Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 146
Filter
Add more filters

Country/Region as subject
Publication year range
1.
J Craniofac Surg ; 33(3): 920-925, 2022 May 01.
Article in English | MEDLINE | ID: mdl-34538794

ABSTRACT

ABSTRACT: The hybrid technique after bilateral sagittal split ramus osteotomy is an internal fixation method using monocortical mini-plates and additional bicortical positional screws. In this study, we analyzed the postoperative stability of 23 patients with mandibular asymmetry who underwent bilateral sagittal split ramus osteotomy and hybrid fixation with or without LeFort I osteotomy. Anatomical landmarks of the deviated and non-deviated sides of the jaw were established to measure the angle and distance to the reference plane in three-dimensional cone beam computed tomography images. We analyzed the positional changes and correlations of the reference points at preoperative (T1), postoperative 2 weeks (T2), and postoperative 1 year (T3). There were significant differences in preoperative position of the upper and lower molar cervix alveolar crest to the reference plane (U6-X and L6-X) and the condylion angles between deviated and non-deviated sides. Postoperatively (T2-T3), each reference point had no statistically significant positional change. Pearson correlation coefficient between the amount of menton deviation (ME-X at T1) and positional change of menton after surgery (T2-T3) was 0.30, and P value was 0.168. The hybrid fixation technique is an effective fixation method for achieving postoperative stability for mandibular asymmetry.


Subject(s)
Osteotomy, Sagittal Split Ramus , Prognathism , Cephalometry/methods , Cone-Beam Computed Tomography/methods , Female , Humans , Imaging, Three-Dimensional , Mandible/diagnostic imaging , Mandible/surgery , Mandibular Condyle , Osteotomy, Sagittal Split Ramus/methods , Prognathism/surgery , Retrospective Studies
2.
Korean J Physiol Pharmacol ; 24(1): 101-110, 2020 Jan.
Article in English | MEDLINE | ID: mdl-31908579

ABSTRACT

Transient receptor potential canonical 4 (TRPC4) channel is a nonselective calcium-permeable cation channels. In intestinal smooth muscle cells, TRPC4 currents contribute more than 80% to muscarinic cationic current (mIcat). With its inward-rectifying current-voltage relationship and high calcium permeability, TRPC4 channels permit calcium influx once the channel is opened by muscarinic receptor stimulation. Polyamines are known to inhibit nonselective cation channels that mediate the generation of mIcat. Moreover, it is reported that TRPC4 channels are blocked by the intracellular spermine through electrostatic interaction with glutamate residues (E728, E729). Here, we investigated the correlation between the magnitude of channel inactivation by spermine and the magnitude of channel conductance. We also found additional spermine binding sites in TRPC4. We evaluated channel activity with electrophysiological recordings and revalidated structural significance based on Cryo-EM structure, which was resolved recently. We found that there is no correlation between magnitude of inhibitory action of spermine and magnitude of maximum current of the channel. In intracellular region, TRPC4 attracts spermine at channel periphery by reducing access resistance, and acidic residues contribute to blocking action of intracellular spermine; channel periphery, E649; cytosolic space, D629, D649, and E687.

3.
J Struct Biol ; 208(3): 107395, 2019 12 01.
Article in English | MEDLINE | ID: mdl-31560999

ABSTRACT

Transaminases are pyridoxal 5'-phosphate-dependent enzymes that reversibly catalyze transamination reactions from an amino group donor substrate to an amino group acceptor substrate. ω-Transaminases (ωTAs) utilize compounds with an amino group not at α-carbon position as their amino group donor substrates. Recently, a novel ωTA with broad substrate specificity and high thermostability from the thermophilic bacterium Sphaerobacter thermophilus (St-ωTA) has been reported. Although St-ωTA has been biochemically characterized, little is known about its determinants of substrate specificity. In the present study, we determined the crystal structure of St-ωTA at 1.9 Šresolution to clarify in detail its mechanism of substrate recognition. The structure of St-ωTA revealed that it has a voluminous active site resulting from the unique spatial arrangement of residues comprising its active site. In addition, our molecular docking simulation results suggest that substrate compounds may bind to active site residues via electrostatic interactions or hydrophobic interactions that can be induced by subtle rearrangements of active site residues. On the basis of these structural analyses, we propose a plausible working model of the enzymatic mechanism of St-ωTA. Our results provide profound structural insights into the substrate specificity of St-ωTA and extend the boundaries of knowledge of TAs.


