off-target effects, including severe peripheral edema, limit their clinical use. Here, we demonstrate that KCOs impair the rhythmic contractions of lymph vessels and attenuate lymph flow, which may promote edema formation. Our finding that the KATP channels in lymphatic muscle cells may be unique from their counterparts in arterial muscle implies that designing arterial-selective KCOs may avoid activation of lymphatic KATP channels and peripheral edema.
Collapse
Affiliation(s)
- Brittney R Garner
- Department of Pharmacology and Toxicology (B.R.G., A.J.S., D.S., Y.L., P.T.P., N.J.R., S.M.) and Arkansas Nanomedicine Center (M.S.), College of Medicine and Department of Pharmaceutical Sciences, College of Pharmacy (A.J.S.), University of Arkansas for Medical Sciences, Little Rock, Arkansas
| | - Amanda J Stolarz
- Department of Pharmacology and Toxicology (B.R.G., A.J.S., D.S., Y.L., P.T.P., N.J.R., S.M.) and Arkansas Nanomedicine Center (M.S.), College of Medicine and Department of Pharmaceutical Sciences, College of Pharmacy (A.J.S.), University of Arkansas for Medical Sciences, Little Rock, Arkansas
| | - Daniel Stuckey
- Department of Pharmacology and Toxicology (B.R.G., A.J.S., D.S., Y.L., P.T.P., N.J.R., S.M.) and Arkansas Nanomedicine Center (M.S.), College of Medicine and Department of Pharmaceutical Sciences, College of Pharmacy (A.J.S.), University of Arkansas for Medical Sciences, Little Rock, Arkansas
| | - Mustafa Sarimollaoglu
- Department of Pharmacology and Toxicology (B.R.G., A.J.S., D.S., Y.L., P.T.P., N.J.R., S.M.) and Arkansas Nanomedicine Center (M.S.), College of Medicine and Department of Pharmaceutical Sciences, College of Pharmacy (A.J.S.), University of Arkansas for Medical Sciences, Little Rock, Arkansas
| | - Yunmeng Liu
- Department of Pharmacology and Toxicology (B.R.G., A.J.S., D.S., Y.L., P.T.P., N.J.R., S.M.) and Arkansas Nanomedicine Center (M.S.), College of Medicine and Department of Pharmaceutical Sciences, College of Pharmacy (A.J.S.), University of Arkansas for Medical Sciences, Little Rock, Arkansas
| | - Philip T Palade
- Department of Pharmacology and Toxicology (B.R.G., A.J.S., D.S., Y.L., P.T.P., N.J.R., S.M.) and Arkansas Nanomedicine Center (M.S.), College of Medicine and Department of Pharmaceutical Sciences, College of Pharmacy (A.J.S.), University of Arkansas for Medical Sciences, Little Rock, Arkansas
| | - Nancy J Rusch
- Department of Pharmacology and Toxicology (B.R.G., A.J.S., D.S., Y.L., P.T.P., N.J.R., S.M.) and Arkansas Nanomedicine Center (M.S.), College of Medicine and Department of Pharmaceutical Sciences, College of Pharmacy (A.J.S.), University of Arkansas for Medical Sciences, Little Rock, Arkansas
| | - Shengyu Mu
- Department of Pharmacology and Toxicology (B.R.G., A.J.S., D.S., Y.L., P.T.P., N.J.R., S.M.) and Arkansas Nanomedicine Center (M.S.), College of Medicine and Department of Pharmaceutical Sciences, College of Pharmacy (A.J.S.), University of Arkansas for Medical Sciences, Little Rock, Arkansas
| |
Collapse
|
4
|
Li Y, Aziz Q, Anderson N, Ojake L, Tinker A. Endothelial ATP-Sensitive Potassium Channel Protects Against the Development of Hypertension and Atherosclerosis. Hypertension 2020; 76:776-784. [PMID: 32654556 PMCID: PMC7418932 DOI: 10.1161/hypertensionaha.120.15355] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2020] [Revised: 05/18/2020] [Accepted: 06/19/2020] [Indexed: 12/28/2022]
Abstract
In the endothelium, ATP-sensitive potassium (KATP) channels are thought to couple cellular metabolism with membrane excitability, calcium entry, and endothelial mediator release. We hypothesized that endothelial KATP channels have a broad role protecting against high blood pressure and atherosclerosis. Endothelial-specific Kir6.1 KO mice (eKO) and eKO mice on an apolipoprotein E KO background were generated (A-eKO) to investigate the role of KATP channels in the endothelium. Basal blood pressure was not elevated in eKO mice. However, when challenged with a high-salt diet and the eNOS inhibitor L-NAME, eKO mice became more hypertensive than their littermate controls. In aorta, NO release at least partly contributes to the endothelium-dependent vasorelaxation induced by pinacidil. In A-eKO mice atherosclerotic plaque density was significantly greater than in their littermate controls when challenged with a high-fat diet, particularly in the aortic arch region. Levels of endothelial dysfunction markers were higher in eKO compared with WT mice; however, these were not significant for A-eKO mice compared with their littermate controls. Furthermore, decreased vascular reactivity was observed in the mesenteric arteries of A-eKO mice, but not in aorta when on a high-fat diet. Our data support a role for endothelial Kir6.1-containing KATP channels in the endothelial protection against environmental stressors: the maintenance of blood pressure homeostasis in response to high salt and endothelial integrity when challenged with a high-fat diet.
Collapse
Affiliation(s)
- Yiwen Li
- The Heart Centre, William Harvey Research Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, United Kingdom
| | - Qadeer Aziz
- The Heart Centre, William Harvey Research Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, United Kingdom
| | - Naomi Anderson
- The Heart Centre, William Harvey Research Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, United Kingdom
| | - Leona Ojake
- The Heart Centre, William Harvey Research Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, United Kingdom
| | - Andrew Tinker
- The Heart Centre, William Harvey Research Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, United Kingdom
| |
Collapse
|
5
|
Tinker A, Aziz Q, Li Y, Specterman M. ATP‐Sensitive Potassium Channels and Their Physiological and Pathophysiological Roles. Compr Physiol 2018; 8:1463-1511. [DOI: 10.1002/cphy.c170048] [Citation(s) in RCA: 72] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
|
6
|
Aziz Q, Finlay M, Montaigne D, Ojake L, Li Y, Anderson N, Ludwig A, Tinker A. ATP-sensitive potassium channels in the sinoatrial node contribute to heart rate control and adaptation to hypoxia. J Biol Chem 2018; 293:8912-8921. [PMID: 29666184 DOI: 10.1074/jbc.ra118.002775] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2018] [Revised: 04/16/2018] [Indexed: 11/06/2022] Open
Abstract
ATP-sensitive potassium channels (KATP) contribute to membrane currents in many tissues, are responsive to intracellular metabolism, and open as ATP falls and ADP rises. KATP channels are widely distributed in tissues and are prominently expressed in the heart. They have generally been observed in ventricular tissue, but they are also expressed in the atria and conduction tissues. In this study, we focused on the contribution and role of the inwardly rectifying KATP channel subunit, Kir6.1, in the sinoatrial node (SAN). To develop a murine, conduction-specific Kir6.1 KO model, we selectively deleted Kir6.1 in the conduction system in adult mice (cKO). Electrophysiological data in single SAN cells indicated that Kir6.1 underlies a KATP current in a significant proportion of cells and influences early repolarization during pacemaking, resulting in prolonged cycle length. Implanted telemetry probes to measure heart rate and electrocardiographic characteristics revealed that the cKO mice have a slow heart rate, with episodes of sinus arrest in some mice. The PR interval (time between the onset of the P wave to the beginning of QRS complex) was increased, suggesting effects on the atrioventricular node. Ex vivo studies of whole heart or dissected heart regions disclosed impaired adaptive responses of the SAN to hypoxia, and this may have had long-term pathological consequences in the cKO mice. In conclusion, Kir6.1-containing KATP channels in the SAN have a role in excitability, heart rate control, and the electrophysiological adaptation of the SAN to hypoxia.
Collapse
Affiliation(s)
- Qadeer Aziz
- From the Heart Centre, William Harvey Research Institute, Barts and the London School of Medicine and Dentistry, London, EC1M 6BQ, United Kingdom
| | - Malcolm Finlay
- From the Heart Centre, William Harvey Research Institute, Barts and the London School of Medicine and Dentistry, London, EC1M 6BQ, United Kingdom
| | - David Montaigne
- the Department of Clinical Physiology & Echocardiography, CHU Lille and the University of Lille, EGID, INSERM UMR1011, F-59000 Lille, France
| | - Leona Ojake
- From the Heart Centre, William Harvey Research Institute, Barts and the London School of Medicine and Dentistry, London, EC1M 6BQ, United Kingdom
| | - Yiwen Li
- From the Heart Centre, William Harvey Research Institute, Barts and the London School of Medicine and Dentistry, London, EC1M 6BQ, United Kingdom
| | - Naomi Anderson
- From the Heart Centre, William Harvey Research Institute, Barts and the London School of Medicine and Dentistry, London, EC1M 6BQ, United Kingdom
| | - Andreas Ludwig
- the Institut für Experimentelle und Klinische Pharmakologie und Toxikologie, Universität Erlangen-Nürnberg, 91054 Erlangen, Germany, and
| | - Andrew Tinker
- From the Heart Centre, William Harvey Research Institute, Barts and the London School of Medicine and Dentistry, London, EC1M 6BQ, United Kingdom,
| |
Collapse
|
7
|
Aziz Q, Li Y, Anderson N, Ojake L, Tsisanova E, Tinker A. Molecular and functional characterization of the endothelial ATP-sensitive potassium channel. J Biol Chem 2017; 292:17587-17597. [PMID: 28893911 DOI: 10.1074/jbc.m117.810325] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2017] [Revised: 09/07/2017] [Indexed: 01/29/2023] Open
Abstract
ATP-sensitive potassium (KATP) channels are widely expressed in the cardiovascular system, where they regulate a range of biological activities by linking cellular metabolism with membrane excitability. KATP channels in vascular smooth muscle have a well-defined role in regulating vascular tone. KATP channels are also thought to be expressed in vascular endothelial cells, but their presence and function in this context are less clear. As a result, we aimed to investigate the molecular composition and physiological role of endothelial KATP channels. We first generated mice with an endothelial specific deletion of the channel subunit Kir6.1 (eKO) using cre-loxP technology. Data from qRT-PCR, patch clamp, ex vivo coronary perfusion Langendorff heart experiments, and endothelial cell Ca2+ imaging comparing eKO and wild-type mice show that Kir6.1-containing KATP channels are indeed present in vascular endothelium. An increase in intracellular [Ca2+], which is central to changes in endothelial function such as mediator release, at least partly contributes to the endothelium-dependent vasorelaxation induced by the KATP channel opener pinacidil. The absence of Kir6.1 did not elevate basal coronary perfusion pressure in eKO mice. However, vasorelaxation was impaired during hypoxia in the coronary circulation, and this resulted in greater cardiac injury during ischemia-reperfusion. The response to adenosine receptor stimulation was impaired in eKO mice in single cells in patch clamp recordings and in the intact coronary circulation. Our data support the existence of an endothelial KATP channel that contains Kir6.1, is involved in vascular reactivity in the coronary circulation, and has a protective role in ischemia reperfusion.
