Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 20
Filter
Add more filters










Publication year range
1.
Function (Oxf) ; 3(3): zqac013, 2022.
Article in English | MEDLINE | ID: mdl-35462614

ABSTRACT

The auxiliary α2δ subunits of voltage-gated calcium (CaV) channels are key to augmenting expression and function of CaV1 and CaV2 channels, and are also important drug targets in several therapeutic areas, including neuropathic pain. The α2δ proteins are translated as preproteins encoding both α2 and δ, and post-translationally proteolyzed into α2 and δ subunits, which remain associated as a complex. In this study, we have identified ADAM17 as a key protease involved in proteolytic processing of pro-α2δ-1 and α2δ-3 subunits. We provide three lines of evidence: First, proteolytic cleavage is inhibited by chemical inhibitors of particular metalloproteases, including ADAM17. Second, proteolytic cleavage of both α2δ-1 and α2δ-3 is markedly reduced in cell lines by knockout of ADAM17 but not ADAM10. Third, proteolytic cleavage is reduced by the N-terminal active domain of TIMP-3 (N-TIMP-3), which selectively inhibits ADAM17. We have found previously that proteolytic cleavage into mature α2δ is essential for the enhancement of CaV function, and in agreement, knockout of ADAM17 inhibited the ability of α2δ-1 to enhance both CaV2.2 and CaV1.2 calcium currents. Finally, our data also indicate that the main site of proteolytic cleavage of α2δ-1 is the Golgi apparatus, although cleavage may also occur at the plasma membrane. Thus, our study identifies ADAM17 as a key protease required for proteolytic maturation of α2δ-1 and α2δ-3, and thus a potential drug target in neuropathic pain.


Subject(s)
Neuralgia , Tissue Inhibitor of Metalloproteinase-3 , Humans , Tissue Inhibitor of Metalloproteinase-3/metabolism , Calcium Channels, N-Type/genetics , Proteolysis , Calcium, Dietary/metabolism , Peptide Hydrolases/metabolism , ADAM17 Protein/genetics
2.
Brain ; 145(8): 2721-2729, 2022 08 27.
Article in English | MEDLINE | ID: mdl-35293990

ABSTRACT

Voltage-gated calcium (CaV) channels form three subfamilies (CaV1-3). The CaV1 and CaV2 channels are heteromeric, consisting of an α1 pore-forming subunit, associated with auxiliary CaVß and α2δ subunits. The α2δ subunits are encoded in mammals by four genes, CACNA2D1-4. They play important roles in trafficking and function of the CaV channel complexes. Here we report biallelic variants in CACNA2D1, encoding the α2δ-1 protein, in two unrelated individuals showing a developmental and epileptic encephalopathy. Patient 1 has a homozygous frameshift variant c.818_821dup/p.(Ser275Asnfs*13) resulting in nonsense-mediated mRNA decay of the CACNA2D1 transcripts, and absence of α2δ-1 protein detected in patient-derived fibroblasts. Patient 2 is compound heterozygous for an early frameshift variant c.13_23dup/p.(Leu9Alafs*5), highly probably representing a null allele and a missense variant c.626G>A/p.(Gly209Asp). Our functional studies show that this amino-acid change severely impairs the function of α2δ-1 as a calcium channel subunit, with strongly reduced trafficking of α2δ-1G209D to the cell surface and a complete inability of α2δ-1G209D to increase the trafficking and function of CaV2 channels. Thus, biallelic loss-of-function variants in CACNA2D1 underlie the severe neurodevelopmental disorder in these two patients. Our results demonstrate the critical importance and non-interchangeability of α2δ-1 and other α2δ proteins for normal human neuronal development.


Subject(s)
Calcium Channels, N-Type , Epilepsy , Age of Onset , Animals , Calcium , Calcium Channels , Calcium Channels, L-Type , Cell Membrane , Humans , Mammals , Neurons
3.
Cell Calcium ; 87: 102180, 2020 05.
Article in English | MEDLINE | ID: mdl-32065986

ABSTRACT

The fight-or-flight response is studied by all students of Physiology as a concerted bodily response to danger. Liu et al (2020) have now revealed its mechanism, after surveying the proteomic neighbourhood around the cardiac calcium channels in a study which is a tour-de-force of modern biological techniques.