Subject(s)
Chloroflexi/enzymology , Transaminases/chemistry , Transaminases/metabolism , Bacterial Proteins/chemistry , Bacterial Proteins/metabolism , Catalytic Domain , Crystallography, X-Ray , Molecular Docking Simulation , Protein Conformation , Pyridoxal Phosphate/metabolism , Spectrophotometry, Ultraviolet , Substrate Specificity
4.
Biochem Biophys Res Commun ; 519(2): 366-371, 2019 11 05.
Article in English | MEDLINE | ID: mdl-31519324

ABSTRACT

Zinc finger homeobox 3 (ZFHX3) is a transcription factor that regulates multiple cellular processes including cell proliferation, differentiation and neoplastic development. It is also involved in the function of steroid hormones estrogen and progesterone and the peptide hormone prolactin in mammary epithelial cells. In this study, we investigated whether and how ZFHX3 regulates intracellular calcium homeostasis in mammary epithelial cells. We found that ZFHX3 affected both store operated calcium entry and store independent calcium entry (SOCE and SICE). Simultaneously, the expression of the calcium channel TRPV6 was regulated by ZFHX3, as demonstrated by expression analysis and luciferase reporter assay. In cells with knockdown of ZFHX3, calcium entry was partially rescued by the overexpression of wild type but not the pore mutants of TRPV6. In addition, overexpression of TRPV6 promoted differentiation of the MCF10A mammary epithelial cells in three-dimensional culture, which is consistent with our previous findings that ZFHX3 is essential for mammary gland differentiation. These findings suggest that ZFHX3 plays an important role in intracellular calcium homeostasis in mammary epithelial cells, at least in part, by regulating TRPV6.


Subject(s)
Calcium Channels/metabolism , Calcium/metabolism , Epithelial Cells/metabolism , Homeodomain Proteins/metabolism , TRPV Cation Channels/metabolism , Calcium Channels/genetics , Cells, Cultured , HEK293 Cells , Humans , TRPV Cation Channels/genetics
5.
J Korean Med Sci ; 39(3): e34, 2024 Jan 22.
Article in English | MEDLINE | ID: mdl-38258366

Subject(s)
Caves , Paintings , Humans
6.
Korean J Physiol Pharmacol ; 23(5): 357-366, 2019 Sep.
Article in English | MEDLINE | ID: mdl-31496873

ABSTRACT

Gαq-coupled receptor stimulation was implied in the activation process of transient receptor potential canonical (TRPC)1/4 and TRPC1/5 heterotetrameric channels. The inactivation occurs due to phosphatidylinositol 4,5-biphosphate (PI(4,5)P2) depletion. When PI(4,5)P2 depletion was induced by muscarinic stimulation or inositol polyphosphate 5-phosphatase (Inp54p), however, the inactivation by muscarinic stimulation was greater compared to that by Inp54p. The aim of this study was to investigate the complete inactivation mechanism of the heteromeric channels upon Gαq-phospholipase C ß (Gαq-PLCß) activation. We evaluated the activity of heteromeric channels with electrophysiological recording in HEK293 cells expressing TRPC channels. TRPC1/4 and TRPC1/5 heteromers undergo further inhibition in PLCß activation and calcium/protein kinase C (PKC) signaling. Nevertheless, the key factors differ. For TRPC1/4, the inactivation process was facilitated by Ca2+ release from the endoplasmic reticulum, and for TRPC1/5, activation of PKC was concerned mostly. We conclude that the subsequent increase in cytoplasmic Ca2+ due to Ca2+ release from the endoplasmic reticulum and activation of PKC resulted in a second phase of channel inhibition following PI(4,5)P2 depletion.

