Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 36
Filter
1.
Biomed Pharmacother ; 174: 116472, 2024 May.
Article in English | MEDLINE | ID: mdl-38531121

ABSTRACT

The Voltage-Gated Calcium Channel (VGCC) auxiliary subunit Cavα2δ-1 (CACNA2D1) is the target/receptor of gabapentinoids which are known therapeutics in epilepsy and neuropathic pain. Following damage to the peripheral sensory nervous system, Cavα2δ-1 is upregulated in dorsal root ganglion (DRG) neurons in several animal models of chronic neuropathic pain. Gabapentinoids, such as gabapentin and pregabalin, engage with Cavα2δ-1 via binding an arginine residue (R241) within an RRR motif located at the N-terminus of human Cavα2δ-1. A novel, next generation gabapentinoid, engineered not to penetrate the brain, was able to generate a strong analgesic response in Chronic Constriction Injury animal model of chronic neuropathic pain and showed binding specificity for Cavα2δ-1 versus the Cavα2δ-2 subunit. This novel non-brain penetrant gabapentinoid, binds to R241 and a novel binding site on Cavα2δ-1, which is located within the VGCC_α2 domain, identified as a lysine residue within an IKAK amino acid motif (K634). The overall whole cell current amplitudes were diminished by the compound, with these inhibitory effects being diminished in R241A mutant Cavα2δ-1 subunits. The functional effects occurred at lower concentrations than those needed for inhibition by gabapentin or pregabalin, which apparently bound the Cavα2δ-1 subunit only on the R241 and not on the K634 residue. Our work sets the stage for the identification and characterisation of novel compounds with therapeutic properties in neuropathic pain and possibly in other disorders and conditions which require engagement of the Cavα2δ-1 target.


Subject(s)
Calcium Channels, L-Type , Neuralgia , Neuralgia/drug therapy , Neuralgia/metabolism , Animals , Ligands , Humans , Male , Calcium Channels/metabolism , Calcium Channels/genetics , Gabapentin/pharmacology , Rats, Sprague-Dawley , Ganglia, Spinal/metabolism , Ganglia, Spinal/drug effects , Rats , Calcium Channel Blockers/pharmacology , Calcium Channels, N-Type/metabolism , Calcium Channels, N-Type/genetics , Analgesics/pharmacology , Disease Models, Animal , Pregabalin/pharmacology
2.
Br J Pharmacol ; 180(12): 1616-1633, 2023 06.
Article in English | MEDLINE | ID: mdl-36647671

ABSTRACT

BACKGROUND AND PURPOSE: Cannabinoids are a promising therapeutic avenue for chronic pain. However, clinical trials often fail to report analgesic efficacy of cannabinoids. Inhibition of voltage gate calcium (Cav ) channels is one mechanism through which cannabinoids may produce analgesia. We hypothesized that cannabinoids and cannabinoid receptor agonists target different types of Cav channels through distinct mechanisms. EXPERIMENTAL APPROACH: Electrophysiological recordings from tsA-201 cells expressing either Cav 3.2 or Cav 2.2 were used to assess inhibition by HU-210 or cannabidiol (CBD) in the absence and presence of the CB1 receptor. Homology modelling assessed potential interaction sites for CBD in both Cav 2.2 and Cav 3.2. Analgesic effects of CBD were assessed in mouse models of inflammatory and neuropathic pain. KEY RESULTS: HU-210 (1 µM) inhibited Cav 2.2 function in the presence of CB1 receptor but had no effect on Cav 3.2 regardless of co-expression of CB1 receptor. By contrast, CBD (3 µM) produced no inhibition of Cav 2.2 and instead inhibited Cav 3.2 independently of CB1 receptors. Homology modelling supported these findings, indicating that CBD binds to and occludes the pore of Cav 3.2, but not Cav 2.2. Intrathecal CBD alleviated thermal and mechanical hypersensitivity in both male and female mice, and this effect was absent in Cav 3.2 null mice. CONCLUSION AND IMPLICATIONS: Our findings reveal differential modulation of Cav 2.2 and Cav 3.2 channels by CB1 receptors and CBD. This advances our understanding of how different cannabinoids produce analgesia through action at different voltage-gated calcium channels and could influence the development of novel cannabinoid-based therapeutics for treatment of chronic pain.


