1
|
Incontro S, Musella ML, Sammari M, Di Scala C, Fantini J, Debanne D. Lipids shape brain function through ion channel and receptor modulations: physiological mechanisms and clinical perspectives. Physiol Rev 2025; 105:137-207. [PMID: 38990068 DOI: 10.1152/physrev.00004.2024] [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: 01/16/2024] [Revised: 05/28/2024] [Accepted: 07/01/2024] [Indexed: 07/12/2024] Open
Abstract
Lipids represent the most abundant molecular type in the brain, with a fat content of ∼60% of the dry brain weight in humans. Despite this fact, little attention has been paid to circumscribe the dynamic role of lipids in brain function and disease. Membrane lipids such as cholesterol, phosphoinositide, sphingolipids, arachidonic acid, and endocannabinoids finely regulate both synaptic receptors and ion channels that ensure critical neural functions. After a brief introduction on brain lipids and their respective properties, we review here their role in regulating synaptic function and ion channel activity, action potential propagation, neuronal development, and functional plasticity and their contribution in the development of neurological and neuropsychiatric diseases. We also provide possible directions for future research on lipid function in brain plasticity and diseases.
Collapse
Affiliation(s)
| | | | - Malika Sammari
- UNIS, INSERM, Aix-Marseille Université, Marseille, France
| | | | | | | |
Collapse
|
2
|
Salim C, Batsaikhan E, Kan AK, Chen H, Jee C. Nicotine Motivated Behavior in C. elegans. Int J Mol Sci 2024; 25:1634. [PMID: 38338915 PMCID: PMC10855306 DOI: 10.3390/ijms25031634] [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: 01/04/2024] [Revised: 01/20/2024] [Accepted: 01/22/2024] [Indexed: 02/12/2024] Open
Abstract
To maximize the advantages offered by Caenorhabditis elegans as a high-throughput (HTP) model for nicotine dependence studies, utilizing its well-defined neuroconnectome as a robust platform, and to unravel the genetic basis of nicotine-motivated behaviors, we established the nicotine conditioned cue preference (CCP) paradigm. Nicotine CCP enables the assessment of nicotine preference and seeking, revealing a parallel to fundamental aspects of nicotine-dependent behaviors observed in mammals. We demonstrated that nicotine-elicited cue preference in worms is mediated by nicotinic acetylcholine receptors and requires dopamine for CCP development. Subsequently, we pinpointed nAChR subunits associated with nicotine preference and validated human GWAS candidates linked to nicotine dependence involved in nAChRs. Functional validation involves assessing the loss-of-function strain of the CACNA2D3 ortholog and the knock-out (KO) strain of the CACNA2D2 ortholog, closely related to CACNA2D3 and sharing human smoking phenotypes. Our orthogonal approach substantiates the functional conservation of the α2δ subunit of the calcium channel in nicotine-motivated behavior. Nicotine CCP in C. elegans serves as a potent affirmation of the cross-species functional relevance of GWAS candidate genes involved in nicotine seeking associated with tobacco abuse, providing a streamlined yet comprehensive system for investigating intricate behavioral paradigms within a simplified and reliable framework.
Collapse
Affiliation(s)
| | | | | | | | - Changhoon Jee
- Department of Pharmacology, Addiction Science and Toxicology, College of Medicine, University of Tennessee Health Science Center, Memphis, TN 38163, USA; (C.S.)
| |
Collapse
|
3
|
Samhan-Arias AK, Poejo J, Marques-da-Silva D, Martínez-Costa OH, Gutierrez-Merino C. Are There Lipid Membrane-Domain Subtypes in Neurons with Different Roles in Calcium Signaling? Molecules 2023; 28:7909. [PMID: 37894616 PMCID: PMC10708093 DOI: 10.3390/molecules28237909] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2023] [Revised: 11/24/2023] [Accepted: 11/29/2023] [Indexed: 12/18/2023] Open
Abstract
Lipid membrane nanodomains or lipid rafts are 10-200 nm diameter size cholesterol- and sphingolipid-enriched domains of the plasma membrane, gathering many proteins with different roles. Isolation and characterization of plasma membrane proteins by differential centrifugation and proteomic studies have revealed a remarkable diversity of proteins in these domains. The limited size of the lipid membrane nanodomain challenges the simple possibility that all of them can coexist within the same lipid membrane domain. As caveolin-1, flotillin isoforms and gangliosides are currently used as neuronal lipid membrane nanodomain markers, we first analyzed the structural features of these components forming nanodomains at the plasma membrane since they are relevant for building supramolecular complexes constituted by these molecular signatures. Among the proteins associated with neuronal lipid membrane nanodomains, there are a large number of proteins that play major roles in calcium signaling, such as ionotropic and metabotropic receptors for neurotransmitters, calcium channels, and calcium pumps. This review highlights a large variation between the calcium signaling proteins that have been reported to be associated with isolated caveolin-1 and flotillin-lipid membrane nanodomains. Since these calcium signaling proteins are scattered in different locations of the neuronal plasma membrane, i.e., in presynapses, postsynapses, axonal or dendritic trees, or in the neuronal soma, our analysis suggests that different lipid membrane-domain subtypes should exist in neurons. Furthermore, we conclude that classification of lipid membrane domains by their content in calcium signaling proteins sheds light on the roles of these domains for neuronal activities that are dependent upon the intracellular calcium concentration. Some examples described in this review include the synaptic and metabolic activity, secretion of neurotransmitters and neuromodulators, neuronal excitability (long-term potentiation and long-term depression), axonal and dendritic growth but also neuronal cell survival and death.
Collapse
Affiliation(s)
- Alejandro K. Samhan-Arias
- Departamento de Bioquímica, Universidad Autónoma de Madrid (UAM), C/Arturo Duperier 4, 28029 Madrid, Spain;
- Instituto de Investigaciones Biomédicas ‘Sols-Morreale’ (CSIC-UAM), C/Arturo Duperier 4, 28029 Madrid, Spain
| | - Joana Poejo
- Instituto de Biomarcadores de Patologías Moleculares, Universidad de Extremadura, 06006 Badajoz, Spain;
| | - Dorinda Marques-da-Silva
- LSRE—Laboratory of Separation and Reaction Engineering and LCM—Laboratory of Catalysis and Materials, School of Management and Technology, Polytechnic Institute of Leiria, Morro do Lena-Alto do Vieiro, 2411-901 Leiria, Portugal;
- ALiCE—Associate Laboratory in Chemical Engineering, Faculty of Engineering, University of Porto, Rua Dr. Roberto Frias, 4200-465 Porto, Portugal
- School of Technology and Management, Polytechnic Institute of Leiria, Morro do Lena-Alto do Vieiro, 2411-901 Leiria, Portugal
| | - Oscar H. Martínez-Costa
- Departamento de Bioquímica, Universidad Autónoma de Madrid (UAM), C/Arturo Duperier 4, 28029 Madrid, Spain;
- Instituto de Investigaciones Biomédicas ‘Sols-Morreale’ (CSIC-UAM), C/Arturo Duperier 4, 28029 Madrid, Spain
| | - Carlos Gutierrez-Merino
- Instituto de Biomarcadores de Patologías Moleculares, Universidad de Extremadura, 06006 Badajoz, Spain;
| |
Collapse
|
4
|
Harada K, Inoue M. Muscarinic Receptor Stimulation Does Not Inhibit Voltage-dependent Ca 2+ Channels in Rat Adrenal Medullary Chromaffin Cells. Acta Histochem Cytochem 2023; 56:67-75. [PMID: 37680574 PMCID: PMC10480484 DOI: 10.1267/ahc.23-00042] [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: 06/05/2023] [Accepted: 07/25/2023] [Indexed: 09/09/2023] Open
Abstract
Adrenal medullary chromaffin (AMC) and sympathetic ganglion cells are derived from the neural crest and show a similar developmental path. Thus, these two cell types have many common properties in membrane excitability and signaling. However, AMC cells function as endocrine cells while sympathetic ganglion cells are neurons. In rat sympathetic ganglion cells, muscarinic M1 and M4 receptors mediate excitation and inhibition via suppression of M-type K+ channels and suppression of voltage-dependent Ca2+ channels, respectively. On the other hand, M1 receptor stimulation in rat AMC cells also produces excitation by suppressing TWIK-related acid sensitive K+ (TASK) channels. However, whether M4 receptors are coupled with voltage-dependent Ca2+ channel suppression is unclear. We explore this issue electrophysiologically and biochemically. Electrical stimulation of nerve fibers in rat adrenal glands trans-synaptically increased the Ca2+ signal in AMC cells. This electrically evoked increased Ca2+ signal was not altered during muscarine-induced increase in Ca2+ signal, whereas it decreased significantly during a GABA-induced increase, due to a shunt effect of increased Cl- conductance. The whole-cell current recordings revealed that voltage-dependent Ca2+ currents in AMC cells were suppressed by adenosine triphosphate, but not by muscarinic agonists. The fractionation analysis and immunocytochemistry indicated that CaV1.2 Ca2+ channels and M4 receptors are located in the raft and non-raft membrane domains, respectively. We concluded that muscarinic stimulation in rat AMC cells does not produce voltage-dependent Ca2+ channel inhibition. This lack of muscarinic inhibition is at least partly due to physical separation of voltage-dependent Ca2+ channels and M4 receptors in the plasma membrane.
Collapse
Affiliation(s)
- Keita Harada
- Department of Cell and Systems Physiology, University of Occupational and Environmental Health School of Medicine, Kitakyushu 807–8555, Japan
| | - Masumi Inoue
- Department of Cell and Systems Physiology, University of Occupational and Environmental Health School of Medicine, Kitakyushu 807–8555, Japan
| |
Collapse
|
5
|
Komatsuya K, Kikuchi N, Hirabayashi T, Kasahara K. The Regulatory Roles of Cerebellar Glycosphingolipid Microdomains/Lipid Rafts. Int J Mol Sci 2023; 24:ijms24065566. [PMID: 36982638 PMCID: PMC10058044 DOI: 10.3390/ijms24065566] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2023] [Revised: 03/09/2023] [Accepted: 03/11/2023] [Indexed: 03/17/2023] Open
Abstract
Lipid rafts are dynamic assemblies of glycosphingolipids, sphingomyelin, cholesterol, and specific proteins which are stabilized into platforms involved in the regulation of vital cellular processes. Cerebellar lipid rafts are cell surface ganglioside microdomains for the attachment of GPI-anchored neural adhesion molecules and downstream signaling molecules such as Src-family kinases and heterotrimeric G proteins. In this review, we summarize our recent findings on signaling in ganglioside GD3 rafts of cerebellar granule cells and several findings by other groups on the roles of lipid rafts in the cerebellum. TAG-1, of the contactin group of immunoglobulin superfamily cell adhesion molecules, is a phosphacan receptor. Phosphacan regulates the radial migration signaling of cerebellar granule cells, via Src-family kinase Lyn, by binding to TAG-1 on ganglioside GD3 rafts. Chemokine SDF-1α, which induces the tangential migration of cerebellar granule cells, causes heterotrimeric G protein Goα translocation to GD3 rafts. Furthermore, the functional roles of cerebellar raft-binding proteins including cell adhesion molecule L1, heterotrimeric G protein Gsα, and L-type voltage-dependent calcium channels are discussed.
Collapse
|
6
|
Pilch KS, Ramgoolam KH, Dolphin AC. Involvement of Ca V 2.2 channels and α 2 δ-1 in homeostatic synaptic plasticity in cultured hippocampal neurons. J Physiol 2022; 600:5333-5351. [PMID: 36377048 PMCID: PMC10107484 DOI: 10.1113/jp283600] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2022] [Accepted: 11/08/2022] [Indexed: 11/16/2022] Open
Abstract
In the mammalian brain, presynaptic CaV 2 channels play a pivotal role in synaptic transmission by mediating fast neurotransmitter exocytosis via influx of Ca2+ into the active zone of presynaptic terminals. However, the distribution and modulation of CaV 2.2 channels at plastic hippocampal synapses remains to be elucidated. Here, we assess CaV 2.2 channels during homeostatic synaptic plasticity, a compensatory form of homeostatic control preventing excessive or insufficient neuronal activity during which extensive active zone remodelling has been described. We show that chronic silencing of neuronal activity in mature hippocampal cultures resulted in elevated presynaptic Ca2+ transients, mediated by increased levels of CaV 2.2 channels at the presynaptic site. This work focused further on the role of α2 δ-1 subunits, important regulators of synaptic transmission and CaV 2.2 channel abundance at the presynaptic membrane. We found that α2 δ-1 overexpression reduces the contribution of CaV 2.2 channels to total Ca2+ flux without altering the amplitude of the Ca2+ transients. Levels of endogenous α2 δ-1 decreased during homeostatic synaptic plasticity, whereas the overexpression of α2 δ-1 prevented homeostatic synaptic plasticity in hippocampal neurons. Together, this study reveals a key role for CaV 2.2 channels and novel roles for α2 δ-1 during synaptic plastic adaptation. KEY POINTS: The roles of CaV 2.2 channels and α2 δ-1 in homeostatic synaptic plasticity in hippocampal neurons in culture were examined. Chronic silencing of neuronal activity resulted in elevated presynaptic Ca2+ transients, mediated by increased levels of CaV 2.2 channels at presynaptic sites. The level of endogenous α2 δ-1 decreased during homeostatic synaptic plasticity, whereas overexpression of α2 δ-1 prevented homeostatic synaptic plasticity in hippocampal neurons. Together, this study reveals a key role for CaV 2.2 channels and novel roles for α2 δ-1 during synaptic plastic adaptation.
