Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 90
1.
Mol Psychiatry ; 22(12): 1735-1745, 2017 Dec.
Article En | MEDLINE | ID: mdl-28194001

Genetic factors significantly influence susceptibility for substance abuse and mood disorders. Rodent studies have begun to elucidate a role of Cav1.3 L-type Ca2+ channels in neuropsychiatric-related behaviors, such as addictive and depressive-like behaviors. Human studies have also linked the CACNA1D gene, which codes for the Cav1.3 protein, with bipolar disorder. However, the neurocircuitry and the molecular mechanisms underlying the role of Cav1.3 in neuropsychiatric phenotypes are not well established. In the present study, we directly manipulated Cav1.3 channels in Cav1.2 dihydropyridine insensitive mutant mice and found that ventral tegmental area (VTA) Cav1.3 channels mediate cocaine-related and depressive-like behavior through a common nucleus accumbens (NAc) shell calcium-permeable α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptor (CP-AMPAR) mechanism that requires GluA1 phosphorylation at S831. Selective activation of VTA Cav1.3 with (±)-BayK-8644 (BayK) enhanced cocaine conditioned place preference and cocaine psychomotor activity while inducing depressive-like behavior, an effect not observed in S831A phospho-mutant mice. Infusion of the CP-AMPAR-specific blocker Naspm into the NAc shell reversed the cocaine and depressive-like phenotypes. In addition, activation of VTA Cav1.3 channels resulted in social behavioral deficits. In contrast to the cocaine- and depression-related phenotypes, GluA1/A2 AMPARs in the NAc core mediated social deficits, independent of S831-GluA1 phosphorylation. Using a candidate gene analysis approach, we also identified single-nucleotide polymorphisms in the CACNA1D gene associated with cocaine dependence in human subjects. Together, our findings reveal novel, overlapping mechanisms through which VTA Cav1.3 mediates cocaine-related, depressive-like and social phenotypes, suggesting that Cav1.3 may serve as a target for the treatment of neuropsychiatric symptoms.


Affect/physiology , Calcium Channels, L-Type/metabolism , Cocaine/pharmacology , Dopamine Uptake Inhibitors/pharmacology , Nucleus Accumbens/metabolism , Ventral Tegmental Area/metabolism , Affect/drug effects , Animals , Calcium Channels, L-Type/genetics , Cocaine-Related Disorders/metabolism , Conditioning, Psychological/drug effects , Conditioning, Psychological/physiology , Depression/metabolism , Disease Models, Animal , Genetic Association Studies , Humans , Male , Mice, Inbred C57BL , Mice, Transgenic , Motor Activity/drug effects , Motor Activity/physiology , Nucleus Accumbens/drug effects , Post-Synaptic Density/drug effects , Post-Synaptic Density/metabolism , Receptors, AMPA/metabolism , Social Behavior , Ventral Tegmental Area/drug effects
2.
Transl Psychiatry ; 6(12): e974, 2016 12 06.
Article En | MEDLINE | ID: mdl-27922638

Extinction-based exposure therapy is used to treat anxiety- and trauma-related disorders; however, there is the need to improve its limited efficacy in individuals with impaired fear extinction learning and to promote greater protection against return-of-fear phenomena. Here, using 129S1/SvImJ mice, which display impaired fear extinction acquisition and extinction consolidation, we revealed that persistent and context-independent rescue of deficient fear extinction in these mice was associated with enhanced expression of dopamine-related genes, such as dopamine D1 (Drd1a) and -D2 (Drd2) receptor genes in the medial prefrontal cortex (mPFC) and amygdala, but not hippocampus. Moreover, enhanced histone acetylation was observed in the promoter of the extinction-regulated Drd2 gene in the mPFC, revealing a potential gene-regulatory mechanism. Although enhancing histone acetylation, via administering the histone deacetylase (HDAC) inhibitor MS-275, does not induce fear reduction during extinction training, it promoted enduring and context-independent rescue of deficient fear extinction consolidation/retrieval once extinction learning was initiated as shown following a mild conditioning protocol. This was associated with enhanced histone acetylation in neurons of the mPFC and amygdala. Finally, as a proof-of-principle, mimicking enhanced dopaminergic signaling by L-dopa treatment rescued deficient fear extinction and co-administration of MS-275 rendered this effect enduring and context-independent. In summary, current data reveal that combining dopaminergic and epigenetic mechanisms is a promising strategy to improve exposure-based behavior therapy in extinction-impaired individuals by initiating the formation of an enduring and context-independent fear-inhibitory memory.


