1
|
Li D, Zhang X, Liu R, Long M, Zhou S, Lin J, Zhang L. Kainic acid induced hyperexcitability in thalamic reticular nucleus that initiates an inflammatory response through the HMGB1/TLR4 pathway. Neurotoxicology 2023; 95:94-106. [PMID: 36669621 DOI: 10.1016/j.neuro.2023.01.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2022] [Revised: 01/11/2023] [Accepted: 01/12/2023] [Indexed: 01/20/2023]
Abstract
OBJECTIVE To explore the relationship between the proinflammatory factor high-mobility group box 1 (HMGB1) and glutamatergic alpha-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA) receptors in the development of epilepsy. METHODS Thalamic reticular nucleus (TRN) slices were treated with kainic acid (KA) to simulate seizures. Action potentials and spontaneous inhibitory postsynaptic currents (sIPSCs) were recorded within TRN slices using whole-cell patch clamp techniques. The translocation of HMGB1 was detected by immunofluorescence. The HMGB1/TLR4 signaling pathway and its downstream inflammatory factors (IL-1β and NF-κB) were detected by RTPCR, Western blot and ELISA. RESULTS KA-evoked spikings were observed in TRN slices and blocked by perampanel. sIPSCs in the TRN were enhanced by KA and reduced by perampanel. The translocation of HMGB1 in the TRN was promoted by KA and inhibited by perampanel. The expression of the HMGB1/TLR4 signaling pathway was promoted by KA and suppressed by perampanel. CONCLUSION KA induced hyperexcitability activates the HMGB1/TLR4 pathway, which potentially leading to neuroinflammation in epilepsy.
Collapse
Affiliation(s)
- Dongbin Li
- Department of Neurology, The First Affiliated Hospital of Harbin Medical University, Harbin, China; First Department of Neurology and Neuroscience Center, Heilongjiang Provincial Hospital, Harbin, China
| | - Xiaosi Zhang
- Metro-Medic Clinic, 1538 sherbrooke Ouest, suite 100, Montreal, QC H3G 1L5, Canada
| | - Ruoshi Liu
- Department of Neurology, The Fourth Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Meixin Long
- Department of Neurology, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Shanshan Zhou
- Department of Neurology, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Jinghan Lin
- Department of Neurology, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Liming Zhang
- Department of Neurology, The First Affiliated Hospital of Harbin Medical University, Harbin, China.
| |
Collapse
|
2
|
Evaluation of potential anticonvulsant fluorinated N-benzamide enaminones as T-type Ca 2+ channel blockers. Bioorg Med Chem 2022; 65:116766. [PMID: 35537326 DOI: 10.1016/j.bmc.2022.116766] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2021] [Revised: 04/06/2022] [Accepted: 04/19/2022] [Indexed: 11/21/2022]
Abstract
Trifluoromethylated N-benzamide enaminones have been identified as potential anticonvulsants for the treatment of drug-resistant epilepsy. T-type Ca2+ channels are an important target for anti-seizure medications. Our laboratory has developed several fluorinated N-benzamide enaminone analogs that were evaluated by their ability to target T-type Ca2+ channels. Using whole cell voltage-clamp recordings, we identified two meta-trifluoromethyl N-benzamide enaminones with a significant inhibitory effect on T-type Ca2+ channels. These compounds had no effect on voltage-activated Na+ channels. We also evaluated the effect of the fluorinated N-benzamide enaminone analogs on the T-type Ca2+ channel subunits Cav3.2 and Cav3.3. The meta-trifluoromethyl N-benzamide enaminone lead analogs altered the steady-state inactivation of Cav3.2 T-type Ca2+ channels, which resulted in a significant increase in the inactivation recovery time of the channels. There was no effect of fluorinated N-benzamide enaminone analogs on the gating mechanism of T-type Ca2+ channels, as proven by the lack of effect on the activation and inactivation time constant of Ca2+ currents. On the contrary, the meta-trifluoromethyl N-benzamide enaminone lead analogs altered the gating mechanism of Cav3.3 T-type Ca2+ channels, as proven by the reduction in the activation and inactivation time constant of the channels. There was no effect on the inactivation kinetics of Cav3.3 T-type Ca2+ channels. The present results demonstrate that meta-substituted trifluoromethyl N-benzamide enaminone analogs target T-type Ca2+ channels by different mechanisms depending on the channel subunit. Meta-trifluoromethyl N-benzamide enaminone analogs can potentially lead to the design of more specific blockers of T-type Ca2+ channels for the treatment of epileptic seizures.
Collapse
|
3
|
Coulter I, Timic Stamenic T, Eggan P, Fine BR, Corrigan T, Covey DF, Yang L, Pan JQ, Todorovic SM. Different roles of T-type calcium channel isoforms in hypnosis induced by an endogenous neurosteroid epipregnanolone. Neuropharmacology 2021; 197:108739. [PMID: 34339750 PMCID: PMC8478885 DOI: 10.1016/j.neuropharm.2021.108739] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2021] [Revised: 07/09/2021] [Accepted: 07/29/2021] [Indexed: 12/31/2022]
Abstract
BACKGROUND Many neuroactive steroids induce sedation/hypnosis by potentiating γ-aminobutyric acid (GABAA) currents. However, we previously demonstrated that an endogenous neuroactive steroid epipregnanolone [(3β,5β)-3-hydroxypregnan-20-one] (EpiP) exerts potent peripheral analgesia and blocks T-type calcium currents while sparing GABAA currents in rat sensory neurons. This study seeks to investigate the behavioral effects elicited by systemic administration of EpiP and to characterize its use as an adjuvant agent to commonly used general anesthetics (GAs). METHODS Here, we utilized electroencephalographic (EEG) recordings to characterize thalamocortical oscillations, as well as behavioral assessment and mouse genetics with wild-type (WT) and different knockout (KO) models of T-channel isoforms to investigate potential sedative/hypnotic and immobilizing properties of EpiP. RESULTS Consistent with increased oscillations in slower EEG frequencies, EpiP induced an hypnotic state in WT mice when injected alone intra-peritoneally (i.p.) and effectively facilitated anesthetic effects of isoflurane (ISO) and sevoflurane (SEVO). The CaV3.1 (Cacna1g) KO mice demonstrated decreased sensitivity to EpiP-induced hypnosis when compared to WT mice, whereas no significant difference was noted between CaV3.2 (Cacna1h), CaV3.3 (Cacna1i) and WT mice. Finally, when compared to WT mice, onset of EpiP-induced hypnosis was delayed in CaV3.2 KO mice but not in CaV3.1 and CaV3.3 KO mice. CONCLUSION We posit that EpiP may have an important role as novel hypnotic and/or adjuvant to volatile anesthetic agents. We speculate that distinct hypnotic effects of EpiP across all three T-channel isoforms is due to their differential expression in thalamocortical circuitry.
Collapse
Affiliation(s)
- Ian Coulter
- Department of Anesthesiology, University of Colorado,
Anschutz Medical Campus, Aurora 80045
| | - Tamara Timic Stamenic
- Department of Anesthesiology, University of Colorado,
Anschutz Medical Campus, Aurora 80045
| | - Pierce Eggan
- Department of Anesthesiology, University of Colorado,
Anschutz Medical Campus, Aurora 80045
| | - Brier R. Fine
- Department of Anesthesiology, University of Colorado,
Anschutz Medical Campus, Aurora 80045
| | - Timothy Corrigan
- Department of Pediatrics, Division of Neurology,
Translational Epilepsy Research Program, University of Colorado, Anschutz Medical
Campus, Aurora, CO 80045, USA
| | - Douglas F. Covey
- Department of Developmental Biology, Washington University
School of Medicine, St. Louis, MO 63110, USA;,Taylor Family Institute for Innovative Psychiatric
Research, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Lingling Yang
- Stanley Center for Psychiatric Research, Broad Institute of
Harvard and MIT
| | - Jen Q. Pan
- Stanley Center for Psychiatric Research, Broad Institute of
Harvard and MIT
| | - Slobodan M. Todorovic
- Department of Anesthesiology, University of Colorado,
Anschutz Medical Campus, Aurora 80045;,Neuroscience, University of Colorado, Anschutz Medical
Campus, Aurora 80045;,Pharmacology Graduate Programs, University of Colorado,
Anschutz Medical Campus, Aurora 80045
| |
Collapse
|
4
|
Global genetic deletion of Ca V3.3 channels facilitates anaesthetic induction and enhances isoflurane-sparing effects of T-type calcium channel blockers. Sci Rep 2020; 10:21510. [PMID: 33299036 PMCID: PMC7725806 DOI: 10.1038/s41598-020-78488-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2020] [Accepted: 11/13/2020] [Indexed: 01/02/2023] Open
Abstract
We previously documented that the CaV3.3 isoform of T-type calcium channels (T-channels) is inhibited by clinically relevant concentrations of volatile anaesthetics, including isoflurane. However, little is understood about the functional role of CaV3.3 channels in anaesthetic-induced hypnosis and underlying neuronal oscillations. To address this issue, we used CaV3.3 knock-out (KO) mice and a panselective T-channel blocker 3,5-dichloro-N-[1-(2,2-dimethyltetrahydro-pyran-4-ylmethyl)-4-fluoro-piperidin-4-ylmethyl]-benzamide (TTA-P2). We found that mutant mice injected with the vehicle showed faster induction of hypnosis than wild-type (WT) mice, while the percent isoflurane at which hypnosis and immobility occurred was not different between two genotypes. Furthermore, we found that TTA-P2 facilitated isoflurane induction of hypnosis in the CaV3.3 KO mice more robustly than in the WT mice. Isoflurane-induced hypnosis following injections of TTA-P2 was accompanied with more prominent delta and theta EEG oscillations in the mutant mice, and reached burst-suppression pattern earlier when compared to the WT mice. Our findings point to a relatively specific value of CaV3.3 channels in anaesthetic induced hypnosis. Furthermore, we propose that T-channel blockers may be further explored as a valuable adjunct to reducing the usage of potent volatile anaesthetics, thereby improving their safety.