Subject(s)
Cannabidiol , Cannabinoids , Chronic Pain , Male , Female , Mice , Animals , Cannabidiol/pharmacology , Calcium Channels , Chronic Pain/drug therapy , Analgesics/pharmacology , Receptor, Cannabinoid, CB1/metabolism , Receptor, Cannabinoid, CB2/metabolism
3.
Mol Brain ; 15(1): 91, 2022 11 17.
Article in English | MEDLINE | ID: mdl-36397158

ABSTRACT

Trigeminal neuralgia (TN) is a rare form of chronic neuropathic pain characterized by spontaneous or elicited paroxysms of electric shock-like or stabbing pain in a region of the face. While most cases occur in a sporadic manner and are accompanied by intracranial vascular compression of the trigeminal nerve root, alteration of ion channels has emerged as a potential exacerbating factor. Recently, whole exome sequencing analysis of familial TN patients identified 19 rare variants in the gene CACNA1H encoding for Cav3.2T-type calcium channels. An initial analysis of 4 of these variants pointed to a pathogenic role. In this study, we assessed the electrophysiological properties of 13 additional TN-associated Cav3.2 variants expressed in tsA-201 cells. Our data indicate that 6 out of the 13 variants analyzed display alteration of their gating properties as evidenced by a hyperpolarizing shift of their voltage dependence of activation and/or inactivation resulting in an enhanced window current supported by Cav3.2 channels. An additional variant enhanced the recovery from inactivation. Simulation of neuronal electrical membrane potential using a computational model of reticular thalamic neuron suggests that TN-associated Cav3.2 variants could enhance neuronal excitability. Altogether, the present study adds to the notion that ion channel polymorphisms could contribute to the etiology of some cases of TN and further support a role for Cav3.2 channels.


Subject(s)
Trigeminal Neuralgia , Humans , Calcium Channels , Membrane Potentials , Neurons , Trigeminal Neuralgia/genetics , Electrophysiological Phenomena
4.
Pain ; 163(12): 2315-2325, 2022 12 01.
Article in English | MEDLINE | ID: mdl-35467587

ABSTRACT

ABSTRACT: Trigeminal neuralgia (TN) is a rare but debilitating disorder characterized by excruciating facial pain, with a higher incidence in women. Recent studies demonstrated that TN patients present mutations in the gene encoding the Ca V 3.2 T-type calcium channel, an important player in peripheral pain pathways. We characterize the role of Ca V 3.2 channels in TN at 2 levels. First, we examined the biophysical properties of CACNA1H variants found in TN patients. Second, we investigated the role of Ca V 3.2 in an animal model of trigeminal neuropathic pain. Whole-cell patch-clamp recordings from 4 different mutants expressed in tsA-201 cells (E286K in the pore loop of domain I, H526Y, G563R, and P566T in the domain I-II linker) identified a loss of function in activation in the E286K mutation and gain of function in the G563R and P566T mutations. Moreover, a loss of function in inactivation was observed with the E286K and H526Y mutations. Cell surface biotinylation revealed no difference in channel trafficking among the variants. The G563R mutant also caused a gain of function in the firing properties of transfected trigeminal ganglion neurons. In female and male mice, constriction of the infraorbital nerve induced facial thermal heat hyperalgesia. Block of T-type channels with Z944 resulted in antihyperalgesia. The effect of Z944 was absent in Ca V 3.2 -/- mice, indicating that Ca V 3.2 is the molecular target of the antihyperalgesic Z944 effect. Finally, enzyme-linked immunosorbent assay analysis revealed increased Ca V 3.2 channel expression in the spinal trigeminal subnucleus caudalis. Altogether, the present study demonstrates an important role of Ca V 3.2 channels in trigeminal pain.


Subject(s)
Calcium Channels, T-Type , Trigeminal Neuralgia , Animals , Female , Male , Mice , Facial Pain , Hyperalgesia , Trigeminal Ganglion/metabolism , Trigeminal Neuralgia/genetics , Calcium Channels, T-Type/metabolism
6.
Pflugers Arch ; 474(4): 447-454, 2022 04.
Article in English | MEDLINE | ID: mdl-34623515

ABSTRACT

This study describes the interaction between CaV3.2 calcium channels and the receptor for activated C kinase 1 (Rack-1), a scaffold protein which has recently been implicated in neuropathic pain. The coexpression of CaV3.2 and Rack-1 in tsA-201 cells led to a reduction in the magnitude of whole-cell CaV3.2 currents and CaV3.2 channel expression at the plasma membrane. Co-immunoprecipitations from transfected cells show the formation of a molecular protein complex between Cav3.2 channels and Rack-1. We determined that the interaction of Rack-1 occurs at the intracellular II-III loop and the C-terminus of the channel. Finally, the coexpression of PKCßII abolished the effect of Rack-1 on current densities. Altogether, our findings show that Rack-1 regulates CaV3.2-mediated calcium entry in a PKC-dependent manner.


