Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 33
Filter
1.
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
2.
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
3.
Muscle Nerve ; 49(3): 378-88, 2014 Mar.
Article in English | MEDLINE | ID: mdl-23761265

ABSTRACT

INTRODUCTION: We investigated the mechanism by which the MERG1a K+ channel increases ubiquitin proteasome proteolysis (UPP). METHODS: Hindlimb suspension and electro-transfer of Merg1a cDNA into mouse gastrocnemius muscles induced atrophy. RESULTS: Atrophic gastrocnemius muscles of hindlimb-suspended mice express Merg1a, Murf1, and Mafbx genes. Electrotransfer of Merg1a significantly decreases muscle fiber size (12.6%) and increases UPP E3 ligase Murf1 mRNA (2.1-fold) and protein (23.7%), but does not affect Mafbx E3 ligase expression. Neither Merg1a-induced decreased fiber size nor Merg1a-induced increased Murf1 expression is curtailed significantly by coexpression of inactive HR-Foxo3a, a gene encoding a transcription factor known to induce Mafbx expression. CONCLUSIONS: The MERG1a K+ channel significantly increases expression of Murf1, but not Mafbx. We explored this expression pattern by expressing inactive Foxo3a and showing that it is not involved in MERG1a-mediated expression of Murf1. These findings suggest that MERG1a may not modulate Murf1 expression through the AKT/FOXO pathway.


Subject(s)
Ether-A-Go-Go Potassium Channels/metabolism , Gene Expression Regulation/genetics , Muscle Proteins/metabolism , SKP Cullin F-Box Protein Ligases/metabolism , Ubiquitin-Protein Ligases/metabolism , Analysis of Variance , Animals , ERG1 Potassium Channel , Ether-A-Go-Go Potassium Channels/genetics , Forkhead Box Protein O3 , Forkhead Transcription Factors/genetics , Forkhead Transcription Factors/metabolism , Functional Laterality , Gene Transfer Techniques , Hindlimb Suspension , Male , Mice , Muscle Proteins/genetics , Muscle, Skeletal , Muscular Atrophy/genetics , RNA, Messenger/metabolism , SKP Cullin F-Box Protein Ligases/genetics , Time Factors , Tripartite Motif Proteins , Ubiquitin-Protein Ligases/genetics
4.
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
5.
J Pharmacol Exp Ther ; 347(2): 276-87, 2013 Nov.
Article in English | MEDLINE | ID: mdl-24008337

ABSTRACT

Adenylyl cyclase (AC) isoforms are implicated in several physiologic processes and disease states, but advancements in the therapeutic targeting of AC isoforms have been limited by the lack of potent and isoform-selective small-molecule modulators. The discovery of AC isoform-selective small molecules is expected to facilitate the validation of AC isoforms as therapeutic targets and augment the study of AC isoform function in vivo. Identification of chemical probes for AC2 is particularly important because there are no published genetic deletion studies and few small-molecule modulators. The present report describes the development and implementation of an intact-cell, small-molecule screening approach and subsequent validation paradigm for the discovery of AC2 inhibitors. The NIH clinical collections I and II were screened for inhibitors of AC2 activity using PMA-stimulated cAMP accumulation as a functional readout. Active compounds were subsequently confirmed and validated as direct AC2 inhibitors using orthogonal and counterscreening assays. The screening effort identified SKF-83566 [8-bromo-2,3,4,5-tetrahydro-3-methyl-5-phenyl-1H-3-benzazepin-7-ol hydrobromide] as a selective AC2 inhibitor with superior pharmacological properties for selective modulation of AC2 compared with currently available AC inhibitors. The utility of SKF-83566 as a small-molecule probe to study the function of endogenous ACs was demonstrated in C2C12 mouse skeletal muscle cells and human bronchial smooth muscle cells.