Collapse
|
5
|
Hao X, Ou M, Zhang D, Zhao W, Yang Y, Liu J, Yang H, Zhu T, Li Y, Zhou C. The Effects of General Anesthetics on Synaptic Transmission. Curr Neuropharmacol 2020; 18:936-965. [PMID: 32106800 PMCID: PMC7709148 DOI: 10.2174/1570159x18666200227125854] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2019] [Revised: 01/20/2020] [Accepted: 02/26/2020] [Indexed: 02/08/2023] Open
Abstract
General anesthetics are a class of drugs that target the central nervous system and are widely used for various medical procedures. General anesthetics produce many behavioral changes required for clinical intervention, including amnesia, hypnosis, analgesia, and immobility; while they may also induce side effects like respiration and cardiovascular depressions. Understanding the mechanism of general anesthesia is essential for the development of selective general anesthetics which can preserve wanted pharmacological actions and exclude the side effects and underlying neural toxicities. However, the exact mechanism of how general anesthetics work is still elusive. Various molecular targets have been identified as specific targets for general anesthetics. Among these molecular targets, ion channels are the most principal category, including ligand-gated ionotropic receptors like γ-aminobutyric acid, glutamate and acetylcholine receptors, voltage-gated ion channels like voltage-gated sodium channel, calcium channel and potassium channels, and some second massager coupled channels. For neural functions of the central nervous system, synaptic transmission is the main procedure for which information is transmitted between neurons through brain regions, and intact synaptic function is fundamentally important for almost all the nervous functions, including consciousness, memory, and cognition. Therefore, it is important to understand the effects of general anesthetics on synaptic transmission via modulations of specific ion channels and relevant molecular targets, which can lead to the development of safer general anesthetics with selective actions. The present review will summarize the effects of various general anesthetics on synaptic transmissions and plasticity.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Yu Li
- Address correspondence to these authors at the Laboratory of Anesthesia & Critical Care Medicine, Translational Neuroscience Center, West China Hospital of Sichuan University, Chengdu 610041, Sichuan, P.R. China; E-mail: and Department of Anesthesiology, West China Hospital of Sichuan University, Chengdu, 610041, Sichuan, P.R. China; E-mail:
| | - Cheng Zhou
- Address correspondence to these authors at the Laboratory of Anesthesia & Critical Care Medicine, Translational Neuroscience Center, West China Hospital of Sichuan University, Chengdu 610041, Sichuan, P.R. China; E-mail: and Department of Anesthesiology, West China Hospital of Sichuan University, Chengdu, 610041, Sichuan, P.R. China; E-mail:
| |
Collapse
|
6
|
Miao QL, Herlitze S, Mark MD, Noebels JL. Adult loss of Cacna1a in mice recapitulates childhood absence epilepsy by distinct thalamic bursting mechanisms. Brain 2020; 143:161-174. [PMID: 31800012 PMCID: PMC6935748 DOI: 10.1093/brain/awz365] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2019] [Revised: 09/09/2019] [Accepted: 09/29/2019] [Indexed: 12/15/2022] Open
Abstract
Inborn errors of CACNA1A-encoded P/Q-type calcium channels impair synaptic transmission, producing early and lifelong neurological deficits, including childhood absence epilepsy, ataxia and dystonia. Whether these impairments owe their pathologies to defective channel function during the critical period for thalamic network stabilization in immature brain remains unclear. Here we show that mice with tamoxifen-induced adult-onset ablation of P/Q channel alpha subunit (iKOp/q) display identical patterns of dysfunction, replicating the inborn loss-of-function phenotypes and, therefore demonstrate that these neurological defects do not rely upon developmental abnormality. Unexpectedly, unlike the inborn model, the adult-onset pattern of excitability changes believed to be pathogenic within the thalamic network is non-canonical. Specifically, adult ablation of P/Q channels does not promote Cacna1g-mediated burst firing or T-type calcium current (IT) in the thalamocortical relay neurons; however, burst firing in thalamocortical relay neurons remains essential as iKOp/q mice generated on a Cacna1g deleted background show substantially diminished seizure generation. Moreover, in thalamic reticular nucleus neurons, burst firing is impaired accompanied by attenuated IT. Interestingly, inborn deletion of thalamic reticular nucleus-enriched, human childhood absence epilepsy-linked gene Cacna1h in iKOp/q mice reduces thalamic reticular nucleus burst firing and promotes rather than reduces seizure, indicating an epileptogenic role for loss-of-function Cacna1h gene variants reported in human childhood absence epilepsy cases. Together, our results demonstrate that P/Q channels remain critical for maintaining normal thalamocortical oscillations and motor control in the adult brain, and suggest that the developmental plasticity of membrane currents regulating pathological rhythmicity is both degenerate and age-dependent.
Collapse
Affiliation(s)
- Qing-Long Miao
- Developmental Neurogenetics Laboratory, Department of Neurology, Baylor College of Medicine, Houston TX, USA
| | - Stefan Herlitze
- Department of Zoology and Neurobiology, Ruhr University of Bochum, Bochum, Germany
| | - Melanie D Mark
- Department of Zoology and Neurobiology, Ruhr University of Bochum, Bochum, Germany
| | - Jeffrey L Noebels
- Developmental Neurogenetics Laboratory, Department of Neurology, Baylor College of Medicine, Houston TX, USA
- Department of Neuroscience, Baylor College of Medicine, Houston TX, USA
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston TX, USA
| |
Collapse
|
7
|
Dumenieu M, Senkov O, Mironov A, Bourinet E, Kreutz MR, Dityatev A, Heine M, Bikbaev A, Lopez-Rojas J. The Low-Threshold Calcium Channel Cav3.2 Mediates Burst Firing of Mature Dentate Granule Cells. Cereb Cortex 2019; 28:2594-2609. [PMID: 29790938 PMCID: PMC5998957 DOI: 10.1093/cercor/bhy084] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2018] [Indexed: 12/11/2022] Open
Abstract
Mature granule cells are poorly excitable neurons that were recently shown to fire action potentials, preferentially in bursts. It is believed that the particularly pronounced short-term facilitation of mossy fiber synapses makes granule cell bursting a very effective means of properly transferring information to CA3. However, the mechanism underlying the unique bursting behavior of mature granule cells is currently unknown. Here, we show that Cav3.2 T-type channels at the axon initial segment are responsible for burst firing of mature granule cells in rats and mice. Accordingly, Cav3.2 knockout mice fire tonic spikes and exhibit impaired bursting, synaptic plasticity and dentate-to-CA3 communication. The data show that Cav3.2 channels are strong modulators of bursting and can be considered a critical molecular switch that enables effective information transfer from mature granule cells to the CA3 pyramids.
Collapse
Affiliation(s)
- Mael Dumenieu
- Research Group Neuroplasticity, Leibniz Institute for Neurobiology, Brenneckestr. 6, Magdeburg, Germany
| | - Oleg Senkov
- Molecular Neuroplasticity Group, German Center for Neurodegenerative Diseases (DZNE), Magdeburg, Germany
| | - Andrey Mironov
- Molecular Neuroplasticity Group, German Center for Neurodegenerative Diseases (DZNE), Magdeburg, Germany.,Lobachevsky State University of Nizhny Novgorod, Nizhny Novgorod, Russia.,Privolzhsky Research Medical University, Nizhny Novgorod, Russia
| | - Emmanuel Bourinet
- Calcium Channel Dynamics & Nociception Group, Institute of Functional Genomics, Montpellier, France
| | - Michael R Kreutz
- Research Group Neuroplasticity, Leibniz Institute for Neurobiology, Brenneckestr. 6, Magdeburg, Germany.,Leibniz Group "Dendritic Organelles and Synaptic Function," University Medical Center Hamburg-Eppendorf, Center for Molecular Neurobiology (ZMNH), Hamburg, Germany
| | - Alexander Dityatev
- Molecular Neuroplasticity Group, German Center for Neurodegenerative Diseases (DZNE), Magdeburg, Germany.,Center for Behavioral Brain Sciences (CBBS), Magdeburg, Germany.,Medical Faculty, Otto-von-Guericke University, Magdeburg, Germany
| | - Martin Heine
- Research Group Molecular Physiology, Leibniz Institute for Neurobiology, Brenneckestr. 6, Magdeburg, Germany
| | - Arthur Bikbaev
- Research Group Molecular Physiology, Leibniz Institute for Neurobiology, Brenneckestr. 6, Magdeburg, Germany
| | - Jeffrey Lopez-Rojas
- Research Group Neuroplasticity, Leibniz Institute for Neurobiology, Brenneckestr. 6, Magdeburg, Germany
| |
Collapse
|
8
|
Cytosolic ATP Relieves Voltage-Dependent Inactivation of T-Type Calcium Channels and Facilitates Excitability of Neurons in the Rat Central Medial Thalamus. eNeuro 2018; 5:eN-NWR-0016-18. [PMID: 29468189 PMCID: PMC5819668 DOI: 10.1523/eneuro.0016-18.2018] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2018] [Accepted: 02/01/2018] [Indexed: 12/21/2022] Open
Abstract
The central medial nucleus (CeM) is a part of the intralaminar thalamus, which is involved in the control of arousal and sensory processing. However, ionic conductances and mechanisms that regulate the activity of the CeM are not well studied. Here, we used in vitro electrophysiology in acute brain slices from adolescent rats to demonstrate that T-type calcium currents (T-currents) are prominent in the majority of the studied CeM neurons and are critical determinants of low-threshold calcium spikes (LTSs), which in turn regulate excitability of these neurons. Using an ATP-free internal solution decreased T-current density and induced a profound hyperpolarizing shift in steady-state inactivation curves while voltage-dependent activation kinetics were spared. Furthermore, selective pharmacological blockade of T-channels or use of an ATP-free solution reduced both tonic action potential (AP) frequency and rebound burst firing in CeM neurons. Our results indicate that T-channels are critical regulators of a thalamocortical circuit output and suggest that cytosolic ATP could be an endogenous regulatory mechanism in which T-channels may functionally gate sensory transmission and arousal in vivo.
Collapse
|
9
|
Cain SM, Garcia E, Waheed Z, Jones KL, Bushell TJ, Snutch TP. GABA B receptors suppress burst-firing in reticular thalamic neurons. Channels (Austin) 2017; 11:574-586. [PMID: 28742985 DOI: 10.1080/19336950.2017.1358836] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022] Open
Abstract
Burst-firing in thalamic neurons is known to play a key role in mediating thalamocortical (TC) oscillations that are associated with non-REM sleep and some types of epileptic seizure. Within the TC system the primary output of GABAergic neurons in the reticular thalamic nucleus (RTN) is thought to induce the de-inactivation of T-type calcium channels in thalamic relay (TR) neurons, promoting burst-firing drive to the cortex and the propagation of TC network activity. However, RTN neurons also project back onto other neurons within the RTN. The role of this putative negative feedback upon the RTN itself is less well understood, although is hypothesized to induce de-synchronization of RTN neuron firing leading to the suppression of TC oscillations. Here we tested two hypotheses concerning possible mechanisms underlying TC oscillation modulation. Firstly, we assessed the burst-firing behavior of RTN neurons in response to GABAB receptor activation using acute brain slices. The selective GABAB receptor agonist baclofen was found to induce suppression of burst-firing concurrent with effects on membrane input resistance. Secondly, RTN neurons express CaV3.2 and CaV3.3 T-type calcium channel isoforms known to contribute toward TC burst-firing and we examined the modulation of these channels by GABAB receptor activation. Utilizing exogenously expressed T-type channels we assessed whether GABAB receptor activation could directly alter T-type calcium channel properties. Overall, GABAB receptor activation had only modest effects on CaV3.2 and CaV3.3 isoforms. The only effect that could be predicted to suppress burst-firing was a hyperpolarized shift in the voltage-dependence of inactivation, potentially causing lower channel availability at membrane potentials critical for burst-firing. Conversely, other effects observed such as a hyperpolarized shift in the voltage-dependence of activation of both CaV3.2 and CaV3.3 as well as increased time constant of activation of the CaV3.3 isoform would be expected to enhance burst-firing. Together, we hypothesize that GABAB receptor activation mediates multiple downstream effectors that combined act to suppress burst-firing within the RTN. It appears unlikely that direct GABAB receptor-mediated modulation of T-type calcium channels is the major mechanistic contributor to this suppression.