Dopamine/physiology , Extinction, Psychological/physiology , Fear/physiology , Histone Acetyltransferases/physiology , Signal Transduction/physiology , Amygdala/physiology , Animals , Benzamides/pharmacology , Combined Modality Therapy , Conditioning, Classical/drug effects , Conditioning, Classical/physiology , Extinction, Psychological/drug effects , Fear/drug effects , Implosive Therapy , Levodopa/pharmacology , Male , Mice , Prefrontal Cortex/physiology , Pyridines/pharmacology , Signal Transduction/drug effects
3.
J Neurochem ; 99(4): 1197-206, 2006 Nov.
Article En | MEDLINE | ID: mdl-17026527

Dopamine D(2) long (D(2)L) and D(2) short (D(2)S) isoforms of the D(2) receptor play an important role in psychostimulant-induced neuronal adaptations. In this study, we used quantitative real-time PCR to specifically amplify these two splice variants to examine their mRNA expression in the dorsal striatum (dStr), nucleus accumbens (NAc) and the ventral tegmental area (VTA) of amphetamine-sensitized C57BL/6 mice. We found a significant increase in D(2)L mRNA in the VTA and dStr of amphetamine-treated mice that positively correlated with the sensitized locomotor response. We also found a significant increase in D(2)S mRNA in the VTA. We further examined the role of the Ca(v)1.3 subtype of L-type Ca(2+) channels in up-regulation of D(2)L and D(2)S mRNA in the VTA. Amphetamine-pretreated Ca(v)1.3 wild-type (Ca(v)1.3(+/+)) mice exhibited sensitized behavior and a significant increase in D(2)L and D(2)S mRNA compared with saline-pretreated mice Amphetamine-pretreated homozygous Ca(v)1.3 knockout (Ca(v)1.3(-/-)) mice did not exhibit sensitized behavior. There was a significant increase in D(2)S mRNA, but not D(2)L mRNA. In conclusion, our results find that amphetamine increases D(2)L mRNA expression in the dStr and the VTA, an adaptation that correlates with expression of sensitized behavior and dependence on Ca(v)1.3 Ca(2+) channels.


Amphetamine/pharmacology , Calcium Channels, L-Type/drug effects , Neostriatum/drug effects , Receptors, Dopamine D2/genetics , Ventral Tegmental Area/drug effects , Amphetamine-Related Disorders/genetics , Amphetamine-Related Disorders/metabolism , Amphetamine-Related Disorders/physiopathology , Animals , Calcium Channels, L-Type/genetics , Calcium Channels, L-Type/metabolism , Central Nervous System Stimulants/pharmacology , Disease Models, Animal , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Motor Activity/drug effects , Motor Activity/genetics , Neostriatum/metabolism , Neostriatum/physiopathology , RNA, Messenger/drug effects , RNA, Messenger/metabolism , Up-Regulation/drug effects , Up-Regulation/genetics , Ventral Tegmental Area/metabolism , Ventral Tegmental Area/physiopathology
4.
Biochem Soc Trans ; 34(Pt 5): 903-9, 2006 Nov.
Article En | MEDLINE | ID: mdl-17052224

Voltage-gated LTCCs (L-type Ca2+ channels) are established drug targets for the treatment of cardiovascular diseases. LTCCs are also expressed outside the cardiovascular system. In the brain, LTCCs control synaptic plasticity in neurons, and DHP (dihydropyridine) LTCC blockers such as nifedipine modulate brain function (such as fear memory extinction and depression-like behaviour). Voltage-sensitive Ca2+ channels Cav1 .2 and Cav1.3 are the predominant brain LTCCs. As DHPs and other classes of organic LTCC blockers inhibit both isoforms, their pharmacological distinction is impossible and their individual contributions to defined brain functions remain largely unknown. Here, we summarize our recent experiments with two genetically modified mouse strains, which we generated to explore the individual biophysical features of Cav1.2 and Cav1.3 LTCCs and to determine their relative contributions to various physiological peripheral and neuronal functions. The results described here also allow predictions about the pharmacotherapeutic potential of isoform-selective LTCC modulators.


Brain/physiology , Calcium Channels, L-Type/physiology , Calcium Channels/physiology , Animals , Calcium Channels/deficiency , Calcium Channels/genetics , Calcium Channels, L-Type/deficiency , Calcium Channels, L-Type/genetics , Hippocampus/physiology , Long-Term Potentiation , Mice , Mice, Knockout , Neurons/physiology , Protein Isoforms/physiology , Receptors, N-Methyl-D-Aspartate/physiology
5.
Neuroscience ; 139(3): 1005-15, 2006.
Article En | MEDLINE | ID: mdl-16542784

Ca(V)1.2 and Ca(V)1.3, are the main dihydropyridine-sensitive L-type calcium channel isoforms in the brain. To reveal the contribution of each isoform to the neuronal activation pattern elicited by the dihydropyridine L-type calcium channel activator BayK 8644, we utilized Fos expression as a marker of neuronal activation in mutant mice (Ca(V)1.2(DHP-/-) mice) expressing dihydropyridine-insensitive Ca(V)1.2 L-type calcium channels. BayK 8644-treated wildtype mice displayed intense and widespread Fos expression throughout the neuroaxis in 77 of 80 brain regions quantified. The Fos response in Ca(V)1.2(DHP-/-) mice was greatly attenuated or absent in most of these areas, suggesting that a major part of the widespread Fos induction including most cortical areas was mediated by Ca(V)1.2 L-type calcium channels. BayK 8644-induced Fos expression in Ca(V)1.2(DHP-/-) mice indicating predominantly Ca(V)1.3 L-type calcium channel-mediated activation was noted in more restricted neuronal populations (20 of 80), in particular in the central amygdala, the bed nucleus of the stria terminalis, paraventricular hypothalamic nucleus, lateral preoptic area, locus coeruleus, lateral parabrachial nucleus, central nucleus of the inferior colliculus, and nucleus of the solitary tract. Our data indicate that selective stimulation of other than Ca(V)1.2 L-type calcium channels, mostly Ca(V)1.3, causes neuronal activation in a specific set of mainly limbic, hypothalamic and brainstem areas, which are associated with functions including integration of emotion-related behavior. Hence, selective modulation of Ca(V)1.3 L-type calcium channels could represent a novel (pharmacotherapeutic) tool to influence these CNS functions.


