Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 75
Filter
1.
Mol Pharmacol ; 89(1): 187-96, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26490245

ABSTRACT

Elucidating the mechanisms that modulate calcium channels via opioid receptor activation is fundamental to our understanding of both pain perception and how opioids modulate pain. Neuronal voltage-gated N-type calcium channels (Cav2.2) are inhibited by activation of G protein-coupled opioid receptors (ORs). However, inhibition of R-type (Cav2.3) channels by µ- or κ-ORs is poorly defined and has not been reported for δ-ORs. To investigate such interactions, we coexpressed human µ-, δ-, or κ-ORs with human Cav2.3 or Cav2.2 in human embryonic kidney 293 cells and measured depolarization-activated Ba(2+) currents (IBa). Selective agonists of µ-, δ-, and κ-ORs inhibited IBa through Cav2.3 channels by 35%. Cav2.2 channels were inhibited to a similar extent by κ-ORs, but more potently (60%) via µ- and δ-ORs. Antagonists of δ- and κ-ORs potentiated IBa amplitude mediated by Cav2.3 and Cav2.2 channels. Consistent with G protein ßγ (Gßγ) interaction, modulation of Cav2.2 was primarily voltage-dependent and transiently relieved by depolarizing prepulses. In contrast, Cav2.3 modulation was voltage-independent and unaffected by depolarizing prepulses. However, Cav2.3 inhibition was sensitive to pertussis toxin and to intracellular application of guanosine 5'-[ß-thio]diphosphate trilithium salt and guanosine 5'-[γ-thio]triphosphate tetralithium salt. Coexpression of Gßγ-specific scavengers-namely, the carboxyl terminus of the G protein-coupled receptor kinase 2 or membrane-targeted myristoylated-phosducin-attenuated or abolished Cav2.3 modulation. Our study reveals the diversity of OR-mediated signaling at Cav2 channels and identifies neuronal Cav2.3 channels as potential targets for opioid analgesics. Their novel modulation is dependent on pre-existing OR activity and mediated by membrane-delimited Gßγ subunits in a voltage-independent manner.


Subject(s)
Calcium Channel Blockers/pharmacology , Calcium Channels, R-Type/physiology , GTP-Binding Protein beta Subunits/physiology , GTP-Binding Protein gamma Subunits/physiology , Receptors, Opioid, delta/physiology , Receptors, Opioid, kappa/physiology , Receptors, Opioid, mu/physiology , Analgesics, Opioid/pharmacology , Enkephalin, Ala(2)-MePhe(4)-Gly(5)-/pharmacology , HEK293 Cells , Humans , Protein Subunits/physiology , Receptors, Opioid, delta/agonists , Receptors, Opioid, kappa/agonists , Receptors, Opioid, mu/agonists
2.
Rev Physiol Biochem Pharmacol ; 167: 115-39, 2014.
Article in English | MEDLINE | ID: mdl-25280639

ABSTRACT

Voltage-gated Ca(2+) channels (VGCCs) are ubiquitous in excitable cells. These channels play key roles in many physiological events like cardiac regulation/pacemaker activity due to intracellular Ca(2+) transients. In the myocardium, the Cav1 subfamily (L-type: Cav1.2 and Cav1.3) is the main contributor to excitation-contraction coupling and/or pacemaking, whereas the Cav3 subfamily (T-type: Cav3.1 and Cav3.2) is important in rhythmically firing of the cardiac nodal cells. No established cardiac function has been attributed to the Cav2 family (E-/R-type: Cav2.3) despite accumulating evidence of cardiac dysregulation observed upon deletion of the Cav2.3 gene, the only member of this family so far detected in cardiomyocytes. In this review, we summarize the pathophysiological changes observed after ablation of the E-/R-type VGCC and propose a cardiac mechanism of action for this channel. Also, considering the role played by this channel in epilepsy and its reported sensitivity to antiepileptic drugs, a putative involvement of this channel in the cardiac mechanism of sudden unexpected death in epilepsy is also discussed.


Subject(s)
Calcium Channels, L-Type/physiology , Calcium Channels, R-Type/physiology , Calcium Channels, T-Type/physiology , Cation Transport Proteins/physiology , Death, Sudden/etiology , Epilepsy/physiopathology , Heart/physiology , Animals , Calcium Channels, L-Type/chemistry , Calcium Channels, R-Type/chemistry , Calcium Channels, T-Type/chemistry , Cation Transport Proteins/chemistry , Epilepsy/complications , Humans
3.
Graefes Arch Clin Exp Ophthalmol ; 253(10): 1713-9, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26104874

ABSTRACT

BACKGROUND: The model of the isolated and superfused retina provides the opportunity to test drugs and toxins. Some chemicals have to be applied using low concentrations of organic solvents as carriers. Recently, E-/R-type (Cav2.3) and T-type (Cav3.2) voltage-gated Ca(2+) channels were identified as participating in reciprocal inhibitory retinal signaling. Their participation is apparent, when low concentrations of NiCl2 (15 µM) are applied during superfusion leading to an increase of the ERG b-wave amplitude, which is explained by a reduction of amacrine GABA-release onto bipolar neurons. During these investigations, differences were observed for the solvent carrier used. METHODS: Recording of the transretinal receptor potentials from the isolated bovine retina. RESULTS: The pretreatment of bovine retina with 0.01 % (v/v) dimethylsulfoxide did not impair the NiCl2-mediated increase of the b-wave amplitude, which was 1.31-fold ± 0.03 of initial value (n = 4). However, pretreatment of the retina with the same concentration of ethanol impaired reciprocal signaling (0.96-fold ± 0.05, n = 4). Further, the implicit time of the b-wave was increased, suggesting that ethanol itself but not DMSO may antagonize GABA-receptors. CONCLUSION: Ethanol itself but not DMSO may block GABA receptors and cause an amplitude increase by itself, so that reciprocal signaling is impaired.


