1
|
Yao X, Gao S, Yan N. Structural biology of voltage-gated calcium channels. Channels (Austin) 2024; 18:2290807. [PMID: 38062897 PMCID: PMC10761187 DOI: 10.1080/19336950.2023.2290807] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2023] [Accepted: 11/27/2023] [Indexed: 12/18/2023] Open
Abstract
Voltage-gated calcium (Cav) channels mediate Ca2+ influx in response to membrane depolarization, playing critical roles in diverse physiological processes. Dysfunction or aberrant regulation of Cav channels can lead to life-threatening consequences. Cav-targeting drugs have been clinically used to treat cardiovascular and neuronal disorders for several decades. This review aims to provide an account of recent developments in the structural dissection of Cav channels. High-resolution structures have significantly advanced our understanding of the working and disease mechanisms of Cav channels, shed light on the molecular basis for their modulation, and elucidated the modes of actions (MOAs) of representative drugs and toxins. The progress in structural studies of Cav channels lays the foundation for future drug discovery efforts targeting Cav channelopathies.
Collapse
Affiliation(s)
- Xia Yao
- TaiKang Center for Life and Medical Sciences, School of Pharmaceutical Sciences, Wuhan University, Wuhan, China
| | - Shuai Gao
- TaiKang Center for Life and Medical Sciences, School of Pharmaceutical Sciences, Wuhan University, Wuhan, China
| | - Nieng Yan
- Beijing Frontier Research Center for Biological Structures, State Key Laboratory of Membrane Biology, Tsinghua-Peking Joint Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing, China
- Shenzhen Medical Academy of Research and Translation, Shenzhen, China
| |
Collapse
|
2
|
Bajaj S, Han J. Venom-Derived Peptide Modulators of Cation-Selective Channels: Friend, Foe or Frenemy. Front Pharmacol 2019; 10:58. [PMID: 30863305 PMCID: PMC6399158 DOI: 10.3389/fphar.2019.00058] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2018] [Accepted: 01/18/2019] [Indexed: 01/31/2023] Open
Abstract
Ion channels play a key role in our body to regulate homeostasis and conduct electrical signals. With the help of advances in structural biology, as well as the discovery of numerous channel modulators derived from animal toxins, we are moving toward a better understanding of the function and mode of action of ion channels. Their ubiquitous tissue distribution and the physiological relevancies of their opening and closing suggest that cation channels are particularly attractive drug targets, and years of research has revealed a variety of natural toxins that bind to these channels and alter their function. In this review, we provide an introductory overview of the major cation ion channels: potassium channels, sodium channels and calcium channels, describe their venom-derived peptide modulators, and how these peptides provide great research and therapeutic value to both basic and translational medical research.
Collapse
Affiliation(s)
- Saumya Bajaj
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore
| | - Jingyao Han
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore
| |
Collapse
|
3
|
Abstract
A central theme in the quest to unravel the genetic basis of epilepsy has been the effort to elucidate the roles played by inherited defects in ion channels. The ubiquitous expression of voltage-gated calcium channels (VGCCs) throughout the central nervous system (CNS), along with their involvement in fundamental processes, such as neuronal excitability and synaptic transmission, has made them attractive candidates. Recent insights provided by the identification of mutations in the P/Q-type calcium channel in humans and rodents with epilepsy and the finding of thalamic T-type calcium channel dysfunction in the absence of seizures have raised expectations of a causal role of calcium channels in the polygenic inheritance of idiopathic epilepsy. In this review, we consider how genetic variation in neuronal VGCCs may influence the development of epilepsy.
Collapse
Affiliation(s)
- Sanjeev Rajakulendran
- UCL-Institute of Neurology, MRC Centre for Neuromuscular Diseases, Queen Square, London WC1N 3BG, United Kingdom
| | - Michael G Hanna
- UCL-Institute of Neurology, MRC Centre for Neuromuscular Diseases, Queen Square, London WC1N 3BG, United Kingdom
| |
Collapse
|
4
|
Kalia J, Milescu M, Salvatierra J, Wagner J, Klint JK, King GF, Olivera BM, Bosmans F. From foe to friend: using animal toxins to investigate ion channel function. J Mol Biol 2014; 427:158-175. [PMID: 25088688 DOI: 10.1016/j.jmb.2014.07.027] [Citation(s) in RCA: 125] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2014] [Revised: 07/18/2014] [Accepted: 07/18/2014] [Indexed: 12/19/2022]
Abstract
Ion channels are vital contributors to cellular communication in a wide range of organisms, a distinct feature that renders this ubiquitous family of membrane-spanning proteins a prime target for toxins found in animal venom. For many years, the unique properties of these naturally occurring molecules have enabled researchers to probe the structural and functional features of ion channels and to define their physiological roles in normal and diseased tissues. To illustrate their considerable impact on the ion channel field, this review will highlight fundamental insights into toxin-channel interactions and recently developed toxin screening methods and practical applications of engineered toxins.
Collapse
Affiliation(s)
- Jeet Kalia
- Indian Institute of Science Education and Research Pune; Pune, Maharashtra 411 008 India
| | - Mirela Milescu
- Division of Biological Sciences; University of Missouri, Columbia, MO 65211 USA
| | - Juan Salvatierra
- Department of Physiology; Johns Hopkins University, School of Medicine, Baltimore, MD 21205 USA
| | - Jordan Wagner
- Department of Physiology; Johns Hopkins University, School of Medicine, Baltimore, MD 21205 USA
| | - Julie K Klint
- Institute for Molecular Bioscience; The University of Queensland, St. Lucia, QLD 4072 Australia
| | - Glenn F King
- Institute for Molecular Bioscience; The University of Queensland, St. Lucia, QLD 4072 Australia
| | | | - Frank Bosmans
- Department of Physiology; Johns Hopkins University, School of Medicine, Baltimore, MD 21205 USA.,Solomon H. Snyder Department of Neuroscience; Johns Hopkins University, School of Medicine, Baltimore, MD 21205 USA
| |
Collapse
|
5
|
Hofmann F, Flockerzi V, Kahl S, Wegener JW. L-type CaV1.2 calcium channels: from in vitro findings to in vivo function. Physiol Rev 2014; 94:303-26. [PMID: 24382889 DOI: 10.1152/physrev.00016.2013] [Citation(s) in RCA: 239] [Impact Index Per Article: 23.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023] Open
Abstract
The L-type Cav1.2 calcium channel is present throughout the animal kingdom and is essential for some aspects of CNS function, cardiac and smooth muscle contractility, neuroendocrine regulation, and multiple other processes. The L-type CaV1.2 channel is built by up to four subunits; all subunits exist in various splice variants that potentially affect the biophysical and biological functions of the channel. Many of the CaV1.2 channel properties have been analyzed in heterologous expression systems including regulation of the L-type CaV1.2 channel by Ca(2+) itself and protein kinases. However, targeted mutations of the calcium channel genes confirmed only some of these in vitro findings. Substitution of the respective serines by alanine showed that β-adrenergic upregulation of the cardiac CaV1.2 channel did not depend on the phosphorylation of the in vitro specified amino acids. Moreover, well-established in vitro phosphorylation sites of the CaVβ2 subunit of the cardiac L-type CaV1.2 channel were found to be irrelevant for the in vivo regulation of the channel. However, the molecular basis of some kinetic properties, such as Ca(2+)-dependent inactivation and facilitation, has been approved by in vivo mutagenesis of the CaV1.2α1 gene. This article summarizes recent findings on the in vivo relevance of well-established in vitro results.
Collapse
|
6
|
Vink S, Alewood PF. Targeting voltage-gated calcium channels: developments in peptide and small-molecule inhibitors for the treatment of neuropathic pain. Br J Pharmacol 2013; 167:970-89. [PMID: 22725651 DOI: 10.1111/j.1476-5381.2012.02082.x] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Chronic pain affects approximately 20% of people worldwide and places a large economic and social burden on society. Despite the availability of a range of analgesics, this condition is inadequately treated, with complete alleviation of symptoms rarely occurring. In the past 30 years, the voltage-gated calcium channels (VGCCs) have been recognized as potential targets for analgesic development. Although the majority of the research has been focused on Ca(v) 2.2 in particular, other VGCC subtypes such as Ca(v) 3.2 have recently come to the forefront of analgesic research. Venom peptides from marine cone snails have been proven to be a valuable tool in neuroscience, playing a major role in the identification and characterization of VGCC subtypes and producing the first conotoxin-based drug on the market, the ω-conotoxin, ziconotide. This peptide potently and selectively inhibits Ca(v) 2.2, resulting in analgesia in chronic pain states. However, this drug is only available via intrathecal administration, and adverse effects and a narrow therapeutic window have limited its use in the clinic. Other Ca(v) 2.2 inhibitors are currently in development and offer the promise of an improved route of administration and safety profile. This review assesses the potential of targeting VGCCs for analgesic development, with a main focus on conotoxins that block Ca(v) 2.2 and the developments made to transform them into therapeutics.
Collapse
Affiliation(s)
- S Vink
- Division of Chemistry and Structural Biology, Institute for Molecular Bioscience, The University of Queensland, Brisbane, Australia
| | | |
Collapse
|
7
|
Abstract
Voltage-gated calcium (Ca(2+)) channels are key transducers of membrane potential changes into intracellular Ca(2+) transients that initiate many physiological events. There are ten members of the voltage-gated Ca(2+) channel family in mammals, and they serve distinct roles in cellular signal transduction. The Ca(V)1 subfamily initiates contraction, secretion, regulation of gene expression, integration of synaptic input in neurons, and synaptic transmission at ribbon synapses in specialized sensory cells. The Ca(V)2 subfamily is primarily responsible for initiation of synaptic transmission at fast synapses. The Ca(V)3 subfamily is important for repetitive firing of action potentials in rhythmically firing cells such as cardiac myocytes and thalamic neurons. This article presents the molecular relationships and physiological functions of these Ca(2+) channel proteins and provides information on their molecular, genetic, physiological, and pharmacological properties.
