Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 302
Filter
1.
Biochem Pharmacol ; 86(6): 824-35, 2013 Sep 15.
Article in English | MEDLINE | ID: mdl-23928187

ABSTRACT

The pregnane X receptor (PXR) regulates drug metabolism by regulating the expression of drug-metabolizing enzymes such as cytochrome P450 3A4 (CYP3A4), which is involved in the metabolism of >50% of clinically prescribed drugs. The activity of PXR can be controlled by the binding of small molecule agonists or antagonists. Because of its unique ligand binding pocket, PXR binds promiscuously to structurally diverse chemicals. To study the structure-activity relationship, novel modulators for PXR are needed. Here we report the virtual screening of ∼25,000 natural product derivatives from the ZINC database using the Molecular Operating Environment docking software tool against the PXR-rifampicin complex X-ray crystal structure. Our screening resulted in identification of compounds based on the lowest S score, which measures Gibbs free energy. Interestingly, we found that the compounds that bind directly to PXR, as revealed in an intrinsic tryptophan fluorescence assay, modulate CYP3A4 promoter activity differentially in HepG2 cells. Mutational analysis and docking studies showed that these compounds bind broadly in the ligand binding pocket but interact with different amino acid residues. We further investigated the mechanism of binding by analyzing the functional groups that are important for distinguishing agonists from antagonists. The approach we used to identify novel modulators that bind to PXR can be useful for finding novel modulators of PXR.


Subject(s)
Biological Products/chemistry , Cytochrome P-450 CYP3A/genetics , Epithelial Cells/metabolism , Receptors, Steroid/genetics , Binding Sites , Biological Products/pharmacology , Cell Line , Cytochrome P-450 CYP3A/metabolism , Databases, Chemical , Epithelial Cells/cytology , Epithelial Cells/drug effects , Escherichia coli/genetics , Escherichia coli/metabolism , Gene Expression Regulation/drug effects , Genes, Reporter , Hep G2 Cells , High-Throughput Screening Assays , Humans , Luciferases , Molecular Docking Simulation , Pregnane X Receptor , Promoter Regions, Genetic , Protein Binding , Receptors, Steroid/agonists , Receptors, Steroid/antagonists & inhibitors , Receptors, Steroid/metabolism , Recombinant Proteins/agonists , Recombinant Proteins/antagonists & inhibitors , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Structure-Activity Relationship
2.
Biochem J ; 454(2): 283-93, 2013 Sep 01.
Article in English | MEDLINE | ID: mdl-23782265

ABSTRACT

The atherogenic cytokine IL-6 (interleukin-6) induces pro-inflammatory gene expression in VECs (vascular endothelial cells) by activating the JAK (Janus kinase)/STAT3 (signal transducer and activator of transcription 3) signalling pathway, which is normally down-regulated by the STAT3-dependent induction of the E3 ubiquitin ligase component SOCS3 (suppressor of cytokine signalling 3). Novel treatments based on the regulation of SOCS3 protein levels could therefore have value in the treatment of diseases with an inflammatory component, such as atherosclerosis. To this end we carried out a screen of 1031 existing medicinal compounds to identify inducers of SOCS3 gene expression and identified the flavanoids naringenin and flavone as effective inducers of SOCS3 protein, mRNA and promoter activity. This was in contrast with the action of traditional JAK/STAT3 inhibitors and the polyphenol resveratrol, which effectively suppress SOCS3 gene expression. Both naringenin and flavone also effectively suppressed IL-6-stimulated phosphorylation of STAT3 (Tyr7°5) which led to suppression of IL-6-induction of the atherogenic STAT3 target gene MCP1 (monocyte chemotactic protein-1), suggesting that their ability to induce SOCS3 gene expression is STAT3-independent. Supporting this idea was the observation that the general kinase inhibitor compound C inhibits flavone- and cAMP-dependent, but not JAK-dependent, SOCS3 induction in VECs. Indeed, the ability of flavanoids to induce SOCS3 expression requires activation of the ERK (extracellular-signal-regulated kinase)-dependent transcription factor SP3, and not STAT3. In the present paper we therefore describe novel molecular actions of flavanoids, which control SOCS3 gene induction and suppression of STAT3 signalling in VECs. These mechanisms could potentially be exploited to develop novel anti-atherogenic therapies.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/metabolism , Cytokine Receptor gp130/antagonists & inhibitors , Endothelium, Vascular/metabolism , Flavonoids/metabolism , Interleukin-6/antagonists & inhibitors , STAT3 Transcription Factor/antagonists & inhibitors , Suppressor of Cytokine Signaling Proteins/agonists , AMP-Activated Protein Kinases/antagonists & inhibitors , AMP-Activated Protein Kinases/metabolism , Animals , Anti-Inflammatory Agents, Non-Steroidal/antagonists & inhibitors , Cell Line , Cells, Cultured , Chlorocebus aethiops , Cytokine Receptor gp130/metabolism , Endothelium, Vascular/cytology , Endothelium, Vascular/drug effects , Flavonoids/antagonists & inhibitors , Gene Expression Regulation/drug effects , Human Umbilical Vein Endothelial Cells/cytology , Human Umbilical Vein Endothelial Cells/drug effects , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Interleukin-6/metabolism , Interleukin-6 Receptor alpha Subunit/metabolism , Mice , Mutant Proteins/agonists , Mutant Proteins/metabolism , Promoter Regions, Genetic/drug effects , Protein Kinase Inhibitors/pharmacology , Recombinant Proteins/agonists , Recombinant Proteins/metabolism , STAT3 Transcription Factor/metabolism , Signal Transduction/drug effects , Suppressor of Cytokine Signaling 3 Protein , Suppressor of Cytokine Signaling Proteins/genetics , Suppressor of Cytokine Signaling Proteins/metabolism
3.
FEBS J ; 280(16): 3840-53, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23745637

