56 results on '"Hisao Yamamura"'
Search Results
2. Upregulated ClC3 Channels/Transporters Elicit Swelling-Activated Cl− Currents and Induce Excessive Cell Proliferation in Idiopathic Pulmonary Arterial Hypertension
- Author
-
Taiki Amano, Aya Yamamura, Moe Fujiwara, Seiji Hirai, Rubii Kondo, Yoshiaki Suzuki, and Hisao Yamamura
- Subjects
Pharmacology ,Pharmaceutical Science ,General Medicine - Published
- 2022
3. Local Ca2+ Signals within Caveolae Cause Nuclear Translocation of CaMK1α in Mouse Vascular Smooth Muscle Cells
- Author
-
Yoshiaki Suzuki, Tomo Kurata, Tsukasa Koide, Itsuki Okada, Nanami Nakajima, Yuji Imaizumi, and Hisao Yamamura
- Subjects
Pharmacology ,Pharmaceutical Science ,General Medicine - Published
- 2022
4. Ca2+ Signaling and Proliferation via Ca2+-Sensing Receptors in Human Hepatic Stellate LX-2 Cells
- Author
-
Rubii Kondo, Naoki Kawata, Yoshiaki Suzuki, and Hisao Yamamura
- Subjects
Pharmacology ,Pharmaceutical Science ,General Medicine - Published
- 2022
5. Involvement of TREK1 channels in the proliferation of human hepatic stellate LX-2 cells
- Author
-
Rubii Kondo, Akari Deguchi, Naoki Kawata, Yoshiaki Suzuki, and Hisao Yamamura
- Subjects
Liver Cirrhosis ,Pharmacology ,Proliferation ,Gene Expression ,Calcium signaling ,RM1-950 ,Collagen Type I ,Cell Line ,Membrane Potentials ,TREK1 ,Potassium Channels, Tandem Pore Domain ,Hepatic Stellate Cells ,Humans ,Molecular Medicine ,Calcium ,Two-pore domain potassium channel ,Therapeutics. Pharmacology ,Hepatic stellate cell ,Cell Proliferation - Abstract
Activation of hepatic stellate cells (HSCs) causes hepatic fibrosis and results in chronic liver diseases. Although activated HSC functions are facilitated by an increase in the cytosolic Ca2+ concentration ([Ca2+]cyt), the pathophysiological roles of ion channels are largely unknown. In the present study, functional analyses of the two-pore domain K+ (K2P) channels, which regulate the resting membrane potential and [Ca2+]cyt, were performed using the human HSC line, LX-2. Expression analyses revealed that TREK1 (also known as KCNK2 and K2P2.1) channels are expressed in LX-2 cells. Whole-cell K+ currents were activated by 10 μM arachidonic acid and the activation was abolished by 100 μM tetrapentylammonium, which are pharmacological characteristics of TREK1 channels. The siRNA knockdown of TREK1 channels caused membrane depolarization and reduced [Ca2+]cyt. In addition, TREK1 knockdown downregulated the gene expression of collage type I and platelet-derived growth factor. Furthermore, TREK1 knockdown inhibited the proliferation of LX-2 cells. In conclusion, the activity of TREK1 channels determines the resting membrane potential and [Ca2+]cyt, which play a role in extracellular matrix production and cell proliferation in HSCs. This study may help elucidate the molecular mechanism underlying hepatic fibrosis in HSCs and provide a potential therapeutic target for hepatic fibrosis.
- Published
- 2022
6. Comparative analysis of age in monocrotaline-induced pulmonary hypertensive rats
- Author
-
Aya Yamamura, Yoshiaki Suzuki, Moe Fujiwara, Naoki Ohara, Hisao Yamamura, Rubii Kondo, Chihiro Horii, Shiho Mori, Akari Hiraku, Akiko Kawade, Kikuo Tsukamoto, Saki Kobayashi, and Sayo Suzumura
- Subjects
Adult ,Male ,0301 basic medicine ,medicine.medical_specialty ,Hypertension, Pulmonary ,RM1-950 ,Pulmonary Artery ,Pulmonary hypertension ,Rats, Sprague-Dawley ,03 medical and health sciences ,0302 clinical medicine ,Age ,Downregulation and upregulation ,Internal medicine ,Animals ,Humans ,Medicine ,Receptor ,Pharmacology ,Monocrotaline ,Age differences ,business.industry ,Age Factors ,Middle Aged ,medicine.disease ,Disease Models, Animal ,030104 developmental biology ,Endocrinology ,Lung disease ,Molecular Medicine ,Female ,Calcium Channels ,Therapeutics. Pharmacology ,business ,Receptors, Calcium-Sensing ,030217 neurology & neurosurgery - Abstract
Pulmonary arterial hypertension (PAH) is a rare, progressive, and fatal cardiovascular/lung disease. The incidence rate is affected by age. Monocrotaline (MCT, 60 mg/kg)-treated rats are widely used as an experimental PAH model. Here, we found that young rats died at a mean of 23.4 days after MCT injection, whereas adult rats survived for over 42 days. However, young (7-week-old) and adult (20-week-old) MCT-treated rats developed PAH, and had upregulated Ca2+-sensing receptor and transient receptor potential canonical subfamily 6 channel expression in pulmonary arteries. The present study provides novel information for elucidating the mechanism underlying the age difference in PAH patients.
- Published
- 2021
7. Downregulation of Ca2+-Activated Cl− Channel TMEM16A Mediated by Angiotensin II in Cirrhotic Portal Hypertensive Mice
- Author
-
Rubii Kondo, Nami Furukawa, Akari Deguchi, Naoki Kawata, Yoshiaki Suzuki, Yuji Imaizumi, and Hisao Yamamura
- Subjects
Pharmacology ,Pharmacology (medical) - Abstract
Portal hypertension is defined as an increased pressure in the portal venous system and occurs as a major complication in chronic liver diseases. The pathological mechanism underlying the pathogenesis and development of portal hypertension has been extensively investigated. Vascular tone of portal vein smooth muscles (PVSMs) is regulated by the activities of several ion channels, including Ca2+-activated Cl− (ClCa) channels. TMEM16A is mainly responsible for ClCa channel conductance in vascular smooth muscle cells, including portal vein smooth muscle cells (PVSMCs). In the present study, the functional roles of TMEM16A channels were examined using two experimental portal hypertensive models, bile duct ligation (BDL) mice with cirrhotic portal hypertension and partial portal vein ligation (PPVL) mice with non-cirrhotic portal hypertension. Expression analyses revealed that the expression of TMEM16A was downregulated in BDL-PVSMs, but not in PPVL-PVSMs. Whole-cell ClCa currents were smaller in BDL-PVSMCs than in sham- and PPVL-PVSMCs. The amplitude of spontaneous contractions was smaller and the frequency was higher in BDL-PVSMs than in sham- and PPVL-PVSMs. Spontaneous contractions sensitive to a specific inhibitor of TMEM16A channels, T16Ainh-A01, were reduced in BDL-PVSMs. Furthermore, in normal PVSMs, the downregulation of TMEM16A expression was mimicked by the exposure to angiotensin II, but not to bilirubin. This study suggests that the activity of ClCa channels is attenuated by the downregulation of TMEM16A expression in PVSMCs associated with cirrhotic portal hypertension, which is partly mediated by increased angiotensin II in cirrhosis.
