1
|
Cairns SP, Lindinger MI. Lactic acidosis: implications for human exercise performance. Eur J Appl Physiol 2025:10.1007/s00421-025-05750-0. [PMID: 40088272 DOI: 10.1007/s00421-025-05750-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2024] [Accepted: 02/22/2025] [Indexed: 03/17/2025]
Abstract
During high-intensity exercise a lactic-acidosis occurs with raised myoplasmic and plasma concentrations of lactate- and protons ([lactate-], [H+] or pH). We critically evaluate whether this causes/contributes to fatigue during human exercise. Increases of [lactate-] per se (to 25 mM in plasma, 50 mM intracellularly) exert little detrimental effect on muscle performance while ingestion/infusion of lactate- can be ergogenic. An exercise-induced intracellular acidosis at the whole-muscle level (pHi falls from 7.1-7.0 to 6.9-6.3), incorporates small changes in slow-twitch fibres (pHi ~ 6.9) and large changes in fast-twitch fibres (pHi ~ 6.2). The relationship between peak force/power and acidosis during fatiguing contractions varies across exercise regimes implying that acidosis is not the sole cause of fatigue. Concomitant changes of other putative fatigue factors include phosphate metabolites, glycogen, ions and reactive oxygen species. Acidosis to pHi 6.7-6.6 at physiological temperatures (during recovery from exercise or induced in non-fatigued muscle), has minimal effect on force/power. Acidosis to pHi ~ 6.5-6.2 per se reduces maximum force (~12%), slows shortening velocity (~5%), and lowers peak power (~22%) in non-fatigued muscles/individuals. A pre-exercise induced-acidosis with ammonium chloride impairs exercise performance in humans and accelerates the decline of force/power (15-40% initial) in animal muscles stimulated repeatedly in situ. Raised [H+]i and diprotonated inorganic phosphate ([H2PO4-]i) act on myofilament proteins to reduce maximum cross-bridge activity, Ca2+-sensitivity, and myosin ATPase activity. Acidosis/[lactate-]o attenuates detrimental effects of large K+-disturbances on action potentials and force in non-fatigued muscle. We propose that depressive effects of acidosis and [H2PO4-]i on myofilament function dominate over the protective effects of acidosis/lactate- on action potentials during fatigue. Raised extracellular [H+]/[lactate-] do not usually cause central fatigue but do contribute to elevated perceived exertion and fatigue sensations by activating group III/IV muscle afferents. Modulation of H+/lactate- regulation (via extracellular H+-buffers, monocarboxylate transporters, carbonic anhydrase, carnosine) supports a role for intracellular acidosis in fatigue. In conclusion, current evidence advocates that severe acidosis in fast-twitch fibres can contribute to force/power fatigue during intense human exercise.
Collapse
Affiliation(s)
- Simeon P Cairns
- Sport and Recreation Research Institute New Zealand, School of Sport and Recreation, Faculty of Health and Environmental Sciences, Auckland University of Technology, Private Bag 92006, Auckland, 1020, New Zealand.
- Health and Rehabilitation Research Institute, Faculty of Health and Environmental Sciences, Auckland University of Technology, Auckland, 1020, New Zealand.
| | - Michael I Lindinger
- Research and Development, The Nutraceutical Alliance Inc, Guelph, ON, L8N 3Z5, Canada
| |
Collapse
|
2
|
Boedtkjer E, Ara T. Strengthening the basics: acids and bases influence vascular structure and function, tissue perfusion, blood pressure, and human cardiovascular disease. Pflugers Arch 2024; 476:623-637. [PMID: 38383822 DOI: 10.1007/s00424-024-02926-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2023] [Revised: 02/08/2024] [Accepted: 02/09/2024] [Indexed: 02/23/2024]
Abstract
Acids and their conjugate bases accumulate in or dissipate from the interstitial space when tissue perfusion does not match the metabolic demand. Extracellular acidosis dilates most arterial beds, but associated acid-base disturbances-e.g., intracellular acidification and decreases in HCO3- concentration-can also elicit pro-contractile influences that diminish vasodilation and even dominate in some vascular beds to cause vasoconstriction. The ensemble activities of the acid-base-sensitive reactions in vascular smooth muscle and endothelial cells optimize vascular resistance for blood pressure control and direct the perfusion towards active tissue. In this review, we describe the mechanisms of intracellular pH regulation in the vascular wall and discuss how vascular smooth muscle and endothelial cells sense acid-base disturbances. We further deliberate on the functional effects of local acid-base disturbances and their integrated cardiovascular consequences under physiological and pathophysiological conditions. Finally, we address how mutations and polymorphisms in the molecular machinery that regulates pH locally and senses acid-base disturbances in the vascular wall can result in cardiovascular disease. Based on the emerging molecular insight, we propose that targeting local pH-dependent effectors-rather than systemic acid-base disturbances-has therapeutic potential to interfere with the progression and reduce the severity of cardiovascular disease.
Collapse
Affiliation(s)
- Ebbe Boedtkjer
- Department of Biomedicine, Aarhus University, Hoegh-Guldbergs Gade 10, DK-8000, Aarhus, Denmark.
| | - Tarannum Ara
- Department of Biomedicine, Aarhus University, Hoegh-Guldbergs Gade 10, DK-8000, Aarhus, Denmark
| |
Collapse
|
3
|
Daher A, Payne S. The conducted vascular response as a mediator of hypercapnic cerebrovascular reactivity: A modelling study. Comput Biol Med 2024; 170:107985. [PMID: 38245966 DOI: 10.1016/j.compbiomed.2024.107985] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2023] [Revised: 12/29/2023] [Accepted: 01/13/2024] [Indexed: 01/23/2024]
Abstract
It is well established that the cerebral blood flow (CBF) shows exquisite sensitivity to changes in the arterial blood partial pressure of CO2 ( [Formula: see text] ), which is reflected by an index termed cerebrovascular reactivity. In response to elevations in [Formula: see text] (hypercapnia), the vessels of the cerebral microvasculature dilate, thereby decreasing the vascular resistance and increasing CBF. Due to the challenges of access, scale and complexity encountered when studying the microvasculature, however, the mechanisms behind cerebrovascular reactivity are not fully understood. Experiments have previously established that the cholinergic release of the Acetylcholine (ACh) neurotransmitter in the cortex is a prerequisite for the hypercapnic response. It is also known that ACh functions as an endothelial-dependent agonist, in which the local administration of ACh elicits local hyperpolarization in the vascular wall; this hyperpolarization signal is then propagated upstream the vascular network through the endothelial layer and is coupled to a vasodilatory response in the vascular smooth muscle (VSM) layer in what is known as the conducted vascular response (CVR). Finally, experimental data indicate that the hypercapnic response is more strongly correlated with the CO2 levels in the tissue than in the arterioles. Accordingly, we hypothesize that the CVR, evoked by increases in local tissue CO2 levels and a subsequent local release of ACh, is responsible for the CBF increase observed in response to elevations in [Formula: see text] . By constructing physiologically grounded dynamic models of CBF and control in the cerebral vasculature, ones that integrate the available knowledge and experimental data, we build a new model of the series of signalling events and pathways underpinning the hypercapnic response, and use the model to provide compelling evidence that corroborates the aforementioned hypothesis. If the CVR indeed acts as a mediator of the hypercapnic response, the proposed mechanism would provide an important addition to our understanding of the repertoire of metabolic feedback mechanisms possessed by the brain and would motivate further in-vivo investigation. We also model the interaction of the hypercapnic response with dynamic cerebral autoregulation (dCA), the collection of mechanisms that the brain possesses to maintain near constant CBF despite perturbations in pressure, and show how the dCA mechanisms, which otherwise tend to be overlooked when analysing experimental results of cerebrovascular reactivity, could play a significant role in shaping the CBF response to elevations in [Formula: see text] . Such in-silico models can be used in tandem with in-vivo experiments to expand our understanding of cerebrovascular diseases, which continue to be among the leading causes of morbidity and mortality in humans.
Collapse
Affiliation(s)
- Ali Daher
- Institute of Biomedical Engineering, Department of Engineering Science, University of Oxford, United Kingdom.
| | - Stephen Payne
- Institute of Applied Mechanics, National Taiwan University, Taiwan
| |
Collapse
|
4
|
McClenaghan C, Nichols CG. Kir6.1 and SUR2B in Cantú syndrome. Am J Physiol Cell Physiol 2022; 323:C920-C935. [PMID: 35876283 PMCID: PMC9467476 DOI: 10.1152/ajpcell.00154.2022] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2022] [Revised: 07/07/2022] [Accepted: 07/07/2022] [Indexed: 12/25/2022]
Abstract
Kir6.1 and SUR2 are subunits of ATP-sensitive potassium (KATP) channels expressed in a wide range of tissues. Extensive study has implicated roles of these channel subunits in diverse physiological functions. Together they generate the predominant KATP conductance in vascular smooth muscle and are the target of vasodilatory drugs. Roles for Kir6.1/SUR2 dysfunction in disease have been suggested based on studies of animal models and human genetic discoveries. In recent years, it has become clear that gain-of-function (GoF) mutations in both genes result in Cantú syndrome (CS)-a complex, multisystem disorder. There is currently no targeted therapy for CS, but studies of mouse models of the disease reveal that pharmacological reversibility of cardiovascular and gastrointestinal pathologies can be achieved by administration of the KATP channel inhibitor, glibenclamide. Here we review the function, structure, and physiological and pathological roles of Kir6.1/SUR2B channels, with a focus on CS. Recent studies have led to much improved understanding of the underlying pathologies and the potential for treatment, but important questions remain: Can the study of genetically defined CS reveal new insights into Kir6.1/SUR2 function? Do these reveal new pathophysiological mechanisms that may be important in more common diseases? And is our pharmacological armory adequately stocked?
Collapse
Affiliation(s)
- Conor McClenaghan
- Department of Cell Biology and Physiology, Center for the Investigation of Membrane Excitability Diseases (CIMED), Washington University, St. Louis, Missouri
| | - Colin G Nichols
- Department of Cell Biology and Physiology, Center for the Investigation of Membrane Excitability Diseases (CIMED), Washington University, St. Louis, Missouri
| |
Collapse
|
5
|
Hostrup M, Cairns SP, Bangsbo J. Muscle Ionic Shifts During Exercise: Implications for Fatigue and Exercise Performance. Compr Physiol 2021; 11:1895-1959. [PMID: 34190344 DOI: 10.1002/cphy.c190024] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Exercise causes major shifts in multiple ions (e.g., K+ , Na+ , H+ , lactate- , Ca2+ , and Cl- ) during muscle activity that contributes to development of muscle fatigue. Sarcolemmal processes can be impaired by the trans-sarcolemmal rundown of ion gradients for K+ , Na+ , and Ca2+ during fatiguing exercise, while changes in gradients for Cl- and Cl- conductance may exert either protective or detrimental effects on fatigue. Myocellular H+ accumulation may also contribute to fatigue development by lowering glycolytic rate and has been shown to act synergistically with inorganic phosphate (Pi) to compromise cross-bridge function. In addition, sarcoplasmic reticulum Ca2+ release function is severely affected by fatiguing exercise. Skeletal muscle has a multitude of ion transport systems that counter exercise-related ionic shifts of which the Na+ /K+ -ATPase is of major importance. Metabolic perturbations occurring during exercise can exacerbate trans-sarcolemmal ionic shifts, in particular for K+ and Cl- , respectively via metabolic regulation of the ATP-sensitive K+ channel (KATP ) and the chloride channel isoform 1 (ClC-1). Ion transport systems are highly adaptable to exercise training resulting in an enhanced ability to counter ionic disturbances to delay fatigue and improve exercise performance. In this article, we discuss (i) the ionic shifts occurring during exercise, (ii) the role of ion transport systems in skeletal muscle for ionic regulation, (iii) how ionic disturbances affect sarcolemmal processes and muscle fatigue, (iv) how metabolic perturbations exacerbate ionic shifts during exercise, and (v) how pharmacological manipulation and exercise training regulate ion transport systems to influence exercise performance in humans. © 2021 American Physiological Society. Compr Physiol 11:1895-1959, 2021.
Collapse
Affiliation(s)
- Morten Hostrup
- Section of Integrative Physiology, Department of Nutrition, Exercise and Sports, University of Copenhagen, Copenhagen, Denmark
| | - Simeon Peter Cairns
- SPRINZ, School of Sport and Recreation, Auckland University of Technology, Auckland, New Zealand.,Health and Rehabilitation Research Institute, Auckland University of Technology, Auckland, New Zealand
| | - Jens Bangsbo
- Section of Integrative Physiology, Department of Nutrition, Exercise and Sports, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
6
|
Bonnet U, Wiemann M. Topiramate Decelerates Bicarbonate-Driven Acid-Elimination of Human Neocortical Neurons: Strategic Significance for its Antiepileptic, Antimigraine and Neuroprotective Properties. CNS & NEUROLOGICAL DISORDERS-DRUG TARGETS 2020; 19:264-275. [PMID: 32496992 DOI: 10.2174/1871527319666200604173208] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/15/2019] [Revised: 04/08/2020] [Accepted: 04/09/2020] [Indexed: 12/27/2022]
Abstract
BACKGROUND Mammalian central neurons regulate their intracellular pH (pHi) strongly and even slight pHi-fluctuations can influence inter-/intracellular signaling, synaptic plasticity and excitability. OBJECTIVE For the first time, we investigated topiramate´s (TPM) influence on pHi-behavior of human central neurons representing a promising target for anticonvulsants and antimigraine drugs. METHODS In slice-preparations of tissue resected from the middle temporal gyrus of five adults with intractable temporal lobe epilepsy, BCECF-AM-loaded neocortical pyramidal-cells were investigated by fluorometry. The pHi-regulation was estimated by using the recovery-slope from intracellular acidification after an Ammonium-Prepulse (APP). RESULTS Among 17 pyramidal neurons exposed to 50 μM TPM, seven (41.24%) responded with an altered resting-pHi (7.02±0.12), i.e., acidification of 0.01-0.03 pH-units. The more alkaline the neurons, the greater the TPM-related acidifications (r=0.7, p=0.001, n=17). The recovery from APPacidification was significantly slowed under TPM (p<0.001, n=5). Further experiments using nominal bicarbonate-free (n=2) and chloride-free (n=2) conditions pointed to a modulation of the HCO3 -- driven pHi-regulation by TPM, favoring a stimulation of the passive Cl-/HCO3 --antiporter (CBT) - an acid-loader predominantly in more alkaline neurons. CONCLUSION TPM modulated the bicarbonate-driven pHi-regulation, just as previously described in adult guinea-pig hippocampal neurons. We discussed the significance of the resulting subtle acidifications for beneficial antiepileptic, antimigraine and neuroprotective effects as well as for unwanted cognitive deficits.
