1
|
Richter-Laskowska M, Trybek P, Delfino DV, Wawrzkiewicz-Jałowiecka A. Flavonoids as Modulators of Potassium Channels. Int J Mol Sci 2023; 24:1311. [PMID: 36674825 PMCID: PMC9861088 DOI: 10.3390/ijms24021311] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2022] [Revised: 01/02/2023] [Accepted: 01/04/2023] [Indexed: 01/12/2023] Open
Abstract
Potassium channels are widely distributed integral proteins responsible for the effective and selective transport of K+ ions through the biological membranes. According to the existing structural and mechanistic differences, they are divided into several groups. All of them are considered important molecular drug targets due to their physiological roles, including the regulation of membrane potential or cell signaling. One of the recent trends in molecular pharmacology is the evaluation of the therapeutic potential of natural compounds and their derivatives, which can exhibit high specificity and effectiveness. Among the pharmaceuticals of plant origin, which are potassium channel modulators, flavonoids appear as a powerful group of biologically active substances. It is caused by their well-documented anti-oxidative, anti-inflammatory, anti-mutagenic, anti-carcinogenic, and antidiabetic effects on human health. Here, we focus on presenting the current state of knowledge about the possibilities of modulation of particular types of potassium channels by different flavonoids. Additionally, the biological meaning of the flavonoid-mediated changes in the activity of K+ channels will be outlined. Finally, novel promising directions for further research in this area will be proposed.
Collapse
Affiliation(s)
- Monika Richter-Laskowska
- The Centre for Biomedical Engineering, Łukasiewicz Research Network—Krakow Institute of Technology, 30-418 Krakow, Poland
| | - Paulina Trybek
- Faculty of Science and Technology, University of Silesia in Katowice, 41-500 Chorzów, Poland
| | | | - Agata Wawrzkiewicz-Jałowiecka
- Department of Physical Chemistry and Technology of Polymers, Silesian University of Technology, 44-100 Gliwice, Poland
| |
Collapse
|
2
|
Lee HM, Hahn SJ, Choi BH. The antidiabetic drug rosiglitazone blocks Kv1.5 potassium channels in an open state. THE KOREAN JOURNAL OF PHYSIOLOGY & PHARMACOLOGY 2022; 26:135-144. [PMID: 35203063 PMCID: PMC8890944 DOI: 10.4196/kjpp.2022.26.2.135] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/03/2022] [Revised: 01/15/2022] [Accepted: 01/17/2022] [Indexed: 11/15/2022]
Abstract
An antidiabetic drug, rosiglitazone is a member of the drug class of thiazolidinedione. Although restrictions on use due to the possibility of heart toxicity have been removed, it is still a drug that is concerned about side effects on the heart. We here examined, using Chinese hamster ovary cells, the action of rosiglitazone on Kv1.5 channels, which is a major determinant of the duration of cardiac action potential. Rosiglitazone rapidly and reversibly inhibited Kv1.5 currents in a concentration-dependent manner (IC50 = 18.9 µM) and accelerated the decay of Kv1.5 currents without modifying the activation kinetics. In addition, the deactivation of Kv1.5 current, assayed with tail current, was slowed by the drug. All of the results as well as the use-dependence of the rosiglitazone-mediated blockade indicate that rosiglitazone acts on Kv1.5 channels as an open channel blocker. This study suggests that the cardiac side effects of rosiglitazone might be mediated in part by suppression of Kv1.5 channels, and therefore, raises a concern of using the drug for diabetic therapeutics.
Collapse
Affiliation(s)
- Hyang Mi Lee
- Department of Pharmacology, Institute for Medical Sciences, Jeonbuk National University Medical School, Jeonju 54097, Korea
| | - Sang June Hahn
- Department of Physiology, Medical Research Center, College of Medicine, The Catholic University of Korea, Seoul 06591, Korea
| | - Bok Hee Choi
- Department of Pharmacology, Institute for Medical Sciences, Jeonbuk National University Medical School, Jeonju 54097, Korea
| |
Collapse
|
3
|
Zhang M, Che C, Cheng J, Li P, Yang Y. Ion channels in stem cells and their roles in stem cell biology and vascular diseases. J Mol Cell Cardiol 2022; 166:63-73. [PMID: 35143836 DOI: 10.1016/j.yjmcc.2022.02.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/11/2021] [Revised: 01/11/2022] [Accepted: 02/01/2022] [Indexed: 10/19/2022]
Abstract
Stem cell therapy may be a promising option for the treatment of vascular diseases. In recent years, significant progress has been made in stem cell research, especially in the mechanism of stem cell activation, homing and differentiation in vascular repair and reconstruction. Current research on stem cells focuses on protein expression and transcriptional networks. Ion channels are considered to be the basis for the generation of bioelectrical signals, which control the proliferation, differentiation and migration of various cell types. Although heterogeneity of multiple ion channels has been found in different types of stem cells, it is unclear whether the heterogeneous expression of ion channels is related to different cell subpopulations and/or different stages of the cell cycle. There is still a long way to go in clinical treatment by using the regulation of stem cell ion channels. In this review, we reviewed the main ion channels found on stem cells, their expression and function in stem cell proliferation, differentiation and migration, and the research status of stem cells' involvement in vascular diseases.
Collapse
Affiliation(s)
- Min Zhang
- Key Laboratory of Medical Electrophysiology of Ministry of Education and Medical Electrophysiological Key Laboratory of Sichuan Province, Collaborative Innovation Center for Prevention and Treatment of Cardiovascular Disease, Institute of Cardiovascular Research, Southwest Medical University, 319 Zhongshan Road, Luzhou 646000, China
| | - Chang Che
- Key Laboratory of Medical Electrophysiology of Ministry of Education and Medical Electrophysiological Key Laboratory of Sichuan Province, Collaborative Innovation Center for Prevention and Treatment of Cardiovascular Disease, Institute of Cardiovascular Research, Southwest Medical University, 319 Zhongshan Road, Luzhou 646000, China
| | - Jun Cheng
- Key Laboratory of Medical Electrophysiology of Ministry of Education and Medical Electrophysiological Key Laboratory of Sichuan Province, Collaborative Innovation Center for Prevention and Treatment of Cardiovascular Disease, Institute of Cardiovascular Research, Southwest Medical University, 319 Zhongshan Road, Luzhou 646000, China
| | - Pengyun Li
- Key Laboratory of Medical Electrophysiology of Ministry of Education and Medical Electrophysiological Key Laboratory of Sichuan Province, Collaborative Innovation Center for Prevention and Treatment of Cardiovascular Disease, Institute of Cardiovascular Research, Southwest Medical University, 319 Zhongshan Road, Luzhou 646000, China.
| | - Yan Yang
- Key Laboratory of Medical Electrophysiology of Ministry of Education and Medical Electrophysiological Key Laboratory of Sichuan Province, Collaborative Innovation Center for Prevention and Treatment of Cardiovascular Disease, Institute of Cardiovascular Research, Southwest Medical University, 319 Zhongshan Road, Luzhou 646000, China.
| |
Collapse
|
4
|
Kamiya T, Omae T, Nakabayashi S, Takahashi K, Tanner A, Yoshida A. Effect of Rho Kinase Inhibitor Ripasudil (K-115) on Isolated Porcine Retinal Arterioles. J Ocul Pharmacol Ther 2020; 37:104-111. [PMID: 33351704 DOI: 10.1089/jop.2020.0082] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
Purpose: To investigate the vasorelaxation effect of ripasudil (K-115), a novel Rho-associated coiled-coil-containing protein kinase (ROCK) inhibitor, on isolated retinal arterioles. We determined whether the actions of ripasudil on the retinal microvascular diameter were dependent on the endothelium and/or potassium channels in the smooth muscle, with the goals of uncovering the signaling mechanisms required for this vasomotor activity and inhibiting the action of endothelin-1 (ET-1). Methods: In this in vitro study, we isolated porcine retinal arterioles, which were cannulated and pressurized without flow. We recorded diametric changes using videomicroscopic techniques. Results: In a dose-dependent (10 nM-30 μM) manner, retinal arterioles were relaxed in response to ripasudil [maximum % resting diameter, 160.3% ± 7.7% (mean ± standard error of the mean)]. The ripasudil-induced vasorelaxation was unaffected by endothelium removal, using nonselective potassium channel blocker tetraethylammonium, Ca2+-activated large-conductance potassium channel blocker iberiotoxin, voltage-gated potassium channel blocker 4-AP, ATP-sensitive potassium channel blocker glibenclamide, and inward rectifier potassium channel blocker BaCl2. Ripasudil prevented ET-1-caused vasoconstriction of the retinal arterioles regardless of the presence of endothelium to a similar extent. Conclusion: The ROCK inhibitor ripasudil elicits endothelium-independent relaxation and inhibits the action of ET-1 on the retinal arterioles. Determining the relaxation properties of ripasudil on the retinal microvasculature will likely support the development of potential therapies for glaucoma.
Collapse
Affiliation(s)
- Takayuki Kamiya
- Department of Ophthalmology, Asahikawa Medical University, Asahikawa, Japan
| | - Tsuneaki Omae
- Department of Ophthalmology, Asahikawa Medical University, Asahikawa, Japan
| | - Seigo Nakabayashi
- Department of Ophthalmology, Asahikawa Medical University, Asahikawa, Japan
| | - Kengo Takahashi
- Department of Ophthalmology, Asahikawa Medical University, Asahikawa, Japan
| | - Akira Tanner
- Department of Ophthalmology, Asahikawa Medical University, Asahikawa, Japan
| | - Akitoshi Yoshida
- Department of Ophthalmology, Asahikawa Medical University, Asahikawa, Japan
| |
Collapse
|
5
|
Adiponectin Exerts Peripheral Inhibitory Effects on the Mouse Gastric Smooth Muscle through the AMPK Pathway. Int J Mol Sci 2020; 21:ijms21249617. [PMID: 33348652 PMCID: PMC7767160 DOI: 10.3390/ijms21249617] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2020] [Revised: 12/10/2020] [Accepted: 12/15/2020] [Indexed: 12/17/2022] Open
Abstract
Some adipokines, such as adiponectin (ADPN), other than being implicated in the central regulation of feeding behavior, may influence gastric motor responses, which are a source of peripheral signals that also influence food intake. The present study aims to elucidate the signaling pathways through which ADPN exerts its actions in the mouse gastric fundus. To this purpose, we used a multidisciplinary approach. The mechanical results showed that ADPN caused a decay of the strip basal tension, which was abolished by the nitric oxide (NO) synthesis inhibitor, L-NG-nitro arginine (L-NNA). The electrophysiological experiments confirmed that all ADPN effects were abolished by L-NNA, except for the reduction of Ca2+ current, which was instead prevented by the inhibitor of AMP-activated protein kinase (AMPK), dorsomorphin. The activation of the AMPK signaling by ADPN was confirmed by immunofluorescence analysis, which also revealed the ADPN R1 receptor (AdipoR1) expression in glial cells of the myenteric plexus. In conclusion, our results indicate that ADPN exerts an inhibitory action on the gastric smooth muscle by acting on AdipoR1 and involving the AMPK signaling pathway at the peripheral level. These findings provide novel bases for considering AMPK as a possible pharmacologic target for the potential treatment of obesity and eating disorders.
Collapse
|
6
|
Tykocki NR, Boerman EM, Jackson WF. Smooth Muscle Ion Channels and Regulation of Vascular Tone in Resistance Arteries and Arterioles. Compr Physiol 2017; 7:485-581. [PMID: 28333380 DOI: 10.1002/cphy.c160011] [Citation(s) in RCA: 228] [Impact Index Per Article: 32.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Vascular tone of resistance arteries and arterioles determines peripheral vascular resistance, contributing to the regulation of blood pressure and blood flow to, and within the body's tissues and organs. Ion channels in the plasma membrane and endoplasmic reticulum of vascular smooth muscle cells (SMCs) in these blood vessels importantly contribute to the regulation of intracellular Ca2+ concentration, the primary determinant of SMC contractile activity and vascular tone. Ion channels provide the main source of activator Ca2+ that determines vascular tone, and strongly contribute to setting and regulating membrane potential, which, in turn, regulates the open-state-probability of voltage gated Ca2+ channels (VGCCs), the primary source of Ca2+ in resistance artery and arteriolar SMCs. Ion channel function is also modulated by vasoconstrictors and vasodilators, contributing to all aspects of the regulation of vascular tone. This review will focus on the physiology of VGCCs, voltage-gated K+ (KV) channels, large-conductance Ca2+-activated K+ (BKCa) channels, strong-inward-rectifier K+ (KIR) channels, ATP-sensitive K+ (KATP) channels, ryanodine receptors (RyRs), inositol 1,4,5-trisphosphate receptors (IP3Rs), and a variety of transient receptor potential (TRP) channels that contribute to pressure-induced myogenic tone in resistance arteries and arterioles, the modulation of the function of these ion channels by vasoconstrictors and vasodilators, their role in the functional regulation of tissue blood flow and their dysfunction in diseases such as hypertension, obesity, and diabetes. © 2017 American Physiological Society. Compr Physiol 7:485-581, 2017.
Collapse
Affiliation(s)
- Nathan R Tykocki
- Department of Pharmacology, University of Vermont, Burlington, Vermont, USA
| | - Erika M Boerman
- Department of Medical Pharmacology and Physiology, University of Missouri, Columbia, Missouri, USA
| | - William F Jackson
- Department of Pharmacology and Toxicology, Michigan State University, East Lansing, Michigan, USA
| |
Collapse
|
7
|
Xiao GS, Zhang YH, Wu W, Sun HY, Wang Y, Li GR. Genistein and tyrphostin AG556 decrease ultra-rapidly activating delayed rectifier K + current of human atria by inhibiting EGF receptor tyrosine kinase. Br J Pharmacol 2017; 174:454-467. [PMID: 28072464 DOI: 10.1111/bph.13710] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2016] [Revised: 12/16/2016] [Accepted: 01/05/2017] [Indexed: 02/02/2023] Open
Abstract
BACKGROUND AND PURPOSE The ultra-rapidly activating delayed rectifier K+ current IKur (encoded by Kv 1.5 or KCNA5) plays an important role in human atrial repolarization. The present study investigates the regulation of this current by protein tyrosine kinases (PTKs). EXPERIMENTAL APPROACH Whole-cell patch voltage clamp technique and immunoprecipitation and Western blotting analysis were used to investigate whether the PTK inhibitors genistein, tyrphostin AG556 (AG556) and PP2 regulate human atrial IKur and hKv1.5 channels stably expressed in HEK 293 cells. KEY RESULTS Human atrial IKur was decreased by genistein (a broad-spectrum PTK inhibitor) and AG556 (a highly selective EGFR TK inhibitor) in a concentration-dependent manner. Inhibition of IKur induced by 30 μM genistein or 10 μM AG556 was significantly reversed by 1 mM orthovanadate (a protein tyrosine phosphatase inhibitor). Similar results were observed in HEK 293 cells stably expressing hKv 1.5 channels. On the other hand, the Src family kinase inhibitor PP2 (1 μM) slightly enhanced IKur and hKv 1.5 current, and the current increase was also reversed by orthovanadate. Immunoprecipitation and Western blotting analysis showed that genistein, AG556, and PP2 decreased tyrosine phosphorylation of hKv 1.5 channels and that the decrease was countered by orthovanadate. CONCLUSION AND IMPLICATIONS The PTK inhibitors genistein and AG556 decrease human atrial IKur and cloned hKv 1.5 channels by inhibiting EGFR TK, whereas the Src kinase inhibitor PP2 increases IKur and hKv 1.5 current. These results imply that EGFR TK and the soluble Src kinases may have opposite effects on human atrial IKur .
Collapse
Affiliation(s)
- Guo-Sheng Xiao
- Xiamen Cardiovascular Hospital, Xiamen University, Xiamen, Fujian, China
| | - Yan-Hui Zhang
- Xiamen Cardiovascular Hospital, Xiamen University, Xiamen, Fujian, China.,Department of Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong, China
| | - Wei Wu
- Department of Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong, China
| | - Hai-Ying Sun
- Department of Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong, China
| | - Yan Wang
- Xiamen Cardiovascular Hospital, Xiamen University, Xiamen, Fujian, China
| | - Gui-Rong Li
- Xiamen Cardiovascular Hospital, Xiamen University, Xiamen, Fujian, China.,Department of Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong, China
| |
Collapse
|
8
|
Le NT, Martin JF, Fujiwara K, Abe JI. Sub-cellular localization specific SUMOylation in the heart. Biochim Biophys Acta Mol Basis Dis 2017; 1863:2041-2055. [PMID: 28130202 DOI: 10.1016/j.bbadis.2017.01.018] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2016] [Revised: 11/21/2016] [Accepted: 01/09/2017] [Indexed: 12/27/2022]
Abstract
Although the majority of SUMO substrates are localized in the nucleus, SUMOylation is not limited to nuclear proteins and can be also detected in extra-nuclear proteins. In this review, we will highlight and discuss how SUMOylation in different cellular compartments regulate biological processes. First, we will discuss the key role of SUMOylation of proteins in the extra-nuclear compartment in cardiomyocytes, which is overwhelmingly cardio-protective. On the other hand, SUMOylation of nuclear proteins is generally detrimental to the cardiac function mainly because of the trans-repressive nature of SUMOylation on many transcription factors. We will also discuss the potential role of SUMOylation in epigenetic regulation. In this review, we will propose a new concept that shuttling of SUMO proteases between the nuclear and extra-nuclear compartments without changing their enzymatic activity regulates the extent of SUMOylation in these compartments and determines the response and fate of cardiomyocytes after cardiac insults. Approaches focused specifically to inhibit this shuttling in cardiomyocytes will be necessary to understand the whole picture of SUMOylation and its pathophysiological consequences in the heart, especially after cardiac insults. This article is part of a Special Issue entitled: Genetic and epigenetic control of heart failure - edited by Jun Ren & Megan Yingmei Zhang.
Collapse
Affiliation(s)
- Nhat-Tu Le
- Department of Cardiology - Research, Division of Internal Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - James F Martin
- Department of Molecular Physiology and Biophysics, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA
| | - Keigi Fujiwara
- Department of Cardiology - Research, Division of Internal Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Jun-Ichi Abe
- Department of Cardiology - Research, Division of Internal Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| |
Collapse
|
9
|
Abe JI, Sandhu UG, Hoang NM, Thangam M, Quintana-Quezada RA, Fujiwara K, Le NT. Coordination of Cellular Localization-Dependent Effects of Sumoylation in Regulating Cardiovascular and Neurological Diseases. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 963:337-358. [PMID: 28197922 PMCID: PMC5716632 DOI: 10.1007/978-3-319-50044-7_20] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Sumoylation, a reversible post-transcriptional modification process, of proteins are involved in cellular differentiation, growth, and even motility by regulating various protein functions. Sumoylation is not limited to cytosolic proteins as recent evidence shows that nuclear proteins, those associated with membranes, and mitochondrial proteins are also sumoylated. Moreover, it is now known that sumoylation plays an important role in the process of major human ailments such as malignant, cardiovascular and neurological diseases. In this chapter, we will highlight and discuss how the localization of SUMO protease and SUMO E3 ligase in different compartments within a cell regulates biological processes that depend on sumoylation. First, we will discuss the key role of sumoylation in the nucleus, which leads to the development of endothelial dysfunction and atherosclerosis . We will then discuss how sumoylation of plasma membrane potassium channel proteins are involved in epilepsy and arrhythmia. Mitochondrial proteins are known to be also sumoylated, and the importance of dynamic-related protein 1 (DRP1) sumoylation on mitochondrial function will be discussed. As we will emphasize throughout this review, sumoylation plays crucial roles in different cellular compartments, which is coordinately regulated by the translocation of various SUMO proteases and SUMO E3 ligase. Comprehensive approach will be necessary to understand the molecular mechanism for efficiently moving around various enzymes that regulate sumoylation within cells.
Collapse
Affiliation(s)
- Jun-Ichi Abe
- Department of Cardiology - Research, Division of Internal Medicine, The University of Texas MD Anderson Cancer Center, 2121 W. Holcombe Blvd, Unit Number: 1101, Room Number: IBT8.803E, Houston, TX, 77030, USA.
| | - Uday G Sandhu
- Department of Cardiology - Research, Division of Internal Medicine, The University of Texas MD Anderson Cancer Center, 2121 W. Holcombe Blvd, Unit Number: 1101, Room Number: IBT8.803E, Houston, TX, 77030, USA
| | - Nguyet Minh Hoang
- Department of Cardiology - Research, Division of Internal Medicine, The University of Texas MD Anderson Cancer Center, 2121 W. Holcombe Blvd, Unit Number: 1101, Room Number: IBT8.803E, Houston, TX, 77030, USA
| | - Manoj Thangam
- Department of Cardiology - Research, Division of Internal Medicine, The University of Texas MD Anderson Cancer Center, 2121 W. Holcombe Blvd, Unit Number: 1101, Room Number: IBT8.803E, Houston, TX, 77030, USA
| | - Raymundo A Quintana-Quezada
- Department of Cardiology - Research, Division of Internal Medicine, The University of Texas MD Anderson Cancer Center, 2121 W. Holcombe Blvd, Unit Number: 1101, Room Number: IBT8.803E, Houston, TX, 77030, USA
| | - Keigi Fujiwara
- Department of Cardiology - Research, Division of Internal Medicine, The University of Texas MD Anderson Cancer Center, 2121 W. Holcombe Blvd, Unit Number: 1101, Room Number: IBT8.803E, Houston, TX, 77030, USA
| | - Nhat Tu Le
- Department of Cardiology - Research, Division of Internal Medicine, The University of Texas MD Anderson Cancer Center, 2121 W. Holcombe Blvd, Unit Number: 1101, Room Number: IBT8.803E, Houston, TX, 77030, USA
| |
Collapse
|
10
|
Role of BK Ca in Stretch-Induced Relaxation of Colonic Smooth Muscle. BIOMED RESEARCH INTERNATIONAL 2016; 2016:9497041. [PMID: 28018918 PMCID: PMC5149602 DOI: 10.1155/2016/9497041] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/22/2016] [Revised: 09/30/2016] [Accepted: 10/23/2016] [Indexed: 12/15/2022]
Abstract
Stretch-induced relaxation has not been clearly identified in gastrointestinal tract. The present study is to explore the role of large conductance calcium-activated potassium channels (BKCa) in stretch-induced relaxation of colon. The expression and currents of BKCa were detected and the basal muscle tone and contraction amplitude of colonic smooth muscle strips were measured. The expression of BKCa in colon is higher than other GI segments (P < 0.05). The density of BKCa currents was very high in colonic smooth muscle cells (SMCs). BKCa in rat colonic SMCs were sensitive to stretch. The relaxation response of colonic SM strips to stretch was attenuated by charybdotoxin (ChTX), a nonspecific BKCa blocker (P < 0.05). After blocking enteric nervous activities by tetrodotoxin (TTX), the stretch-induced relaxation did not change (P > 0.05). Still, ChTX and iberiotoxin (IbTX, a specific BKCa blocker) attenuated the relaxation of the colonic muscle strips enduring stretch (P < 0.05). These results suggest stretch-activation of BKCa in SMCs was involved in the stretch-induced relaxation of colon. Our study highlights the role of mechanosensitive ion channels in SMCs in colon motility regulation and their physiological and pathophysiological significance is worth further study.
Collapse
|
11
|
De Mello WC. Intracellular angiotensin II as a regulator of muscle tone in vascular resistance vessels. Pathophysiological implications. Peptides 2016; 78:87-90. [PMID: 26944358 DOI: 10.1016/j.peptides.2016.02.006] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/03/2015] [Revised: 01/19/2016] [Accepted: 02/23/2016] [Indexed: 12/26/2022]
Abstract
The influence of intracellular angiotensin II on the regulation of potassium current and membrane potential of smooth muscle cells of mesenteric arteries and its relevance for the regulation of vascular tone was reviewed. The presence of components of the renin angiotensin system (RAS) in different cells of the cardiovascular system, was discussed including their presence in the nuclei and mitochondria. Emphasis was given to the opposite effects of intracellular and extracellular angiotensin II (Ang II) on the regulation of potassium current, membrane potential and contractility of vascular resistance vessels and its implication to vascular physiology and pathology and the possible role of epigenetic factors on the expression of angiotensin II (Ang II) and renin in vascular resistance vessels as well as its possible pathophysiological role in hypertension and other cardiovascular diseases.
Collapse
Affiliation(s)
- Walmor C De Mello
- School of Medicine, Medical Sciences Campus, UPR, San Juan, PR 00936-5067, USA.
| |
Collapse
|
12
|
Angiotensin (1-7) increases the potassium current and the resting potential of arterial myocytes from vascular resistance vessels of normal adult rats: Pathophysiological implications. ACTA ACUST UNITED AC 2013; 8:14-20. [PMID: 24220548 DOI: 10.1016/j.jash.2013.08.003] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2013] [Accepted: 08/20/2013] [Indexed: 11/21/2022]
Abstract
The influence of angiotensin (Ang) (1-7) on potassium current (Kv) and resting potential of smooth muscle cells isolated from mesenteric artery of Sprague Dawley rats was investigated. Measurements of potassium current were performed using the whole cell configuration of pCLAMP. The results indicated that Ang (1-7) (10(-9) M) increased the potassium current by 120% ± 2.6% (P < .05) and the resting potential of smooth muscle cells by 8 ± 2.8 mV (n = 23; P < .05). Ang II (10(-9) M) administered to the bath reduced the potassium current by 35% ± 3.6% (n = 23; P < .05) and depolarized the arterial myocytes by 7.8 ± 2.1 mV (n = 25; P < .05). The effect of the heptapeptide on potassium current was inhibited by a Mas receptor inhibitor (A779; 10(-8) M) as well as by a protein kinase A (PKA) inhibitor (10(-9) M) dialyzed into the cell. Intracellular dialysis of the catalytic subunit of PKA (5 × 10(-8) M) enhanced the potassium current by 38% ± 3.4% (n = 14; P < .05) but did not abolish the effect of Ang (1-7). On the other hand, Bis-1 (10(-9) M), which is a specific inhibitor of PKC, suppressed the effect of Ang (1-7) on potassium current. In conclusion, Ang (1-7) counteracts the effect of Ang II on potassium current and membrane potential of smooth muscle cells from mesenteric arteries, which are resistance vessels involved in the regulation of peripheral resistance and blood pressure. The activation of the cAMP/PKA cascade is essential for the effect of the heptapeptide. Pathophysiological implications are discussed.
Collapse
|
13
|
De Mello WC. Intracellular angiotensin II increases the total potassium current and the resting potential of arterial myocytes from vascular resistance vessels of the rat. Physiological and pathological implications. ACTA ACUST UNITED AC 2013; 7:192-7. [PMID: 23538141 DOI: 10.1016/j.jash.2013.02.003] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2012] [Revised: 02/08/2013] [Accepted: 02/11/2013] [Indexed: 11/18/2022]
Abstract
The influence of intracellular and extracellular administration of angiotensin II (Ang II; 10(-9) M) on total potassium current of arterial myocytes isolated from mesenteric arteries of Sprague Dawley rats was investigated. Measurements of total potassium current were performed using the voltage clamp whole cell configuration while the effect of intracellular Ang II on the resting potential of arterial myocytes was measured using the current clamp configuration of pCLAMP. The results indicated that: 1) intracellular Ang II (10(-9) M) increased the total potassium current by 73% ± 2.6% (n = 22; P < .05) within 5 minutes; 2) concurrently with the increment of potassium current, the resting potential was increased by 7 ± 1.5 mV (n = 23; P < .05); 3) extracellular administration of Ang II (10(-9) M) reduced the total potassium current by 20% ± 1.6% (n = 21; P < .05) within 5 minutes and depolarized the smooth muscle cells by 9 ± 2.3 mV (n = 26; P < .05); 4) the effects of intracellular Ang II on potassium current and membrane potential were inhibited by dialyzing a PKA inhibitor (10(-9) M) inside the cell together with Ang II (10(-9) M; P > .05); 5) valsartan (10(-9) M) dialyzed into the cell together with Ang II (10(-9) M) abolished the effect of the peptide on potassium current and membrane potential. The presence of endogenous or internalized intracellular Ang II in vascular resistance vessels and its effect on potassium current and resting potential indicates that the peptide present inside the arterial myocytes plays an important role on the regulation of vascular tone and consequently on peripheral resistance, which is a determining factor in the regulation of arterial blood pressure.
Collapse
Affiliation(s)
- Walmor C De Mello
- School of Medicine, Medical Sciences Campus, University of Puerto Rico, San Juan, Puerto Rico 00936, USA.
| |
Collapse
|
14
|
Mia S, Munoz C, Pakladok T, Siraskar G, Voelkl J, Alesutan I, Lang F. Downregulation of Kv1.5 K channels by the AMP-activated protein kinase. Cell Physiol Biochem 2012; 30:1039-50. [PMID: 23221389 DOI: 10.1159/000341480] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/03/2012] [Indexed: 12/28/2022] Open
Abstract
BACKGROUND The voltage gated K(+) channel Kv1.5 participates in the repolarization of a wide variety of cell types. Kv1.5 is downregulated during hypoxia, which is known to stimulate the energy-sensing AMP-activated serine/threonine protein kinase (AMPK). AMPK is a powerful regulator of nutrient transport and metabolism. Moreover, AMPK is known to downregulate several ion channels, an effect at least in part due to stimulation of the ubiquitin ligase Nedd4- 2. The present study explored whether AMPK regulates Kv1.5. METHODS cRNA encoding Kv1.5 was injected into Xenopus oocytes with and without additional injection of wild-type AMPK (α1 β 1γ1), of constitutively active (γR70Q)AMPK (α1 β 1γ1(R70Q)), of inactive mutant (αK45R)AMPK (α1(K45R)β1γ1), or of Nedd4-2. Kv1.5 activity was determined by two-electrode voltage-clamp. Moreover, Kv1.5 protein abundance in the cell membrane was determined by chemiluminescence and immunostaining with subsequent confocal microscopy. RESULTS Coexpression of wild-type AMPK(WT) and constitutively active AMPK(γR70Q), but not of inactive AMPK(αK45R) significantly reduced Kv1.5-mediated currents. Coexpression of constitutively active AMPKγR70Q further reduced Kv1.5 K(+) channel protein abundance in the cell membrane. Co-expression of Nedd4-2 similarly downregulated Kv1.5-mediated currents. CONCLUSION AMPK is a potent regulator of Kv1.5. AMPK inhibits Kv1.5 presumably in part by activation of Nedd4- 2 with subsequent clearance of channel protein from the cell membrane.
Collapse
Affiliation(s)
- Sobuj Mia
- Department of Physiology, University of Tübingen, Tübingen, Germany
| | | | | | | | | | | | | |
Collapse
|
15
|
Jeong I, Yoon SH, Hahn SJ. Effects of dapoxetine on cloned Kv1.5 channels expressed in CHO cells. Naunyn Schmiedebergs Arch Pharmacol 2012; 385:707-16. [PMID: 22538641 DOI: 10.1007/s00210-012-0754-y] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2012] [Accepted: 04/10/2012] [Indexed: 11/25/2022]
Abstract
The effects of dapoxetine were examined on cloned Kv1.5 channels stably expressed in Chinese hamster ovary cells using the whole-cell patch clamp technique. Dapoxetine decreased the peak amplitude of Kv1.5 currents and accelerated the decay rate of current inactivation in a concentration-dependent manner with an IC ( 50 ) of 11.6 μM. Kinetic analysis of the time-dependent effects of dapoxetine on Kv1.5 current decay yielded the apparent association (k (+1 )) and dissociation (k (-1 )) rate constants of 2.8 μM(-1) s(-1) and 34.2 s(-1), respectively. The theoretical K ( D ) value, derived by k (-1 )/k (+1 ), yielded 12.3 μM, which was reasonably similar to the IC ( 50 ) value obtained from the concentration-response curve. Dapoxetine decreased the tail current amplitude and slowed the deactivation process of Kv1.5, which resulted in a tail crossover phenomenon. The block by dapoxetine is voltage-dependent and steeply increased at potentials between -10 and +10 mV, which correspond to the voltage range of channel activation. At more depolarized potentials, a weaker voltage dependence was observed (δ=0.31). Dapoxetine had no effect on the steady-state activation of Kv1.5 but shifted the steady-state inactivation curves in a hyperpolarizing direction. Dapoxetine produced a use-dependent block of Kv1.5 at frequencies of 1 and 2 Hz and slowed the time course for recovery of inactivation. These effects were reversible after washout of the drug. Our results indicate that dapoxetine blocks Kv1.5 currents by interacting with the channel in both the open and inactivated states of the channel.
Collapse
Affiliation(s)
- Imju Jeong
- Department of Physiology, Medical Research Center, College of Medicine, The Catholic University of Korea, 505 Banpo-dong, Socho-gu, Seoul 137-701, South Korea
| | | | | |
Collapse
|
16
|
Beyder A, Farrugia G. Targeting ion channels for the treatment of gastrointestinal motility disorders. Therap Adv Gastroenterol 2012; 5:5-21. [PMID: 22282704 PMCID: PMC3263980 DOI: 10.1177/1756283x11415892] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
Gastrointestinal (GI) functional and motility disorders are highly prevalent and responsible for long-term morbidity and sometimes mortality in the affected patients. It is estimated that one in three persons has a GI functional or motility disorder. However, diagnosis and treatment of these widespread conditions remains challenging. This partly stems from the multisystem pathophysiology, including processing abnormalities in the central and peripheral (enteric) nervous systems and motor dysfunction in the GI wall. Interstitial cells of Cajal (ICCs) are central to the generation and propagation of the cyclical electrical activity and smooth muscle cells (SMCs) are responsible for electromechanical coupling. In these and other excitable cells voltage-sensitive ion channels (VSICs) are the main molecular units that generate and regulate electrical activity. Thus, VSICs are potential targets for intervention in GI motility disorders. Research in this area has flourished with advances in the experimental methods in molecular and structural biology and electrophysiology. However, our understanding of the molecular mechanisms responsible for the complex and variable electrical behavior of ICCs and SMCs remains incomplete. In this review, we focus on the slow waves and action potentials in ICCs and SMCs. We describe the constituent VSICs, which include voltage-gated sodium (Na(V)), calcium (Ca(V)), potassium (K(V), K(Ca)), chloride (Cl(-)) and nonselective ion channels (transient receptor potentials [TRPs]). VSICs have significant structural homology and common functional mechanisms. We outline the approaches and limitations and provide examples of targeting VSICs at the pores, voltage sensors and alternatively spliced sites. Rational drug design can come from an integrated view of the structure and mechanisms of gating and activation by voltage or mechanical stress.
Collapse
Affiliation(s)
- Arthur Beyder
- Enteric Neuroscience Program, Division of Gastroenterology and Hepatology, Department of Medicine, Mayo Clinic, Rochester, MN, USA
| | | |
Collapse
|
17
|
Munoz C, Tóvolli RH, Sopjani M, Alesutan I, Lam RS, Seebohm G, Föller M, Lang F. Activation of voltage gated K⁺ channel Kv1.5 by β-catenin. Biochem Biophys Res Commun 2011; 417:692-6. [PMID: 22166221 DOI: 10.1016/j.bbrc.2011.11.156] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2011] [Accepted: 11/30/2011] [Indexed: 01/16/2023]
Abstract
Voltage-gated Kv1.5 channels are expressed in a wide variety of tissues including cardiac myocytes, smooth muscle and tumor cells. Kv1.5 channel activity is modified by N-cadherin, which in turn binds the multifunctional oncogenic protein β-catenin. The present experiments explored the effect of β-catenin on Kv1.5 channel activity. To this end, Kv1.5 was expressed in Xenopus oocytes with or without β-catenin and the voltage-gated Kv current determined by dual electrode voltage clamp. As a result, expression of β-catenin significantly increased the voltage-gated Kv current at positive potentials. The stimulating effect of β-catenin on Kv1.5 was not dependent on the stimulation of transcription since it was observed even in the presence of the transcription inhibitor actinomycin D. Specific antibody binding to surface Kv1.5 in Xenopus oocytes revealed that β-catenin enhances the membrane abundance of Kv1.5. Further experiments with brefeldin A showed that β-catenin fosters the insertion of Kv1.5 into rather than delaying the retrieval from the plasma membrane. According to electrophysiological recordings with mutant β-catenin, the effect on Kv1.5 requires the same protein domains that are required for association of β-catenin with cadherin. The experiments disclose a completely novel function of β-catenin, i.e. the regulation of Kv1.5 channel activity.
Collapse
Affiliation(s)
- Carlos Munoz
- Department of Physiology, University of Tübingen, Gmelinstr. 5, D-72076 Tübingen, Germany
| | | | | | | | | | | | | | | |
Collapse
|
18
|
Ko EA, Park WS, Firth AL, Kim N, Yuan JXJ, Han J. Pathophysiology of voltage-gated K+ channels in vascular smooth muscle cells: Modulation by protein kinases. PROGRESS IN BIOPHYSICS AND MOLECULAR BIOLOGY 2010; 103:95-101. [DOI: 10.1016/j.pbiomolbio.2009.10.001] [Citation(s) in RCA: 63] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/01/2009] [Accepted: 10/07/2009] [Indexed: 10/20/2022]
|
19
|
Abstract
The human genome encodes 40 voltage-gated K(+) channels (K(V)), which are involved in diverse physiological processes ranging from repolarization of neuronal and cardiac action potentials, to regulating Ca(2+) signalling and cell volume, to driving cellular proliferation and migration. K(V) channels offer tremendous opportunities for the development of new drugs to treat cancer, autoimmune diseases and metabolic, neurological and cardiovascular disorders. This Review discusses pharmacological strategies for targeting K(V) channels with venom peptides, antibodies and small molecules, and highlights recent progress in the preclinical and clinical development of drugs targeting the K(V)1 subfamily, the K(V)7 subfamily (also known as KCNQ), K(V)10.1 (also known as EAG1 and KCNH1) and K(V)11.1 (also known as HERG and KCNH2) channels.
Collapse
|
20
|
Jepps TA, Greenwood IA, Moffatt JD, Sanders KM, Ohya S. Molecular and functional characterization of Kv7 K+ channel in murine gastrointestinal smooth muscles. Am J Physiol Gastrointest Liver Physiol 2009; 297:G107-15. [PMID: 19389803 PMCID: PMC2711751 DOI: 10.1152/ajpgi.00057.2009] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Members of the K(v)7 voltage-gated K(+) channel family are important determinants of cardiac and neuronal membrane excitability. Recently, we and others have shown that K(v)7 channels are also crucial regulators of smooth muscle activity. The aim of the present study was to assess the K(v)7 expression in different parts of the murine gastrointestinal (GI) tract and to assess their functional roles by use of pharmacological agents. Of KCNQ/K(v)7 members, both KCNQ4/K(v)7.4 and KCNQ5/K(v)7.5 genes and proteins were the most abundantly expressed K(v)7 channels in smooth muscles throughout the GI tract. Immunohistochemical staining also revealed that K(v)7.4 and K(v)7.5 but not K(v)7.1 were expressed in the circular muscle layer of the colon. In segments of distal colon circular muscle exhibiting spontaneous phasic contractions, the nonselective K(v)7 blockers XE991 and linopirdine increased the integral of tension. Increases in the integral of tension were also observed under conditions of neuronal blockade. Similar effects, although less marked, were observed in the proximal colon. As expected, the K(v)7.1-selective blocker chromanol 293B had no effect in either type of segment. These data show that K(v)7.x especially K(v)7.4 and K(v)7.5 are expressed in different regions of the murine gastrointestinal tract and blockers of K(v)7 channels augment inherent contractile activity. Drugs that selectively block K(v)7.4/7.5 might be promising therapeutics for the treatment of motility disorders such as constipation associated with irritable bowel syndrome.
Collapse
Affiliation(s)
- Thomas A. Jepps
- Division of Basic Medical Sciences, St George's, University of London, London, United Kingdom; Department of Physiology and Cell Biology, University of Nevada School of Medicine, Reno, Nevada; and Department of Molecular and Cellular Pharmacology, Graduate School of Pharmaceutical Sciences, Nagoya City University, Nagoya, Japan
| | - Iain A. Greenwood
- Division of Basic Medical Sciences, St George's, University of London, London, United Kingdom; Department of Physiology and Cell Biology, University of Nevada School of Medicine, Reno, Nevada; and Department of Molecular and Cellular Pharmacology, Graduate School of Pharmaceutical Sciences, Nagoya City University, Nagoya, Japan
| | - James D. Moffatt
- Division of Basic Medical Sciences, St George's, University of London, London, United Kingdom; Department of Physiology and Cell Biology, University of Nevada School of Medicine, Reno, Nevada; and Department of Molecular and Cellular Pharmacology, Graduate School of Pharmaceutical Sciences, Nagoya City University, Nagoya, Japan
| | - Kenton M. Sanders
- Division of Basic Medical Sciences, St George's, University of London, London, United Kingdom; Department of Physiology and Cell Biology, University of Nevada School of Medicine, Reno, Nevada; and Department of Molecular and Cellular Pharmacology, Graduate School of Pharmaceutical Sciences, Nagoya City University, Nagoya, Japan
| | - Susumu Ohya
- Division of Basic Medical Sciences, St George's, University of London, London, United Kingdom; Department of Physiology and Cell Biology, University of Nevada School of Medicine, Reno, Nevada; and Department of Molecular and Cellular Pharmacology, Graduate School of Pharmaceutical Sciences, Nagoya City University, Nagoya, Japan
| |
Collapse
|
21
|
Abstract
Papaverine, a vasodilator used as a therapeutic agent for a range of diseases, has been reported to increase the risk of occasional serious ventricular arrhythmias. To examine the mechanism for this effect, we herein tested the effects of papaverine on human ether-a-go-go (HERG) K channels expressed in HEK293 cells and Xenopus oocytes. Our results revealed that papaverine dose-dependently decreased the tail currents of HERG channel expressed in HEK293 cells with the IC50 and the Hill coefficient of 0.58 microM and 0.58, respectively, at +20 mV and 36 degrees C. The IC50 for the papaverine-induced blockade of HERG current in Xenopus oocytes was found to decrease progressively relative to depolarization (38.8, 30.0, and 24.8 microM at -10, +20, and +40 mV, respectively). The papaverine-induced blockade of HERG current was time-dependent; the fractional current was 0.92 +/- 0.03 of the control at the beginning of the pulse, but it declined to 0.18 +/- 0.06 after 6 seconds at a test potential of 0 mV. These results collectively indicate that papaverine blocks HERG channel in a concentration-, voltage-, and time-dependent manner. Two S6 domain mutations, Y652A and F656A, partially attenuated (Y652A) or abolished (F656A) the hERG current blockade, suggesting that papaverine blocks HERG channel at the pore of the channel. This was consistent with the computational simulation that showed papaverine interacts with Tyr652 and Phe656. Therefore, ventricular arrhythmias induced by papaverine could be resulted from the blockage of the HERG channel at the cardiac myocytes.
Collapse
|
22
|
Heitzmann D, Warth R. Physiology and pathophysiology of potassium channels in gastrointestinal epithelia. Physiol Rev 2008; 88:1119-82. [PMID: 18626068 DOI: 10.1152/physrev.00020.2007] [Citation(s) in RCA: 104] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Epithelial cells of the gastrointestinal tract are an important barrier between the "milieu interne" and the luminal content of the gut. They perform transport of nutrients, salts, and water, which is essential for the maintenance of body homeostasis. In these epithelia, a variety of K(+) channels are expressed, allowing adaptation to different needs. This review provides an overview of the current literature that has led to a better understanding of the multifaceted function of gastrointestinal K(+) channels, thereby shedding light on pathophysiological implications of impaired channel function. For instance, in gastric mucosa, K(+) channel function is a prerequisite for acid secretion of parietal cells. In epithelial cells of small intestine, K(+) channels provide the driving force for electrogenic transport processes across the plasma membrane, and they are involved in cell volume regulation. Fine tuning of salt and water transport and of K(+) homeostasis occurs in colonic epithelia cells, where K(+) channels are involved in secretory and reabsorptive processes. Furthermore, there is growing evidence for changes in epithelial K(+) channel expression during cell proliferation, differentiation, apoptosis, and, under pathological conditions, carcinogenesis. In the future, integrative approaches using functional and postgenomic/proteomic techniques will help us to gain comprehensive insights into the role of K(+) channels of the gastrointestinal tract.
Collapse
Affiliation(s)
- Dirk Heitzmann
- Institute of Physiology and Clinic and Policlinic for Internal Medicine II, Regensburg, Germany
| | | |
Collapse
|
23
|
Abstract
Smooth muscle cells (SMC) make up the muscular portion of the gastrointestinal (GI) tract from the distal oesophagus to the internal anal sphincter. Coordinated contractions of these cells produce the motor patterns of GI motility. Considerable progress was made during the last 20 years to understand the basic mechanisms controlling excitation-contraction (E-C) coupling. The smooth muscle motor is now understood in great molecular detail, and much has been learned about the mechanisms that deliver and recover Ca2+ during contractions. The majority of Ca2+ that initiates contractions comes from the external solution and is supplied by voltage-dependent Ca2+ channels (VDCC). VDCC are regulated largely by the effects of K+ and non-selective cation conductances (NSCC) on cell membrane potential and excitability. Ca2+ entry is supplemented by release of Ca2+ from IP(3) receptor-operated stores and by mechanisms that alter the sensitivity of the contractile apparatus to changes in cytoplasmic Ca2+. Molecular studies of the regulation of smooth muscle have been complicated by the plasticity of SMC and difficulties in culturing these cells without dramatic phenotypic changes. Major questions remain to be resolved regarding the details of E-C coupling in human GI smooth muscles. New discoveries regarding molecular expression that give GI smooth muscle their unique properties, the phenotypic changes that occur in SMC in GI motor disorders, tissue engineering approaches to repair or replace defective muscular regions, and molecular manipulations of GI smooth muscles in animals models and in cell culture will be topics for exciting investigations in the future.
Collapse
Affiliation(s)
- K M Sanders
- Department of Physiology and Cell Biology, University of Nevada School of Medicine, Reno, NV 89557, USA.
| |
Collapse
|
24
|
Voltage-gated K+ channel dysfunction in myocytes from a dog model of subarachnoid hemorrhage. J Cereb Blood Flow Metab 2008; 28:797-811. [PMID: 17987046 DOI: 10.1038/sj.jcbfm.9600577] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Delayed cerebral vasospasm after subarachnoid hemorrhage is primarily due to sustained contraction of arterial smooth muscle cells. Its pathogenesis remains unclear. The degree of arterial constriction is regulated by membrane potential that in turn is determined predominately by K+ conductance (GK). Here, we identified the main voltage-gated K+ (Kv) channels contributing to outward delayed rectifier currents in dog basilar artery smooth muscle as Kv2 class through a combination of electrophysiological and pharmacological methods. Kv2 current density was nearly halved in vasospastic myocytes after subarachnoid hemorrhage (SAH) in dogs, and Kv2.1 and Kv2.2 were downregulated in vasospastic myocytes when examined by quantitative mRNA, Western blotting, and immunohistochemistry. Vasospastic myocytes were depolarized and had a smaller contribution of GK toward maintenance of their membrane potential. Pharmacological block of Kv current in control myocytes mimicked the depolarization observed in vasospastic arteries. The degree of membrane depolarization was found to be compatible with the amount of vasoconstriction observed after SAH. We conclude that Kv2 dysfunction after SAH contributes to the pathogenesis of delayed cerebral vasospasm. This may confer a novel target for treatment of delayed cerebral vasospasm.
Collapse
|
25
|
Jindal HK, Folco EJ, Liu GX, Koren G. Posttranslational modification of voltage-dependent potassium channel Kv1.5: COOH-terminal palmitoylation modulates its biological properties. Am J Physiol Heart Circ Physiol 2008; 294:H2012-21. [PMID: 18344374 DOI: 10.1152/ajpheart.01374.2007] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The physiological function of ion channels is affected by protein-protein and protein-membrane interactions that modulate their activity and/or localization. Palmitoylation modulates protein function by facilitating targeted membrane association, interaction with other proteins, and determining subcellular localization. In this study, we demonstrate that the voltage-dependent potassium (Kv) channel Kv1.5 is palmitoylated and that the mutation of COOH-terminal cysteines is sufficient to abolish the palmitoylation of the Kv1.5 polypeptide in Chinese hamster ovary (CHO) cells. The labeling represented the thioester linkage of the labeled palmitic acid to cysteine rather than amide and oxygen ester linkages as judged by the release of the palmitic acid upon the treatment of the gel with hydroxylamine at a neutral pH. Site-directed mutagenesis and radiolabeling studies revealed that C593 was the sole site of palmitoylation. The elucidation of the biological function of palmitoylation revealed that the expression of the FLAG-Kv1.5 palmitoylation-deficient mutant (FL-Kv1.5(Palm-)) in stable CHO cells increased membrane expression as determined by the biotinylation of surface proteins and quantitative immunofluorescence analyses of these cells, in turn enhancing the outward potassium current. This enhanced surface expression and the currents were consequential to the slower rate of internalization, causing an increased localization of FL-Kv1.5(Palm-) in the plasma membrane compared with the wild-type FL-Kv1.5 channels. We conclude that the Kv1.5 channel is palmitoylated and that its palmitoylation modulates its biological functions and, therefore, might provide a physiological link between the metabolic state and the expression of Kv1.5 on the plasma membrane.
Collapse
Affiliation(s)
- Hitesh K Jindal
- Cardiovascular Research Center, Rhode Island Hospital, Brown University School of Medicine, 1 Hoppin Street, Providence, RI 02903, USA
| | | | | | | |
Collapse
|
26
|
Bonnet S, Archer SL. Potassium channel diversity in the pulmonary arteries and pulmonary veins: implications for regulation of the pulmonary vasculature in health and during pulmonary hypertension. Pharmacol Ther 2007; 115:56-69. [PMID: 17583356 DOI: 10.1016/j.pharmthera.2007.03.014] [Citation(s) in RCA: 70] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2007] [Accepted: 03/28/2007] [Indexed: 12/15/2022]
Abstract
This review describes the ionic heterogeneity manifest in the pulmonary circulation, particularly as it pertains to hypoxic pulmonary vasoconstriction (HPV) and pulmonary arterial hypertension (PAH). Heterogeneity in potassium (K(+)) channels, key regulators of vascular tone, cell proliferation, and apoptosis rates, contribute to the diverse response of vascular segments to hypoxia and to the localization of pathological changes in PAH. Pulmonary artery (PA) and pulmonary vein (PV) smooth muscle cells (SMC) express several K(+) channel families, including calcium-sensitive (KCa), voltage-gated (K(v)), inward rectifier (Kir), and 2-pore channels. Diversity is created by heterogeneous occurrence of alternatively spliced, mRNA species, assembly of heterotetrameric channels from diverse alpha-subunits, and association of channels with regulatory beta-subunits. Local heterogeneity in transcription factor activity may underlie differences in channel expression. Enrichment of resistance PASMCs with O(2)-sensitive K(+) channels, such as K(v)1.5, partially explains the greater HPV in resistance versus conduit PAs. In addition, resistance PAs are unique in having mitochondria which dynamically alter production of reactive O(2) species (ROS) in proportion to PO(2), thereby regulating K(+) channel activity and controlling expression through transcription factors, such as HIF-1alpha. In intraparenchymal PVs, a coaxial layer of cardiomyocytes encompasses a media of typical vascular SMCs. PV cardiomyocytes have rhythmic contraction and their Kir-enriched channels may be relevant to genesis of atrial arrhythmias and pulmonary edema. K(v) channel expression is decreased in PAH, leading to elevations of cytosolic K(+) and Ca(2+) that impair apoptosis and increase proliferation. Understanding ionic diversity may allow development of therapies that locally increase K(+) channel current and expression to treat PHT.
Collapse
Affiliation(s)
- Sébastien Bonnet
- Department of Medicine (Cardiology), University of Alberta, Edmonton, Canada
| | | |
Collapse
|
27
|
Remillard CV, Tigno DD, Platoshyn O, Burg ED, Brevnova EE, Conger D, Nicholson A, Rana BK, Channick RN, Rubin LJ, O'connor DT, Yuan JXJ. Function of Kv1.5 channels and genetic variations of KCNA5 in patients with idiopathic pulmonary arterial hypertension. Am J Physiol Cell Physiol 2007; 292:C1837-53. [PMID: 17267549 DOI: 10.1152/ajpcell.00405.2006] [Citation(s) in RCA: 119] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The pore-forming alpha-subunit, Kv1.5, forms functional voltage-gated K(+) (Kv) channels in human pulmonary artery smooth muscle cells (PASMC) and plays an important role in regulating membrane potential, vascular tone, and PASMC proliferation and apoptosis. Inhibited Kv channel expression and function have been implicated in PASMC from patients with idiopathic pulmonary arterial hypertension (IPAH). Here, we report that overexpression of the Kv1.5 channel gene (KCNA5) in human PASMC and other cell lines produced a 15-pS single channel current and a large whole cell current that was sensitive to 4-aminopyridine. Extracellular application of nicotine, bepridil, correolide, and endothelin-1 (ET-1) all significantly and reversibly reduced the Kv1.5 currents, while nicotine and bepridil also accelerated the inactivation kinetics of the currents. Furthermore, we sequenced KCNA5 from IPAH patients and identified 17 single-nucleotide polymorphisms (SNPs); 7 are novel SNPs. There are 12 SNPs in the upstream 5' region, 2 of which may alter transcription factor binding sites in the promoter, 2 nonsynonymous SNPs in the coding region, 2 SNPs in the 3'-untranslated region, and 1 SNP in the 3'-flanking region. Two SNPs may correlate with the nitric oxide-mediated decrease in pulmonary arterial pressure. Allele frequency of two other SNPs in patients with a history of fenfluramine and phentermine use was significantly different from patients who have never taken the anorexigens. These results suggest that 1) Kv1.5 channels are modulated by various agonists (e.g., nicotine and ET-1); 2) novel SNPs in KCNA5 are present in IPAH patients; and 3) SNPs in the promoter and translated regions of KCNA5 may underlie the altered expression and/or function of Kv1.5 channels in PASMC from IPAH patients.
Collapse
Affiliation(s)
- Carmelle V Remillard
- Department of Medicine, University of California--San Diego, 9500 Gilman Dr., MC 0725, La Jolla, CA 92093-0725, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Benson MD, Li QJ, Kieckhafer K, Dudek D, Whorton MR, Sunahara RK, Iñiguez-Lluhí JA, Martens JR. SUMO modification regulates inactivation of the voltage-gated potassium channel Kv1.5. Proc Natl Acad Sci U S A 2007; 104:1805-10. [PMID: 17261810 PMCID: PMC1794304 DOI: 10.1073/pnas.0606702104] [Citation(s) in RCA: 120] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
The voltage-gated potassium (Kv) channel Kv1.5 mediates the I(Kur) repolarizing current in human atrial myocytes and regulates vascular tone in multiple peripheral vascular beds. Understanding the complex regulation of Kv1.5 function is of substantial interest because it represents a promising pharmacological target for the treatment of atrial fibrillation and hypoxic pulmonary hypertension. Herein we demonstrate that posttranslational modification of Kv1.5 by small ubiquitin-like modifier (SUMO) proteins modulates Kv1.5 function. We have identified two membrane-proximal and highly conserved cytoplasmic sequences in Kv1.5 that conform to established SUMO modification sites in transcription factors. We find that Kv1.5 interacts specifically with the SUMO-conjugating enzyme Ubc9 and is a target for modification by SUMO-1, -2, and -3 in vivo. In addition, purified recombinant Kv1.5 serves as a substrate in a minimal in vitro reconstituted SUMOylation reaction. The SUMO-specific proteases SENP2 and Ulp1 efficiently deconjugate SUMO from Kv1.5 in vivo and in vitro, and disruption of the two identified target motifs results in a loss of the major SUMO-conjugated forms of Kv1.5. In whole-cell patch-clamp electrophysiological studies, loss of Kv1.5 SUMOylation, by either disruption of the conjugation sites or expression of the SUMO protease SENP2, leads to a selective approximately 15-mV hyperpolarizing shift in the voltage dependence of steady-state inactivation. Reversible control of voltage-sensitive channels through SUMOylation constitutes a unique and likely widespread mechanism for adaptive tuning of the electrical excitability of cells.
Collapse
Affiliation(s)
- Mark D. Benson
- Department of Pharmacology, University of Michigan Medical School, Ann Arbor, MI 48109-0632
| | - Qiu-Ju Li
- Department of Pharmacology, University of Michigan Medical School, Ann Arbor, MI 48109-0632
| | - Katherine Kieckhafer
- Department of Pharmacology, University of Michigan Medical School, Ann Arbor, MI 48109-0632
| | - David Dudek
- Department of Pharmacology, University of Michigan Medical School, Ann Arbor, MI 48109-0632
| | - Matthew R. Whorton
- Department of Pharmacology, University of Michigan Medical School, Ann Arbor, MI 48109-0632
| | - Roger K. Sunahara
- Department of Pharmacology, University of Michigan Medical School, Ann Arbor, MI 48109-0632
| | - Jorge A. Iñiguez-Lluhí
- Department of Pharmacology, University of Michigan Medical School, Ann Arbor, MI 48109-0632
- To whom correspondence may be addressed. E-mail:
or
| | - Jeffrey R. Martens
- Department of Pharmacology, University of Michigan Medical School, Ann Arbor, MI 48109-0632
- To whom correspondence may be addressed. E-mail:
or
| |
Collapse
|
29
|
Kwak YG, Kim DK, Ma TZ, Park SA, Park H, Jung YH, Yoo DJ, Eun JS. Torilin fromTorilis japonica (Houtt.) DC. Blocks hKv1.5 channel current. Arch Pharm Res 2006; 29:834-9. [PMID: 17121176 DOI: 10.1007/bf02973902] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
Torilin was purified from Torilis japonica (Houtt.) DC., and its effects on a rapidly activating delayed rectifier K+ channel (hKv1.5), cloned from human heart and stably expressed in Ltk- cells, as well as the corresponding K+ current (the ultrarapid delayed rectifier, I(KUR)) were assessed in human atrial myocytes. Using the whole cell configuration of the patch-clamp technique, torilin was found to inhibit the hKv1.5 current in time and voltage-dependent manners, with an IC50 value of 2.51+/-0.34 microM at +60 mV. Torilin accelerated the inactivation kinetics of the hKv1.5 channel, and slowed the deactivation kinetics of the hKv1.5 current, resulting in a tail crossover phenomenon. Additionally, torilin inhibited the hKv1.5 current in a use-dependent manner. These results strongly suggest that torilin is a type of open-channel blocker of the hKv1.5 channel.
Collapse
Affiliation(s)
- Yong Geun Kwak
- Department of Pharmacology, Chonbuk National University Medical School, Chonju, 561-756, Korea
| | | | | | | | | | | | | | | |
Collapse
|
30
|
Hayama E, Imamura SI, Wu C, Nakazawa M, Matsuoka R, Nakanishi T. Analysis of voltage-gated potassium channel beta1 subunits in the porcine neonatal ductus arteriosus. Pediatr Res 2006; 59:167-74. [PMID: 16439573 DOI: 10.1203/01.pdr.0000196736.89742.c1] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
The voltage-gated potassium channels (Kv) are partially responsible for the contraction/relaxation of blood vessels in response to changes in the Po(2) level. The present study determined the expression of Kvbeta1 and four oxygen-sensitive Kvalpha subunits (Kv1.2, Kv1.5, Kv2.1, and Kv9.3) in the ductus arteriosus (DA), the aorta (Ao), and the pulmonary artery (PA) in porcine neonates immediately after birth. We cloned three Kvbeta1 transcript variants (Kvbeta1.2, Kvbeta1.3, and Kvbeta1.4), Kv1.2, Kv1.5, and Kv9.3 from piglets. Three Kvbeta1 transcripts, Kv1.2, Kv1.5, and Kv9.3, encode predicted proteins of 401, 408, 202, 499, 600, and 491 residues. These Kv showed a high degree of sequence conservation with the corresponding Kv in human. Northern and quantitative real-time PCR (qr-PCR) analyses showed that Kvbeta1.2 expression was high in the DA and Ao but low in the PA. Kv1.5 expression was high in the Ao and PA but low in the DA. Expression of Kvbeta1.3, Kvbeta1.4, Kv1.2, Kv2.1, and Kv9.3 was low in these blood vessels. The inactivation property of Kvbeta1.2 against Kv1.5 was confirmed using Xenopus laevis oocytes. Our findings suggest that the molecular basis for the differential electrophysiological characteristics including opposing response to oxygen in the DA and the PA are partially due to diversity in expression of Kv1.5 and Kvbeta1.2 subunits. The high expression of Kvbeta1.2 and relatively low expression of Kv1.5 in the DA might be partially responsible for the ductal closure after birth.
Collapse
Affiliation(s)
- Emiko Hayama
- Department of Pediatric Cardiology, The Heart Institute of Japan, Tokyo Women's Medical University
| | | | | | | | | | | |
Collapse
|
31
|
Plante I, Fournier D, Ricard G, Drolet B, O'Hara G, Champagne J, Mathieu P, Baillot R, Daleau P. Electrophysiological characterization of three non-synonymous single nucleotide polymorphisms (R87Q, A251T, and P307S) found in hKv1.5. Pflugers Arch 2006; 452:316-23. [PMID: 16411137 DOI: 10.1007/s00424-005-0031-8] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2005] [Accepted: 12/01/2005] [Indexed: 10/25/2022]
Abstract
Non-synonymous single nucleotide polymorphisms (SNPs) in the KCNA5/hKv1.5 gene, which encodes for a voltage-gated K+ channel responsible for the I (Kur) current in the human atria, have been recently reported. To gain further knowledge on potential influence of hKv1.5 SNPs, we searched for their presence in a specific population of 96 French-Canadians and characterized electrophysiological properties of the variants in two cell lines. The presumed promoter (-83 bp) and coding regions were sequenced. We found three heterozygous SNPs: R87Q, A251T, and P307S. Functional analysis of SNPs transfected in Chinese hamster ovary (CHO) cells showed that both R87Q and P307S diminished the inactivation amplitude (e.g., at +60 mV, amplitudes were 89+/-26, 23+/-4, and 22+/-7 pA/pF for the wild type, R87Q and P307S, respectively; n=8, 6, and 8, respectively). Inactivation was slowed with these variants (e.g., tau (fast) at +50 mV were 270+/-48, 490+/-66, and 340+/-45 ms for the wild type, R87Q, and P307S, respectively) while R87Q additionally accelerated the rate of hKv1.5 channel opening. A dominant-negative effect was observed for R87Q but not for P307S. SNPs properties were not reproduced when expressed in the HEK293 cell line, suggesting that the regulatory beta-subunit present in CHO cells (and the human heart) is essential for the SNPs effects that we have observed.
Collapse
Affiliation(s)
- Isabelle Plante
- Quebec Heart Institute, Laval Hospital Research Centre, and Faculty of Pharmacy, Laval University, Sainte-Foy, Quebec, G1V 4G5, Canada
| | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Rezazadeh S, Claydon TW, Fedida D. KN-93 (2-[N-(2-Hydroxyethyl)]-N-(4-methoxybenzenesulfonyl)]amino-N-(4-chlorocinnamyl)-N-methylbenzylamine), a Calcium/Calmodulin-Dependent Protein Kinase II Inhibitor, Is a Direct Extracellular Blocker of Voltage-Gated Potassium Channels. J Pharmacol Exp Ther 2005; 317:292-9. [PMID: 16368898 DOI: 10.1124/jpet.105.097618] [Citation(s) in RCA: 93] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
The effect of Ca(2+)/calmodulin-dependent protein kinase II (CaMK II) on voltage-gated ion channels is widely studied through the use of specific CaMK II blockers such as 2-[N-(2-hydroxyethyl)]-N-(4methoxybenzenesulfonyl)]amino-N-(4-chlorocinnamyl)-N-methylbenzylamine (KN-93). The present study demonstrates that KN-93 is a direct extracellular blocker of a wide range of cloned Kv channels from a number of different subfamilies. In all channels tested, the effect of 1 microM KN-93 was independent of CaMK II because 1 microM2-[N-(4-methoxybenzenesulfonyl)]amino-N-(4-chlorocinnamyl)-N-methylbenzylamine, phosphate (KN-92), an inactive analog of KN-93, caused similar inhibition of currents. In addition, dialysis of cells with 10 microM CaMK II inhibitory peptide fragment 281-301 (CIP) had no effect on current kinetics and did not prevent the inhibitory effect of KN-93. The IC(50) for block of the Kv1.5 channel (used as an example to determine the nature of KN-93 block) was 307 +/- 12 nM. KN-93 blocked open channels with little voltage dependence that did not alter the V(1/2) of channel activation. Removal of P/C-type inactivation by mutation of arginine 487 to valine in the outer pore region of Kv1.5 (R487V) greatly reduced KN-93 block, whereas enhancement of inactivation induced by mutation of threonine 462 to cysteine (T462C) increased the potency of KN-93 by 4-fold. This suggested that KN-93 acted through promotion and stabilization of C-type inactivation. Importantly, KN-93 was ineffective as a blocker when applied intracellularly, suggesting that CaMK II-independent effects of KN-93 on Kv channels can be circumvented by intracellular application of KN-93.
Collapse
Affiliation(s)
- Saman Rezazadeh
- Department of Cellular and Physiological Sciences, University of British Columbia, Vancouver, Canada
| | | | | |
Collapse
|
33
|
Liu L, Hansen DR, Kim I, Gilbertson TA. Expression and characterization of delayed rectifying K+channels in anterior rat taste buds. Am J Physiol Cell Physiol 2005; 289:C868-80. [PMID: 15930148 DOI: 10.1152/ajpcell.00115.2005] [Citation(s) in RCA: 49] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Delayed rectifying K+(DRK) channels in taste cells have been implicated in the regulation of cell excitability and as potential targets for direct and indirect modulation by taste stimuli. In the present study, we have used patch-clamp recording to determine the biophysical properties and pharmacological sensitivity of DRK channels in isolated rat fungiform taste buds. Molecular biological assays at the taste bud and single-cell levels are consistent with the interpretation that taste cells express a variety of DRK channels, including members from each of the three major subfamilies: KCNA, KCNB, and KCNC. Real-time PCR assays were used to quantify expression of the nine DRK channel subtypes. While taste cells express a number of DRK channels, the electrophysiological and molecular biological assays indicate that the Shaker Kv1.5 channel (KCNA5) is the major functional DRK channel expressed in the anterior rat tongue.
Collapse
Affiliation(s)
- Lidong Liu
- Department of Biology and The Center for Integrated BioSystems, Utah State University, 5305 Old Main Hill, Logan, Utah 84322-5305, USA
| | | | | | | |
Collapse
|
34
|
Li B, Gallin WJ. Computational identification of residues that modulate voltage sensitivity of voltage-gated potassium channels. BMC STRUCTURAL BIOLOGY 2005; 5:16. [PMID: 16111489 PMCID: PMC1208917 DOI: 10.1186/1472-6807-5-16] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/19/2005] [Accepted: 08/19/2005] [Indexed: 01/29/2023]
Abstract
Background Studies of the structure-function relationship in proteins for which no 3D structure is available are often based on inspection of multiple sequence alignments. Many functionally important residues of proteins can be identified because they are conserved during evolution. However, residues that vary can also be critically important if their variation is responsible for diversity of protein function and improved phenotypes. If too few sequences are studied, the support for hypotheses on the role of a given residue will be weak, but analysis of large multiple alignments is too complex for simple inspection. When a large body of sequence and functional data are available for a protein family, mature data mining tools, such as machine learning, can be applied to extract information more easily, sensitively and reliably. We have undertaken such an analysis of voltage-gated potassium channels, a transmembrane protein family whose members play indispensable roles in electrically excitable cells. Results We applied different learning algorithms, combined in various implementations, to obtain a model that predicts the half activation voltage of a voltage-gated potassium channel based on its amino acid sequence. The best result was obtained with a k-nearest neighbor classifier combined with a wrapper algorithm for feature selection, producing a mean absolute error of prediction of 7.0 mV. The predictor was validated by permutation test and evaluation of independent experimental data. Feature selection identified a number of residues that are predicted to be involved in the voltage sensitive conformation changes; these residues are good target candidates for mutagenesis analysis. Conclusion Machine learning analysis can identify new testable hypotheses about the structure/function relationship in the voltage-gated potassium channel family. This approach should be applicable to any protein family if the number of training examples and the sequence diversity of the training set that are necessary for robust prediction are empirically validated. The predictor and datasets can be found at the VKCDB web site [1].
Collapse
Affiliation(s)
- Bin Li
- Department of Biological Sciences, University of Alberta, Edmonton, Canada T6G 2E9
- Partners AIDS Research Center, Massachusetts General Hospital, Harvard Medical School, 149 13th Street 6th floor, Charlestown MA USA 02129
| | - Warren J Gallin
- Department of Biological Sciences, University of Alberta, Edmonton, Canada T6G 2E9
- Department of Cell Biology, University of Alberta, Edmonton, Alberta, Canada
| |
Collapse
|
35
|
Choi BH, Park JA, Kim KR, Lee GI, Lee YT, Choe H, Ko SH, Kim MH, Seo YH, Kwak YG. Direct block of cloned hKv1.5 channel by cytochalasins, actin-disrupting agents. Am J Physiol Cell Physiol 2005; 289:C425-36. [PMID: 15800051 DOI: 10.1152/ajpcell.00450.2004] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The action of cytochalasins, actin-disrupting agents on human Kv1.5 channel (hKv1.5) stably expressed in Ltk−cells was investigated using the whole cell patch-clamp technique. Cytochalasin B inhibited hKv1.5 currents rapidly and reversibly at +60 mV in a concentration-dependent manner with an IC50of 4.2 μM. Cytochalasin A, which has a structure very similar to cytochalasin B, inhibited hKv1.5 (IC50of 1.4 μM at +60 mV). Pretreatment with other actin filament disruptors cytochalasin D and cytochalasin J, and an actin filament stabilizing agent phalloidin had no effect on the cytochalasin B-induced inhibition of hKv1.5 currents. Cytochalasin B accelerated the decay rate of inactivation for the hKv1.5 currents. Cytochalasin B-induced inhibition of the hKv1.5 channels was voltage dependent with a steep increase over the voltage range of the channel's opening. However, the inhibition exhibited voltage independence over the voltage range in which channels are fully activated. Cytochalasin B produced no significant effect on the steady-state activation or inactivation curves. The rate constants for association and dissociation of cytochalasin B were 3.7 μM/s and 7.5 s−1, respectively. Cytochalasin B produced a use-dependent inhibition of hKv1.5 current that was consistent with the slow recovery from inactivation in the presence of the drug. Cytochalasin B (10 μM) also inhibited an ultrarapid delayed rectifier K+current ( IK,ur) in human atrial myocytes. These results indicate that cytochalasin B primarily blocks activated hKv1.5 channels and endogenous IK,urin a cytoskeleton-independent manner as an open-channel blocker.
Collapse
Affiliation(s)
- Bok Hee Choi
- Department of Pharmacology, Chonbuk National University Medical School, Chonju, Chonbuk 561-180, Republic of Korea
| | | | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Cole WC, Chen TT, Clément-Chomienne O. Myogenic regulation of arterial diameter: role of potassium channels with a focus on delayed rectifier potassium current. Can J Physiol Pharmacol 2005; 83:755-65. [PMID: 16333377 DOI: 10.1139/y05-082] [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] [Indexed: 01/12/2023]
Abstract
The phenomenon of myogenic constriction of arterial resistance vessels in response to increased intraluminal pressure has been known for over 100 years, yet our understanding of the molecular mechanisms involved remains incomplete. The focus of this paper concerns the potassium (K+) channels that provide a negative feedback control of the myogenic depolarization of vascular smooth muscle cells that is provoked by elevations in intraluminal pressure, and specifically, the contribution of delayed rectifier (KDR) channels. Our knowledge of the important role played by KDR channels, as well as their molecular identity and acute modulation via changes in gating, has increased dramatically in recent years. Several lines of evidence point to a crucial contribution by heteromultimeric KV1 subunit-containing KDR channels in the control of arterial diameter and myogenic reactivity, but other members of the KV superfamily are also expressed by vascular myocytes, and less is known concerning their specific functions. The effect of pharmacological modulation of KDR channels is discussed, with particular reference to the actions of anorexinogens on KV1- and KV2-containing KDR channels. Finally, the need for a greater understanding of the mechanisms that control KDR channel gene expression is stressed in light of evidence indicating that there is a reduced expression of KDR channels in diseases associated with abnormal myogenic reactivity and vascular remodelling.Key words: resistance arteries, myogenic response, potassium channels, delayed rectifier K+ current, KV channels, KV1, KV2.
Collapse
Affiliation(s)
- William C Cole
- The Smooth Muscle Research Group, Department of Pharmacology & Therapeutics, Universityk of Calgary, Calgary, Canada.
| | | | | |
Collapse
|
37
|
Cho SY, Beckett EA, Baker SA, Han I, Park KJ, Monaghan K, Ward SM, Sanders KM, Koh SD. A pH-sensitive potassium conductance (TASK) and its function in the murine gastrointestinal tract. J Physiol 2005; 565:243-59. [PMID: 15774516 PMCID: PMC1464505 DOI: 10.1113/jphysiol.2005.084574] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
Abstract
The excitability of smooth muscles is regulated, in part, by background K+ conductances that determine resting membrane potential. However, the K+ conductances so far described in gastrointestinal (GI) muscles are not sufficient to explain the negative resting potentials of these cells. Here we describe expression of two-pore K+ channels of the TASK family in murine small and large intestinal muscles. TASK-2, cloned from murine intestinal muscles, resulted in a pH-sensitive, time-dependent, non-inactivating K+ conductance with slow activation kinetics. A similar conductance was found in native intestinal myocytes using whole-cell patch-clamp conditions. The pH-sensitive current was blocked by local anaesthetics. Lidocaine, bupivacaine and acidic pH depolarized circular muscle cells in intact muscles and decreased amplitude and frequency of slow waves. The effects of lidocaine were not blocked by tetraethylammonium chloride, 4-aminopyridine, glibenclamide, apamin or MK-499. However, depolarization by acidic pH was abolished by pre-treatment with lidocaine, suggesting that lidocaine-sensitive K+ channels were responsible for pH-sensitive changes in membrane potential. The kinetics of activation, sensitivity to pH, and pharmacology of the conductance in intestinal myocytes and the expression of TASK-1 and TASK-2 in these cells suggest that the pH-sensitive background conductance is encoded by TASK genes. This conductance appears to contribute significantly to resting potential and may regulate excitability of GI muscles.
Collapse
Affiliation(s)
- Sang Yun Cho
- Department of Physiology and Cell Biology, University of Nevada Reno, School of Medicine, Reno, NV 89557, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Hebert SC, Desir G, Giebisch G, Wang W. Molecular diversity and regulation of renal potassium channels. Physiol Rev 2005; 85:319-71. [PMID: 15618483 PMCID: PMC2838721 DOI: 10.1152/physrev.00051.2003] [Citation(s) in RCA: 236] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
K(+) channels are widely distributed in both plant and animal cells where they serve many distinct functions. K(+) channels set the membrane potential, generate electrical signals in excitable cells, and regulate cell volume and cell movement. In renal tubule epithelial cells, K(+) channels are not only involved in basic functions such as the generation of the cell-negative potential and the control of cell volume, but also play a uniquely important role in K(+) secretion. Moreover, K(+) channels participate in the regulation of vascular tone in the glomerular circulation, and they are involved in the mechanisms mediating tubuloglomerular feedback. Significant progress has been made in defining the properties of renal K(+) channels, including their location within tubule cells, their biophysical properties, regulation, and molecular structure. Such progress has been made possible by the application of single-channel analysis and the successful cloning of K(+) channels of renal origin.
Collapse
Affiliation(s)
- Steven C Hebert
- Department of Cellular and Molecular Physiology, Yale University School of Medicine, New Haven, Connecticut 06520-8026, USA.
| | | | | | | |
Collapse
|
39
|
Archer SL, Wu XC, Thébaud B, Nsair A, Bonnet S, Tyrrell B, McMurtry MS, Hashimoto K, Harry G, Michelakis ED. Preferential expression and function of voltage-gated, O2-sensitive K+ channels in resistance pulmonary arteries explains regional heterogeneity in hypoxic pulmonary vasoconstriction: ionic diversity in smooth muscle cells. Circ Res 2004; 95:308-18. [PMID: 15217912 DOI: 10.1161/01.res.0000137173.42723.fb] [Citation(s) in RCA: 141] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Hypoxic pulmonary vasoconstriction (HPV) is initiated by inhibition of O2-sensitive, voltage-gated (Kv) channels in pulmonary arterial smooth muscle cells (PASMCs). Kv inhibition depolarizes membrane potential (E(M)), thereby activating Ca2+ influx via voltage-gated Ca2+ channels. HPV is weak in extrapulmonary, conduit pulmonary arteries (PA) and strong in precapillary resistance arteries. We hypothesized that regional heterogeneity in HPV reflects a longitudinal gradient in the function/expression of PASMC O2-sensitive Kv channels. In adult male Sprague Dawley rats, constrictions to hypoxia, the Kv blocker 4-aminopyridine (4-AP), and correolide, a Kv1.x channel inhibitor, were endothelium-independent and greater in resistance versus conduit PAs. Moreover, HPV was dependent on Kv-inhibition, being completely inhibited by pretreatment with 4-AP. Kv1.2, 1.5, Kv2.1, Kv3.1b, Kv4.3, and Kv9.3. mRNA increased as arterial caliber decreased; however, only Kv1.5 protein expression was greater in resistance PAs. Resistance PASMCs had greater K+ current (I(K)) and a more hyperpolarized E(M) and were uniquely O2- and correolide-sensitive. The O2-sensitive current (active at -65 mV) was resistant to iberiotoxin, with minimal tityustoxin sensitivity. In resistance PASMCs, 4-AP and hypoxia inhibited I(K) 57% and 49%, respectively, versus 34% for correolide. Intracellular administration of anti-Kv1.5 antibodies inhibited correolide's effects. The hypoxia-sensitive, correolide-insensitive I(K) (15%) was conducted by Kv2.1. Anti-Kv1.5 and anti-Kv2.1 caused additive depolarization in resistance PASMCs (Kv1.5>Kv2.1) and inhibited hypoxic depolarization. Heterologously expressed human PASMC Kv1.5 generated an O2- and correolide-sensitive I(K) like that in resistance PASMCs. In conclusion, Kv1.5 and Kv2.1 account for virtually all the O2-sensitive current. HPV occurs in a Kv-enriched resistance zone because resistance PASMCs preferentially express O2-sensitive Kv-channels.
Collapse
MESH Headings
- 4-Aminopyridine/pharmacology
- Acetylcholine/pharmacology
- Animals
- Cell Hypoxia
- Cells, Cultured/drug effects
- Cells, Cultured/physiology
- Gene Expression Regulation
- Humans
- Hypoxia/physiopathology
- Ion Channel Gating/drug effects
- Ion Transport/drug effects
- Kv1.5 Potassium Channel
- Male
- Membrane Potentials/drug effects
- Muscle, Smooth, Vascular/pathology
- Myocytes, Smooth Muscle/drug effects
- Myocytes, Smooth Muscle/physiology
- Oxygen/pharmacology
- Patch-Clamp Techniques
- Peptides/pharmacology
- Potassium/metabolism
- Potassium Channels, Voltage-Gated/biosynthesis
- Potassium Channels, Voltage-Gated/genetics
- Potassium Channels, Voltage-Gated/physiology
- Pulmonary Artery/pathology
- Pulmonary Circulation/drug effects
- Pulmonary Circulation/physiology
- Rats
- Rats, Sprague-Dawley
- Recombinant Fusion Proteins/physiology
- Scorpion Venoms/pharmacology
- Shab Potassium Channels
- Transduction, Genetic
- Triterpenes/pharmacology
- Vascular Resistance/drug effects
- Vascular Resistance/physiology
- Vasoconstriction/drug effects
- Vasoconstriction/physiology
Collapse
Affiliation(s)
- Stephen L Archer
- Heart and Stroke Chair in Cardiovascular Research, Cardiology Division, Department of Medicine, University of Alberta, WMC 2C2.36, 8440 112th St, Edmonton, Alberta T6G 2B7, Canada.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Archer SL, Wu XC, Thébaud B, Moudgil R, Hashimoto K, Michelakis ED. O2 sensing in the human ductus arteriosus: redox-sensitive K+ channels are regulated by mitochondria-derived hydrogen peroxide. Biol Chem 2004; 385:205-16. [PMID: 15134333 DOI: 10.1515/bc.2004.014] [Citation(s) in RCA: 48] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
The ductus arteriosus (DA) is a fetal artery that allows blood ejected from the right ventricle to bypass the pulmonary circulation in utero. At birth, functional closure of the DA is initiated by an O2-induced, vasoconstrictor mechanism which, though modulated by endothelial-derived endothelin and prostaglandins, is intrinsic to the smooth muscle cell (DASMC) [Michelakis et al., Circ. Res. 91 (2002); pp. 478-486]. As pO2 increases, a mitochondrial O2-sensor (electron transport chain complexes I or III) is activated, which generates a diffusible redox mediator (H2O2). H2O2 inhibits voltage-gated K+ channels (Kv) in DASMC. The resulting membrane depolarization activates L-type Ca2+ channels, thereby promoting vasoconstriction. Conversely, inhibiting mitochondrial ETC complexes I or III mimics hypoxia, depolarizing mitochondria, and decreasing H2O2 levels. The resulting increase in K+ current hyperpolarizes the DASMC and relaxes the DA. We have developed two models for study of the DA's O2-sensor pathway, both characterized by decreased O2-constriction and Kv expression: (i) preterm rabbit DA, (ii) ionically-remodeled, human term DA. The O2-sensitive channels Kv1.5 and Kv2.1 are important to DA O2-constriction and overexpression of either channel enhances DA constriction in these models. Understanding this O2-sensing pathway offers therapeutic targets to modulate the tone and patency of human DA in vivo, thereby addressing a common form of congenital heart disease in preterm infants.
Collapse
Affiliation(s)
- Stephen L Archer
- Vascular Biology Group, University of Alberta, WMC 2C2.36, 8440 112th Street, Edmonton, Alberta T6G 2B7, Canada.
| | | | | | | | | | | |
Collapse
|
41
|
Aihara Y, Jahromi BS, Yassari R, Nikitina E, Agbaje-Williams M, Macdonald RL. Molecular profile of vascular ion channels after experimental subarachnoid hemorrhage. J Cereb Blood Flow Metab 2004; 24:75-83. [PMID: 14688619 DOI: 10.1097/01.wcb.0000095803.98378.d8] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Cerebral vasospasm is a transient, delayed constriction of cerebral arteries that occurs after subarachnoid hemorrhage (SAH). Smooth muscle cells show impaired relaxation after SAH, which may be caused by a defect in the ionic mechanisms regulating smooth muscle membrane potential and Ca(2+) permeability. We tested this hypothesis by examining changes in expression of mRNA and protein for ion channels in the basilar arteries of dogs after SAH using quantitative real-time polymerase chain reaction (PCR) and western blotting. SAH was associated with a significant reduction in basilar artery diameter to 41 +/- 8% of pre-SAH diameter (P < 0.001) after 7 days. There was significant downregulation of the voltage-gated K(+) channel K(v) 2.2 (65% reduction in mRNA, P < 0.001; 49% reduction in protein, P < 0.05) and the beta1 subunit of the large-conductance, Ca(2+) - activated K(+) (BK) channel (53% reduction in mRNA, P < 0.02). There was no change in BK beta1 subunit protein. Changes in mRNA levels of K(v) 2.2 and the BK-beta1 subunit correlated with the degree of vasospasm (r(2) = 0.490 and 0.529 respectively, P < 0.05). The inwardly rectifying K(+) (K(ir)) channel K(ir) 2.1 was upregulated (234% increase in mRNA, P < 0.001; 350% increase in protein, P < 0.001). There was no significant change in mRNA expression of L- type Ca(2+) channels and the BK-alpha subunit. These data suggest that K(+) channel dysfunction may contribute to the pathogenesis of cerebral vasospasm.
Collapse
Affiliation(s)
- Yasuo Aihara
- Department of Surgery, University of Chicago and Pritzker School of Medicine, Chicago, IL 60637, USA
| | | | | | | | | | | |
Collapse
|
42
|
Abstract
Most antiarrhythmic drugs are ion channel blockers, and to date, those tested in large randomized placebo-controlled clinical trials have shown no decrease in mortality outcome. This apparent lack of survival benefit may result from the significant liabilities associated with these agents that offset any long-term benefit. Despite the current success of implantable defibrillators and the future promise of gene therapy, there is still a pressing need for new antiarrhythmic drugs. An improved understanding of cardiac ion channels and novel approaches to target selection and compound screening will provide new opportunities for drug discovery in the near future. Here, we briefly review the multiple mechanisms of arrhythmia, the history of drug failures, and the possibilities that evolving technologies may provide in the search for more efficacious and safer antiarrhythmic drugs.
Collapse
Affiliation(s)
- Michael C Sanguinetti
- Department of Physiology, Eccles Institute of Human Genetics, University of Utah, 15 N 2030 E, Room 4220, Salt Lake City, UT 84112, USA.
| | | |
Collapse
|
43
|
Thorneloe KS, Nelson MT. Properties and molecular basis of the mouse urinary bladder voltage-gated K+ current. J Physiol 2003; 549:65-74. [PMID: 12679374 PMCID: PMC2342925 DOI: 10.1113/jphysiol.2003.039859] [Citation(s) in RCA: 62] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Potassium channels play an important role in controlling the excitability of urinary bladder smooth muscle (UBSM). Here we describe the biophysical, pharmacological and molecular properties of the mouse UBSM voltage-gated K+ current (IK(V)). The IK(V) activated, deactivated and inactivated slowly with time constants of 29.9 ms at +30 mV, 131 ms at -40 mV and 3.4 s at +20 mV. The midpoints of steady-state activation and inactivation curves were 1.1 mV and -61.4 mV, respectively. These properties suggest that IK(V) plays a role in regulating the resting membrane potential and contributes to the repolarization and after-hyperpolarization phases of action potentials. The IK(V) was blocked by tetraethylammonium ions with an IC50 of 5.2 mM and was unaffected by 1 mM 4-aminopyridine. RT-PCR for voltage-gated K+ channel (KV) subunits revealed the expression of Kv2.1, Kv5.1, Kv6.1, Kv6.2 and Kv6.3 in isolated UBSM myocytes. A comparison of the biophysical properties of UBSM IK(V) with those reported for Kv2.1 and Kv5.1 and/or Kv6 heteromultimeric channels demonstrated a marked similarity. We propose that heteromultimeric channel complexes composed of Kv2.1 and Kv5.1 and/or Kv6 subunits form the molecular basis of the mouse UBSM IK(V).
Collapse
Affiliation(s)
- Kevin S Thorneloe
- Department of Pharmacology, College of Medicine, University of Vermont, Burlington 05405, USA
| | | |
Collapse
|
44
|
Choe H, Lee YK, Lee YT, Choe H, Ko SH, Joo CU, Kim MH, Kim GS, Eun JS, Kim JH, Chae SW, Kwak YG. Papaverine blocks hKv1.5 channel current and human atrial ultrarapid delayed rectifier K+ currents. J Pharmacol Exp Ther 2003; 304:706-12. [PMID: 12538825 DOI: 10.1124/jpet.102.042770] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Papaverine, 1-[(3,4-dimethoxyphenyl)methyl]-6,-7-dimethoxyisoquinoline, has been used as a vasodilator agent and a therapeutic agent for cerebral vasospasm, renal colic, and penile impotence. We examined the effects of papaverine on a rapidly activating delayed rectifier K(+) channel (hKv1.5) cloned from human heart and stably expressed in Ltk(-) cells as well as a corresponding K(+) current (the ultrarapid delayed rectifier, I(Kur)) in human atrial myocytes. Using the whole cell configuration of the patch-clamp technique, we found that papaverine inhibited hKv1.5 current in a time- and voltage-dependent manner with an IC(50) value of 43.4 microM at +60 mV. Papaverine accelerated the kinetics of the channel inactivation, suggesting the blockade of open channels. Papaverine (100 microM) also blocked I(Kur) in human atrial myocytes. These results indicate that papaverine blocks hKv1.5 channels and native hKv1.5 channels in a concentration-, voltage-, state-, and time-dependent manner. This interaction suggests that papaverine could alter cardiac excitability in vivo.
Collapse
Affiliation(s)
- Han Choe
- Department of Physiology, Ulsan University College of Medicine, Seoul, South Korea
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Adegunloye B, Lamarre E, Moreland RS. Quinine inhibits vascular contraction independent of effects on calcium or myosin phosphorylation. J Pharmacol Exp Ther 2003; 304:294-300. [PMID: 12490604 DOI: 10.1124/jpet.102.042101] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
This report contains results of studies designed to determine whether quinine has direct effects on myofilament Ca2+ sensitization in addition to effects on Ca2+. Quinine decreased the EC50 value and maximal contraction of intact arterial strips to histamine. Incubation of arterial strips with indomethacin or 1H-[1,2,4]oxadiazole[4,3-alpha]quinoxalin-1-one did not alter quinine inhibition, suggesting that the effect is not mediated via cyclooxygenase or cGMP. Pretreatment of strips with quinine had no effect on the histamine-dependent increases in myosin light chain phosphorylation levels. Quinine inhibited Ca2+-induced contraction in alpha-toxin permeabilized strips, but not the Ca2+-induced contraction in Triton X-100 permeabilized strips. Pretreatment of the alpha-toxin permeabilized strips with quinine before stimulation with guanosine-5'-O-(3-thio)triphosphate (GTPgammaS) did not have any effect on the response. In conclusion, quinine inhibited Ca2+-dependent contractions of the alpha-toxin permeabilized strips, which retain modulatory pathways both upstream and downstream from the contractile proteins but did not inhibit GTPgammaS-dependent contraction of the alpha-toxin permeabilized preparation important in upstream modulation of the contraction. Moreover, quinine did not inhibit the Ca2+-dependent contractions of the Triton X-100 permeabilized strips, which are devoid of all modulatory pathways. This suggests that quinine does not act upstream from or directly on the contractile proteins. A more likely site of action may be downstream of the contractile proteins and specifically at the coupling of the contractile proteins with the physiological endpoint of force development.
Collapse
Affiliation(s)
- Banji Adegunloye
- Department of Pharmacology and Physiology, Drexel University College of Medicine, Philadelphia, Pennsylvania 19102, USA
| | | | | |
Collapse
|
46
|
Hogg DS, Davies ARL, McMurray G, Kozlowski RZ. K(V)2.1 channels mediate hypoxic inhibition of I(KV) in native pulmonary arterial smooth muscle cells of the rat. Cardiovasc Res 2002; 55:349-60. [PMID: 12123774 DOI: 10.1016/s0008-6363(02)00411-x] [Citation(s) in RCA: 19] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
OBJECTIVE To determine whether, in native pulmonary arterial smooth muscle cells (PASMC), K(V)2.1 delayed-rectifying K(+) channels are central to the process of hypoxic pulmonary vasoconstriction. METHODS In this study, we tested for the presence of K(V)2.1 channel transcripts in rat small pulmonary arteries using RT-PCR, and for the protein itself using immunolocalisation. The contribution of K(V)2.1 channels to whole-cell K(V) currents (I(KV)) and their role in hypoxic inhibition of I(KV) in native PASMC was investigated utilising patch-clamp recordings. RESULTS K(V)2.1 mRNA expression and AbK(V)2.1 (anti-K(V)2.1 antibody) protein immunoreactivity were both present in small pulmonary arteries. Dialysis of PASMC with AbK(V)2.1 significantly attenuated I(KV) by 67% at +50 mV. Hypoxia ( approximately 20-30 mmHg) inhibited I(KV) by approximately 70% at +50 mV. Ablation of currents associated with K(V)2.1 using AbK(V)2.1 caused a marked reduction in the amplitude of I(KV). Hypoxia in the presence of the antibody did not affect the magnitude of I(KV). CONCLUSIONS These results indicate that K(V)2.1 channel subunits exist within small pulmonary arteries and conduct a significant part of I(KV) within native PASMC. Furthermore, application of AbK(V)2.1 abolishes hypoxic inhibition of I(KV) in native PASMC suggesting that K(V)2.1 channels play a pivotal role in mediating hypoxic pulmonary vasoconstriction.
Collapse
Affiliation(s)
- Dayle S Hogg
- Department of Pharmacology, School of Medical Sciences, University of Bristol, University Walk, Bristol, BS8 1TD, UK
| | | | | | | |
Collapse
|
47
|
|
48
|
Ji J, Salapatek AMF, Lau H, Wang G, Gaisano HY, Diamant NE. SNAP-25, a SNARE protein, inhibits two types of K channels in esophageal smooth muscle. Gastroenterology 2002; 122:994-1006. [PMID: 11910352 DOI: 10.1053/gast.2002.32412] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/02/2022]
Abstract
BACKGROUND & AIMS The plasma membrane-associated soluble N-ethylmaleimide-sensitive factors attachment protein receptors (SNAREs), synaptosome-associated protein of 25 kilodaltons (SNAP-25), and syntaxin 1A, have been found to physically interact with and functionally modify membrane-spanning ion channels. Studies were performed in cat esophageal body and lower esophageal sphincter (LES) smooth muscle to (1) show the presence of SNAP-25, and (2) determine whether SNAP-25 affects K+ channel activity. METHODS Single circular muscle cells from the esophageal body and sphincter were studied. Cellular localization of SNAP-25 and K+ channel activity were assessed. RESULTS SNAP-25 was found in the plasma membrane of all regions examined. Outward K+ currents in body circular muscle were mainly composed of large conductance Ca2+-activated channel currents (K(Ca), 40.1%) and delayed rectifier K+ channel currents (K(V), 54.2%). Microinjection of SNAP-25 into muscle cells caused a dose-dependent inhibition of both outward K+ currents, maximal 44% at 10(-8) mol/L. Cleavage of endogenous SNAP-25 by dialyzing botulinum neurotoxin A into the cell interior resulted in a 35% increase in outward currents. CONCLUSIONS SNAP-25 protein is present in esophageal smooth muscle cells, and inhibits both K(V) and K(Ca) currents in circular muscle cells. The findings suggest a role for SNAP-25 in regulation of esophageal muscle cell excitability and contractility, and point to potential new targets for treatment of esophageal motor disorders.
Collapse
Affiliation(s)
- Junzhi Ji
- Department of Medicine, University of Toronto, Ontario, Canada
| | | | | | | | | | | |
Collapse
|
49
|
Davies A, Batchelor T, Eardley I, Beech D. Potassium Channel K
V
α1 Subunit Expression And Function In Human Detrusor Muscle. J Urol 2002. [DOI: 10.1016/s0022-5347(05)65254-8] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Affiliation(s)
- A.M. Davies
- From the School of Biomedical Sciences, University of Leeds and Department of Urology, St James’s Hospital, Leeds, United Kingdom
| | - T.J.P. Batchelor
- From the School of Biomedical Sciences, University of Leeds and Department of Urology, St James’s Hospital, Leeds, United Kingdom
| | - I. Eardley
- From the School of Biomedical Sciences, University of Leeds and Department of Urology, St James’s Hospital, Leeds, United Kingdom
| | - D.J. Beech
- From the School of Biomedical Sciences, University of Leeds and Department of Urology, St James’s Hospital, Leeds, United Kingdom
| |
Collapse
|
50
|
Mason HS, Latten MJ, Godoy LD, Horowitz B, Kenyon JL. Modulation of Kv1.5 currents by protein kinase A, tyrosine kinase, and protein tyrosine phosphatase requires an intact cytoskeleton. Mol Pharmacol 2002; 61:285-93. [PMID: 11809852 DOI: 10.1124/mol.61.2.285] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
The regulation of cardiac delayed rectifier potassium (Kv) currents by cAMP-dependent protein kinase (PKA) contributes to the control of blood pressure and heart rate. We investigated the modulation by PKA and protein phosphatases of cloned Kv1.5 channels expressed in Xenopus laevis oocytes. Exposure of oocytes to activators of PKA (100 nM forskolin, 1 mM 8-bromo-cAMP, or 1 mM 3-isobutyl-1-methylxanthine) had no effect on the amplitude of Kv1.5 currents. Inhibition of PKA by injection of protein kinase A inhibitor peptide or exposure to myristoylated protein kinase A inhibitor peptide (M-PKI; 100 nM) reduced currents mediated by Kv1.5. M-PKI also reduced the amplitude of currents mediated by mutated Kv1.5 channels in which the COOH terminal PKA phosphorylation sites and PSD-95, Disc-large, and ZO-1-binding domain were removed. The reduction of Kv1.5 currents by M-PKI was attenuated by inhibition of actin polymerization by 1 microM cytochalasins B and D, but was not affected by 10 microM phalloidin (stabilizes actin filaments) or 50 microM colchicine (disrupts microtubules). Treatment of oocytes with antisense oligonucleotides against alpha-actinin-2 abolished the reduction in Kv1.5 current by M-PKI. These observations suggest that Kv1.5 currents are activated by endogenous PKA in "resting" oocytes and that inhibition of PKA activity reveals the action of endogenous phosphatases. Indeed, injection of alkaline phosphatase reduced currents mediated by Kv1.5. Further preincubation of oocytes with 1 mM sodium orthovanadate (a protein tyrosine phosphatase inhibitor) abolished the reduction in Kv1.5 currents by M-PKI. We conclude that currents encoded by Kv1.5 are regulated by PKA and protein tyrosine phosphatase and that this regulation requires an intact actin cytoskeleton and alpha-actinin-2.
Collapse
Affiliation(s)
- H S Mason
- Department of Physiology and Cell Biology, University of Nevada School of Medicine, Reno, Nevada 89557-0046, USA
| | | | | | | | | |
Collapse
|