ABSTRACT

The cyclin-dependent kinase inhibitor p21(CIP1/WAF1) is a regulatory factor of the cell cycle. Its transcriptional activation and protein stability are tightly controlled by several distinct mechanisms. S100A11 is a member of the S100 family of Ca²âº-binding proteins involved in several biological processes, including cell cycle progression and signal transduction. In the present study, we show that down-regulation of S100A11 results in the reduction of p21 protein in human HaCaT keratinocytes. It appears that a ubiquitin-independent proteasomal degradation process is involved in p21 degradation in S100A11 down-regulated cells. The application of a proteasome inhibitor stabilized p21 protein in these cells. Analysis of distinct signal transduction pathways revealed a disturbed phosphatidylinositol-3-kinase/Akt pathway after S100A11 knockdown. We determined that the glycogen synthase kinase-3, which is negatively regulated by phosphatidylinositol 3-kinase/Akt, was activated in cells possessing knocked-down S100A11 and appears to be involved in p21 protein destabilization. The application of a specific inhibitor of glycogen synthase kinase 3 resulted in an increase of the p21 protein level in S100A11 down-regulated HaCaT cells. Glycogen synthase kinase 3 is able to phosphorylate p21 at T57, which induces p21 proteasomal turnover. Mutation of the glycogen synthase kinase 3 site threonine 57 into alanine (T57A) stabilizes p21 in HaCaT cells lacking S100A11. Beside decreased p21 protein, down-regulation of S100A11 triggered the induction of apoptosis in HaCaT cells. These observations suggest that S100A11 is involved in the maintenance of p21 protein stability and appears to function as an inhibitor of apoptosis in human HaCaT keratinocyte cells. Thus, the data shed light on a novel pathway regulating p21 protein stability.


Subject(s)
Cyclin-Dependent Kinase Inhibitor p21/metabolism , Down-Regulation , Keratinocytes/metabolism , S100 Proteins/metabolism , Up-Regulation , Apoptosis/drug effects , Cell Line , Cyclin-Dependent Kinase Inhibitor p21/agonists , Cyclin-Dependent Kinase Inhibitor p21/antagonists & inhibitors , Cyclin-Dependent Kinase Inhibitor p21/genetics , Down-Regulation/drug effects , Gene Silencing , Glycogen Synthase Kinase 3/antagonists & inhibitors , Glycogen Synthase Kinase 3/chemistry , Glycogen Synthase Kinase 3/genetics , Glycogen Synthase Kinase 3/metabolism , Humans , Keratinocytes/drug effects , Mutant Proteins/agonists , Mutant Proteins/antagonists & inhibitors , Mutant Proteins/metabolism , Phosphatidylinositol 3-Kinase/metabolism , Phosphorylation/drug effects , Proteasome Endopeptidase Complex/chemistry , Proteasome Endopeptidase Complex/metabolism , Proteasome Inhibitors/pharmacology , Protein Kinase Inhibitors/pharmacology , Protein Processing, Post-Translational/drug effects , Protein Stability/drug effects , Proto-Oncogene Proteins c-akt/metabolism , Recombinant Proteins/agonists , Recombinant Proteins/antagonists & inhibitors , Recombinant Proteins/metabolism , S100 Proteins/antagonists & inhibitors , S100 Proteins/genetics , Signal Transduction/drug effects , Up-Regulation/drug effects
4.
Biochem J ; 454(2): 323-32, 2013 Sep 01.
Article in English | MEDLINE | ID: mdl-23758196

ABSTRACT

Ras GTPases undergo post-translational modifications that govern their subcellular trafficking and localization. In particular, palmitoylation of the Golgi tags N-Ras and H-Ras for exocytotic transport and residency at the PM (plasma membrane). Following depalmitoylation, PM-Ras redistributes to all subcellular membranes causing an accumulation of palmitate-free Ras at endomembranes, including the Golgi and endoplasmic reticulum. Palmitoylation is unanimously regarded as a critical modification at the crossroads of Ras activity and trafficking control, but its precise relevance to native wild-type Ras function in growth factor signalling is unknown. We show in the present study by use of palmitoylation-deficient N-Ras mutants and via the analysis of palmitate content of agonist-activated GTP-loaded N-Ras that only palmitoylated N-Ras becomes activated by agonists. In line with an essential role of palmitoylation in Ras activation, dominant-negative RasS17N loses its blocking potency if rendered devoid of palmitoylation. Live-cell Ras-GTP imaging shows that N-Ras activation proceeds only at the PM, consistent with activated N-Ras-GTP being palmitoylated. Finally, palmitoylation-deficient N-Ras does not sustain EGF (epidermal growth factor) or serum-elicited mitogenic signalling, confirming that palmitoylation is essential for signal transduction by N-Ras. These findings document that N-Ras activation proceeds at the PM and suggest that depalmitoylation, by removing Ras from the PM, may contribute to the shutdown of Ras signalling.


Subject(s)
Cell Membrane/metabolism , Down-Regulation , Epidermal Growth Factor/metabolism , GTP Phosphohydrolases/metabolism , Membrane Proteins/metabolism , Palmitic Acid/metabolism , Signal Transduction , ras Proteins/metabolism , Amino Acid Substitution , Animals , Cell Line , Cells, Cultured , Chlorocebus aethiops , Embryo, Mammalian/cytology , Enzyme Activation , GTP Phosphohydrolases/genetics , Humans , Lipoylation , Membrane Proteins/agonists , Membrane Proteins/genetics , Mice , Mice, Knockout , Mutant Proteins/agonists , Mutant Proteins/metabolism , Protein Processing, Post-Translational , Protein Transport , Recombinant Proteins/agonists , Recombinant Proteins/metabolism , ras Proteins/genetics
5.
J Biol Chem ; 288(29): 21082-21095, 2013 Jul 19.
Article in English | MEDLINE | ID: mdl-23754289

ABSTRACT

Understanding the molecular basis of drug action can facilitate development of more potent and selective drugs. Here, we explore the molecular basis for action of a unique small molecule ligand that is a type 1 cholecystokinin (CCK) receptor agonist and type 2 CCK receptor antagonist, GI181771X. We characterize its binding utilizing structurally related radioiodinated ligands selective for CCK receptor subtypes that utilize the same allosteric ligand-binding pocket, using wild-type receptors and chimeric constructs exchanging the distinct residues lining this pocket. Intracellular calcium assays were performed to determine biological activity. Molecular models for docking small molecule agonists to the type 1 CCK receptor were developed using a ligand-guided refinement approach. The optimal model was distinct from the previous antagonist model for the same receptor and was mechanistically consistent with the current mutagenesis data. This study revealed a key role for Leu(7.39) that was predicted to interact with the isopropyl group in the N1 position of the benzodiazepine that acts as a "trigger" for biological activity. The molecular model was predictive of binding of other small molecule agonists, effectively distinguishing these from 1065 approved drug decoys with an area under curve value of 99%. The model also selectively enriched for agonist compounds, with 130 agonists identified by ROC analysis when seeded in 2175 non-agonist ligands of the type 1 CCK receptor (area under curve 78%). Benzodiazepine agonists in this series docked in consistent pose within this pocket, with a key role played by Leu(7.39), whereas the role of this residue was less clear for chemically distinct agonists.


