Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters










Database
Language
Publication year range
1.
EMBO Rep ; 20(11): e47967, 2019 11 05.
Article in English | MEDLINE | ID: mdl-31566294

ABSTRACT

Dystroglycan, an extracellular matrix receptor, has essential functions in various tissues. Loss of α-dystroglycan-laminin interaction due to defective glycosylation of α-dystroglycan underlies a group of congenital muscular dystrophies often associated with brain malformations, referred to as dystroglycanopathies. The lack of isogenic human dystroglycanopathy cell models has limited our ability to test potential drugs in a human- and neural-specific context. Here, we generated induced pluripotent stem cells (iPSCs) from a severe dystroglycanopathy patient with homozygous FKRP (fukutin-related protein gene) mutation. We showed that CRISPR/Cas9-mediated gene correction of FKRP restored glycosylation of α-dystroglycan in iPSC-derived cortical neurons, whereas targeted gene mutation of FKRP in wild-type cells disrupted this glycosylation. In parallel, we screened 31,954 small molecule compounds using a mouse myoblast line for increased glycosylation of α-dystroglycan. Using human FKRP-iPSC-derived neural cells for hit validation, we demonstrated that compound 4-(4-bromophenyl)-6-ethylsulfanyl-2-oxo-3,4-dihydro-1H-pyridine-5-carbonitrile (4BPPNit) significantly augmented glycosylation of α-dystroglycan, in part through upregulation of LARGE1 glycosyltransferase gene expression. Together, isogenic human iPSC-derived cells represent a valuable platform for facilitating dystroglycanopathy drug discovery and therapeutic development.


Subject(s)
Drug Evaluation, Preclinical , Dystroglycans/metabolism , Induced Pluripotent Stem Cells/metabolism , Base Sequence , CRISPR-Cas Systems , Cells, Cultured , Drug Evaluation, Preclinical/methods , Dystroglycans/genetics , Gene Editing , Gene Targeting , Genetic Loci , Glycosylation/drug effects , High-Throughput Nucleotide Sequencing , Humans , Molecular Imaging , Muscular Dystrophies/drug therapy , Muscular Dystrophies/etiology , Muscular Dystrophies/metabolism , Mutation , N-Acetylglucosaminyltransferases/genetics , N-Acetylglucosaminyltransferases/metabolism , Neural Stem Cells/metabolism , Neurons/metabolism , Pentosyltransferases/genetics , Pentosyltransferases/metabolism
2.
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
3.
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
4.
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
5.
Pain ; 155(3): 522-533, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24315988

ABSTRACT

The α2δ-1 protein is an auxiliary subunit of voltage-gated calcium channels, critical for neurotransmitter release. It is upregulated in dorsal root ganglion (DRG) neurons following sensory nerve injury, and is also the therapeutic target of the gabapentinoid drugs, which are efficacious in both experimental and human neuropathic pain conditions. α2δ-1 has 3 spliced regions: A, B, and C. A and C are cassette exons, whereas B is introduced via an alternative 3' splice acceptor site. Here we have examined the presence of α2δ-1 splice variants in DRG neurons, and have found that although the main α2δ-1 splice variant in DRG is the same as that in brain (α2δ-1 ΔA+B+C), there is also another α2δ-1 splice variant (ΔA+BΔC), which is expressed in DRG neurons and is differentially upregulated compared to the main DRG splice variant α2δ-1 ΔA+B+C following spinal nerve ligation. Furthermore, this differential upregulation occurs preferentially in a small nonmyelinated DRG neuron fraction, obtained by density gradient separation. The α2δ-1 ΔA+BΔC splice variant supports CaV2 calcium currents with unaltered properties compared to α2δ-1 ΔA+B+C, but shows a significantly reduced affinity for gabapentin. This variant could therefore play a role in determining the efficacy of gabapentin in neuropathic pain.


Subject(s)
Amines/metabolism , Calcium Channels/biosynthesis , Cyclohexanecarboxylic Acids/metabolism , Ganglia, Spinal/metabolism , Peripheral Nerve Injuries/metabolism , Protein Isoforms/biosynthesis , Up-Regulation/physiology , gamma-Aminobutyric Acid/metabolism , Animals , Calcium Channels/chemistry , Calcium Channels/genetics , Calcium Channels, L-Type , Gabapentin , Male , Peripheral Nerve Injuries/genetics , Protein Binding/physiology , Protein Isoforms/chemistry , Protein Isoforms/genetics , Rabbits , Rats , Rats, Sprague-Dawley
6.
Nature ; 486(7401): 122-5, 2012 May 13.
Article in English | MEDLINE | ID: mdl-22678293

ABSTRACT

Synaptic neurotransmitter release is driven by Ca(2+) influx through active zone voltage-gated calcium channels (VGCCs). Control of active zone VGCC abundance and function remains poorly understood. Here we show that a trafficking step probably sets synaptic VGCC levels in rats, because overexpression of the pore-forming α1(A) VGCC subunit fails to change synaptic VGCC abundance or function. α2δs are a family of glycosylphosphatidylinositol (GPI)-anchored VGCC-associated subunits that, in addition to being the target of the potent neuropathic analgesics gabapentin and pregabalin (α2δ-1 and α2δ-2), were also identified in a forward genetic screen for pain genes (α2δ-3). We show that these proteins confer powerful modulation of presynaptic function through two distinct molecular mechanisms. First, α2δ subunits set synaptic VGCC abundance, as predicted from their chaperone-like function when expressed in non-neuronal cells. Second, α2δs configure synaptic VGCCs to drive exocytosis through an extracellular metal ion-dependent adhesion site (MIDAS), a conserved set of amino acids within the predicted von Willebrand A domain of α2δ. Expression of α2δ with an intact MIDAS motif leads to an 80% increase in release probability, while simultaneously protecting exocytosis from blockade by an intracellular Ca(2+) chelator. α2δs harbouring MIDAS site mutations still drive synaptic accumulation of VGCCs; however, they no longer change release probability or sensitivity to intracellular Ca(2+) chelators. Our data reveal dual functionality of these clinically important VGCC subunits, allowing synapses to make more efficient use of Ca(2+) entry to drive neurotransmitter release.


Subject(s)
Calcium Channels/genetics , Calcium Channels/metabolism , Exocytosis , Neurotransmitter Agents/metabolism , Presynaptic Terminals/metabolism , Action Potentials , Animals , Calcium Channels/biosynthesis , Calcium Channels, L-Type , Calcium Signaling , Mice , Probability , Rats
SELECTION OF CITATIONS
SEARCH DETAIL
...