Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters










Database
Language
Publication year range
1.
Sci Rep ; 12(1): 7713, 2022 05 11.
Article in English | MEDLINE | ID: mdl-35562179

ABSTRACT

The role of ER Ca2+ release via ryanodine receptors (RyR) in pancreatic ß-cell function is not well defined. Deletion of RyR2 from the rat insulinoma INS-1 (RyR2KO) enhanced IP3 receptor activity stimulated by 7.5 mM glucose, coincident with reduced levels of the protein IP3 Receptor Binding protein released with Inositol 1,4,5 Trisphosphate (IRBIT). Insulin content, basal (2.5 mM glucose) and 7.5 mM glucose-stimulated insulin secretion were reduced in RyR2KO and IRBITKO cells compared to controls. INS2 mRNA levels were reduced in both RyR2KO and IRBITKO cells, but INS1 mRNA levels were specifically decreased in RyR2KO cells. Nuclear localization of S-adenosylhomocysteinase (AHCY) was increased in RyR2KO and IRBITKO cells. DNA methylation of the INS1 and INS2 gene promotor regions was very low, and not different among RyR2KO, IRBITKO, and controls, but exon 2 of the INS1 and INS2 genes was more extensively methylated in RyR2KO and IRBITKO cells. Exploratory proteomic analysis revealed that deletion of RyR2 or IRBIT resulted in differential regulation of 314 and 137 proteins, respectively, with 41 in common. These results suggest that RyR2 regulates IRBIT levels and activity in INS-1 cells, and together maintain insulin content and secretion, and regulate the proteome, perhaps via DNA methylation.


Subject(s)
Insulinoma , Pancreatic Neoplasms , Animals , Cell Line , Glucose , Insulin/metabolism , Insulinoma/genetics , Pancreatic Neoplasms/genetics , Proteomics , RNA, Messenger , Rats , Ryanodine Receptor Calcium Release Channel/metabolism
2.
PLoS One ; 14(8): e0215188, 2019.
Article in English | MEDLINE | ID: mdl-31442224

ABSTRACT

Pancreatic ß-cells express multiple phosphodiesterase (PDE) subtypes, but the specific roles for each in ß-cell function, particularly in humans, is not clear. We evaluated the cellular role of PDE1, PDE3, and PDE4 activity in the rat insulinoma cell line INS-1 and in primary human ß-cells using subtype-selective PDE inhibitors. Using a genetically encoded, FRET-based cAMP sensor, we found that the PDE1 inhibitor 8MM-IBMX, elevated cAMP levels in the absence of glucose to a greater extent than either the PDE3 inhibitor cilostamide or the PDE4 inhibitor rolipram. In 18 mM glucose, PDE1 inhibition elevated cAMP levels to a greater extent than PDE3 inhibition in INS-1 cells, while PDE4 inhibition was without effect. Inhibition of PDE1 or PDE4, but not PDE3, potentiated glucose-stimulated insulin secretion in INS-1 cells. PDE1 inhibition, but not PDE3 or PDE4 inhibition, reduced palmitate-induced caspase-3/7 activation, and enhanced CREB phosphorylation in INS-1 cells. In human ß-cells, only PDE3 or PDE4 inhibition increased cAMP levels in 1.7 mM glucose, but PDE1, PDE3, or PDE4 inhibition potentiated cAMP levels in 16.7 mM glucose. Inhibition of PDE1 or PDE4 increased cAMP levels to a greater extent in 16.7 mM glucose than in 1.7 mM glucose in human ß-cells. In contrast, elevation of cAMP levels by PDE3 inhibition was not different at these glucose concentrations. PDE1 inhibition also potentiated insulin secretion from human islets, suggesting that the role of PDE1 may be conserved between INS-1 cells and human pancreatic ß-cells. Our results suggest that inhibition of PDE1 may be a useful strategy to potentiate glucose-stimulated insulin secretion, and to protect ß-cells from the toxic effects of excess fatty acids.


Subject(s)
3',5'-Cyclic-AMP Phosphodiesterases/metabolism , Cyclic AMP/metabolism , Insulin-Secreting Cells/metabolism , 3',5'-Cyclic-AMP Phosphodiesterases/antagonists & inhibitors , Adult , Calcium/metabolism , Cell Line , Cell Survival/drug effects , Cytosol/drug effects , Cytosol/metabolism , Female , Humans , Insulin-Secreting Cells/cytology , Insulin-Secreting Cells/drug effects , Male , Middle Aged , Phosphodiesterase Inhibitors/pharmacology , Stress, Physiological/drug effects
3.
Mol Pharmacol ; 94(3): 973-983, 2018 09.
Article in English | MEDLINE | ID: mdl-29980657