- Published
- 2022
8. TMEM16A Ca2+-Activated Cl- Channel Regulates the Proliferation and Migration of Brain Capillary Endothelial Cells
- Author
-
Yoshiaki Suzuki, Hisao Yamamura, Kiyofumi Asai, Yuji Imaizumi, Takahisa Suzuki, and Miki Yasumoto
- Subjects
0301 basic medicine ,Pharmacology ,Membrane potential ,Chemistry ,Cell growth ,HEK 293 cells ,Niflumic acid ,Hyperpolarization (biology) ,Cell biology ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,medicine ,Molecular Medicine ,Channel blocker ,Viability assay ,030217 neurology & neurosurgery ,Ion channel ,medicine.drug - Abstract
The blood-brain barrier (BBB) is essential for the maintenance of homeostasis in the brain. Brain capillary endothelial cells (BCECs) comprise the BBB, and thus a delicate balance between their proliferation and death is required. Although the activity of ion channels in BCECs is involved in BBB functions, the underlying molecular mechanisms remain unclear. In the present study, the molecular components of Ca2+-activated Cl- (ClCa) channels and their physiological roles were examined using mouse BCECs (mBCECs) and a cell line derived from bovine BCECs, t-BBEC117. Expression analyses revealed that TMEM16A was strongly expressed in mBCECs and t-BBEC117 cells. In t-BBEC117 cells, whole-cell Cl- currents were sensitive to the ClCa channel blockers, 100 μM niflumic acid and 10 μM T16Ainh-A01, and were also reduced markedly by small-interfering RNA (siRNA) knockdown of TMEM16A. Importantly, block of ClCa currents with ClCa channel blockers or TMEM16A siRNA induced membrane hyperpolarization. Moreover, treatment with TMEM16A siRNA caused an increase in resting cytosolic Ca2+ concentration ([Ca2+]cyt). T16Ainh-A01 reduced cell viability in a concentration-dependent manner. Either ClCa channel blockers or TMEM16A siRNA also curtailed cell proliferation and migration. Furthermore, ClCa channel blockers attenuated the trans-endothelial permeability. In combination, these results strongly suggest that TMEM16A contributes to ClCa channel conductance and can regulate both the resting membrane potential and [Ca2+]cyt in BCECs. Our data also reveal how these BCECs may be involved in the maintenance of BBB functions, as both the proliferation and migration are altered following changes in channel activity. SIGNIFICANCE STATEMENT: In brain capillary endothelial cells (BCECs) of the blood-brain barrier (BBB), TMEM16A is responsible for Ca2+-activated Cl- channels and can regulate both the resting membrane potential and cytosolic Ca2+ concentration, contributing to the proliferation and migration of BCECs. The present study provides novel information on the molecular mechanisms underlying the physiological functions of BCECs in the BBB and a novel target for therapeutic drugs for disorders associated with dysfunctions in the BBB.
- Published
- 2020
9. Pathophysiological roles of TRPC6 channels in pulmonary arterial hypertension
- Author
-
Hisao Yamamura, Yoshiaki Suzuki, and Aya Yamamura
- Subjects
medicine.medical_specialty ,Hypertension, Pulmonary ,Pulmonary Artery ,Muscle, Smooth, Vascular ,TRPC6 ,Pathogenesis ,Transient receptor potential channel ,Transient Receptor Potential Channels ,medicine.artery ,Internal medicine ,TRPC6 Cation Channel ,medicine ,Animals ,Humans ,Channel blocker ,Cell Proliferation ,Pharmacology ,SOC channels ,Pulmonary Arterial Hypertension ,business.industry ,medicine.anatomical_structure ,Pulmonary artery ,Vascular resistance ,Cardiology ,Calcium ,medicine.symptom ,business ,Vasoconstriction - Abstract
Pulmonary arterial hypertension (PAH) is a progressive and lethal disease of the pulmonary artery. The pathogenesis of PAH is mainly sustained vasoconstriction and vascular remodeling of the pulmonary artery. These pathogeneses cause progressive elevations in pulmonary vascular resistance and pulmonary arterial pressure in PAH patients. Elevated pulmonary arterial pressure leads to right heart failure and finally death. The vascular remodeling is caused by the enhanced proliferation and reduced apoptosis of pulmonary arterial smooth muscle cells (PASMCs). Excitable abnormality in the pulmonary artery of PAH patients is mostly mediated by an elevated cytosolic Ca2+ concentration. PASMCs express several Ca2+-permeable channels including voltage-dependent Ca2+ channels, store-operated Ca2+ (SOC) channels, and receptor-operated Ca2+ (ROC) channels. The activation and upregulation of these Ca2+ channels have been reported in PASMCs from PAH patients. Here, we analyzed pathophysiological functions of enhanced Ca2+ signaling mediated by SOC and ROC channels using PASMCs from idiopathic PAH patients and animal PAH models. Notch signal enhanced transient receptor potential canonical 6 (TRPC6) "SOC" channels via direct (non-genomic and stimulatory) and indirect (genomic and upregulating) effects in PAH. On the other hand, the activation of Ca2+-sensing receptors evoked Ca2+ influx through TRPC6 "ROC" channels in PAH. In addition, TRPC6 channel blocker and TRPC6 gene deletion inhibited the development of PAH. Specifically, TRPC6 channels potentially form both ROC and SOC channels in PASMCs, which are involved in the pathophysiological events in PAH. Therefore, targeting TRPC6 channels in PASMCs may help develop novel therapeutic approach for PAH.
- Published
- 2020
10. 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
11. Physiological and Pathological Functions of Cl− Channels in Chondrocytes
- Author
-
Hisao Yamamura, Yoshiaki Suzuki, and Yuji Imaizumi
- Subjects
0301 basic medicine ,Pharmacology ,Membrane potential ,Chemistry ,Cartilage ,Pharmaceutical Science ,General Medicine ,Chondrocyte ,Cell biology ,03 medical and health sciences ,Transient receptor potential channel ,030104 developmental biology ,medicine.anatomical_structure ,Chloride channel ,medicine ,Mechanosensitive channels ,Mechanotransduction ,Ion channel - Abstract
Articular chondrocytes are embedded in the cartilage of diarthrodial joints and responsible for the synthesis and secretion of extracellular matrix. The extracellular matrix mainly contains collagens and proteoglycans, and covers the articular cartilage to protect from mechanical and biochemical stresses. In mammalian chondrocytes, various types of ion channels have been identified: e.g., voltage-dependent K+ channels, Ca2+-activated K+ channels, ATP-sensitive K+ channels, two-pore domain K+ channels, voltage-dependent Ca2+ channels, store-operated Ca2+ channels, epithelial Na+ channels, acid-sensing ion channels, transient receptor potential channels, and mechanosensitive channels. These channels play important roles for the regulation of resting membrane potential, Ca2+ signaling, pH sensing, mechanotransduction, and cell proliferation in articular chondrocytes. In addition to these cation channels, Cl- channels are known to be expressed in mammalian chondrocytes: e.g., voltage-dependent Cl- channels, cystic fibrosis transmembrane conductance regulator channels, swelling-activated Cl- channels, and Ca2+-activated Cl- channels. Although these chondrocyte Cl- channels are thought to contribute to the regulation of resting membrane potential, Ca2+ signaling, cell volume, cell survival, and endochondral bone formation, the physiological functions have not been fully clarified. Osteoarthritis (OA) is caused by the degradation of articular cartilage, resulting in inflammation and pain in the joints. Therefore the pathophysiological roles of Cl- channels in OA chondrocytes are of considerable interest. Elucidating the physiological and pathological functions of chondrocyte Cl- channels will provide us a more comprehensive understanding of chondrocyte functions and may suggest novel molecular targets of drug development for OA.