Subject(s)
Calcium Channels, T-Type , Neuralgia , Calcium Channels, T-Type/metabolism , Cell Membrane/metabolism , Humans , Receptors for Activated C Kinase/metabolism
7.
Mol Brain ; 14(1): 145, 2021 09 20.
Article in English | MEDLINE | ID: mdl-34544471

ABSTRACT

The CACNA1H gene encodes the α1 subunit of the low voltage-activated Cav3.2 T-type calcium channel, an important regulator of neuronal excitability. Alternative mRNA splicing can generate multiple channel variants with distinct biophysical properties and expression patterns. Two major splice variants, containing or lacking exon 26 (± 26) have been found in different human tissues. In this study, we report splice variant specific effects of a Cav3.2 mutation found in patients with autosomal dominant writer's cramp, a specific type of focal dystonia. We had previously reported that the R481C missense mutation caused a gain of function effect when expressed in Cav3.2 (+ 26) by accelerating its recovery from inactivation. Here, we show that when the mutation is expressed in the short variant of the channel (- 26), we observe a significant increase in current density when compared to wild-type Cav3.2 (- 26) but the effect on the recovery from inactivation is lost. Our data add to growing evidence that the functional expression of calcium channel mutations depends on which splice variant is being examined.


Subject(s)
Calcium Channels, T-Type/genetics , Dystonic Disorders/genetics , Action Potentials , Alternative Splicing , Barium/metabolism , Calcium Channels, T-Type/physiology , Dystonic Disorders/physiopathology , Exons/genetics , Gain of Function Mutation , Humans , Ion Transport , Models, Molecular , Mutation, Missense , Point Mutation , Protein Isoforms/genetics , Protein Isoforms/physiology
8.
Mol Brain ; 14(1): 126, 2021 08 16.
Article in English | MEDLINE | ID: mdl-34399820

ABSTRACT

Developmental and epileptic encephalopathies (DEEs) are a group of severe epilepsies that are characterized by seizures and developmental delay. DEEs are primarily attributed to genetic causes and an increasing number of cases have been correlated with variants in ion channel genes. In this study, we report a child with an early severe DEE. Whole exome sequencing showed a de novo heterozygous variant (c.4873-4881 duplication) in the SCN8A gene and an inherited heterozygous variant (c.952G > A) in the CACNA1H gene encoding for Nav1.6 voltage-gated sodium and Cav3.2 voltage-gated calcium channels, respectively. In vitro functional analysis of human Nav1.6 and Cav3.2 channel variants revealed mild but significant alterations of their gating properties that were in general consistent with a gain- and loss-of-channel function, respectively. Although additional studies will be required to confirm the actual pathogenic involvement of SCN8A and CACNA1H, these findings add to the notion that rare ion channel variants may contribute to the etiology of DEEs.


Subject(s)
Developmental Disabilities/genetics , Drug Resistant Epilepsy/genetics , Epilepsy, Tonic-Clonic/genetics , NAV1.6 Voltage-Gated Sodium Channel/genetics , Abnormalities, Multiple/genetics , Calcium Channels, T-Type/genetics , Calcium Channels, T-Type/physiology , Female , Gain of Function Mutation , Gene Duplication , Genetic Predisposition to Disease , Humans , Infant, Newborn , Ion Channel Gating/genetics , Ion Channel Gating/physiology , Mutation, Missense , NAV1.6 Voltage-Gated Sodium Channel/physiology , Pedigree , Point Mutation , Scoliosis/genetics
9.
Mol Brain ; 14(1): 27, 2021 02 08.
Article in English | MEDLINE | ID: mdl-33557884