Collapse
Affiliation(s)
- Kjara S. Pilch
- Department of NeurosciencePhysiology and PharmacologyUniversity College LondonLondonUK
| | - Krishma H. Ramgoolam
- Department of NeurosciencePhysiology and PharmacologyUniversity College LondonLondonUK
| | - Annette C. Dolphin
- Department of NeurosciencePhysiology and PharmacologyUniversity College LondonLondonUK
| |
Collapse
|
7
|
Self-organization and surface properties of hBest1 in models of biological membranes. Adv Colloid Interface Sci 2022; 302:102619. [PMID: 35276535 DOI: 10.1016/j.cis.2022.102619] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2021] [Revised: 02/19/2022] [Accepted: 02/20/2022] [Indexed: 11/22/2022]
Abstract
The transmembrane Ca2+ - activated Cl- channel - human bestrophin-1 (hBest1) is expressed in retinal pigment epithelium and mutations of BEST1 gene cause ocular degenerative diseases colectivelly referred to as "bestrophinopathies". A large number of genetical, biochemical, biophysical and molecular biological studies have been performed to understand the relationship between structure and function of the hBest1 protein and its pathophysiological significance. Here, we review the current understanding of hBest1 surface organization, interactions with membrane lipids in model membranes, and its association with microdomains of cellular membranes. These highlights are significant for modulation of channel activity in cells.
Collapse
|
8
|
Beeson KA, Westbrook GL, Schnell E. α2δ-2 is required for depolarization-induced suppression of excitation in Purkinje cells. J Physiol 2022; 600:111-122. [PMID: 34783012 PMCID: PMC8724408 DOI: 10.1113/jp282438] [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: 09/30/2021] [Accepted: 11/11/2021] [Indexed: 01/03/2023] Open
Abstract
α2δ proteins (CACNA2D1-4) are required for normal neurological function and contribute to membrane trafficking of voltage-gated calcium channels, through which calcium entry initiates numerous physiological processes. However, it remains unclear how α2δ proteins influence calcium-mediated signalling to control neuronal output. Using whole-cell recordings of mouse Purkinje cells, we show that α2δ-2 is required for functional coupling of postsynaptic voltage-dependent calcium entry with calcium-dependent effector mechanisms controlling two different outputs, depolarization-induced suppression of excitation and spike afterhyperpolarization. Our findings indicate an important role for α2δ-2 proteins in regulating functional postsynaptic calcium channel coupling in neurons, providing new context for understanding the effects of α2δ mutations on neuronal circuit function and presenting additional potential avenues to manipulate α2δ-mediated signalling for therapeutic gain. KEY POINTS: Calcium influx, via voltage-dependent calcium channels, drives numerous neuronal signalling processes with precision achieved in part by tight coupling between calcium entry and calcium-dependent effectors. α2δ proteins are important for neurological function and contribute to calcium channel membrane trafficking, although how α2δ proteins influence postsynaptic calcium-dependent signalling is largely unexplored. Here it is shown that loss of α2δ-2 proteins disrupts functional calcium coupling to two different postsynaptic calcium-dependent signals in mouse Purkinje cell neurons, retrograde endocannabinoid signalling and the action potential afterhyperpolarization. The findings provide new insights into the control of calcium coupling as well as new roles for α2δ-2 proteins in neurons.
Collapse
Affiliation(s)
- Kathleen A. Beeson
- Neuroscience Graduate Program, OHSU, Portland, OR, 97239,Department of Anesthesiology and Perioperative Medicine, OHSU, Portland, OR, 97239
| | | | - Eric Schnell
- Department of Anesthesiology and Perioperative Medicine, OHSU, Portland, OR, 97239,Operative Care Division, Portland VA Health Care System, Portland, OR, 97239,Eric Schnell, MD, PhD,
| |
Collapse
|
9
|
Sharma N, Tan MA, An SSA. Phytosterols: Potential Metabolic Modulators in Neurodegenerative Diseases. Int J Mol Sci 2021; 22:ijms222212255. [PMID: 34830148 PMCID: PMC8618769 DOI: 10.3390/ijms222212255] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2021] [Revised: 11/03/2021] [Accepted: 11/09/2021] [Indexed: 12/12/2022] Open
Abstract
Phytosterols constitute a class of natural products that are an important component of diet and have vast applications in foods, cosmetics, and herbal medicines. With many and diverse isolated structures in nature, they exhibit a broad range of biological and pharmacological activities. Among over 200 types of phytosterols, stigmasterol and β-sitosterol were ubiquitous in many plant species, exhibiting important aspects of activities related to neurodegenerative diseases. Hence, this mini-review presented an overview of the reported studies on selected phytosterols related to neurodegenerative diseases. It covered the major phytosterols based on biosynthetic considerations, including other phytosterols with significant in vitro and in vivo biological activities.
Collapse
Affiliation(s)
- Niti Sharma
- Bionano Research Institute, Gachon University, 1342 Seongnam-daero, Sujeong-gu, Seongnam-si 461-701, Gyeonggi-do, Korea;
| | - Mario A. Tan
- Research Center for the Natural and Applied Sciences, College of Science, University of Santo Tomas, Manila 1015, Philippines;
| | - Seong Soo A. An
- Bionano Research Institute, Gachon University, 1342 Seongnam-daero, Sujeong-gu, Seongnam-si 461-701, Gyeonggi-do, Korea;
- Correspondence: ; Tel.: +82-31-750-8755
| |
Collapse
|
10
|
Brito BE, García MA, De Gouveia YM, Bolaños P, Devis S, Bernal G, Tortorici-Brito VA, Baute L, Díaz-Serrano G, Tortorici V. Concomitant Antihyperalgesic and Antitumor Effects of Gabapentin in a Murine Cancer Pain Model. Int J Mol Sci 2021; 22:ijms22189671. [PMID: 34575835 PMCID: PMC8471802 DOI: 10.3390/ijms22189671] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 08/10/2021] [Accepted: 08/12/2021] [Indexed: 12/30/2022] Open
Abstract
Cancer pain may be the consequence of physical nerve compression by a growing tumor. We employed a murine model to study whether gabapentin was able to regulate tumor growth, in addition to controlling hyperalgesic symptoms. A fluorescent melanoma cell line (B16-BL6/Zs green) was inoculated into the proximity of the sciatic nerve in male C57BL/6 mice. The tumor gradually compressed the nerve, causing hypersensitivity. Tumor growth was characterized via in vivo imaging techniques. Every other day, gabapentin (100 mg/Kg) or saline was IP administered to each animal. In the therapeutic protocol, gabapentin was administered once the tumor had induced increased nociception. In the preventive protocol, gabapentin was administered before the appearance of the positive signs. Additionally, in vitro experiments were performed to determine gabapentin's effects on cell-line proliferation, the secretion of the chemokine CCL2, and calcium influx. In the therapeutically treated animals, baseline responses to noxious stimuli were recovered, and tumors were significantly reduced. Similarly, gabapentin reduced tumor growth during the preventive treatment, but a relapse was noticed when the administration stopped. Gabapentin also inhibited cell proliferation, the secretion of CCL2, and calcium influx. These results suggest that gabapentin might represent a multivalent strategy to control cancer-associated events in painful tumors.
Collapse
Affiliation(s)
- Beatriz Elena Brito
- Laboratorio de Patología Celular y Molecular, Centro de Medicina Experimental, Instituto Venezolano de Investigaciones Científicas (IVIC), Caracas 1020A, Venezuela; (B.E.B.); (M.A.G.); (Y.M.D.G.); (G.B.); (V.A.T.-B.); (L.B.)
| | - María Alejandra García
- Laboratorio de Patología Celular y Molecular, Centro de Medicina Experimental, Instituto Venezolano de Investigaciones Científicas (IVIC), Caracas 1020A, Venezuela; (B.E.B.); (M.A.G.); (Y.M.D.G.); (G.B.); (V.A.T.-B.); (L.B.)
| | - Yetsenia María De Gouveia
- Laboratorio de Patología Celular y Molecular, Centro de Medicina Experimental, Instituto Venezolano de Investigaciones Científicas (IVIC), Caracas 1020A, Venezuela; (B.E.B.); (M.A.G.); (Y.M.D.G.); (G.B.); (V.A.T.-B.); (L.B.)
| | - Pura Bolaños
- Laboratorio de Fisiología Celular, Centro de Biofísica y Bioquímica, Instituto Venezolano de Investigaciones Científicas (IVIC), Caracas 1020A, Venezuela;
| | - Sindy Devis
- Laboratorio de Neurofisiología, Centro de Biofísica y Bioquímica, Instituto Venezolano de Investigaciones Científicas (IVIC), Caracas 1020A, Venezuela; (S.D.); (G.D.-S.)
| | - Geraldinee Bernal
- Laboratorio de Patología Celular y Molecular, Centro de Medicina Experimental, Instituto Venezolano de Investigaciones Científicas (IVIC), Caracas 1020A, Venezuela; (B.E.B.); (M.A.G.); (Y.M.D.G.); (G.B.); (V.A.T.-B.); (L.B.)
| | - Víctor Alejandro Tortorici-Brito
- Laboratorio de Patología Celular y Molecular, Centro de Medicina Experimental, Instituto Venezolano de Investigaciones Científicas (IVIC), Caracas 1020A, Venezuela; (B.E.B.); (M.A.G.); (Y.M.D.G.); (G.B.); (V.A.T.-B.); (L.B.)
| | - Leslie Baute
- Laboratorio de Patología Celular y Molecular, Centro de Medicina Experimental, Instituto Venezolano de Investigaciones Científicas (IVIC), Caracas 1020A, Venezuela; (B.E.B.); (M.A.G.); (Y.M.D.G.); (G.B.); (V.A.T.-B.); (L.B.)
| | - Gabriel Díaz-Serrano
- Laboratorio de Neurofisiología, Centro de Biofísica y Bioquímica, Instituto Venezolano de Investigaciones Científicas (IVIC), Caracas 1020A, Venezuela; (S.D.); (G.D.-S.)
| | - Víctor Tortorici
- Laboratorio de Neurofisiología, Centro de Biofísica y Bioquímica, Instituto Venezolano de Investigaciones Científicas (IVIC), Caracas 1020A, Venezuela; (S.D.); (G.D.-S.)
- Laboratorio de Neurociencia, Departamento de Ciencias del Comportamiento, Escuela de Psicología, Universidad Metropolitana (UNIMET), Caracas 1073, Venezuela
- Correspondence: ; Tel.: +58-(212)-240-3788
| |
Collapse
|
11
|
Meyer JO, Dolphin AC. Rab11-dependent recycling of calcium channels is mediated by auxiliary subunit α 2δ-1 but not α 2δ-3. Sci Rep 2021; 11:10256. [PMID: 33986433 PMCID: PMC8119971 DOI: 10.1038/s41598-021-89820-1] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Accepted: 04/16/2021] [Indexed: 11/18/2022] Open
Abstract
N-type voltage-gated calcium channels (CaV2.2) are predominantly expressed at presynaptic terminals, and their function is regulated by auxiliary α2δ and β subunits. All four mammalian α2δ subunits enhance calcium currents through CaV1 and CaV2 channels, and this increase is attributed, in part, to increased CaV expression at the plasma membrane. In the present study we provide evidence that α2δ-1, like α2δ-2, is recycled to the plasma membrane through a Rab11a-dependent endosomal recycling pathway. Using a dominant-negative Rab11a mutant, Rab11a(S25N), we show that α2δ-1 increases plasma membrane CaV2.2 expression by increasing the rate and extent of net forward CaV2.2 trafficking in a Rab11a-dependent manner. Dominant-negative Rab11a also reduces the ability of α2δ-1 to increase CaV2.2 expression on the cell-surface of hippocampal neurites. In contrast, α2δ-3 does not enhance rapid forward CaV2.2 trafficking, regardless of whether Rab11a(S25N) is present. In addition, whole-cell CaV2.2 currents are reduced by co-expression of Rab11a(S25N) in the presence of α2δ-1, but not α2δ-3. Taken together these data suggest that α2δ subtypes participate in distinct trafficking pathways which in turn influence the localisation and function of CaV2.2.
