Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 25
Filter
Add more filters










Publication year range
1.
Mol Brain ; 14(1): 144, 2021 09 20.
Article in English | MEDLINE | ID: mdl-34544455

ABSTRACT

Astrocytes express a plethora of G protein-coupled receptors (GPCRs) that are crucial for shaping synaptic activity. Upon GPCR activation, astrocytes can respond with transient variations in intracellular Ca2+. In addition, Ca2+-dependent and/or Ca2+-independent release of gliotransmitters can occur, allowing them to engage in bidirectional neuron-astrocyte communication. The development of designer receptors exclusively activated by designer drugs (DREADDs) has facilitated many new discoveries on the roles of astrocytes in both physiological and pathological conditions. They are an excellent tool, as they can target endogenous GPCR-mediated intracellular signal transduction pathways specifically in astrocytes. With increasing interest and accumulating research on this topic, several discrepancies on astrocytic Ca2+ signalling and astrocyte-mediated effects on synaptic plasticity have emerged, preventing a clear-cut consensus about the downstream effects of DREADDs in astrocytes. In the present study, we performed a side-by-side evaluation of the effects of bath application of the DREADD agonist, clozapine-N-oxide (10 µM), on Gq- and Gi-DREADD activation in mouse CA1 hippocampal astrocytes. In doing so, we aimed to avoid confounding factors, such as differences in experimental procedures, and to directly compare the actions of both DREADDs on astrocytic intracellular Ca2+ dynamics and synaptic plasticity in acute hippocampal slices. We used an adeno-associated viral vector approach to transduce dorsal hippocampi of male, 8-week-old C57BL6/J mice, to drive expression of either the Gq-DREADD or Gi-DREADD in CA1 astrocytes. A viral vector lacking the DREADD construct was used to generate controls. Here, we show that agonism of Gq-DREADDs, but not Gi-DREADDs, induced consistent increases in spontaneous astrocytic Ca2+ events. Moreover, we demonstrate that both Gq-DREADD as well as Gi-DREADD-mediated activation of CA1 astrocytes induces long-lasting synaptic potentiation in the hippocampal CA1 Schaffer collateral pathway in the absence of a high frequency stimulus. Moreover, we report for the first time that astrocytic Gi-DREADD activation is sufficient to elicit de novo potentiation. Our data demonstrate that activation of either Gq or Gi pathways drives synaptic potentiation through Ca2+-dependent and Ca2+-independent mechanisms, respectively.


Subject(s)
Astrocytes/physiology , CA1 Region, Hippocampal/physiology , Calcium Signaling/physiology , Clozapine/analogs & derivatives , Designer Drugs/pharmacology , GTP-Binding Protein alpha Subunits, Gi-Go/physiology , GTP-Binding Protein alpha Subunits, Gq-G11/physiology , Receptors, G-Protein-Coupled/physiology , Animals , Astrocytes/drug effects , CA1 Region, Hippocampal/cytology , Clozapine/pharmacology , GTP-Binding Protein alpha Subunits, Gi-Go/drug effects , GTP-Binding Protein alpha Subunits, Gq-G11/drug effects , Genetic Vectors/pharmacology , Long-Term Potentiation/drug effects , Long-Term Potentiation/physiology , Male , Mice , Mice, Inbred C57BL , Neuronal Plasticity/physiology , Receptors, G-Protein-Coupled/drug effects
2.
J Pharm Pharmacol ; 70(2): 214-222, 2018 Feb.
Article in English | MEDLINE | ID: mdl-29148061

ABSTRACT

OBJECTIVES: The endocannabinoid (eCB) system centrally and peripherally regulates cardiovascular parameters, including blood pressure, in health and disease. The relationship between Gq/11 protein-coupled receptor activation, regulation of eCBs release (mainly 2-arachidonoylglycerol) and subsequent CB1 receptor activation was initially observed in the central nervous system. Here, we review the latest findings from systemic physiological studies which include for the first time data from pulmonary arteries. We present evidence for direct CB1 -dependent cannabinoid ligand-induced vasorelaxation, vascular expression of eCBs along with their degradation enzymes, and indicate the location of the described interaction. KEY FINDINGS: Endocannabinoids (mainly 2-arachidonoylglycerol), acting via CB1 receptors, evoke vasodilatory effects and may modulate responses of vasoconstrictors for Gq/11 protein-coupled receptors including angiotensin II, thromboxane A2 , phenylephrine, noradrenaline in systemic or pulmonary arteries. However, the role of the endothelium in this interaction is not well-established, and the precise vascular location of eCB system components remains unclear, which contributes to discrepancies in the interpretation of results when describing the above-mentioned relationship. SUMMARY: Endocannabinoid's negative feedback is responsible for diminishing agonist-induced vasoconstriction, which may be clinically important in the treatment of arterial and pulmonary hypertension. Further research is required to establish the importance of the eCB system and its downstream signalling pathways.


Subject(s)
Arteries/drug effects , Endocannabinoids/metabolism , GTP-Binding Protein alpha Subunits, Gq-G11/metabolism , Receptor, Cannabinoid, CB1/metabolism , Vasoconstriction , Vasodilation , Angiotensin II/metabolism , Animals , Antihypertensive Agents/pharmacology , Arteries/metabolism , Dinoprost/metabolism , Feedback, Physiological , GTP-Binding Protein alpha Subunits, Gq-G11/drug effects , Humans , Ligands , Receptor Cross-Talk , Receptor, Cannabinoid, CB1/drug effects , Signal Transduction , Thromboxane A2/metabolism , Vasoconstriction/drug effects , Vasoconstrictor Agents/pharmacology , Vasodilation/drug effects , Vasodilator Agents/pharmacology
3.
JCI Insight ; 2(20)2017 10 19.
Article in English | MEDLINE | ID: mdl-29046478

ABSTRACT

Loss-of-function mutations of GNA11, which encodes G-protein subunit α11 (Gα11), a signaling partner for the calcium-sensing receptor (CaSR), result in familial hypocalciuric hypercalcemia type 2 (FHH2). FHH2 is characterized by hypercalcemia, inappropriately normal or raised parathyroid hormone (PTH) concentrations, and normal or low urinary calcium excretion. A mouse model for FHH2 that would facilitate investigations of the in vivo role of Gα11 and the evaluation of calcimimetic drugs, which are CaSR allosteric activators, is not available. We therefore screened DNA from > 10,000 mice treated with the chemical mutagen N-ethyl-N-nitrosourea (ENU) for GNA11 mutations and identified a Gα11 variant, Asp195Gly (D195G), which downregulated CaSR-mediated intracellular calcium signaling in vitro, consistent with it being a loss-of-function mutation. Treatment with the calcimimetic cinacalcet rectified these signaling responses. In vivo studies showed mutant heterozygous (Gna11+/195G) and homozygous (Gna11195G/195G) mice to be hypercalcemic with normal or increased plasma PTH concentrations and normal urinary calcium excretion. Cinacalcet (30mg/kg orally) significantly reduced plasma albumin-adjusted calcium and PTH concentrations in Gna11+/195G and Gna11195G/195G mice. Thus, our studies have established a mouse model with a germline loss-of-function Gα11 mutation that is representative for FHH2 in humans and demonstrated that cinacalcet can correct the associated abnormalities of plasma calcium and PTH.