Subject(s)
2,3,4,5-Tetrahydro-7,8-dihydroxy-1-phenyl-1H-3-benzazepine/analogs & derivatives , Adenylyl Cyclase Inhibitors , Enzyme Inhibitors/pharmacology , High-Throughput Screening Assays/methods , Small Molecule Libraries/pharmacology , 2,3,4,5-Tetrahydro-7,8-dihydroxy-1-phenyl-1H-3-benzazepine/chemistry , 2,3,4,5-Tetrahydro-7,8-dihydroxy-1-phenyl-1H-3-benzazepine/pharmacology , Adenylyl Cyclases/genetics , Animals , Cell Membrane/enzymology , Cell Membrane/immunology , Cyclic AMP/metabolism , Enzyme Inhibitors/chemistry , HEK293 Cells , Humans , Mice , Myocytes, Smooth Muscle/enzymology , Myocytes, Smooth Muscle/immunology , Sf9 Cells , Small Molecule Libraries/chemistry , Spodoptera , Transfection
6.
PLoS One ; 18(5): e0285316, 2023.
Article in English | MEDLINE | ID: mdl-37141277

ABSTRACT

The ER Ca2+ channel ryanodine receptor 2 (RyR2) is required for maintenance of insulin content and glucose-stimulated insulin secretion, in part, via regulation of the protein IRBIT in the insulinoma cell line INS-1. Here, we examined store-operated and depolarization-dependent Ca2+entry using INS-1 cells in which either RyR2 or IRBIT were deleted. Store-operated Ca2+ entry (SOCE) stimulated with thapsigargin was reduced in RyR2KO cells compared to controls, but was unchanged in IRBITKO cells. STIM1 protein levels were not different between the three cell lines. Basal and stimulated (500 µM carbachol) phospholipase C (PLC) activity was also reduced specifically in RyR2KO cells. Insulin secretion stimulated by tolbutamide was reduced in RyR2KO and IRBITKO cells compared to controls, but was potentiated by an EPAC-selective cAMP analog in all three cell lines. Cellular PIP2 levels were increased and cortical f-actin levels were reduced in RyR2KO cells compared to controls. Whole-cell Cav channel current density was increased in RyR2KO cells compared to controls, and barium current was reduced by acute activation of the lipid phosphatase pseudojanin preferentially in RyR2KO cells over control INS-1 cells. Action potentials stimulated by 18 mM glucose were more frequent in RyR2KO cells compared to controls, and insensitive to the SK channel inhibitor apamin. Taken together, these results suggest that RyR2 plays a critical role in regulating PLC activity and PIP2 levels via regulation of SOCE. RyR2 also regulates ß-cell electrical activity by controlling Cav current density and SK channel activation.


Subject(s)
Insulinoma , Pancreatic Neoplasms , Humans , Ryanodine Receptor Calcium Release Channel/metabolism , Calcium/metabolism , Cell Line , Glucose/pharmacology , Type C Phospholipases/metabolism
7.
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
8.
Diagnostics (Basel) ; 11(10)2021 Oct 12.
Article in English | MEDLINE | ID: mdl-34679577

ABSTRACT

BACKGROUND: The potassium channel encoded by the ether-a-gogo-related gene 1A (erg1a) has been detected in the atrophying skeletal muscle of mice experiencing either muscle disuse or cancer cachexia and further evidenced to contribute to muscle deterioration by enhancing ubiquitin proteolysis; however, to our knowledge, ERG1A has not been reported in human skeletal muscle. METHODS AND RESULTS: Here, using immunohistochemistry, we detect ERG1A immunofluorescence in human Rectus abdominis skeletal muscle sarcolemma. Further, using single point brightness data, we report the detection of ERG1A immunofluorescence at low levels in the Rectus abdominis muscle sarcolemma of young adult humans and show that it trends toward greater levels (10.6%) in healthy aged adults. Interestingly, we detect ERG1A immunofluorescence at a statistically greater level (53.6%; p < 0.05) in the skeletal muscle of older cancer patients than in age-matched healthy adults. Importantly, using immunoblot, we reveal that lower mass ERG1A protein is 61.5% (p < 0.05) more abundant in the skeletal muscle of cachectic older adults than in healthy age-matched controls. Additionally, we report that the ERG1A protein is detected in a cultured human rhabdomyosarcoma line that may be a good in vitro model for the study of ERG1A in muscle. CONCLUSIONS: The data demonstrate that ERG1A is detected more abundantly in the atrophied skeletal muscle of cancer patients, suggesting it may be related to muscle loss in humans as it has been shown to be in mice experiencing muscle atrophy as a result of malignant tumors.