Brain/metabolism , Calcium Channels, L-Type/metabolism , Neurons/metabolism , 3-Pyridinecarboxylic acid, 1,4-dihydro-2,6-dimethyl-5-nitro-4-(2-(trifluoromethyl)phenyl)-, Methyl ester/pharmacology , Animals , Brain/drug effects , Calcium Channel Agonists/pharmacology , Calcium Channels, L-Type/drug effects , Glial Fibrillary Acidic Protein/metabolism , Immunohistochemistry , Male , Mice , Neurons/drug effects , Protein Isoforms/drug effects , Protein Isoforms/metabolism , Proto-Oncogene Proteins c-fos/drug effects , Proto-Oncogene Proteins c-fos/metabolism
6.
Neuroscience ; 120(2): 435-42, 2003.
Article En | MEDLINE | ID: mdl-12890513

Neuronal L-type calcium channels have been implicated in pain perception and neuronal synaptic plasticity. To investigate this we have examined the effect of disrupting the gene encoding the CaV1.3 (alpha 1D) alpha subunit of L-type Ca2+ channels on neurological function, acute nociceptive behavior, and hippocampal synaptic function in mice. CaV1.3 alpha 1 subunit knockout (CaV1.3 alpha 1(-/-)) mice had relatively normal neurological function with the exception of reduced auditory evoked behavioral responses and lower body weight. Baseline thermal and mechanical thresholds were unaltered in these animals. CaV1.3 alpha 1(-/-) mice were also examined for differences in N-methyl-D-aspartate (NMDA) receptor-dependent (100 Hz tetanization for 1 s) and NMDA receptor-independent (200 Hz in 100 microM DL-2-amino-5-phosphopentanoic acid) long-term potentiation within the CA1 region of the hippocampus. Both NMDA receptor-dependent and NMDA receptor-independent forms of long-term potentiation were expressed normally. Radioligand binding studies revealed that the density of (+)[3H]isradipine binding sites in brain homogenates was reduced by 20-25% in CaV1.3 alpha 1(-/-) mice, without any detectable change in CaV1.2 (alpha 1C) protein levels as detected using Western blot analysis. Taken together these data indicate that following loss of CaV1.3 alpha 1 subunit expression there is sufficient residual activity of other Ca2+ channel subtypes to support NMDA receptor-independent long-term potentiation and some forms of sensory behavior/function.


Calcium Channels, L-Type/metabolism , Neurons/physiology , Phenotype , Synapses/physiology , Valine/analogs & derivatives , Animals , Behavior, Animal , Binding Sites , Body Weight , Calcium Channel Blockers/pharmacokinetics , Calcium Channels , Calcium Channels, L-Type/genetics , Calcium Channels, L-Type/physiology , Case-Control Studies , Deoxyadenine Nucleotides/pharmacology , Dose-Response Relationship, Drug , Drinking , Ear/physiology , Eating , Electric Stimulation , Excitatory Postsynaptic Potentials/drug effects , Excitatory Postsynaptic Potentials/physiology , Hippocampus/cytology , Hippocampus/physiology , Immunohistochemistry/methods , In Situ Hybridization/methods , Isotope Labeling/methods , Isradipine/pharmacokinetics , Long-Term Potentiation/physiology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Motor Activity , Oligonucleotide Probes , Pain Threshold , Rats , Rotation , Time Factors , Valine/pharmacology
7.
J Mol Biol ; 332(1): 171-82, 2003 Sep 05.
Article En | MEDLINE | ID: mdl-12946355

The three-dimensional structure of the skeletal muscle voltage-gated L-type calcium channel (Ca(v)1.1; dihydropyridine receptor, DHPR) was determined using electron cryo-microscopy and single-particle averaging. The structure shows a single channel complex with an approximate total molecular mass of 550 kDa, corresponding to the five known subunits of the DHPR, and bound detergent and lipid. Features visible in our structure together with antibody labeling of the beta and alpha(2) subunits allowed us to assign locations for four of the five subunits within the structure. The most striking feature of the structure is the extra-cellular alpha(2) subunit that protrudes from the membrane domain in close proximity to the alpha(1) subunit. The cytosolic beta subunit is located close to the membrane and adjacent to subunits alpha(1), gamma and delta. Our structure correlates well with the functional and biochemical data available for this channel and suggests a three-dimensional model for the excitation-contraction coupling complex consisting of DHPR tetrads and the calcium release channel.