Collapse
Affiliation(s)
- William A Catterall
- Department of Pharmacology, University of Washington, Seattle, Washington 98195-7280, USA.
| |
Collapse
|
8
|
Whorlow SL, Angus JA, Wright CE. SELECTIVITY OF ω-CONOTOXIN GVIA FOR N-TYPE CALCIUM CHANNELS IN RAT ISOLATED SMALL MESENTERIC ARTERIES. Clin Exp Pharmacol Physiol 2010. [DOI: 10.1111/j.1440-1681.1996.tb03056.x] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
9
|
Fox AP, Cahill AL, Currie KPM, Grabner C, Harkins AB, Herring B, Hurley JH, Xie Z. N- and P/Q-type Ca2+ channels in adrenal chromaffin cells. Acta Physiol (Oxf) 2008; 192:247-61. [PMID: 18021320 DOI: 10.1111/j.1748-1716.2007.01817.x] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
Abstract
Ca2+ is the most ubiquitous second messenger found in all cells. Alterations in [Ca2+]i contribute to a wide variety of cellular responses including neurotransmitter release, muscle contraction, synaptogenesis and gene expression. Voltage-dependent Ca2+ channels, found in all excitable cells (Hille 1992), mediate the entry of Ca2+ into cells following depolarization. Ca2+ channels are composed of a large pore-forming subunit, called the alpha1 subunit, and several accessory subunits. Ten different alpha1 subunit genes have been identified and classified into three families, Ca(v1-3) (Dunlap et al. 1995, Catterall 2000). Each alpha1 gene produces a unique Ca2+ channel. Although chromaffin cells express several different types of Ca2+ channels, this review will focus on the Cav(2.1) and Cav(2.2) channels, also known as P/Q- and N-type respectively (Nowycky et al. 1985, Llinas et al. 1989b, Wheeler et al. 1994). These channels exhibit physiological and pharmacological properties similar to their neuronal counterparts. N-, P/Q and to a lesser extent R-type Ca2+ channels are known to regulate neurotransmitter release (Hirning et al. 1988, Horne & Kemp 1991, Uchitel et al. 1992, Luebke et al. 1993, Takahashi & Momiyama 1993, Turner et al. 1993, Regehr & Mintz 1994, Wheeler et al. 1994, Wu & Saggau 1994, Waterman 1996, Wright & Angus 1996, Reid et al. 1997). N- and P/Q-type Ca2+ channels are abundant in nerve terminals where they colocalize with synaptic vesicles. Similarly, these channels play a role in neurotransmitter release in chromaffin cells (Garcia et al. 2006). N- and P/Q-type channels are subject to many forms of regulation (Ikeda & Dunlap 1999). This review pays particular attention to the regulation of N- and P/Q-type channels by heterotrimeric G-proteins, interaction with SNARE proteins, and channel inactivation in the context of stimulus-secretion coupling in adrenal chromaffin cells.
Collapse
Affiliation(s)
- A P Fox
- Department of Neurobiology, Pharmacology and Physiology, University of Chicago, Chicago, IL 60637, USA.
| | | | | | | | | | | | | | | |
Collapse
|
10
|
Chen Y, Sharp AH, Hata K, Yunker AMR, Polo-Parada L, Landmesser LT, McEnery MW. Site-directed antibodies to low-voltage-activated calcium channel CaV3.3 (alpha1I) subunit also target neural cell adhesion molecule-180. Neuroscience 2007; 145:981-96. [PMID: 17317015 DOI: 10.1016/j.neuroscience.2006.12.060] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2006] [Revised: 12/18/2006] [Accepted: 12/19/2006] [Indexed: 11/19/2022]
Abstract
Synthetic peptides of defined amino acid sequence are commonly used as unique antigens for production of antibodies to more complex target proteins. We previously showed that an affinity-purified, site-directed polyclonal antibody (CW90) raised against a peptide antigen (CNGRMPNIAKDVFTKM) anticipated to be specific to a T-type voltage-dependent Ca(2+) channel subunit identified recombinant rat alpha1I/Ca(V)3.3 and two endogenous mouse proteins distinct in their developmental expression and apparent molecular mass (neonatal form 260 kDa, mature form 190 kDa) [Yunker AM, Sharp AH, Sundarraj S, Ranganathan V, Copeland TD, McEnery MW (2003) Immunological characterization of T-type voltage-dependent calcium channel Ca(V)3.1 (alpha 1G) and Ca(V)3.3 (alpha 1I) isoforms reveal differences in their localization, expression, and neural development. Neuroscience 117:321-335]. In the present study, we further characterize the biochemical properties of the CW90 antigens. We show for the first time that recombinant alpha1I/Ca(V)3.3 is modified by N-glycosylation. Using peptide:N-glycosidase F (PNGase F), an enzyme that removes polysaccharides attached at Asn residues, and endoneuraminidase-N (Endo-N), which specifically removes polysialic acid modifications, we reveal that differential glycosylation fully accounts for the large difference in apparent molecular mass between neonatal and adult CW90 antigens and that the neonatal form is polysialylated. As very few proteins are substrates for Endo-N, we carried out extensive analyses and herein present evidence that CW90 reacts with recombinant alpha1I/Ca(V)3.3 as well as endogenous neural cell adhesion molecule-180 (NCAM-180). We demonstrate the basis for CW90 cross-reactivity is a five amino acid epitope (AKDVF) present in both alpha1I/Ca(V)3.3 and NCAM-180. To extend these findings, we introduce a novel polyclonal anti-peptide antibody (CW678) that uniquely recognizes NCAM-180 and a new antibody (CW109) against alpha1I/Ca(V)3.3. Western blot analyses obtained with CW678, CW109 and CW90 on a variety of samples confirm that the endogenous CW90 signals are fully attributed to the two developmental forms of NCAM-180. Using CW678, we present novel data on differentiation-dependent NCAM-180 expression in human neuroblastoma IMR32 cells. These results strongly suggest the need for careful analyses to validate anti-peptide antibodies when targeting membrane proteins of low abundance.
Collapse
Affiliation(s)
- Y Chen
- Department of General Medical Sciences, Case Western Reserve University, School of Medicine, 10900 Euclid Avenue, Cleveland, OH 44106, USA
| | | | | | | | | | | | | |
Collapse
|
11
|
HELL JOHANNESW, WESTENBROEK RUTHE, ELLIOTT ELICIAM, CATTERALL WILLIAMA. Differential Phosphorylation, Localization, and Function of Distinct α1 Subunits of Neuronal Calcium Channels. Ann N Y Acad Sci 2006. [DOI: 10.1111/j.1749-6632.1994.tb44416.x] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
12
|
Benjamin ER, Pruthi F, Olanrewaju S, Shan S, Hanway D, Liu X, Cerne R, Lavery D, Valenzano KJ, Woodward RM, Ilyin VI. Pharmacological characterization of recombinant N-type calcium channel (Cav2.2) mediated calcium mobilization using FLIPR. Biochem Pharmacol 2006; 72:770-82. [PMID: 16844100 DOI: 10.1016/j.bcp.2006.06.003] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2006] [Revised: 06/04/2006] [Accepted: 06/06/2006] [Indexed: 11/24/2022]
Abstract
The N-type voltage-gated calcium channel (Ca(v)2.2) functions in neurons to regulate neurotransmitter release. It comprises a clinically relevant target for chronic pain. We have validated a calcium mobilization approach to assessing Ca(v)2.2 pharmacology in two stable Ca(v)2.2 cell lines: alpha1(B), alpha2delta, beta(3)-HEK-293 and alpha1(B), beta(3)-HEK-293. Ca(v)2.2 channels were opened by addition of KCl and Ca(2+) mobilization was measured by Fluo-4 fluorescence on a fluorescence imaging plate reader (FLIPR(96)). Ca(v)2.2 expression and biophysics were confirmed by patch-clamp electrophysiology (EP). Both cell lines responded to KCl with adequate signal-to-background. Signals from both cell lines were inhibited by omega-conotoxin (ctx)-MVIIa and omega-conotoxin (ctx)-GVIa with IC(50) values of 1.8 and 1nM, respectively, for the three-subunit stable, and 0.9 and 0.6nM, respectively, for the two-subunit stable. Other known Ca(v)2.2 blockers were characterized including cadmium, flunarizine, fluspirilene, and mibefradil. IC(50) values correlated with literature EP-derived values. Novel Ca(v)2.2 pharmacology was identified in classes of compounds with other primary pharmacological activities, including Na(+) channel inhibitors and antidepressants. Novel Na(+) channel compounds with high potency at Ca(v)2.2 were identified in the phenoxyphenyl pyridine, phenoxyphenyl pyrazole, and other classes. The highest potency at Ca(v)2.2 tricyclic antidepressant identified was desipramine.
Collapse
Affiliation(s)
- Elfrida R Benjamin
- Purdue Pharma Discovery Research, 6 Cedarbrook Drive, Cranbury, NJ 08512, USA.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
13
|
Ichida S, Abe J, Komoike K, Imanishi T, Wada T, Masuko T, Minami T. Characteristics of Omega-conotoxin GVI A and MVIIC Binding to Cav 2.1 and Cav 2.2 Channels Captured by Anti-Ca2+ Channel Peptide Antibodies. Neurochem Res 2005; 30:457-66. [PMID: 16076016 DOI: 10.1007/s11064-005-2681-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
A New Binding Method (NBM) was used to investigate the characteristics of the specific binding of 125I-omega-conotoxin (omega-CTX) GVIA and 125I-omega-CTX MVIIC to Cav2.1 and Cav2.2 channels captured from chick brain membranes by antibodies against B1Nt (a peptide sequence in Car2.1 and Cav2.2 channels). The results for the NBM were as follows. (1) The ED50 values for specific binding of 125I-omega-CTX GVIA and 125I-omega-CTX MVIIC to Cav2.1 and Cav2.2 channels were about 68 and 60 pM, respectively, and very similar to those (87 and 35 pM, respectively) to crude membranes from chick brain. (2) The specific 125I-omega-CTX GVIA (100 pM) binding was inhibited by omega-CTX GVIA (0.5 nM), dynorphine A (Dyn), gentamicin (Gen), neomycin (Neo) and tobramicin (Tob) (100 microM each), but not by omega-agaconotoxin (Aga) IVA, calciseptine, omega-CTX SVIB, omega-CTX MVIIC (0.5 nM each), PN200-110 (PN), diltiazem (Dil) or verapamil (Ver) (100 microM each). Calmodulin (CaM) inhibited the specific binding in a dose-dependent manner (IC50 value of about 100 microg protein/ml). (3) The specific 125I-omega-CTX MVIIC (60 pM) binding was inhibited by omega-CTX MVIIC, omega-CTX GVIA, omega-CTX SVIB (0.5 nM each), Dyn, Neo and Tob (100 microM, each), but not by omega-Aga IVA, calciseptine (0.5 nM each), PN, Dil, Ver (100 microM each) or 100 microg protein/ml CaM. These results suggested that the characteristics of the specific binding of 125I-omega-CTX GVIA and 125I-omega-CTX MVIIC to Cav2.1 and Cav2.2 channels in the NBM were very similar to those to crude membranes from chick brain, although the IC50 values for CaM and free Ca2+ of CaM were about 33- and 5000-fold higher, respectively, than those for the specific binding of 125I-omega-CTX GVIA and 125I-omega-CTX MVIIC to crude membranes.
Collapse
Affiliation(s)
- Seiji Ichida
- Department of Biological Chemistry, School of Pharmaceutical Sciences, Kinki University, Kowakae 3-4-1, 577-8502, Higashio-saka, Japan.
| | | | | | | | | | | | | |
Collapse
|
14
|
Herlitze S, Xie M, Han J, Hümmer A, Melnik-Martinez KV, Moreno RL, Mark MD. Targeting mechanisms of high voltage-activated Ca2+ channels. J Bioenerg Biomembr 2004; 35:621-37. [PMID: 15000523 DOI: 10.1023/b:jobb.0000008027.19384.c0] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
Functional voltage-dependent Ca2+ channel complexes are assembled by three to four subunits: alpha1, beta, alpha2delta subunits (C. Leveque et al., 1994, J. Biol Chem. 269, 6306-6312; M. W. McEnery et al., 1991, Proc. Natl. Acad. Sci. U.S.A. 88, 11095-11099) and at least in muscle cells also y subunits (B. M. Curtis and W. A. Catterall, 1984, Biochemistry 23, 2113-2118). Ca2+ channels mediate the voltage-dependent Ca2+ influx in subcellular compartments, triggering such diverse processes as neurotransmitter release, dendritic action potentials, excitation-contraction, and excitation-transcription coupling. The targeting of biophysically defined Ca2+ channel complexes to the correct subcellular structures is, thus, critical to proper cell and physiological functioning. Despite their importance, surprisingly little is known about the targeting mechanisms by which Ca2+ channel complexes are transported to their site of function. Here we summarize what we know about the targeting of Ca2+ channel complexes through the cell to the plasma membrane and subcellular structures.