Collapse
Affiliation(s)
- Qadeer Aziz
- From the Heart Centre, William Harvey Research Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, Charterhouse Square, London, EC1M 6BQ, United Kingdom
| | - Yiwen Li
- From the Heart Centre, William Harvey Research Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, Charterhouse Square, London, EC1M 6BQ, United Kingdom
| | - Naomi Anderson
- From the Heart Centre, William Harvey Research Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, Charterhouse Square, London, EC1M 6BQ, United Kingdom
| | - Leona Ojake
- From the Heart Centre, William Harvey Research Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, Charterhouse Square, London, EC1M 6BQ, United Kingdom
| | - Elena Tsisanova
- From the Heart Centre, William Harvey Research Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, Charterhouse Square, London, EC1M 6BQ, United Kingdom
| | - Andrew Tinker
- From the Heart Centre, William Harvey Research Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, Charterhouse Square, London, EC1M 6BQ, United Kingdom
| |
Collapse
|
8
|
Aziz Q, Thomas AM, Gomes J, Ang R, Sones WR, Li Y, Ng KE, Gee L, Tinker A. The ATP-Sensitive Potassium Channel Subunit, Kir6.1, in Vascular Smooth Muscle Plays a Major Role in Blood Pressure Control. Hypertension 2014; 64:523-9. [DOI: 10.1161/hypertensionaha.114.03116] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Affiliation(s)
- Qadeer Aziz
- From The Heart Centre, William Harvey Research Institute, Barts and the London School of Medicine and Dentistry, London, United Kingdom (Q.A., A.M.T., Y.L., K.-E.N., L.G., A.T.); and Department of Medicine, University College London, London, United Kingdom (J.G., R.A., W.R.S., A.T.)
| | - Alison M. Thomas
- From The Heart Centre, William Harvey Research Institute, Barts and the London School of Medicine and Dentistry, London, United Kingdom (Q.A., A.M.T., Y.L., K.-E.N., L.G., A.T.); and Department of Medicine, University College London, London, United Kingdom (J.G., R.A., W.R.S., A.T.)
| | - John Gomes
- From The Heart Centre, William Harvey Research Institute, Barts and the London School of Medicine and Dentistry, London, United Kingdom (Q.A., A.M.T., Y.L., K.-E.N., L.G., A.T.); and Department of Medicine, University College London, London, United Kingdom (J.G., R.A., W.R.S., A.T.)
| | - Richard Ang
- From The Heart Centre, William Harvey Research Institute, Barts and the London School of Medicine and Dentistry, London, United Kingdom (Q.A., A.M.T., Y.L., K.-E.N., L.G., A.T.); and Department of Medicine, University College London, London, United Kingdom (J.G., R.A., W.R.S., A.T.)
| | - William R. Sones
- From The Heart Centre, William Harvey Research Institute, Barts and the London School of Medicine and Dentistry, London, United Kingdom (Q.A., A.M.T., Y.L., K.-E.N., L.G., A.T.); and Department of Medicine, University College London, London, United Kingdom (J.G., R.A., W.R.S., A.T.)
| | - Yiwen Li
- From The Heart Centre, William Harvey Research Institute, Barts and the London School of Medicine and Dentistry, London, United Kingdom (Q.A., A.M.T., Y.L., K.-E.N., L.G., A.T.); and Department of Medicine, University College London, London, United Kingdom (J.G., R.A., W.R.S., A.T.)
| | - Keat-Eng Ng
- From The Heart Centre, William Harvey Research Institute, Barts and the London School of Medicine and Dentistry, London, United Kingdom (Q.A., A.M.T., Y.L., K.-E.N., L.G., A.T.); and Department of Medicine, University College London, London, United Kingdom (J.G., R.A., W.R.S., A.T.)
| | - Lorna Gee
- From The Heart Centre, William Harvey Research Institute, Barts and the London School of Medicine and Dentistry, London, United Kingdom (Q.A., A.M.T., Y.L., K.-E.N., L.G., A.T.); and Department of Medicine, University College London, London, United Kingdom (J.G., R.A., W.R.S., A.T.)
| | - Andrew Tinker
- From The Heart Centre, William Harvey Research Institute, Barts and the London School of Medicine and Dentistry, London, United Kingdom (Q.A., A.M.T., Y.L., K.-E.N., L.G., A.T.); and Department of Medicine, University College London, London, United Kingdom (J.G., R.A., W.R.S., A.T.)
| |
Collapse
|
9
|
Tinker A, Aziz Q, Thomas A. The role of ATP-sensitive potassium channels in cellular function and protection in the cardiovascular system. Br J Pharmacol 2014; 171:12-23. [PMID: 24102106 DOI: 10.1111/bph.12407] [Citation(s) in RCA: 83] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2013] [Revised: 07/30/2013] [Accepted: 08/26/2013] [Indexed: 12/14/2022] Open
Abstract
ATP-sensitive potassium channels (K(ATP)) are widely distributed and present in a number of tissues including muscle, pancreatic beta cells and the brain. Their activity is regulated by adenine nucleotides, characteristically being activated by falling ATP and rising ADP levels. Thus, they link cellular metabolism with membrane excitability. Recent studies using genetically modified mice and genomic studies in patients have implicated K(ATP) channels in a number of physiological and pathological processes. In this review, we focus on their role in cellular function and protection particularly in the cardiovascular system.
Collapse
Affiliation(s)
- Andrew Tinker
- William Harvey Heart Centre, Barts and The London School of Medicine and Dentistry, London, UK
| | | | | |
Collapse
|
10
|
Zayas-Santiago A, Agte S, Rivera Y, Benedikt J, Ulbricht E, Karl A, Dávila J, Savvinov A, Kucheryavykh Y, Inyushin M, Cubano LA, Pannicke T, Veh RW, Francke M, Verkhratsky A, Eaton MJ, Reichenbach A, Skatchkov SN. Unidirectional photoreceptor-to-Müller glia coupling and unique K+ channel expression in Caiman retina. PLoS One 2014; 9:e97155. [PMID: 24831221 PMCID: PMC4022631 DOI: 10.1371/journal.pone.0097155] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2014] [Accepted: 04/15/2014] [Indexed: 02/07/2023] Open
Abstract
Background Müller cells, the principal glial cells of the vertebrate retina, are fundamental for the maintenance and function of neuronal cells. In most vertebrates, including humans, Müller cells abundantly express Kir4.1 inwardly rectifying potassium channels responsible for hyperpolarized membrane potential and for various vital functions such as potassium buffering and glutamate clearance; inter-species differences in Kir4.1 expression were, however, observed. Localization and function of potassium channels in Müller cells from the retina of crocodiles remain, hitherto, unknown. Methods We studied retinae of the Spectacled caiman (Caiman crocodilus fuscus), endowed with both diurnal and nocturnal vision, by (i) immunohistochemistry, (ii) whole-cell voltage-clamp, and (iii) fluorescent dye tracing to investigate K+ channel distribution and glia-to-neuron communications. Results Immunohistochemistry revealed that caiman Müller cells, similarly to other vertebrates, express vimentin, GFAP, S100β, and glutamine synthetase. In contrast, Kir4.1 channel protein was not found in Müller cells but was localized in photoreceptor cells. Instead, 2P-domain TASK-1 channels were expressed in Müller cells. Electrophysiological properties of enzymatically dissociated Müller cells without photoreceptors and isolated Müller cells with adhering photoreceptors were significantly different. This suggests ion coupling between Müller cells and photoreceptors in the caiman retina. Sulforhodamine-B injected into cones permeated to adhering Müller cells thus revealing a uni-directional dye coupling. Conclusion Our data indicate that caiman Müller glial cells are unique among vertebrates studied so far by predominantly expressing TASK-1 rather than Kir4.1 K+ channels and by bi-directional ion and uni-directional dye coupling to photoreceptor cells. This coupling may play an important role in specific glia-neuron signaling pathways and in a new type of K+ buffering.
Collapse
Affiliation(s)
- Astrid Zayas-Santiago
- Departments of Pathology, Biochemistry and Physiology, Universidad Central Del Caribe, Bayamón, Puerto Rico, United States of America
| | - Silke Agte
- Paul Flechsig Institute of Brain Research, Faculty of Medicine, University of Leipzig, Leipzig, Germany
- Division of Soft Matter Physics, Department of Physics, University of Leipzig, Leipzig, Germany
| | - Yomarie Rivera
- Departments of Pathology, Biochemistry and Physiology, Universidad Central Del Caribe, Bayamón, Puerto Rico, United States of America
| | - Jan Benedikt
- Departments of Pathology, Biochemistry and Physiology, Universidad Central Del Caribe, Bayamón, Puerto Rico, United States of America
| | - Elke Ulbricht
- Paul Flechsig Institute of Brain Research, Faculty of Medicine, University of Leipzig, Leipzig, Germany
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, United Kingdom
| | - Anett Karl
- Paul Flechsig Institute of Brain Research, Faculty of Medicine, University of Leipzig, Leipzig, Germany
| | - José Dávila
- Departments of Pathology, Biochemistry and Physiology, Universidad Central Del Caribe, Bayamón, Puerto Rico, United States of America
| | - Alexey Savvinov
- Department of Physical Sciences, Universidad de Puerto Rico, Recinto de Río Piedras, Río Piedras, Puerto Rico, United States of America
| | - Yuriy Kucheryavykh
- Departments of Pathology, Biochemistry and Physiology, Universidad Central Del Caribe, Bayamón, Puerto Rico, United States of America
| | - Mikhail Inyushin
- Departments of Pathology, Biochemistry and Physiology, Universidad Central Del Caribe, Bayamón, Puerto Rico, United States of America
| | - Luis A. Cubano
- Departments of Pathology, Biochemistry and Physiology, Universidad Central Del Caribe, Bayamón, Puerto Rico, United States of America
| | - Thomas Pannicke
- Paul Flechsig Institute of Brain Research, Faculty of Medicine, University of Leipzig, Leipzig, Germany
| | | | - Mike Francke
- Paul Flechsig Institute of Brain Research, Faculty of Medicine, University of Leipzig, Leipzig, Germany
- Translational Centre for Regenerative Medicine (TRM) University of Leipzig, Leipzig, Germany
| | - Alexei Verkhratsky
- Faculty of Life Sciences, University of Manchester, Manchester, United Kingdom
| | - Misty J. Eaton
- Departments of Pathology, Biochemistry and Physiology, Universidad Central Del Caribe, Bayamón, Puerto Rico, United States of America
| | - Andreas Reichenbach
- Paul Flechsig Institute of Brain Research, Faculty of Medicine, University of Leipzig, Leipzig, Germany
| | - Serguei N. Skatchkov
- Departments of Pathology, Biochemistry and Physiology, Universidad Central Del Caribe, Bayamón, Puerto Rico, United States of America
- * E-mail:
| |
Collapse
|
11
|
Imaging energy status in live cells with a fluorescent biosensor of the intracellular ATP-to-ADP ratio. Nat Commun 2014; 4:2550. [PMID: 24096541 PMCID: PMC3852917 DOI: 10.1038/ncomms3550] [Citation(s) in RCA: 312] [Impact Index Per Article: 31.2] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2013] [Accepted: 09/04/2013] [Indexed: 02/07/2023] Open
Abstract
The ATP:ADP ratio is a critical parameter of cellular energy status that regulates many metabolic activities. Here we report an optimized genetically-encoded fluorescent biosensor, PercevalHR, that senses the ATP:ADP ratio. PercevalHR is tuned to the range of intracellular ATP:ADP expected in mammalian cells, and it can be used with one- or two-photon microscopy in live samples. We use PercevalHR to visualize activity-dependent changes in ATP:ADP when neurons are exposed to multiple stimuli, demonstrating that it is a sensitive reporter of physiological changes in energy consumption and production. We also use PercevalHR to visualize intracellular ATP:ADP while simultaneously recording currents from ATP-sensitive potassium (KATP) channels in single cells, showing that PercevalHR enables the study of coordinated variation in ATP:ADP and KATP channel open probability in intact cells. With its ability to monitor changes in cellular energetics within seconds, PercevalHR should be a versatile tool for metabolic research.
Collapse
|
12
|
Burley DS, Cox CD, Zhang J, Wann KT, Baxter GF. Natriuretic peptides modulate ATP-sensitive K(+) channels in rat ventricular cardiomyocytes. Basic Res Cardiol 2014; 109:402. [PMID: 24477916 PMCID: PMC3951884 DOI: 10.1007/s00395-014-0402-4] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/25/2013] [Revised: 12/10/2013] [Accepted: 01/10/2014] [Indexed: 11/28/2022]
Abstract
B-type natriuretic peptide (BNP) and C-type natriuretic peptide (CNP), and (Cys-18)-atrial natriuretic factor (4–23) amide (C-ANF), are cytoprotective under conditions of ischemia–reperfusion, limiting infarct size. ATP-sensitive K+ channel (KATP) opening is also cardioprotective, and although the KATP activation is implicated in the regulation of cardiac natriuretic peptide release, no studies have directly examined the effects of natriuretic peptides on cardiac KATP activity. Normoxic cardiomyocytes were patch clamped in the cell-attached configuration to examine sarcolemmal KATP (sKATP) activity. The KATP opener pinacidil (200 μM) increased the open probability of the patch (NPo; values normalized to control) at least twofold above basal value, and this effect was abolished by HMR1098 10 μM, a selective KATP blocker (5.23 ± 1.20 versus 0.89 ± 0.18; P < 0.001). We then examined the effects of BNP, CNP, C-ANF and 8Br-cGMP on the sKATP current. Bath application of BNP (≥10 nM) or CNP (≥0.01 nM) suppressed basal NPo (BNP: 1.00 versus 0.56 ± 0.09 at 10 nM, P < 0.001; CNP: 1.0 versus 0.45 ± 0.16, at 0.01 nM, P < 0.05) and also abolished the pinacidil-activated current at concentrations ≥10 nM. C-ANF (≥10 nM) enhanced KATP activity (1.00 versus 3.85 ± 1.13, at 100 nM, P < 0.05). The cGMP analog 8Br-cGMP 10 nM dampened the pinacidil-activated current (2.92 ± 0.60 versus 1.53 ± 0.32; P < 0.05). Natriuretic peptides modulate sKATP current in ventricular cardiomyocytes. This may be at least partially associated with their ability to augment intracellular cGMP concentrations via NPR-A/B, or their ability to bind NPR-C with high affinity. Although the mechanism of modulation requires elucidation, these preliminary data give new insights into the relationship between natriuretic peptide signaling and sKATP in the myocardium.