Subject(s)
Dimethyl Sulfoxide/administration & dosage , Electroretinography/drug effects , Ethanol/administration & dosage , Retina/drug effects , Signal Transduction/physiology , Solvents/administration & dosage , Animals , Calcium Channels, R-Type/physiology , Calcium Channels, T-Type/physiology , Cattle , GABA-A Receptor Antagonists/administration & dosage , Nickel/pharmacology , Photic Stimulation , Retina/metabolism
4.
J Neurosci ; 33(50): 19396-405, 2013 Dec 11.
Article in English | MEDLINE | ID: mdl-24336706

ABSTRACT

Small-conductance calcium-activated potassium (SK) channels play an important role in regulating neuronal excitability. While SK channels at the soma have long been known to contribute to the medium afterhyperpolarization (mAHP), recent evidence indicates they also regulate NMDA receptor activation in dendritic spines. Here we investigate the activation of SK channels in spines and dendrites of rat cortical pyramidal neurons during action potentials (APs), and compare this to SK channel activation at the soma. Using confocal calcium imaging, we demonstrate that the inhibition of SK channels with apamin results in a location-dependent increase in calcium influx into dendrites and spines during backpropagating APs (average increase, ~40%). This effect was occluded by block of R-type voltage-dependent calcium channels (VDCCs), but not by inhibition of N- or P/Q-type VDCCs, or block of calcium release from intracellular stores. During these experiments, we noticed that the calcium indicator (Oregon Green BAPTA-1) blocked the mAHP. Subsequent experiments using low concentrations of EGTA (1 mm) produced the same result, suggesting that somatic SK channels are not tightly colocalized with their calcium source. Consistent with this idea, all known subtypes of VDCCs except R-type were calcium sources for the apamin-sensitive mAHP at the soma. We conclude that SK channels in spines and dendrites of cortical pyramidal neurons regulate calcium influx during backpropagating APs in a distance-dependent manner, and are tightly coupled to R-type VDCCs. In contrast, SK channels activated by APs at the soma of these neurons are weakly coupled to a variety of VDCCs.


Subject(s)
Action Potentials/physiology , Calcium/metabolism , Dendrites/physiology , Pyramidal Cells/physiology , Small-Conductance Calcium-Activated Potassium Channels/physiology , Animals , Calcium Channels, R-Type/physiology , Cerebral Cortex/cytology , Cerebral Cortex/drug effects , Cerebral Cortex/physiology , Dendrites/drug effects , Egtazic Acid/pharmacology , Organic Chemicals/pharmacology , Pyramidal Cells/cytology , Pyramidal Cells/drug effects , Rats
5.
Nat Cell Biol ; 9(4): 453-60, 2007 Apr.
Article in English | MEDLINE | ID: mdl-17369816

ABSTRACT

Pancreatic islets have a central role in blood glucose homeostasis. In addition to insulin-producing beta-cells and glucagon-secreting alpha-cells, the islets contain somatostatin-releasing delta-cells. Somatostatin is a powerful inhibitor of insulin and glucagon secretion. It is normally secreted in response to glucose and there is evidence suggesting its release becomes perturbed in diabetes. Little is known about the control of somatostatin release. Closure of ATP-regulated K(+)-channels (K(ATP)-channels) and a depolarization-evoked increase in cytoplasmic free Ca(2+) concentration ([Ca(2+)](i)) have been proposed to be essential. Here, we report that somatostatin release evoked by high glucose (>or=10 mM) is unaffected by the K(ATP)-channel activator diazoxide and proceeds normally in K(ATP)-channel-deficient islets. Glucose-induced somatostatin secretion is instead primarily dependent on Ca(2+)-induced Ca(2+)-release (CICR). This constitutes a novel mechanism for K(ATP)-channel-independent metabolic control of pancreatic hormone secretion.


Subject(s)
Calcium Channels, R-Type/physiology , Calcium/metabolism , Glucose/pharmacology , Somatostatin/metabolism , Animals , Calcium/pharmacology , Calcium Channels, R-Type/genetics , Cytophotometry , Diazoxide/pharmacology , Dose-Response Relationship, Drug , Electrophysiology , Immunohistochemistry , In Vitro Techniques , Islets of Langerhans/cytology , Islets of Langerhans/drug effects , Islets of Langerhans/metabolism , Isradipine/pharmacology , Macrocyclic Compounds/pharmacology , Mannoheptulose/pharmacology , Membrane Potentials/drug effects , Mice , Mice, Inbred C57BL , Mice, Inbred Strains , Mice, Knockout , Microscopy, Confocal , Oxazoles/pharmacology , Potassium/pharmacology , Potassium Channel Blockers/pharmacology , Potassium Channels/physiology , Ryanodine/pharmacology , Somatostatin-Secreting Cells/drug effects , Somatostatin-Secreting Cells/metabolism
6.
PLoS Biol ; 8(11): e1000534, 2010 Nov 16.
Article in English | MEDLINE | ID: mdl-21103408

ABSTRACT

Activation of group I metabotropic glutamate receptors (subtypes mGluR1 and mGluR5) regulates neural activity in a variety of ways. In CA1 pyramidal neurons, activation of group I mGluRs eliminates the post-burst afterhyperpolarization (AHP) and produces an afterdepolarization (ADP) in its place. Here we show that upregulation of Ca(v)2.3 R-type calcium channels is responsible for a component of the ADP lasting several hundred milliseconds. This medium-duration ADP is rapidly and reversibly induced by activation of mGluR5 and requires activation of phospholipase C (PLC) and release of calcium from internal stores. Effects of mGluR activation on subthreshold membrane potential changes are negligible but are large following action potential firing. Furthermore, the medium ADP exhibits a biphasic activity dependence consisting of short-term facilitation and longer-term inhibition. These findings suggest that mGluRs may dramatically alter the firing of CA1 pyramidal neurons via a complex, activity-dependent modulation of Ca(v)2.3 R-type channels that are activated during spiking at physiologically relevant rates and patterns.


Subject(s)
Action Potentials , Calcium Channels, R-Type/physiology , Cation Transport Proteins/physiology , Neurons/physiology , Pyramidal Cells/physiology , Receptors, Metabotropic Glutamate/physiology , Up-Regulation/physiology , Animals , Female , In Vitro Techniques , Ion Channel Gating , Male , Methoxyhydroxyphenylglycol/analogs & derivatives , Methoxyhydroxyphenylglycol/pharmacology , Mice , Mice, Inbred C57BL , Patch-Clamp Techniques , Rats , Rats, Wistar , Xenopus
7.
Nat Commun ; 14(1): 516, 2023 01 31.
Article in English | MEDLINE | ID: mdl-36720859

ABSTRACT

High-voltage-activated R-type CaV2.3 channel plays pivotal roles in many physiological activities and is implicated in epilepsy, convulsions, and other neurodevelopmental impairments. Here, we determine the high-resolution cryo-electron microscopy (cryo-EM) structure of human CaV2.3 in complex with the α2δ1 and ß1 subunits. The VSDII is stabilized in the resting state. Electrophysiological experiments elucidate that the VSDII is not required for channel activation, whereas the other VSDs are essential for channel opening. The intracellular gate is blocked by the W-helix. A pre-W-helix adjacent to the W-helix can significantly regulate closed-state inactivation (CSI) by modulating the association and dissociation of the W-helix with the gate. Electrostatic interactions formed between the negatively charged domain on S6II, which is exclusively conserved in the CaV2 family, and nearby regions at the alpha-interacting domain (AID) and S4-S5II helix are identified. Further functional analyses indicate that these interactions are critical for the open-state inactivation (OSI) of CaV2 channels.


Subject(s)
Calcium Channels, R-Type , Cation Transport Proteins , Humans , Cryoelectron Microscopy , Calcium Channels, R-Type/physiology , Cation Transport Proteins/physiology
8.
Adv Exp Med Biol ; 740: 1219-47, 2012.
Article in English | MEDLINE | ID: mdl-22453990

ABSTRACT

Voltage- and ligand-gated ion channels are key elements in the etiopathogenesis of various forms of epilepsy. In this chapter, we present an overview of the functional implications of voltage-gated Ca(2+) channels in modulating internal Ca(2+) level fluctuations and generating ictiform/epileptiform cellular electrophysiological activity. A specific focus will be on the fascinating and evolving field of high-voltage activated (HVA) Non-L-type Ca(v)2.3 R-type channels and low-voltage activated (LVA) Ca(v)3.1-3.3 T-type Ca(2+) channels in the genesis of plateau potentials and excessive rebound bursting. Plateau potentials have been characterised in the hippocampus and were shown to be triggered by Ca(v)2.3 which subsequently activate CNG channels that mediate long-lasting plateaus. In the thalamocortical network, a complex ion channel armamentarium is involved in regulating a complex balance of burst and tonic mode activity. Recent findings point to an outstanding role of R- and T-type channels in both thalamocortical eurhythmia and pathophysiological -aberrations. Thus, pharmacological modulation of voltage-gated Ca(2+)-channels might prove more and more important in treatment of neurological and psychiatric disorder such as schizophrenia, mania, dementia and epilepsy.