Collapse
Affiliation(s)
- Stefan Herlitze
- Department of Neurosciences, Case Western Reserve University, School of Medicine, Room E604, 10900 Euclid Avenue, Cleveland, Ohio 44106-4975, USA.
| | | | | | | | | | | | | |
Collapse
|
15
|
Mould J, Yasuda T, Schroeder CI, Beedle AM, Doering CJ, Zamponi GW, Adams DJ, Lewis RJ. The α2δ Auxiliary Subunit Reduces Affinity of ω-Conotoxins for Recombinant N-type (Cav2.2) Calcium Channels. J Biol Chem 2004; 279:34705-14. [PMID: 15166237 DOI: 10.1074/jbc.m310848200] [Citation(s) in RCA: 64] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The omega-conotoxins from fish-hunting cone snails are potent inhibitors of voltage-gated calcium channels. The omega-conotoxins MVIIA and CVID are selective N-type calcium channel inhibitors with potential in the treatment of chronic pain. The beta and alpha(2)delta-1 auxiliary subunits influence the expression and characteristics of the alpha(1B) subunit of N-type channels and are differentially regulated in disease states, including pain. In this study, we examined the influence of these auxiliary subunits on the ability of the omega-conotoxins GVIA, MVIIA, CVID and analogues to inhibit peripheral and central forms of the rat N-type channels. Although the beta3 subunit had little influence on the on- and off-rates of omega-conotoxins, coexpression of alpha(2)delta with alpha(1B) significantly reduced on-rates and equilibrium inhibition at both the central and peripheral isoforms of the N-type channels. The alpha(2)delta also enhanced the selectivity of MVIIA, but not CVID, for the central isoform. Similar but less pronounced trends were also observed for N-type channels expressed in human embryonic kidney cells. The influence of alpha(2)delta was not affected by oocyte deglycosylation. The extent of recovery from the omega-conotoxin block was least for GVIA, intermediate for MVIIA, and almost complete for CVID. Application of a hyperpolarizing holding potential (-120 mV) did not significantly enhance the extent of CVID recovery. Interestingly, [R10K]MVIIA and [O10K]GVIA had greater recovery from the block, whereas [K10R]CVID had reduced recovery from the block, indicating that position 10 had an important influence on the extent of omega-conotoxin reversibility. Recovery from CVID block was reduced in the presence of alpha(2)delta in human embryonic kidney cells and in oocytes expressing alpha(1B-b). These results may have implications for the antinociceptive properties of omega-conotoxins, given that the alpha(2)delta subunit is up-regulated in certain pain states.
Collapse
Affiliation(s)
- Jorgen Mould
- Institute for Molecular Bioscience, University of Queensland, Queensland 4072, Australia
| | | | | | | | | | | | | | | |
Collapse
|
16
|
Moss FJ, Dolphin AC, Clare JJ. Human neuronal stargazin-like proteins, gamma2, gamma3 and gamma4; an investigation of their specific localization in human brain and their influence on CaV2.1 voltage-dependent calcium channels expressed in Xenopus oocytes. BMC Neurosci 2003; 4:23. [PMID: 14505496 PMCID: PMC270087 DOI: 10.1186/1471-2202-4-23] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2003] [Accepted: 09/23/2003] [Indexed: 11/15/2022] Open
Abstract
BACKGROUND Stargazin (gamma2) and the closely related gamma3, and gamma4 transmembrane proteins are part of a family of proteins that may act as both neuronal voltage-dependent calcium channel (VDCC) gamma subunits and transmembrane alpha-amino-3-hydroxy-5-methyl-4-isoxazoleproponinc (AMPA) receptor regulatory proteins (TARPs). In this investigation, we examined the distribution patterns of the stargazin-like proteins gamma2, gamma3, and gamma4 in the human central nervous system (CNS). In addition, we investigated whether human gamma2 or gamma4 could modulate the electrophysiological properties of a neuronal VDCC complex transiently expressed in Xenopus oocytes. RESULTS The mRNA encoding human gamma2 is highly expressed in cerebellum, cerebral cortex, hippocampus and thalamus, whereas gamma3 is abundant in cerebral cortex and amygdala and gamma4 in the basal ganglia. Immunohistochemical analysis of the cerebellum determined that both gamma2 and gamma4 are present in the molecular layer, particularly in Purkinje cell bodies and dendrites, but have an inverse expression pattern to one another in the dentate cerebellar nucleus. They are also detected in the interneurons of the granule cell layer though only gamma2 is clearly detected in granule cells. The hippocampus stains for gamma2 and gamma4 throughout the layers of the every CA region and the dentate gyrus, whilst gamma3 appears to be localized particularly to the pyramidal and granule cell bodies. When co-expressed in Xenopus oocytes with a CaV2.1/beta4 VDCC complex, either in the absence or presence of an alpha2delta2 subunit, neither gamma2 nor gamma4 significantly modulated the VDCC peak current amplitude, voltage-dependence of activation or voltage-dependence of steady-state inactivation. CONCLUSION The human gamma2, gamma3 and gamma4 stargazin-like proteins are detected only in the CNS and display differential distributions among brain regions and several cell types in found in the cerebellum and hippocampus. These distribution patterns closely resemble those reported by other laboratories for the rodent orthologues of each protein. Whilst the fact that neither gamma2 nor gamma4 modulated the properties of a VDCC complex with which they could associate in vivo in Purkinje cells adds weight to the hypothesis that the principal role of these proteins is not as auxiliary subunits of VDCCs, it does not exclude the possibility that they play another role in VDCC function.
Collapse
Affiliation(s)
- Fraser J Moss
- Department of Pharmacology, University College London, Gower Street, London, WC1E 6BT, UK
- Current address: Division of Biology, M/C 156-29, California Institute of Technology, 1200 E. California Blvd., Pasadena, CA 91125, USA
| | - Annette C Dolphin
- Department of Pharmacology, University College London, Gower Street, London, WC1E 6BT, UK
| | - Jeffrey J Clare
- Gene Expression and Protein Biochemistry, GlaxoSmithKline, Medicines Research Center, Gunnels Wood Road, Stevenage, Herts, SG1 2NY, UK
| |
Collapse
|
17
|
Anantharam A, Diversé-Pierluissi MA. Biochemical approaches to study interaction of calcium channels with RGS12 in primary neuronal cultures. Methods Enzymol 2002; 345:60-70. [PMID: 11665642 DOI: 10.1016/s0076-6879(02)45007-0] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/22/2023]
Affiliation(s)
- Arun Anantharam
- Department of Pharmacology, Mount Sinai School of Medicine, New York, New York 10029, USA
| | | |
Collapse
|
18
|
Sharp AH, Black JL, Dubel SJ, Sundarraj S, Shen JP, Yunker AM, Copeland TD, McEnery MW. Biochemical and anatomical evidence for specialized voltage-dependent calcium channel gamma isoform expression in the epileptic and ataxic mouse, stargazer. Neuroscience 2001; 105:599-617. [PMID: 11516827 DOI: 10.1016/s0306-4522(01)00220-2] [Citation(s) in RCA: 59] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Inherited forms of ataxia and absence seizures in mice have been linked to defects in voltage-dependent calcium channel subunits. However, a correlation between the sites of neuronal dysfunction and the impact of the primary lesion upon calcium channel subunit expression or function has not been clearly established. For example, the mutation in stargazer mice has pleiotropic consequences including synaptic alterations in cerebellar granule cells, hippocampal CA3/mossy fibers, and cortical neurons in layer V that, presumably, lead to ataxia and seizures. Genetic analysis of stargazer mice determined that the defective gene encodes a protein expressed in brain (gamma2) with limited homology to the skeletal muscle L-type calcium channel gamma1 subunit. Although additional gamma isoforms have been subsequently identified primarily in neural tissue, little was known about the proteins they encode. Therefore, this study explored the distribution and biochemical properties of gamma2 and other gamma isoforms in wild-type and stargazer brain. We cloned human gamma2, gamma3, and gamma4 isoforms, produced specific anti-peptide antibodies to gamma isoforms and characterized both heterologously expressed and endogenous gamma. We identified regional specificity in the expression of gamma isoforms by western analysis and immunohistochemistry. We report for the first time that the mutation in the stargazer gene resulted in the loss of gamma2 protein. Furthermore, no compensatory changes in the expression of gamma3 or gamma4 protein were evident in stargazer brain. In contrast to other voltage-dependent calcium channel subunits, gamma immunostaining was striking in that it was primarily detected in regions highly enriched in excitatory glutamatergic synapses and faintly detected in cell bodies, suggesting a role for gamma in synaptic functions. Sites of known synaptic dysfunction in stargazer (the hippocampal CA3 region, dentate gyrus, and cerebellar molecular layer) were revealed as relying primarily upon gamma2, as total gamma isoform expression was dramatically decreased in these regions. Electron microscopy localized anti-gamma antibody immunostaining to dendritic structures of hippocampal mossy fiber synapses, with enrichment at postsynaptic densities. To assess the association of native gamma with voltage-dependent calcium channel or alpha-amino-3-hydroxy-5-methyl-4-isoxazole propionic acid (AMPA) receptor subunits, gamma isoforms (gamma2, gamma3 and gamma4) were detergent solubilized from mouse forebrain. Antibodies against a highly conserved C-terminal epitope present in gamma2, gamma3 and gamma4 immunoprecipitated voltage-dependent calcium channel subunits (alpha1B), providing the first in vivo evidence that gamma and voltage-dependent calcium channels form stable complexes. Furthermore, both anti-gamma2 antibodies and anti-alpha1B antibodies independently immunoprecipitated the AMPA receptor subunit, GluR1, from mouse forebrain homogenates. In summary, loss of gamma2 immunoreactivity in stargazer is precisely localized so as to contribute to previously characterized synaptic defects. The data in this paper provide compelling evidence that gamma isoforms form complexes in vivo with voltage-dependent calcium channels as well as AMPA receptors, are selectively and differentially expressed in neuronal processes, and localize primarily to dendritic structures in the hippocampal mossy fiber region.