Calcium Channels, L-Type/chemistry , Cryoelectron Microscopy/methods , Protein Structure, Quaternary , Animals , Calcium Channels, L-Type/isolation & purification , Calcium Channels, L-Type/ultrastructure , Models, Molecular , Molecular Weight , Protein Subunits/chemistry , Rabbits , Ryanodine Receptor Calcium Release Channel/chemistry
8.
Hear Res ; 178(1-2): 95-105, 2003 Apr.
Article En | MEDLINE | ID: mdl-12684182

Voltage-gated Ca(2+) channels formed by subunits (class D Ca(2+) channels) tightly regulate neurotransmitter release from cochlear inner hair cells (IHCs) by controlling the majority of depolarisation-induced Ca(2+) entry. We have recently shown that the absence of these channels can cause deafness and degeneration of outer hair cells (OHCs) and IHCs in alpha1D-deficient mice (alpha1D(-/-)) (Platzer et al., 2000. Cell 102, 89-97). We investigated the time-dependent patterns of degeneration during postnatal development in the alpha1D(-/-) mouse cochlea using light and electron microscopy. At postnatal day 3 (P3), electron microscopy revealed no morphological aberrations in sensory cells, in afferent as well as in efferent nerve endings. But at P7 we observed a beginning degeneration of afferent nerve fibres by electron microscopy. By P15, we found a loss of OHCs in apical turns but electron microscopy revealed no ultrastructural changes in IHCs and efferent axons as compared to C57 black control animals (C57BL). We demonstrated by serial ultrathin sectioning of 15 days old alpha1D(-/-) mice that intact efferent nerve fibres formed direct contacts with IHCs as the degeneration of afferent nerve fibres progressed. We also saw a notable degeneration of spiral ganglion cells at P15. By 8 months, nearly all spiral ganglion and sensory cells of the organ of Corti were absent. Random ultrathin sectioning gave the impression that synaptic bodies abundant in wild-type animals were absent in nearly all alpha1D(-/-) mice investigated. We conclude that besides presumably reduced synaptic bodies the absence of class D L-type Ca(2+) channels does not prevent morphological development of the cochlea until P3 but may cause cochlear degeneration thereafter. The observed pattern of degeneration involves afferent nerve fibres (P7) followed by cell bodies in the spiral ganglion (P15), OHCs (P15) and IHCs (after P15).


Calcium Channels, L-Type/physiology , Cochlea/ultrastructure , Aging/metabolism , Animals , Animals, Newborn/anatomy & histology , Animals, Newborn/growth & development , Animals, Newborn/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Microscopy, Electron
9.
Biophys J ; 81(6): 3308-23, 2001 Dec.
Article En | MEDLINE | ID: mdl-11720994

The association of L-type Ca(2+) channels to the secretory granules and its functional significance to secretion was investigated in mouse pancreatic B cells. Nonstationary fluctuation analysis showed that the B cell is equipped with <500 alpha1(C) L-type Ca(2+) channels, corresponding to a Ca(2+) channel density of 0.9 channels per microm(2). Analysis of the kinetics of exocytosis during voltage-clamp depolarizations revealed an early component that reached a peak rate of 1.1 pFs(-1) (approximately 650 granules/s) 25 ms after onset of the pulse and is completed within approximately 100 ms. This component represents a subset of approximately 60 granules situated in the immediate vicinity of the L-type Ca(2+) channels, corresponding to approximately 10% of the readily releasable pool of granules. Experiments involving photorelease of caged Ca(2+) revealed that the rate of exocytosis was half-maximal at a cytoplasmic Ca(2+) concentration of 17 microM, and concentrations >25 microM are required to attain the rate of exocytosis observed during voltage-clamp depolarizations. The rapid component of exocytosis was not affected by inclusion of millimolar concentrations of the Ca(2+) buffer EGTA but abolished by addition of exogenous L(C753-893), the 140 amino acids of the cytoplasmic loop connecting the 2(nd) and 3(rd) transmembrane region of the alpha1(C) L-type Ca(2+) channel, which has been proposed to tether the Ca(2+) channels to the secretory granules. In keeping with the idea that secretion is determined by Ca(2+) influx through individual Ca(2+) channels, exocytosis triggered by brief (15 ms) depolarizations was enhanced 2.5-fold by the Ca(2+) channel agonist BayK8644 and 3.5-fold by elevating extracellular Ca(2+) from 2.6 to 10 mM. Recordings of single Ca(2+) channel activity revealed that patches predominantly contained no channels or many active channels. We propose that several Ca(2+) channels associate with a single granule thus forming a functional unit. This arrangement is important in a cell with few Ca(2+) channels as it ensures maximum usage of the Ca(2+) entering the cell while minimizing the influence of stochastic variations of the Ca(2+) channel activity.


B-Lymphocytes/metabolism , Calcium Channels/metabolism , Exocytosis , Insulin/metabolism , Pancreas/metabolism , Animals , Calcium/metabolism , Cell Membrane/metabolism , Electrophysiology , Mice , Mice, Knockout , Microscopy, Fluorescence , Protein Binding , Reverse Transcriptase Polymerase Chain Reaction , Time Factors
11.
J Biol Chem ; 276(25): 22100-6, 2001 Jun 22.
Article En | MEDLINE | ID: mdl-11285265