Collapse
Affiliation(s)
- Stuart M Cain
- a Michael Smith Laboratories and Djavad Mowafaghian Centre for Brain Health , University of British Columbia , Vancouver , Canada
| | - Esperanza Garcia
- a Michael Smith Laboratories and Djavad Mowafaghian Centre for Brain Health , University of British Columbia , Vancouver , Canada
| | - Zeina Waheed
- a Michael Smith Laboratories and Djavad Mowafaghian Centre for Brain Health , University of British Columbia , Vancouver , Canada
| | - Karen L Jones
- a Michael Smith Laboratories and Djavad Mowafaghian Centre for Brain Health , University of British Columbia , Vancouver , Canada
| | - Trevor J Bushell
- b Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde , Glasgow , UK
| | - Terrance P Snutch
- a Michael Smith Laboratories and Djavad Mowafaghian Centre for Brain Health , University of British Columbia , Vancouver , Canada
| |
Collapse
|
10
|
Coulon P, Landisman CE. The Potential Role of Gap Junctional Plasticity in the Regulation of State. Neuron 2017; 93:1275-1295. [PMID: 28334604 DOI: 10.1016/j.neuron.2017.02.041] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2016] [Revised: 01/20/2017] [Accepted: 02/22/2017] [Indexed: 11/19/2022]
Abstract
Electrical synapses are the functional correlate of gap junctions and allow transmission of small molecules and electrical current between coupled neurons. Instead of static pores, electrical synapses are actually plastic, similar to chemical synapses. In the thalamocortical system, gap junctions couple inhibitory neurons that are similar in their biochemical profile, morphology, and electrophysiological properties. We postulate that electrical synaptic plasticity among inhibitory neurons directly interacts with the switching between different firing patterns in a state-dependent and type-dependent manner. In neuronal networks, electrical synapses may function as a modifiable resonance feedback system that enables stable oscillations. Furthermore, the plasticity of electrical synapses may play an important role in regulation of state, synchrony, and rhythmogenesis in the mammalian thalamocortical system, similar to chemical synaptic plasticity. Based on their plasticity, rich diversity, and specificity, electrical synapses are thus likely to participate in the control of consciousness and attention.
Collapse
Affiliation(s)
- Philippe Coulon
- Seattle Children's Research Institute, Center for Integrative Brain Research, Seattle, WA 98101, USA.
| | - Carole E Landisman
- Seattle Children's Research Institute, Center for Integrative Brain Research, Seattle, WA 98101, USA.
| |
Collapse
|
11
|
Abstract
The role of T-type calcium currents is rarely considered in the extensive literature covering the mechanisms of long-term synaptic plasticity. This situation reflects the lack of suitable T-type channel antagonists that till recently has hampered investigations of the functional roles of these channels. However, with the development of new pharmacological and genetic tools, a clear involvement of T-type channels in synaptic plasticity is starting to emerge. Here, we review a number of studies showing that T-type channels participate to numerous homo- and hetero-synaptic plasticity mechanisms that involve different molecular partners and both pre- and post-synaptic modifications. The existence of T-channel dependent and independent plasticity at the same synapse strongly suggests a subcellular localization of these channels and their partners that allows specific interactions. Moreover, we illustrate the functional importance of T-channel dependent synaptic plasticity in neocortex and thalamus.
Collapse
Affiliation(s)
- Nathalie Leresche
- a Sorbonne Universités, Université Pierre et Marie Curie (UPMC) UM119, CNRS UMR8246, INSERM U1130, Neuroscience Paris Seine (NPS) , Paris , France
| | - Régis C Lambert
- a Sorbonne Universités, Université Pierre et Marie Curie (UPMC) UM119, CNRS UMR8246, INSERM U1130, Neuroscience Paris Seine (NPS) , Paris , France
| |
Collapse
|
12
|
Pellegrini C, Lecci S, Lüthi A, Astori S. Suppression of Sleep Spindle Rhythmogenesis in Mice with Deletion of CaV3.2 and CaV3.3 T-type Ca(2+) Channels. Sleep 2016; 39:875-85. [PMID: 26612388 DOI: 10.5665/sleep.5646] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2015] [Accepted: 10/28/2015] [Indexed: 11/03/2022] Open
Abstract
STUDY OBJECTIVES Low-threshold voltage-gated T-type Ca(2+) channels (T-channels or CaV3 channels) sustain oscillatory discharges of thalamocortical (TC) and nucleus Reticularis thalami (nRt) cells. The CaV3.3 subtype dominates nRt rhythmic bursting and mediates a substantial fraction of spindle power in the NREM sleep EEG. CaV3.2 channels are also found in nRt, but whether these contribute to nRt-dependent spindle generation is unexplored. We investigated thalamic rhythmogenesis in mice lacking this subtype in isolation (CaV3.2KO mice) or in concomitance with CaV3.3 deletion (CaV3.double-knockout (DKO) mice). METHODS We examined discharge characteristics of thalamic cells and intrathalamic evoked synaptic transmission in brain slices from wild-type, CaV3.2KO and CaV3.DKO mice through patch-clamp recordings. The sleep profile of freely behaving CaV3.2KO and CaV3.DKO mice was assessed by polysomnographic recordings. RESULTS CaV3.2 channel deficiency left nRt discharge properties largely unaltered, but additional deletion of CaV3.3 channels fully abolished low-threshold whole-cell Ca(2+) currents and bursting, and suppressed burst-mediated inhibitory responses in TC cells. CaV3.DKO mice had more fragmented sleep, with shorter NREM sleep episodes and more frequent microarousals. The NREM sleep EEG power spectrum displayed a relative suppression of the σ frequency band (10-15 Hz), which was accompanied by an increase in the δ band (1-4 Hz). CONCLUSIONS Consistent with previous findings, CaV3.3 channels dominate nRt rhythmogenesis, but the lack of CaV3.2 channels further aggravates neuronal, synaptic, and EEG deficits. Therefore, CaV3.2 channels can boost intrathalamic synaptic transmission, and might play a modulatory role adjusting the relative presence of NREM sleep EEG rhythms.
Collapse
Affiliation(s)
- Chiara Pellegrini
- Department of Fundamental Neurosciences, University of Lausanne, Lausanne, Switzerland
| | - Sandro Lecci
- Department of Fundamental Neurosciences, University of Lausanne, Lausanne, Switzerland
| | - Anita Lüthi
- Department of Fundamental Neurosciences, University of Lausanne, Lausanne, Switzerland
| | - Simone Astori
- Department of Fundamental Neurosciences, University of Lausanne, Lausanne, Switzerland
| |
Collapse
|
13
|
Todorovic SM, Jevtovic-Todorovic V. Redox regulation of neuronal voltage-gated calcium channels. Antioxid Redox Signal 2014; 21:880-91. [PMID: 24161125 PMCID: PMC4116091 DOI: 10.1089/ars.2013.5610] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
SIGNIFICANCE Voltage-gated calcium channels are ubiquitously expressed in neurons and are key regulators of cellular excitability and synaptic transmitter release. There is accumulating evidence that multiple subtypes of voltage-gated calcium channels may be regulated by oxidation and reduction. However, the redox mechanisms involved in the regulation of channel function are not well understood. RECENT ADVANCES Several studies have established that both T-type and high-voltage-activated subtypes of voltage-gated calcium channel can be redox-regulated. This article reviews different mechanisms that can be involved in redox regulation of calcium channel function and their implication in neuronal function, particularly in pain pathways and thalamic oscillation. CRITICAL ISSUES A current critical issue in the field is to decipher precise mechanisms of calcium channel modulation via redox reactions. In this review we discuss covalent post-translational modification via oxidation of cysteine molecules and chelation of trace metals, and reactions involving nitric oxide-related molecules and free radicals. Improved understanding of the roles of redox-based reactions in regulation of voltage-gated calcium channels may lead to improved understanding of novel redox mechanisms in physiological and pathological processes. FUTURE DIRECTIONS Identification of redox mechanisms and sites on voltage-gated calcium channel may allow development of novel and specific ion channel therapies for unmet medical needs. Thus, it may be possible to regulate the redox state of these channels in treatment of pathological process such as epilepsy and neuropathic pain.
Collapse
Affiliation(s)
- Slobodan M Todorovic
- 1 Department of Anesthesiology, University of Virginia School of Medicine , Charlottesville, Virginia
| | | |
Collapse
|
14
|
Thalamocortical neurons display suppressed burst-firing due to an enhanced Ih current in a genetic model of absence epilepsy. Pflugers Arch 2014; 467:1367-82. [PMID: 24953239 PMCID: PMC4435665 DOI: 10.1007/s00424-014-1549-4] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2014] [Revised: 05/28/2014] [Accepted: 05/30/2014] [Indexed: 10/25/2022]
Abstract
Burst-firing in distinct subsets of thalamic relay (TR) neurons is thought to be a key requirement for the propagation of absence seizures. However, in the well-regarded Genetic Absence Epilepsy Rats from Strasbourg (GAERS) model as yet there has been no link described between burst-firing in TR neurons and spike-and-wave discharges (SWDs). GAERS ventrobasal (VB) neurons are a specific subset of TR neurons that do not normally display burst-firing during absence seizures in the GAERS model, and here, we assessed the underlying relationship of VB burst-firing with Ih and T-type calcium currents between GAERS and non-epileptic control (NEC) animals. In response to 200-ms hyperpolarizing current injections, adult epileptic but not pre-epileptic GAERS VB neurons displayed suppressed burst-firing compared to NEC. In response to longer duration 1,000-ms hyperpolarizing current injections, both pre-epileptic and epileptic GAERS VB neurons required significantly more hyperpolarizing current injection to burst-fire than those of NEC animals. The current density of the Hyperpolarization and Cyclic Nucleotide-activated (HCN) current (Ih) was found to be increased in GAERS VB neurons, and the blockade of Ih relieved the suppressed burst-firing in both pre-epileptic P15-P20 and adult animals. In support, levels of HCN-1 and HCN-3 isoform channel proteins were increased in GAERS VB thalamic tissue. T-type calcium channel whole-cell currents were found to be decreased in P7-P9 GAERS VB neurons, and also noted was a decrease in CaV3.1 mRNA and protein levels in adults. Z944, a potent T-type calcium channel blocker with anti-epileptic properties, completely abolished hyperpolarization-induced VB burst-firing in both NEC and GAERS VB neurons.
Collapse
|
15
|
T-type Ca2+ channels in absence epilepsy. Pflugers Arch 2014; 466:719-34. [DOI: 10.1007/s00424-014-1461-y] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2014] [Accepted: 01/22/2014] [Indexed: 11/25/2022]
|
16
|
Abstract
Low-voltage-activated T-type Ca(2+) channels are widely expressed in various types of neurons. Once deinactivated by hyperpolarization, T-type channels are ready to be activated by a small depolarization near the resting membrane potential and, therefore, are optimal for regulating the excitability and electroresponsiveness of neurons under physiological conditions near resting states. Ca(2+) influx through T-type channels engenders low-threshold Ca(2+) spikes, which in turn trigger a burst of action potentials. Low-threshold burst firing has been implicated in the synchronization of the thalamocortical circuit during sleep and in absence seizures. It also has been suggested that T-type channels play an important role in pain signal transmission, based on their abundant expression in pain-processing pathways in peripheral and central neurons. In this review, we will describe studies on the role of T-type Ca(2+) channels in the physiological as well as pathological generation of brain rhythms in sleep, absence epilepsy, and pain signal transmission. Recent advances in studies of T-type channels in the control of cognition will also be briefly discussed.
