Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
1.
Molecules ; 26(21)2021 Oct 28.
Article in English | MEDLINE | ID: mdl-34770935

ABSTRACT

Catharanthus roseus is a well-known traditional herbal medicine for the treatment of cancer, hypertension, scald, and sore in China. Phytochemical investigation on the twigs and leaves of this species led to the isolation of two new monoterpene indole alkaloids, catharanosines A (1) and B (2), and six known analogues (3-8). Structures of 1 and 2 were established by 1H-, 13C- and 2D-NMR, and HREIMS data. The absolute configuration of 1 was confirmed by single-crystal X-ray diffraction analysis. Compound 2 represented an unprecedented aspidosperma-type alkaloid with a 2-piperidinyl moiety at C-10. Compounds 6-8 exhibited remarkable Cav3.1 low voltage-gated calcium channel (LVGCC) inhibitory activity with IC50 values of 11.83 ± 1.02, 14.3 ± 1.20, and 14.54 ± 0.99 µM, respectively.


Subject(s)
Calcium Channel Blockers/pharmacology , Calcium Channels, T-Type/chemistry , Catharanthus/chemistry , Indole Alkaloids/pharmacology , Monoterpenes/pharmacology , Plant Extracts/pharmacology , Calcium Channel Blockers/chemistry , Calcium Channels, T-Type/metabolism , Dose-Response Relationship, Drug , Indole Alkaloids/chemistry , Molecular Conformation , Molecular Docking Simulation , Molecular Dynamics Simulation , Molecular Structure , Monoterpenes/chemistry , Plant Extracts/chemistry , Structure-Activity Relationship
3.
Biochem Pharmacol ; 89(4): 536-44, 2014 Jun 15.
Article in English | MEDLINE | ID: mdl-24726441

ABSTRACT

Activation of T-type Ca²âº channels contributes to nociceptive signaling by facilitating action potential bursting and modulation of membrane potentials during periods of neuronal hyperexcitability. The role of T-type Ca²âº channels in chronic pain is supported by gene knockdown studies showing that decreased Ca(v)3.2 channel expression results in the loss of low voltage-activated (LVA) currents in dorsal root ganglion (DRG) neurons and attenuation of neuropathic pain in the chronic constriction injury (CCI) model. ABT-639 is a novel, peripherally acting, selective T-type Ca²âº channel blocker. ABT-639 blocks recombinant human T-type (Ca(v)3.2) Ca²âº channels in a voltage-dependent fashion (IC50 = 2 µM) and attenuates LVA currents in rat DRG neurons (IC50 = 8 µM). ABT-639 was significantly less active at other Ca²âº channels (e.g. Ca(v)1.2 and Ca(v)2.2) (IC50 > 30 µM). ABT-639 has high oral bioavailability (%F = 73), low protein binding (88.9%) and a low brain:plasma ratio (0.05:1) in rodents. Following oral administration ABT-639 produced dose-dependent antinociception in a rat model of knee joint pain (ED50 = 2 mg/kg, p.o.). ABT-639 (10-100 mg/kg, p.o.) also increased tactile allodynia thresholds in multiple models of neuropathic pain (e.g. spinal nerve ligation, CCI, and vincristine-induced). [corrected]. ABT-639 did not attenuate hyperalgesia in inflammatory pain models induced by complete Freund's adjuvant or carrageenan. At higher doses (e.g. 100-300 mg/kg) ABT-639 did not significantly alter hemodynamic or psychomotor function. The antinociceptive profile of ABT-639 provides novel insights into the role of peripheral T-type (Ca(v)3.2) channels in chronic pain states.