Collapse
Affiliation(s)
- James O Meyer
- Department of Neuroscience, Physiology and Pharmacology, University College London, Gower Street, London, WC1E 6BT, UK
| | - Annette C Dolphin
- Department of Neuroscience, Physiology and Pharmacology, University College London, Gower Street, London, WC1E 6BT, UK.
| |
Collapse
|
12
|
Wright CS, Robling AG, Farach-Carson MC, Thompson WR. Skeletal Functions of Voltage Sensitive Calcium Channels. Curr Osteoporos Rep 2021; 19:206-221. [PMID: 33721180 PMCID: PMC8216424 DOI: 10.1007/s11914-020-00647-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 12/16/2020] [Indexed: 12/15/2022]
Abstract
Voltage-sensitive calcium channels (VSCCs) are ubiquitous multimeric protein complexes that are necessary for the regulation of numerous physiological processes. VSCCs regulate calcium influx and various intracellular processes including muscle contraction, neurotransmission, hormone secretion, and gene transcription, with function specificity defined by the channel's subunits and tissue location. The functions of VSCCs in bone are often overlooked since bone is not considered an electrically excitable tissue. However, skeletal homeostasis and adaptation relies heavily on VSCCs. Inhibition or deletion of VSCCs decreases osteogenesis, impairs skeletal structure, and impedes anabolic responses to mechanical loading. RECENT FINDINGS: While the functions of VSCCs in osteoclasts are less clear, VSCCs have distinct but complementary functions in osteoblasts and osteocytes. PURPOSE OF REVIEW: This review details the structure, function, and nomenclature of VSCCs, followed by a comprehensive description of the known functions of VSCCs in bone cells and their regulation of bone development, bone formation, and mechanotransduction.
Collapse
Affiliation(s)
- Christian S Wright
- Department of Physical Therapy, School of Health and Rehabilitation Sciences, Indiana University, Indianapolis, IN, 46202, USA
- Indiana Center for Musculoskeletal Health, Indiana University, Indianapolis, IN, 46202, USA
| | - Alexander G Robling
- Indiana Center for Musculoskeletal Health, Indiana University, Indianapolis, IN, 46202, USA
- Department of Anatomy & Cell Biology, Indiana University, Indianapolis, IN, 46202, USA
| | - Mary C Farach-Carson
- Department of Diagnostic & Biomedical Sciences, University of Texas Health Science Center at Houston School of Dentistry, Houston, TX, 77054, USA
| | - William R Thompson
- Department of Physical Therapy, School of Health and Rehabilitation Sciences, Indiana University, Indianapolis, IN, 46202, USA.
- Indiana Center for Musculoskeletal Health, Indiana University, Indianapolis, IN, 46202, USA.
- Department of Anatomy & Cell Biology, Indiana University, Indianapolis, IN, 46202, USA.
| |
Collapse
|
13
|
Fu M, Liu F, Zhang YY, Lin J, Huang CL, Li YL, Wang H, Zhou C, Li CJ, Shen JF. The α2δ-1-NMDAR1 interaction in the trigeminal ganglion contributes to orofacial ectopic pain following inferior alveolar nerve injury. Brain Res Bull 2021; 171:162-171. [PMID: 33811955 DOI: 10.1016/j.brainresbull.2021.03.019] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Revised: 03/21/2021] [Accepted: 03/25/2021] [Indexed: 02/05/2023]
Abstract
Orofacial ectopic pain can often arise following nerve injury. However, the exact mechanism responsible for orofacial ectopic pain induced by trigeminal nerve injury remains unknown. The α2δ-1 and glutamate N-methyl-d-aspartic acid receptor (NMDAR) interactions have been demonstrated to participate in neuropathic pain regulation in the spinal cord. In this study, a rat model of inferior alveolar nerve transection (IANX) was used to investigate the role of α2δ-1-NMDAR1 interaction in the trigeminal ganglion (TG) in regard to the regulation of orofacial ectopic pain. Western blot (WB) analysis indicated that α2δ-1 and NMDAR1 in the TG were substantially higher in IANX rats than they were in sham/naive rats. Additionally, immunofluorescence (IF) results revealed that α2δ-1 and NMDAR1 were co-expressed and distributed within neurons and activated satellite glial cells in the TG. Co-immunoprecipitation (Co-IP) results indicated that α2δ-1-NMDAR1 complex levels in the TG were higher in IANX rats than they were in sham rats. Furthermore, the results of behavioral tests demonstrated that intra-TG injection of gabapentin (α2δ-1 inhibitory ligand) or memantine hydrochloride (NMDAR antagonist) reversed the decrease in mechanical head-withdrawal threshold (HWT) in IANX rats. Moreover, inhibition of α2δ-1 by intra-TG administration of gabapentin suppressed the upregulation of the NMDAR1 protein, and the inhibition of NMDAR by intra-TG administration of memantine hydrochloride inhibited the increased expression of α2δ-1 protein induced by IANX. In conclusion, the physical and functional interaction between α2δ-1 and NMDAR1 is critical for the development of orofacial ectopic pain, indicating that α2δ-1, NMDAR1, and the α2δ-1-NMDAR1 complex may represent potential targets for the treatment of orofacial ectopic pain.
Collapse
Affiliation(s)
- Min Fu
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China; Department of Prosthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Fei Liu
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China; Department of Prosthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Yan-Yan Zhang
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China; Department of Prosthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Jiu Lin
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China; Department of Prosthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Chao-Lan Huang
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China; Department of Prosthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Yue-Ling Li
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China; Department of Prosthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Hang Wang
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China; Department of Prosthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Cheng Zhou
- Laboratory of Anesthesia and Critical Care Medicine, Translational Neuroscience Center, West China Hospital of Sichuan University, Chengdu, China
| | - Chun-Jie Li
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China; Department of Head and Neck Oncology, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Jie-Fei Shen
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China; Department of Prosthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China.
| |
Collapse
|
14
|
Coin JT, Vance JM. Gabapentin Relieves Vertigo of Periodic Vestibulocerebellar Ataxia: 3 Cases and Possible Mechanism. Mov Disord 2021; 36:1264-1267. [PMID: 33452831 DOI: 10.1002/mds.28491] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2020] [Revised: 11/25/2020] [Accepted: 12/21/2020] [Indexed: 11/10/2022] Open
Abstract
OBJECTIVE The aim of this study was to report relief of optokinetic-triggered vertigo (OKTV) with low-dose gabapentin in three patients with periodic vestibulocerebellar ataxia [episodic ataxia type 4 (EA4); OMIM 606552]. METHODS Clinical observations and analysis of video-recorded eye movements were used before and after gabapentin. RESULTS Gabapentin relieved vertigo of all three treated patients with EA4, particularly during activities that typically would induce vertiginous symptoms. Two patients reported 8-12 hours of sustained relief after the first 100 mg dose. One has benefited from 100-200 mg TID for 7 years. Video analysis of nystagmus revealed improved target tracking on smooth pursuit and a steadier gaze hold. CONCLUSIONS Gabapentin effectively relieved the optokinetic-triggered vertigo in our patients with EA4. Mechanisms are postulated in terms of known tight gabapentin binding to the Purkinje cell voltage-gated calcium channel. The observations may offer insight into this rare disease's neuropathology. © 2021 International Parkinson and Movement Disorder Society.
Collapse
Affiliation(s)
| | - Jeffery M Vance
- Dr. John T. Macdonald Foundation Department of Human Genetics, Department of Neurology, Hussman Institute for Human Genomics, Miller School of Medicine, University of Miami, Miami, Florida, USA
| |
Collapse
|
15
|
Different functions of two putative Drosophila α 2δ subunits in the same identified motoneurons. Sci Rep 2020; 10:13670. [PMID: 32792569 PMCID: PMC7426832 DOI: 10.1038/s41598-020-69748-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2020] [Accepted: 07/15/2020] [Indexed: 11/24/2022] Open
Abstract
Voltage gated calcium channels (VGCCs) regulate neuronal excitability and translate activity into calcium dependent signaling. The α1 subunit of high voltage activated (HVA) VGCCs associates with α2δ accessory subunits, which may affect calcium channel biophysical properties, cell surface expression, localization and transport and are thus important players in calcium-dependent signaling. In vertebrates, the functions of the different combinations of the four α2δ and the seven HVA α1 subunits are incompletely understood, in particular with respect to partially redundant or separate functions in neurons. This study capitalizes on the relatively simpler situation in the Drosophila genetic model containing two neuronal putative α2δ subunits, straightjacket and CG4587, and one Cav1 and Cav2 homolog each, both with well-described functions in different compartments of identified motoneurons. Straightjacket is required for normal Cav1 and Cav2 current amplitudes and correct Cav2 channel function in all neuronal compartments. By contrast, CG4587 does not affect Cav1 or Cav2 current amplitudes or presynaptic function, but is required for correct Cav2 channel allocation to the axonal versus the dendritic domain. We suggest that the two different putative α2δ subunits are required in the same neurons to regulate different functions of VGCCs.
Collapse
|
16
|
Sun W, Larson MJ, Kiyoshi CM, Annett AJ, Stalker WA, Peng J, Tedeschi A. Gabapentinoid treatment promotes corticospinal plasticity and regeneration following murine spinal cord injury. J Clin Invest 2020; 130:345-358. [PMID: 31793909 DOI: 10.1172/jci130391] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2019] [Accepted: 10/02/2019] [Indexed: 02/06/2023] Open
Abstract
Axon regeneration failure causes neurological deficits and long-term disability after spinal cord injury (SCI). Here, we found that the α2δ2 subunit of voltage-gated calcium channels negatively regulates axon growth and regeneration of corticospinal neurons, the cells that originate the corticospinal tract. Increased α2δ2 expression in corticospinal neurons contributed to loss of corticospinal regrowth ability during postnatal development and after SCI. In contrast, α2δ2 pharmacological blockade through gabapentin administration promoted corticospinal structural plasticity and regeneration in adulthood. Using an optogenetic strategy combined with in vivo electrophysiological recording, we demonstrated that regenerating corticospinal axons functionally integrate into spinal circuits. Mice administered gabapentin recovered upper extremity function after cervical SCI. Importantly, such recovery relies on reorganization of the corticospinal pathway, as chemogenetic silencing of injured corticospinal neurons transiently abrogated recovery. Thus, targeting α2δ2 with a clinically relevant treatment strategy aids repair of motor circuits after SCI.
Collapse
Affiliation(s)
- Wenjing Sun
- Department of Neuroscience, Wexner Medical Center
| | | | | | | | | | - Juan Peng
- Center for Biostatistics and Bioinformatics, and
| | - Andrea Tedeschi
- Department of Neuroscience, Wexner Medical Center.,Chronic Brain Injury Program, The Ohio State University, Columbus, Ohio, USA
| |
Collapse
|
17
|
Ablinger C, Geisler SM, Stanika RI, Klein CT, Obermair GJ. Neuronal α 2δ proteins and brain disorders. Pflugers Arch 2020; 472:845-863. [PMID: 32607809 PMCID: PMC7351808 DOI: 10.1007/s00424-020-02420-2] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2020] [Revised: 06/08/2020] [Accepted: 06/12/2020] [Indexed: 01/31/2023]
Abstract
α2δ proteins are membrane-anchored extracellular glycoproteins which are abundantly expressed in the brain and the peripheral nervous system. They serve as regulatory subunits of voltage-gated calcium channels and, particularly in nerve cells, regulate presynaptic and postsynaptic functions independently from their role as channel subunits. α2δ proteins are the targets of the widely prescribed anti-epileptic and anti-allodynic drugs gabapentin and pregabalin, particularly for the treatment of neuropathic pain conditions. Recently, the human genes (CACNA2D1-4) encoding for the four known α2δ proteins (isoforms α2δ-1 to α2δ-4) have been linked to a large variety of neurological and neuropsychiatric disorders including epilepsy, autism spectrum disorders, bipolar disorders, schizophrenia, and depressive disorders. Here, we provide an overview of the hitherto identified disease associations of all known α2δ genes, hypothesize on the pathophysiological mechanisms considering their known physiological roles, and discuss the most immanent future research questions. Elucidating their specific physiological and pathophysiological mechanisms may open the way for developing entirely novel therapeutic paradigms for treating brain disorders.