Subject(s)
Benzodiazepines/pharmacology , Receptor, Cholecystokinin A/agonists , Amino Acid Sequence , Animals , Benzodiazepines/chemistry , CHO Cells , Cricetinae , Cricetulus , Models, Molecular , Molecular Sequence Data , Mutant Proteins/agonists , Mutant Proteins/chemistry , Mutant Proteins/metabolism , ROC Curve , Receptor, Cholecystokinin A/chemistry , Receptor, Cholecystokinin A/metabolism , Receptor, Cholecystokinin B/chemistry , Receptor, Cholecystokinin B/metabolism , Recombinant Proteins/agonists , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Sequence Alignment
6.
Biochem J ; 454(2): 267-74, 2013 Sep 01.
Article in English | MEDLINE | ID: mdl-23767959

ABSTRACT

DGKs (diacylglycerol kinases) catalyse the conversion of diacylglycerol into PA (phosphatidic acid), a positive modulator of mTOR (mammalian target of rapamycin). We have found that chenodeoxycholic acid and the synthetic FXR (farnesoid X receptor) ligand GW4064 induce the mRNA and protein expression of DGKθ in the HepG2 cell line and in primary human hepatocytes. Reporter gene studies using 1.5 kB of the DGKθ promoter fused to the luciferase gene revealed that bile acids increase DGKθ transcriptional activity. Mutation of putative FXR-binding sites attenuated the ability of GW4046 to increase DGKθ luciferase activity. Consistent with this finding, ChIP (chromatin immunoprecipitation) assays demonstrated that bile acid signalling increased the recruitment of FXR to the DGKθ promoter. Furthermore, GW4064 evoked a time-dependent increase in the cellular concentration of PA. We also found that GW4064 and PA promote the phosphorylation of mTOR, Akt and FoxO1 (forkhead box O1), and that silencing DGKθ expression significantly abrogated the ability of GW4046 to promote the phosphorylation of these PA-regulated targets. DGKθ was also required for bile-acid-dependent decreased glucose production. Taken together, our results establish DGKθ as a key mediator of bile-acid-stimulated modulation of mTORC2 (mTOR complex 2), the Akt pathway and glucose homoeostasis.


Subject(s)
Chenodeoxycholic Acid/metabolism , Diacylglycerol Kinase/metabolism , Glucose/metabolism , Hepatocytes/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Receptors, Cytoplasmic and Nuclear/metabolism , Signal Transduction , Cells, Cultured , Diacylglycerol Kinase/antagonists & inhibitors , Diacylglycerol Kinase/chemistry , Diacylglycerol Kinase/genetics , Gene Expression Regulation/drug effects , Gene Silencing , Genes, Reporter , Hep G2 Cells , Hepatocytes/cytology , Hepatocytes/drug effects , Humans , Isoenzymes/antagonists & inhibitors , Isoenzymes/chemistry , Isoenzymes/genetics , Isoenzymes/metabolism , Isoxazoles/pharmacology , Mechanistic Target of Rapamycin Complex 2 , Multiprotein Complexes/metabolism , Mutation , Phosphatidic Acids/metabolism , Phosphorylation/drug effects , Promoter Regions, Genetic/drug effects , Protein Processing, Post-Translational/drug effects , Receptors, Cytoplasmic and Nuclear/agonists , Receptors, Cytoplasmic and Nuclear/genetics , Recombinant Proteins/agonists , Recombinant Proteins/antagonists & inhibitors , Recombinant Proteins/metabolism , Signal Transduction/drug effects , TOR Serine-Threonine Kinases/metabolism
7.
J Clin Endocrinol Metab ; 98(7): 2876-86, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23678037

ABSTRACT

CONTEXT: The clinical effectiveness of ablative radioiodine treatment of thyroid tumors is limited by the availability of the sodium iodide symporter (NIS) at the plasma membrane (PM) for uptake of ¹³¹I. A significant proportion of well-differentiated thyroid tumors are unable to concentrate sufficient radioiodine for effective therapy, and in other tumor models such as breast tumors, where radioiodine uptake would be an attractive therapeutic option, uptake is insufficient. OBJECTIVE: Pituitary tumor-transforming gene-binding factor (PBF; PTTG1IP) is overexpressed in multiple cancers and significantly decreases NIS expression at the PM. The goal of this study was to identify a method by which PBF repression of NIS may be overcome in human tumors. RESULTS: Here, we identify PBF as a tyrosine phosphoprotein that specifically binds the proto-oncogene tyrosine protein kinase Src in mass spectrometry, glutathione S-transferase pulldown and coimmunoprecipitation assays. Src induction leads to phosphorylation at PBF residue Y174. Abrogation of this residue results in PM retention and a markedly reduced ability to bind NIS. The Src inhibitor PP1 inhibits PBF phosphorylation in multiple cell lines in vitro, including human primary thyroid cells. Of direct clinical importance to the treatment of thyroid cancer, PP1 stimulates iodide uptake by transfected NIS in TPC1 thyroid carcinoma cells and entirely overcomes PBF repression of iodide uptake in human primary thyroid cells. CONCLUSIONS: We propose that targeting PBF phosphorylation at residue Y174 via tyrosine kinase inhibitors may be a novel therapeutic strategy to enhance the efficacy of ablative radioiodine treatment in thyroid and other endocrine and endocrine-related tumors.