7.
Korean J Physiol Pharmacol ; 23(3): 191-201, 2019 May.
Article in English | MEDLINE | ID: mdl-31080350

ABSTRACT

The transient receptor potential canonical (TRPC) 5 channel, known as a nonselective cation channel, has a crucial role in calcium influx. TRPC5 has been reported to be activated by muscarinic receptor activation and extracellular pH change and inhibited by the protein kinase C pathway. Recent studies have also suggested that TRPC5 is extracellularly activated by englerin A (EA), but the mechanism remains unclear. The purpose of this study is to identify the EA-interaction sites in TRPC5 and thereby clarify the mechanism of TRPC5 activation. TRPC5 channels are over-expressed in human embryonic kidney (HEK293) cells. TRPC5 mutants were generated by site-directed mutagenesis. The whole-cell patch-clamp configuration was used to record TRPC5 currents. Western analysis was also performed to observe the expression of TRPC5 mutants. To identify the EA-interaction site in TRPC5, we first generated pore mutants. When screening the mutants with EA, we observed the EA-induced current increases of TRPC5 abolished in K554N, H594N, and E598Q mutants. The current increases of other mutants were reduced in different levels. We also examined the functional intactness of the mutants that had no effect by EA with TRPC5 agonists, such as carbachol or GTPγS. Our results suggest that the three residues, Lys-554, His-594, and Glu-598, in TRPC5 might be responsible for direct interaction with EA, inducing the channel activation. We also suggest that although other pore residues are not critical, they could partly contribute to the EA-induced channel activation.

8.
Biochem Biophys Res Commun ; 483(1): 129-134, 2017 01 29.
Article in English | MEDLINE | ID: mdl-28039055

ABSTRACT

Aberrant transforming growth factor ß1 (TGFß1) signaling plays a crucial role in the pathogenesis of vascular fibrosis. On the other hand, deregulated transient receptor potential canonical 6 (TRPC6) channel expression shows impaired vascular physiology and wound healing. However, it has little been known about the functional association between TGFß1 and TRPC6 in vascular smooth muscle cells (VSMCs). In this study, we analyzed the microarray data obtained from TGFß1-treated A7r5 VSMCs. We found that TGFß1 specifically elevates the expression level of TRPC6 mainly through Smad-dependent canonical pathway. The siRNA against TRPC6 abolished TGFß1-induced molecular and cellular phenotype changes, including myosin light chain phosphorylation, actin stress fiber formation, and cell migration. These results demonstrate that TRPC6 is an important component of TGFß1 signaling pathway in VSMCs. Therefore, our findings provide a basis for future investigation aimed at developing novel therapeutic strategies for treatment of vascular fibrosis.


Subject(s)
Muscle, Smooth, Vascular/metabolism , Stress Fibers/metabolism , TRPC Cation Channels/metabolism , Transforming Growth Factor beta1/metabolism , Actins/metabolism , Animals , Cell Line , Cell Movement , Fibrosis , Muscle, Smooth, Vascular/pathology , Myocytes, Smooth Muscle/metabolism , Myocytes, Smooth Muscle/pathology , Myosin Light Chains/metabolism , Phosphorylation , RNA, Small Interfering/genetics , Rats , Signal Transduction , Smad Proteins/metabolism , Stress Fibers/pathology , TRPC Cation Channels/antagonists & inhibitors , TRPC Cation Channels/genetics , Up-Regulation
9.
Biochim Biophys Acta ; 1853(3): 619-31, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25549939

ABSTRACT

Cystamine and its reduced form cysteamine showed protective effects in various models of neurodegenerative disease, including Huntington's disease and Parkinson's disease. Other lines of evidence demonstrated the cytotoxic effect of cysteamine on duodenal mucosa leading to ulcer development. However, the mechanism for cystamine cytotoxicity remains poorly understood. Here, we report a new pathway in which cystamine induces apoptosis by targeting apoptosis-inducing factor (AIF). By screening of various cell lines, we observed that cystamine and cysteamine induce cell death in a cell type-specific manner. Comparison between cystamine-sensitive and cystamine-resistant cell lines revealed that cystamine cytotoxicity is not associated with unfolded protein response, reactive oxygen species generation and transglutaminase or caspase activity; rather, it is associated with the ability of cystamine to trigger AIF nuclear translocation. In cystamine-sensitive cells, cystamine suppresses the levels of intracellular glutathione by inhibiting γ-glutamylcysteine synthetase expression that triggers AIF translocation. Conversely, glutathione supplementation completely prevents cystamine-induced AIF translocation and apoptosis. In rats, cysteamine administration induces glutathione depletion and AIF translocation leading to apoptosis of duodenal epithelium. These results indicate that AIF translocation through glutathione depletion is the molecular mechanism of cystamine toxicity, and provide important implications for cystamine in the neurodegenerative disease therapeutics as well as in the regulation of AIF-mediated cell death.