9.
Skelet Muscle ; 10(1): 1, 2020 01 16.
Article in English | MEDLINE | ID: mdl-31948476

ABSTRACT

BACKGROUND: Skeletal muscle atrophy is the net loss of muscle mass that results from an imbalance in protein synthesis and protein degradation. It occurs in response to several stimuli including disease, injury, starvation, and normal aging. Currently, there is no truly effective pharmacological therapy for atrophy; therefore, exploration of the mechanisms contributing to atrophy is essential because it will eventually lead to discovery of an effective therapeutic target. The ether-a-go-go related gene (ERG1A) K+ channel has been shown to contribute to atrophy by upregulating ubiquitin proteasome proteolysis in cachectic and unweighted mice and has also been implicated in calcium modulation in cancer cells. METHODS: We transduced C2C12 myotubes with either a human ERG1A encoded adenovirus or an appropriate control virus. We used fura-2 calcium indicator to measure intracellular calcium concentration and Calpain-Glo assay kits (ProMega) to measure calpain activity. Quantitative PCR was used to monitor gene expression and immunoblot evaluated protein abundances in cell lysates. Data were analyzed using either a Student's t test or two-way ANOVAs and SAS software as indicated. RESULTS: Expression of human ERG1A in C2C12 myotubes increased basal intracellular calcium concentration 51.7% (p < 0.0001; n = 177). Further, it increased the combined activity of the calcium-activated cysteine proteases, calpain 1 and 2, by 31.9% (p < 0.08; n = 24); these are known to contribute to degradation of myofilaments. The increased calcium levels are likely a contributor to the increased calpain activity; however, the change in calpain activity may also be attributable to increased calpain protein abundance and/or a decrease in levels of the native calpain inhibitor, calpastatin. To explore the enhanced calpain activity further, we evaluated expression of calpain and calpastatin genes and observed no significant differences. There was no change in calpain 1 protein abundance; however, calpain 2 protein abundance decreased 40.7% (p < 0.05; n = 6). These changes do not contribute to an increase in calpain activity; however, we detected a 31.7% decrease (p < 0.05; n = 6) in calpastatin which could contribute to enhanced calpain activity. CONCLUSIONS: Human ERG1A expression increases both intracellular calcium concentration and combined calpain 1 and 2 activity. The increased calpain activity is likely a result of the increased calcium levels and decreased calpastatin abundance.


Subject(s)
Calcium/metabolism , Calpain/metabolism , ERG1 Potassium Channel/metabolism , Muscle Fibers, Skeletal/metabolism , Animals , Calcium-Binding Proteins/genetics , Calcium-Binding Proteins/metabolism , Calpain/genetics , Cell Line , Male , Mice
10.
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
11.
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
12.
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
13.
FASEB J ; 20(9): 1531-3, 2006 Jul.
Article in English | MEDLINE | ID: mdl-16723379

ABSTRACT

Skeletal muscle atrophy results from an imbalance in protein degradation and protein synthesis and occurs in response to injury, various disease states, disuse, and normal aging. Current treatments for this debilitating condition are inadequate. More information about mechanisms involved in the onset and progression of muscle atrophy is necessary for development of more effective therapies. Here we show that expression of the mouse ether-a-go-go related gene (Merg1a) K+ channel is up-regulated in skeletal muscle of mice experiencing atrophy as a result of both malignant tumor expression and disuse. Further, ectopic expression of Merg1a in vivo induces atrophy in healthy wt-bearing mice, while expression of a dysfunctional Merg1a mutant suppresses atrophy in hindlimb-suspended mice. Treatment of hindlimb-suspended mice with astemizole, a known Merg1a channel blocker, inhibits atrophy in these animals. Importantly, in vivo expression of Merg1a in mouse skeletal muscle activates the ubiquitin proteasome pathway that is responsible for the majority of protein degradation that causes muscle atrophy, yet expression of a dysfunctional Merg1a mutant decreases levels of ubiquitin-proteasome proteolysis. Thus, expression of Merg1a likely initiates atrophy by activating ubiquitin-proteasome proteolysis. This gene and its product are potential targets for prevention and treatment of muscle atrophy.