Subject(s)
Cell Membrane/metabolism , Membrane Proteins/metabolism , Neoplasm Proteins/metabolism , Symporters/metabolism , Thyroid Gland/metabolism , Thyroid Neoplasms/metabolism , Amino Acid Substitution , Animals , Biological Transport/drug effects , COS Cells , Cell Line, Tumor , Cell Membrane/drug effects , Cell Membrane/pathology , Cells, Cultured , Chlorocebus aethiops , Humans , Intracellular Signaling Peptides and Proteins , Iodine Radioisotopes/metabolism , Membrane Proteins/genetics , Mutant Proteins/metabolism , Neoplasm Proteins/agonists , Neoplasm Proteins/antagonists & inhibitors , Neoplasm Proteins/genetics , Phosphorylation/drug effects , Protein Kinase Inhibitors/pharmacology , Protein Processing, Post-Translational/drug effects , Proto-Oncogene Mas , Proto-Oncogene Proteins pp60(c-src)/antagonists & inhibitors , Proto-Oncogene Proteins pp60(c-src)/metabolism , Radiopharmaceuticals/metabolism , Recombinant Proteins/agonists , Recombinant Proteins/antagonists & inhibitors , Recombinant Proteins/metabolism , Symporters/agonists , Symporters/genetics , Thyroid Gland/cytology , Thyroid Gland/drug effects , Thyroid Gland/pathology , Thyroid Neoplasms/drug therapy , Thyroid Neoplasms/pathology , Thyroid Neoplasms/radiotherapy
8.
Cancer Chemother Pharmacol ; 72(1): 189-99, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23673445

ABSTRACT

PURPOSE: Specific tyrosine kinase inhibitors were recently reported to modulate the activity of ABC transporters, leading to an increase in the intracellular concentration of their substrate drugs. In this study, we determine whether PD173074, a specific fibroblast growth factor receptor (FGFR) inhibitor, could reverse ABC transporter-mediated multidrug resistance. METHODS: 3-(4,5-Dimethylthiazol-yl)-2,5-diphenyllapatinibrazolium bromide assay was used to determine the effect of PD173074 on reversal of ABC transporter-mediated multidrug resistance (MDR). In addition, [³H]-paclitaxel accumulation/efflux assay, western blotting analysis, ATPase, and photoaffinity labeling assays were done to study the interaction of PD173074 on ABC transporters. RESULTS: PD173074 significantly sensitized both ABCB1-transfected and drug-selected cell lines overexpressing this transporter to substrate anticancer drugs colchicine, paclitaxel, and vincristine. This effect of PD173074 is specific to ABCB1, as no significant interaction was detected with other ABC transporters such as ABCC1 and ABCG2. The observed reversal effect seems to be primarily due to the decreased active efflux of [³H]-paclitaxel in ABCB1 overexpressing cells observed in efflux assay. In addition, no significant change in the ABCB1 expression was observed when ABCB1 overexpressing cells were exposed to 5 µM PD173074 for up to 3 days, thereby further suggesting its role in modulating the function of the transporter. In addition, PD173074 stimulated the ATPase activity of ABCB1 in a concentration-dependent manner, indicating a direct interaction with the transporter. Interestingly, PD173074 did not inhibit photolabeling of ABCB1 with [¹²5I]-iodoarylazidoprazosin (IAAP), showing that it binds at a site different from that of IAAP in the drug-binding pocket. CONCLUSIONS: Here, we report for the first time, PD173074, an inhibitor of the FGFR, to selectively reverse ABCB1 transporter-mediated MDR by directly blocking the efflux function of the transporter.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 1/antagonists & inhibitors , Antineoplastic Agents/agonists , Drug Resistance, Multiple/drug effects , Drug Resistance, Neoplasm/drug effects , Neoplasms/drug therapy , Pyrimidines/pharmacology , Receptors, Fibroblast Growth Factor/antagonists & inhibitors , ATP Binding Cassette Transporter, Subfamily B , ATP Binding Cassette Transporter, Subfamily B, Member 1/agonists , ATP Binding Cassette Transporter, Subfamily B, Member 1/genetics , ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , Adenosine Triphosphate/metabolism , Affinity Labels/pharmacology , Allosteric Regulation , Antineoplastic Agents/metabolism , Antineoplastic Agents/pharmacology , Biological Transport/drug effects , Cell Line, Tumor , Colchicine/agonists , Colchicine/pharmacology , HEK293 Cells , Humans , Hydrolysis/drug effects , Neoplasm Proteins/agonists , Neoplasm Proteins/antagonists & inhibitors , Neoplasm Proteins/metabolism , Neoplasms/enzymology , Neoplasms/metabolism , Paclitaxel/agonists , Paclitaxel/metabolism , Paclitaxel/pharmacology , Protein Kinase Inhibitors/pharmacology , Recombinant Proteins/agonists , Recombinant Proteins/antagonists & inhibitors , Recombinant Proteins/metabolism , Tubulin Modulators/agonists , Tubulin Modulators/metabolism , Tubulin Modulators/pharmacology , Vincristine/agonists , Vincristine/pharmacology
9.
J Clin Invest ; 123(4): 1750-62, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23478411

ABSTRACT

Type 2 diabetes (T2D) has emerged as a major threat to human health in most parts of the world. Therapeutic strategies aimed at improving pancreatic ß cell function are predicted to prove beneficial for the treatment of T2D. In the present study, we demonstrate that drug-mediated, chronic, and selective activation of ß cell G(q) signaling greatly improve ß cell function and glucose homeostasis in mice. These beneficial metabolic effects were accompanied by the enhanced expression of many genes critical for ß cell function, maintenance, and differentiation. By employing a combination of in vivo and in vitro approaches, we identified a novel ß cell pathway through which receptor-activated G(q) leads to the sequential activation of ERK1/2 and IRS2 signaling, thus triggering a series of events that greatly improve ß cell function. Importantly, we found that chronic stimulation of a designer G(q)-coupled receptor selectively expressed in ß cells prevented both streptozotocin-induced diabetes and the metabolic deficits associated with the consumption of a high-fat diet in mice. Since ß cells are endowed with numerous receptors that mediate their cellular effects via activation of G(q)-type G proteins, our findings provide a rational basis for the development of novel antidiabetic drugs targeting this class of receptors.


