Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
Add more filters










Publication year range
1.
Front Physiol ; 14: 1155976, 2023.
Article in English | MEDLINE | ID: mdl-37654674

ABSTRACT

Voltage-gated Ca2+ channels (VGCC) directly control muscle contraction and neurotransmitter release, and slower processes such as cell differentiation, migration, and death. They are potently inhibited by RGK GTP-ases (Rem, Rem2, Rad, and Gem/Kir), which decrease Ca2+ channel membrane expression, as well as directly inhibit membrane-resident channels. The mechanisms of membrane-resident channel inhibition are difficult to study because RGK-overexpression causes complete or near complete channel inhibition. Using titrated levels of Gem expression in Xenopus oocytes to inhibit WT P/Q-type calcium channels by ∼50%, we show that inhibition is dependent on channel inactivation. Interestingly, fast-inactivating channels, including Familial Hemiplegic Migraine mutants, are more potently inhibited than WT channels, while slow-inactivating channels, such as those expressed with the Cavß2a auxiliary subunit, are spared. We found similar results in L-type channels, and, remarkably, Timothy Syndrome mutant channels were insensitive to Gem inhibition. Further results suggest that RGKs slow channel recovery from inactivation and further implicate RGKs as likely modulating factors in channelopathies.

2.
PLoS One ; 14(9): e0217733, 2019.
Article in English | MEDLINE | ID: mdl-31479461

ABSTRACT

Human ether-à-go-go-related gene (Kv11.1, or hERG) is a potassium channel that conducts the delayed rectifier potassium current (IKr) during the repolarization phase of cardiac action potentials. hERG channels have a larger pore than other K+channels and can trap many unintended drugs, often resulting in acquired LQTS (aLQTS). R-roscovitine is a cyclin-dependent kinase (CDK) inhibitor that induces apoptosis in colorectal, breast, prostate, multiple myeloma, other cancer cell lines, and tumor xenografts, in micromolar concentrations. It is well tolerated in phase II clinical trials. R-roscovitine inhibits open hERG channels but does not become trapped in the pore. Two-electrode voltage clamp recordings from Xenopus oocytes expressing wild-type (WT) or hERG pore mutant channels (T623A, S624A, Y652A, F656A) demonstrated that compared to WT hERG, T623A, Y652A, and F656A inhibition by 200 µM R-roscovitine was ~ 48%, 29%, and 73% weaker, respectively. In contrast, S624A hERG was inhibited more potently than WT hERG, with a ~ 34% stronger inhibition. These findings were further supported by the IC50 values, which were increased for T623A, Y652A and F656A (by ~5.5, 2.75, and 42 fold respectively) and reduced 1.3 fold for the S624A mutant. Our data suggest that while T623, Y652, and F656 are critical for R-roscovitine-mediated inhibition, S624 may not be. Docking studies further support our findings. Thus, R-roscovitine's relatively unique features, coupled with its tolerance in clinical trials, could guide future drug screens.


Subject(s)
Ether-A-Go-Go Potassium Channels/antagonists & inhibitors , Ether-A-Go-Go Potassium Channels/chemistry , Potassium Channel Blockers/chemistry , Potassium Channel Blockers/pharmacology , Animals , Dose-Response Relationship, Drug , Drug Discovery , Ether-A-Go-Go Potassium Channels/genetics , Humans , Molecular Docking Simulation , Molecular Dynamics Simulation , Molecular Structure , Mutation , Oocytes/drug effects , Oocytes/metabolism , Protein Conformation , Structure-Activity Relationship
3.
Physiol Rep ; 7(8): e14047, 2019 04.
Article in English | MEDLINE | ID: mdl-31008554