In cochlea inner hair cells (IHCs), L-type Ca(2+) channels (LTCCs) formed by alpha1D subunits (D-LTCCs) possess biophysical and pharmacological properties distinct from those of alpha1C containing C-LTCCs. We investigated to which extent these differences are determined by alpha1D itself by analyzing the biophysical and pharmacological properties of cloned human alpha1D splice variants in tsA-201 cells. Variant alpha1D(8A,) containing exon 8A sequence in repeat I, yielded alpha1D protein and L-type currents, whereas no intact protein and currents were observed after expression with exon 8B. In whole cell patch-clamp recordings (charge carrier 15-20 mm Ba(2+)), alpha1D(8A) - mediated currents activated at more negative voltages (activation threshold, -45.7 versus -31.5 mV, p < 0.05) and more rapidly (tau(act) for maximal inward currents 0.8 versus 2.3 ms; p < 0.05) than currents mediated by rabbit alpha1C. Inactivation during depolarizing pulses was slower than for alpha1C (current inactivation after 5-s depolarizations by 90 versus 99%, p < 0.05) but faster than for LTCCs in IHCs. The sensitivity for the dihydropyridine (DHP) L-type channel blocker isradipine was 8.5-fold lower than for alpha1C. Radioligand binding experiments revealed that this was not due to a lower affinity for the DHP binding pocket, suggesting that differences in the voltage-dependence of DHP block account for decreased sensitivity of D-LTCCs. Our experiments show that alpha1D(8A) subunits can form slowly inactivating LTCCs activating at more negative voltages than alpha1C. These properties should allow D-LTCCs to control physiological processes, such as diastolic depolarization in sinoatrial node cells, neurotransmitter release in IHCs and neuronal excitability.


Calcium Channels, L-Type/metabolism , Amino Acid Sequence , Calcium Channel Blockers/pharmacology , Calcium Channels, L-Type/genetics , Cell Line , Cloning, Molecular , DNA, Complementary , Humans , Isradipine/pharmacology , Molecular Sequence Data
12.
J Biol Chem ; 276(16): 12730-5, 2001 Apr 20.
Article En | MEDLINE | ID: mdl-11278630

We investigated the mechanism of interaction of individual L-type channel amino acid residues with dihydropyridines within a dihydropyridine-sensitive alpha1A subunit (alpha1A(DHP)). Mutation of individual residues in repeat III and expression in Xenopus oocytes revealed that Thr(1393) is not required for dihydropyridine interaction but that bulky side chains (tyrosine, phenylalanine) in this position sterically inhibit dihydropyridine coordination. In position 1397 a side chain carbonyl group was required for high antagonist sensitivity. Agonist function required the complete amide group of a glutamine residue. Val(1516) and Met(1512) side chains were required for agonist (Val(1516)) and antagonist (Val(1516), Met(1512)) sensitivity. Replacement of Ile(1504) and Ile(1507) by alpha1A phenylalanines was tolerated. Substitution of Thr(1393) by phenylalanine or Val(1516) by alanine introduced voltage dependence of antagonist action into alpha1A(DHP), suggesting that these residues form part of a mechanism mediating voltage dependence of dihydropyridine sensitivity. Our data provide important insight into dihydropyridine binding to alpha1A(DHP) which could facilitate the development of alpha1A-selective modulators. By modulating P/Q-type Ca(2+) channels such drugs could serve as new anti-migraine therapeutics.


Calcium Channels, L-Type/chemistry , Calcium Channels, L-Type/physiology , Dihydropyridines/pharmacology , Oocytes/physiology , Alanine , Amino Acid Sequence , Amino Acid Substitution , Animals , Binding Sites , Calcium Channel Blockers/pharmacology , Dihydropyridines/pharmacokinetics , Female , Isoleucine , Isradipine/pharmacology , Kinetics , Methionine , Models, Molecular , Molecular Sequence Data , Mutagenesis, Site-Directed , Oocytes/drug effects , Phenylalanine , Protein Structure, Secondary , Protein Subunits , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Valine , Xenopus laevis
13.
Gastroenterology ; 120(2): 480-9, 2001 Feb.
Article En | MEDLINE | ID: mdl-11159888

BACKGROUND & AIMS: Circular smooth muscle phasic contractions and tone are suppressed during colonic inflammation, but the contributing factors are poorly understood. This study investigated if the expression level of voltage-gated long-lasting (L-type) Ca(2+) channel protein and functional Ca(2+) channel current are down-regulated in the circular muscle cells of the inflamed canine colon. METHODS: L-type Ca(2+) channel expression was compared between normal and inflamed smooth muscle cells by Western immunoblots using an antibody directed against the pore-forming alpha 1C-subunit, and patch-clamp methods were used to evaluate Ca(2+) channel current density. RESULTS: The expression of the L-type Ca(2+) channel protein was significantly reduced in inflamed compared with normal circular smooth muscle cell membranes, and this finding was associated with suppressed levels of Ca(2+) channel current in patch-clamped cells. The L-type Ca(2+) channel current in normal and inflamed cells increased proportionately in response to Bay K 8644, but the maximal current density was still lower in the inflamed cells. Acetylcholine increased the L-type Ca(2+) channel current in normal but not in inflamed cells. CONCLUSIONS: The expression level of L-type Ca(2+) channels is down-regulated in the circular smooth muscle cell membranes of the inflamed colon, which may result in reduced Ca(2+) influx. The functional and pharmacologic properties of the channels seem normal. Although some Ca(2+) channels are still present in the inflamed cells, acetylcholine does not activate these channels, which may be caused by additional upstream defects in the receptor signaling cascade. The down-regulation of L-type Ca(2+) channel expression may suppress circular smooth muscle contractions in the inflamed colon and contribute to the abnormalities in motility and digestion observed during inflammatory disorders.


