Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
Sci Signal ; 10(485)2017 Jun 27.
Article in English | MEDLINE | ID: mdl-28655862

ABSTRACT

Constitutive WNT activity drives the growth of various human tumors, including nearly all colorectal cancers (CRCs). Despite this prominence in cancer, no WNT inhibitor is currently approved for use in the clinic largely due to the small number of druggable signaling components in the WNT pathway and the substantial toxicity to normal gastrointestinal tissue. We have shown that pyrvinium, which activates casein kinase 1α (CK1α), is a potent inhibitor of WNT signaling. However, its poor bioavailability limited the ability to test this first-in-class WNT inhibitor in vivo. We characterized a novel small-molecule CK1α activator called SSTC3, which has better pharmacokinetic properties than pyrvinium, and found that it inhibited the growth of CRC xenografts in mice. SSTC3 also attenuated the growth of a patient-derived metastatic CRC xenograft, for which few therapies exist. SSTC3 exhibited minimal gastrointestinal toxicity compared to other classes of WNT inhibitors. Consistent with this observation, we showed that the abundance of the SSTC3 target, CK1α, was decreased in WNT-driven tumors relative to normal gastrointestinal tissue, and knocking down CK1α increased cellular sensitivity to SSTC3. Thus, we propose that distinct CK1α abundance provides an enhanced therapeutic index for pharmacological CK1α activators to target WNT-driven tumors.


Subject(s)
Antineoplastic Agents/pharmacology , Benzoates/pharmacology , Casein Kinase Ialpha/metabolism , Enzyme Activators/pharmacology , Neoplasms/drug therapy , Wnt Proteins/metabolism , Animals , Enzyme Activation , Gene Expression Regulation, Neoplastic , HCT116 Cells , Humans , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Nude , Neoplasm Metastasis , Organ Culture Techniques , Phosphorylation , Pyrvinium Compounds/chemistry , Signal Transduction , Surface Plasmon Resonance , Wnt Signaling Pathway , Xenograft Model Antitumor Assays , Xenopus laevis
2.
FEBS Open Bio ; 7(2): 174-186, 2017 02.
Article in English | MEDLINE | ID: mdl-28174684

ABSTRACT

The sweetener sucralose can signal through its GPCR receptor to induce insulin secretion from pancreatic ß cells, but the downstream signaling pathways involved are not well-understood. Here we measure responses to sucralose, glucagon-like peptide 1, and amino acids in MIN6 ß cells. Our data suggest a signaling axis, whereby sucralose induces calcium and cAMP, activation of ERK1/2, and site-specific phosphorylation of ribosomal protein S6. Interestingly, sucralose acted independently of mTORC1 or ribosomal S6 kinase (RSK). These results suggest that sweeteners like sucralose can influence ß-cell responses to secretagogues like glucose through metabolic as well as GPCR-mediated pathways. Future investigation of novel sweet taste receptor signaling pathways in ß cells will have implications for diabetes and other emergent fields involving these receptors.

3.
Mol Cell Endocrinol ; 419: 60-71, 2016 Jan 05.
Article in English | MEDLINE | ID: mdl-26435461

ABSTRACT

We previously reported that INS-1 cells expressing the intracellular II-III loop of the L-type Ca(2+) channel Cav1.2 (Cav1.2/II-III cells) are deficient in Ca(2+)-induced Ca(2+) release (CICR). Here we show that glucose-stimulated ERK 1/2 phosphorylation (GSEP) is slowed and reduced in Cav1.2/II-III cells compared to INS-1 cells. This parallels a decrease in glucose-stimulated cAMP accumulation (GS-cAMP) in Cav1.2/II-III cells. Influx of Ca(2+) via L-type Ca(2+) channels and CICR play roles in both GSEP and GS-cAMP in INS-1 cells since both are inhibited by nicardipine or ryanodine. Further, the Epac1-selective inhibitor CE3F4 abolishes glucose-stimulated ERK activation in INS-1 cells, as measured using the FRET-based sensor EKAR. The non-selective Epac antagonist ESI-09 but not the Epac2-selective antagonist ESI-05 nor the PKA antagonist Rp-cAMPs inhibits GSEP in both INS-1 and Cav1.2/II-III cells. We conclude that L-type Ca(2+) channel-dependent cAMP accumulation, that's amplified by CICR, activates Epac1 and drives GSEP in INS-1 cells.


