Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
1.
Channels (Austin) ; 12(1): 378-387, 2018.
Article in English | MEDLINE | ID: mdl-30403912

ABSTRACT

Contributions of voltage sensing S4 segments in domains I - IV of CaV3.1 channel to channel activation were analyzed. Neutralization of the uppermost charge in individual S4 segments by exchange of arginine for cysteine was employed. Mutant channels with single exchange in domains I - IV, in two adjacent domains, and in all four domains were constructed and expressed in HEK 293 cells. Changes in maximal gating charge Qmax and the relation between Qmax and maximal conductance Gmax were evaluated. Qmax was the most affected by single mutation in domain I and by double mutations in domains I + II and I + IV. The ratio Gmax/Qmax proportional to opening probability of the channel was significantly decreased by the mutation in domain III and increased by mutations in domains I and II. In channels containing double mutations Gmax/Qmax ratio increased significantly when the mutation in domain I was included. Mutations in domains II and III zeroed each other. Mutation in domain IV prevented the decrease caused by the mutation in domain III. Neither ion current nor gating current was observed when channels with quadruple mutations were expressed. Immunocytochemistry analysis did not reveal the presence of channel protein in the cell membrane. Likely, quadruple mutation results in a structural change that affects the channel's trafficking mechanism. Altogether, S4 segments in domains I-IV of the CaV3.1 channel unequally contribute to channel gating by voltage. We suggest the most important role of the voltage sensor in the domain I and lesser roles of voltage sensors in domains II and III.


Subject(s)
Calcium Channels, T-Type/metabolism , Animals , Calcium Channels, T-Type/analysis , Calcium Channels, T-Type/genetics , Cell Membrane/chemistry , Cell Membrane/metabolism , HEK293 Cells , Humans , Mice , Mutation
2.
Biochem Biophys Res Commun ; 473(2): 396-402, 2016 Apr 29.
Article in English | MEDLINE | ID: mdl-26944020

ABSTRACT

T-type Ca(2+) channels are important regulators of peripheral sensory neuron excitability. Accordingly, T-type Ca(2+) currents are often increased in various pathological pain conditions, such as inflammation or nerve injury. Here we investigated effects of inflammation on functional expression of T-type Ca(2+) channels in small-diameter cultured dorsal root ganglion (DRG) neurons. We found that overnight treatment of DRG cultures with a cocktail of inflammatory mediators bradykinin (BK), adenosine triphosphate (ATP), norepinephrine (NE) and prostaglandin E2 (PGE2) strongly increased the population size of the small-diameter neurons displaying low-voltage activated (LVA, T-type) Ca(2+) currents while having no effect on the peak LVA current amplitude. When applied individually, BK and ATP also increased the population size of LVA-positive neurons while NE and PGE2 had no effect. The PLC inhibitor U-73122 and B2 receptor antagonist, Hoe-140, both abolished the increase of the population of LVA-positive DRG neurons. Inflammatory treatment did not affect CaV3.2 mRNA or protein levels in DRG cultures. Furthermore, an ubiquitination inhibitor, MG132, did not increase the population of LVA-positive neurons. Our data suggest that inflammatory mediators BK and ATP increase the abundance of LVA-positive DRG neurons in total neuronal population by stimulating the recruitment of a 'reserve pool' of CaV3.2 channels, particularly in neurons that do not display measurable LVA currents under control conditions.


Subject(s)
Bradykinin/immunology , Calcium Channels, T-Type/immunology , Ganglia, Spinal/cytology , Sensory Receptor Cells/immunology , Adenosine Triphosphate/immunology , Animals , Calcium Channels, T-Type/analysis , Cells, Cultured , Dinoprostone/immunology , Ganglia, Spinal/immunology , Inflammation/immunology , Norepinephrine/immunology , Rats, Sprague-Dawley , Sensory Receptor Cells/cytology
3.
Adv Exp Med Biol ; 860: 291-300, 2015.
Article in English | MEDLINE | ID: mdl-26303493