ABSTRACT

Hypothalamic magnocellular neurosecretory cells (MNCs) undergo dramatic structural reorganization during lactation in female rats that is thought to contribute to the pulsatile secretion of oxytocin critical for milk ejection. MNCs from male rats generate robust bursts of GABAergic synaptic currents, a subset of which are onset-synchronized between MNC pairs, but the functional role of the IPSC bursts is not known. To determine the physiological relevance of IPSC bursts, we compared MNCs from lactating and non-lactating female rats using whole-cell recordings in brain slices. We recorded a sixfold increase in the incidence of IPSC bursts in oxytocin (OT)-MNCs from lactating rats compared to non-lactating rats, whereas there was no change in IPSC bursts in vasopressin (VP)-MNCs. Synchronized bursts of IPSCs were observed in pairs of MNCs in slices from lactating rats. Our data indicate, therefore, that IPSC bursts are upregulated specifically in OT-MNCs during lactation, and may, therefore, contribute via rebound depolarization to the spike trains in OT neurons that lead to reflex milk ejection.


Subject(s)
Inhibitory Postsynaptic Potentials , Lactation/physiology , Neuroendocrine Cells/physiology , Oxytocin/metabolism , Animals , Female , Hypothalamus/cytology , Hypothalamus/metabolism , Hypothalamus/physiology , Lactation/metabolism , Neuroendocrine Cells/metabolism , Rats , Rats, Wistar , Vasopressins/metabolism
4.
Article in English | MEDLINE | ID: mdl-28904646

ABSTRACT

This two-year study describes the assessment of student learning gains arising from participation in a year-long curriculum consisting of a classroom undergraduate research experience (CURE) embedded into second-year, major core Genetics and Cellular and Molecular Biology (CMB) laboratory courses. For the first course in our CURE, students used micro-array or RNAseq analyses to identify genes important for environmental stress responses by Saccharomyces cerevisiae. The students were tasked with creating overexpressing mutants of their genes and designing their own original experiments to investigate the functions of those genes using the overexpression and null mutants in the second CURE course. In order to evaluate student learning gains, we employed three validated concept inventories in a pretest/posttest format and compared gains on the posttest versus the pretest with student laboratory final grades. Our results demonstrated that there was a significant correlation between students earning lower grades in the Genetics laboratory for both years of this study and gains on the Genetics Concept Assessment (GCA). We also demonstrated a correlation between students earning lower grades in the Genetics laboratory and gains on the Introductory Molecular and Cell Biology Assessment (IMCA) for year 1 of the study. Students furthermore demonstrated significant gains in identifying the variable properties of experimental subjects when assessed using the Rubric for Experimental (RED) design tool. Results from the administration of the CURE survey support these findings. Our results suggest that a year-long CURE enables lower performing students to experience greater gains in their foundational skills for success in the STEM disciplines.

5.
Curr Mol Pharmacol ; 8(2): 180-7, 2015.
Article in English | MEDLINE | ID: mdl-25966691

ABSTRACT

Due to their essential biological roles, voltage-gated calcium channels (VGCCs) are regulated by a myriad of molecules and mechanisms. Fifteen years ago, RGK proteins were discovered to bind the VGCC ß subunit (Cavß) and potently inhibit high-voltage activated Ca(2+) channels. RGKs (Rad, Rem, Rem2 and Gem/Kir) are a family of monomeric small GTPases belonging to the superfamily of Ras GTPases. They exert dual inhibitory effects on VGCCs, decreasing surface expression and suppressing surface channels through immobilization of the voltage sensor or reduction of channel open probability. While Cavß is required for all forms of RGK inhibition, not all inhibition is mediated by the RGK-Cavß interaction. Some RGK proteins also interact directly with the pore-forming α1 subunit of some types of VGCCs (Cavα1). Importantly, RGK proteins tonically inhibit VGCCs in native cells, regulating cardiac and neural functions. This minireview summarizes the mechanisms, molecular determinants, and physiological impact of RGK inhibition of VGCCs.