Subject(s)
Apoptosis Inducing Factor/physiology , Apoptosis/drug effects , Cystamine/pharmacology , Glutathione/metabolism , Animals , Apoptosis/genetics , Duodenal Ulcer/metabolism , Duodenal Ulcer/pathology , Female , HeLa Cells , Humans , MCF-7 Cells , Rats , Rats, Sprague-Dawley , Reactive Oxygen Species/metabolism , Tumor Cells, Cultured , Up-Regulation/drug effects
10.
Pflugers Arch ; 468(4): 551-61, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26631167

ABSTRACT

Transient receptor potential canonical (TRPC) 4 channels are calcium-permeable, nonselective cation channels and are widely expressed in mammalian tissue, especially in the GI tract and brain. TRPC4 channels are known to be involved in neurogenic contraction of ileal smooth muscle cells via generating cationic current after muscarinic stimulation (muscarinic cationic current (mIcat)). Polyamines exist in numerous tissues and are believed to be involved in cell proliferation, differentiation, scar formation, wound healing, and carcinogenesis. Besides, physiological polyamines are essential to maintain inward rectification of cardiac potassium channels (Kir2.1). At membrane potentials more positive than equilibrium potential, intracellular polyamines plug the cytosolic surface of the Kir2.1 so that potassium ions cannot pass through the pore. Recently, it was reported that polyamines inhibit not only cardiac potassium channels but also nonselective cation channels that mediate the generation of mIcat. Here, we report that TRPC4, a definite mIcat mediator, is inhibited by intracellular spermine with great extent. The inhibition was specific to TRPC4 and TRPC5 channels but was not effective to TRPC1/4, TRPC1/5, and TRPC3 channels. For this inhibition to occur, we found that glutamates at 728th and 729th position of TRPC4 channels are essential whereby we conclude that spermine blocks the TRPC4 channel with electrostatic interaction between negative amino acids at the C-terminus of the channel.


Subject(s)
Spermine/metabolism , TRPC Cation Channels/metabolism , Action Potentials , Animals , Binding Sites , Glutamic Acid/chemistry , Glutamic Acid/metabolism , HEK293 Cells , Humans , Mice , Potassium Channels, Inwardly Rectifying/metabolism , Protein Binding , Static Electricity , TRPC Cation Channels/chemistry
11.
Biochem Biophys Res Commun ; 474(3): 476-481, 2016 06 03.
Article in English | MEDLINE | ID: mdl-27131740

ABSTRACT

Transient receptor potential canonical (TRPC) family contains a non-selective cation channel, and four TRPC subunits form a functional tetrameric channel. TRPC4/5 channels form not only the homotetrameric channel but also a heterotetrameric channel with TRPC1. We investigated the interaction domain required for TRPC1/4 or TRPC1/5 heteromultimeric channels using FRET and the patch-clamp technique. TRPC1 only localized at the plasma membrane (PM) when it was coexpressed with TRPC4 or TRPC5. The TRPC1/4 or TRPC1/5 heteromultimeric showed the typical outward rectifying I/V curve. When TRPC1 and TRPC4 form a heteromeric channel, the N-terminal coiled-coil domain (CCD) and C-terminal 725-745 region of TRPC1 interact with the N-terminal CCD and C-terminal 700-728 region of TRPC4. However, when TRPC1 and TRPC5 form a heteromeric channel, the N-terminal CCD and C-terminal 673-725 region of TRPC1 interact with the N-terminal CCD and C-terminal 707-735 region of TRPC5. In conclusion, the N-terminal CCD of TRPC channels is essential for the heteromultimeric structure of TRPC channels, whereas specific C-terminal regions are required for unique heteromerization between subgroups of TRPC channels.


Subject(s)
TRPC Cation Channels/chemistry , TRPC Cation Channels/metabolism , Binding Sites , Protein Binding , Protein Domains , Protein Interaction Mapping/methods , Protein Multimerization/physiology
12.
Protein Expr Purif ; 120: 153-9, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26767484

ABSTRACT

The Ras superfamily of small G proteins is a family of guanosine triphosphatases (GTPases) and each GTPase has conserved amino acid sequences in the enzymatic active site that are responsible for specific interactions with GDP and GTP molecules. Rab GTPases, which belong to the Ras superfamily, are key regulators of intracellular vesicle trafficking via the recruitment of effector molecules. Here, we purified wild type, active mutant and inactive mutant of Rab11A. In this process, we found that the inactive mutant (Rab11A S25N) had low stability compared with wild type and other mutants. Further analysis revealed that the stability of Rab11A S25N is dependent on the occupation of GDP in the nucleotide binding pocket of the protein. We found that the stability of Rab11A S25N is affected by the presence of GDP, not other nucleotides, and is independent of pH or salt in FPLC buffer. Our results provide a better understanding of how GTPase can be stable under in vitro conditions without effector proteins and how proper substrate/cofactor coordination is crucial to the stability of Rab11A. Successful purification and proposed purification methods will provide a valuable guide for investigation of other small GTPase proteins.