Subject(s)
Cinacalcet/therapeutic use , GTP-Binding Protein alpha Subunits, Gq-G11/drug effects , GTP-Binding Protein alpha Subunits, Gq-G11/genetics , Hypercalcemia/drug therapy , Mutation/drug effects , Administration, Oral , Animals , Calcium/blood , Calcium/urine , Cinacalcet/administration & dosage , Disease Models, Animal , Ethylnitrosourea/pharmacology , Female , GTP-Binding Protein alpha Subunits, Gq-G11/chemistry , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Mutant Strains , Models, Molecular , Parathyroid Hormone/blood , Parathyroid Hormone/metabolism , Receptors, Calcium-Sensing/metabolism , Sequence Alignment , Sequence Analysis, DNA , Serum Albumin , Signal Transduction
4.
Am J Physiol Endocrinol Metab ; 312(3): E183-E189, 2017 03 01.
Article in English | MEDLINE | ID: mdl-28049625

ABSTRACT

It has been demonstrated that the neuropeptide oxytocin (OT) attenuates oxidative stress and inflammation in macrophages. In the current study, we examined the role of inflammation on the expression of the oxytocin receptor (OXTR). We hypothesized that OXTR expression is increased during the inflammation through a nuclear factor-κB (NF-κB)-mediated pathway, thus responding as an acute-phase protein. Inflammation was induced by treating macrophages (human primary, THP-1, and murine) with lipopolysaccharide (LPS) and monitored by expression of IL-6. Expression of OXTR and vasopressin receptors was assessed by qPCR, and OXTR expression was confirmed by immunoblotting. Inflammation upregulated OXTR transcription 10- to 250-fold relative to control in THP-1 and human primary macrophages and increased OXTR protein expression. In contrast, vasopressin receptor-2 mRNA expression was reduced following LPS treatment. Blocking NF-κB activation prevented the increase in OXTR transcription. OT treatment of control cells and LPS-treated cells increased ERK1/2 phosphorylation, demonstrating activation of the OXTR/Gαq/11 signaling pathway. OT activation of OXTR reduced secretion of IL-6 in LPS-activated macrophages. Collectively, these findings suggest that OXTR is an acute-phase protein and that its increased expression is regulated by NF-κB and functions to attenuate cellular inflammatory responses in macrophages.


Subject(s)
Macrophages/metabolism , Receptors, Oxytocin/genetics , Animals , Blotting, Western , GTP-Binding Protein alpha Subunits, Gq-G11/drug effects , GTP-Binding Protein alpha Subunits, Gq-G11/metabolism , Gene Expression Regulation/drug effects , Humans , Interleukin-6/genetics , Interleukin-6/metabolism , Lipopolysaccharides/pharmacology , Macrophages/drug effects , Macrophages, Peritoneal , Male , Mice , Mitogen-Activated Protein Kinase 1/drug effects , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/drug effects , Mitogen-Activated Protein Kinase 3/metabolism , NF-kappa B/drug effects , NF-kappa B/metabolism , Oxytocics/pharmacology , Oxytocin/pharmacology , Phosphorylation/drug effects , Polymerase Chain Reaction , Real-Time Polymerase Chain Reaction , Receptors, Oxytocin/drug effects , Receptors, Vasopressin/drug effects , Receptors, Vasopressin/genetics
5.
Life Sci ; 152: 99-106, 2016 May 01.
Article in English | MEDLINE | ID: mdl-27012764

ABSTRACT

Gαq signals with phospholipase C-ß (PLC-ß) to modify behavior in response to an agonist-bound GPCR. While the fundamental steps which prime Gαq to interact with PLC-ß have been identified, questions remain concerning signal strength with PLC-ß and other effectors. Gαq is generally viewed to function as a simple ON and OFF switch for its effector, dependent on the binding of GTP or GDP. However, Gαq does not have a single effector, Gαq has many different effectors. Furthermore, select effectors also regulate Gαq activity. PLC-ß is a lipase and a GTPase activating protein (GAP) selective for Gαq. The contribution of G protein regulating activity to signal amplitude remains unclear. The unique PLC-ß coiled-coil domain is essential for maximum Gαq response, both lipase and GAP. Nonetheless, coiled-coil domain associations necessary to maximum response have not been revealed by the structural approach. This review discusses progress towards understanding the basis for signal strength with PLC-ß and other effectors. Shared and effector-specific interactions have been identified. Finally, the evidence for allosteric regulation of lipase stimulation by protein kinase C, the membrane, phosphatidic acid, phosphatidylinositol-4, 5-bisphosphate and GPCR is explored. Endogenous allosteric regulators can suppress or enhance maximum lipase stimulation dependent on the PLC-ß coiled-coil domain. A better understanding of allosteric modulation may therefore identify a wealth of new targets to regulate signal strength and behavior.