Subject(s)
Calcium Channels/metabolism , Ion Channel Gating/physiology , Monomeric GTP-Binding Proteins/metabolism , Animals , Calcium Channels/chemistry , Humans , Models, Biological , Models, Molecular , Monomeric GTP-Binding Proteins/chemistry , Protein Binding , Protein Structure, Tertiary , Protein Subunits/chemistry , Protein Subunits/metabolism
6.
Channels (Austin) ; 9(1): 50-5, 2015.
Article in English | MEDLINE | ID: mdl-25664681

ABSTRACT

The delivery of Ca2+ into cells by CaV channels provides the trigger for many cellular actions, such as cardiac muscle contraction and neurotransmitter release. Thus, a full understanding of Ca2+ permeation through these channels is critical. Using whole-cell voltage-clamp recordings, we recently demonstrated that voltage modulates the apparent affinity of N-type (CaV2.2) channels for permeating Ca2+ and Ba2+ ions. While we took many steps to ensure the high fidelity of our recordings, problems can occur when CaV currents become large and fast, or when currents run down. Thus, we use here single channel recordings to further test the hypothesis that permeating ions interact with N-type channels in a voltage-dependent manner. We also examined L-type (CaV1.2) channels to determine if these channels also exhibit voltage-dependent permeation. Like our whole-cell data, we find that voltage modulates N-channel affinity for Ba2+ at voltages>0 mV, but has little or no effect at voltages<0 mV. Furthermore, we demonstrate that permeation through L-channel is also modulated by voltage. Thus, voltage-dependence may be a common feature of divalent cation permeation through CaV1 and CaV2 channels (i.e. high-voltage activated CaV channels). The voltage dependence of CaV1 channel permeation is likely a mechanism mediating sustained Ca2+ influx during the plateau phase of the cardiac action potential.


Subject(s)
Calcium Channels, L-Type/metabolism , Calcium Channels, N-Type/metabolism , Cell Membrane Permeability , Electrophysiology
7.
Sci China Life Sci ; 58(1): 28-38, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25576452

ABSTRACT

Voltage-gated calcium channels (VGCCs) play critical roles in cardiac and skeletal muscle contractions, hormone and neurotransmitter release, as well as slower processes such as cell proliferation, differentiation, migration and death. Mutations in VGCCs lead to numerous cardiac, muscle and neurological disease, and their physiological function is tightly regulated by kinases, phosphatases, G-proteins, calmodulin and many other proteins. Fifteen years ago, RGK proteins were discovered as the most potent endogenous regulators of VGCCs. They are a family of monomeric GTPases (Rad, Rem, Rem2, and Gem/Kir), in the superfamily of Ras GTPases, and they have two known functions: regulation of cytoskeletal dynamics including dendritic arborization and inhibition of VGCCs. Here we review the mechanisms and molecular determinants of RGK-mediated VGCC inhibition, the physiological impact of this inhibition, and recent evidence linking the two known RGK functions.


Subject(s)
Calcium Channels/physiology , GTP Phosphohydrolases/physiology , Ion Channel Gating/physiology , Humans
8.
J Gen Physiol ; 144(3): 207-20, 2014 Sep.
Article in English | MEDLINE | ID: mdl-25114024