ABSTRACT

T-type Ca(2+) channels regulate proliferation in a number of tissue types, including vascular smooth muscle and various cancers. In such tissues, up-regulation of the inducible enzyme heme oxygenase-1 (HO-1) is often observed, and hypoxia is a key factor in its induction. HO-1 degrades heme to generate carbon monoxide (CO) along with Fe(2+) and biliverdin. Since CO is increasingly recognized as a regulator of ion channels (Peers et al. 2015), we have explored the possibility that it may regulate proliferation via modulation of T-type Ca(2+) channels.Whole-cell patch-clamp recordings revealed that CO (applied as the dissolved gas or via CORM donors) inhibited all 3 isoforms of T-type Ca(2+) channels (Cav3.1-3.3) when expressed in HEK293 cells with similar IC(50) values, and induction of HO-1 expression also suppressed T-type currents (Boycott et al. 2013). CO/HO-1 induction also suppressed the elevated basal [Ca(2+) ](i) in cells expressing these channels and reduced their proliferative rate to levels seen in non-transfected control cells (Duckles et al. 2015).Proliferation of vascular smooth muscle cells (both A7r5 and human saphenous vein cells) was also suppressed either by T-type Ca(2+) channel inhibitors (mibefradil and NNC 55-0396), HO-1 induction or application of CO. Effects of these blockers and CO were non additive. Although L-type Ca(2+) channels were also sensitive to CO (Scragg et al. 2008), they did not influence proliferation. Our data suggest that HO-1 acts to control proliferation via CO modulation of T-type Ca(2+) channels.


Subject(s)
Calcium Channels, T-Type/physiology , Carbon Monoxide/pharmacology , Calcium/metabolism , Calcium Channels, T-Type/analysis , Cell Proliferation , HEK293 Cells , Heme Oxygenase-1/physiology , Humans , Muscle, Smooth, Vascular/cytology , Myocytes, Smooth Muscle/physiology
4.
Channels (Austin) ; 2(6): 404-6, 2008.
Article in English | MEDLINE | ID: mdl-19098452

ABSTRACT

Cellular defects in ankyrin-based ion channels and transporter targeting pathways have previously been linked with abnormal vertebrate physiology and human disease. In a recent study, our group linked dysfunction in cardiac ankyrin-B function with human sinus node disease. Ankyrin-B deficient mice displayed bradycardia and heart rate variability similar to individuals harboring an ANK2 variant. Isolated sinoatrial node (SAN) cells from ankyrin-B-deficient animals displayed abnormal membrane expression of Na+/Ca2+ exchanger (NCX1), Na+/K+ ATPase (NKA), IP3 receptor (IP3R) and, surprisingly, Ca(V)1.3. Loss of ankyrin-B promoted slow and irregular Ca2+ release, as well as afterdepolarizations in isolated SAN cardiomyocytes. Our findings suggest that ankyrin-B serves as a critical focal point for channels and transporters important for sarcoplasmic reticulum (SR) calcium homeostasis as well as membrane depolarization in SAN cells. The severity and penetrance of human ANK2 sinus node dysfunction likely reflects the essential role of ankyrin-B for orchestrating membrane function of multiple SAN ion channel and transporters within a single functional pathway. Therefore, ankyrin-based pathways may serve as ideal therapeutic targets in SAN cardiomyocytes where a "multi-hit" approach is necessary to impact a complex process such as SAN cell automaticity. In summary, our new findings define a novel genetic basis for human SND and expand our understanding of the critical role that ankyrin-based targeting pathways play in excitable cell physiology.


Subject(s)
Ankyrins/physiology , Sinoatrial Node/physiology , Animals , Ankyrins/deficiency , Ankyrins/genetics , Calcium/metabolism , Calcium Channels, T-Type/analysis , Drug Delivery Systems , Humans , Inositol 1,4,5-Trisphosphate Receptors/analysis , Mice , Mice, Knockout , Myocytes, Cardiac , Sodium-Potassium-Exchanging ATPase/analysis
5.
J Mol Diagn ; 9(3): 305-14, 2007 Jul.
Article in English | MEDLINE | ID: mdl-17591929

ABSTRACT

The CpG island methylator phenotype (CIMP or CIMP-high) with extensive promoter methylation is a distinct phenotype in colorectal cancer. However, a choice of markers for CIMP has been controversial. A recent extensive investigation has selected five methylation markers (CACNA1G, IGF2, NEUROG1, RUNX3, and SOCS1) as surrogate markers for epigenomic aberrations in tumor. The use of these markers as a CIMP-specific panel needs to be validated by an independent, large dataset. Using MethyLight assays on 920 colorectal cancers from two large prospective cohort studies, we quantified DNA methylation in eight CIMP-specific markers [the above five plus CDKN2A (p16), CRABP1, and MLH1]. A CIMP-high cutoff was set at > or = 6/8 or > or = 5/8 methylated promoters, based on tumor distribution and BRAF/KRAS mutation frequencies. All but two very specific markers [MLH1 (98% specific) and SOCS1 (93% specific)] demonstrated > or = 85% sensitivity and > or = 80% specificity, indicating overall good concordance in methylation patterns and good performance of these markers. Based on sensitivity, specificity, and false positives and negatives, the eight markers were ranked in order as: RUNX3, CACNA1G, IGF2, MLH1, NEUROG1, CRABP1, SOCS1, and CDKN2A. In conclusion, a panel of markers including at least RUNX3, CACNA1G, IGF2, and MLH1 can serve as a sensitive and specific marker panel for CIMP-high.


Subject(s)
Biomarkers, Tumor/analysis , Carcinoma/diagnosis , Carcinoma/genetics , Colorectal Neoplasms/diagnosis , Colorectal Neoplasms/genetics , CpG Islands , DNA Methylation , Adaptor Proteins, Signal Transducing/analysis , Adaptor Proteins, Signal Transducing/genetics , Calcium Channels, T-Type/analysis , Calcium Channels, T-Type/genetics , Carcinoma/pathology , Cohort Studies , Colorectal Neoplasms/pathology , Core Binding Factor Alpha 3 Subunit/analysis , Core Binding Factor Alpha 3 Subunit/genetics , Female , Follow-Up Studies , Genetic Testing/methods , Genetics, Population , Humans , Insulin-Like Growth Factor II , Male , MutL Protein Homolog 1 , Nuclear Proteins/analysis , Nuclear Proteins/genetics , Phenotype , Promoter Regions, Genetic , Proteins/analysis , Proteins/genetics , Sensitivity and Specificity
6.
J Physiol ; 577(Pt 2): 513-23, 2006 Dec 01.
Article in English | MEDLINE | ID: mdl-17008378

ABSTRACT

T-type Ca2+ channels play essential roles in numerous cellular processes. Recently, we reported that phorbol-12-myristate-13-acetate (PMA) potently enhanced the current amplitude of Cav3.2 T-type channels reconstituted in Xenopus oocytes. Here, we have compared PMA modulation of the activities of Cav3.1, Cav3.2 and Cav3.3 channels, and have investigated the underlying mechanism. PMA augmented the current amplitudes of the three T-type channel isoforms, but the fold stimulations and time courses differed. The augmentation effects were not mimicked by 4alpha-PMA, an inactive stereoisomer of PMA, but were abolished by preincubation with protein kinase C (PKC) inhibitors, indicating that PMA augmented T-type channel currents via activation of oocyte PKC. The stimulation effect on Cav3.1 channel activity by PKC was mimicked by endothelin when endothelin receptor type A was coexpressed with Cav3.1 in the Xenopus oocyte system. Pharmacological studies combined with fluorescence imaging revealed that the surface density of Cav3.1 T-type channels was not significantly changed by activation of PKC. The PKC effect on Cav3.1 was localized to the cytoplasmic II-III loop using chimeric channels with individual cytoplasmic loops of Cav3.1 replaced by those of Cav2.1.


Subject(s)
Calcium Channels, T-Type/drug effects , Protein Kinase C/metabolism , Tetradecanoylphorbol Acetate/pharmacology , Animals , Calcium/metabolism , Calcium Channels, T-Type/analysis , Calcium Channels, T-Type/genetics , Calcium Channels, T-Type/metabolism , Dose-Response Relationship, Drug , Endothelin-1/pharmacology , Enzyme Activation/drug effects , Membrane Potentials/drug effects , Membrane Transport Proteins/metabolism , Microinjections , Mutation , Oocytes/chemistry , Oocytes/metabolism , Patch-Clamp Techniques , Protein Kinase C/antagonists & inhibitors , Protein Kinase Inhibitors/pharmacology , Receptor, Endothelin A/drug effects , Recombinant Fusion Proteins/drug effects , Recombinant Fusion Proteins/metabolism , Time Factors , Xenopus laevis
7.
J Pharmacol Sci ; 99(3): 205-10, 2005 Nov.
Article in English | MEDLINE | ID: mdl-16272790

ABSTRACT

Re-expression of fetal genes has been considered to underlie ionic remodeling in diseased heart. T-type Ca(2+) channels have been reported to be functionally expressed in embryonic hearts. In this review, we summarize developmental changes of T-type Ca(2+) channels in mouse ventricles from 9.5 days postcoitum (dpc) to adulthood, using patch clamp and quantitative PCR. In addition, we introduced T-type Ca(2+) channel expression in hypertrophied ventricles caused by myocardial infarction (MI) and aortic banding (AOB). Substantial T-type Ca(2+) channel current was recorded at both 9.5 and 18 dpc. The currents were inhibited by Ni(2+) at low concentrations. The current was not detectable in the adult stage. Ca(v)3.2 (alpha(1H)) mRNA is expressed dominantly at both 9.5 and 18 dpc. Ca(v)3.1 (alpha(1G)) increases from 9.5 to 18 dpc, but remains at low level compared with Ca(v)3.2. In contrast, Ca(v)3.1 is greater than Ca(v)3.2 at the adult stage. In MI, Ca(v)3.1 mRNA correlates negatively with brain natriuretic peptide (BNP) mRNA, whereas Ca(v)3.2 mRNA correlates positively with BNP mRNA. In AOB, these correlations are weak. We also analyzed the neuron-restrictive silencer factor (NRSF) in these hearts because it is the suppressor of transcription of the fetal cardiac gene program. The negative correlation between NRSF and BNP was stronger in MI than in AOB. Our findings show that Ca(v)3.2 underlies the functional T-type Ca(2+) channel in embryonic heart and suggest that NRSF may regulate Ca(v)3.2 expression in diseased hearts.


Subject(s)
Calcium Channels, T-Type/physiology , Fetal Heart/metabolism , Myocardial Infarction/metabolism , Myocardium/metabolism , Animals , Calcium Channels, T-Type/analysis , Humans
8.
Circulation ; 111(25): 3420-8, 2005 Jun 28.
Article in English | MEDLINE | ID: mdl-15967848

ABSTRACT

BACKGROUND: Cardiac responses to beta-adrenergic receptor stimulation are depressed with pressure overload-induced cardiac hypertrophy. We investigated whether exercise training could modify beta-adrenergic receptor responsiveness in a model of spontaneous hypertension by modifying the beta-adrenergic receptor desensitizing kinase GRK2 and the abundance and phosphorylation of some key Ca2+ cycling proteins. METHODS AND RESULTS: Female spontaneously hypertensive rats (SHR; age, 4 months) were placed into a treadmill running (SHR-TRD; 20 m/min, 1 h/d, 5 d/wk, 12 weeks) or sedentary group (SHR-SED). Age-matched Wistar Kyoto (WKY) rats were controls. Mean blood pressure was higher in SHR versus WKY (P<0.01) and unaltered with exercise. Left ventricular (LV) diastolic anterior and posterior wall thicknesses were greater in SHR than WKY (P<0.001) and augmented with training (P<0.01). Langendorff LV performance was examined during isoproterenol (ISO) infusions (1x10(-10) to 1x10(-7) mol/L) and pacing stress (8.5 Hz). The peak LV developed pressure/ISO dose response was shifted rightward 100-fold in SHR relative to WKY. The peak ISO LV developed pressure response was similar between WKY and SHR-SED and increased in SHR-TRD (P<0.05). SHR-TRD showed the greatest lusitropic response to ISO (P<0.05) and offset the pacing-induced increase in LV end-diastolic pressure and the time constant of isovolumic relaxation (tau) observed in WKY and SHR-SED. Improved cardiac responses to ISO in SHR-TRD were associated with normalized myocardial levels of GRK2 (P<0.05). SHR displayed increased L-type Ca2+ channel and sodium calcium exchanger abundance compared with WKY (P<0.001). Training increased ryanodine receptor phosphorylation and phospholamban phosphorylation at both the Ser16 and Thr17 residues (P<0.05). CONCLUSIONS: Exercise training in hypertension improves the inotropic and lusitropic responsiveness to beta-adrenergic receptor stimulation despite augmenting LV wall thickness. A lower GRK2 abundance and an increased phosphorylation of key Ca2+ cycling proteins may be responsible for the above putative effects.


Subject(s)
Adrenergic beta-Agonists/pharmacology , Exercise Therapy/methods , Hypertension/therapy , Animals , Blood Pressure , Calcium Channels, T-Type/analysis , Female , G-Protein-Coupled Receptor Kinase 2 , Heart Ventricles/chemistry , Hypertrophy, Left Ventricular , In Vitro Techniques , Isoproterenol/pharmacology , Myocardial Contraction , Phosphorylation , Rats , Rats, Inbred SHR , Rats, Wistar , Sodium-Calcium Exchanger/analysis , beta-Adrenergic Receptor Kinases/analysis
9.
Circ Res ; 96(8): 864-72, 2005 Apr 29.
Article in English | MEDLINE | ID: mdl-15774856

ABSTRACT

While Ca2+ influx is essential for activation of the cell cycle machinery, the processes that regulate Ca2+ influx in this context have not been fully elucidated. Electrophysiological and molecular studies have identified multiple Ca2+ channel genes expressed in mammalian cells. Ca(v)3.x gene family members, encoding low voltage-activated (LVA) or T-type channels, were first identified in the central nervous system and subsequently in non-neuronal tissue. Reports of a potential role for T-type Ca2+ channels in controlling cell proliferation conflict. The present study tested the hypothesis that T-type Ca2+ channels, encoded by Ca(v)3.x genes, control pulmonary artery smooth muscle cell proliferation and cell cycle progression. Using quantitative RT/PCR, immunocytochemistry, and immunohistochemistry we found that Ca(v)3.1 was the predominant Ca(v)3.x channel expressed in early passage human pulmonary artery smooth muscle cells in vitro and in the media of human pulmonary arteries, in vivo. Selective blockade of Ca(v)3.1 expression with small interfering RNA (siRNA) and pharmacological blockade of T-type channels completely inhibited proliferation in response to 5% serum and prevented cell cycle entry. These studies establish that T-type voltage-operated Ca2+ channels are required for cell cycle progression and proliferation of human PA SMC.


Subject(s)
Calcium Channels, T-Type/physiology , Muscle, Smooth, Vascular/cytology , Myocytes, Smooth Muscle/cytology , Pulmonary Artery/cytology , Calcium Channels, T-Type/analysis , Calcium Channels, T-Type/genetics , Cell Proliferation , Cells, Cultured , Diltiazem/pharmacology , Humans , Lung/metabolism , Mibefradil/pharmacology , Muscle, Smooth, Vascular/metabolism , Myocytes, Smooth Muscle/metabolism , RNA, Small Interfering/pharmacology , Reverse Transcriptase Polymerase Chain Reaction
10.
J Histochem Cytochem ; 48(6): 807-19, 2000 Jun.
Article in English | MEDLINE | ID: mdl-10820154

ABSTRACT

The calcium channel alpha1E subunit was originally cloned from mammalian brain. A new splice variant was recently identified in rat islets of Langerhans and in human kidney by the polymerase chain reaction. The same isoform of alpha1E was detected in rat and guinea pig heart by amplifying indicative cDNA fragments and by immunostaining using peptide-specific antibodies. The apparent molecular size of cardiac alpha1E was determined by SDS-PAGE and immunoblotting (218 +/- 6 kD; n = 3). Compared to alpha1E from stably transfected HEK-293 cells, this is smaller by 28 kD. The distribution of alpha1E in cardiac muscle cells of the conducting system and in the cardiomyoblast cell line H9c2 was compared to the distribution of chromogranin, a marker of neuroendocrine cells, and to the distribution of atrial natriuretic peptide (ANP). In serial sections from atrial and ventricular regions of rat heart, co-localization of alpha1E with ANP was detected in atrium and with chromogranin A/B in Purkinje fibers of the conducting system in both rat atrium and ventricle. The kidney is another organ in which natriuretic peptide hormones are secreted. The detection of alpha1E in the distal tubules of human kidney, where urodilatin is stored and secreted, led to the conclusion that the expression of alpha1E in rat heart and human kidney is linked to regions with endocrine functions and therefore is involved in the Ca(2+)-dependent secretion of peptide hormones such as ANP and urodilatin.