Subject(s)
Clozapine/analogs & derivatives , GTP-Binding Protein alpha Subunits, Gq-G11/metabolism , Hypoglycemic Agents/pharmacology , Insulin-Secreting Cells/drug effects , Receptors, G-Protein-Coupled/agonists , Animals , Cell Line, Tumor , Cell Proliferation , Clozapine/pharmacology , Diabetes Mellitus, Experimental/pathology , Diabetes Mellitus, Experimental/prevention & control , Drug Evaluation, Preclinical , Female , GTP-Binding Protein alpha Subunits, Gq-G11/genetics , Gene Expression/drug effects , Insulin Receptor Substrate Proteins/genetics , Insulin Receptor Substrate Proteins/metabolism , Insulin Receptor Substrate Proteins/physiology , Insulin-Secreting Cells/metabolism , Insulin-Secreting Cells/physiology , MAP Kinase Signaling System , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Molecular Targeted Therapy , Muscarinic Agonists/pharmacology , Protein Engineering , Receptor, Muscarinic M3/agonists , Receptor, Muscarinic M3/physiology , Receptors, G-Protein-Coupled/genetics , Receptors, G-Protein-Coupled/metabolism , Recombinant Proteins/agonists , Recombinant Proteins/genetics , Recombinant Proteins/metabolism
10.
Eur J Pharmacol ; 704(1-3): 15-22, 2013 Mar 15.
Article in English | MEDLINE | ID: mdl-23461853

ABSTRACT

The anti-diabetic drug glibenclamide inhibits K(ATP) channels in pancreatic ß-cells and stimulates insulin release. It also causes adverse effects, among which are abdominal pain, gastrointestinal disturbances and nocturia. We report that glibenclamide activates human TRPA1 in a concentration range that is commonly used to induce inhibition of K(ATP) channels in vitro. Glibenclamide generates calcium transients in HEK293t cells transiently transfected with human TRPA1, which are inhibited by the selective TRPA1 antagonist HC030031 and also evokes outwardly rectifying currents mediated by recombinant TRPA1. Glibenclamide activates a subpopulation of mouse primary sensory neurons, most of which are also sensitive to the selective TRPA1 agonist mustard oil. This glibenclamide sensitivity is completely abolished by genetic ablation of TRPA1. Taken together, our data demonstrate that glibenclamide is an agonist of human TRPA1, which may explain some of the adverse effects of the drug.


Subject(s)
Glyburide/pharmacology , Hypoglycemic Agents/pharmacology , Nerve Tissue Proteins/agonists , Transient Receptor Potential Channels/agonists , Acetanilides/pharmacology , Animals , Calcium/physiology , Calcium Channels/physiology , Cells, Cultured , Cysteine/physiology , Diazoxide/pharmacology , Ganglia, Spinal/cytology , HEK293 Cells , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Nerve Tissue Proteins/physiology , Purines/pharmacology , Recombinant Proteins/agonists , Sensory Receptor Cells/drug effects , Sensory Receptor Cells/physiology , TRPA1 Cation Channel , Transient Receptor Potential Channels/physiology
11.
Methods Mol Biol ; 995: 43-54, 2013.
Article in English | MEDLINE | ID: mdl-23494371

ABSTRACT

Calcium signaling plays a major role in the function of cells. Measurement of intracellular calcium mobilization is a robust assay that can be performed in a high-throughput manner to study the effect of compounds on potential drug targets. Pharmaceutical companies frequently use calcium signaling assays to screen compound libraries on G-protein-coupled receptors (GPCRs). In this chapter we describe the application of FLIPR technology to the evaluation of GPCR-induced calcium mobilization. We also include the implications of GPCR hetero-oligomerization and the identification of heteromeric receptors as novel drug targets on high-throughput calcium screening.


Subject(s)
Benzylidene Compounds/pharmacology , Drug Evaluation, Preclinical/methods , Enkephalin, D-Penicillamine (2,5)-/pharmacology , High-Throughput Screening Assays/methods , Naltrexone/analogs & derivatives , Receptors, Opioid, delta/agonists , Calcium Signaling , Cell Culture Techniques , HEK293 Cells , Humans , Naltrexone/pharmacology , Receptors, G-Protein-Coupled/agonists , Receptors, G-Protein-Coupled/antagonists & inhibitors , Receptors, G-Protein-Coupled/biosynthesis , Receptors, Opioid, delta/antagonists & inhibitors , Receptors, Opioid, delta/biosynthesis , Recombinant Proteins/agonists , Recombinant Proteins/antagonists & inhibitors , Recombinant Proteins/biosynthesis , Spectrometry, Fluorescence , Transfection
12.
Bioorg Med Chem ; 21(7): 2045-55, 2013 Apr 01.
Article in English | MEDLINE | ID: mdl-23395112

ABSTRACT

Selective CB2 agonists have the potential for treating pain without central CB1-mediated adverse effects. Screening efforts identified 1,2-dihydro-3-isoquinolone 1; however, this compound has the drawbacks of being difficult to synthesize with two asymmetric carbons on an isoquinolone scaffold and of having a highly lipophilic physicochemical property. To address these two major problems, we designed the 2-pyridone-based lead 15a, which showed moderate affinity for CB2. Optimization of 15a led to identification of 39f with high affinity for CB2 and selectivity over CB1. Prediction of the binding mode of 39f in complex with an active-state CB2 homology model provided structural insights into its high affinity for CB2.


Subject(s)
Drug Design , Pyridones/chemistry , Pyridones/pharmacology , Receptor, Cannabinoid, CB2/agonists , Receptor, Cannabinoid, CB2/metabolism , Catalytic Domain , Humans , Molecular Docking Simulation , Pyridones/chemical synthesis , Receptor, Cannabinoid, CB2/chemistry , Recombinant Proteins/agonists , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Structure-Activity Relationship
13.
Endocrinology ; 154(3): 1105-16, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23372019

ABSTRACT

Cinacalcet is predominantly used to treat secondary hyperparathyroidism due to end-stage renal failure, but, more recently, its potential clinical efficacy in treating patients with loss-of-function mutations in the calcium-sensing receptor (CaSR) has been recognized. Many clinically relevant CaSR mutations are located in the heptahelical membrane spanning and extracellular loop regions of the receptor, where allosteric modulators are predicted to bind. The aim of the present study was to investigate the impact of such mutations on the pharmacoregulation of the CaSR by the positive and negative allosteric modulators, cinacalcet and NPS-2143, respectively. Both cinacalcet and NPS-2143 effectively rescued mutants whose cell surface expression was substantially impaired, suggesting that both classes of drug can stabilize a receptor conformation that is trafficked more effectively to the cell surface. In addition, functional impairments in almost all mutant CaSRs were rescued by either cinacalcet or NPS-2143 via restoration of intracellular signaling. There was a significantly greater ability of both compounds to modulate agonist-stimulated intracellular Ca(2+) mobilization than ERK1/2 phosphorylation, indicating that the allosteric modulators engender bias in agonist-stimulated CaSR signaling to different pathways. Three mutations (G(670)R, P(748)R, and L(773)R) altered the binding affinity of allosteric modulators to the CaSR, and 3 mutations (V(817)I, L(773)R, and E(767)K) altered the cooperativity between the allosteric modulator and Ca(2+)(o). These findings have important implications for the treatment of diseases associated with CaSR mutations using allosteric CaSR modulators and for analyzing the effects of mutations on the function and pharmacoregulation of the CaSR.