ABSTRACT

Voltage-gated calcium (Ca(V)) channels deliver Ca(2+) to trigger cellular functions ranging from cardiac muscle contraction to neurotransmitter release. The mechanism by which these channels select for Ca(2+) over other cations is thought to involve multiple Ca(2+)-binding sites within the pore. Although the Ca(2+) affinity and cation preference of these sites have been extensively investigated, the effect of voltage on these sites has not received the same attention. We used a neuronal preparation enriched for N-type calcium (Ca(V)2.2) channels to investigate the effect of voltage on Ca(2+) flux. We found that the EC50 for Ca(2+) permeation increases from 13 mM at 0 mV to 240 mM at 60 mV, indicating that, during permeation, Ca(2+) ions sense the electric field. These data were nicely reproduced using a three-binding-site step model. Using roscovitine to slow Ca(V)2.2 channel deactivation, we extended these measurements to voltages <0 mV. Permeation was minimally affected at these hyperpolarized voltages, as was predicted by the model. As an independent test of voltage effects on permeation, we examined the Ca(2+)-Ba(2+) anomalous mole fraction (MF) effect, which was both concentration and voltage dependent. However, the Ca(2+)-Ba(2+) anomalous MF data could not be reproduced unless we added a fourth site to our model. Thus, Ca(2+) permeation through Ca(V)2.2 channels may require at least four Ca(2+)-binding sites. Finally, our results suggest that the high affinity of Ca(2+) for the channel helps to enhance Ca(2+) influx at depolarized voltages relative to other ions (e.g., Ba(2+) or Na(+)), whereas the absence of voltage effects at negative potentials prevents Ca(2+) from becoming a channel blocker. Both effects are needed to maximize Ca(2+) influx over the voltages spanned by action potentials.


Subject(s)
Calcium Channels, N-Type/metabolism , Calcium/metabolism , Membrane Potentials , Animals , Barium/pharmacology , Binding Sites , Calcium Channel Blockers/pharmacology , Calcium Channels, N-Type/chemistry , Cells, Cultured , Ion Transport , Neurons/drug effects , Neurons/metabolism , Neurons/physiology , Purines/pharmacology , Rana catesbeiana , Roscovitine
9.
J Neurosci ; 34(17): 5738-46, 2014 Apr 23.
Article in English | MEDLINE | ID: mdl-24760834

ABSTRACT

Progressive neurodegenerative diseases are among the most frequently occurring aging-associated human pathologies. In a screen for Caenorhabditis elegans mutant animals that lack their normal complement of dopaminergic neurons, we identified two strains with progressive loss of dopaminergic neurons during postembryonic life. Through whole-genome sequencing we show that both strains harbor dominant (d), gain-of-function mutations in the Transient Receptor Potential (TRP) mechanosensory channel trp-4, a member of the invertebrate and vertebrate TRPN-type of the TRP family channels. Gain-of-function mutations in TRP channels have not been previously implicated in dopaminergic neuronal degeneration. We show that trp-4(d) induces cell death in dopamine neurons through a defined, calcium-related downstream pathway.


Subject(s)
Caenorhabditis elegans Proteins/metabolism , Cell Death/physiology , Dopaminergic Neurons/pathology , Nerve Degeneration/pathology , TRPC Cation Channels/metabolism , Animals , Caenorhabditis elegans , Caenorhabditis elegans Proteins/genetics , Dopaminergic Neurons/metabolism , Movement/physiology , Mutation , Nerve Degeneration/genetics , Nerve Degeneration/metabolism , TRPC Cation Channels/genetics
10.
Biochim Biophys Acta ; 1828(7): 1530-40, 2013 Jul.
Article in English | MEDLINE | ID: mdl-22981275

ABSTRACT

The voltage-gated Ca²âº channel ß subunit (Ca(v)ß) is a cytosolic auxiliary subunit that plays an essential role in regulating the surface expression and gating properties of high-voltage activated (HVA) Ca²âº channels. It is also crucial for the modulation of HVA Ca²âº channels by G proteins, kinases, Ras-related RGK GTPases, and other proteins. There are indications that Ca(v)ß may carry out Ca²âº channel-independent functions. Ca(v)ß knockouts are either non-viable or result in a severe pathophysiology, and mutations in Ca(v)ß have been implicated in disease. In this article, we review the structure and various biological functions of Ca(v)ß, as well as recent advances. This article is part of a Special Issue entitled: Calcium channels.


