Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
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
2.
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
3.
Biochim Biophys Acta Mol Cell Res ; 1868(1): 118865, 2021 01.
Article in English | MEDLINE | ID: mdl-32980354

ABSTRACT

Zinc (Zn2+) is an essential micronutrient that is required for a wide variety of cellular processes. Tools and methods have been instrumental in revealing the myriad roles of Zn2+ in cells. This review highlights recent developments fluorescent sensors to measure the labile Zn2+ pool, chelators to manipulate Zn2+ availability, and fluorescent tools and proteomics approaches for monitoring Zn2+-binding proteins in cells. Finally, we close with some highlights on the role of Zn2+ in regulating cell function and in cell signaling.


Subject(s)
Biosensing Techniques , Carrier Proteins/isolation & purification , Signal Transduction/genetics , Zinc/isolation & purification , Carrier Proteins/chemistry , Fluorescent Dyes/chemistry , Fluorescent Dyes/isolation & purification , Humans , Micronutrients/chemistry , Micronutrients/metabolism , Zinc/chemistry , Zinc/metabolism
4.
ACS Sens ; 5(12): 3879-3891, 2020 12 24.
Article in English | MEDLINE | ID: mdl-33305939

ABSTRACT

Genetically encoded fluorescent sensors have been widely used to illuminate secretory vesicle dynamics and the vesicular lumen, including Zn2+ and pH, in living cells. However, vesicular sensors have a tendency to mislocalize and are susceptible to the acidic intraluminal pH. In this study, we performed a systematic comparison of five different vesicular proteins to target the fluorescent protein mCherry and a Zn2+ Förster resonance energy transfer (FRET) sensor to secretory vesicles. We found that motifs derived from vesicular cargo proteins, including chromogranin A (CgA), target vesicular puncta with greater efficacy than transmembrane proteins. To characterize vesicular Zn2+ levels, we developed CgA-Zn2+ FRET sensor fusions with existing sensors ZapCY1 and eCALWY-4 and characterized subcellular localization and the influence of pH on sensor performance. We simultaneously monitored Zn2+ and pH in individual secretory vesicles by leveraging the acceptor fluorescent protein as a pH sensor and found that pH influenced FRET measurements in situ. While unable to characterize vesicular Zn2+ at the single-vesicle level, we were able to monitor Zn2+ dynamics in populations of vesicles and detected high vesicular Zn2+ in MIN6 cells compared to lower levels in the prostate cancer cell line LnCaP. The combination of CgA-ZapCY1 and CgA-eCALWY-4 allows for measurement of Zn2+ from pM to nM ranges.


Subject(s)
Fluorescence Resonance Energy Transfer , Zinc , Cell Line , Hydrogen-Ion Concentration , Male , Secretory Vesicles
5.
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
6.
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
7.
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
8.
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
9.
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
10.
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
11.
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
...