Subject(s)
Escape Reaction/physiology , Proteomics , Animals , Calcium Channels/metabolism , HEK293 Cells , Humans , Mice, Transgenic , Models, Biological , Monomeric GTP-Binding Proteins/metabolism
4.
Cell Rep ; 29(1): 22-33.e5, 2019 Oct 01.
Article in English | MEDLINE | ID: mdl-31577951

ABSTRACT

Voltage-gated calcium channels are exquisitely Ca2+ selective, conferred primarily by four conserved pore-loop glutamate residues contributing to the selectivity filter. There has been little previous work directly measuring whether the trafficking of calcium channels requires their ability to bind Ca2+ in the selectivity filter or to conduct Ca2+. Here, we examine trafficking of neuronal CaV2.1 and 2.2 channels with mutations in their selectivity filter and find reduced trafficking to the cell surface in cell lines. Furthermore, in hippocampal neurons, there is reduced trafficking to the somatic plasma membrane, into neurites, and to presynaptic terminals. However, the CaV2.2 selectivity filter mutants are still influenced by auxiliary α2δ subunits and, albeit to a reduced extent, by ß subunits, indicating the channels are not grossly misfolded. Our results indicate that Ca2+ binding in the pore of CaV2 channels may promote their correct trafficking, in combination with auxiliary subunits. Furthermore, physiological studies utilizing selectivity filter mutant CaV channels should be interpreted with caution.


Subject(s)
Binding Sites/physiology , Calcium Channels, N-Type/metabolism , Calcium/metabolism , Neurons/metabolism , Protein Transport/physiology , Animals , Cell Line , Cell Membrane/metabolism , Female , Hippocampus/metabolism , Humans , Male , Mice , Neurites/metabolism , Rats , Rats, Sprague-Dawley
5.
Cell Rep ; 25(6): 1610-1621.e5, 2018 11 06.
Article in English | MEDLINE | ID: mdl-30404013

ABSTRACT

Voltage-gated calcium channel auxiliary α2δ subunits are important for channel trafficking and function. Here, we compare the effects of α2δ-1 and an α2δ-like protein called Cachd1 on neuronal N-type (CaV2.2) channels, which are important in neurotransmission. Previous structural studies show the α2δ-1 VWA domain interacting with the first loop in CaV1.1 domain-I via its metal ion-dependent adhesion site (MIDAS) motif and additional Cache domain interactions. Cachd1 has a disrupted MIDAS motif. However, Cachd1 increases CaV2.2 currents substantially (although less than α2δ-1) and increases CaV2.2 cell surface expression by reducing endocytosis. Although the effects of α2δ-1 are abolished by mutation of Asp122 in CaV2.2 domain-I, which mediates interaction with its VWA domain, the Cachd1 responses are unaffected. Furthermore, Cachd1 co-immunoprecipitates with CaV2.2 and inhibits co-immunoprecipitation of α2δ-1 by CaV2.2. Cachd1 also competes with α2δ-1 for effects on trafficking. Thus, Cachd1 influences both CaV2.2 trafficking and function and can inhibit responses to α2δ-1.


Subject(s)
Calcium Channels, N-Type/metabolism , Calcium Channels/metabolism , Cell Membrane/metabolism , Ion Channel Gating , Membrane Proteins/metabolism , Animals , Calcium Channels/genetics , Calcium Channels, N-Type/genetics , Hippocampus/metabolism , Male , Mutation/genetics , Neurites/metabolism , Protein Binding , Rats, Sprague-Dawley
6.
Elife ; 72018 06 19.
Article in English | MEDLINE | ID: mdl-29916807

ABSTRACT

Auxiliary α2δ subunits are important proteins for trafficking of voltage-gated calcium channels (CaV) at the active zones of synapses. We have previously shown that the post-translational proteolytic cleavage of α2δ is essential for their modulatory effects on the trafficking of N-type (CaV2.2) calcium channels (Kadurin et al., 2016). We extend these results here by showing that the probability of presynaptic vesicular release is reduced when an uncleaved α2δ is expressed in rat neurons and that this inhibitory effect is reversed when cleavage of α2δ is restored. We also show that asynchronous release is influenced by the maturation of α2δ-1, highlighting the role of CaV channels in this component of vesicular release. We present additional evidence that CaV2.2 co-immunoprecipitates preferentially with cleaved wild-type α2δ. Our data indicate that the proteolytic maturation increases the association of α2δ-1 with CaV channel complex and is essential for its function on synaptic release.


Subject(s)
Calcium Channels, N-Type/metabolism , Neurons/metabolism , Protein Subunits/metabolism , Synaptic Vesicles/metabolism , Animals , Animals, Newborn , Biological Transport , Calcium Channels, N-Type/genetics , Gene Expression , Genes, Reporter , Hippocampus/metabolism , Hippocampus/ultrastructure , Immunoprecipitation , Luminescent Proteins/genetics , Luminescent Proteins/metabolism , Neurons/ultrastructure , Primary Cell Culture , Probability , Protein Subunits/genetics , Proteolysis , Rats , Rats, Sprague-Dawley , Synaptic Vesicles/ultrastructure , Red Fluorescent Protein
7.
Sci Rep ; 7: 43802, 2017 03 03.
Article in English | MEDLINE | ID: mdl-28256585