Collapse
MESH Headings
- Animals
- Antibody Specificity
- Ataxia/genetics
- Ataxia/metabolism
- Ataxia/physiopathology
- Brain/metabolism
- Brain/physiopathology
- Brain/ultrastructure
- Calcium Channels, L-Type/genetics
- Calcium Channels, L-Type/metabolism
- Calcium Channels, N-Type/genetics
- Calcium Channels, N-Type/metabolism
- Calcium Signaling/genetics
- Dendrites/metabolism
- Dendrites/ultrastructure
- Epilepsy/genetics
- Epilepsy/metabolism
- Epilepsy/physiopathology
- Gene Expression/physiology
- Hippocampus/metabolism
- Hippocampus/ultrastructure
- Immunohistochemistry/methods
- Mice
- Mice, Neurologic Mutants/abnormalities
- Mice, Neurologic Mutants/metabolism
- Microscopy, Electron
- Molecular Sequence Data
- Protein Isoforms/genetics
- Protein Isoforms/metabolism
- Receptors, AMPA/genetics
- Receptors, AMPA/metabolism
- Sequence Homology, Amino Acid
- Synapses/metabolism
- Synapses/ultrastructure
Collapse
Affiliation(s)
- A H Sharp
- Department of Physiology and Biophysics, Case Western Reserve University, School of Medicine, Cleveland, OH 44106-4970, USA
| | | | | | | | | | | | | | | |
Collapse
|
19
|
Abstract
Voltage-gated Ca(2+) channels mediate Ca(2+) entry into cells in response to membrane depolarization. Electrophysiological studies reveal different Ca(2+) currents designated L-, N-, P-, Q-, R-, and T-type. The high-voltage-activated Ca(2+) channels that have been characterized biochemically are complexes of a pore-forming alpha1 subunit of approximately 190-250 kDa; a transmembrane, disulfide-linked complex of alpha2 and delta subunits; an intracellular beta subunit; and in some cases a transmembrane gamma subunit. Ten alpha1 subunits, four alpha2delta complexes, four beta subunits, and two gamma subunits are known. The Cav1 family of alpha1 subunits conduct L-type Ca(2+) currents, which initiate muscle contraction, endocrine secretion, and gene transcription, and are regulated primarily by second messenger-activated protein phosphorylation pathways. The Cav2 family of alpha1 subunits conduct N-type, P/Q-type, and R-type Ca(2+) currents, which initiate rapid synaptic transmission and are regulated primarily by direct interaction with G proteins and SNARE proteins and secondarily by protein phosphorylation. The Cav3 family of alpha1 subunits conduct T-type Ca(2+) currents, which are activated and inactivated more rapidly and at more negative membrane potentials than other Ca(2+) current types. The distinct structures and patterns of regulation of these three families of Ca(2+) channels provide a flexible array of Ca(2+) entry pathways in response to changes in membrane potential and a range of possibilities for regulation of Ca(2+) entry by second messenger pathways and interacting proteins.
Collapse
Affiliation(s)
- W A Catterall
- Department of Pharmacology, Box 357280, University of Washington, Seattle, Washington 98195-7280, USA.
| |
Collapse
|
20
|
Schiff ML, Siderovski DP, Jordan JD, Brothers G, Snow B, De Vries L, Ortiz DF, Diversé-Pierluissi M. Tyrosine-kinase-dependent recruitment of RGS12 to the N-type calcium channel. Nature 2000; 408:723-7. [PMID: 11130074 DOI: 10.1038/35047093] [Citation(s) in RCA: 126] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Gamma-aminobutyric acid (GABA)B receptors couple to Go to inhibit N-type calcium channels in embryonic chick dorsal root ganglion neurons. The voltage-independent inhibition, mediated by means of a tyrosine-kinase pathway, is transient and lasts up to 100 seconds. Inhibition of endogenous RGS12, a member of the family of regulators of G-protein signalling, selectively alters the time course of voltage-independent inhibition. The RGS12 protein, in addition to the RGS domain, contains PDZ and PTB domains. Fusion proteins containing the PTB domain of RGS12 alter the rate of termination of the GABA(B) signal, whereas the PDZ or RGS domains of RGS 12 have no observable effects. Using primary dorsal root ganglion neurons in culture, here we show an endogenous agonist-induced tyrosine-kinase-dependent complex of RGS12 and the calcium channel. These results indicate that RGS12 is a multifunctional protein capable of direct interactions through its PTB domain with the tyrosine-phosphorylated calcium channel. Recruitment of RGS proteins to G-protein effectors may represent an additional mechanism for signal termination in G-protein-coupled pathways.
Collapse
Affiliation(s)
- M L Schiff
- Department of Pharmacology, Mount Sinai School of Medicine, New York, New York 10029, USA
| | | | | | | | | | | | | | | |
Collapse
|
21
|
Yan L, Adams ME. The spider toxin omega-Aga IIIA defines a high affinity site on neuronal high voltage-activated calcium channels. J Biol Chem 2000; 275:21309-16. [PMID: 10787403 DOI: 10.1074/jbc.m000212200] [Citation(s) in RCA: 19] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The spider toxin omega-agatoxin IIIA (omega-Aga-IIIA) is a potent inhibitor of high voltage-activated calcium currents in the mammalian brain. To establish the biochemical parameters governing its action, we radiolabeled the toxin and examined its binding to native and recombinant calcium channels. In experiments with purified rat synaptosomal membranes, both kinetic and equilibrium data demonstrate one-to-one binding of omega-Aga-IIIA to a single population of high affinity sites, with K(d) = approximately 9 pm and B(max) = approximately 1.4 pmol/mg protein. Partial inhibition of omega-Aga-IIIA binding by omega-conotoxins GVIA, MVIIA, and MVIIC identifies N and P/Q channels as components of this population. omega-Aga-IIIA binds to recombinant alpha(1B) and alpha(1E) calcium channels with a similar high affinity (K(d) = approximately 5-9 pm) in apparent one-to-one fashion. Results from recombinant alpha(1B) binding experiments demonstrate virtually identical B(max) values for omega-Aga-IIIA and omega-conotoxin MVIIA, providing further evidence for a one-to-one stoichiometry of agatoxin binding to calcium channels. The combined evidence suggests that omega-Aga-IIIA defines a unique, high affinity binding site on N-, P/Q-, and R-type calcium channels.
Collapse
Affiliation(s)
- L Yan
- Environmental Toxicology Graduate Program and Departments of Entomology and Neuroscience, University of California, Riverside, California 92521, USA
| | | |
Collapse
|
22
|
Gao B, Sekido Y, Maximov A, Saad M, Forgacs E, Latif F, Wei MH, Lerman M, Lee JH, Perez-Reyes E, Bezprozvanny I, Minna JD. Functional properties of a new voltage-dependent calcium channel alpha(2)delta auxiliary subunit gene (CACNA2D2). J Biol Chem 2000; 275:12237-42. [PMID: 10766861 PMCID: PMC3484885 DOI: 10.1074/jbc.275.16.12237] [Citation(s) in RCA: 118] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
We have positionally cloned and characterized a new calcium channel auxiliary subunit, alpha(2)delta-2 (CACNA2D2), which shares 56% amino acid identity with the known alpha(2)delta-1 subunit. The gene maps to the critical human tumor suppressor gene region in chromosome 3p21.3, showing very frequent allele loss and occasional homozygous deletions in lung, breast, and other cancers. The tissue distribution of alpha(2)delta-2 expression is different from alpha(2)delta-1, and alpha(2)delta-2 mRNA is most abundantly expressed in lung and testis and well expressed in brain, heart, and pancreas. In contrast, alpha(2)delta-1 is expressed predominantly in brain, heart, and skeletal muscle. When co-expressed (via cRNA injections) with alpha(1B) and beta(3) subunits in Xenopus oocytes, alpha(2)delta-2 increased peak size of the N-type Ca(2+) currents 9-fold, and when co-expressed with alpha(1C) or alpha(1G) subunits in Xenopus oocytes increased peak size of L-type channels 2-fold and T-type channels 1.8-fold, respectively. Anti-peptide antibodies detect the expression of a 129-kDa alpha(2)delta-2 polypeptide in some but not all lung tumor cells. We conclude that the alpha(2)delta-2 gene encodes a functional auxiliary subunit of voltage-gated Ca(2+) channels. Because of its chromosomal location and expression patterns, CACNA2D2 needs to be explored as a potential tumor suppressor gene linking Ca(2+) signaling and lung, breast, and other cancer pathogenesis. The homologous location on mouse chromosome 9 is also the site of the mouse neurologic mutant ducky (du), and thus, CACNA2D2 is also a candidate gene for this inherited idiopathic generalized epilepsy syndrome.
Collapse
Affiliation(s)
- Boning Gao
- Hamon Center for Therapeutic Oncology Research, Department of Internal Medicine, Pharmacology, University of Texas, Southwestern Medical Center, Dallas, Texas 75390
| | - Yoshitaka Sekido
- Hamon Center for Therapeutic Oncology Research, Department of Internal Medicine, Pharmacology, University of Texas, Southwestern Medical Center, Dallas, Texas 75390
| | - Anton Maximov
- Department of Physiology, University of Texas, Southwestern Medical Center, Dallas, Texas 75390
| | - Mohamad Saad
- Hamon Center for Therapeutic Oncology Research, Department of Internal Medicine, Pharmacology, University of Texas, Southwestern Medical Center, Dallas, Texas 75390
| | - Eva Forgacs
- Hamon Center for Therapeutic Oncology Research, Department of Internal Medicine, Pharmacology, University of Texas, Southwestern Medical Center, Dallas, Texas 75390
| | - Farida Latif
- University of Birmingham, Birmingham B15 2TT, United Kingdom
| | - Ming H. Wei
- Laboratory of Immunobiology, NCI-Frederick Cancer Research and Development Center, Frederick, Maryland 21702
| | - Michael Lerman
- Laboratory of Immunobiology, NCI-Frederick Cancer Research and Development Center, Frederick, Maryland 21702
| | - Jung-Ha Lee
- Department of Pharmacology, University of Virginia, Charlottesville, Virginia 22908
| | - Edward Perez-Reyes
- Department of Pharmacology, University of Virginia, Charlottesville, Virginia 22908
| | - Ilya Bezprozvanny
- Department of Physiology, University of Texas, Southwestern Medical Center, Dallas, Texas 75390
| | - John D. Minna
- Hamon Center for Therapeutic Oncology Research, Department of Internal Medicine, Pharmacology, University of Texas, Southwestern Medical Center, Dallas, Texas 75390
- To whom correspondence should be addressed: Hamon Center for Therapeutic Oncology Research, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd., Dallas, TX 75390-8593. Tel.: 214-648-4900; Fax: 214-648-4940;
| |
Collapse
|
23
|
Wakamori M, Mikala G, Mori Y. Auxiliary subunits operate as a molecular switch in determining gating behaviour of the unitary N-type Ca2+ channel current in Xenopus oocytes. J Physiol 1999; 517 ( Pt 3):659-72. [PMID: 10358108 PMCID: PMC2269381 DOI: 10.1111/j.1469-7793.1999.0659s.x] [Citation(s) in RCA: 48] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
Abstract
1. We systematically examined the biophysical properties of omega-conotoxin GVIA-sensitive neuronal N-type channels composed of various combinations of the alpha1B, alpha2/delta and beta1b subunits in Xenopus oocytes. 2. Whole-cell recordings demonstrated that coexpression of the beta1b subunit decelerated inactivation, whereas the alpha2/delta accelerated both activation and inactivation, and cancelled the kinetic effects of the beta1b. The alpha2/delta and the beta1b controlled voltage dependence of activation differently: the beta1b significantly shifted the current-voltage relationship towards the hyperpolarizing direction; however, the alpha2/delta shifted the relationship only slightly in the depolarizing direction. The extent of voltage-dependent inactivation was modified solely by the beta1b. 3. Unitary currents measured using a cell-attached patch showed stable patterns of opening that were markedly different among subunit combinations in their kinetic parameters. The alpha2/delta and the beta1b subunits also acted antagonistically in regulating gating patterns of unitary N-type channels. Open time was shortened by the alpha2/delta, while the fraction of long opening was enhanced by the beta1b. The alpha2/delta decreased opening probability (Po), while the beta1b increased Po. alpha1Balpha2/deltabeta1b produced unitary activity with an open time distribution value in between those of alpha1Balpha2/delta and alpha1Bbeta1b. However, both the alpha2/delta and the beta1b subunits reduced the number of null traces. 4. These results suggest that the auxiliary subunits alone and in combination contribute differently in forming gating apparatuses in the N-type channel, raising the possibility that subunit interaction contributes to the generation of functional diversity of N-type channels in native neuronal preparations also.