Collapse
Affiliation(s)
- Cornelia Ablinger
- Institute of Physiology, Medical University Innsbruck, 6020, Innsbruck, Austria
| | - Stefanie M Geisler
- Department of Pharmacology and Toxicology, University of Innsbruck, 6020, Innsbruck, Austria
| | - Ruslan I Stanika
- Division Physiology, Karl Landsteiner University of Health Sciences, 3500, Krems, Austria
| | - Christian T Klein
- Department of Life Sciences, IMC University of Applied Sciences, 3500, Krems, Austria
| | - Gerald J Obermair
- Institute of Physiology, Medical University Innsbruck, 6020, Innsbruck, Austria.
- Division Physiology, Karl Landsteiner University of Health Sciences, 3500, Krems, Austria.
| |
Collapse
|
18
|
Taylor CP, Harris EW. Analgesia with Gabapentin and Pregabalin May Involve N-Methyl-d-Aspartate Receptors, Neurexins, and Thrombospondins. J Pharmacol Exp Ther 2020; 374:161-174. [DOI: 10.1124/jpet.120.266056] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2020] [Accepted: 04/17/2020] [Indexed: 11/22/2022] Open
|
19
|
Dolphin AC, Lee A. Presynaptic calcium channels: specialized control of synaptic neurotransmitter release. Nat Rev Neurosci 2020; 21:213-229. [PMID: 32161339 PMCID: PMC7873717 DOI: 10.1038/s41583-020-0278-2] [Citation(s) in RCA: 126] [Impact Index Per Article: 31.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/12/2020] [Indexed: 11/09/2022]
Abstract
Chemical synapses are heterogeneous junctions formed between neurons that are specialized for the conversion of electrical impulses into the exocytotic release of neurotransmitters. Voltage-gated Ca2+ channels play a pivotal role in this process as they are the major conduits for the Ca2+ ions that trigger the fusion of neurotransmitter-containing vesicles with the presynaptic membrane. Alterations in the intrinsic function of these channels and their positioning within the active zone can profoundly alter the timing and strength of synaptic output. Advances in optical and electron microscopic imaging, structural biology and molecular techniques have facilitated recent breakthroughs in our understanding of the properties of voltage-gated Ca2+ channels that support their presynaptic functions. Here we examine the nature of these channels, how they are trafficked to and anchored within presynaptic boutons, and the mechanisms that allow them to function optimally in shaping the flow of information through neural circuits.
Collapse
Affiliation(s)
- Annette C Dolphin
- Department of Neuroscience, Physiology and Pharmacology, University College London, London, UK.
| | - Amy Lee
- Department of Molecular Physiology and Biophysics, University of Iowa, Iowa City, IA, USA.
| |
Collapse
|
20
|
Moshal KS, Roder K, Kabakov AY, Werdich AA, Yi-Eng Chiang D, Turan NN, Xie A, Kim TY, Cooper LL, Lu Y, Zhong M, Li W, Terentyev D, Choi BR, Karma A, MacRae CA, Koren G. LITAF (Lipopolysaccharide-Induced Tumor Necrosis Factor) Regulates Cardiac L-Type Calcium Channels by Modulating NEDD (Neural Precursor Cell Expressed Developmentally Downregulated Protein) 4-1 Ubiquitin Ligase. CIRCULATION. GENOMIC AND PRECISION MEDICINE 2019; 12:407-420. [PMID: 31462068 PMCID: PMC6750970 DOI: 10.1161/circgen.119.002641] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/13/2019] [Accepted: 07/19/2019] [Indexed: 12/13/2022]
Abstract
BACKGROUND The turnover of cardiac ion channels underlying action potential duration is regulated by ubiquitination. Genome-wide association studies of QT interval identified several single-nucleotide polymorphisms located in or near genes involved in protein ubiquitination. A genetic variant upstream of LITAF (lipopolysaccharide-induced tumor necrosis factor) gene prompted us to determine its role in modulating cardiac excitation. METHODS Optical mapping was performed in zebrafish hearts to determine Ca2+ transients. Live-cell confocal calcium imaging was performed on adult rabbit cardiomyocytes to determine intracellular Ca2+handling. L-type calcium channel (LTCC) current (ICa,L) was measured using whole-cell recording. To study the effect of LITAF on Cav1.2 (L-type voltage-gated calcium channel 1.2) channel expression, surface biotinylation, and Westerns were performed. LITAF interactions were studied using coimmunoprecipitation and in situ proximity ligation assay. RESULTS LITAF knockdown in zebrafish resulted in a robust increase in calcium transients. Overexpressed LITAF in 3-week-old rabbit cardiomyocytes resulted in a decrease in ICa,L and Cavα1c abundance, whereas LITAF knockdown increased ICa,L and Cavα1c protein. LITAF-overexpressing decreases calcium transients in adult rabbit cardiomyocytes, which was associated with lower Cavα1c levels. In tsA201 cells, overexpressed LITAF downregulated total and surface pools of Cavα1c via increased Cavα1c ubiquitination and its subsequent lysosomal degradation. We observed colocalization between LITAF and LTCC in tsA201 and cardiomyocytes. In tsA201, NEDD (neural precursor cell expressed developmentally downregulated protein) 4-1, but not its catalytically inactive form NEDD4-1-C867A, increased Cavα1c ubiquitination. Cavα1c ubiquitination was further increased by coexpressed LITAF and NEDD4-1 but not NEDD4-1-C867A. NEDD4-1 knockdown abolished the negative effect of LITAF on ICa,L and Cavα1c levels in 3-week-old rabbit cardiomyocytes. Computer simulations demonstrated that a decrease of ICa,L current associated with LITAF overexpression simultaneously shortened action potential duration and decreased calcium transients in rabbit cardiomyocytes. CONCLUSIONS LITAF acts as an adaptor protein promoting NEDD4-1-mediated ubiquitination and subsequent degradation of LTCC, thereby controlling LTCC membrane levels and function and thus cardiac excitation.
Collapse
Affiliation(s)
- Karni S. Moshal
- Cardiovascular Research Center, Division of Cardiology, Dept of Medicine, Rhode Island Hospital, The Warren Alpert Medical School, Brown Univ, Providence, RI
| | - Karim Roder
- Cardiovascular Research Center, Division of Cardiology, Dept of Medicine, Rhode Island Hospital, The Warren Alpert Medical School, Brown Univ, Providence, RI
| | - Anatoli Y. Kabakov
- Cardiovascular Research Center, Division of Cardiology, Dept of Medicine, Rhode Island Hospital, The Warren Alpert Medical School, Brown Univ, Providence, RI
| | - Andreas A. Werdich
- Cardiovascular Division, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA
| | - David Yi-Eng Chiang
- Cardiovascular Division, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA
| | - Nilüfer N. Turan
- Cardiovascular Research Center, Division of Cardiology, Dept of Medicine, Rhode Island Hospital, The Warren Alpert Medical School, Brown Univ, Providence, RI
| | - An Xie
- Cardiovascular Research Center, Division of Cardiology, Dept of Medicine, Rhode Island Hospital, The Warren Alpert Medical School, Brown Univ, Providence, RI
| | - Tae Yun Kim
- Cardiovascular Research Center, Division of Cardiology, Dept of Medicine, Rhode Island Hospital, The Warren Alpert Medical School, Brown Univ, Providence, RI
| | | | - Yichun Lu
- Cardiovascular Research Center, Division of Cardiology, Dept of Medicine, Rhode Island Hospital, The Warren Alpert Medical School, Brown Univ, Providence, RI
| | - Mingwang Zhong
- Physics Dept & Center for Interdisciplinary Research in Complex Systems, Northeastern Univ, Boston, MA
| | - Weiyan Li
- Cardiovascular Research Center, Division of Cardiology, Dept of Medicine, Rhode Island Hospital, The Warren Alpert Medical School, Brown Univ, Providence, RI
| | - Dmitry Terentyev
- Cardiovascular Research Center, Division of Cardiology, Dept of Medicine, Rhode Island Hospital, The Warren Alpert Medical School, Brown Univ, Providence, RI
| | - Bum-Rak Choi
- Cardiovascular Research Center, Division of Cardiology, Dept of Medicine, Rhode Island Hospital, The Warren Alpert Medical School, Brown Univ, Providence, RI
| | - Alain Karma
- Physics Dept & Center for Interdisciplinary Research in Complex Systems, Northeastern Univ, Boston, MA
| | - Calum A. MacRae
- Cardiovascular Division, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA
| | - Gideon Koren
- Cardiovascular Research Center, Division of Cardiology, Dept of Medicine, Rhode Island Hospital, The Warren Alpert Medical School, Brown Univ, Providence, RI
| |
Collapse
|
21
|
Abstract
Voltage-gated calcium (CaV) channels are associated with β and α2δ auxiliary subunits. This review will concentrate on the function of the α2δ protein family, which has four members. The canonical role for α2δ subunits is to convey a variety of properties on the CaV1 and CaV2 channels, increasing the density of these channels in the plasma membrane and also enhancing their function. More recently, a diverse spectrum of non-canonical interactions for α2δ proteins has been proposed, some of which involve competition with calcium channels for α2δ or increase α2δ trafficking and others which mediate roles completely unrelated to their calcium channel function. The novel roles for α2δ proteins which will be discussed here include association with low-density lipoprotein receptor-related protein 1 (LRP1), thrombospondins, α-neurexins, prion proteins, large conductance (big) potassium (BK) channels, and N-methyl-d-aspartate (NMDA) receptors.
Collapse
Affiliation(s)
- Annette C Dolphin
- Department of Neuroscience, Physiology and Pharmacology, University College London, Gower Street, London, WC1E 6BT, UK
| |
Collapse
|
22
|
|
23
|
Kato AS, Witkin JM. Protein complexes as psychiatric and neurological drug targets. Biochem Pharmacol 2018; 151:263-281. [PMID: 29330067 DOI: 10.1016/j.bcp.2018.01.018] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2017] [Accepted: 01/05/2018] [Indexed: 12/25/2022]
Abstract
The need for improved medications for psychiatric and neurological disorders is clear. Difficulties in finding such drugs demands that all strategic means be utilized for their invention. The discovery of forebrain specific AMPA receptor antagonists, which selectively block the specific combinations of principal and auxiliary subunits present in forebrain regions but spare targets in the cerebellum, was recently disclosed. This discovery raised the possibility that other auxiliary protein systems could be utilized to help identify new medicines. Discussion of the TARP-dependent AMPA receptor antagonists has been presented elsewhere. Here we review the diversity of protein complexes of neurotransmitter receptors in the nervous system to highlight the broad range of protein/protein drug targets. We briefly outline the structural basis of protein complexes as drug targets for G-protein-coupled receptors, voltage-gated ion channels, and ligand-gated ion channels. This review highlights heterodimers, subunit-specific receptor constructions, multiple signaling pathways, and auxiliary proteins with an emphasis on the later. We conclude that the use of auxiliary proteins in chemical compound screening could enhance the detection of specific, targeted drug searches and lead to novel and improved medicines for psychiatric and neurological disorders.
Collapse
Affiliation(s)
- Akihiko S Kato
- Neuroscience Discovery, Lilly Research Labs, Eli Lilly and Company, Indianapolis, IN, USA.
| | - Jeffrey M Witkin
- Neuroscience Discovery, Lilly Research Labs, Eli Lilly and Company, Indianapolis, IN, USA
| |
Collapse
|
24
|
3D nerve cell cultures and complex physiological relevance. Drug Discov Today 2017; 23:22-25. [PMID: 29074438 DOI: 10.1016/j.drudis.2017.10.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2017] [Revised: 10/01/2017] [Accepted: 10/16/2017] [Indexed: 11/20/2022]
Abstract
The field of tissue engineering has not yet provided knowledge on which a consensus for the complex physiological relevance (CPR) of neuronal cultures could be established. The CPR of 3D neuronal cultures can have a profound impact on the drug discovery process through the validation of in vitro models for the study of neuropsychiatric and degenerative diseases, as well as screening for neurotoxicity during drug development. Herein, we assemble evidence in support of the potential of [Ca2+]i oscillation frequency as a CPR outcome that can demonstrate the in vivo-like behavior of 3D cultures and differentiate them from 2D monolayers. We demonstrate that [Ca2+]i oscillation frequencies in 2D cultures are significantly higher than those found in 3D cultures, and provide a possible molecular explanation.
Collapse
|
25
|
Scott MB, Kammermeier PJ. Ca V2 channel subtype expression in rat sympathetic neurons is selectively regulated by α 2δ subunits. Channels (Austin) 2017; 11:555-573. [PMID: 28837380 DOI: 10.1080/19336950.2017.1356954] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
Type two voltage gated calcium (CaV2) channels are the primary mediators of neurotransmission at neuronal presynapses, but their function at neural soma is also important in regulating excitability. 1 Mechanisms that regulate CaV2 channel expression at synapses have been studied extensively, which motivated us to perform similar studies in the soma. Rat sympathetic neurons from the superior cervical ganglion (SCG) natively express CaV2.2 and CaV2.3. 2 We noted previously that heterologous expression of CaV2.1 but not CaV2.2 results in increased calcium current in SCG neurons. 3 In the present study, we extended these observations to show that both CaV2.1 and CaV2.3 expression resulted in increased calcium currents while CaV2.2 expression did not. Further, CaV2.1 could displace native CaV2.2 channels, but CaV2.3 expression could not. Heterologous expression of the individual accessory subunits α2δ-1, α2δ-2, α2δ-3, or β4 alone failed to increase current density, suggesting that the calcium current ceiling when CaV2.2 was over-expressed was not due to lack of these subunits. Interestingly, introduction of recombinant α2δ subunits produced surprising effects on displacement of native CaV2.2 by recombinant channels. Both α2δ-1 and α2δ-2 seemed to promote CaV2.2 displacement by recombinant channel expression, while α2δ-3 appeared to protect CaV2.2 from displacement. Thus, we observe a selective prioritization of CaV channel functional expression in neurons by specific α2δ subunits. These data highlight a new function for α2δ subtypes that could shed light on subtype selectivity of CaV2 membrane expression.
Collapse
Affiliation(s)
- Mallory B Scott
- a Departments of Biochemistry and Biophysics , University of Rochester Medical Center , Rochester , NY , USA
| | - Paul J Kammermeier
- b Departments of Pharmacology and Physiology , University of Rochester Medical Center , Rochester , NY , USA
| |
Collapse
|
26
|
Dorgans K, Salvi J, Bertaso F, Bernard L, Lory P, Doussau F, Mezghrani A. Characterization of the dominant inheritance mechanism of Episodic Ataxia type 2. Neurobiol Dis 2017; 106:110-123. [PMID: 28688851 DOI: 10.1016/j.nbd.2017.07.004] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2016] [Revised: 06/30/2017] [Accepted: 07/04/2017] [Indexed: 12/01/2022] Open
Abstract
Episodic Ataxia type 2 (EA2) is an autosomal dominant neuronal disorder linked to mutations in the Cav2.1 subunit of P/Q-type calcium channels. In vitro studies have established that EA2 mutations induce loss of channel activity and that EA2 mutants can exert a dominant negative effect, suppressing normal Cav2.1 activity through protein misfolding and trafficking defects. To date, the role of this mechanism in the disease pathogenesis is unknown because no animal model exists. To address this issue, we have generated a mouse bearing the R1497X nonsense mutation in Cav2.1 (Cav2.1R1497X). Phenotypic analysis of heterozygous Cav2.1R1497X mice revealed ataxia associated with muscle weakness and generalized absence epilepsy. Electrophysiological studies of the cerebellar circuits in heterozygous Cav2.1R1497X mice highlighted severe dysregulations in synaptic transmission of the two major excitatory inputs as well as alteration of the spontaneous activity of Purkinje cells. Moreover, these neuronal dysfunctions were associated with a strong suppression of Cav2.1 channel expression in the cerebellum of heterozygous Cav2.1R1497X mice. Finally, the presence of Cav2.1 in cerebellar lipid raft microdomains was strongly impaired in heterozygous Cav2.1R1497X mice. Altogether, these results reveal a pathogenic mechanism for EA2 based on a dominant negative activity of mutant channels.
Collapse
Affiliation(s)
- Kevin Dorgans
- Institut des Neurosciences Cellulaires et Intégratives-INCI CNRS-UPR 3212, 5 rue Blaise Pascal, 67084 Strasbourg, France
| | - Julie Salvi
- Institut de Génomique Fonctionnelle, CNRS UMR 5203, INSERM U1191, Université de Montpellier, LabEx 'Ion Channel Science and Therapeutics', 141 rue de la Cardonille, 34094 Montpellier, France
| | - Federica Bertaso
- Institut de Génomique Fonctionnelle, CNRS UMR 5203, INSERM U1191, Université de Montpellier, LabEx 'Ion Channel Science and Therapeutics', 141 rue de la Cardonille, 34094 Montpellier, France
| | - Ludivine Bernard
- Institut de Génomique Fonctionnelle, CNRS UMR 5203, INSERM U1191, Université de Montpellier, LabEx 'Ion Channel Science and Therapeutics', 141 rue de la Cardonille, 34094 Montpellier, France
| | - Philippe Lory
- Institut de Génomique Fonctionnelle, CNRS UMR 5203, INSERM U1191, Université de Montpellier, LabEx 'Ion Channel Science and Therapeutics', 141 rue de la Cardonille, 34094 Montpellier, France.
| | - Frederic Doussau
- Institut des Neurosciences Cellulaires et Intégratives-INCI CNRS-UPR 3212, 5 rue Blaise Pascal, 67084 Strasbourg, France
| | - Alexandre Mezghrani
- Institut des Neurosciences de Montpellier, INSERM U1051, Hôpital Saint Eloi - Bâtiment INM, 80 rue Augustin Fliche, 34091 Montpellier, France.
| |
Collapse
|
27
|
Unravelling the complexities of vascular smooth muscle ion channels: Fine tuning of activity by ancillary subunits. Pharmacol Ther 2017; 178:57-66. [PMID: 28336473 DOI: 10.1016/j.pharmthera.2017.03.010] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Which ion channel is the most important for regulating vascular tone? Which one is responsible for controlling the resting membrane potential or repolarization? Which channels are recruited by different intracellular signalling pathways or change in certain vascular diseases? Many different ion channels have been identified in the vasculature over the years and claimed as future therapeutic targets. Unfortunately, several of these ion channels are not just found in the vasculature, with many of them also found to have prominent functional roles in different organs of the body, which then leads to off-target effects. As cardiovascular diseases are expected to increase worldwide to epidemic proportions, ion channel research and the hunt for the next major therapeutic target to treat different vascular diseases has never been more important. However, I believe that the question we should now be asking is: which ancillary subunits are involved in regulating specific ion channels in the vasculature and do they have the potential to be new therapeutic targets?
Collapse
|
28
|
LRP1 influences trafficking of N-type calcium channels via interaction with the auxiliary α 2δ-1 subunit. Sci Rep 2017; 7:43802. [PMID: 28256585 PMCID: PMC5335561 DOI: 10.1038/srep43802] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2016] [Accepted: 01/30/2017] [Indexed: 01/18/2023] Open
Abstract
Voltage-gated Ca2+ (CaV) channels consist of a pore-forming α1 subunit, which determines the main functional and pharmacological attributes of the channel. The CaV1 and CaV2 channels are associated with auxiliary β- and α2δ-subunits. The molecular mechanisms involved in α2δ subunit trafficking, and the effect of α2δ subunits on trafficking calcium channel complexes remain poorly understood. Here we show that α2δ-1 is a ligand for the Low Density Lipoprotein (LDL) Receptor-related Protein-1 (LRP1), a multifunctional receptor which mediates trafficking of cargoes. This interaction with LRP1 is direct, and is modulated by the LRP chaperone, Receptor-Associated Protein (RAP). LRP1 regulates α2δ binding to gabapentin, and influences calcium channel trafficking and function. Whereas LRP1 alone reduces α2δ-1 trafficking to the cell-surface, the LRP1/RAP combination enhances mature glycosylation, proteolytic processing and cell-surface expression of α2δ-1, and also increase plasma-membrane expression and function of CaV2.2 when co-expressed with α2δ-1. Furthermore RAP alone produced a small increase in cell-surface expression of CaV2.2, α2δ-1 and the associated calcium currents. It is likely to be interacting with an endogenous member of the LDL receptor family to have these effects. Our findings now provide a key insight and new tools to investigate the trafficking of calcium channel α2δ subunits.
Collapse
|
29
|
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: 2.7] [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
|
30
|
Yoshimoto H, Takeo T, Irie T, Nakagata N. Fertility of cold-stored mouse sperm is recovered by promoting acrosome reaction and hyperactivation after cholesterol efflux by methyl-beta-cyclodextrin. Biol Reprod 2017; 96:446-455. [DOI: 10.1095/biolreprod.116.142901] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2016] [Revised: 12/24/2016] [Accepted: 01/04/2017] [Indexed: 12/17/2022] Open
|
31
|
Kadurin I, Ferron L, Rothwell SW, Meyer JO, Douglas LR, Bauer CS, Lana B, Margas W, Alexopoulos O, Nieto-Rostro M, Pratt WS, Dolphin AC. Proteolytic maturation of α 2δ represents a checkpoint for activation and neuronal trafficking of latent calcium channels. eLife 2016; 5. [PMID: 27782881 PMCID: PMC5092059 DOI: 10.7554/elife.21143] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2016] [Accepted: 10/25/2016] [Indexed: 12/23/2022] Open
Abstract
The auxiliary α2δ subunits of voltage-gated calcium channels are extracellular membrane-associated proteins, which are post-translationally cleaved into disulfide-linked polypeptides α2 and δ. We now show, using α2δ constructs containing artificial cleavage sites, that this processing is an essential step permitting voltage-dependent activation of plasma membrane N-type (CaV2.2) calcium channels. Indeed, uncleaved α2δ inhibits native calcium currents in mammalian neurons. By inducing acute cell-surface proteolytic cleavage of α2δ, voltage-dependent activation of channels is promoted, independent from the trafficking role of α2δ. Uncleaved α2δ does not support trafficking of CaV2.2 channel complexes into neuronal processes, and inhibits Ca2+ entry into synaptic boutons, and we can reverse this by controlled intracellular proteolytic cleavage. We propose a model whereby uncleaved α2δ subunits maintain immature calcium channels in an inhibited state. Proteolytic processing of α2δ then permits voltage-dependent activation of the channels, acting as a checkpoint allowing trafficking only of mature calcium channel complexes into neuronal processes. DOI:http://dx.doi.org/10.7554/eLife.21143.001
Collapse
Affiliation(s)
- Ivan Kadurin
- Department of Neuroscience, Physiology and Pharmacology, University College London, London, United Kingdom
| | - Laurent Ferron
- Department of Neuroscience, Physiology and Pharmacology, University College London, London, United Kingdom
| | - Simon W Rothwell
- Department of Neuroscience, Physiology and Pharmacology, University College London, London, United Kingdom
| | - James O Meyer
- Department of Neuroscience, Physiology and Pharmacology, University College London, London, United Kingdom
| | - Leon R Douglas
- Department of Neuroscience, Physiology and Pharmacology, University College London, London, United Kingdom
| | - Claudia S Bauer
- Department of Neuroscience, Physiology and Pharmacology, University College London, London, United Kingdom
| | - Beatrice Lana
- Department of Neuroscience, Physiology and Pharmacology, University College London, London, United Kingdom
| | - Wojciech Margas
- Department of Neuroscience, Physiology and Pharmacology, University College London, London, United Kingdom
| | - Orpheas Alexopoulos
- Department of Neuroscience, Physiology and Pharmacology, University College London, London, United Kingdom
| | - Manuela Nieto-Rostro
- Department of Neuroscience, Physiology and Pharmacology, University College London, London, United Kingdom
| | - Wendy S Pratt
- Department of Neuroscience, Physiology and Pharmacology, University College London, London, United Kingdom
| | - Annette C Dolphin
- Department of Neuroscience, Physiology and Pharmacology, University College London, London, United Kingdom
| |
Collapse
|
32
|
Tedeschi A, Dupraz S, Laskowski CJ, Xue J, Ulas T, Beyer M, Schultze JL, Bradke F. The Calcium Channel Subunit Alpha2delta2 Suppresses Axon Regeneration in the Adult CNS. Neuron 2016; 92:419-434. [PMID: 27720483 DOI: 10.1016/j.neuron.2016.09.026] [Citation(s) in RCA: 169] [Impact Index Per Article: 21.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2016] [Revised: 07/21/2016] [Accepted: 09/01/2016] [Indexed: 12/22/2022]
Abstract
Injuries to the adult CNS often result in permanent disabilities because neurons lose the ability to regenerate their axon during development. Here, whole transcriptome sequencing and bioinformatics analysis followed by gain- and loss-of-function experiments identified Cacna2d2, the gene encoding the Alpha2delta2 subunit of voltage-gated calcium channels (VGCCs), as a developmental switch that limits axon growth and regeneration. Cacna2d2 gene deletion or silencing promoted axon growth in vitro. In vivo, Alpha2delta2 pharmacological blockade through Pregabalin (PGB) administration enhanced axon regeneration in adult mice after spinal cord injury (SCI). As PGB is already an established treatment for a wide range of neurological disorders, our findings suggest that targeting Alpha2delta2 may be a novel treatment strategy to promote structural plasticity and regeneration following CNS trauma.
Collapse
Affiliation(s)
- Andrea Tedeschi
- Axonal Growth and Regeneration, German Center for Neurodegenerative Diseases, 53175 Bonn, Germany
| | - Sebastian Dupraz
- Axonal Growth and Regeneration, German Center for Neurodegenerative Diseases, 53175 Bonn, Germany
| | - Claudia J Laskowski
- Axonal Growth and Regeneration, German Center for Neurodegenerative Diseases, 53175 Bonn, Germany
| | - Jia Xue
- Genomics and Immunoregulation, LIMES-Institute, University of Bonn, 53115 Bonn, Germany
| | - Thomas Ulas
- Genomics and Immunoregulation, LIMES-Institute, University of Bonn, 53115 Bonn, Germany
| | - Marc Beyer
- Genomics and Immunoregulation, LIMES-Institute, University of Bonn, 53115 Bonn, Germany
| | - Joachim L Schultze
- Genomics and Immunoregulation, LIMES-Institute, University of Bonn, 53115 Bonn, Germany; Platform for Single Cell Genomics and Epigenomics, German Center for Neurodegenerative Diseases, 53175 Bonn, Germany
| | - Frank Bradke
- Axonal Growth and Regeneration, German Center for Neurodegenerative Diseases, 53175 Bonn, Germany.
| |
Collapse
|
33
|
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: 218] [Impact Index Per Article: 27.3] [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
|
34
|
Lana B, Page KM, Kadurin I, Ho S, Nieto-Rostro M, Dolphin AC. Thrombospondin-4 reduces binding affinity of [(3)H]-gabapentin to calcium-channel α2δ-1-subunit but does not interact with α2δ-1 on the cell-surface when co-expressed. Sci Rep 2016; 6:24531. [PMID: 27076051 PMCID: PMC4830977 DOI: 10.1038/srep24531] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2016] [Accepted: 03/30/2016] [Indexed: 01/09/2023] Open
Abstract
The α2δ proteins are auxiliary subunits of voltage-gated calcium channels, and influence their trafficking and biophysical properties. The α2δ ligand gabapentin interacts with α2δ-1, and inhibits calcium channel trafficking. However, α2-1 has also been proposed to play a synaptogenic role, independent of calcium channel function. In this regard, α2δ-1 was identified as a ligand of thrombospondins, with the interaction involving the thrombospondin synaptogenic domain and the α2δ-1 von-Willebrand-factor domain. Co-immunoprecipitation between α2δ-1 and the synaptogenic domain of thrombospondin-2 was prevented by gabapentin. We therefore examined whether interaction of thrombospondin with α2δ-1 might reciprocally influence (3)H-gabapentin binding. We concentrated on thrombospondin-4, because, like α2δ-1, it is upregulated in neuropathic pain models. We found that in membranes from cells co-transfected with α2δ-1 and thrombospondin-4, there was a Mg(2+) -dependent reduction in affinity of (3)H-gabapentin binding to α2δ-1. This effect was lost for α2δ-1 with mutations in the von-Willebrand-factor-A domain. However, the effect on (3)H-gabapentin binding was not reproduced by the synaptogenic EGF-domain of thrombospondin-4. Partial co-immunoprecipitation could be demonstrated between thrombospondin-4 and α2δ-1 when co-transfected, but there was no co-immunoprecipitation with thrombospondin-4-EGF domain. Furthermore, we could not detect any association between these two proteins on the cell-surface, indicating the demonstrated interaction occurs intracellularly.
Collapse
Affiliation(s)
- Beatrice Lana
- Department of Neuroscience, Physiology and Pharmacology, University College London, Gower St., London WC1E 6BT, United Kingdom
| | - Karen M Page
- Department of Neuroscience, Physiology and Pharmacology, University College London, Gower St., London WC1E 6BT, United Kingdom
| | - Ivan Kadurin
- Department of Neuroscience, Physiology and Pharmacology, University College London, Gower St., London WC1E 6BT, United Kingdom
| | - Shuxian Ho
- Department of Neuroscience, Physiology and Pharmacology, University College London, Gower St., London WC1E 6BT, United Kingdom
| | - Manuela Nieto-Rostro
- Department of Neuroscience, Physiology and Pharmacology, University College London, Gower St., London WC1E 6BT, United Kingdom
| | - Annette C Dolphin
- Department of Neuroscience, Physiology and Pharmacology, University College London, Gower St., London WC1E 6BT, United Kingdom
| |
Collapse
|
35
|
Patel R, Dickenson AH. Mechanisms of the gabapentinoids and α 2 δ-1 calcium channel subunit in neuropathic pain. Pharmacol Res Perspect 2016; 4:e00205. [PMID: 27069626 PMCID: PMC4804325 DOI: 10.1002/prp2.205] [Citation(s) in RCA: 161] [Impact Index Per Article: 20.1] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2015] [Revised: 11/17/2015] [Accepted: 11/19/2015] [Indexed: 12/18/2022] Open
Abstract
The gabapentinoid drugs gabapentin and pregabalin are key front‐line therapies for various neuropathies of peripheral and central origin. Originally designed as analogs of GABA, the gabapentinoids bind to the α2δ‐1 and α2δ‐2 auxiliary subunits of calcium channels, though only the former has been implicated in the development of neuropathy in animal models. Transgenic approaches also identify α2δ‐1 as key in mediating the analgesic effects of gabapentinoids, however the precise molecular mechanisms remain unclear. Here we review the current understanding of the pathophysiological role of the α2δ‐1 subunit, the mechanisms of analgesic action of gabapentinoid drugs and implications for efficacy in the clinic. Despite widespread use, the number needed to treat for gabapentin and pregabalin averages from 3 to 8 across neuropathies. The failure to treat large numbers of patients adequately necessitates a novel approach to treatment selection. Stratifying patients by sensory profiles may imply common underlying mechanisms, and a greater understanding of these mechanisms could lead to more direct targeting of gabapentinoids.
Collapse
Affiliation(s)
- Ryan Patel
- Department of Neuroscience, Physiology and Pharmacology University College London Gower Street London WC1E 6BT UK
| | - Anthony H Dickenson
- Department of Neuroscience, Physiology and Pharmacology University College London Gower Street London WC1E 6BT UK
| |
Collapse
|
36
|
Alternative Splicing in Ca(V)2.2 Regulates Neuronal Trafficking via Adaptor Protein Complex-1 Adaptor Protein Motifs. J Neurosci 2016; 35:14636-52. [PMID: 26511252 DOI: 10.1523/jneurosci.3034-15.2015] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
N-type voltage-gated calcium (Ca(V)2.2) channels are expressed in neurons and targeted to the plasma membrane of presynaptic terminals, facilitating neurotransmitter release. Here, we find that the adaptor protein complex-1 (AP-1) mediates trafficking of Ca(V)2.2 from the trans-Golgi network to the cell surface. Examination of splice variants of Ca(V)2.2, containing either exon 37a (selectively expressed in nociceptors) or 37b in the proximal C terminus, reveal that canonical AP-1 binding motifs, YxxΦ and [DE]xxxL[LI], present only in exon 37a, enhance intracellular trafficking of exon 37a-containing Ca(V)2.2 to the axons and plasma membrane of rat DRG neurons. Finally, we identify differential effects of dopamine-2 receptor (D2R) and its agonist-induced activation on trafficking of Ca(V)2.2 isoforms. D2R slowed the endocytosis of Ca(V)2.2 containing exon 37b, but not exon 37a, and activation by the agonist quinpirole reversed the effect of the D2R. Our work thus reveals key mechanisms involved in the trafficking of N-type calcium channels.
Collapse
|
37
|
Tétreault MP, Bourdin B, Briot J, Segura E, Lesage S, Fiset C, Parent L. Identification of Glycosylation Sites Essential for Surface Expression of the CaVα2δ1 Subunit and Modulation of the Cardiac CaV1.2 Channel Activity. J Biol Chem 2016; 291:4826-43. [PMID: 26742847 DOI: 10.1074/jbc.m115.692178] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2015] [Indexed: 12/15/2022] Open
Abstract
Alteration in the L-type current density is one aspect of the electrical remodeling observed in patients suffering from cardiac arrhythmias. Changes in channel function could result from variations in the protein biogenesis, stability, post-translational modification, and/or trafficking in any of the regulatory subunits forming cardiac L-type Ca(2+) channel complexes. CaVα2δ1 is potentially the most heavily N-glycosylated subunit in the cardiac L-type CaV1.2 channel complex. Here, we show that enzymatic removal of N-glycans produced a 50-kDa shift in the mobility of cardiac and recombinant CaVα2δ1 proteins. This change was also observed upon simultaneous mutation of the 16 Asn sites. Nonetheless, the mutation of only 6/16 sites was sufficient to significantly 1) reduce the steady-state cell surface fluorescence of CaVα2δ1 as characterized by two-color flow cytometry assays and confocal imaging; 2) decrease protein stability estimated from cycloheximide chase assays; and 3) prevent the CaVα2δ1-mediated increase in the peak current density and voltage-dependent gating of CaV1.2. Reversing the N348Q and N812Q mutations in the non-operational sextuplet Asn mutant protein partially restored CaVα2δ1 function. Single mutation N663Q and double mutations N348Q/N468Q, N348Q/N812Q, and N468Q/N812Q decreased protein stability/synthesis and nearly abolished steady-state cell surface density of CaVα2δ1 as well as the CaVα2δ1-induced up-regulation of L-type currents. These results demonstrate that Asn-663 and to a lesser extent Asn-348, Asn-468, and Asn-812 contribute to protein stability/synthesis of CaVα2δ1, and furthermore that N-glycosylation of CaVα2δ1 is essential to produce functional L-type Ca(2+) channels.
Collapse
Affiliation(s)
| | - Benoîte Bourdin
- From the Départment de Physiologie Moléculaire et Intégrative, Faculté de Médecine, and
| | - Julie Briot
- From the Départment de Physiologie Moléculaire et Intégrative, Faculté de Médecine, and
| | - Emilie Segura
- From the Départment de Physiologie Moléculaire et Intégrative, Faculté de Médecine, and
| | - Sylvie Lesage
- Départment de Microbiologie, Infectiologie, and Immunologie, Faculté de Médecine, Centre de Recherche de l'Hôpital Maisonneuve-Rosemont, Université de Montréal, Montréal, Québec H3C 3J7, Canada
| | - Céline Fiset
- Faculté de Pharmacie, Institut de Cardiologie de Montréal and
| | - Lucie Parent
- From the Départment de Physiologie Moléculaire et Intégrative, Faculté de Médecine, and
| |
Collapse
|
38
|
Qi Y, Andolfi L, Frattini F, Mayer F, Lazzarino M, Hu J. Membrane stiffening by STOML3 facilitates mechanosensation in sensory neurons. Nat Commun 2015; 6:8512. [PMID: 26443885 PMCID: PMC4633829 DOI: 10.1038/ncomms9512] [Citation(s) in RCA: 111] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2015] [Accepted: 09/01/2015] [Indexed: 12/21/2022] Open
Abstract
Sensing force is crucial to maintain the viability of all living cells. Despite its fundamental importance, how force is sensed at the molecular level remains largely unknown. Here we show that stomatin-like protein-3 (STOML3) controls membrane mechanics by binding cholesterol and thus facilitates force transfer and tunes the sensitivity of mechano-gated channels, including Piezo channels. STOML3 is detected in cholesterol-rich lipid rafts. In mouse sensory neurons, depletion of cholesterol and deficiency of STOML3 similarly and interdependently attenuate mechanosensitivity while modulating membrane mechanics. In heterologous systems, intact STOML3 is required to maintain membrane mechanics to sensitize Piezo1 and Piezo2 channels. In C57BL/6N, but not STOML3−/− mice, tactile allodynia is attenuated by cholesterol depletion, suggesting that membrane stiffening by STOML3 is essential for mechanical sensitivity. Targeting the STOML3–cholesterol association might offer an alternative strategy for control of chronic pain. To maintain viability, cells must be able to sense and respond to mechanical stimuli. Here, Qi et al. show that the STOML3 protein acts in mechanosensation by binding cholesterol and regulating membrane stiffness which can in turn regulate ion flux through mechanosensitive channels.
Collapse
Affiliation(s)
- Yanmei Qi
- Sensory Mechanotransduction, Centre for Integrative Neuroscience, Otfried-Mueller-Strasse 25, 72076 Tuebingen, Germany
| | - Laura Andolfi
- Istituto Officina dei Materiali Consiglio Nazionale delle Ricerche, Laboratorio TASC, 34149 Basovizza, Trieste, Italy
| | - Flavia Frattini
- Sensory Mechanotransduction, Centre for Integrative Neuroscience, Otfried-Mueller-Strasse 25, 72076 Tuebingen, Germany
| | - Florian Mayer
- Sensory Mechanotransduction, Centre for Integrative Neuroscience, Otfried-Mueller-Strasse 25, 72076 Tuebingen, Germany
| | - Marco Lazzarino
- Istituto Officina dei Materiali Consiglio Nazionale delle Ricerche, Laboratorio TASC, 34149 Basovizza, Trieste, Italy
| | - Jing Hu
- Sensory Mechanotransduction, Centre for Integrative Neuroscience, Otfried-Mueller-Strasse 25, 72076 Tuebingen, Germany
| |
Collapse
|
39
|
Sághy É, Szőke É, Payrits M, Helyes Z, Börzsei R, Erostyák J, Jánosi TZ, Sétáló Jr G, Szolcsányi J. Evidence for the role of lipid rafts and sphingomyelin in Ca2+-gating of Transient Receptor Potential channels in trigeminal sensory neurons and peripheral nerve terminals. Pharmacol Res 2015; 100:101-16. [DOI: 10.1016/j.phrs.2015.07.028] [Citation(s) in RCA: 58] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/13/2015] [Revised: 07/27/2015] [Accepted: 07/28/2015] [Indexed: 10/23/2022]
|
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: 728] [Impact Index Per Article: 80.9] [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
|
Doroudi M, Olivares-Navarrete R, Boyan BD, Schwartz Z. A review of 1α,25(OH)2D3 dependent Pdia3 receptor complex components in Wnt5a non-canonical pathway signaling. J Steroid Biochem Mol Biol 2015; 152:84-8. [PMID: 25845934 DOI: 10.1016/j.jsbmb.2015.04.002] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/12/2014] [Revised: 03/16/2015] [Accepted: 04/02/2015] [Indexed: 11/17/2022]
Abstract
Wnt5a and 1α,25-dihydroxyvitamin D3 [1α,25(OH)2D3] regulate endochondral ossification. 1α,25(OH)2D3 initiates its calcium-dependent effects via its membrane-associated receptor, protein disulfide isomerase A3 (Pdia3). 1α,25(OH)2D3 binding to Pdia3 triggers the interaction between Pdia3 and phospholipase A2 (PLA2)-activating protein (PLAA), resulting in downstream activation of calcium/calmodulin-dependent protein kinase II (CaMKII), PLA2, and protein kinase C (PKC). Wnt5a initiates its calcium-dependent effects via binding its receptors Frizzled2 (FZD2) and Frizzled5 (FZD5) and receptor tyrosine kinase-like orphan receptor 2 (ROR2), activating intracellular calcium release and stimulating PKC and CaMKII. Recent efforts to determine the inter-relation between Wnt5a and 1α,25(OH)2D3 signaling pathways have demonstrated that Wnt5a signals through a CaMKII/PLA2/PGE2/PKC cascade in chondrocytes and osteoblasts in which the components of the Pdia3 receptor complex were required. Furthermore, ROR2, but not FZD2 or FZD5, was required to mediate the calcium-dependent actions of 1α,25(OH)2D3. This review provides evidence that 1α,25(OH)2D3 and Wnt5a mediate their calcium-dependent pathways via similar receptor components and proposes that these pathways may interact since they are competing for the same receptor complex components.
Collapse
Affiliation(s)
- Maryam Doroudi
- School of Biology, Georgia Institute of Technology, Atlanta, GA 30332, USA
| | - Rene Olivares-Navarrete
- Department of Biomedical Engineering, Virginia Commonwealth University, Richmond, VA 23284, USA
| | - Barbara D Boyan
- School of Biology, Georgia Institute of Technology, Atlanta, GA 30332, USA; Department of Biomedical Engineering, Virginia Commonwealth University, Richmond, VA 23284, USA; Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA 30332, USA.
| | - Zvi Schwartz
- Department of Biomedical Engineering, Virginia Commonwealth University, Richmond, VA 23284, USA; Department of Periodontics, The University of Texas Health Science Center at San Antonio, San Antonio, TX 78284, USA
| |
Collapse
|
42
|
Hotta S, Nakatani Y, Kambe T, Abe K, Masuda Y, Utsumomiya I, Taguchi K. Effects of IgG anti-GM1 monoclonal antibodies on neuromuscular transmission and calcium channel binding in rat neuromuscular junctions. Exp Ther Med 2015; 10:535-540. [PMID: 26622350 DOI: 10.3892/etm.2015.2575] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2014] [Accepted: 05/01/2015] [Indexed: 01/24/2023] Open
Abstract
Guillain-Barré syndrome is a type of acute inflammatory neuropathy that causes ataxia and is associated with the IgG anti-GM1 antibody. However, the pathogenic role of the IgG anti-GM1 antibody and calcium channels in neuromuscular junctions (NMJs) remains unclear. Thus, the aim of the present study was to investigate the effects of the IgG anti-GM1 monoclonal antibody (mAb) on spontaneous muscle action potentials (SMAPs), and the effects of calcium channel blockers, in a rat spinal cord-muscle co-culture system. In addition, the binding of IgG anti-GM1 mAb to calcium channels was investigated in the rat hemidiaphragm. The frequency of SMAPs in the innervated muscle cells was acutely inhibited by the IgG anti-GM1 mAb; however, this effect was blocked by the N-type calcium channel blocker, ω-conotoxin GVIA (30 nM). Furthermore, the P/Q-type calcium channel blocker, ω-agatoxin IVA (10 nM), was found to partially block the IgG anti-GM1 mAb-induced inhibitory effect in the spinal cord-muscle co-culture system. Immunohistochemical analysis of the rat hemidiaphragm indicated that IgG anti-GM1 mAb binding overlapped with anti-Cav2.2 (α1B) antibody binding in the nerve terminal. In addition, IgG anti-GM1 mAb binding partially overlapped with anti-Cav2.1 (α1A) antibody binding. Thus, the results demonstrated that the IgG anti-GM1 mAb binds to calcium channels in the nerve terminals of NMJs. Therefore, the inhibitory effect of IgG anti-GM1 mAb on SMAPs may involve N-type and P/Q-type calcium channels in motor nerve terminals at the NMJ.
Collapse
Affiliation(s)
- Sayako Hotta
- Department of Medicinal Pharmacology, Showa Pharmaceutical University, Machida, Tokyo 194-8543, Japan
| | - Yoshihiko Nakatani
- Department of Medicinal Pharmacology, Showa Pharmaceutical University, Machida, Tokyo 194-8543, Japan
| | - Toshie Kambe
- Department of Pharmacology, Showa Pharmaceutical University, Machida, Tokyo 194-8543, Japan
| | - Kenji Abe
- Department of Pharmacology, School of Pharmaceutical Sciences, Ohu University, Koriyama, Fukushima 963-8611, Japan
| | - Yutaka Masuda
- Laboratory of Clinical Pharmacy, Showa Pharmaceutical University, Machida, Tokyo 194-8543, Japan
| | - Iku Utsumomiya
- Department of Developmental Education, Showa Pharmaceutical University, Machida, Tokyo 194-8543, Japan
| | - Kyoji Taguchi
- Department of Medicinal Pharmacology, Showa Pharmaceutical University, Machida, Tokyo 194-8543, Japan
| |
Collapse
|
43
|
Yue HY, Xu J. Cholesterol regulates multiple forms of vesicle endocytosis at a mammalian central synapse. J Neurochem 2015; 134:247-60. [PMID: 25893258 DOI: 10.1111/jnc.13129] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2014] [Revised: 03/01/2015] [Accepted: 03/31/2015] [Indexed: 01/10/2023]
Abstract
Endocytosis in synapses sustains neurotransmission by recycling vesicle membrane and maintaining the homeostasis of synaptic membrane. A role of membrane cholesterol in synaptic endocytosis remains controversial because of conflicting observations, technical limitations in previous studies, and potential interference from non-specific effects after cholesterol manipulation. Furthermore, it remains unclear whether cholesterol participates in distinct forms of endocytosis that function under different activity levels. In this study, applying the whole-cell membrane capacitance measurement to monitor endocytosis in real time at the rat calyx of Held terminals, we found that disrupting cholesterol with dialysis of cholesterol oxidase or methyl-β-cyclodextrin impaired three different forms of endocytosis, including slow endocytosis, rapid endocytosis, and endocytosis of the retrievable membrane that exists at the surface before stimulation. The effects were observed when disruption of cholesterol was mild enough not to change Ca(2+) channel current or vesicle exocytosis, indicative of stringent cholesterol requirement in synaptic endocytosis. Extracting cholesterol with high concentrations of methyl-β-cyclodextrin reduced exocytosis, mainly by decreasing the readily releasable pool and the vesicle replenishment after readily releasable pool depletion. Our study suggests that cholesterol is an important, universal regulator in multiple forms of vesicle endocytosis at mammalian central synapses.
Collapse
Affiliation(s)
- Hai-Yuan Yue
- Department of Neuroscience and Regenerative Medicine, Georgia Regents University, Augusta, Georgia, USA
| | - Jianhua Xu
- Department of Neuroscience and Regenerative Medicine, Georgia Regents University, Augusta, Georgia, USA.,Department of Neurology, Medical College of Georgia, Georgia Regents University, Augusta, Georgia, USA
| |
Collapse
|
44
|
Sosial E, Nussinovitch I. Multiple Ca2+ channel-dependent components in growth hormone secretion from rat anterior pituitary somatotrophs. J Neuroendocrinol 2015; 27:166-76. [PMID: 25442738 DOI: 10.1111/jne.12240] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/24/2014] [Revised: 11/25/2014] [Accepted: 11/25/2014] [Indexed: 12/22/2022]
Abstract
The involvement of L-type Ca(2+) channels in both 'basal' and 'stimulated' growth hormone (GH) secretion is well established; however, knowledge regarding the involvement of non-L-type Ca(2+) channels is lacking. We investigated whether non-L-type Ca(2+) channels regulate GH secretion from anterior pituitary (AP) cells. To this end, GH secretion was monitored from dissociated AP cells, which were incubated for 15 min with 2 mm K(+) ('basal' secretion) or 60 mm K(+) ('stimulated' secretion). The role of non-L-type Ca(2+) influx was investigated using specific channel blockers, including ω-agatoxin-IVA, ω-conotoxin GVIA or SNX-482, to block P/Q-, N- or R-type Ca(2+) channels, respectively. Our results demonstrate that P/Q-, N- and R-type Ca(2+) channels contributed 21.2 ± 1.9%, 20.2 ± 7.6% and 11.4 ± 1.8%, respectively, to 'basal' GH secretion and 18.3 ± 1.0%, 24.4 ± 5.4% and 14.2 ± 4.8%, respectively, to 'stimulated' GH secretion. After treatment with a 'cocktail' that comprised the previously described non-L-type blockers, non-L-type Ca(2+) channels contributed 50.9 ± 0.4% and 45.5 ± 2.0% to 'basal' and 'stimulated' GH secretion, respectively. Similarly, based on the effects of nifedipine (10 μM), L-type Ca(2+) channels contributed 34.2 ± 3.7% and 54.7 ± 4.1% to 'basal' and 'stimulated' GH secretion, respectively. Interestingly, the relative contributions of L-type/non-L-type Ca(2+) channels to 'stimulated' GH secretion were well correlated with the relative contributions of L-type/non-L-type Ca(2+) channels to voltage-gated Ca(2+) influx in AP cells. Finally, we demonstrated that compartmentalisation of Ca(2+) channels is important for GH secretion. Lipid raft disruption (methyl-β-cyclodextrin, 10 mm) abrogated the compartmentalisation of Ca(2+) channels and substantially reduced 'basal' and 'stimulated' GH secretion by 43.2 ± 3.4% and 58.4 ± 4.0%, respectively. In summary, we have demonstrated that multiple Ca(2+) channel-dependent pathways regulate GH secretion. The proper function of these pathways depends on their compartmentalisation within AP cell membranes.
Collapse
Affiliation(s)
- E Sosial
- Department of Medical Neurobiology, Institute for Medical Research-Israel-Canada, The Hebrew University Faculty of Medicine, Jerusalem, Israel
| | | |
Collapse
|
45
|
Abstract
The α2δ auxiliary subunits of voltage-gated Ca2+ channels (VGCCs) are important modulators of VGCC function. Gabapentin interacts with α2δ1 and α2δ2 subunits and is reported to reduce Ca2+ channel current amplitude (ICa). This study aimed to determine the effects of gabapentin on VGCCs in retinal ganglion cells (RGCs). Whole cell patch clamp was used to record ICa in isolated RGCs, and calcium imaging was used to measure Ca2+ transients from RGCs in situ. Immunohistochemistry was used to detect the presence of α2δ1-containing VGCCs in isolated RGCs in the absence and presence of gabapentin pretreatment. Acute administration of gabapentin reduced ICa and Ca2+ transients compared to control conditions. In isolated RGCs, pretreatment with gabapentin (4-18 h) reduced ICa, and cell surface α2δ1 staining was reduced compared to nonpretreated cells. Acute administration of gabapentin to isolated RGCs that had been pretreated further reduced ICa. These results show that gabapentin has both short-term and long-term mechanisms to reduce ICa in isolated RGCs. Some Ca2+ channel blockers have been shown to protect RGCs in retinal trauma suggesting that modulation of VGCCs by gabapentin may prevent the deleterious effects of elevated Ca2+ levels in RGCs in trauma and disease.
Collapse
|
46
|
Nieto-Rostro M, Sandhu G, Bauer CS, Jiruska P, Jefferys JGR, Dolphin AC. Altered expression of the voltage-gated calcium channel subunit α₂δ-1: a comparison between two experimental models of epilepsy and a sensory nerve ligation model of neuropathic pain. Neuroscience 2014; 283:124-37. [PMID: 24641886 PMCID: PMC4259901 DOI: 10.1016/j.neuroscience.2014.03.013] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2014] [Revised: 03/03/2014] [Accepted: 03/09/2014] [Indexed: 12/20/2022]
Abstract
The auxiliary α2δ-1 subunit of voltage-gated calcium channels is up-regulated in dorsal root ganglion neurons following peripheral somatosensory nerve damage, in several animal models of neuropathic pain. The α2δ-1 protein has a mainly presynaptic localization, where it is associated with the calcium channels involved in neurotransmitter release. Relevant to the present study, α2δ-1 has been shown to be the therapeutic target of the gabapentinoid drugs in their alleviation of neuropathic pain. These drugs are also used in the treatment of certain epilepsies. In this study we therefore examined whether the level or distribution of α2δ-1 was altered in the hippocampus following experimental induction of epileptic seizures in rats, using both the kainic acid model of human temporal lobe epilepsy, in which status epilepticus is induced, and the tetanus toxin model in which status epilepticus is not involved. The main finding of this study is that we did not identify somatic overexpression of α2δ-1 in hippocampal neurons in either of the epilepsy models, unlike the upregulation of α2δ-1 that occurs following peripheral nerve damage to both somatosensory and motor neurons. However, we did observe local reorganization of α2δ-1 immunostaining in the hippocampus only in the kainic acid model, where it was associated with areas of neuronal cell loss, as indicated by absence of NeuN immunostaining, dendritic loss, as identified by areas where microtubule-associated protein-2 immunostaining was missing, and reactive gliosis, determined by regions of strong OX42 staining.
Collapse
Affiliation(s)
- M Nieto-Rostro
- Department of Neuroscience, Physiology and Pharmacology, University College London, London WC1E 6BT, UK
| | - G Sandhu
- Department of Neuroscience, Physiology and Pharmacology, University College London, London WC1E 6BT, UK
| | - C S Bauer
- Department of Neuroscience, Physiology and Pharmacology, University College London, London WC1E 6BT, UK
| | - P Jiruska
- Neuronal Networks Group, School of Clinical and Experimental Medicine, University of Birmingham, Birmingham B15 2TT, UK
| | - J G R Jefferys
- Neuronal Networks Group, School of Clinical and Experimental Medicine, University of Birmingham, Birmingham B15 2TT, UK
| | - A C Dolphin
- Department of Neuroscience, Physiology and Pharmacology, University College London, London WC1E 6BT, UK.
| |
Collapse
|
47
|
Exogenous α-synuclein decreases raft partitioning of Cav2.2 channels inducing dopamine release. J Neurosci 2014; 34:10603-15. [PMID: 25100594 DOI: 10.1523/jneurosci.0608-14.2014] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
α-Synuclein is thought to regulate neurotransmitter release through multiple interactions with presynaptic proteins, cytoskeletal elements, ion channels, and synaptic vesicles membrane. α-Synuclein is abundant in the presynaptic compartment, and its release from neurons and glia has been described as responsible for spreading of α-synuclein-derived pathology. α-Synuclein-dependent dysregulation of neurotransmitter release might occur via its action on surface-exposed calcium channels. Here, we provide electrophysiological and biochemical evidence to show that α-synuclein, applied to rat neurons in culture or striatal slices, selectively activates Cav2.2 channels, and said activation correlates with increased neurotransmitter release. Furthermore, in vivo perfusion of α-synuclein into the striatum also leads to acute dopamine release. We further demonstrate that α-synuclein reduces the amount of plasma membrane cholesterol and alters the partitioning of Cav2.2 channels, which move from raft to cholesterol-poor areas of the plasma membrane. We provide evidence for a novel mechanism through which α-synuclein acts from the extracellular milieu to modulate neurotransmitter release and propose a unifying hypothesis for the mechanism of α-synuclein action on multiple targets: the reorganization of plasma membrane microdomains.
Collapse
|
48
|
In vivo single-molecule imaging identifies altered dynamics of calcium channels in dystrophin-mutant C. elegans. Nat Commun 2014; 5:4974. [PMID: 25232639 PMCID: PMC4199201 DOI: 10.1038/ncomms5974] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2014] [Accepted: 08/12/2014] [Indexed: 01/15/2023] Open
Abstract
Single-molecule (SM) fluorescence microscopy allows the imaging of biomolecules in cultured cells with a precision of a few nanometres but has yet to be implemented in living adult animals. Here we used split-GFP (green fluorescent protein) fusions and complementation-activated light microscopy (CALM) for subresolution imaging of individual membrane proteins in live Caenorhabditis elegans (C. elegans). In vivo tissue-specific SM tracking of transmembrane CD4 and voltage-dependent Ca2+ channels (VDCC) was achieved with a precision of 30 nm within neuromuscular synapses and at the surface of muscle cells in normal and dystrophin-mutant worms. Through diffusion analyses, we reveal that dystrophin is involved in modulating the confinement of VDCC within sarcolemmal membrane nanodomains in response to varying tonus of C. elegans body-wall muscles. CALM expands the applications of SM imaging techniques beyond the petri dish and opens the possibility to explore the molecular basis of homeostatic and pathological cellular processes with subresolution precision, directly in live animals. Single molecule fluorescence microscopy is a powerful technique to study protein dynamics in cells, but it has not been applied to adult animals. The authors use complementation-activated light microscopy in C. elegansto discover that dystrophin regulates the diffusion properties of voltage-dependent calcium ion channels at the surface of body-wall muscle cells.![]()
Collapse
|
49
|
Buys MJ, Alphonso C. Novel use of perineural pregabalin infusion for analgesia in a rat neuropathic pain model. Anesth Analg 2014; 119:481-488. [PMID: 24914629 DOI: 10.1213/ane.0000000000000291] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
BACKGROUND The anticonvulsant drugs pregabalin and gabapentin are often used systemically to treat some forms of chronic neuropathic pain. However, many patients report side effects serious enough to cause discontinuation of the drug. Here we present evidence that pregabalin may block neuropathic pain when applied to the site of nerve injury in a rat neuropathic pain model. METHODS Forty male Sprague Dawley rats were randomized into 4 groups: sciatic nerve crush injury with perineural pregabalin treatment (treatment), crush injury with perineural saline treatment (saline control), crush injury with subcutaneous pregabalin treatment (systemic drug control), and sham surgery (sham surgery control). Animals received either continuous infusions of 1% pregabalin for 7 days (treatment and systemic control) or saline (saline control) and were tested for pain behaviors using incapacitance meter, guarding scores, and radiant heat withdrawal latency (Hargreaves method). Nerves were studied using histology and immunohistochemistry for α(2)δ-1 receptors thought to mediate the central analgesic action of pregabalin. RESULTS Treatment rats had significantly better guarding scores than systemic drug controls or saline controls (P < 0.0001) and had significantly better incapacitance scores than systemic drug controls and saline controls (P ≤ 0.001). Hargreaves method data showed hypoalgesia in all injured animals with no difference among injured groups (P = 0.80). Qualitatively, immunohistochemistry likely showed equivalent expression of the α(2)δ-1 calcium channel at the injured nerve site in all nerve-injured animals. CONCLUSIONS Perineural pregabalin administration produced superior analgesia compared with that of systemic pregabalin in this neuropathic pain model. Perineural pregabalin treatment may provide a useful alternative to systemic pregabalin treatment for neuropathic pain.
Collapse
Affiliation(s)
- Michael J Buys
- From the Department of Anesthesiology, San Antonio Military Medical Centers, Lackland AFB, USAF
| | | |
Collapse
|
50
|
Meriney SD, Umbach JA, Gundersen CB. Fast, Ca2+-dependent exocytosis at nerve terminals: shortcomings of SNARE-based models. Prog Neurobiol 2014; 121:55-90. [PMID: 25042638 DOI: 10.1016/j.pneurobio.2014.07.001] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2013] [Revised: 04/14/2014] [Accepted: 07/03/2014] [Indexed: 11/30/2022]
Abstract
Investigations over the last two decades have made major inroads in clarifying the cellular and molecular events that underlie the fast, synchronous release of neurotransmitter at nerve endings. Thus, appreciable progress has been made in establishing the structural features and biophysical properties of the calcium (Ca2+) channels that mediate the entry into nerve endings of the Ca2+ ions that trigger neurotransmitter release. It is now clear that presynaptic Ca2+ channels are regulated at many levels and the interplay of these regulatory mechanisms is just beginning to be understood. At the same time, many lines of research have converged on the conclusion that members of the synaptotagmin family serve as the primary Ca2+ sensors for the action potential-dependent release of neurotransmitter. This identification of synaptotagmins as the proteins which bind Ca2+ and initiate the exocytotic fusion of synaptic vesicles with the plasma membrane has spurred widespread efforts to reveal molecular details of synaptotagmin's action. Currently, most models propose that synaptotagmin interfaces directly or indirectly with SNARE (soluble, N-ethylmaleimide sensitive factor attachment receptors) proteins to trigger membrane fusion. However, in spite of intensive efforts, the field has not achieved consensus on the mechanism by which synaptotagmins act. Concurrently, the precise sequence of steps underlying SNARE-dependent membrane fusion remains controversial. This review considers the pros and cons of the different models of SNARE-mediated membrane fusion and concludes by discussing a novel proposal in which synaptotagmins might directly elicit membrane fusion without the intervention of SNARE proteins in this final fusion step.
Collapse
Affiliation(s)
- Stephen D Meriney
- Department of Neuroscience, Center for Neuroscience, University of Pittsburgh, Pittsburgh, PA 15260, USA
| | - Joy A Umbach
- Department of Molecular and Medical Pharmacology, UCLA David Geffen School of Medicine, Los Angeles, CA 90095, USA
| | - Cameron B Gundersen
- Department of Molecular and Medical Pharmacology, UCLA David Geffen School of Medicine, Los Angeles, CA 90095, USA.
| |
Collapse
|