Calcium Channels, L-Type/metabolism , Colitis/metabolism , Colon/metabolism , Muscle, Smooth/metabolism , 3-Pyridinecarboxylic acid, 1,4-dihydro-2,6-dimethyl-5-nitro-4-(2-(trifluoromethyl)phenyl)-, Methyl ester/pharmacology , Acetylcholine/pharmacology , Animals , Calcium/metabolism , Calcium Channel Agonists/pharmacology , Calcium Channel Blockers/pharmacology , Calcium Channels, L-Type/analysis , Colitis/immunology , Colon/chemistry , Colon/immunology , Dogs , Down-Regulation/physiology , Female , Immunoblotting , Male , Membrane Potentials/drug effects , Membrane Potentials/physiology , Muscle, Smooth/chemistry , Muscle, Smooth/immunology , Nifedipine/pharmacology , Patch-Clamp Techniques , Potassium Channels/analysis , Potassium Channels/metabolism
14.
J Biol Chem ; 275(50): 39193-9, 2000 Dec 15.
Article En | MEDLINE | ID: mdl-10973973

The L-type alpha(1C) (Ca(v)1.2) calcium channel is the major calcium entry pathway in cardiac and smooth muscle. We inactivated the Ca(v)1.2 gene in two independent mouse lines that had indistinguishable phenotypes. Homozygous knockout embryos (Ca(v)1. 2-/-) died before day 14.5 postcoitum (p.c.). At day 12.5 p.c., the embryonic heart contracted with identical frequency in wild type (+/+), heterozygous (+/-), and homozygous (-/-) Ca(v)1.2 embryos. Beating of isolated embryonic cardiomyocytes depended on extracellular calcium and was blocked by 1 microm nisoldipine. In (+/+), (+/-), and (-/-) cardiomyocytes, an L-type Ba(2+) inward current (I(Ba)) was present that was stimulated by Bay K 8644 in all genotypes. At a holding potential of -80 mV, nisoldipine blocked I(Ba) of day 12.5 p.c. (+/+) and (+/-) cells with two IC(50) values of approximately 0.1 and approximately 1 microm. Inhibition of I(Ba) of (-/-) cardiomyocytes was monophasic with an IC(50) of approximately 1 microm. The low affinity I(Ba) was also present in cardiomyocytes of homozygous alpha(1D) (Ca(v)1.3) knockout embryos at day 12.5 p.c. These results indicate that, up to day 14 p.c., contraction of murine embryonic hearts requires an unidentified, low affinity L-type like calcium channel.


Calcium Channels, L-Type/genetics , Calcium Channels, L-Type/physiology , Heart/embryology , Myocardium/cytology , 3-Pyridinecarboxylic acid, 1,4-dihydro-2,6-dimethyl-5-nitro-4-(2-(trifluoromethyl)phenyl)-, Methyl ester/pharmacology , Animals , Barium/metabolism , Calcium Channel Agonists/pharmacology , Calcium Channel Blockers/pharmacology , Cell Line , DNA, Complementary/metabolism , Dose-Response Relationship, Drug , Electrophysiology , Exons , Genetic Vectors , Genotype , Homozygote , Inhibitory Concentration 50 , Ions , Kinetics , Mice , Mice, Knockout , Models, Genetic , Nisoldipine/pharmacology , Phenotype , RNA/metabolism , Time Factors
15.
Cell ; 102(1): 89-97, 2000 Jul 07.
Article En | MEDLINE | ID: mdl-10929716

Voltage-gated L-type Ca2+ channels (LTCCs) containing a pore-forming alpha1D subunit (D-LTCCs) are expressed in neurons and neuroendocrine cells. Their relative contribution to total L-type Ca2+ currents and their physiological role and significance as a drug target remain unknown. Therefore, we generated D-LTCC deficient mice (alpha1D-/-) that were viable with no major disturbances of glucose metabolism. alpha1D-/-mice were deaf due to the complete absence of L-type currents in cochlear inner hair cells and degeneration of outer and inner hair cells. In wild-type controls, D-LTCC-mediated currents showed low activation thresholds and slow inactivation kinetics. Electrocardiogram recordings revealed sinoatrial node dysfunction (bradycardia and arrhythmia) in alpha1D-/- mice. We conclude that alpha1D can form LTCCs with negative activation thresholds essential for normal auditory function and control of cardiac pacemaker activity.


Calcium Channels, L-Type/physiology , Deafness/congenital , Sinoatrial Node/physiopathology , Acoustic Stimulation , Animals , Calcium Channels, L-Type/genetics , Deafness/etiology , Electroencephalography , Electrophysiology , Hair Cells, Auditory, Inner/physiology , Hair Cells, Auditory, Outer/physiology , Heart/physiopathology , Heart Atria/metabolism , Heart Atria/physiopathology , Mice , Mice, Knockout , Rabbits
16.
Naunyn Schmiedebergs Arch Pharmacol ; 361(6): 578-83, 2000 Jun.
Article En | MEDLINE | ID: mdl-10882031