Collapse
Affiliation(s)
- M Wakamori
- Department of Information Physiology, National Institute for Physiological Sciences, Okazaki, Aichi 444-8585, Japan
| | | | | |
Collapse
|
24
|
Vance CL, Begg CM, Lee WL, Dubel SJ, Copeland TD, Sönnichsen FD, McEnery MW. N-type calcium channel/syntaxin/SNAP-25 complex probed by antibodies to II-III intracellular loop of the alpha1B subunit. Neuroscience 1999; 90:665-76. [PMID: 10215168 DOI: 10.1016/s0306-4522(98)00420-5] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Neuronal voltage-dependent calcium channels are integral components of cellular excitation and neurosecretion. In addition to mediating the entry of calcium across the plasma membrane, both N-type and P/Q-type voltage-dependent calcium channels have been shown to form stable complexes with synaptic vesicle and presynaptic membrane proteins, indicating a structural role for the voltage-dependent calcium channels in secretion. Recently, detailed structural analyses of N-type calcium channels have identified residues amino acids 718-963 as the site in the rat alpha1B subunit that mediates binding to syntaxin, synaptosome-associated protein of 25,000 mol. wt and synaptotagmin [Sheng et al. (1996) Nature 379, 451-454]. The purpose of this study was to employ site-directed antibodies to target domains within and outside of the interaction site on the rat alpha1B to probe potential binding sites for syntaxin/SNAP-25/synaptotagmin. Our results demonstrate that both antibodies employed in this study have access to their epitopes on the alpha1B as evidenced by equivalent immunoprecipitation of native [125I]omega-conotoxin GVIA-labeled alpha1B protein from CHAPS-solubilized preparations. The N-type voltage-dependent calcium channel immunoprecipitated by Ab CW14, the antibody directed to a domain outside of the synprint site, is associated with syntaxin and SNAP-25 with the recovery of these proteins, increasing in parallel to the recovery of alpha1B. However, when we used the antibody raised to an epitope within the synprint site (Ab CW8) to immunoprecipitate N-type calcium channels, the alpha1B was depleted of more than 65% of syntaxin and 80% of SNAP-25 when compared to the recovery of these proteins using Ab CW14. This is the first report of a defined epitope on the alpha1B subunit II-III loop (amino acids 863-875) whose perturbation by a site-directed antibody influences the dissociation of SNAP-25 and syntaxin.
Collapse
Affiliation(s)
- C L Vance
- Department of Physiology and Biophysics, Case Western Reserve University School of Medicine, Cleveland, OH 44106-4970, USA
| | | | | | | | | | | | | |
Collapse
|
25
|
Seagar M, Lévêque C, Charvin N, Marquèze B, Martin-Moutot N, Boudier JA, Boudier JL, Shoji-Kasai Y, Sato K, Takahashi M. Interactions between proteins implicated in exocytosis and voltage-gated calcium channels. Philos Trans R Soc Lond B Biol Sci 1999; 354:289-97. [PMID: 10212477 PMCID: PMC1692480 DOI: 10.1098/rstb.1999.0380] [Citation(s) in RCA: 64] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Neurotransmitter release from synaptic vesicles is triggered by voltage-gated calcium influx through P/Q-type or N-type calcium channels. Purification of N-type channels from rat brain synaptosomes initially suggested molecular interactions between calcium channels and two key proteins implicated in exocytosis: synaptotagmin I and syntaxin 1. Co-immunoprecipitation experiments were consistent with the hypothesis that both N- and P/Q-type calcium channels, but not L-type channels, are associated with the 7S complex containing syntaxin 1, SNAP-25, VAMP and synaptotagmin I or II. Immunofluorescence confocal microscopy at the frog neuromuscular junction confirmed that calcium channels, syntaxin 1 and SNAP-25 are co-localized at active zones of the presynaptic plasma membrane where transmitter release occurs. Experiments with recombinant proteins were performed to map synaptic protein interaction sites on the alpha 1A subunit, which forms the pore of the P/Q-type calcium channel. In vitro-translated 35S-synaptotagmin I bound to a site located on the cytoplasmic loop linking homologous domains II and III of the alpha 1A subunit. This direct link would target synaptotagmin, a putative calcium sensor for exocytosis, to a microdomain of calcium influx close to the channel mouth. Cysteine string proteins (CSPs) contain a J-domain characteristic of molecular chaperones that cooperate with Hsp70. They are located on synaptic vesicles and thought to be involved in modulating the activity of presynaptic calcium channels. CSPs were found to bind to the same domain of the calcium channel as synaptotagmin, and also to associate with VAMP. CSPs may act as molecular chaperones in association with Hsp70 to direct assembly or dissociation of multiprotein complexes at the calcium channel.
Collapse
Affiliation(s)
- M Seagar
- INSERM Unité 464, Institut Jean Roche, Faculté de Médecine Secteur Nord, Marseille, France.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
26
|
Mochida S, Yokoyama CT, Kim DK, Itoh K, Catterall WA. Evidence for a voltage-dependent enhancement of neurotransmitter release mediated via the synaptic protein interaction site of N-type Ca2+ channels. Proc Natl Acad Sci U S A 1998; 95:14523-8. [PMID: 9826733 PMCID: PMC24406 DOI: 10.1073/pnas.95.24.14523] [Citation(s) in RCA: 70] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Secretion of neurotransmitters is initiated by voltage-gated calcium influx through presynaptic, voltage-gated N-type calcium channels. These channels interact with the SNARE proteins, which are core components of the exocytosis process, via the synaptic protein interaction (synprint) site in the intracellular loop connecting domains II and III of their alpha1B subunit. Interruption of this interaction by competing synprint peptides inhibits fast, synchronous transmitter release. Here we identify a voltage-dependent, but calcium-independent, enhancement of transmitter release that is elicited by trains of action potentials in the presence of a hyperosmotic extracellular concentration of sucrose. This enhancement of transmitter release requires interaction of SNARE proteins with the synprint site. Our results provide evidence for a voltage-dependent signal that is transmitted by protein-protein interactions from the N-type calcium channel to the SNARE proteins and enhances neurotransmitter release by altering SNARE protein function.
Collapse
Affiliation(s)
- S Mochida
- Department of Physiology, Tokyo Medical College, Tokyo 160, Japan
| | | | | | | | | |
Collapse
|
27
|
McEnery MW, Copeland TD, Vance CL. Altered expression and assembly of N-type calcium channel alpha1B and beta subunits in epileptic lethargic (lh/lh) mouse. J Biol Chem 1998; 273:21435-8. [PMID: 9705268 DOI: 10.1074/jbc.273.34.21435] [Citation(s) in RCA: 56] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Voltage-dependent calcium channels (VDCC) are multisubunit complexes whose expression and targeting require the assembly of the pore-forming alpha1 with auxiliary beta and alpha2/delta subunits. The developmentally regulated expression and differential assembly of beta isoforms with the alpha1B subunit to form N-type VDCC suggested a unique role for the beta4 isoform in VDCC maturation (Vance, C. L., Begg, C. M., Lee, W.-L., Haase, H., Copeland, T. D., and McEnery, M. W. (1998) J. Biol. Chem. 273, 14495-14502). The focus of this study is the expression and assembly of alpha1B and beta isoforms in the epileptic mouse, lethargic (lh/lh), a mutant anticipated to produce a truncated beta4 subunit (Burgess, D. L., Jones, J. M., Meisler, M. H., and Noebels, J. L. (1997) Cell 88, 385-392). In this report, we demonstrate that neither full-length nor truncated beta4 protein is expressed in lh/lh mice. The absence of beta4 in lh/lh mice is associated with decreased expression of N-type VDCC in forebrain and cerebellum. The most surprising characteristic of the lh/lh mouse is increased expression of beta1b protein. This result suggests a previously unidentified cellular mechanism wherein expression of the total pool of available beta subunits is under tight metabolic regulation. As a consequence of increased beta1b expression, the beta1b is increased in its incorporation into alpha1B/beta complexes relative to wild type. Thus, in striking similarity to the population of N-type VDCC present in immature rat brain, the population of N-type VDCC present in adult lh/lh mice is characterized by the absence of beta4 with increased beta1b expression and assembly into N-type VDCC. It is intriguing to speculate that the increased excitability and susceptibility to seizures observed in the lh/lh mouse arises from the inappropriate expression of an immature population of N-type VDCC throughout neuronal development.
Collapse
Affiliation(s)
- M W McEnery
- Department of Physiology and Biophysics, Case Western Reserve University, School of Medicine, Cleveland, Ohio 44106-4970, USA.
| | | | | |
Collapse
|
28
|
McEnery MW, Vance CL, Begg CM, Lee WL, Choi Y, Dubel SJ. Differential expression and association of calcium channel subunits in development and disease. J Bioenerg Biomembr 1998; 30:409-18. [PMID: 9758336 DOI: 10.1023/a:1021997924473] [Citation(s) in RCA: 46] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
Voltage-gated calcium channels (VDCC) are essential to neuronal maturation and differentiation. It is believed that important signaling information is encoded by VDCC-mediated calcium influx that has both spatial and temporal components. VDCC are multimeric complexes comprised of a pore-forming alpha1 subunit and auxiliary beta and alpha2/delta subunits. Changes in the fractional contribution of distinct calcium conductances to the total calcium current have been noted in developing and differentiating neurons. These changes are anticipated to reflect the differential expression and localization of the pore-forming alpha1 subunits. However, as in vitro studies have established that beta regulates the channel properties and targeting of alpha1, attention has been directed toward the developmental expression and assembly of beta isoforms. Recently, changes in the beta component of the omega-conotoxin GVIA (CTX)-sensitive N-type VDCC have indicated differential assembly of alpha1B with beta in postnatal rat brain. In addition, unique properties of beta4 have been noted with respect to its temporal pattern of expression and incorporation into N-type VDCC complexes. Therefore, the expression and assembly of specific alpha1/beta complexes may reflect an elaborate cellular strategy for regulating VDCC diversity. The importance of these developmental findings is bolstered by a recent study which identified mutations in the beta4 as the molecular defect in the mutant epileptic mouse (lethargic; lh/lh). As beta4 is normally expressed in both forebrain and cerebellum, one may consider the impact of the loss of beta4 upon VDCC assembly and activity. The importance of the beta1b and beta4 isoforms to calcium channel maturation and assembly is discussed.