Subject(s)
GTP-Binding Protein alpha Subunits, Gq-G11/physiology , Signal Transduction/physiology , Animals , GTP-Binding Protein alpha Subunits, Gq-G11/drug effects , Humans , Phospholipase C beta/metabolism , Receptors, G-Protein-Coupled/agonists , Signal Transduction/drug effects
6.
J Pharmacol Sci ; 127(1): 127-34, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25704028

ABSTRACT

The human ether-a-go-go-related gene (HERG) potassium current (IHERG) has been shown to decrease in amplitude following stimulation with Gq protein-coupled receptors (GqRs), such as α1-adrenergic and M1-muscarinic receptors (α1R and M1R, respectively), at least partly via the reduction of membrane phosphatidylinositol 4,5-bisphosphate (PI(4,5)P2). The present study was designed to investigate the modulation of HERG channels by PI(4,5)P2 and phosphatidylinositol4-phosphate 5-kinase (PI(4)P5-K), a synthetic enzyme of PI(4,5)P2. Whole-cell patch-clamp recordings were used to examine the activity of HERG channels expressed heterologously in Chinese Hamster Ovary cells. The stimulation of α1R with phenylephrine or M1R with acetylcholine decreased the amplitude of IHERG accompanied by a significant acceleration of deactivation kinetics and the effects on IHERG were significantly attenuated in cells expressing PI(4)P5-K. The density of IHERG in cells expressing GqRs alone was significantly increased by the coexpression of PI(4)P5-K without significant differences in the voltage dependence of activation and deactivation kinetics. The kinase-deficient substitution mutant, PI(4)P5-K-K138A did not have these counteracting effects on the change in IHERG by M1R stimulation. These results suggest that the current density of IHERG is closely dependent on the membrane PI(4,5)P2 level, which is regulated by PI(4)P5-K and GqRs and that replenishing PI(4,5)P2 by PI(4)P5-K recovers IHERG.


Subject(s)
Ether-A-Go-Go Potassium Channels/drug effects , Phosphotransferases (Alcohol Group Acceptor)/physiology , Acetylcholine/pharmacology , Animals , CHO Cells , Cricetinae , Cricetulus , Ether-A-Go-Go Potassium Channels/physiology , GTP-Binding Protein alpha Subunits, Gq-G11/drug effects , Humans , Membrane Potentials/drug effects , Membrane Potentials/physiology , Mutation , Phenylephrine/pharmacology , Phosphatidylinositol 4,5-Diphosphate/physiology , Phosphotransferases (Alcohol Group Acceptor)/genetics , Transfection
7.
Acta Pharmacol Sin ; 33(5): 701-9, 2012 May.
Article in English | MEDLINE | ID: mdl-22555372

ABSTRACT

AIM: To investigate the effects of (-)-epigallocatechin-3-gallate (EGCG), an active compound in green tea, on prostaglandin E(2) (PGE(2))-induced proliferation and migration, and the expression of prostanoid EP(1) receptors in hepatocellular carcinoma (HCC) cells. METHODS: HCC cell line HepG2, human hepatoma cell lines MHCC-97L, MHCC-97H and human hepatocyte cell line L02 were used. Cell viability was analyzed using MTT assay. PGE(2) production was determined with immunoassay. Wound healing assay and transwell filter assay were employed to assess the extent of HCC cell migration. The expression of EP(1) receptor and Gq protein were examined using Western blot assay. RESULTS: PGE(2) (4-40000 nmol/L) or the EP(1) receptor agonist ONO-DI-004 (400-4000 nmol/L) increased the viability and migration of HepG2 cells in concentration-dependent manners. EGCG (100 µg/mL) significantly inhibited the viability and migration of HepG2 cells induced by PGE(2) or ONO-DI-004. HepG2 cells secreted an abundant amount of PGE(2) into the medium, and EGCG (100 µg/mL) significantly inhibited the PGE(2)production and EP(1) receptor expression in HepG2 cells. EGCG (100 µg/mL) also inhibited the viability of MHCC-97L cells, but not that of MHCC-97H cells. Both EGCG (100 µg/mL) and EP(1) receptor antagonist ONO-8711 inhibited PGE(2) 4 µmol/L and ONO-DI-004 400 nmol/L-induced growth and migration of HepG2 cells. Both EGCG (100 µg/mL) and ONO-8711 210 nmol/L inhibited PGE(2)- and ONO-DI-004-induced EP(1) expression. EGCG and ONO-8711 had synergistic effects in inhibiting EP(1) receptor expression. PGE(2), ONO-DI-004, ONO-8711, and EGCG had no effects on Gq expression in HepG2 cells, respectively. CONCLUSION: These findings suggest that the anti-HCC effects of EGCG might be mediated, at least partially, through the suppressing EP(1) receptor expression and PGE(2) production.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Carcinoma, Hepatocellular/metabolism , Catechin/analogs & derivatives , Cell Movement/drug effects , Cell Proliferation/drug effects , Liver Neoplasms/metabolism , Receptors, Prostaglandin E, EP1 Subtype/drug effects , Alprostadil/analogs & derivatives , Alprostadil/pharmacology , Blotting, Western , Bridged Bicyclo Compounds/pharmacology , Caproates/pharmacology , Carcinoma, Hepatocellular/pathology , Catechin/pharmacology , Cell Survival/drug effects , Dinoprostone/metabolism , Dose-Response Relationship, Drug , GTP-Binding Protein alpha Subunits, Gq-G11/drug effects , GTP-Binding Protein alpha Subunits, Gq-G11/metabolism , Hep G2 Cells , Humans , Immunoassay , Liver Neoplasms/pathology , Neoplasm Invasiveness , Receptors, Prostaglandin E, EP1 Subtype/metabolism , Time Factors
8.
Exp Cell Res ; 315(18): 3210-9, 2009 Nov 01.
Article in English | MEDLINE | ID: mdl-19563802

ABSTRACT

We previously reported that the hypothalamic hormone oxytocin (OT), best known for its uterotonic activity, also stimulates migration and invasion in human umbilical vein endothelial cells (HUVECs), thus suggesting a possible role for the peptide in the regulation of angiogenesis. We identified the Gq coupling of OT receptors (OTRs) and phospholipase C (PLC) as the main effectors of OT's action in HUVECs. Moreover, the pro-migratory effect of OT required the OTR-induced activation of the phosphatidylinositol-3-kinase (PI-3-K)/AKT/endothelial nitric oxide synthase (eNOS) pathway. To better characterize the proposed pro-angiogenic effect of OT in HUVECs, we have now utilized a three-dimensional (3-D) in vitro angiogenesis assay, and demonstrated that OT stimulates the outgrowth of capillary-like structures from HUVEC spheroids to an extent comparable to that of vascular endothelial growth factor (VEGF). This OT effect was abolished by inhibitors of PLC, PI-3-K and Src kinase. It was also found that OT phosphorylates proline-rich tyrosine kinase-2 (Pyk-2) and Src kinase in a PLC- and calcium-dependent manner. Furthermore, knockdown of Pyk-2 expression by RNA interference markedly impaired Src phosphorylation, migration and endothelial cell sprouting induced by OT. In conclusion, by using a pharmacological and genetic approach, the OT pro-angiogenic action and the cascade of intracellular signals responsible for it were defined by showing for the first time that OT, by interacting with its Gq-coupled receptor, induces HUVEC capillary outgrowth via Pyk-2 phosphorylation, which activates Src which in turn activates the PI-3-K/AKT pathway.