Briefly after withdrawal of the (T-type) calcium channel blocker mibefradil from the market, four cases of life-threatening interaction of mibefradil with dihydropyridines were reported. We investigated in vitro whether mibefradil interacts with a dihydropyridine, as described for other non-dihydropyridine compounds. Rat working hearts were used to examine functional interactions between amlodipine and mibefradil. Gallopamil and another T-type-channel blocker, ethosuximide, were included for comparison. Effects of mibefradil, (+)- and (-)-gallopamil on [3H](+)-isradipine binding were studied in membranes from tsA201-cells transfected with alpha(1c)-, alpha(2)delta-, and beta(1a)- or beta(2a)-calcium channel subunits. Mibefradil increased negative inotropic effect of amlodipine, but not of gallopamil. Gallopamil and ethosuximide showed no influence on contractile effects of amlodipine. Furthermore, mibefradil concentration-dependently caused bradycardic rhythm disturbance. The same type of arrhythmia was observed combining low concentrations of mibefradil with amlodipine, or with gallopamil, respectively. Amlodipine alone, or the combination of gallopamil or ethosuximide with amlodipine did not cause any arrhythmia. Binding studies showed a concentration-dependent positive allosteric interaction between [3H](+)-isradipine and mibefradil, but not with [3H](+)-isradipine and gallopamil enantiomers. Molecular and functional evidence points to an interaction between a dihydropyridine and mibefradil. Mibefradil caused rhythm disturbances and potentiation of negative inotropy when combined with amlodipine.


Arrhythmias, Cardiac/chemically induced , Calcium Channel Blockers/pharmacology , Calcium Channels, L-Type/drug effects , Dihydropyridines/pharmacology , Mibefradil/pharmacology , Myocardial Contraction/drug effects , Ventricular Pressure/drug effects , Amlodipine/pharmacology , Animals , Calcium Channel Blockers/metabolism , Calcium Channels, L-Type/genetics , Cell Line , Cell Membrane/metabolism , Dihydropyridines/metabolism , Drug Interactions , Ethosuximide/pharmacology , Female , Gallopamil/pharmacology , Humans , In Vitro Techniques , Isradipine/metabolism , Male , Mibefradil/metabolism , Perfusion , Radioligand Assay , Rats , Rats, Wistar
17.
Br J Pharmacol ; 130(3): 669-77, 2000 Jun.
Article En | MEDLINE | ID: mdl-10821797

Mibefradil is a novel Ca(2+) antagonist which blocks both high-voltage activated and low voltage-activated Ca(2+) channels. Although L-type Ca(2+) channel block was demonstrated in functional experiments its molecular interaction with the channel has not yet been studied. We therefore investigated the binding of [(3)H]-mibefradil and a series of mibefradil analogues to L-type Ca(2+) channels in different tissues. [(3)H]-Mibefradil labelled a single class of high affinity sites on skeletal muscle L-type Ca(2+) channels (K(D) of 2.5+/-0.4 nM, B(max)=56.4+/-2.3 pmol mg(-1) of protein). Mibefradil (and a series of analogues) partially inhibited (+)-[(3)H]-isradipine binding to skeletal muscle membranes but stimulated binding to brain L-type Ca(2+) channels and alpha1C-subunits expressed in tsA201 cells indicating a tissue-specific, non-competitive interaction between the dihydropyridine and mibefradil binding domain. [(3)H]-Mibefradil also labelled a heterogenous population of high affinity sites in rabbit brain which was inhibited by a series of nonspecific Ca(2+) and Na(+)-channel blockers. Mibefradil and its analogue RO40-6040 had high affinity for neuronal voltage-gated Na(+)-channels as confirmed in binding (apparent K(i) values of 17 and 1.0 nM, respectively) and functional experiments (40% use-dependent inhibition of Na(+)-channel current by 1 microM mibefradil in GH3 cells). Our data demonstrate that mibefradil binds to voltage-gated L-type Ca(2+) channels with very high affinity and is also a potent blocker of voltage-gated neuronal Na(+)-channels. More lipophilic mibefradil analogues may possess neuroprotective properties like other nonselective Ca(2+)-/Na(+)-channel blockers.


Calcium Channels, L-Type/drug effects , Ion Channel Gating/drug effects , Mibefradil/pharmacology , Sodium Channel Blockers , Animals , Calcium Channel Blockers/pharmacology , Electric Stimulation , Electrophysiology , Guinea Pigs , In Vitro Techniques , Muscle, Skeletal/drug effects , Muscle, Skeletal/metabolism , Patch-Clamp Techniques , Rabbits , Radioligand Assay
18.
Biochem J ; 347 Pt 3: 829-36, 2000 May 01.
Article En | MEDLINE | ID: mdl-10769189

Sensitivity to 1,4-dihydropyridines (DHPs) can be transferred from L-type (alpha1C) to non-L-type (alpha1A) Ca(2+) channel alpha1 subunits by the mutation of nine pore-associated non-conserved amino acid residues, yielding mutant alpha1A(DHP). To determine whether the hallmarks of reversible DHP binding to L-type Ca(2+) channels (nanomolar dissociation constants, stereoselectivity and modulation by other chemical classes of Ca(2+) antagonist drugs) were maintained in alpha1A(DHP), we analysed the pharmacological properties of (+)-[(3)H]isradipine-labelled alpha1A(DHP) Ca(2+) channels after heterologous expression. Binding of (+)-isradipine (K(i) 7.4 nM) and the non-benzoxadiazole DHPs nifedipine (K(i) 86 nM), (+/-)-nitrendipine (K(i) 33 nM) and (+/-)-nimodipine (K(i) 67 nM) to alpha1A(DHP) occurred at low nanomolar K(i) values. DHP binding was highly stereoselective [25-fold higher affinity for (+)-isradipine]. As with native channels it was stimulated by (+)-cis-diltiazem, (+)-tetrandrine and mibefradil. This suggested that the three-dimensional architecture of the channel pore was maintained within the non-L-type alpha1A subunit. To predict the three-dimensional arrangement of the DHP-binding residues we exploited the X-ray structure of a recently crystallized bacterial K(+) channel (KcsA) as a template. Our model is based on the assumption that the Ca(2+) channel S5 and S6 segments closely resemble the KcsA transmembrane folding architecture. In the absence of three-dimensional structural data for the alpha1 subunit this is currently the most reasonable approach for modelling this drug-interaction domain. Our model predicts that the previously identified DHP-binding residues form a binding pocket large enough to co-ordinate a single DHP molecule. It also implies that the four homologous Ca(2+) channel repeats are arranged in a clockwise manner.