Collapse
Affiliation(s)
- M W McEnery
- Department of Physiology and Biophysics, Case Western Reserve University, School of Medicine, Cleveland, Ohio 44106-4970, USA
| | | | | | | | | | | |
Collapse
|
29
|
Seagar M, Takahashi M. Interactions between presynaptic calcium channels and proteins implicated in synaptic vesicle trafficking and exocytosis. J Bioenerg Biomembr 1998; 30:347-56. [PMID: 9758331 DOI: 10.1023/a:1021937605818] [Citation(s) in RCA: 42] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
Monoclonal antibodies were generated by immunizing mice with chick brain synaptic membranes and screening for immunoprecipitation of solubilized omega conotoxin GVIA receptors (N-type calcium channels). Antibodies against two synaptic proteins (p35--syntaxin 1 and p58--synaptotagmin) were produced and used to purify and characterize a ternary complex containing N-type channels associated with these two proteins. These results provided the first evidence for a specific interaction between presynaptic calcium channels and SNARE proteins involved in synaptic vesicle docking and calcium-dependent exocytosis. Immunoprecipitation experiments supported the conclusion that syntaxin 1/SNAP-25/VAMP/synaptotagmin I or II complexes associate with N-type, P/Q-type, but not L-type calcium channels from rat brain nerve terminals. Immunofluorescent confocal microscopy at the frog neuromuscular junction was consistent with the co-localization of syntaxin 1, SNAP-25, and calcium channels, all of which are predominantly expressed at active zones of the presynaptic plasma membrane facing post-synaptic folds rich in acetylcholine receptors. The interaction of proteins implicated in calcium-dependent exocytosis with presynaptic calcium channels may locate the sensor(s) that trigger vesicle fusion within a microdomain of calcium entry.
Collapse
Affiliation(s)
- M Seagar
- INSERM Unité 464, Institut Jean Roche, Faculté de Médecine, Secteur Nord, Marseille, France
| | | |
Collapse
|
30
|
Sher E, Rosa P, Francolini M, Codignola A, Morlacchi E, Taverna E, Giovannini F, Brioschi A, Clementi F, McEnery MW, Passafaro M. Metabolism and trafficking of N-type voltage-operated calcium channels in neurosecretory cells. J Bioenerg Biomembr 1998; 30:399-407. [PMID: 9758335 DOI: 10.1023/a:1021945907635] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
The N-type voltage-operated calcium channel has been characterized over the years as a high-threshold channel, with variable inactivation kinetics, and a unique ability to bind with high affinity and specificity omega-conotoxin GVIA and related toxins. This channel is particularly expressed in some neurons and endocrine cells, where it participates in several calcium-dependent processes, including secretion. Omega-conotoxin GVIA was instrumental not only for the biophysical and pharmacological characterization of N-type channels but also for the development of in vitro assays for studying N-type VOCC subcellular localization, biosynthesis, turnover, as well as short-and long-term regulation of its expression. We here summarize our studies on N-type VOCC expression in neurosecretory cells, with a major emphasis on recent data demonstrating the presence of N-type channels in intracellular secretory organelles and their recruitment to the cell surface during regulated exocytosis.
Collapse
Affiliation(s)
- E Sher
- CNR Center of Cellular and Molecular Pharmacology, Department of Medical Pharmacology, University of Milan, Italy
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Colston JT, Valdes JJ, Chambers JP. Ca2+ channel alpha 1-subunit transcripts are differentially expressed in rat pheochromocytoma (PC12) cells following nerve growth factor treatment. Int J Dev Neurosci 1998; 16:379-89. [PMID: 9829174 DOI: 10.1016/s0736-5748(98)00036-7] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
In this report, we describe the effect of nerve growth factor (NGF) on the transcriptional expression of voltage-dependent Ca2+ channel alpha 1 subunits, i.e., alpha 1A, alpha 1B, alpha 1C, alpha 1D, and alpha 1E in rat pheochromocytoma (PC12) cells. Using reverse transcriptase-coupled polymerase chain reaction (RT-PCR) and class-specific Ca2+ channel oligonucleotide probes, messenger RNA levels were measured and compared to Histone H3.3 transcript which remained relatively constant over the duration of NGF treatment. Although no statistically significant differences in P-type (alpha 1A) Ca2+ channel transcript levels were observed, N-type (alpha 1B) Ca2+ channel transcript levels increased 50% over control values (P values < 0.05) at days 7 and 14. In contrast, NGF treatment resulted in decreased levels of L-type (alpha 1C and alpha 1D) transcripts with alpha 1C decreasing steadily to approximately 50% of control (P value < 0.01) by 2 weeks, while alpha 1D decreased to approximately 20% of control (P value < 0.01) after 2 days treatment. No alpha 1E Ca2+ channel transcripts were detected in PC12 cells. For comparison, PC12 cells were also treated with another differentiative growth factor, i.e., basic fibroblast growth factor (bFGF) and a nondifferentiative growth factor epidermal growth factor (EGF). In contrast to NGF, bFGF and EGF treatment had no inhibitory effect on L-type (alpha 1C and alpha 1D) channel transcript levels after 3 days. Like NGF, EGF treatment had no statistically significant effect upon P-type (alpha 1A) transcript levels but increased in a biphasic manner following bFGF treatment. Presynaptic-associated alpha 1B (N-type) Ca2+ channel transcripts were observed decreased following EGF treatment (2 days) while L-type alpha 1C transcripts decreased after 7 days (P value < 0.01). Although a varied response to differentiative growth factors NGF and bFGF was observed, data presented here indicate that NGF treatment of PC12 cells results in 'late' increased expression of N-type Ca2+ channel transcripts, while L-type (alpha 1C and alpha 1D) Ca2+ channel transcripts appear to be down regulated.
Collapse
Affiliation(s)
- J T Colston
- Brain Research Laboratory of Biochemistry, University of Texas at San Antonio 78249, USA
| | | | | |
Collapse
|
32
|
Vance CL, Begg CM, Lee WL, Haase H, Copeland TD, McEnery MW. Differential expression and association of calcium channel alpha1B and beta subunits during rat brain ontogeny. J Biol Chem 1998; 273:14495-502. [PMID: 9603963 DOI: 10.1074/jbc.273.23.14495] [Citation(s) in RCA: 79] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Calcium functions as an essential second messenger during neuronal development and synapse acquisition. Voltage-dependent calcium channels (VDCC), which are critical to these processes, are heteromultimeric complexes composed of alpha1, alpha2/delta, and beta subunits. beta subunits function to direct the VDCC complex to the plasma membrane as well as regulate its channel properties. The importance of beta to neuronal functioning was recently underscored by the identification of a truncated beta4 isoform in the epileptic mouse lethargic (lh) (Burgess, D. L., Jones, J. M., Meisler, M. H., and Noebels, J. L. (1997) Cell 88, 385-392). The goal of our study was to investigate the role of individual beta isoforms (beta1b, beta2, beta3, and beta4) in the assembly of N-type VDCC during rat brain development. By using quantitative Western blot analysis with anti-alpha1B-directed antibodies and [125I-Tyr22]omega-conotoxin GVIA (125I-CTX) radioligand binding assays, we observed that only a small fraction of the total alpha1B protein present in embryonic and early postnatal brain expressed high affinity 125I-CTX-binding sites. These results suggested that subsequent maturation of alpha1B or its assembly with auxiliary subunits was required to exhibit high affinity 125I-CTX binding. The temporal pattern of expression of beta subunits and their assembly with alpha1B indicated a developmental pattern of expression of beta isoforms: beta1b increased 3-fold from P0 to adult, beta4 increased 10-fold, and both beta2 and beta3 expression remained unchanged. As the beta component of N-type VDCC changed during postnatal development, we were able to identify both immature and mature forms of N-type VDCC. At P2, the relative contribution of beta is beta1b > beta3 >> beta2, whereas at P14 and adult the distribution is beta3 > beta1b = beta4. Although we observed no beta4 associated with the alpha1B at P2, beta4 accounted for 14 and 25% of total alpha1B/beta subunit complexes in P14 and adult, respectively. Thus, of the beta isoforms analyzed, only the beta4 was assembled with the rat alpha1B to form N-type VDCC with a time course that paralleled its level of expression during rat brain development. These results suggest a role for the beta4 isoform in the assembly and maturation of the N-type VDCC.
Collapse
Affiliation(s)
- C L Vance
- Department of Physiology and Biophysics, Case Western Reserve University School of Medicine, Cleveland, Ohio 44106-4970, USA
| | | | | | | | | | | |
Collapse
|
33
|
Evidence for a 95 kDa short form of the alpha1A subunit associated with the omega-conotoxin MVIIC receptor of the P/Q-type Ca2+ channels. J Neurosci 1998. [PMID: 9425006 DOI: 10.1523/jneurosci.18-02-00641.1998] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Neuronal voltage-dependent Ca2+ channels have been isolated previously and shown to contain a primary alpha1 pore-forming subunit as well as auxiliary alpha2delta and beta subunits, in addition to an uncharacterized 95 kDa protein. In the present study, using multiple approaches, we have extensively characterized the molecular structure of the 95 kDa protein. Separation of the P/Q- and N-type neuronal Ca2+ channels showed that the 95 kDa protein is associated exclusively with the omega-Conotoxin MVIIC receptor of the P/Q-type channels. Analysis of purified synaptic plasma membranes and the isolated P/Q-type channels, using alpha1A-specific antibodies, suggested a structural relationship between the alpha1A subunit and the 95 kDa protein. This finding was supported by protein-protein interaction data, which revealed that the beta subunit can associate with the 95 kDa protein in addition to the alpha1A subunit. Changes in electrophoretic mobility after enzymatic treatment with Endo F indicated that the 95 kDa protein is glycosylated. Furthermore, microsequencing of the 95 kDa protein yielded 13 peptide sequences, all of which are present in the first half of the alpha1A subunit up to amino acid 829 of the cytoplasmic linker between repeats II and III. Taken together, our results strongly suggest that the 95 kDa glycoprotein associated with the P/Q-type Ca2+ channels is a short form of the alpha1A subunit.
Collapse
|
34
|
Scott VE, Felix R, Arikkath J, Campbell KP. Evidence for a 95 kDa short form of the alpha1A subunit associated with the omega-conotoxin MVIIC receptor of the P/Q-type Ca2+ channels. J Neurosci 1998; 18:641-7. [PMID: 9425006 PMCID: PMC6792535] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Neuronal voltage-dependent Ca2+ channels have been isolated previously and shown to contain a primary alpha1 pore-forming subunit as well as auxiliary alpha2delta and beta subunits, in addition to an uncharacterized 95 kDa protein. In the present study, using multiple approaches, we have extensively characterized the molecular structure of the 95 kDa protein. Separation of the P/Q- and N-type neuronal Ca2+ channels showed that the 95 kDa protein is associated exclusively with the omega-Conotoxin MVIIC receptor of the P/Q-type channels. Analysis of purified synaptic plasma membranes and the isolated P/Q-type channels, using alpha1A-specific antibodies, suggested a structural relationship between the alpha1A subunit and the 95 kDa protein. This finding was supported by protein-protein interaction data, which revealed that the beta subunit can associate with the 95 kDa protein in addition to the alpha1A subunit. Changes in electrophoretic mobility after enzymatic treatment with Endo F indicated that the 95 kDa protein is glycosylated. Furthermore, microsequencing of the 95 kDa protein yielded 13 peptide sequences, all of which are present in the first half of the alpha1A subunit up to amino acid 829 of the cytoplasmic linker between repeats II and III. Taken together, our results strongly suggest that the 95 kDa glycoprotein associated with the P/Q-type Ca2+ channels is a short form of the alpha1A subunit.
Collapse
Affiliation(s)
- V E Scott
- Howard Hughes Medical Institute, Department of Physiology, University of Iowa College of Medicine, Iowa City, Iowa 52242, USA
| | | | | | | |
Collapse
|
35
|
McEnery MW, Haase H, Vance CL, Dubel SJ, Morano I, Copeland TD, Choi Y. Beta1B subunit of voltage-dependent Ca2+ channels is predominant isoform expressed in human neuroblastoma cell line IMR32. FEBS Lett 1997; 420:74-8. [PMID: 9450553 DOI: 10.1016/s0014-5793(97)01490-7] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Human neuroblastoma cells (IMR32) respond to treatment with either dibutyryl-cAMP or nerve factor by acquiring a neuronal phenotype which is accompanied by a marked increase in the density of neuronal (N-type) VDCC currents. Using IMR32 cells as a model for neuronal differentiation, we were interested in examining possible changes in the level of expression of the alpha1B subunit of N-type calcium channels as well as beta subunit isoforms. Upon differentiation with dibutyryl-cAMP and 5-bromo-2-deoxyuridine for 16 days, we observed a dramatic increase in alpha1B protein which initiated between day 8 and 10. Day 10 evidenced maximal expression of alpha1B protein, which was followed by an interval of relatively constant expression of alpha1B (day 12 to day 16). Monitoring beta subunit expression using a pan specific anti-beta antibody (Ab CW20), we observed an increase in expression of a single 82 kDa beta subunit. The predominant 82 kDa beta subunit expressed throughout the course of differentiation was identified as the beta1b isoform using a panel of beta subunit specific antibodies. Of significance, neither the beta2 nor beta3 isoforms were detected in full differentiated IMR32 cells. Contrary to a previous report on the absence of neurotypic expression of VDCC beta subunits in a second model for in vitro differentiation, NGF-treated rat pheochromocytoma cells (PC12 cells) [1], we report the regulated expression of the beta1b protein in differentiated IMR32 cells suggesting a cell specific function for this beta subunit which parallels the acquisition of the neuronal phenotype. The restrictive expression of the beta1b in IMR32 cells may reflect a cell-type specific function that extends beyond its role as an auxiliary subunit of VDCC complexes.
Collapse
Affiliation(s)
- M W McEnery
- Department of Physiology and Biophysics, Case Western Reserve University, School of Medicine, Cleveland, OH 44106-4970, USA.
| | | | | | | | | | | | | |
Collapse
|
36
|
Choi Y, Dubel SJ, Pacioaiou ML, Omori A, Ito T, Copeland TD, Takahashi M, McEnery MW. Parallel detection of Na,K-ATPase alpha subunit isoforms by pan-specific monoclonal mAb 9A7. Arch Biochem Biophys 1997; 344:165-75. [PMID: 9244394 DOI: 10.1006/abbi.1997.0183] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
While emphasis has been placed upon those proteins which either mediate or respond to the rapid influx of calcium following depolarization, there has been little emphasis upon those proteins which aid in the reequilibration of the membrane potential. In an effort to identify presynaptic membrane proteins implicated in neurosecretion, monoclonal antibodies were screened against proteins which cosegregated with neuronal voltage-dependent calcium channels (VDCC) following immunoprecipitation. One monoclonal antibody (mAb 9A7) identified a 110-kDa protein. Micropeptide sequencing of (i) the mAb 9A7 immunoaffinity purified antigen and (ii) the 110-kDa protein present in the neuronal (N-type) VDCC preparation (McEnery et al., 1991, Proc. Natl. Acad. Sci. 88, 11095-11099) indicated identity with the alpha subunit(s) of the Na,K-ATPase. Further characterization by Western blotting, immunochemical localization, and immunoaffinity purification indicated that mAb 9A7 not only recognized the alpha3 isoform which is predominant in neuronal tissues but also identified the alpha1 and alpha2 isoforms. mAb 9A7 exhibited a wide cross-species reactivity and recognized human, rat, and mouse alpha subunit isoforms at an internal epitope. The pan-specificity of mAb 9A7 and the differential mobility of the alpha1 isoform relative to the alpha2 and alpha3 permitted parallel detection of multiple alpha isoforms. Western blot analysis of undifferentiated rat pheochromocytoma cell line (PC12) and human neuroblastoma (IMR32) cells indicated coexpression of the alpha1 and alpha3 isozymes. Upon differentiation of IMR32 cells by dibutrylyl-cAMP, a substantial increase in the alpha3 relative to the alpha1 isoform was observed. While the enrichment of total Na,K-ATPase may reflect the increased demand for ATP-dependent ion transport as IMR32 cells become more excitable, the specific increase in the alpha3 isoform suggests a unique role of this isoform during IMR32 cell differentiation.
Collapse
Affiliation(s)
- Y Choi
- Department of Physiology and Biophysics, Case Western Reserve University School of Medicine, Cleveland, Ohio 44106, USA
| | | | | | | | | | | | | | | |
Collapse
|
37
|
Abstract
The selective block of N-type Ca2+ channels by omega-conotoxins has been a hallmark of these channels, critical in delineating their biological roles and molecular characteristics. Here we report that the omega-conotoxin-channel interaction depends strongly on channel gating. N-type channels (alpha1B, alpha2, and beta1) expressed in Xenopus oocytes were blocked with a variety of omega-conotoxins, including omega-CTx-GVIA, omega-CTx-MVIIA, and SNX-331, a derivative of omega-CTx-MVIIC. Changes in holding potential (HP) markedly altered the severity of toxin block and the kinetics of its onset and removal. Notably, strong hyperpolarization renders omega-conotoxin block completely reversible. These effects could be accounted for by a modulated receptor model, in which toxin dissociation from the inactivated state is approximately 60-fold slower than from the resting state. Because omega-conotoxins act exclusively outside cells, our results suggest that voltage-dependent inactivation of Ca2+ channels must be associated with an externally detectable conformational change.
Collapse
|
38
|
Stocker JW, Nadasdi L, Aldrich RW, Tsien RW. Preferential interaction of omega-conotoxins with inactivated N-type Ca2+ channels. J Neurosci 1997; 17:3002-13. [PMID: 9096136 PMCID: PMC6573648] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
The selective block of N-type Ca2+ channels by omega-conotoxins has been a hallmark of these channels, critical in delineating their biological roles and molecular characteristics. Here we report that the omega-conotoxin-channel interaction depends strongly on channel gating. N-type channels (alpha1B, alpha2, and beta1) expressed in Xenopus oocytes were blocked with a variety of omega-conotoxins, including omega-CTx-GVIA, omega-CTx-MVIIA, and SNX-331, a derivative of omega-CTx-MVIIC. Changes in holding potential (HP) markedly altered the severity of toxin block and the kinetics of its onset and removal. Notably, strong hyperpolarization renders omega-conotoxin block completely reversible. These effects could be accounted for by a modulated receptor model, in which toxin dissociation from the inactivated state is approximately 60-fold slower than from the resting state. Because omega-conotoxins act exclusively outside cells, our results suggest that voltage-dependent inactivation of Ca2+ channels must be associated with an externally detectable conformational change.
Collapse
Affiliation(s)
- J W Stocker
- Department of Molecular and Cellular Physiology, Stanford University, Stanford, California 94305, USA
| | | | | | | |
Collapse
|
39
|
A Drosophila calcium channel alpha1 subunit gene maps to a genetic locus associated with behavioral and visual defects. J Neurosci 1997. [PMID: 8987815 DOI: 10.1523/jneurosci.16-24-07868.1996] [Citation(s) in RCA: 127] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
We have cloned cDNAs that encode a complete open reading frame for a calcium channel alpha1 subunit from Drosophila melanogaster. The deduced 1851 amino acid protein belongs to the superfamily of voltage-gated sodium and calcium channels. Phylogenetic analysis shows that the sequence of this subunit is relatively distant from sodium channel alpha subunits and most similar to genes encoding the A, B, and E isoforms of calcium channel alpha1 subunits. To indicate its similarity to this subfamily of vertebrate isoforms, we name this protein Dmca1A, for Drosophila melanogaster calcium channel alpha1 subunit, type A. Northern blot analysis detected a single 10. 5 kb transcript class that is regulated developmentally, with expression peaks in the first larval instar, midpupal, and late pupal stages. In late-stage embryos, Dmca1A is expressed preferentially in the nervous system. Variant transcripts are generated by alternative splicing. In addition, single nucleotide variations between cDNAs and genomic sequence are consistent with RNA editing. Dmca1A maps to a chromosomal region implicated in, and is the likely candidate for, the gene involved in the generation of behavioral, physiological, and lethal phenotypes of the cacophony, nightblind-A, and lethal(1)L13 mutants.
Collapse
|
40
|
Abstract
Nerve growth factor (NGF)-induced differentiation in PC12 cells is accompanied by changes in the expression of voltage-dependent Ca2+ channels. Ca2+ channels are multimeric complexes composed of at least three subunits (alpha1, beta, and alpha2delta) and are involved in neuronal migration, gene expression, and neurotransmitter release. Although attempts have been undertaken to elucidate NGF regulation of Ca2+ channel expression, the changes in subunit composition of these channels during differentiation still remain uncertain. In the present study, patch-clamp recordings show that in addition to the previously documented L-type and N-type Ca2+ currents, undifferentiated PC12 cells also express an omega-agatoxin-IVA-sensitive (P/Q-type) component. In addition, the corresponding mRNA encoding the pore-forming alpha1 subunits for these channels (C, B, and A, respectively) was detected. Likewise, mRNA for three distinct auxiliary beta subunits (1, 2, 3) were also found, beta3 protein being dominantly expressed. Immunoprecipitation experiments show that the N-type Ca2+ channel is associated with either a beta2 or beta3 subunit and that NGF increases the channel expression without affecting its beta subunit association. These results (1) indicate that the diversity of Ca2+ currents in PC12 cells arise from the expression of three distinct alpha1 and three different beta subunit genes; (2) support a model for heterogenous beta subunit association of the N-type Ca2+ channel in a single cell type; and (3) suggest that the regulation of the N-type Ca2+ channel during NGF-mediated differentiation involves an increase in the number of functional channels with no apparent changes in subunit composition.
Collapse
|
41
|
Ohizumi Y. Application of Physiologically Active Substances Isolated from Natural Resources to Pharmacological Studies. ACTA ACUST UNITED AC 1997. [DOI: 10.1254/jjp.60.263] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
|
42
|
Smith LA, Wang X, Peixoto AA, Neumann EK, Hall LM, Hall JC. A Drosophila calcium channel alpha1 subunit gene maps to a genetic locus associated with behavioral and visual defects. J Neurosci 1996; 16:7868-79. [PMID: 8987815 PMCID: PMC6579206] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
We have cloned cDNAs that encode a complete open reading frame for a calcium channel alpha1 subunit from Drosophila melanogaster. The deduced 1851 amino acid protein belongs to the superfamily of voltage-gated sodium and calcium channels. Phylogenetic analysis shows that the sequence of this subunit is relatively distant from sodium channel alpha subunits and most similar to genes encoding the A, B, and E isoforms of calcium channel alpha1 subunits. To indicate its similarity to this subfamily of vertebrate isoforms, we name this protein Dmca1A, for Drosophila melanogaster calcium channel alpha1 subunit, type A. Northern blot analysis detected a single 10. 5 kb transcript class that is regulated developmentally, with expression peaks in the first larval instar, midpupal, and late pupal stages. In late-stage embryos, Dmca1A is expressed preferentially in the nervous system. Variant transcripts are generated by alternative splicing. In addition, single nucleotide variations between cDNAs and genomic sequence are consistent with RNA editing. Dmca1A maps to a chromosomal region implicated in, and is the likely candidate for, the gene involved in the generation of behavioral, physiological, and lethal phenotypes of the cacophony, nightblind-A, and lethal(1)L13 mutants.
Collapse
Affiliation(s)
- L A Smith
- Department of Biology, Brandeis University, Waltham, Massachusetts 02254, USA
| | | | | | | | | | | |
Collapse
|
43
|
Liu H, Felix R, Gurnett CA, De Waard M, Witcher DR, Campbell KP. Expression and subunit interaction of voltage-dependent Ca2+ channels in PC12 cells. J Neurosci 1996; 16:7557-65. [PMID: 8922412 PMCID: PMC6579093] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
Nerve growth factor (NGF)-induced differentiation in PC12 cells is accompanied by changes in the expression of voltage-dependent Ca2+ channels. Ca2+ channels are multimeric complexes composed of at least three subunits (alpha1, beta, and alpha2delta) and are involved in neuronal migration, gene expression, and neurotransmitter release. Although attempts have been undertaken to elucidate NGF regulation of Ca2+ channel expression, the changes in subunit composition of these channels during differentiation still remain uncertain. In the present study, patch-clamp recordings show that in addition to the previously documented L-type and N-type Ca2+ currents, undifferentiated PC12 cells also express an omega-agatoxin-IVA-sensitive (P/Q-type) component. In addition, the corresponding mRNA encoding the pore-forming alpha1 subunits for these channels (C, B, and A, respectively) was detected. Likewise, mRNA for three distinct auxiliary beta subunits (1, 2, 3) were also found, beta3 protein being dominantly expressed. Immunoprecipitation experiments show that the N-type Ca2+ channel is associated with either a beta2 or beta3 subunit and that NGF increases the channel expression without affecting its beta subunit association. These results (1) indicate that the diversity of Ca2+ currents in PC12 cells arise from the expression of three distinct alpha1 and three different beta subunit genes; (2) support a model for heterogenous beta subunit association of the N-type Ca2+ channel in a single cell type; and (3) suggest that the regulation of the N-type Ca2+ channel during NGF-mediated differentiation involves an increase in the number of functional channels with no apparent changes in subunit composition.
Collapse
Affiliation(s)
- H Liu
- Program in Neuroscience, University of Iowa College of Medicine, Iowa City 52242, USA
| | | | | | | | | | | |
Collapse
|
44
|
Naciff JM, Behbehani MM, Kaetzel MA, Dedman JR. Annexin VI modulates Ca2+ and K+ conductances of spinal cord and dorsal root ganglion neurons. THE AMERICAN JOURNAL OF PHYSIOLOGY 1996; 271:C2004-15. [PMID: 8997203 DOI: 10.1152/ajpcell.1996.271.6.c2004] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
Annexin VI is a member of a Ca(2+)-dependent phospholipid-binding protein family that participates in the transduction of the intracellular Ca2+ signal. We have identified annexin VI as one of the major annexins expressed differentially by sensory neurons of dorsal root ganglia (DRG) and by neurons of spinal cord (SC) of the rat and the mouse. This annexin shows a preferential localization at the plasma membrane of the soma and cellular processes, particularly in motoneurons of the SC. This finding suggests an active role of annexin VI in the Ca(2+)-dependent regulation of plasma membrane functions. To test this possibility, the neuronal function of annexin VI was evaluated by whole cell electrophysiology of mouse embryo SC and DRG neurons. An antibody was developed that has the property of neutralizing annexin VI-phospholipid interactions. The intracellular perfusion of individual neurons in culture, either from SC or DRG, with monospecific affinity-purified anti-annexin VI antibodies resulted in an increase in the magnitude of the K+ current and in an increase in the Ca2+ current in sensory neurons. Our results suggest that the endogenous annexin VI regulates the Ca2+ conductance, which indirectly modifies Ca(2+)-dependent ionic conductances in SC and DRG neurons.
Collapse
Affiliation(s)
- J M Naciff
- Department of Molecular and Cellular Physiology, University of Cincinnati College of Medicine, Ohio 45267-0576, USA
| | | | | | | |
Collapse
|
45
|
Arsac C, Raymond C, Martin-Moutot N, Dargent B, Couraud F, Pouget J, Seagar M. Immunoassays fail to detect antibodies against neuronal calcium channels in amyotrophic lateral sclerosis serum. Ann Neurol 1996; 40:695-700. [PMID: 8957009 DOI: 10.1002/ana.410400504] [Citation(s) in RCA: 44] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
Recent studies suggested that autoantibodies that bind to voltage-dependent calcium channels and activate calcium entry may play a role in the progressive degeneration of motoneurons in sporadic amyotrophic lateral sclerosis. Immunoassays were performed to assess autoantibody titer in patients with amyotrophic lateral sclerosis or Lambert-Eaton myasthenic syndrome, a disease in which the presence of anti-calcium channel antibodies is well documented. Based on immunoprecipitation assays for antibodies against N-type calcium channels, only 8% (2/25) of amyotrophic lateral sclerosis patients had marginally positive titers, whereas 58% (18/31) of patients with Lambert-Eaton myasthenic syndrome had positive titers. Enzyme-linked immunosorbent assays with purified neuronal N-type calcium channels revealed immunoreactivity in 2 of 25 amyotrophic lateral sclerosis sera and 12 of 31 Lambert-Eaton myasthenic syndrome sera, which is not compatible with suggestions that enzyme-linked immunosorbent assay is a more sensitive technique for the detection of autoantibodies in amyotrophic lateral sclerosis. Furthermore, based on immunoprecipitation assays, amyotrophic lateral sclerosis sera were totally negative for antibodies against L-type calcium channels from skeletal muscle or brain. These data do not support the hypothesis that an autoimmune response against calcium channels plays a primary role in amyotrophic lateral sclerosis.
Collapse
Affiliation(s)
- C Arsac
- INSERM U374, Institut Jean Roche, Faculté de Médecine Secteur Nord, Marseille, France
| | | | | | | | | | | | | |
Collapse
|
46
|
Prado MA, Guatimosim C, Gomez MV, Diniz CR, Cordeiro MN, Romano-Silva MA. A novel tool for the investigation of glutamate release from rat cerebrocortical synaptosomes: the toxin Tx3-3 from the venom of the spider Phoneutria nigriventer. Biochem J 1996; 314 ( Pt 1):145-50. [PMID: 8660275 PMCID: PMC1217017 DOI: 10.1042/bj3140145] [Citation(s) in RCA: 53] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
The present experiments investigated the effect of some of the toxic components present in the venom of the spider Phoneutria nigriventer on the release of neurotransmitter. The toxic fraction, Phoneutria nigriventer toxin-3 (PhTx3), abolished Ca2+-dependent glutamate release, but did not alter Ca2+-independent secretion of glutamate when rat brain cortical synaptosomes were depolarized with 33 mM KCl. This effect was most likely due to interference with the entry of calcium through voltage-gated calcium channels, because PhTx3 reduced by 50% the increase in intrasynaptosomal free calcium induced by membrane depolarization, and did not affect the release of glutamate evoked by a calcium ionophore (ionomycin). A polypeptide (Tx3-3) present in the PhTx3 fraction reproduced the effects of the PhTx3 fraction on transmitter release and intrasynaptosomal free calcium in the low nanomolar range. We compared the alterations produced by the Tx3-3 with the actions of toxins known to block calcium channels coupled to exocytosis: the results indicated that the Tx3-3 inhibition of glutamate release and intrasynaptosomal calcium resemble that observed with omega-conotoxin MVIIC. We suggest that the Tx3-3 is a calcium-channel antagonist that blocked glutamate exocytosis.
Collapse
Affiliation(s)
- M A Prado
- Departamento de Farmacologia, ICB, Universidade Federal de Minas Gerais, Belo Horizonte-MG, Brazil
| | | | | | | | | | | |
Collapse
|
47
|
Yu AS. Identification and localization of calcium channel alpha 1 and beta subunit isoforms in the kidney. Kidney Int 1995; 48:1097-101. [PMID: 8569071 DOI: 10.1038/ki.1995.393] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Affiliation(s)
- A S Yu
- Department of Medicine, Brigham & Women's Hospital, Boston, Massachusetts
| |
Collapse
|
48
|
Abstract
Pharmacological and electrophysiological studies have established that there are multiple types of voltage-gated Ca2+ channels. Molecular biology has uncovered an even greater number of channel molecules. Thus, the molecular diversity of Ca2+ channels has its basis in the expression of many alpha 1 and beta genes, and also in the splice variants produced from these genes. This ability to mix and match subunits provides the cell with yet another mechanism to control the influx of calcium. Future studies will describe new subunits, the subunit composition of each type of channel, and the cloning of new Ca2+ channel types.
Collapse
Affiliation(s)
- E Perez-Reyes
- Department of Physiology, Loyola University Medical Center, Maywood, Illinois, USA
| | | |
Collapse
|
49
|
Adelman JP. Proteins that interact with the pore-forming subunits of voltage-gated ion channels. Curr Opin Neurobiol 1995; 5:286-95. [PMID: 7580150 DOI: 10.1016/0959-4388(95)80040-9] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
Voltage-gated ion channels are composed of pore-forming subunits, as well as auxiliary subunits that modify the functions of these channels. In addition, the channels interact with other modulatory proteins in a more transient manner, although with significant effects on channel activity. Even though many second-messenger systems influence the voltage-gated ion channels, only in a few cases has clear evidence for direct protein-protein interactions been demonstrated. Recent biochemical and genetic studies have helped to elucidate the scope of the interactions between these ion channels and various modulatory proteins by determining the structures and functions of nonpore-forming subunits.
Collapse
Affiliation(s)
- J P Adelman
- Vollum Institute, Oregon Health Sciences University, Portland, USA
| |
Collapse
|
50
|
Ichida S, Wada T, Akimoto T, Kasamatsu Y, Tahara M, Hasimoto K. Characteristics of [125I]omega-conotoxin labeling using bifunctional cross linker DSP in crude membranes from chick brain. Neurochem Res 1995; 20:467-73. [PMID: 7651585 DOI: 10.1007/bf00973104] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
Characteristic of [125I]omega-conotoxin (omega-CgTX) labeling using bifunctional cross linker (dithio bis[succinimidyl propionate]:DSP) was systematically investigated in crude membranes from chick whole brain. [125I]omega-CgTX specifically labeled 216 kDa as a main and 236 kDa as a minor bands in the crude membranes under non-reduced condition, but not labeled under reduced condition. We investigated the effect of various Ca channel antagonists on [125I]omega-CgTX labeling with DSP in detail, and found that there is a strong correlation between the effects of Ca channel antagonists on [125I]omega-CgTX labeling of the 216 kDa band and specific [125I]omega-CgTX binding. These results suggest that labeling of the 216 kDa band under non-reduced condition with [125I]omega-CgTX using DSP involves the specific binding sites of [125I]omega-CgTX, perhaps including one of the neuronal N-type Ca channel subunits in the crude membranes.
Collapse
Affiliation(s)
- S Ichida
- Department of Biological Chemistry, Faculty of Pharmacy, Kinki University, Higashi-Osaka, Japan
| | | | | | | | | | | |
Collapse
|