Subject(s)
Mutation , Receptors, Calcium-Sensing/genetics , Receptors, Calcium-Sensing/metabolism , Allosteric Regulation , Amino Acid Substitution , Calcium Signaling/drug effects , Cinacalcet , Extracellular Signal-Regulated MAP Kinases/metabolism , HEK293 Cells , Humans , Mutant Proteins/agonists , Mutant Proteins/antagonists & inhibitors , Mutant Proteins/genetics , Mutant Proteins/metabolism , Naphthalenes/pharmacology , Receptors, Calcium-Sensing/agonists , Receptors, Calcium-Sensing/antagonists & inhibitors , Recombinant Proteins/agonists , Recombinant Proteins/antagonists & inhibitors , Recombinant Proteins/genetics , Recombinant Proteins/metabolism
14.
FEBS Lett ; 587(4): 339-44, 2013 Feb 14.
Article in English | MEDLINE | ID: mdl-23333295

ABSTRACT

Xylella fastidiosa is responsible for a wide range of economically important plant diseases. We report here the crystal structure and kinetic data of Xylellain, the first cysteine protease characterized from the genome of the pathogenic X. fastidiosa strain 9a5c. Xylellain has a papain-family fold, and part of the N-terminal sequence blocks the enzyme active site, thereby mediating protein activity. One novel feature identified in the structure is the presence of a ribonucleotide bound outside the active site. We show that this ribonucleotide plays an important regulatory role in Xylellain enzyme kinetics, possibly functioning as a physiological mediator.


Subject(s)
Bacterial Proteins/chemistry , Cysteine Proteases/chemistry , Models, Molecular , Xylella/enzymology , Amino Acid Substitution , Bacterial Proteins/agonists , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Biocatalysis , Catalytic Domain , Crystallography, X-Ray , Cysteine Proteases/genetics , Cysteine Proteases/metabolism , Cysteine Proteinase Inhibitors/pharmacology , Enzyme Activation , Kinetics , Mutagenesis, Site-Directed , Mutant Proteins/agonists , Mutant Proteins/antagonists & inhibitors , Mutant Proteins/chemistry , Mutant Proteins/metabolism , Point Mutation , Protein Folding , Protein Structure, Quaternary , Recombinant Proteins/agonists , Recombinant Proteins/antagonists & inhibitors , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Uridine Diphosphate/chemistry , Uridine Diphosphate/metabolism
15.
Diabetes ; 62(5): 1453-63, 2013 May.
Article in English | MEDLINE | ID: mdl-23305646

ABSTRACT

Glucagon, an essential regulator of glucose homeostasis, also modulates lipid metabolism and promotes weight loss, as reflected by the wasting observed in glucagonoma patients. Recently, coagonist peptides that include glucagon agonism have emerged as promising therapeutic candidates for the treatment of obesity and diabetes. We developed a novel stable and soluble glucagon receptor (GcgR) agonist, which allowed for in vivo dissection of glucagon action. As expected, chronic GcgR agonism in mice resulted in hyperglycemia and lower body fat and plasma cholesterol. Notably, GcgR activation also raised hepatic expression and circulating levels of fibroblast growth factor 21 (FGF21). This effect was retained in isolated primary hepatocytes from wild-type (WT) mice, but not GcgR knockout mice. We confirmed this link in healthy human volunteers, where injection of natural glucagon increased plasma FGF21 within hours. Functional relevance was evidenced in mice with genetic deletion of FGF21, where GcgR activation failed to induce the body weight loss and lipid metabolism changes observed in WT mice. Taken together, these data reveal for the first time that glucagon controls glucose, energy, and lipid metabolism at least in part via FGF21-dependent pathways.


Subject(s)
Fibroblast Growth Factors/metabolism , Glucagon/metabolism , Hepatocytes/metabolism , Receptors, Glucagon/metabolism , Adult , Animals , Anti-Obesity Agents/chemical synthesis , Anti-Obesity Agents/pharmacokinetics , Anti-Obesity Agents/pharmacology , Anti-Obesity Agents/therapeutic use , Cells, Cultured , Cross-Over Studies , Diabetes Mellitus, Type 2/blood , Diabetes Mellitus, Type 2/drug therapy , Diabetes Mellitus, Type 2/metabolism , Double-Blind Method , Female , Fibroblast Growth Factors/blood , Fibroblast Growth Factors/genetics , Glucagon/agonists , Glucagon/pharmacology , HEK293 Cells , Hepatocytes/drug effects , Hepatocytes/pathology , Humans , Hypoglycemic Agents/chemical synthesis , Hypoglycemic Agents/pharmacokinetics , Hypoglycemic Agents/pharmacology , Hypoglycemic Agents/therapeutic use , Insulin Resistance , Male , Mice , Mice, Knockout , Mice, Mutant Strains , Molecular Targeted Therapy , Obesity/blood , Obesity/drug therapy , Obesity/metabolism , Peptides/chemical synthesis , Peptides/pharmacokinetics , Peptides/physiology , Peptides/therapeutic use , Rats , Receptors, Glucagon/agonists , Receptors, Glucagon/genetics , Recombinant Proteins/agonists , Recombinant Proteins/metabolism
16.
Mol Cell Endocrinol ; 365(1): 36-43, 2013 Jan 05.
Article in English | MEDLINE | ID: mdl-22975079

ABSTRACT

Endocrine disrupting chemicals (EDCs) have emerged as a major public health issue because of their potentially disruptive effects on physiological hormonal actions. SXR (steroid xenobiotic receptor), also known as NR1I2, regulates CYP3A expression in response to exogenous chemicals, such as EDCs, after binding to SXRE (SXR response element). In our study, luciferase assay showed that 14 out of 55 EDCs could enhance SXR-mediated rat or human CYP3A gene transcription nearly evenly, and could also activate rat CYP7A1 gene transcription by cross-interaction of SXR and LXRE (LXRα response element). SXR diffused in the nucleus without ligand, whereas intranuclear foci of liganded SXR were produced. Furthermore, endogenous mRNA expression of CYP3A4 gene was enhanced by the 14 positive EDCs. Our results suggested a probable mechanism of EDCs disrupting the steroid or xenobiotic metabolism homeostasis via SXR.