ABSTRACT

CACNA1A pathogenic variants have been linked to several neurological disorders including familial hemiplegic migraine and cerebellar conditions. More recently, de novo variants have been associated with severe early onset developmental encephalopathies. CACNA1A is highly expressed in the central nervous system and encodes the pore-forming CaVα1 subunit of P/Q-type (Cav2.1) calcium channels. We have previously identified a patient with a de novo missense mutation in CACNA1A (p.Y1384C), characterized by hemiplegic migraine, cerebellar atrophy and developmental delay. The mutation is located at the transmembrane S5 segment of the third domain. Functional analysis in two predominant splice variants of the neuronal Cav2.1 channel showed a significant loss of function in current density and changes in gating properties. Moreover, Y1384 variants exhibit differential splice variant-specific effects on recovery from inactivation. Finally, structural analysis revealed structural damage caused by the tyrosine substitution and changes in electrostatic potentials.


Subject(s)
Calcium Channels, N-Type/genetics , Cerebellum/pathology , Developmental Disabilities/genetics , Genetic Predisposition to Disease , Migraine with Aura/genetics , Mutation/genetics , Adolescent , Adult , Alternative Splicing/genetics , Atrophy , Biophysical Phenomena , Calcium Channels, N-Type/chemistry , Calcium Channels, N-Type/metabolism , Cell Line , Child, Preschool , Developmental Disabilities/complications , Female , Humans , Infant, Newborn , Ion Channel Gating , Male , Migraine with Aura/complications , Models, Molecular , Mutant Proteins/chemistry , Mutant Proteins/metabolism , Protein Isoforms/genetics , Protein Structure, Secondary , Structural Homology, Protein
10.
Mol Brain ; 14(1): 18, 2021 01 21.
Article in English | MEDLINE | ID: mdl-33478561

ABSTRACT

Writer's cramp (WC) is a task-specific focal dystonia that occurs selectively in the hand and arm during writing. Previous studies have shown a role for genetics in the pathology of task-specific focal dystonia. However, to date, no causal gene has been reported for task-specific focal dystonia, including WC. In this study, we investigated the genetic background of a large Dutch family with autosomal dominant‒inherited WC that was negative for mutations in known dystonia genes. Whole exome sequencing identified 4 rare variants of unknown significance that segregated in the family. One candidate gene was selected for follow-up, Calcium Voltage-Gated Channel Subunit Alpha1 H, CACNA1H, due to its links with the known dystonia gene Potassium Channel Tetramerization Domain Containing 17, KCTD17, and with paroxysmal movement disorders. Targeted resequencing of CACNA1H in 82 WC cases identified another rare, putative damaging variant in a familial WC case that did not segregate. Using structural modelling and functional studies in vitro, we show that both the segregating p.Arg481Cys variant and the non-segregating p.Glu1881Lys variant very likely cause structural changes to the Cav3.2 protein and lead to similar gains of function, as seen in an accelerated recovery from inactivation. Both mutant channels are thus available for re-activation earlier, which may lead to an increase in intracellular calcium and increased neuronal excitability. Overall, we conclude that rare functional variants in CACNA1H need to be interpreted very carefully, and additional studies are needed to prove that the p.Arg481Cys variant is the cause of WC in the large Dutch family.


Subject(s)
Calcium Channels, T-Type/genetics , Dystonic Disorders/genetics , Genetic Predisposition to Disease , Mutation, Missense/genetics , Chromosome Segregation , Female , Humans , Male , Pedigree , Phenotype
11.
Mol Brain ; 14(1): 4, 2021 01 07.
Article in English | MEDLINE | ID: mdl-33413531

ABSTRACT

A novel missense mutation in the CACNA1A gene that encodes the pore forming α1 subunit of the CaV2.1 voltage-gated calcium channel was identified in a patient with trigeminal neuralgia. This mutation leads to a substitution of proline 2455 by histidine (P2455H) in the distal C-terminus region of the channel. Due to the well characterized role of this channel in neurotransmitter release, our aim was to characterize the biophysical properties of the P2455H variant in heterologously expressed CaV2.1 channels. Whole-cell patch clamp recordings of wild type and mutant CaV2.1 channels expressed in tsA-201 cells reveal that the mutation mediates a depolarizing shift in the voltage-dependence of activation and inactivation. Moreover, the P2455H mutant strongly reduced calcium-dependent inactivation of the channel that is consistent with an overall gain of function. Hence, the P2455H CaV2.1 missense mutation alters the gating properties of the channel, suggesting that associated changes in CaV2.1-dependent synaptic communication in the trigeminal system may contribute to the development of trigeminal neuralgia.


Subject(s)
Calcium Channels/genetics , Genetic Association Studies , Genetic Predisposition to Disease , Genetic Variation , Ion Channel Gating/genetics , Trigeminal Neuralgia/genetics , Cell Line , Humans , Mutation/genetics
12.
Mol Brain ; 13(1): 149, 2020 11 11.
Article in English | MEDLINE | ID: mdl-33176830

ABSTRACT

Low-voltage-activated T-type calcium channels are important contributors to nervous system function. Post-translational modification of these channels has emerged as an important mechanism to control channel activity. Previous studies have documented the importance of asparagine (N)-linked glycosylation and identified several asparagine residues within the canonical consensus sequence N-X-S/T that is essential for the expression and function of Cav3.2 channels. Here, we explored the functional role of non-canonical N-glycosylation motifs in the conformation N-X-C based on site directed mutagenesis. Using a combination of electrophysiological recordings and surface biotinylation assays, we show that asparagines N345 and N1780 located in the motifs NVC and NPC, respectively, are essential for the expression of the human Cav3.2 channel in the plasma membrane. Therefore, these newly identified asparagine residues within non-canonical motifs add to those previously reported in canonical sites and suggest that N-glycosylation of Cav3.2 may also occur at non-canonical motifs to control expression of the channel in the plasma membrane. It is also the first study to report the functional importance of non-canonical N-glycosylation motifs in an ion channel.


Subject(s)
Calcium Channels, T-Type/metabolism , Amino Acid Motifs , Asparagine/metabolism , Calcium Channels, T-Type/chemistry , Glycosylation , Humans , Structure-Activity Relationship
13.
Mol Brain ; 13(1): 33, 2020 03 06.
Article in English | MEDLINE | ID: mdl-32143681

ABSTRACT

Amyotrophic lateral sclerosis (ALS) is a neurodegenerative disorder characterized by the progressive loss of cortical, brain stem and spinal motor neurons that leads to muscle weakness and death. A previous study implicated CACNA1H encoding for Cav3.2 calcium channels as a susceptibility gene in ALS. In the present study, two heterozygous CACNA1H variants were identified by whole genome sequencing in a small cohort of ALS patients. These variants were functionally characterized using patch clamp electrophysiology, biochemistry assays, and molecular modeling. A previously unreported c.454GTAC > G variant produced an inframe deletion of a highly conserved isoleucine residue in Cav3.2 (p.ΔI153) and caused a complete loss-of-function of the channel, with an additional dominant-negative effect on the wild-type channel when expressed in trans. In contrast, the c.3629C > T variant caused a missense substitution of a proline with a leucine (p.P1210L) and produced a comparatively mild alteration of Cav3.2 channel activity. The newly identified ΔI153 variant is the first to be reported to cause a complete loss of Cav3.2 channel function. These findings add to the notion that loss-of-function of Cav3.2 channels associated with rare CACNA1H variants may be risk factors in the complex etiology of ALS.


Subject(s)
Amyotrophic Lateral Sclerosis/genetics , Calcium Channels, T-Type/genetics , Genetic Association Studies , Genetic Predisposition to Disease , Mutation/genetics , Amino Acid Sequence , Animals , Calcium Channels, T-Type/chemistry , Genes, Dominant , Heterozygote , Male , Rats , Structural Homology, Protein , Whole Genome Sequencing
14.
Mol Brain ; 12(1): 98, 2019 11 27.
Article in English | MEDLINE | ID: mdl-31775826