ABSTRACT

Voltage-gated Ca2+ (CaV) channels consist of a pore-forming α1 subunit, which determines the main functional and pharmacological attributes of the channel. The CaV1 and CaV2 channels are associated with auxiliary ß- and α2δ-subunits. The molecular mechanisms involved in α2δ subunit trafficking, and the effect of α2δ subunits on trafficking calcium channel complexes remain poorly understood. Here we show that α2δ-1 is a ligand for the Low Density Lipoprotein (LDL) Receptor-related Protein-1 (LRP1), a multifunctional receptor which mediates trafficking of cargoes. This interaction with LRP1 is direct, and is modulated by the LRP chaperone, Receptor-Associated Protein (RAP). LRP1 regulates α2δ binding to gabapentin, and influences calcium channel trafficking and function. Whereas LRP1 alone reduces α2δ-1 trafficking to the cell-surface, the LRP1/RAP combination enhances mature glycosylation, proteolytic processing and cell-surface expression of α2δ-1, and also increase plasma-membrane expression and function of CaV2.2 when co-expressed with α2δ-1. Furthermore RAP alone produced a small increase in cell-surface expression of CaV2.2, α2δ-1 and the associated calcium currents. It is likely to be interacting with an endogenous member of the LDL receptor family to have these effects. Our findings now provide a key insight and new tools to investigate the trafficking of calcium channel α2δ subunits.


Subject(s)
Calcium Channels, N-Type/metabolism , Cell Membrane/metabolism , LDL-Receptor Related Protein-Associated Protein/metabolism , Low Density Lipoprotein Receptor-Related Protein-1/metabolism , Amines/metabolism , Animals , Calcium Channels, N-Type/genetics , Cell Line , Cyclohexanecarboxylic Acids/metabolism , Gabapentin , Humans , LDL-Receptor Related Protein-Associated Protein/genetics , Low Density Lipoprotein Receptor-Related Protein-1/genetics , Mice, Inbred C57BL , Mice, Knockout , Mutation , Protein Binding , Protein Subunits/genetics , Protein Subunits/metabolism , Protein Transport , Radioligand Assay , gamma-Aminobutyric Acid/metabolism
8.
Elife ; 52016 10 26.
Article in English | MEDLINE | ID: mdl-27782881

ABSTRACT

The auxiliary α2δ subunits of voltage-gated calcium channels are extracellular membrane-associated proteins, which are post-translationally cleaved into disulfide-linked polypeptides α2 and δ. We now show, using α2δ constructs containing artificial cleavage sites, that this processing is an essential step permitting voltage-dependent activation of plasma membrane N-type (CaV2.2) calcium channels. Indeed, uncleaved α2δ inhibits native calcium currents in mammalian neurons. By inducing acute cell-surface proteolytic cleavage of α2δ, voltage-dependent activation of channels is promoted, independent from the trafficking role of α2δ. Uncleaved α2δ does not support trafficking of CaV2.2 channel complexes into neuronal processes, and inhibits Ca2+ entry into synaptic boutons, and we can reverse this by controlled intracellular proteolytic cleavage. We propose a model whereby uncleaved α2δ subunits maintain immature calcium channels in an inhibited state. Proteolytic processing of α2δ then permits voltage-dependent activation of the channels, acting as a checkpoint allowing trafficking only of mature calcium channel complexes into neuronal processes.


Subject(s)
Calcium Channels, N-Type/metabolism , Neurons/enzymology , Protein Processing, Post-Translational , Animals , Mice , Models, Biological , Protein Transport , Proteolysis , Rabbits , Rats
9.
Sci Rep ; 6: 24531, 2016 Apr 14.
Article in English | MEDLINE | ID: mdl-27076051

ABSTRACT

The α2δ proteins are auxiliary subunits of voltage-gated calcium channels, and influence their trafficking and biophysical properties. The α2δ ligand gabapentin interacts with α2δ-1, and inhibits calcium channel trafficking. However, α2-1 has also been proposed to play a synaptogenic role, independent of calcium channel function. In this regard, α2δ-1 was identified as a ligand of thrombospondins, with the interaction involving the thrombospondin synaptogenic domain and the α2δ-1 von-Willebrand-factor domain. Co-immunoprecipitation between α2δ-1 and the synaptogenic domain of thrombospondin-2 was prevented by gabapentin. We therefore examined whether interaction of thrombospondin with α2δ-1 might reciprocally influence (3)H-gabapentin binding. We concentrated on thrombospondin-4, because, like α2δ-1, it is upregulated in neuropathic pain models. We found that in membranes from cells co-transfected with α2δ-1 and thrombospondin-4, there was a Mg(2+) -dependent reduction in affinity of (3)H-gabapentin binding to α2δ-1. This effect was lost for α2δ-1 with mutations in the von-Willebrand-factor-A domain. However, the effect on (3)H-gabapentin binding was not reproduced by the synaptogenic EGF-domain of thrombospondin-4. Partial co-immunoprecipitation could be demonstrated between thrombospondin-4 and α2δ-1 when co-transfected, but there was no co-immunoprecipitation with thrombospondin-4-EGF domain. Furthermore, we could not detect any association between these two proteins on the cell-surface, indicating the demonstrated interaction occurs intracellularly.