Subject(s)
Endothelial Cells/drug effects , Focal Adhesion Kinase 2/metabolism , Neovascularization, Physiologic , Oxytocin/pharmacology , Type C Phospholipases/metabolism , src-Family Kinases/metabolism , Cell Movement/drug effects , Cell Movement/physiology , Cells, Cultured , Chromones/pharmacology , Endothelial Cells/cytology , Endothelial Cells/metabolism , Enzyme Inhibitors/pharmacology , Estrenes/pharmacology , Focal Adhesion Kinase 2/drug effects , Focal Adhesion Kinase 2/genetics , GTP-Binding Protein alpha Subunits, Gq-G11/drug effects , GTP-Binding Protein alpha Subunits, Gq-G11/metabolism , Humans , Morpholines/pharmacology , Phosphatidylinositol 3-Kinases/metabolism , Phosphodiesterase Inhibitors/pharmacology , Phosphoinositide-3 Kinase Inhibitors , Phosphorylation/drug effects , Phosphorylation/physiology , Phosphotransferases (Alcohol Group Acceptor)/antagonists & inhibitors , Phosphotransferases (Alcohol Group Acceptor)/metabolism , Proto-Oncogene Proteins c-akt/drug effects , Proto-Oncogene Proteins c-akt/metabolism , Pyrimidines/pharmacology , Pyrrolidinones/pharmacology , RNA, Small Interfering/metabolism , Receptors, Oxytocin/drug effects , Receptors, Oxytocin/metabolism , Signal Transduction/drug effects , Signal Transduction/physiology , Sphingosine/analogs & derivatives , Sphingosine/pharmacology , Type C Phospholipases/antagonists & inhibitors , Umbilical Veins/cytology , Umbilical Veins/drug effects , Vascular Endothelial Growth Factor A/pharmacology , src-Family Kinases/antagonists & inhibitors
9.
J Biomol Screen ; 14(3): 246-55, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19211780

ABSTRACT

G-protein-coupled receptors can couple to different signal transduction pathways in different cell types (termed cell-specific signaling) and can activate different signaling pathways depending on the receptor conformation(s) stabilized by the activating ligand (functional selectivity). These concepts offer potential for developing pathway-specific drugs that increase efficacy and reduce side effects. Despite significant interest, functional selectivity has been difficult to exploit in drug discovery, in part due to the burden of multiple assays. Cellular impedance assays use an emerging technology that can qualitatively distinguish Gs, Gi/o, and Gq signaling in a single assay and is thereby suited for studying these pharmacological concepts. Cellular impedance confirmed cell-specific Gs and Gq coupling for the melanocortin-4 receptor and dual Gi and Gs signaling with the cannabinoid-1 (CB1) receptor. The balance of Gi versus Gs signaling depended on the cell line. In CB1-HEKs, Giand Gs-like responses combined to yield a novel impedance profile demonstrating the dynamic nature of these traces. Cellspecific signaling was observed with endogenous D1 receptor in U-2 cells and SK-N-MC cells, yet the pharmacological profile of partial and full agonists was similar in both cell lines. We conclude that the dynamic impedance profile encodes valuable relative signaling information and is sufficiently robust to help evaluate cell-specific signaling and functional selectivity.


Subject(s)
Biological Assay/methods , GTP-Binding Protein alpha Subunits, Gi-Go/metabolism , GTP-Binding Protein alpha Subunits, Gq-G11/metabolism , GTP-Binding Protein alpha Subunits, Gs/metabolism , Receptors, G-Protein-Coupled/metabolism , Animals , Brain Neoplasms/metabolism , Brain Neoplasms/pathology , CHO Cells , Cell Culture Techniques , Cell Line, Tumor , Cells, Cultured , Cricetinae , Cricetulus , Cytochalasin D/pharmacology , Dopamine Agonists/pharmacology , Dose-Response Relationship, Drug , Electric Impedance , GTP-Binding Protein alpha Subunits, Gi-Go/drug effects , GTP-Binding Protein alpha Subunits, Gq-G11/drug effects , GTP-Binding Protein alpha Subunits, Gs/drug effects , Humans , Inhibitory Concentration 50 , Kidney/cytology , Neuroectodermal Tumors, Primitive, Peripheral/metabolism , Neuroectodermal Tumors, Primitive, Peripheral/pathology , Osteosarcoma/metabolism , Osteosarcoma/pathology , Pertussis Toxin/pharmacology , Receptor, Muscarinic M1/metabolism , Receptors, Dopamine D2/metabolism , Receptors, Dopamine D5/metabolism , Sensitivity and Specificity , Signal Transduction/drug effects , Signal Transduction/physiology , alpha-MSH/agonists , alpha-MSH/analogs & derivatives
10.
Pharmacology ; 82(4): 245-9, 2008.
Article in English | MEDLINE | ID: mdl-18818509

ABSTRACT

GPR35 is a Gi/o- and G16-coupled receptor abundantly expressed in gastrointestinal tissues and immune cells. Kynurenic acid (a tryptophan metabolite and ionotropic glutamate receptor antagonist) and zaprinast (a phosphodiesterase inhibitor) are GPR35 agonists. Here, we show that the chloride channel blocker 5-nitro-2-(3-phenylpropylamino) benzoic acid (NPPB) is also a GPR35 agonist. NPPB activates the GPR35-Gi/o and GPR35-G16 pathways in human embryonic kidney 293 (HEK293) cells and induces intracellular calcium mobilization in a concentration-dependent manner in HEK293 cells coexpressing human, rat or mouse GPR35 and the chimeric G protein G(qi5). These results suggest a novel pharmacological activity of NPPB and will provide useful information to search for more potent and selective GPR35 agonists.


