59 results on '"Hisao Yamamura"'
Search Results
2. Involvement of small-conductance Ca2+-activated K+ (SKCa2) channels in spontaneous Ca2+ oscillations in rat pinealocytes
- Author
-
Shunsuke Ando, Hiroya Mizutani, Makoto Muramatsu, Yumiko Hagihara, Hiroki Mishima, Rubii Kondo, Yoshiaki Suzuki, Yuji Imaizumi, and Hisao Yamamura
- Subjects
Biophysics ,Cell Biology ,Molecular Biology ,Biochemistry - Published
- 2022
3. Swelling-activated ClC-3 activity regulates prostaglandin E2 release in human OUMS-27 chondrocytes
- Author
-
Eiva Bernotiene, Satoshi Yamada, Yuji Imaizumi, Wayne R. Giles, Hisao Yamamura, and Yoshiaki Suzuki
- Subjects
0301 basic medicine ,Gene knockdown ,urogenital system ,Cartilage ,Niflumic acid ,Biophysics ,Stimulation ,Cell Biology ,Biochemistry ,Chondrocyte ,Cell biology ,03 medical and health sciences ,chemistry.chemical_compound ,030104 developmental biology ,0302 clinical medicine ,medicine.anatomical_structure ,chemistry ,DIDS ,030220 oncology & carcinogenesis ,medicine ,Channel blocker ,Prostaglandin E2 ,Molecular Biology ,medicine.drug - Abstract
Articular chondrocytes are exposed to dynamic osmotic environments during normal joint loading, and thus, require effective volume regulatory mechanisms. A regulatory volume decrease (RVD) is one of the mechanisms for protecting chondrocytes from swelling and damage. Swelling-activated Cl− currents (ICl,swell) are responsible for the RVD, but the molecular identity in chondrocytes is largely unknown. In this study, we reveal that in human OUMS-27 chondrocytes, ICl,swell can be elicited by hypoosmotic stimulation (180 mOsm) and be inhibited by classical Cl− channel blockers, 4,4′-diisothiocyano-2,2′-stilbenedisulfonic acid (DIDS) and niflumic acid, and be attenuated by siRNA knockdown of ClC-3. Our molecular analyses revealed that ClC-3A is expressed as a major splice variant in both human articular chondrocytes and OUMS-27 cells. The onset and early phase of RVD following hypoosmotic stress in OUMS-27 cells were affected by DIDS and ClC-3 knockdown. Hypoosmotic stimulation caused Ca2+ influx and subsequent release of prostaglandin E2 (PGE2) in OUMS-27 cells, and both of these responses were reduced by DIDS and ClC-3 knockdown. These results strongly suggest that ClC-3 is responsible for ICl,swell and RVD under the hypoosmotic environments. It is likely that ClC-3 is associated with the pathogenesis of cartilage degenerative diseases including osteoarthritis via PGE2 release.
- Published
- 2021
4. Involvement of the γ1 subunit of the large-conductance Ca2+-activated K+ channel in the proliferation of human somatostatinoma cells
- Author
-
Yuji Imaizumi, Hisao Yamamura, Yoshiaki Suzuki, Kana Chikazawa, and Sayuri Noda
- Subjects
0301 basic medicine ,Cell growth ,Protein subunit ,Biophysics ,Depolarization ,Cell Biology ,Somatostatinoma ,medicine.disease ,Biochemistry ,Cell biology ,03 medical and health sciences ,chemistry.chemical_compound ,030104 developmental biology ,0302 clinical medicine ,Somatostatin ,chemistry ,030220 oncology & carcinogenesis ,Cancer cell ,medicine ,Secretion ,Paxilline ,Molecular Biology - Abstract
Pancreatic neuroendocrine tumors (pNETs) occur due to the abnormal growth of pancreatic islet cells and predominantly develop in the duodenal-pancreatic region. Somatostatinoma is one of the pNETs associated with tumors of pancreatic δ cells, which produce and secrete somatostatin. Limited information is currently available on the pathogenic mechanisms of somatostatinoma. The large-conductance Ca2+-activated K+ (BKCa) channel is expressed in several types of cancer cells and regulates cell proliferation, migration, invasion, and metastasis. In the present study, the functional expression of the BKCa channel was examined in a human somatostatinoma QGP-1 cell line. In QGP-1 cells, outward currents were elicited by membrane depolarization at pCa 6.5 (300 nM) in the pipette solution and inhibited by the specific BKCa channel blocker, paxilline. Paxilline-sensitive currents were detected, even at pCa 8.0 (10 nM) in the pipette solution, in QGP-1 cells. In addition to the α and β2-4 subunits of the BKCa channel, the novel regulatory γ1 subunit (BKCaγ1) was co-localized with the α subunit in QGP-1 cells. Paxilline-sensitive currents at pCa 8.0 in the pipette solution were reduced by the siRNA knockdown of BKCaγ1. Store-operated Ca2+ entry was smaller in BKCaγ1 siRNA-treated QGP-1 cells. The proliferation of QGP-1 cells was attenuated by paxilline or the siRNA knockdown of BKCaγ1. These results strongly suggest that BKCaγ1 facilitates the proliferation of human somatostatinoma cells. Therefore, BKCaγ1 may be a novel therapeutic target for somatostatinoma.
- Published
- 2020
5. Oxidative stress facilitates cell death by inhibiting Orai1-mediated Ca2+ entry in brain capillary endothelial cells
- Author
-
Kiyofumi Asai, Hideto Yamamura, Yuji Imaizumi, Yoshiaki Suzuki, and Hisao Yamamura
- Subjects
0301 basic medicine ,chemistry.chemical_classification ,Reactive oxygen species ,Programmed cell death ,ORAI1 ,Chemistry ,Cell growth ,Biophysics ,Cell Biology ,Oxidative phosphorylation ,medicine.disease_cause ,Biochemistry ,Cell biology ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,030220 oncology & carcinogenesis ,medicine ,Viability assay ,Molecular Biology ,Intracellular ,Oxidative stress - Abstract
Brain capillary endothelial cells (BCECs) form the blood-brain barrier (BBB) and play an essential role in the regulation of its functions. Oxidative stress accumulates excessive reactive oxygen species (ROS) and facilitates the death of BCECs, leading to a dysfunctional BBB. However, the mechanisms underlying the death of BCECs under oxidative stress remain unclear. In the present study, the effects of oxidative stress on cell viability, ROS production, intracellular Ca2+ concentration, and protein expression were examined using a cell line derived from bovine BCECs, t-BBEC117. When t-BBEC117 cells were exposed to oxidative stress induced by hydrogen peroxide (H2O2, 10–100 μM), cell growth was inhibited in a dose-dependent manner. Oxidative stress by 30 μM H2O2 increased the production of ROS and its effects were blocked by the ROS scavenger, 10 mM N-acetyl- l -cysteine (NAC). In addition, oxidative stress reduced store-operated Ca2+ entry (SOCE) and this decrease was recovered by NAC or the Orai channel activator, 5 μM 2-aminoethyl diphenylborinate (2-APB). The siRNA knockdown of Orai1 revealed that Orai1 was mainly responsible for SOCE channels and its activity was decreased by oxidative stress. However, the protein expression of Orai1 and STIM1 was not affected by oxidative stress. Oxidative stress-induced cell death was rescued by 2-APB, NAC, or the STIM-Orai activating region. In conclusion, oxidative stress reduces Orai1-mediated SOCE and, thus, facilitates the death of BCECs.
- Published
- 2020
6. Roles of LRRC26 as an auxiliary γ1-subunit of large-conductance Ca2+-activated K+ channels in bronchial smooth muscle cells
- Author
-
Yuji Imaizumi, Yoshiaki Suzuki, Sayuri Noda, Wayne R. Giles, and Hisao Yamamura
- Subjects
0301 basic medicine ,Pulmonary and Respiratory Medicine ,BK channel ,biology ,Physiology ,Chemistry ,Protein subunit ,Conductance ,Cell Biology ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,Smooth muscle ,Physiology (medical) ,Negative feedback ,biology.protein ,Biophysics ,030217 neurology & neurosurgery ,K channels ,Communication channel - Abstract
In visceral smooth muscle cells (SMCs), the large-conductance Ca2+-activated K+ (BK) channel is one of the key elements underlying a negative feedback mechanism that is essential for the regulation of intracellular Ca2+ concentration. Although leucine-rich repeat-containing (LRRC) proteins have been identified as novel auxiliary γ-subunits of the BK channel (BKγ) in several cell types, its physiological roles in SMCs are unclear. The BKγ expression patterns in selected SM tissues were examined using real-time PCR analyses and Western blotting. The functional contribution of BKγ1 to BK channel activity was examined by whole cell patch-clamp in SMCs and heterologous expression systems. BKγ1 expression in mouse bronchial SMCs (mBSMCs) was higher than in other several SMC types. Coimmunoprecipitation and total internal reflection fluorescence imaging analyses revealed molecular interaction between BKα and BKγ1 in mBSMCs. Under voltage-clamp, steady-state activation of BK channel currents at pCa 8.0 in mBSMCs occurred in a voltage range comparable to that of reconstituted BKα/BKγ1 complex. However, this range was much more negative than in mouse aortic SMCs (mASMCs) or in HEK293 cells expressing BKα alone and β-subunit (BKβ1). Mallotoxin, a selective activator of BK channel that lacks BKγ1, dose-dependently activated BK currents in mASMCs but not in mBSMCs. The abundant expression of BKγ1 in mBSMCs extensively facilitates BK channel activity to keep the resting membrane potential at negative values and prevents contraction under physiological conditions.