Subject(s)
Cholesterol 7-alpha-Hydroxylase/biosynthesis , Cytochrome P-450 CYP3A/biosynthesis , Endocrine Disruptors/pharmacology , Kidney/drug effects , Liver/drug effects , Receptors, Steroid/agonists , Transcriptional Activation/drug effects , Animals , Cell Line , Chlorocebus aethiops , Cholesterol 7-alpha-Hydroxylase/genetics , Cytochrome P-450 CYP3A/genetics , Endocrine Disruptors/toxicity , Genes, Reporter/drug effects , Hep G2 Cells , Humans , Kidney/cytology , Kidney/enzymology , Kidney/metabolism , Liver/enzymology , Liver/metabolism , Liver X Receptors , Membrane Proteins/biosynthesis , Membrane Proteins/genetics , Orphan Nuclear Receptors/agonists , Orphan Nuclear Receptors/chemistry , Orphan Nuclear Receptors/genetics , Orphan Nuclear Receptors/metabolism , Pregnane X Receptor , Promoter Regions, Genetic/drug effects , Protein Transport/drug effects , Rats , Receptors, Steroid/chemistry , Receptors, Steroid/genetics , Receptors, Steroid/metabolism , Recombinant Proteins/agonists , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Response Elements/drug effects
17.
Nucleic Acid Ther ; 23(1): 35-43, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23113766

ABSTRACT

Induction of an effective immune response that can target and eliminate malignant cells or virus-infected cells requires the stimulation of antigen-specific effector T cells. A productive and long-lasting memory response requires 2 signals: a specific signal provided by antigen recognition through engagement of the T cell receptor and a secondary signal via engagement of costimulatory molecules (such as OX40) on these newly activated T cells. The OX40-OX40-ligand interaction is critical for the generation of productive effector and memory T cell functions. Thus agonistic antibodies that stimulate OX40 on activated T cells have been used as adjuvants to augment immune responses. We previously demonstrated that an aptamer modified to stimulate murine OX40 enhanced vaccine-mediated immune responses in a murine melanoma model. In this study, we describe the development of an agonistic aptamer that targets human OX40 (hOX40). This hOX40 aptamer was isolated using systematic evolution of ligands by exponential enrichment and binds the target purified protein with high affinity [dissociation constants (K(d))<10 nM]. Moreover, the hOX40 aptamer-streptavidin complex has an apparent binding affinity of ~50 nM for hOX40 on activated T cells as determined by flow cytometry and specifically binds activated human T cells. A multivalent version of the aptamer, but not a mutant version of the aptamer, was able to stimulate OX40 on T cells and enhance cell proliferation and interferon-gamma production. Future studies will assess the therapeutic potential of hOX40 aptamers for ex vivo stimulation of antigen specific T cells in conjunction with dendritic cell-based vaccines for adoptive cellular therapy.


Subject(s)
Aptamers, Nucleotide/pharmacology , Receptors, OX40/agonists , Adjuvants, Immunologic/pharmacology , Aptamers, Nucleotide/genetics , Aptamers, Nucleotide/immunology , Base Sequence , CD4-Positive T-Lymphocytes/drug effects , CD4-Positive T-Lymphocytes/immunology , Humans , Immunotherapy, Adoptive , Nucleic Acid Conformation , OX40 Ligand/metabolism , Receptors, OX40/genetics , Receptors, OX40/metabolism , Recombinant Proteins/agonists , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , SELEX Aptamer Technique , Signal Transduction
18.
Bioorg Med Chem ; 21(2): 436-47, 2013 Jan 15.
Article in English | MEDLINE | ID: mdl-23245803

ABSTRACT

A series of 2-hydrazinyladenosine derivatives was synthesized and investigated in radioligand binding studies for their affinity at the adenosine receptor subtypes with the goal to obtain potent and A(2A)AR selective agonists and to explore the structure-activity relationships of this class of compounds at A(2A)AR. Modifications included introduction of a second sugar moiety at position 2 of adenosine to form new bis-sugar nucleosides and/or modifications of the 2-position linker in different ways. The performed modifications were found to produce compounds with relatively high A(2A)AR affinity and very high selectivity toward A(2A)AR. The most potent bis-sugar nucleoside was obtained with the D-galactose derivative 16 which exhibited a K(i) value of 329 nM at A(2A)AR with marked selectivity against the other AR subtypes. In another set of compounds, compound 3 was modified via replacement of its cyclic structure with mono- and disubstituted phenyl moieties and the resulting hydrazones 10-14 were found to have low nanomolar affinity for A(2A)AR. In addition to 3, compounds 10, 11 and 13 have been identified as the most potent compounds in the present series with K(i) values of 16.1, 24.4, and 12.0 nM, respectively, at rat A(2A)AR. Species differences were tested and found to exist in different rates. Functional properties of the most potent compounds 10, 11, 13 and 16 were assessed showing that the compounds acted as agonists at A(2A)AR.


Subject(s)
Adenosine A2 Receptor Agonists/chemical synthesis , Adenosine/analogs & derivatives , Hydrazones/chemistry , Receptor, Adenosine A2A/metabolism , Adenosine/chemical synthesis , Adenosine/metabolism , Adenosine A2 Receptor Agonists/chemistry , Adenosine A2 Receptor Agonists/metabolism , Animals , Brain/metabolism , Humans , Kinetics , Ligands , Protein Binding , Rats , Receptor, Adenosine A2A/chemistry , Receptor, Adenosine A2A/genetics , Recombinant Proteins/agonists , Recombinant Proteins/biosynthesis , Recombinant Proteins/genetics , Structure-Activity Relationship
19.
Br J Pharmacol ; 168(6): 1421-9, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23083095

ABSTRACT

BACKGROUND AND PURPOSE: Previous work in our laboratory showed opioid agents inhibit cytokine expression in astrocytes. Recently, Watkins and colleagues hypothesized that opioid agonists activate toll-like receptor 4 (TLR4) signalling, which leads to neuroinflammation. To test this hypothesis, we characterized LPS and opioid effects on TLR4 signalling in reporter cells. EXPERIMENTAL APPROACH: NF-κB reporter cells expressing high levels of TLR4 were used to compare LPS and opioid effects on NF-κB activation, a pathway activated by TLR4 stimulation. KEY RESULTS: LPS increased TLR4 signalling in a concentration-dependent manner and was antagonized by LPS antagonist (LPS-RS, from Rhodobacter sphaeroides). A concentration ratio analysis showed that LPS-RS was a competitive antagonist. The opioid agonists, morphine and fentanyl, produced minor activation of TLR4 signalling when given alone. When tested following LPS stimulation, opioid agonists inhibited NF-κB activation but this inhibition was not blocked by the general opioid antagonist, naloxone, nor by the selective µ opioid receptor antagonist, ß-FNA. Indeed, both naloxone and ß-FNA also inhibited NF-κB activation in reporter cells. Further examination of fentanyl and ß-FNA effects revealed that both opioid agents inhibited LPS signalling in a non-competitive fashion. CONCLUSIONS AND IMPLICATIONS: These results show that LPS-RS is a competitive antagonist at the TLR4 complex, and that both opioid agonists and antagonists inhibit LPS signalling in a non-competitive fashion through a non-GPCR, opioid site(s) in the TLR4 signalling pathway. If confirmed, existing opioid agents or other drug molecules more selective at this novel site may provide a new therapeutic approach to the treatment of neuroinflammation.


Subject(s)
Analgesics, Opioid/pharmacology , Lipopolysaccharides/pharmacology , Nerve Tissue Proteins/metabolism , Signal Transduction/drug effects , Toll-Like Receptor 4/metabolism , Analgesics, Opioid/agonists , Analgesics, Opioid/antagonists & inhibitors , Binding, Competitive , Drug Antagonism , Escherichia coli K12/metabolism , Genes, Reporter/drug effects , HEK293 Cells , Humans , Kinetics , Ligands , Lipopolysaccharide Receptors/genetics , Lipopolysaccharide Receptors/metabolism , Lipopolysaccharides/antagonists & inhibitors , Lymphocyte Antigen 96/genetics , Lymphocyte Antigen 96/metabolism , NF-kappa B p50 Subunit/agonists , NF-kappa B p50 Subunit/genetics , NF-kappa B p50 Subunit/metabolism , Narcotic Antagonists/chemistry , Narcotic Antagonists/pharmacology , Nerve Tissue Proteins/agonists , Nerve Tissue Proteins/antagonists & inhibitors , Nerve Tissue Proteins/genetics , Receptors, Opioid, mu/agonists , Receptors, Opioid, mu/antagonists & inhibitors , Receptors, Opioid, mu/metabolism , Recombinant Proteins/agonists , Recombinant Proteins/antagonists & inhibitors , Recombinant Proteins/metabolism , Rhodobacter sphaeroides/metabolism , Toll-Like Receptor 4/agonists , Toll-Like Receptor 4/antagonists & inhibitors , Toll-Like Receptor 4/genetics
20.
Br J Pharmacol ; 168(6): 1430-44, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23083124

ABSTRACT

BACKGROUND AND PURPOSE: Palmitoylethanolamide (PEA) is an endogenous fatty acid amide displaying anti-inflammatory and analgesic actions. To investigate the molecular mechanism responsible for these effects, the ability of PEA and of pain-inducing stimuli such as capsaicin (CAP) or bradykinin (BK) to influence intracellular calcium concentrations ([Ca²âº](i)) in peripheral sensory neurons, has been assessed in the present study. The potential involvement of the transcription factor PPARα and of TRPV1 channels in PEA-induced effects was also studied. EXPERIMENTAL APPROACH: [Ca²âº](i) was evaluated by single-cell microfluorimetry in differentiated F11 cells. Activation of TRPV1 channels was assessed by imaging and patch-clamp techniques in CHO cells transiently-transfected with rat TRPV1 cDNA. KEY RESULTS: In F11 cells, PEA (1-30 µM) dose-dependently increased [Ca²âº](i). The TRPV1 antagonists capsazepine (1 µM) and SB-366791 (1 µM), as well as the PPARα antagonist GW-6471 (10 µM), inhibited PEA-induced [Ca²âº](i) increase; blockers of cannabinoid receptors were ineffective. PEA activated TRPV1 channels heterologously expressed in CHO cells; this effect appeared to be mediated at least in part by PPARα. When compared with CAP, PEA showed similar potency and lower efficacy, and caused stronger TRPV1 currents desensitization. Sub-effective PEA concentrations, closer to those found in vivo, counteracted CAP- and BK-induced [Ca²âº](i) transients, as well as CAP-induced TRPV1 activation. CONCLUSIONS AND IMPLICATIONS: Activation of PPARα and TRPV1 channels, rather than of cannabinoid receptors, largely mediate PEA-induced [Ca²âº](i) transients in sensory neurons. Differential TRPV1 activation and desensitization by CAP and PEA might contribute to their distinct pharmacological profile, possibly translating into potentially relevant clinical differences.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Endocannabinoids/pharmacology , Ethanolamines/pharmacology , Nerve Tissue Proteins/agonists , PPAR alpha/agonists , Palmitic Acids/pharmacology , Sensory Receptor Cells/drug effects , TRPV Cation Channels/agonists , Tachyphylaxis , Action Potentials/drug effects , Amides , Animals , Anti-Inflammatory Agents, Non-Steroidal/antagonists & inhibitors , CHO Cells , Calcium Signaling/drug effects , Cell Line , Cricetinae , Cricetulus , Drug Antagonism , Endocannabinoids/antagonists & inhibitors , Ethanolamines/antagonists & inhibitors , Membrane Transport Modulators/antagonists & inhibitors , Membrane Transport Modulators/pharmacology , Mice , Nerve Tissue Proteins/antagonists & inhibitors , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , PPAR alpha/antagonists & inhibitors , PPAR alpha/metabolism , Palmitic Acids/antagonists & inhibitors , Rats , Recombinant Proteins/agonists , Recombinant Proteins/antagonists & inhibitors , Recombinant Proteins/metabolism , Sensory Receptor Cells/cytology , Sensory Receptor Cells/metabolism , TRPV Cation Channels/antagonists & inhibitors , TRPV Cation Channels/genetics , TRPV Cation Channels/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL