1
|
Wang D, Herzig V, Dekan Z, Rosengren KJ, Payne CD, Hasan MM, Zhuang J, Bourinet E, Ragnarsson L, Alewood PF, Lewis RJ. Novel Scorpion Toxin ω-Buthitoxin-Hf1a Selectively Inhibits Calcium Influx via Ca V3.3 and Ca V3.2 and Alleviates Allodynia in a Mouse Model of Acute Postsurgical Pain. Int J Mol Sci 2024; 25:4745. [PMID: 38731963 PMCID: PMC11084959 DOI: 10.3390/ijms25094745] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Revised: 04/23/2024] [Accepted: 04/24/2024] [Indexed: 05/13/2024] Open
Abstract
Venom peptides have evolved to target a wide range of membrane proteins through diverse mechanisms of action and structures, providing promising therapeutic leads for diseases, including pain, epilepsy, and cancer, as well as unique probes of ion channel structure-function. In this work, a high-throughput FLIPR window current screening assay on T-type CaV3.2 guided the isolation of a novel peptide named ω-Buthitoxin-Hf1a from scorpion Hottentotta franzwerneri crude venom. At only 10 amino acid residues with one disulfide bond, it is not only the smallest venom peptide known to target T-type CaVs but also the smallest structured scorpion venom peptide yet discovered. Synthetic Hf1a peptides were prepared with C-terminal amidation (Hf1a-NH2) or a free C-terminus (Hf1a-OH). Electrophysiological characterization revealed Hf1a-NH2 to be a concentration-dependent partial inhibitor of CaV3.2 (IC50 = 1.18 μM) and CaV3.3 (IC50 = 0.49 μM) depolarized currents but was ineffective at CaV3.1. Hf1a-OH did not show activity against any of the three T-type subtypes. Additionally, neither form showed activity against N-type CaV2.2 or L-type calcium channels. The three-dimensional structure of Hf1a-NH2 was determined using NMR spectroscopy and used in docking studies to predict its binding site at CaV3.2 and CaV3.3. As both CaV3.2 and CaV3.3 have been implicated in peripheral pain signaling, the analgesic potential of Hf1a-NH2 was explored in vivo in a mouse model of incision-induced acute post-surgical pain. Consistent with this role, Hf1a-NH2 produced antiallodynia in both mechanical and thermal pain.
Collapse
Affiliation(s)
- Dan Wang
- Department of Chinese Medicine and Pharmacy, School of Pharmacy, Jiangsu University, Zhenjiang 212013, China;
- Division of Chemistry and Structural Biology, Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD 4072, Australia (L.R.); (P.F.A.)
| | - Volker Herzig
- Centre for Bioinnovation, University of the Sunshine Coast, Sippy Downs, QLD 4556, Australia;
- School of Science, Technology and Engineering, University of the Sunshine Coast, Sippy Downs, QLD 4556, Australia
| | - Zoltan Dekan
- Division of Chemistry and Structural Biology, Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD 4072, Australia (L.R.); (P.F.A.)
| | - K. Johan Rosengren
- School of Biomedical Sciences, The University of Queensland, Brisbane, QLD 4072, Australia; (K.J.R.); (C.D.P.)
| | - Colton D. Payne
- School of Biomedical Sciences, The University of Queensland, Brisbane, QLD 4072, Australia; (K.J.R.); (C.D.P.)
| | - Md. Mahadhi Hasan
- Pharmacy Discipline, Life Science School, Khulna University, Khulna 9208, Bangladesh;
| | - Jiajie Zhuang
- Department of Chinese Medicine and Pharmacy, School of Pharmacy, Jiangsu University, Zhenjiang 212013, China;
| | - Emmanuel Bourinet
- Institute of Functional Genomics, Montpellier University, CNRS, INSERM, 34090 Montpellier, France;
| | - Lotten Ragnarsson
- Division of Chemistry and Structural Biology, Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD 4072, Australia (L.R.); (P.F.A.)
| | - Paul F. Alewood
- Division of Chemistry and Structural Biology, Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD 4072, Australia (L.R.); (P.F.A.)
| | - Richard J. Lewis
- Division of Chemistry and Structural Biology, Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD 4072, Australia (L.R.); (P.F.A.)
| |
Collapse
|
2
|
Weiss N, Zamponi GW. The T-type calcium channelosome. Pflugers Arch 2024; 476:163-177. [PMID: 38036777 DOI: 10.1007/s00424-023-02891-z] [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: 10/27/2023] [Revised: 11/21/2023] [Accepted: 11/23/2023] [Indexed: 12/02/2023]
Abstract
T-type calcium channels perform crucial physiological roles across a wide spectrum of tissues, spanning both neuronal and non-neuronal system. For instance, they serve as pivotal regulators of neuronal excitability, contribute to cardiac pacemaking, and mediate the secretion of hormones. These functions significantly hinge upon the intricate interplay of T-type channels with interacting proteins that modulate their expression and function at the plasma membrane. In this review, we offer a panoramic exploration of the current knowledge surrounding these T-type channel interactors, and spotlight certain aspects of their potential for drug-based therapeutic intervention.
Collapse
Affiliation(s)
- Norbert Weiss
- Department of Pathophysiology, Third Faculty of Medicine, Charles University, Prague, Czech Republic.
| | - Gerald W Zamponi
- Department of Clinical Neurosciences, Alberta Children's Hospital Research Institute, Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Calgary, Canada
| |
Collapse
|
3
|
Mustafá ER, Weiß K, Weiss N. Secretory carrier-associated membrane protein 5 regulates cell-surface targeting of T-type calcium channels. Channels (Austin) 2023; 17:2230776. [PMID: 37389974 PMCID: PMC10316736 DOI: 10.1080/19336950.2023.2230776] [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: 05/03/2023] [Revised: 06/15/2023] [Accepted: 06/21/2023] [Indexed: 07/02/2023] Open
Abstract
Missense mutations in the human secretary carrier-associated membrane protein 5 (SCAMP5) cause a variety of neurological disorders including neurodevelopmental delay, epilepsy, and Parkinson's disease. We recently documented the importance of SCAMP2 in the regulation of T-type calcium channel expression in the plasma membrane. Here, we show that similar to SCAMP2, the co-expression of SCAMP5 in tsA-201 cells expressing recombinant Cav3.1, Cav3.2, and Cav3.3 channels nearly abolished whole-cell T-type currents. Recording of intramembrane charge movements revealed that SCAMP5-induced inhibition of T-type currents is primarily caused by the reduced expression of functional channels in the plasma membrane. Moreover, we show that SCAMP5-mediated downregulation of Cav3.2 channels is essentially preserved with disease-causing SCAMP5 R91W and G180W mutations. Hence, this study extends our previous findings with SCAMP2 and indicates that SCAMP5 also contributes to repressing the expression of T-type channels in the plasma membrane.
Collapse
Affiliation(s)
- Emilio R. Mustafá
- Department of Pathophysiology, Third Faculty of Medicine, Charles University, Prague, Czech Republic
- Electrophysiology Laboratory of the Multidisciplinary Institute of Cell Biology (Argentine Research Council CONICET, Scientific Research Commission of the Buenos Aires Province and National University of La Plata, La Plata, Buenos Aires, Argentina
| | - Konstantin Weiß
- Department of Pathophysiology, Third Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Norbert Weiss
- Department of Pathophysiology, Third Faculty of Medicine, Charles University, Prague, Czech Republic
| |
Collapse
|
4
|
Debreczeni D, Baukál D, Pergel E, Veres I, Czirják G. Critical contribution of the intracellular C-terminal region to TRESK channel activity is revealed by the epithelial Na + current ratio (ENaR) method. J Biol Chem 2023; 299:104737. [PMID: 37084812 DOI: 10.1016/j.jbc.2023.104737] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2023] [Revised: 04/15/2023] [Accepted: 04/17/2023] [Indexed: 04/23/2023] Open
Abstract
TRESK (K2P18.1) possesses unique structural proportions within the K2P background potassium channel family. The previously described TRESK regulatory mechanisms are based on the long intracellular loop between the second and third transmembrane segments (TMS). However, the functional significance of the exceptionally short intracellular C-terminal region (iCtr) following the fourth TMS has not yet been examined. In the present study, we investigated TRESK constructs modified at the iCtr by two-electrode voltage clamp and the newly developed epithelial sodium current ratio (ENaR) method in Xenopus oocytes. The ENaR method allowed the evaluation of channel activity by exclusively using electrophysiology, and provided data that are otherwise not readily available under whole-cell conditions. TRESK homodimer was connected with two ENaC (epithelial Na+ channel) heterotrimers and the Na+ current was measured as an internal reference, proportional to the number of channels in the plasma membrane. Modifications of TRESK iCtr resulted in diverse functional effects, indicating a complex contribution of this region to K+ channel activity. Mutations of positive residues in proximal iCtr locked TRESK in a low activity, calcineurin-insensitive state, although this phosphatase binds to distant motifs in the loop region. Accordingly, mutations in proximal iCtr may prevent the transmission of modulation to the gating machinery. Replacing distal iCtr with a sequence designed to interact with the inner surface of the plasma membrane increased the activity of the channel to unprecedented levels, as indicated by ENaR and single channel measurements. In conclusion, the distal iCtr is a major positive determinant of TRESK function.
Collapse
Affiliation(s)
| | - Dóra Baukál
- Department of Physiology, Semmelweis University, Budapest, Hungary
| | - Enikő Pergel
- Department of Physiology, Semmelweis University, Budapest, Hungary
| | - Irén Veres
- Department of Physiology, Semmelweis University, Budapest, Hungary
| | - Gábor Czirják
- Department of Physiology, Semmelweis University, Budapest, Hungary.
| |
Collapse
|
5
|
Zong P, Yue L. Regulation of Presynaptic Calcium Channels. ADVANCES IN NEUROBIOLOGY 2023; 33:171-202. [PMID: 37615867 DOI: 10.1007/978-3-031-34229-5_7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/25/2023]
Abstract
Voltage-gated calcium channels (VGCCs), especially Cav2.1 and Cav2.2, are the major mediators of Ca2+ influx at the presynaptic membrane in response to neuron excitation, thereby exerting a predominant control on synaptic transmission. To guarantee the timely and precise release of neurotransmitters at synapses, the activity of presynaptic VGCCs is tightly regulated by a variety of factors, including auxiliary subunits, membrane potential, G protein-coupled receptors (GPCRs), calmodulin (CaM), Ca2+-binding proteins (CaBP), protein kinases, various interacting proteins, alternative splicing events, and genetic variations.
Collapse
Affiliation(s)
- Pengyu Zong
- Department of Cell Biology, Calhoun Cardiology Center, University of Connecticut School of Medicine, Farmington, CT, USA
| | - Lixia Yue
- Department of Cell Biology, Calhoun Cardiology Center, University of Connecticut School of Medicine, Farmington, CT, USA.
| |
Collapse
|
6
|
El Ghaleb Y, Flucher BE. Ca V3.3 Channelopathies. Handb Exp Pharmacol 2023; 279:263-288. [PMID: 36592228 DOI: 10.1007/164_2022_631] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
CaV3.3 is the third member of the low-voltage-activated calcium channel family and the last to be recognized as disease gene. Previously, CACNA1I, the gene encoding CaV3.3, had been described as schizophrenia risk gene. More recently, de novo missense mutations in CACNA1I were identified in patients with variable degrees of neurodevelopmental disease with and without epilepsy. Their functional characterization indicated gain-of-function effects resulting in increased calcium load and hyperexcitability of neurons expressing CaV3.3. The amino acids mutated in the CaV3.3 disease variants are located in the vicinity of the channel's activation gate and thus are classified as gate-modifying channelopathy mutations. A persistent calcium leak during rest and prolonged calcium spikes due to increased voltage sensitivity of activation and slowed kinetics of channel inactivation, respectively, may be causal for the neurodevelopmental defects. The prominent expression of CaV3.3 in thalamic reticular nucleus neurons and its essential role in generating the rhythmic thalamocortical network activity are consistent with a role of the mutated channels in the etiology of epileptic seizures and thus suggest T-type channel blockers as a viable treatment option.
Collapse
Affiliation(s)
- Yousra El Ghaleb
- Institute of Physiology, Medical University Innsbruck, Innsbruck, Austria
| | - Bernhard E Flucher
- Institute of Physiology, Medical University Innsbruck, Innsbruck, Austria.
| |
Collapse
|
7
|
Mäki-Marttunen T, Mäki-Marttunen V. Excitatory and inhibitory effects of HCN channel modulation on excitability of layer V pyramidal cells. PLoS Comput Biol 2022; 18:e1010506. [PMID: 36099307 PMCID: PMC9506642 DOI: 10.1371/journal.pcbi.1010506] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Revised: 09/23/2022] [Accepted: 08/19/2022] [Indexed: 11/19/2022] Open
Abstract
Dendrites of cortical pyramidal cells are densely populated by hyperpolarization-activated cyclic nucleotide-gated (HCN) channels, a.k.a. Ih channels. Ih channels are targeted by multiple neuromodulatory pathways, and thus are one of the key ion-channel populations regulating the pyramidal cell activity. Previous observations and theories attribute opposing effects of the Ih channels on neuronal excitability due to their mildly hyperpolarized reversal potential. These effects are difficult to measure experimentally due to the fine spatiotemporal landscape of the Ih activity in the dendrites, but computational models provide an efficient tool for studying this question in a reduced but generalizable setting. In this work, we build upon existing biophysically detailed models of thick-tufted layer V pyramidal cells and model the effects of over- and under-expression of Ih channels as well as their neuromodulation. We show that Ih channels facilitate the action potentials of layer V pyramidal cells in response to proximal dendritic stimulus while they hinder the action potentials in response to distal dendritic stimulus at the apical dendrite. We also show that the inhibitory action of the Ih channels in layer V pyramidal cells is due to the interactions between Ih channels and a hot zone of low voltage-activated Ca2+ channels at the apical dendrite. Our simulations suggest that a combination of Ih-enhancing neuromodulation at the proximal part of the apical dendrite and Ih-inhibiting modulation at the distal part of the apical dendrite can increase the layer V pyramidal excitability more than either of the two alone. Our analyses uncover the effects of Ih-channel neuromodulation of layer V pyramidal cells at a single-cell level and shed light on how these neurons integrate information and enable higher-order functions of the brain.
Collapse
Affiliation(s)
- Tuomo Mäki-Marttunen
- Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
- Department of Biosciences, University of Oslo, Oslo, Norway
- Simula Research Laboratory, Oslo, Norway
- * E-mail:
| | - Verónica Mäki-Marttunen
- Cognitive Psychology Unit, Faculty of Social Sciences, University of Leiden, Leiden, Netherlands
| |
Collapse
|
8
|
King DR, Sedovy MW, Eaton X, Dunaway LS, Good ME, Isakson BE, Johnstone SR. Cell-To-Cell Communication in the Resistance Vasculature. Compr Physiol 2022; 12:3833-3867. [PMID: 35959755 DOI: 10.1002/cphy.c210040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
The arterial vasculature can be divided into large conduit arteries, intermediate contractile arteries, resistance arteries, arterioles, and capillaries. Resistance arteries and arterioles primarily function to control systemic blood pressure. The resistance arteries are composed of a layer of endothelial cells oriented parallel to the direction of blood flow, which are separated by a matrix layer termed the internal elastic lamina from several layers of smooth muscle cells oriented perpendicular to the direction of blood flow. Cells within the vessel walls communicate in a homocellular and heterocellular fashion to govern luminal diameter, arterial resistance, and blood pressure. At rest, potassium currents govern the basal state of endothelial and smooth muscle cells. Multiple stimuli can elicit rises in intracellular calcium levels in either endothelial cells or smooth muscle cells, sourced from intracellular stores such as the endoplasmic reticulum or the extracellular space. In general, activation of endothelial cells results in the production of a vasodilatory signal, usually in the form of nitric oxide or endothelial-derived hyperpolarization. Conversely, activation of smooth muscle cells results in a vasoconstriction response through smooth muscle cell contraction. © 2022 American Physiological Society. Compr Physiol 12: 1-35, 2022.
Collapse
Affiliation(s)
- D Ryan King
- Fralin Biomedical Research Institute at Virginia Tech Carilion, Center for Vascular and Heart Research, Virginia Tech, Roanoke, Virginia, USA
| | - Meghan W Sedovy
- Fralin Biomedical Research Institute at Virginia Tech Carilion, Center for Vascular and Heart Research, Virginia Tech, Roanoke, Virginia, USA.,Translational Biology, Medicine, and Health Graduate Program, Virginia Tech, Blacksburg, Virginia, USA
| | - Xinyan Eaton
- Fralin Biomedical Research Institute at Virginia Tech Carilion, Center for Vascular and Heart Research, Virginia Tech, Roanoke, Virginia, USA
| | - Luke S Dunaway
- Robert M. Berne Cardiovascular Research Centre, University of Virginia School of Medicine, Charlottesville, Virginia, USA
| | - Miranda E Good
- Molecular Cardiology Research Institute, Tufts Medical Center, Boston, Massachusetts, USA
| | - Brant E Isakson
- Robert M. Berne Cardiovascular Research Centre, University of Virginia School of Medicine, Charlottesville, Virginia, USA.,Department of Molecular Physiology and Biophysics, University of Virginia School of Medicine, Charlottesville, Virginia, USA
| | - Scott R Johnstone
- Fralin Biomedical Research Institute at Virginia Tech Carilion, Center for Vascular and Heart Research, Virginia Tech, Roanoke, Virginia, USA.,Department of Biological Sciences, Virginia Tech, Blacksburg, Virginia, USA
| |
Collapse
|
9
|
Ferron L, Guderyan SD, Smith EJ, Zamponi GW. CaVβ-subunit dependence of forward and reverse trafficking of CaV1.2 calcium channels. Mol Brain 2022; 15:43. [PMID: 35534894 PMCID: PMC9082888 DOI: 10.1186/s13041-022-00930-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2022] [Accepted: 05/02/2022] [Indexed: 11/10/2022] Open
Abstract
AbstractAuxiliary CaVβ subunits interact with the pore forming CaVα1 subunit to promote the plasma membrane expression of high voltage-activated calcium channels and to modulate the biophysical properties of Ca2+ currents. However, the effect of CaVβ subunits on channel trafficking to and from the plasma membrane is still controversial. Here, we have investigated the impact of CaVβ1b and CaVβ2a subunits on plasma membrane trafficking of CaV1.2 using a live-labeling strategy. We show that the CaVβ1b subunit is more potent in increasing CaV1.2 expression at the plasma membrane than the CaVβ2a subunit and that this effect is not related to modification of intracellular trafficking of the channel (i.e. neither forward trafficking, nor recycling, nor endocytosis). We conclude that the differential effect of CaVβ subunit subtypes on CaV1.2 surface expression is likely due to their differential ability to protect CaV1.2 from degradation.
Collapse
|
10
|
Dixon RE, Navedo MF, Binder MD, Santana LF. Mechanisms and Physiological Implications of Cooperative Gating of Ion Channels Clusters. Physiol Rev 2021; 102:1159-1210. [PMID: 34927454 DOI: 10.1152/physrev.00022.2021] [Citation(s) in RCA: 59] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Ion channels play a central role in the regulation of nearly every cellular process. Dating back to the classic 1952 Hodgkin-Huxley model of the generation of the action potential, ion channels have always been thought of as independent agents. A myriad of recent experimental findings exploiting advances in electrophysiology, structural biology, and imaging techniques, however, have posed a serious challenge to this long-held axiom as several classes of ion channels appear to open and close in a coordinated, cooperative manner. Ion channel cooperativity ranges from variable-sized oligomeric cooperative gating in voltage-gated, dihydropyridine-sensitive Cav1.2 and Cav1.3 channels to obligatory dimeric assembly and gating of voltage-gated Nav1.5 channels. Potassium channels, transient receptor potential channels, hyperpolarization cyclic nucleotide-activated channels, ryanodine receptors (RyRs), and inositol trisphosphate receptors (IP3Rs) have also been shown to gate cooperatively. The implications of cooperative gating of these ion channels range from fine tuning excitation-contraction coupling in muscle cells to regulating cardiac function and vascular tone, to modulation of action potential and conduction velocity in neurons and cardiac cells, and to control of pace-making activity in the heart. In this review, we discuss the mechanisms leading to cooperative gating of ion channels, their physiological consequences and how alterations in cooperative gating of ion channels may induce a range of clinically significant pathologies.
Collapse
Affiliation(s)
- Rose Ellen Dixon
- Department of Physiology and Membrane Biology, University of California, Davis, CA, United States
| | - Manuel F Navedo
- Department of Pharmacology, University of California, Davis, CA, United States
| | - Marc D Binder
- Department of Physiology and Biophysics, University of Washington, Seattle, WA, United States
| | - L Fernando Santana
- Department of Physiology and Membrane Biology, University of California, Davis, CA, United States
| |
Collapse
|
11
|
Iacovides S, Kamerman P, Baker FC, Mitchell D. Why It Is Important to Consider the Effects of Analgesics on Sleep: A Critical Review. Compr Physiol 2021; 11:2589-2619. [PMID: 34558668 DOI: 10.1002/cphy.c210006] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
We review the known physiological mechanisms underpinning all of pain processing, sleep regulation, and pharmacology of analgesics prescribed for chronic pain. In particular, we describe how commonly prescribed analgesics act in sleep-wake neural pathways, with potential unintended impact on sleep and/or wake function. Sleep disruption, whether pain- or drug-induced, negatively impacts quality of life, mental and physical health. In the context of chronic pain, poor sleep quality heightens pain sensitivity and may affect analgesic function, potentially resulting in further analgesic need. Clinicians already have to consider factors including efficacy, abuse potential, and likely side effects when making analgesic prescribing choices. We propose that analgesic-related sleep disruption should also be considered. The neurochemical mechanisms underlying the reciprocal relationship between pain and sleep are poorly understood, and studies investigating sleep in those with specific chronic pain conditions (including those with comorbidities) are lacking. We emphasize the importance of further work to clarify the effects (intended and unintended) of each analgesic class to inform personalized treatment decisions in patients with chronic pain. © 2021 American Physiological Society. Compr Physiol 11:1-31, 2021.
Collapse
Affiliation(s)
- Stella Iacovides
- Brain Function Research Group, School of Physiology, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Peter Kamerman
- Brain Function Research Group, School of Physiology, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Fiona C Baker
- Brain Function Research Group, School of Physiology, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa.,Human Sleep Research Program, SRI International, Menlo Park, California, USA
| | - Duncan Mitchell
- Brain Function Research Group, School of Physiology, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| |
Collapse
|
12
|
Evaluating the impact of gabapentinoids on sleep health in patients with chronic neuropathic pain: a systematic review and meta-analysis. Pain 2021; 161:476-490. [PMID: 31693543 DOI: 10.1097/j.pain.0000000000001743] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Chronic neuropathic pain (NP) is debilitating and impacts sleep health and quality of life. Treatment with gabapentinoids (GBs) has been shown to reduce pain, but its effects on sleep health have not been systematically evaluated. The objective of this systematic review and meta-analysis was to assess the relationship between GB therapy dose and duration on sleep quality, daytime somnolence, and intensity of pain in patients with NP. Subgroup comparisons were planned for high- vs low-dose GBs, where 300 mg per day or more of pregabalin was used to classify high-dose therapy. Trial data were segregated by duration less than 6 weeks and 6 weeks or greater. Twenty randomized controlled trials were included. Primary outcome measures included pain-related sleep interference and incidence of daytime somnolence. Secondary outcomes included daily pain scores (numerical rating scale 0-10) and patient global impression of change. Significant improvement in sleep quality was observed after 6 weeks of GB treatment when compared with placebo (standardized mean difference 0.39, 95% confidence interval 0.32-0.46 P < 0.001). Increased daytime somnolence was observed among all GB-treated groups when compared with placebo. Treated patients were also more likely to report improvement of patient global impression of change scores. Pain scores decreased significantly in patients both after 6 weeks of treatment (P < 0.001) and in trials less than 6 weeks (P = 0.017) when compared with placebo. Our data demonstrate that GBs have a positive impact on sleep health, quality of life, and pain in patients with NP syndromes. However, these benefits come at the expense of daytime somnolence.
Collapse
|
13
|
Lory P, Nicole S, Monteil A. Neuronal Cav3 channelopathies: recent progress and perspectives. Pflugers Arch 2020; 472:831-844. [PMID: 32638069 PMCID: PMC7351805 DOI: 10.1007/s00424-020-02429-7] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2020] [Revised: 06/08/2020] [Accepted: 06/26/2020] [Indexed: 12/22/2022]
Abstract
T-type, low-voltage activated, calcium channels, now designated Cav3 channels, are involved in a wide variety of physiological functions, especially in nervous systems. Their unique electrophysiological properties allow them to finely regulate neuronal excitability and to contribute to sensory processing, sleep, and hormone and neurotransmitter release. In the last two decades, genetic studies, including exploration of knock-out mouse models, have greatly contributed to elucidate the role of Cav3 channels in normal physiology, their regulation, and their implication in diseases. Mutations in genes encoding Cav3 channels (CACNA1G, CACNA1H, and CACNA1I) have been linked to a variety of neurodevelopmental, neurological, and psychiatric diseases designated here as neuronal Cav3 channelopathies. In this review, we describe and discuss the clinical findings and supporting in vitro and in vivo studies of the mutant channels, with a focus on de novo, gain-of-function missense mutations recently discovered in CACNA1G and CACNA1H. Overall, the studies of the Cav3 channelopathies help deciphering the pathogenic mechanisms of corresponding diseases and better delineate the properties and physiological roles Cav3 channels.
Collapse
Affiliation(s)
- Philippe Lory
- Institut de Génomique Fonctionnelle, CNRS, INSERM, University Montpellier, 141, rue de la Cardonille, 34094, Montpellier, France. .,LabEx 'Ion Channel Science and Therapeutics' (ICST), Montpellier, France.
| | - Sophie Nicole
- Institut de Génomique Fonctionnelle, CNRS, INSERM, University Montpellier, 141, rue de la Cardonille, 34094, Montpellier, France.,LabEx 'Ion Channel Science and Therapeutics' (ICST), Montpellier, France
| | - Arnaud Monteil
- Institut de Génomique Fonctionnelle, CNRS, INSERM, University Montpellier, 141, rue de la Cardonille, 34094, Montpellier, France.,LabEx 'Ion Channel Science and Therapeutics' (ICST), Montpellier, France
| |
Collapse
|
14
|
Carter MT, McMillan HJ, Tomin A, Weiss N. Compound heterozygous CACNA1H mutations associated with severe congenital amyotrophy. Channels (Austin) 2020; 13:153-161. [PMID: 31070086 PMCID: PMC6527065 DOI: 10.1080/19336950.2019.1614415] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023] Open
Abstract
Neuromuscular disorders encompass a wide range of conditions often associated with a genetic component. In the present study, we report a patient with severe infantile-onset amyotrophy in whom two compound heterozygous variants in the gene CACNA1H encoding for Cav3.2 T-type calcium channels were identified. Functional analysis of Cav3.2 variants revealed several alterations of the gating properties of the channel that were in general consistent with a loss-of-channel function. Taken together, these findings suggest that severe congenital amyoplasia may be related to CACNA1H and would represent a new phenotype associated with mutations in this gene.