Subject(s)
Ether-A-Go-Go Potassium Channels/physiology , Muscle, Skeletal/pathology , Proteasome Endopeptidase Complex/metabolism , Ubiquitin/metabolism , Animals , Atrophy , Brain/physiology , ERG1 Potassium Channel , Esophageal Neoplasms , Hindlimb , Humans , KB Cells , Mice , Weight-Bearing
14.
Eur J Pharmacol ; 565(1-3): 7-16, 2007 Jun 22.
Article in English | MEDLINE | ID: mdl-17397827

ABSTRACT

Cardiac voltage-gated Ca2+ channels regulate the intracellular Ca2+ concentration and are therefore essential for muscle contraction, second messenger activation, gene expression and electrical signaling. As a first step in accessing the structural versus functional properties of the L-type Ca2+ channel in the heart, we have expressed a dihydropyridine (DHP)-insensitive CaV1.2 channel in rat ventricular myocytes and fibroblasts. Following isolation and culture, cells were infected with adenovirus expressing either LacZ or a mutant CaV1.2 channel (CaV1.2DHPi) containing the double mutation (T1039Y & Q1043M). This mutation renders the channel insensitive to neutral DHP compounds such as nisoldipine. The whole-cell, L-type Ca2+ current (ICa) measured in control myocytes was inhibited in a concentration-dependent manner by nisoldipine with an IC50 of 66 nM and complete block at 250 nM. In contrast, ICa in cells infected with AdCaV1.2DHPi was inhibited by only 35% by 500 nM nisoldipine but completely blocked by 50 microM diltiazem. In order to study CaV1.2DHPi in isolation, myocytes infected with AdCaV1.2DHPi were incubated with nisoldipine. Under this condition the cells expressed a large ICa (12 pA/pF) and displayed Ca2+ transients during field stimulation. Furthermore, addition of 2 microM forskolin and 100 microM 3-isobutyl-1-methylxanthine (IBMX), to stimulate protein kinase A, strongly increased IBa in the AdCaV1.2DHPi-infected cells. A Cd2+-sensitive IBa was also recorded in cardiac fibroblasts infected with AdCaV1.2DHPi. Thus, expression of CaV1.2DHPi will provide an important tool in studies of cardiac myocyte and fibroblast function.


Subject(s)
Adenoviridae/genetics , Calcium Channels, L-Type/genetics , Fibroblasts/metabolism , Myocytes, Cardiac/metabolism , Nisoldipine/pharmacology , Animals , Calcium/metabolism , Calcium Channels, L-Type/drug effects , Calcium Channels, L-Type/physiology , Cells, Cultured , Cyclic AMP-Dependent Protein Kinases/physiology , Diltiazem/pharmacology , Dose-Response Relationship, Drug , Heart Ventricles , Myocardial Contraction/drug effects , Rats
15.
J Mol Endocrinol ; 36(3): 503-15, 2006 Jun.
Article in English | MEDLINE | ID: mdl-16720719

ABSTRACT

SNARE-proteins (soluble NSF-attachment protein receptor) are important for Ca(2+)-dependent exocytosis. We have used capacitance measurements and confocal imaging to dissect the role of synaptosomal protein of 25 kDa (SNAP-25) and syntaxin 1 in rapid exocytosis in insulin-secreting pancreatic beta-cells. Following immunoneutralization of syntaxin 1 and SNAP-25, exocytosis was strongly reduced and associated with a marked reduction in the size of the readily releasable pool (RRP) by 65% and 86% in the presence of the anti-SNAP-25 and anti-syntaxin 1 antibodies respectively. The size of the immediately releasable pool (IRP), a subset of RRP in close association with the voltage-dependent Ca(2+)-channels, was reduced to an equal extent. The reduction in IRP correlated with slowed release kinetics and the time constant (tau) increased from a control value of 16 to 36 ms and 51 ms after inclusion of anti-SNAP-25 and anti-syntaxin 1 antibodies respectively in the pipette solution. We further show that SNAP-25 and syntaxin 1 aggregate in clusters along the plasma membrane. The size of these clusters was estimated to be approximately 300 nm and every beta-cell contained approximately 400 SNAP-25/syntaxin 1 clusters. Whereas the inhibitory action of the anti-syntaxin 1 antibody on exocytosis could be attributed almost entirely to suppression of the voltage-dependent Ca(2+)-current (-40%), the effect of the anti-SNAP-25 antibody was not mediated by decreased Ca(2+)-entry and is more likely due to a direct interference with the exocytotic machinery. Our data are consistent with the concept that both syntaxin 1 and SNAP-25 are required for rapid exocytosis in beta-cells.