Amino Acid Substitution/genetics , Bacterial Proteins , Calcium Channel Blockers/metabolism , Calcium Channels/chemistry , Calcium Channels/metabolism , Dihydropyridines/metabolism , Protein Folding , Amino Acid Sequence , Animals , Binding Sites , Calcium Channels/genetics , Calcium Channels, L-Type/chemistry , Calcium Channels, L-Type/genetics , Calcium Channels, L-Type/metabolism , Cell Line , Cell Membrane/metabolism , Humans , Models, Molecular , Molecular Sequence Data , Potassium Channels/chemistry , Protein Binding , Protein Structure, Tertiary , Rabbits , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Sequence Alignment , Stereoisomerism , Substrate Specificity , Thermodynamics
19.
J Biol Chem ; 275(13): 9239-43, 2000 Mar 31.
Article En | MEDLINE | ID: mdl-10734061

Missense mutations in the pore-forming human alpha(1A) subunit of neuronal P/Q-type Ca(2+) channels are associated with familial hemiplegic migraine. We studied the functional consequences on P/Q-type Ca(2+) channel function of three recently identified mutations, R583Q, D715E, and V1457L after introduction into rabbit alpha(1A) and expression in Xenopus laevis oocytes. The potential for half-maximal channel activation of Ba(2+) inward currents was shifted by > 9 mV to more negative potentials in all three mutants. The potential for half-maximal channel inactivation was shifted by > 7 mV in the same direction in R583Q and D715E. Biexponential current inactivation during 3-s test pulses was significantly faster in D715E and slower in V1457L than in wild type. Mutations R583Q and V1457L delayed the time course of recovery from channel inactivation. The decrease of peak current through R583Q (30.2%) and D715E (30. 1%) but not V1457L (18.7%) was more pronounced during 1-Hz trains of 15 100-ms pulses than in wild type (18.2%). Our data demonstrate that the mutations R583Q, D715E, and V1457L, like the previously reported mutations T666M, V714A, and I1819L, affect P/Q-type Ca(2+) channel gating. We therefore propose that altered channel gating represents a common pathophysiological mechanism in familial hemiplegic migraine.


Calcium Channels/metabolism , Migraine Disorders/genetics , Mutation , Animals , DNA, Complementary , Functional Laterality , Humans , Kinetics , Migraine Disorders/physiopathology , Xenopus laevis
20.
FEBS Lett ; 467(1): 65-9, 2000 Feb 04.
Article En | MEDLINE | ID: mdl-10664458

Various beta subunit isoforms stabilize different gating properties of voltage-gated L-type Ca(2+) channels. We therefore investigated the expression of Ca(2+) channel beta subunit isoforms in different smooth muscle types on the protein level by immunoblotting and immunoprecipitation employing beta subunit-selective sequence-directed antibodies. From the four known beta subunit isoforms only beta2 and beta3 were detected in porcine uterus, bovine trachea and bovine aorta membranes. Multiple immunoreactive beta2 bands were detected in a tissue-selective manner indicating structural heterogeneity of beta2. Immunoprecipitation of (+)-[(3)H]isradipine-prelabeled channels revealed that beta2 and beta3 participate in Ca(2+) channel formation in uterus and trachea, and beta3 in aortic smooth muscle. We conclude that beta2 and beta3 subunits form L-type Ca(2+) channels in smooth muscle tissues. This subunit heterogeneity may be important to fine-tune channel function.


Calcium Channels, L-Type/analysis , Calcium Channels, L-Type/chemistry , Muscle, Smooth/chemistry , Animals , Antibodies/immunology , Aorta , Blotting, Western , Calcium Channel Blockers/metabolism , Calcium Channels, L-Type/immunology , Calcium Channels, L-Type/metabolism , Cattle , Cell Membrane/metabolism , Cerebral Cortex/chemistry , Female , Isradipine/metabolism , Molecular Weight , Muscle, Smooth/immunology , Muscle, Smooth, Vascular/chemistry , Muscle, Smooth, Vascular/immunology , Myocardium/chemistry , Myometrium/chemistry , Myometrium/cytology , Myometrium/immunology , Organ Specificity , Precipitin Tests , Protein Isoforms/analysis , Protein Isoforms/chemistry , Protein Isoforms/immunology , Protein Isoforms/metabolism , Rabbits , Swine , Trachea/chemistry , Trachea/immunology
...