Subject(s)
Calcium Channels/physiology , Calcium/metabolism , Epilepsy/etiology , Animals , Calcium Channels, R-Type/physiology , Calcium Channels, T-Type/physiology , Cation Transport Proteins/physiology , Epilepsy/drug therapy , Epilepsy/metabolism , Humans
9.
Neuron ; 53(2): 249-60, 2007 Jan 18.
Article in English | MEDLINE | ID: mdl-17224406

ABSTRACT

The roles of voltage-sensitive sodium (Na) and calcium (Ca) channels located on dendrites and spines in regulating synaptic signals are largely unknown. Here we use 2-photon glutamate uncaging to stimulate individual spines while monitoring uncaging-evoked excitatory postsynaptic potentials (uEPSPs) and Ca transients. We find that, in CA1 pyramidal neurons in acute mouse hippocampal slices, CaV(2.3) voltage-sensitive Ca channels (VSCCs) are found selectively on spines and act locally to dampen uncaging-evoked Ca transients and somatic potentials. These effects are mediated by a regulatory loop that requires opening of CaV(2.3) channels, voltage-gated Na channels, small conductance Ca-activated potassium (SK) channels, and NMDA receptors. Ca influx through CaV(2.3) VSCCs selectively activates SK channels, revealing the presence of functional Ca microdomains within the spine. Our results suggest that synaptic strength can be modulated by mechanisms that regulate voltage-gated conductances within the spine but do not alter the properties or numbers of synaptic glutamate receptors.


Subject(s)
Calcium Channels, R-Type/physiology , Cation Transport Proteins/physiology , Dendritic Spines/metabolism , Hippocampus/physiology , Pyramidal Cells/physiology , Synapses/physiology , Animals , Calcium/metabolism , Calcium Channels, R-Type/chemistry , Cation Transport Proteins/chemistry , Electrophysiology , Excitatory Postsynaptic Potentials , Hippocampus/metabolism , In Vitro Techniques , Ion Channel Gating , Mice , Mice, Inbred C57BL , Protein Structure, Tertiary , Pyramidal Cells/metabolism , Receptors, N-Methyl-D-Aspartate/metabolism , Small-Conductance Calcium-Activated Potassium Channels/metabolism , Sodium Channels/metabolism , Temperature
10.
J Biol Chem ; 285(46): 35878-88, 2010 Nov 12.
Article in English | MEDLINE | ID: mdl-20826800

ABSTRACT

The ß subunit of high voltage-activated Ca(2+) (Ca(v)) channels targets the pore-forming α(1) subunit to the plasma membrane and tunes the biophysical phenotype of the Ca(v) channel complex. We used a combination of molecular biology and whole-cell patch clamp to investigate the functional role of a long N-terminal polyacidic motif (NPAM) in a Ca(v)ß subunit of the human parasite Schistosoma mansoni (ß(Sm)), a motif that does not occur in other known Ca(v)ß subunits. When expressed in human embryonic kidney cells stably expressing Ca(v)2.3, ß(Sm) accelerates Ca(2+)/calmodulin-independent inactivation of Ca(v)2.3. Deleting the first 44 amino acids of ß(Sm), a region that includes NPAM, significantly slows the predominant time constant of inactivation (τ(fast)) under conditions that prevent Ca(2+)/CaM-dependent inactivation (ß(Sm): τ(fast) = 66 ms; ß(SmΔ2-44): τ(fast) = 111 ms, p < 0.01). Interestingly, deleting the amino acids that are N-terminal to NPAM (2-24 or 2-17) results in faster inactivation than with an intact N terminus (τ(fast) = 42 ms with ß(SmΔ2-17); τ(fast) = 40 ms with ß(SmΔ2-24), p < 0.01). This suggests that NPAM is the structural determinant for accelerating Ca(2+)/calmodulin-independent inactivation. We also created three chimeric subunits that contain the first 44 amino acids of ß(Sm) attached to mammalian ß(1b), ß(2a), and ß(3) subunits. For any given mammalian ß subunit, inactivation was faster if it contained the N terminus of ß(Sm) than if it did not. Co-expression of the mammalian α(2)δ-1 subunit resulted in doubling of the inactivation rate, but the effects of NPAM persisted. Thus, it appears that the schistosome Ca(v) channel complex has acquired a new function that likely contributes to reducing the amount of Ca(2+) that enters the cells in vivo. This feature is of potential interest as a target for new antihelminthics.


Subject(s)
Calcium Channels/physiology , Helminth Proteins/physiology , Ion Channel Gating/physiology , Schistosoma mansoni/metabolism , Amino Acid Sequence , Animals , Blotting, Western , Calcium/metabolism , Calcium Channels/chemistry , Calcium Channels/genetics , Calcium Channels, R-Type/genetics , Calcium Channels, R-Type/metabolism , Calcium Channels, R-Type/physiology , Calmodulin/metabolism , Cation Transport Proteins/genetics , Cation Transport Proteins/metabolism , Cation Transport Proteins/physiology , Chelating Agents/pharmacology , Egtazic Acid/pharmacology , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , HEK293 Cells , Helminth Proteins/genetics , Helminth Proteins/metabolism , Humans , Ion Channel Gating/genetics , Kinetics , Membrane Potentials/drug effects , Microscopy, Confocal , Models, Biological , Molecular Sequence Data , Mutation , Patch-Clamp Techniques , Protein Subunits/genetics , Protein Subunits/physiology , Schistosoma mansoni/genetics , Transfection
11.
J Neurosci ; 29(5): 1434-45, 2009 Feb 04.
Article in English | MEDLINE | ID: mdl-19193890

ABSTRACT

Because inhibitory synaptic transmission is a major mechanism of general anesthesia, we examined the effects of isoflurane on properties of GABAergic inhibitory currents in the reticular thalamic nucleus (nRT) in brain slices. The evoked IPSCs (eIPSCs) and spontaneous miniature synaptic currents (mIPSCs) of visualized nRT cells in young and adult rats were recorded. Consistent with postsynaptic effects on GABA(A) receptors, isoflurane prolonged the decay-time constants of both eIPSCs and mIPCSs. Surprisingly, isoflurane completely inhibited the amplitude of eIPSCs at clinically relevant concentrations (IC(50) of 240+/-20 microm), increased the paired-pulse ratio, and decreased the frequency of mIPSCs, indicating that presynaptic mechanisms may also contribute to the effects of isoflurane on IPSCs. The overall effect of isoflurane on eIPSCs in nRT cells was a decrease of net charge-transfer across the postsynaptic membrane. The application of 100 microm nickel (Ni(2+)) and the more specific R-type Ca(2+) channel blocker SNX-482 (0.5 microm) decreased eIPSC amplitudes, increased the paired-pulse ratio, and attenuated isoflurane-induced inhibition of eIPSCs. In addition, isoflurane potently blocked currents in recombinant human Ca(V)2.3 (alpha1E) channels with an IC(50) of 206 +/- 22 mum. Importantly, in vivo electroencephalographic (EEG) recordings in adult Ca(V)2.3 knock-out mice demonstrated alterations in isoflurane-induced burst-suppression activity. Because the thalamus has a key function in processing sensory information, sleep, and cognition, modulation of its GABAergic tone by presynaptic R-type Ca(2+) channels may contribute to the clinical effects of general anesthesia.


Subject(s)
Calcium Channels, R-Type/physiology , Inhibitory Postsynaptic Potentials/physiology , Isoflurane/pharmacology , Presynaptic Terminals/physiology , Thalamus/physiology , Animals , Cell Line , Dose-Response Relationship, Drug , Humans , In Vitro Techniques , Inhibitory Postsynaptic Potentials/drug effects , Mice , Mice, Inbred C57BL , Mice, Knockout , Presynaptic Terminals/drug effects , Rats , Rats, Sprague-Dawley , Synaptic Transmission/drug effects , Synaptic Transmission/physiology , Thalamus/drug effects
12.
J Physiol ; 588(Pt 8): 1281-92, 2010 Apr 15.
Article in English | MEDLINE | ID: mdl-20194128

ABSTRACT

Synaptic activity in the medial prefrontal cortex (mPFC) is fundamental for higher cognitive functions such as working memory. The present study shows that small conductance (SK) calcium-activated potassium channels attenuate excitatory synaptic transmission at layer 2/3 and layer 5 inputs to layer 5 pyramidal neurons in the mPFC. SK channels are located postsynaptically at synapses where they are activated during synaptic transmission by calcium influx through NMDA receptors, L-type calcium channels, R-type calcium channels and by calcium release from IP(3)-sensitive stores. Removal of the SK channel-mediated shunt of synaptic transmission reveals significant NMDA receptor-mediated activation during basal synaptic transmission, which is greater at layer 5 inputs (approximately 30%) than at layer 2/3 inputs (approximately 20%). These findings show that interactions between NMDA receptors, SK channels and voltage-gated calcium channels play a critical role in regulating excitatory synaptic transmission in layer 5 pyramidal neurons in the mPFC.


Subject(s)
Potassium Channels, Voltage-Gated/physiology , Prefrontal Cortex/physiology , Receptors, N-Methyl-D-Aspartate/physiology , Small-Conductance Calcium-Activated Potassium Channels/physiology , Synapses/physiology , Animals , Calcium/metabolism , Calcium Channels, L-Type/physiology , Calcium Channels, R-Type/physiology , Cell Communication , Female , Male , Models, Animal , Pyramidal Cells/cytology , Pyramidal Cells/physiology , Rats , Rats, Wistar , Synaptic Transmission/physiology
13.
J Pharmacol Exp Ther ; 334(2): 545-55, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20439438

ABSTRACT

Voltage-gated calcium channel (Ca(v))2.2 (N-type calcium channels) are key components in nociceptive transmission pathways. Ziconotide, a state-independent peptide inhibitor of Ca(v)2.2 channels, is efficacious in treating refractory pain but exhibits a narrow therapeutic window and must be administered intrathecally. We have discovered an N-triazole oxindole, (3R)-5-(3-chloro-4-fluorophenyl)-3-methyl-3-(pyrimidin-5-ylmethyl)-1-(1H-1,2,4-triazol-3-yl)-1,3-dihydro-2H-indol-2-one (TROX-1), as a small-molecule, state-dependent blocker of Ca(v)2 channels, and we investigated the therapeutic advantages of this compound for analgesia. TROX-1 preferentially inhibited potassium-triggered calcium influx through recombinant Ca(v)2.2 channels under depolarized conditions (IC(50) = 0.27 microM) compared with hyperpolarized conditions (IC(50) > 20 microM). In rat dorsal root ganglion (DRG) neurons, TROX-1 inhibited omega-conotoxin GVIA-sensitive calcium currents (Ca(v)2.2 channel currents), with greater potency under depolarized conditions (IC(50) = 0.4 microM) than under hyperpolarized conditions (IC(50) = 2.6 microM), indicating state-dependent Ca(v)2.2 channel block of native as well as recombinant channels. TROX-1 fully blocked calcium influx mediated by a mixture of Ca(v)2 channels in calcium imaging experiments in rat DRG neurons, indicating additional block of all Ca(v)2 family channels. TROX-1 reversed inflammatory-induced hyperalgesia with maximal effects equivalent to nonsteroidal anti-inflammatory drugs, and it reversed nerve injury-induced allodynia to the same extent as pregabalin and duloxetine. In contrast, no significant reversal of hyperalgesia was observed in Ca(v)2.2 gene-deleted mice. Mild impairment of motor function in the Rotarod test and cardiovascular functions were observed at 20- to 40-fold higher plasma concentrations than required for analgesic activities. TROX-1 demonstrates that an orally available state-dependent Ca(v)2 channel blocker may achieve a therapeutic window suitable for the treatment of chronic pain.


Subject(s)
Analgesics/pharmacology , Calcium Channel Blockers/pharmacology , Calcium Channels, N-Type/physiology , Indoles/pharmacology , Triazoles/pharmacology , Analgesics/adverse effects , Analgesics/pharmacokinetics , Animals , Baroreflex/drug effects , Biological Availability , Calcium Channel Blockers/adverse effects , Calcium Channel Blockers/pharmacokinetics , Calcium Channels, N-Type/genetics , Calcium Channels, R-Type/physiology , Cation Transport Proteins/physiology , Cell Line , Dogs , Ganglia, Spinal/drug effects , Ganglia, Spinal/physiology , Hyperalgesia/drug therapy , Hypotension, Orthostatic/chemically induced , Indoles/adverse effects , Indoles/pharmacokinetics , Male , Mice , Mice, Knockout , Neurons/drug effects , Neurons/physiology , Pain/drug therapy , Pain/etiology , Patch-Clamp Techniques , Rats , Rats, Sprague-Dawley , Triazoles/adverse effects , Triazoles/pharmacokinetics
14.
Sci Rep ; 10(1): 15993, 2020 10 02.
Article in English | MEDLINE | ID: mdl-33009476

ABSTRACT

The mating of 77 heterozygous pairs (Cav3.2[+|-] x Cav3.2[+|-]) revealed a significant deviation of genotype distribution from Mendelian inheritance in weaned pups. The mating of 14 pairs (Cav3.2[-|-] female x Cav3.2[+|-] male) and 8 pairs (Cav3.2[+|-] female x Cav3.2[-|-] male) confirmed the significant reduction of deficient homozygous Cav3.2[-|-] pups, leading to the conclusion that prenatal lethality may occur, when one or both alleles, encoding the Cav3.2T-type Ca2+ channel, are missing. Also, the mating of 63 heterozygous pairs (Cav2.3[+|-] x Cav2.3[+|-]) revealed a significant deviation of genotype distribution from Mendelian inheritance in weaned pups, but only for heterozygous male mice, leading to the conclusion that compensation may only occur for Cav2.3[-|-] male mice lacking both alleles of the R-type Ca2+ channel. During the mating of heterozygous parents, the number of female mice within the weaned population does not deviate from the expected Mendelian inheritance. During prenatal development, both, T- and R-type Ca2+ currents are higher expressed in some tissues than postnatally. It will be discussed that the function of voltage-gated Ca2+ channels during prenatal development must be investigated in more detail, not least to understand devastative diseases like developmental epileptic encephalopathies (DEE).


Subject(s)
Calcium Channels, R-Type/physiology , Calcium Channels, T-Type/physiology , Cation Transport Proteins/physiology , Chromosomes/genetics , Genomic Instability , Inbreeding/methods , Quantitative Trait Loci , Animals , Female , Male , Mice , Mice, Inbred C57BL , Mice, Knockout
15.
Neuron ; 39(3): 483-96, 2003 Jul 31.
Article in English | MEDLINE | ID: mdl-12895422

ABSTRACT

Ca2+ influx into presynaptic terminals via voltage-dependent Ca2+ channels triggers fast neurotransmitter release as well as different forms of synaptic plasticity. Using electrophysiological and genetic techniques we demonstrate that presynaptic Ca2+ entry through Cav2.3 subunits contributes to the induction of mossy fiber LTP and posttetanic potentiation by brief trains of presynaptic action potentials while they do not play a role in fast synaptic transmission, paired-pulse facilitation, or frequency facilitation. This functional specialization is most likely achieved by a localization remote from the release machinery and by a Cav2.3 channel-dependent facilitation of presynaptic Ca2+ influx. Thus, the presence of Cav2.3 channels boosts the accumulation of presynaptic Ca2+ triggering presynaptic LTP and posttetanic potentiation without affecting the low release probability that is a prerequisite for the enormous plasticity displayed by mossy fiber synapses.