Subject(s)
Nitrobenzoates/pharmacology , Receptors, G-Protein-Coupled/agonists , Animals , Cell Line , Chloride Channels/antagonists & inhibitors , Dose-Response Relationship, Drug , GTP-Binding Protein alpha Subunits, Gi-Go/drug effects , GTP-Binding Protein alpha Subunits, Gi-Go/metabolism , GTP-Binding Protein alpha Subunits, Gq-G11/drug effects , GTP-Binding Protein alpha Subunits, Gq-G11/metabolism , Humans , Kidney/cytology , Kidney/metabolism , Mice , Nitrobenzoates/administration & dosage , Purinones/administration & dosage , Purinones/pharmacology , Rats
11.
J Pept Sci ; 14(8): 998-1002, 2008 Aug.
Article in English | MEDLINE | ID: mdl-18383439

ABSTRACT

G(q), a heterotrimeric guanine nucleotide-binding protein, plays important roles such as the regulation of calcium mobilization and cell proliferation. This protein is considered as a promising drug target for the treatment of cardiac hypertrophy. Selective activation of G(q) would be quite useful for analyzing the role of G(q) in signaling pathways. We synthesized m3i3c-a peptide with 16 amino acid residues that corresponds to the junction between the C-terminus of the third intracellular loop and the sixth transmembrane helix (TM-VI) of human m3 muscarinic acetylcholine receptor, which couples to G(q) but not G(i2). At micromolar concentrations, this peptide was found to activate G(q) but not G(i2). This peptide is the first small compound that selectively activates G(q) but not G(i2).


Subject(s)
GTP-Binding Protein alpha Subunits, Gq-G11/drug effects , Peptide Fragments/pharmacology , Receptor, Muscarinic M3/chemistry , Amino Acid Sequence , Animals , Binding Sites , Brain , Cattle , Cell Membrane/chemistry , Dose-Response Relationship, Drug , GTP-Binding Protein alpha Subunit, Gi2/chemistry , GTP-Binding Protein alpha Subunit, Gi2/drug effects , GTP-Binding Protein alpha Subunits, Gq-G11/chemistry , GTP-Binding Protein alpha Subunits, Gq-G11/isolation & purification , Humans , Molecular Sequence Data , Peptide Fragments/chemical synthesis , Peptide Fragments/chemistry , Protein Folding/drug effects , Protein Structure, Secondary , Protein Structure, Tertiary , Structure-Activity Relationship
12.
Assay Drug Dev Technol ; 6(1): 39-53, 2008 Feb.
Article in English | MEDLINE | ID: mdl-18315499

ABSTRACT

Abstract: Compared to biochemical high-throughput screening (HTS) assays, cell-based functional assays are generally thought to be more time consuming and complex because of additional efforts for running continuous cell cultures as well as the numerous assay steps when transferring media and compounds. A common strategy to compensate the anticipated reduction in overall throughput is to implement highly automated cell culture and screening systems. However, such systems require substantial investments in sophisticated hardware and highly specialized personnel. In trying to set up alternatives to increasing throughput in functional cell-based screening, we combined several approaches. By using (1) cryopreserved cell aliquots instead of continuous cell culture, (2) cells in suspension instead of adherent cells, and (3) "ready-to-screen" assay plates with nanoliter aliquots of test compounds, an assay procedure was developed that very much resembles a standard biochemical, enzymatic assay comprising only a few dispense steps. Chinese hamster ovary cells stably overexpressing a Galphaq-coupled receptor were used as a model system to measure receptor activation by detection of intracellular D-myo-inositol 1-phosphate with the help of homogeneous time-resolved fluorescence (HTRF, CISbio International, Bagnols-sur-Cèze, France). Initially established in 384-well adherent cell format, the assay was successfully transferred to 1,536-well format. The assay quality was sufficient to run HTS campaigns in both formats with good Z'-factors and excellent reproducibility of antagonists. Subsequently, the assay procedure was optimized for usage of suspension cells. The influences of cell culture media, plate type, cell number, and incubation time were assessed. Finally, the suspension cell assay was applied to pharmacological characterization of a small molecule antagonist by Schild plot analysis. Our data demonstrate not only the application of the IP-One HTRF assay (CISbio International) for HTS in a high-density format, but furthermore the successful use of cryopreserved and suspension cells in a one-day functional cell-based assay.


Subject(s)
Drug Evaluation, Preclinical/methods , GTP-Binding Protein alpha Subunits, Gq-G11/drug effects , Animals , Automation , CHO Cells , Calibration , Cell Adhesion , Cells, Cultured , Cricetinae , Cricetulus , GTP-Binding Protein alpha Subunits, Gq-G11/antagonists & inhibitors , Gene Library , Indicators and Reagents , Kinetics , Linear Models , Robotics
13.
Cell Biochem Funct ; 26(2): 264-74, 2008.
Article in English | MEDLINE | ID: mdl-18041110

ABSTRACT

The molecular mechanisms involved in GPCR-initiated signaling cascades where the two receptors share the same signaling cascade, such as thyrotropin-releasing hormone (TRH) and angiotensin II (ANG II), are still far from being understood. Here, we analyzed hormone-induced Ca(2+) responses and the process of desensitization in HEK-293 cells, which express endogenous ANG II receptors. These cells were transfected to express exogenously high levels of TRH receptors (clone E2) or both TRH receptors and G(11)alpha protein (clone E2M11). We observed that the characteristics of the Ca(2+) response, as well as the process of desensitization, were both strongly dependent on receptor number and G(11)alpha protein level. Whereas treatment of E2 cells with TRH or ANG II led to significant desensitization of the Ca(2+) response to subsequent addition of either hormone, the response was not desensitized in E2M11 cells expressing high levels of G(11)alpha. In addition, stimulation of both cell lines with THR elicited a clear heterologous desensitization to subsequent stimulation with ANG II. On the other hand, ANG II did not affect a subsequent response to TRH. ANG II-mediated signal transduction was strongly dependent on plasma membrane integrity modified by cholesterol depletion, but signaling through TRH receptors was altered only slightly under these conditions. It may be concluded that the level of expression of G-protein-coupled receptors and their cognate G-proteins strongly influences not only the magnitude of the Ca(2+) response but also the process of desensitization and resistance to subsequent hormone addition.