ABSTRACT

Nifedipine and FPL 64176 (FPL), which block and potentiate L-type voltage-gated Ca2+ channels, respectively, modulate Cav1.2 more potently than Cav1.3. To identify potential strategies for developing subtype-selective inhibitors, we investigated the role of divergent amino acid residues in transmembrane domains IIIS5 and the extracellular IIIS5-3P loop region in modulation of these channels by nifedipine and FPL. Insertion of the extracellular IIIS5-3P loop from Cav1.2 into Cav1.3 (Cav1.3+) reduced the IC50 of nifedipine from 289 to 101 nM, and substitution of S1100 with an A residue, as in Cav1.2, accounted for this difference. Substituting M1030 in IIIS5 to V in Cav1.3+ (Cav1.3+V) further reduced the IC50 of nifedipine to 42 nM. FPL increased current amplitude with an EC50 of 854 nM in Cav1.3, 103 nM in Cav1.2, and 99 nM in Cav1.3+V. In contrast to nifedipine block, substitution of M1030 to V in Cav1.3 had no effect on potency of FPL potentiation of current amplitude, but slowed deactivation in the presence and absence of 10 µM FPL. FPL had no effect on deactivation of Cav1.3/dihydropyridine-insensitive (DHPi), a channel with very low sensitivity to nifedipine block (IC50 ∼93 µM), but did shift the voltage-dependence of activation by ∼-10 mV. We conclude that the M/V variation in IIIS5 and the S/A variation in the IIIS5-3P loop of Cav1.2 and Cav1.3 largely determine the difference in nifedipine potency between these two channels, but the difference in FPL potency is determined by divergent amino acids in the IIIS5-3P loop.


Subject(s)
Calcium Channel Agonists/pharmacology , Calcium Channel Blockers/pharmacology , Calcium Channels, L-Type/physiology , Nifedipine/pharmacology , Pyrroles/pharmacology , Amino Acid Sequence , Calcium Channel Agonists/metabolism , Calcium Channel Blockers/metabolism , Calcium Channels, L-Type/chemistry , Dose-Response Relationship, Drug , HEK293 Cells , Humans , Nifedipine/metabolism , Protein Structure, Secondary , Pyrroles/metabolism
4.
Bioorg Med Chem Lett ; 28(16): 2697-2700, 2018 09 01.
Article in English | MEDLINE | ID: mdl-29657102

ABSTRACT

ß-Hydroxy difluoromethyl ketones represent the newest class of agonists of the GABA-B receptor, and they are structurally distinct from all other known agonists at this receptor because they do not display the carboxylic acid or amino group of γ-aminobutyric acid (GABA). In this report, the design, synthesis, and biological evaluation of additional analogues of ß-hydroxy difluoromethyl ketones characterized the critical nature of the substituted aromatic group on the lead compound. The importance of these new data is interpreted by docking studies using the X-ray structure of the GABA-B receptor. Moreover, we also report that the synthesis and biological evaluation of ß-amino difluoromethyl ketones provided the most potent compound across these two series.


Subject(s)
GABA-B Receptor Agonists/pharmacology , Ketones/pharmacology , Propylamines/pharmacology , Binding Sites , GABA-B Receptor Agonists/chemical synthesis , GABA-B Receptor Agonists/chemistry , HEK293 Cells , Humans , Ketones/chemical synthesis , Ketones/chemistry , Molecular Docking Simulation , Propylamines/chemical synthesis , Propylamines/chemistry , Receptors, GABA-B/chemistry , Stereoisomerism , Structure-Activity Relationship
5.
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
6.
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
7.
J Med Chem ; 56(6): 2456-65, 2013 Mar 28.
Article in English | MEDLINE | ID: mdl-23428109

ABSTRACT

The design, synthesis, biological evaluation, and in vivo studies of difluoromethyl ketones as GABAB agonists that are not structurally analogous to known GABAB agonists, such as baclofen or 3-aminopropyl phosphinic acid, are presented. The difluoromethyl ketones were assembled in three synthetic steps using a trifluoroacetate-release aldol reaction. Following evaluation at clinically relevant GABA receptors, we have identified a difluoromethyl ketone that is a potent GABAB agonist, obtained its X-ray structure, and presented preliminary in vivo data in alcohol-preferring mice. The behavioral studies in mice demonstrated that this compound tended to reduce the acoustic startle response, which is consistent with an anxiolytic profile. Structure-activity investigations determined that replacing the fluorines of the difluoromethyl ketone with hydrogens resulted in an inactive analogue. Resolution of the individual enantiomers of the difluoromethyl ketone provided a compound with full biological activity at concentrations less than an order of magnitude greater than the pharmaceutical, baclofen.


Subject(s)
GABA-B Receptor Agonists/chemistry , GABA-B Receptor Agonists/pharmacology , Ketones/chemistry , Ketones/pharmacology , Receptors, GABA-B/metabolism , Animals , Behavior, Animal/drug effects , Female , Halogenation , Male , Mice , Models, Molecular , Protein Conformation , Receptors, GABA-B/chemistry
8.
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
SELECTION OF CITATIONS
SEARCH DETAIL
...