Collapse
Affiliation(s)
- Eunji Cheong
- Department of Biotechnology, Translational Research Center for Protein Function Control, College of Life Science and Biotechnology, Yonsei University, Seoul, Republic of Korea.
| | | |
Collapse
|
17
|
Lambert RC, Bessaïh T, Crunelli V, Leresche N. The many faces of T-type calcium channels. Pflugers Arch 2013; 466:415-23. [PMID: 24043572 DOI: 10.1007/s00424-013-1353-6] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2013] [Revised: 09/06/2013] [Accepted: 09/07/2013] [Indexed: 01/27/2023]
Abstract
Since the discovery of low-voltage-activated T-type calcium channels in sensory neurons and the initial characterization of their physiological function mainly in inferior olive and thalamic neurons, studies on neuronal T-type currents have predominantly focused on the generation of low-threshold spike (and associated action potential burst firing) which is strictly conditioned by a preceding hyperpolarization. This T-type current mediated activity has become an archetype of the function of these channels, constraining our view of the potential physiological and pathological roles that they may play in controlling the excitability of single cells and neural networks. However, greatly helped by the recent availability of the first potent and selective antagonists for this class of calcium channels, novel T-type current functions are rapidly being uncovered, including their surprising involvement in neuronal excitability at depolarized membrane potentials and their complex control of dendritic integration and neurotransmitter release. These and other data summarized in this short review clearly indicate a much wider physiological involvement of T-type channels in neuronal activity than previously expected.
Collapse
|
18
|
Shan HQ, Hammarback JA, Godwin DW. Ethanol inhibition of a T-type Ca²+ channel through activity of protein kinase C. Alcohol Clin Exp Res 2013; 37:1333-42. [PMID: 23488970 DOI: 10.1111/acer.12098] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2012] [Accepted: 01/02/2013] [Indexed: 11/28/2022]
Abstract
BACKGROUND T-type calcium channels (T-channels) are widely distributed in the central and peripheral nervous system, where they mediate calcium entry and regulate the intrinsic excitability of neurons. T-channels are dysregulated in response to alcohol administration and withdrawal. We therefore investigated acute ethanol (EtOH) effects and the underlying mechanism of action in human embryonic kidney (HEK) 293 cell lines, as well as effects on native currents recorded from dorsal root ganglion (DRG) neurons cultured from Long-Evans rats. METHODS Whole-cell voltage-clamp recordings were performed at 32 to 34°C in both HEK cell lines and DRG neurons. The recordings were taken after a 10-minute application of EtOH or protein kinase C (PKC) activator (phorbol 12-myristate 13-acetate [PMA]). RESULTS We recorded T-type Ca²⁺ currents (T-currents) from 3 channel isoforms (CaV3.1, CaV3.2, and CaV3.3) before and during administration of EtOH. We found that only 1 isoform, CaV3.2, was significantly affected by EtOH. EtOH reduced current density as well as producing a hyperpolarizing shift in steady-state inactivation of both CaV3.2 currents from HEK 293 cell lines and in native T-currents from DRG neurons that are known to be enriched in CaV3.2. A myristoylated PKC peptide inhibitor (MPI) blocked the major EtOH effects, in both the cell lines and the DRG neurons. However, PMA effects were more complex. Lower concentration PMA (100 nM) replicated the major effects of EtOH, while higher concentration PMA (1 μM) did not, suggesting that the EtOH effects operate through activation of PKC and were mimicked by lower concentration of PMA. CONCLUSIONS EtOH primarily affects the CaV3.2 isoform of T-type Ca²⁺ channels acting through PKC, highlighting a novel target and mechanism for EtOH effects on excitable membranes.
Collapse
Affiliation(s)
- Hong Qu Shan
- Department of Neurobiology and Anatomy, Wake Forest University School of Medicine, Winston-Salem, NC 27157, USA
| | | | | |
Collapse
|
19
|
Cheong E, Shin HS. T-type Ca²⁺ channels in absence epilepsy. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2013; 1828:1560-71. [PMID: 23416255 DOI: 10.1016/j.bbamem.2013.02.002] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/27/2012] [Revised: 01/15/2013] [Accepted: 02/01/2013] [Indexed: 11/28/2022]
Abstract
Low-voltage-activated T-type Ca²⁺ channels are highly expressed in the thalamocortical circuit, suggesting that they play a role in this brain circuit. Indeed, low-threshold burst firing mediated by T-type Ca²⁺ channels has long been implicated in the synchronization of the thalamocortical circuit. Over the past few decades, the conventional view has been that rhythmic burst firing mediated by T-type channels in both thalamic reticular nuclie (TRN) and thalamocortical (TC) neurons are equally critical in the generation of thalamocortical oscillations during sleep rhythms and spike-wave-discharges (SWDs). This review broadly investigates recent studies indicating that even though both TRN and TC nuclei are required for thalamocortical oscillations, the contributions of T-type channels to TRN and TC neurons are not equal in the genesis of sleep spindles and SWDs. T-type channels in TC neurons are an essential component of SWD generation, whereas the requirement for TRN T-type channels in SWD generation remains controversial at least in the GBL model of absence seizures. Therefore, a deeper understanding of the functional consequences of modulating each T-type channel subtype could guide the development of therapeutic tools for absence seizures while minimizing side effects on physiological thalamocortical oscillations. This article is part of a Special Issue entitled: Calcium channels.
Collapse
Affiliation(s)
- Eunji Cheong
- Department of Biotechnology, Translational Research Center for Protein Function Control, College of Life Science and Biotechnology, Yonsei University, Seoul, Republic of Korea.
| | | |
Collapse
|
20
|
Tringham E, Powell KL, Cain SM, Kuplast K, Mezeyova J, Weerapura M, Eduljee C, Jiang X, Smith P, Morrison JL, Jones NC, Braine E, Rind G, Fee-Maki M, Parker D, Pajouhesh H, Parmar M, O'Brien TJ, Snutch TP. T-type calcium channel blockers that attenuate thalamic burst firing and suppress absence seizures. Sci Transl Med 2012; 4:121ra19. [PMID: 22344687 DOI: 10.1126/scitranslmed.3003120] [Citation(s) in RCA: 145] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Absence seizures are a common seizure type in children with genetic generalized epilepsy and are characterized by a temporary loss of awareness, arrest of physical activity, and accompanying spike-and-wave discharges on an electroencephalogram. They arise from abnormal, hypersynchronous neuronal firing in brain thalamocortical circuits. Currently available therapeutic agents are only partially effective and act on multiple molecular targets, including γ-aminobutyric acid (GABA) transaminase, sodium channels, and calcium (Ca(2+)) channels. We sought to develop high-affinity T-type specific Ca(2+) channel antagonists and to assess their efficacy against absence seizures in the Genetic Absence Epilepsy Rats from Strasbourg (GAERS) model. Using a rational drug design strategy that used knowledge from a previous N-type Ca(2+) channel pharmacophore and a high-throughput fluorometric Ca(2+) influx assay, we identified the T-type Ca(2+) channel blockers Z941 and Z944 as candidate agents and showed in thalamic slices that they attenuated burst firing of thalamic reticular nucleus neurons in GAERS. Upon administration to GAERS animals, Z941 and Z944 potently suppressed absence seizures by 85 to 90% via a mechanism distinct from the effects of ethosuximide and valproate, two first-line clinical drugs for absence seizures. The ability of the T-type Ca(2+) channel antagonists to inhibit absence seizures and to reduce the duration and cycle frequency of spike-and-wave discharges suggests that these agents have a unique mechanism of action on pathological thalamocortical oscillatory activity distinct from current drugs used in clinical practice.
Collapse
Affiliation(s)
- Elizabeth Tringham
- Zalicus Pharmaceuticals Ltd., Suite 301, 2389 Health Sciences Mall, Vancouver, British Columbia V6T 1Z3, Canada
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
21
|
Eckle VS, Digruccio MR, Uebele VN, Renger JJ, Todorovic SM. Inhibition of T-type calcium current in rat thalamocortical neurons by isoflurane. Neuropharmacology 2012; 63:266-73. [PMID: 22491022 DOI: 10.1016/j.neuropharm.2012.03.018] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2011] [Revised: 02/27/2012] [Accepted: 03/16/2012] [Indexed: 10/28/2022]
Abstract
Thalamocortical (TC) neurons provide the major sensory input to the mammalian somatosensory cortex. Decreased activity of these cells may be pivotal in the ability of general anesthetics to induce loss of consciousness and promote sleep (hypnosis). T-type voltage-gated calcium currents (T-currents) have a key function regulating the cellular excitability of TC neurons and previous studies have indicated that volatile general anesthetics may alter the excitability of these neurons. Using a patch-clamp technique, we investigated the mechanisms whereby isoflurane, a common volatile anesthetic, modulates isolated T-currents and T-current-dependent excitability of native TC neurons in acute brain slices of the rat. In voltage-clamp experiments, we found that isoflurane strongly inhibited peak amplitude of T-current, yielding an IC(50) of 1.1 vol-% at physiological membrane potentials. Ensuing biophysical studies demonstrated that inhibition was more prominent at depolarized membrane potentials as evidenced by hyperpolarizing shifts in channel availability curves. In current-clamp experiments we found that isoflurane decreased the rate of depolarization of low-threshold-calcium spikes (LTCSs) and consequently increased the latency of rebound spike firing at the same concentrations that inhibited isolated T-currents. This effect was mimicked by a novel selective T-channel blocker 3,5-dichloro-N-[1-(2,2-dimethyl-tetrahydro-pyran-4-ylmethyl)-4-fluoro-piperidin-4-ylmethyl]-benzamide (TTA-P2). In contrast, isoflurane and TTA-P2 had minimal effect on resting membrane potential and cell input resistance. We propose that the clinical properties of isoflurane may at least partly be provided by depression of thalamic T-currents.
Collapse
Affiliation(s)
- Veit-Simon Eckle
- Department of Anesthesiology, University of Virginia Health System, School of Medicine, Charlottesville, VA, USA
| | | | | | | | | |
Collapse
|
22
|
Weiss N, Hameed S, Fernández-Fernández JM, Fablet K, Karmazinova M, Poillot C, Proft J, Chen L, Bidaud I, Monteil A, Huc-Brandt S, Lacinova L, Lory P, Zamponi GW, De Waard M. A Ca(v)3.2/syntaxin-1A signaling complex controls T-type channel activity and low-threshold exocytosis. J Biol Chem 2011; 287:2810-8. [PMID: 22130660 DOI: 10.1074/jbc.m111.290882] [Citation(s) in RCA: 95] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
T-type calcium channels represent a key pathway for Ca(2+) entry near the resting membrane potential. Increasing evidence supports a unique role of these channels in fast and low-threshold exocytosis in an action potential-independent manner, but the underlying molecular mechanisms have remained unknown. Here, we report the existence of a syntaxin-1A/Ca(v)3.2 T-type calcium channel signaling complex that relies on molecular determinants that are distinct from the synaptic protein interaction site (synprint) found in synaptic high voltage-activated calcium channels. This interaction potently modulated Ca(v)3.2 channel activity, by reducing channel availability. Other members of the T-type calcium channel family were also regulated by syntaxin-1A, but to a smaller extent. Overexpression of Ca(v)3.2 channels in MPC 9/3L-AH chromaffin cells induced low-threshold secretion that could be prevented by uncoupling the channels from syntaxin-1A. Altogether, our findings provide compelling evidence for the existence of a syntaxin-1A/T-type Ca(2+) channel signaling complex and provide new insights into the molecular mechanism by which these channels control low-threshold exocytosis.
Collapse
Affiliation(s)
- Norbert Weiss
- Department of Physiology and Pharmacology, Hotchkiss Brain Institute, University of Calgary, Calgary T2N4N1, Canada
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
23
|
The sleep relay--the role of the thalamus in central and decentral sleep regulation. Pflugers Arch 2011; 463:53-71. [PMID: 21912835 DOI: 10.1007/s00424-011-1014-6] [Citation(s) in RCA: 60] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2011] [Revised: 08/08/2011] [Accepted: 08/11/2011] [Indexed: 10/17/2022]
Abstract
Surprisingly, the concept of sleep, its necessity and function, the mechanisms of action, and its elicitors are far from being completely understood. A key to sleep function is to determine how and when sleep is induced. The aim of this review is to merge the classical concepts of central sleep regulation by the brainstem and hypothalamus with the recent findings on decentral sleep regulation in local neuronal assemblies and sleep regulatory substances that create a scenario in which sleep is both local and use dependent. The interface between these concepts is provided by thalamic cellular and network mechanisms that support rhythmogenesis of sleep-related activity. The brainstem and the hypothalamus centrally set the pace for sleep-related activity throughout the brain. Decentral regulation of the sleep-wake cycle was shown in the cortex, and the homeostat of non-rapid-eye-movement sleep is made up by molecular networks of sleep regulatory substances, allowing individual neurons or small neuronal assemblies to enter sleep-like states. Thalamic neurons provide state-dependent gating of sensory information via their ability to produce different patterns of electrogenic activity during wakefulness and sleep. Many mechanisms of sleep homeostasis or sleep-like states of neuronal assemblies, e.g. by the action of adenosine, can also be found in thalamic neurons, and we summarize cellular and network mechanisms of the thalamus that may elicit non-REM sleep. It is argued that both central and decentral regulators ultimately target the thalamus to induce global sleep-related oscillatory activity. We propose that future studies should integrate ideas of central, decentral, and thalamic sleep generation.
Collapse
|
24
|
The Ca(V)3.3 calcium channel is the major sleep spindle pacemaker in thalamus. Proc Natl Acad Sci U S A 2011; 108:13823-8. [PMID: 21808016 DOI: 10.1073/pnas.1105115108] [Citation(s) in RCA: 166] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Low-threshold (T-type) Ca(2+) channels encoded by the Ca(V)3 genes endow neurons with oscillatory properties that underlie slow waves characteristic of the non-rapid eye movement (NREM) sleep EEG. Three Ca(V)3 channel subtypes are expressed in the thalamocortical (TC) system, but their respective roles for the sleep EEG are unclear. Ca(V)3.3 protein is expressed abundantly in the nucleus reticularis thalami (nRt), an essential oscillatory burst generator. We report the characterization of a transgenic Ca(V)3.3(-/-) mouse line and demonstrate that Ca(V)3.3 channels are indispensable for nRt function and for sleep spindles, a hallmark of natural sleep. The absence of Ca(V)3.3 channels prevented oscillatory bursting in the low-frequency (4-10 Hz) range in nRt cells but spared tonic discharge. In contrast, adjacent TC neurons expressing Ca(V)3.1 channels retained low-threshold bursts. Nevertheless, the generation of synchronized thalamic network oscillations underlying sleep-spindle waves was weakened markedly because of the reduced inhibition of TC neurons via nRt cells. T currents in Ca(V)3.3(-/-) mice were <30% compared with those in WT mice, and the remaining current, carried by Ca(V)3.2 channels, generated dendritic [Ca(2+)](i) signals insufficient to provoke oscillatory bursting that arises from interplay with Ca(2+)-dependent small conductance-type 2 K(+) channels. Finally, naturally sleeping Ca(V)3.3(-/-) mice showed a selective reduction in the power density of the σ frequency band (10-12 Hz) at transitions from NREM to REM sleep, with other EEG waves remaining unaltered. Together, these data identify a central role for Ca(V)3.3 channels in the rhythmogenic properties of the sleep-spindle generator and provide a molecular target to elucidate the roles of sleep spindles for brain function and development.
Collapse
|
25
|
Ku WH, Schneider SP. Multiple T-type Ca2+ current subtypes in electrophysiologically characterized hamster dorsal horn neurons: possible role in spinal sensory integration. J Neurophysiol 2011; 106:2486-98. [PMID: 21795620 DOI: 10.1152/jn.01083.2010] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Whole cell patch-clamp recordings were used to investigate the contribution of transient, low-threshold calcium currents (I(T)) to firing properties of hamster spinal dorsal horn neurons. I(T) was widely, though not uniformly, expressed by cells in Rexed's laminae I-IV and correlated with the pattern of action potential discharge evoked under current-clamp conditions: I(T) in neurons responding to constant membrane depolarization with one or two action potentials was nearly threefold larger than I(T) in cells responding to the same activation with continuous firing. I(T) was evoked by depolarizing voltage ramps exceeding 46 mV/s and increased with ramp slope (240-2,400 mV/s). Bath application of 200 μM Ni(2+) depressed ramp-activated I(T). Phasic firing recorded in current clamp could only be activated by membrane depolarizations exceeding ∼43-46 mV/s and was blocked by Ni(2+) and mibefradil, suggesting I(T) as an underlying mechanism. Two components of I(T), "fast" and "slow," were isolated based on a difference in time constant of inactivation (12 ms and 177 ms, respectively). The amplitude of the fast subtype depended on the slope of membrane depolarization and was twice as great in burst-firing cells than in cells having a tonic discharge. Post hoc single-cell RT-PCR analyses suggested that the fast component is associated with the Ca(V)3.1 channel subtype. I(T) may enhance responses of phasic-firing dorsal horn neurons to rapid membrane depolarizations and contribute to an ability to discriminate between afferent sensory inputs that encode high- and low-frequency stimulus information.
Collapse
Affiliation(s)
- Wen-hsin Ku
- Dept. of Physiology, Michigan State Univ., East Lansing, MI 48824-3320, USA
| | | |
Collapse
|
26
|
Masurkar AV, Chen WR. Calcium currents of olfactory bulb juxtaglomerular cells: profile and multiple conductance plateau potential simulation. Neuroscience 2011; 192:231-46. [PMID: 21704681 DOI: 10.1016/j.neuroscience.2011.06.016] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2011] [Revised: 05/06/2011] [Accepted: 06/03/2011] [Indexed: 11/25/2022]
Abstract
The olfactory glomerulus is the locus of information transfer between olfactory sensory neurons and output neurons of the olfactory bulb. Juxtaglomerular cells (JGCs) may influence intraglomerular processing by firing plateau potentials that support multiple spikes. It is unclear what inward currents mediate this firing pattern. In previous work, we characterized potassium currents of JGCs. We focus here on the inward currents using whole cell current clamp and voltage recording in a rat in vitro slice preparation, as well as computer simulation. We first showed that sodium current was not required to mediate plateau potentials. Voltage clamp characterization of calcium current (I(Ca)) determined that I(Ca) consisted of a slow activating, rapidly inactivating (τ(10%-90% rise) 6-8 ms, τ(inactivation) 38-77 ms) component I(cat1), similar to T-type currents, and a sustained (τ(inactivation)>>500 ms) component I(cat2), likely composed of L-type and P/Q-type currents. We used computer simulation to test their roles in plateau potential firing. We robustly modeled I(cat1) and I(cat2) to Hodgkin-Huxley schemes (m(3)h and m(2), respectively) and simulated a JGC plateau potential with six conductances: calcium currents as above, potassium currents from our prior study (A-type I(kt1), D-type I(kt2), delayed rectifier I(kt3)), and a fast sodium current (I(Na)). We demonstrated that I(cat1) was required for mediating the plateau potential, unlike I(Na) and I(cat2), and its τ(inactivation) determined plateau duration. We also found that I(kt1) dictated plateau potential shape more than I(kt2) and I(kt3). The influence of these two transient and opposing conductances suggests a unique mechanism of plateau potential physiology.
Collapse
Affiliation(s)
- A V Masurkar
- Department of Neurobiology, Yale University School of Medicine, New Haven, CT 06520, USA.
| | | |
Collapse
|
27
|
Cav2.3 channels are critical for oscillatory burst discharges in the reticular thalamus and absence epilepsy. Neuron 2011; 70:95-108. [PMID: 21482359 DOI: 10.1016/j.neuron.2011.02.042] [Citation(s) in RCA: 62] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/21/2011] [Indexed: 11/27/2022]
Abstract
Neurons of the reticular thalamus (RT) display oscillatory burst discharges that are believed to be critical for thalamocortical network oscillations related to absence epilepsy. Ca²+-dependent mechanisms underlie such oscillatory discharges. However, involvement of high-voltage activated (HVA) Ca²+ channels in this process has been discounted. We examined this issue closely using mice deficient for the HVA Ca(v)2.3 channels. In brain slices of Ca(v)2.3⁻/⁻, a hyperpolarizing current injection initiated a low-threshold burst of spikes in RT neurons; however, subsequent oscillatory burst discharges were severely suppressed, with a significantly reduced slow afterhyperpolarization (AHP). Consequently, the lack of Ca(v)2.3 resulted in a marked decrease in the sensitivity of the animal to γ-butyrolactone-induced absence epilepsy. Local blockade of Ca(v)2.3 channels in the RT mimicked the results of Ca(v)2.3⁻/⁻ mice. These results provide strong evidence that Ca(v)2.3 channels are critical for oscillatory burst discharges in RT neurons and for the expression of absence epilepsy.
Collapse
|
28
|
Liao YF, Tsai ML, Chen CC, Yen CT. Involvement of the Cav3.2 T-type calcium channel in thalamic neuron discharge patterns. Mol Pain 2011; 7:43. [PMID: 21639922 PMCID: PMC3127773 DOI: 10.1186/1744-8069-7-43] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2010] [Accepted: 06/04/2011] [Indexed: 11/30/2022] Open
Abstract
Background Mice that have defects in their low-threshold T-type calcium channel (T-channel) genes show altered pain behaviors. The changes in the ratio of nociceptive neurons and the burst firing property of reticular thalamic (RT) and ventroposterior (VP) neurons in Cav3.2 knockout (KO) mice were studied to test the involvement of thalamic T-channel and burst firing activity in pain function. Results Under pentobarbital or urethane anesthesia, the patterns of tonic and burst firings were recorded in functionally characterized RT and VPL neurons of Cav3.2 KO mice. Many RT neurons were nociceptive (64% under pentobarbital anesthesia and 50% under urethane anesthesia). Compared to their wild-type (WT) controls, fewer nociceptive RT neurons were found in Cav3.2 KO mice. Both nociceptive and tactile RT neurons showed fewer bursts in Cav3.2 KO mice. Within a burst, RT neurons of Cav3.2 KO mice had a lower spike frequency and less-prominent accelerando-decelerando change. In contrast, VP neurons of Cav3.2 KO mice showed a higher ratio of bursts and a higher discharge rate within a burst than those of the WT control. In addition, the long-lasting tonic firing episodes in RT neurons of the Cav3.2 KO had less stereotypic regularity than their counterparts in WT mice. Conclusions RT might be important in nociception of the mouse. In addition, we showed an important role of Cav3.2 subtype of T-channel in RT burst firing pattern. The decreased occurrence and slowing of the bursts in RT neurons might cause the increased VP bursts. These changes would be factors contributing to alternation of pain behavior in the Cav3.2 KO mice.
Collapse
Affiliation(s)
- Yi-Fang Liao
- Institute of Zoology, National Taiwan University, Roosevelt Road, Taipei, Taiwan
| | | | | | | |
Collapse
|
29
|
Cain SM, Snutch TP. Contributions of T-type calcium channel isoforms to neuronal firing. Channels (Austin) 2011; 4:475-82. [PMID: 21139420 DOI: 10.4161/chan.4.6.14106] [Citation(s) in RCA: 138] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
Low voltage-activated (LVA) T-type calcium channels play critical roles in the excitability of many cell types and are a focus of research aimed both at understanding the physiological basis of calcium channel-dependent signaling and the underlying pathophysiology associated with hyperexcitability disorders such as epilepsy. These channels play a critical role towards neuronal firing in both conducting calcium ions during action potentials and also in switching neurons between distinct modes of firing. In this review the properties of the CaV3.1, CaV3.2 and CaV3.3 T-type channel isoforms is discussed in relation to their individual contributions to action potentials during burst and tonic firing states as well their roles in switching between firing states.
Collapse
Affiliation(s)
- Stuart M Cain
- Michael Smith Laboratories, University of British Columbia, Vancouver, BC, Canada.
| | | |
Collapse
|
30
|
Parajuli LK, Fukazawa Y, Watanabe M, Shigemoto R. Subcellular distribution of α1G subunit of T-type calcium channel in the mouse dorsal lateral geniculate nucleus. J Comp Neurol 2011; 518:4362-74. [PMID: 20853512 DOI: 10.1002/cne.22461] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
T-type calcium channels play a pivotal role in regulating neural membrane excitability in the nervous system. However, the precise subcellular distributions of T-type channel subunits and their implication for membrane excitability are not well understood. Here we investigated the subcellular distribution of the α1G subunit of the calcium channel which is expressed highly in the mouse dorsal lateral geniculate nucleus (dLGN). Light microscopic analysis demonstrated that dLGN exhibits intense immunoperoxidase reactivity for the α1G subunit. Electron microscopic observation showed that the labeling was present in both the relay cells and interneurons and was found in the somatodendritic, but not axonal, domains of these cells. Most of the immunogold particles for the α1G subunit were either associated with the plasma membrane or the intracellular membranes. Reconstruction analysis of serial electron microscopic images revealed that the intensity of the intracellular labeling exhibited a gradient such that the labeling density was higher in the proximal dendrite and progressively decreased towards the distal dendrite. In contrast, the plasma membrane-associated particles were distributed with a uniform density over the somatodendritic surface of dLGN cells. The labeling density in the relay cell plasma membrane was about 3-fold higher than that of the interneurons. These results provide ultrastructural evidence for cell-type-specific expression levels and for uniform expression density of the α1G subunit over the plasma membrane of dLGN cells.
Collapse
Affiliation(s)
- Laxmi Kumar Parajuli
- Division of Cerebral Structure, National Institute for Physiological Sciences, Okazaki 444-8787, Japan
| | | | | | | |
Collapse
|
31
|
Low-threshold Ca2+ current amplifies distal dendritic signaling in thalamic reticular neurons. J Neurosci 2010; 30:15419-29. [PMID: 21084598 DOI: 10.1523/jneurosci.3636-10.2010] [Citation(s) in RCA: 77] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
The low-threshold transient calcium current (I(T)) plays a critical role in modulating the firing behavior of thalamic neurons; however, the role of I(T) in the integration of afferent information within the thalamus is virtually unknown. We have used two-photon laser scanning microscopy coupled with whole-cell recordings to examine calcium dynamics in the neurons of the strategically located thalamic reticular nucleus (TRN). We now report that a single somatic burst discharge evokes large-magnitude calcium responses, via I(T), in distal TRN dendrites. The magnitude of the burst-evoked calcium response was larger than those observed in thalamocortical projection neurons under the same conditions. We also demonstrate that direct stimulation of distal TRN dendrites, via focal glutamate application and synaptic activation, can locally activate distal I(T), producing a large distal calcium response independent of the soma/proximal dendrites. These findings strongly suggest that distally located I(T) may function to amplify afferent inputs. Boosting the magnitude ensures integration at the somatic level by compensating for attenuation that would normally occur attributable to passive cable properties. Considering the functional architecture of the TRN, elongated nature of their dendrites, and robust dendritic signaling, these distal dendrites could serve as sites of intense intra-modal/cross-modal integration and/or top-down modulation, leading to focused thalamocortical communication.
Collapse
|
32
|
Orestes P, Todorovic SM. Are neuronal voltage-gated calcium channels valid cellular targets for general anesthetics? Channels (Austin) 2010; 4:518-22. [PMID: 21164281 DOI: 10.4161/chan.4.6.12873] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
The effects of anesthetics and analgesics on ion channels have been the subject of intense research since recent reports of direct actions of anesthetic molecules on ion channel proteins. It is now known that ligand-gated channels, particularly γ-amino-butyric acid (GABAA) and N-methyl-D-aspartate (NMDA) receptors, play a key role in mediating anesthetic actions, but these channels are unable to account for all aspects of clinical anesthesia such as loss of consciousness, immobility, analgesia, amnesia, and muscle relaxation. Furthermore, an assortment of voltage-gated and background channels also display anesthetic sensitivity and a key question arises: What role do these other channels play in clinical anesthesia? These channels have overlapping physiological roles and pharmacological profiles, making it difficult to assign aspects of the anesthetic state to individual channel types. Here, we will focus on the function of neuronal voltage-gated calcium channels in mediating the effects of general anesthetics.
Collapse
Affiliation(s)
- Peihan Orestes
- Department of Anesthesiology, University of Virginia School of Medicine and Health System, Charlottesville, VA, USA
| | | |
Collapse
|
33
|
Joksovic PM, Todorovic SM. Isoflurane modulates neuronal excitability of the nucleus reticularis thalami in vitro. Ann N Y Acad Sci 2010; 1199:36-42. [PMID: 20633107 DOI: 10.1111/j.1749-6632.2009.05172.x] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
The thalamus has a key function in processing sensory information, sleep, and cognition. We examined the effects of a common volatile anesthetic, isoflurane, on modulation of neuronal excitability in reticular thalamic nucleus (nRT) in intact brain slices from immature rats. In current-clamp recordings, isoflurane (300-600 micromol/L) consistently depolarized membrane potential, decreased input resistance, and inhibited both rebound burst firing and tonic spike firing modes of nRT neurons. The isoflurane-induced depolarization persisted not only in the presence of tetrodotoxin, but after replacement of Ca(2+) with Ba(2+) ions in external solution; it was abolished by partial replacement of extracellular Na(+) ions with N-methyl-D-glucamine. In voltage-clamp recordings, we found that isoflurane slowed recovery from inactivation of T-type Ca(2+) current. Thus, at clinically relevant concentrations, isoflurane inhibits neuronal excitability of nRT neurons in developing brain via multiple ion channels. Inhibition of the neuronal excitability of thalamic cells may contribute to impairment of sensory information transfer in the thalamocortical network by general anesthetics. The findings may be important for understanding cellular mechanisms of anesthesia, such as loss of consciousness and potentially damaging consequences of general anesthetics on developing mammalian brains.
Collapse
Affiliation(s)
- Pavle M Joksovic
- Department of Anesthesiology, University of Virginia School of Medicine, Charlottesville, Virginia, USA
| | | |
Collapse
|
34
|
Eckle VS, Todorovic SM. Mechanisms of inhibition of CaV3.1 T-type calcium current by aliphatic alcohols. Neuropharmacology 2010; 59:58-69. [PMID: 20363234 DOI: 10.1016/j.neuropharm.2010.03.016] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2010] [Revised: 03/25/2010] [Accepted: 03/26/2010] [Indexed: 11/30/2022]
Abstract
Many aliphatic alcohols modulate activity of various ion channels involved in sensory processing and also exhibit anesthetic capacity in vivo. Although the interaction of one such compound, 1-octanol (octanol) with different T-type calcium channels (T-channels) has been described, the mechanisms of current modulation and its functional significance are not well studied. Using patch-clamp technique, we investigated the mechanisms of inhibition of T-currents by a series of aliphatic alcohols in recombinant human Ca(V)3.1 (alpha1G) T-channel isoform expressed in human embryonic kidney (HEK) 293 cells and thalamocortical (TC) relay neurons in brain slices of young rats. Octanol, 1-heptanol (heptanol) and 1-hexanol (hexanol) inhibited the recombinant Ca(V)3.1 currents in concentration-dependent manner yielding IC(50) values of 362 microM, 1063 microM and 3167 microM, respectively. Octanol similarly inhibited native thalamic Ca(V)3.1 T-currents with an IC(50) of 287 microM and diminished burst firing without significant effect on passive membrane properties of these neurons. Inhibitory effect of octanol on T-currents in both native and recombinant cells was accompanied with accelerated macroscopic inactivation kinetics and hyperpolarizing shift in the steady-state inactivation curve. Additionally, octanol induced a depolarizing shift in steady-state activation curves of T-current in TC neurons. Surprisingly, the recovery from fast inactivation at hyperpolarized membrane potentials was accelerated by octanol up 3-fold in native but not recombinant channels. Given the importance of thalamocortical pathways in providing sleep, arousal, and anesthetic states, modulation of thalamic T-currents may at least contribute to the pharmacological effects of aliphatic alcohols.
Collapse
Affiliation(s)
- Veit-Simon Eckle
- Department of Anesthesiology, University of Virginia Health System, School of Medicine, Charlottesville, VA 22908-0710, USA
| | | |
Collapse
|
35
|
Eckle VS, Hucklenbruch C, Todorovic SM. [What do we know about anesthetic mechanisms?: hypnosis, unresponsiveness to surgical incision and amnesia]. Anaesthesist 2010; 58:1144-9. [PMID: 19760252 DOI: 10.1007/s00101-009-1618-9] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Despite the increase of molecular knowledge in anesthesia research over the past decades there is still ongoing discussion about the mechanisms of anesthesia. This article focuses on presenting anesthetic sensitive ligand and voltage gated ion channels. The impact on anesthetic modulated ion channels is summarized for clinically commonly used anesthetics isoflurane, propofol and ketamine. Furthermore, the anesthetic features hypnosis, unresponsiveness to surgical incision and amnesia and their putative relevant anatomical sites in the central nervous system are briefly introduced.
Collapse
Affiliation(s)
- V-S Eckle
- Department of Anesthesiology, University of Virginia, 22909, Charlottesville, VA, USA.
| | | | | |
Collapse
|
36
|
Selective T-type calcium channel block in thalamic neurons reveals channel redundancy and physiological impact of I(T)window. J Neurosci 2010; 30:99-109. [PMID: 20053892 DOI: 10.1523/jneurosci.4305-09.2010] [Citation(s) in RCA: 156] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Although it is well established that low-voltage-activated T-type Ca(2+) channels play a key role in many neurophysiological functions and pathological states, the lack of selective and potent antagonists has so far hampered a detailed analysis of the full impact these channels might have on single-cell and neuronal network excitability as well as on Ca(2+) homeostasis. Recently, a novel series of piperidine-based molecules has been shown to selectively block recombinant T-type but not high-voltage-activated (HVA) Ca(2+) channels and to affect a number of physiological and pathological T-type channel-dependent behaviors. Here we directly show that one of these compounds, 3,5-dichloro-N-[1-(2,2-dimethyl-tetrahydro-pyran-4-ylmethyl)-4-fluoro-piperidin-4-ylmethyl]-benzamide (TTA-P2), exerts a specific, potent (IC(50) = 22 nm), and reversible inhibition of T-type Ca(2+) currents of thalamocortical and reticular thalamic neurons, without any action on HVA Ca(2+) currents, Na(+) currents, action potentials, and glutamatergic and GABAergic synaptic currents. Thus, under current-clamp conditions, the low-threshold Ca(2+) potential (LTCP)-dependent high-frequency burst firing of thalamic neurons is abolished by TTA-P2, whereas tonic firing remains unaltered. Using TTA-P2, we provide the first direct demonstration of the presence of a window component of Ca(2+) channels in neurons and its contribution to the resting membrane potential of thalamic neurons and to the Up state of their intrinsically generated slow (<1 Hz) oscillation. Moreover, we demonstrate that activation of only a small fraction of the T-type channel population is required to generate robust LTCPs, suggesting that LTCP-driven bursts of action potentials can be evoked at depolarized potentials where the vast majority of T-type channels are inactivated.