Subject(s)
Calcium Channels, L-Type/chemistry , Calcium Channels, L-Type/physiology , Calcium/metabolism , Ion Channel Gating/physiology , Animals , Humans , Protein Subunits
11.
Structure ; 20(12): 1991-2, 2012 Dec 05.
Article in English | MEDLINE | ID: mdl-23217679

ABSTRACT

HCN channels and their modulation by cAMP play a key role in cardiac pacemaking. In this issue of Structure, Xu and colleagues reveal that an arrhythmia-causing mutation of an HCN channel weakens cAMP binding to the channel by altering the local structure of its entry-exit pathway.

12.
J Biol Chem ; 287(27): 22749-58, 2012 Jun 29.
Article in English | MEDLINE | ID: mdl-22589533

ABSTRACT

The RGK family of monomeric GTP-binding proteins potently inhibits high voltage-activated Ca(2+) channels. The molecular mechanisms of this inhibition are largely unclear. In Xenopus oocytes, Gem suppresses the activity of P/Q-type Ca(2+) channels on the plasma membrane. This is presumed to occur through direct interactions of one or more Gem inhibitory sites and the pore-forming Ca(v)2.1 subunit in a manner dependent on the Ca(2+) channel subunit ß (Ca(v)ß). In this study we investigated the molecular determinants in Gem that are critical for this inhibition. Like other RGK proteins, Gem contains a conserved Ras-like core and extended N and C termini. A 12-amino acid fragment in the C terminus was found to be crucial for and sufficient to produce Ca(v)ß-dependent inhibition, suggesting that this region forms an inhibitory site. A three-amino acid motif in the core was also found to be critical, possibly forming another inhibitory site. Mutating either site individually did not hamper Gem inhibition, but mutating both sites together completely abolished Gem inhibition without affecting Gem protein expression level or disrupting Gem interaction with Ca(v)2.1 or Ca(v)ß. Mutating Gem residues that are crucial for interactions with previously demonstrated RGK modulators such as calmodulin, 14-3-3, and phosphatidylinositol lipids did not significantly affect Gem inhibition. These results suggest that Gem contains two candidate inhibitory sites, each capable of producing full inhibition of P/Q-type Ca(2+) channels.


Subject(s)
Calcium Channels, N-Type/genetics , Calcium Channels, N-Type/physiology , Monomeric GTP-Binding Proteins/genetics , Monomeric GTP-Binding Proteins/physiology , Amino Acid Motifs/physiology , Amino Acid Sequence , Animals , Binding Sites/physiology , Calcium Channels, N-Type/chemistry , Calmodulin/metabolism , Cell Membrane/physiology , Escherichia coli/genetics , HEK293 Cells , Humans , Molecular Sequence Data , Monomeric GTP-Binding Proteins/chemistry , Oocytes/physiology , Patch-Clamp Techniques , Peptide Mapping , Protein Structure, Tertiary/physiology , Rabbits , Rats , Signal Transduction/physiology , Xenopus laevis
13.
Physiol Rev ; 90(4): 1461-506, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20959621

ABSTRACT

Calcium regulates a wide spectrum of physiological processes such as heartbeat, muscle contraction, neuronal communication, hormone release, cell division, and gene transcription. Major entryways for Ca(2+) in excitable cells are high-voltage activated (HVA) Ca(2+) channels. These are plasma membrane proteins composed of several subunits, including α(1), α(2)δ, ß, and γ. Although the principal α(1) subunit (Ca(v)α(1)) contains the channel pore, gating machinery and most drug binding sites, the cytosolic auxiliary ß subunit (Ca(v)ß) plays an essential role in regulating the surface expression and gating properties of HVA Ca(2+) channels. Ca(v)ß is also crucial for the modulation of HVA Ca(2+) channels by G proteins, kinases, and the Ras-related RGK GTPases. New proteins have emerged in recent years that modulate HVA Ca(2+) channels by binding to Ca(v)ß. There are also indications that Ca(v)ß may carry out Ca(2+) channel-independent functions, including directly regulating gene transcription. All four subtypes of Ca(v)ß, encoded by different genes, have a modular organization, consisting of three variable regions, a conserved guanylate kinase (GK) domain, and a conserved Src-homology 3 (SH3) domain, placing them into the membrane-associated guanylate kinase (MAGUK) protein family. Crystal structures of Ca(v)ßs reveal how they interact with Ca(v)α(1), open new research avenues, and prompt new inquiries. In this article, we review the structure and various biological functions of Ca(v)ß, with both a historical perspective as well as an emphasis on recent advances.