Subject(s)
Catalytic Domain , Guanosine Diphosphate/metabolism , rab GTP-Binding Proteins/isolation & purification , Humans , Mutation , Protein Stability , Protein Structure, Tertiary , Recombinant Proteins/isolation & purification , rab GTP-Binding Proteins/genetics , rab GTP-Binding Proteins/metabolism
13.
Brain ; 138(Pt 10): 3030-47, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26133660

ABSTRACT

Aberrant glutathione or Ca(2+) homeostasis due to oxidative stress is associated with the pathogenesis of neurodegenerative disorders. The Ca(2+)-permeable transient receptor potential cation (TRPC) channel is predominantly expressed in the brain, which is sensitive to oxidative stress. However, the role of the TRPC channel in neurodegeneration is not known. Here, we report a mechanism of TRPC5 activation by oxidants and the effect of glutathionylated TRPC5 on striatal neurons in Huntington's disease. Intracellular oxidized glutathione leads to TRPC5 activation via TRPC5 S-glutathionylation at Cys176/Cys178 residues. The oxidized glutathione-activated TRPC5-like current results in a sustained increase in cytosolic Ca(2+), activated calmodulin-dependent protein kinase and the calpain-caspase pathway, ultimately inducing striatal neuronal cell death. We observed an abnormal glutathione pool indicative of an oxidized state in the striatum of Huntington's disease transgenic (YAC128) mice. Increased levels of endogenous TRPC5 S-glutathionylation were observed in the striatum in both transgenic mice and patients with Huntington's disease. Both knockdown and inhibition of TRPC5 significantly attenuated oxidation-induced striatal neuronal cell death. Moreover, a TRPC5 blocker improved rearing behaviour in Huntington's disease transgenic mice and motor behavioural symptoms in littermate control mice by increasing striatal neuron survival. Notably, low levels of TRPC1 increased the formation of TRPC5 homotetramer, a highly Ca(2+)-permeable channel, and stimulated Ca(2+)-dependent apoptosis in Huntington's disease cells (STHdh(Q111/111)). Taken together, these novel findings indicate that increased TRPC5 S-glutathionylation by oxidative stress and decreased TRPC1 expression contribute to neuronal damage in the striatum and may underlie neurodegeneration in Huntington's disease.


Subject(s)
Corpus Striatum/pathology , Glutathione/metabolism , Green Fluorescent Proteins/metabolism , Huntington Disease/pathology , Neurons/metabolism , TRPC Cation Channels/metabolism , Analysis of Variance , Animals , Calcium/metabolism , Green Fluorescent Proteins/genetics , HEK293 Cells , Humans , Huntingtin Protein , Mice , Mice, Transgenic , Mutation/genetics , Nerve Tissue Proteins/genetics , RNA, Small Interfering/metabolism , TRPC Cation Channels/genetics , Transfection
14.
Proc Natl Acad Sci U S A ; 110(31): 12673-8, 2013 Jul 30.
Article in English | MEDLINE | ID: mdl-23858470