- Published
- 2020
7. SKF96365 activates calcium-sensing receptors in pulmonary arterial smooth muscle cells
- Author
-
Riko Miyaki, Aya Yamamura, Akiko Kawade, Moe Fujiwara, Rubii Kondo, Yoshiaki Suzuki, and Hisao Yamamura
- Subjects
Hypertension, Pulmonary ,Myocytes, Smooth Muscle ,Biophysics ,Imidazoles ,Cell Biology ,Pulmonary Artery ,Biochemistry ,Humans ,Calcium ,Familial Primary Pulmonary Hypertension ,Molecular Biology ,Receptors, Calcium-Sensing ,Cells, Cultured ,Cell Proliferation - Abstract
In pulmonary arterial smooth muscle cells (PASMCs), an increase in the cytosolic Ca
- Published
- 2021
8. A junctophilin-caveolin interaction enables efficient coupling between ryanodine receptors and BKCa channels in the Ca2+ microdomain of vascular smooth muscle
- Author
-
Takanori Saeki, Yuji Imaizumi, Hiroshi Takeshima, Yoshiaki Suzuki, and Hisao Yamamura
- Subjects
0301 basic medicine ,Vascular smooth muscle ,030102 biochemistry & molecular biology ,Ryanodine receptor ,Chemistry ,Calcium channel ,Lipid microdomain ,Cell Biology ,Membrane hyperpolarization ,musculoskeletal system ,Biochemistry ,Potassium channel ,03 medical and health sciences ,030104 developmental biology ,Caveolae ,cardiovascular system ,Biophysics ,Patch clamp ,tissues ,Molecular Biology - Abstract
Functional coupling between large-conductance Ca2+-activated K+ (BKCa) channels in the plasma membrane (PM) and ryanodine receptors (RyRs) in the sarcoplasmic reticulum (SR) is an essential mechanism for regulating mechanical force in most smooth muscle (SM) tissues. Spontaneous Ca2+ release through RyRs (Ca2+ sparks) and subsequent BKCa channel activation occur within the PM-SR junctional sites. We report here that a molecular interaction of caveolin-1 (Cav1), a caveola-forming protein, with junctophilin-2 (JP2), a bridging protein between PM and SR, positions BKCa channels near RyRs in SM cells (SMCs) and thereby contributes to the formation of a molecular complex essential for Ca2+ microdomain function. Approximately half of all Ca2+ sparks occurred within a close distance (
- Published
- 2019
9. Development of a Novel Cell-Based Assay System for High-Throughput Screening of Compounds Acting on Background Two-Pore Domain K+ Channels
- Author
-
Yuji Imaizumi, Hisao Yamamura, Keisuke Kawasaki, and Yoshiaki Suzuki
- Subjects
Programmed cell death ,High-throughput screening ,Genetic Vectors ,Cell ,Cell Culture Techniques ,Drug Evaluation, Preclinical ,Models, Biological ,01 natural sciences ,Biochemistry ,Analytical Chemistry ,03 medical and health sciences ,Potassium Channels, Tandem Pore Domain ,Potassium Channel Blockers ,medicine ,Humans ,Viability assay ,Ion channel ,030304 developmental biology ,Membrane potential ,0303 health sciences ,Dose-Response Relationship, Drug ,Chemistry ,HEK 293 cells ,Reproducibility of Results ,Depolarization ,Electrophysiological Phenomena ,High-Throughput Screening Assays ,0104 chemical sciences ,010404 medicinal & biomolecular chemistry ,HEK293 Cells ,medicine.anatomical_structure ,Biophysics ,Molecular Medicine ,Ion Channel Gating ,Biotechnology - Abstract
Two-pore domain K+ (K2P) channels are thought to be druggable targets. However, only a few agents specific for K2P channels have been identified, presumably due to the lack of an efficient screening system. To develop a new high-throughput screening (HTS) system targeting these channels, we have established a HEK293-based "test cell" expressing a mutated Na+ channel (Nav1.5) with markedly slowed inactivation, as well as a K+ channel (Kir2.1) that sets the membrane potential quite negative, close to K+ equilibrium potential. We found in this system that Kir2.1 block by 100 μM Ba2+ application consistently elicited a large depolarization like a long-lasting action potential. This maneuver resulted in cell death, presumably due to the sustained Na+ influx. When either the TWIK-related acid-sensitive K+ (TASK)-1 or TASK-3 channel was expressed in the test cells, Ba2+-induced cell death was markedly weakened. Stronger activation of TASK-1 by extracellular acidification further decreased the cell death. In contrast, the presence of K2P channel blockers enhanced cell death. IC50 values for TASK-1 and/or TASK-3 blockers acquired by measurements of relative cell viability were comparable to those obtained using patch-clamp recordings. Both blockers and openers of K2P channels can be accurately assessed with high efficiency and throughput by this novel HTS system.
- Published
- 2019
10. Rapid Na+ accumulation by a sustained action potential impairs mitochondria function and induces apoptosis in HEK293 cells expressing non-inactivating Na+ channels
- Author
-
Hisao Yamamura, Yoshiaki Suzuki, Yuji Imaizumi, and Keisuke Kawasaki
- Subjects
0301 basic medicine ,Membrane potential ,Programmed cell death ,Biophysics ,Depolarization ,Cell Biology ,Phosphatidylserine ,Mitochondrion ,Biochemistry ,Cell biology ,03 medical and health sciences ,chemistry.chemical_compound ,030104 developmental biology ,0302 clinical medicine ,chemistry ,Cell culture ,Apoptosis ,030220 oncology & carcinogenesis ,Molecular Biology ,Intracellular - Abstract
The mechanisms underlying neuronal cell death induced by the rise of intracellular Na+ concentration ([Na+]i) following abnormal hyperexcitation are not fully understood. Previously, we have established a recombinant cell line derived from HEK293 cells, in which the occurrence of a sustained action potential (AP) induces cell death. Mutated voltage-gated Nav1.5 channel (IFM/QQQ) lacking inactivation, and inward rectifying K+ channel (Kir2.1) were co-expressed in HEK293 cells (IFM/QQQ + Kir2.1 cells). In this cell line, the rise of [Na+]i due to a sustained AP reached maximum within 15 min without concomitant [Ca2+]i rise, and then elicited significant externalization of phosphatidylserine and enhancement of caspase activity. Marked decreases in mitochondrial transmembrane potential and ATP concentration were also detected. The significant cell death occurred at 3 h from the AP onset and reached a steady state at around 12 h. The significant release of lactate dehydrogenase was not detected even after 12 h. These results provide novel findings that Na+ accumulation or/and possibly concomitant K+ loss elicits apoptosis presumably due to the mitochondrial dysfunction, which is attributable to neither the membrane depolarization nor [Ca2+]i change. This apoptotic mechanism may be involved, at least in part, in neuronal cell death under pathophysiological settings with abnormal hyperexcitability.
- Published
- 2019
11. Conversion of Ca2+ oscillation into propagative electrical signals by Ca2+-activated ion channels and connexin as a reconstituted Ca2+ clock model for the pacemaker activity
- Author
-
Shinsuke Nakayama, Susumu Ohya, Yuji Imaizumi, Yoshiaki Suzuki, Hisao Yamamura, Keigo Hashidume, Takashi Murayama, Taisuke Kimura, and Takanori Saeki
- Subjects
0301 basic medicine ,Cell type ,Ryanodine receptor ,Chemistry ,Biophysics ,Gap junction ,Connexin ,Cell Biology ,Biochemistry ,Intracellular signal transduction ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,030220 oncology & carcinogenesis ,Oscillation (cell signaling) ,Molecular Biology ,Ion channel ,Intracellular - Abstract
Conversion of intracellular Ca2+ signals to electrical activity results in multiple and differing physiological impacts depending on cell types. In some organs such as gastrointestinal and urinary systems, spontaneous Ca2+ oscillation in pacermaker cells can function essentially as a Ca2+ clock mechanism, which has been originally found in pacemaking in sinoatrial node cell of the heart. The conversion of discrete Ca2+ clock events to spontaneous electrical activity is an essential step for the initiation and propagation of pacemaker activity through the multicellular organs resulting in synchronized physiological functions. Here, a model of intracellular signal transduction from a Ca2+ oscillation to initiation of electrical slow waves and their propagation were reconstituted in HEK293 cells. This was accomplished based on ryanodine receptor (RyR) type 3, Ca2+-activated ion channels, i.e. small conductance Ca2+-activated K+ channel (SK2) or Ca2+-activated Cl− channel (TMEM16A), and connexin43 being heterologously co-expressed. The propagation of electrical waves was abolished or substantially reduced by treatment with selective blockers of the expressed channels and 18β-glycyrrhetinic acid, a gap junction inhibitor, respectively. Thus, we demonstrated that the conversion of Ca2+ oscillation to electrical signals with cell to cell propagation can be reconstituted as a model of Ca2+ clock pacemaker activity by combinational expression of critical elements in heterologous expression system.
- Published
- 2019
12. Single Molecule Fluorescence Imaging Reveals the Stoichiometry of BKγ1 Subunit in Living HEK293 Cell Expression System
- Author
-
Sayuri Noda, Yuji Imaizumi, Yoshiaki Suzuki, and Hisao Yamamura
- Subjects
0301 basic medicine ,Pharmacology ,Total internal reflection fluorescence microscope ,Chemistry ,Protein subunit ,HEK 293 cells ,Optical Imaging ,Pharmaceutical Science ,General Medicine ,Single-molecule experiment ,Single Molecule Imaging ,Green fluorescent protein ,03 medical and health sciences ,Protein Subunits ,030104 developmental biology ,0302 clinical medicine ,HEK293 Cells ,030220 oncology & carcinogenesis ,Biophysics ,Humans ,Patch clamp ,Large-Conductance Calcium-Activated Potassium Channels ,Intracellular - Abstract
Large conductance Ca2+-activated K+ (BKCa) channels are ubiquitously expressed in plasma membrane of both excitable and non-excitable cells and possess significant physiological functions. A tetrameric complex of α subunit (BKα) forms a functional pore of BKCa channel. The properties of BKCa channel, such as voltage-dependence, Ca2+ sensitivity and pharmacological responses, are extensively modulated by co-expressing accessory β subunits (BKβ), which can associate with BKα in one to one manner. Although the functional significance of newly identified γ subunits (BKγ) has been revealed, the stoichiometry between BKα and BKγ1 remains unclear. In the present study, we utilized a single molecule fluorescence imaging with a total internal reflection fluorescence (TIRF) microscope to directly count the number of green fluorescent protein (GFP)-tagged BKγ1 (BKγ1-GFP) within a single BKCa channel complex in HEK293 cell expression system. BKγ1-GFP significantly enhanced the BK channel activity even when the intracellular Ca2+ concentration was kept lower, i.e., 10 nM, than the physiological resting level. BKγ1-GFP stably formed molecular complexes with BKα-mCherry in the plasma membrane. Counting of GFP bleaching steps revealed that a BKCa channel can contain up to four BKγ1 per channel at the maximum. These results suggest that BKγ1 forms a BKCa channel complex with BKα in a 1 : 1 stoichiometry in a human cell line.
- Published
- 2020
13. Heterodimerization of two pore domain K+ channel TASK1 and TALK2 in HEK293 heterologous expression systems
- Author
-
Tatsuya Miyamoto, Hisao Yamamura, Kanako Tsutsumi, Yoshiaki Suzuki, and Yuji Imaizumi
- Subjects
Physics ,Applied Mathematics ,General Mathematics ,HEK 293 cells ,Biophysics ,Heterologous expression ,K channels ,Domain (software engineering) - Published
- 2018
14. Local Ca2+ coupling between mitochondria and sarcoplasmic reticulum following depolarization in guinea pig urinary bladder smooth muscle cells
- Author
-
Yuji Imaizumi, Sou Inagaki, Hisao Yamamura, Yoshiaki Suzuki, and Keisuke Kawasaki
- Subjects
0301 basic medicine ,Membrane potential ,Physiology ,Ryanodine receptor ,Endoplasmic reticulum ,Depolarization ,Cell Biology ,03 medical and health sciences ,chemistry.chemical_compound ,030104 developmental biology ,chemistry ,Biochemistry ,Biophysics ,Repolarization ,Carbonyl cyanide-p-trifluoromethoxyphenylhydrazone ,Ion channel ,Calcium signaling - Abstract
Spatiotemporal changes in cytosolic Ca2+ concentration ([Ca2+]c) trigger a number of physiological functions in smooth muscle cells (SMCs). We previously imaged Ca2+-induced Ca2+ release following membrane depolarization as local Ca2+ transients, Ca2+ hotspots, in subplasmalemmal regions. In this study, the physiological significance of mitochondria on local Ca2+ signaling was examined. Cytosolic and mitochondrial Ca2+ images following depolarization or action potentials were recorded in single SMCs from the guinea pig urinary bladder using a fast-scanning confocal fluorescent microscope. Depolarization- and action potential-induced [Ca2+]c transients occurred at several discrete sites in subplasmalemmal regions, peaked within 30 ms, and then spread throughout the whole-cell. In contrast, Ca2+ concentration in the mitochondria matrix ([Ca2+]m) increased after a delay of ~50 ms from the start of depolarization, and then peaked within 500 ms. Following repolarization, [Ca2+]c returned to the resting level with a half-decay time of ~500 ms, while [Ca2+]m recovered more slowly (∼1.5 s). Carbonyl cyanide 4-(trifluoromethoxy)phenylhydrazone, a mitochondrial uncoupler, abolished depolarization-induced [Ca2+]m elevations and slowed [Ca2+]c changes. Importantly, short depolarization-induced changes in [Ca2+]m and transmembrane potential in mitochondria coupled to Ca2+ hotspots were significantly larger than those in other mitochondria. Total internal reflection fluorescence imaging revealed that a subset of mitochondria closely localized with ryanodine receptors and voltage-dependent Ca2+ channels. These results indicate that particular mitochondria are functionally coupled to ion channels and sarcoplasmic reticulum fragments within the local Ca2+ microdomain, and thus, strongly contribute to [Ca2+]c regulation in SMCs.