- Published
- 2018
12. 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
13. Up-Regulation of the Voltage-Gated KV2.1 K+ Channel in the Renal Arterial Myocytes of Dahl Salt-Sensitive Hypertensive Rats
- Author
-
Susumu Ohya, Hisao Yamamura, Kazunobu Ogiwara, Yuji Imaizumi, and Yoshiaki Suzuki
- Subjects
0301 basic medicine ,medicine.medical_specialty ,Contraction (grammar) ,Pharmaceutical Science ,030204 cardiovascular system & hematology ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,medicine.artery ,Internal medicine ,medicine ,Myocyte ,Channel blocker ,cardiovascular diseases ,Renal artery ,Pharmacology ,Tetraethylammonium ,Voltage-gated ion channel ,General Medicine ,030104 developmental biology ,medicine.anatomical_structure ,Endocrinology ,chemistry ,Renal blood flow ,cardiovascular system ,Vascular resistance ,circulatory and respiratory physiology - Abstract
Salt-sensitive hypertension induces renal injury via decreased blood flow in the renal artery (RA), and ion channel dysfunction in RA myocytes (RAMs) may be involved in the higher renal vascular resistance. We examined the effects of several voltage-gated K+ (KV) channel blockers on the resting tension in endothelium-denuded RA strips and delayed-rectifier K+ currents in RAMs of Dahl salt-sensitive hypertensive rats (Dahl-S) fed with low- (Dahl-LS) and high-salt diets (Dahl-HS). The tetraethylammonium (TEA)-induced contraction in RA strips were significantly larger in Dahl-HS than Dahl-LS. Correspondingly, TEA-sensitive KV currents were significantly larger in the RAMs of Dahl-HS than Dahl-LS. Among the TEA-sensitive KV channel subtypes, the expression levels of KV2.1 transcript and protein were significantly higher in the RA of Dahl-HS than Dahl-LS, while those of KV1.5, KV7.1, and KV7.4 transcripts was comparable in two groups. KV2.1 currents detected as the guangxitoxin-1E-sensitive component were larger in the RAMs of Dahl-HS than Dahl-LS. These suggest that the up-regulation of the KV2.1 channel in RAMs may be involved in the compensatory mechanisms against decreased renal blood flow in salt-sensitive hypertension.
- Published
- 2017
14. Physiological roles of mitochondria and mitofusins on Ca2+ signaling in smooth muscles
- Author
-
Hisao Yamamura, Yuji Imaizumi, and Yoshiaki Suzuki
- Subjects
Pharmacology ,chemistry ,Endoplasmic reticulum ,chemistry.chemical_element ,Calcium ,Mitochondrion ,Ca2 signaling ,Calcium signaling ,Cell biology - Published
- 2017
15. Fingolimod ameliorates the development of PAH in monocrotaline-induced pulmonary hypertensive rats
- Author
-
Yoshiaki Suzuki, Moe Fujiwara, Hisao Yamamura, and Aya Yamamura
- Subjects
business.industry ,Applied Mathematics ,General Mathematics ,Medicine ,Pharmacology ,business ,Fingolimod ,medicine.drug - Published
- 2021
16. CRAC channel blockers attenuate the activation of human hepatic stellate cells
- Author
-
Rubii Kondo, Hisao Yamamura, and Yoshiaki Suzuki
- Subjects
Chemistry ,Applied Mathematics ,General Mathematics ,Hepatic stellate cell ,Channel blocker ,Pharmacology - Published
- 2021
17. Pimaric acid inhibits contraction of pulmonary artery via BKCa channel activation
- Author
-
Yoshiaki Suzuki, Ishida Masashi, and Hisao Yamamura
- Subjects
chemistry.chemical_compound ,Contraction (grammar) ,Bkca channel ,Chemistry ,Applied Mathematics ,General Mathematics ,medicine.artery ,Pulmonary artery ,medicine ,Pharmacology ,Pimaric acid - Published
- 2020
18. The ClC-7 Chloride Channel Is Downregulated by Hypoosmotic Stress in Human Chondrocytes
- Author
-
Yoshiaki Suzuki, Hisao Yamamura, Takashi Kurita, Wayne R. Giles, and Yuji Imaizumi
- Subjects
medicine.medical_specialty ,Programmed cell death ,Apoptosis ,4,4'-Diisothiocyanostilbene-2,2'-Disulfonic Acid ,Biology ,Cell Line ,Membrane Potentials ,chemistry.chemical_compound ,Chondrocytes ,Chlorides ,Chloride Channels ,Osmotic Pressure ,Internal medicine ,Osteoarthritis ,medicine ,Humans ,Pharmacology ,Membrane potential ,Gene knockdown ,Hyperpolarization (biology) ,Culture Media ,Endocrinology ,Gene Expression Regulation ,chemistry ,DIDS ,Cell culture ,Gene Knockdown Techniques ,Chloride channel ,Molecular Medicine ,Calcium - Abstract
Articular chondrocytes in osteoarthritis (OA) patients are exposed to hypoosmotic stress because the osmolality of this synovial fluid is significantly decreased. Hypoosmotic stress can cause an efflux of Cl(-) and an associated decrease of cell volume. We have previously reported that a Cl(-) conductance contributes to the regulation of resting membrane potential and thus can alter intracellular Ca(2+) concentration ([Ca(2+)]i) in human chondrocytes. The molecular identity and pathologic function of these Cl(-) channels, however, remained to be determined. Here, we show that the ClC-7 Cl(-) channel is strongly expressed in a human chondrocyte cell line (OUMS-27) and that it is responsible for Cl(-) currents that are activated by extracellular acidification (pH 5.0). These acid-sensitive currents are inhibited by 4,4'-diisothiocyanatostilbene-2,2'-disulfonic acid (DIDS; IC50 = 13 μM) and are markedly reduced by small-interfering RNA-induced knockdown of ClC-7. DIDS hyperpolarized these chondrocytes, and this was followed by an increase in [Ca(2+)]i. ClC-7 knockdown caused a similar hyperpolarization of the membrane potential. Short-term culture (48 hours) in hypoosmotic medium (270 mOsm) reduced the expression of ClC-7 and decreased the acid-sensitive currents. Interestingly, these hypoosmotic culture conditions, or ClC-7 knockdown, resulted in enhanced cell death. Taken together, our results show that the significant hyperpolarization due to ClC-7 impairment in chondrocytes can significantly increase [Ca(2+)]i and cell death. Thus, downregulation of ClC-7 channels during the hypoosmotic stress that accompanies OA progression is one important concept of the complex etiology of OA. These findings suggest novel targets for therapeutic intervention(s) and drug development for OA.
- Published
- 2015
19. 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
20. Tadalafil induces antiproliferation, apoptosis, and phosphodiesterase type 5 downregulation in idiopathic pulmonary arterial hypertension in vitro
- Author
-
Kikuo Tsukamoto, Aya Yamamura, Hisao Yamamura, Eri Fujitomi, Naoki Ohara, and Motohiko Sato
- Subjects
0301 basic medicine ,medicine.medical_specialty ,Sildenafil ,Myocytes, Smooth Muscle ,Down-Regulation ,Caspase 3 ,Apoptosis ,030204 cardiovascular system & hematology ,Pharmacology ,Pulmonary Artery ,Cell Line ,Tadalafil ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,medicine.artery ,Internal medicine ,medicine ,Humans ,Familial Primary Pulmonary Hypertension ,Cell Proliferation ,Cyclic Nucleotide Phosphodiesterases, Type 5 ,business.industry ,Phosphodiesterase 5 Inhibitors ,medicine.disease ,Pulmonary hypertension ,030104 developmental biology ,Endocrinology ,chemistry ,Vardenafil ,cGMP-specific phosphodiesterase type 5 ,Pulmonary artery ,medicine.symptom ,business ,Vasoconstriction ,medicine.drug - Abstract
Idiopathic pulmonary arterial hypertension (IPAH) is a fatal disease of the pulmonary artery resulting from a currently unidentified etiology. IPAH is pathologically characterized as sustained vasoconstriction and vascular remodeling of the pulmonary artery. Vascular remodeling is mediated by enhanced proliferation and reduced apoptosis in pulmonary arterial smooth muscle cells (PASMCs). Based on its pathological mechanism, specific phosphodiesterase type 5 (PDE5) inhibitors have been used in the treatment of IPAH. In addition to sildenafil, tadalafil has been approved for the treatment of IPAH. However, the effects of tadalafil on excessive proliferation of IPAH-PASMCs currently remain unknown. In the present study, the in vitro pharmacological profiles of tadalafil for cell proliferation and apoptosis were assessed in IPAH-PASMCs using MTT, BrdU incorporation, and caspase 3/7 assays. Expression analyses revealed that PDE5 mRNA and protein expression levels were markedly higher in IPAH-PASMCs than in normal-PASMCs. The treatment with tadalafil inhibited the excessive proliferation of IPAH-PASMCs in a concentration-dependent manner with an IC50 value of 4.5μM. On the other hand, tadalafil (0.03-100μM) did not affect cell growth of PASMCs from normal subjects and patients with chronic thromboembolic pulmonary hypertension (CTEPH). In addition, tadalafil induced apoptosis in IPAH-PASMCs. The antiproliferative and apoptotic effects of tadalafil were markedly stronger than those of sildenafil and vardenafil. The upregulated expression of PDE5 in IPAH-PASMCs was significantly attenuated by a long-term treatment with tadalafil. Taken together, these results indicate that tadalafil attenuates vascular remodeling by inhibiting cell proliferation, promoting apoptosis, and downregulating PDE5 in IPAH-PASMCs, thereby ameliorating IPAH.
- Published
- 2017
21. Modulation of TMEM16A-Channel Activity as Ca2+ Activated Cl− Conductance via the Interaction With Actin Cytoskeleton in Murine Portal Vein
- Author
-
Yuji Imaizumi, Junya Ohshiro, Yoshiaki Suzuki, and Hisao Yamamura
- Subjects
Pharmacology ,lcsh:RM1-950 ,HEK 293 cells ,Actin remodeling ,macromolecular substances ,Transfection ,Biology ,Actin cytoskeleton ,Cell biology ,chemistry.chemical_compound ,Actin remodeling of neurons ,lcsh:Therapeutics. Pharmacology ,chemistry ,Molecular Medicine ,Cytochalasin ,Actin ,Cytochalasin D - Abstract
TMEM16A is a major component of Ca2+-activated Cl− channel (CaCC) conductance in murine portal vein smooth muscle cells (mPVSMCs). Here, the regulation of CaCC activity by the actin cytoskeleton was examined in mPVSMCs. Actin disruption by cytochalasin D did not affect the current density, but increased the deactivation time constant in mPVSMCs. The elongated deactivation was recovered by jasplakinolide. When murine TMEM16A was transfected into HEK293 cells that have a poorly developed actin cytoskeleton, electrophysiological properties of CaCC currents were not changed by cytochalasin D. In conclusion, the CaCC activity in mPVSMCs is modified by the interaction of TMEM16A with abundant actin cytoskeleton. Keywords:: TMEM16A, actin cytoskeleton, portal vein
- Published
- 2014
22. Enhanced Ca2+-sensing Receptor Function in Pulmonary Hypertension
- Author
-
Hisao Yamamura, Jason X.-J. Yuan, and Aya Yamamura
- Subjects
Pharmacology ,medicine.medical_specialty ,business.industry ,Pharmaceutical Science ,medicine.disease ,Pulmonary hypertension ,medicine.anatomical_structure ,Right ventricular hypertrophy ,medicine.artery ,Hypoxic pulmonary vasoconstriction ,Internal medicine ,Pathophysiology of hypertension ,Pulmonary artery ,medicine ,Vascular resistance ,Cardiology ,medicine.symptom ,Receptor ,business ,Vasoconstriction - Abstract
Pulmonary arterial hypertension (PAH) is a rare, progressive, and fetal disease. The five-year survival rate after diagnosis is ~50%. In Japan, PAH is listed in the Specified Rare and Intractable Diseases. Pulmonary vascular remodeling and sustained pulmonary vasoconstriction are the major causes for the elevated pulmonary vascular resistance (PVR) in PAH. The pathogenic mechanisms involved in the pulmonary vascular abnormalities in PAH remain unclear. Sustained vasoconstriction and vascular remodeling owing to proliferation of pulmonary arterial smooth muscle cells (PASMCs) are key pathogenic events that lead to early morbidity and mortality. These events have been closely linked to Ca(2+) mobilization and signaling in PASMCs. An increase in cytosolic Ca(2+) concentration ([Ca(2+)]cyt) in PASMCs is an important stimulus for pulmonary vasoconstriction and cell proliferation which subsequently cause pulmonary vascular wall thickening followed by the increase in PVR. Increased resting [Ca(2+)]cyt and enhanced Ca(2+) influx have been implicated in PASMCs from PAH patients, but precise therapeutic targets to interrupt these signal pathways have not been identified. We recently found that the extracellular Ca(2+)-sensing receptor (CaSR), a G protein-coupled receptor (GPCR), is upregulated in PASMCs from patients with idiopathic pulmonary arterial hypertension (IPAH). In addition, blockage of the CaSR with an antagonist (NPS2143) prevents the development of pulmonary hypertension and right ventricular hypertrophy in animal models of pulmonary hypertension. The functionally upregulated CaSR in PASMCs is a novel pathogenic mechanism contributing to the augmented Ca(2+) signaling and excessive cell proliferation in IPAH. Targeting CaSR in PASMCs may help develop novel therapeutic approach for PAH.
- Published
- 2013
23. 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
24. Role of the KCa3.1 K+channel in auricular lymph node CD4+T-lymphocyte function of the delayed-type hypersensitivity model
- Author
-
Susumu Ohya, Yuji Imaizumi, Miki Matsui, Hisao Yamamura, Sayuri Horiba, Erina Nakamura, and Hiroaki Kito
- Subjects
Pharmacology ,Membrane potential ,medicine.diagnostic_test ,Repressor ,Potassium channel blocker ,Biology ,Molecular biology ,Flow cytometry ,Blot ,Oxazolone ,Pathogenesis ,chemistry.chemical_compound ,chemistry ,Immunology ,medicine ,Patch clamp ,medicine.drug - Abstract
Background and purpose The intermediate-conductance Ca(2+)-activated K(+) channel (K(Ca)3.1) modulates the Ca(2+) response through the control of the membrane potential in the immune system. We investigated the role of K(Ca)3.1 on the pathogenesis of delayed-type hypersensitivity (DTH) in auricular lymph node (ALN) CD4(+) T-lymphocytes of oxazolone (Ox)-induced DTH model mice. Experimental approach The expression patterns of K(Ca)3.1 and its possible transcriptional regulators were compared among ALN T-lymphocytes of three groups [non-sensitized (Ox-/-), Ox-sensitized, but non-challenged (Ox+/-) and Ox-sensitized and -challenged (Ox+/+)] using real-time polymerase chain reaction, Western blotting and flow cytometry. KCa 3.1 activity was measured by whole-cell patch clamp and the voltage-sensitive dye imaging. The effects of K(Ca)3.1 blockade were examined by the administration of selective K(Ca)3.1 blockers. Key results Significant up-regulation of K(Ca)3.1a was observed in CD4(+) T-lymphocytes of Ox+/- and Ox+/+, without any evident changes in the expression of the dominant-negative form, K(Ca)3.1b. Negatively correlated with this, the repressor element-1 silencing transcription factor (REST) was significantly down-regulated. Pharmacological blockade of K(Ca)3.1 resulted in an accumulation of the CD4(+) T-lymphocytes of Ox+/+ at the G0/G1 phase of the cell cycle, and also significantly recovered not only the pathogenesis of DTH, but also the changes in the K(Ca)3.1 expression and activity in the CD4(+) T-lymphocytes of Ox+/- and Ox+/+. Conclusions and implications The up-regulation of K(Ca)3.1a in conjunction with the down-regulation of REST may be involved in CD4(+) T-lymphocyte proliferation in the ALNs of DTH model mice; and K(Ca)3.1 may be an important target for therapeutic intervention in allergy diseases such as DTH.