Subject(s)
Angiotensin II/pharmacology , Calcium Signaling/drug effects , Calcium/metabolism , Cell Membrane/metabolism , GTP-Binding Protein alpha Subunits, Gq-G11/biosynthesis , Thyrotropin-Releasing Hormone/pharmacology , Calcium/pharmacology , Calcium Signaling/physiology , Cell Membrane/drug effects , Cells, Cultured , Cholesterol/analysis , Cholesterol/metabolism , Dose-Response Relationship, Drug , GTP-Binding Protein alpha Subunits, Gq-G11/drug effects , GTP-Binding Protein alpha Subunits, Gq-G11/genetics , Humans , Receptors, Thyrotropin-Releasing Hormone/biosynthesis , Receptors, Thyrotropin-Releasing Hormone/drug effects , Temperature , Time Factors , Transfection
14.
Arch Pharm (Weinheim) ; 340(1): 9-16, 2007 Jan.
Article in English | MEDLINE | ID: mdl-17206612

ABSTRACT

Imidazolylpropylguanidines derived from impromidine and arpromidine are more potent and efficacious agonists at the guinea pig histamine H2 receptor (gpH2R) than at the human H2R (hH2R) in the GTPase assay. Additionally, such guanidines are histamine H1 receptor (H1R) antagonists with preference for the human relative to the guinea pig receptor. The purpose of this study was to examine structure-activity relationships of guanidines at human and guinea pig H1R and H2R species isoforms expressed in Sf9 insect cells. Three impromidine analogues and six arpromidine analogues exhibited agonistic activity at H2R and antagonistic activity at H1R as assessed in the steady-state GTPase assay. Species selectivity of derivatives was similar as compared with the parent compounds. None of the structural modifications examined (different aromatic ring systems and different ring substituents) was superior in terms of H2R potency and efficacy relative to impromidine and arpromidine, respectively. These data point to substantial structural constraints at the agonist binding site of H2R. Guanidines exhibited distinct structure-activity relationships for H1R antagonism in a radioligand competition binding assay and the GTPase assay and for H1R inverse agonism. Our data indicate that it is difficult to obtain guanidine-type agonists with high potency and high efficacy for hH2R, but those compounds may be useful tools for exploring the antagonist binding site and constitutive activity of H1R.


Subject(s)
Guanidines/pharmacology , Histamine Agonists/pharmacology , Histamine H1 Antagonists/pharmacology , Imidazoles/pharmacology , Impromidine/pharmacology , Receptors, Histamine H1/drug effects , Receptors, Histamine H2/drug effects , Animals , Binding, Competitive , Cell Line , GTP Phosphohydrolases/metabolism , GTP-Binding Protein alpha Subunits, Gq-G11/drug effects , GTP-Binding Protein alpha Subunits, Gq-G11/genetics , GTP-Binding Protein alpha Subunits, Gs/drug effects , GTP-Binding Protein alpha Subunits, Gs/genetics , Guanidines/chemistry , Guanidines/metabolism , Guinea Pigs , Histamine Agonists/chemistry , Histamine Agonists/metabolism , Histamine H1 Antagonists/chemistry , Histamine H1 Antagonists/metabolism , Humans , Imidazoles/chemistry , Imidazoles/metabolism , Impromidine/analogs & derivatives , Impromidine/chemistry , Impromidine/metabolism , Insecta , Molecular Structure , Pyrilamine/metabolism , Receptors, Histamine H1/genetics , Receptors, Histamine H1/metabolism , Receptors, Histamine H2/genetics , Receptors, Histamine H2/metabolism , Recombinant Fusion Proteins/drug effects , Structure-Activity Relationship , Transfection
15.
Glia ; 55(3): 317-27, 2007 Feb.
Article in English | MEDLINE | ID: mdl-17115416

ABSTRACT

The Pituitary adenylate cyclase-activating peptide (PACAP) ligand/type 1 receptor (PAC1) system regulates neurogenesis and gliogenesis. It has been well established that the PACAP/PAC1 system induces differentiation of neural progenitor cells (NPCs) through the Gs-mediated cAMP-dependent signaling pathway. However, it is unknown whether this ligand/receptor system has a function in proliferation of NPCs. In this study, we identified that PACAP and PAC1 were highly expressed and co-localized in NPCs of mouse cortex at embryonic day 14.5 (E14.5) and found that the PACAP/PAC1 system potentiated growth factor-induced proliferation of mouse cortical NPCs at E14.5 via Gq-, but not Gs-, mediated PLC/IP3-dependent signaling pathway in an autocrine manner. Moreover, PAC1 activation induced elongation of cellular processes and a stellate morphology in astrocytes that had the bromodeoxyuridine (BrdU)-incorporating ability of NPCs. Consistent with this notion, we determined that the most BrdU positive NPCs differentiated to astrocytes through PAC1 signaling. These results suggest that the PACAP/PAC1 system may play a dual role in neural/glial progenitor cells not only differentiation but also proliferation in the cortical astrocyte lineage via Ca2+-dependent signaling pathways through PAC1.


Subject(s)
Astrocytes/metabolism , Cell Differentiation/physiology , Cell Proliferation , Pituitary Adenylate Cyclase-Activating Polypeptide/metabolism , Receptors, Pituitary Adenylate Cyclase-Activating Polypeptide/metabolism , Stem Cells/metabolism , Animals , Astrocytes/cytology , Autocrine Communication/physiology , Bromodeoxyuridine , Calcium Signaling/physiology , Cell Lineage/physiology , Cell Shape/drug effects , Cell Shape/physiology , Cells, Cultured , Cerebral Cortex/cytology , Cerebral Cortex/embryology , Cerebral Cortex/metabolism , GTP-Binding Protein alpha Subunits, Gq-G11/drug effects , GTP-Binding Protein alpha Subunits, Gq-G11/metabolism , Intercellular Signaling Peptides and Proteins/metabolism , Intercellular Signaling Peptides and Proteins/pharmacology , Mice , Mice, Inbred C57BL , Neurons/cytology , Neurons/metabolism , Signal Transduction/drug effects , Signal Transduction/physiology , Stem Cells/cytology , Type C Phospholipases/drug effects , Type C Phospholipases/metabolism
16.
Eur J Pharmacol ; 551(1-3): 10-8, 2006 Dec 03.
Article in English | MEDLINE | ID: mdl-17027743

ABSTRACT

With respect to the discovery and characterization of neuropeptide Y(2) receptor ligands as pharmacological tools or potential drugs, fluorescence- and luminescence-based assays were developed to determine both the affinity and the activity of receptor agonists and antagonists. A flow cytometric binding assay is described for the hY(2) receptor stably expressed in CHO cells using cy5-labeled porcine neuropeptide Y and compared with a radioligand binding assay. Binding of the fluorescent ligand was visualized by confocal microscopy. Stable co-transfection with the chimeric G protein Gq(i5) enabled the establishment of a spectrofluorimetric fura-2 and a flow cytometric fluo-4 calcium assay. Further stable expression of apoaequorin targeted to the mitochondria allowed the establishment of an aequorin assay which could be performed in the 96-well format. The shape of the concentration-response curves of porcine neuropeptide Y in the presence of the Y(2)-selective receptor antagonist BIIE0246, characteristic of either competitive or insurmountable antagonism, depended on the period of incubation with the cells. Functional data of Y(2) receptor agonists and antagonists determined in the fluorescence- and luminescence-based assays were in good agreement.