Collapse
|
37
|
Kovács K, SÃk A, Ricketts C, Timofeev I. Subcellular distribution of low-voltage activated T-type Ca2+ channel subunits (Cav3.1 and Cav3.3) in reticular thalamic neurons of the cat. J Neurosci Res 2010; 88:448-60. [DOI: 10.1002/jnr.22200] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
|
38
|
Coulon P, Herr D, Kanyshkova T, Meuth P, Budde T, Pape HC. Burst discharges in neurons of the thalamic reticular nucleus are shaped by calcium-induced calcium release. Cell Calcium 2009; 46:333-46. [PMID: 19913909 DOI: 10.1016/j.ceca.2009.09.005] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2009] [Revised: 09/01/2009] [Accepted: 09/27/2009] [Indexed: 11/18/2022]
Abstract
The nucleus reticularis thalami (NRT) is a layer of inhibitory neurons that surrounds the dorsal thalamus. It appears to be the 'pacemaker' of certain forms of slow oscillations in the thalamus and was proposed to be a key determinant of the internal attentional searchlight as well as the origin of hypersynchronous activity during absence seizures. Neurons of the NRT exhibit a transient depolarization termed low threshold spike (LTS) following sustained hyperpolarization. This is caused by the activation of low-voltage-activated Ca2+ channels (LVACC). Although the role of these channels in thalamocortical oscillations was studied in great detail, little is known about the downstream intracellular Ca2+ signalling pathways and their feedback onto the oscillations. A signalling triad consisting of the sarco(endo)plasmic reticulum calcium ATPase (SERCA), Ca2+ activated K+ channels (SK2), and LVACC is active in dendrites of NRT neurons and shapes rhythmic oscillations. The aim of our study was to find out (i) if and how Ca2+-induced Ca2+ release (CICR) via ryanodine receptors (RyR) can be evoked in NRT neurons and (ii) how the released Ca2+ affects burst activity. Combining electrophysiological, immunohistochemical, and two-photon Ca2+ imaging techniques, we show that CICR in NRT neurons takes place by a cell-type specific coupling of LVACC and RyR. CICR could be evoked by the application of caffeine, by activation of LVACC, or by repetitive LTS generation. During the latter, CICR contributed 30% to the resulting build-up of [Ca2+]i. CICR was abolished by cyclopiazonic acid, a specific blocker for SERCA, or by high concentrations of ryanodine (50 microM). Unlike other thalamic nuclei, in the NRT the activation of high-voltage-activated Ca2+ channels failed to evoke CICR. While action potentials contributed little to the build-up of [Ca2+]i upon repetitive LTS generation, the Ca2+ released via RyR significantly reduced the number of action potentials during an LTS and reduced the neurons' low threshold activity, thus potentially reducing hypersynchronicity. This effect persisted in the presence of the SK2 channel blocker apamin. We conclude that the activation of LVACC specifically causes CICR via RyR in neurons of the NRT, thereby adding a Ca2+-dependent intracellular route to the mechanisms determining rhythmic oscillatory bursting in this nucleus.
Collapse
Affiliation(s)
- Philippe Coulon
- Institut für Physiologie I, Westfälische Wilhelms-Universität Münster, Robert-Koch-Str. 27a, 48149 Münster, Germany.
| | | | | | | | | | | |
Collapse
|
39
|
Joksovic PM, Choe WJ, Nelson MT, Orestes P, Brimelow BC, Todorovic SM. Mechanisms of inhibition of T-type calcium current in the reticular thalamic neurons by 1-octanol: implication of the protein kinase C pathway. Mol Pharmacol 2009; 77:87-94. [PMID: 19846748 DOI: 10.1124/mol.109.059931] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
Abstract
Recent studies indicate that T-type calcium channels (T-channels) in the thalamus are cellular targets for general anesthetics. Here, we recorded T-currents and underlying low-threshold calcium spikes from neurons of nucleus reticularis thalami (nRT) in brain slices from young rats and investigated the mechanisms of their modulation by an anesthetic alcohol, 1-octanol. We found that 1-octanol inhibited native T-currents at subanesthetic concentrations with an IC(50) of approximately 4 muM. In contrast, 1-octanol was up to 30-fold less potent in inhibiting recombinant Ca(V)3.3 T-channels heterologously expressed in human embryonic kidney cells. Inhibition of both native and recombinant T-currents was accompanied by a hyperpolarizing shift in steady-state inactivation, indicating that 1-octanol stabilized inactive states of the channel. To explore the mechanisms underlying higher 1-octanol potency in inhibiting native nRT T-currents, we tested the effect of the protein kinase C (PKC) activator phorbol 12-myristate 13-acetate (PMA) and PKC inhibitors. We found that PMA caused a modest increase of T-current, whereas the inactive PMA analog 4alpha-PMA failed to affect T-current in nRT neurons. In contrast, 12-(2-cyanoethyl)-6,7,12,13-tetrahydro-13-methyl-5-oxo-5H-indolo(2,3-a)pyrrolo(3,4-c)-carbazole (Go 6976), an inhibitor of calcium-dependent PKC, decreased baseline T-current amplitude in nRT cells and abolished the effects of subsequently applied 1-octanol. The effects of 1-octanol were also abolished by chelation of intracellular calcium ions with 1,2-bis(2-aminophenoxy)ethane-N,N,N',N'-tetraacetic acid. Taken together, these results suggest that inhibition of calcium-dependent PKC signaling is a possible molecular substrate for modulation of T-channels in nRT neurons by 1-octanol.
Collapse
Affiliation(s)
- Pavle M Joksovic
- Department of Anesthesiology, University of Virginia Health System, Charlottesville, VA 22908-0710, USA
| | | | | | | | | | | |
Collapse
|
40
|
Joksovic PM, Weiergräber M, Lee W, Struck H, Schneider T, Todorovic SM. Isoflurane-sensitive presynaptic R-type calcium channels contribute to inhibitory synaptic transmission in the rat thalamus. J Neurosci 2009; 29:1434-45. [PMID: 19193890 PMCID: PMC2659547 DOI: 10.1523/jneurosci.5574-08.2009] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2008] [Revised: 12/29/2008] [Accepted: 01/06/2009] [Indexed: 11/21/2022] Open
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.
Collapse
Affiliation(s)
| | - Marco Weiergräber
- Department of Neurophysiology and
- Center for Molecular Medicine Cologne, University of Cologne, D-50931 Cologne, Germany
| | - WooYong Lee
- Departments of Anesthesiology and
- Department of Anesthesiology and Pain Medicine, InJe University, Sanggyepaik Hospital, Seoul 139-707, South Korea, and
| | - Henrik Struck
- Department of Neurophysiology and
- Center for Molecular Medicine Cologne, University of Cologne, D-50931 Cologne, Germany
| | - Toni Schneider
- Department of Neurophysiology and
- Center for Molecular Medicine Cologne, University of Cologne, D-50931 Cologne, Germany
| | - Slobodan M. Todorovic
- Departments of Anesthesiology and
- Neuroscience, University of Virginia School of Medicine, Charlottesville, Virginia 22908-0710
| |
Collapse
|
41
|
Orestes P, Bojadzic D, Chow RM, Todorovic SM. Mechanisms and functional significance of inhibition of neuronal T-type calcium channels by isoflurane. Mol Pharmacol 2008; 75:542-54. [PMID: 19038845 DOI: 10.1124/mol.108.051664] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Previous data have indicated that T-type calcium channels (low-voltage activated T-channels) are potently inhibited by volatile anesthetics. Although the interactions of T-channels with a number of anesthetics have been described, the mechanisms by which these agents modulate channel activity, and the functional consequences of such interactions, are not well studied. Here, we used patch-clamp recordings to explore the actions of a prototypical volatile anesthetic, isoflurane (Iso), on recombinant human Ca(V)3.1 and Ca(V)3.2 isoforms of T-channels. We also performed behavioral testing of anesthetic endpoints in mice lacking Ca(V)3.2. Iso applied at resting channel states blocked current through both isoforms in a similar manner at clinically relevant concentrations (1 minimum alveolar concentration, MAC). Inhibition was more prominent at depolarized membrane potentials (-65 versus -100 mV) as evidenced by hyperpolarizing shifts in channel availability curves and a 2.5-fold decrease in IC(50) values. Iso slowed recovery from inactivation and enhanced deactivation in both Ca(V)3.1 and Ca(V)3.2 in a comparable manner but caused a depolarizing shift in activation curves and greater use-dependent block of Ca(V)3.2 channels. In behavioral tests, Ca(V)3.2 knockout (KO) mice showed significantly decreased MAC in comparison with wild-type (WT) litter mates. KO and WT mice did not differ in loss of righting reflex, but mutant mice displayed a delayed onset of anesthetic induction. We conclude that state-dependent inhibition of T-channel isoforms in the central and peripheral nervous systems may contribute to isoflurane's important clinical effects.
Collapse
Affiliation(s)
- Peihan Orestes
- Department of Anesthesiology, University of Virginia Health System, Charlottesville, Virginia 22908-0710, USA
| | | | | | | |
Collapse
|
42
|
Himmel HM. Mechanisms Involved in Cardiac Sensitization by Volatile Anesthetics: General Applicability to Halogenated Hydrocarbons? Crit Rev Toxicol 2008; 38:773-803. [DOI: 10.1080/10408440802237664] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
|
43
|
Cueni L, Canepari M, Adelman JP, Lüthi A. Ca2+ signaling by T-type Ca2+ channels in neurons. Pflugers Arch 2008; 457:1161-72. [DOI: 10.1007/s00424-008-0582-6] [Citation(s) in RCA: 52] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2008] [Accepted: 08/15/2008] [Indexed: 10/21/2022]
|
44
|
Cueni L, Canepari M, Luján R, Emmenegger Y, Watanabe M, Bond CT, Franken P, Adelman JP, Lüthi A. T-type Ca2+ channels, SK2 channels and SERCAs gate sleep-related oscillations in thalamic dendrites. Nat Neurosci 2008; 11:683-92. [DOI: 10.1038/nn.2124] [Citation(s) in RCA: 163] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2008] [Accepted: 04/16/2008] [Indexed: 11/09/2022]
|
45
|
Joksovic PM, Covey DF, Todorovic SM. Inhibition of T-type calcium current in the reticular thalamic nucleus by a novel neuroactive steroid. Ann N Y Acad Sci 2008; 1122:83-94. [PMID: 18077566 DOI: 10.1196/annals.1403.006] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
Neurons of the nucleus reticularis of the thalamus (nRT) serve as an important inhibitory gate that controls trafficking of thalamocortical sensory signals and states of sleep, arousal, and epilepsy. T-type calcium channels in nRT play a crucial role in the subthreshold excitability of these neurons, but their modulation by neuroactive steroids has not been previously studied. Here we explored the effect of (3beta,5beta,17beta)-3-hydroxyandrostane-17-carbonitrile (3beta-OH), a novel neuroactive steroid on T-type currents recorded from nRT neurons in intact brain slices of young rats. We found in voltage-clamp experiments that 3beta-OH potently and reversibly decreased T-type Ca(2+) current amplitude and stabilized inactive states of the channels. In current-clamp experiments, 3beta-OH significantly decreased the frequency of action potential firing from negative membrane potentials and minimally changed passive membrane properties. Our results indicate that 5beta-reduced neuroactive steroids, through the mechanisms of inhibition of T-type Ca(2+) currents and diminished spike firing in nRT neurons, may be important agents in control of sensory information processing in physiological conditions and possibly pathological brain states associated with increased cellular excitability such as epilepsy and/or tissue ischemia/hypoxia.
Collapse
Affiliation(s)
- Pavle M Joksovic
- Department of Anesthesiology, University of Virginia Health System, P.O. Box 800710, Charlottesville, VA 22908-0710, USA
| | | | | |
Collapse
|
46
|
Cataldi M, Lariccia V, Marzaioli V, Cavaccini A, Curia G, Viggiano D, Canzoniero LMT, di Renzo G, Avoli M, Annunziato L. Zn2+ Slows Down CaV3.3 Gating Kinetics: Implications for Thalamocortical Activity. J Neurophysiol 2007; 98:2274-84. [PMID: 17699699 DOI: 10.1152/jn.00889.2006] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
We employed whole cell patch-clamp recordings to establish the effect of Zn2+ on the gating the brain specific, T-type channel isoform CaV3.3 expressed in HEK-293 cells. Zn2+ (300 μM) modified the gating kinetics of this channel without influencing its steady-state properties. When inward Ca2+ currents were elicited by step depolarizations at voltages above the threshold for channel opening, current inactivation was significantly slowed down while current activation was moderately affected. In addition, Zn2+ slowed down channel deactivation but channel recovery from inactivation was only modestly changed. Zn2+ also decreased whole cell Ca2+ permeability to 45% of control values. In the presence of Zn2+, Ca2+ currents evoked by mock action potentials were more persistent than in its absence. Furthermore, computer simulation of action potential generation in thalamic reticular cells performed to model the gating effect of Zn2+ on T-type channels (while leaving the kinetic parameters of voltage-gated Na+ and K+ unchanged) revealed that Zn2+ increased the frequency and the duration of burst firing, which is known to depend on T-type channel activity. In line with this finding, we discovered that chelation of endogenous Zn2+ decreased the frequency of occurrence of ictal-like epileptiform discharges in rat thalamocortical slices perfused with medium containing the convulsant 4-aminopyridine (50 μM). These data demonstrate that Zn2+ modulates CaV3.3 channel gating thus leading to increased neuronal excitability. We also propose that endogenous Zn2+ may have a role in controlling thalamocortical oscillations.
Collapse
Affiliation(s)
- M Cataldi
- Divisione di Farmacologia, Dipartimento di Neuroscienze, Facoltà di Medicina e Chirurgia, Università di Napoli Federico II, Naples, Italy
| | | | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Hildebrand ME, David LS, Hamid J, Mulatz K, Garcia E, Zamponi GW, Snutch TP. Selective Inhibition of Cav3.3 T-type Calcium Channels by Gαq/11-coupled Muscarinic Acetylcholine Receptors. J Biol Chem 2007; 282:21043-55. [PMID: 17535809 DOI: 10.1074/jbc.m611809200] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
T-type calcium channels play critical roles in controlling neuronal excitability, including the generation of complex spiking patterns and the modulation of synaptic plasticity, although the mechanisms and extent to which T-type Ca(2+) channels are modulated by G-protein-coupled receptors (GPCRs) remain largely unexplored. To examine specific interactions between T-type Ca(2+) channel subtypes and muscarinic acetylcholine receptors (mAChRS), the Cav3.1 (alpha(1G)), Cav3.2 (alpha(1H)), and Cav3.3 (alpha) T-type Ca(2+)(1I)channels were co-expressed with the M1 Galpha(q/11)-coupled mAChR. Perforated patch recordings demonstrate that activation of M1 receptors has a strong inhibitory effect on Cav3.3 T-type Ca(2+) currents but either no effect or a moderate stimulating effect on Cav3.1 and Cav3.2 peak current amplitudes. This differential modulation was observed for both rat and human T-type Ca(2+) channel variants. The inhibition of Cav3.3 channels by M1 receptors is reversible, use-independent, and associated with a concomitant increase in inactivation kinetics. Loss-of-function experiments with genetically encoded antagonists of Galpha and Gbetagamma proteins and gain-of-function experiments with genetically encoded Galpha subtypes indicate that M1 receptor-mediated inhibition of Cav3.3 occurs through Galpha(q/11). This is supported by experiments showing that activation of the M3 and M5 Galpha(q/11)-coupled mAChRs also causes inhibition of Cav3.3 currents, although Galpha(i)-coupled mAChRs (M2 and M4) have no effect. Examining Cav3.1-Cav3.3 chimeric channels demonstrates that two distinct regions of the Cav3.3 channel are necessary and sufficient for complete M1 receptor-mediated channel inhibition and represent novel sites not previously implicated in T-type channel modulation.
Collapse
Affiliation(s)
- Michael E Hildebrand
- Michael Smith Laboratories, University of British Columbia, Vancouver, British Columbia V6T 1Z4
| | | | | | | | | | | | | |
Collapse
|
48
|
Zhou L, Zhao S, Nadim F. Neuromodulation of short-term synaptic dynamics examined in a mechanistic model based on kinetics of calcium currents. Neurocomputing 2007; 70:2050-2054. [PMID: 18516212 DOI: 10.1016/j.neucom.2006.10.084] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
Network plasticity arises in large part due to the effects of exogenous neuromodulators. We investigate the neuromodulatory effects on short-term synaptic dynamics. The synapse from the lateral pyloric (LP) to the pyloric dilator (PD) neuron in the pyloric network of the crab C. borealis has both spike-mediated and non-spike-mediated (graded) components. Previous studies have shown that the graded component of this synapse exhibits short-term depression. Recent results from our lab indicate that in the presence of neuromodulatory peptide proctolin, low-amplitude presynaptic stimuli switch the short-term dynamics of this graded component from depression to facilitation. In this study, we show that this facilitation is correlated with the activation of a presynaptic inward current that is blocked by Mn(2+) suggesting that it is a slowly-accumulating Ca(2+) current. We modify a mechanistic model of synaptic release by assuming that the low-voltage-activating Ca(2+) current in our system is composed of two currents with fast (I(CaF)) and slow (I(CaS)) kinetics. We show that if proctolin adjusts the activation rate of I(CaS), this leads to accumulation of local intracellular Ca(2+) in response to multiple presynaptic voltage stimuli which, in turn, results in synaptic facilitation. Additionally, we assume that proctolin increases the maximal conductances of Ca(2+) currents in the model, consistent with the increased synaptic release found in the experiments. We find that these two presynaptic actions of proctolin in the model are sufficient to describe its actions on the short-term dynamics of the LP to PD synapse.
Collapse
Affiliation(s)
- Lian Zhou
- Department of Biological Sciences, Rutgers University, 101 Warren St., Newark, NJ 07102, USA
| | | | | |
Collapse
|
49
|
Joksovic PM, Doctor A, Gaston B, Todorovic SM. Functional regulation of T-type calcium channels by s-nitrosothiols in the rat thalamus. J Neurophysiol 2007; 97:2712-21. [PMID: 17287440 DOI: 10.1152/jn.00926.2006] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Although T-type Ca(2+) channels in the reticular thalamic nucleus (nRT) have a central function in tuning neuronal excitability and are implicated in sensory processing, sleep, and epilepsy, the mechanisms involved in their regulation are poorly understood. Here we recorded T-type Ca(2+) currents from intact nRT neurons in brain slices from young rats and investigated the mechanisms of T-type channel modulation by S-nitrosothiols (SNOs). We found that extracellular application of S-nitrosoglutathione (GSNO), S-nitrosocysteine (CSNO) and S-nitroso-N-acetyl-penicillamin (SNAP) rapidly and reversibly reduced T-type currents. The effects of SNOs are strongly stereoselective at physiological concentrations: (L)-CSNO was fourfold more effective in inhibiting T-type current than was (D)-CSNO. The effects of GSNO were abolished if cells had been treated with free hemoglobin or N-ethylmaleimide, an irreversible alkylating agent but not by 8-bromoguanosine-3',5'-cyclomonophosphate sodium salt, a membrane-permeant cGMP analogue or 1H-(1,2,4) oxadiazolo (4,3-a) quinoxalin-1-one, a specific soluble guanylyl cyclase inhibitor. In addition, bath applications of GSNO inhibited T-type currents in nucleated outside-out patches and whole cell recordings to a similar extent, with minimal effect on cell-attached recordings, suggesting a direct effect of GSNO on putative extracellular thiol residues on T-type channels. Biophysical studies indicate that GSNO decreased the availability of T-type channels at physiological potentials by modifying gating and stabilizing inactive states of the channels. In current-clamp experiments, GSNO diminished the amplitude of low-threshold calcium spikes and frequency of spike firing with minimal effects on the passive membrane properties. Collectively, the results indicate that SNOs may be a class of endogenous agents that control the functional states of the thalamus.
Collapse
Affiliation(s)
- Pavle M Joksovic
- Dept. of Anesthesiology, University of Virginia Health System, Mail Box 800710, Charlottesville, VA 22908-0710, USA
| | | | | | | |
Collapse
|
50
|
McKay BE, McRory JE, Molineux ML, Hamid J, Snutch TP, Zamponi GW, Turner RW. CaV3 T-type calcium channel isoforms differentially distribute to somatic and dendritic compartments in rat central neurons. Eur J Neurosci 2006; 24:2581-94. [PMID: 17100846 DOI: 10.1111/j.1460-9568.2006.05136.x] [Citation(s) in RCA: 155] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Spike output in many neuronal cell types is affected by low-voltage-activated T-type calcium currents arising from the Ca(v)3.1, Ca(v)3.2 and Ca(v)3.3 channel subtypes and their splice isoforms. The contributions of T-type current to cell output is often proposed to reflect a differential distribution of channels to somatic and dendritic compartments, but the subcellular distribution of the various rat T-type channel isoforms has not been fully determined. We used subtype-specific Ca(v)3 polyclonal antibodies to determine their distribution in key regions of adult Sprague-Dawley rat brain thought to exhibit T-type channel expression, and in particular, dendritic low-voltage-activated responses. We found a selective subcellular distribution of Ca(v)3 channel proteins in cell types of the neocortex and hippocampus, thalamus, and cerebellar input and output neurons. In general, the Ca(v)3.1 T-type channel immunolabel is prominent in the soma/proximal dendritic region and Ca(v)3.2 immunolabel in the soma and proximal-mid dendrites. Ca(v)3.3 channels are distinct in distributing to the soma and over extended lengths of the dendritic arbor of particular cell types. Ca(v)3 distribution overlaps with cell types previously established to exhibit rebound burst discharge as well as those not recognized for this activity. Additional immunolabel in the region of the nucleus in particular cell types was verified as corresponding to Ca(v)3 antigen through analysis of isolated protein fractions. These results provide evidence that different Ca(v)3 channel isoforms may contribute to low-voltage-activated calcium-dependent responses at the somatic and dendritic level, and the potential for T-type calcium channels to contribute to multiple aspects of neuronal activity.
Collapse
Affiliation(s)
- Bruce E McKay
- Hotchkiss Brain Institute, University of Calgary, 3330 Hospital Dr, N.W., Calgary, Alberta, T2N 4N1, Canada
| | | | | | | | | | | | | |
Collapse
|