Subject(s)
Amines/metabolism , Analgesics/metabolism , Calcium Channels/metabolism , Cyclohexanecarboxylic Acids/metabolism , Thrombospondins/metabolism , gamma-Aminobutyric Acid/metabolism , Gabapentin , Immunoprecipitation , Protein Binding
10.
Proc Natl Acad Sci U S A ; 111(24): 8979-84, 2014 Jun 17.
Article in English | MEDLINE | ID: mdl-24889613

ABSTRACT

CaV1 and CaV2 voltage-gated calcium channels are associated with ß and α2δ accessory subunits. However, examination of cell surface-associated CaV2 channels has been hampered by the lack of antibodies to cell surface-accessible epitopes and of functional exofacially tagged CaV2 channels. Here we report the development of fully functional CaV2.2 constructs containing inserted surface-accessible exofacial tags, which allow visualization of only those channels at the plasma membrane, in both a neuronal cell line and neurons. We first examined the effect of the auxiliary subunits. Although α2δ subunits copurify with CaV2 channels, it has recently been suggested that this interaction is easily disrupted and nonquantitative. We have now tested whether α2δ subunits are associated with these channels at the cell surface. We found that, whereas α2δ-1 is readily observed at the plasma membrane when expressed alone, it appears absent when coexpressed with CaV2.2/ß1b, despite our finding that α2δ-1 increases plasma-membrane CaV2.2 expression. However, this was due to occlusion of the antigenic epitope by association with CaV2.2, as revealed by antigen retrieval; thus, our data provide evidence for a tight interaction between α2δ-1 and the α1 subunit at the plasma membrane. We further show that, although CaV2.2 cell-surface expression is reduced by gabapentin in the presence of wild-type α2δ-1 (but not a gabapentin-insensitive α2δ-1 mutant), the interaction between CaV2.2 and α2δ-1 is not disrupted by gabapentin. Altogether, these results demonstrate that CaV2.2 and α2δ-1 are intimately associated at the plasma membrane and allow us to infer a region of interaction.


Subject(s)
Calcium Channels, N-Type/metabolism , Calcium Channels/metabolism , Amines/chemistry , Animals , Calcium/chemistry , Calcium Channels, L-Type , Cell Line, Tumor , Cell Membrane/metabolism , Cyclohexanecarboxylic Acids/chemistry , Electrophysiology , Epitopes/chemistry , Gabapentin , Ganglia, Spinal/metabolism , Ligands , Mice , Neuroblastoma/metabolism , Neurons/metabolism , Protein Structure, Tertiary , Rabbits , Rats , Rats, Sprague-Dawley , gamma-Aminobutyric Acid/chemistry
11.
Biochem J ; 458(2): 365-74, 2014 Mar 01.
Article in English | MEDLINE | ID: mdl-24329154

ABSTRACT

It has been shown recently that PrP (prion protein) and the calcium channel auxiliary α2δ subunits interact in neurons and expression systems [Senatore, Colleoni, Verderio, Restelli, Morini, Condliffe, Bertani, Mantovani, Canovi, Micotti, Forloni, Dolphin, Matteoli, Gobbi and Chiesa (2012) Neuron 74, 300-313]. In the present study we examined whether there was an effect of PrP on calcium currents. We have shown that when PrP is co-expressed with calcium channels formed from CaV2.1/ß and α2δ-1 or α2δ-2, there is a consistent decrease in calcium current density. This reduction was absent when a PrP construct was used lacking its GPI (glycosylphosphatidylinositol) anchor. We have reported previously that α2δ subunits are able to form GPI-anchored proteins [Davies, Kadurin, Alvarez-Laviada, Douglas, Nieto-Rostro, Bauer, Pratt and Dolphin (2010) Proc. Natl. Acad. Sci. U.S.A. 107, 1654-1659] and show further evidence in the present paper. We have characterized recently a C-terminally truncated α2δ-1 construct, α2δ-1ΔC, and found that, despite loss of its membrane anchor, it still shows a partial ability to increase calcium currents [Kadurin, Alvarez-Laviada, Ng, Walker-Gray, D'Arco, Fadel, Pratt and Dolphin (2012) J. Biol. Chem. 1287, 33554-33566]. We now find that PrP does not inhibit CaV2.1/ß currents formed with α2δ-1ΔC, rather than α2δ-1. It is possible that PrP and α2δ-1 compete for GPI-anchor intermediates or trafficking pathways, or that interaction between PrP and α2δ-1 requires association in cholesterol-rich membrane microdomains. Our additional finding that CaV2.1/ß1b/α2δ-1 currents were inhibited by GPI-GFP, but not cytosolic GFP, indicates that competition for limited GPI-anchor intermediates or trafficking pathways may be involved in PrP suppression of α2δ subunit function.


Subject(s)
Calcium Channels, N-Type/metabolism , Calcium Channels/metabolism , Gene Expression Regulation , Glycosylphosphatidylinositols/metabolism , Prions/biosynthesis , Animals , Binding, Competitive/genetics , Female , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Protein Binding/physiology , Protein Transport/genetics , Rats , Signal Transduction/genetics , Xenopus
12.
J Biol Chem ; 287(40): 33554-66, 2012 Sep 28.
Article in English | MEDLINE | ID: mdl-22869375

ABSTRACT

The accessory α(2)δ subunits of voltage-gated calcium channels are membrane-anchored proteins, which are highly glycosylated, possess multiple disulfide bonds, and are post-translationally cleaved into α(2) and δ. All α(2)δ subunits have a C-terminal hydrophobic, potentially trans-membrane domain and were described as type I transmembrane proteins, but we found evidence that they can be glycosylphosphatidylinositol-anchored. To probe further the function of membrane anchoring in α(2)δ subunits, we have now examined the properties of α(2)δ-1 constructs truncated at their putative glycosylphosphatidylinositol anchor site, located before the C-terminal hydrophobic domain (α(2)δ-1ΔC-term). We find that the majority of α(2)δ-1ΔC-term is soluble and secreted into the medium, but unexpectedly, some of the protein remains associated with detergent-resistant membranes, also termed lipid rafts, and is extrinsically bound to the plasma membrane. Furthermore, heterologous co-expression of α(2)δ-1ΔC-term with Ca(V)2.1/ß1b results in a substantial enhancement of the calcium channel currents, albeit less than that produced by wild-type α(2)δ-1. These results call into question the role of membrane anchoring of α(2)δ subunits for calcium current enhancement.


Subject(s)
Calcium Channels, N-Type/chemistry , Calcium/metabolism , Animals , Cell Membrane/metabolism , DNA, Complementary/metabolism , Electrophysiology/methods , Ganglia, Spinal/metabolism , Hydrogen-Ion Concentration , Immunohistochemistry/methods , Protein Binding , Protein Structure, Tertiary , Protein Subunits/chemistry , Rabbits , Rats , Rats, Sprague-Dawley , Sequence Analysis, DNA
13.
Nat Neurosci ; 14(4): 478-86, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21358644

ABSTRACT

The hyperpolarization-activated cyclic nucleotide-gated (HCN) channels are subthreshold, voltage-gated ion channels that are highly expressed in hippocampal and cortical pyramidal cell dendrites, where they are important for regulating synaptic potential integration and plasticity. We found that HCN1 subunits are also localized to the active zone of mature asymmetric synaptic terminals targeting mouse entorhinal cortical layer III pyramidal neurons. HCN channels inhibited glutamate synaptic release by suppressing the activity of low-threshold voltage-gated T-type (Ca(V)3.2) Ca²(+) channels. Consistent with this, electron microscopy revealed colocalization of presynaptic HCN1 and Ca(V)3.2 subunit. This represents a previously unknown mechanism by which HCN channels regulate synaptic strength and thereby neural information processing and network excitability.


Subject(s)
Calcium Channels, T-Type/physiology , Cyclic Nucleotide-Gated Cation Channels/physiology , Entorhinal Cortex/physiology , Potassium Channels/physiology , Presynaptic Terminals/physiology , Pyramidal Cells/physiology , Synaptic Transmission/physiology , Animals , Calcium Signaling/genetics , Calcium Signaling/physiology , Entorhinal Cortex/metabolism , Entorhinal Cortex/ultrastructure , Excitatory Amino Acid Antagonists , Glutamates/metabolism , Hyperpolarization-Activated Cyclic Nucleotide-Gated Channels , Mice , Mice, Knockout , Mice, Transgenic , Neural Inhibition/physiology , Organ Culture Techniques , Presynaptic Terminals/metabolism , Presynaptic Terminals/ultrastructure , Protein Subunits/physiology , Pyramidal Cells/metabolism , Pyramidal Cells/ultrastructure , Synaptic Transmission/genetics
14.
Curr Opin Neurobiol ; 20(5): 563-71, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20579869

ABSTRACT

The classical roles of α(2)δ proteins are as accessory calcium channel subunits, enhancing channel trafficking. They were thought to have type-I transmembrane topology, but we find that they can form GPI-anchored proteins. Moreover α(2)δ-1 and α(2)δ-3 have been shown to have novel functions in synaptogenesis, independent of their effect on calcium channels. In neurons, the α(2)δ-1 subunits are present mainly in presynaptic terminals. Peripheral sensory nerve injury results in the up-regulation of α(2)δ-1 in dorsal root ganglion (DRG) neurons, and there is a consequent increase in trafficking of α(2)δ-1 to their terminals. Furthermore, gabapentinoid drugs, which bind to α(2)δ-1 and α(2)δ-2, not only impair their trafficking, but also affect α(2)δ-1-dependent synaptogenesis. These drugs may interfere with α(2)δ function at several different levels.


Subject(s)
Calcium Channels/chemistry , Calcium Channels/physiology , Synaptic Transmission/physiology , Amino Acid Sequence , Animals , Calcium Channels/biosynthesis , Ganglia, Spinal/chemistry , Ganglia, Spinal/cytology , Ganglia, Spinal/pathology , Humans , Molecular Sequence Data , Presynaptic Terminals/chemistry , Presynaptic Terminals/pathology , Presynaptic Terminals/physiology , Protein Transport/physiology , Sensory Receptor Cells/chemistry , Sensory Receptor Cells/pathology , Sensory Receptor Cells/physiology , Up-Regulation/physiology
15.
Proc Natl Acad Sci U S A ; 107(4): 1654-9, 2010 Jan 26.
Article in English | MEDLINE | ID: mdl-20080692

ABSTRACT

Voltage-gated calcium channels are thought to exist in the plasma membrane as heteromeric proteins, in which the alpha1 subunit is associated with two auxiliary subunits, the intracellular beta subunit and the alpha(2)delta subunit; both of these subunits influence the trafficking and properties of Ca(V)1 and Ca(V)2 channels. The alpha(2)delta subunits have been described as type I transmembrane proteins, because they have an N-terminal signal peptide and a C-terminal hydrophobic and potentially transmembrane region. However, because they have very short C-terminal cytoplasmic domains, we hypothesized that the alpha(2)delta proteins might be associated with the plasma membrane through a glycosylphosphatidylinositol (GPI) anchor attached to delta rather than a transmembrane domain. Here, we provide biochemical, immunocytochemical, and mutational evidence to show that all of the alpha(2)delta subunits studied, alpha(2)delta-1, alpha(2)delta-2, and alpha(2)delta-3, show all of the properties expected of GPI-anchored proteins, both when heterologously expressed and in native tissues. They are substrates for prokaryotic phosphatidylinositol-phospholipase C (PI-PLC) and trypanosomal GPI-PLC, which release the alpha(2)delta proteins from membranes and intact cells and expose a cross-reacting determinant epitope. PI-PLC does not affect control transmembrane or membrane-associated proteins. Furthermore, mutation of the predicted GPI-anchor sites markedly reduced plasma membrane and detergent-resistant membrane localization of alpha(2)delta subunits. We also show that GPI anchoring of alpha(2)delta subunits is necessary for their function to enhance calcium currents, and PI-PLC treatment only reduces calcium current density when alpha(2)delta subunits are coexpressed. In conclusion, this study redefines our understanding of alpha(2)delta subunits, both in terms of their role in calcium-channel function and other roles in synaptogenesis.


Subject(s)
Calcium Channels/metabolism , Glycosylphosphatidylinositols/metabolism , Protein Processing, Post-Translational , Amino Acid Sequence , Animals , COS Cells , Calcium Channels/chemistry , Calcium Channels/genetics , Calcium Channels, L-Type , Chlorocebus aethiops , Mice , Molecular Sequence Data , Mutation , Protein Binding , Protein Subunits/chemistry , Protein Subunits/genetics , Protein Subunits/metabolism , Rats
16.
J Neurosci ; 29(13): 4076-88, 2009 Apr 01.
Article in English | MEDLINE | ID: mdl-19339603

ABSTRACT

Neuropathic pain results from damage to the peripheral sensory nervous system, which may have a number of causes. The calcium channel subunit alpha(2)delta-1 is upregulated in dorsal root ganglion (DRG) neurons in several animal models of neuropathic pain, and this is causally related to the onset of allodynia, in which a non-noxious stimulus becomes painful. The therapeutic drugs gabapentin and pregabalin (PGB), which are both alpha(2)delta ligands, have antiallodynic effects, but their mechanism of action has remained elusive. To investigate this, we used an in vivo rat model of neuropathy, unilateral lumbar spinal nerve ligation (SNL), to characterize the distribution of alpha(2)delta-1 in DRG neurons, both at the light- and electron-microscopic level. We found that, on the side of the ligation, alpha(2)delta-1 was increased in the endoplasmic reticulum of DRG somata, in intracellular vesicular structures within their axons, and in the plasma membrane of their presynaptic terminals in superficial layers of the dorsal horn. Chronic PGB treatment of SNL animals, at a dose that alleviated allodynia, markedly reduced the elevation of alpha(2)delta-1 in the spinal cord and ascending axon tracts. In contrast, it had no effect on the upregulation of alpha(2)delta-1 mRNA and protein in DRGs. In vitro, PGB reduced plasma membrane expression of alpha(2)delta-1 without affecting endocytosis. We conclude that the antiallodynic effect of PGB in vivo is associated with impaired anterograde trafficking of alpha(2)delta-1, resulting in its decrease in presynaptic terminals, which would reduce neurotransmitter release and spinal sensitization, an important factor in the maintenance of neuropathic pain.


Subject(s)
Anticonvulsants/therapeutic use , Neuralgia/pathology , Presynaptic Terminals/metabolism , gamma-Aminobutyric Acid/analogs & derivatives , Analysis of Variance , Animals , Behavior, Animal/drug effects , Calcium Channels/metabolism , Calcium Channels, L-Type , Disease Models, Animal , Endocytosis/drug effects , Functional Laterality , Ganglia, Spinal/drug effects , Ganglia, Spinal/metabolism , Ganglia, Spinal/ultrastructure , Male , Microscopy, Electron, Transmission/methods , Neuralgia/drug therapy , Pain Measurement/methods , Pregabalin , Presynaptic Terminals/drug effects , Presynaptic Terminals/ultrastructure , Protein Transport/drug effects , Rats , Rats, Sprague-Dawley , Reaction Time/drug effects , Time Factors , Up-Regulation/drug effects , Up-Regulation/physiology , gamma-Aminobutyric Acid/therapeutic use
17.
Biochem J ; 418(3): 587-94, 2009 Mar 15.
Article in English | MEDLINE | ID: mdl-19032151

ABSTRACT

Stomatin is an integral membrane protein which is widely expressed in many cell types. It is accepted that stomatin has a unique hairpin-loop topology: it is anchored to the membrane with an N-terminal hydrophobic domain and the N- and C-termini are cytoplasmically localized. Stomatin is a prototype for a family of related proteins, containing among others MEC-2 (mechanosensory protein 2) from Caenorhabditis elegans, SLP (stomatin-like protein)-3 and podocin, all of which interact with ion channels to regulate their activity. Members of the stomatin family partly localize in DRMs (detergent-resistant membrane domains) enriched in cholesterol and sphingolipids. It has been proposed that a highly conserved proline residue in the middle of the hydrophobic domain directly binds cholesterol and that cholesterol binding is necessary for the regulation of ion channels. In the present study we show that a small part of the stomatin pool exists as a single-pass transmembrane protein rather than a hairpin-loop protein. The highly conserved proline residue is crucial for adopting the hairpin-loop topology: substitution of this proline residue by serine transfers the whole stomatin pool to the single-pass transmembrane form, which no longer localizes to DRMs. These results suggest that formation of the hairpin loop is inefficient and that the conserved proline residue is indispensable for formation of the hairpin loop. The single-pass transmembrane form exists also for SLP-3 and it should be considered that it mediates part of the physiological functions of stomatin and related proteins.


Subject(s)
Blood Proteins/chemistry , Membrane Proteins/chemistry , Proline/chemistry , Animals , Blood Proteins/genetics , Cell Membrane/chemistry , Cell Membrane/ultrastructure , Cholesterol/chemistry , Female , Glycosylation , Membrane Proteins/genetics , Mice , Mutagenesis, Site-Directed , Nerve Tissue Proteins/chemistry , Oocytes/metabolism , Xenopus laevis
18.
Biochem J ; 412(3): 469-75, 2008 Jun 15.
Article in English | MEDLINE | ID: mdl-18307415

ABSTRACT

ASICs (acid-sensing ion channels) are H(+)-gated Na(+) channels with a widespread expression pattern in the central and the peripheral nervous system. ASICs have a simple topology with two transmembrane domains, cytoplasmic termini and a large ectodomain between the transmembrane domains; this topology has been confirmed by the crystal structure of chicken ASIC1. ASIC1a and ASIC1b are two variants encoded by the asic1 gene. The variable part of the protein includes the cytoplasmic N-terminus, the first transmembrane domain and approximately the first third of the ectodomain. Both variants contain two consensus sequences for N-linked glycosylation in the common, distal part of the ectodomain. In contrast with ASIC1a, ASIC1b contains two additional consensus sequences in the variable, proximal part of the ectodomain. Here we show that all the extracellular asparagine residues within the putative consensus sequences for N-glycosylation carry glycans. The two common distal glycans increase surface expression of the channels, but are no absolute requirement for channel activity. In sharp contrast, the presence of at least one of the two proximal glycans, which are specific to ASIC1b, is an absolute requirement for surface expression of ASIC1b. This result suggests substantial differences in the structure of the proximal ectodomain between the two ASIC1 variants.