Subject(s)
Calcium Channels/metabolism , Ion Channel Gating/physiology , Amino Acid Sequence , Animals , Calcium Channels/chemistry , Calcium Channels/genetics , Humans , Protein Conformation , Protein Subunits
14.
Proc Natl Acad Sci U S A ; 107(33): 14887-92, 2010 Aug 17.
Article in English | MEDLINE | ID: mdl-20679232

ABSTRACT

The Rem, Rem2, Rad, and Gem/Kir (RGK) family of small GTP-binding proteins potently inhibits high voltage-activated (HVA) Ca(2+) channels, providing a powerful means of modulating neural, endocrine, and muscle functions. The molecular mechanisms of this inhibition are controversial and remain largely unclear. RGK proteins associate directly with Ca(2+) channel beta subunits (Ca(v)beta), and this interaction is widely thought to be essential for their inhibitory action. In this study, we investigate the molecular underpinnings of Gem inhibition of P/Q-type Ca(2+) channels. We find that a purified Gem protein markedly and acutely suppresses P/Q channel activity in inside-out membrane patches, that this action requires Ca(v)beta but not the Gem/Ca(v)beta interaction, and that Gem coimmunoprecipitates with the P/Q channel alpha(1) subunit (Ca(v)alpha(1)) in a Ca(v)beta-independent manner. By constructing chimeras between P/Q channels and Gem-insensitive low voltage-activated T-type channels, we identify a region encompassing transmembrane segments S1, S2, and S3 in the second homologous repeat of Ca(v)alpha(1) critical for Gem inhibition. Exchanging this region between P/Q and T channel Ca(v)alpha(1) abolishes Gem inhibition of P/Q channels and confers Ca(v)beta-dependent Gem inhibition to a chimeric T channel that also carries the P/Q I-II loop (a cytoplasmic region of Ca(v)alpha(1) that binds Ca(v)beta). Our results challenge the prevailing view regarding the role of Ca(v)beta in RGK inhibition of high voltage-activated Ca(2+) channels and prompt a paradigm in which Gem directly binds and inhibits Ca(v)beta-primed Ca(v)alpha(1) on the plasma membrane.


Subject(s)
Calcium Channels, P-Type/physiology , Calcium Channels, Q-Type/physiology , Cell Membrane/drug effects , Monomeric GTP-Binding Proteins/pharmacology , Amino Acid Sequence , Animals , Blotting, Western , Calcium Channels, P-Type/genetics , Calcium Channels, P-Type/metabolism , Calcium Channels, Q-Type/genetics , Calcium Channels, Q-Type/metabolism , Cell Line , Cell Membrane/physiology , Female , Humans , Immunoprecipitation , Kinetics , Membrane Potentials/drug effects , Models, Biological , Molecular Sequence Data , Monomeric GTP-Binding Proteins/genetics , Monomeric GTP-Binding Proteins/metabolism , Oocytes/drug effects , Oocytes/metabolism , Oocytes/physiology , Protein Binding , Protein Subunits/genetics , Protein Subunits/metabolism , Protein Subunits/physiology , Sequence Homology, Amino Acid , Xenopus laevis
15.
Proc Natl Acad Sci U S A ; 106(28): 11558-63, 2009 Jul 14.
Article in English | MEDLINE | ID: mdl-19556541

ABSTRACT

Mutations in PKD1 and TRPP2 account for nearly all cases of autosomal dominant polycystic kidney disease (ADPKD). These 2 proteins form a receptor/ion channel complex on the cell surface. Using a combination of biochemistry, crystallography, and a single-molecule method to determine the subunit composition of proteins in the plasma membrane of live cells, we find that this complex contains 3 TRPP2 and 1 PKD1. A newly identified coiled-coil domain in the C terminus of TRPP2 is critical for the formation of this complex. This coiled-coil domain forms a homotrimer, in both solution and crystal structure, and binds to a single coiled-coil domain in the C terminus of PKD1. Mutations that disrupt the TRPP2 coiled-coil domain trimer abolish the assembly of both the full-length TRPP2 trimer and the TRPP2/PKD1 complex and diminish the surface expression of both proteins. These results have significant implications for the assembly, regulation, and function of the TRPP2/PKD1 complex and the pathogenic mechanism of some ADPKD-producing mutations.


Subject(s)
Models, Molecular , Multiprotein Complexes/genetics , TRPP Cation Channels/genetics , Amino Acid Sequence , Animals , Base Sequence , Crystallization , Humans , Molecular Sequence Data , Multiprotein Complexes/metabolism , Protein Binding , Protein Structure, Tertiary , Sequence Analysis, DNA , TRPP Cation Channels/metabolism , Xenopus
16.
J Neurochem ; 105(4): 1450-61, 2008 May.
Article in English | MEDLINE | ID: mdl-18221369

ABSTRACT

Dihydropyridines can affect L-type calcium channels (CaV1) as either agonists or antagonists. Seliciclib or R-roscovitine, a 2,6,9-trisubstituted purine, is a potent cyclin-dependent kinase inhibitor that induces both agonist and antagonist effects on CaV2 channels (N-, P/Q- and R-type). We studied the effects induced by various trisubstituted purines on CaV2.2 (N-type) channels to learn about chemical structure-function relationships. We found that S-roscovitine and R-roscovitine showed similar potency to inhibit, but agonist activity of S-roscovitine required at least a 20-fold higher concentration, suggesting stereospecificity of the agonist-binding site. The testing of other trisubstituted purines showed a correlation between CaV2.2 inhibition and cyclin-dependent kinase affinity that broke down after determining that a chemically unrelated inhibitor, kenpaullone, was a poor CaV2.2 inhibitor, and a kinase inactive analog (dimethylamino-olomoucine; DMAO) was a strong inhibitor, which together support a kinase independent effect. In fact, like dihydropyridine-induced L-channel inhibition, R-roscovitine left-shifted the closed-state inactivation versus voltage relationship, which suggests that inhibition results from CaV2 channels moving into the inactivated state. Trisubstituted purine antagonists could become clinically important drugs to treat diseases, such as heart failure and neuropathic pain that result from elevated CaV2 channel activity.


Subject(s)
Calcium Channel Blockers/chemistry , Calcium Channel Blockers/pharmacology , Purines/chemistry , Purines/pharmacology , Xenopus Proteins/agonists , Xenopus Proteins/antagonists & inhibitors , Animals , Calcium Channel Blockers/metabolism , Calcium Channels, N-Type/metabolism , Purines/metabolism , Rana catesbeiana , Roscovitine , Structure-Activity Relationship , Xenopus Proteins/metabolism
17.
Neuropharmacology ; 52(3): 883-94, 2007 Mar.
Article in English | MEDLINE | ID: mdl-17125805

ABSTRACT

Roscovitine potently inhibits cyclin-dependent kinases (CDK) and can independently slow the closing of neuronal (CaV2.2) calcium channels. We were interested if this drug could affect other ion channels similarly. Using whole cell recordings, we found that roscovitine specifically slows deactivation of all CaV2 channels (N, P/Q and R) by binding to the open state. This effect had a rapid onset and EC(50)=54, 120 and 54microM for N-, P/Q-, and R-type channels, respectively. Deactivation of other channel types was not slowed, including L-type calcium channels (CaV1.2, CaV1.3), potassium channels (native, Kv4.2, Kv2.1 and Kv1.3), and native sodium channels. However, most of the channels tested were inhibited by roscovitine. The inhibition was characterized by slow development and a lower affinity (EC(50)=100-300microM). Surprisingly, potassium channels were rapidly inhibited with an EC(50)=23microM, which is similar to the EC(50) for roscovitine block of cell division [Meijer, L., Borgne, A., Mulner, O., Chong, J., Blow, J., Inagaki, N., Inagaki, M., Delcros, J., Moulinoux, J., 1997. Biochemical and cellular effects of roscovitine, a potent and selective inhibitor of the cyclin-dependent kinases cdc2, cdk2 and cdk5. Eur. J. Biochem. 243, 527-536]. Potassium current inhibition seemed to result from open channel block. The high potency of these two rapid onset effects makes them complicating factors for ongoing clinical trials and research using roscovitine. Thus, the physiology and pharmacology of slow CaV2 deactivation and potassium channel block must be explored.


Subject(s)
Calcium Channels, L-Type/physiology , Caveolin 2/physiology , Ion Channel Gating/drug effects , Membrane Potentials/drug effects , Protein Kinase Inhibitors/pharmacology , Purines/pharmacology , Cell Line, Transformed , Dose-Response Relationship, Drug , Dose-Response Relationship, Radiation , Electric Stimulation/methods , Humans , Membrane Potentials/genetics , Membrane Potentials/radiation effects , Patch-Clamp Techniques/methods , Roscovitine , Transfection/methods
18.
Biophys J ; 89(3): 1681-91, 2005 Sep.
Article in English | MEDLINE | ID: mdl-15951378

ABSTRACT

The lack of a calcium channel agonist (e.g., BayK8644) for CaV2 channels has impeded their investigation. Roscovitine, a potent inhibitor of cyclin-dependent kinases 1, 2, and 5, has recently been reported to slow the deactivation of P/Q-type calcium channels (CaV2.1). We show that roscovitine also slows deactivation (EC(50) approximately 53 microM) of N-type calcium channels (CaV2.2) and investigate gating alterations induced by roscovitine. The onset of slowed deactivation was rapid ( approximately 2 s), which contrasts with a slower effect of roscovitine to inhibit N-current (EC(50) approximately 300 microM). Slow deactivation was specific to roscovitine, since it could not be induced by a closely related cyclin-dependent kinase inhibitor, olomoucine (300 microM). Intracellularly applied roscovitine failed to slow deactivation, which implies an extracellular binding site. The roscovitine-induced slow deactivation was accompanied by a slight left shift in the activation-voltage relationship, slower activation at negative potentials, and increased inactivation. Additional data showed that roscovitine preferentially binds to the open channel to slow deactivation. A model where roscovitine reduced a backward rate constant between two open states was able to reproduce the effect of roscovitine on both activation and deactivation.


Subject(s)
Calcium Channels, N-Type/physiology , Calcium Channels, P-Type/physiology , Calcium Channels, Q-Type/physiology , Purines/pharmacology , Action Potentials , Animals , Calcium/chemistry , Calcium/metabolism , Calcium Channels, P-Type/drug effects , Calcium Channels, Q-Type/drug effects , Computer Simulation , Cyclin-Dependent Kinases/metabolism , Dose-Response Relationship, Drug , Electrophysiology , Enzyme Inhibitors/pharmacology , Ganglia, Sympathetic/metabolism , Ion Channel Gating , Kinetics , Kinetin/chemistry , Kinetin/pharmacology , Markov Chains , Membrane Potentials , Models, Chemical , Neurons/metabolism , Neurotransmitter Agents , Protein Kinase Inhibitors/pharmacology , Protein Structure, Tertiary , Purines/chemistry , Rana catesbeiana , Roscovitine
SELECTION OF CITATIONS
SEARCH DETAIL
...