Subject(s)
Calcium Channel Blockers/therapeutic use , Calcium Channels, T-Type/metabolism , Disease Models, Animal , Heterocyclic Compounds, 2-Ring/therapeutic use , Nerve Tissue Proteins/antagonists & inhibitors , Neuralgia/drug therapy , Nociceptive Pain/drug therapy , Peripheral Nerves/drug effects , Sulfonamides/therapeutic use , Animals , Behavior, Animal/drug effects , Biological Availability , Blood-Brain Barrier/drug effects , Blood-Brain Barrier/metabolism , Calcium Channel Blockers/adverse effects , Calcium Channel Blockers/pharmacokinetics , Calcium Channel Blockers/pharmacology , Calcium Channels, T-Type/chemistry , Calcium Channels, T-Type/genetics , Cells, Cultured , Chronic Pain/drug therapy , Chronic Pain/metabolism , Dose-Response Relationship, Drug , Heterocyclic Compounds, 2-Ring/adverse effects , Heterocyclic Compounds, 2-Ring/pharmacokinetics , Heterocyclic Compounds, 2-Ring/pharmacology , Humans , Male , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Neuralgia/metabolism , Nociceptive Pain/metabolism , Peripheral Nerves/cytology , Peripheral Nerves/metabolism , Rats , Rats, Sprague-Dawley , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Sulfonamides/adverse effects , Sulfonamides/pharmacokinetics , Sulfonamides/pharmacology
4.
Pflugers Arch ; 466(4): 635-44, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24449276

ABSTRACT

Ca(2+) entry is indispensable part of intracellular Ca(2+) signaling, which is vital for most of cellular functions. Low voltage-activated (LVA or T-type) calcium channels belong to the family of voltage-gated calcium channels (VGCCs) which provide Ca(2+) entry in response to membrane depolarization. VGCCs are generally characterized by exceptional Ca(2+) selectivity combined with high permeation rate, thought to be determined by the presence in their selectivity filter of a versatile Ca(2+) binding site formed by four glutamate residues (EEEE motif). The subfamily of LVA channels includes three members, Cav3.1, Cav3.2 and Cav3.3. They all possess two aspartates instead of glutamates (i.e., EEDD motif) in their selectivity filter and are the least Ca(2+)-selective of all VGCCs. They also have the lowest conductance, weakly discriminate Ca(2+), Sr(2+) and Ba(2+) and demonstrate channel-specific sensitivity to divalent metal blockers, such as Ni(2+). The available data suggest that EEDD binding site of LVA channels is more rigid compared to EEEE one, and their selectivity permeation and block are determined by two supplementary low-affinity intrapore Ca(2+) binding sites located above and below EEDD locus. In addition, LVA channels have extracellular metal binding site that allosterically regulates channel's gating, permeation and block depending on trace metals concentration.


Subject(s)
Calcium Channels, T-Type/genetics , Calcium Channels, T-Type/metabolism , Calcium/metabolism , Models, Molecular , Amino Acid Sequence , Animals , Calcium Channels, T-Type/chemistry , Humans , Molecular Sequence Data , Permeability , Protein Structure, Secondary
5.
Bioorg Med Chem Lett ; 23(1): 119-24, 2013 Jan 01.
Article in English | MEDLINE | ID: mdl-23200256

ABSTRACT

The T-type calcium channel inhibitor Mibefradil was reported to protect the heart from atrial remodeling, a key process involved in the development of atrial fibrillation and arrhythmias. Mibefradil is not a selective T-type calcium channel inhibitor and also affects the function of different ion channels. Our aim was to develop a selective T-type calcium channel inhibitor to validate the importance of T-type-related pharmacology in atrial fibrillation. Structural optimisation of a previously disclosed hit series focussed on minimising exposure to the central nervous system and improving pharmacokinetic properties, while maintain adequate potency and selectivity. This resulted in the design of N-[[1-[2-(tert-butylcarbamoylamino)ethyl]-4-(hydroxymethyl)-4-piperidyl]methyl]-3,5-dichloro-benzamide, a novel, selective, peripherally restricted chemical probe to verify the role of T-type calcium channel inhibition on atrial fibrillation protection.