Subject(s)
Nerve Tissue Proteins/chemistry , Nerve Tissue Proteins/metabolism , Polysaccharides/metabolism , Sodium Channels/chemistry , Sodium Channels/metabolism , Acid Sensing Ion Channels , Animals , Cell Membrane/metabolism , Electrophysiology , Glycosylation , Membrane Potentials , Models, Molecular , Mutagenesis, Site-Directed , Protein Conformation , Rats , Xenopus laevis
19.
J Biol Chem ; 282(42): 30406-13, 2007 Oct 19.
Article in English | MEDLINE | ID: mdl-17686779

ABSTRACT

There are four genes for acid-sensing ion channels (ASICs) in the genome of mammalian species. Whereas ASIC1 to ASIC3 form functional H+-gated Na+ channels, ASIC4 is not gated by H+, and its function is unknown. Zebrafish has two ASIC4 paralogs: zASIC4.1 and zASIC4.2. Whereas zASIC4.1 is gated by extracellular H+, zASIC4.2 is not. This differential response to H+ makes zASIC4 paralogs a good model to study the properties of this ion channel. In this study, we found that surface expression of homomeric zASIC4.2 is higher than that of zASIC4.1. Surface expression of zASIC4.1 was much increased by formation of heteromeric channels, suggesting that zASIC4.1 contributes to heteromeric ASICs in zebrafish neurons. Robust surface expression of H+-insensitive zASIC4.2 suggests that zASIC4.2 functions as a homomer and is gated by an as yet unknown stimulus, different from H+. Moreover, we identified a small region just distal to the first transmembrane domain that is crucial for the differential H+ response of the two paralogs. This post-TM1 domain may have a general role in gating of members of this gene family.


Subject(s)
Ion Channel Gating/physiology , Membrane Proteins/metabolism , Nerve Tissue Proteins/metabolism , Neurons/metabolism , Protons , Sodium Channels/metabolism , Zebrafish Proteins/metabolism , Zebrafish/metabolism , Acid Sensing Ion Channels , Animals , Dimerization , Female , Gene Expression , Membrane Proteins/genetics , Models, Biological , Multigene Family , Nerve Tissue Proteins/genetics , Protein Structure, Tertiary/physiology , Sodium Channels/genetics , Xenopus laevis , Zebrafish/genetics , Zebrafish Proteins/genetics
20.
Neuropharmacology ; 50(8): 964-74, 2006 Jun.
Article in English | MEDLINE | ID: mdl-16549075

ABSTRACT

Acid-sensing ion channels are excitatory receptors for extracellular H+. Since the extracellular H+ concentration can significantly increase during an inflammation, one of the proposed functions for ASICs is peripheral perception of pain. The ASIC1b and ASIC3 subunits are specifically expressed in sensory ganglia neurons and are candidate sensors of peripheral acidosis. However, the function of these ASIC subunits is limited by their steady-state desensitization during a small but persistent increase of the H+ concentration and by their desensitization after stronger H+ stimuli. Here we show that ASIC1b and ASIC3 form a heteromeric channel that, at steady-state, desensitizes at more acidic values than either homomeric ASIC1b or homomeric ASIC3 alone. Moreover, we show that RFamide neuropeptides, putative modulators of ASIC activity during inflammation, drastically slow down the desensitization of the ASIC1b/3 heteromer with an apparent dissociation constant of approximately 24microM. The apparent affinity for RFamide-induced effects was about 3-fold higher at low extracellular calcium concentrations. Our results suggest that the ASIC1b/3 heteromer is a possible target for RFamide neuropeptides in the peripheral nervous system.


Subject(s)
Calcium/metabolism , Extracellular Space/drug effects , Hydrogen-Ion Concentration , Membrane Proteins/metabolism , Nerve Tissue Proteins/metabolism , Neuropeptides/pharmacology , Sodium Channels/metabolism , Acid Sensing Ion Channels , Animals , Binding Sites/drug effects , Dose-Response Relationship, Drug , Ion Channel Gating/drug effects , Ion Channel Gating/physiology , Ion Channel Gating/radiation effects , Membrane Potentials/drug effects , Membrane Potentials/physiology , Membrane Potentials/radiation effects , Membrane Proteins/chemistry , Nerve Tissue Proteins/chemistry , Oocytes , Patch-Clamp Techniques/methods , Sodium Channels/chemistry , Xenopus laevis
SELECTION OF CITATIONS
SEARCH DETAIL
...