Subject(s)
Calcium Channels, R-Type/physiology , Presynaptic Terminals/physiology , Animals , Calcium/physiology , Calcium Channels/genetics , Calcium Channels/physiology , Calcium Channels, R-Type/genetics , Excitatory Postsynaptic Potentials/physiology , Hippocampus/physiology , Long-Term Potentiation/physiology , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Neuronal Plasticity/physiology , Nickel/physiology
16.
J Neurosci ; 27(49): 13420-9, 2007 Dec 05.
Article in English | MEDLINE | ID: mdl-18057200

ABSTRACT

Voltage-gated Ca2+ channels in presynaptic terminals initiate the Ca2+ inflow necessary for transmitter release. At a variety of synapses, multiple Ca2+ channel subtypes are involved in synaptic transmission and plasticity. However, it is unknown whether presynaptic Ca2+ channels differ in gating properties and whether they are differentially activated by action potentials or subthreshold voltage signals. We examined Ca2+ channels in hippocampal mossy fiber boutons (MFBs) by presynaptic recording, using the selective blockers omega-agatoxin IVa, omega-conotoxin GVIa, and SNX-482 to separate P/Q-, N-, and R-type components. Nonstationary fluctuation analysis combined with blocker application revealed a single MFB contained on average approximately 2000 channels, with 66% P/Q-, 26% N-, and 8% R-type channels. Whereas both P/Q-type and N-type Ca2+ channels showed high activation threshold and rapid activation and deactivation, R-type Ca2+ channels had a lower activation threshold and slower gating kinetics. To determine the efficacy of activation of different Ca2+ channel subtypes by physiologically relevant voltage waveforms, a six-state gating model reproducing the experimental observations was developed. Action potentials activated P/Q-type Ca2+ channels with high efficacy, whereas N- and R-type channels were activated less efficiently. Action potential broadening selectively recruited N- and R-type channels, leading to an equalization of the efficacy of channel activation. In contrast, subthreshold presynaptic events activated R-type channels more efficiently than P/Q- or N-type channels. In conclusion, single MFBs coexpress multiple types of Ca2+ channels, which are activated differentially by subthreshold and suprathreshold presynaptic voltage signals.


Subject(s)
Calcium Channels, P-Type/physiology , Calcium Channels, R-Type/physiology , Ion Channel Gating/physiology , Mossy Fibers, Hippocampal/physiology , Recruitment, Neurophysiological/physiology , Animals , Calcium Channels/physiology , Calcium Channels, N-Type/physiology , Electric Stimulation/methods , Presynaptic Terminals/physiology , Rats , Rats, Wistar , Synaptic Transmission/physiology
17.
J Physiol ; 586(22): 5503-20, 2008 Nov 15.
Article in English | MEDLINE | ID: mdl-18832426

ABSTRACT

Recent studies have suggested that transmitter release facilitation at synapses is largely mediated by presynaptic Ca(2+) current facilitation, but the exact contribution of Ca(2+) current facilitation has not been determined quantitatively. Here, we determine the contribution of Ca(2+) current facilitation, and of an increase in the residual free Ca(2+) concentration ([Ca(2+)](i)) in the nerve terminal, to paired-pulse facilitation of transmitter release at the calyx of Held. Under conditions of low release probability imposed by brief presynaptic voltage-clamp steps, transmitter release facilitation at short interstimulus intervals (4 ms) was 227 +/- 31% of control, Ca(2+) current facilitation was 113 +/- 4% of control, and the peak residual [Ca(2+)](i) was 252 +/- 18 nm over baseline. By inferring the 'local' [Ca(2+)](i) transients that drive transmitter release during these voltage-clamp stimuli with the help of a kinetic release model, we estimate that Ca(2+) current facilitation contributes to approximately 40% to paired-pulse facilitation of transmitter release. The remaining component of facilitation strongly depends on the build-up, and on the decay of the residual free [Ca(2+)](i), but cannot be explained by linear summation of the residual free [Ca(2+)](i), and the back-calculated 'local' [Ca(2+)](i) signal, which only accounts for approximately 10% of the total release facilitation. Further voltage-clamp experiments designed to compensate for Ca(2+) current facilitation demonstrated that about half of the observed transmitter release facilitation remains in the absence of Ca(2+) current facilitation. Our results indicate that paired-pulse facilitation of transmitter release at the calyx of Held is driven by at least two distinct mechanisms: Ca(2+) current facilitation, and a mechanism independent of Ca(2+) current facilitation that closely tracks the time course of residual free [Ca(2+)](i).


Subject(s)
Auditory Pathways/physiology , Calcium Signaling , Neurotransmitter Agents/metabolism , Action Potentials , Animals , Brain Stem/metabolism , Brain Stem/physiology , Calcium Channels, N-Type/physiology , Calcium Channels, R-Type/physiology , Electrophysiology , In Vitro Techniques , Models, Neurological , Patch-Clamp Techniques , Rats , Rats, Wistar , Synapses/metabolism
18.
Acta Neurochir Suppl ; 104: 99-102, 2008.
Article in English | MEDLINE | ID: mdl-18456998

ABSTRACT

Voltage-dependent potassium (Kv) and calcium (VDCC) channels play an important role in the regulation of membrane potential and intracellular calcium concentration in cerebral artery myocytes. Recent evidence suggests VDCC activity is increased and Kv channel activity is decreased in cerebral arteries following subarachnoid hemorrhage (SAH), promoting enhanced constriction. We have examined the impact of the blood component oxyhemoglobin on Kv and VDCC function in small (100-200 microm) diameter cerebral arteries. Acute (10 min) exposure of oxyhemoglobin caused cerebral artery constriction and Kv current suppression that was abolished by tyrosine kinase inhibitors and a Kv channel blocker. Although short-term oxyhemoglobin application did not directly alter VDCC activity, five-day exposure to oxyhemoglobin was associated with enhanced expression of voltage-dependent calcium channels. This work suggests that acute and chronic effects of oxyhemoglobin act synergistically to promote membrane depolarization and increased VDCC activity in cerebral arteries. These actions of oxyhemoglobin may contribute to the development of cerebral vasospasm following aneurysmal subarachnoid hemorrhage.


Subject(s)
Cerebral Arteries/physiology , Ion Channels/physiology , Oxyhemoglobins/pharmacology , Animals , Calcium Channels, R-Type/drug effects , Calcium Channels, R-Type/physiology , Cerebral Arteries/drug effects , Ion Channels/drug effects , Models, Animal , Organ Culture Techniques , Rabbits , Vasoconstriction/drug effects
19.
Nat Neurosci ; 6(9): 948-55, 2003 Sep.
Article in English | MEDLINE | ID: mdl-12937422

ABSTRACT

Voltage-sensitive Ca2+ channels (VSCCs) constitute a major source of calcium ions in dendritic spines, but their function is unknown. Here we show that R-type VSCCs in spines of rat CA1 pyramidal neurons are depressed for at least 30 min after brief trains of back-propagating action potentials. Populations of channels in single spines are depressed stochastically and synchronously, independent of channels in the parent dendrite and other spines, implying that depression is the result of signaling restricted to individual spines. Induction of VSCC depression blocks theta-burst-induced long-term potentiation (LTP), indicating that postsynaptic action potentials can modulate synaptic plasticity by tuning VSCCs. Induction of depression requires [Ca2+] elevations and activation of L-type VSCCs, which activate Ca2+/calmodulin-dependent kinase II (CaMKII) and a cyclic adenosine monophosphate (cAMP)-dependent pathway. Given that L-type VSCCs do not contribute measurably to Ca2+ influx in spines, they must activate downstream effectors either directly through voltage-dependent conformational changes or via [Ca2+] microdomains.


Subject(s)
Action Potentials/physiology , Calcium Channels, R-Type/physiology , Dendrites/physiology , Neuronal Plasticity/physiology , Action Potentials/drug effects , Animals , Calcium/pharmacology , Calcium/physiology , Dendrites/drug effects , Hippocampus/cytology , Hippocampus/drug effects , Hippocampus/physiology , In Vitro Techniques , Neuronal Plasticity/drug effects , Rats
20.
J Neurosci ; 26(23): 6249-58, 2006 Jun 07.
Article in English | MEDLINE | ID: mdl-16763032

ABSTRACT

The "toxin-resistant" R-type Ca2+ channels are expressed widely in the CNS and distributed mainly in apical dendrites and spines. They play important roles in regulating signal transduction and intrinsic properties of neurons, but the modulation of these channels in the mammalian CNS has not been studied. In this study we used whole-cell patch-clamp recordings and found that muscarinic activation enhances R-type, but does not affect T-type, Ca2+ currents in hippocampal CA1 pyramidal neurons after N, P/Q, and L-type Ca2+ currents selectively were blocked. M1/M3 cholinergic receptors mediated the muscarinic stimulation of R-type Ca2+ channels. The signaling pathway underlying the R-type enhancement was independent of intracellular [Ca2+] changes and required the activation of a Ca(2+)-independent PKC pathway. Furthermore, we found that the enhancement of R-type Ca2+ currents resulted in the de novo appearance of Ca2+ spikes and in remarkable changes in the firing pattern of R-type Ca2+ spikes, which could fire repetitively in the theta frequency. Therefore, muscarinic enhancement of R-type Ca2+ channels could play an important role in modifying the dendritic response to synaptic inputs and in the intrinsic resonance properties of neurons.


Subject(s)
Calcium Channels, R-Type/physiology , Hippocampus/metabolism , Muscarine/metabolism , Pyramidal Cells/metabolism , Animals , Calcium/metabolism , Calcium Channels, R-Type/drug effects , Calcium Channels, T-Type/drug effects , Calcium Channels, T-Type/physiology , Carbachol/pharmacology , Cholinergic Agents/pharmacology , Electric Conductivity , Electrophysiology , Hippocampus/cytology , In Vitro Techniques , Patch-Clamp Techniques , Protein Kinase C/metabolism , Rats , Rats, Sprague-Dawley , Receptor, Muscarinic M1/physiology , Receptor, Muscarinic M3/physiology , Signal Transduction/physiology , Theta Rhythm
SELECTION OF CITATIONS
SEARCH DETAIL