Collapse
Affiliation(s)
- Udo Bonnet
- Department of Psychiatry, Psychotherapy, and Psychosomatic Medicine, Evangelisches Krankenhaus Castrop-Rauxel, Academic Teaching Hospital of the University Duisburg-Essen, Castrop-Rauxel, Germany.,Department of Psychiatry and Psychotherapy, Faculty of Medicine, LVR-Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Martin Wiemann
- Institute of Physiology, University of Duisburg-Essen, Essen, Germany.,IBE R&D gGmbH, Institute for Lung Health, D-48149 Munster, Germany
| |
Collapse
|
7
|
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: 10.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
|
8
|
Boedtkjer E. Acid-base regulation and sensing: Accelerators and brakes in metabolic regulation of cerebrovascular tone. J Cereb Blood Flow Metab 2018; 38:588-602. [PMID: 28984162 PMCID: PMC5888856 DOI: 10.1177/0271678x17733868] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/08/2017] [Revised: 08/10/2017] [Accepted: 09/06/2017] [Indexed: 12/29/2022]
Abstract
Metabolic regulation of cerebrovascular tone directs blood flow to areas of increased neuronal activity and during disease states partially compensates for insufficient perfusion by enhancing blood flow in collateral blood vessels. Acid-base disturbances frequently occur as result of enhanced metabolism or insufficient blood supply, but despite definitive evidence that acid-base disturbances alter arterial tone, effects of individual acid-base equivalents and the underlying signaling mechanisms are still being debated. H+ is an important intra- and extracellular messenger that modifies cerebrovascular tone. In addition, low extracellular [HCO3-] promotes cerebrovascular contraction through an endothelium-dependent mechanism. CO2 alters arterial tone development via changes in intra- and extracellular pH but it is still controversial whether CO2 also has direct vasomotor effects. Vasocontractile responses to low extracellular [HCO3-] and acute CO2-induced decreases in intracellular pH can counteract H+-mediated vasorelaxation during metabolic and respiratory acidosis, respectively, and may thereby reduce the risk of capillary damage and cerebral edema that could be consequences of unopposed vasodilation. In this review, the signaling mechanisms for acid-base equivalents in cerebral arteries and the mechanisms of intracellular pH control in the arterial wall are discussed in the context of metabolic regulation of cerebrovascular tone and local perfusion.
Collapse
Affiliation(s)
- Ebbe Boedtkjer
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
| |
Collapse
|
9
|
Smith ZM, Krizay E, Sá RC, Li ET, Scadeng M, Powell FL, Dubowitz DJ. Evidence from high-altitude acclimatization for an integrated cerebrovascular and ventilatory hypercapnic response but different responses to hypoxia. J Appl Physiol (1985) 2017; 123:1477-1486. [PMID: 28705997 DOI: 10.1152/japplphysiol.00341.2017] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Abstract
Ventilation and cerebral blood flow (CBF) are both sensitive to hypoxia and hypercapnia. To compare chemosensitivity in these two systems, we made simultaneous measurements of ventilatory and cerebrovascular responses to hypoxia and hypercapnia in 35 normal human subjects before and after acclimatization to hypoxia. Ventilation and CBF were measured during stepwise changes in isocapnic hypoxia and iso-oxic hypercapnia. We used MRI to quantify actual cerebral perfusion. Measurements were repeated after 2 days of acclimatization to hypoxia at 3,800 m altitude (partial pressure of inspired O2 = 90 Torr) to compare plasticity in the chemosensitivity of these two systems. Potential effects of hypoxic and hypercapnic responses on acute mountain sickness (AMS) were assessed also. The pattern of CBF and ventilatory responses to hypercapnia were almost identical. CO2 responses were augmented to a similar degree in both systems by concomitant acute hypoxia or acclimatization to sustained hypoxia. Conversely, the pattern of CBF and ventilatory responses to hypoxia were markedly different. Ventilation showed the well-known increase with acute hypoxia and a progressive decline in absolute value over 25 min of sustained hypoxia. With acclimatization to hypoxia for 2 days, the absolute values of ventilation and O2 sensitivity increased. By contrast, O2 sensitivity of CBF or its absolute value did not change during sustained hypoxia for up to 2 days. The results suggest a common or integrated control mechanism for CBF and ventilation by CO2 but different mechanisms of O2 sensitivity and plasticity between the systems. Ventilatory and cerebrovascular responses were the same for all subjects irrespective of AMS symptoms. NEW & NOTEWORTHY Ventilatory and cerebrovascular hypercapnic response patterns show similar plasticity in CO2 sensitivity following hypoxic acclimatization, suggesting an integrated control mechanism. Conversely, ventilatory and cerebrovascular hypoxic responses differ. Ventilation initially increases but adapts with prolonged hypoxia (hypoxic ventilatory decline), and ventilatory sensitivity increases following acclimatization. In contrast, cerebral blood flow hypoxic sensitivity remains constant over a range of hypoxic stimuli, with no cerebrovascular acclimatization to sustained hypoxia, suggesting different mechanisms for O2 sensitivity in the two systems.
Collapse
Affiliation(s)
- Zachary M Smith
- Department of Radiology, Center for Functional MRI, University of California San Diego School of Medicine , La Jolla, California
| | - Erin Krizay
- Department of Radiology, Center for Functional MRI, University of California San Diego School of Medicine , La Jolla, California
| | - Rui Carlos Sá
- Division of Physiology, Department of Medicine, University of California San Diego School of Medicine , La Jolla, California
| | - Ethan T Li
- Department of Radiology, Center for Functional MRI, University of California San Diego School of Medicine , La Jolla, California
| | - Miriam Scadeng
- Department of Radiology, Center for Functional MRI, University of California San Diego School of Medicine , La Jolla, California
| | - Frank L Powell
- Division of Physiology, Department of Medicine, University of California San Diego School of Medicine , La Jolla, California.,White Mountain Research Station, University of California , Bishop, California
| | - David J Dubowitz
- Department of Radiology, Center for Functional MRI, University of California San Diego School of Medicine , La Jolla, California
| |
Collapse
|
10
|
Zhang B, Novitskaya T, Wheeler DG, Xu Z, Chepurko E, Huttinger R, He H, Varadharaj S, Zweier JL, Song Y, Xu M, Harrell FE, Su YR, Absi T, Kohr MJ, Ziolo MT, Roden DM, Shaffer CM, Galindo CL, Wells QS, Gumina RJ. Kcnj11 Ablation Is Associated With Increased Nitro-Oxidative Stress During Ischemia-Reperfusion Injury: Implications for Human Ischemic Cardiomyopathy. Circ Heart Fail 2017; 10:CIRCHEARTFAILURE.116.003523. [PMID: 28209764 DOI: 10.1161/circheartfailure.116.003523] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/03/2015] [Accepted: 01/06/2017] [Indexed: 11/16/2022]
Abstract
BACKGROUND Despite increased secondary cardiovascular events in patients with ischemic cardiomyopathy (ICM), the expression of innate cardiac protective molecules in the hearts of patients with ICM is incompletely characterized. Therefore, we used a nonbiased RNAseq approach to determine whether differences in cardiac protective molecules occur with ICM. METHODS AND RESULTS RNAseq analysis of human control and ICM left ventricular samples demonstrated a significant decrease in KCNJ11 expression with ICM. KCNJ11 encodes the Kir6.2 subunit of the cardioprotective KATP channel. Using wild-type mice and kcnj11-deficient (kcnj11-null) mice, we examined the effect of kcnj11 expression on cardiac function during ischemia-reperfusion injury. Reactive oxygen species generation increased in kcnj11-null hearts above that found in wild-type mice hearts after ischemia-reperfusion injury. Continuous left ventricular pressure measurement during ischemia and reperfusion demonstrated a more compromised diastolic function in kcnj11-null compared with wild-type mice during reperfusion. Analysis of key calcium-regulating proteins revealed significant differences in kcnj11-null mice. Despite impaired relaxation, kcnj11-null hearts increased phospholamban Ser16 phosphorylation, a modification that results in the dissociation of phospholamban from sarcoendoplasmic reticulum Ca2+, thereby increasing sarcoendoplasmic reticulum Ca2+-mediated calcium reuptake. However, kcnj11-null mice also had increased 3-nitrotyrosine modification of the sarcoendoplasmic reticulum Ca2+-ATPase, a modification that irreversibly impairs sarcoendoplasmic reticulum Ca2+ function, thereby contributing to diastolic dysfunction. CONCLUSIONS KCNJ11 expression is decreased in human ICM. Lack of kcnj11 expression increases peroxynitrite-mediated modification of the key calcium-handling protein sarcoendoplasmic reticulum Ca2+-ATPase after myocardial ischemia-reperfusion injury, contributing to impaired diastolic function. These data suggest a mechanism for ischemia-induced diastolic dysfunction in patients with ICM.
Collapse
Affiliation(s)
- Bo Zhang
- From the Division of Cardiovascular Medicine, Davis Heart and Lung Research Institute (B.Z., D.G.W., Z.X., R.H., H.H., S.V., J.L.Z.), Department of Physiology and Cell Biology (B.Z., J.L.Z., M.J.K., M.T.Z.), The Ohio State University, Columbus; Institute of Organ Transplantation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, HuBei, China (B.Z.); Department of Biostatistics (Y.S., M.X., F.E.H.), Division of Clinical Pharmacology, Department of Medicine (D.M.R., C.M.S.), Division of Cardiac Surgery, Department of Surgery (T.A.), Division of Cardiovascular Medicine (T. N., E. C., Y.R.S., D.R., C.L.G., Q.S.W, R.J.G.), Department of Pharmacology and Department of Pathology, Immunology, and Microbiology (R.J.G.), Vanderbilt University Medical Center, Nashville, TN
| | - Tatiana Novitskaya
- From the Division of Cardiovascular Medicine, Davis Heart and Lung Research Institute (B.Z., D.G.W., Z.X., R.H., H.H., S.V., J.L.Z.), Department of Physiology and Cell Biology (B.Z., J.L.Z., M.J.K., M.T.Z.), The Ohio State University, Columbus; Institute of Organ Transplantation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, HuBei, China (B.Z.); Department of Biostatistics (Y.S., M.X., F.E.H.), Division of Clinical Pharmacology, Department of Medicine (D.M.R., C.M.S.), Division of Cardiac Surgery, Department of Surgery (T.A.), Division of Cardiovascular Medicine (T. N., E. C., Y.R.S., D.R., C.L.G., Q.S.W, R.J.G.), Department of Pharmacology and Department of Pathology, Immunology, and Microbiology (R.J.G.), Vanderbilt University Medical Center, Nashville, TN
| | - Debra G Wheeler
- From the Division of Cardiovascular Medicine, Davis Heart and Lung Research Institute (B.Z., D.G.W., Z.X., R.H., H.H., S.V., J.L.Z.), Department of Physiology and Cell Biology (B.Z., J.L.Z., M.J.K., M.T.Z.), The Ohio State University, Columbus; Institute of Organ Transplantation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, HuBei, China (B.Z.); Department of Biostatistics (Y.S., M.X., F.E.H.), Division of Clinical Pharmacology, Department of Medicine (D.M.R., C.M.S.), Division of Cardiac Surgery, Department of Surgery (T.A.), Division of Cardiovascular Medicine (T. N., E. C., Y.R.S., D.R., C.L.G., Q.S.W, R.J.G.), Department of Pharmacology and Department of Pathology, Immunology, and Microbiology (R.J.G.), Vanderbilt University Medical Center, Nashville, TN
| | - Zhaobin Xu
- From the Division of Cardiovascular Medicine, Davis Heart and Lung Research Institute (B.Z., D.G.W., Z.X., R.H., H.H., S.V., J.L.Z.), Department of Physiology and Cell Biology (B.Z., J.L.Z., M.J.K., M.T.Z.), The Ohio State University, Columbus; Institute of Organ Transplantation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, HuBei, China (B.Z.); Department of Biostatistics (Y.S., M.X., F.E.H.), Division of Clinical Pharmacology, Department of Medicine (D.M.R., C.M.S.), Division of Cardiac Surgery, Department of Surgery (T.A.), Division of Cardiovascular Medicine (T. N., E. C., Y.R.S., D.R., C.L.G., Q.S.W, R.J.G.), Department of Pharmacology and Department of Pathology, Immunology, and Microbiology (R.J.G.), Vanderbilt University Medical Center, Nashville, TN
| | - Elena Chepurko
- From the Division of Cardiovascular Medicine, Davis Heart and Lung Research Institute (B.Z., D.G.W., Z.X., R.H., H.H., S.V., J.L.Z.), Department of Physiology and Cell Biology (B.Z., J.L.Z., M.J.K., M.T.Z.), The Ohio State University, Columbus; Institute of Organ Transplantation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, HuBei, China (B.Z.); Department of Biostatistics (Y.S., M.X., F.E.H.), Division of Clinical Pharmacology, Department of Medicine (D.M.R., C.M.S.), Division of Cardiac Surgery, Department of Surgery (T.A.), Division of Cardiovascular Medicine (T. N., E. C., Y.R.S., D.R., C.L.G., Q.S.W, R.J.G.), Department of Pharmacology and Department of Pathology, Immunology, and Microbiology (R.J.G.), Vanderbilt University Medical Center, Nashville, TN
| | - Ryan Huttinger
- From the Division of Cardiovascular Medicine, Davis Heart and Lung Research Institute (B.Z., D.G.W., Z.X., R.H., H.H., S.V., J.L.Z.), Department of Physiology and Cell Biology (B.Z., J.L.Z., M.J.K., M.T.Z.), The Ohio State University, Columbus; Institute of Organ Transplantation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, HuBei, China (B.Z.); Department of Biostatistics (Y.S., M.X., F.E.H.), Division of Clinical Pharmacology, Department of Medicine (D.M.R., C.M.S.), Division of Cardiac Surgery, Department of Surgery (T.A.), Division of Cardiovascular Medicine (T. N., E. C., Y.R.S., D.R., C.L.G., Q.S.W, R.J.G.), Department of Pharmacology and Department of Pathology, Immunology, and Microbiology (R.J.G.), Vanderbilt University Medical Center, Nashville, TN
| | - Heng He
- From the Division of Cardiovascular Medicine, Davis Heart and Lung Research Institute (B.Z., D.G.W., Z.X., R.H., H.H., S.V., J.L.Z.), Department of Physiology and Cell Biology (B.Z., J.L.Z., M.J.K., M.T.Z.), The Ohio State University, Columbus; Institute of Organ Transplantation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, HuBei, China (B.Z.); Department of Biostatistics (Y.S., M.X., F.E.H.), Division of Clinical Pharmacology, Department of Medicine (D.M.R., C.M.S.), Division of Cardiac Surgery, Department of Surgery (T.A.), Division of Cardiovascular Medicine (T. N., E. C., Y.R.S., D.R., C.L.G., Q.S.W, R.J.G.), Department of Pharmacology and Department of Pathology, Immunology, and Microbiology (R.J.G.), Vanderbilt University Medical Center, Nashville, TN
| | - Saradhadevi Varadharaj
- From the Division of Cardiovascular Medicine, Davis Heart and Lung Research Institute (B.Z., D.G.W., Z.X., R.H., H.H., S.V., J.L.Z.), Department of Physiology and Cell Biology (B.Z., J.L.Z., M.J.K., M.T.Z.), The Ohio State University, Columbus; Institute of Organ Transplantation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, HuBei, China (B.Z.); Department of Biostatistics (Y.S., M.X., F.E.H.), Division of Clinical Pharmacology, Department of Medicine (D.M.R., C.M.S.), Division of Cardiac Surgery, Department of Surgery (T.A.), Division of Cardiovascular Medicine (T. N., E. C., Y.R.S., D.R., C.L.G., Q.S.W, R.J.G.), Department of Pharmacology and Department of Pathology, Immunology, and Microbiology (R.J.G.), Vanderbilt University Medical Center, Nashville, TN
| | - Jay L Zweier
- From the Division of Cardiovascular Medicine, Davis Heart and Lung Research Institute (B.Z., D.G.W., Z.X., R.H., H.H., S.V., J.L.Z.), Department of Physiology and Cell Biology (B.Z., J.L.Z., M.J.K., M.T.Z.), The Ohio State University, Columbus; Institute of Organ Transplantation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, HuBei, China (B.Z.); Department of Biostatistics (Y.S., M.X., F.E.H.), Division of Clinical Pharmacology, Department of Medicine (D.M.R., C.M.S.), Division of Cardiac Surgery, Department of Surgery (T.A.), Division of Cardiovascular Medicine (T. N., E. C., Y.R.S., D.R., C.L.G., Q.S.W, R.J.G.), Department of Pharmacology and Department of Pathology, Immunology, and Microbiology (R.J.G.), Vanderbilt University Medical Center, Nashville, TN
| | - Yanna Song
- From the Division of Cardiovascular Medicine, Davis Heart and Lung Research Institute (B.Z., D.G.W., Z.X., R.H., H.H., S.V., J.L.Z.), Department of Physiology and Cell Biology (B.Z., J.L.Z., M.J.K., M.T.Z.), The Ohio State University, Columbus; Institute of Organ Transplantation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, HuBei, China (B.Z.); Department of Biostatistics (Y.S., M.X., F.E.H.), Division of Clinical Pharmacology, Department of Medicine (D.M.R., C.M.S.), Division of Cardiac Surgery, Department of Surgery (T.A.), Division of Cardiovascular Medicine (T. N., E. C., Y.R.S., D.R., C.L.G., Q.S.W, R.J.G.), Department of Pharmacology and Department of Pathology, Immunology, and Microbiology (R.J.G.), Vanderbilt University Medical Center, Nashville, TN
| | - Meng Xu
- From the Division of Cardiovascular Medicine, Davis Heart and Lung Research Institute (B.Z., D.G.W., Z.X., R.H., H.H., S.V., J.L.Z.), Department of Physiology and Cell Biology (B.Z., J.L.Z., M.J.K., M.T.Z.), The Ohio State University, Columbus; Institute of Organ Transplantation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, HuBei, China (B.Z.); Department of Biostatistics (Y.S., M.X., F.E.H.), Division of Clinical Pharmacology, Department of Medicine (D.M.R., C.M.S.), Division of Cardiac Surgery, Department of Surgery (T.A.), Division of Cardiovascular Medicine (T. N., E. C., Y.R.S., D.R., C.L.G., Q.S.W, R.J.G.), Department of Pharmacology and Department of Pathology, Immunology, and Microbiology (R.J.G.), Vanderbilt University Medical Center, Nashville, TN
| | - Frank E Harrell
- From the Division of Cardiovascular Medicine, Davis Heart and Lung Research Institute (B.Z., D.G.W., Z.X., R.H., H.H., S.V., J.L.Z.), Department of Physiology and Cell Biology (B.Z., J.L.Z., M.J.K., M.T.Z.), The Ohio State University, Columbus; Institute of Organ Transplantation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, HuBei, China (B.Z.); Department of Biostatistics (Y.S., M.X., F.E.H.), Division of Clinical Pharmacology, Department of Medicine (D.M.R., C.M.S.), Division of Cardiac Surgery, Department of Surgery (T.A.), Division of Cardiovascular Medicine (T. N., E. C., Y.R.S., D.R., C.L.G., Q.S.W, R.J.G.), Department of Pharmacology and Department of Pathology, Immunology, and Microbiology (R.J.G.), Vanderbilt University Medical Center, Nashville, TN
| | - Yan Ru Su
- From the Division of Cardiovascular Medicine, Davis Heart and Lung Research Institute (B.Z., D.G.W., Z.X., R.H., H.H., S.V., J.L.Z.), Department of Physiology and Cell Biology (B.Z., J.L.Z., M.J.K., M.T.Z.), The Ohio State University, Columbus; Institute of Organ Transplantation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, HuBei, China (B.Z.); Department of Biostatistics (Y.S., M.X., F.E.H.), Division of Clinical Pharmacology, Department of Medicine (D.M.R., C.M.S.), Division of Cardiac Surgery, Department of Surgery (T.A.), Division of Cardiovascular Medicine (T. N., E. C., Y.R.S., D.R., C.L.G., Q.S.W, R.J.G.), Department of Pharmacology and Department of Pathology, Immunology, and Microbiology (R.J.G.), Vanderbilt University Medical Center, Nashville, TN
| | - Tarek Absi
- From the Division of Cardiovascular Medicine, Davis Heart and Lung Research Institute (B.Z., D.G.W., Z.X., R.H., H.H., S.V., J.L.Z.), Department of Physiology and Cell Biology (B.Z., J.L.Z., M.J.K., M.T.Z.), The Ohio State University, Columbus; Institute of Organ Transplantation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, HuBei, China (B.Z.); Department of Biostatistics (Y.S., M.X., F.E.H.), Division of Clinical Pharmacology, Department of Medicine (D.M.R., C.M.S.), Division of Cardiac Surgery, Department of Surgery (T.A.), Division of Cardiovascular Medicine (T. N., E. C., Y.R.S., D.R., C.L.G., Q.S.W, R.J.G.), Department of Pharmacology and Department of Pathology, Immunology, and Microbiology (R.J.G.), Vanderbilt University Medical Center, Nashville, TN
| | - Mark J Kohr
- From the Division of Cardiovascular Medicine, Davis Heart and Lung Research Institute (B.Z., D.G.W., Z.X., R.H., H.H., S.V., J.L.Z.), Department of Physiology and Cell Biology (B.Z., J.L.Z., M.J.K., M.T.Z.), The Ohio State University, Columbus; Institute of Organ Transplantation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, HuBei, China (B.Z.); Department of Biostatistics (Y.S., M.X., F.E.H.), Division of Clinical Pharmacology, Department of Medicine (D.M.R., C.M.S.), Division of Cardiac Surgery, Department of Surgery (T.A.), Division of Cardiovascular Medicine (T. N., E. C., Y.R.S., D.R., C.L.G., Q.S.W, R.J.G.), Department of Pharmacology and Department of Pathology, Immunology, and Microbiology (R.J.G.), Vanderbilt University Medical Center, Nashville, TN
| | - Mark T Ziolo
- From the Division of Cardiovascular Medicine, Davis Heart and Lung Research Institute (B.Z., D.G.W., Z.X., R.H., H.H., S.V., J.L.Z.), Department of Physiology and Cell Biology (B.Z., J.L.Z., M.J.K., M.T.Z.), The Ohio State University, Columbus; Institute of Organ Transplantation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, HuBei, China (B.Z.); Department of Biostatistics (Y.S., M.X., F.E.H.), Division of Clinical Pharmacology, Department of Medicine (D.M.R., C.M.S.), Division of Cardiac Surgery, Department of Surgery (T.A.), Division of Cardiovascular Medicine (T. N., E. C., Y.R.S., D.R., C.L.G., Q.S.W, R.J.G.), Department of Pharmacology and Department of Pathology, Immunology, and Microbiology (R.J.G.), Vanderbilt University Medical Center, Nashville, TN
| | - Dan M Roden
- From the Division of Cardiovascular Medicine, Davis Heart and Lung Research Institute (B.Z., D.G.W., Z.X., R.H., H.H., S.V., J.L.Z.), Department of Physiology and Cell Biology (B.Z., J.L.Z., M.J.K., M.T.Z.), The Ohio State University, Columbus; Institute of Organ Transplantation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, HuBei, China (B.Z.); Department of Biostatistics (Y.S., M.X., F.E.H.), Division of Clinical Pharmacology, Department of Medicine (D.M.R., C.M.S.), Division of Cardiac Surgery, Department of Surgery (T.A.), Division of Cardiovascular Medicine (T. N., E. C., Y.R.S., D.R., C.L.G., Q.S.W, R.J.G.), Department of Pharmacology and Department of Pathology, Immunology, and Microbiology (R.J.G.), Vanderbilt University Medical Center, Nashville, TN
| | - Christian M Shaffer
- From the Division of Cardiovascular Medicine, Davis Heart and Lung Research Institute (B.Z., D.G.W., Z.X., R.H., H.H., S.V., J.L.Z.), Department of Physiology and Cell Biology (B.Z., J.L.Z., M.J.K., M.T.Z.), The Ohio State University, Columbus; Institute of Organ Transplantation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, HuBei, China (B.Z.); Department of Biostatistics (Y.S., M.X., F.E.H.), Division of Clinical Pharmacology, Department of Medicine (D.M.R., C.M.S.), Division of Cardiac Surgery, Department of Surgery (T.A.), Division of Cardiovascular Medicine (T. N., E. C., Y.R.S., D.R., C.L.G., Q.S.W, R.J.G.), Department of Pharmacology and Department of Pathology, Immunology, and Microbiology (R.J.G.), Vanderbilt University Medical Center, Nashville, TN
| | - Cristi L Galindo
- From the Division of Cardiovascular Medicine, Davis Heart and Lung Research Institute (B.Z., D.G.W., Z.X., R.H., H.H., S.V., J.L.Z.), Department of Physiology and Cell Biology (B.Z., J.L.Z., M.J.K., M.T.Z.), The Ohio State University, Columbus; Institute of Organ Transplantation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, HuBei, China (B.Z.); Department of Biostatistics (Y.S., M.X., F.E.H.), Division of Clinical Pharmacology, Department of Medicine (D.M.R., C.M.S.), Division of Cardiac Surgery, Department of Surgery (T.A.), Division of Cardiovascular Medicine (T. N., E. C., Y.R.S., D.R., C.L.G., Q.S.W, R.J.G.), Department of Pharmacology and Department of Pathology, Immunology, and Microbiology (R.J.G.), Vanderbilt University Medical Center, Nashville, TN
| | - Quinn S Wells
- From the Division of Cardiovascular Medicine, Davis Heart and Lung Research Institute (B.Z., D.G.W., Z.X., R.H., H.H., S.V., J.L.Z.), Department of Physiology and Cell Biology (B.Z., J.L.Z., M.J.K., M.T.Z.), The Ohio State University, Columbus; Institute of Organ Transplantation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, HuBei, China (B.Z.); Department of Biostatistics (Y.S., M.X., F.E.H.), Division of Clinical Pharmacology, Department of Medicine (D.M.R., C.M.S.), Division of Cardiac Surgery, Department of Surgery (T.A.), Division of Cardiovascular Medicine (T. N., E. C., Y.R.S., D.R., C.L.G., Q.S.W, R.J.G.), Department of Pharmacology and Department of Pathology, Immunology, and Microbiology (R.J.G.), Vanderbilt University Medical Center, Nashville, TN
| | - Richard J Gumina
- From the Division of Cardiovascular Medicine, Davis Heart and Lung Research Institute (B.Z., D.G.W., Z.X., R.H., H.H., S.V., J.L.Z.), Department of Physiology and Cell Biology (B.Z., J.L.Z., M.J.K., M.T.Z.), The Ohio State University, Columbus; Institute of Organ Transplantation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, HuBei, China (B.Z.); Department of Biostatistics (Y.S., M.X., F.E.H.), Division of Clinical Pharmacology, Department of Medicine (D.M.R., C.M.S.), Division of Cardiac Surgery, Department of Surgery (T.A.), Division of Cardiovascular Medicine (T. N., E. C., Y.R.S., D.R., C.L.G., Q.S.W, R.J.G.), Department of Pharmacology and Department of Pathology, Immunology, and Microbiology (R.J.G.), Vanderbilt University Medical Center, Nashville, TN.
| |
Collapse
|
11
|
Hostrup M, Bangsbo J. Limitations in intense exercise performance of athletes - effect of speed endurance training on ion handling and fatigue development. J Physiol 2016; 595:2897-2913. [PMID: 27673449 DOI: 10.1113/jp273218] [Citation(s) in RCA: 59] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2016] [Accepted: 09/21/2016] [Indexed: 01/10/2023] Open
Abstract
Mechanisms underlying fatigue development and limitations for performance during intense exercise have been intensively studied during the past couple of decades. Fatigue development may involve several interacting factors and depends on type of exercise undertaken and training level of the individual. Intense exercise (½-6 min) causes major ionic perturbations (Ca2+ , Cl- , H+ , K+ , lactate- and Na+ ) that may reduce sarcolemmal excitability, Ca2+ release and force production of skeletal muscle. Maintenance of ion homeostasis is thus essential to sustain force production and power output during intense exercise. Regular speed endurance training (SET), i.e. exercise performed at intensities above that corresponding to maximum oxygen consumption (V̇O2, max ), enhances intense exercise performance. However, most of the studies that have provided mechanistic insight into the beneficial effects of SET have been conducted in untrained and recreationally active individuals, making extrapolation towards athletes' performance difficult. Nevertheless, recent studies indicate that only a few weeks of SET enhances intense exercise performance in highly trained individuals. In these studies, the enhanced performance was not associated with changes in V̇O2, max and muscle oxidative capacity, but rather with adaptations in muscle ion handling, including lowered interstitial concentrations of K+ during and in recovery from intense exercise, improved lactate- -H+ transport and H+ regulation, and enhanced Ca2+ release function. The purpose of this Topical Review is to provide an overview of the effect of SET and to discuss potential mechanisms underlying enhancements in performance induced by SET in already well-trained individuals with special emphasis on ion handling in skeletal muscle.
Collapse
Affiliation(s)
- Morten Hostrup
- Section of Integrated Physiology, Department of Nutrition, Exercise and Sports, University of Copenhagen, Denmark.,Department of Respiratory Research, Bispebjerg University Hospital, Denmark
| | - Jens Bangsbo
- Section of Integrated Physiology, Department of Nutrition, Exercise and Sports, University of Copenhagen, Denmark
| |
Collapse
|
12
|
van der Baan A, Kortekaas KA, van Es E, Meier S, Klautz RJM, Engbers FHM. Sevoflurane-enriched blood cardioplegia: the intramyocardial delivery of a volatile anesthetic. Perfusion 2014; 30:295-301. [DOI: 10.1177/0267659114545666] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Myocardial ischemia/reperfusion injury is a major problem in cardiac surgery, characterized by an enhanced inflammatory response postoperatively. Sevoflurane has anti-inflammatory effects and may attenuate this injury. This study describes a novel approach to using sevoflurane as a local anti-inflammatory drug and not as an anesthetic. Therefore, a pediatric oxygenator with a sevoflurane vaporizer was integrated into the blood cardioplegia system of an adult bypass system. In addition, a gas blender was implemented to regulate pO2 and pCO2 concentrations in the cardioplegia. This proof-of-principle study was tested in vivo and shows that it is feasible to deliver sevoflurane locally while regulating O2 and CO2 concentrations. Moreover, this set-up enables one to use only the specific cardioprotective features of sevoflurane. Inflammatory responses were attenuated, both locally (i.e. the heart) as well as systemically through intramyocardial delivery of sevoflurane.
Collapse
Affiliation(s)
- A van der Baan
- Department of Cardiothoracic Surgery, Leiden University Medical Center, Leiden, The Netherlands
| | - K A Kortekaas
- Department of Cardiothoracic Surgery, Leiden University Medical Center, Leiden, The Netherlands
| | - E van Es
- Department of Cardiothoracic Surgery, Leiden University Medical Center, Leiden, The Netherlands
| | - S Meier
- Department of Anesthesiology, Leiden University Medical Center, Leiden, The Netherlands
| | - R J M Klautz
- Department of Cardiothoracic Surgery, Leiden University Medical Center, Leiden, The Netherlands
| | - F H M Engbers
- Department of Anesthesiology, Leiden University Medical Center, Leiden, The Netherlands
| |
Collapse
|
13
|
Abstract
The field of mitochondrial ion channels has recently seen substantial progress, including the molecular identification of some of the channels. An integrative approach using genetics, electrophysiology, pharmacology, and cell biology to clarify the roles of these channels has thus become possible. It is by now clear that many of these channels are important for energy supply by the mitochondria and have a major impact on the fate of the entire cell as well. The purpose of this review is to provide an up-to-date overview of the electrophysiological properties, molecular identity, and pathophysiological functions of the mitochondrial ion channels studied so far and to highlight possible therapeutic perspectives based on current information.
Collapse
|
14
|
Mogi C, Nakakura T, Okajima F. Role of extracellular proton-sensing OGR1 in regulation of insulin secretion and pancreatic β-cell functions. Endocr J 2014; 61:101-10. [PMID: 24088601 DOI: 10.1507/endocrj.ej13-0380] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
Insulin secretion with respect to pH environments has been investigated for a long time but its mechanism remains largely unknown. Extracellular pH is usually maintained at around 7.4 and, its change has been thought to occur in non-physiological situations. Acidification takes place under ischemic and inflammatory microenvironments, where stimulation of anaerobic glycolysis results in the production of lactic acid. In addition to ionotropic ion channels, such as transient receptor potential V1 (TRPV1) and acid-sensing ion channels (ASICs), metabotropic proton-sensing G protein-coupled receptors (GPCRs) have also been identified recently as proton-sensing machineries. While ionotropic ion channels usually sense strong acidic pH, proton-sensing GPCRs sense pH of 7.6 to 6.0 and have been shown to mediate a variety of biological actions in neutral and mildly acidic pH environments. Studies with receptor knockout mice have revealed that proton-sensing receptors, including ovarian cancer G protein-coupled receptor 1 (OGR1), a proton-sensing GPCRs, play a role in the regulation of insulin secretion and glucose metabolism under physiological conditions. Small molecule 3,5-disubstituted isoxazoles have recently been identified as OGR1 agonists working at neutral pH and have been shown to stimulate pancreatic β-cell differentiation and insulin synthesis. Thus, proton-sensing OGR1 may be an important player for insulin secretion and a potential target for improving β-cell function.
Collapse
Affiliation(s)
- Chihiro Mogi
- Laboratory of Signal Transduction, Institute for Molecular and Cellular Regulation, Gunma University, Maebashi 371-8512, Japan
| | | | | |
Collapse
|
15
|
Regulation of inflammation by extracellular acidification and proton-sensing GPCRs. Cell Signal 2013; 25:2263-71. [PMID: 23917207 DOI: 10.1016/j.cellsig.2013.07.022] [Citation(s) in RCA: 140] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2013] [Accepted: 07/26/2013] [Indexed: 12/14/2022]
Abstract
Under ischemic and inflammatory circumstances, such as allergic airway asthma, rheumatoid arthritis, atherosclerosis, and tumors, extracellular acidification occurs due to the stimulation of anaerobic glycolysis. An acidic microenvironment has been shown to modulate pro-inflammatory or anti-inflammatory responses, including cyclooxygenase-2 (COX-2) expression, prostaglandin synthesis, and cytokine expression, in a variety of cell types, and thereby to exacerbate or ameliorate inflammation. However, molecular mechanisms underlying extracellular acidic pH-induced actions have not been fully understood. Recent studies have shown that ovarian cancer G protein-coupled receptor 1 (OGR1)-family G protein-coupled receptors (GPCRs) can sense extracellular pH or protons, which in turn stimulates intracellular signaling pathways and subsequent diverse cellular responses. In the present review, I discuss extracellular acidic pH-induced inflammatory responses and related responses in inflammatory cells, such as macrophages and neutrophils, and non-inflammatory cells, such as smooth muscle cells and endothelial cells, focusing especially on proton-sensing GPCRs.
Collapse
|
16
|
Parker MD, Boron WF. The divergence, actions, roles, and relatives of sodium-coupled bicarbonate transporters. Physiol Rev 2013; 93:803-959. [PMID: 23589833 PMCID: PMC3768104 DOI: 10.1152/physrev.00023.2012] [Citation(s) in RCA: 208] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
The mammalian Slc4 (Solute carrier 4) family of transporters is a functionally diverse group of 10 multi-spanning membrane proteins that includes three Cl-HCO3 exchangers (AE1-3), five Na(+)-coupled HCO3(-) transporters (NCBTs), and two other unusual members (AE4, BTR1). In this review, we mainly focus on the five mammalian NCBTs-NBCe1, NBCe2, NBCn1, NDCBE, and NBCn2. Each plays a specialized role in maintaining intracellular pH and, by contributing to the movement of HCO3(-) across epithelia, in maintaining whole-body pH and otherwise contributing to epithelial transport. Disruptions involving NCBT genes are linked to blindness, deafness, proximal renal tubular acidosis, mental retardation, and epilepsy. We also review AE1-3, AE4, and BTR1, addressing their relevance to the study of NCBTs. This review draws together recent advances in our understanding of the phylogenetic origins and physiological relevance of NCBTs and their progenitors. Underlying these advances is progress in such diverse disciplines as physiology, molecular biology, genetics, immunocytochemistry, proteomics, and structural biology. This review highlights the key similarities and differences between individual NCBTs and the genes that encode them and also clarifies the sometimes confusing NCBT nomenclature.
Collapse
Affiliation(s)
- Mark D Parker
- Dept. of Physiology and Biophysics, Case Western Reserve University School of Medicine, 10900 Euclid Ave., Cleveland, OH 44106-4970, USA.
| | | |
Collapse
|
17
|
Shibayama J, Taylor TG, Venable PW, Rhodes NL, Gil RB, Warren M, Wende AR, Abel ED, Cox J, Spitzer KW, Zaitsev AV. Metabolic determinants of electrical failure in ex-vivo canine model of cardiac arrest: evidence for the protective role of inorganic pyrophosphate. PLoS One 2013; 8:e57821. [PMID: 23520482 PMCID: PMC3592894 DOI: 10.1371/journal.pone.0057821] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2012] [Accepted: 01/26/2013] [Indexed: 12/14/2022] Open
Abstract
RATIONALE Deterioration of ventricular fibrillation (VF) into asystole or severe bradycardia (electrical failure) heralds a fatal outcome of cardiac arrest. The role of metabolism in the timing of electrical failure remains unknown. OBJECTIVE To determine metabolic factors of early electrical failure in an ex-vivo canine model of cardiac arrest (VF+global ischemia). METHODS AND RESULTS Metabolomic screening was performed in left ventricular biopsies collected before and after 0.3, 2, 5, 10 and 20 min of VF and global ischemia. Electrical activity was monitored via plunge needle electrodes and pseudo-ECG. Four out of nine hearts exhibited electrical failure at 10.1±0.9 min (early-asys), while 5/9 hearts maintained VF for at least 19.7 min (late-asys). As compared to late-asys, early-asys hearts had more ADP, less phosphocreatine, and higher levels of lactate at some time points during VF/ischemia (all comparisons p<0.05). Pre-ischemic samples from late-asys hearts contained ∼25 times more inorganic pyrophosphate (PPi) than early-asys hearts. A mechanistic role of PPi in cardioprotection was then tested by monitoring mitochondrial membrane potential (ΔΨ) during 20 min of simulated-demand ischemia using potentiometric probe TMRM in rabbit adult ventricular myocytes incubated with PPi versus control group. Untreated myocytes experienced significant loss of ΔΨ while in the PPi-treated myocytes ΔΨ was relatively maintained throughout 20 min of simulated-demand ischemia as compared to control (p<0.05). CONCLUSIONS High tissue level of PPi may prevent ΔΨm loss and electrical failure at the early phase of ischemic stress. The link between the two protective effects may involve decreased rates of mitochondrial ATP hydrolysis and lactate accumulation.
Collapse
Affiliation(s)
- Junko Shibayama
- Nora Eccles Harrison Cardiovascular Research and Training Institute, University of Utah, Salt Lake City, Utah, United States of America
| | - Tyson G. Taylor
- Nora Eccles Harrison Cardiovascular Research and Training Institute, University of Utah, Salt Lake City, Utah, United States of America
| | - Paul W. Venable
- Nora Eccles Harrison Cardiovascular Research and Training Institute, University of Utah, Salt Lake City, Utah, United States of America
| | - Nathaniel L. Rhodes
- Nora Eccles Harrison Cardiovascular Research and Training Institute, University of Utah, Salt Lake City, Utah, United States of America
| | - Ryan B. Gil
- Nora Eccles Harrison Cardiovascular Research and Training Institute, University of Utah, Salt Lake City, Utah, United States of America
| | - Mark Warren
- Nora Eccles Harrison Cardiovascular Research and Training Institute, University of Utah, Salt Lake City, Utah, United States of America
| | - Adam R. Wende
- School of Medicine, Division of Endocrinology, Metabolism and Diabetes, Program in Molecular Medicine, University of Utah, Salt Lake City, Utah, United States of America
| | - E. Dale Abel
- School of Medicine, Division of Endocrinology, Metabolism and Diabetes, Program in Molecular Medicine, University of Utah, Salt Lake City, Utah, United States of America
| | - James Cox
- Metabolomics Core Research Facility, University of Utah, Salt Lake City, Utah, United States of America
- Department of Biochemistry, University of Utah, Salt Lake City, Utah, United States of America
| | - Kenneth W. Spitzer
- Nora Eccles Harrison Cardiovascular Research and Training Institute, University of Utah, Salt Lake City, Utah, United States of America
| | - Alexey V. Zaitsev
- Nora Eccles Harrison Cardiovascular Research and Training Institute, University of Utah, Salt Lake City, Utah, United States of America
- * E-mail:
| |
Collapse
|
18
|
Lin BR, Hsieh YH, Jiang C, Tai PC. Escherichia coli Membranes Depleted of SecYEG Elicit SecA-Dependent Ion-Channel Activity but Lose Signal Peptide Specificity. J Membr Biol 2012; 245:747-57. [DOI: 10.1007/s00232-012-9477-8] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2011] [Accepted: 06/30/2012] [Indexed: 11/29/2022]
|
19
|
Parsons MP, Belanger-Willoughby N, Linehan V, Hirasawa M. ATP-sensitive potassium channels mediate the thermosensory response of orexin neurons. J Physiol 2012; 590:4707-15. [PMID: 22802589 DOI: 10.1113/jphysiol.2012.236497] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
High body temperatures are generally associated with somnolence, lethargy, hypophagia and anhedonia. Orexin neurons have been suggested to play a role in such sickness behaviours due to their known functions in appetite, behavioural and autonomic activation. Furthermore, the activity of orexin neurons is inhibited by lipopolysaccharide that induces fever. However, the cellular mechanism(s) underlying this suppression of orexin neurons was unknown. We used patch-clamp recordings in acute rat brain slices to demonstrate that orexin neurons, including those projecting to the wake-promoting locus coeruleus, are inhibited by increasing the ambient temperature by a 2-4°C increment between 26 and 40°C. This effect was not mediated by conventional thermosensing mechanisms but instead involved the activation of ATP-sensitive potassium (KATP) channels. Since KATP channels can also sense energy substrate levels and cellular metabolism, our results suggest that orexin neurons can integrate the state of energy balance and body temperature, and adjust their output accordingly. Thus, the thermosensitivity of orexin neurons may be an important part of maintaining energy homeostasis during hyperthermia and fever.
Collapse
Affiliation(s)
- Matthew P Parsons
- Division of Biomedical Sciences, Faculty of Medicine, Memorial University, 300 Prince Philip Drive, St John's, NL, A1B 3V6, Canada
| | | | | | | |
Collapse
|
20
|
Chao CC, Mihic A, Tsushima RG, Gaisano HY. SNARE protein regulation of cardiac potassium channels and atrial natriuretic factor secretion. J Mol Cell Cardiol 2011; 50:401-7. [DOI: 10.1016/j.yjmcc.2010.11.018] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/21/2010] [Revised: 11/17/2010] [Accepted: 11/19/2010] [Indexed: 01/28/2023]
|
21
|
Ye B, Kroboth SL, Pu JL, Sims JJ, Aggarwal NT, McNally EM, Makielski JC, Shi NQ. Molecular identification and functional characterization of a mitochondrial sulfonylurea receptor 2 splice variant generated by intraexonic splicing. Circ Res 2009; 105:1083-93. [PMID: 19797704 DOI: 10.1161/circresaha.109.195040] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
RATIONALE Cardioprotective pathways may involve a mitochondrial ATP-sensitive potassium (mitoK(ATP)) channel but its composition is not fully understood. OBJECTIVE We hypothesized that the mitoK(ATP) channel contains a sulfonylurea receptor (SUR)2 regulatory subunit and aimed to identify the molecular structure. METHODS AND RESULTS Western blot analysis in cardiac mitochondria detected a 55-kDa mitochondrial SUR2 (mitoSUR2) short form, 2 additional short forms (28 and 68 kDa), and a 130-kDa long form. RACE (Rapid Amplification of cDNA Ends) identified a 1.5-Kb transcript, which was generated by a nonconventional intraexonic splicing (IES) event within the 4th and 29th exons of the SUR2 mRNA. The translated product matched the predicted size of the 55-kDa short form. In a knockout mouse (SUR2KO), in which the SUR2 gene was disrupted, the 130-kDa mitoSUR2 was absent, but the short forms remained expressed. Diazoxide failed to induce increased fluorescence of flavoprotein oxidation in SUR2KO cells, indicating that the diazoxide-sensitive mitoK(ATP) channel activity was associated with 130-kDa-based channels. However, SUR2KO mice displayed similar infarct sizes to preconditioned wild type, suggesting a protective role for the remaining short form-based channels. Heterologous coexpression of the SUR2 IES variant and Kir6.2 in a K(+) transport mutant Escherichia coli strain permitted improved cell growth under acidic pH conditions. The SUR2 IES variant was localized to mitochondria, and removal of a predicted mitochondrial targeting sequence allowed surface expression and detection of an ATP-sensitive current when coexpressed with Kir6.2. CONCLUSIONS We identify a novel SUR2 IES variant in cardiac mitochondria and provide evidence that the variant-based channel can form an ATP-sensitive conductance and may contribute to cardioprotection.
Collapse
Affiliation(s)
- Bin Ye
- Department of Medicine, University of Wisconsin, Madison, WI 53706, USA
| | | | | | | | | | | | | | | |
Collapse
|
22
|
Differential responses of porcine anterior spinal and middle cerebral arteries to carbon dioxide and pH. Crit Care Med 2009; 37:987-92. [PMID: 19237908 DOI: 10.1097/ccm.0b013e3181961330] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
OBJECTIVE Dysfunction of the anterior spinal arteries (ASAs) may induce paresis or paraplegia after thoracoabdominal aortic aneurysm or spine surgery. However, there have been no reports of the effects of CO2 and pH on ASAs. Information on these effects on ASAs might contribute to the perioperative management or critical care of spinal cord function. Thus, we investigated the effects of CO2 and pH on the vasomotor tone of ASAs and the third branch of the middle cerebral artery (bMCA). DESIGN Prospective study of the effects of CO2 and pH on vasomotor response of porcine ASA and bMCA in vitro. SETTING University laboratories. SUBJECTS Porcine heads and spinal cords obtained from a slaughterhouse. INTERVENTION ASAs and bMCAs were isolated, and changes in the intraluminal region of these pressurized arteries ( approximately 80 mm Hg) were observed for 30 minutes after perfusion with a solution saturated with various concentrations of CO2 and pH. MEASUREMENTS AND MAIN RESULTS Respiratory acidosis (pH/Pco2 approximately 7.10-7.15/ approximately 60-80 mm Hg) constricted the ASAs, followed by a partial but gradual decrease in tone, whereas the bMCAs were exclusively dilated. The respiratory alkalosis (pH/Pco2 approximately 7.60/ approximately 20 mm Hg) did not influence ASA tone. Vasoconstriction of the ASAs induced by respiratory acidosis was abolished by removal of the endothelium, but not by N-nitro-L-arginine (1 microM). Respiratory acidosis dilated the ASAs in all preparations treated with ONO-3708 (1 microM), a specific thromboxane A2 receptor antagonist, and OKY-046 (1 microM), a specific thromboxane synthase inhibitor. Metabolic acidosis (pH/Pco2 approximately 7.10/ approximately 40 mm Hg) caused dilation of both bMCAs and ASAs, which was abolished by glibenclamide (1 microM). CONCLUSIONS CO2-induced endothelium-dependent constriction in porcine ASAs through releasing thromboxane A2-like substance(s). Thus, hypercarbia might not be favorable for the perioperative or critical care management of spinal cord function during thoracoabdominal aortic aneurysm and spine surgery.
Collapse
|
23
|
Ainslie PN, Duffin J. Integration of cerebrovascular CO2 reactivity and chemoreflex control of breathing: mechanisms of regulation, measurement, and interpretation. Am J Physiol Regul Integr Comp Physiol 2009; 296:R1473-95. [PMID: 19211719 DOI: 10.1152/ajpregu.91008.2008] [Citation(s) in RCA: 416] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Cerebral blood flow (CBF) and its distribution are highly sensitive to changes in the partial pressure of arterial CO(2) (Pa(CO(2))). This physiological response, termed cerebrovascular CO(2) reactivity, is a vital homeostatic function that helps regulate and maintain central pH and, therefore, affects the respiratory central chemoreceptor stimulus. CBF increases with hypercapnia to wash out CO(2) from brain tissue, thereby attenuating the rise in central Pco(2), whereas hypocapnia causes cerebral vasoconstriction, which reduces CBF and attenuates the fall of brain tissue Pco(2). Cerebrovascular reactivity and ventilatory response to Pa(CO(2)) are therefore tightly linked, so that the regulation of CBF has an important role in stabilizing breathing during fluctuating levels of chemical stimuli. Indeed, recent reports indicate that cerebrovascular responsiveness to CO(2), primarily via its effects at the level of the central chemoreceptors, is an important determinant of eupneic and hypercapnic ventilatory responsiveness in otherwise healthy humans during wakefulness, sleep, and exercise and at high altitude. In particular, reductions in cerebrovascular responsiveness to CO(2) that provoke an increase in the gain of the chemoreflex control of breathing may underpin breathing instability during central sleep apnea in patients with congestive heart failure and on ascent to high altitude. In this review, we summarize the major factors that regulate CBF to emphasize the integrated mechanisms, in addition to Pa(CO(2)), that control CBF. We discuss in detail the assessment and interpretation of cerebrovascular reactivity to CO(2). Next, we provide a detailed update on the integration of the role of cerebrovascular CO(2) reactivity and CBF in regulation of chemoreflex control of breathing in health and disease. Finally, we describe the use of a newly developed steady-state modeling approach to examine the effects of changes in CBF on the chemoreflex control of breathing and suggest avenues for future research.
Collapse
Affiliation(s)
- Philip N Ainslie
- Department of Physiology, University of Otago, Dunedin, New Zealand.
| | | |
Collapse
|
24
|
Abstract
Acidosis is a noxious condition associated with inflammation, ischaemia or defective acid containment. As a consequence, acid sensing has evolved as an important property of afferent neurons with unmyelinated and thinly myelinated nerve fibres. Protons evoke multiple currents in primary afferent neurons, which are carried by several acid-sensitive ion channels. Among these, acid-sensing ion channels (ASICs) and transient receptor potential (TRP) vanilloid-1 (TRPV1) ion channels have been most thoroughly studied. ASICs survey moderate decreases in extracellular pH, whereas TRPV1 is activated only by severe acidosis resulting in pH values below 6. Two-pore-domain K(+) (K(2P)) channels are differentially regulated by small deviations of extra- or intracellular pH from physiological levels. Other acid-sensitive channels include TRPV4, TRPC4, TRPC5, TRPP2 (PKD2L1), ionotropic purinoceptors (P2X), inward rectifier K(+) channels, voltage-activated K(+) channels, L-type Ca(2+) channels, hyperpolarization-activated cyclic nucleotide gated channels, gap junction channels, and Cl(-) channels. In addition, acid-sensitive G protein coupled receptors have also been identified. Most of these molecular acid sensors are expressed by primary sensory neurons, although to different degrees and in various combinations. Emerging evidence indicates that many of the acid-sensitive ion channels and receptors play a role in acid sensing, acid-induced pain and acid-evoked feedback regulation of homeostatic reactions. The existence and apparent redundancy of multiple pH surveillance systems attests to the concept that acid-base regulation is a vital issue for cell and tissue homeostasis. Since upregulation and overactivity of acid sensors appear to contribute to various forms of chronic pain, acid-sensitive ion channels and receptors are considered as targets for novel analgesic drugs. This approach will only be successful if the pathological implications of acid sensors can be differentiated pharmacologically from their physiological function.
Collapse
Affiliation(s)
- Peter Holzer
- Research Unit of Translational Neurogastroenterology, Institute of Experimental and Clinical Pharmacology, Medical University of Graz, Universitätsplatz 4, 8010, Graz, Austria.
| |
Collapse
|
25
|
Kawano T, Tanaka K, Nazari H, Oshita S, Takahashi A, Nakaya Y. The effects of extracellular pH on vasopressin inhibition of ATP-sensitive K+ channels in vascular smooth muscle cells. Anesth Analg 2007; 105:1714-9, table of contents. [PMID: 18042872 DOI: 10.1213/01.ane.0000290334.91624.2f] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
BACKGROUND Arginine vasopressin (AVP) inhibits ATP-sensitive potassium (K(ATP)) channels and may help to restore vascular tone in patients with vasodilatory shock. In the present study, we investigated whether extracellular acidification modifies the inhibition of vascular K(ATP) channels by AVP. METHODS We used a cell-attached patch-clamp configuration to investigate the effects of extracellular pH (pH(o)) on AVP-K(ATP) channel interaction in rat aortic smooth muscle cells. RESULTS Bath application of AVP significantly inhibited extracellular acidification (pH(o) = 6.5)-induced K(ATP) channel activity in a concentration-dependent manner, with an half-maximal inhibitory concentration (IC50) value of 16.8 pM. Furthermore, bath application of AVP significantly inhibited pinacidil-induced K(ATP) channel activity at mild (pH(o) = 7.0) and severe (pH(o) = 6.5) extracellular acidification, with IC50 values of 266.7 and 21.4 pM, respectively, but failed to significantly inhibit at normal pH (pH(o) = 7.4) or under alkalosis (pH(o) = 9.0). Augmentation of AVP inhibition of vascular K(ATP) channels during extracellular acidification was eliminated by pretreatment with OPC-21268, a specific blocker of the V1 receptor, but not by a V2 blocker, OPC-31260. AVP-induced inhibition was also suppressed by pretreatment with a protein kinase C inhibitor, calphostin C. CONCLUSIONS Our results suggest that AVP inhibits extracellular acidification-induced vascular K(ATP) channel activity, and that the inhibitory effects of AVP on vascular K(ATP) channels are enhanced by extracellular acidification via the V1 receptor-protein kinase C cell-signaling pathway. The potent inhibition of vascular K(ATP) channels by AVP under acidic conditions may make it suitable for management of vasodilatory shock.
Collapse
Affiliation(s)
- Takashi Kawano
- Department of Anesthesiology, Institute of Health Biosciences, The University of Tokushima Graduate School, Tokushima, Japan.
| | | | | | | | | | | |
Collapse
|
26
|
Hughes BA, Swaminathan A. Modulation of the Kir7.1 potassium channel by extracellular and intracellular pH. Am J Physiol Cell Physiol 2007; 294:C423-31. [PMID: 18094146 DOI: 10.1152/ajpcell.00393.2007] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Inwardly rectifying K(+) (K(ir)) channels in the apical membrane of the retinal pigment epithelium (RPE) contribute to extracellular K(+) homeostasis in the distal retina by mediating K(+) secretion. Multiple lines of evidence suggest that these channels are composed of Kir7.1. Previously, we showed that native K(ir) channels in bovine RPE are modulated by changes in intracellular pH in the physiological range. In the present study, we used the Xenopus laevis oocyte expression system to investigate the pH dependence of cloned human Kir7.1 channels and several point mutants involving histidine residues in the NH(2) and COOH termini. Kir7.1 channels were inhibited by strong extracellular acidification and modulated by intracellular pH in a biphasic manner, with maximal activity at about intracellular pH (pH(i)) 7.0 and inhibition by acidification or alkalinization. Replacement of histidine 26 (H26) in the NH(2) terminus with alanine eliminated the requirement of protons for channel activity and increased sensitivity to proton-induced inhibition, resulting in maximal channel activity at alkaline pH(i) and smaller whole cell currents at resting pH(i) compared with wild-type Kir7.1. When H26 was replaced with arginine, the pH(i) sensitivity profile was similar to that of the H26A mutant but with the pK(a) shifted to a more acidic value, giving rise to whole cell current amplitude at resting pH(i) that was comparable to that of wild-type Kir7.1. These results indicate that Kir7.1 channels are modulated by intracellular protons by diverse mechanisms and suggest that H26 is important for channel activation at physiological pH(i) and that it influences an unidentified proton-induced inhibitory mechanism.
Collapse
Affiliation(s)
- Bret A Hughes
- Department of Ophthalmology, University of Michigan, Ann Arbor, MI 48105, USA.
| | | |
Collapse
|
27
|
Shi Y, Cui N, Shi W, Jiang C. A short motif in Kir6.1 consisting of four phosphorylation repeats underlies the vascular KATP channel inhibition by protein kinase C. J Biol Chem 2007; 283:2488-94. [PMID: 18048350 DOI: 10.1074/jbc.m708769200] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Vascular ATP-sensitive K(+) channels are inhibited by multiple vasoconstricting hormones via the protein kinase C (PKC) pathway. However, the molecular substrates for PKC phosphorylation remain unknown. To identify the PKC sites, Kir6.1/SUR2B and Kir6.2/SUR2B were expressed in HEK293 cells. Following channel activation by pinacidil, the catalytic fragment of PKC inhibited the Kir6.1/SUR2B currents but not the Kir6.2/SUR2B currents. Phorbol 12-myristate 13-acetate (a PKC activator) had similar effects. Using Kir6.1-Kir6.2 chimeras, two critical protein domains for the PKC-dependent channel inhibition were identified. The proximal N terminus of Kir6.1 was necessary for channel inhibition. Because there was no PKC phosphorylation site in the N-terminal region, our results suggest its potential involvement in channel gating. The distal C terminus of Kir6.1 was crucial where there are several consensus PKC sites. Mutation of Ser-354, Ser-379, Ser-385, Ser-391, or Ser-397 to nonphosphorylatable alanine reduced PKC inhibition moderately but significantly. Combined mutations of these residues had greater effects. The channel inhibition was almost completely abolished when 5 of them were jointly mutated. In vitro phosphorylation assay showed that 4 of the serine residues were necessary for the PKC-dependent (32)P incorporation into the distal C-terminal peptides. Thus, a motif containing four phosphorylation repeats is identified in the Kir6.1 subunit underlying the PKC-dependent inhibition of the Kir6.1/SUR2B channel. The presence of the phosphorylation motif in Kir6.1, but not in its close relative Kir6.2, suggests that the vascular K(ATP) channel may have undergone evolutionary optimization, allowing it to be regulated by a variety of vasoconstricting hormones and neurotransmitters.
Collapse
Affiliation(s)
- Yun Shi
- Department of Biology, Georgia State University, 33 Gilmer Street, Atlanta, GA 30302-4010, USA
| | | | | | | |
Collapse
|
28
|
Effects of intracellular MgADP and acidification on the inhibition of cardiac sarcolemmal ATP-sensitive potassium channels by propofol. J Anesth 2007; 21:472-9. [PMID: 18008114 DOI: 10.1007/s00540-007-0551-9] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2006] [Accepted: 06/29/2007] [Indexed: 10/22/2022]
Abstract
PURPOSE Propofol inhibits adenosine triphosphate-sensitive potassium (K(ATP)) channels, which may result in the blocking of ischemic preconditioning in the heart. During cardiac ischemia, sarcolemmal K(ATP) channel activity is regulated by the increased levels of cytosolic metabolites, such as adenosine diphosphate (ADP) and protons. However, it remains unclear whether these cytosolic metabolites modulate the inhibitory action of propofol. The aim of this study was to investigate the effects of intracellular MgADP and acidification on K(ATP) channel inhibition by propofol. METHODS We used inside-out patch-clamp configurations to investigate the effects of propofol on the activities of recombinant cardiac sarcolemmal K(ATP) channels, which are reassociated by expressed subunits, sulfonylurea receptor (SUR) 2A, and inwardly rectifying potassium channels (Kir6.2). RESULTS In the absence of MgADP, propofol inhibited the SUR2A/Kir6.2 channel currents in a concentration-dependent manner, and an IC(50) of 78 microM. Increasing the intracellular MgADP concentrations to 0.1 and 0.3 mM markedly attenuated the inhibitory potency of propofol, and shifted the IC(50) to 183 and 265 microM, respectively. Moreover, decreasing the intracellular pH from 7.4 to 6.5 attenuated the inhibitory potency of propofol, and shifted the IC(50) to 277 microM. In addition, propofol-induced inhibition of truncated Kir6.2DeltaC36 currents, which form a functional channel without SUR2A, was not affected by an increase in intracellular MgADP. However, intracellular acidification (pH 6.5) significantly reduced the propofol sensitivity of Kir6.2DeltaC36 channels. CONCLUSION Our results demonstrated that the existence of intracellular MgADP and protons attenuated the direct inhibitory potency of propofol on recombinant cardiac sarcolemmal K(ATP) channels, via SUR2A and Kir6.2 subunits, respectively.
Collapse
|
29
|
Wang R, Zhang X, Cui N, Wu J, Piao H, Wang X, Su J, Jiang C. Subunit-stoichiometric evidence for kir6.2 channel gating, ATP binding, and binding-gating coupling. Mol Pharmacol 2007; 71:1646-56. [PMID: 17369308 DOI: 10.1124/mol.106.030528] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
ATP-sensitive K(+) channels are gated by intracellular ATP, allowing them to couple intermediary metabolism to cellular excitability, whereas the gating mechanism remains unclear. To understand subunit stoichiometry for the ATP-dependent channel gating, we constructed tandem-multimeric Kir6.2 channels by selective disruption of the binding or gating mechanism in certain subunits. Stepwise disruptions of channel gating caused graded losses in ATP sensitivity and increases in basal P(open), with no effect on maximum ATP inhibition. Prevention of ATP binding lowered the ATP sensitivity and maximum inhibition without affecting basal P(open). The ATP-dependent gating required a minimum of two functional subunits. Two adjacent subunits are more favorable for ATP binding than two diagonal ones. Subunits showed negative cooperativity in ATP binding and positive cooperativity in channel gating. Joint disruptions of the binding and gating mechanisms in the same or alternate subunits of a concatemer revealed that both intra- and intersubunit couplings contributed to channel gating, although the binding-gating coupling preferred the intrasubunit to intersubunit configuration within the C terminus. No such preference was found between the C and N termini. These phenomena are well-described with the operational model used widely for ligand-receptor interactions.
Collapse
Affiliation(s)
- Runping Wang
- Department of Biology, Georgia State University, Atlanta, GA 30302-4010, USA
| | | | | | | | | | | | | | | |
Collapse
|
30
|
Lin BR, Gierasch LM, Jiang C, Tai PC. Electrophysiological studies in Xenopus oocytes for the opening of Escherichia coli SecA-dependent protein-conducting channels. J Membr Biol 2007; 214:103-13. [PMID: 17530158 PMCID: PMC2896742 DOI: 10.1007/s00232-006-0079-1] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2006] [Revised: 11/16/2006] [Indexed: 11/25/2022]
Abstract
Protein translocation in Escherichia coli requires protein-conducting channels in cytoplasmic membranes to allow precursor peptides to pass through with adenosine triphosphate (ATP) hydrolysis. Here, we report a novel, sensitive method that detects the opening of the SecA-dependent protein-conducting channels at the nanogram level. E. coli inverted membrane vesicles were injected into Xenopus oocytes, and ionic currents were recorded using the two-electrode voltage clamp. Currents were observed only in the presence of E. coli SecA in conjunction with E. coli membranes. Observed currents showed outward rectification in the presence of KCl as permeable ions and were significantly enhanced by coinjection with the precursor protein proOmpA or active LamB signal peptide. Channel activity was blockable with sodium azide or adenylyl 5'-(beta,gamma-methylene)-diphosphonate, a nonhydrolyzable ATP analogue, both of which are known to inhibit SecA protein activity. Endogenous oocyte precursor proteins also stimulated ion current activity and can be inhibited by puromycin. In the presence of puromycin, exogenous proOmpA or LamB signal peptides continued to enhance ionic currents. Thus, the requirement of signal peptides and ATP hydrolysis for the SecA-dependent currents resembles biochemical protein translocation assay with E. coli membrane vesicles, indicating that the Xenopus oocyte system provides a sensitive assay to study the role of Sec and precursor proteins in the formation of protein-conducting channels using electrophysiological methods.
Collapse
Affiliation(s)
- Bor-Ruei Lin
- Department of Biology, Georgia State University, 24 Peachtree Center Avenue, Atlanta, GA 30303, USA
| | - Lila M. Gierasch
- Departments of Biochemistry and Molecular Biology and of Chemistry, University of Massachusetts, 710 N. Pleasant Street, Amherst, MA 01003, USA
| | - Chun Jiang
- Department of Biology, Georgia State University, 24 Peachtree Center Avenue, Atlanta, GA 30303, USA
| | - Phang C. Tai
- Department of Biology, Georgia State University, 24 Peachtree Center Avenue, Atlanta, GA 30303, USA
| |
Collapse
|
31
|
Wang R, Su J, Zhang X, Shi Y, Cui N, Onyebuchi VA, Jiang C. Kir6.2 channel gating by intracellular protons: subunit stoichiometry for ligand binding and channel gating. J Membr Biol 2007; 213:155-64. [PMID: 17468960 DOI: 10.1007/s00232-006-0038-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2006] [Accepted: 09/17/2006] [Indexed: 10/23/2022]
Abstract
The adenosine triphosphate-sensitive K(+) (K(ATP)) channels are gated by several metabolites, whereas the gating mechanism remains unclear. Kir6.2, a pore-forming subunit of the K(ATP) channels, has all machineries for ligand binding and channel gating. In Kir6.2, His175 is the protonation site and Thr71 and Cys166 are involved in channel gating. Here, we show how individual subunits act in proton binding and channel gating by selectively disrupting functional subunits using these residues. All homomeric dimers and tetramers showed pH sensitivity similar to the monomeric channels. Concatenated construction of wild type with disrupted subunits revealed that none of these residues had a dominant-negative effect on the proton-dependent channel gating. Subunit action in proton binding was almost identical to that for channel gating involving Cys166, suggesting a one-to-one coupling from the C terminus to the M2 helix. This was significantly different from the effect of T71Y heteromultimers, suggesting distinct contributions of M1 and M2 helices to channel gating. Subunits underwent concerted rather than independent action. Two wild-type subunits appeared to act as a functional dimer in both cis and trans configurations. The understanding of K(ATP) channel gating by intracellular pH has a profound impact on cellular responses to metabolic stress as a significant drop in intracellular pH is more frequently seen under a number of physiological and pathophysiological conditions than a sole decrease in intracellular ATP levels.
Collapse
Affiliation(s)
- Runping Wang
- Department of Biology, Georgia State University, 24 Peachtree Center Avenue, Atlanta, Georgia, 30303-4010, USA
| | | | | | | | | | | | | |
Collapse
|
32
|
Manning Fox JE, Karaman G, Wheeler MB. Alkali pH directly activates ATP-sensitive K+ channels and inhibits insulin secretion in beta-cells. Biochem Biophys Res Commun 2006; 350:492-7. [PMID: 17011513 DOI: 10.1016/j.bbrc.2006.09.084] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2006] [Accepted: 09/19/2006] [Indexed: 11/20/2022]
Abstract
Glucose stimulation of pancreatic beta-cells is reported to lead to sustained alkalization, while extracellular application of weak bases is reported to inhibit electrical activity and decrease insulin secretion. We hypothesize that beta-cell K(ATP) channel activity is modulated by alkaline pH. Using the excised patch-clamp technique, we demonstrate a direct stimulatory action of alkali pH on recombinant SUR1/Kir6.2 channels due to increased open probability. Bath application of alkali pH similarly activates native islet beta-cell K(ATP) channels, leading to an inhibition of action potentials, and hyperpolarization of membrane potential. In situ pancreatic perfusion confirms that these cellular effects of alkali pH are observable at a functional level, resulting in decreases in both phase 1 and phase 2 glucose-stimulated insulin secretion. Our data are the first to report a stimulatory effect of a range of alkali pH on K(ATP) channel activity and link this to downstream effects on islet beta-cell function.
Collapse
Affiliation(s)
- Jocelyn E Manning Fox
- Department of Physiology, 3352 Medical Sciences Building, 1 King's College Circle, University of Toronto, Toronto, Ont., Canada.
| | | | | |
Collapse
|
33
|
Rojas A, Wu J, Wang R, Jiang C. Gating of the ATP-sensitive K+ channel by a pore-lining phenylalanine residue. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2006; 1768:39-51. [PMID: 16970907 DOI: 10.1016/j.bbamem.2006.06.027] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/17/2006] [Revised: 06/27/2006] [Accepted: 06/28/2006] [Indexed: 11/26/2022]
Abstract
ATP-sensitive K(+) (K(ATP)) channels are gated by intracellular ATP, proton and phospholipids. The pore-forming Kir6.2 subunit has all essential machineries for channel gating by these ligands. It is known that channel gating involves the inner helix bundle of crossing in which a phenylalanine residue (Phe168) is found in the TM2 at the narrowest region of the ion-conduction pathway in the Kir6.2. Here we present evidence that Phe168-Kir6.2 functions as an ATP- and proton-activated gate via steric hindrance and hydrophobic interactions. Site-specific mutations of Phe168 to a small amino acid resulted in losses of the ATP- and proton-dependent gating, whereas the channel gating was well maintained after mutation to a bulky tryptophan, supporting the steric hindrance effect. The steric hindrance effect, though necessary, was insufficient for the gating, as mutating Phe168 to a bulky hydrophilic residue severely compromised the channel gating. Single-channel kinetics of the F168W mutant resembled the wild-type channel. Small residues increased P(open), and displayed long-lasting closures and long-lasting openings. Kinetic modeling showed that these resulted from stabilization of the channel to open and long-lived closed states, suggesting that a bulky and hydrophobic residue may lower the energy barrier for the switch between channel openings and closures. Thus, it is likely that the Phe168 acts as not only a steric hindrance gate but also potentially a facilitator of gating transitions in the Kir6.2 channel.
Collapse
Affiliation(s)
- Asheebo Rojas
- Department of Biology, Georgia State University, 24 Peachtree Center Avenue, Atlanta, GA 30302-4010, USA
| | | | | | | |
Collapse
|
34
|
Oyamada Y, Yamaguchi K, Murai M, Ishizaka A, Okada Y. Potassium channels in the central control of breathing. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2006; 580:339-44; discussion 351-9. [PMID: 16683741 DOI: 10.1007/0-387-31311-7_52] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/09/2023]
Affiliation(s)
- Yoshitaka Oyamada
- Department of Pulmonary Medicine, School of Medicine, Keio University, Shinjuku, Tokyo, Japan
| | | | | | | | | |
Collapse
|
35
|
Kang Y, Ng B, Leung YM, He Y, Xie H, Lodwick D, Norman RI, Tinker A, Tsushima RG, Gaisano HY. Syntaxin-1A actions on sulfonylurea receptor 2A can block acidic pH-induced cardiac K(ATP) channel activation. J Biol Chem 2006; 281:19019-28. [PMID: 16672225 DOI: 10.1074/jbc.m513160200] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
During cardiac ischemia, ATP stores are depleted, and cardiomyocyte intracellular pH lowers to <7.0. The acidic pH acts on the Kir6.2 subunit of K(ATP) channels to reduce its sensitivity to ATP, causing channel opening. We recently reported that syntaxin-1A (Syn-1A) binds nucleotide binding folds (NBF)-1 and NBF2 of sulfonylurea receptor 2A (SUR2A) to inhibit channel activity (Kang, Y., Leung, Y. M., Manning-Fox, J. E., Xia, F., Xie, H., Sheu, L., Tsushima, R. G., Light, P. E., and Gaisano, H. Y. (2004) J. Biol. Chem. 279, 47125-47131). Here, we examined Syn-1A actions on SUR2A to influence the pH regulation of cardiac K(ATP) channels. K(ATP) channel currents from inside-out patches excised from Kir6.2/SUR2A expressing HEK293 cells and freshly isolated cardiac myocytes were increased by reducing intracellular pH from 7.4 to 6.8, which could be blocked by increasing concentrations of Syn-1A added to the cytoplasmic surface. Syn-1A had no effect on C-terminal truncated Kir6.2 (Kir6.2-deltaC26) channels expressed in TSA cells without the SUR subunit. In vitro binding and co-immunoprecipitation studies show that Syn-1A binding to SUR2A or its NBF-1 and NBF-2 domain proteins increased progressively as pH was reduced from 7.4 to 6.0. The enhancement of Syn-1A binding to SUR2A by acidic pH was further regulated by Mg2+ and ATP. Therefore, pH regulates Kir.6.2/SUR2A channels not only by its direct actions on the Kir6.2 subunit but also by modulation of Syn-1A binding to SUR2A. The increased Syn-1A binding to the SUR2A at acidic pH would assert some inhibition of the K(ATP) channels, which may serve as a "brake" to temper the fluctuation of low pH-induced K(ATP) channel opening that could induce fatal reentrant arrhythmias.
Collapse
Affiliation(s)
- Youhou Kang
- Department of Medicine, University of Toronto, Toronto, Canada M5S 1A8
| | | | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Wu SN, Chang HD. Diethyl pyrocarbonate, a histidine-modifying agent, directly stimulates activity of ATP-sensitive potassium channels in pituitary GH(3) cells. Biochem Pharmacol 2006; 71:615-623. [PMID: 16375866 DOI: 10.1016/j.bcp.2005.11.016] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2005] [Revised: 11/12/2005] [Accepted: 11/16/2005] [Indexed: 10/25/2022]
Abstract
The ATP-sensitive K(+) (K(ATP)) channels are composed of sulfonylurea receptor and inwardly rectifying K(+) channel (Kir6.2) subunit. These channels are regulated by intracellular ADP/ATP ratio and play a role in cellular metabolism. Diethyl pyrocarbonate (DEPC), a histidine-specific alkylating reagent, is known to modify the histidine residues of the structure of proteins. The objective of this study was to determine whether DEPC modifies K(ATP)-channel activity in pituitary GH(3) cells. Steady-state fluctuation analyses of macroscopic K(+) current at -120 mV produced power spectra that could be fitted with a single Lorentzian curve in these cells. The time constants in the presence of DEPC were increased. Consistent with fluctuation analyses, the mean open time of K(ATP)-channels was significantly increased during exposure to DEPC. However, DEPC produced no change in single-channel conductance, despite the ability of this compound to enhance K(ATP)-channel activity in a concentration-dependent manner with an EC(50) value of 16 microM. DEPC-stimulated K(ATP)-channel activity was attenuated by pretreatment with glibenclamide. In current-clamp configuration, DEPC decreased the firing of action potentials in GH(3) cells. A further application of glibenclamide reversed DEPC-induced inhibition of spontaneous action potentials. Intracellullar Ca(2+) measurements revealed the ability of DEPC to decrease Ca(2+) oscillations in GH(3) cells. Simulation studies also demonstrated that the increased conductance of K(ATP)-channels used to mimic DEPC actions reduced the frequency of spontaneous action potentials and fluctuation of intracellular Ca(2+). The results indicate that chemical modification with DEPC enhances K(ATP)-channel activity and influences functional activities of pituitary GH(3) cells.
Collapse
Affiliation(s)
- Sheng-Nan Wu
- Department of Physiology, National Cheng Kung University Medical College, No 1, University Road, Tainan 70101, Taiwan.
| | | |
Collapse
|
37
|
Cui N, Li L, Wang X, Shi Y, Shi W, Jiang C. Elimination of allosteric modulation of myocardial KATP channels by ATP and protons in two Kir6.2 polymorphisms found in sudden cardiac death. Physiol Genomics 2006; 25:105-15. [PMID: 16403845 DOI: 10.1152/physiolgenomics.00106.2005] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
The major cause of sudden cardiac death (SCD) is ventricular arrhythmias due to unstable myocardial electrical activity in which the ATP-sensitive K+ (KATP) channels play a role. Genetic disruption of these channels predisposes the myocardium to arrhythmias. Two point mutations in the Kir6.2 subunit are found in SCD with acute myocardial infarction. Here we show evidence for the functional consequences of the P266T and R371H variants. Baseline single-channel properties, expression density, and channel modulations were studied in patch clamp. We focused on channel modulations by intracellular ATP and protons, as the concentration of these two important KATP channel regulators changes widely with hypoxic ischemia. We found that both variants expressed functional currents even though they occur at two highly conserved regions. The open state probability of P266T was twice as high as the wild-type (WT) channel, whereas its channel density was only approximately 20% of the WT channel. Although the outward current was not affected by these two mutations at neutral pH, it was approximately 20% lower at acidic pH in the P266T than in the WT channel. Both P266T and R371H mutations significantly reduced ATP sensitivity and increased pH sensitivity. More dramatically, allosteric regulation by intracellular ATP and protons was almost completely eliminated in the polymorphic P266T and R371H channels. Such an abnormality was seen in both inward and outward currents. Given the importance and beneficial effects of allosteric regulation in cellular responses to metabolic stress, the loss of such a regulatory mechanism in the P266T and R371H variants appears consistent with the adverse consequences occurring during acute myocardial infarction in patients.
Collapse
Affiliation(s)
- Ningren Cui
- Department of Biology, Georgia State University, Atlanta, Georgia 30302-4010, USA
| | | | | | | | | | | |
Collapse
|
38
|
Komaru T, Shirato K. Myocardial ischemia and coronary microvascular vasomotion: the impact of hypercholesterolemia. Future Cardiol 2005; 1:637-47. [PMID: 19804104 DOI: 10.2217/14796678.1.5.637] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Coronary microvascular dilation in response to myocardial ischemia is an important defense mechanism for minimizing heart injury. Since myocardial ischemia is an intense stimulus for a living body, many biologic signals that affect the vascular tone are activated. Recent evidence demonstrated that among them, ischemic myocardium-derived factors play dominant roles as mediators of ischemic vasodilation, and that ischemic myocardium releases vasoconstrictor signals simultaneously. Hypercholesterolemia, a major risk factor for coronary atherosclerosis, is known to produce microvascular dysfunction, although it does not produce atherosclerotic lesions at the microvascular level. A recent bioassay study demonstrated that the vasodilator signal transduction from the myocardium to coronary microvessels is severely impaired in hypercholesterolemia. It is likely that redox-sensitive mechanisms play a major role in the impairment of the defensive responses. The present understanding of the mechanism of ischemic vasodilation and the impact of hypercholesterolemia on coronary microvascular regulation shall be discussed in this review.
Collapse
Affiliation(s)
- Tatsuya Komaru
- Tohoku University Graduate School of Medicine, Department of Cardiovascular Medicine, 1-1 Seiryo-machi, Aoba-ku, Sendai 980-8574, Japan.
| | | |
Collapse
|
39
|
Wang R, Rojas A, Wu J, Piao H, Adams CY, Xu H, Shi Y, Wang Y, Jiang C. Determinant role of membrane helices in K ATP channel gating. J Membr Biol 2005; 204:1-10. [PMID: 16007498 DOI: 10.1007/s00232-005-0741-z] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2004] [Revised: 03/10/2005] [Indexed: 11/25/2022]
Abstract
The ATP-sensitive K(+) (K(ATP)) channels couple chemical signals to cellular activity, in which the control of channel opening and closure (i.e., channel gating) is crucial. Transmembrane helices play an important role in channel gating. Here we report that the gating of Kir6.2, the core subunit of pancreatic and cardiac K(ATP) channels, can be switched by manipulating the interaction between two residues located in transmembrane domains (TM) 1 and 2 of the channel protein. The Kir6.2 channel is gated by ATP and proton, which inhibit and activate the channel, respectively. The channel gating involves two residues, namely, Thr71 and Cys166, located at the interface of the TM1 and TM2. Creation of electrostatic attraction between these sites reverses the channel gating, which makes the ATP an activator and proton an inhibitor of the channel. Electrostatic repulsion with two acidic residues retains or even enhances the wild-type channel gating. A similar switch of the pH-dependent channel gating was observed in the Kir2.1 channel, which is normally pH- insensitive. Thus, the manner in which the TM1 and TM2 helices interact appears to determine whether the channels are open or closed following ligand binding.
Collapse
Affiliation(s)
- R Wang
- Department of Biology, Georgia State University, 24 Peachtree Center Avenue, Atlanta, GA 30303-4010, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Tomura H, Wang JQ, Komachi M, Damirin A, Mogi C, Tobo M, Kon J, Misawa N, Sato K, Okajima F. Prostaglandin I(2) production and cAMP accumulation in response to acidic extracellular pH through OGR1 in human aortic smooth muscle cells. J Biol Chem 2005; 280:34458-64. [PMID: 16087674 DOI: 10.1074/jbc.m505287200] [Citation(s) in RCA: 80] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
Ovarian cancer G-protein-coupled receptor 1 (OGR1) and GPR4 have recently been identified as proton-sensing or extracellular pH-responsive G-protein-coupled receptors stimulating inositol phosphate production and cAMP accumulation, respectively. In the present study, we found that OGR1 and GPR4 mRNAs were expressed in human aortic smooth muscle cells (AoSMCs). Acidic extracellular pH induced inositol phosphate production, a transient increase in intracellular Ca(2+) concentration ([Ca(2+)](i)), and cAMP accumulation in these cells. When small interfering RNAs (siRNAs) targeted for OGR1 and GPR4 were transfected to the cells, the acid-induced inositol phosphate production and [Ca(2+)](i) increase were markedly inhibited by the OGR1 siRNA but not by the GPR4 siRNA. Unexpectedly, the acid-induced cAMP accumulation was also largely inhibited by OGR1 siRNA but only slightly by GPR4 siRNA. Acidic extracellular pH also stimulated prostaglandin I2 (PGI(2)) production, which was again inhibited by OGR1 siRNA. The specific inhibitors for extracellular signal-regulated kinase kinase and cyclooxygenase attenuated the acid-induced PGI(2) production and cAMP accumulation without changes in the inositol phosphate production. A specific inhibitor of phospholipase C also inhibited the acid-induced cAMP accumulation. In conclusion, OGR1 is a major receptor involved in the extracellular acid-induced stimulation of PGI(2) production and cAMP accumulation in AoSMCs. The cAMP accumulation may occur through OGR1-mediated stimulation of the phospholipase C/cyclooxygenase/PGI(2) pathway.
Collapse
Affiliation(s)
- Hideaki Tomura
- Laboratory of Signal Transduction, Institute for Molecular and Cellular Regulation, Gunma University, Maebashi 371-8512, Japan.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Rohra DK, Sharif HM, Zubairi HS, Sarfraz K, Ghayur MN, Gilani AH. Acidosis-induced relaxation of human internal mammary artery is due to activation of ATP-sensitive potassium channels. Eur J Pharmacol 2005; 514:175-81. [PMID: 15910804 DOI: 10.1016/j.ejphar.2005.02.041] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2004] [Revised: 02/14/2005] [Accepted: 02/24/2005] [Indexed: 11/17/2022]
Abstract
Metabolic acidosis is associated with various clinical situations including diabetes mellitus and renal diseases. The aim of this study was to investigate the effects of acidosis on the resting as well as precontracted human left internal mammary artery. The vessels were obtained from the patients undergoing coronary artery bypass grafting surgery at The Aga Khan University Hospital, Karachi. Left internal mammary artery was cut into rings and isometric tension recording experiments were performed. Decrease in pH of the bathing solution from 7.4 to 6.8 had no effect on the resting tension of left internal mammary artery, whereas, acidic pH markedly relaxed the contractions to 24.8 mM KCl and 300 nM phenylephrine. Interestingly, when the KCl- or phenylephrine-contracted rings were treated with 3 microM glibenclamide; an inhibitor of ATP-sensitive potassium (K(ATP)) channels, the relaxant effect of acidosis was abolished. Similarly, acidosis failed to cause relaxation of 100 nM endothelin-1-induced contraction in Ca2+-free bathing solution or in the presence of a voltage-dependent Ca2+ channel inhibitor, verapamil (10 microM), whereas, endothelin-1-induced contraction was attenuated by acidosis in Ca2+-containing normal solution. From all these data, it is concluded that under the acidic pH conditions, opening of K(ATP) channels occurs; resulting in the hyperpolarization, decrease in Ca2+ influx via voltage-dependent Ca2+ channels and subsequent relaxation of human left internal mammary artery.
Collapse
Affiliation(s)
- Dileep Kumar Rohra
- Department of Biological and Biomedical Sciences, Faculty of Health Sciences, The Aga Khan University, Stadium Road, P.O. Box 3500, Karachi 74800, Pakistan.
| | | | | | | | | | | |
Collapse
|
42
|
Li L, Shi Y, Wang X, Shi W, Jiang C. Single nucleotide polymorphisms in K(ATP) channels: muscular impact on type 2 diabetes. Diabetes 2005; 54:1592-7. [PMID: 15855351 DOI: 10.2337/diabetes.54.5.1592] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
ATP-sensitive K+ channels (K(ATP) channels) play an important role in glucose homeostasis. A single nucleotide polymorphism (SNP) in the Kir6.2 subunit causes a point mutation of Glu23 to lysine and reduces the ATP sensitivity of pancreatic K(ATP) channels. The SNP found in 58% of Caucasians accounts for 15% of type 2 diabetes. Here we show evidence for dysregulations of muscular K(ATP) channels with the E23K variation. We were particularly interested in the channel modulation by intracellular protons, as pH changes widely and frequently in skeletal muscles. Surprisingly, we found that the defect of the E23K variant was more related to pH than ATP. A level of intracellular acidification seen during exercise not only activated the E23K channel more readily than the wild type, but also relieved the channel inhibition by ATP, leading to a vast increase in the channel open-state probability by approximately sevenfold at pH 6.8 over the wild-type channel at pH 7.4. Considering the reduction in sarcolemmal excitability, muscle fatigue, and impairment of muscular glucose uptake found previously by genetically disrupting K(ATP) channels, it is likely that the E23K variant in muscular K(ATP) channels affects systemic glucose homeostasis and poses an important risk factor for type 2 diabetes and obesity.
Collapse
Affiliation(s)
- Li Li
- Department of Biology, Georgia State University, 24 Peachtree Center Ave., Atlanta, Georgia 30302-4010, USA
| | | | | | | | | |
Collapse
|
43
|
Wang R, Su J, Wang X, Piao H, Zhang X, Adams CY, Cui N, Jiang C. Subunit stoichiometry of the Kir1.1 channel in proton-dependent gating. J Biol Chem 2005; 280:13433-41. [PMID: 15691840 DOI: 10.1074/jbc.m411895200] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Kir1.1 channel regulates membrane potential and K+ secretion in renal tubular cells. This channel is gated by intracellular protons, in which a lysine residue (Lys80) plays a critical role. Mutation of the Lys80 to a methionine (K80M) disrupts pH-dependent channel gating. To understand how an individual subunit in a tetrameric channel is involved in pH-dependent channel gating, we performed these studies by introducing K80M-disrupted subunits to tandem tetrameric channels. The pH sensitivity was studied in whole-cell voltage clamp and inside-out patches. Homomeric tetramers of the wild-type (wt) and K80M-disrupted channels showed a pH sensitivity almost identical to that of their monomeric counterparts. In heteromeric tetramers and dimers, pH sensitivity was a function of the number of wt subunits. Recruitment of the first single wt subunit shifts the pK(a) greatly, whereas additions of any extra wt subunit had smaller effects. Single-channel analysis revealed that the tetrameric channel with two or more wt subunits showed one substate conductance at approximately 40% of the full conductance, suggesting that four subunits act as two pairs. However, three and four substates of conductance were seen in the tetrameric wt-3K80M and 4K80M channels. Acidic pH increased long-time closures when there were two or more wt subunits. Disruption of more than two subunits led to flicking activity with appearance of a new opening event and loss of the long period of closures. Interestingly, the channel with two wt subunits at diagonal and adjacent configurations showed the same pH sensitivity, substate conductance, and long-time closure. These results thus suggest that one functional subunit is sufficient to act in the pH-dependent gating of the Kir1.1 channel, the channel sensitivity to pH increases with additional subunits, the full pH sensitivity requires contributions of all four subunits, and two subunits may be coordinated in functional dimers of either trans or cis configuration.
Collapse
Affiliation(s)
- Runping Wang
- Department of Biology, Georgia State University, Atlanta, Georgia 30302-4010, USA
| | | | | | | | | | | | | | | |
Collapse
|
44
|
Jiang C, Rojas A, Wang R, Wang X. CO2 central chemosensitivity: why are there so many sensing molecules? Respir Physiol Neurobiol 2005; 145:115-26. [PMID: 15705527 DOI: 10.1016/j.resp.2004.07.005] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/12/2004] [Indexed: 11/18/2022]
Abstract
CO2 central chemoreceptors (CCRs) play a critical role in respiratory and cardiovascular controls. Although the primary sensory cells and their neuronal networks remain elusive, recent studies have begun to shed insight into the molecular mechanisms of several pH sensitive proteins. These putative CO2/pH-sensing molecules are expressed in the brainstem, detect P(CO2) at physiological levels, and couple the P(CO2) to membrane excitability. Functional analysis suggests that multiple CO2/pH-sensing molecules are needed to achieve high sensitivity and broad bandwidth of the CCRs. In contrast to the diversity of pH sensitive molecules, molecular mechanisms for CO2 sensing are rather general. The sensing molecules detect pH changes rather than molecular CO2. One or a few titratable amino acid residues in these proteins are usually involved. Protonation of these residues may lead to a change in protein conformation that is coupled to a change in channel activity. Depending on the location of the protonation sites, a membrane protein can detect extra- and/or intracellular pH.
Collapse
Affiliation(s)
- Chun Jiang
- Department of Biology, Georgia State University, 24 Peachtree Center Avenue, Atlanta GA 30302-4010, USA.
| | | | | | | |
Collapse
|
45
|
Kluess HA, Buckwalter JB, Hamann JJ, Clifford PS. Acidosis attenuates P2X purinergic vasoconstriction in skeletal muscle arteries. Am J Physiol Heart Circ Physiol 2004; 288:H129-32. [PMID: 15374827 DOI: 10.1152/ajpheart.00574.2004] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Vasoconstriction via alpha(2)-receptors is known to be sensitive to acidic pH, but little is known about the pH sensitivity of P2X receptors. ATP is a cotransmitter released with norepinephrine from the sympathetic nerves and causes vasoconstriction via P2X purinergic receptors on vascular smooth muscle. We hypothesized that reductions in pH would attenuate P2X-mediated vasoconstriction in iliofemoral artery rings. Twenty-five rats were killed, and the iliac and femoral arteries were dissected out and placed in modified Krebs-Henseleit buffer. The arteries were cut into 2-mm sections and mounted in an organ tissue bath. Tension (g) was measured during a potassium chloride and norepinephrine challenge (maximal tension). The arteries were then exposed to alpha,beta-methylene ATP (10(-7)-10(-3) M; n = 13) or phenylephrine (10(-7)-10(-4) M; n = 6) with a tissue bath pH of 7.8, 7.4, and 7.0. Dose-response curves were fit with nonlinear regression analysis to calculate the EC(50) and slope. The peak tension with alpha,beta-methylene ATP was lower during pH 7.0 (1.37 +/- 0.09 g) compared with pH 7.8 (1.90 +/- 0.12 g). EC(50) was highest with pH 7.4 (-5.38 +/- 0.18 log M alpha,beta-methylene ATP) and lowest with pH 7.0 (-4.9 +/- 0.10 log M alpha,beta-methylene ATP). The slopes of the dose-response curves were not different. Pyridoxal phosphate-6-azo(benzene-2,4-disulfonic acid) abolished contraction caused by the addition of alpha,beta-methylene ATP (n = 6). There was no effect of pH on phenylephrine dose-response curves. These data indicate that the vasoconstrictor response to alpha,beta-methylene ATP is sensitive to pH and that lower pH attenuates the response of P2X purinergic receptors.
Collapse
Affiliation(s)
- Heidi A Kluess
- Department of Anesthesiology and Physiology, Medical College of Wisconsin, Milwaukee, WI , USA
| | | | | | | |
Collapse
|
46
|
Du X, Zhang H, Lopes C, Mirshahi T, Rohacs T, Logothetis DE. Characteristic interactions with phosphatidylinositol 4,5-bisphosphate determine regulation of kir channels by diverse modulators. J Biol Chem 2004; 279:37271-81. [PMID: 15155739 DOI: 10.1074/jbc.m403413200] [Citation(s) in RCA: 149] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
The activity of specific inwardly rectifying potassium (Kir) channels is regulated by any of a number of different modulators, such as protein kinase C, G(q) -coupled receptor stimulation, pH, intracellular Mg(2+) or the betagamma-subunits of G proteins. Phosphatidylinositol 4,5-bisphosphate (PIP(2)) is an essential factor for maintenance of the activity of all Kir channels. Here, we demonstrate that the strength of channel-PIP(2) interactions determines the sensitivity of Kir channels to regulation by the various modulators. Furthermore, our results suggest that differences among Kir channels in their specific regulation by a given modulator may reflect differences in their apparent affinity of interactions with PIP(2).
Collapse
Affiliation(s)
- Xiaona Du
- Department of Pharmacology, Hebei Medical University, Shijiazhuang, China
| | | | | | | | | | | |
Collapse
|
47
|
Abstract
Potassium (K+) channels exist in all three domains of organisms: eubacteria, archaebacteria, and eukaryotes. In higher animals, these membrane proteins participate in a multitude of critical physiological processes, including food and fluid intake, locomotion, stress response, and cognitive functions. Metabolic regulatory factors such as O2, CO2/pH, redox equivalents, glucose/ATP/ADP, hormones, eicosanoids, cell volume, and electrolytes regulate a diverse group of K+ channels to maintain homeostasis.
Collapse
Affiliation(s)
- Xiang Dong Tang
- Department of Physiology, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA.
| | | | | | | |
Collapse
|
48
|
Rosenblum WI, Wei EP, Kontos HA. Vasodilation of brain surface arterioles by blockade of Na–H+ antiport and its inhibition by inhibitors of KATP channel openers. Brain Res 2004; 1005:77-83. [PMID: 15044067 DOI: 10.1016/j.brainres.2004.01.035] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/28/2004] [Indexed: 11/19/2022]
Abstract
Pial artrioles of rats were monitored in vivo and found to dilate in dose-dependent fashion upon application of either benzamil or ethyl isopropyl amiloride, both of which are inhibitors of the sodium-hydrogen antiport. Antiport blockade is known to decrease the internal pH of vascular smooth muscle (VSM). The dilation was blocked by 1 microm glibenclamide, which in that dose is a selective inhibitor of ATP sensitive potassium channels (K(ATP)). The nitric oxide synthase inhibitor nitro-l arginine (l-NNA) also blocked the response. Previous studies of this preparation under the same experimental conditions showed that l-NNA inhibited dilation by K(ATP) openers and that nitric oxide had no permissive action in this setting. Moreover, one study by others has demonstrated a pH sensitive site on the internal surface of K(ATP) while another study by others has demonstrated that sodium propionate, a direct acidifier of the cell, dilates rat basilar artery in K(ATP)-dependent fashion. Therefore, the present data support the following conclusions: decrease of internal pH dilates brain arterioles; the response is K(ATP) dependent; in some situations, inhibitors of nitric oxide synthase can inhibit K(ATP) and K(ATP)-dependent dilations including those produced by decrease of internal pH.
Collapse
Affiliation(s)
- William I Rosenblum
- Department of Pathology (Neuropathology), Virginia Commonwealth University Medical Center-Medical College of Virginia Campus, Richmond VA 23298-0017, USA.
| | | | | |
Collapse
|
49
|
Wu J, Piao H, Rojas A, Wang R, Wang Y, Cui N, Shi Y, Chen F, Jiang C. Critical protein domains and amino acid residues for gating the KIR6.2 channel by intracellular ATP. J Cell Physiol 2004; 198:73-81. [PMID: 14584046 DOI: 10.1002/jcp.10388] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
K(ATP) channels couple intermediary metabolism to cellular excitability. Such a property relies on the inherent ATP-sensing mechanism known to be located in the Kir6 subunit. However, the molecular basis for the ATP sensitivity remains unclear. Here we showed evidence for protein domains and amino acid residues essential for the channel gating by intracellular ATP. Chimerical channels were constructed using protein domains of Kir6.2 and Kir1.1, expressed in HEK293 cells, and studied in inside-out patches. The N and C termini, although important, were inadequate for channel gating by intracellular ATP. Full ATP sensitivity also required M1 and M2 helices. Cytosolic portions of the M1 and M2 sequences were crucial, in which six amino acid residues were identified, i.e., Thr76, Met77, Ala161, Iso162, Leu164, and Cys166. Site-specific mutation of any of them reduced the ATP sensitivity. Construction of these residues together with the N/C termini produced ATP sensitivity identical to the wild-type channels. The requirement for specific membrane helices suggests that the Kir6.2 gating by ATP is not shared by even two closest relatives in the K(+) channel family, although the general gating mechanisms involving membrane helices appear to be conserved in all K(+) channels.
Collapse
Affiliation(s)
- Jianping Wu
- Department of Biology, Georgia State University, 24 Peachtree Center Avenue, Atlanta, Georgia 30302, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Paajanen V, Vornanen M. Regulation of action potential duration under acute heat stress by I(K,ATP) and I(K1) in fish cardiac myocytes. Am J Physiol Regul Integr Comp Physiol 2003; 286:R405-15. [PMID: 14592934 DOI: 10.1152/ajpregu.00500.2003] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The mechanism underlying temperature-dependent shortening of action potential (AP) duration was examined in the fish (Carassius carassius L.) heart ventricle. Acute temperature change from +5 to +18 degrees C (heat stress) shortened AP duration from 2.8 +/- 0.3 to 1.3 +/- 0.1 s in intact ventricles. In 56% (18 of 32) of enzymatically isolated myocytes, heat stress also induced reversible opening of ATP-sensitive K+ channels and increased their single-channel conductance from 37 +/- 12 pS at +8 degrees C to 51 +/- 13 pS at +18 degrees C (Q10 = 1.38) (P < 0.01; n = 12). The ATP-sensitive K+ channels of the crucian carp ventricle were characterized by very low affinity to ATP both at +8 degrees C [concentration of Tris-ATP that produces half-maximal inhibition of the channel (K1/2)= 1.35 mM] and +18 degrees C (K1/2 = 1.85 mM). Although acute heat stress induced ATP-sensitive K+ current (IK,ATP) in patch-clamped myocytes, similar heat stress did not cause any glibenclamide (10 microM)-sensitive changes in AP duration in multicellular ventricular preparations. Examination of APs and K+ currents from the same myocytes by alternate recording under current-clamp and voltage-clamp modes revealed that changes in AP duration were closely correlated with temperature-specific changes in the voltage-dependent rectification of the background inward rectifier K+ current IK1. In approximately 15% of myocytes (4 out of 27), IK,ATP-dependent shortening of AP followed the IK1-induced AP shortening. Thus heat stress-induced shortening of AP duration in crucian carp ventricle is primarily dependent on IK1. IK,ATP is induced only in response to prolonged temperature elevation or perhaps in the presence of additional stressors.
Collapse
Affiliation(s)
- Vesa Paajanen
- Univ. of Joensuu, Dept. of Biology, P.O. Box 111, 80101 Joensuu, Finland.
| | | |
Collapse
|