Subject(s)
Calcium Channels, L-Type/metabolism , Calcium/metabolism , Cyclic AMP/metabolism , Guanine Nucleotide Exchange Factors/metabolism , MAP Kinase Signaling System , Animals , Benzene Derivatives/pharmacology , Glucose/pharmacology , MAP Kinase Signaling System/drug effects , Nicardipine/pharmacology , Phosphorylation/drug effects , Quinolines/pharmacology , Rats , Ryanodine/pharmacology , Sulfones/pharmacology
4.
Mol Endocrinol ; 29(8): 1114-22, 2015 Aug.
Article in English | MEDLINE | ID: mdl-26168033

ABSTRACT

The MAPKs ERK1/2 respond to nutrients and other insulin secretagogues in pancreatic ß-cells and mediate nutrient-dependent insulin gene transcription. Nutrients also stimulate the mechanistic target of rapamycin complex 1 (mTORC1) to regulate protein synthesis. We showed previously that activation of both ERK1/2 and mTORC1 in the MIN6 pancreatic ß-cell-derived line by extracellular amino acids (AAs) is at least in part mediated by the heterodimeric T1R1/T1R3, a G protein-coupled receptor. We show here that AAs differentially activate these two signaling pathways in MIN6 cells. Pretreatment with pertussis toxin did not prevent the activation of either ERK1/2 or mTORC1 by AAs, indicating that G(I) is not central to either pathway. Although glucagon-like peptide 1, an agonist for a G(s-)coupled receptor, activated ERK1/2 well and mTORC1 to a small extent, AAs had no effect on cytosolic cAMP accumulation. Ca(2+) entry is required for ERK1/2 activation by AAs but is dispensable for AA activation of mTORC1. Pretreatment with UBO-QIC, a selective G(q) inhibitor, reduced the activation of ERK1/2 but had little effect on the activation of mTORC1 by AAs, suggesting a differential requirement for G(q). Inhibition of G(12/13) by the overexpression of the regulator of G protein signaling domain of p115 ρ-guanine nucleotide exchange factor had no effect on mTORC1 activation by AAs, suggesting that these G proteins are also not involved. We conclude that AAs regulate ERK1/2 and mTORC1 through distinct signaling pathways.


Subject(s)
Amino Acids/chemistry , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Multiprotein Complexes/metabolism , Receptors, G-Protein-Coupled/metabolism , Signal Transduction , TOR Serine-Threonine Kinases/metabolism , Animals , Calcium/metabolism , Cyclic AMP/metabolism , Endosomes/metabolism , Gene Expression Regulation , HeLa Cells , Humans , Insulin-Secreting Cells/cytology , Lysosomes/metabolism , Mechanistic Target of Rapamycin Complex 1 , Mice , Neurons/metabolism , Protein Multimerization
5.
Mol Biol Cell ; 25(11): 1782-92, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24719457

ABSTRACT

Cigarette smoking is a major risk factor for acquisition of small cell lung cancer (SCLC). A role has been demonstrated for the basic helix-loop-helix transcription factor NeuroD1 in the pathogenesis of neural and neuroendocrine lung cancer, including SCLC. In the present study we investigate the possible function of NeuroD1 in established tumors, as well as actions early on in pathogenesis, in response to nicotine. We demonstrate that nicotine up-regulates NeuroD1 in immortalized normal bronchial epithelial cells and a subset of undifferentiated carcinomas. Increased expression of NeuroD1 subsequently leads to regulation of expression and function of the nicotinic acetylcholine receptor subunit cluster of α3, α5, and ß4. In addition, we find that coordinated expression of these subunits by NeuroD1 leads to enhanced nicotine-induced migration and invasion, likely through changes in intracellular calcium. These findings suggest that aspects of the pathogenesis of neural and neuroendocrine lung cancers may be affected by a nicotine- and NeuroD1-induced positive feedback loop.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/metabolism , Carcinoma, Neuroendocrine/metabolism , Carcinoma, Neuroendocrine/pathology , Cell Movement/drug effects , Nerve Tissue Proteins/metabolism , Nicotine/pharmacology , Protein Subunits/metabolism , Receptors, Nicotinic/metabolism , Bronchi/pathology , Calcium/metabolism , Cell Differentiation/drug effects , Cell Line, Tumor , Clone Cells , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Extracellular Signal-Regulated MAP Kinases/metabolism , Humans , Lung Neoplasms/pathology , Models, Biological , Mutation , Neoplasm Invasiveness , Receptor, trkB/metabolism , Tumor Suppressor Protein p53/metabolism , Up-Regulation/drug effects
6.
J Biol Chem ; 289(20): 14370-9, 2014 May 16.
Article in English | MEDLINE | ID: mdl-24695728

ABSTRACT

We have shown recently that the class C G protein-coupled receptor T1R1/T1R3 taste receptor complex is an early amino acid sensor in MIN6 pancreatic ß cells. Amino acids are unable to activate ERK1/2 in ß cells in which T1R3 has been depleted. The muscarinic receptor agonist carbachol activated ERK1/2 better in T1R3-depleted cells than in control cells. Ligands that activate certain G protein-coupled receptors in pancreatic ß cells potentiate glucose-stimulated insulin secretion. Among these is the M3 muscarinic acetylcholine receptor, the major muscarinic receptor in ß cells. We found that expression of M3 receptors increased in T1R3-depleted MIN6 cells and that calcium responses were altered. To determine whether these changes were related to impaired amino acid signaling, we compared responses in cells exposed to reduced amino acid concentrations. M3 receptor expression was increased, and some, but not all, changes in calcium signaling were mimicked. These findings suggest that M3 acetylcholine receptors are increased in ß cells as a mechanism to compensate for amino acid deficiency.


Subject(s)
Amino Acids/metabolism , Insulin-Secreting Cells/metabolism , Receptor, Muscarinic M3/metabolism , Signal Transduction , Animals , Calcium/metabolism , Carbachol/pharmacology , Cell Line, Tumor , Enzyme Activation/drug effects , Gene Expression Regulation/drug effects , Insulin-Secreting Cells/cytology , Intracellular Space/drug effects , Intracellular Space/metabolism , Mice , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Phosphorylation/drug effects , Receptor, Muscarinic M3/genetics , Receptors, G-Protein-Coupled/deficiency , Receptors, G-Protein-Coupled/metabolism , Signal Transduction/drug effects
7.
Mol Endocrinol ; 28(4): 458-76, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24506535

ABSTRACT

We investigated the role of Cav1.2 in pancreatic ß-cell function by expressing a Cav1.2 II-III loop/green fluorescent protein fusion in INS-1 cells (Cav1.2/II-III cells) to disrupt channel-protein interactions. Neither block of KATP channels nor stimulation of membrane depolarization by tolbutamide was different in INS-1 cells compared with Cav1.2/II-III cells, but whole-cell Cav current density was significantly increased in Cav1.2/II-III cells. Tolbutamide (200 µM) stimulated insulin secretion and Ca(2+) transients in INS-1 cells, and Cav1.2/II-III cells were completely blocked by nicardipine (2 µM), but thapsigargin (1 µM) blocked tolbutamide-stimulated secretion and Ca(2+) transients only in INS-1 cells. Tolbutamide-stimulated endoplasmic reticulum [Ca(2+)] decrease was reduced in Cav1.2/II-III cells compared with INS-1 cells. However, Ca(2+) transients in both INS-1 cells and Cav1.2/II-III cells were significantly potentiated by 8-pCPT-2'-O-Me-cAMP (5 µM), FPL-64176 (0.5 µM), or replacement of extracellular Ca(2+) with Sr(2+). Glucose (10 mM) + glucagon-like peptide-1 (10 nM) stimulated discrete spikes in [Ca(2+)]i in the presence of verapamil at a higher frequency in INS-1 cells than in Cav1.2/II-II cells. Glucose (18 mM) stimulated more frequent action potentials in Cav1.2/II-III cells and primary rat ß-cells expressing the Cav1.2/II-II loop than in control cells. Further, apamin (1 µM) increased glucose-stimulated action potential frequency in INS-1 cells, but not Cav1.2/II-III cells, suggesting that SK channels were not activated under these conditions in Cav1.2/II-III loop-expressing cells. We propose the II-III loop of Cav1.2 as a key molecular determinant that couples the channel to Ca(2+)-induced Ca(2+) release and activation of SK channels in pancreatic ß-cells.


Subject(s)
Calcium Channels, L-Type/metabolism , Calcium/metabolism , Calcium/pharmacology , Insulin-Secreting Cells/drug effects , Insulin-Secreting Cells/metabolism , Small-Conductance Calcium-Activated Potassium Channels/metabolism , Action Potentials/drug effects , Animals , Calcium Channels, L-Type/chemistry , Cell Fractionation , Centrifugation, Density Gradient , Cyclic AMP/analogs & derivatives , Endocytosis/drug effects , Endoplasmic Reticulum/drug effects , Endoplasmic Reticulum/metabolism , Eukaryotic Initiation Factor-3/metabolism , Glucagon-Like Peptide 1/metabolism , Glucose/pharmacology , Immunoprecipitation , Insulin/metabolism , Insulin Secretion , Intracellular Space/drug effects , Intracellular Space/metabolism , Ion Channel Gating/drug effects , Male , Protein Structure, Secondary , Rats , Rats, Wistar , Tolbutamide/pharmacology , Verapamil/pharmacology , ras GTPase-Activating Proteins/metabolism
8.
Biochemistry ; 52(31): 5164-6, 2013 Aug 06.
Article in English | MEDLINE | ID: mdl-23848362

ABSTRACT

The mitogen-activated protein kinases (MAPKs) ERK1/2 regulate numerous cellular processes, including gene transcription, proliferation, and differentiation. The only known substrates of the MAP2Ks MEK1/2 are ERK1/2; thus, MEK inhibitors PD98059, U0126, and PD0325901 have been important tools in determining the functions of ERK1/2. By using these inhibitors and genetically manipulating MEK, we found that ERK1/2 activation is neither sufficient nor necessary for regulated secretion of insulin from pancreatic ß cells or secretion of epinephrine from chromaffin cells. We show that both PD98059 and U0126 reduce agonist-induced entry of calcium into cells in a manner independent of their ability to inhibit ERK1/2. Caution should be used when interpreting results from experiments using these compounds.


Subject(s)
Chromaffin Cells/drug effects , Insulin-Secreting Cells/drug effects , MAP Kinase Kinase 1/antagonists & inhibitors , MAP Kinase Kinase 2/antagonists & inhibitors , Protein Kinase Inhibitors/pharmacology , Animals , Benzamides/pharmacology , Butadienes/pharmacology , Calcium/metabolism , Cell Line , Chromaffin Cells/metabolism , Diphenylamine/analogs & derivatives , Diphenylamine/pharmacology , Epinephrine/metabolism , Flavonoids/pharmacology , Insulin/metabolism , Insulin Secretion , Insulin-Secreting Cells/metabolism , MAP Kinase Kinase 1/metabolism , MAP Kinase Kinase 2/metabolism , Mice , Mitogen-Activated Protein Kinase 3/metabolism , Nitriles/pharmacology
9.
Mol Pharmacol ; 83(1): 191-205, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23071106

ABSTRACT

Tolbutamide and gliclazide block the K(ATP) channel K(ir)6.2/Sur1, causing membrane depolarization and stimulating insulin secretion in pancreatic beta cells. We examined the ability of the EPAC-selective cAMP analog 8-pCPT-2'-O-Me-cAMP-AM to potentiate the action of these drugs and the mechanism that might account for it. Insulin secretion stimulated by both 200 µM tolbutamide and 20 µM gliclazide, concentrations that had equivalent effects on membrane potential, was inhibited by thapsigargin (1 µM) or the L-type Ca(2+) channel blocker nicardipine (2 µM) and was potentiated by 8-pCPT-2'-O-Me-cAMP-AM at concentrations ≥2 µM in INS-1 cells. Ca(2+) transients stimulated by either tolbutamide or gliclazide were inhibited by thapsigargin or nicardipine and were significantly potentiated by 8-pCPT-2'-O-Me-cAMP-AM at 5 µM but not 1 µM. Both tolbutamide and gliclazide stimulated phospholipase C activity; however, only gliclazide did so independently of its activity at K(ATP) channels, and this activity was partially inhibited by pertussis toxin. 8-pCPT-2'-O-Me-cAMP-AM alone (5 µM) did not stimulate insulin secretion, but did increase intracellular Ca(2+) concentration significantly, and this activity was inhibited by 25 µM 2-aminoethoxydiphenylborate (2-APB) or the removal of extracellular Ca(2+). 8-pCPT-2'-O-Me-cAMP-AM potentiation of insulin secretion stimulated by tolbutamide was markedly inhibited by 2-APB (25 µM) and enhanced by the PKC inhibitor bisindolylmaleimide I (1 µM). Our data demonstrate that the actions of both tolbutamide and gliclazide are strongly potentiated by 8-pCPT-2'-O-Me-cAMP-AM, that gliclazide can stimulate phospholipase C activity via a partially pertussis toxin-sensitive mechanism, and that 8-pCPT-2'-O-Me-cAMP-AM potentiation of tolbutamide action may involve activation of a 2-APB-sensitive Ca(2+) influx.


Subject(s)
Boron Compounds/pharmacology , Cyclic AMP/analogs & derivatives , Gliclazide/pharmacology , Guanine Nucleotide Exchange Factors/metabolism , Hypoglycemic Agents/pharmacology , Tolbutamide/pharmacology , Animals , Calcium/metabolism , Calcium Channels, L-Type/physiology , Cell Line, Tumor , Cyclic AMP/pharmacology , Drug Synergism , Enzyme Activation , GTP-Binding Protein alpha Subunits, Gi-Go/physiology , Indoles/pharmacology , Insulin/metabolism , Insulin Secretion , Intracellular Space/metabolism , KATP Channels/physiology , Maleimides/pharmacology , Membrane Potentials/drug effects , Patch-Clamp Techniques , Rats , Type C Phospholipases/antagonists & inhibitors , Type C Phospholipases/metabolism
10.
Mol Cell ; 47(6): 851-62, 2012 Sep 28.
Article in English | MEDLINE | ID: mdl-22959271

ABSTRACT

Cells continually assess their energy and nutrient state to maintain growth and survival and engage necessary homeostatic mechanisms. Cell-autonomous responses to the fed state require the surveillance of the availability of amino acids and other nutrients. The mammalian target of rapamycin complex 1 (mTORC1) integrates information on nutrient and amino acid availability to support protein synthesis and cell growth. We identify the G protein-coupled receptor (GPCR) T1R1/T1R3 as a direct sensor of the fed state and amino acid availability. Knocking down this receptor, which is found in most tissues, reduces the ability of amino acids to signal to mTORC1. Interfering with this receptor alters localization of mTORC1, downregulates expression of pathway inhibitors, upregulates key amino acid transporters, blocks translation initiation, and induces autophagy. These findings reveal a mechanism for communicating amino acid availability through a GPCR to mTORC1 in mammals.


Subject(s)
Autophagy , Insulin-Secreting Cells/metabolism , Proteins/metabolism , Receptors, G-Protein-Coupled/genetics , Receptors, G-Protein-Coupled/metabolism , Amino Acids/metabolism , Animals , Down-Regulation , Extracellular Signal-Regulated MAP Kinases/metabolism , Insulin/metabolism , Mechanistic Target of Rapamycin Complex 1 , Mice , Mice, Knockout , Multiprotein Complexes , Protein Biosynthesis , RNA Interference , RNA, Small Interfering , Signal Transduction , TOR Serine-Threonine Kinases
11.
J Pharmacol Exp Ther ; 331(2): 724-32, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19710366

ABSTRACT

The incretin peptides, glucose-dependent insulinotropic polypeptide (GIP) and glucagon-like peptide-1 (GLP-1), potentiate glucose-stimulated insulin secretion (GSIS) and beta-cell proliferation and differentiation. Ca(2+) influx via voltage-gated L-type Ca(2+) channels is required for GLP-1 and GIP potentiation of GSIS. We investigated the role of the L-type Ca(2+) channels Ca(v)1.2 and Ca(v)1.3 in mediating GLP-1- and GIP-stimulated events in INS-1 cells and INS-1 cell lines expressing dihydropyridine-insensitive (DHPi) mutants of either Ca(v)1.2 or Ca(v)1.3. Ca(v)1.3/DHPi channels supported full potentiation of GSIS by GLP-1 (50 nM) compared with untransfected INS-1 cells. However, GLP-1-potentiated GSIS mediated by Ca(v)1.2/DHPi channels was markedly reduced compared with untransfected INS-1 cells. In contrast, GIP (10 nM) potentiation of GSIS mediated by both Ca(v)1.2/DHPi and Ca(v)1.3/DHPi channels was similar to that observed in untransfected INS-1 cells. Disruption of intracellular Ca(2+) release with thapsigargin, ryanodine, or 2-aminoethyldiphenylborate and inhibition of protein kinase A (PKA) or protein kinase C (PKC) significantly reduced GLP-1 potentiation of GSIS by Ca(v)1.3/DHPi channels and by endogenous L-type channels in INS-1 cells, but not by Ca(v)1.2/DHPi channels. Inhibition of glucose-stimulated phospholipase C activity with 1-(6-((17b-3-methoxyestra-1,3,5(10)-trien-17-yl)amino)hexyl)-1H-pyrrole-2,5-dione (U73122) did not inhibit potentiation of GSIS by GLP-1 in INS-1 cells. In contrast, wortmannin, an inhibitor of phosphatidylinositol 3-kinase, and 2'-amino-3'-methoxyflavone (PD98059), an inhibitor of mitogen-activated protein kinase/extracellular signal-regulated kinase (ERK) kinase, both markedly inhibited GLP-1 potentiation of GSIS by endogenous channels in INS-1 cells and Ca(v)1.3/DHPi channels, but not by Ca(v)1.2/DHPi channels. Thus, Ca(v)1.3 is preferentially coupled to GLP-1 potentiation of GSIS in INS-1 cells via a mechanism that requires intact intracellular Ca(2+) stores, PKA and PKC activity, and activation of ERK1/2.


Subject(s)
Calcium Channels, L-Type/drug effects , Calcium Channels, T-Type/drug effects , Glucagon-Like Peptide 1/pharmacology , Glucose/pharmacology , Insulin-Secreting Cells/metabolism , Insulin/metabolism , Animals , Calcium Channel Blockers/pharmacology , Calcium Channels, L-Type/genetics , Calcium Channels, T-Type/genetics , Cell Line , Cyclic AMP-Dependent Protein Kinases/metabolism , Dihydropyridines/pharmacology , Gastric Inhibitory Polypeptide/pharmacology , Indicators and Reagents , Insulin-Secreting Cells/drug effects , Mutation , Phosphatidylinositol 4,5-Diphosphate/metabolism , Phosphatidylinositol 4,5-Diphosphate/physiology , Plasmids/genetics , Potassium Chloride/pharmacology , Protein Kinase C/metabolism , Rats , Signal Transduction/drug effects , Stimulation, Chemical
12.
J Pharmacol Exp Ther ; 330(1): 283-93, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19351867

ABSTRACT

L-type Ca(2+) channels play a key role in the integration of physiological signals regulating insulin secretion that probably requires their localization to specific subdomains of the plasma membrane. We investigated the role of the intracellular II-III loop domains of the L-type channels Ca(v)1.2 and 1.3 in coupling of Ca(2+) influx with glucose-stimulated insulin secretion (GSIS) potentiated by the incretin hormone glucagon-like peptide (GLP)-1. In INS-1 cell lines expressing the Ca(v)1.2/II-III or Ca(v)1.3/II-III peptides, GLP-1 potentiation of GSIS was inhibited markedly, coincident with a decrease in GLP-1-stimulated cAMP accumulation and the redistribution of Ca(v)1.2 and Ca(v)1.3 out of lipid rafts. Neither the Ca(v)1.2/II-III nor the Ca(v)1.3/II-III peptide decreased L-type current density compared with untransfected INS-1 cells. GLP-1 potentiation of GSIS was restored by the L-type channel agonist 2,5-dimethyl-4-[2-(phenylmethyl)benzoyl]-1H-pyrrole-3-carboxylic acid methyl ester (FPL-64176). In contrast, potentiation of GSIS by 8-bromo-cAMP (8-Br-cAMP) was inhibited in Ca(v)1.2/II-III but not Ca(v)1.3/II-III cells. These differences may involve unique protein-protein interactions because the Ca(v)1.2/II-III peptide, but not the Ca(v)1.3/II-III peptide, immunoprecipitates Rab3-interacting molecule (RIM) 2 from INS-1 cell lysates. RIM2, and its binding partner Piccolo, localize to lipid rafts, and they may serve as anchors for Ca(v)1.2 localization to lipid rafts in INS-1 cells. These findings suggest that the II-III interdomain loops of Ca(v)1.2, and possibly Ca(v)1.3, direct these channels to membrane microdomains in which the proteins that mediate potentiation of GSIS by GLP-1 and 8-Br-cAMP assemble.


Subject(s)
Calcium Channels, L-Type/physiology , Glucagon-Like Peptide 1/metabolism , Insulin/metabolism , Intracellular Fluid/physiology , Membrane Microdomains/metabolism , Animals , Calcium Channels, L-Type/biosynthesis , Calcium Channels, L-Type/genetics , Calcium Channels, L-Type/metabolism , Cell Line, Tumor , Glucagon-Like Peptide 1/physiology , Glucagon-Like Peptide-1 Receptor , Glucose/physiology , Humans , Insulin Secretion , Intracellular Fluid/chemistry , Intracellular Fluid/metabolism , Membrane Microdomains/physiology , Protein Interaction Mapping , Protein Structure, Secondary/physiology , Protein Structure, Tertiary/physiology , Rats , Receptors, Glucagon/metabolism , Receptors, Glucagon/physiology
13.
Mol Pharmacol ; 72(5): 1124-31, 2007 Nov.
Article in English | MEDLINE | ID: mdl-17673571

ABSTRACT

p21-Activated kinases (PAKs) are regulators of cell motility and proliferation. PAK activity is regulated in part by phosphoinositide-dependent kinase 1 (PDK1). We hypothesized that reduced PAK activity was involved in the effects of 2-amino-N-{4-[5-(2-phenanthrenyl)-3-(trifluoromethyl)-1H-pyrazol-1-yl]-phenyl} acetamide (OSU-03012), a previously characterized PDK1 inhibitor derived from celecoxib. In three human thyroid cancer cell lines, OSU-03012 inhibited cell proliferation with reduced AKT phosphorylation by PDK1. OSU-03012 unexpectedly inhibited PAK phosphorylation at lower concentrations than PDK1-dependent AKT phosphorylation in two of the three lines. In cell-free kinase assays, OSU-03012 was shown to inhibit PAK activity and compete with ATP binding. In addition, computer modeling predicted a docking site for OSU-03012 in the ATP binding motif of PAK1. Finally, overexpression of constitutively activated PAK1 partially rescued the ability of motile NPA thyroid cancer cells to migrate during OSU-03012 treatment, suggesting that inhibition of PAK may be involved in the cellular effects of OSU-03012 in these cells. In summary, OSU-03012 is a direct inhibitor of PAK, and inhibition of PAK, either directly or indirectly, may be involved in its biological effects in vitro.


Subject(s)
Antineoplastic Agents/pharmacology , Protein Kinase Inhibitors/pharmacology , Pyrazoles/pharmacology , Sulfonamides/pharmacology , Thyroid Neoplasms/enzymology , p21-Activated Kinases/antagonists & inhibitors , Adenosine Triphosphate/metabolism , Antineoplastic Agents/chemistry , Antineoplastic Agents/metabolism , Celecoxib , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Humans , Protein Kinase Inhibitors/chemistry , Protein Kinase Inhibitors/metabolism , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Proto-Oncogene Proteins c-akt/metabolism , Pyrazoles/chemistry , Pyrazoles/metabolism , Sulfonamides/chemistry , Sulfonamides/metabolism , p21-Activated Kinases/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...