Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 20
Filter
1.
Acta Neuropathol ; 144(3): 437-464, 2022 09.
Article in English | MEDLINE | ID: mdl-35876881

ABSTRACT

Dysfunction and degeneration of synapses is a common feature of amyotrophic lateral sclerosis and frontotemporal dementia (ALS/FTD). A GGGGCC hexanucleotide repeat expansion in the C9ORF72 gene is the main genetic cause of ALS/FTD (C9ALS/FTD). The repeat expansion leads to reduced expression of the C9orf72 protein. How C9orf72 haploinsufficiency contributes to disease has not been resolved. Here we identify the synapsin family of synaptic vesicle proteins, the most abundant group of synaptic phosphoproteins, as novel interactors of C9orf72 at synapses and show that C9orf72 plays a cell-autonomous role in the regulation of excitatory synapses. We mapped the interaction of C9orf72 and synapsin to the N-terminal longin domain of C9orf72 and the conserved C domain of synapsin, and show interaction of the endogenous proteins in synapses. Functionally, C9orf72 deficiency reduced the number of excitatory synapses and decreased synapsin levels at remaining synapses in vitro in hippocampal neuron cultures and in vivo in the hippocampal mossy fibre system of C9orf72 knockout mice. Consistent with synaptic dysfunction, electrophysiological recordings identified impaired excitatory neurotransmission and network function in hippocampal neuron cultures with reduced C9orf72 expression, which correlated with a severe depletion of synaptic vesicles from excitatory synapses in the hippocampus of C9orf72 knockout mice. Finally, neuropathological analysis of post-mortem sections of C9ALS/FTD patient hippocampus with C9orf72 haploinsufficiency revealed a marked reduction in synapsin, indicating that disruption of the interaction between C9orf72 and synapsin may contribute to ALS/FTD pathobiology. Thus, our data show that C9orf72 plays a cell-autonomous role in the regulation of neurotransmission at excitatory synapses by interaction with synapsin and modulation of synaptic vesicle pools, and identify a novel role for C9orf72 haploinsufficiency in synaptic dysfunction in C9ALS/FTD.


Subject(s)
Amyotrophic Lateral Sclerosis , C9orf72 Protein/metabolism , Frontotemporal Dementia , Synapsins/metabolism , Amyotrophic Lateral Sclerosis/metabolism , Amyotrophic Lateral Sclerosis/pathology , Animals , C9orf72 Protein/genetics , DNA Repeat Expansion , Frontotemporal Dementia/metabolism , Frontotemporal Dementia/pathology , Mice , Mice, Knockout , Synapses/pathology
2.
EMBO J ; 35(15): 1656-76, 2016 08 01.
Article in English | MEDLINE | ID: mdl-27334615

ABSTRACT

A GGGGCC hexanucleotide repeat expansion in the C9orf72 gene is the most common genetic cause of amyotrophic lateral sclerosis and frontotemporal dementia (C9ALS/FTD). C9orf72 encodes two C9orf72 protein isoforms of unclear function. Reduced levels of C9orf72 expression have been reported in C9ALS/FTD patients, and although C9orf72 haploinsufficiency has been proposed to contribute to C9ALS/FTD, its significance is not yet clear. Here, we report that C9orf72 interacts with Rab1a and the Unc-51-like kinase 1 (ULK1) autophagy initiation complex. As a Rab1a effector, C9orf72 controls initiation of autophagy by regulating the Rab1a-dependent trafficking of the ULK1 autophagy initiation complex to the phagophore. Accordingly, reduction of C9orf72 expression in cell lines and primary neurons attenuated autophagy and caused accumulation of p62-positive puncta reminiscent of the p62 pathology observed in C9ALS/FTD patients. Finally, basal levels of autophagy were markedly reduced in C9ALS/FTD patient-derived iNeurons. Thus, our data identify C9orf72 as a novel Rab1a effector in the regulation of autophagy and indicate that C9orf72 haploinsufficiency and associated reductions in autophagy might be the underlying cause of C9ALS/FTD-associated p62 pathology.


Subject(s)
Autophagy-Related Protein-1 Homolog/metabolism , Autophagy , Cell Physiological Phenomena , Intracellular Signaling Peptides and Proteins/metabolism , Proteins/metabolism , rab1 GTP-Binding Proteins/metabolism , C9orf72 Protein , Cells, Cultured , Frontotemporal Dementia/pathology , Humans , Neurons/chemistry , Neurons/metabolism
3.
Brain ; 142(3): 586-605, 2019 03 01.
Article in English | MEDLINE | ID: mdl-30698736

ABSTRACT

As clinical evidence supports a negative impact of dysfunctional energy metabolism on the disease progression in amyotrophic lateral sclerosis, it is vital to understand how the energy metabolic pathways are altered and whether they can be restored to slow disease progression. Possible approaches include increasing or rerouting catabolism of alternative fuel sources to supplement the glycolytic and mitochondrial pathways such as glycogen, ketone bodies and nucleosides. To analyse the basis of the catabolic defect in amyotrophic lateral sclerosis we used a novel phenotypic metabolic array. We profiled fibroblasts and induced neuronal progenitor-derived human induced astrocytes from C9orf72 amyotrophic lateral sclerosis patients compared to normal controls, measuring the rates of production of reduced nicotinamide adenine dinucleotides from 91 potential energy substrates. This approach shows for the first time that C9orf72 human induced astrocytes and fibroblasts have an adenosine to inosine deamination defect caused by reduction of adenosine deaminase, which is also observed in induced astrocytes from sporadic patients. Patient-derived induced astrocyte lines were more susceptible to adenosine-induced toxicity, which could be mimicked by inhibiting adenosine deaminase in control lines. Furthermore, adenosine deaminase inhibition in control induced astrocytes led to increased motor neuron toxicity in co-cultures, similar to the levels observed with patient derived induced astrocytes. Bypassing metabolically the adenosine deaminase defect by inosine supplementation was beneficial bioenergetically in vitro, increasing glycolytic energy output and leading to an increase in motor neuron survival in co-cultures with induced astrocytes. Inosine supplementation, in combination with modulation of the level of adenosine deaminase may represent a beneficial therapeutic approach to evaluate in patients with amyotrophic lateral sclerosis.


Subject(s)
Adenosine Deaminase/metabolism , Amyotrophic Lateral Sclerosis/metabolism , Motor Neurons/metabolism , Adenosine Deaminase/physiology , Adult , Amyotrophic Lateral Sclerosis/physiopathology , Animals , Astrocytes/metabolism , C9orf72 Protein/metabolism , Cell Death , Cell Survival , Cells, Cultured , Coculture Techniques , Disease Progression , Energy Metabolism/physiology , Female , Fibroblasts/metabolism , Humans , Inosine/metabolism , Male , Mice , Mice, Inbred C57BL , Middle Aged , Rats , Rats, Sprague-Dawley , Stem Cells/metabolism
4.
Hum Mol Genet ; 26(23): 4668-4679, 2017 12 01.
Article in English | MEDLINE | ID: mdl-28973175

ABSTRACT

Defective axonal transport is an early neuropathological feature of amyotrophic lateral sclerosis (ALS). We have previously shown that ALS-associated mutations in Cu/Zn superoxide dismutase 1 (SOD1) impair axonal transport of mitochondria in motor neurons isolated from SOD1 G93A transgenic mice and in ALS mutant SOD1 transfected cortical neurons, but the underlying mechanisms remained unresolved. The outer mitochondrial membrane protein mitochondrial Rho GTPase 1 (Miro1) is a master regulator of mitochondrial axonal transport in response to cytosolic calcium (Ca2+) levels ([Ca2+]c) and mitochondrial damage. Ca2+ binding to Miro1 halts mitochondrial transport by modifying its interaction with kinesin-1 whereas mitochondrial damage induces Phosphatase and Tensin Homolog (PTEN)-induced Putative Kinase 1 (PINK1) and Parkin-dependent degradation of Miro1 and consequently stops transport. To identify the mechanism underlying impaired axonal transport of mitochondria in mutant SOD1-related ALS we investigated [Ca2+]c and Miro1 levels in ALS mutant SOD1 expressing neurons. We found that expression of ALS mutant SOD1 reduced the level of endogenous Miro1 but did not affect [Ca2+]c. ALS mutant SOD1 induced reductions in Miro1 levels were Parkin dependent. Moreover, both overexpression of Miro1 and ablation of PINK1 rescued the mitochondrial axonal transport deficit in ALS mutant SOD1-expressing cortical and motor neurons. Together these results provide evidence that ALS mutant SOD1 inhibits axonal transport of mitochondria by inducing PINK1/Parkin-dependent Miro1 degradation.


Subject(s)
Amyotrophic Lateral Sclerosis/metabolism , Axonal Transport/physiology , Mitochondrial Proteins/metabolism , Superoxide Dismutase-1/genetics , Superoxide Dismutase-1/metabolism , rho GTP-Binding Proteins/metabolism , Amyotrophic Lateral Sclerosis/enzymology , Amyotrophic Lateral Sclerosis/genetics , Animals , Axons/metabolism , Calcium/metabolism , Cytosol/metabolism , Disease Models, Animal , HEK293 Cells , HeLa Cells , Humans , Mice, Transgenic , Mitochondria/metabolism , Motor Neurons/metabolism , Mutation , Protein Kinases/metabolism , Rats
5.
J Neurosci ; 33(42): 16412-26, 2013 Oct 16.
Article in English | MEDLINE | ID: mdl-24133248

ABSTRACT

The α2δ-1 subunit of voltage-gated calcium channels is upregulated after sensory nerve injury and is also the therapeutic target of gabapentinoid drugs. It is therefore likely to play a key role in the development of neuropathic pain. In this study, we have examined mice in which α2δ-1 gene expression is disrupted, to determine whether α2δ-1 is involved in various modalities of nociception, and for the development of behavioral hypersensitivity after partial sciatic nerve ligation (PSNL). We find that naive α2δ-1(-/-) mice show a marked behavioral deficit in mechanical and cold sensitivity, but no change in thermal nociception threshold. The lower mechanical sensitivity is mirrored by a reduced in vivo electrophysiological response of dorsal horn wide dynamic range neurons. The CaV2.2 level is reduced in brain and spinal cord synaptosomes from α2δ-1(-/-) mice, and α2δ-1(-/-) DRG neurons exhibit lower calcium channel current density. Furthermore, a significantly smaller number of DRG neurons respond to the TRPM8 agonist menthol. After PSNL, α2δ-1(-/-) mice show delayed mechanical hypersensitivity, which only develops at 11 d after surgery, whereas in wild-type littermates it is maximal at the earliest time point measured (3 d). There is no compensatory upregulation of α2δ-2 or α2δ-3 after PSNL in α2δ-1(-/-) mice, and other transcripts, including neuropeptide Y and activating transcription factor-3, are upregulated normally. Furthermore, the ability of pregabalin to alleviate mechanical hypersensitivity is lost in PSNL α2δ-1(-/-) mice. Thus, α2δ-1 is essential for rapid development of mechanical hypersensitivity in a nerve injury model of neuropathic pain.


Subject(s)
Calcium Channels/genetics , Hyperalgesia/genetics , Neuralgia/genetics , Peripheral Nerve Injuries/genetics , Sensory Receptor Cells/physiology , Animals , Calcium Channels/metabolism , Cold Temperature , Ganglia, Spinal/metabolism , Ganglia, Spinal/physiopathology , Hyperalgesia/metabolism , Hyperalgesia/physiopathology , Mice , Mice, Knockout , Neuralgia/metabolism , Neuralgia/physiopathology , Pain Threshold/physiology , Peripheral Nerve Injuries/metabolism , Peripheral Nerve Injuries/physiopathology , Physical Stimulation , Sciatic Nerve/injuries , Sciatic Nerve/metabolism , Sciatic Nerve/physiopathology
6.
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
7.
Sci Transl Med ; 15(685): eabo3823, 2023 03.
Article in English | MEDLINE | ID: mdl-36857431

ABSTRACT

Hexanucleotide repeat expansions in C9ORF72 are the most common genetic cause of familial amyotrophic lateral sclerosis (ALS) and frontotemporal dementia (FTD). Studies have shown that the hexanucleotide expansions cause the noncanonical translation of C9ORF72 transcripts into neurotoxic dipeptide repeat proteins (DPRs) that contribute to neurodegeneration. We show that a cell-penetrant peptide blocked the nuclear export of C9ORF72-repeat transcripts in HEK293T cells by competing with the interaction between SR-rich splicing factor 1 (SRSF1) and nuclear export factor 1 (NXF1). The cell-penetrant peptide also blocked the translation of toxic DPRs in neurons differentiated from induced neural progenitor cells (iNPCs), which were derived from individuals carrying C9ORF72-linked ALS mutations. This peptide also increased survival of iNPC-differentiated C9ORF72-ALS motor neurons cocultured with astrocytes. Oral administration of the cell-penetrant peptide reduced DPR translation and rescued locomotor deficits in a Drosophila model of mutant C9ORF72-mediated ALS/FTD. Intrathecal injection of this peptide into the brains of ALS/FTD mice carrying a C9ORF72 mutation resulted in reduced expression of DPRs in mouse brains. These findings demonstrate that disrupting the production of DPRs in cellular and animal models of ALS/FTD might be a strategy to ameliorate neurodegeneration in these diseases.


Subject(s)
Amyotrophic Lateral Sclerosis , Frontotemporal Dementia , Humans , Animals , Mice , Dipeptides , C9orf72 Protein , Active Transport, Cell Nucleus , HEK293 Cells , Peptides , Motor Neurons , RNA , Serine-Arginine Splicing Factors
8.
Front Cell Neurosci ; 16: 1061559, 2022.
Article in English | MEDLINE | ID: mdl-36619668

ABSTRACT

Disruption to protein homeostasis caused by lysosomal dysfunction and associated impairment of autophagy is a prominent pathology in amyotrophic lateral sclerosis and frontotemporal dementia (ALS/FTD). The most common genetic cause of ALS/FTD is a G4C2 hexanucleotide repeat expansion in C9orf72 (C9ALS/FTD). Repeat-associated non-AUG (RAN) translation of G4C2 repeat transcripts gives rise to dipeptide repeat (DPR) proteins that have been shown to be toxic and may contribute to disease etiology. Genetic variants in TMEM106B have been associated with frontotemporal lobar degeneration with TDP-43 pathology and disease progression in C9ALS/FTD. TMEM106B encodes a lysosomal transmembrane protein of unknown function that is involved in various aspects of lysosomal biology. How TMEM106B variants affect C9ALS/FTD is not well understood but has been linked to changes in TMEM106B protein levels. Here, we investigated TMEM106B function in the context of C9ALS/FTD DPR pathology. We report that knockdown of TMEM106B expression exacerbates the accumulation of C9ALS/FTD-associated cytotoxic DPR proteins in cell models expressing RAN-translated or AUG-driven DPRs as well as in C9ALS/FTD-derived iAstrocytes with an endogenous G4C2 expansion by impairing autophagy. Loss of TMEM106B caused a block late in autophagy by disrupting autophagosome to autolysosome maturation which coincided with impaired lysosomal acidification, reduced cathepsin activity, and juxtanuclear clustering of lysosomes. Lysosomal clustering required Rab7A and coincided with reduced Arl8b-mediated anterograde transport of lysosomes to the cell periphery. Increasing Arl8b activity in TMEM106B-deficient cells not only restored the distribution of lysosomes, but also fully rescued autophagy and DPR protein accumulation. Thus, we identified a novel function of TMEM106B in autophagosome maturation via Arl8b. Our findings indicate that TMEM106B variants may modify C9ALS/FTD by regulating autophagic clearance of DPR proteins. Caution should therefore be taken when considering modifying TMEM106B expression levels as a therapeutic approach in ALS/FTD.

9.
Anesthesiology ; 115(1): 144-52, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21602662

ABSTRACT

BACKGROUND: Opioid-induced hyperalgesia is recognized in the laboratory and the clinic, generating central hyperexcitability in the absence of peripheral pathology. We investigated pregabalin, indicated for neuropathic pain, and ondansetron, a drug that disrupts descending serotonergic processing in the central nervous system, on spinal neuronal hyperexcitability and visceral hypersensitivity in a rat model of opioid-induced hyperalgesia. METHODS: Male Sprague-Dawley rats (180-200 g) were implanted with osmotic mini-pumps filled with morphine (90 µg · µl⁻¹ · h⁻¹) or saline (0.9% w/v). On days 7-10 in isoflurane anesthetized animals, we evaluated the effects of (1) systemic pregabalin on spinal neuronal and visceromotor responses, and (2) spinal ondansetron on dorsal horn neuronal response. Messenger ribonucleic acid concentrations of α2δ-1, 5HT3A, and µ-opioid receptor in the dorsal root ganglia of all animals were analyzed. RESULTS: In morphine-treated animals, evoked spinal neuronal responses were enhanced to a subset of thermal and mechanical stimuli. This activity was attenuated by pregabalin (by at least 71%) and ondansetron (37%); the visceromotor response to a subset of colorectal distension pressures was attenuated by pregabalin (52.8%; n = 8 for all measures, P < 0.05). Messenger ribonucleic acid concentrations were unchanged. CONCLUSIONS: The inhibitory action of pregabalin in opioid-induced hyperalgesia animals is neither neuropathy-dependent nor reliant on up-regulation of the α2δ-1 subunit of voltage-gated calcium channels-mechanisms proposed as being essential for pregabalin's efficacy in neuropathy. In opioid-induced hyperalgesia, which extends to colonic distension, a serotonergic facilitatory system may be up-regulated, creating an environment that is permissive for pregabalin-mediated analgesia without peripheral pathology.


Subject(s)
Analgesics/pharmacology , Hyperalgesia/drug therapy , Neurons/drug effects , Spinal Cord/cytology , Spinal Cord/drug effects , gamma-Aminobutyric Acid/analogs & derivatives , Analgesics/therapeutic use , Analgesics, Opioid , Animals , Dilatation , Electrophysiology , Hyperalgesia/chemically induced , Male , Morphine , Ondansetron/pharmacology , Pain/drug therapy , Pain Measurement , Pregabalin , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Rats , Rats, Sprague-Dawley , Receptors, Opioid, mu/drug effects , Receptors, Opioid, mu/metabolism , Receptors, Serotonin, 5-HT3/drug effects , Receptors, Serotonin, 5-HT3/metabolism , Rectum/physiology , Serotonin Antagonists/pharmacology , gamma-Aminobutyric Acid/pharmacology , gamma-Aminobutyric Acid/therapeutic use
10.
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
11.
Biochem Soc Trans ; 38(2): 525-8, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20298215

ABSTRACT

Neuropathic pain is caused by lesion or dysfunction of the peripheral sensory nervous system. Up-regulation of the voltage-gated Ca(2+) channel subunit alpha(2)delta-1 in DRG (dorsal root ganglion) neurons and the spinal cord correlates with the onset of neuropathic pain symptoms such as allodynia in several animal models of neuropathic pain. The clinically important anti-allodynic drugs gabapentin and pregabalin are alpha(2)delta-1 ligands, but how these drugs alleviate neuropathic pain is poorly understood. In the present paper, we review recent advances in our understanding of their molecular mechanisms.


Subject(s)
Calcium Channels/metabolism , Presynaptic Terminals/drug effects , Presynaptic Terminals/metabolism , gamma-Aminobutyric Acid/analogs & derivatives , Amines/pharmacology , Amines/therapeutic use , Analgesics/pharmacology , Analgesics/therapeutic use , Animals , Calcium Channels/physiology , Cyclohexanecarboxylic Acids/pharmacology , Cyclohexanecarboxylic Acids/therapeutic use , Gabapentin , Humans , Ligands , Models, Biological , Neuralgia/drug therapy , Pregabalin , Protein Transport/drug effects , gamma-Aminobutyric Acid/metabolism , gamma-Aminobutyric Acid/pharmacology , gamma-Aminobutyric Acid/therapeutic use
12.
Mol Pain ; 5: 45, 2009 Aug 07.
Article in English | MEDLINE | ID: mdl-19664204

ABSTRACT

BACKGROUND: Descending facilitation, from the brainstem, promotes spinal neuronal hyperexcitability and behavioural hypersensitivity in many chronic pain states. We have previously demonstrated enhanced descending facilitation onto dorsal horn neurones in a neuropathic pain model, and shown this to enable the analgesic effectiveness of gabapentin. Here we have tested if this hypothesis applies to other pain states by using a combination of approaches in a rat model of osteoarthritis (OA) to ascertain if 1) a role for descending 5HT mediated facilitation exists, and 2) if pregabalin (a newer analogue of gabapentin) is an effective antinociceptive agent in this model. Further, quantitative-PCR experiments were undertaken to analyse the alpha 2 delta-1 and 5-HT3A subunit mRNA levels in L3-6 DRG in order to assess whether changes in these molecular substrates have a bearing on the pharmacological effects of ondansetron and pregabalin in OA. RESULTS: Osteoarthritis was induced via intra-articular injection of monosodium iodoacetate (MIA) into the knee joint. Control animals were injected with 0.9% saline. Two weeks later in vivo electrophysiology was performed, comparing the effects of spinal ondansetron (10-100 microg/50 microl) or systemic pregabalin (0.3 - 10 mg/kg) on evoked responses of dorsal horn neurones to electrical, mechanical and thermal stimuli in MIA or control rats. In MIA rats, ondansetron significantly inhibited the evoked responses to both innocuous and noxious natural evoked neuronal responses, whereas only inhibition of noxious evoked responses was seen in controls. Pregabalin significantly inhibited neuronal responses in the MIA rats only; this effect was blocked by a pre-administration of spinal ondansetron. Analysis of alpha 2 delta-1 and 5-HT3A subunit mRNA levels in L3-6 DRG revealed a significant increase in alpha 2 delta-1 levels in ipsilateral L3&4 DRG in MIA rats. 5-HT3A subunit mRNA levels were unchanged. CONCLUSION: These data suggest descending serotonergic facilitation plays a role in mediating the brush and innocuous mechanical punctate evoked neuronal responses in MIA rats, suggesting an adaptive change in the excitatory serotonergic drive modulating low threshold evoked neuronal responses in MIA-induced OA pain. This alteration in excitatory serotonergic drive, alongside an increase in alpha 2 delta-1 mRNA levels, may underlie pregabalin's state dependent effects in this model of chronic pain.


Subject(s)
Analgesics/therapeutic use , Osteoarthritis/drug therapy , Osteoarthritis/metabolism , gamma-Aminobutyric Acid/analogs & derivatives , Analgesics/pharmacology , Animals , Behavior, Animal/drug effects , Calcium Channels/genetics , Calcium Channels/physiology , Calcium Channels, L-Type , Disease Models, Animal , Electrophysiology , Osteoarthritis/pathology , Polymerase Chain Reaction , Pregabalin , Rats , Rats, Sprague-Dawley , Serotonin/genetics , Serotonin/physiology , gamma-Aminobutyric Acid/pharmacology , gamma-Aminobutyric Acid/therapeutic use
13.
J Neurosci ; 27(1): 212-9, 2007 Jan 03.
Article in English | MEDLINE | ID: mdl-17202488

ABSTRACT

The vesicle priming protein Munc13-1 is regulated by diacylglycerol (DAG) and is therefore hypothesized to play a role in the control of neurotransmitter release by phospholipase C (PLC)-coupled receptors. We combined voltage-clamp recordings of voltage-gated Ca2+ channels (VGCCs) and high-resolution capacitance measurements to investigate the mechanism of receptor-mediated modulation of exocytosis in bovine chromaffin cells. Activation of endogenous H1 G(q)-protein-coupled receptors (G(q)PCRs) by histamine potentiated stimulus-coupled secretion despite concurrently inhibiting Ca2+ influx through VGCCs. Histamine increased the size of the readily releasable pool of vesicles and in particular a subpool of fusion-competent vesicles localized in close proximity to VGCCs. Pharmacological characterization showed that potentiation of exocytosis depended on the activation of PLC but not protein kinase C. Overexpression of wild-type Munc13-1 by adenoviral infection had no effect on histamine-induced potentiation per se, whereas DAG-insensitive Munc13-1(H567K) completely abolished it. This is the first endogenous mammalian G(q)PCR signaling pathway identified that engages Munc13-1 to increase stimulus-coupled secretion by recruiting vesicles to the immediately releasable pool. G(q)PCRs are therefore able to control exocytosis at the level of SNARE (soluble N-ethylmaleimide-sensitive factor attachment protein receptor) complex formation to produce rapid, short-term potentiation of the secretory output of neurons and endocrine cells.


Subject(s)
Chromaffin Cells/physiology , Exocytosis/physiology , Long-Term Potentiation/physiology , Nerve Tissue Proteins/metabolism , Receptors, G-Protein-Coupled/metabolism , Type C Phospholipases/metabolism , Animals , Calcium Channels/physiology , Cattle , Cells, Cultured
14.
J Neurosci ; 27(19): 5236-48, 2007 May 09.
Article in English | MEDLINE | ID: mdl-17494710

ABSTRACT

P/Q-type (Ca(V)2.1) and N-type (Ca(V)2.2) Ca2+ channels are critical to stimulus-secretion coupling in the nervous system; feedback regulation of these channels by Ca2+ is therefore predicted to profoundly influence neurotransmission. Here we report divergent regulation of Ca2+-dependent inactivation (CDI) of native N- and P/Q-type Ca2+ channels by calmodulin (CaM) in adult chromaffin cells. Robust CDI of N-type channels was observed in response to prolonged step depolarizations, as well as repetitive stimulation with either brief step depolarizations or action potential-like voltage stimuli. Adenoviral expression of Ca2+-insensitive calmodulin mutants eliminated CDI of N-type channels. This is the first demonstration of CaM-dependent CDI of a native N-type channel. CDI of P/Q-type channels was by comparison modest and insensitive to expression of CaM mutants. Cloning of the C terminus of the Ca(V)2.1 alpha1 subunit from chromaffin cells revealed multiple splice variants lacking structural motifs required for CaM-dependent CDI. The physiological relevance of CDI on stimulus-coupled exocytosis was revealed by combining perforated-patch voltage-clamp recordings of pharmacologically isolated Ca2+ currents with membrane capacitance measurements of exocytosis. Increasing stimulus intensity to invoke CDI resulted in a significant decrease in the exocytotic efficiency of N-type channels compared with P/Q-type channels. Our results reveal unexpected diversity in CaM regulation of native Ca(V)2 channels and suggest that the ability of individual Ca2+ channel subtypes to undergo CDI may be tailored by alternative splicing to meet the specific requirements of a particular cellular function.


Subject(s)
Calcium Channels, N-Type/metabolism , Calcium Channels, P-Type/metabolism , Calcium Signaling/physiology , Calmodulin/metabolism , Chromaffin Cells/metabolism , Exocytosis/physiology , Animals , Calcium/metabolism , Calcium/pharmacology , Calcium Channels, N-Type/chemistry , Calcium Channels, N-Type/genetics , Calcium Channels, P-Type/chemistry , Calcium Channels, P-Type/drug effects , Calcium Signaling/drug effects , Cattle , Cell Line , Cell Membrane/chemistry , Cell Membrane/drug effects , Cell Membrane/metabolism , Cells, Cultured , Chromaffin Cells/drug effects , Electric Capacitance , Exocytosis/drug effects , Green Fluorescent Proteins/genetics , Humans , Ion Channel Gating/drug effects , Ion Channel Gating/genetics , Membrane Potentials/drug effects , Membrane Potentials/physiology , Patch-Clamp Techniques , Protein Structure, Tertiary/genetics
15.
Nat Commun ; 8: 16063, 2017 07 05.
Article in English | MEDLINE | ID: mdl-28677678

ABSTRACT

Hexanucleotide repeat expansions in the C9ORF72 gene are the commonest known genetic cause of amyotrophic lateral sclerosis and frontotemporal dementia. Expression of repeat transcripts and dipeptide repeat proteins trigger multiple mechanisms of neurotoxicity. How repeat transcripts get exported from the nucleus is unknown. Here, we show that depletion of the nuclear export adaptor SRSF1 prevents neurodegeneration and locomotor deficits in a Drosophila model of C9ORF72-related disease. This intervention suppresses cell death of patient-derived motor neuron and astrocytic-mediated neurotoxicity in co-culture assays. We further demonstrate that either depleting SRSF1 or preventing its interaction with NXF1 specifically inhibits the nuclear export of pathological C9ORF72 transcripts, the production of dipeptide-repeat proteins and alleviates neurotoxicity in Drosophila, patient-derived neurons and neuronal cell models. Taken together, we show that repeat RNA-sequestration of SRSF1 triggers the NXF1-dependent nuclear export of C9ORF72 transcripts retaining expanded hexanucleotide repeats and reveal a novel promising therapeutic target for neuroprotection.


Subject(s)
Amyotrophic Lateral Sclerosis/metabolism , C9orf72 Protein/metabolism , Frontotemporal Dementia/metabolism , Nucleocytoplasmic Transport Proteins/metabolism , RNA-Binding Proteins/metabolism , Serine-Arginine Splicing Factors/metabolism , Adult , Aged , Amyotrophic Lateral Sclerosis/etiology , Animals , Astrocytes/physiology , Cell Line , Coculture Techniques , Disease Models, Animal , Drosophila , Female , Frontotemporal Dementia/etiology , Humans , Male , Mice , Middle Aged , Nuclear Proteins/metabolism , Rats , Transcription Factors/metabolism
16.
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
17.
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
18.
Channels (Austin) ; 6(2): 124-32, 2012.
Article in English | MEDLINE | ID: mdl-22627148

ABSTRACT

In this study, we have examined the properties of synaptic transmission between dorsal root ganglion (DRG) and dorsal horn (DH) neurons, placed in co-culture. We also examined the effect of the anti-hyperalgesic gabapentinoid drug pregabalin (PGB) at this pharmacologically relevant synapse. The main method used was electrophysiological recording of excitatory post synaptic currents (EPSCs) in DH neurons. Synaptic transmission between DRG and DH neurons was stimulated by capsaicin, which activates transient receptor potential vanilloid-1 (TRPV1) receptors on small diameter DRG neurons. Capsaicin (1 µM) application increased the frequency of EPSCs recorded in DH neurons in DRG-DH co-cultures, by about 3-fold, but had no effect on other measured properties of the EPSCs. There was also no effect of capsaicin in the absence of co-cultured DRGs. Application of PGB (100 µM) for 40-48 h caused a reduction in the capsaicin-induced increase in EPSC frequency by 57%. In contrast, brief preincubation of PGB had no significant effect on the capsaicin-induced increase in EPSC frequency. In conclusion, this study shows that PGB applied for 40-48 h, but not acute application inhibits excitatory synaptic transmission at DRG-DH synapses, in response to nociceptive stimulation, most likely by a presynaptic effect on neurotransmitter release from DRG presynaptic terminals.


Subject(s)
Analgesics/pharmacology , Ganglia, Spinal/physiology , Posterior Horn Cells/physiology , Synaptic Potentials/drug effects , Synaptic Transmission/drug effects , gamma-Aminobutyric Acid/analogs & derivatives , Animals , Calcium/metabolism , Capsaicin/pharmacology , Coculture Techniques , Ganglia, Spinal/cytology , Microscopy, Fluorescence , Patch-Clamp Techniques , Posterior Horn Cells/cytology , Pregabalin , Rats , Rats, Sprague-Dawley , Sensory System Agents/pharmacology , TRPV Cation Channels/agonists , gamma-Aminobutyric Acid/pharmacology
19.
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
20.
Nature ; 419(6910): 947-52, 2002 Oct 31.
Article in English | MEDLINE | ID: mdl-12410316

ABSTRACT

Voltage-gated calcium channels (VGCCs) conduct calcium into cells after membrane depolarization and are vital for diverse biological events. They are regulated by various signalling pathways, which has profound functional consequences. The activity of VGCCs decreases with time in whole-cell and inside-out patch-clamp recordings. This rundown reflects persistent intrinsic modulation of VGCCs in intact cells. Although several mechanisms have been reported to contribute to rundown of L-type channels, the mechanism of rundown of other types of VGCC is poorly understood. Here we show that phosphatidylinositol-4,5-bisphosphate (PtdIns(4,5)P2), an essential regulator of ion channels and transporters, is crucial for maintaining the activity of P/Q- and N-type channels. Activation of membrane receptors that stimulate hydrolysis of PtdIns(4,5)P2 causes channel inhibition in oocytes and neurons. PtdIns(4,5)P2 also inhibits P/Q-type channels by altering the voltage dependence of channel activation and making the channels more difficult to open. This inhibition is alleviated by phosphorylation by protein kinase A. The dual actions of PtdIns(4,5)P2 and the crosstalk between PtdIns(4,5)P2 and protein kinase A set up a dynamic mechanism through which the activity of VGCCs can be finely tuned by various neurotransmitters, hormones and trophic factors.


Subject(s)
Calcium Channels/metabolism , Ion Channel Gating , Phosphatidylinositol 4,5-Diphosphate/metabolism , Adenosine Triphosphate/pharmacology , Animals , Calcium Channels/drug effects , Cyclic AMP-Dependent Protein Kinases/metabolism , Electric Conductivity , Enzyme Activation , Hydrolysis , Ion Channel Gating/drug effects , Models, Biological , Mutation , Neurons/drug effects , Neurons/enzymology , Neurons/metabolism , Oocytes/drug effects , Oocytes/enzymology , Oocytes/metabolism , Patch-Clamp Techniques , Phosphatidylinositol 4,5-Diphosphate/deficiency , Phosphatidylinositol 4,5-Diphosphate/pharmacology , Phosphorylation , Rabbits , Rana catesbeiana , Rats , Receptor, trkA/genetics , Receptor, trkA/metabolism , Xenopus laevis
SELECTION OF CITATIONS
SEARCH DETAIL