ABSTRACT

Leptin is a pivotal regulator of energy and glucose homeostasis, and defects in leptin signaling result in obesity and diabetes. The ATP-sensitive potassium (K(ATP)) channels couple glucose metabolism to insulin secretion in pancreatic ß-cells. In this study, we provide evidence that leptin modulates pancreatic ß-cell functions by promoting K(ATP) channel translocation to the plasma membrane via AMP-activated protein kinase (AMPK) signaling. K(ATP) channels were localized mostly to intracellular compartments of pancreatic ß-cells in the fed state and translocated to the plasma membrane in the fasted state. This process was defective in leptin-deficient ob/ob mice, but restored by leptin treatment. We discovered that the molecular mechanism of leptin-induced AMPK activation involves canonical transient receptor potential 4 and calcium/calmodulin-dependent protein kinase kinase ß. AMPK activation was dependent on both leptin and glucose concentrations, so at optimal concentrations of leptin, AMPK was activated sufficiently to induce K(ATP) channel trafficking and hyperpolarization of pancreatic ß-cells in a physiological range of fasting glucose levels. There was a close correlation between phospho-AMPK levels and ß-cell membrane potentials, suggesting that AMPK-dependent K(ATP) channel trafficking is a key mechanism for regulating ß-cell membrane potentials. Our results present a signaling pathway whereby leptin regulates glucose homeostasis by modulating ß-cell excitability.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Insulin-Secreting Cells/metabolism , Leptin/metabolism , Membrane Potentials/physiology , Signal Transduction/physiology , Sodium-Potassium-Exchanging ATPase/metabolism , AMP-Activated Protein Kinases/genetics , Animals , Calcium-Calmodulin-Dependent Protein Kinase Type 2/genetics , Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Glucose/metabolism , Homeostasis/physiology , Insulin-Secreting Cells/cytology , Leptin/genetics , Mice , Mice, Obese , Protein Transport/physiology , Sodium-Potassium-Exchanging ATPase/genetics , TRPC Cation Channels/genetics , TRPC Cation Channels/metabolism
15.
J Korean Med Sci ; 31(6): 829-35, 2016 Jun.
Article in English | MEDLINE | ID: mdl-27247489

ABSTRACT

The authors conducted a survey on essential humanistic competency that medical students should have, and on teaching methods that will effectively develop such attributes. The participants consisted of 154 medical school professors, 589 medical students at Seoul National University College of Medicine, 228 parents, and 161 medical school and university hospital staff. They answered nine questions that the authors created. According to the results, all groups chose "morality and a sense of ethics," a "sense of accountability," "communication skills," and "empathic ability" were selected as essential qualities. According to the evaluation on the extent to which students possess each quality, participants believed students had a high "sense of accountability" and "morality," whereas they thought students had low "empathic ability," "communicate," or "collaborate with others". In terms of effective teaching methods, all sub-groups preferred extracurricular activities including small group activities, debates, and volunteer services. With regard to the speculated effect of humanism education and the awareness of the need for colleges to offer it, all sub-groups had a positive response. However the professors and students expressed a relatively passive stance on introducing humanism education as a credited course. Most participants responded that they preferred a grading method based on their rate of participation, not a relative evaluation. In order to reap more comprehensive and lasting effects of humanism education courses in medical school, it is necessary to conduct faculty training, and continuously strive to develop new teaching methods.


Subject(s)
Health Personnel/psychology , Humanism , Students, Medical/psychology , Adult , Curriculum , Female , Humans , Male , Middle Aged , Parents/psychology , Surveys and Questionnaires , Young Adult
16.
Am J Physiol Cell Physiol ; 308(11): C879-89, 2015 Jun 01.
Article in English | MEDLINE | ID: mdl-25788576

ABSTRACT

TPRC channels are Ca(2+)-permeable, nonselective cation channels that are activated by a wide variety of stimuli, including G protein-coupled receptors (GPCRs). TRPC4 is commonly assumed to be activated by Gq/phospholipase C-coupled receptors. However, the other molecular mechanisms by which Gα proteins regulate TRPC4 remain unclear. Here, we found that Gαi2 regulates TRPC4 activation by direct binding. To investigate this mechanism, we used whole patch clamp and fluorescence resonance energy transfer (FRET). We tagged an isoform of mTRPC4 and G protein with CFP and YFP, respectively, and transiently transfected cells with the FRET pair. The FRET efficiency between TRPC4ß-CFP and the constitutively active mutant form of Gαi2 was nearly 15% and was greater than that observed with wild-type Gαi2 (nearly 5%). Gßγ and the TRPC4 channel showed a FRET efficiency lower than 6%. In HEK293 cells transfected with the M2 muscarinic receptor, the application of carbachol increased the FRET efficiency between TRPC4ß-CFP and Gαi2(WT)-YFP from 4.7 ± 0.4% (n = 7) to 12.6 ± 1.4% (n = 7). We also found that the TRPC4 channel directly interacts with Gαi2, but not with Gαq, when the channel is open. We analyzed the calcium levels in HEK293 cells expressing the channels and Gαi2 or Gαq using the calcium indicator YC6.1 (Yellow Cameleon 6.1). In response to the muscarinic agonist carbachol, M2-, Gαi2-, and TRPC4-expressing cells showed a prolonged Ca(2+) influx compared with cells expressing only M2. Together, these data suggest that Gαi2 activates the TRPC4 channel by direct binding, which then induces Ca(2+) entry.


Subject(s)
Calcium Signaling/genetics , Calcium/metabolism , GTP-Binding Protein alpha Subunit, Gi2/genetics , GTP-Binding Protein alpha Subunits, Gq-G11/genetics , TRPC Cation Channels/genetics , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Calcium Signaling/drug effects , Calcium-Binding Proteins/chemistry , Carbachol/pharmacology , Cholinergic Agonists/pharmacology , Fluorescence Resonance Energy Transfer , GTP-Binding Protein alpha Subunit, Gi2/metabolism , GTP-Binding Protein alpha Subunits, Gq-G11/metabolism , Gene Expression Regulation , Genes, Reporter , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , HEK293 Cells , Humans , Luminescent Proteins/chemistry , Luminescent Proteins/genetics , Luminescent Proteins/metabolism , Protein Binding , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , TRPC Cation Channels/metabolism
17.
J Biol Chem ; 289(50): 34990-5002, 2014 Dec 12.
Article in English | MEDLINE | ID: mdl-25349210

ABSTRACT

Canonical transient receptor potential (TRPC) channels are Ca(2+)-permeable nonselective cation channels that are activated by a wide variety of stimuli, including G protein-coupled receptors (GPCRs). The TRPC4 channel is expressed in a punctate distribution in the membrane. To identify the regulating region of the channel trafficking to the membrane, we generated deletion mutants of the TRPC4 channel. We determined that when either region that was downstream of the 20 amino acids of the N terminus or the 700-730 amino acids was deleted, the mutants were retained in the endoplasmic reticulum. By coexpression of the wild-type TRPC4 with deletion mutants, we found that the 23-29 amino acids of the N terminus regulate a membrane trafficking. Additionally, by the fluorescence resonance energy transfer (FRET) method, we found that the regions downstream of the 99 amino acid region of the N terminus and upstream of the 730 amino acid region in the C terminus produce assembly of the TRPC4 tetramers. We inferred the candidate proteins that regulate or interact with the 23-29 domain of TRPC4.


Subject(s)
Cell Membrane/metabolism , Protein Multimerization , TRPC Cation Channels/chemistry , TRPC Cation Channels/metabolism , Amino Acid Sequence , Gene Expression Regulation , HEK293 Cells , Humans , Molecular Sequence Data , Protein Structure, Quaternary , Protein Structure, Tertiary , Protein Transport , Sequence Deletion , TRPC Cation Channels/genetics
18.
Pflugers Arch ; 467(10): 2081-91, 2015 Oct.
Article in English | MEDLINE | ID: mdl-25502319

ABSTRACT

Canonical transient receptor potential 4 (TRPC4) channels are calcium-permeable, nonselective cation channels that are widely distributed in mammalian cells. It is generally speculated that TRPC4 channels are activated by Gq/11-PLC pathway or directly activated by Gi/o proteins. Although many mechanistic studies regarding TRPC4 have dealt with heterotrimeric G proteins, here, we first report the functional relationship between TRPC4 and small GTPase, Rasd1. Rasd1 selectively activated TRPC4 channels, and it was the only Ras protein among Ras protein family that can activate TRPC4 channels. For this to occur, it was found that certain population of functional Gαi1 and Gαi3 proteins are essential. Meanwhile, dexamethasone, a synthetic glucocorticoid and anti-inflammatory drug was known to increase messenger RNA (mRNA) level of Rasd1 in pancreatic ß-cells. We have found that dexamethasone triggers TRPC4-like cationic current in INS-1 cells via increasing protein expression level of Rasd1. This relationship among dexamethasone, Rasd1, and TRPC4 could suggest a new therapeutic agent for hospitalized diabetes mellitus (DM) patients with prolonged dexamethasone prescription.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Dexamethasone/pharmacology , TRPC Cation Channels/metabolism , ras Proteins/metabolism , Animals , GTP-Binding Protein alpha Subunits, Gi-Go/metabolism , HEK293 Cells , Humans , Insulin-Secreting Cells/drug effects , Insulin-Secreting Cells/metabolism , Rats , ras Proteins/genetics
19.
Pflugers Arch ; 467(4): 703-12, 2015 Apr.
Article in English | MEDLINE | ID: mdl-24859801