Collapse
Affiliation(s)
- Melissa T Carter
- a Children's Hospital of Eastern Ontario Research Institute , University of Ottawa , Ottawa , Ontario , Canada
| | - Hugh J McMillan
- a Children's Hospital of Eastern Ontario Research Institute , University of Ottawa , Ottawa , Ontario , Canada
| | - Andriy Tomin
- b Institute of Organic Chemistry and Biochemistry , Czech Academy of Sciences , Prague , Czech Republic
| | - Norbert Weiss
- b Institute of Organic Chemistry and Biochemistry , Czech Academy of Sciences , Prague , Czech Republic
| |
Collapse
|
15
|
Stringer RN, Lazniewska J, Weiss N. Transcriptomic analysis of glycan-processing genes in the dorsal root ganglia of diabetic mice and functional characterization on Ca v3.2 channels. Channels (Austin) 2020; 14:132-140. [PMID: 32233724 PMCID: PMC7153791 DOI: 10.1080/19336950.2020.1745406] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
Cav3.2 T-type calcium channels play an essential role in the transmission of peripheral nociception in the dorsal root ganglia (DRG) and alteration of Cav3.2 expression is associated with the development of peripheral painful diabetic neuropathy (PDN). Several studies have previously documented the role of glycosylation in the expression and functioning of Cav3.2 and suggested that altered glycosylation of the channel may contribute to the aberrant expression of the channel in diabetic conditions. In this study, we aimed to analyze the expression of glycan-processing genes in DRG neurons from a leptin-deficient genetic mouse model of diabetes (db/db). Transcriptomic analysis revealed that several glycan-processing genes encoding for glycosyltransferases and sialic acid-modifying enzymes were upregulated in diabetic conditions. Functional analysis of these enzymes on recombinant Cav3.2 revealed an unexpected loss-of-function of the channel. Collectively, our data indicate that diabetes is associated with an alteration of the glycosylation machinery in DRG neurons. However, individual action of these enzymes when tested on recombinant Cav3.2 cannot explain the observed upregulation of T-type channels under diabetic conditions. Abbreviations: Galnt16: Polypeptide N-acetylgalactosaminyltransferase 16; B3gnt8: UDP-GlcNAc:betaGal beta-1,3-N-acetylglucosaminyltransferase 8; B4galt1: Beta-1,4-galactosyltransferase 1; St6gal1: Beta-galactoside alpha-2,6-sialyltransferase 1; Neu3: Sialidase-3
Collapse
Affiliation(s)
- Robin N Stringer
- Institute of Organic Chemistry and Biochemistry, Czech Academy of Sciences, Prague, Czech Republic.,Third Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Joanna Lazniewska
- Institute of Organic Chemistry and Biochemistry, Czech Academy of Sciences, Prague, Czech Republic
| | - Norbert Weiss
- Institute of Organic Chemistry and Biochemistry, Czech Academy of Sciences, Prague, Czech Republic
| |
Collapse
|
16
|
Stringer RN, Jurkovicova-Tarabova B, Huang S, Haji-Ghassemi O, Idoux R, Liashenko A, Souza IA, Rzhepetskyy Y, Lacinova L, Van Petegem F, Zamponi GW, Pamphlett R, Weiss N. A rare CACNA1H variant associated with amyotrophic lateral sclerosis causes complete loss of Ca v3.2 T-type channel activity. Mol Brain 2020; 13:33. [PMID: 32143681 PMCID: PMC7060640 DOI: 10.1186/s13041-020-00577-6] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2019] [Accepted: 03/02/2020] [Indexed: 12/13/2022] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a neurodegenerative disorder characterized by the progressive loss of cortical, brain stem and spinal motor neurons that leads to muscle weakness and death. A previous study implicated CACNA1H encoding for Cav3.2 calcium channels as a susceptibility gene in ALS. In the present study, two heterozygous CACNA1H variants were identified by whole genome sequencing in a small cohort of ALS patients. These variants were functionally characterized using patch clamp electrophysiology, biochemistry assays, and molecular modeling. A previously unreported c.454GTAC > G variant produced an inframe deletion of a highly conserved isoleucine residue in Cav3.2 (p.ΔI153) and caused a complete loss-of-function of the channel, with an additional dominant-negative effect on the wild-type channel when expressed in trans. In contrast, the c.3629C > T variant caused a missense substitution of a proline with a leucine (p.P1210L) and produced a comparatively mild alteration of Cav3.2 channel activity. The newly identified ΔI153 variant is the first to be reported to cause a complete loss of Cav3.2 channel function. These findings add to the notion that loss-of-function of Cav3.2 channels associated with rare CACNA1H variants may be risk factors in the complex etiology of ALS.
Collapse
Affiliation(s)
- Robin N. Stringer
- Institute of Organic Chemistry and Biochemistry, Czech Academy of Sciences, Flemingovo nam 2, 16610 Prague, Czech Republic
- Third Faculty of Medicine, Charles University, Prague, Czech Republic
| | | | - Sun Huang
- Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, Canada
| | - Omid Haji-Ghassemi
- Department of Biochemistry and Molecular Biology, University of British Columbia, Vancouver, Canada
| | - Romane Idoux
- Institute of Organic Chemistry and Biochemistry, Czech Academy of Sciences, Flemingovo nam 2, 16610 Prague, Czech Republic
| | - Anna Liashenko
- Institute of Organic Chemistry and Biochemistry, Czech Academy of Sciences, Flemingovo nam 2, 16610 Prague, Czech Republic
| | - Ivana A. Souza
- Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, Canada
| | - Yuriy Rzhepetskyy
- Institute of Organic Chemistry and Biochemistry, Czech Academy of Sciences, Flemingovo nam 2, 16610 Prague, Czech Republic
| | - Lubica Lacinova
- Center of Biosciences, Institute of Molecular Physiology and Genetics, Academy of Sciences, Bratislava, Slovakia
| | - Filip Van Petegem
- Department of Biochemistry and Molecular Biology, University of British Columbia, Vancouver, Canada
| | - Gerald W. Zamponi
- Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, Canada
| | - Roger Pamphlett
- Discipline of Pathology, Brain and Mind Centre, The University of Sydney, Sydney, NSW Australia
| | - Norbert Weiss
- Institute of Organic Chemistry and Biochemistry, Czech Academy of Sciences, Flemingovo nam 2, 16610 Prague, Czech Republic
| |
Collapse
|
17
|
A potential role for T-type calcium channels in homocysteinemia-induced peripheral neuropathy. Pain 2019; 160:2798-2810. [DOI: 10.1097/j.pain.0000000000001669] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
|
18
|
Qu YS, Lazzerini PE, Capecchi PL, Laghi-Pasini F, El Sherif N, Boutjdir M. Autoimmune Calcium Channelopathies and Cardiac Electrical Abnormalities. Front Cardiovasc Med 2019; 6:54. [PMID: 31119135 PMCID: PMC6507622 DOI: 10.3389/fcvm.2019.00054] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2018] [Accepted: 04/16/2019] [Indexed: 12/24/2022] Open
Abstract
Patients with autoimmune diseases are at increased risk for developing cardiovascular diseases, and abnormal electrocardiographic findings are common. Voltage-gated calcium channels play a major role in the cardiovascular system and regulate cardiac excitability and contractility. Particularly, by virtue of their localization and expression in the heart, calcium channels modulate pace making at the sinus node, conduction at the atrioventricular node and cardiac repolarization in the working myocardium. Consequently, emerging evidence suggests that calcium channels are targets to autoantibodies in autoimmune diseases. Autoimmune-associated cardiac calcium channelopathies have been recognized in both sinus node dysfunction atrioventricular block in patients positive for anti-Ro/La antibodies, and ventricular arrhythmias in patients with dilated cardiomyopathy. In this review, we discuss mechanisms of autoimmune-associated calcium channelopathies and their relationship with the development of cardiac electrical abnormalities.
Collapse
Affiliation(s)
- Yongxia Sarah Qu
- Department of Cardiology, New York Presbyterian Brooklyn Methodist Hospital, Brooklyn, NY, United States.,VA New York Harbor Healthcare System and State University of New York Downstate Medical Center, Brooklyn, NY, United States
| | - Pietro Enea Lazzerini
- Department of Medical Sciences, Surgery and Neurosciences, University of Siena, Siena, Italy
| | - Pier Leopoldo Capecchi
- Department of Medical Sciences, Surgery and Neurosciences, University of Siena, Siena, Italy
| | - Franco Laghi-Pasini
- Department of Medical Sciences, Surgery and Neurosciences, University of Siena, Siena, Italy
| | - Nabil El Sherif
- VA New York Harbor Healthcare System and State University of New York Downstate Medical Center, Brooklyn, NY, United States
| | - Mohamed Boutjdir
- VA New York Harbor Healthcare System and State University of New York Downstate Medical Center, Brooklyn, NY, United States.,NYU School of Medicine, New York, NY, United States
| |
Collapse
|
19
|
Calcium Channel Subunit α2δ4 Is Regulated by Early Growth Response 1 and Facilitates Epileptogenesis. J Neurosci 2019; 39:3175-3187. [PMID: 30792272 DOI: 10.1523/jneurosci.1731-18.2019] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2018] [Revised: 12/03/2018] [Accepted: 01/08/2019] [Indexed: 12/17/2022] Open
Abstract
Transient brain insults, including status epilepticus (SE), can trigger a period of epileptogenesis during which functional and structural reorganization of neuronal networks occurs resulting in the onset of focal epileptic seizures. In recent years, mechanisms that regulate the dynamic transcription of individual genes during epileptogenesis and thereby contribute to the development of a hyperexcitable neuronal network have been elucidated. Our own results have shown early growth response 1 (Egr1) to transiently increase expression of the T-type voltage-dependent Ca2+ channel (VDCC) subunit CaV3.2, a key proepileptogenic protein. However, epileptogenesis involves complex and dynamic transcriptomic alterations; and so far, our understanding of the transcriptional control mechanism of gene regulatory networks that act in the same processes is limited. Here, we have analyzed whether Egr1 acts as a key transcriptional regulator for genes contributing to the development of hyperexcitability during epileptogenesis. We found Egr1 to drive the expression of the VDCC subunit α2δ4, which was augmented early and persistently after pilocarpine-induced SE. Furthermore, we show that increasing levels of α2δ4 in the CA1 region of the hippocampus elevate seizure susceptibility of mice by slightly decreasing local network activity. Interestingly, we also detected increased expression levels of Egr1 and α2δ4 in human hippocampal biopsies obtained from epilepsy surgery. In conclusion, Egr1 controls the abundance of the VDCC subunits CaV3.2 and α2δ4, which act synergistically in epileptogenesis, and thereby contributes to a seizure-induced "transcriptional Ca2+ channelopathy."SIGNIFICANCE STATEMENT The onset of focal recurrent seizures often occurs after an epileptogenic process induced by transient insults to the brain. Recently, transcriptional control mechanisms for individual genes involved in converting neurons hyperexcitable have been identified, including early growth response 1 (Egr1), which activates transcription of the T-type Ca2+ channel subunit CaV3.2. Here, we find Egr1 to regulate also the expression of the voltage-dependent Ca2+ channel subunit α2δ4, which was augmented after pilocarpine- and kainic acid-induced status epilepticus. In addition, we observed that α2δ4 affected spontaneous network activity and the susceptibility for seizure induction. Furthermore, we detected corresponding dynamics in human biopsies from epilepsy patients. In conclusion, Egr1 orchestrates a seizure-induced "transcriptional Ca2+ channelopathy" consisting of CaV3.2 and α2δ4, which act synergistically in epileptogenesis.
Collapse
|
20
|
T-type Calcium Channels in Cancer. Cancers (Basel) 2019; 11:cancers11020134. [PMID: 30678110 PMCID: PMC6407089 DOI: 10.3390/cancers11020134] [Citation(s) in RCA: 50] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2018] [Revised: 01/10/2019] [Accepted: 01/17/2019] [Indexed: 12/21/2022] Open
Abstract
Although voltage-activated Ca2+ channels are a common feature in excitable cells, their expression in cancer tissue is less understood. T-type Ca2+ channels are particularly overexpressed in various cancers. Because of their activation profile at membrane potentials close to rest and the generation of a window current, T-type Ca2+ channels may regulate a variety of Ca2+-dependent cellular processes, including cell proliferation, survival, and differentiation. The expression of T-type Ca2+ channels is of special interest as a target for therapeutic interventions.
Collapse
|
21
|
Arteaga-Tlecuitl R, Sanchez-Sandoval AL, Ramirez-Cordero BE, Rosendo-Pineda MJ, Vaca L, Gomora JC. Increase of Ca V3 channel activity induced by HVA β1b-subunit is not mediated by a physical interaction. BMC Res Notes 2018; 11:810. [PMID: 30428904 PMCID: PMC6236959 DOI: 10.1186/s13104-018-3917-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2018] [Accepted: 11/09/2018] [Indexed: 01/13/2023] Open
Abstract
Objective Low voltage-activated (LVA) calcium channels are crucial for regulating oscillatory behavior in several types of neurons and other excitable cells. LVA channels dysfunction has been implicated in epilepsy, neuropathic pain, cancer, among other diseases. Unlike for High Voltage-Activated (HVA) channels, voltage-dependence and kinetics of currents carried by recombinant LVA, i.e., CaV3 channels, are quite similar to those observed in native currents. Therefore, whether these channels are regulated by HVA auxiliary subunits, remain controversial. Here, we used the α1-subunits of CaV3.1, CaV3.2, and CaV3.3 channels, together with HVA auxiliary β-subunits to perform electrophysiological, confocal microscopy and immunoprecipitation experiments, in order to further explore this possibility. Results Functional expression of CaV3 channels is up-regulated by all four β-subunits, although most consistent effects were observed with the β1b-subunit. The biophysical properties of CaV3 channels were not modified by any β-subunit. Furthermore, although β1b-subunits increased colocalization of GFP-tagged CaV3 channels and the plasma membrane of HEK-293 cells, western blots analysis revealed the absence of physical interaction between CaV3.3 and β1b-subunits as no co-immunoprecipitation was observed. These results provide solid evidence that the up-regulation of LVA channels in the presence of HVA-β1b subunit is not mediated by a high affinity interaction between both proteins. Electronic supplementary material The online version of this article (10.1186/s13104-018-3917-1) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Rogelio Arteaga-Tlecuitl
- Departamento de Neuropatología Molecular, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, 04510, Mexico City, Mexico
| | - Ana Laura Sanchez-Sandoval
- Departamento de Neuropatología Molecular, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, 04510, Mexico City, Mexico
| | - Belen Ernestina Ramirez-Cordero
- Departamento de Neuropatología Molecular, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, 04510, Mexico City, Mexico
| | - Margarita Jacaranda Rosendo-Pineda
- Departamento de Biología Celular y del Desarrollo, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, 04510, Mexico City, Mexico
| | - Luis Vaca
- Departamento de Biología Celular y del Desarrollo, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, 04510, Mexico City, Mexico
| | - Juan Carlos Gomora
- Departamento de Neuropatología Molecular, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, 04510, Mexico City, Mexico.
| |
Collapse
|
22
|
CACHD1 is an α2δ-Like Protein That Modulates Ca V3 Voltage-Gated Calcium Channel Activity. J Neurosci 2018; 38:9186-9201. [PMID: 30181139 DOI: 10.1523/jneurosci.3572-15.2018] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2015] [Revised: 06/03/2018] [Accepted: 06/13/2018] [Indexed: 11/21/2022] Open
Abstract
The putative cache (Ca2+ channel and chemotaxis receptor) domain containing 1 (CACHD1) protein has predicted structural similarities to members of the α2δ voltage-gated Ca2+ channel auxiliary subunit family. CACHD1 mRNA and protein were highly expressed in the male mammalian CNS, in particular in the thalamus, hippocampus, and cerebellum, with a broadly similar tissue distribution to CaV3 subunits, in particular CaV3.1. In expression studies, CACHD1 increased cell-surface localization of CaV3.1, and these proteins were in close proximity at the cell surface, consistent with the formation of CACHD1-CaV3.1 complexes. In functional electrophysiological studies, coexpression of human CACHD1 with CaV3.1, CaV3.2, and CaV3.3 caused a significant increase in peak current density and corresponding increases in maximal conductance. By contrast, α2δ-1 had no effect on peak current density or maximal conductance in CaV3.1, CaV3.2, or CaV3.3. A comparison of CACHD1-mediated increases in CaV3.1 current density and gating currents revealed an increase in channel open probability. In hippocampal neurons from male and female embryonic day 19 rats, CACHD1 overexpression increased CaV3-mediated action potential firing frequency and neuronal excitability. These data suggest that CACHD1 is structurally an α2δ-like protein that functionally modulates CaV3 voltage-gated calcium channel activity.SIGNIFICANCE STATEMENT This is the first study to characterize the Ca2+ channel and chemotaxis receptor domain containing 1 (CACHD1) protein. CACHD1 is widely expressed in the CNS, in particular in the thalamus, hippocampus, and cerebellum. CACHD1 distribution is similar to that of low voltage-activated (CaV3, T-type) calcium channels, in particular to CaV3.1, a protein that regulates neuronal excitability and is a potential therapeutic target in conditions such as epilepsy and pain. CACHD1 is structurally an α2δ-like protein that functionally increases CaV3 calcium current. CACHD1 increases the presence of CaV3.1 at the cell surface, forms complexes with CaV3.1 at the cell surface, and causes an increase in channel open probability. In hippocampal neurons, CACHD1 causes increases in neuronal firing. Thus, CACHD1 represents a novel protein that modulates CaV3 activity.
Collapse
|
23
|
Gong N, Park J, Luo ZD. Injury-induced maladaptation and dysregulation of calcium channel α 2 δ subunit proteins and its contribution to neuropathic pain development. Br J Pharmacol 2018; 175:2231-2243. [PMID: 28646556 PMCID: PMC5980513 DOI: 10.1111/bph.13930] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2017] [Revised: 06/05/2017] [Accepted: 06/12/2017] [Indexed: 01/12/2023] Open
Abstract
Voltage-gated calcium channels (VGCCs) play important roles in physiological functions including the modulation of neurotransmitter release, neuronal network activities, intracellular signalling pathways and gene expression. Some pathological conditions, including nerve injuries, can cause the dysregulation of VGCCs and their subunits. This in turn can lead to a functional maladaptation of VGCCs and their subunits, which can contribute to the development of disorders such as pain sensations. This review has summarized recent findings related to maladaptive changes in the dysregulated VGCC α2 δ1 subunit (Cav α2 δ1 ) with a focus on exploring the mechanisms underlying the contribution of Cav α2 δ1 to pain signal transduction. At least under neuropathic pain conditions, the dysregulated Cav α2 δ1 can modulate VGCC functions as well as other plasticity changes. The latter includes abnormal excitatory synaptogenesis resulting from its interactions with injury-induced extracellular matrix glycoprotein molecule thrombospondins, which is independent of the VGCC functions. Blocking Cav α2 δ1 with gabapentinoids can reverse neuropathic pain significantly with relatively mild side effects, but only in a small population of neuropathic pain patients due to reasons yet to be explored. There are emerging data suggesting that early preventive treatment with gabapentinoids can prevent aberrant excitatory synapse formation and the development of chronic pain. If these findings are confirmed clinically, this could be an attractive approach for neuropathic pain management. LINKED ARTICLES This article is part of a themed section on Recent Advances in Targeting Ion Channels to Treat Chronic Pain. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v175.12/issuetoc.
Collapse
Affiliation(s)
- Nian Gong
- Department of Anesthesiology & Perioperative CareSchool of Medicine, University of California IrvineIrvineCAUSA
| | - John Park
- Department of Pharmacology, School of MedicineUniversity of California IrvineIrvineCAUSA
| | - Z David Luo
- Department of Anesthesiology & Perioperative CareSchool of Medicine, University of California IrvineIrvineCAUSA
- Department of Pharmacology, School of MedicineUniversity of California IrvineIrvineCAUSA
| |
Collapse
|
24
|
Celli R, Santolini I, Guiducci M, van Luijtelaar G, Parisi P, Striano P, Gradini R, Battaglia G, Ngomba RT, Nicoletti F. The α2δ Subunit and Absence Epilepsy: Beyond Calcium Channels? Curr Neuropharmacol 2018; 15:918-925. [PMID: 28290248 PMCID: PMC5652034 DOI: 10.2174/1570159x15666170309105451] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2016] [Revised: 01/16/2017] [Accepted: 03/06/2017] [Indexed: 02/08/2023] Open
Abstract
Background: Spike-wave discharges, underlying absence seizures, are generated within a cortico-thalamo-cortical network that involves the somatosensory cortex, the reticular thalamic nucleus, and the ventrobasal thalamic nuclei. Activation of T-type voltage-sensitive calcium channels (VSCCs) contributes to the pathological oscillatory activity of this network, and some of the first-line drugs used in the treatment of absence epilepsy inhibit T-type calcium channels. The α2δ subunit is a component of high voltage-activated VSCCs (i.e., L-, N-, P/Q-, and R channels) and studies carried out in heterologous expression systems suggest that it may also associate with T channels. The α2δ subunit is also targeted by thrombospondins, which regulate synaptogenesis in the central nervous system. Objective: To discuss the potential role for the thrombospondin/α2δ axis in the pathophysiology of absence epilepsy. Methods: We searched PubMed articles for the terms “absence epilepsy”, “T-type voltage-sensitive calcium channels”, “α2δ subunit”, “ducky mice”, “pregabalin”, “gabapentin”, “thrombospondins”, and included papers focusing this Review's scope. Results: We moved from the evidence that mice lacking the α2δ-2 subunit show absence seizures and α2δ ligands (gabapentin and pregabalin) are detrimental in the treatment of absence epilepsy. This suggests that α2δ may be protective against absence epilepsy via a mechanism that does not involve T channels. We discuss the interaction between thrombospondins and α2δ and its potential relevance in the regulation of excitatory synaptic formation in the cortico-thalamo-cortical network. Conclusion: We speculate on the possibility that the thrombospondin/α2δ axis is critical for the correct functioning of the cortico-thalamo-cortical network, and that abnormalities in this axis may play a role in the pathophysiology of absence epilepsy.
Collapse
Affiliation(s)
- Roberta Celli
- I.R.C.C.S. Neuromed, Neuropharmacology Unit, Pozzilli, (IS), Italy
| | - Ines Santolini
- I.R.C.C.S. Neuromed, Neuropharmacology Unit, Pozzilli, (IS), Italy
| | - Michela Guiducci
- Departments of Neurosciences, Mental Health and Sensory Organs, Experimental Medicine, and Physiology and Pharmacology, University Sapienza, Rome, Italy
| | - Gilles van Luijtelaar
- Donders Centre for Cognition, Donders Institute for Brain, Cognition and Behaviour, Radboud University, Nijmegen. Netherlands
| | - Pasquale Parisi
- Departments of Neurosciences, Mental Health and Sensory Organs, Experimental Medicine, and Physiology and Pharmacology, University Sapienza, Rome, Italy
| | - Pasquale Striano
- Pediatric Neurology and Muscular Diseases Unit, Department of Neurosciences, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health, University of Genoa, "G. Gaslini" Institute, Genova, Italy
| | - Roberto Gradini
- I.R.C.C.S. Neuromed, Neuropharmacology Unit, Pozzilli, (IS), Italy
| | | | - Richard T Ngomba
- University of Lincoln, School of Pharmacy, Lincoln, United Kingdom
| | - Ferdinando Nicoletti
- Department of Physiology and Pharmacology, University Sapienza, Piazzale Aldo Moro, 5, 00185 Rome, Italy
| |
Collapse
|
25
|
Chemin J, Taiakina V, Monteil A, Piazza M, Guan W, Stephens RF, Kitmitto A, Pang ZP, Dolphin AC, Perez-Reyes E, Dieckmann T, Guillemette JG, Spafford JD. Calmodulin regulates Ca v3 T-type channels at their gating brake. J Biol Chem 2017; 292:20010-20031. [PMID: 28972185 PMCID: PMC5723990 DOI: 10.1074/jbc.m117.807925] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2017] [Revised: 09/19/2017] [Indexed: 01/10/2023] Open
Abstract
Calcium (Cav1 and Cav2) and sodium channels possess homologous CaM-binding motifs, known as IQ motifs in their C termini, which associate with calmodulin (CaM), a universal calcium sensor. Cav3 T-type channels, which serve as pacemakers of the mammalian brain and heart, lack a C-terminal IQ motif. We illustrate that T-type channels associate with CaM using co-immunoprecipitation experiments and single particle cryo-electron microscopy. We demonstrate that protostome invertebrate (LCav3) and human Cav3.1, Cav3.2, and Cav3.3 T-type channels specifically associate with CaM at helix 2 of the gating brake in the I-II linker of the channels. Isothermal titration calorimetry results revealed that the gating brake and CaM bind each other with high-nanomolar affinity. We show that the gating brake assumes a helical conformation upon binding CaM, with associated conformational changes to both CaM lobes as indicated by amide chemical shifts of the amino acids of CaM in 1H-15N HSQC NMR spectra. Intact Ca2+-binding sites on CaM and an intact gating brake sequence (first 39 amino acids of the I-II linker) were required in Cav3.2 channels to prevent the runaway gating phenotype, a hyperpolarizing shift in voltage sensitivities and faster gating kinetics. We conclude that the presence of high-nanomolar affinity binding sites for CaM at its universal gating brake and its unique form of regulation via the tuning of the voltage range of activity could influence the participation of Cav3 T-type channels in heart and brain rhythms. Our findings may have implications for arrhythmia disorders arising from mutations in the gating brake or CaM.
Collapse
Affiliation(s)
- Jean Chemin
- Institut de Génomique Fonctionnelle, CNRS, INSERM, Université de Montpellier, Montpellier F-34094, France
| | | | - Arnaud Monteil
- Institut de Génomique Fonctionnelle, CNRS, INSERM, Université de Montpellier, Montpellier F-34094, France
| | - Michael Piazza
- Departments of Chemistry, Waterloo, Ontario N2L 3G1, Canada
| | - Wendy Guan
- Biology, University of Waterloo, Waterloo, Ontario N2L 3G1, Canada
| | | | - Ashraf Kitmitto
- Division of Cardiovascular Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester M13 9NT, United Kingdom
| | - Zhiping P Pang
- Department of Neuroscience and Cell Biology, Rutgers Robert Wood Johnson Medical School, New Brunswick, New Jersey 08901
| | - Annette C Dolphin
- Department of Neuroscience, Physiology and Pharmacology, University College London, London WC1E 6BT, United Kingdom
| | - Edward Perez-Reyes
- Department of Pharmacology, University of Virginia, Charlottesville, Virginia 22908
| | | | | | - J David Spafford
- Biology, University of Waterloo, Waterloo, Ontario N2L 3G1, Canada.
| |
Collapse
|
26
|
Mustafá ER, López Soto EJ, Martínez Damonte V, Rodríguez SS, Lipscombe D, Raingo J. Constitutive activity of the Ghrelin receptor reduces surface expression of voltage-gated Ca 2+ channels in a Ca Vβ-dependent manner. J Cell Sci 2017; 130:3907-3917. [PMID: 29038230 DOI: 10.1242/jcs.207886] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2017] [Accepted: 10/04/2017] [Indexed: 12/15/2022] Open
Abstract
Voltage-gated Ca2+ (CaV) channels couple membrane depolarization to Ca2+ influx, triggering a range of Ca2+-dependent cellular processes. CaV channels are, therefore, crucial in shaping neuronal activity and function, depending on their individual temporal and spatial properties. Furthermore, many neurotransmitters and drugs that act through G protein coupled receptors (GPCRs), modulate neuronal activity by altering the expression, trafficking, or function of CaV channels. GPCR-dependent mechanisms that downregulate CaV channel expression levels are observed in many neurons but are, by comparison, less studied. Here we show that the growth hormone secretagogue receptor type 1a (GHSR), a GPCR, can inhibit the forwarding trafficking of several CaV subtypes, even in the absence of agonist. This constitutive form of GPCR inhibition of CaV channels depends on the presence of a CaVβ subunit. CaVβ subunits displace CaVα1 subunits from the endoplasmic reticulum. The actions of GHSR on CaV channels trafficking suggest a role for this signaling pathway in brain areas that control food intake, reward, and learning and memory.
Collapse
Affiliation(s)
- Emilio R Mustafá
- Electrophysiology Laboratory, Multidisciplinary Institute of Cell Biology (IMBICE), Universidad Nacional de La Plata - Consejo Nacional de Investigaciones Científicas y Técnicas, CONICET, and Comisión de Investigaciones de la Provincia de buenos Aires (CIC) Calle 526 1499-1579, B1906APM Tolosa, Buenos Aires, Argentina
| | - Eduardo J López Soto
- Department of Neuroscience, Brown University; Sidney E. Frank Hall for Life Sciences, 185 Meeting Street, Providence, Rhode Island 02912, USA
| | - Valentina Martínez Damonte
- Electrophysiology Laboratory, Multidisciplinary Institute of Cell Biology (IMBICE), Universidad Nacional de La Plata - Consejo Nacional de Investigaciones Científicas y Técnicas, CONICET, and Comisión de Investigaciones de la Provincia de buenos Aires (CIC) Calle 526 1499-1579, B1906APM Tolosa, Buenos Aires, Argentina
| | - Silvia S Rodríguez
- Electrophysiology Laboratory, Multidisciplinary Institute of Cell Biology (IMBICE), Universidad Nacional de La Plata - Consejo Nacional de Investigaciones Científicas y Técnicas, CONICET, and Comisión de Investigaciones de la Provincia de buenos Aires (CIC) Calle 526 1499-1579, B1906APM Tolosa, Buenos Aires, Argentina
| | - Diane Lipscombe
- Department of Neuroscience, Brown University; Sidney E. Frank Hall for Life Sciences, 185 Meeting Street, Providence, Rhode Island 02912, USA
| | - Jesica Raingo
- Electrophysiology Laboratory, Multidisciplinary Institute of Cell Biology (IMBICE), Universidad Nacional de La Plata - Consejo Nacional de Investigaciones Científicas y Técnicas, CONICET, and Comisión de Investigaciones de la Provincia de buenos Aires (CIC) Calle 526 1499-1579, B1906APM Tolosa, Buenos Aires, Argentina
| |
Collapse
|
27
|
Santolini I, Celli R, Cannella M, Imbriglio T, Guiducci M, Parisi P, Schubert J, Iacomino M, Zara F, Lerche H, Moyanova S, Ngomba RT, van Luijtelaar G, Battaglia G, Bruno V, Striano P, Nicoletti F. Alterations in the α 2 δ ligand, thrombospondin-1, in a rat model of spontaneous absence epilepsy and in patients with idiopathic/genetic generalized epilepsies. Epilepsia 2017; 58:1993-2001. [PMID: 28913875 DOI: 10.1111/epi.13898] [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] [Accepted: 08/18/2017] [Indexed: 11/28/2022]
Abstract
OBJECTIVES Thrombospondins, which are known to interact with the α2 δ subunit of voltage-sensitive calcium channels to stimulate the formation of excitatory synapses, have recently been implicated in the process of epileptogenesis. No studies have been so far performed on thrombospondins in models of absence epilepsy. We examined whether expression of the gene encoding for thrombospondin-1 was altered in the brain of WAG/Rij rats, which model absence epilepsy in humans. In addition, we examined the frequency of genetic variants of THBS1 in a large cohort of children affected by idiopathic/genetic generalized epilepsies (IGE/GGEs). METHODS We measured the transcripts of thrombospondin-1 and α2 δ subunit, and protein levels of α2 δ, Rab3A, and the vesicular glutamate transporter, VGLUT1, in the somatosensory cortex and ventrobasal thalamus of presymptomatic and symptomatic WAG/Rij rats and in two control strains by real-time polymerase chain reaction (PCR) and immunoblotting. We examined the genetic variants of THBS1 and CACNA2D1 in two independent cohorts of patients affected by IGE/GGE recruited through the Genetic Commission of the Italian League Against Epilepsy (LICE) and the EuroEPINOMICS-CoGIE Consortium. RESULTS Thrombospondin-1 messenger RNA (mRNA) levels were largely reduced in the ventrobasal thalamus of both presymptomatic and symptomatic WAG/Rij rats, whereas levels in the somatosensory cortex were unchanged. VGLUT1 protein levels were also reduced in the ventrobasal thalamus of WAG/Rij rats. Genetic variants of THBS1 were significantly more frequent in patients affected by IGE/GGE than in nonepileptic controls, whereas the frequency of CACNA2D1 was unchanged. SIGNIFICANCE These findings suggest that thrombospondin-1 may have a role in the pathogenesis of IGE/GGEs.
Collapse
Affiliation(s)
| | | | | | | | - Michela Guiducci
- Departments of Neurosciences, Mental Health and Sensory Organs, University Sapienza, Rome, Italy
| | - Pasquale Parisi
- Departments of Neurosciences, Mental Health and Sensory Organs, University Sapienza, Rome, Italy
| | - Julian Schubert
- Department of Neurology and Epileptology, Hertie Institute for Clinical Brain Research, University of Tübingen, Tübingen, Germany
| | - Michele Iacomino
- Laboratory of Neurogenetics, "G. Gaslini" Institute, Genova, Italy
| | - Federico Zara
- Laboratory of Neurogenetics, "G. Gaslini" Institute, Genova, Italy
| | - Holger Lerche
- Department of Neurology and Epileptology, Hertie Institute for Clinical Brain Research, University of Tübingen, Tübingen, Germany
| | | | | | | | | | | | | | - Valeria Bruno
- I.R.C.C.S. Neuromed, Pozzilli, Italy.,Departments of Physiology and Pharmacology, University Sapienza, Rome, Italy
| | - Pasquale Striano
- Pediatric Neurology and Muscular Diseases Unit, Departments of Neurosciences, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health, "G. Gaslini" Institute, University of Genoa, Genova, Italy
| | - Ferdinando Nicoletti
- I.R.C.C.S. Neuromed, Pozzilli, Italy.,Departments of Physiology and Pharmacology, University Sapienza, Rome, Italy
| |
Collapse
|
28
|
Proft J, Rzhepetskyy Y, Lazniewska J, Zhang FX, Cain SM, Snutch TP, Zamponi GW, Weiss N. The Cacna1h mutation in the GAERS model of absence epilepsy enhances T-type Ca 2+ currents by altering calnexin-dependent trafficking of Ca v3.2 channels. Sci Rep 2017; 7:11513. [PMID: 28912545 PMCID: PMC5599688 DOI: 10.1038/s41598-017-11591-5] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2017] [Accepted: 08/29/2017] [Indexed: 12/12/2022] Open
Abstract
Low-voltage-activated T-type calcium channels are essential contributors to the functioning of thalamocortical neurons by supporting burst-firing mode of action potentials. Enhanced T-type calcium conductance has been reported in the Genetic Absence Epilepsy Rat from Strasbourg (GAERS) and proposed to be causally related to the overall development of absence seizure activity. Here, we show that calnexin, an endoplasmic reticulum integral membrane protein, interacts with the III-IV linker region of the Cav3.2 channel to modulate the sorting of the channel to the cell surface. We demonstrate that the GAERS missense mutation located in the Cav3.2 III-IV linker alters the Cav3.2/calnexin interaction, resulting in an increased surface expression of the channel and a concomitant elevation in calcium influx. Our study reveals a novel mechanism that controls the expression of T-type channels, and provides a molecular explanation for the enhancement of T-type calcium conductance in GAERS.
Collapse
Affiliation(s)
- Juliane Proft
- Institute of Organic Chemistry and Biochemistry, Academy of Sciences of the Czech Republic, v.v.i., Prague, Czech Republic
| | - Yuriy Rzhepetskyy
- Institute of Organic Chemistry and Biochemistry, Academy of Sciences of the Czech Republic, v.v.i., Prague, Czech Republic
| | - Joanna Lazniewska
- Institute of Organic Chemistry and Biochemistry, Academy of Sciences of the Czech Republic, v.v.i., Prague, Czech Republic
| | - Fang-Xiong Zhang
- Department of Physiology and Pharmacology, Hotchkiss Brain Institute and Alberta Children's Hospital Research Institute, Cumming School of Medicine, University of Calgary, Calgary, T2N 4N1, Canada
| | - Stuart M Cain
- Michael Smith Laboratories and the Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, BC, V6T 1Z4, Canada
| | - Terrance P Snutch
- Michael Smith Laboratories and the Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, BC, V6T 1Z4, Canada
| | - Gerald W Zamponi
- Department of Physiology and Pharmacology, Hotchkiss Brain Institute and Alberta Children's Hospital Research Institute, Cumming School of Medicine, University of Calgary, Calgary, T2N 4N1, Canada.
| | - Norbert Weiss
- Institute of Organic Chemistry and Biochemistry, Academy of Sciences of the Czech Republic, v.v.i., Prague, Czech Republic.
| |
Collapse
|
29
|
Smith CL, Abdallah S, Wong YY, Le P, Harracksingh AN, Artinian L, Tamvacakis AN, Rehder V, Reese TS, Senatore A. Evolutionary insights into T-type Ca 2+ channel structure, function, and ion selectivity from the Trichoplax adhaerens homologue. J Gen Physiol 2017; 149:483-510. [PMID: 28330839 PMCID: PMC5379919 DOI: 10.1085/jgp.201611683] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2016] [Accepted: 02/07/2017] [Indexed: 12/31/2022] Open
Abstract
The role of T-type calcium channels in animals without nervous systems is unknown. Smith et al. characterize TCav3 from Trichoplax adhaerens, finding expression in neurosecretory-like cells and preference for Ca2+ over Na+ via strong extracellular Ca2+ block, despite low selectivity for Ca2+ in the pore. Four-domain voltage-gated Ca2+ (Cav) channels play fundamental roles in the nervous system, but little is known about when or how their unique properties and cellular roles evolved. Of the three types of metazoan Cav channels, Cav1 (L-type), Cav2 (P/Q-, N- and R-type) and Cav3 (T-type), Cav3 channels are optimized for regulating cellular excitability because of their fast kinetics and low activation voltages. These same properties permit Cav3 channels to drive low-threshold exocytosis in select neurons and neurosecretory cells. Here, we characterize the single T-type calcium channel from Trichoplax adhaerens (TCav3), an early diverging animal that lacks muscle, neurons, and synapses. Co-immunolocalization using antibodies against TCav3 and neurosecretory cell marker complexin labeled gland cells, which are hypothesized to play roles in paracrine signaling. Cloning and in vitro expression of TCav3 reveals that, despite roughly 600 million years of divergence from other T-type channels, it bears the defining structural and biophysical features of the Cav3 family. We also characterize the channel’s cation permeation properties and find that its pore is less selective for Ca2+ over Na+ compared with the human homologue Cav3.1, yet it exhibits a similar potent block of inward Na+ current by low external Ca2+ concentrations (i.e., the Ca2+ block effect). A comparison of the permeability features of TCav3 with other cloned channels suggests that Ca2+ block is a locus of evolutionary change in T-type channel cation permeation properties and that mammalian channels distinguish themselves from invertebrate ones by bearing both stronger Ca2+ block and higher Ca2+ selectivity. TCav3 is the most divergent metazoan T-type calcium channel and thus provides an evolutionary perspective on Cav3 channel structure–function properties, ion selectivity, and cellular physiology.
Collapse
Affiliation(s)
- Carolyn L Smith
- National Institute of Neurological Diseases and Stroke, National Institutes of Health, Bethesda, MD 20892
| | - Salsabil Abdallah
- University of Toronto Mississauga, Mississauga, Ontario L5L 1C6, Canada
| | - Yuen Yan Wong
- University of Toronto Mississauga, Mississauga, Ontario L5L 1C6, Canada
| | - Phuong Le
- University of Toronto Mississauga, Mississauga, Ontario L5L 1C6, Canada
| | | | | | | | | | - Thomas S Reese
- National Institute of Neurological Diseases and Stroke, National Institutes of Health, Bethesda, MD 20892
| | - Adriano Senatore
- University of Toronto Mississauga, Mississauga, Ontario L5L 1C6, Canada
| |
Collapse
|
30
|
Thrombospondin-4 divergently regulates voltage-gated Ca2+ channel subtypes in sensory neurons after nerve injury. Pain 2017; 157:2068-2080. [PMID: 27168360 DOI: 10.1097/j.pain.0000000000000612] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Loss of high-voltage-activated (HVA) calcium current (ICa) and gain of low-voltage-activated (LVA) ICa after painful peripheral nerve injury cause elevated excitability in sensory neurons. Nerve injury is also accompanied by increased expression of the extracellular matrix glycoprotein thrombospondin-4 (TSP4), and interruption of TSP4 function can reverse or prevent behavioral hypersensitivity after injury. We therefore investigated TSP4 regulation of ICa in dorsal root ganglion (DRG) neurons. During depolarization adequate to activate HVA ICa, TSP4 decreases both N- and L-type ICa and the associated intracellular calcium transient. In contrast, TSP4 increases ICa and the intracellular calcium signal after low-voltage depolarization, which we confirmed is due to ICa through T-type channels. These effects are blocked by gabapentin, which ameliorates neuropathic pain by targeting the α2δ1 calcium subunit. Injury-induced changes of HVA and LVA ICa are attenuated in TSP4 knockout mice. In the neuropathic pain model of spinal nerve ligation, TSP4 application did not further regulate ICa of injured DRG neurons. Taken together, these findings suggest that elevated TSP4 after peripheral nerve injury may contribute to hypersensitivity of peripheral sensory systems by decreasing HVA and increasing LVA in DRG neurons by targeting the α2δ1 calcium subunit. Controlling TSP4 overexpression in peripheral sensory neurons may be a target for analgesic drug development for neuropathic pain.
Collapse
|
31
|
Trafficking of neuronal calcium channels. Neuronal Signal 2017; 1:NS20160003. [PMID: 32714572 PMCID: PMC7373241 DOI: 10.1042/ns20160003] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2017] [Revised: 01/20/2017] [Accepted: 01/19/2017] [Indexed: 12/18/2022] Open
Abstract
Neuronal voltage-gated calcium channels (VGCCs) serve complex yet essential physiological functions via their pivotal role in translating electrical signals into intracellular calcium elevations and associated downstream signalling pathways. There are a number of regulatory mechanisms to ensure a dynamic control of the number of channels embedded in the plasma membrane, whereas alteration of the surface expression of VGCCs has been linked to various disease conditions. Here, we provide an overview of the mechanisms that control the trafficking of VGCCs to and from the plasma membrane, and discuss their implication in pathophysiological conditions and their potential as therapeutic targets.
Collapse
|
32
|
Lazniewska J, Weiss N. Glycosylation of voltage-gated calcium channels in health and disease. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2017; 1859:662-668. [PMID: 28109749 DOI: 10.1016/j.bbamem.2017.01.018] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/09/2016] [Revised: 01/10/2017] [Accepted: 01/16/2017] [Indexed: 12/26/2022]
Abstract
Voltage-gated calcium channels (VGCCs) are transmembrane proteins that translate electrical activities into intracellular calcium elevations and downstream signaling pathways. They serve essential physiological functions including communication between nerve cells, muscle contraction, cardiac activity, and release of hormones and neurotransmitters. Asparagine-linked glycosylation has emerged as an essential post-translational modification to control the number of channels embedded in the plasma membrane but also their functional gating properties. This review provides a comprehensive overview about the current state of knowledge on the role of glycosylation in the expression and functioning of VGCCs, and discusses how variations in the glycosylation of the channel proteins can contribute to pathological conditions.
Collapse
Affiliation(s)
- Joanna Lazniewska
- Institute of Organic Chemistry and Biochemistry, Czech Academy of Sciences, Prague, Czech Republic
| | - Norbert Weiss
- Institute of Organic Chemistry and Biochemistry, Czech Academy of Sciences, Prague, Czech Republic.
| |
Collapse
|
33
|
Gerardi MC, Atzeni F, Batticciotto A, Di Franco M, Rizzi M, Sarzi-Puttini P. The safety of pregabalin in the treatment of fibromyalgia. Expert Opin Drug Saf 2016; 15:1541-1548. [DOI: 10.1080/14740338.2016.1242575] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Affiliation(s)
- Maria Chiara Gerardi
- Rheumatology Unit, ASST-Fatebenefratelli-L, Sacco University Hospital, Milan, Italy
| | - Fabiola Atzeni
- Rheumatology Unit, ASST-Fatebenefratelli-L, Sacco University Hospital, Milan, Italy
| | - Alberto Batticciotto
- Rheumatology Unit, ASST-Fatebenefratelli-L, Sacco University Hospital, Milan, Italy
| | - Manuela Di Franco
- Rheumatology Unit, Department of Internal Medicine and Medical Specialities, Sapienza, University of Rome, Rome, Italy
| | - Maurizio Rizzi
- Pulmonary Department, ASST-Fatebenefratelli-L, Sacco University Hospital, Milan, Italy
| | | |
Collapse
|
34
|
Daniil G, Fernandes-Rosa FL, Chemin J, Blesneac I, Beltrand J, Polak M, Jeunemaitre X, Boulkroun S, Amar L, Strom TM, Lory P, Zennaro MC. CACNA1H Mutations Are Associated With Different Forms of Primary Aldosteronism. EBioMedicine 2016; 13:225-236. [PMID: 27729216 PMCID: PMC5264314 DOI: 10.1016/j.ebiom.2016.10.002] [Citation(s) in RCA: 109] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2016] [Revised: 09/22/2016] [Accepted: 10/03/2016] [Indexed: 01/10/2023] Open
Abstract
Primary aldosteronism (PA) is the most common form of secondary hypertension. Mutations in KCNJ5, ATP1A1, ATP2B3 and CACNA1D are found in aldosterone producing adenoma (APA) and familial hyperaldosteronism (FH). A recurrent mutation in CACNA1H (coding for Cav3.2) was identified in a familial form of early onset PA. Here we performed whole exome sequencing (WES) in patients with different types of PA to identify new susceptibility genes. Four different heterozygous germline CACNA1H variants were identified. A de novo Cav3.2 p.Met1549Ile variant was found in early onset PA and multiplex developmental disorder. Cav3.2 p.Ser196Leu and p.Pro2083Leu were found in two patients with FH, and p.Val1951Glu was identified in one patient with APA. Electrophysiological analysis of mutant Cav3.2 channels revealed significant changes in the Ca2+ current properties for all mutants, suggesting a gain of function phenotype. Transfections of mutant Cav3.2 in H295R-S2 cells led to increased aldosterone production and/or expression of genes coding for steroidogenic enzymes after K+ stimulation. Identification of CACNA1H mutations associated with early onset PA, FH, and APA suggests that CACNA1H might be a susceptibility gene predisposing to PA with different phenotypic presentations, opening new perspectives for genetic diagnosis and management of patients with PA.
Collapse
Affiliation(s)
- Georgios Daniil
- INSERM, UMRS_970, Paris Cardiovascular Research Center, Paris, France; Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - Fabio L Fernandes-Rosa
- INSERM, UMRS_970, Paris Cardiovascular Research Center, Paris, France; Université Paris Descartes, Sorbonne Paris Cité, Paris, France; Assistance Publique-Hôpitaux de Paris, Hôpital Européen Georges Pompidou, Service de Génétique, Paris, France.
| | - Jean Chemin
- Institut de Génomique Fonctionnelle, Université de Montpellier, CNRS UMR 5203, INSERM U 1191, Montpellier F-34094, France; LabEx Ion Channel Science and Therapeutics, Montpellier F-34094, France
| | - Iulia Blesneac
- Institut de Génomique Fonctionnelle, Université de Montpellier, CNRS UMR 5203, INSERM U 1191, Montpellier F-34094, France; LabEx Ion Channel Science and Therapeutics, Montpellier F-34094, France
| | - Jacques Beltrand
- Université Paris Descartes, Sorbonne Paris Cité, Paris, France; Assistance Publique-Hôpitaux de Paris, Hôpital Necker Enfants Malades, Service d'Endocrinologie, Paris, France; Inserm UMR_1016, Institut Cochin, Paris, France; Institut Imagine, Paris Descartes - Université Sorbonne Paris Cité, Paris, France
| | - Michel Polak
- Université Paris Descartes, Sorbonne Paris Cité, Paris, France; Assistance Publique-Hôpitaux de Paris, Hôpital Necker Enfants Malades, Service d'Endocrinologie, Paris, France; Inserm UMR_1016, Institut Cochin, Paris, France; Institut Imagine, Paris Descartes - Université Sorbonne Paris Cité, Paris, France
| | - Xavier Jeunemaitre
- INSERM, UMRS_970, Paris Cardiovascular Research Center, Paris, France; Université Paris Descartes, Sorbonne Paris Cité, Paris, France; Assistance Publique-Hôpitaux de Paris, Hôpital Européen Georges Pompidou, Service de Génétique, Paris, France
| | - Sheerazed Boulkroun
- INSERM, UMRS_970, Paris Cardiovascular Research Center, Paris, France; Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - Laurence Amar
- INSERM, UMRS_970, Paris Cardiovascular Research Center, Paris, France; Université Paris Descartes, Sorbonne Paris Cité, Paris, France; Assistance Publique-Hôpitaux de Paris, Hôpital Européen Georges Pompidou, Unité Hypertension artérielle, Paris, France
| | - Tim M Strom
- Institute of Human Genetics, Helmholtz Zentrum München, Neuherberg, Germany; Institute of Human Genetics, Technische Universität München, Munich, Germany
| | - Philippe Lory
- Institut de Génomique Fonctionnelle, Université de Montpellier, CNRS UMR 5203, INSERM U 1191, Montpellier F-34094, France; LabEx Ion Channel Science and Therapeutics, Montpellier F-34094, France
| | - Maria-Christina Zennaro
- INSERM, UMRS_970, Paris Cardiovascular Research Center, Paris, France; Université Paris Descartes, Sorbonne Paris Cité, Paris, France; Assistance Publique-Hôpitaux de Paris, Hôpital Européen Georges Pompidou, Service de Génétique, Paris, France.
| |
Collapse
|
35
|
Voigt A, Freund R, Heck J, Missler M, Obermair GJ, Thomas U, Heine M. Dynamic association of calcium channel subunits at the cellular membrane. NEUROPHOTONICS 2016; 3:041809. [PMID: 27872869 PMCID: PMC5093230 DOI: 10.1117/1.nph.3.4.041809] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/15/2016] [Accepted: 10/10/2016] [Indexed: 05/25/2023]
Abstract
High voltage gated calcium channels (VGCCs) are composed of at least three subunits, one pore forming [Formula: see text]-subunit, an intracellular [Formula: see text]-variant, and a mostly extracellular [Formula: see text]-variant. Interactions between these subunits determine the kinetic properties of VGCCs. It is unclear whether these interactions are stable over time or rather transient. Here, we used single-molecule tracking to investigate the surface diffusion of [Formula: see text]- and [Formula: see text]-subunits at the cell surface. We found that [Formula: see text]-subunits show higher surface mobility than [Formula: see text]-subunits, and that they are only transiently confined together, suggesting a weak association between [Formula: see text]- and [Formula: see text]-subunits. Moreover, we observed that different [Formula: see text]-subunits engage in different degrees of association with the [Formula: see text]-subunit, revealing the tighter interaction of [Formula: see text] with [Formula: see text]. These data indicate a distinct regulation of the [Formula: see text] interaction in VGCC subtypes. We modeled their membrane dynamics in a Monte Carlo simulation using experimentally determined diffusion constants. Our modeling predicts that the ratio of associated [Formula: see text]- and [Formula: see text]-subunits mainly depends on their expression density and confinement in the membrane. Based on the different motilities of particular [Formula: see text]-subunit combinations, we propose that their dynamic assembly and disassembly represent an important mechanism to regulate the signaling properties of VGCC.
Collapse
Affiliation(s)
- Andreas Voigt
- Otto-von-Guericke-University of Magdeburg, Lehrstuhl Systemverfahrenstechnik, Universitätsplatz 2, Magdeburg D-39106, Germany
| | - Romy Freund
- Leibniz-Institute of Neurobiology, Research Group Molecular Physiology, Brenneckestrasse 6, Magdeburg D-39118, Germany
| | - Jennifer Heck
- Leibniz-Institute of Neurobiology, Research Group Molecular Physiology, Brenneckestrasse 6, Magdeburg D-39118, Germany
| | - Markus Missler
- Westfälische Wilhelms-University, Institute of Anatomy and Molecular Neurobiology, Vesaliusweg 2, Münster 48149, Germany
| | - Gerald J. Obermair
- Medical University Innsbruck, Division of Physiology, Department of Physiology and Medical Physics, Schöpfstrasse 41, Innsbruck 6020, Austria
| | - Ulrich Thomas
- Leibniz-Institute of Neurobiology, Department Neurochemistry, Brenneckestrasse 6, Magdeburg D-39118, Germany
| | - Martin Heine
- Leibniz-Institute of Neurobiology, Research Group Molecular Physiology, Brenneckestrasse 6, Magdeburg D-39118, Germany
- Otto-von-Guericke-University Magdeburg, Center for Behavioral Brain Sciences (CBBS), Universitätsplatz 2, Magdeburg D-39106, Germany
| |
Collapse
|
36
|
Cooperative roles of glucose and asparagine-linked glycosylation in T-type calcium channel expression. Pflugers Arch 2016; 468:1837-1851. [DOI: 10.1007/s00424-016-1881-y] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2016] [Revised: 09/04/2016] [Accepted: 09/07/2016] [Indexed: 12/15/2022]
|
37
|
Dolphin AC. Voltage-gated calcium channels and their auxiliary subunits: physiology and pathophysiology and pharmacology. J Physiol 2016; 594:5369-90. [PMID: 27273705 PMCID: PMC5043047 DOI: 10.1113/jp272262] [Citation(s) in RCA: 237] [Impact Index Per Article: 26.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2016] [Accepted: 05/09/2016] [Indexed: 12/22/2022] Open
Abstract
Voltage‐gated calcium channels are essential players in many physiological processes in excitable cells. There are three main subdivisions of calcium channel, defined by the pore‐forming α1 subunit, the CaV1, CaV2 and CaV3 channels. For all the subtypes of voltage‐gated calcium channel, their gating properties are key for the precise control of neurotransmitter release, muscle contraction and cell excitability, among many other processes. For the CaV1 and CaV2 channels, their ability to reach their required destinations in the cell membrane, their activation and the fine tuning of their biophysical properties are all dramatically influenced by the auxiliary subunits that associate with them. Furthermore, there are many diseases, both genetic and acquired, involving voltage‐gated calcium channels. This review will provide a general introduction and then concentrate particularly on the role of auxiliary α2δ subunits in both physiological and pathological processes involving calcium channels, and as a therapeutic target.
![]()
Collapse
Affiliation(s)
- Annette C Dolphin
- Department of Neuroscience, Physiology and Pharmacology, University College London, Gower Street, London, WC1E 6BT, UK.
| |
Collapse
|
38
|
|
39
|
Wang G, Bochorishvili G, Chen Y, Salvati KA, Zhang P, Dubel SJ, Perez-Reyes E, Snutch TP, Stornetta RL, Deisseroth K, Erisir A, Todorovic SM, Luo JH, Kapur J, Beenhakker MP, Zhu JJ. CaV3.2 calcium channels control NMDA receptor-mediated transmission: a new mechanism for absence epilepsy. Genes Dev 2015. [PMID: 26220996 PMCID: PMC4526737 DOI: 10.1101/gad.260869.115] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
CaV3.2 T-type calcium channels, encoded by CACNA1H, are expressed throughout the brain, yet their general function remains unclear. We discovered that CaV3.2 channels control NMDA-sensitive glutamatergic receptor (NMDA-R)-mediated transmission and subsequent NMDA-R-dependent plasticity of AMPA-R-mediated transmission at rat central synapses. Interestingly, functional CaV3.2 channels primarily incorporate into synapses, replace existing CaV3.2 channels, and can induce local calcium influx to control NMDA transmission strength in an activity-dependent manner. Moreover, human childhood absence epilepsy (CAE)-linked hCaV3.2(C456S) mutant channels have a higher channel open probability, induce more calcium influx, and enhance glutamatergic transmission. Remarkably, cortical expression of hCaV3.2(C456S) channels in rats induces 2- to 4-Hz spike and wave discharges and absence-like epilepsy characteristic of CAE patients, which can be suppressed by AMPA-R and NMDA-R antagonists but not T-type calcium channel antagonists. These results reveal an unexpected role of CaV3.2 channels in regulating NMDA-R-mediated transmission and a novel epileptogenic mechanism for human CAE.
Collapse
Affiliation(s)
- Guangfu Wang
- Department of Pharmacology, University of Virginia, Charlottesville, Virginia 22908, USA
| | - Genrieta Bochorishvili
- Department of Pharmacology, University of Virginia, Charlottesville, Virginia 22908, USA
| | - Yucai Chen
- Department of Pharmacology, University of Virginia, Charlottesville, Virginia 22908, USA
| | - Kathryn A Salvati
- Department of Pharmacology, University of Virginia, Charlottesville, Virginia 22908, USA
| | - Peng Zhang
- Department of Pharmacology, University of Virginia, Charlottesville, Virginia 22908, USA
| | - Steve J Dubel
- Laboratoire de Génomique Fonctionnelle, Département de Physiologie, Unité Propre de Recherche 2580, Centre National de la Recherche Scientifique, 34396 Montpellier, France
| | - Edward Perez-Reyes
- Department of Pharmacology, University of Virginia, Charlottesville, Virginia 22908, USA
| | - Terrance P Snutch
- Michael Smith Laboratories, University of British Columbia, Vancouver, British Columbia V6T 1Z4, Canada; Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, British Columbia V6T 1Z4, Canada
| | - Ruth L Stornetta
- Department of Pharmacology, University of Virginia, Charlottesville, Virginia 22908, USA
| | - Karl Deisseroth
- Department of Bioengineering, Stanford University, Stanford, California 94305, USA; Howard Hughes Medical Institute, Stanford University, Stanford, California 94305, USA
| | - Alev Erisir
- Department of Psychology, University of Virginia, Charlottesville, Virginia 22908, USA
| | - Slobodan M Todorovic
- Department of Anesthesiology, University of Virginia, Charlottesville, Virginia 22908, USA; Department of Neuroscience, University of Virginia, Charlottesville, Virginia 22908, USA
| | - Jian-Hong Luo
- Department of Neurobiology, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310058, China
| | - Jaideep Kapur
- Department of Neurology, University of Virginia, Charlottesville, Virginia 22908, USA
| | - Mark P Beenhakker
- Department of Pharmacology, University of Virginia, Charlottesville, Virginia 22908, USA
| | - J Julius Zhu
- Department of Pharmacology, University of Virginia, Charlottesville, Virginia 22908, USA; Department of Neuroscience, University of Virginia, Charlottesville, Virginia 22908, USA
| |
Collapse
|
40
|
Zamponi GW, Striessnig J, Koschak A, Dolphin AC. The Physiology, Pathology, and Pharmacology of Voltage-Gated Calcium Channels and Their Future Therapeutic Potential. Pharmacol Rev 2015; 67:821-70. [PMID: 26362469 PMCID: PMC4630564 DOI: 10.1124/pr.114.009654] [Citation(s) in RCA: 790] [Impact Index Per Article: 79.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Voltage-gated calcium channels are required for many key functions in the body. In this review, the different subtypes of voltage-gated calcium channels are described and their physiologic roles and pharmacology are outlined. We describe the current uses of drugs interacting with the different calcium channel subtypes and subunits, as well as specific areas in which there is strong potential for future drug development. Current therapeutic agents include drugs targeting L-type Ca(V)1.2 calcium channels, particularly 1,4-dihydropyridines, which are widely used in the treatment of hypertension. T-type (Ca(V)3) channels are a target of ethosuximide, widely used in absence epilepsy. The auxiliary subunit α2δ-1 is the therapeutic target of the gabapentinoid drugs, which are of value in certain epilepsies and chronic neuropathic pain. The limited use of intrathecal ziconotide, a peptide blocker of N-type (Ca(V)2.2) calcium channels, as a treatment of intractable pain, gives an indication that these channels represent excellent drug targets for various pain conditions. We describe how selectivity for different subtypes of calcium channels (e.g., Ca(V)1.2 and Ca(V)1.3 L-type channels) may be achieved in the future by exploiting differences between channel isoforms in terms of sequence and biophysical properties, variation in splicing in different target tissues, and differences in the properties of the target tissues themselves in terms of membrane potential or firing frequency. Thus, use-dependent blockers of the different isoforms could selectively block calcium channels in particular pathologies, such as nociceptive neurons in pain states or in epileptic brain circuits. Of important future potential are selective Ca(V)1.3 blockers for neuropsychiatric diseases, neuroprotection in Parkinson's disease, and resistant hypertension. In addition, selective or nonselective T-type channel blockers are considered potential therapeutic targets in epilepsy, pain, obesity, sleep, and anxiety. Use-dependent N-type calcium channel blockers are likely to be of therapeutic use in chronic pain conditions. Thus, more selective calcium channel blockers hold promise for therapeutic intervention.
Collapse
Affiliation(s)
- Gerald W Zamponi
- Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada (G.W.Z.); Department of Pharmacology and Toxicology, Institute of Pharmacy, Center for Molecular Biosciences, University of Innsbruck, Innsbruck, Austria (J.S., A.K.); and Department of Neuroscience, Physiology, and Pharmacology, Division of Biosciences, University College London, London, United Kingdom (A.C.D.)
| | - Joerg Striessnig
- Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada (G.W.Z.); Department of Pharmacology and Toxicology, Institute of Pharmacy, Center for Molecular Biosciences, University of Innsbruck, Innsbruck, Austria (J.S., A.K.); and Department of Neuroscience, Physiology, and Pharmacology, Division of Biosciences, University College London, London, United Kingdom (A.C.D.)
| | - Alexandra Koschak
- Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada (G.W.Z.); Department of Pharmacology and Toxicology, Institute of Pharmacy, Center for Molecular Biosciences, University of Innsbruck, Innsbruck, Austria (J.S., A.K.); and Department of Neuroscience, Physiology, and Pharmacology, Division of Biosciences, University College London, London, United Kingdom (A.C.D.)
| | - Annette C Dolphin
- Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada (G.W.Z.); Department of Pharmacology and Toxicology, Institute of Pharmacy, Center for Molecular Biosciences, University of Innsbruck, Innsbruck, Austria (J.S., A.K.); and Department of Neuroscience, Physiology, and Pharmacology, Division of Biosciences, University College London, London, United Kingdom (A.C.D.)
| |
Collapse
|
41
|
Abstract
The ubiquitous pannexin 1 (Panx1) ion- and metabolite-permeable channel mediates the release of ATP, a potent signalling molecule. In the present study, we provide striking evidence that ATP, in turn, stimulates internalization of Panx1 to intracellular membranes. These findings hold important implications for understanding the regulation of Panx1 when extracellular ATP is elevated. In the nervous system, this includes phenomena such as synaptic plasticity, pain, precursor cell development and stroke; outside of the nervous system, this includes things like skeletal and smooth muscle activity and inflammation. Within 15 min, ATP led to significant Panx1-EGFP internalization. In a series of experiments, we determined that hydrolysable ATP is the most potent stimulator of Panx1 internalization. We identified two possible mechanisms for Panx1 internalization, including activation of ionotropic purinergic (P2X) receptors and involvement of a putative ATP-sensitive residue in the first extracellular loop of Panx1 (Trp(74)). Internalization was cholesterol-dependent, but clathrin, caveolin and dynamin independent. Detailed analysis of Panx1 at specific endosome sub-compartments confirmed that Panx1 is expressed in endosome membranes of the classical degradation pathway under basal conditions and that elevation of ATP levels diverts a sub-population to recycling endosomes. This is the first report detailing endosome localization of Panx1 under basal conditions and the potential for ATP regulation of its surface expression. Given the ubiquitous expression profile of Panx1 and the importance of ATP signalling, these findings are of critical importance for understanding the role of Panx1 in health and disease.
Collapse
|
42
|
Duzhyy DE, Viatchenko-Karpinski VY, Khomula EV, Voitenko NV, Belan PV. Upregulation of T-type Ca2+ channels in long-term diabetes determines increased excitability of a specific type of capsaicin-insensitive DRG neurons. Mol Pain 2015; 11:29. [PMID: 25986602 PMCID: PMC4490764 DOI: 10.1186/s12990-015-0028-z] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2015] [Accepted: 05/13/2015] [Indexed: 01/15/2023] Open
Abstract
Background Previous studies have shown that increased excitability of capsaicin-sensitive DRG neurons and thermal hyperalgesia in rats with short-term (2–4 weeks) streptozotocin-induced diabetes is mediated by upregulation of T-type Ca2+ current. In longer–term diabetes (after the 8th week) thermal hyperalgesia is changed to hypoalgesia that is accompanied by downregulation of T-type current in capsaicin-sensitive small-sized nociceptors. At the same time pain symptoms of diabetic neuropathy other than thermal persist in STZ-diabetic animals and patients during progression of diabetes into later stages suggesting that other types of DRG neurons may be sensitized and contribute to pain. In this study, we examined functional expression of T-type Ca2+ channels in capsaicin-insensitive DRG neurons and excitability of these neurons in longer-term diabetic rats and in thermally hypoalgesic diabetic rats. Results Here we have demonstrated that in STZ-diabetes T-type current was upregulated in capsaicin-insensitive low-pH-sensitive small-sized nociceptive DRG neurons of longer-term diabetic rats and thermally hypoalgesic diabetic rats. This upregulation was not accompanied by significant changes in biophysical properties of T-type channels suggesting that a density of functionally active channels was increased. Sensitivity of T-type current to amiloride (1 mM) and low concentration of Ni2+ (50 μM) implicates prevalence of Cav3.2 subtype of T-type channels in the capsaicin-insensitive low-pH-sensitive neurons of both naïve and diabetic rats. The upregulation of T-type channels resulted in the increased neuronal excitability of these nociceptive neurons revealed by a lower threshold for action potential initiation, prominent afterdepolarizing potentials and burst firing. Sodium current was not significantly changed in these neurons during long-term diabetes and could not contribute to the diabetes-induced increase of neuronal excitability. Conclusions Capsaicin-insensitive low-pH-sensitive type of DRG neurons shows diabetes-induced upregulation of Cav3.2 subtype of T-type channels. This upregulation results in the increased excitability of these neurons and may contribute to nonthermal nociception at a later-stage diabetes.
Collapse
Affiliation(s)
- Dmytro E Duzhyy
- Department of General Physiology of the CNS and State Key Laboratory of Molecular and Cellular Biology, Bogomoletz Institute of Physiology of National Academy of Science of Ukraine, 4 Bogomoletz street, 01024, Kyiv, Ukraine.
| | - Viacheslav Y Viatchenko-Karpinski
- Department of General Physiology of the CNS and State Key Laboratory of Molecular and Cellular Biology, Bogomoletz Institute of Physiology of National Academy of Science of Ukraine, 4 Bogomoletz street, 01024, Kyiv, Ukraine.
| | - Eugen V Khomula
- International Center of Molecular Physiology of National Academy of Science of Ukraine, 4 Bogomoletz street, 01024, Kyiv, Ukraine.
| | - Nana V Voitenko
- Department of General Physiology of the CNS and State Key Laboratory of Molecular and Cellular Biology, Bogomoletz Institute of Physiology of National Academy of Science of Ukraine, 4 Bogomoletz street, 01024, Kyiv, Ukraine.
| | - Pavel V Belan
- Department of General Physiology of the CNS and State Key Laboratory of Molecular and Cellular Biology, Bogomoletz Institute of Physiology of National Academy of Science of Ukraine, 4 Bogomoletz street, 01024, Kyiv, Ukraine.
| |
Collapse
|
43
|
Monteil A, Chausson P, Boutourlinsky K, Mezghrani A, Huc-Brandt S, Blesneac I, Bidaud I, Lemmers C, Leresche N, Lambert RC, Lory P. Inhibition of Cav3.2 T-type Calcium Channels by Its Intracellular I-II Loop. J Biol Chem 2015; 290:16168-76. [PMID: 25931121 DOI: 10.1074/jbc.m114.634261] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2014] [Indexed: 11/06/2022] Open
Abstract
Voltage-dependent calcium channels (Cav) of the T-type family (Cav3.1, Cav3.2, and Cav3.3) are activated by low threshold membrane depolarization and contribute greatly to neuronal network excitability. Enhanced T-type channel activity, especially Cav3.2, contributes to disease states, including absence epilepsy. Interestingly, the intracellular loop connecting domains I and II (I-II loop) of Cav3.2 channels is implicated in the control of both surface expression and channel gating, indicating that this I-II loop plays an important regulatory role in T-type current. Here we describe that co-expression of this I-II loop or its proximal region (Δ1-Cav3.2; Ser(423)-Pro(542)) together with recombinant full-length Cav3.2 channel inhibited T-type current without affecting channel expression and membrane incorporation. Similar T-type current inhibition was obtained in NG 108-15 neuroblastoma cells that constitutively express Cav3.2 channels. Of interest, Δ1-Cav3.2 inhibited both Cav3.2 and Cav3.1 but not Cav3.3 currents. Efficacy of Δ1-Cav3.2 to inhibit native T-type channels was assessed in thalamic neurons using viral transduction. We describe that T-type current was significantly inhibited in the ventrobasal neurons that express Cav3.1, whereas in nucleus reticularis thalami neurons that express Cav3.2 and Cav3.3 channels, only the fast inactivating T-type current (Cav3.2 component) was significantly inhibited. Altogether, these data describe a new strategy to differentially inhibit Cav3 isoforms of the T-type calcium channels.
Collapse
Affiliation(s)
- Arnaud Monteil
- From the Université de Montpellier, CNRS UMR 5203, Département de Physiologie, Institut de Génomique Fonctionnelle, Montpellier, F-34094 France, INSERM, U1191, Montpellier, F-34094 France, Plateforme de Vectorologie, Biocampus Montpellier CNRS UMS 3426, INSERM US009, Montpellier, F-34094 France, LabEx "Ion Channel Science and Therapeutics," Montpellier, F-34094 France
| | - Patrick Chausson
- Sorbonne Universités, Université Pierre et Marie Curie Université Paris 06, UM 119, Neuroscience Paris Seine (NPS), Paris F-75005, France, CNRS UMR 8246, NPS, Paris F-75005, France, and INSERM, U1130, NPS, Paris F-75005, France
| | - Katia Boutourlinsky
- Sorbonne Universités, Université Pierre et Marie Curie Université Paris 06, UM 119, Neuroscience Paris Seine (NPS), Paris F-75005, France, CNRS UMR 8246, NPS, Paris F-75005, France, and INSERM, U1130, NPS, Paris F-75005, France
| | - Alexandre Mezghrani
- From the Université de Montpellier, CNRS UMR 5203, Département de Physiologie, Institut de Génomique Fonctionnelle, Montpellier, F-34094 France, INSERM, U1191, Montpellier, F-34094 France, LabEx "Ion Channel Science and Therapeutics," Montpellier, F-34094 France
| | - Sylvaine Huc-Brandt
- From the Université de Montpellier, CNRS UMR 5203, Département de Physiologie, Institut de Génomique Fonctionnelle, Montpellier, F-34094 France, INSERM, U1191, Montpellier, F-34094 France, LabEx "Ion Channel Science and Therapeutics," Montpellier, F-34094 France
| | - Iulia Blesneac
- From the Université de Montpellier, CNRS UMR 5203, Département de Physiologie, Institut de Génomique Fonctionnelle, Montpellier, F-34094 France, INSERM, U1191, Montpellier, F-34094 France, LabEx "Ion Channel Science and Therapeutics," Montpellier, F-34094 France
| | - Isabelle Bidaud
- From the Université de Montpellier, CNRS UMR 5203, Département de Physiologie, Institut de Génomique Fonctionnelle, Montpellier, F-34094 France, INSERM, U1191, Montpellier, F-34094 France, LabEx "Ion Channel Science and Therapeutics," Montpellier, F-34094 France
| | - Céline Lemmers
- From the Université de Montpellier, CNRS UMR 5203, Département de Physiologie, Institut de Génomique Fonctionnelle, Montpellier, F-34094 France, INSERM, U1191, Montpellier, F-34094 France, Plateforme de Vectorologie, Biocampus Montpellier CNRS UMS 3426, INSERM US009, Montpellier, F-34094 France, LabEx "Ion Channel Science and Therapeutics," Montpellier, F-34094 France
| | - Nathalie Leresche
- Sorbonne Universités, Université Pierre et Marie Curie Université Paris 06, UM 119, Neuroscience Paris Seine (NPS), Paris F-75005, France, CNRS UMR 8246, NPS, Paris F-75005, France, and INSERM, U1130, NPS, Paris F-75005, France
| | - Régis C Lambert
- Sorbonne Universités, Université Pierre et Marie Curie Université Paris 06, UM 119, Neuroscience Paris Seine (NPS), Paris F-75005, France, CNRS UMR 8246, NPS, Paris F-75005, France, and INSERM, U1130, NPS, Paris F-75005, France
| | - Philippe Lory
- From the Université de Montpellier, CNRS UMR 5203, Département de Physiologie, Institut de Génomique Fonctionnelle, Montpellier, F-34094 France, INSERM, U1191, Montpellier, F-34094 France, LabEx "Ion Channel Science and Therapeutics," Montpellier, F-34094 France,
| |
Collapse
|
44
|
Dawson TF, Boone AN, Senatore A, Piticaru J, Thiyagalingam S, Jackson D, Davison A, Spafford JD. Gene splicing of an invertebrate beta subunit (LCavβ) in the N-terminal and HOOK domains and its regulation of LCav1 and LCav2 calcium channels. PLoS One 2014; 9:e92941. [PMID: 24690951 PMCID: PMC3972191 DOI: 10.1371/journal.pone.0092941] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2013] [Accepted: 02/27/2014] [Indexed: 01/31/2023] Open
Abstract
The accessory beta subunit (Ca(v)β) of calcium channels first appear in the same genome as Ca(v)1 L-type calcium channels in single-celled coanoflagellates. The complexity of this relationship expanded in vertebrates to include four different possible Ca(v)β subunits (β1, β2, β3, β4) which associate with four Ca(v)1 channel isoforms (Ca(v)1.1 to Ca(v)1.4) and three Ca(v)2 channel isoforms (Ca(v)2.1 to Ca(v)2.3). Here we assess the fundamentally-shared features of the Ca(v)β subunit in an invertebrate model (pond snail Lymnaea stagnalis) that bears only three homologous genes: (LCa(v)1, LCa(v)2, and LCa(v)β). Invertebrate Ca(v)β subunits (in flatworms, snails, squid and honeybees) slow the inactivation kinetics of Ca(v)2 channels, and they do so with variable N-termini and lacking the canonical palmitoylation residues of the vertebrate β2a subunit. Alternative splicing of exon 7 of the HOOK domain is a primary determinant of a slow inactivation kinetics imparted by the invertebrate LCa(v)β subunit. LCa(v)β will also slow the inactivation kinetics of LCa(v)3 T-type channels, but this is likely not physiologically relevant in vivo. Variable N-termini have little influence on the voltage-dependent inactivation kinetics of differing invertebrate Ca(v)β subunits, but the expression pattern of N-terminal splice isoforms appears to be highly tissue specific. Molluscan LCa(v)β subunits have an N-terminal "A" isoform (coded by exons: 1a and 1b) that structurally resembles the muscle specific variant of vertebrate β1a subunit, and has a broad mRNA expression profile in brain, heart, muscle and glands. A more variable "B" N-terminus (exon 2) in the exon position of mammalian β3 and has a more brain-centric mRNA expression pattern. Lastly, we suggest that the facilitation of closed-state inactivation (e.g. observed in Ca(v)2.2 and Ca(v)β3 subunit combinations) is a specialization in vertebrates, because neither snail subunit (LCa(v)2 nor LCa(v)β) appears to be compatible with this observed property.
Collapse
Affiliation(s)
- Taylor F. Dawson
- Department of Biology, University of Waterloo, Waterloo, Ontario, Canada
| | - Adrienne N. Boone
- Department of Biology, University of Waterloo, Waterloo, Ontario, Canada
| | - Adriano Senatore
- Department of Biology, University of Waterloo, Waterloo, Ontario, Canada
| | - Joshua Piticaru
- Department of Biology, University of Waterloo, Waterloo, Ontario, Canada
| | | | - Daniel Jackson
- Institute of Genetics, School of Biology, University of Nottingham, Nottingham, United Kingdom
| | - Angus Davison
- Institute of Genetics, School of Biology, University of Nottingham, Nottingham, United Kingdom
| | - J. David Spafford
- Department of Biology, University of Waterloo, Waterloo, Ontario, Canada
| |
Collapse
|
45
|
T-type calcium channels in chronic pain: mouse models and specific blockers. Pflugers Arch 2014; 466:707-17. [PMID: 24590509 DOI: 10.1007/s00424-014-1484-4] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2014] [Accepted: 02/13/2014] [Indexed: 02/01/2023]
Abstract
Pain is a quite frequent complaint accompanying numerous pathologies. Among these pathological cases, neuropathies are retrieved with identified etiologies (chemotherapies, diabetes, surgeries…) and also more diffuse syndromes such as fibromyalgia. More broadly, pain is one of the first consequences of the majority of inherited diseases. Despite its importance for the quality of life, current pain management is limited to drugs that are either old or with a limited efficacy or that possess a bad benefit/risk ratio. As no new pharmacological concept has led to new analgesics in the last decades, the discovery of medications is needed, and to this aim the identification of new druggable targets in pain transmission is a first step. Therefore, studies of ion channels in pain pathways are extremely active. This is particularly true with ion channels in peripheral sensory neurons in dorsal root ganglia (DRG) known now to express unique sets of these channels. Moreover, both spinal and supraspinal levels are clearly important in pain modulation. Among these ion channels, we and others revealed the important role of low voltage-gated calcium channels in cellular excitability in different steps of the pain pathways. These channels, by being activated nearby resting membrane potential have biophysical characteristics suited to facilitate action potential generation and rhythmicity. In this review, we will review the current knowledge on the role of these channels in the perception and modulation of pain.
Collapse
|
46
|
Bourinet E, Altier C, Hildebrand ME, Trang T, Salter MW, Zamponi GW. Calcium-permeable ion channels in pain signaling. Physiol Rev 2014; 94:81-140. [PMID: 24382884 DOI: 10.1152/physrev.00023.2013] [Citation(s) in RCA: 240] [Impact Index Per Article: 21.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
The detection and processing of painful stimuli in afferent sensory neurons is critically dependent on a wide range of different types of voltage- and ligand-gated ion channels, including sodium, calcium, and TRP channels, to name a few. The functions of these channels include the detection of mechanical and chemical insults, the generation of action potentials and regulation of neuronal firing patterns, the initiation of neurotransmitter release at dorsal horn synapses, and the ensuing activation of spinal cord neurons that project to pain centers in the brain. Long-term changes in ion channel expression and function are thought to contribute to chronic pain states. Many of the channels involved in the afferent pain pathway are permeable to calcium ions, suggesting a role in cell signaling beyond the mere generation of electrical activity. In this article, we provide a broad overview of different calcium-permeable ion channels in the afferent pain pathway and their role in pain pathophysiology.
Collapse
|
47
|
Role of T-type channels in vasomotor function: team player or chameleon? Pflugers Arch 2014; 466:767-79. [PMID: 24482062 DOI: 10.1007/s00424-013-1430-x] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2013] [Accepted: 12/19/2013] [Indexed: 01/28/2023]
Abstract
Low-voltage-activated T-type calcium channels play an important role in regulating cellular excitability and are implicated in conditions, such as epilepsy and neuropathic pain. T-type channels, especially Cav3.1 and Cav3.2, are also expressed in the vasculature, although patch clamp studies of isolated vascular smooth muscle cells have in general failed to demonstrate these low-voltage-activated calcium currents. By contrast, the channels which are blocked by T-type channel antagonists are high-voltage activated but distinguishable from their L-type counterparts by their T-type biophysical properties and small negative shifts in activation and inactivation voltages. These changes in T-channel properties may result from vascular-specific expression of splice variants of Cav3 genes, particularly in exon 25/26 of the III-IV linker region. Recent physiological studies suggest that T-type channels make a small contribution to vascular tone at low intraluminal pressures, although the relevance of this contribution is unclear. By contrast, these channels play a larger role in vascular tone of small arterioles, which would be expected to function at lower intra-vascular pressures. Upregulation of T-type channel function following decrease in nitric oxide bioavailability and increase in oxidative stress, which occurs during cardiovascular disease, suggests that a more important role could be played by these channels in pathophysiological situations. The ability of T-type channels to be rapidly recruited to the plasma membrane, coupled with their subtype-specific localisation in signalling microdomains where they could modulate the function of calcium-dependent ion channels and pathways, provides a mechanism for rapid up- and downregulation of vasoconstriction. Future investigation into the molecules which govern these changes may illuminate novel targets for the treatment of conditions such as therapy-resistant hypertension and vasospasm.
Collapse
|
48
|
Perez-Reyes E, Lee JH. Ins and outs of T-channel structure function. Pflugers Arch 2013; 466:627-33. [PMID: 24337909 DOI: 10.1007/s00424-013-1419-5] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2013] [Accepted: 12/03/2013] [Indexed: 02/07/2023]
Abstract
We review the ins and outs of T-channel structure, focusing on the extracellular high-affinity metal-binding site and intracellular loops. The high-affinity metal-binding site was localized to repeat I of Cav3.2. Interestingly, a similar binding site was found in the high voltage-activated Cav2.3 channel where it controls the channels' voltage dependence. Histidine at position 191 has a particularly interesting role in the high-affinity binding site, and its modification plays an important role in channel regulation by pharmacological agents that alter redox reactions. The intracellular loop connecting repeats I and II plays two important roles in Cav3.2 properties: one, its gating; and two, its surface expression. These studies have also identified a highly conserved intracellular gating brake that is predicted to form a helix-loop-helix structure. We conclude that the gating brake establishes important contacts with the gating machinery, thereby stabilizing a closed state of T-channels. This interaction is disrupted by depolarization, allowing the S6 segments to open and allowing Ca(2+) ions to flow through. Studies in cultured hippocampal neurons provided novel insights into how mutations found in idiopathic generalized epilepsy patients increase seizure susceptibility by both altering T-current pacemaker currents and by activating Ca-activated transcription factors that regulate dendritic arborization. These studies reveal novel roles for T-channels to control cellular physiology.
Collapse
Affiliation(s)
- Edward Perez-Reyes
- Department of Pharmacology, University of Virginia, Charlottesville, VA, USA,
| | | |
Collapse
|
49
|
Eckle VS, Shcheglovitov A, Vitko I, Dey D, Yap CC, Winckler B, Perez-Reyes E. Mechanisms by which a CACNA1H mutation in epilepsy patients increases seizure susceptibility. J Physiol 2013; 592:795-809. [PMID: 24277868 DOI: 10.1113/jphysiol.2013.264176] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
T-type calcium channels play essential roles in regulating neuronal excitability and network oscillations in the brain. Mutations in the gene encoding Cav3.2 T-type Ca(2+) channels, CACNA1H, have been found in association with various forms of idiopathic generalized epilepsy. We and others have found that these mutations may influence neuronal excitability either by altering the biophysical properties of the channels or by increasing their surface expression. The goals of the present study were to investigate the excitability of neurons expressing Cav3.2 with the epilepsy mutation, C456S, and to elucidate the mechanisms by which it influences neuronal properties. We found that expression of the recombinant C456S channels substantially increased the excitability of cultured neurons by increasing the spontaneous firing rate and reducing the threshold for rebound burst firing. Additionally, we found that molecular determinants in the I-II loop (the region in which most childhood absence epilepsy-associated mutations are found) substantially increase the surface expression of T-channels but do not alter the relative distribution of channels into dendrites of cultured hippocampal neurons. Finally, we discovered that expression of C456S channels promoted dendritic growth and arborization. These effects were reversed to normal by either the absence epilepsy drug ethosuximide or a novel T-channel blocker, TTA-P2. As Ca(2+)-regulated transcription factors also increase dendritic development, we tested a transactivator trap assay and found that the C456S variant can induce changes in gene transcription. Taken together, our findings suggest that gain-of-function mutations in Cav3.2 T-type Ca(2+) channels increase seizure susceptibility by directly altering neuronal electrical properties and indirectly by changing gene expression.
Collapse
Affiliation(s)
- Veit-Simon Eckle
- Jordan Hall 800735, 1340 Jefferson Park Avenue, Charlottesville, VA 22908, USA.
| | | | | | | | | | | | | |
Collapse
|
50
|
Buda P, Reinbothe T, Nagaraj V, Mahdi T, Luan C, Tang Y, Axelsson AS, Li D, Rosengren AH, Renström E, Zhang E. Eukaryotic translation initiation factor 3 subunit e controls intracellular calcium homeostasis by regulation of cav1.2 surface expression. PLoS One 2013; 8:e64462. [PMID: 23737983 PMCID: PMC3667822 DOI: 10.1371/journal.pone.0064462] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2012] [Accepted: 04/15/2013] [Indexed: 01/09/2023] Open
Abstract
Inappropriate surface expression of voltage-gated Ca2+channels (CaV) in pancreatic ß-cells may contribute to the development of type 2 diabetes. First, failure to increase intracellular Ca2+ concentrations at the sites of exocytosis impedes insulin release. Furthermore, excessive Ca2+ influx may trigger cytotoxic effects. The regulation of surface expression of CaV channels in the pancreatic β-cells remains unknown. Here, we used real-time 3D confocal and TIRFM imaging, immunocytochemistry, cellular fractionation, immunoprecipitation and electrophysiology to study trafficking of L-type CaV1.2 channels upon β-cell stimulation. We found decreased surface expression of CaV1.2 and a corresponding reduction in L-type whole-cell Ca2+ currents in insulin-secreting INS-1 832/13 cells upon protracted (15–30 min) stimulation. This internalization occurs by clathrin-dependent endocytosis and could be prevented by microtubule or dynamin inhibitors. eIF3e (Eukaryotic translation initiation factor 3 subunit E) is part of the protein translation initiation complex, but its effect on translation are modest and effects in ion channel trafficking have been suggested. The factor interacted with CaV1.2 and regulated CaV1.2 traffic bidirectionally. eIF3e silencing impaired CaV1.2 internalization, which resulted in an increased intracellular Ca2+ load upon stimulation. These findings provide a mechanism for regulation of L-type CaV channel surface expression with consequences for β-cell calcium homeostasis, which will affect pancreatic β-cell function and insulin production.
Collapse
Affiliation(s)
- Pawel Buda
- Lund University Diabetes Center, Malmö, Sweden
| | | | | | - Taman Mahdi
- Lund University Diabetes Center, Malmö, Sweden
| | - Cheng Luan
- Lund University Diabetes Center, Malmö, Sweden
| | - Yunzhao Tang
- Lund University Diabetes Center, Malmö, Sweden
- Key Lab of Hormones and Development, Ministry of Health, and Metabolic Diseases Hospital, Tianjin Medical University, Tianjin, China
| | | | - Daiqing Li
- Key Lab of Hormones and Development, Ministry of Health, and Metabolic Diseases Hospital, Tianjin Medical University, Tianjin, China
| | | | - Erik Renström
- Lund University Diabetes Center, Malmö, Sweden
- * E-mail: (ER); (EZ)
| | - Enming Zhang
- Lund University Diabetes Center, Malmö, Sweden
- * E-mail: (ER); (EZ)
| |
Collapse
|