Subject(s)
Calcium Signaling , Flow Cytometry/methods , Microscopy, Confocal/methods , Neuropeptide Y/metabolism , Receptors, Neuropeptide Y/metabolism , Spectrometry, Fluorescence/methods , Aequorin/drug effects , Aequorin/metabolism , Animals , Apoproteins/drug effects , Apoproteins/metabolism , Arginine/analogs & derivatives , Arginine/metabolism , Arginine/pharmacology , Benzazepines/metabolism , Benzazepines/pharmacology , Binding, Competitive , CHO Cells , Calcium Signaling/drug effects , Cricetinae , Cricetulus , Dose-Response Relationship, Drug , Fluorescent Dyes , GTP-Binding Protein alpha Subunits, Gq-G11/drug effects , GTP-Binding Protein alpha Subunits, Gq-G11/metabolism , Ligands , Neuropeptide Y/analogs & derivatives , Neuropeptide Y/pharmacology , Peptide Fragments/metabolism , Peptide Fragments/pharmacology , Peptide YY/metabolism , Peptide YY/pharmacology , Protein Binding , Radioligand Assay , Receptors, Neuropeptide Y/drug effects , Recombinant Fusion Proteins/drug effects , Recombinant Fusion Proteins/metabolism , Recombinant Proteins/drug effects , Recombinant Proteins/metabolism , Reproducibility of Results , Swine , Transfection
17.
J Physiol ; 577(Pt 2): 497-511, 2006 Dec 01.
Article in English | MEDLINE | ID: mdl-16990402

ABSTRACT

Thyrotropin-releasing hormone (TRH) is a tripeptide that is widely distributed in the brain including the hippocampus where TRH receptors are also expressed. TRH has anti-epileptic effects and regulates arousal, sleep, cognition, locomotion and mood. However, the cellular mechanisms underlying such effects remain to be determined. We examined the effects of TRH on GABAergic transmission in the hippocampus and found that TRH increased the frequency of GABAA receptor-mediated spontaneous IPSCs in each region of the hippocampus but had no effects on miniature IPSCs or evoked IPSCs. TRH increased the action potential firing frequency recorded from GABAergic interneurons in CA1 stratum radiatum and induced membrane depolarization suggesting that TRH increases the excitability of interneurons to facilitate GABA release. TRH-induced inward current had a reversal potential close to the K+ reversal potential suggesting that TRH inhibits resting K+ channels. The involved K+ channels were sensitive to Ba2+ but resistant to other classical K+ channel blockers, suggesting that TRH inhibits the two-pore domain K+ channels. Because the effects of TRH were mediated via Galphaq/11, but were independent of its known downstream effectors, a direct coupling may exist between Galphaq/11 and K+ channels. Inhibition of the function of dynamin slowed the desensitization of TRH responses. TRH inhibited seizure activity induced by Mg2+ deprivation, but not that generated by picrotoxin, suggesting that TRH-mediated increase in GABA release contributes to its anti-epileptic effects. Our results demonstrate a novel mechanism to explain some of the hippocampal actions of TRH.


Subject(s)
Anticonvulsants/pharmacology , Hippocampus/drug effects , Neural Inhibition/drug effects , Receptors, GABA-A/metabolism , Synaptic Transmission/drug effects , Thyrotropin-Releasing Hormone/pharmacology , gamma-Aminobutyric Acid/metabolism , Action Potentials/drug effects , Animals , Dynamins/metabolism , GTP-Binding Protein alpha Subunits, Gq-G11/drug effects , GTP-Binding Protein alpha Subunits, Gq-G11/metabolism , Hippocampus/cytology , Hippocampus/metabolism , In Vitro Techniques , Inhibitory Postsynaptic Potentials/drug effects , Interneurons/drug effects , Interneurons/metabolism , Isoenzymes/genetics , Isoenzymes/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , Patch-Clamp Techniques , Phospholipase C beta , Potassium/metabolism , Potassium Channels, Tandem Pore Domain/drug effects , Potassium Channels, Tandem Pore Domain/metabolism , Rats , Rats, Sprague-Dawley , Time Factors , Type C Phospholipases/genetics , Type C Phospholipases/metabolism
18.
Anesthesiology ; 105(2): 313-24, 2006 Aug.
Article in English | MEDLINE | ID: mdl-16871065

ABSTRACT

BACKGROUND: Halothane inhibits airway smooth muscle contraction in part by inhibiting the functional coupling between muscarinic receptors and one of its cognate heterotrimeric G proteins, Galphaq. Based on previous studies indicating a more potent effect of halothane and sevoflurane on airway smooth muscle contraction compared with isoflurane, the current study hypothesized that at anesthetic concentrations of 2 minimum alveolar concentration (MAC) or less, halothane and sevoflurane but not isoflurane inhibit acetylcholine-promoted Galphaq guanosine nucleotide exchange. METHODS: Galphaq guanosine nucleotide exchange was measured in crude membranes prepared from COS-7 cells transiently coexpressing the human M3 muscarinic receptor and human Galphaq. A radioactive, nonhydrolyzable analog of guanosine-5'-triphosphate, [35S]GTPgammaS, was used as a reporter for nucleotide exchange at Galphaq. RESULTS: Acetylcholine caused a concentration-dependent increase in Galphaq [35S]GTPgammaS-GDP exchange. Neither anesthetic affected constitutive Galphaq [35S]GTPgammaS-GDP exchange in the absence of acetylcholine. Conversely, each anesthetic caused a concentration-dependent and reversible inhibition of Galphaq [35S]GTPgammaS-GDP exchange when promoted by acetylcholine. At concentrations of 3 MAC or less, the effect of halothane and sevoflurane were significantly greater than that of isoflurane, with only a minimal inhibition by isoflurane observed at 2 MAC. CONCLUSION: The differential effects of volatile anesthetics on acetylcholine-promoted guanosine nucleotide exchange at Galphaq are consistent with the apparent more potent direct effect of halothane and sevoflurane compared with isoflurane on muscarinic receptor-mediated contraction of isolated airway smooth muscle. These differential effects also suggest a mode of anesthetic action that could be due to anesthetic-protein interactions and not simply anesthetic accumulation in the lipid membrane.


Subject(s)
Anesthetics, Inhalation/pharmacology , GTP-Binding Protein alpha Subunits, Gq-G11/drug effects , Receptor, Muscarinic M3/drug effects , Acetylcholine/pharmacology , Algorithms , Animals , COS Cells , Calcium/metabolism , Cell Membrane/drug effects , Cell Membrane/metabolism , Cells, Cultured , Chlorocebus aethiops , Cytoplasm/metabolism , Data Interpretation, Statistical , Guanine Nucleotides/metabolism , Halothane/pharmacology , Hexanols/pharmacology , Humans , Immunoblotting , Isoflurane/pharmacology , Isometric Contraction , Methyl Ethers/pharmacology , Sevoflurane
19.
Neuroscience ; 137(2): 545-53, 2006.
Article in English | MEDLINE | ID: mdl-16289891

ABSTRACT

Diverse excitatory and inhibitory neuronal responses are mediated via Gq-coupled receptors, but the lack of a systematic comparison of different receptors or neurons has hindered a better understanding of these responses. Such a comparison may be provided by an exogenous receptor that is activated by compounds that have no effect on endogenous receptors. We therefore expressed an invertebrate biogenic amine receptor, the Drosophila octopamine receptor, in rat cortical neurons and compared octopamine receptor-mediated responses with those mediated by the group I metabotropic glutamate receptor, the endogenous Gq-coupled receptor in rat cortical neurons. Stimulation of either receptor did not result in a calcium response in octopamine receptor-expressing neurons, although octopamine preferentially elicited a calcium increase in octopamine receptor-expressing PC12h cells, while enhancing the neuronal depolarization-induced calcium increase and the electrical excitability. The increased excitability was caused by inward currents resulting from a reduction in the leak current, which was voltage-independent and blocked by genistein, a non-selective tyrosine kinase inhibitor. These results show that, in cortical neurons, exogenous octopamine receptor in mushroom bodies activated the same cell signaling pathway as endogenous metabotropic glutamate receptor, suggesting that the diverse neuronal responses mediated by Gq-coupled receptors are due to the properties of different neurons, rather than to the properties of the receptors.


Subject(s)
Drosophila Proteins/genetics , Drosophila/genetics , GTP-Binding Protein alpha Subunits, Gq-G11/genetics , Mammals/genetics , Neurons/metabolism , Receptors, G-Protein-Coupled/genetics , Receptors, Neurotransmitter/genetics , Signal Transduction/genetics , Action Potentials/drug effects , Action Potentials/genetics , Animals , Calcium Channels/drug effects , Calcium Channels/metabolism , Calcium Signaling/drug effects , Calcium Signaling/genetics , Cells, Cultured , Cerebral Cortex/drug effects , Cerebral Cortex/metabolism , Enzyme Inhibitors/pharmacology , GTP-Binding Protein alpha Subunits, Gq-G11/drug effects , Gene Transfer Techniques , Mushroom Bodies/metabolism , Neurons/drug effects , PC12 Cells , Patch-Clamp Techniques , Phosphatidylinositols/metabolism , Protein-Tyrosine Kinases/antagonists & inhibitors , Protein-Tyrosine Kinases/metabolism , Rats , Receptors, G-Protein-Coupled/drug effects , Receptors, Metabotropic Glutamate/drug effects , Receptors, Metabotropic Glutamate/metabolism , Signal Transduction/drug effects , Synaptic Transmission/drug effects , Synaptic Transmission/genetics
20.
Proc Natl Acad Sci U S A ; 102(12): 4643-8, 2005 Mar 22.
Article in English | MEDLINE | ID: mdl-15767570

ABSTRACT

Phosphatidylionsitol 4,5-bisphosphate (PIP(2)), a substrate of phospholipase C, has recently been recognized to regulate membrane-associated proteins and act as a signal molecule in phospholipase C-linked Gq-coupled receptor (GqPCR) pathways. However, it is not known whether PIP(2) depletion induced by GqPCRs can act as receptor-specific signals in native cells. We investigated this issue in cardiomyocytes where PIP(2)-dependent ion channels, G protein-gated inwardly rectifying K(+) (GIRK) and inwardly rectifying background K(+) (IRK) channels, and various GqPCRs are present. The GIRK current was recorded by using the patch-clamp technique during the application of 10 microM acetylcholine. The extent of receptor-mediated inhibition was estimated as the current decrease over 4 min while taking the GIRK current (I(GIRK)) value during a previous stimulation as the control. Each GqPCR agonist inhibited I(GIRK) with different potencies and kinetics. The extents of inhibition induced by phenylephrine, angiotensin II, endothelin-1, prostaglandin F2alpha, and bradykinin at supramaximal concentrations were (mean +/- SE) 32.1 +/- 0.6%, 21.9 +/- 1.4%, 86.4 +/- 1.6%, 63.7 +/- 4.9%, and 5.7 +/- 1.9%, respectively. GqPCR-induced inhibitions of I(GIRK) were not affected by protein kinase C inhibitor (calphostin C) but potentiated and became irreversible when the replenishment of PIP(2) was blocked by wortmannin (phosphatidylinositol kinase inhibitor). Loading the cells with PIP(2) significantly reduced endothelin-1 and prostaglandin F2alpha-induced inhibition of I(GIRK). On the contrary, GqPCR-mediated inhibitions of inwardly rectifying background K(+) currents were observed only when GqPCR agonists were applied with wortmannin, and the effects were not parallel with those on I(GIRK). These results indicate that GqPCR-induced inhibition of ion channels by means of PIP(2) depletion occurs in a receptor-specific manner.


Subject(s)
GTP-Binding Protein alpha Subunits, Gq-G11/metabolism , Phosphatidylinositol 4,5-Diphosphate/metabolism , Potassium Channels, Inwardly Rectifying/antagonists & inhibitors , Acetylcholine/pharmacology , Androstadienes/pharmacology , Angiotensin II/pharmacology , Animals , Bradykinin/pharmacology , Dinoprost/pharmacology , Endothelin-1/pharmacology , G Protein-Coupled Inwardly-Rectifying Potassium Channels , GTP-Binding Protein alpha Subunits, Gq-G11/drug effects , In Vitro Techniques , Mice , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , Naphthalenes/pharmacology , Patch-Clamp Techniques , Phenylephrine/pharmacology , Potassium Channels, Inwardly Rectifying/drug effects , Potassium Channels, Inwardly Rectifying/metabolism , Wortmannin
SELECTION OF CITATIONS
SEARCH DETAIL