- Published
- 2018
15. Molecular mechanisms underlying pimaric acid-induced modulation of voltage-gated K+ channels
- Author
-
Yuji Imaizumi, Susumu Ohya, Katsuhiko Muraki, Hisao Yamamura, Kazuho Sakamoto, and Yoshiaki Suzuki
- Subjects
0301 basic medicine ,Rosin acid ,endocrine system diseases ,Action Potentials ,Voltage-Gated K+ Channels ,Pimaric acid ,Kv channel ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,Drug Discovery ,Humans ,Point Mutation ,Amino Acid Sequence ,Molecular Targeted Therapy ,Peptide sequence ,Voltage-gated K+ channel ,Pharmacology ,Point mutation ,K+ channel opener ,lcsh:RM1-950 ,Mutagenesis ,nutritional and metabolic diseases ,Conductance ,lcsh:Therapeutics. Pharmacology ,HEK293 Cells ,030104 developmental biology ,chemistry ,Biochemistry ,Potassium Channels, Voltage-Gated ,Modulation ,Mutagenesis, Site-Directed ,Biophysics ,Molecular Medicine ,Calcium ,Diterpenes ,Channel gating ,Ca2+-activated K+ channel ,030217 neurology & neurosurgery - Abstract
Voltage-gated K+ (KV) channels, which control firing and shape of action potentials in excitable cells, are supposed to be potential therapeutic targets in many types of diseases. Pimaric acid (PiMA) is a unique opener of large conductance Ca2+-activated K+ channel. Here, we report that PiMA modulates recombinant rodent KV channel activity. The enhancement was significant at low potentials (
- Published
- 2017
16. Expression analysis of Ca2+-sensing receptor and TRPC6 channel in monocrotaline-induced pulmonary hypertensive rats
- Author
-
Aya Yamamura, Yoshiaki Suzuki, Hisao Yamamura, and Kawade Akiko
- Subjects
Chemistry ,Applied Mathematics ,General Mathematics ,Expression analysis ,Biophysics ,Channel (broadcasting) ,Receptor ,TRPC6 - Published
- 2021
17. Functional expression of two-pore domain K+ channels in pulmonary arterial hypertension
- Author
-
Aya Yamamura, Hisao Yamamura, Yoshiaki Suzuki, and Natsumi Shima
- Subjects
Physics ,Functional expression ,Applied Mathematics ,General Mathematics ,Biophysics ,Domain (software engineering) ,K channels - Published
- 2021
18. Inhibitory mechanism of melatonin on voltage-dependent Kv4.2 channels
- Author
-
Hisao Yamamura, Yoshiaki Suzuki, and Hiroki Mishima
- Subjects
Melatonin ,Chemistry ,Applied Mathematics ,General Mathematics ,Biophysics ,medicine ,Inhibitory postsynaptic potential ,Mechanism (sociology) ,Voltage ,medicine.drug - Published
- 2021
19. Protein-Coupled Fluorescent Probe To Visualize Potassium Ion Transition on Cellular Membranes
- Author
-
Manabu Shimonishi, Tetsuo Nagano, Tomoya Hirata, Takuya Terai, Kenjiro Hanaoka, Tasuku Ueno, Yasuteru Urano, Yuji Imaizumi, Hisao Yamamura, and Toru Komatsu
- Subjects
Boron Compounds ,BK channel ,Analytical chemistry ,010402 general chemistry ,01 natural sciences ,Potassium Ionophores ,Analytical Chemistry ,Cell membrane ,Crown Ethers ,Extracellular ,Fluorescence microscope ,medicine ,Humans ,Large-Conductance Calcium-Activated Potassium Channel alpha Subunits ,Fluorescent Dyes ,Total internal reflection fluorescence microscope ,biology ,010405 organic chemistry ,Chemistry ,Cell Membrane ,Cations, Monovalent ,Fluorescence ,0104 chemical sciences ,HEK293 Cells ,Membrane ,medicine.anatomical_structure ,Potassium ,Biophysics ,biology.protein ,HT29 Cells ,HeLa Cells - Abstract
K(+) is the most abundant metal ion in cells, and changes of [K(+)] around cell membranes play important roles in physiological events. However, there is no practical method to selectively visualize [K(+)] at the surface of cells. To address this issue, we have developed a protein-coupled fluorescent probe for K(+), TLSHalo. TLSHalo is responsive to [K(+)] in the physiological range, with good selectivity over Na(+) and retains its K(+)-sensing properties after covalent conjugation with HaloTag protein. By using cells expressing HaloTag on the plasma membrane, we successfully directed TLSHalo specifically to the outer surface of target cells. This enabled us to visualize localized extracellular [K(+)] change with TLSHalo under a fluorescence microscope in real time. To confirm the experimental value of this system, we used TLSHalo to monitor extracellular [K(+)] change induced by K(+) ionophores or by activation of a native Ca(2+)-dependent K(+) channel (BK channel). Further, we show that K(+) efflux via BK channel induced by electrical stimulation at the bottom surface of the cells can be visualized with TLSHalo by means of total internal reflection fluorescence microscope (TIRFM) imaging. Our methodology should be useful to analyze physiological K(+) dynamics with high spatiotemporal resolution.
- Published
- 2016
20. Inhibitory effect of melatonin on voltage-dependent potassium (Kv4.2) channels
- Author
-
Mishima Hiroki, Hisao Yamamura, and Yoshiaki Suzuki
- Subjects
Melatonin ,Chemistry ,Applied Mathematics ,General Mathematics ,Potassium ,Biophysics ,medicine ,chemistry.chemical_element ,Inhibitory effect ,Voltage ,medicine.drug - Published
- 2020
21. LRRC26 is functional as an Auxiliary Subunit of Large‐Conductance Ca 2+ ‐Activated K + (BK) Channel and regulates BK channel activity in Bronchial Smooth Muscle Cells
- Author
-
Hisao Yamamura, Yuji Imaizumi, Yoshiaki Suzuki, and Sayuri Noda
- Subjects
BK channel ,biology ,Smooth muscle ,Chemistry ,Protein subunit ,Genetics ,biology.protein ,Biophysics ,Conductance ,Molecular Biology ,Biochemistry ,Biotechnology ,BK channel activity - Published
- 2018
22. Functional molecular complexes of junctophilin‐2 and caveolin‐1 provide a structural/functional basis for Ca 2+ ‐microdomain formation between BK Ca channels and RyRs in vascular smooth muscle cells
- Author
-
Hiroshi Takeshima, Hisao Yamamura, Takanori Saeki, Yoshiaki Suzuki, and Yuji Imaizumi
- Subjects
Vascular smooth muscle ,Chemistry ,Caveolin 1 ,Lipid microdomain ,Genetics ,Biophysics ,Molecular Biology ,Biochemistry ,Ca channel ,Biotechnology - Published
- 2018
23. New light on ion channel imaging by total internal reflection fluorescence (TIRF) microscopy
- Author
-
Yuji Imaizumi, Hisao Yamamura, and Yoshiaki Suzuki
- Subjects
TIRF imaging ,Microscope ,Biophysics ,Cellular functions ,Nanotechnology ,Fluorescence ,Ion Channels ,law.invention ,Cell membrane ,law ,Subunit stoichiometry ,Drug Discovery ,Fluorescence Resonance Energy Transfer ,medicine ,Microscopy, Interference ,Calcium Signaling ,Single-molecule imaging ,Ion channel ,Pharmacology ,Total internal reflection fluorescence microscope ,Chemistry ,Cell Membrane ,lcsh:RM1-950 ,Single Molecule Imaging ,Molecular Imaging ,Membrane ,medicine.anatomical_structure ,Förster resonance energy transfer ,lcsh:Therapeutics. Pharmacology ,Microscopy, Fluorescence ,FRET ,Molecular Medicine - Abstract
Ion channels play pivotal roles in a wide variety of cellular functions; therefore, their physiological characteristics, pharmacological responses, and molecular structures have been extensively investigated. However, the mobility of an ion channel itself in the cell membrane has not been examined in as much detail. A total internal reflection fluorescence (TIRF) microscope allows fluorophores to be imaged in a restricted region within an evanescent field of less than 200 nm from the interface of the coverslip and plasma membrane in living cells. Thus the TIRF microscope is useful for selectively visualizing the plasmalemmal surface and subplasmalemmal zone. In this review, we focused on a single-molecule analysis of the dynamic movement of ion channels in the plasma membrane using TIRF microscopy. We also described two single-molecule imaging techniques under TIRF microscopy: fluorescence resonance energy transfer (FRET) for the identification of molecules that interact with ion channels, and subunit counting for the determination of subunit stoichiometry in a functional channel. TIRF imaging can also be used to analyze spatiotemporal Ca 2+ events in the subplasmalemma. Single-molecule analyses of ion channels and localized Ca 2+ signals based on TIRF imaging provide beneficial pharmacological and physiological information concerning the functions of ion channels.
- Published
- 2015
24. Regulation of store-operated Ca2+ entry activity by cell cycle dependent up-regulation of Orai2 in brain capillary endothelial cells
- Author
-
Yuji Imaizumi, Kiyofumi Asai, Hiroaki Kito, Hisao Yamamura, Hideto Yamamura, Yoshiaki Suzuki, and Susumu Ohya
- Subjects
Small interfering RNA ,Biophysics ,Biology ,Biochemistry ,Cell Line ,Downregulation and upregulation ,Animals ,Calcium Signaling ,RNA, Small Interfering ,Receptor ,Molecular Biology ,Cell Proliferation ,Membrane Glycoproteins ,Cell growth ,ORAI1 ,Brain ,Endothelial Cells ,Cell Cycle Checkpoints ,Cell Biology ,STIM2 ,Cell cycle ,Up-Regulation ,Cell biology ,Endothelial stem cell ,Blood-Brain Barrier ,Gene Knockdown Techniques ,Cattle ,Calcium Channels - Abstract
Store-operated Ca(2+) entry (SOCE) via Orai1 and STIM1 complex is supposed to have obligatory roles in the regulation of cellular functions of vascular endothelial cells, while little is known about the contribution of Orai2. Quantitative PCR and Western blot analyses indicated the expression of Orai2 and STIM2, in addition to Orai1 and STIM1 in bovine brain capillary endothelial cell line, t-BBEC117. During the exponential growth of t-BBEC117, the knockdown of Orai1 and STIM1 significantly reduced the SOCE activity, whereas Orai2 and STIM2 siRNAs had no effect. To examine whether endogenous SOCE activity contributes to the regulation of cell cycle progression, t-BBEC117 were synchronized using double thymidine blockage. At the G2/M phase, Ca(2+) influx via SOCE was decreased and Orai2 expression was increased compared to the G0/G1 phase. When Orai2 was knocked down at the G2/M phase, the decrease in SOCE was removed, and cell proliferation was partly attenuated. Taken together, Orai1 significantly contributes to cell proliferation via the functional expression, which is presumably independent of the cell cycle phases. In construct, Orai2 is specifically up-regulated during the G2/M phase, negatively modulates the SOCE activity, and may contribute to the regulation of cell cycle progression in brain capillary endothelial cells.