Subject(s)
Calcium Channels, T-Type/analysis , Chromogranins/metabolism , Ion Channel Gating , Kidney Tubules, Distal/chemistry , Myocardium/chemistry , Alternative Splicing , Amino Acid Sequence , Animals , Antibody Specificity , Brain/metabolism , Brain/pathology , Calcium Channels, T-Type/genetics , Calcium Channels, T-Type/immunology , Cell Line, Transformed , Female , Guinea Pigs , Humans , Immunoblotting/methods , Immunohistochemistry/methods , Intracellular Membranes/chemistry , Kidney Tubules, Distal/cytology , Membrane Proteins/analysis , Microsomes/chemistry , Molecular Sequence Data , Myocardium/cytology , Rats , Rats, Wistar , Sequence Homology, Amino Acid , Tumor Cells, Cultured
11.
Eur J Neurosci ; 12(4): 1217-26, 2000 Apr.
Article in English | MEDLINE | ID: mdl-10762351

ABSTRACT

The auxiliary calcium channel alpha2delta subunit comprises a family of three genes, alpha2delta-1 to 3, which are expressed in a tissue-specific manner. alpha2delta-2 mRNA is found in the heart, skeletal muscle, brain, kidney, liver and pancreas. We report here for the first time the identification and functional characterization of alpha2delta-2 splice variants and their mRNA distribution in the mouse brain. The splice variants differ in the alpha2 and delta protein by eight and three amino acid residues, respectively, and are differentially expressed in cardiac tissue and human medullary thyroid carcinoma (hMTC) cells. In situ hybridization of mouse brain sections revealed the highest expression of alpha2delta-2 mRNA in the Purkinje cell layer of the cerebellum, habenulae and septal nuclei, and a lower expression in the cerebral cortex, olfactory bulb, thalamic and hypothalamic nuclei, as well as the inferior and superior colliculus. As the in situ data did not suggest a specific colocalization with any alpha1 subunit, coexpression studies of alpha2delta-2 were carried out either with the high-voltage-gated calcium channels, alpha1C, alpha1E or alpha1A, or with the low-voltage-gated calcium channel, alpha1G. Coexpression of alpha2delta-2 increased the current density, shifted the voltage dependence of channel activation and inactivation of alpha1C, alpha1E and alpha1A subunits in a hyperpolarizing direction, and accelerated the decay and shifted the steady-state inactivation of the alpha1G current.


Subject(s)
Calcium Channels, L-Type/analysis , Calcium Channels, L-Type/genetics , Calcium Channels, T-Type/analysis , Calcium Channels, T-Type/genetics , Neurons/chemistry , Alternative Splicing/physiology , Animals , Blotting, Northern , Calcium Channels, L-Type/metabolism , Calcium Channels, T-Type/metabolism , Cell Line , Cloning, Molecular , DNA, Complementary , Electrophysiology , Gene Expression/physiology , Humans , In Situ Hybridization , Ion Channel Gating/physiology , Kidney/cytology , Membrane Potentials/physiology , Mice , Neurons/physiology , RNA, Messenger/analysis , Transfection
12.
Endocrine ; 13(3): 251-62, 2000 Dec.
Article in English | MEDLINE | ID: mdl-11216635

ABSTRACT

Ca2+ influx through voltage-dependent Ca2+ channels plays a crucial role in stimulus-secretion coupling in pancreatic islet beta-cells. Molecular and physiologic studies have identified multiple Ca2+ channel subtypes in rodent islets and insulin-secreting cell lines. The differential targeting of Ca2+ channel subtypes to the vicinity of the insulin secretory apparatus is likely to account for their selective coupling to glucose-dependent insulin secretion. In this article, I review these studies. In addition, I discuss temporal and spatial aspects of Ca2+ signaling in beta-cells, the former involving the oscillatory activation of Ca2+ channels during glucose-induced electrical bursting, and the latter involving [Ca2+]i elevation in restricted microscopic "domains," as well as direct interactions between Ca2+ channels and secretory SNARE proteins. Finally, I review the evidence supporting a possible role for Ca2+ release from the endoplasmic reticulum in glucose-dependent insulin secretion, and evidence to support the existence of novel Ca2+ entry pathways. I also show that the beta-cell has an elaborate and complex set of [Ca2+]i signaling mechanisms that are capable of generating diverse and extremely precise [Ca2+]i patterns. These signals, in turn, are exquisitely coupled in space and time to the beta-cell secretory machinery to produce the precise minute-to-minute control of insulin secretion necessary for body energy homeostasis.


Subject(s)
Calcium/metabolism , Islets of Langerhans/metabolism , Animals , Calcium/pharmacology , Calcium Channels, L-Type/analysis , Calcium Channels, L-Type/physiology , Calcium Channels, T-Type/analysis , Calcium Channels, T-Type/physiology , Electric Conductivity , Electrophysiology , Exocytosis , Glucose/pharmacology , Humans , Insulin/metabolism , Insulin Secretion , Ion Channel Gating , Islets of Langerhans/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...