Collapse
Affiliation(s)
- Dwaine S Burley
- Cardiff School of Pharmacy and Pharmaceutical Sciences, Cardiff University, King Edward VII Avenue, Cardiff, CF10 3NB, UK,
| | | | | | | | | |
Collapse
|
13
|
Differential gene expression of cardiac ion channels in human dilated cardiomyopathy. PLoS One 2013; 8:e79792. [PMID: 24339868 PMCID: PMC3855055 DOI: 10.1371/journal.pone.0079792] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2013] [Accepted: 09/25/2013] [Indexed: 11/23/2022] Open
Abstract
Background Dilated cardiomyopathy (DCM) is characterized by idiopathic dilation and systolic contractile dysfunction of the cardiac chambers. The present work aimed to study the alterations in gene expression of ion channels involved in cardiomyocyte function. Methods and Results Microarray profiling using the Affymetrix Human Gene® 1.0 ST array was performed using 17 RNA samples, 12 from DCM patients undergoing cardiac transplantation and 5 control donors (CNT). The analysis focused on 7 cardiac ion channel genes, since this category has not been previously studied in human DCM. SCN2B was upregulated, while KCNJ5, KCNJ8, CLIC2, CLCN3, CACNB2, and CACNA1C were downregulated. The RT-qPCR (21 DCM and 8 CNT samples) validated the gene expression of SCN2B (p < 0.0001), KCNJ5 (p < 0.05), KCNJ8 (p < 0.05), CLIC2 (p < 0.05), and CACNB2 (p < 0.05). Furthermore, we performed an IPA analysis and we found a functional relationship between the different ion channels studied in this work. Conclusion This study shows a differential expression of ion channel genes involved in cardiac contraction in DCM that might partly underlie the changes in left ventricular function observed in these patients. These results could be the basis for new genetic therapeutic approaches.
Collapse
|
14
|
Abstract
ATP-sensitive potassium (KATP) channels were first discovered in the heart 30 years ago. Reconstitution of KATP channel activity by coexpression of members of the pore-forming inward rectifier gene family (Kir6.1, KCNJ8, and Kir6.2 KCNJ11) with sulfonylurea receptors (SUR1, ABCC8, and SUR2, ABCC9) of the ABCC protein subfamily has led to the elucidation of many details of channel gating and pore properties. In addition, the essential roles of Kir6.x and SURx subunits in generating cardiac and vascular KATP(2) and the detrimental consequences of genetic deletions or mutations in mice have been recognized. However, despite this extensive body of knowledge, there has been a paucity of defined roles of KATP subunits in human cardiovascular diseases, although there are reports of association of a single Kir6.1 variant with the J-wave syndrome in the ECG, and 2 isolated studies have reported association of loss of function mutations in SUR2 with atrial fibrillation and heart failure. Two new studies convincingly demonstrate that mutations in the SUR2 gene are associated with Cantu syndrome, a complex multi-organ disorder characterized by hypertrichosis, craniofacial dysmorphology, osteochondrodysplasia, patent ductus arteriosus, cardiomegaly, pericardial effusion, and lymphoedema. This realization of previously unconsidered consequences provides significant insight into the roles of the KATP channel in the cardiovascular system and suggests novel therapeutic possibilities.
Collapse
Affiliation(s)
- Colin G Nichols
- Center for the Investigation of Membrane Excitability Diseases and Department of Cell Biology and Physiology, Washington University School of Medicine, 660 South Euclid Avenue, St. Louis, MO 63110, USA.
| | | | | |
Collapse
|
15
|
Yang ZW, Chen JK, Ni M, Zhao T, Deng YP, Tao X, Jiang GJ, Shen FM. Role of Kir6.2 subunits of ATP-sensitive potassium channels in endotoxemia-induced cardiac dysfunction. Cardiovasc Diabetol 2013; 12:75. [PMID: 23659427 PMCID: PMC3654940 DOI: 10.1186/1475-2840-12-75] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/08/2013] [Accepted: 05/02/2013] [Indexed: 12/15/2022] Open
Abstract
Background Cardiac dysfunction is well-described in endotoxemia and diagnosed in up to 60% of patients with endotoxic shock. ATP-sensitive potassium (KATP) channels are critical to cardiac function. This study investigates the role of Kir6.2 subunits of KATP channels on cardiac dysfunction in lipopolysaccharide (LPS)-induced endotoxemia. Methods Kir6.2 subunits knockout (Kir6.2−/−) and wild-type (WT) mice were injected with LPS to induce endotoxemia. Cardiac function was monitored by echocardiography. Left ventricles were taken for microscopy (both light and electron) and TUNEL examination. Serum lactate dehydrogenase (LDH) and creatine kinase (CK) activities, and tumor necrosis factor-α (TNF-α) levels in both serum and left ventricular tissues were determined. Results Compared to WT, Kir6.2−/− mice showed significantly declined cardiac function 360 min after LPS administration, aggravated myocardial damage and elevated serum LDH and CK activities. Apoptotic cells were obviously increased in heart tissues from Kir6.2−/− mice at 90, 180 and 360 min. TNF-α expression in both serum and heart tissues of Kir6.2−/− mice was significantly increased. Conclusions We conclude that Kir6.2 subunits are critical in resistance to endotoxemia-induced cardiac dysfunction through reducing myocardial damage by inhibition of apoptosis and inflammation. KATP channels blockers are extensively used in the treatment of diabetes, their potential role should therefore be considered in the clinic when patients treated with antidiabetic sulfonylureas are complicated by endotoxemia.
Collapse
Affiliation(s)
- Zhong-Wei Yang
- Department of Pharmacology, School of Pharmacy, Second Military Medical University, Shanghai 200433, China
| | | | | | | | | | | | | | | |
Collapse
|
16
|
Cardiac KATP channel alterations associated with acclimation to hypoxia in goldfish (Carassius auratus L.). Comp Biochem Physiol A Mol Integr Physiol 2013; 164:554-64. [DOI: 10.1016/j.cbpa.2012.12.020] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2012] [Revised: 12/24/2012] [Accepted: 12/25/2012] [Indexed: 01/21/2023]
|
17
|
Fatima N, Schooley JF, Claycomb WC, Flagg TP. Promoter DNA methylation regulates murine SUR1 (Abcc8) and SUR2 (Abcc9) expression in HL-1 cardiomyocytes. PLoS One 2012; 7:e41533. [PMID: 22844491 PMCID: PMC3402388 DOI: 10.1371/journal.pone.0041533] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2011] [Accepted: 06/27/2012] [Indexed: 01/28/2023] Open
Abstract
Two mammalian genes encode the SURx (SUR1, Abcc8 and SUR2, Abcc9) subunits that combine with Kir6.2 (Kcnj11) subunits to form the ATP-sensitive potassium (KATP) channel in cardiac myocytes. Different isoform combinations endow the channel with distinct physiological and pharmacological properties, and we have recently reported that the molecular composition of sarcolemmal KATP channels is chamber specific in the mouse heart. KATP channel composition is determined by what subunits are expressed in a cell or tissue. In the present study, we explore the role of CpG methylation in regulating SUR1 and SUR2 expression. In HL-1 cardiomyocytes, as in atrial myocytes, SUR1 expression is markedly greater than SUR2. Consistent with CpG methylation-dependent silencing of SUR2 expression, bisulfite sequencing of genomic DNA isolated from HL-1 cells demonstrates that 57.6% of the CpGs in the promoter region of the SUR2 gene are methylated, compared with 0.14% of the the CpG residues in the SUR1 sequence. Moreover, treatment with 10 µM 5-aza-2'-deoxycytidine (Aza-dC) significantly increased both the unmethylated fraction of the SUR2 CpG island and mRNA expression. However, we cannot rule out additional mechanisms of Aza-dC action, as Aza-dC also causes a decrease in SUR1 expression and lower doses of Aza-dC do not alter the unmethylated DNA fraction but do elicit a small increase in SUR2 expression. The conclusion that DNA methylation alone is not the only regulator of SUR subunit expression is also consistent with observations in native myocytes, where the CpG islands of both SUR genes are essentially unmethylated in both atrial and ventricular myocytes. Collectively, these data demonstrate the potential for CpG methylation to regulate SURx subunit expression and raises the possibility that regulated or aberrant CpG methylation might play a role in controlling channel structure and function under different physiological conditions or different species.
Collapse
Affiliation(s)
- Naheed Fatima
- Department of Anatomy, Physiology and Genetics, Uniformed Services University of the Health Sciences, F. Edward Hebert School of Medicine, Bethesda, Maryland, United States of America
| | - James F. Schooley
- Department of Anatomy, Physiology and Genetics, Uniformed Services University of the Health Sciences, F. Edward Hebert School of Medicine, Bethesda, Maryland, United States of America
| | - Willliam C. Claycomb
- Department of Biochemistry and Molecular Biology, LSU Health Sciences Center, New Orleans, Louisiana, United States of America
| | - Thomas P. Flagg
- Department of Anatomy, Physiology and Genetics, Uniformed Services University of the Health Sciences, F. Edward Hebert School of Medicine, Bethesda, Maryland, United States of America
- * E-mail:
| |
Collapse
|
18
|
Nie L, Tang M, Zeng Y, Jiang H, Shi H, Luo H, Hu X, Gao L, Xi J, Liu A, Reppel M, Hescheler J, Liang H. Properties and functions of KATP during mouse perinatal development. Biochem Biophys Res Commun 2012; 418:74-80. [PMID: 22252295 DOI: 10.1016/j.bbrc.2011.12.133] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2011] [Accepted: 12/26/2011] [Indexed: 10/14/2022]
Abstract
BACKGROUND Prevailing data suggest that ATP-sensitive potassium channels (K(ATP)) contribute to a surprising resistance to hypoxia in mammalian embryos, thus we aimed to characterize the developmental changes of K(ATP) channels in murine fetal ventricular cardiomyocytes. METHODS Patch clamp was applied to investigate the functions of K(ATP). RT-PCR, Western blot were used to further characterize the molecular properties of K(ATP) channels. RESULTS Similar K(ATP) current density was detected in ventricular cardiomyocytes of late development stage (LDS) and early development stage (EDS). Molecular-biological study revealed the upregulation of Kir6.1/SUR2A in membrane and Kir6.2 remained constant during development. Kir6.1, Kir6.2, and SUR1 were detectable in the mitochondria without marked difference between EDS and LDS. Acute hypoxia-ischemia led to cessation of APs in 62.5% of tested EDS cells and no APs cessation was observed in LDS cells. SarcK(ATP) blocker glibenclamide rescued 47% of EDS cells but converted 42.8% of LDS cells to APs cessations under hypoxia-ischemic condition. MitoK(ATP) blocker 5-HD did not significantly influence the response to acute hypoxia-ischemia at either EDS or LDS. In summary, sarcK(ATP) played distinct functional roles under acute hypoxia-ischemic condition in EDS and LDS fetal ventricular cardiomyocytes, with developmental changes in sarcK(ATP) subunits. MitoK(ATP) were not significantly involved in the response of fetal cardiomyocytes to acute hypoxia-ischemia and no developmental changes of K(ATP) subunits were found in mitochondria.
Collapse
Affiliation(s)
- Li Nie
- Department of Physiology, Tongji Medical College, Huazhong University of Science and Technology, Hangkong Road 13, Wuhan 430030, China
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
19
|
Aziz Q, Thomas AM, Khambra T, Tinker A. Regulation of the ATP-sensitive potassium channel subunit, Kir6.2, by a Ca2+-dependent protein kinase C. J Biol Chem 2011; 287:6196-207. [PMID: 22207763 DOI: 10.1074/jbc.m111.243923] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The activity of ATP-sensitive potassium (K(ATP)) channels is governed by the concentration of intracellular ATP and ADP and is thus responsive to the metabolic status of the cell. Phosphorylation of K(ATP) channels by protein kinase A (PKA) or protein kinase C (PKC) results in the modulation of channel activity and is particularly important in regulating smooth muscle tone. At the molecular level the smooth muscle channel is composed of a sulfonylurea subunit (SUR2B) and a pore-forming subunit Kir6.1 and/or Kir6.2. Previously, Kir6.1/SUR2B channels have been shown to be inhibited by PKC, and Kir6.2/SUR2B channels have been shown to be activated or have no response to PKC. In this study we have examined the modulation of channel complexes formed of the inward rectifier subunit, Kir6.2, and the sulfonylurea subunit, SUR2B. Using a combination of biochemical and electrophysiological techniques we show that this complex can be inhibited by protein kinase C in a Ca(2+)-dependent manner and that this inhibition is likely to be as a result of internalization. We identify a residue in the distal C terminus of Kir6.2 (Ser-372) whose phosphorylation leads to down-regulation of the channel complex. This inhibitory effect is distinct from activation which is seen with low levels of channel activity.
Collapse
Affiliation(s)
- Qadeer Aziz
- William Harvey Heart Centre, Barts and the London School of Medicine and Dentistry, Charterhouse Square, London EC1M 6BQ, United Kingdom
| | | | | | | |
Collapse
|
20
|
Increased expression of adenosine triphosphate-sensitive K+ channels in mitral dysfunction: mechanically stimulated transcription and hypoxia-induced protein stability? J Am Coll Cardiol 2011; 59:390-6. [PMID: 22133355 DOI: 10.1016/j.jacc.2011.08.077] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/06/2011] [Revised: 07/26/2011] [Accepted: 08/17/2011] [Indexed: 01/08/2023]
Abstract
OBJECTIVES The aim of this study was to test whether adenosine triphosphate-sensitive K(+) (KATP) channel expression relates to mechanical and hypoxic stress within the left human heart. BACKGROUND The KATP channels play a vital role in preserving the metabolic integrity of the stressed heart. However, the mechanisms that govern the expression of their subunits (e.g., potassium inward rectifier [Kir] 6.2) in adult pathologies are mostly unknown. METHODS We collected biopsies from the 4 cardiac chambers and 50 clinical parameters from 30 surgical patients with severe mitral dysfunction. Proteins and messenger ribonucleic acids (mRNAs) of KATP pore subunits and mRNAs of their known transcriptional regulators (forkhead box [FOX] F2, FOXO1, FOXO3, and hypoxia inducible factor [HIF]-1α) were measured respectively by Western blotting, immunohistochemistry, and quantitative real-time polymerase chain reaction, and submitted to statistical analysis. RESULTS In all heart chambers, Kir6.2 mRNA correlated with HIF-1α mRNA. Neither Kir6.1 nor Kir6.2 proteins positively correlated with their respective mRNAs. The HIF-1α mRNA related in the left ventricle to aortic pressure, in the left atrium to left atrial pressure, and in all heart chambers to a decreased Kir6.2 protein/mRNA ratio. Interestingly, in the left heart, Kir6.2 protein and its immunohistochemical detection in myocytes were maximal at low venous PO(2). In the left ventricle, the Kir6.2 protein/mRNA ratio was also significantly higher at low venous PO(2), suggesting that tissue hypoxia might stabilize the Kir6.2 protein. CONCLUSIONS Results suggest that post-transcriptional events determine Kir6.2 protein expression in the left ventricle of patients with severe mitral dysfunction and low venous PO(2). Mechanical stress mainly affects transcription of HIF-1α and Kir6.2. This study implies that new therapies could aim at the proteasome for stabilizing the left ventricular Kir6.2 protein.
Collapse
|
21
|
Tester DJ, Tan BH, Medeiros-Domingo A, Song C, Makielski JC, Ackerman MJ. Loss-of-function mutations in the KCNJ8-encoded Kir6.1 K(ATP) channel and sudden infant death syndrome. ACTA ACUST UNITED AC 2011; 4:510-5. [PMID: 21836131 DOI: 10.1161/circgenetics.111.960195] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND Approximately 10% of sudden infant death syndrome (SIDS) may stem from cardiac channelopathies. The KCNJ8-encoded Kir6.1 (K(ATP)) channel critically regulates vascular tone and cardiac adaptive response to systemic metabolic stressors, including sepsis. KCNJ8-deficient mice are prone to premature sudden death, particularly with infection. We determined the spectrum, prevalence, and function of KCNJ8 mutations in a large SIDS cohort. METHODS AND RESULTS Using polymerase chain reaction, denaturing high-performance liquid chromatography, and DNA sequencing, comprehensive open reading frame/splice-site mutational analysis of KCNJ8 was performed on genomic DNA isolated from necropsy tissue on 292 unrelated SIDS cases (178 males, 204 white; age, 2.9±1.9 months). KCNJ8 mutations were coexpressed heterologously with SUR2A in COS-1 cells and characterized using whole-cell patch-clamp. Two novel KCNJ8 mutations were identified. A 5-month-old white male had an in-frame deletion (E332del) and a 2-month-old black female had a missense mutation (V346I). Both mutations localized to Kir6.1's C-terminus, involved conserved residues and were absent in 400 and 200 ethnic-matched reference alleles respectively. Both cases were negative for mutations in established channelopathic genes. Compared with WT, the pinacidil-activated K(ATP) current was decreased 45% to 68% for Kir6.1-E332del and 40% to 57% for V346I between -20 mV and 40 mV. CONCLUSIONS Molecular and functional evidence implicated loss-of-function KCNJ8 mutations as a novel pathogenic mechanism in SIDS, possibly by predisposition of a maladaptive cardiac response to systemic metabolic stressors akin to the mouse models of KCNJ8 deficiency.
Collapse
Affiliation(s)
- David J Tester
- Department of Medicine, Division of Cardiovascular Diseases, Windland Smith Rice Sudden Death Genomics Laboratory, Mayo Clinic, Rochester, MN 55905, USA
| | | | | | | | | | | |
Collapse
|
22
|
Leptihn S, Thompson JR, Ellory JC, Tucker SJ, Wallace MI. In vitro reconstitution of eukaryotic ion channels using droplet interface bilayers. J Am Chem Soc 2011; 133:9370-5. [PMID: 21591742 DOI: 10.1021/ja200128n] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
The ability to routinely study eukaryotic ion channels in a synthetic lipid environment would have a major impact on our understanding of how different lipids influence ion channel function. Here, we describe a straightforward, detergent-free method for the in vitro reconstitution of eukaryotic ion channels and ionotropic receptors into droplet interface bilayers and measure their electrical activity at both the macroscopic and single-channel level. We explore the general applicability of this method by reconstitution of channels from a wide range of sources including recombinant cell lines and native tissues, as well as preparations that are difficult to study by conventional methods including erythrocytes and mitochondria.
Collapse
Affiliation(s)
- Sebastian Leptihn
- Department of Chemistry, University of Oxford, 12 Mansfield Road, Oxford OX1 3TA, United Kingdom
| | | | | | | | | |
Collapse
|
23
|
Flagg TP, Enkvetchakul D, Koster JC, Nichols CG. Muscle KATP channels: recent insights to energy sensing and myoprotection. Physiol Rev 2010; 90:799-829. [PMID: 20664073 DOI: 10.1152/physrev.00027.2009] [Citation(s) in RCA: 208] [Impact Index Per Article: 14.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
ATP-sensitive potassium (K(ATP)) channels are present in the surface and internal membranes of cardiac, skeletal, and smooth muscle cells and provide a unique feedback between muscle cell metabolism and electrical activity. In so doing, they can play an important role in the control of contractility, particularly when cellular energetics are compromised, protecting the tissue against calcium overload and fiber damage, but the cost of this protection may be enhanced arrhythmic activity. Generated as complexes of Kir6.1 or Kir6.2 pore-forming subunits with regulatory sulfonylurea receptor subunits, SUR1 or SUR2, the differential assembly of K(ATP) channels in different tissues gives rise to tissue-specific physiological and pharmacological regulation, and hence to the tissue-specific pharmacological control of contractility. The last 10 years have provided insights into the regulation and role of muscle K(ATP) channels, in large part driven by studies of mice in which the protein determinants of channel activity have been deleted or modified. As yet, few human diseases have been correlated with altered muscle K(ATP) activity, but genetically modified animals give important insights to likely pathological roles of aberrant channel activity in different muscle types.
Collapse
Affiliation(s)
- Thomas P Flagg
- Department of Cell Biology and Physiology, Washington University School of Medicine, St. Louis, Missouri 63110, USA.
| | | | | | | |
Collapse
|
24
|
Ng KE, Schwarzer S, Duchen MR, Tinker A. The intracellular localization and function of the ATP-sensitive K+ channel subunit Kir6.1. J Membr Biol 2010; 234:137-47. [PMID: 20306027 DOI: 10.1007/s00232-010-9241-x] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2009] [Accepted: 03/04/2010] [Indexed: 10/19/2022]
Abstract
Our aim was to determine the subcellular localization and functional roles of the K(ATP) channel subunit Kir6.1 in intracellular membranes. Specifically, we focused on the potential role of Kir6.1 as a subunit of the mitochondrial ATP-sensitive K+ channel. Cell imaging showed that a major proportion of heterologously expressed Kir6.1-GFP and endogenously expressed Kir6.1 was distributed in the endoplasmic reticulum with little in the mitochondria or plasma membrane. We used pharmacological and molecular tools to investigate the functional significance of this distribution. The K(ATP) channel opener diazoxide increased reactive oxygen species production, and glibenclamide abolished this effect. However, in cells lacking Kir6.1 or expressing siRNA or dominant negative constructs of Kir6.1, the same effect was seen. Ca2+ handling was examined in the muscle cell line C2C12. Transfection of the dominant negative constructs of Kir6.1 significantly reduced the amplitude and rate of rise of [Ca2+]( c ) transients elicited by ATP. This study suggests that Kir6.1 is located in the endoplasmic reticulum and plays a role in modifying Ca2+ release from intracellular stores.
Collapse
Affiliation(s)
- Keat-Eng Ng
- Deparment of Medicine, The Rayne Institute, University College London, Room 107, University Street, London, WC1E 6JF, UK
| | | | | | | |
Collapse
|
25
|
Sukhodub A, Du Q, Jovanović S, Jovanović A. Nicotinamide-rich diet protects the heart against ischaemia-reperfusion in mice: a crucial role for cardiac SUR2A. Pharmacol Res 2010; 61:564-70. [PMID: 20083200 PMCID: PMC2859200 DOI: 10.1016/j.phrs.2010.01.008] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/22/2009] [Revised: 01/11/2010] [Accepted: 01/11/2010] [Indexed: 11/15/2022]
Abstract
It is a consensus view that a strategy to increase heart resistance to ischaemia-reperfusion is a warranted. Here, based on our previous study, we have hypothesized that a nicotinamide-rich diet could increase myocardial resistance to ischaemia-reperfusion. Therefore, the purpose of this study was to determine whether nicotinamide-rich diet would increase heart resistance to ischaemia-reperfusion and what is the underlying mechanism. Experiments have been done on mice on control and nicotinamide-rich diet (mice were a week on nicotinamide-rich diet) as well as on transgenic mice overexpressing SUR2A (SUR2A mice), a regulatory subunit of cardioprotective ATP-sensitive K(+) (K(ATP)) channels and their littermate controls (WT). The levels of mRNA in heart tissue were measured by real-time RT-PCR, whole heart and single cell resistance to ischaemia-reperfusion and severe hypoxia was measured by TTC staining and laser confocal microscopy, respectively. Nicotinamide-rich diet significantly decreased the size of myocardial infarction induced by ischaemia-reperfusion (from 42.5+/-4.6% of the area at risk zone in mice on control diet to 26.8+/-1.8% in mice on nicotinamide-rich diet, n=6-12, P=0.031). The cardioprotective effect of nicotinamide-rich diet was associated with 11.46+/-1.22 times (n=6) increased mRNA levels of SUR2A in the heart. HMR1098, a selective inhibitor of the sarcolemmal K(ATP) channels opening, abolished cardioprotection afforded by nicotinamide-rich diet. Transgenic mice with a sole increase in SUR2A expression had also increased cardiac resistance to ischaemia-reperfusion. We conclude that nicotinamide-rich diet up-regulate SUR2A and increases heart resistance to ischaemia-reperfusion.
Collapse
Affiliation(s)
- Andriy Sukhodub
- Division of Medical Sciences, Centre for Cardiovascular and Lung Biology, Ninewells Hospital & Medical School, University of Dundee, Dundee DD1 9SY, UK
| | | | | | | |
Collapse
|
26
|
Akrouh A, Halcomb SE, Nichols CG, Sala-Rabanal M. Molecular biology of K(ATP) channels and implications for health and disease. IUBMB Life 2009; 61:971-8. [PMID: 19787700 DOI: 10.1002/iub.246] [Citation(s) in RCA: 56] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
The ATP-sensitive potassium (K(ATP)) channel is expressed in most excitable tissues and plays a critical role in numerous physiological processes by coupling intracellular energetics to electrical activity. The channel is comprised of four Kir6.x subunits associated with four regulatory sulfonylurea receptors (SUR). Intracellular ATP acts on Kir6.x to inhibit channel activity, while MgADP stimulates channel activity through SUR. Changes in the cytosolic [ATP] to [ADP] ratio thus determine channel activity. Multiple mutations in Kir6.x and SUR genes have implicated K(ATP) channels in various diseases ranging from diabetes and hyperinsulinism to cardiac arrhythmias and cardiovascular disease. Continuing studies of channel physiology and pathology will bring new insights to the molecular basis of K(ATP) channel function, leading to a better understanding of the role that K(ATP) channels play in both health and disease.
Collapse
Affiliation(s)
- Alejandro Akrouh
- Department of Cell Biology and Physiology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | | | | | | |
Collapse
|
27
|
Isidoro Tavares N, Philip-Couderc P, Baertschi AJ, Lerch R, Montessuit C. Angiotensin II and tumour necrosis factor alpha as mediators of ATP-dependent potassium channel remodelling in post-infarction heart failure. Cardiovasc Res 2009; 83:726-36. [PMID: 19460779 DOI: 10.1093/cvr/cvp162] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
AIMS Angiotensin II (Ang II) and tumour necrosis factor alpha (TNFalpha) are involved in the progression from compensated hypertrophy to heart failure. Here, we test their role in the remodelling of ATP-dependent potassium channel (K(ATP)) in heart failure, conferring increased metabolic and diazoxide sensitivity. METHODS AND RESULTS We observed increased expression of both angiotensinogen and TNFalpha in the failing rat myocardium, with a regional gradient matching that of the K(ATP) subunit Kir6.1 expression. Both angiotensinogen and TNFalpha expression correlated positively with Kir6.1 and negatively with Kir6.2 expression across the post-infarction myocardium. To further identify a causal relationship, cardiomyocytes isolated from normal rat hearts were exposed in vitro to Ang II or TNFalpha. We observed increased Kir6.1 and SUR subunit and reduced Kir6.2 subunit mRNA expression in cardiomyocytes cultured with Ang II or TNFalpha, similar to what was observed in failing hearts. In patch-clamp experiments, cardiomyocytes cultured with Ang II or TNFalpha exhibited responsiveness to diazoxide, in terms of both K(ATP) current and action potential shortening. This was not observed in untreated cardiomyocytes and resembles the diazoxide sensitivity of failing cardiomyocytes that also overexpress Kir6.1. Ang II exerted its effect through induction of TNFalpha expression, because TNFalpha-neutralizing antibody abolished the effect of Ang II, and in failing hearts, regional expression of angiotensinogen matched TNFalpha expression. Finally, Ang II and TNFalpha regulated K(ATP) subunit expression, possibly through differential expression of Forkhead box transcription factors. CONCLUSION This study identifies Ang II and TNFalpha as mediators of the remodelling of K(ATP) channels in heart failure.
Collapse
|
28
|
Orie NN, Thomas AM, Perrino BA, Tinker A, Clapp LH. Ca2+/calcineurin regulation of cloned vascular K ATP channels: crosstalk with the protein kinase A pathway. Br J Pharmacol 2009; 157:554-64. [PMID: 19422382 DOI: 10.1111/j.1476-5381.2009.00221.x] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
Abstract
BACKGROUND AND PURPOSE Vascular ATP-sensitive potassium (K(ATP)) channels are activated by cyclic AMP elevating vasodilators through protein kinase A (PKA). Direct channel phosphorylation is a critical mechanism, though the phosphatase opposing these effects is unknown. Previously, we reported that calcineurin, a Ca(2+)-dependent phosphatase, inhibits K(ATP) channels, though neither the site nor the calcineurin isoform involved is established. Given that the type-2 regulatory (RII) subunit of PKA is a substrate for calcineurin we considered whether calcineurin regulates channel activity through interacting with PKA. EXPERIMENTAL APPROACH Whole-cell recordings were made in HEK-293 cells stably expressing the vascular K(ATP) channel (K(IR)6.1/SUR2B). The effect of intracellular Ca(2+) and modulators of the calcineurin and PKA pathway on glibenclamide-sensitive currents were examined. KEY RESULTS Constitutively active calcineurin A alpha but not A beta significantly attenuated K(ATP) currents activated by low intracellular Ca(2+), whereas calcineurin inhibitors had the opposite effect. PKA inhibitors reduced basal K(ATP) currents and responses to calcineurin inhibitors, consistent with the notion that some calcineurin action involves inhibition of PKA. However, raising intracellular Ca(2+) (equivalent to increasing calcineurin activity), almost completely inhibited K(ATP) channel activation induced by the catalytic subunit of PKA, whose enzymatic activity is independent of the RII subunit. In vitro phosphorylation experiments showed calcineurin could directly dephosphorylate a site in Kir6.1 that was previously phosphorylated by PKA. CONCLUSIONS AND IMPLICATIONS Calcineurin A alpha regulates K(IR)6.1/SUR2B by inhibiting PKA-dependent phosphorylation of the channel as well as PKA itself. Such a mechanism is likely to directly oppose the action of vasodilators on the K(ATP) channel.
Collapse
Affiliation(s)
- N N Orie
- BHF Laboratories, Department of Medicine, University College, London, UK
| | | | | | | | | |
Collapse
|
29
|
Jovanović A, Jovanović S. SURA2 targeting for cardioprotection? Curr Opin Pharmacol 2008; 9:189-93. [PMID: 19084477 DOI: 10.1016/j.coph.2008.11.003] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2008] [Accepted: 11/03/2008] [Indexed: 11/29/2022]
Abstract
SUR2A is an ATP-binding protein known to serve as a regulatory subunit of metabolic-sensing, cardioprotective sarcolemmal ATP-sensitive K(+) (K(ATP)) channels. It has been recently found that a moderate increase in expression of SUR2A protects the heart against different types of metabolic stresses, including ischaemia/reperfusion and hypoxia. Although the sarcolemmal K(ATP) channel is a multiprotein complex composed of many proteins in vivo, it seems that an increase in SUR2A levels is sufficient to increase the number of sarcolemmal K(ATP) channels. This effect of SUR2A could be due to SUR2A being the rate-limiting factor in generating fully composed sarcolemmal K(ATP) channels. An increased number of sarcolemmal K(ATP) channels seems to protect the heart by regulating action membrane potential, inhibiting Ca(2+) influx and preventing Ca(2+) overload, although an additional yet to be recognised mechanism independent of K(ATP) channels activity cannot be excluded.
Collapse
Affiliation(s)
- Aleksandar Jovanović
- Division of Medical Sciences, Ninewells Hospital & Medical School, University of Dundee, Dundee DD1 9SY, UK.
| | | |
Collapse
|
30
|
Teramoto N, Zhu HL, Shibata A, Aishima M, Walsh EJ, Nagao M, Cole WC. ATP-sensitive K+ channels in pig urethral smooth muscle cells are heteromultimers of Kir6.1 and Kir6.2. Am J Physiol Renal Physiol 2008; 296:F107-17. [PMID: 18945825 DOI: 10.1152/ajprenal.90440.2008] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
The inwardly rectifying properties and molecular basis of ATP-sensitive K(+) channels (K(ATP) channels) have now been established for several cell types. However, these aspects of nonvascular smooth muscle K(ATP) channels still remain to be defined. In this study, we investigated the molecular basis of the pore of K(ATP) channels of pig urethral smooth muscle cells through a comparative study of the inwardly rectifying properties, conductance, and regulation by PKC of native and homo- and heteroconcatemeric recombinant Kir6.x channels coexpressed with sulfonylurea receptor subunit SUR2B in human embryonic kidney (HEK) 293 cells by the patch-clamp technique (conventional whole-cell and cell-attached modes). In conventional whole-cell clamp recordings, levcromakalim (> or = 1 microM) caused a concentration-dependent increase in current that demonstrated strong inward rectification at positive membrane potentials. In cell-attached mode, the unitary amplitude of levcromakalim-induced native and recombinant heteroconcatemeric Kir6.1-Kir6.2 K(ATP) channels also showed strong inward rectification at positive membrane potentials. Phorbol 12,13-dibutyrate, but not the inactive phorbol ester, 4alpha-phorbol 12,13-didecanoate, enhanced the activity of native and heteroconcatemeric K(ATP) channels at -50 mV. The conductance of the native channels at approximately 43 pS was consistent with that of heteroconcatemeric channels with a pore-forming subunit composition of (Kir6.1)(3)-(Kir6.2). RT-PCR analysis revealed the expression of Kir6.1 and Kir6.2 transcripts in pig urethral myocytes. Our findings provide the first evidence that the predominant K(ATP) channel expressed in pig urethral smooth muscle possesses a unique, heteromeric pore structure that differs from the homomeric Kir6.1 channels of vascular myocytes and is responsible for the differences in inward rectification, conductance, and PKC regulation exhibited by the channels in these smooth muscle cell types.
Collapse
Affiliation(s)
- Noriyoshi Teramoto
- Dept. of Pharmacology, Graduate School of Medical Sciences, Kyushu Univ., 3-1-1 Maidashi, Higashi Ward, Fukuoka, 812-8582, Japan.
| | | | | | | | | | | | | |
Collapse
|
31
|
Wheeler A, Wang C, Yang K, Fang K, Davis K, Styer AM, Mirshahi U, Moreau C, Revilloud J, Vivaudou M, Liu S, Mirshahi T, Chan KW. Coassembly of different sulfonylurea receptor subtypes extends the phenotypic diversity of ATP-sensitive potassium (KATP) channels. Mol Pharmacol 2008; 74:1333-44. [PMID: 18723823 DOI: 10.1124/mol.108.048355] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
K(ATP) channels are metabolic sensors and targets of potassium channel openers (KCO; e.g., diazoxide and pinacidil). They comprise four sulfonylurea receptors (SUR) and four potassium channel subunits (Kir6) and are critical in regulating insulin secretion. Different SUR subtypes (SUR1, SUR2A, SUR2B) largely determine the metabolic sensitivities and the pharmacological profiles of K(ATP) channels. SUR1- but not SUR2-containing channels are highly sensitive to metabolic inhibition and diazoxide, whereas SUR2 channels are sensitive to pinacidil. It is generally believed that SUR1 and SUR2 are incompatible in channel coassembly. We used triple tandems, T1 and T2, each containing one SUR (SUR1 or SUR2A) and two Kir6.2Delta26 (last 26 residues are deleted) to examine the coassembly of different SUR. When T1 or T2 was expressed in Xenopus laevis oocytes, small whole-cell currents were activated by metabolic inhibition (induced by azide) plus a KCO (diazoxide for T1, pinacidil for T2). When coexpressed with any SUR subtype, the activated-currents were increased by 2- to 13-fold, indicating that different SUR can coassemble. Consistent with this, heteromeric SUR1+SUR2A channels were sensitive to azide, diazoxide, and pinacidil, and their single-channel burst duration was 2-fold longer than that of the T1 channels. Furthermore, SUR2A was coprecipitated with SUR1. Using whole-cell recording and immunostaining, heteromeric channels could also be detected when T1 and SUR2A were coexpressed in mammalian cells. Finally, the response of the SUR1+SUR2A channels to azide was found to be intermediate to those of the homomeric channels. Therefore, different SUR subtypes can coassemble into K(ATP) channels with distinct metabolic sensitivities and pharmacological profiles.
Collapse
Affiliation(s)
- Adam Wheeler
- Department of Physiology and Biophysics, Case Western Reserve University, School of Medicine, Cleveland, Ohio, USA
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Farzaneh T, Tinker A. Differences in the mechanism of metabolic regulation of ATP-sensitive K+ channels containing Kir6.1 and Kir6.2 subunits. Cardiovasc Res 2008; 79:621-31. [PMID: 18522960 DOI: 10.1093/cvr/cvn138] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
AIMS ATP sensitive K(+) channels (K(ATP)) sense adenine nucleotide concentrations and thus couple the metabolic state of the cell to membrane potential. The hetero-octameric complex of a sulphonylurea receptor (SUR2B) and an inwardly rectifying K(+) channel (Kir6.1) and the corresponding native channel in smooth muscle are relatively insensitive to variations in intracellular ATP. Activation of these channels in blood vessels during hypoxia/ischaemia is thought to be mediated via hormonal regulation such as cellular adenosine release or the release of mediators from the endothelium. In contrast, intracellular ATP prominently inhibits Kir6.2 containing complexes, such as those present in cardiac myocytes. Thus, we investigated differences in the mechanism of metabolic regulation of Kir6.1 and Kir6.2 containing K(ATP) channels. METHODS AND RESULTS We have heterologously expressed K(ATP) channel subunits in HEK293 and CHO cells and studied their function using (86)Rb efflux and patch clamping. We show that rodent Kir6.1/SUR2B has direct intrinsic metabolic sensitivity independent of any regulation by protein kinase A. In contrast to Kir6.2 containing complexes, this was not endowed by the ATP sensitivity of the pore forming subunit but was instead a property of the SUR2B subunit. Mutagenesis of key residues within the nucleotide-binding domains (NBD) implicated both domains in governing the metabolic sensitivity. CONCLUSION Kir6.1\SUR2B has intrinsic sensitivity to metabolism endowed by the likely processing of adenine nucleotides at the NBD of SUR2B.
Collapse
Affiliation(s)
- Tabasum Farzaneh
- BHF Laboratories, Department of Medicine, The Rayne Institute, University College London, Room 107, 5 University Street, London WC1E 6JJ, UK
| | | |
Collapse
|
33
|
Effects of Aβ1–42 on the Subunits of KATP Expression in Cultured Primary Rat Basal Forebrain Neurons. Neurochem Res 2008; 33:1419-24. [DOI: 10.1007/s11064-008-9603-2] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2007] [Accepted: 01/23/2008] [Indexed: 10/22/2022]
|
34
|
Chan KW, Wheeler A, Csanády L. Sulfonylurea receptors type 1 and 2A randomly assemble to form heteromeric KATP channels of mixed subunit composition. ACTA ACUST UNITED AC 2007; 131:43-58. [PMID: 18079561 PMCID: PMC2174157 DOI: 10.1085/jgp.200709894] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/03/2022]
Abstract
ATP-sensitive potassium (KATP) channels play important roles in regulating insulin secretion, controlling vascular tone, and protecting cells against metabolic stresses. KATP channels are heterooctamers of four pore-forming inwardly rectifying (Kir6.2) subunits and four sulfonylurea receptor (SUR) subunits. KATP channels containing SUR1 (e.g. pancreatic) and SUR2A (e.g. cardiac) display distinct metabolic sensitivities and pharmacological profiles. The reported expression of both SUR1 and SUR2 together with Kir6.2 in some cells raises the possibility that heteromeric channels containing both SUR subtypes might exist. To test whether SUR1 can coassemble with SUR2A to form functional KATP channels, we made tandem constructs by fusing SUR to either a wild-type (WT) or a mutant N160D Kir6.2 subunit. The latter mutation greatly increases the sensitivity of KATP channels to block by intracellular spermine. We expressed, individually and in combinations, tandem constructs SUR1-Kir6.2 (S1-WT), SUR1-Kir6.2[N160D] (S1-ND), and SUR2A-Kir6.2[N160D] (S2-ND) in Xenopus oocytes, and studied the voltage dependence of spermine block in inside-out macropatches over a range of spermine concentrations and RNA mixing ratios. Each tandem construct expressed alone supported macroscopic K+ currents with pharmacological properties indistinguishable from those of the respective native channel types. Spermine sensitivity was low for S1-WT but high for S1-ND and S2-ND. Coexpression of S1-WT and S1-ND generated current components with intermediate spermine sensitivities indicating the presence of channel populations containing both types of Kir subunits at all possible stoichiometries. The relative abundances of these populations, determined by global fitting over a range of conditions, followed binomial statistics, suggesting that WT and N160D Kir6.2 subunits coassemble indiscriminately. Coexpression of S1-WT with S2-ND also yielded current components with intermediate spermine sensitivities, suggesting that SUR1 and SUR2A randomly coassemble into functional KATP channels. Further pharmacological characterization confirmed coassembly of not only S1-WT and S2-ND, but also of coexpressed free SUR1, SUR2A, and Kir6.2 into functional heteromeric channels.
Collapse
Affiliation(s)
- Kim W Chan
- Department of Physiology and Biophysics, Case Western Reserve University, Cleveland, OH 44106, USA.
| | | | | |
Collapse
|
35
|
Jabr RI, Wilson AJ, Riddervold MH, Jenkins AH, Perrino BA, Clapp LH. Nuclear translocation of calcineurin Aβ but not calcineurin Aα by platelet-derived growth factor in rat aortic smooth muscle. Am J Physiol Cell Physiol 2007; 292:C2213-25. [PMID: 17303652 DOI: 10.1152/ajpcell.00139.2005] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
Calcineurin regulates the proliferation of many cell types through activation of the nuclear factor of activated T cells (NFAT). Two main isoforms of the calcineurin catalytic subunit [calcineurin A (CnA)α and CnAβ] have been identified, although their expression and function are largely unknown in smooth muscle. Western blot analysis and confocal imaging were performed in freshly isolated and cultured rat aortic myocytes to identify these CnA isoforms and elucidate the effect of PDGF on their cellular distribution and interaction with NFAT isoforms. CnAα and CnAβ isoforms displayed differential cellular distribution, with CnAα being evenly distributed between the nucleus and cytosol and CnAβ being restricted to the cytosol. In contrast with the rat brain, we found no evidence for particulate/membrane localization of calcineurin. PDGF caused significant nuclear translocation of CnAβ and induced smooth muscle cell proliferation, with both effects being abrogated by the calcineurin inhibitor cyclosporin A, the novel NFAT inhibitors A-285222 and inhibitor of NFAT-calcineurin association-6, and the adenylyl cyclase activator forskolin. PDGF also caused cyclosporin A-sensitive translocation of NFATc3, with no apparent effect on either CnAα or NFATc1 distribution. Moreover, ∼87% of nuclear CnAβ was found to colocalize with NFATc3, consistent with the finding that CnAβ bound more avidly than CnAα to a glutathione S-transferase-NFATc3 fusion protein. Based on their differential distribution in aortic muscle, our results suggest that CnAα and CnAβ are likely to have different cellular functions. However, CnAβ appears to be specifically activated by PDGF, and we postulate that calcineurin-dependent nuclear translocation of NFATc3 is involved in smooth muscle proliferation induced by this mitogen.
Collapse
Affiliation(s)
- Rita I Jabr
- BHF Laboratories, Rayne Bldg., Dept. of Medicine, University College London, London WC1E 6JF, UK
| | | | | | | | | | | |
Collapse
|
36
|
Isidoro Tavares N, Philip-Couderc P, Papageorgiou I, Baertschi AJ, Lerch R, Montessuit C. Expression and function of ATP-dependent potassium channels in late post-infarction remodeling. J Mol Cell Cardiol 2007; 42:1016-25. [PMID: 17512536 DOI: 10.1016/j.yjmcc.2007.04.008] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/07/2007] [Revised: 04/11/2007] [Accepted: 04/12/2007] [Indexed: 10/23/2022]
Abstract
Myocardial remodeling late after infarction is associated with increased incidence of fatal arrhythmias. Heterogeneous prolongation of the action potential in the surviving myocardium is one of the predominant causes. Sarcolemmal ATP-dependent potassium (K(ATP)) channels are important metabolic sensors regulating electrical activity of cardiomyocytes and are capable of considerably shortening the action potential. We determined whether ATP-dependent potassium channels generate or, on the contrary prevent the heterogeneity in action potential prolongation. Cardiomyocytes were obtained from the infarct border zone, the septum and the right ventricle of rat hearts 20 weeks after coronary occlusion when rats developed signs of heart failure. Expression of the conductance subunit Kir6.1, but not Kir6.2, and of all SUR regulatory subunits was increased up to 3-fold in cardiomyocytes from the infarct border zone. Concomitantly, there was a prominent response of the K(ATP) current to diazoxide that was not detectable in control cardiomyocytes. The action potential was prolonged in cardiomyocytes from the infarct border zone (74 ms) relative to sham (41 ms). However, activation of the K(ATP) channels by diazoxide reduced action potential duration to 42 ms. In myocytes of the septum and right ventricle, expression of channel subunits, duration of action potential, and sensitivity to diazoxide were only slightly increased relative to shams. In conclusion, the myocardium remodeled after infarction displays alterations of K(ATP) expression and function with spatial heterogeneity matching that of the action potential prolongation. Drugs selectively activating diazoxide-sensitive sarcolemmal K(ATP) channels should be considered in the prevention of arrhythmias in post-infarction heart failure.
Collapse
Affiliation(s)
- Nadia Isidoro Tavares
- Division of Cardiology, Geneva University Hospitals, 24 Micheli-du-Crest, 1211 Geneva 14, Switzerland
| | | | | | | | | | | |
Collapse
|
37
|
Tennant BP, Cui Y, Tinker A, Clapp LH. Functional expression of inward rectifier potassium channels in cultured human pulmonary smooth muscle cells: evidence for a major role of Kir2.4 subunits. J Membr Biol 2007; 213:19-29. [PMID: 17347781 PMCID: PMC1973150 DOI: 10.1007/s00232-006-0037-y] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2006] [Revised: 10/04/2006] [Indexed: 10/23/2022]
Abstract
Strong inwardly rectifying K(+) (K(IR)) channels that contribute to maintaining the resting membrane potential are encoded by the Kir2.0 family (Kir2.1-2.4). In smooth muscle, K(IR) currents reported so far have the characteristics of Kir2.1. However, Kir2.4, which exhibits unique characteristics of barium block, has been largely overlooked. Using patch-clamp techniques, we characterized K(IR) channels in cultured human pulmonary artery smooth muscle (HPASM) cells and compared them to cloned Kir2.1 and Kir2.4 channels. In a physiological K(+) gradient, inwardly rectifying currents were observed in HPASM cells, the magnitude and reversal potential of which were sensitive to extracellular K(+) concentration. Ba(2+) (100 microM ) significantly inhibited inward currents and depolarized HPASM cells by approximately 10 mV. In 60 mM extracellular K(+), Ba(2+) blocked K(IR) currents in HPASM cells with a 50% inhibitory concentration of 39.1 microM at -100 mV compared to 3.9 microM and 65.6 microM for Kir2.1 and Kir2.4, respectively. Cloned Kir2.4 and K(IR) currents in HPASM cells showed little voltage dependence to Ba(2+) inhibition, which blocked at a more superficial site than for Kir2.1. Single-channel recordings revealed strong inwardly rectifying channels with an average conductance of 21 pS in HPASM cells, not significantly different from either Kir2.1 (19.6 pS) or Kir2.4 (19.4 pS). Reverse-transcription polymerase chain reaction detected products corresponding to Kir2.1, Kir2.2 and Kir2.4 but not Kir2.3. We demonstrate that cultured HPASM cells express K(IR) channels and suggest both Kir2.1 and Kir2.4 subunits contribute to these channels, although the whole-cell current characteristics described share more similarity with Kir2.4.
Collapse
MESH Headings
- Animals
- Barium/pharmacology
- Base Sequence
- Biophysical Phenomena
- Biophysics
- Cells, Cultured
- DNA Primers/genetics
- Humans
- Membrane Potentials/drug effects
- Mice
- Muscle, Smooth, Vascular/cytology
- Muscle, Smooth, Vascular/metabolism
- Myocytes, Smooth Muscle/drug effects
- Myocytes, Smooth Muscle/metabolism
- Patch-Clamp Techniques
- Potassium Channels, Inwardly Rectifying/genetics
- Potassium Channels, Inwardly Rectifying/metabolism
- Pulmonary Artery/cytology
- Pulmonary Artery/metabolism
- RNA, Messenger/genetics
- RNA, Messenger/metabolism
- Rats
- Reverse Transcriptase Polymerase Chain Reaction
- Transfection
Collapse
Affiliation(s)
| | | | | | - Lucie H. Clapp
- Author for Correspondence, BHF Laboratories, Department of Medicine, 4 Floor Rayne Institute, University College, 5 University Street, London WC1E 6JJ, Tel 020 7679 6180, Fax 020 7679 6205, Email
| |
Collapse
|
38
|
Wu SN, Wu AZ, Sung RJ. Identification of two types of ATP-sensitive K+ channels in rat ventricular myocytes. Life Sci 2006; 80:378-87. [PMID: 17097686 DOI: 10.1016/j.lfs.2006.09.042] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2006] [Revised: 09/18/2006] [Accepted: 09/20/2006] [Indexed: 11/24/2022]
Abstract
The ATP-sensitive K(+) (K(ATP)) channels are known to provide a functional linkage between the electrical activity of the cell membrane and metabolism. Two types of inwardly rectifying K(+) channel subunits (i.e., Kir6.1 and Kir6.2) with which sulfonylurea receptors are associated were reported to constitute the K(ATP) channels. In this study, we provide evidence to show two types of K(ATP) channels with different biophysical properties functionally expressed in isolated rat ventricular myocytes. Using patch-clamp technique, we found that single-channel conductance for the different two types of K(ATP) channels in these cells was 57 and 21 pS. The kinetic properties, including mean open time and bursting kinetics, did not differ between these two types of K(ATP) channels. Diazoxide only activated the small-conductance K(ATP) channel, while pinacidil and dinitrophenol stimulated both channels. Both of these K(ATP) channels were sensitive to block by glibenclamide. Additionally, western blotting, immunochemistry, and RT-PCR revealed two types of Kir6.X channels, i.e., Kir6.1 and Kir6.2, in rat ventricular myocytes. Single-cell Ca(2+) imaging also revealed that similar to dinitrophenol, diazoxide reduced the concentration of intracellular Ca(2+). The present results suggest that these two types of K(ATP) channels may functionally be related to the activity of heart cells.
Collapse
MESH Headings
- Animals
- Blotting, Western
- Calcium/metabolism
- Diazoxide/pharmacology
- Dinitrophenols/pharmacology
- Dose-Response Relationship, Drug
- Fluorescent Antibody Technique, Direct
- Gene Expression/drug effects
- Glyburide/pharmacology
- Heart Ventricles/drug effects
- Heart Ventricles/metabolism
- Heart Ventricles/pathology
- KATP Channels
- Male
- Membrane Potentials/drug effects
- Myocytes, Cardiac/drug effects
- Myocytes, Cardiac/metabolism
- Myocytes, Cardiac/pathology
- Patch-Clamp Techniques
- Pinacidil/pharmacology
- Potassium Channels, Inwardly Rectifying/drug effects
- Potassium Channels, Inwardly Rectifying/genetics
- Potassium Channels, Inwardly Rectifying/metabolism
- RNA, Messenger/metabolism
- Rats
- Rats, Sprague-Dawley
- Reverse Transcriptase Polymerase Chain Reaction
Collapse
Affiliation(s)
- Sheng-Nan Wu
- Department of Physiology, National Cheng Kung University Medical College, Tainan, Taiwan.
| | | | | |
Collapse
|
39
|
Abstract
Potassium channels that are inhibited by intracellular ATP (ATP(i)) were first identified in ventricular myocytes, and are referred to as ATP-sensitive K+ channels (i.e. K(ATP) channels). Subsequently, K+ channels with similar characteristics have been demonstrated in many other tissues (pancreatic beta-cells, skeletal muscle, central neurones, smooth muscle). Approximately one decade ago, K(ATP) channels were cloned and were found to be composed of at least two subunits: an inwardly rectifying K+ channel six family (Kir6.x) that forms the ion conducting pore and a modulatory sulphonylurea receptor (SUR) that accounts for several pharmacological properties. Various types of native K(ATP) channels have been identified in a number of visceral and vascular smooth muscles in single-channel recordings. However, little attention has been paid to the molecular properties of the subunits in K(ATP) channels and it is important to determine the relative expression of K(ATP) channel components which give rise to native K(ATP) channels in smooth muscle. The aim of this review is to briefly discuss the current knowledge available for K(ATP) channels with the main interest in the molecular basis of native K(ATP) channels, and to discuss their possible linkage with physiological functions in smooth muscle.
Collapse
Affiliation(s)
- Noriyoshi Teramoto
- Department of Pharmacology, Graduate School of Medical Sciences, Kyushu University, Fukuoka 812-8582, Japan.
| |
Collapse
|
40
|
Acevedo JJ, Mendoza-Lujambio I, de la Vega-Beltrán JL, Treviño CL, Felix R, Darszon A. KATP channels in mouse spermatogenic cells and sperm, and their role in capacitation. Dev Biol 2005; 289:395-405. [PMID: 16343479 DOI: 10.1016/j.ydbio.2005.11.002] [Citation(s) in RCA: 48] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2005] [Revised: 11/01/2005] [Accepted: 11/04/2005] [Indexed: 11/21/2022]
Abstract
Mammalian sperm must undergo a series of physiological changes after leaving the testis to become competent for fertilization. These changes, collectively known as capacitation, occur in the female reproductive tract where the sperm plasma membrane is modified in terms of its components and ionic permeability. Among other events, mouse sperm capacitation leads to an increase in the intracellular Ca(2+) and pH as well as to a hyperpolarization of the membrane potential. It is well known that ion channels play a crucial role in these events, though the molecular identity of the particular channels involved in capacitation is poorly defined. In the present work, we report the identification and potential functional role of K(ATP) channels in mouse spermatogenic cells and sperm. By using whole-cell patch clamp recordings in mouse spermatogenic cells, we found K(+) inwardly rectifying (K(ir)) currents that are sensitive to Ba(2+), glucose and the sulfonylureas (tolbutamide and glibenclamide) that block K(ATP) channels. The presence of these channels was confirmed using inhibitors of the ATP synthesis and K(ATP) channel activators. Furthermore, RT-PCR assays allowed us to detect transcripts for the K(ATP) subunits SUR1, SUR2, K(ir)6.1 and K(ir)6.2 in total RNA from elongated spermatids. In addition, immunoconfocal microscopy revealed the presence of these K(ATP) subunits in mouse spermatogenic cells and sperm. Notably, incubation of sperm with tolbutamide during capacitation abolished hyperpolarization and significantly decreased the percentage of AR in a dose-dependent fashion. Together, our results provide evidence for the presence of K(ATP) channels in mouse spermatogenic cells and sperm and disclose the contribution of these channels to the capacitation-associated hyperpolarization.
Collapse
Affiliation(s)
- Juan José Acevedo
- Department of Developmental Genetics and Molecular Physiology, Institute of Biotechnology, UNAM, Cuernavaca, Mexico
| | | | | | | | | | | |
Collapse
|
41
|
Dhar-Chowdhury P, Harrell MD, Han SY, Jankowska D, Parachuru L, Morrissey A, Srivastava S, Liu W, Malester B, Yoshida H, Coetzee WA. The glycolytic enzymes, glyceraldehyde-3-phosphate dehydrogenase, triose-phosphate isomerase, and pyruvate kinase are components of the K(ATP) channel macromolecular complex and regulate its function. J Biol Chem 2005; 280:38464-70. [PMID: 16170200 PMCID: PMC4667781 DOI: 10.1074/jbc.m508744200] [Citation(s) in RCA: 98] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The regulation of ATP-sensitive potassium (K(ATP)) channel activity is complex and a multitude of factors determine their open probability. Physiologically and pathophysiologically, the most important of these are intracellular nucleotides, with a long-recognized role for glycolytically derived ATP in regulating channel activity. To identify novel regulatory subunits of the K(ATP) channel complex, we performed a two-hybrid protein-protein interaction screen, using as bait the mouse Kir6.2 C terminus. Screening a rat heart cDNA library, we identified two potential interacting proteins to be the glycolytic enzymes, glyceraldehyde-3-phosphate dehydrogenase (GAPDH) and triose-phosphate isomerase. The veracity of interaction was verified by co-immunoprecipitation techniques in transfected mammalian cells. We additionally demonstrated that pyruvate kinase also interacts with Kir6.2 subunits. The physiological relevance of these interactions is illustrated by the demonstration that native Kir6.2 protein similarly interact with GAPDH and pyruvate kinase in rat heart membrane fractions and that Kir6.2 protein co-localize with these glycolytic enzymes in rat ventricular myocytes. The functional relevance of our findings is demonstrated by the ability of GAPDH or pyruvate kinase substrates to directly block the K(ATP) channel under patch clamp recording conditions. Taken together, our data provide direct evidence for the concept that key enzymes involved in glycolytic ATP production are part of a multisubunit K(ATP) channel protein complex. Our data are consistent with the concept that the activity of these enzymes (possibly by ATP formation in the immediate intracellular microenvironment of this macromolecular K(ATP) channel complex) causes channel closure.
Collapse
Affiliation(s)
- Piyali Dhar-Chowdhury
- Department of Pediatrics, New York University School of Medicine, New York, New York 10016
| | - Maddison D. Harrell
- Department of Pediatrics, New York University School of Medicine, New York, New York 10016
| | - Sandra Y. Han
- Department of Pediatrics, New York University School of Medicine, New York, New York 10016
| | - Danuta Jankowska
- Department of Pediatrics, New York University School of Medicine, New York, New York 10016
| | - Lavanya Parachuru
- Department of Pediatrics, New York University School of Medicine, New York, New York 10016
| | - Alison Morrissey
- Department of Pediatrics, New York University School of Medicine, New York, New York 10016
| | - Shekhar Srivastava
- Department of Pediatrics, New York University School of Medicine, New York, New York 10016
| | - Weixia Liu
- Department of Pediatrics, New York University School of Medicine, New York, New York 10016
| | - Brian Malester
- Department of Pediatrics, New York University School of Medicine, New York, New York 10016
| | - Hidetada Yoshida
- Department of Pediatrics, New York University School of Medicine, New York, New York 10016
| | - William A. Coetzee
- Department of Pediatrics, New York University School of Medicine, New York, New York 10016
- Department of Physiology & Neuroscience, New York University School of Medicine, New York, New York 10016
- Department of Pharmacology, New York University School of Medicine, New York, New York 10016
| |
Collapse
|
42
|
Thomzig A, Laube G, Prüss H, Veh RW. Pore-forming subunits of K-ATP channels, Kir6.1 and Kir6.2, display prominent differences in regional and cellular distribution in the rat brain. J Comp Neurol 2005; 484:313-30. [PMID: 15739238 DOI: 10.1002/cne.20469] [Citation(s) in RCA: 102] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
K-ATP channels consist of two structurally different subunits: a pore-forming subunit of the Kir6.0-family (Kir6.1 or Kir6.2) and a sulfonylurea receptor (SUR1, SUR2, SUR2A, SUR2B) with regulatory activity. The functional diversity of K-ATP channels in brain is broad and of fundamental importance for neuronal activity. Here, using immunocytochemistry with monospecific antibodies against the Kir6.1 and Kir6.2 subunits, we analyze the regional and cellular distribution of both proteins in the adult rat brain. We find Kir6.2 to be widely expressed in all brain regions, suggesting that the Kir6.2 subunit forms the pore of the K-ATP channels in most neurons, presumably protecting the cells during cellular stress conditions such as hypoglycemia or ischemia. Especially in hypothalamic nuclei, in particular the ventromedial and arcuate nucleus, neurons display Kir6.2 immunoreactivity only, suggesting that Kir6.2 is the pore-forming subunit of the K-ATP channels in the glucose-responsive neurons of the hypothalamus. In contrast, Kir6.1-like immunolabeling is restricted to astrocytes (Thomzig et al. [2001] Mol Cell Neurosci 18:671-690) in most areas of the rat brain and very weak or absent in neurons. Only in distinct nuclei or neuronal subpopulations is a moderate or even strong Kir6.1 staining detected. The biological functions of these K-ATP channels still need to be elucidated.
Collapse
Affiliation(s)
- Achim Thomzig
- Centrum für Anatomie, Charité, Universitätsmedizin Berlin, D-10115 Berlin, Germany
| | | | | | | |
Collapse
|
43
|
Jin X, Malykhina AP, Lupu F, Akbarali HI. Altered gene expression and increased bursting activity of colonic smooth muscle ATP-sensitive K+ channels in experimental colitis. Am J Physiol Gastrointest Liver Physiol 2004; 287:G274-85. [PMID: 14962845 DOI: 10.1152/ajpgi.00472.2003] [Citation(s) in RCA: 52] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
The ATP-sensitive K(+) channel (K(ATP)) is a complex composed of an inwardly rectifying, pore-forming subunit (Kir 6.1 and Kir 6.2) and the sulfonylurea receptor (SUR1 and SUR2). In gastrointestinal smooth muscle, these channels are important in regulating cell excitability. We examined the molecular composition of the K(ATP) channel in mouse colonic smooth muscle and determined its activity in the pathophysiological setting of experimental colitis. Following 7 days of dextran sulfate sodium (DSS) treatment in drinking water, colonic inflammation was scored by histology and physical signs. In whole cell recordings, levcromakalim-induced currents were significantly larger in inflamed cells. In cell-attached patch recordings of single-channel events, levcromakalim enhanced the bursting duration in inflamed cells. The single-channel conductance of approximately 42 pS was not altered with inflammation. mRNA for both Kir 6.1 and 6.2 were detected by RT-PCR. Kir 6.1 was localized to the plasma membrane, whereas Kir 6.2 was mainly detected in the cytosol by immunohistochemistry. Quantitative PCR showed that Kir 6.1 gene expression was upregulated by almost 22-fold, whereas SUR2B was downregulated by threefold after inflammation. Thus decreased motility of the colon during inflammation may be associated with changes in the transcriptional regulation of Kir 6.1 and SUR2B gene expression.
Collapse
Affiliation(s)
- Xiaochun Jin
- Dept. of Physiology, University of Oklahoma Health Science Center, 940 Stanton L. Young Boulevard, Oklahoma City, OK 73104, USA
| | | | | | | |
Collapse
|
44
|
Quinn KV, Giblin JP, Tinker A. Multisite phosphorylation mechanism for protein kinase A activation of the smooth muscle ATP-sensitive K+ channel. Circ Res 2004; 94:1359-66. [PMID: 15087422 DOI: 10.1161/01.res.0000128513.34817.c4] [Citation(s) in RCA: 65] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
The activation of ATP-sensitive K+ channels by protein kinase A in vascular smooth muscle is an important component of the action of vasodilators. In this study, we examine the molecular mechanisms of regulation of the cloned equivalent of this channel comprising the sulfonylurea receptor 2B and the inward rectifier 6.1 subunit (SUR2B/Kir6.1). Specifically, we focus on whether the channel is directly phosphorylated and the sites at which this occurs in the protein complex. We identify one site in Kir6.1 (S385) and two sites in SUR2B (T633 and S1465) using a combination of biochemical and functional assays. Our work supports a model in which multiple sites in the channel complex have to be phosphorylated before activation occurs.
Collapse
Affiliation(s)
- Kathryn V Quinn
- British Heart Foundation Laboratories and the Department of Medicine, University College London, UK
| | | | | |
Collapse
|
45
|
van Bever L, Poitry S, Faure C, Norman RI, Roatti A, Baertschi AJ. Pore loop-mutated rat KIR6.1 and KIR6.2 suppress KATP current in rat cardiomyocytes. Am J Physiol Heart Circ Physiol 2004; 287:H850-9. [PMID: 15044189 DOI: 10.1152/ajpheart.00054.2004] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Cardiomyocytes express mRNA for all major subunits of ATP-sensitive potassium (K(ATP)) channels: KIR6.1, KIR6.2, SUR1A, SUR2A, and SUR2B. It has remained controversial as to whether KIR6.1 may associate with KIR6.2 to form the tetrameric pore of K(ATP) channels in cardiomyocytes. To explore this possibility, cultured rat cardiomyocytes were examined for an inhibition of K(ATP) current by overexpression of pore loop-mutated (inactive) KIR6.x. Bicistronic plasmids were constructed encoding loop-mutated (AFA or SFG for GFG) rat KIR6.x followed by EGFP. In ventricular myocytes, the overexpression of KIR6.1SFG-pIRES(2)-EGFP or KIR6.2AFA-pIRES(2)-EGFP DNA caused, after 72 h, a major decrease of K(ATP) current density of 85.8% and 82.7%, respectively (P < 0.01), relative to EGFP controls (59 +/- 9 pA/pF). In atrial myocytes, overexpression of these pore-mutated KIR6.x by 6.0-fold and 10.6-fold, as assessed by quantitative immunohistochemistry, caused a decrease of K(ATP) current density of 73.7% and 58.5%, respectively (P < 0.01). Expression of wild-type rat KIR6.2 increased the ventricular and atrial K(ATP) current density by 58.3% and 42.9%, respectively (P < 0.01), relative to corresponding EGFP controls, indicating a reserve of SUR to accommodate increased KIR6.x trafficking to the sarcolemma. The results favor the view that KIR6.1 may associate with KIR6.2 to form heterotetrameric pores of native K(ATP) channels in cardiomyocytes.
Collapse
Affiliation(s)
- Laurianne van Bever
- Department of Physiology, Centre Médical Universitarie, Geneva 1204, Switzerland
| | | | | | | | | | | |
Collapse
|
46
|
Teramoto N, Zhu HL, Ito Y. Blocking actions of glibenclamide on ATP-sensitive K+ channels in pig urethral myocytes. J Pharm Pharmacol 2004; 56:395-9. [PMID: 15025866 DOI: 10.1211/0022357022755] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022]
Abstract
The inhibitory effects of glibenclamide on the levcromakalim-induced ATP-sensitive K+ (K(ATP)) channels were investigated with cell-attached configuration in pig proximal urethra. Application of 10 microM glibenclamide reversibly inhibited the activity of the 100 microM levcromakalim-induced K(ATP) channel, decreasing not only the channel open probability but also the amplitude of unitary current. The inhibitory concentration-response curve of the glibenclamide-induced sublevel conductance of K(ATP) channel was shifted to the right (IC50 = 4.7 microM), compared with the levcromakalim-induced K(ATP) channel (full conductance, IC50 = 0.5 microM). Glibenclamide is the first reported sulphonylurea to selectively block K(ATP) channel, not only by decreasing the channel activity but also by reducing the unitary amplitude in smooth muscle.
Collapse
Affiliation(s)
- Noriyoshi Teramoto
- Department of Pharmacology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, 812-8582, Japan.
| | | | | |
Collapse
|
47
|
Sun X, Cao K, Yang G, Huang Y, Hanna ST, Wang R. Selective expression of Kir6.1 protein in different vascular and non-vascular tissues. Biochem Pharmacol 2004; 67:147-56. [PMID: 14667937 DOI: 10.1016/j.bcp.2003.08.041] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
K(ATP) channels are composed of pore-forming subunits Kir6.x and auxiliary subunits SURx. These channels play important roles in modulating the contractility of vascular smooth muscle cells (SMCs) by altering membrane potentials. The molecular basis of K(ATP) channels in vascular SMCs is unclear and the expression of different K(ATP) channel subunits at protein level in various tissues still undetermined. In this study, using an anti-Kir6.1 antibody, we detected the expression of Kir6.1 proteins in rat vascular tissues including mesenteric artery, pulmonary artery, aorta, and tail artery. Kir6.1 proteins were also identified in heart and other non-vascular tissues including spleen and brain, but they were undetectable in liver and kidney. Immunocytochemical study revealed the expression of Kir6.1 proteins in cultured rat thoracic aortic SMCs. Using the whole-cell patch-clamp technique, it was found that the intracellularly applied anti-Kir6.1 antibody significantly inhibited K(ATP) channel currents in HEK-293 cells that were stably transfected with Kir6.1 cDNA. A better understanding of differential expression of Kir6.1 proteins in various vascular and non-vascular tissues may help discern different molecular basis and functions of K(ATP) channel complexes in these tissues.
Collapse
Affiliation(s)
- Xianfeng Sun
- Department of Physiology, College of Medicine, University of Saskatchewan, Sask., S7N 5E5, Saskatoon, Canada
| | | | | | | | | | | |
Collapse
|
48
|
Kuniyasu A, Kaneko K, Kawahara K, Nakayama H. Molecular assembly and subcellular distribution of ATP-sensitive potassium channel proteins in rat hearts. FEBS Lett 2003; 552:259-63. [PMID: 14527696 DOI: 10.1016/s0014-5793(03)00936-0] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
Cardiac ATP-sensitive K(+) (K(ATP)) channels are proposed to contribute to cardio-protection and ischemic preconditioning. Although mRNAs for all subunits of K(ATP) channels (Kir6.0 and sulfonylurea receptors SURs) were detected in hearts, subcellular localization of their proteins and the subunit combination are not well elucidated. We address these questions in rat hearts, using anti-peptide antibodies raised against each subunit. By immunoblot analysis, all of the subunits were detected in microsomal fractions including sarcolemmal membranes, while they were not detected in mitochondrial fractions at all. Immunoprecipitation and sucrose gradient sedimentation of the digitonin-solubilized microsomes indicated that Kir6.2 exclusively assembled with SUR2A. The molecular mass of the Kir6.2-SUR2A complex estimated by sucrose sedimentation was 1150 kDa, significantly larger than the calculated value for (Kir6.2)(4)-(SUR2A)(4), suggesting a potential formation of micellar complex with digitonin but no indication of hybrid channel formation under the conditions. These findings provide additional information on the structural and functional relationships of cardiac K(ATP) channel proteins involving subcellular localization and roles for cardioprotection and ischemic preconditioning.
Collapse
Affiliation(s)
- Akihiko Kuniyasu
- Department of Molecular Cell Function, Graduate School of Medical and Pharmaceutical Sciences, Kumamoto University, 5-1 Ohe-Honmachi, 862-0973 Kumamoto, Japan
| | | | | | | |
Collapse
|
49
|
Quinn KV, Cui Y, Giblin JP, Clapp LH, Tinker A. Do anionic phospholipids serve as cofactors or second messengers for the regulation of activity of cloned ATP-sensitive K+ channels? Circ Res 2003; 93:646-55. [PMID: 12970116 DOI: 10.1161/01.res.0000095247.81449.8e] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
The regulation of ion channels by anionic phospholipids is currently very topical. An outstanding issue is whether phosphatidylinositol 4,5-diphosphate and related species act as true second messengers in signaling or behave in a manner analogous to an enzymatic cofactor. This question is especially pertinent regarding ATP-sensitive K+ channels in smooth muscle, for which there is substantial literature supporting inhibitory regulation by hormones. In this study, we have examined regulation of the potential cloned equivalents of the smooth muscle ATP-sensitive K+ channel (SUR2B/Kir6.1 and SUR2B/Kir6.2). We find that both can be inhibited via the Gq/11-coupled muscarinic M3 receptor but that the pathways by which this occurs are different. Our data show that SUR2B/Kir6.1 is inhibited by protein kinase C and binds anionic phospholipids with high affinity, such that potential physiological fluctuations in their levels do not influence channel activity. In contrast, Kir6.2 is not regulated by protein kinase C but binds anionic phospholipids with low affinity. In this case, phosphatidylinositol 4,5-diphosphate and related species have the potential to act as second messengers in signaling. Thus, Kir6.1 and Kir6.2 are regulated by distinct inhibitory mechanisms.
Collapse
Affiliation(s)
- Kathryn V Quinn
- BHF Laboratories and Department of Medicine, University College London, 5 University St, London WC1E 6JJ, UK
| | | | | | | | | |
Collapse
|
50
|
Yunoki T, Teramoto N, Ito Y. Functional involvement of sulphonylurea receptor (SUR) type 1 and 2B in the activity of pig urethral ATP-sensitive K+ channels. Br J Pharmacol 2003; 139:652-60. [PMID: 12788825 PMCID: PMC1573868 DOI: 10.1038/sj.bjp.0705268] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
(1) We have investigated the possible roles of sulphonylurea receptor (SUR) type 1 and 2B in the activity of pig urethral ATP-sensitive K(+) channels (K(ATP) channels) by use of patch-clamp and reverse transcriptase-polymerase chain reaction (RT-PCR) techniques. (2) In voltage-clamp experiments, not only diazoxide, a SUR1 and weak SUR2B activator, but also pinacidil, a selective SUR2 activator, caused an inward current at a holding potential of -50 mV in symmetrical 140 mM K(+) conditions. (3) Gliclazide (</=1 micro M), a selective SUR1 blocker, inhibited the 10 micro M pinacidil-induced currents (K(i)=177 micro M) and the 500 micro M diazoxide-induced currents (high-affinity site, K(i1)=5 nM; low-affinity site, K(i2)=108 micro M) at -50 mV. (4) Application of tolbutamide (</=100 micro M) reversibly caused an inhibition of the 500 micro M diazoxide-induced current at -50 mV. (5) MCC-134, a SUR type-specific K(ATP) channel regulator (1-100 micro M), produced a concentration-dependent inward K(+) current, which was suppressed by the application of glibenclamide at -50 mV. The amplitude of the MCC-134 (100 micro M)-induced current was approximately 50% of that of the 100 micro M pinacidil-induced currents. (6) Using cell-attached configuration, MCC-134 activated a glibenclamide-sensitive K(ATP) channel which was also activated by pinacidil. (7) RT-PCR analysis demonstrated the presence of SUR1 and SUR2B transcripts in pig urethra. 8 These results indicate that both SUR1 and SUR2B subunits play a functional role in regulating the activity of pig urethral K(ATP) channels and that SUR1 contributes less than 25% to total K(ATP) currents.
Collapse
Affiliation(s)
- Takakazu Yunoki
- Department of Pharmacology, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi Ward, Fukuoka 812-8582, Japan
- Japanese Society for the Promotion of Sciences, Tokyo 102-8471, Japan
| | - Noriyoshi Teramoto
- Department of Pharmacology, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi Ward, Fukuoka 812-8582, Japan
- Author for correspondence:
| | - Yushi Ito
- Department of Pharmacology, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi Ward, Fukuoka 812-8582, Japan
| |
Collapse
|