- Published
- 2015
25. The multiple expression of Ca2+-activated Cl− channels via homo- and hetero-dimer formation of TMEM16A splicing variants in murine portal vein
- Author
-
Takanori Saeki, Yoshiaki Suzuki, Junya Ohshiro, Hisao Yamamura, and Yuji Imaizumi
- Subjects
Gene isoform ,Total internal reflection fluorescence microscope ,HEK 293 cells ,Biophysics ,Cell Biology ,Biology ,Biochemistry ,Molecular biology ,Cell biology ,Cell membrane ,medicine.anatomical_structure ,Förster resonance energy transfer ,RNA splicing ,medicine ,Molecular Biology ,Immunostaining ,Intracellular - Abstract
Ca 2+ -activated Cl − channel (CaCC) often plays substantial roles in the regulation of membrane excitability in smooth muscle cells (SMCs). TMEM16A, a member of the TMEM16 family, has been suggested as the molecular entity responsible for CaCC in several types of SMCs. In this study, the expression of TMEM16A splicing variants and their contribution to CaCC activity were examined in murine portal vein SMCs (mPVSMCs). Four transcripts of TMEM16A splicing variants, which include four alternatively spliced segments (“ a ” and “ b ” in N-terminus and “ c ” and “ d ” in the first intracellular loop), were identified; the expression ratio of four transcripts of “ abc ”, “ acd ”, “ abcd ” and “ ac ” was 64.5, 25.8, 4.8 and 4.8%, respectively. The immunostaining of isolated mPVSMCs with anti-TMEM16A antibody indicates the abundant expression of TMEM16A on the cell membrane. CaCC currents recorded in mPVSMCs were markedly reduced by T16A inh -A01, a specific TMEM16A inhibitor. When the two major TMEM16A splicing variants, abc and acd isoforms, were expressed separately in HEK293 cells, the CaCC currents, which possess similar electrophysiological characteristics to those in mPVSMCs were observed. The single-molecule photobleaching analyses using total internal reflection fluorescence (TIRF) microscope indicated that the distribution of stepwise photobleaching events was fit well with a binomial distribution for homodimer. Additionally, the heterodimer formation was suggested by fluorescence resonance energy transfer (FRET) analyses in HEK293 cells co-expressing CFP- or YFP-tagged variants. In conclusion, alternatively spliced variants of TMEM16A abc and acd in mPVSMCs are two major molecular entities of CaCC and may form hetero-/homo-dimers to be functional as CaCC in the regulation of membrane excitability and contractility in mPVSMCs.
- Published
- 2014
26. Ca2+ signaling and ion channels in pulmonary arterial hypertension
- Author
-
Hisao Yamamura
- Subjects
Chemistry ,Applied Mathematics ,General Mathematics ,Biophysics ,Ion channel - Published
- 2019
27. Enhancement of Ca2+Influx and Ciliary Beating by Membrane Hyperpolarization due to ATP-Sensitive K+Channel Opening in Mouse Airway Epithelial Cells
- Author
-
Susumu Ohya, Hisao Yamamura, Yoshiaki Suzuki, Yuji Imaizumi, Teruya Ohba, Eiji Sawada, and Hiroyuki Tsuda
- Subjects
Male ,medicine.medical_specialty ,Protein subunit ,Respiratory Mucosa ,Biology ,Sulfonylurea Receptors ,Glibenclamide ,Mice ,KATP Channels ,Internal medicine ,Diazoxide ,medicine ,Extracellular ,Animals ,Cilia ,Potassium Channels, Inwardly Rectifying ,Pharmacology ,Calcium metabolism ,Membrane hyperpolarization ,Potassium channel ,Mice, Inbred C57BL ,Endocrinology ,Biophysics ,Molecular Medicine ,Respiratory epithelium ,Calcium ,Ion Channel Gating ,medicine.drug - Abstract
Among the several types of cells composing the airway epithelium, the ciliary cells are responsible for one of the most important defense mechanisms of the airway epithelium: the transport of inhaled particles back up into the throat by coordinated ciliary movement. Changes in the cytoplasmic Ca(2+) concentration ([Ca(2+)]i) are the main driving force controlling the ciliary activity. In mouse ciliary cells, membrane hyperpolarization from -20 to -60 mV under whole-cell voltage-clamp induced a slow but significant [Ca(2+)]i rise in a reversible manner. This rise was completely inhibited by the removal of Ca(2+) from the extracellular solution. Application of diazoxide, an ATP-dependent K(+) channel opener, dose-dependently induced a membrane hyperpolarization (EC50 = 2.3 μM), which was prevented by the addition of 5 μM glibenclamide. An inwardly rectifying current was elicited by the application of 10 μM diazoxide and suppressed by subsequent addition of 5 μM glibenclamide. Moreover, the application of 10 μM diazoxide induced a significant [Ca(2+)]i rise and facilitated ciliary movement. Multi-cell reverse-transcription polymerase chain reaction analyses and immunocytochemical staining suggested that the subunit combination of Kir6.2/SUR2B and possibly also Kir6.1/SUR2B is expressed in ciliary cells. The confocal Ca(2+) imaging analyses suggested that the [Ca(2+)]i rise induced by diazoxide occurred preferentially in the apical submembrane region. In conclusion, the application of a KATP channel opener to airway ciliary cells induces membrane hyperpolarization and thereby induces a [Ca(2+)]i rise via the facilitation of Ca(2+) influx through the non-voltage-dependent Ca(2+) permeable channels. Therefore, a KATP opener may be beneficial in facilitating ciliary movement.
- Published
- 2013
28. Overactive bladder mediated by accelerated Ca2+influx mode of Na+/Ca2+exchanger in smooth muscle
- Author
-
Yuji Imaizumi, William C. Cole, Shingo Hotta, Satomi Kita, Takahiro Iwamoto, Hisao Yamamura, Yoshiaki Suzuki, Susumu Ohya, and Hidemichi Murata
- Subjects
Male ,medicine.medical_specialty ,Physiology ,Urinary Bladder ,Biological Transport, Active ,Mice, Transgenic ,Sodium-Calcium Exchanger ,Mice ,Smooth muscle ,Internal medicine ,Benzyl Compounds ,medicine ,Animals ,Kb r7943 ,Sodium-calcium exchanger ,Urinary Bladder, Overactive ,Chemistry ,Thiourea ,Ca2 influx ,Muscle, Smooth ,Cell Biology ,medicine.disease ,Cytosol ,Endocrinology ,Overactive bladder ,Biophysics ,Thiazolidines ,Calcium ,Anti-Arrhythmia Agents ,Muscle Contraction - Abstract
The Na+/Ca2+exchanger (NCX) is thought to be a key molecule in the regulation of cytosolic Ca2+dynamics. The relative importance of the two Ca2+transport modes of NCX activity leading to Ca2+efflux (forward) and influx (reverse) in smooth muscle, however, remains unclear. Unexpectedly, spontaneous contractions of urinary bladder smooth muscle (UBSM) were enhanced in transgenic mice overexpressing NCX1.3 (NCX1.3tg/tg). The enhanced activity was attenuated by KB-R7943 or SN-6. Whole cell outward NCX current sensitive to KB-R7943 or Ni2+was readily detected in UBSM cells from NCX1.3tg/tgbut not wild-type mice. Spontaneous Ca2+transients in myocytes of NCX1.3tg/tgwere larger and frequently resulted in propagating events and global elevations in cytosolic Ca2+concentration. Significantly, NCX1.3tg/tgmice exhibited a pattern of more frequent urination of smaller volumes and this phenotype was reversed by oral administration of KB-R7943. On the other hand, KB-R7943 did not improve it in KB-R7943-insensitive (G833C-)NCX1.3tg/tgmice. We conclude that NCX1.3 overexpression is associated with abnormal urination owing to enhanced Ca2+influx via reverse mode NCX leading to prolonged, propagating spontaneous Ca2+release events and a potentiation of spontaneous UBSM contraction. These findings suggest the possibility that NCX is a candidate molecular target for overactive bladder therapy.
- Published
- 2013
29. Direct molecular interaction of caveolin-3 with KCa1.1 channel in living HEK293 cell expression system
- Author
-
Susumu Ohya, Yuji Imaizumi, Yoshiaki Suzuki, and Hisao Yamamura
- Subjects
Caveolin 3 ,Green Fluorescent Proteins ,Molecular Sequence Data ,Biophysics ,Biology ,Caveolae ,Biochemistry ,Cell membrane ,Caveolin ,Fluorescence Resonance Energy Transfer ,medicine ,Humans ,Immunoprecipitation ,Large-Conductance Calcium-Activated Potassium Channel alpha Subunits ,Molecular Biology ,Total internal reflection fluorescence microscope ,Cell Membrane ,Cell Biology ,Photobleaching ,Protein Structure, Tertiary ,Cell biology ,HEK293 Cells ,medicine.anatomical_structure ,Förster resonance energy transfer ,mCherry - Abstract
Caveolin family is supposed to be essential molecules for the formation of not only caveola structure on cell membrane but also functional molecular complexes in them with direct and/or indirect interaction with other membrane and/or submembrane associated proteins. The direct coupling of caveolin-1 (cav1) with large conductance Ca(2+)-activated K(+) channel, KCa1.1 has been established in several types of cells and in expression system as well. The possible interaction of caveolin-3 (cav3), which shows expression in some differential tissues from cav1, with KCa1.1 remains to be determined. In the present study, the density of KCa1.1 current expressed in HEK293 cells was significantly reduced by the co-expression of cav3, as well as cav1. The co-localization and direct interaction of GFP- or CFP-labeled cav3 (GFP/CFP-cav3) with YFP- or mCherry-labeled KCa1.1 (KCa1.1-YFP/mCherry) were clearly demonstrated by single molecular image analyses using total internal reflection fluorescence (TIRF) microscopy and fluorescence resonance energy transfer (FRET) analyses with acceptor photobleaching method. The deletion of suggested cav1-binding motif in C terminus region of KCa1.1 (KCa1.1ΔCB-YFP) resulted in the marked decrease in cell surface expression, co-localization and FRET efficiency with CFP-cav3 and CFP-cav1. The FLAG-KCa1.1 co-immunoprecipitation with GFP-cav3 or GFP-cav1 also supported their direct molecular interaction. These results strongly suggest that cav3 possesses direct interaction with KCa1.1, presumably at the same domain for cav1 binding. This interaction regulates KCa1.1 expression to cell surface and the formation of functional molecular complex in caveolae in living cells.
- Published
- 2013
30. Hypoxic stress up-regulates Kir2.1 expression and facilitates cell proliferation in brain capillary endothelial cells
- Author
-
Hisao Yamamura, Yoshiaki Suzuki, Kiyofumi Asai, Hideto Yamamura, and Yuji Imaizumi
- Subjects
0301 basic medicine ,Biophysics ,Biology ,Biochemistry ,Membrane Potentials ,Brain ischemia ,03 medical and health sciences ,Transient receptor potential channel ,Stress, Physiological ,medicine ,Animals ,Potassium Channels, Inwardly Rectifying ,Molecular Biology ,Cells, Cultured ,Cell Proliferation ,Membrane potential ,SOC channels ,Inward-rectifier potassium ion channel ,Brain ,Endothelial Cells ,Cell Biology ,Anatomy ,Hypoxia (medical) ,Hyperpolarization (biology) ,medicine.disease ,Cell Hypoxia ,Cell biology ,Up-Regulation ,Endothelial stem cell ,030104 developmental biology ,Calcium ,Cattle ,medicine.symptom - Abstract
The blood-brain barrier (BBB) is mainly composed of brain capillary endothelial cells (BCECs), astrocytes and pericytes. Brain ischemia causes hypoxic encephalopathy and damages BBB. However, it remains still unclear how hypoxia affects BCECs. In the present study, t-BBEC117 cells, an immortalized bovine brain endothelial cell line, were cultured under hypoxic conditions at 4-5% oxygen for 72 h. This hypoxic stress caused hyperpolarization of resting membrane potential. Patch-clamp recordings revealed a marked increase in Ba(2+)-sensitive inward rectifier K(+) current in t-BBEC117 cells after hypoxic culture. Western blot and real-time PCR analyses showed that Kir2.1 expression was significantly up-regulated at protein level but not at mRNA level after the hypoxic culture. Ca(2+) imaging study revealed that the hypoxic stress enhanced store-operated Ca(2+) (SOC) entry, which was significantly reduced in the presence of 100 μM Ba(2+). On the other hand, the expression of SOC channels such as Orai1, Orai2, and transient receptor potential channels was not affected by hypoxic stress. MTT assay showed that the hypoxic stress significantly enhanced t-BBEC117 cell proliferation, which was inhibited by approximately 60% in the presence of 100 μM Ba(2+). We first show here that moderate cellular stress by cultivation under hypoxic conditions hyperpolarizes membrane potential via the up-regulation of functional Kir2.1 expression and presumably enhances Ca(2+) entry, resulting in the facilitation of BCEC proliferation. These findings suggest potential roles of Kir2.1 expression in functional changes of BCECs in BBB following ischemia.
- Published
- 2016
31. Total internal reflection fluorescence imaging of Ca2+-induced Ca2+ release in mouse urinary bladder smooth muscle cells
- Author
-
Hisao Yamamura and Yuji Imaizumi
- Subjects
Myocytes, Smooth Muscle ,Urinary Bladder ,Biophysics ,Analytical chemistry ,Cell Separation ,Biochemistry ,Mice ,Animals ,Calcium Signaling ,Large-Conductance Calcium-Activated Potassium Channels ,Molecular Biology ,Cells, Cultured ,Total internal reflection fluorescence microscope ,Ryanodine receptor ,Chemistry ,Endoplasmic reticulum ,Lipid microdomain ,Ryanodine Receptor Calcium Release Channel ,Depolarization ,Cell Biology ,Fluorescence ,Molecular Imaging ,Mouse Urinary Bladder ,Membrane ,Microscopy, Fluorescence ,cardiovascular system ,Calcium ,Calcium Channels - Abstract
In smooth muscles (SMs), cytosolic Ca 2+ ([Ca 2+ ] cyt ) dynamics during an action potential are triggered by Ca 2+ influx through voltage-dependent Ca 2+ channels (VDCCs) in the plasma membrane. The physiological significance of Ca 2+ amplification by subsequent Ca 2+ release through ryanodine receptors (RyRs) from the sarcoplasmic reticulum (SR) is still a matter of topics in SMs. In the present study, depolarization-evoked local Ca 2+ dynamics in Ca 2+ microdomain were imaged using total internal reflection fluorescence (TIRF) microscopy in mouse urinary bladder SM cells (UBSMCs). Upon depolarization under whole-cell voltage-clamp, the rapid and local elevation of [Ca 2+ ] cyt was followed by larger [Ca 2+ ] cyt increase with propagation occurred in a limited TIRF zone within ∼200 nm from cell surface. The depolarization-evoked [Ca 2+ ] cyt increase in a TIRF zone was abolished or greatly reduced by the pretreatment with Cd 2+ or ryanodine, respectively. The initial local [Ca 2+ ] cyt increases were mediated by Ca 2+ influx through single or clustered VDCCs as Ca 2+ sparklets, and the following step was elicited by Ca 2+ -induced Ca 2+ release (CICR) through RyR from SR. The depolarization-induced outward currents, mainly due to large-conductance Ca 2+ -activated K + channel activation, were also markedly reduced by Cd 2+ and ryanodine. In addition, TIRF analyses showed that the fluorescent signals of individual or clustered VDCC distributed in relatively uniform fashion and that a subset of RyRs in the subplasmalemmal SR also located in TIRF zone. In conclusion, fast TIRF imaging successfully demonstrated two step Ca 2+ events upon depolarization in Ca 2+ microdomain of UBSMCs; the initial Ca 2+ influx as Ca 2+ sparklets through discrete VDCC or their clusters and the following CICR via the activation of loosely coupled RyRs in SR located in the Ca 2+ microdomains.
- Published
- 2012
32. Involvement of Inositol 1,4,5-Trisphosphate Formation in the Voltage-Dependent Regulation of the Ca2+Concentration in Porcine Coronary Arterial Smooth Muscle Cells
- Author
-
Katsuhiko Muraki, Yuji Imaizumi, Hisao Yamamura, and Susumu Ohya
- Subjects
Potassium Channels ,Swine ,Voltage clamp ,Myocytes, Smooth Muscle ,Inositol 1,4,5-Trisphosphate ,Muscle, Smooth, Vascular ,Membrane Potentials ,chemistry.chemical_compound ,Animals ,Inositol 1,4,5-Trisphosphate Receptors ,Myocyte ,Inositol ,Evans Blue ,Pharmacology ,Phospholipase C ,Heparin ,Chemistry ,Depolarization ,Membrane hyperpolarization ,3-Pyridinecarboxylic acid, 1,4-dihydro-2,6-dimethyl-5-nitro-4-(2-(trifluoromethyl)phenyl)-, Methyl ester ,Hyperpolarization (biology) ,Coronary Vessels ,Biochemistry ,Biophysics ,Molecular Medicine ,Calcium ,Calcium Channels - Abstract
The involvement of inositol 1,4,5-trisphosphate (IP(3)) formation in the voltage-dependent regulation of intracellular Ca(2+) concentration ([Ca(2+)](i)) was examined in smooth muscle cells of the porcine coronary artery. Slow ramp depolarization from -90 to 0 mV induced progressive [Ca(2+)](i) increase. The slope was reduced or increased in the presence of Cd(2+) or (±)-1,4-dihydro-2,6-dimethyl-5-nitro-4-(2-[trifluoromethyl]-phenyl)pyridine-3-carboxlic acid methyl ester (Bay K 8644), respectively. The decrease in [Ca(2+)](i) via the membrane hyperpolarization induced by K(+) channel openers (levcromakalim and Evans blue) under current clamp was identical to that under voltage clamp. The step hyperpolarization from -40 to -80 mV reduced [Ca(2+)](i) uniformly over the whole-cell area with a time constant of ∼10 s. The [Ca(2+)](i) at either potential was unaffected by heparin, an inhibitor of IP(3) receptors. Alternatively, [Ca(2+)](i) rapidly increased in the peripheral regions by depolarization from -80 to 0 mV and stayed at that level (∼400 nM) during a 60-s pulse. When the pipette solution contained IP(3) pathway blockers [heparin, 2-aminoethoxydiphenylborate, xestospongin C, or 1-[6-[((17β)-3-methoxyestra-1,3,5[10]-trien-17-yl)amino]hexyl]-1H-pyrrole-2,5-dione (U73122)], the peak [Ca(2+)](i) was unchanged, but the sustained [Ca(2+)](i) was gradually reduced by ∼250 nM within 30 s. In the presence of Cd(2+), a long depolarization period slightly increased the [Ca(2+)](i), which was lower than that in the presence of heparin alone. In coronary arterial myocytes, the sustained increase in the [Ca(2+)](i) during depolarization was partly caused by the Ca(2+) release mediated by the enhanced formation of IP(3). The initial [Ca(2+)](i) elevation triggered by the Ca(2+) influx though voltage-dependent Ca(2+) channels may be predominantly responsible for the activation of phospholipase C for IP(3) formation.
- Published
- 2012
33. Up-regulation of Kir2.1 by ER stress facilitates cell death of brain capillary endothelial cells
- Author
-
Hisao Yamamura, Hiroaki Kito, Yuji Imaizumi, Susumu Ohya, Daiju Yamazaki, and Kiyofumi Asai
- Subjects
Membrane potential ,Programmed cell death ,Inward-rectifier potassium ion channel ,Cell growth ,Tunicamycin ,Endoplasmic reticulum ,Biophysics ,Brain ,Cell Biology ,Membrane hyperpolarization ,Biology ,Endoplasmic Reticulum ,Biochemistry ,Capillaries ,Cell Line ,Up-Regulation ,Cell biology ,Stress, Physiological ,Apoptosis ,Unfolded protein response ,Animals ,Cattle ,Endothelium, Vascular ,Potassium Channels, Inwardly Rectifying ,Molecular Biology - Abstract
Brain capillary endothelial cells (BCECs) form blood brain barrier (BBB) to maintain brain homeostasis. Cell turnover of BCECs by the balance of cell proliferation and cell death is critical for maintaining the integrity of BBB. Here we found that stimuli with tunicamycin, endoplasmic reticulum (ER) stress inducer, up-regulated inward rectifier K(+) channel (K(ir)2.1) and facilitated cell death in t-BBEC117, a cell line derived from bovine BCECs. The activation of K(ir) channels contributed to the establishment of deeply negative resting membrane potential in t-BBEC117. The deep resting membrane potential increased the resting intracellular Ca(2+) concentration due to Ca(2+) influx through non-selective cation channels and thereby partly but significantly regulated cell death in t-BBEC117. The present results suggest that the up-regulation of K(ir)2.1 is, at least in part, responsible for cell death/cell turnover of BCECs induced by a variety of cellular stresses, particularly ER stress, under pathological conditions.
- Published
- 2011
34. Accelerated Ca2+entry by membrane hyperpolarization due to Ca2+-activated K+channel activation in response to histamine in chondrocytes
- Author
-
Kenji Funabashi, Susumu Ohya, Noriyuki Hatano, Yuji Imaizumi, Katsuhiko Muraki, Hisao Yamamura, and Wayne R. Giles
- Subjects
medicine.medical_specialty ,Physiology ,Histamine H1 receptor ,Ranitidine ,Chondrocyte ,Cell Line ,Membrane Potentials ,Histamine Agonists ,Potassium Channels, Calcium-Activated ,chemistry.chemical_compound ,Chondrocytes ,Internal medicine ,Potassium Channel Blockers ,medicine ,Extracellular ,Animals ,Humans ,Channel blocker ,Chemistry ,Cell Biology ,Membrane hyperpolarization ,Hyperpolarization (biology) ,Potassium channel ,Diphenhydramine ,medicine.anatomical_structure ,Endocrinology ,Histamine H2 Antagonists ,Histamine H1 Antagonists ,Biophysics ,Calcium ,Histamine - Abstract
In articular cartilage inflammation, histamine release from mast cells is a key event. It can enhance cytokine production and matrix synthesis and also promote cell proliferation by stimulating chondrocytes. In this study, the functional impact of Ca2+-activated K+(KCa) channels in the regulation of intracellular Ca2+concentration ([Ca2+]i) in chondrocytes in response to histamine was examined using OUMS-27 cells, as a model of chondrocytes derived from human chondrosarcoma. Application of histamine induced a significant [Ca2+]irise and also membrane hyperpolarization, and both effects were mediated by the stimulation of H1receptors. The histamine-induced membrane hyperpolarization was attenuated to ∼50% by large-conductance KCa(BK) channel blockers, and further reduced by intermediate (IK) and small conductance KCa(SK) channel blockers. The tonic component of histamine-induced [Ca2+]irise strongly depended on the presence of extracellular Ca2+([Ca2+]o) and was markedly reduced by La3+or Gd3+but not by nifedipine. It was significantly attenuated by BK channel blockers, and further blocked by the cocktail of BK, IK, and SK channel blockers. The KCablocker cocktail also significantly reduced the store-operated Ca2+entry (SOCE), which was induced by Ca2+addition after store-depletion by thapsigargin in [Ca2+]ofree solution. Our results demonstrate that the histamine-induced membrane hyperpolarization in chondrocytes due to KCachannel activation contributes to sustained Ca2+entry mainly through SOCE channels in OUMS-27 cells. Thus, KCachannels appear to play an important role in the positive feedback mechanism of [Ca2+]iregulation in chondrocytes in the presence of articular cartilage inflammation.
- Published
- 2010
35. Contribution of Chloride Channel Conductance to the Regulation of Resting Membrane Potential in Chondrocytes
- Author
-
Kenji Funabashi, Yuji Imaizumi, Susumu Ohya, Hisao Yamamura, and Masato Fujii
- Subjects
medicine.medical_specialty ,Cell Line ,Membrane Potentials ,Chondrocytes ,Chloride Channels ,Internal medicine ,medicine ,Humans ,Pharmacology ,Membrane potential ,Chemistry ,lcsh:RM1-950 ,Niflumic acid ,Niflumic Acid ,Conductance ,Membrane hyperpolarization ,Hyperpolarization (biology) ,Resting potential ,lcsh:Therapeutics. Pharmacology ,Endocrinology ,Chloride channel ,Biophysics ,Molecular Medicine ,Calcium ,Intracellular ,Histamine ,medicine.drug - Abstract
The contribution of Cl− conductance relative to that of K+ in the regulation of membrane potential was examined using OUMS-27 cells, a model cell-line of human chondrocytes. Application of 100 μM niflumic acid (NFA) and other anion-channel blockers induced significant membrane hyperpolarization. The NFA-sensitive membrane current under voltage-clamp was predominantly Cl− current. Application of NFA induced small but significant increase in intracellular Ca2+ concentration ([Ca2+]i) and markedly enhanced the late component of [Ca2+]i rise induced by 1 μM histamine. In conclusion, Cl− conductance substantially contributes to the regulation of resting membrane potential and [Ca2+]i in OUMS-27 cells. Keywords:: chondrocyte, chloride channel, niflumic acid
- Published
- 2010
36. Gender difference in BK channel expression in amygdala complex of rat brain
- Author
-
Susumu Ohya, Hisao Yamamura, Akitoshi Ohno, and Yuji Imaizumi
- Subjects
Male ,medicine.medical_specialty ,BK channel ,Population ,Biophysics ,Stimulation ,Biochemistry ,Amygdala ,chemistry.chemical_compound ,Sex Factors ,Internal medicine ,medicine ,Animals ,Large-Conductance Calcium-Activated Potassium Channels ,education ,Receptor ,Molecular Biology ,education.field_of_study ,biology ,Chemistry ,Pyramidal Cells ,Cell Biology ,Mycotoxins ,Rats ,Up-Regulation ,Endocrinology ,medicine.anatomical_structure ,Castration ,nervous system ,Receptors, Androgen ,Penitrem A ,Androgens ,biology.protein ,Female ,Neuron - Abstract
The expression of large-conductance Ca(2+)-activated K(+) (BK) channel protein in amygdala complex was higher in adult (8-10 weeks old) male rats than in female. Castration at 4-6 weeks old significantly reduced BK channel expression in amygdala to the level similar to that in female. Immunocytochemical analyses of pyramidal-like neurons isolated from amygdala revealed that somas with relatively large size were highly immunoreactive to both anti-androgen receptor (AR) and anti-BK channel antibodies, while those with smaller size were not. The double-immunopositive neurons were dominant (60%) among pyramidal-like neurons isolated from amygdala of male rats but rare among those from female. The membrane current sensitive to penitrem A, a BK channel blocker, was the major K(+) current component in large neurons and showed higher current-density than that in smaller ones. These results suggest the gender-dependent cell population expressing BK channels in amygdala complex and its up-regulation by AR stimulation.
- Published
- 2009
37. Expression analysis of the epithelial Na+ channel δ subunit in human melanoma G-361 cells
- Author
-
Shoichi Shimada, Shinya Ugawa, Takashi Ueda, and Hisao Yamamura
- Subjects
Epithelial sodium channel ,Messenger RNA ,Gene Expression Profiling ,Melanoma ,Protein subunit ,Immunocytochemistry ,Biophysics ,Cell Biology ,In situ hybridization ,Biology ,medicine.disease ,Biochemistry ,Molecular biology ,Neoplasm Proteins ,Cell Line, Tumor ,Immunology ,medicine ,Humans ,Skin cancer ,Epithelial Sodium Channels ,Molecular Biology ,Ion channel - Abstract
Malignant melanoma is the most deadly form of skin cancer and its incidence is steadily increasing worldwide. The plasma membrane in melanoma cells possesses a variety of ion channels, so its profile is thought to lead to a novel target for medical treatment for malignant melanoma. Here we showed that human melanoma G-361 cells expressed the epithelial Na(+) channel delta subunit (ENaC delta), which is largely unknown in physiological and pathological functions in non-neuronal tissues. Expression analyses at the level of mRNA clearly revealed that ENaC delta transcript was strongly expressed in human melanoma cells using reverse transcription-polymerase chain reaction and cell-based in situ hybridization techniques. Other ENaC subunits (alpha, beta, and gamma) were also distributed in human melanoma cells. In addition, human melanoma cells possessed an abundant expression of ENaC delta protein by immunocytochemistry. These results provide an attractive target for drug development of malignant melanoma.
- Published
- 2008
38. Voltage-Dependent Ca2+-Channel Block by Openers of Intermediate and Small Conductance Ca2+-Activated K+ Channels in Urinary Bladder Smooth Muscle Cells
- Author
-
Hisao Yamamura, Kozo Morimura, Yuji Imaizumi, and Susumu Ohya
- Subjects
Indoles ,Calcium Channels, L-Type ,Small-Conductance Calcium-Activated Potassium Channels ,Myocytes, Smooth Muscle ,Urinary Bladder ,In Vitro Techniques ,SK channel ,Mice ,Smooth muscle ,Oximes ,medicine ,Animals ,K channels ,Pharmacology ,Urinary bladder ,Dose-Response Relationship, Drug ,Chemistry ,lcsh:RM1-950 ,Conductance ,Intermediate-Conductance Calcium-Activated Potassium Channels ,Mouse Urinary Bladder ,Mice, Inbred C57BL ,medicine.anatomical_structure ,lcsh:Therapeutics. Pharmacology ,Biophysics ,Molecular Medicine ,Ca2 channels ,Benzimidazoles ,Calcium ,Whole cell ,Ion Channel Gating - Abstract
We examined effects of small and intermediate conductance Ca2+-activated K+ (SK and IK) channel openers, DCEBIO (5,6-dichloro-1-ethyl-1,3-dihydro-2H-benzimidazol-2-one) and NS309 (3-oxime-6,7-dichloro-1H-indole-2,3-dione), on L-type Ca2+ channel current (ICa) that was measured in smooth muscle cells isolated from mouse urinary bladder under whole cell voltage-clamp. The ICa was concentration-dependently inhibited by DCEBIO and NS309; half inhibition was obtained at 71.6 and 10.6 µM, respectively. The specificity of NS309 to the IK channel over the Ca2+ channel appears to be high and higher than that of DCEBIO. DCEBIO and even NS309 may, however, substantially block Ca2+ channels when used as SK channel openers. Keywords:: voltage-dependent Ca2+ channel, DCEBIO, NS309
- Published
- 2006
39. Capsazepine Is a Novel Activator of the δ Subunit of the Human Epithelial Na+ Channel
- Author
-
Hisao Yamamura, Shoichi Shimada, Takashi Ueda, Shinya Ugawa, and Masataka Nagao
- Subjects
Epithelial sodium channel ,Dopamine ,Receptors, Drug ,Xenopus ,Neurotoxins ,Resiniferatoxin ,Pharmacology ,Biochemistry ,Sodium Channels ,Vanilloids ,Amiloride ,Transient receptor potential channel ,chemistry.chemical_compound ,Benzamil ,medicine ,Animals ,Humans ,Diuretics ,Epithelial Sodium Channels ,Molecular Biology ,Dose-Response Relationship, Drug ,Anti-Inflammatory Agents, Non-Steroidal ,Cell Biology ,Hydrogen-Ion Concentration ,Protein Structure, Tertiary ,Electrophysiology ,chemistry ,Competitive antagonist ,Oocytes ,Biophysics ,Capsaicin ,Diterpenes ,Protons ,Capsazepine ,medicine.drug - Abstract
The amiloride-sensitive epithelial Na+ channel (ENaC) regulates Na+ homeostasis into cells and across epithelia. So far, four homologous subunits of mammalian ENaC have been isolated and are denoted as alpha, beta, gamma, and delta. The chemical agents acting on ENaC are, however, largely unknown, except for amiloride and benzamil as ENaC inhibitors. In particular, there are no agonists currently known that are selective for ENaCdelta, which is mainly expressed in the brain. Here we demonstrate that capsazepine, a competitive antagonist for transient receptor potential vanilloid subfamily 1, potentiates the activity of human ENaCdeltabetagamma (hENaCdeltabetagamma) heteromultimer expressed in Xenopus oocytes. The inward currents at a holding potential of -60 mV in hENaCdeltabetagamma-expressing oocytes were markedly enhanced by the application of capsazepine (or =1 microM), and the capsazepine-induced current was mostly abolished by the addition of 100 microM amiloride. The stimulatory effects of capsazepine on the inward current were concentration-dependent with an EC50 value of 8 microM. Neither the application of other vanilloid compounds (capsaicin, resiniferatoxin, and olvanil) nor a structurally related compound (dopamine) modulated the inward current. Although hENaCdelta homomer was also significantly activated by capsazepine, unexpectedly, capsazepine had no effect on hENaCalpha and caused a slight decrease on the hENaCalphabetagamma current. In conclusion, capsazepine acts on ENaCdelta and acts together with protons. Other vanilloids tested do not have any effect. These findings identify capsazepine as the first known chemical activator of ENaCdelta.
- Published
- 2004
40. Local Ca2+transients and distribution of BK channels and ryanodine receptors in smooth muscle cells of guinea-pig vas deferens and urinary bladder
- Author
-
Yuji Imaizumi, Norihiro Nagano, Yoshiaki Ohi, Minoru Watanabe, Katsuhiko Muraki, Hisao Yamamura, and Susumu Ohya
- Subjects
Male ,medicine.medical_specialty ,BK channel ,Patch-Clamp Techniques ,Potassium Channels ,Contraction (grammar) ,Physiology ,Voltage clamp ,Guinea Pigs ,Urinary Bladder ,Membrane Potentials ,Potassium Channels, Calcium-Activated ,Vas Deferens ,Internal medicine ,medicine ,Animals ,Myocyte ,Calcium Signaling ,Large-Conductance Calcium-Activated Potassium Channels ,Large-Conductance Calcium-Activated Potassium Channel alpha Subunits ,Fluorescent Dyes ,Aniline Compounds ,Microscopy, Confocal ,biology ,Chemistry ,Ryanodine receptor ,Endoplasmic reticulum ,Cell Membrane ,Vas deferens ,Muscle, Smooth ,Ryanodine Receptor Calcium Release Channel ,Depolarization ,Original Articles ,Immunohistochemistry ,Kinetics ,Sarcoplasmic Reticulum ,Endocrinology ,medicine.anatomical_structure ,Xanthenes ,cardiovascular system ,Biophysics ,biology.protein ,Calcium ,tissues - Abstract
1. The relationship between Ca(2+) sparks spontaneously occurring at rest and local Ca(2+) transients elicited by depolarization was analysed using two-dimensional confocal Ca(2+) images of single smooth muscle cells isolated from guinea-pig vas deferens and urinary bladder. The current activation by these Ca(2+) events was also recorded simultaneously under whole-cell voltage clamp. 2. Spontaneous transient outward currents (STOCs) and Ca(2+) sparks were simultaneously detected at -40 mV in approximately 50 % of myocytes of either type. Ca(2+) sparks and corresponding STOCs occurred repetitively in several discrete sites in the subplasmalemmal area. Large conductance Ca(2+)-dependent K(+) (BK) channel density in the plasmalemma near the Ca(2+) spark sites generating STOCs was calculated to be 21 channels microm(-2). 3. When myocytes were depolarized from -60 to 0 mV, several local Ca(2+) transients were elicited within 20 ms in exactly the same peripheral sites where sparks occurred at rest. The local Ca(2+) transients often lasted over 300 ms and spread into other areas. The appearance of local Ca(2+) transients occurred synchronously with the activation of Ca(2+)-dependent K(+) current (I(K,Ca)). 4. Immunofluorescence staining of the BK channel alpha-subunit (BKalpha) revealed a spot-like pattern on the plasmalemma, in contrast to the uniform staining of voltage-dependent Ca(2+) channel alpha1C subunits along the plasmalemma. Ryanodine receptor (RyR) immunostaining also suggested punctate localization predominantly in the periphery. Double staining of BKalpha and RyRs revealed spot-like co-localization on/beneath the plasmalemma. 5. Using pipettes of relatively low resistance, inside-out patches that included both clustered BK channels at a density of over 20 channels microm(-2) and functional Ca(2+) storage sites were obtained at a low probability of approximately 5%. The averaged BK channel density was 3-4 channels microm(-2) in both types of myocyte. 6. These results support the idea that a limited number of discrete sarcoplasmic reticulum (SR) fragments in the subplasmalemmal area play key roles in the control of BK channel activity in two ways: (i) by generating Ca(2+) sparks at rest to activate STOCs and (ii) by generating Ca(2+) transients presumably triggered by sparks during an action potential to activate a large I(K,Ca) and also induce a contraction. BK channels and RyRs may co-localize densely at the junctional areas of plasmalemma and SR fragments, where Ca(2+) sparks occur to elicit STOCs.
- Published
- 2001
41. BK channel activation by NS-1619 is partially mediated by intracellular Ca2+ release in smooth muscle cells of porcine coronary artery
- Author
-
Yoshiaki Ohi, Yuji Imaizumi, Minoru Watanabe, Hisao Yamamura, and Katsuhiko Muraki
- Subjects
Pharmacology ,Membrane potential ,BK channel ,biology ,Chemistry ,Ryanodine receptor ,Anatomy ,Membrane hyperpolarization ,Iberiotoxin ,Hyperpolarization (biology) ,Potassium channel ,biology.protein ,Biophysics ,Intracellular - Abstract
1. Effects of NS-1619, an opener of large conductance Ca2+-activated K+ (BK) channel, on intracellular Ca2+ concentration ([Ca2+]i) and membrane potential were examined in single myocytes freshly isolated from porcine coronary artery. 2. Under current clamp mode, the application of 1-30 microM NS-1619 hyperpolarized the membrane in concentration-dependent manner. The NS-1619-induced hyperpolarization was abolished by the presence of 100 nM iberiotoxin. 3. Application of 1-10 microM NS-1619 hyperpolarized the membrane by approximately 6 mV or less but did not change significantly the [Ca2+]i. When membrane hyperpolarization of 12 mV or so was caused by 30 microM NS-1619, [Ca2+]i was unexpectedly increased by approximately 200 nM. This increase in [Ca2+]i and the concomitant outward current activation were also observed under voltage-clamp at holding potential of -40 mV. 4. The increase in [Ca2+]i by 30 microM NS-1619 occurred mainly in peripheral regions than in the centre of the myocytes. The removal of extracellular Ca2+ affected neither the membrane hyperpolarization nor the increase in [Ca2+]i. 5. In the presence of 10 mM caffeine and 10 microM ryanodine, the increase in [Ca2+]i by 30 microM NS-1619 was not observed and the membrane hyperpolarization was reduced to approximately 67% of the control. 6. These results indicate that the opening of BK channels by NS-1619 at 30 microM, which is the most frequently used concentration of this agent, is partly due to Ca2+ release from caffeine/ryanodine-sensitive intracellular storage sites but is mainly due to the direct activation of the channels.
- Published
- 2001
42. Caveolin-1 Facilitates the Direct Coupling between Large Conductance Ca2+-activated K+ (BKCa) and Cav1.2 Ca2+ Channels and Their Clustering to Regulate Membrane Excitability in Vascular Myocytes*
- Author
-
Yuji Imaizumi, Hisao Yamamura, Yoshiaki Suzuki, and Susumu Ohya
- Subjects
medicine.medical_specialty ,Vascular smooth muscle ,Calcium Channels, L-Type ,Caveolin 1 ,Myocytes, Smooth Muscle ,Action Potentials ,Biochemistry ,Cav1.2 ,Muscle, Smooth, Vascular ,Mice ,Internal medicine ,Caveolae ,Membrane Biology ,Caveolin ,medicine ,Myocyte ,Animals ,Humans ,Patch clamp ,Large-Conductance Calcium-Activated Potassium Channels ,Molecular Biology ,Calcium signaling ,Mice, Knockout ,biology ,Chemistry ,Ryanodine receptor ,Ryanodine Receptor Calcium Release Channel ,Cell Biology ,Rats ,Mice, Inbred C57BL ,Protein Subunits ,Endocrinology ,HEK293 Cells ,biology.protein ,Biophysics ,Calcium ,Muscle Contraction ,Protein Binding - Abstract
L-type voltage-dependent Ca(2+) channels (LVDCC) and large conductance Ca(2+)-activated K(+) channels (BKCa) are the major factors defining membrane excitability in vascular smooth muscle cells (VSMCs). The Ca(2+) release from sarcoplasmic reticulum through ryanodine receptor significantly contributes to BKCa activation in VSMCs. In this study direct coupling between LVDCC (Cav1.2) and BKCa and the role of caveoline-1 on their interaction in mouse mesenteric artery SMCs were examined. The direct activation of BKCa by Ca(2+) influx through coupling LVDCC was demonstrated by patch clamp recordings in freshly isolated VSMCs. Using total internal reflection fluorescence microscopy, it was found that a large part of yellow fluorescent protein-tagged BKCa co-localized with the cyan fluorescent protein-tagged Cav1.2 expressed in the plasma membrane of primary cultured mouse VSMCs and that the two molecules often exhibited FRET. It is notable that each BKα subunit of a tetramer in BKCa can directly interact with Cav1.2 and promotes Cav1.2 cluster in the molecular complex. Furthermore, caveolin-1 deficiency in knock-out (KO) mice significantly reduced not only the direct coupling between BKCa and Cav1.2 but also the functional coupling between BKCa and ryanodine receptor in VSMCs. The measurement of single cell shortening by 40 mm K(+) revealed enhanced contractility in VSMCs from KO mice than wild type. Taken together, caveolin-1 facilitates the accumulation/clustering of BKCa-LVDCC complex in caveolae, which effectively regulates spatiotemporal Ca(2+) dynamics including the negative feedback, to control the arterial excitability and contractility.
- Published
- 2013
43. [TMEM16 as calcium-activated chloride channels]
- Author
-
Hisao Yamamura
- Subjects
Pharmacology ,Chemistry ,Anoctamins ,Membrane Proteins ,Neoplasm Proteins ,Mice ,Membrane protein ,Chloride Channels ,Calcium-Activated Chloride Channels ,Chloride channel ,Biophysics ,Animals ,Humans ,Anoctamin-1 - Published
- 2013
44. Functional Characterization Of Voltage-Dependent Ca2+ Channels In Mouse Pulmonary Arterial Smooth Muscle Cells
- Author
-
Hisao Yamamura, Amy Zeifman, Jun Wan, Hae Young Yoo, Eun A. Ko, and Jason X.-J. Yuan
- Subjects
Chemistry ,Biophysics ,Ca2 channels ,Arterial smooth muscle cells ,Voltage - Published
- 2012
45. Molecular mechanisms for Kv1.3 potassium channel current inhibition by CD3/CD28 stimulation in Jurkat T cells
- Author
-
Haruna Itoda, Hisao Yamamura, Yuichiro Matsushita, Takuya Kimura, Susumu Ohya, Yuji Imaizumi, and Yoshiaki Suzuki
- Subjects
Kv1.3 Potassium Channel ,CD3 Complex ,Cell growth ,T-Lymphocytes ,Biophysics ,CD28 ,hemic and immune systems ,Stimulation ,Cell Biology ,Protein tyrosine phosphatase ,T lymphocyte ,Biology ,Biochemistry ,Jurkat cells ,Potassium channel ,Antibodies ,Cell biology ,Jurkat Cells ,CD28 Antigens ,Cell culture ,Humans ,Leukocyte Common Antigens ,Protein Tyrosine Phosphatases ,Molecular Biology - Abstract
In T lymphocyte, activation of Kv1.3 channel, the major voltage-dependent K(+) channel, is an essential step for cell proliferation in immune responses. Here, effects of anti-CD3 and anti-CD28 antibodies on Kv1.3 current were examined in three types of human T lymphocyte derived cell lines, Jurkat E6-1, p56lck-kinase deficient mutant JCaM.1, and CD45-phosphatase deficient mutant J45.01. Kv1.3 current was partly reduced by CD3 stimulation and more strongly by addition of anti-CD28 antibody in E6-1. In JCaM.1, Kv1.3 current responses to anti-CD28/CD3 antibodies were similar to those in E6-1. In J45.01, CD3 stimulation partly inhibited Kv1.3 current, but the additive reduction by CD28 stimulation was not significant. The inhibition of tyrosine phosphatase in E6-1 abolished the additional inhibition by anti-CD28 antibody in a similar manner as in J45.01. In conclusion, the stimulation of CD28 in addition to CD3 strongly inhibits Kv1.3 current and this additive inhibition is mediated by CD45 activation.
- Published
- 2008
46. Epithelial Na+ channel delta subunit mediates acid-induced ATP release in the human skin
- Author
-
Masataka Nagao, Hisao Yamamura, Takashi Ueda, Shinya Ugawa, and Shoichi Shimada
- Subjects
Epithelial sodium channel ,Keratinocytes ,Protein subunit ,Biophysics ,Human skin ,Biology ,Biochemistry ,Amiloride ,chemistry.chemical_compound ,Adenosine Triphosphate ,Benzamil ,medicine ,Epithelial Sodium Channel Blockers ,Humans ,Viability assay ,RNA, Messenger ,Epithelial Sodium Channels ,Molecular Biology ,Skin ,Cell Biology ,Hydrogen-Ion Concentration ,Cell biology ,medicine.anatomical_structure ,chemistry ,Epidermal Cells ,Epidermis ,Keratinocyte ,Acids ,Homeostasis ,medicine.drug - Abstract
The amiloride-sensitive epithelial Na(+) channel (ENaC) regulates Na(+) homeostasis in cells and across epithelia. Although we described that ENaCdelta is a candidate molecule for a pH sensor in the human brain, the physiological and pathological roles of ENaCdelta in non-neuronal tissues are still unknown. Here we show a novel physiological function of ENaCdelta in peripheral tissues in humans. Expression analyses at the level of mRNA clearly revealed that ENaCdelta was abundantly expressed in human epidermis and keratinocytes. In addition, ENaCdelta protein was detected in there. In cultured keratinocytes, acidic stress (pH 5.0) evoked ATP release, which was significantly reduced in the presence of 100 microM amiloride or 10 microM benzamil. In conclusion, ENaCdelta may be involved in the mechanism underlying pH sensing followed by the regulation of cell viability in the human skin.
- Published
- 2008
47. The CaV3.1 T-type Ca2+channel contributes to voltage-dependent calcium currents in rat outer hair cells
- Author
-
Shoichi Shimada, Shingo Murakami, Shinya Ugawa, Hisao Yamamura, and Akira Inagaki
- Subjects
Male ,Pathology ,medicine.medical_specialty ,Patch-Clamp Techniques ,chemistry.chemical_element ,In situ hybridization ,Biology ,Calcium ,Mechanotransduction, Cellular ,Membrane Potentials ,Calcium Channels, T-Type ,Organ Culture Techniques ,Western blot ,Hearing ,medicine ,Animals ,Calcium Signaling ,RNA, Messenger ,Rats, Wistar ,Molecular Biology ,Organ of Corti ,Cochlea ,Mibefradil ,medicine.diagnostic_test ,General Neuroscience ,Cell Membrane ,Depolarization ,Immunohistochemistry ,Rats ,Electrophysiology ,Hair Cells, Auditory, Outer ,chemistry ,Biophysics ,sense organs ,Neurology (clinical) ,Intracellular ,Developmental Biology ,medicine.drug ,Subcellular Fractions - Abstract
Calcium currents through voltage-dependent Ca2+channels (VDCCs) in mammalian outer hair cells (OHCs) are generally considered to possess the pharmacological properties of L-type (dihydropyridine-sensitive) currents. However, the OHCs' low resting potentials and their slight depolarization upon sound stimuli suggest that the low voltage-activated channels may contribute to Ca2+regulation. We present morphological and electrophysiological evidence for the presence of the CaV3.1 T-type Ca2+channels, one of the low voltage-activated Ca2+channels, in mature rat OHCs. PCR experiments revealed the expression of CaV3.1, but not CaV3.2 or CaV3.3, in the mature rat cochlea. In situ hybridization and immunohistochemistry revealed expression of CaV3.1 in both inner and outer hair cells at the mRNA level, but only in the OHCs at the protein level. Western blot analysis of anti-CaV3.1 antibody showed a 242 kDa band in mature rat cochlear lysates. Patch-clamp recordings of OHCs isolated from rat cochleae after the onset of hearing revealed that whole-cell voltage-dependent Ca2+ currents were significantly increased in depolarizing steps from a holding potential of − 100 mV when compared with those from − 70 mV. Only the currents from − 100 mV manifested a distinct transient inward Ca2+ current, and this transient component was effectively blocked by 1 μM of the T-type-specific antagonist, mibefradil. Our data suggest an involvement of CaV3.1 in intracellular Ca2+regulation in mature OHCs.
- Published
- 2007
48. Methyl-beta-cyclodextrin prevents Ca2+-induced Ca2+ release in smooth muscle cells of mouse urinary bladder
- Author
-
Susumu Ohya, Shingo Hotta, Yuji Imaizumi, and Hisao Yamamura
- Subjects
Male ,medicine.medical_specialty ,Contraction (grammar) ,Myocytes, Smooth Muscle ,Urinary Bladder ,chemistry.chemical_element ,Stimulation ,Beta-Cyclodextrins ,Calcium ,Mice ,Internal medicine ,medicine ,Animals ,Pharmacology ,Voltage-dependent calcium channel ,Chemistry ,Ryanodine receptor ,lcsh:RM1-950 ,beta-Cyclodextrins ,Depolarization ,Ryanodine Receptor Calcium Release Channel ,Mice, Inbred C57BL ,lcsh:Therapeutics. Pharmacology ,Endocrinology ,cardiovascular system ,Biophysics ,Molecular Medicine ,Calcium Channels ,medicine.symptom ,Muscle contraction ,Muscle Contraction - Abstract
We examined the effects of methyl-β-cyclodextrin (MβCD) on Ca2+-induced Ca2+ release (CICR) in smooth muscle cells (SMCs) of mouse urinary bladder (UB). Short depolarization of UBSMCs under voltage-clamp elicited several local Ca2+ transients (Ca2+ hot spots) via CICR within 20 ms in discrete sub-sarcolemmal areas. Then, the Ca2+ wave spread to whole areas. The pretreatment with 10 mM MβCD significantly attenuated Ca2+ hot spots in UBSMCs and reduced contraction by single direct electrical pulse stimulation in UBSM strips. MβCD may prevent CICR by attenuating the coupling between voltage-dependent Ca2+ channels and ryanodine receptors in Ca2+ hot spot areas. Keywords:: methyl-β-cyclodextrin, Ca2+-induced Ca2+ release, smooth muscle
- Published
- 2007
49. A novel spliced variant of the epithelial Na+ channel delta-subunit in the human brain
- Author
-
Takashi Ueda, Shoichi Shimada, Hisao Yamamura, Shinya Ugawa, and Masataka Nagao
- Subjects
Epithelial sodium channel ,Protein subunit ,Molecular Sequence Data ,Biophysics ,Biology ,Biochemistry ,Sodium Channels ,Amiloride ,Xenopus laevis ,medicine ,Animals ,Humans ,Amino Acid Sequence ,Epithelial Sodium Channels ,Molecular Biology ,Genetics ,Sequence Homology, Amino Acid ,Brain ,Cell Biology ,Human brain ,Cell biology ,Protein Structure, Tertiary ,Alternative Splicing ,medicine.anatomical_structure ,Ph sensing ,Oocytes ,Homeostasis ,medicine.drug ,Protein Binding - Abstract
The amiloride-sensitive epithelial Na+ channel regulates Na+ homeostasis in cells. Recently, we described that the delta-subunit is a candidate molecule for a pH sensor in the human brain. Here, an N-terminal spliced variant of the delta-subunit is cloned from human brain, and designated as the delta2-subunit, which is expressed with the original delta-subunit (delta1-subunit) at the same level in the human brain. Functional analyses revealed that the physiological and pharmacological properties (interaction with accessory betagamma-subunits, activation by acidic pH, amiloride sensitivity) of the delta2-subunit were similar to those of the delta1-subunit. In conclusion, the activities of both subunits may be involved in the mechanism underlying pH sensing in the human brain.
- Published
- 2006
50. Two-step Ca2+ intracellular release underlies excitation-contraction coupling in mouse urinary bladder myocytes
- Author
-
Susumu Ohya, Hisao Yamamura, Yuji Imaizumi, Kozo Morimura, Katsuhiko Muraki, and Yoshiaki Ohi
- Subjects
Male ,Indoles ,Macrocyclic Compounds ,Patch-Clamp Techniques ,Physiology ,Two step ,Myocytes, Smooth Muscle ,Urinary Bladder ,Action Potentials ,Mice ,Repolarization ,Myocyte ,Animals ,Calcium Signaling ,Enzyme Inhibitors ,Oxazoles ,Cells, Cultured ,Microscopy, Confocal ,Chemistry ,Ryanodine ,Excitation–contraction coupling ,Cell Biology ,Anatomy ,Mouse Urinary Bladder ,Coupling (electronics) ,Mice, Inbred C57BL ,Biophysics ,Calcium ,Female ,Calcium Channels ,Intracellular ,Muscle Contraction - Abstract
The relative contributions of Ca2+-induced Ca2+ release (CICR) versus Ca2+ influx through voltage-dependent Ca2+ channels (VDCCs) to excitation-contraction coupling has not been defined in most smooth muscle cells (SMCs). The present study was undertaken to address this issue in mouse urinary bladder (UB) smooth muscle cells (UBSMCs). Confocal Ca2+ images were obtained under voltage- or current-clamp conditions. When UBSMCs were activated by a 30-ms depolarization to 0 mV, intracellular Ca2+ concentration ([Ca2+]i) increased in several small, discrete areas just beneath the cell membrane. These Ca2+ “hot spots” then spread slowly through the myoplasm as Ca2+ waves, which continued even after repolarization. Shorter depolarizations (5 ms) elicited only a few Ca2+ sparks, which declined quickly. The number of Ca2+ sparks, or hot spots, was closely related to the depolarization duration in the range of ∼5–20 ms. There was an apparent threshold depolarization duration of ∼10 ms within which to induce enough Ca2+ transients to spread globally and then induce a contraction. Application of 100 μM ryanodine to the pipette solution did not change the resting [Ca2+]i or the VDCC current, but it did abolish Ca2+ hot spots elicited by depolarization. Application of 3 μM xestospongin C reduced ACh-induced Ca2+ release but did not affect depolarization-induced Ca2+ events. The addition of 100 μM ryanodine to tissue segments markedly reduced the amplitude of contractions triggered by direct electrical stimulation. In conclusion, global [Ca2+]i rise triggered by a single action potential is not due mainly to Ca2+ influx through VDCCs but is attributable to the subsequent two-step CICR.
- Published
- 2005
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.