- Published
- 2013
25. Imaging analyses of ion channel_molecule functions
- Author
-
Hisao Yamamura
- Subjects
Pharmacology ,Microscopy, Confocal ,Patch-Clamp Techniques ,Materials science ,Microscopy, Fluorescence ,Humans ,Calcium Channels ,In Vitro Techniques ,Molecular Imaging - Published
- 2013
26. 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
27. Down-Regulation of the Large-Conductance Ca2+-Activated K+ Channel, KCa1.1 in the Prostatic Stromal Cells of Benign Prostate Hyperplasia
- Author
-
Motomu Sakuragi, Hisao Yamamura, Susumu Ohya, Yoshiyuki Kojima, Yuji Imaizumi, Satomi Niwa, Kenjiro Kohri, and Shoichi Sasaki
- Subjects
Pharmacology ,Membrane potential ,medicine.medical_specialty ,Stromal cell ,medicine.diagnostic_test ,business.industry ,Pharmaceutical Science ,Depolarization ,General Medicine ,Hyperplasia ,medicine.disease ,Endocrinology ,Downregulation and upregulation ,Western blot ,Internal medicine ,Medicine ,Immunohistochemistry ,Channel blocker ,business - Abstract
Large-conductance Ca(2+)-activated K(+) (BK(Ca)) channel encoded by K(Ca)1.1 plays an important role in the control of smooth muscle tone by modulating membrane potential and intracellular Ca(2+) mobilization. BK(Ca) channel is functionally expressed in prostatic smooth muscle cells, and is activated by α(1)-adrenoceptor agonists. The main objective of this study was to elucidate the pathophysiological significance of changes in prostatic K(Ca)1.1 expressions in benign prostatic hyperplasia (BPH). Our previous study has shown that K(Ca)3.1 encoding intermediate-conductance K(Ca) (IK(Ca)) channel is up-regulated in stromal cells of implanted urogenital sinuses (UGSs) of stromal hyperplasia BPH model rats and in those of prostatic tissues from BPH patients. In the present study, the results from real-time polymerase chain reaction (PCR), Western blot, and immunohistochemical analyses showed significant down-regulation of K(Ca)1.1 transcripts and proteins and negative correlation between K(Ca)1.1 and K(Ca)3.1 transcript expressions in prostatic stromal cells of both BPH model rats and BPH patients. Corresponding to down-regulation of K(Ca)1.1 expression in stromal cells of implanted UGSs, membrane depolarization by application of the BK(Ca) channel blocker was disappeared. Down-regulation of K(Ca)1.1 may be involved in the phenotype switch from contractile profile to proliferative one in prostatic stromal cells of BPH patients.
- Published
- 2012
28. Downregulation of Ca2+-activated Cl- channel TMEM16A in cirrhotic portal hypertension
- Author
-
Hisao Yamamura, Yuji Imaizumi, and Yoshiaki Suzuki
- Subjects
Downregulation and upregulation ,Chemistry ,Applied Mathematics ,General Mathematics ,medicine ,Portal hypertension ,Channel (broadcasting) ,Pharmacology ,medicine.disease - Published
- 2018
29. Cellular Ca2+ Dynamics in Urinary Bladder Smooth Muscle From Transgenic Mice Overexpressing Na+-Ca2+ Exchanger
- Author
-
Eiji Sawada, Susumu Ohya, Yuji Imaizumi, Hidemichi Murata, Shingo Hotta, Hisao Yamamura, Takahiro Iwamoto, and Satomi Kita
- Subjects
Genetically modified mouse ,Male ,medicine.medical_specialty ,Transgene ,Myocytes, Smooth Muscle ,Urinary Bladder ,Stimulation ,Mice, Transgenic ,Sodium-Calcium Exchanger ,chemistry.chemical_compound ,Mice ,Smooth muscle ,Internal medicine ,medicine ,Myocyte ,Animals ,Pharmacology ,Urinary bladder ,Chemistry ,lcsh:RM1-950 ,medicine.anatomical_structure ,Endocrinology ,lcsh:Therapeutics. Pharmacology ,Gene Expression Regulation ,cardiovascular system ,Molecular Medicine ,Calcium ,Caffeine ,Acetylcholine ,medicine.drug - Abstract
The rise of Ca2+ concentration ([Ca2+] i ) by reducing external Na+ in urinary bladder smooth muscle cells (UBSMCs) from transgenic mice overexpressing Na+/Ca2+ exchanger type-1.3 (NCX1.3tg/tg) was about 4 times as large as that in the wild-type (WT). NCX1 protein expression in UB increased about 4-fold in NCX1.3tg/tg. The Ca2+ release by caffeine in UBSMCs was comparable between NCX1.3tg/tg and WT, but [Ca2+]i decay was faster in NCX1.3tg/tg. Contractions induced by acetylcholine, 60 mM K+, or electrical stimulation were significantly smaller in UB segments of NCX1.3tg/tg. NCX worked in Ca2+-extrusion mode during these contractions in UBSMCs of both WT and NCX1.3tg/tg. Keywords:: Na+-Ca2+ exchanger, urinary bladder smooth muscle, transgenic mouse
- Published
- 2010
30. 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
31. TRPV4-Like Non-selective Cation Currents in Cultured Aortic Myocytes
- Author
-
Yuji Imaizumi, Yuka Itoh, Susumu Ohya, Hisao Yamamura, Ryoichi Tanaka, Katsuhiko Muraki, and Noriyuki Hatano
- Subjects
Male ,TRPV4 ,Agonist ,medicine.medical_specialty ,Ruthenium red ,Time Factors ,medicine.drug_class ,Myocytes, Smooth Muscle ,TRPV Cation Channels ,Pulmonary Artery ,Biology ,Muscle, Smooth, Vascular ,Membrane Potentials ,chemistry.chemical_compound ,Internal medicine ,medicine.artery ,Phorbol Esters ,medicine ,Animals ,Humans ,Myocyte ,RNA, Messenger ,Rats, Wistar ,Reversal potential ,Aorta ,Cells, Cultured ,Pharmacology ,Membrane potential ,Dose-Response Relationship, Drug ,lcsh:RM1-950 ,Ruthenium Red ,Molecular biology ,Rats ,Endocrinology ,lcsh:Therapeutics. Pharmacology ,chemistry ,Cell culture ,Molecular Medicine ,Calcium - Abstract
In this study, we provide evidence of critical changes in the expression of non-selective cation currents (NSCC) during culture in rat aortic myocytes. A selective TRPV4 agonist, 4α-phorbol 12,13-didecanoate (4αPDD), had little effect on membrane currents and intracellular Ca2+ (Ca2+i) in freshly isolated cells from the aorta. In contrast, in cultured aortic myocytes with and without serum, 4αPDD at a concentration range between 0.3 and 3 μM effectively elevated Ca2+i, which was abolished in the absence of external Ca2+. Application of 4αPDD to cultured aortic myocytes also activated NSCC, which had a reversal potential of +3 mV. Both of these signals were blocked by ruthenium red (RuR), an effective blocker of TRPVs. Although the expression of TRPV4 mRNA transcript was found in cultured as well as non-cultured aortic myocytes, significant immunoreactivity to TRPV4 protein was only detected in cultured rat aortic myocytes. Moreover, cultured human pulmonary arterial smooth muscle cells (hPASM) had a substantial response to 4αPDD, which was susceptible to the removal of external Ca2+ and application of RuR. These results provide a strong basis for our proposal that endogenous TRPV4 functions as an important regulator of Ca2+i in vascular myocytes under some physiological and pathophysiological conditions. Keywords:: TRPV4, 4α-phorbol 12,13-didecanoate, cell-culture
- Published
- 2008
32. 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
33. Evans Blue Is a Specific Antagonist of the Human Epithelial Na+ Channel δ-Subunit
- Author
-
Takashi Ueda, Shoichi Shimada, Shinya Ugawa, and Hisao Yamamura
- Subjects
Epithelial sodium channel ,medicine.medical_specialty ,Patch-Clamp Techniques ,Xenopus ,Protein subunit ,Vascular permeability ,Sodium Channels ,Membrane Potentials ,chemistry.chemical_compound ,Internal medicine ,medicine ,Animals ,Humans ,Epithelial Sodium Channels ,Evans Blue ,Pharmacology ,Dose-Response Relationship, Drug ,biology ,Antagonist ,biology.organism_classification ,Amiloride ,Cell biology ,Endocrinology ,chemistry ,Oocytes ,Molecular Medicine ,Homeostasis ,Sodium Channel Blockers ,medicine.drug - Abstract
The epithelial Na(+) channel (ENaC) regulates Na(+) homeostasis in cells and across epithelia. Four homologous ENaC subunits (alpha, beta, gamma, and delta) have been isolated in mammals. Combination of alpha-, beta-, and gamma-subunits or delta-, beta-, and gamma-subunits forms fully functional channels. Amiloride is a well known blocker of the ENaC family that inhibits both channel complexes. However, no specific antagonists are currently known that distinguish them. Here, we show that Evans blue, a diagnostic aid for the measurement of blood volume and vascular permeability, inhibits the activity of the delta-subunit expressed in Xenopus oocytes. The inward currents at a holding potential of -60 mV in human ENaCdeltabetagamma-expressing oocytes were inhibited by the application of Evans blue in a concentration-dependent manner with an IC(50) value of 143 muM. Evans blue markedly inhibited the delta-subunit current but did not block the alpha-subunit current. In conclusion, Evans blue is the first known delta-subunit-specific antagonist of ENaC. This finding provides us with a key compound for elucidating the physiological and pathological functions of ENaCdelta in humans and for drug development in the ENaC family.