Subject(s)
Benzamides/chemistry , Calcium Channel Blockers/chemistry , Calcium Channels, T-Type/chemistry , Animals , Benzamides/chemical synthesis , Benzamides/pharmacokinetics , Calcium Channel Blockers/chemical synthesis , Calcium Channel Blockers/pharmacokinetics , Calcium Channels, T-Type/metabolism , Dogs , Drug Evaluation, Preclinical , Half-Life , Heart Rate/drug effects , Humans , Structure-Activity Relationship
6.
J Neurosci ; 30(14): 4957-69, 2010 Apr 07.
Article in English | MEDLINE | ID: mdl-20371816

ABSTRACT

beta-Catenin, together with LEF1/TCF transcription factors, activates genes involved in the proliferation and differentiation of neuronal precursor cells. In mature neurons, beta-catenin participates in dendritogenesis and synaptic function as a component of the cadherin cell adhesion complex. However, the transcriptional activity of beta-catenin in these cells remains elusive. In the present study, we found that in the adult mouse brain, beta-catenin and LEF1 accumulate in the nuclei of neurons specifically in the thalamus. The particular electrophysiological properties of thalamic neurons depend on T-type calcium channels. Cav3.1 is the predominant T-type channel subunit in the thalamus, and we hypothesized that the Cacna1g gene encoding Cav3.1 is a target of the LEF1/beta-catenin complex. We demonstrated that the expression of Cacna1g is high in the thalamus and is further increased in thalamic neurons treated in vitro with LiCl or WNT3A, activators of beta-catenin. Luciferase reporter assays confirmed that the Cacna1G promoter is activated by LEF1 and beta-catenin, and footprinting analysis revealed four LEF1 binding sites in the proximal region of this promoter. Chromatin immunoprecipitation demonstrated that the Cacna1g proximal promoter is occupied by beta-catenin in vivo in the thalamus, but not in the hippocampus. Moreover, WNT3A stimulation enhanced T-type current in cultured thalamic neurons. Together, our data indicate that the LEF1/beta-catenin complex regulates transcription of Cacna1g and uncover a novel function for beta-catenin in mature neurons. We propose that beta-catenin contributes to neuronal excitability not only by a local action at the synapse but also by activating gene expression in thalamic neurons.


Subject(s)
Calcium Channels, T-Type/genetics , Lymphoid Enhancer-Binding Factor 1/physiology , Neurons/physiology , Thalamus/physiology , Transcriptional Activation/physiology , beta Catenin/physiology , Age Factors , Animals , Calcium Channels, T-Type/biosynthesis , Calcium Channels, T-Type/chemistry , Cells, Cultured , Female , HeLa Cells , Humans , Lymphoid Enhancer-Binding Factor 1/chemistry , Male , Mice , Rats , Synapses/chemistry , Synapses/genetics , Synapses/metabolism , beta Catenin/chemistry
7.
Proteins ; 64(2): 320-42, 2006 Aug 01.
Article in English | MEDLINE | ID: mdl-16671074

ABSTRACT

We describe the regulated transcriptome of CACNA1G, a human gene for T-type Ca(v)3.1 calcium channels that is subject to extensive alternative RNA splicing. Fifteen sites of transcript variation include 2 alternative 5'-UTR promoter sites, 2 alternative 3'-UTR polyadenylation sites, and 11 sites of alternative splicing within the open reading frame. A survey of 1580 fetal and adult human brain full-length complementary DNAs reveals a family of 30 distinct transcripts, including multiple functional forms that vary in expression with development. Statistical analyses of fetal and adult transcript populations reveal patterns of linkages among intramolecular splice site configurations that change dramatically with development. A shift from nearly independent, biased splicing in fetal transcripts to strongly concerted splicing in adult transcripts suggests progressive activation of multiple "programs" of splicing regulation that reorganize molecular structures in differentiating cells. Patch-clamp studies of nine selected variants help relate splicing regulation to permutations of the gating parameters most likely to modify T-channel physiology in expressing neurons. Gating behavior reflects combinatorial interactions between variable domains so that molecular phenotype depends on ensembles of coselected domains, consistent with the observed emergence of concerted splicing during development. We conclude that the structural gene and networks of splicing regulatory factors define an integrated system for the phenotypic variation of Ca(v)3.1 biophysics during nervous system development.


Subject(s)
Calcium Channels, T-Type/physiology , Gene Expression Regulation, Developmental , Alternative Splicing , Biophysics/methods , Brain/embryology , Brain/metabolism , Calcium Channels, T-Type/chemistry , DNA, Complementary/metabolism , Genetic Variation , Humans , Kinetics , Open Reading Frames , Patch-Clamp Techniques , Protein Conformation , Protein Structure, Tertiary
8.
J Physiol ; 571(Pt 3): 519-36, 2006 Mar 15.
Article in English | MEDLINE | ID: mdl-16423851

ABSTRACT

T-type Ca2+ channels give rise to low-threshold inward currents that are central determinants of neuronal excitability. The availability of T-type Ca2+ channels is strongly influenced by voltage-dependent inactivation and recovery from inactivation. Here, we show that native and cloned T-type Ca2+ channel subunits selectively encode specific aspects of prior membrane potential changes via a powerful modulation of the rates with which these channels recover from inactivation. Increasing the duration of subthreshold (-70 to -55 mV) conditioning depolarizations caused a pronounced slowing of subsequent recovery from inactivation of both cloned (Ca(v)3.1-3.3) and native T-type channels (thalamic neurones). The scaling of recovery rates with increasing duration of conditioning depolarizations could be well described by a power law function. Different T-type channel isoforms exhibited overlapping but complementary ranges of recovery rates. Intriguingly, scaling of recovery rates was dramatically reduced in Ca(v)3.2 and Ca(v)3.3, but not Ca(v)3.1 subunits, when mock action potentials were superimposed on conditioning depolarizations. Our results suggest that different T-type channel subunits exhibit dramatic differences in scaling relationships, in addition to well-described differences in other biophysical properties. Furthermore, the availability of T-type channels is powerfully modulated over time, depending on the patterns of prior activity that these channels have encountered. These data provide a novel mechanism for cellular short-term plasticity on the millisecond to second time scale that relies on biophysical properties of specific T-type Ca2+ channel subunits.


Subject(s)
Calcium Channels, T-Type/metabolism , Geniculate Bodies/metabolism , Neuronal Plasticity , Action Potentials , Animals , Calcium/metabolism , Calcium Channels, T-Type/chemistry , Calcium Channels, T-Type/genetics , Cell Line , Humans , Kinetics , Male , Membrane Transport Proteins/chemistry , Membrane Transport Proteins/genetics , Membrane Transport Proteins/metabolism , Models, Biological , Neurons/metabolism , Patch-Clamp Techniques , Rats , Rats, Wistar , Transfection
9.
J Biol Chem ; 281(8): 4823-30, 2006 Feb 24.
Article in English | MEDLINE | ID: mdl-16377633

ABSTRACT

Molecular cloning studies have revealed that heterogeneity of T-type Ca2+ currents in native tissues arises from the three isoforms of Ca(v)3 channels: Ca(v)3.1, Ca(v)3.2, and Ca(v)3.3. From pharmacological analysis of the recombinant T-type channels, low concentrations (<50 microM) of nickel were found to selectively block the Ca(v)3.2 over the other isoforms. To date, however, the structural element(s) responsible for the nickel block on the Ca(v)3.2 T-type Ca2+ channel remain unknown. Thus, we constructed chimeric channels between the nickel-sensitive Ca(v)3.2 and the nickel-insensitive Ca(v)3.1 to localize the region interacting with nickel. Systematic assaying of serial chimeras suggests that the region preceding domain I S4 of Ca(v)3.2 contributes to nickel block. Point mutations of potential nickel-interacting sites revealed that H191Q in the S3-S4 loop of domain I significantly attenuated the nickel block of Ca(v)3.2, mimicking the nickel-insensitive blocking potency of Ca(v)3.1. These findings indicate that His-191 in the S3-S4 loop is a critical residue conferring nickel block to Ca(v)3.2 and reveal a novel role for the S3-S4 loop to control ion permeation through T-type Ca2+ channels.


Subject(s)
Calcium Channels, T-Type/genetics , Calcium Channels, T-Type/physiology , Nickel/pharmacology , Amino Acid Sequence , Animals , Binding Sites , Calcium Channels, T-Type/chemistry , Cloning, Molecular , Electrophysiology , Histidine/chemistry , Humans , Hydrogen-Ion Concentration , Inhibitory Concentration 50 , Ions , Kinetics , Molecular Sequence Data , Mutagenesis, Site-Directed , Mutation , Nickel/chemistry , Oocytes/metabolism , Plasmids/metabolism , Point Mutation , Protein Binding , Protein Isoforms , Protein Structure, Tertiary , RNA, Complementary/metabolism , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/metabolism , Recombinant Proteins/chemistry , Sequence Homology, Amino Acid , Xenopus
10.
J Neurophysiol ; 92(6): 3399-407, 2004 Dec.
Article in English | MEDLINE | ID: mdl-15254077

ABSTRACT

Low-voltage-activated T-type (Cav3) Ca2+ channels produce low-threshold spikes that trigger burst firing in many neurons. The CACNA1I gene encodes the Cav3.3 isoform, which activates and inactivates much more slowly than the other Cav3 channels. These distinctive kinetic features, along with its brain-region-specific expression, suggest that Cav3.3 channels endow neurons with the ability to generate long-lasting bursts of firing. The human CACNA1I gene contains two regions of alternative splicing: variable inclusion of exon 9 and an alternative acceptor site within exon 33, which leads to deletion of 13 amino acids (Delta33). The goal of this study is to determine the functional consequences of these variations in the full-length channel. The cDNA encoding these regions were cloned using RT-PCR from human brain, and currents were recorded by whole cell patch clamp. Introduction of the Delta33 deletion slowed the rate of channel opening. Addition of exon 9 had little effect on kinetics, whereas its addition to Delta33 channels unexpectedly slowed both activation and inactivation kinetics. Modeling of neuronal firing showed that exon 9 or Delta33 alone reduced burst firing, whereas the combination enhanced firing. The major conclusions of this study are that the intracellular regions after repeats I and IV play a role in channel gating, that their effects are interdependent, suggesting a direct interaction, and that splice variation of Cav3.3 channels provides a mechanism for fine-tuning the latency and duration of low-threshold spikes.


Subject(s)
Action Potentials/physiology , Alternative Splicing/physiology , Calcium Channels, T-Type/genetics , Calcium Channels, T-Type/physiology , Amino Acid Sequence , Calcium Channels, T-Type/chemistry , Cells, Cultured , DNA, Complementary , Exons , Humans , Ion Channel Gating/genetics , Kidney/cytology , Membrane Transport Proteins , Models, Neurological , Molecular Sequence Data , Neurons/physiology , Patch-Clamp Techniques , Structure-Activity Relationship , Thalamus/cytology , Thalamus/physiology , Transfection
11.
Neuroreport ; 15(9): 1451-5, 2004 Jun 28.
Article in English | MEDLINE | ID: mdl-15194872

ABSTRACT

We previously formulated dynamics of ion channel gates by the path probability method. In this study, we apply that theoretical approach to derive the activation rate kinetics of T-type calcium channel in thalamic relay neurons. We derive explicit expressions of the forward and backward rate constants and show that the proposed rate constants accurately capture form of the empirical time constant, and that they also provide its saturation to a constant value at depolarized membrane potentials. We also compare our derivations with linear and nonlinear thermodynamic models of rate kinetics obtained from the same calcium channel, and show that it is possible to capture saturation of the time constant for the depolarized membrane potentials by the only proposed rate constants.


Subject(s)
Calcium Channels, T-Type/chemistry , Calcium Channels, T-Type/physiology , Ion Channel Gating/physiology , Models, Neurological , Thermodynamics , Calcium/metabolism , Membrane Potentials/physiology , Neurons/physiology , Probability , Thalamus/cytology
12.
J Biol Chem ; 279(21): 21707-13, 2004 May 21.
Article in English | MEDLINE | ID: mdl-15016809

ABSTRACT

Molecular cloning and expression studies established the existence of three T-type Ca(2+) channel (Ca(v)3) alpha(1) subunits: Ca(v)3.1 (alpha(1G)), Ca(v)3.2 (alpha(1H)), and Ca(v)3.3 (alpha(1I)). Although all three channels are low voltage-activated, they display considerable differences in their kinetics, with Ca(v)3.1 and Ca(v)3.2 channels activating and inactivating much faster than Ca(v)3.3 channels. The goal of the present study was to determine the structural elements that confer the distinctively slow kinetics of Ca(v)3.3 channels. To address this question, a series of chimeric channels between Ca(v)3.1 and Ca(v)3.3 channels were constructed and expressed in Xenopus oocytes. Kinetic analysis showed that the slow activation and inactivation kinetics of the Ca(v)3.3 channel were not completely abolished by substitution with any one portion of the Ca(v)3.1 channel. Likewise, the Ca(v)3.1 channel failed to acquire the slow kinetics by simply adopting one portion of the Ca(v)3.3 channel. These findings suggest that multiple structural elements contribute to the slow kinetics of Ca(v)3.3 channels.


Subject(s)
Calcium Channels, T-Type/chemistry , Animals , Calcium Channels, T-Type/metabolism , Cloning, Molecular , DNA, Complementary/metabolism , Electrophysiology , Kinetics , Membrane Transport Proteins , Oocytes/metabolism , Plasmids/metabolism , Polymerase Chain Reaction , Protein Structure, Tertiary , Rats , Recombinant Proteins/chemistry , Time Factors , Xenopus
13.
Biophys J ; 83(1): 229-41, 2002 Jul.
Article in English | MEDLINE | ID: mdl-12080115

ABSTRACT

The full-length human Ca(v)3.3 (alpha(1I)) T-type channel was cloned, and found to be longer than previously reported. Comparison of the cDNA sequence to the human genomic sequence indicates the presence of an additional 4-kb exon that adds 214 amino acids to the carboxyl terminus and encodes the 3' untranslated region. The electrophysiological properties of the full-length channel were studied after transient transfection into 293 human embryonic kidney cells using 5 mM Ca(2+) as charge carrier. From a holding potential of -100 mV, step depolarizations elicited inward currents with an apparent threshold of -70 mV, a peak of -30 mV, and reversed at +40 mV. The kinetics of channel activation, inactivation, deactivation, and recovery from inactivation were very similar to those reported previously for rat Ca(v)3.3. Similar voltage-dependent gating and kinetics were found for truncated versions of human Ca(v)3.3, which lack either 118 or 288 of the 490 amino acids that compose the carboxyl terminus. A major difference between these constructs was that the full-length isoform generated twofold more current. These results suggest that sequences in the distal portion of Ca(v)3.3 play a role in channel expression. Studies on the voltage-dependence of activation revealed that a fraction of channels did not gate as low voltage-activated channels, requiring stronger depolarizations to open. A strong depolarizing prepulse (+100 mV, 200 ms) increased the fraction of channels that gated at low voltages. In contrast, human Ca(v)3.3 isoforms with shorter carboxyl termini were less affected by a prepulse. Therefore, Ca(v)3.3 is similar to high voltage-activated Ca(2+) channels in that depolarizing prepulses can regulate their activity, and their carboxy termini play a role in modulating channel activity.


Subject(s)
Calcium Channels, T-Type/chemistry , Calcium Channels, T-Type/genetics , Amino Acid Sequence , Animals , Biophysical Phenomena , Biophysics , Brain/metabolism , Cell Line , Cloning, Molecular , DNA, Complementary/metabolism , Electrophysiology , Gene Library , Humans , Kinetics , Membrane Transport Proteins , Models, Genetic , Molecular Sequence Data , Polymerase Chain Reaction , Rats , Time Factors , Transfection
14.
Biophys J ; 82(4): 1894-906, 2002 Apr.
Article in English | MEDLINE | ID: mdl-11916848

ABSTRACT

Recovery from inactivation of T-type Ca channels is slow and saturates at moderate hyperpolarizing voltage steps compared with Na channels. To explore this unique kinetic pattern we measured gating and ionic currents in two closely related isoforms of T-type Ca channels. Gating current recovers from inactivation much faster than ionic current, and recovery from inactivation is much more voltage dependent for gating current than for ionic current. There is a lag in the onset of gating current recovery at -80 mV, but no lag is discernible at -120 mV. The delay in recovery from inactivation of ionic current is much more evident at all voltages. The time constant for the decay of off gating current is very similar to the time constant of deactivation of open channels (ionic tail current), and both are strongly voltage dependent over a wide voltage range. Apparently, the development of inactivation has little influence on the initial deactivation step. These results suggest that movement of gating charge occurs for inactivated states very quickly. In contrast, the transitions from inactivated to available states are orders of magnitude slower, not voltage dependent, and are rate limiting for ionic recovery. These findings support a deactivation-first path for T-type Ca channel recovery from inactivation. We have integrated these concepts into an eight-state kinetic model, which can account for the major characteristics of T-type Ca channel inactivation.


Subject(s)
Calcium Channels, T-Type/chemistry , Calcium Channels, T-Type/metabolism , Calcium Channels/metabolism , Amino Acids/chemistry , Calcium/metabolism , Cell Line , Computer Simulation , DNA, Complementary/metabolism , Electrophysiology , Humans , Ions , Kinetics , Protein Isoforms , Time Factors , Transfection
15.
J Biol Chem ; 277(10): 8449-56, 2002 Mar 08.
Article in English | MEDLINE | ID: mdl-11751928

ABSTRACT

Low voltage activated, voltage-operated Ca(2+) channels are expressed in rodent male germ cells and are believed to be pivotal in induction of the acrosome reaction in mouse spermatozoa. However, in humans, very little is known about expression of voltage-operated Ca(2+) channels in male germ cells or their function. We have used reverse transcription-polymerase chain reaction, in situ hybridization, and patch clamp recording to investigate the expression of low voltage activated voltage-operated Ca(2+) channels in human male germ cells. We report that full-length transcripts for both alpha(1G) and alpha(1H) low voltage activated channel subunits are expressed in human testis. Multiple isoforms of alpha(1G) are present in the testis and at least two isoforms of alpha(1H), including a splice variant not previously described in the human. Transcripts for all the isoforms of both alpha(1G) and alpha(1H) were detected by reverse transcription-polymerase chain reaction on mRNA isolated from human spermatogenic cells. In situ hybridization for alpha(1G) and alpha(1H) localized transcripts both in germ cells and in other cell types in the testis. Within the seminiferous tubules, alpha(1H) was detected primarily in germ cells. Using the whole cell patch clamp technique, we detected T-type voltage-operated Ca(2+) channel currents in isolated human male germ cells, although the current amplitude and frequency of occurrence were low in comparison to the occurrence of T-currents in murine male germ cells. We conclude that low voltage activated voltage-operated Ca(2+) channels are expressed in cells of the human male germ line.


Subject(s)
Calcium Channels, T-Type/biosynthesis , Calcium Channels, T-Type/chemistry , Spermatozoa/metabolism , Animals , Calcium/metabolism , Cells, Cultured , DNA, Complementary/metabolism , Humans , In Situ Hybridization , Male , Mice , Models, Genetic , Molecular Sequence Data , Patch-Clamp Techniques , Polymerase Chain Reaction , Protein Binding , Protein Isoforms/metabolism , RNA, Messenger/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Seminiferous Tubules/metabolism , Testis/metabolism
16.
J Physiol ; 533(Pt 2): 467-78, 2001 Jun 01.
Article in English | MEDLINE | ID: mdl-11389205

ABSTRACT

1. Voltage-sensitive Ca(2+) channels (VSCCs) are often heteromultimeric complexes. The VSCC subtype specifically expressed by skeletal muscle has long been known to contain a gamma subunit, gamma(1), that is only expressed in this tissue. Recent work, initiated by the identification of the mutation present in the stargazer mouse, has led to the identification of a series of novel potential Ca(2+) channel gamma subunits expressed in the CNS. 2. Based on bioinformatic techniques we identified and cloned the human gamma(2), gamma(3) and gamma(4) subunits. 3. TaqMan analysis was used to quantitatively characterise the mRNA expression patterns of all the gamma subunits. All three subunits were extensively expressed in adult brain with overlapping but subunit-specific distributions. gamma(2) and gamma(3) were almost entirely restricted to the brain, but gamma(4) expression was seen in a broad range of peripheral tissues. 4. Using a myc epitope the gamma(2) subunit was tagged both intracellularly at the C-terminus and on a predicted extracellular site between the first and second transmembrane domains. The cellular distribution was then examined immunocytochemically, which indicated that a substantial proportion of the cellular pool of the gamma(2) subunit was present on the plasma membrane and provided initial evidence for the predicted transmembrane topology of the gamma subunits. 5. Using co-transfection techniques we investigated the functional effects of each of the gamma subunits on the biophysics of the T-type VSCC encoded by the alpha(1I) subunit. This revealed a substantially slowed rate of deactivation in the presence of gamma(2). In contrast, there was no significant corresponding effect of either gamma(3) or gamma(4) on alpha(1I) subunit-mediated currents.


Subject(s)
Calcium Channels, T-Type/genetics , Calcium Channels, T-Type/metabolism , Calcium Channels/genetics , Calcium Channels/metabolism , Neurons/chemistry , Animals , Calcium Channels/chemistry , Calcium Channels, T-Type/chemistry , Cell Line , Cloning, Molecular , DNA, Complementary , Electrophysiology , Gene Expression/physiology , Humans , Kidney/cytology , Kinetics , Membrane Potentials/physiology , Mice , Mice, Neurologic Mutants , Molecular Sequence Data , RNA, Messenger/analysis , Sequence Homology, Amino Acid , Taq Polymerase , Transfection
17.
J Biol Chem ; 276(6): 3999-4011, 2001 Feb 09.
Article in English | MEDLINE | ID: mdl-11073957

ABSTRACT

Voltage-gated calcium channels represent a heterogenous family of calcium-selective channels that can be distinguished by their molecular, electrophysiological, and pharmacological characteristics. We report here the molecular cloning and functional expression of three members of the low voltage-activated calcium channel family from rat brain (alpha(1G), alpha(1H), and alpha(1I)). Northern blot and reverse transcriptase-polymerase chain reaction analyses show alpha(1G), alpha(1H), and alpha(1I) to be expressed throughout the newborn and juvenile rat brain. In contrast, while alpha(1G) and alpha(1H) mRNA are expressed in all regions in adult rat brain, alpha(1I) mRNA expression is restricted to the striatum. Expression of alpha(1G), alpha(1H), and alpha(1I) subunits in HEK293 cells resulted in calcium currents with typical T-type channel characteristics: low voltage activation, negative steady-state inactivation, strongly voltage-dependent activation and inactivation, and slow deactivation. In addition, the direct electrophysiological comparison of alpha(1G), alpha(1H), and alpha(1I) under identical recording conditions also identified unique characteristics including activation and inactivation kinetics and permeability to divalent cations. Simulation of alpha(1G), alpha(1H), and alpha(1I) T-type channels in a thalamic neuron model cell produced unique firing patterns (burst versus tonic) typical of different brain nuclei and suggests that the three channel types make distinct contributions to neuronal physiology.


Subject(s)
Calcium Channels, T-Type/genetics , Calcium Channels, T-Type/physiology , Alternative Splicing , Amino Acid Sequence , Animals , Barium/metabolism , Base Sequence , Brain/metabolism , Calcium/metabolism , Calcium Channels, T-Type/chemistry , Calcium Channels, T-Type/metabolism , Cell Line , Cloning, Molecular , DNA, Complementary , Expressed Sequence Tags , Humans , Ion Channel Gating , Kinetics , Molecular Sequence Data , Permeability , RNA, Messenger/genetics , Rats , Sequence Homology, Amino Acid
18.
Cell Mol Life Sci ; 56(7-8): 660-9, 1999 Nov 15.
Article in English | MEDLINE | ID: mdl-11212313

ABSTRACT

Despite the wealth of information on voltage-gated calcium channels, little is known about low voltage-activated, T-type channels. The ability of the antihypertensive drug mibefradil to selectively block T-type channels has generated much interest in their structure, physiology and pharmacology. This review covers the cloning of a new family of calcium channels, their putative structure, the electrophysiological evidence that demonstrated that these complementary DNAs encoded low voltage-activated, T-type channels, the tissue expression of these genes, and concludes with a discussion of their possible physiological roles.


Subject(s)
Calcium Channels, T-Type/metabolism , Ion Channel Gating , Animals , Calcium Channels, T-Type/chemistry , Calcium Channels, T-Type/genetics , Cloning, Molecular , Electrophysiology , Evolution, Molecular , Gene Expression Profiling , Humans , Phylogeny , RNA, Messenger/analysis , RNA, Messenger/genetics
SELECTION OF CITATIONS
SEARCH DETAIL