ABSTRACT

Crucial cysteine residues can be involved in the modulation of protein activity via the modification of thiol (-SH) groups. Among these reactions, disulfide bonds (S-S) play a key role in the folding, stability, and activity of membrane proteins. However, the regulation of extracellular cysteines in classical transient receptor potential (TRPC) channels remains controversial. Here, we examine the functional importance of the extracellular disulfide bond in TRPC5 in modulating channel gating and trafficking. Specifically, we investigated TRPC5 activity in transiently transfected HEK293 cells with wild-type (WT) or cysteine (C553 and C558) mutants in the pore loop. Using reducing agents, we determined that a disulfide linkage mediates the tetrameric formation of the TRPC5 channel. By measuring the TRPC5 current, we observed that C553S or C558S mutants completely lose channel activity induced by lanthanides or receptor stimulation. Co-expression of TRPC5 (WT) with mutants demonstrated a dominant-negative function in mutants, which inhibited the activity of TRPC5 (WT). We generated TRPC5-TRPC5 dimers and observed reduced activity of WT-mutant (C553S or C558S) dimers compared to WT-WT dimers. When pretreated with reducing agents for 12 h, the TRPC5 current decreased due to a reduction in membrane TRPC5 distribution. In addition, we identified a reduced expression of C553S mutant in plasma membrane. We analyzed a dimeric interaction of wild-type and mutant TRPC5 using co-immunoprecipitation and FRET method, indicating a weak interaction between dimeric partners. These results indicated that the disulfide bond between conserved extracellular cysteines, especially C553, is essential for functional TRPC5 activity by channel multimerization and trafficking.


Subject(s)
Cystine/chemistry , Protein Multimerization , TRPC Cation Channels/chemistry , Animals , HEK293 Cells , Humans , Mice , Protein Stability , Protein Structure, Tertiary , Protein Transport , TRPC Cation Channels/metabolism
20.
Basic Res Cardiol ; 110(3): 21, 2015 May.
Article in English | MEDLINE | ID: mdl-25804308

ABSTRACT

Angiotensin II (Ang II), a potent precursor of hypertrophy and heart failure, upregulates neuronal nitric oxide synthase (nNOS or NOS1) in the myocardium. Here, we investigate the involvement of type 1 and 2 angiotensin receptors (AT1R and AT2R) and molecular mechanisms mediating Ang II-upregulation of nNOS. Our results showed that pre-treatment of left ventricular (LV) myocytes with antagonists of AT1R or AT2R (losartan, PD123319) and ROS scavengers (apocynin, tiron or PEG-catalase) blocked Ang II-upregulation of nNOS. Surface biotinylation or immunocytochemistry experiments demonstrated that AT1R expression in plasma membrane was progressively decreased (internalization), whereas AT2R was increased (membrane trafficking) by Ang II. Inhibition of AT1R or ROS scavengers prevented Ang II-induced translocation of AT2R to plasma membrane, suggesting an alignment of AT1R-ROS-AT2R. Furthermore, Ang II increased eNOS-Ser(1177) but decreased eNOS-Thr(495), indicating concomitant activation of eNOS. Intriguingly, ROS scavengers but not AT2R antagonist prevented Ang II-activation of eNOS. NOS inhibitor (L-NG-Nitroarginine Methyl Ester, L-NAME) or eNOS gene deletion (eNOS(-/-)) abolished Ang II-induced membrane trafficking of AT2R, nNOS protein expression and activity. Mechanistically, S-nitrosation of AT2R was increased by sodium nitroprusside (SNP), a NO donor. Site-specific mutagenesis analysis reveals that C-terminal cysteine 349 in AT2R is essential in AT2R translocation to plasma membrane. Taken together, we demonstrate, for the first time, that Ang II upregulates nNOS protein expression and activity via AT1R/ROS/eNOS-dependent S-nitrosation and membrane translocation of AT2R. Our results suggest a novel crosstalk between AT1R and AT2R in regulating nNOS via eNOS in the myocardium under pathogenic stimuli.


Subject(s)
Angiotensin II/metabolism , Myocytes, Cardiac/metabolism , Nitric Oxide Synthase Type III/metabolism , Nitric Oxide Synthase Type I/metabolism , Reactive Oxygen Species/metabolism , Receptor, Angiotensin, Type 2/metabolism , Animals , HEK293 Cells , Humans , Immunoblotting , Immunohistochemistry , Male , Microscopy, Confocal , Protein Transport/physiology , Rats , Rats, Sprague-Dawley , Receptor Cross-Talk , Reverse Transcriptase Polymerase Chain Reaction , Transfection
SELECTION OF CITATIONS
SEARCH DETAIL