- Published
- 2005
34. 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
35. Membrane hyperpolarization induced by endoplasmic reticulum stress facilitates ca(2+) influx to regulate cell cycle progression in brain capillary endothelial cells
- Author
-
Susumu Ohya, Hiroaki Kito, Yuji Imaizumi, Kiyofumi Asai, Hisao Yamamura, and Yoshiaki Suzuki
- Subjects
Cell Line ,Membrane Potentials ,Downregulation and upregulation ,Animals ,Potassium Channels, Inwardly Rectifying ,TRPC ,Calcium signaling ,Pharmacology ,Cell Death ,Chemistry ,Endoplasmic reticulum ,lcsh:RM1-950 ,Cell Cycle ,Brain ,Endothelial Cells ,STIM1 ,Membrane hyperpolarization ,Endoplasmic Reticulum Stress ,Store-operated calcium entry ,Cell biology ,lcsh:Therapeutics. Pharmacology ,Unfolded protein response ,cardiovascular system ,Molecular Medicine ,Calcium ,Cattle - Abstract
Upregulation of the Kir2.1 channel during endoplasmic reticulum (ER) stress in t-BBEC117, an immortalized bovine brain endothelial cell line, caused a sustained increase in intracellular Ca2+ concentration ([Ca2+]i) and a facilitation of cell death. Expressions of Ca2+ influx channels (TRPC, Orai1, STIM1) were unchanged by ER stress. The ER stress–induced [Ca2+]i increase was mainly attributed to the deeper resting membrane potential due to Kir2.1 upregulation. ER stress arrested at the G2/M phase and it was attenuated by an inhibitor of Kir2.1. These results indicate that Kir2.1 upregulation by ER stress facilitates cell death via regulation of cell cycle progression in t-BBEC117. Keywords:: brain capillary endothelial cell, store operated calcium entry, endoplasmic reticulum stress
- Published
- 2014
36. 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
37. [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
38. Dihydropyridine Ca2+ Channel Blockers Increase Cytosolic [Ca2+] by Activating Ca2+-sensing Receptors in Pulmonary Arterial Smooth Muscle Cells
- Author
-
Ayako Makino, Jason X.-J. Yuan, Nicole M. Pohl, Aya Yamamura, Eun A. Ko, Amy Zeifman, Qiang Guo, Kimberly A. Smith, Jun Wan, Adriana M. Zimnicka, Shanshan Song, and Hisao Yamamura
- Subjects
Male ,medicine.medical_specialty ,Calcium Channels, L-Type ,Nifedipine ,Physiology ,medicine.drug_class ,Hypertension, Pulmonary ,Inositol Phosphates ,Recombinant Fusion Proteins ,Nicardipine ,Myocytes, Smooth Muscle ,Calcium channel blocker ,Pharmacology ,Naphthalenes ,Pulmonary Artery ,Transfection ,Article ,Rats, Sprague-Dawley ,chemistry.chemical_compound ,Cytosol ,Internal medicine ,medicine ,Animals ,Humans ,Channel blocker ,Calcium Signaling ,Cells, Cultured ,Monocrotaline ,Voltage-dependent calcium channel ,Chemistry ,Dihydropyridine ,medicine.disease ,Calcium Channel Blockers ,Bay K8644 ,Pulmonary hypertension ,Rats ,Up-Regulation ,Endocrinology ,Vasoconstriction ,Disease Progression ,Cardiology and Cardiovascular Medicine ,Receptors, Calcium-Sensing ,medicine.drug ,Signal Transduction - Abstract
Rationale: An increase in cytosolic free Ca 2+ concentration ([Ca 2+ ] cyt ) in pulmonary arterial smooth muscle cells (PASMC) is a major trigger for pulmonary vasoconstriction and an important stimulus for PASMC proliferation and pulmonary vascular remodeling. The dihydropyridine Ca 2+ channel blockers, such as nifedipine, have been used for treatment of idiopathic pulmonary arterial hypertension (IPAH). Objective: Our previous study demonstrated that the Ca 2+ -sensing receptor (CaSR) was upregulated and the extracellular Ca 2+ -induced increase in [Ca 2+ ] cyt was enhanced in PASMC from patients with IPAH and animals with experimental pulmonary hypertension. Here, we report that the dihydropyridines (eg, nifedipine) increase [Ca 2+ ] cyt by activating CaSR in PASMC from IPAH patients (in which CaSR is upregulated), but not in normal PASMC. Methods and Results: The nifedipine-mediated increase in [Ca 2+ ] cyt in IPAH-PASMC was concentration dependent with a half maximal effective concentration of 0.20 µmol/L. Knockdown of CaSR with siRNA in IPAH-PASMC significantly inhibited the nifedipine-induced increase in [Ca 2+ ] cyt , whereas overexpression of CaSR in normal PASMC conferred the nifedipine-induced rise in [Ca 2+ ] cyt . Other dihydropyridines, nicardipine and Bay K8644, had similar augmenting effects on the CaSR-mediated increase in [Ca 2+ ] cyt in IPAH-PASMC; however, the nondihydropyridine blockers, such as diltiazem and verapamil, had no effect on the CaSR-mediated rise in [Ca 2+ ] cyt . Conclusions: The dihydropyridine derivatives increase [Ca 2+ ] cyt by potentiating the activity of CaSR in PASMC independently of their blocking (or activating) effect on Ca 2+ channels; therefore, it is possible that the use of dihydropyridine Ca 2+ channel blockers (eg, nifedipine) to treat IPAH patients with upregulated CaSR in PASMC may exacerbate pulmonary hypertension.
- Published
- 2013
39. Novel spliced variants of large-conductance Ca(2+)-activated K(+)-channel β2-subunit in human and rodent pancreas
- Author
-
Takuya Kimura, Susumu Ohya, Tomohiro Fujimori, Hisao Yamamura, and Yuji Imaizumi
- Subjects
Male ,medicine.medical_specialty ,BK channel ,Potassium Channels ,Large-Conductance Calcium-Activated Potassium Channel beta Subunits ,Protein subunit ,RNA Splicing ,Mice, Inbred Strains ,Biology ,Membrane Potentials ,Cell membrane ,Mice ,Potassium Channels, Calcium-Activated ,Downregulation and upregulation ,Internal medicine ,Cell Line, Tumor ,medicine ,Animals ,Humans ,Protein Isoforms ,Rats, Wistar ,Insulinoma ,Pancreas ,Pharmacology ,lcsh:RM1-950 ,HEK 293 cells ,Cell Membrane ,medicine.disease ,Molecular biology ,Potassium channel ,Rats ,Up-Regulation ,Pancreatic Neoplasms ,Protein Subunits ,lcsh:Therapeutics. Pharmacology ,Endocrinology ,medicine.anatomical_structure ,HEK293 Cells ,biology.protein ,Molecular Medicine - Abstract
Large-conductance Ca2+-activated K+ (BK) channel regulates action potential firing in pancreatic β-cells. We cloned novel spliced variants of the BK-channel β2-subunit (BKβ2b), which consisted of 36 amino acids including the N-terminal in the original human BKβ2 (BKβ2a), from human and rodent pancreas. Real-time PCR analysis showed the abundant expression of BKβ2b transcripts in human and rodent pancreas and also in the RINm5f insulinoma cell line. In addition, up-regulation of both BK-channel α-subunit (BKα) and BKβ2b transcripts was observed in pancreas tissues from diabetes mellitus patients. In HEK293 cells co-expressing BKα and BKβ2b, the inactivation of BK-channel currents, which is typical for BKα + BKβ2a, was not observed, and electrophysiological and pharmacological properties of BKα + BKβ2b were almost identical to those of BKα alone. In HEK293 cells stably expressing BKα, the transient co-expression of yellow fluorescence protein (YFP)-tagged BKβ2a proteins resulted in their distribution along the cell membrane. In contrast, the co-expression of YFP-tagged BKβ2b with BKα showed diffusely distributed fluorescence signals throughout the cell body. Taken together, the predominant splicing of BKβ2b versus that of BKβ2a presumably enhances the contribution of BK channels to membrane potential and may possibly be a factor modulating insulin secretion in a suppressive manner in pancreatic β-cells. Keywords:: Ca2+-activated K+ channel, β2-subunit, spliced variant, pancreatic islet β-cell, diabetes mellitus
- Published
- 2010
40. Regulation of ryanodine receptor-mediated Ca(2+) release in vas deferens smooth muscle cells
- Author
-
Yuji Imaizumi, Hisao Yamamura, Susumu Ohya, and Akitoshi Ohno
- Subjects
Male ,medicine.medical_specialty ,Patch-Clamp Techniques ,Phosphodiesterase Inhibitors ,Blotting, Western ,Myocytes, Smooth Muscle ,Biology ,In Vitro Techniques ,Ryanodine receptor 2 ,Flutamide ,chemistry.chemical_compound ,Vas Deferens ,Internal medicine ,Caffeine ,medicine ,Myocyte ,Animals ,Testosterone ,Patch clamp ,Calcium Signaling ,Rats, Wistar ,Receptor ,DNA Primers ,Pharmacology ,Ryanodine receptor ,Reverse Transcriptase Polymerase Chain Reaction ,lcsh:RM1-950 ,Vas deferens ,Androgen Antagonists ,Ryanodine Receptor Calcium Release Channel ,Rats ,Electrophysiology ,lcsh:Therapeutics. Pharmacology ,Endocrinology ,medicine.anatomical_structure ,chemistry ,Molecular Medicine ,RNA ,Calcium ,medicine.symptom ,Muscle contraction ,Muscle Contraction - Abstract
Ca2+ release from intracellular store sites via the ryanodine receptor (RyR) and hormonal regulation by flutamide, an androgen-receptor (AR) antagonist, on it were examined in vas deferens (VD) smooth muscle cells (SMCs). VD and VDSMCs were obtained from two groups of male rats that were treated p.o. with 100 mg/kg flutamide (Flu) or vehicle (Vehicle). Both spontaneous and caffeine-induced Ca2+ releases were markedly smaller in single VDSMCs from Flu than in those from Vehicle. Interestingly, [Ca2+]i rise by 100 μM norepinephrine in VDSMCs from Flu was larger than that in those from Vehicle. The contractions induced by direct electrical stimulation in tissue preparations from Flu showed lower susceptibility to 30 μM ryanodine than those from Vehicle. Real-time PCR analyses revealed that the transcripts of ryanodine receptor (RyR) type 2 and type 3 (RyR2 and RyR3) were expressed in VD and markedly reduced in Flu. The protein expression of total RyR was significantly reduced by flutamide treatment, but that of inositol 1,4,5-trisphosphate receptor (IP3R) was not affected. It can be strongly suggested that long term block of AR by flutamide reduced the expression of RyR and its contribution to the contraction, but not those of IP3R in VDSMCs. Keywords:: ryanodine receptor, Ca2+ release, flutamide, vas deferens, smooth muscle
- Published
- 2009
41. 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
42. Icilin activates the delta-subunit of the human epithelial Na+ channel
- Author
-
Shoichi Shimada, Takashi Ueda, Hisao Yamamura, Shinya Ugawa, and Masataka Nagao
- Subjects
Epithelial sodium channel ,Xenopus ,Pyrimidinones ,Pharmacology ,Sodium Channels ,Amiloride ,chemistry.chemical_compound ,Xenopus laevis ,TRPM8 ,medicine ,Homomeric ,Animals ,Humans ,Epithelial Sodium Channels ,biology ,Activator (genetics) ,Icilin ,biology.organism_classification ,Sodium Channel Agonists ,Cell biology ,chemistry ,Molecular Medicine ,Capsaicin ,Protons ,Capsazepine ,medicine.drug - Abstract
The amiloride-sensitive epithelial Na(+) channel (ENaC) regulates Na(+) homeostasis in cells and across epithelia. Four homologous ENaC subunits (alpha, beta, gamma, and delta) have been isolated in mammals. The chemical activators acting on ENaC, however, are largely unknown. More recently, we have found that capsazepine activates human ENaCdelta (hENaCdelta), which is mainly expressed in the brain. In addition, here we show that icilin, which is a tetrahydropyrimidine-2-one derivative unrelated structurally to capsazepine, markedly enhanced the activity of hENaCdeltabetagamma heteromultimer expressed in Xenopus laevis oocytes. The inward currents at a holding potential of -60 mV in hENaCdeltabetagamma-expressing oocytes were increased by the application of icilin in a concentration-dependent manner with an EC(50) value of 33 microM. The icilin-elicited current was mostly abolished by the addition of 100 microM amiloride or by the removal of external Na(+). Homomeric hENaCdelta was also significantly activated by icilin, whereas hENaCalpha activity was not affected by icilin, and icilin caused a slight inhibition of the hENaCalphabetagamma current. Furthermore, icilin acted together with protons or capsazepine on hENaCdeltabetagamma. These findings identify icilin as a novel chemical activator of ENaCdelta, providing us with a lead compound for drug development in the degenerin/ENaC superfamily.
- Published
- 2005
43. Coexpression of vanilloid receptor subtype-1 and acid-sensing ion channel genes in the human trigeminal ganglion neurons
- Author
-
Shoichi Shimada, Takashi Ueda, Hisao Yamamura, Masataka Nagao, and Shinya Ugawa
- Subjects
Physiology ,Xenopus ,TRPV1 ,Pain ,TRPV Cation Channels ,Nerve Tissue Proteins ,Pharmacology ,Models, Biological ,Ion Channels ,Sodium Channels ,Behavioral Neuroscience ,Trigeminal ganglion ,Transient receptor potential channel ,chemistry.chemical_compound ,Dorsal root ganglion ,Physiology (medical) ,medicine ,Animals ,Humans ,Acid-sensing ion channel ,In Situ Hybridization ,Skin ,Ions ,Neurons ,Sensory Systems ,Acid Sensing Ion Channels ,Nociception ,medicine.anatomical_structure ,chemistry ,Gene Expression Regulation ,Trigeminal Ganglion ,Nociceptor ,Oocytes ,Capsazepine ,Neuroscience - Abstract
Previous psychophysical experiments have shown that repeated applications of high concentrations of acids on one side of the dorsal surface of the human tongue evoke irritation or pain (Dessirier et al., 2000). Under acidification, protons dissociated from the acids probably activate excitatory cation channels expressed in local nociceptors that originate from trigeminal ganglia, leading to the generation of such sensations. Recent molecular investigations into sensory neurons have revealed that a transient receptor potential/vanilloid receptor subtype-1 (TRPV1) and an acid-sensing ion channel (ASIC) mediate the greater part of proton-induced irritation or nociception in mammals (Julius and Basbaum, 2001; Ugawa et al., 2003). Here we provide evidence for involvement of both channels in acid-evoked pain in humans and show their relative contributions to acid-evoked nociception. In our human pain model (approved by the Ethics Committee of Nagoya City University and conducted in accordance with the Declaration of Helsinki), direct infusion of acidic solutions (pH ≥ 6.0) into human skin caused localized pain, which was blocked by amiloride, an inhibitor of ASICs, but not by capsazepine, an inhibitor of TRPV1. Although the efficacy of amiloride was only partially attenuated under more severe acidification (pH 5.0), capsazepine produced some blocking effect on pH 5-evoked pain. Amiloride itself neither blocked capsaicin-evoked localized pain in human skin nor inhibited proton-induced currents in TRPV1expressing Xenopus oocytes (Ugawa et al., 2003). In situ hybridization histochemistry demonstrated that more than half of TRPV1-expressing dorsal root ganglion neurons were ASIC1a- or ASIC3-positive in the rat, and that approximately half of TRPV1expressing human trigeminal neurons were ASIC-positive (S. Ugawa, T. Ueda and S. Shimada, submitted for publication). These results suggest that ASICs are the leading acid sensors in human nociceptors and that both TRPV1 and ASIC channels are involved in acid-evoked oral irritation or pain.
- Published
- 2005
44. Mechanisms underlying the activation of large conductance Ca2+-activated K+ channels by nordihydroguaiaretic acid
- Author
-
Susumu Ohya, Yuji Imaizumi, Kazuho Sakamoto, Hisao Yamamura, and Katsuhiko Muraki
- Subjects
BK channel ,Patch-Clamp Techniques ,Swine ,In Vitro Techniques ,Muscle, Smooth, Vascular ,Cell Line ,Membrane Potentials ,chemistry.chemical_compound ,Potassium Channels, Calcium-Activated ,Animals ,Humans ,Masoprocol ,Patch clamp ,Large-Conductance Calcium-Activated Potassium Channels ,Pharmacology ,biology ,Chemistry ,Ryanodine receptor ,Depolarization ,respiratory system ,Hyperpolarization (biology) ,Iberiotoxin ,Coronary Vessels ,Potassium channel ,Mitochondria ,Rats ,Nordihydroguaiaretic acid ,Biochemistry ,Biophysics ,biology.protein ,Calcium - Abstract
The mechanisms underlying the activation of large conductance Ca2+-activated K+ (BK) channel by nordihydroguaiaretic acid (NDGA) were examined in human embryonic kidney (HEK293) cells, where BK channel alpha (BKalpha) or a plus beta1 subunit (BKalphabeta1) was heterologously expressed, and also in freshly isolated porcine coronary arterial smooth muscle cells (PCASMCs). The activity of both BKalpha and BKalphabeta1 channels was increased by 10 microM NDGA in similar manners, indicating the selective action on the a subunit to increase Ca2+ sensitivity. The application of NDGA to PCASMCs induced outward current and hyperpolarization under voltage and current clamp, respectively, in a concentration-dependent manner (> or = 3 microM). These effects were blocked by 100 nM iberiotoxin. Electrical events induced by NDGA (> or = 10 microM) were, unexpectedly, associated with the increase in [Ca2+]i. After the treatment with caffeine and ryanodine, the [Ca2+]i increase by NDGA was markedly reduced and the hyperpolarization by NDGA was attenuated. The Ca2+ release by 10 microM NDGA was preceded by membrane depolarization of mitochondria. These results indicate that BK channel opening by NDGA in PCASMCs is due to the direct action on a subunit and also to Ca2+ release from sarcoplasmic reticulum, presumably via, at least in part, the inhibition of mitochondria respiration.
- Published
- 2002
45. Ca2+ spark as a regulator of ion channel activity
- Author
-
Katsuhiko Muraki, Susumu Ohya, Yoshiaki Ohi, Minoru Watanabe, Hisao Yamamura, and Yuji Imaizumi
- Subjects
Pharmacology ,Potassium Channels ,Chemistry ,Ryanodine receptor ,Cardiac muscle ,Action Potentials ,Afterhyperpolarization ,Muscle, Smooth ,Ryanodine Receptor Calcium Release Channel ,Membrane hyperpolarization ,Anatomy ,musculoskeletal system ,Potassium channel ,Ion Channels ,medicine.anatomical_structure ,Muscle relaxation ,cardiovascular system ,medicine ,Biophysics ,Myocyte ,Repolarization ,Humans ,Calcium - Abstract
Ca2+ spark is a local and transient Ca2+ release from sarcoplasmic reticulum (SR) through the ryanodine receptor Ca2+-releasing channel (RyR). In cardiac myocytes, Ca2+ spark is an elementary unit of Ca2+-induced Ca2+ release (CICR) by opening of RyR(s) in junctional SR (jSR), which is triggered by Ca2+-influx through L-type Ca2+ channels to the narrow space between a transverse tubule and jSR. Ca2+ spark has, therefore, been described as the evidence for "the local control of excitation-contraction coupling". In contrast, Ca2+ sparks in smooth muscle have been reported in relation to Ca2+-dependent K+ (K(Ca)) channel activation and muscle relaxation. A spontaneous Ca2+ spark in a superficial area activates 10-100 K(Ca) channels nearby and induces membrane hyperpolarization, which reduces Ca2+ channel activity. In several types of smooth muscle cells, which have relatively high membrane excitability, an action potential (AP) elicits 5-20 Ca2+ hot spots (evoked sparks with long life) in the early stage via CICR in discrete superficial SR elements and activates K(Ca)-channel current highly responsible for AP repolarization and afterhyperpolarization. CICR available for contraction may occur more slowly by the propagation of CICR from superficial SR to deeper ones. The regulatory mechanism of ion channel activity on plasma membrane by superficial SR via Ca2+ spark generation in smooth muscle cells may be analogously common in several types of cells including neurons.
- Published
- 1999
46. Direct regulation of ion channel by PIP2
- Author
-
Hisao Yamamura
- Subjects
Phosphatidylinositol 4,5-Diphosphate ,Pharmacology ,Membrane Microdomains ,Materials science ,Chemical physics ,Animals ,Humans ,Ion Channel Gating ,Second Messenger Systems ,Ion Channels ,Ion channel ,Signal Transduction - Published
- 2013
47. Mitochondrial transmembrane potential and Ca2+ uptake and ATP-dependent K+ channel activity in smooth muscle cells
- Author
-
Minoru Watanabe, Yuji Imaizumi, Yoshiaki Ohi, Katuhiko Muraki, and Hisao Yamamura
- Subjects
Pharmacology ,Membrane potential ,Ca2 uptake ,Smooth muscle ,Chemistry ,Biophysics ,K channels - Published
- 2000
48. BK channel activation by NS-1619 partially mediates intracellular Ca2+ release in porcine coronary arterial smooth muscle cells
- Author
-
Minoru Watanabe, Yoshiaki Ohi, Hisao Yamamura, Katsuhiko Muraki, and Yuji Imaizumi
- Subjects
Pharmacology ,BK channel ,biology ,Chemistry ,biology.protein ,Ca2 release ,Intracellular ,Cell biology ,Arterial smooth muscle cells - Published
- 2000
49. Heterogeneity of activation threshold for Ca2+ hot spots in guinea pig vas deferens smooth muscle cell
- Author
-
Yuji Imaizumi, Minoru Watanabe, Hisao Yamamura, and Voshifllri Oi
- Subjects
Pharmacology ,Andrology ,Guinea pig ,medicine.anatomical_structure ,Smooth muscle ,Chemistry ,Cell ,Vas deferens ,medicine - Published
- 2000
50. Modulation of cytosolic Ca2+ concentration by nordihydroguaiaretic acid in porcine coronary arterial smooth muscle cells
- Author
-
Hisao Yamamura, Minoru Watanabe, Katsuhiko Muraki, and Yuji Imaizumi
- Subjects
Pharmacology ,Nordihydroguaiaretic acid ,Cytosol ,chemistry.chemical_compound ,chemistry ,Ca2 concentration ,Biophysics ,Arterial smooth muscle cells - Published
- 1999
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.