Subject(s)
Antibodies/metabolism , Exocytosis/physiology , Insulin-Secreting Cells/metabolism , Synaptosomal-Associated Protein 25/antagonists & inhibitors , Synaptosomal-Associated Protein 25/metabolism , Syntaxin 1/antagonists & inhibitors , Syntaxin 1/metabolism , Animals , Calcium/metabolism , Calcium Channels/metabolism , Cells, Cultured , Insulin/metabolism , Insulin-Secreting Cells/cytology , Mice , Patch-Clamp Techniques , Rats , Synaptosomal-Associated Protein 25/genetics , Syntaxin 1/genetics
16.
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
17.
Methods Mol Biol ; 1474: 153-70, 2016.
Article in English | MEDLINE | ID: mdl-27515079

ABSTRACT

Bimolecular fluorescence complementation (BiFC) is a fluorescence imaging technique used to visualize protein-protein interactions (PPIs) in live cells and animals. One unique application of BiFC is to reveal subcellular localization of PPIs. The superior signal-to-noise ratio of BiFC in comparison with fluorescence resonance energy transfer or bioluminescence resonance energy transfer enables its wide applications. Here, we describe how confocal microscopy can be used to detect and quantify PPIs and their subcellular localization. We use basic leucine zipper transcription factor proteins as an example to provide a step-by-step BiFC protocol using a Nikon A1 confocal microscope and NIS-Elements imaging software. The protocol given below can be readily adapted for use with other confocal microscopes or imaging software.


Subject(s)
Microscopy, Confocal/statistics & numerical data , Optical Imaging/methods , Protein Interaction Mapping/methods , Activating Transcription Factor 2/genetics , Activating Transcription Factor 2/metabolism , Animals , COS Cells , Chlorocebus aethiops , Fluorescence Resonance Energy Transfer , Gene Expression , Nuclear Localization Signals/genetics , Nuclear Localization Signals/metabolism , Plasmids/chemistry , Plasmids/metabolism , Protein Binding , Proto-Oncogene Proteins c-fos/genetics , Proto-Oncogene Proteins c-fos/metabolism , Proto-Oncogene Proteins c-jun/genetics , Proto-Oncogene Proteins c-jun/metabolism , Proto-Oncogene Proteins c-myc/genetics , Proto-Oncogene Proteins c-myc/metabolism , Signal-To-Noise Ratio , Software
18.
Biochem Pharmacol ; 120: 46-55, 2016 11 15.
Article in English | MEDLINE | ID: mdl-27638414

ABSTRACT

The substituted amphetamine, 3,4-methylenedioxy-methamphetamine (MDMA, ecstasy), is a widely used drug of abuse that induces non-exocytotic release of serotonin, dopamine, and norepinephrine through their cognate transporters as well as blocking the reuptake of neurotransmitter by the same transporters. The resulting dramatic increase in volume transmission and signal duration of neurotransmitters leads to psychotropic, stimulant, and entactogenic effects. The mechanism by which amphetamines drive reverse transport of the monoamines remains largely enigmatic, however, promising outcomes for the therapeutic utility of MDMA for post-traumatic stress disorder and the long-time use of the dopaminergic and noradrenergic-directed amphetamines in treatment of attention-deficit hyperactivity disorder and narcolepsy increases the importance of understanding this phenomenon. Previously, we identified functional differences between the human and Drosophila melanogaster serotonin transporters (hSERT and dSERT, respectively) revealing that MDMA is an effective substrate for hSERT but not dSERT even though serotonin is a potent substrate for both transporters. Chimeric dSERT/hSERT transporters revealed that the molecular components necessary for recognition of MDMA as a substrate was linked to regions of the protein flanking transmembrane domains (TM) V through IX. Here, we performed species-scanning mutagenesis of hSERT, dSERT and C. elegans SERT (ceSERT) along with biochemical and electrophysiological analysis and identified a single amino acid in TM10 (Glu394, hSERT; Asn484, dSERT, Asp517, ceSERT) that is primarily responsible for the differences in MDMA recognition. Our findings reveal that an acidic residue is necessary at this position for MDMA recognition as a substrate and serotonin releaser.


Subject(s)
Caenorhabditis elegans Proteins/metabolism , Drosophila Proteins/metabolism , Hallucinogens/metabolism , N-Methyl-3,4-methylenedioxyamphetamine/metabolism , Serotonin Agents/metabolism , Serotonin Plasma Membrane Transport Proteins/metabolism , Amino Acid Substitution , Animals , Caenorhabditis elegans Proteins/chemistry , Caenorhabditis elegans Proteins/genetics , Drosophila Proteins/chemistry , Drosophila Proteins/genetics , Drosophila melanogaster , HEK293 Cells , Hallucinogens/pharmacology , Humans , Mutagenesis, Site-Directed , Mutation , N-Methyl-3,4-methylenedioxyamphetamine/pharmacology , Oocytes/drug effects , Oocytes/metabolism , Patch-Clamp Techniques , Peptide Fragments/chemistry , Peptide Fragments/genetics , Peptide Fragments/metabolism , Protein Interaction Domains and Motifs , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/metabolism , Serotonin/metabolism , Serotonin Agents/pharmacology , Serotonin Plasma Membrane Transport Proteins/chemistry , Serotonin Plasma Membrane Transport Proteins/genetics , Species Specificity , Substrate Specificity , Xenopus laevis
19.
J Med Chem ; 58(16): 6336-47, 2015 Aug 27.
Article in English | MEDLINE | ID: mdl-25856547

ABSTRACT

Since the discovery of the GABA(B) agonist and muscle relaxant baclofen, there have been substantial advancements in the development of compounds that activate the GABA(B) receptor as agonists or positive allosteric modulators. For the agonists, most of the existing structure-activity data apply to understanding the role of substituents on the backbone of GABA as well as replacing the carboxylic acid and amine groups. In the cases of the positive allosteric modulators, the allosteric binding site(s) and structure-activity relationships are less well-defined; however, multiple classes of molecules have been discovered. The recent report of the X-ray structure of the GABA(B) receptor with bound agonists and antagonists provides new insights for the development of compounds that bind the orthosteric site of this receptor. From a therapeutic perspective, these data have enabled efforts in drug discovery in areas of addiction-related behavior, the treatment of anxiety, and the control of muscle contractility.


Subject(s)
GABA Agonists/pharmacology , GABA Modulators/pharmacology , Receptors, GABA-B/drug effects , Animals , GABA Agonists/chemistry , GABA Antagonists/chemistry , GABA Antagonists/pharmacology , GABA Modulators/chemical synthesis , Humans , Molecular Conformation , Receptors, GABA-B/chemistry , Structure-Activity Relationship
20.
Chem Commun (Camb) ; (7): 784-5, 2004 Apr 07.
Article in English | MEDLINE | ID: mdl-15045062

ABSTRACT

Submicron-sized Au particles and Au/SiO(2) nanocomposites (superparticles) as large as 670 nm have been introduced into tsA201 cells with minimal cell trauma by cationic transfection systems. Successful implantations can be characterized by the expression of co-transfected DNA.


Subject(s)
Cations/chemistry , DNA/genetics , Nanotechnology/methods , Transfection/methods , Animals , Cell Line , Composite Resins/chemistry , DNA/chemistry , DNA/metabolism , Female , Fluorescence , Gold/chemistry , Humans , Microscopy, Electron/methods , Particle Size , Silicon Dioxide/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL