1
|
Ma R, Seifi M, Papanikolaou M, Brown JF, Swinny JD, Lewis A. TREK-1 Channel Expression in Smooth Muscle as a Target for Regulating Murine Intestinal Contractility: Therapeutic Implications for Motility Disorders. Front Physiol 2018; 9:157. [PMID: 29563879 PMCID: PMC5845753 DOI: 10.3389/fphys.2018.00157] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2017] [Accepted: 02/16/2018] [Indexed: 11/24/2022] Open
Abstract
Gastrointestinal (GI) motility disorders such as irritable bowel syndrome (IBS) can occur when coordinated smooth muscle contractility is disrupted. Potassium (K+) channels regulate GI smooth muscle tone and are key to GI tract relaxation, but their molecular and functional phenotypes are poorly described. Here we define the expression and functional roles of mechano-gated K2P channels in mouse ileum and colon. Expression and distribution of the K2P channel family were investigated using quantitative RT-PCR (qPCR), immunohistochemistry and confocal microscopy. The contribution of mechano-gated K2P channels to mouse intestinal muscle tension was studied pharmacologically using organ bath. Multiple K2P gene transcripts were detected in mouse ileum and colon whole tissue preparations. Immunohistochemistry confirmed TREK-1 expression was smooth muscle specific in both ileum and colon, whereas TREK-2 and TRAAK channels were detected in enteric neurons but not smooth muscle. In organ bath, mechano-gated K2P channel activators (Riluzole, BL-1249, flufenamic acid, and cinnamyl 1-3,4-dihydroxy-alpha-cyanocinnamate) induced relaxation of KCl and CCh pre-contracted ileum and colon tissues and reduced the amplitude of spontaneous contractions. These data reveal the specific expression of mechano-gated K2P channels in mouse ileum and colon tissues and highlight TREK-1, a smooth muscle specific K2P channel in GI tract, as a potential therapeutic target for combating motility pathologies arising from hyper-contractility.
Collapse
Affiliation(s)
- Ruolin Ma
- School of Pharmacy and Biomedical Sciences, Institute of Biological and Biomedical Sciences, University of Portsmouth, Portsmouth, United Kingdom
| | - Mohsen Seifi
- School of Pharmacy and Biomedical Sciences, Institute of Biological and Biomedical Sciences, University of Portsmouth, Portsmouth, United Kingdom
| | - Maria Papanikolaou
- School of Pharmacy and Biomedical Sciences, Institute of Biological and Biomedical Sciences, University of Portsmouth, Portsmouth, United Kingdom
| | - James F Brown
- School of Pharmacy and Biomedical Sciences, Institute of Biological and Biomedical Sciences, University of Portsmouth, Portsmouth, United Kingdom
| | - Jerome D Swinny
- School of Pharmacy and Biomedical Sciences, Institute of Biological and Biomedical Sciences, University of Portsmouth, Portsmouth, United Kingdom
| | - Anthony Lewis
- School of Pharmacy and Biomedical Sciences, Institute of Biological and Biomedical Sciences, University of Portsmouth, Portsmouth, United Kingdom
| |
Collapse
|
2
|
Chai S, Wan X, Nassal DM, Liu H, Moravec CS, Ramirez-Navarro A, Deschênes I. Contribution of two-pore K + channels to cardiac ventricular action potential revealed using human iPSC-derived cardiomyocytes. Am J Physiol Heart Circ Physiol 2017; 312:H1144-H1153. [PMID: 28341634 DOI: 10.1152/ajpheart.00107.2017] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/15/2017] [Revised: 03/16/2017] [Accepted: 03/22/2017] [Indexed: 01/12/2023]
Abstract
Two-pore K+ (K2p) channels have been described in modulating background conductance as leak channels in different physiological systems. In the heart, the expression of K2p channels is heterogeneous with equivocation regarding their functional role. Our objective was to determine the K2p expression profile and their physiological and pathophysiological contribution to cardiac electrophysiology. Induced pluripotent stem cells (iPSCs) generated from humans were differentiated into cardiomyocytes (iPSC-CMs). mRNA was isolated from these cells, commercial iPSC-CM (iCells), control human heart ventricular tissue (cHVT), and ischemic (iHF) and nonischemic heart failure tissues (niHF). We detected 10 K2p channels in the heart. Comparing quantitative PCR expression of K2p channels between human heart tissue and iPSC-CMs revealed K2p1.1, K2p2.1, K2p5.1, and K2p17.1 to be higher expressed in cHVT, whereas K2p3.1 and K2p13.1 were higher in iPSC-CMs. Notably, K2p17.1 was significantly lower in niHF tissues compared with cHVT. Action potential recordings in iCells after K2p small interfering RNA knockdown revealed prolongations in action potential depolarization at 90% repolarization for K2p2.1, K2p3.1, K2p6.1, and K2p17.1. Here, we report the expression level of 10 human K2p channels in iPSC-CMs and how they compared with cHVT. Importantly, our functional electrophysiological data in human iPSC-CMs revealed a prominent role in cardiac ventricular repolarization for four of these channels. Finally, we also identified K2p17.1 as significantly reduced in niHF tissues and K2p4.1 as reduced in niHF compared with iHF. Thus, we advance the notion that K2p channels are emerging as novel players in cardiac ventricular electrophysiology that could also be remodeled in cardiac pathology and therefore contribute to arrhythmias.NEW & NOTEWORTHY Two-pore K+ (K2p) channels are traditionally regarded as merely background leak channels in myriad physiological systems. Here, we describe the expression profile of K2p channels in human-induced pluripotent stem cell-derived cardiomyocytes and outline a salient role in cardiac repolarization and pathology for multiple K2p channels.
Collapse
Affiliation(s)
- Sam Chai
- Department of Physiology and Biophysics, Case Western Reserve University, Cleveland, Ohio.,Heart and Vascular Research Center, Department of Medicine, MetroHealth Campus, Case Western Reserve University, Cleveland, Ohio; and
| | - Xiaoping Wan
- Heart and Vascular Research Center, Department of Medicine, MetroHealth Campus, Case Western Reserve University, Cleveland, Ohio; and
| | - Drew M Nassal
- Department of Physiology and Biophysics, Case Western Reserve University, Cleveland, Ohio.,Heart and Vascular Research Center, Department of Medicine, MetroHealth Campus, Case Western Reserve University, Cleveland, Ohio; and
| | - Haiyan Liu
- Heart and Vascular Research Center, Department of Medicine, MetroHealth Campus, Case Western Reserve University, Cleveland, Ohio; and
| | | | - Angelina Ramirez-Navarro
- Heart and Vascular Research Center, Department of Medicine, MetroHealth Campus, Case Western Reserve University, Cleveland, Ohio; and
| | - Isabelle Deschênes
- Department of Physiology and Biophysics, Case Western Reserve University, Cleveland, Ohio; .,Heart and Vascular Research Center, Department of Medicine, MetroHealth Campus, Case Western Reserve University, Cleveland, Ohio; and
| |
Collapse
|
3
|
Zhu MH, Sung TS, Kurahashi M, O'Kane LE, O'Driscoll K, Koh SD, Sanders KM. Na+-K+-Cl- cotransporter (NKCC) maintains the chloride gradient to sustain pacemaker activity in interstitial cells of Cajal. Am J Physiol Gastrointest Liver Physiol 2016; 311:G1037-G1046. [PMID: 27742704 PMCID: PMC5206290 DOI: 10.1152/ajpgi.00277.2016] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/02/2016] [Accepted: 10/11/2016] [Indexed: 01/31/2023]
Abstract
Interstitial cells of Cajal (ICC) generate electrical slow waves by coordinated openings of ANO1 channels, a Ca2+-activated Cl- (CaCC) conductance. Efflux of Cl- during slow waves must be significant, as there is high current density during slow-wave currents and slow waves are of sufficient magnitude to depolarize the syncytium of smooth muscle cells and PDGFRα+ cells to which they are electrically coupled. We investigated how the driving force for Cl- current is maintained in ICC. We found robust expression of Slc12a2 (which encodes an Na+-K+-Cl- cotransporter, NKCC1) and immunohistochemical confirmation that NKCC1 is expressed in ICC. With the use of the gramicidin permeabilized-patch technique, which is reported to not disturb [Cl-]i, the reversal potential for spontaneous transient inward currents (ESTICs) was -10.5 mV. This value corresponds to the peak of slow waves when they are recorded directly from ICC in situ. Inhibition of NKCC1 with bumetanide shifted ESTICs to more negative potentials within a few minutes and reduced pacemaker activity. Bumetanide had no direct effects on ANO1 or CaV3.2 channels expressed in HEK293 cells or L-type Ca2+ currents. Reducing extracellular Cl- to 10 mM shifted ESTICs to positive potentials as predicted by the Nernst equation. The relatively rapid shift in ESTICs when NKCC1 was blocked suggests that significant changes in the transmembrane Cl- gradient occur during the slow-wave cycle, possibly within microdomains formed between endoplasmic reticulum and the plasma membrane in ICC. Recovery of Cl- via NKCC1 might have additional consequences on shaping the waveforms of slow waves via Na+ entry into microdomains.
Collapse
Affiliation(s)
- Mei Hong Zhu
- Department of Physiology and Cell Biology, University of Nevada School of Medicine, Reno, Nevada
| | - Tae Sik Sung
- Department of Physiology and Cell Biology, University of Nevada School of Medicine, Reno, Nevada
| | - Masaaki Kurahashi
- Department of Physiology and Cell Biology, University of Nevada School of Medicine, Reno, Nevada
| | - Lauren E. O'Kane
- Department of Physiology and Cell Biology, University of Nevada School of Medicine, Reno, Nevada
| | - Kate O'Driscoll
- Department of Physiology and Cell Biology, University of Nevada School of Medicine, Reno, Nevada
| | - Sang Don Koh
- Department of Physiology and Cell Biology, University of Nevada School of Medicine, Reno, Nevada
| | - Kenton M. Sanders
- Department of Physiology and Cell Biology, University of Nevada School of Medicine, Reno, Nevada
| |
Collapse
|
4
|
Telles CJ, Decker SE, Motley WW, Peters AW, Mehr AP, Frizzell RA, Forrest JN. Functional and molecular identification of a TASK-1 potassium channel regulating chloride secretion through CFTR channels in the shark rectal gland: implications for cystic fibrosis. Am J Physiol Cell Physiol 2016; 311:C884-C894. [PMID: 27653983 PMCID: PMC5206301 DOI: 10.1152/ajpcell.00030.2016] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2016] [Accepted: 09/18/2016] [Indexed: 11/22/2022]
Abstract
In the shark rectal gland (SRG), apical chloride secretion through CFTR channels is electrically coupled to a basolateral K+ conductance whose type and molecular identity are unknown. We performed studies in the perfused SRG with 17 K+ channel inhibitors to begin this search. Maximal chloride secretion was markedly inhibited by low-perfusate pH, bupivicaine, anandamide, zinc, quinidine, and quinine, consistent with the properties of an acid-sensitive, four-transmembrane, two-pore-domain K+ channel (4TM-K2P). Using PCR with degenerate primers to this family, we identified a TASK-1 fragment in shark rectal gland, brain, gill, and kidney. Using 5' and 3' rapid amplification of cDNA ends PCR and genomic walking, we cloned the full-length shark gene (1,282 bp), whose open reading frame encodes a protein of 375 amino acids that was 80% identical to the human TASK-1 protein. We expressed shark and human TASK-1 cRNA in Xenopus oocytes and characterized these channels using two-electrode voltage clamping. Both channels had identical current-voltage relationships (outward rectifying) and a reversal potential of -90 mV. Both were inhibited by quinine, bupivicaine, and acidic pH. The pKa for current inhibition was 7.75 for shark TASK-1 vs. 7.37 for human TASK-1, values similar to the arterial pH for each species. We identified this protein in SRG by Western blot and confocal immunofluorescent microscopy and detected the protein in SRG and human airway cells. Shark TASK-1 is the major K+ channel coupled to chloride secretion in the SRG, is the oldest 4TM 2P family member identified, and is the first TASK-1 channel identified to play a role in setting the driving force for chloride secretion in epithelia. The detection of this potassium channel in mammalian lung tissue has implications for human biology and disease.
Collapse
Affiliation(s)
- Connor J Telles
- Nephrology Division, Department of Medicine, Yale University School of Medicine, New Haven, Connecticut
- Mount Desert Island Biological Laboratory, Salisbury Cove, Maine
| | - Sarah E Decker
- Nephrology Division, Department of Medicine, Yale University School of Medicine, New Haven, Connecticut
- Mount Desert Island Biological Laboratory, Salisbury Cove, Maine
| | - William W Motley
- Nephrology Division, Department of Medicine, Yale University School of Medicine, New Haven, Connecticut
- Mount Desert Island Biological Laboratory, Salisbury Cove, Maine
| | - Alexander W Peters
- Nephrology Division, Department of Medicine, Yale University School of Medicine, New Haven, Connecticut
- Mount Desert Island Biological Laboratory, Salisbury Cove, Maine
| | - Ali Poyan Mehr
- Nephrology Division, Department of Medicine, Yale University School of Medicine, New Haven, Connecticut
- Mount Desert Island Biological Laboratory, Salisbury Cove, Maine
| | - Raymond A Frizzell
- Department of Cell Biology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania; and
- Mount Desert Island Biological Laboratory, Salisbury Cove, Maine
| | - John N Forrest
- Nephrology Division, Department of Medicine, Yale University School of Medicine, New Haven, Connecticut;
- Mount Desert Island Biological Laboratory, Salisbury Cove, Maine
| |
Collapse
|
5
|
Currò D. The Modulation of Potassium Channels in the Smooth Muscle as a Therapeutic Strategy for Disorders of the Gastrointestinal Tract. ADVANCES IN PROTEIN CHEMISTRY AND STRUCTURAL BIOLOGY 2016; 104:263-305. [PMID: 27038377 DOI: 10.1016/bs.apcsb.2015.12.002] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Alterations of smooth muscle contractility contribute to the pathophysiology of important functional gastrointestinal disorders (FGIDs) such as functional dyspepsia and irritable bowel syndrome. Consequently, drugs that decrease smooth muscle contractility are effective treatments for these diseases. Smooth muscle contraction is mainly triggered by Ca(2+) influx through voltage-dependent channels located in the plasma membrane. Thus, the modulation of the membrane potential results in the regulation of Ca(2+) influx and cytosolic levels. K(+) channels play fundamental roles in these processes. The open probability of K(+) channels increases in response to various stimuli, including membrane depolarization (voltage-gated K(+) [K(V)] channels) and the increase in cytosolic Ca(2+) levels (Ca(2+)-dependent K(+) [K(Ca)] channels). K(+) channel activation is mostly associated with outward K(+) currents that hyperpolarize the membrane and reduce cell excitability and contractility. In addition, some K(+) channels are open at the resting membrane potential values of the smooth muscle cells in some gut segments and contribute to set the resting membrane potential itself. The closure of these channels induces membrane depolarization and smooth muscle contraction. K(V)1.2, 1.5, 2.2, 4.3, 7.4 and 11.1, K(Ca)1.1 and 2.3, and inwardly rectifying type 6K(+) (K(ir)6) channels play the most important functional roles in the gastrointestinal smooth muscle. Activators of all these channels may theoretically relax the gastrointestinal smooth muscle and could therefore be promising new therapeutic options for FGID. The challenge of future drug research and development in this area will be to synthesize molecules selective for the channel assemblies expressed in the gastrointestinal smooth muscle.
Collapse
Affiliation(s)
- Diego Currò
- Institute of Pharmacology, School of Medicine, Catholic University of the Sacred Heart, Rome, Italy.
| |
Collapse
|
6
|
Kubota K, Ohtake N, Ohbuchi K, Mase A, Imamura S, Sudo Y, Miyano K, Yamamoto M, Kono T, Uezono Y. Hydroxy-α sanshool induces colonic motor activity in rat proximal colon: a possible involvement of KCNK9. Am J Physiol Gastrointest Liver Physiol 2015; 308:G579-90. [PMID: 25634809 PMCID: PMC4385894 DOI: 10.1152/ajpgi.00114.2014] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/27/2014] [Accepted: 01/22/2015] [Indexed: 01/31/2023]
Abstract
Various colonic motor activities are thought to mediate propulsion and mixing/absorption of colonic content. The Japanese traditional medicine daikenchuto (TU-100), which is widely used for postoperative ileus in Japan, accelerates colonic emptying in healthy humans. Hydroxy-α sanshool (HAS), a readily absorbable active ingredient of TU-100 and a KCNK3/KCNK9/KCNK18 blocker as well as TRPV1/TRPA1 agonist, has been investigated for its effects on colonic motility. Motility was evaluated by intraluminal pressure and video imaging of rat proximal colons in an organ bath. Distribution of KCNKs was investigated by RT-PCR, in situ hybridization, and immunohistochemistry. Current and membrane potential were evaluated with use of recombinant KCNK3- or KCNK9-expressing Xenopus oocytes and Chinese hamster ovary cells. Defecation frequency in rats was measured. HAS dose dependently induced strong propulsive "squeezing" motility, presumably as long-distance contraction (LDC). TRPV1/TRPA1 agonists induced different motility patterns. The effect of HAS was unaltered by TRPV1/TRPA1 antagonists and desensitization. Lidocaine (a nonselective KCNK blocker) and hydroxy-β sanshool (a geometrical isomer of HAS and KCNK3 blocker) also induced colonic motility as a rhythmic propagating ripple (RPR) and a LDC-like motion, respectively. HAS-induced "LDC," but not lidocaine-induced "RPR," was abrogated by a neuroleptic agent tetrodotoxin. KCNK3 and KCNK9 were located mainly in longitudinal smooth muscle cells and in neural cells in the myenteric plexus, respectively. Administration of HAS or TU-100 increased defecation frequency in normal and laparotomy rats. HAS may evoke strong LDC possibly via blockage of the neural KCNK9 channel in the colonic myenteric plexus.
Collapse
Affiliation(s)
| | - Nobuhiro Ohtake
- 1Tsumura Research Laboratories, Tsumura & Co., Ibaraki, Japan;
| | - Katsuya Ohbuchi
- 1Tsumura Research Laboratories, Tsumura & Co., Ibaraki, Japan; ,2Division of Cancer Pathophysiology, National Cancer Center Research Institute, Tokyo, Japan;
| | - Akihito Mase
- 1Tsumura Research Laboratories, Tsumura & Co., Ibaraki, Japan;
| | - Sachiko Imamura
- 1Tsumura Research Laboratories, Tsumura & Co., Ibaraki, Japan;
| | - Yuka Sudo
- 2Division of Cancer Pathophysiology, National Cancer Center Research Institute, Tokyo, Japan;
| | - Kanako Miyano
- 2Division of Cancer Pathophysiology, National Cancer Center Research Institute, Tokyo, Japan;
| | | | - Toru Kono
- 3Department of Gastroenterology, Hokkaido University Graduate School of Medicine, Sapporo, Japan; and ,4Center for Clinical and Biomedical Research, Sapporo Higashi Tokushukai Hospital, Sapporo, Japan
| | - Yasuhito Uezono
- Division of Cancer Pathophysiology, National Cancer Center Research Institute, Tokyo, Japan;
| |
Collapse
|
7
|
Contraction of gut smooth muscle cells assessed by fluorescence imaging. J Pharmacol Sci 2015; 127:344-51. [DOI: 10.1016/j.jphs.2015.02.002] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2014] [Revised: 01/14/2015] [Accepted: 02/02/2015] [Indexed: 11/20/2022] Open
|
8
|
Kelley CA, Decker SE, Silva P, Forrest JN. Gastric inhibitory peptide, serotonin, and glucagon are unexpected chloride secretagogues in the rectal gland of the skate (Leucoraja erinacea). Am J Physiol Regul Integr Comp Physiol 2014; 306:R674-80. [PMID: 24553297 PMCID: PMC4010662 DOI: 10.1152/ajpregu.00531.2013] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2013] [Accepted: 02/17/2014] [Indexed: 11/22/2022]
Abstract
Since the discovery of the rectal gland of the dogfish shark 50 years ago, experiments with this tissue have greatly aided our understanding of secondary active chloride secretion and the secretagogues responsible for this function. In contrast, very little is known about the rectal gland of skates. In the present experiments, we performed the first studies in the perfused rectal gland of the little skate (Leucoraja erinacea), an organ weighing less than one-tenth of the shark rectal gland. Our results indicate that the skate gland can be studied by modified perfusion techniques and in primary culture monolayers, and that secretion is blocked by the inhibitors of membrane proteins required for secondary active chloride secretion. Our major finding is that three G protein-coupled receptor agonists, the incretin gastric inhibitory polypeptide (GIP), also known as glucose-dependent insulinotropic peptide, as well as glucagon and serotonin, are unexpected potent chloride secretagogues in the skate but not the shark. Glucagon stimulated chloride secretion to a mean value of 1,661 ± 587 μeq·h(-1)·g(-1) and serotonin stimulated to 2,893 ± 699 μeq·h(-1)·g(-1). GIP stimulated chloride secretion to 3,733 ± 679 μeq·h(-1)·g(-1) and significantly increased tissue cAMP content compared with basal conditions. This is the first report of GIP functioning as a chloride secretagogue in any species or tissue.
Collapse
Affiliation(s)
- Catherine A Kelley
- Nephrology Division, Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut
| | | | | | | |
Collapse
|
9
|
Kazmierczak M, Zhang X, Chen B, Mulkey DK, Shi Y, Wagner PG, Pivaroff-Ward K, Sassic JK, Bayliss DA, Jegla T. External pH modulates EAG superfamily K+ channels through EAG-specific acidic residues in the voltage sensor. ACTA ACUST UNITED AC 2013; 141:721-35. [PMID: 23712551 PMCID: PMC3664700 DOI: 10.1085/jgp.201210938] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
The Ether-a-go-go (EAG) superfamily of voltage-gated K+ channels consists of three functionally distinct gene families (Eag, Elk, and Erg) encoding a diverse set of low-threshold K+ currents that regulate excitability in neurons and muscle. Previous studies indicate that external acidification inhibits activation of three EAG superfamily K+ channels, Kv10.1 (Eag1), Kv11.1 (Erg1), and Kv12.1 (Elk1). We show here that Kv10.2, Kv12.2, and Kv12.3 are similarly inhibited by external protons, suggesting that high sensitivity to physiological pH changes is a general property of EAG superfamily channels. External acidification depolarizes the conductance–voltage (GV) curves of these channels, reducing low threshold activation. We explored the mechanism of this high pH sensitivity in Kv12.1, Kv10.2, and Kv11.1. We first examined the role of acidic voltage sensor residues that mediate divalent cation block of voltage activation in EAG superfamily channels because protons reduce the sensitivity of Kv12.1 to Zn2+. Low pH similarly reduces Mg2+ sensitivity of Kv10.1, and we found that the pH sensitivity of Kv11.1 was greatly attenuated at 1 mM Ca2+. Individual neutralizations of a pair of EAG-specific acidic residues that have previously been implicated in divalent block of diverse EAG superfamily channels greatly reduced the pH response in Kv12.1, Kv10.2, and Kv11.1. Our results therefore suggest a common mechanism for pH-sensitive voltage activation in EAG superfamily channels. The EAG-specific acidic residues may form the proton-binding site or alternatively are required to hold the voltage sensor in a pH-sensitive conformation. The high pH sensitivity of EAG superfamily channels suggests that they could contribute to pH-sensitive K+ currents observed in vivo.
Collapse
Affiliation(s)
- Marcin Kazmierczak
- Department of Biology, The Pennsylvania State University, University Park, PA 16802, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
10
|
Cid LP, Roa-Rojas HA, Niemeyer MI, González W, Araki M, Araki K, Sepúlveda FV. TASK-2: a K2P K(+) channel with complex regulation and diverse physiological functions. Front Physiol 2013; 4:198. [PMID: 23908634 PMCID: PMC3725403 DOI: 10.3389/fphys.2013.00198] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2013] [Accepted: 07/10/2013] [Indexed: 11/13/2022] Open
Abstract
TASK-2 (K2P5.1) is a two-pore domain K(+) channel belonging to the TALK subgroup of the K2P family of proteins. TASK-2 has been shown to be activated by extra- and intracellular alkalinization. Extra- and intracellular pH-sensors reside at arginine 224 and lysine 245 and might affect separate selectivity filter and inner gates respectively. TASK-2 is modulated by changes in cell volume and a regulation by direct G-protein interaction has also been proposed. Activation by extracellular alkalinization has been associated with a role of TASK-2 in kidney proximal tubule bicarbonate reabsorption, whilst intracellular pH-sensitivity might be the mechanism for its participation in central chemosensitive neurons. In addition to these functions TASK-2 has been proposed to play a part in apoptotic volume decrease in kidney cells and in volume regulation of glial cells and T-lymphocytes. TASK-2 is present in chondrocytes of hyaline cartilage, where it is proposed to play a central role in stabilizing the membrane potential. Additional sites of expression are dorsal root ganglion neurons, endocrine and exocrine pancreas and intestinal smooth muscle cells. TASK-2 has been associated with the regulation of proliferation of breast cancer cells and could become target for breast cancer therapeutics. Further work in native tissues and cells together with genetic modification will no doubt reveal the details of TASK-2 functions that we are only starting to suspect.
Collapse
Affiliation(s)
- L Pablo Cid
- Centro de Estudios Científicos Valdivia, Chile
| | | | | | | | | | | | | |
Collapse
|
11
|
Nam JH, Shin DH, Zheng H, Lee DS, Park SJ, Park KS, Kim SJ. Expression of TASK-2 and its upregulation by B cell receptor stimulation in WEHI-231 mouse immature B cells. Am J Physiol Cell Physiol 2011; 300:C1013-22. [DOI: 10.1152/ajpcell.00475.2010] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Stimulation of B cell receptors (BCR ligation) induces apoptosis of immature B cells, which is critical to the elimination of self-reactive clones. In the mouse immature B cell line WEHI-231, the authors previously reported two types of background K+ channels with large (∼300 pS, LKbg) and medium (∼100 pS, MKbg) conductance in divalent cation-free conditions. While the authors have recently identified LKbg as TREK-2, the molecular nature of MKbg is unknown yet. In the present study, the authors found that BCR ligation markedly increased the background K+ conductance of WEHI-231. A single-channel study revealed that MKbg activity is increased by BCR ligation and that the biophysical properties (unitary conductance and pH sensitivity) of MKbg are consistent with those of TWIK-related acid-sensitive K+ channel 2 (TASK-2). The expression of TASK-2 and its upregulation by BCR ligation were confirmed by RT-PCR and immunoblot assays in WEHI-231. The BCR ligation-induced increase of K+ current was prevented by calcineurin inhibitors (cyclosporine A or FK506), and also by TASK-2-specific small interfering RNA (siRNA) transfection (si-TASK-2). Furthermore, si-TASK-2 attenuated the apoptosis of WEHI-231 caused by BCR ligation. TASK-2 activity and its mRNA were also confirmed in the primary splenic B cells of mouse. Summarizing, the authors report for the first time the expression of TASK-2 in B cells and surmise that the upregulation of TASK-2 by BCR ligation is associated with the apoptosis of immature B cells.
Collapse
Affiliation(s)
- Joo Hyun Nam
- Department of Physiology and
- Department of Physiology, Dongguk University College of Medicine; and
| | | | | | - Dong-Sup Lee
- Department of Anatomy, Seoul National University College of Medicine
| | | | - Kyung Sun Park
- Department of Physiology and
- Ischemia/Hypoxia Disease Institute, Medical Research Center, Seoul National University, Seoul, Korea
| | - Sung Joon Kim
- Department of Physiology and
- Ischemia/Hypoxia Disease Institute, Medical Research Center, Seoul National University, Seoul, Korea
| |
Collapse
|
12
|
Sanders KM. Regulation of smooth muscle excitation and contraction. NEUROGASTROENTEROLOGY AND MOTILITY : THE OFFICIAL JOURNAL OF THE EUROPEAN GASTROINTESTINAL MOTILITY SOCIETY 2008. [PMID: 18402641 DOI: 10.1111/j.1365-2982.2008.01108.x/abstract] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
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
|
13
|
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
|
14
|
Hwang SJ, O'Kane N, Singer C, Ward SM, Sanders KM, Koh SD. Block of inhibitory junction potentials and TREK-1 channels in murine colon by Ca2+ store-active drugs. J Physiol 2008; 586:1169-84. [PMID: 18187470 DOI: 10.1113/jphysiol.2007.148718] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
Abstract
Post-junctional enteric inhibitory responses are composed of at least two components attributed to the release of a purine and nitric oxide (NO). The nitrergic component is characterized by membrane potential hyperpolarization; however, the conductances involved and the role of Ca(2+) stores in regulating these conductances are controversial. Conventional microelectrode recordings were performed in intact muscle strips and whole-cell voltage clamp experiments were performed on freshly dispersed cells and COS7 cells stably transfected with TREK-1 channels. Here we show that several Ca(2+) store-active compounds, including caffeine, ryanodine, and cyclopiazonic acid, reduce inhibitory junction potentials and responses to sodium nitroprusside in murine colonic muscles. We previously proposed that two-pore K(+) channels of the TREK family mediate a portion of the hyperpolarization response to NO in colonic muscles. We tested the effects of Ca(2+) store-active drugs in COS cells expressing murine TREK-1 channels and found these compounds block TREK-1 currents. These effects were greatly attenuated by dialysing cells with protein kinase A inhibitory peptide (PKAI). Caffeine also blocked stretch-dependent K(+) (SDK) channels, thought to be due to expression of TREK channels, in colonic myocytes, but these effects were not apparent in excised patches. Taken together our data show that Ca(2+) store-active compounds inhibit TREK-1 channels, native SDK channels, and nitrergic inhibitory junction potentials. These effects appear to be due, in part, to the cAMP/PKA stimulatory actions of these drugs and inhibitory effects of TREK channels.
Collapse
Affiliation(s)
- Sung Jin Hwang
- Department of Physiology and Cell Biology, University of Nevada School of Medicine, Reno, NV 89557, USA
| | | | | | | | | | | |
Collapse
|
15
|
Chen H, Ordög T, Chen J, Young DL, Bardsley MR, Redelman D, Ward SM, Sanders KM. Differential gene expression in functional classes of interstitial cells of Cajal in murine small intestine. Physiol Genomics 2007; 31:492-509. [PMID: 17895395 DOI: 10.1152/physiolgenomics.00113.2007] [Citation(s) in RCA: 99] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Interstitial cells of Cajal (ICC) have important functions in regulation of motor activity in the gastrointestinal tract. In murine small intestine, ICC are gathered in the regions of the myenteric plexus (ICC-MY) and the deep muscular plexus (ICC-DMP). These two classes of ICC have different physiological functions. ICC-MY are pacemaker cells and generate the slow-wave electrical rhythmicity of gastrointestinal organs. ICC-DMP form synaptic connections with the varicose nerve terminals of enteric motor neurons and are involved in reception and transduction of motor neurotransmission. Gene expression underlying specific functions of ICC classes is incompletely understood. In the present study, we used recently developed highly selective techniques to isolate the two functional ICC classes from enzymatically dispersed intestinal muscles by fluorescence-activated cell sorting. The transcriptomes of ICC-MY and ICC-DMP were investigated using oligonucleotide microarray analysis. Differential expression of functional groups of genes defined by standard gene ontology terms was also studied. There were substantial numbers of genes expressed more abundantly in ICC than in the tunica muscularis, and we also detected marked phenotypic differences between ICC-MY and ICC-DMP. Notably, genes related to cell junction, process guidance, and vesicle trafficking were upregulated in ICC. Consistent with their specific functions, metabolic and Ca(2+) transport genes were relatively upregulated in ICC-MY, whereas genes for signaling proteins involved in transduction of neurotransmitter functions were relatively upregulated in ICC-DMP. Our results may lead to the identification of novel biomarkers for ICC and provide directions for further studies designed to understand ICC function in health and disease.
Collapse
Affiliation(s)
- Hui Chen
- Department of Physiology and Cell Biology, University of Nevada, School of Medicine, Reno, NV 89557, USA
| | | | | | | | | | | | | | | |
Collapse
|
16
|
Lotshaw DP. Biophysical, pharmacological, and functional characteristics of cloned and native mammalian two-pore domain K+ channels. Cell Biochem Biophys 2007; 47:209-56. [PMID: 17652773 DOI: 10.1007/s12013-007-0007-8] [Citation(s) in RCA: 142] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/1999] [Revised: 11/30/1999] [Accepted: 11/30/1999] [Indexed: 12/12/2022]
Abstract
The mammalian family of two-pore domain K+ (K2P) channel proteins are encoded by 15 KCNK genes and subdivided into six subfamilies on the basis of sequence similarities: TWIK, TREK, TASK, TALK, THIK, and TRESK. K2P channels are expressed in cells throughout the body and have been implicated in diverse cellular functions including maintenance of the resting potential and regulation of excitability, sensory transduction, ion transport, and cell volume regulation, as well as metabolic regulation and apoptosis. In recent years K2P channel isoforms have been identified as important targets of several widely employed drugs, including: general anesthetics, local anesthetics, neuroprotectants, and anti-depressants. An important goal of future studies will be to identify the basis of drug actions and channel isoform selectivity. This goal will be facilitated by characterization of native K2P channel isoforms, their pharmacological properties and tissue-specific expression patterns. To this end the present review examines the biophysical, pharmacological, and functional characteristics of cloned mammalian K2P channels and compares this information with the limited data available for native K2P channels in order to determine criteria which may be useful in identifying ionic currents mediated by native channel isoforms and investigating their pharmacological and functional characteristics.
Collapse
Affiliation(s)
- David P Lotshaw
- Department of Biological Sciences, Northern Illinois University, DeKalb, IL 60115, USA.
| |
Collapse
|
17
|
Kito Y, Suzuki H. Role of K+ channels in the regulation of electrical spontaneous activity of the mouse small intestine. Pflugers Arch 2007; 455:505-14. [PMID: 17602242 DOI: 10.1007/s00424-007-0306-3] [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: 04/23/2007] [Revised: 05/25/2007] [Accepted: 06/01/2007] [Indexed: 10/23/2022]
Abstract
The roles of K(+) channels in the regulation of slow waves and pacemaker potentials recorded from mouse small intestine were investigated using intracellular recording techniques in the presence of nifedipine. Iberiotoxin (0.1 microM) and charybdotoxin (0.1 microM) had no effect on the generation of slow waves recorded from circular smooth muscle cells. Apamin (0.3 microM) depolarized the membrane and decreased the amplitude of early, rapid repolarization of slow waves, without altering the amplitude, frequency, duration, or maximum rate of rise of the initial upstroke phase (dV/dt(max)). The early, rapid repolarization was enhanced by phenylephrine (15 microM). 4-Aminopyridine (4-AP, 5 mM) depolarized the membrane and increased the amplitude and dV/dt(max) of slow waves. Both apamin and 4-AP depolarized the membrane and decreased the amplitude and dV/dt(max) of pacemaker potentials recorded from interstitial cells of Cajal distributed in the myenteric region (ICC-MY). Membrane depolarization with a high-K(+) solution decreased the amplitude and dV/dt(max) of slow waves. These results suggest that apamin-sensitive K(+) conductance and 4-AP-sensitive K(+) conductance may contribute to the resting membrane potential of circular smooth muscle cells. The early, rapid repolarization of slow waves appears to result from the opening of apamin-sensitive K(+) conductance. 4-AP-sensitive K(+) conductance is likely to be activated in the initial upstroke component (primary component) of slow waves. In ICC-MY, membrane depolarization induced by apamin or 4-AP may result from electrotonic spread from smooth muscle cells.
Collapse
Affiliation(s)
- Yoshihiko Kito
- Department of Physiology, Nagoya City University Medical School, Nagoya, Japan.
| | | |
Collapse
|
18
|
Kim YC, Sim JH, Kang TM, Suzuki H, Kim SR, Kwon SC, Xu WX, Lee SJ, Kim KW. Sodium-activated potassium current in guinea pig gastric myocytes. J Korean Med Sci 2007; 22:57-62. [PMID: 17297252 PMCID: PMC2693569 DOI: 10.3346/jkms.2007.22.1.57] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
This study was designed to identify and characterize Na+-activated K+ current (I(K(Na))) in guinea pig gastric myocytes under whole-cell patch clamp. After whole-cell configuration was established under 110 mM intracellular Na+ concentration ([Na+]i) at holding potential of -60 mV, a large inward current was produced by external 60 mM K+([K+]degrees). This inward current was not affected by removal of external Ca2+. K+ channel blockers had little effects on the current (p>0.05). Only TEA (5 mM) inhibited steady-state current to 68+/-2.7% of the control (p<0.05). In the presence of K+ channel blocker cocktail (mixture of Ba2+, glibenclamide, 4-AP, apamin, quinidine and TEA), a large inward current was activated. However, the amplitude of the steady-state current produced under [K+]degrees (140 mM) was significantly smaller when Na+ in pipette solution was replaced with K+- and Li+ in the presence of K+ channel blocker cocktail than under 110 mM [Na+]i. In the presence of K+ channel blocker cocktail under low Cl- pipette solution, this current was still activated and seemed K+-selective, since reversal potentials (E(rev)) of various concentrations of [K+]degrees-induced current in current/voltage (I/V) relationship were nearly identical to expected values. R-56865 (10-20 microM), a blocker of I(K(Na)), completely and reversibly inhibited this current. The characteristics of the current coincide with those of I(K(Na)) of other cells. Our results indicate the presence of I(K(Na)) in guinea pig gastric myocytes.
Collapse
Affiliation(s)
- Young Chul Kim
- Department of Physiology, Chungbuk National University, College of Medicine, Cheongju, Korea.
| | | | | | | | | | | | | | | | | |
Collapse
|
19
|
Boddy G, Willis A, Galante G, Daniel EE. Sodium-, chloride-, and mibefradil-sensitive calcium channels in intestinal pacing in wild-type and W/WV mice. Can J Physiol Pharmacol 2006; 84:589-99. [PMID: 16900243 DOI: 10.1139/y06-009] [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] [Indexed: 01/29/2023]
Abstract
Pacing of intestinal smooth muscle is driven by a network of cells found in the myenteric plexus called the interstitial cells of Cajal (ICC-MP), which produce a rhythmic pacemaker current. Using intact segments of circular (CM) and longitudinal (LM) muscle from wild-type and W/WV mice, we found that sodium-, chloride-, and mibefradil-sensitive ion channel currents are required for normal pacing to occur. Application of 30 µmol/L and 300 µmol/L lidocaine, 1 mmol/L 4,4′-diisothiocyanatostilbene-2,2′-disulfonic acid (DIDS), 50 nmol/L and 500 nmol/L mibefradil, or low sodium Krebs significantly reduced pacing frequency in LM and CM. However, simultaneously applying DIDS and lidocaine or low sodium Krebs solution did not completely block pacing nor did it have an additive effect. Lidocaine and low sodium Krebs solution also abolished the gradient of pacing frequencies (higher proximally) found throughout the intestine, resulting in a uniform contraction frequency of 30–40/min. In W/WV mice, which lack ICC-MP, application of DIDS and lidocaine had no effect on the robust pacing in LM segments. In conclusion we found that sodium-, chloride-, and mibefradil-sensitive channel activities were required for normal pacing and to maintain the pacing gradient found throughout the intestines in wild-type but not W/WV mice.
Collapse
Affiliation(s)
- Geoffrey Boddy
- Department of Pharmacology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB T6G 2H7, Canada
| | | | | | | |
Collapse
|
20
|
Gönczi M, Szentandrássy N, Johnson IT, Heagerty AM, Weston AH. Investigation of the role of TASK-2 channels in rat pulmonary arteries; pharmacological and functional studies following RNA interference procedures. Br J Pharmacol 2006; 147:496-505. [PMID: 16432512 PMCID: PMC1616980 DOI: 10.1038/sj.bjp.0706649] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
Abstract
In the present study, we investigated the ability of RNA interference technology to suppress TASK-2 potassium channel expression in human embryonic kidney (HEK293) cells stably transfected with TASK-2 cDNA and in rat isolated intact pulmonary arteries. Lipofectamine-induced transfection of a specific siRNA sequence targeted against TASK-2 resulted in a dose- and time-dependent decrease in TASK-2 channel protein expression. In siRNA-transfected cells the TASK-2 peak currents were significantly smaller than in control cells at every investigated pH, while the pH sensitivity was not altered. Using scrambled siRNA as a negative control, there were no significant changes in TASK-2 protein expression or current compared to mock-transfected cells. In TASK-2 siRNA-transfected small pulmonary arteries, but not in scrambled siRNA-treated vessels, myocyte resting membrane potential at pH 7.4 was significantly less negative and the hyperpolarisations in response to increasing pH from 6.4 to 8.4 were significantly smaller compared with control. The application of levcromakalim (10 microM), NS1619 (33 microM) and a potassium channel inhibitor cocktail (5 mM 4-aminopyridine, 10 mM tetraethylammonium chloride, 30 microM Ba2+ and 10 microM glibenclamide) had similar effects in control and in siRNA-transfected vessels. The TASK-1 (anandamide-sensitive) contribution to resting membrane potential was comparable in each group. Clofilium (100 microM) generated significantly smaller responses in transfected artery segments. These results suggest that RNA interference techniques are effective at inhibiting TASK-2 channel expression in cultured cells and in intact vessels and that TASK-2 channels have a functional role in setting the membrane potential of pulmonary artery myocytes.
Collapse
Affiliation(s)
- Mónika Gönczi
- Faculty of Life Sciences, University of Manchester, Manchester, M13 PT
| | - Norbert Szentandrássy
- Department of Medicine, Stopford Building, University of Manchester, Manchester M13 9PT
| | - Ian T Johnson
- Faculty of Life Sciences, University of Manchester, Manchester, M13 PT
| | - Anthony M Heagerty
- Department of Medicine, Manchester Royal Infirmary, Oxford Road, Manchester M13 9WL
| | - Arthur H Weston
- Faculty of Life Sciences, University of Manchester, Manchester, M13 PT
- Author for correspondence:
| |
Collapse
|
21
|
Renigunta V, Yuan H, Zuzarte M, Rinné S, Koch A, Wischmeyer E, Schlichthörl G, Gao Y, Karschin A, Jacob R, Schwappach B, Daut J, Preisig-Müller R. The Retention Factor p11 Confers an Endoplasmic Reticulum-Localization Signal to the Potassium Channel TASK-1. Traffic 2005; 7:168-81. [PMID: 16420525 DOI: 10.1111/j.1600-0854.2005.00375.x] [Citation(s) in RCA: 80] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
The interaction of the adaptor protein p11, also denoted S100A10, with the C-terminus of the two-pore-domain K+ channel TASK-1 was studied using yeast two-hybrid analysis, glutathione S-transferase pull-down, and co-immunoprecipitation. We found that p11 interacts with a 40 amino-acid region in the proximal C-terminus of the channel. In heterologous expression systems, deletion of the p11-interacting domain enhanced surface expression of TASK-1. Attachment of the p11-interacting domain to the cytosolic tail of the reporter protein CD8 caused retention/retrieval of the construct in the endoplasmic reticulum (ER). Attachment of the last 36 amino acids of p11 to CD8 also caused ER localization, which was abolished by removal or mutation of a putative retention motif (H/K)xKxxx, at the C-terminal end of p11. Imaging of EGFP-tagged TASK-1 channels in COS cells suggested that wild-type TASK-1 was largely retained in the ER. Knockdown of p11 with siRNA enhanced trafficking of TASK-1 to the surface membrane. Our results suggest that binding of p11 to TASK-1 retards the surface expression of the channel, most likely by virtue of a di-lysine retention signal at the C-terminus of p11. Thus, the cytosolic protein p11 may represent a 'retention factor' that causes localization of the channel to the ER.
Collapse
Affiliation(s)
- Vijay Renigunta
- Institute of Physiology, Marburg University, Deutschhausstr. 2, 35037 Marburg, Germany
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
22
|
Brazier SP, Mason HS, Bateson AN, Kemp PJ. Cloning of the human TASK-2 (KCNK5) promoter and its regulation by chronic hypoxia. Biochem Biophys Res Commun 2005; 336:1251-8. [PMID: 16168386 DOI: 10.1016/j.bbrc.2005.09.007] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2005] [Accepted: 09/02/2005] [Indexed: 10/25/2022]
Abstract
The tandem P domain potassium channel family includes five members of the acid-sensing subfamily, TASK. TASK channels are active at resting potential and are inhibited by extracellular protons, suggesting they function as acid sensors and control excitability/ion homeostasis. Indeed, TASK-2 (KCNK5) has been shown to control excitability, volume regulation, bicarbonate handling, and apoptosis in a variety of tissues. With such diverse functions being ascribed to TASK-2, it is important to understand long-term as well as short-term regulation of this important channel. Thus, we have cloned the TASK-2 promoter, demonstrated that its transcriptional activity is dependent upon pO(2), shown that deletion of overlapping consensus binding sites for NF-kappaB/Elk-1 ablates this O(2) sensitivity, and proved that Elk-1 binds preferentially to this site. Furthermore, the consequences of chronic hypoxia on natively expressed TASK-2 are decreased steady-state mRNA and cell depolarization showing that TASK-2 contributes to the excitability of this important lung cell type.
Collapse
|
23
|
Sanders KM, Koh SD. Two-pore-domain potassium channels in smooth muscles: new components of myogenic regulation. J Physiol 2005; 570:37-43. [PMID: 16239268 PMCID: PMC1464292 DOI: 10.1113/jphysiol.2005.098897] [Citation(s) in RCA: 59] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Gastrointestinal (GI) smooth muscles are influenced by many levels of regulation, including those provided by enteric motor neurones, hormones and paracrine substances. The integrated contractile responses to these regulatory mechanisms depend heavily on the state of excitability of smooth muscle cells. Resting ionic conductances and myogenic responses to agonists and physical parameters, such as stretch, are important in establishing basal excitability. This review discusses the role of 2-pore-domain K+ channels in contributing to background conductances and in mediating responses of GI muscles to enteric inhibitory nerve stimulation and stretch. Murine GI muscles express TREK-1 channels and display a stretch-dependent K+ (SDK) conductance that is also activated by nitric oxide via a cGMP-dependent mechanism. Cloning and expression of mTREK-1 produced an SDK conductance that was activated by cGMP-dependent phosphorylation at serine-351. GI muscle cells also express TASK-1 and TASK-2 channels that are inhibited by lidocaine and external acidification. These conductances appear to provide significant background K+ permeability that contributes to the negative resting potentials of GI muscles.
Collapse
Affiliation(s)
- Kenton M Sanders
- Department of Physiology and Cell Biology, University of Nevada School of Medicine, Reno, NV 89557, USA.
| | | |
Collapse
|