ABSTRACT

We have examined the regulation of mutually exclusive Cav2.2 exon 37a and b variants by the mouse µ-opioid receptor (mMOR) C-terminal splice variants 1, 1C and 1O in tsA-201 cells. Electrophysiological analyses revealed that both channel isoforms exhibit DAMGO-induced voltage-dependent (Gßγ-mediated) inhibition and its recovery by voltage pre-pulses, as well as a voltage-independent component. However, the two channel isoforms differ in their relative extent of voltage-dependent and independent inhibition, with Cav2.2-37b showing significantly more voltage-dependent inhibition upon activation of the three mMOR receptors studied. In addition, coexpression of either mMOR1 or mMOR1C results in an agonist-independent reduction in the peak current density of Cav2.2-37a channels, whereas the peak current density of Cav2.2-37b does not appear to be affected. Interestingly, this decrease is not due to an effect on channel expression at the plasma membrane, as demonstrated by biotinylation experiments. We further examined the mechanism underlying the agonist-independent modulation of Cav2.2-37a by mMOR1C. Incubation of cells with pertussis toxin did not affect the mMOR1C mediated inhibition of Cav2.2-37a currents, indicating a lack of involvement of Gi/o signaling. However, when a Src tyrosine kinase inhibitor was applied, the effect of mMOR1C was lost. Moreover, when we recorded currents using a Cav2.2-37a mutant in which tyrosine 1747 was replaced with phenylalanine (Y1747F), the agonist independent effects of mMOR1C were abolished. Altogether our findings show that Cav2.2-37a and Cav2.2-37b isoforms are subject to differential regulation by C-terminal splice variants of mMORs, and that constitutive mMOR1C activity and downstream tyrosine kinase activity exert a selective inhibition of the Cav2.2-37a splice variant, an N-type channel isoform that is highly enriched in nociceptors. Our study provides new insights into the roles of the MOR full-length C-terminal variants in modulating Cav2.2 channel isoform activities.


Subject(s)
Alternative Splicing/genetics , Calcium Channels, N-Type/genetics , Exons/genetics , Receptors, Opioid, mu/genetics , Animals , Calcium Channels, N-Type/metabolism , Enkephalin, Ala(2)-MePhe(4)-Gly(5)-/pharmacology , GTP-Binding Proteins/metabolism , Ion Channel Gating/genetics , Mice , Protein-Tyrosine Kinases/metabolism , Rats , Receptors, Opioid, mu/metabolism
15.
Mol Brain ; 12(1): 86, 2019 10 24.
Article in English | MEDLINE | ID: mdl-31651342

ABSTRACT

Two paternally-inherited missense variants in CACNA1H were identified and characterized in a 6-year-old child with generalized epilepsy. Febrile and unprovoked seizures were present in this child. Both variants were expressed in cis or isolation using human recombinant Cav3.2 calcium channels in tsA-201 cells. Whole-cell patch-clamp recordings indicated that one variant (c.3844C > T; p.R1282W) caused a significant increase in current density consistent with a pathogenic gain-of-function phenotype; while the other cis-related variant (c.5294C > T; p.A1765V) had a benign profile.


Subject(s)
Calcium Channels, T-Type/genetics , Epilepsy, Generalized/genetics , Mutation/genetics , Biophysical Phenomena , Child , Female , Humans , Infant , Infant, Newborn
16.
Mol Brain ; 12(1): 75, 2019 09 02.
Article in English | MEDLINE | ID: mdl-31477143

ABSTRACT

Ankyrin B (AnkB) is an adaptor and scaffold for motor proteins and various ion channels that is ubiquitously expressed, including in the brain. AnkB has been associated with neurological disorders such as epilepsy and autism spectrum disorder, but understanding of the underlying mechanisms is limited. Cav2.1, the pore-forming subunit of P/Q type voltage gated calcium channels, is a known interactor of AnkB and plays a crucial role in neuronal function. Here we report that wildtype AnkB increased overall Cav2.1 levels without impacting surface Cav2.1 levels in HEK293T cells. An AnkB variant, p.S646F, which we recently discovered to be associated with seizures, further increased overall Cav2.1 levels, again with no impact on surface Cav2.1 levels. AnkB p.Q879R, on the other hand, increased surface Cav2.1 levels in the presence of accessory subunits α2δ1 and ß4. Additionally, AnkB p.E1458G decreased surface Cav2.1 irrespective of the presence of accessory subunits. In addition, we found that partial deletion of AnkB in cortex resulted in a decrease in overall Cav2.1 levels, with no change to the levels of Cav2.1 detected in synaptosome fractions. Our work suggests that depending on the particular variant, AnkB regulates intracellular and surface Cav2.1. Notably, expression of the AnkB variant associated with seizure (AnkB p.S646F) caused further increase in intracellular Cav2.1 levels above that of even wildtype AnkB. These novel findings have important implications for understanding the role of AnkB and Cav2.1 in the regulation of neuronal function in health and disease.


Subject(s)
Ankyrins/metabolism , Calcium Channels, N-Type/metabolism , Cell Membrane/metabolism , Intracellular Space/metabolism , Mutant Proteins/metabolism , Animals , Ankyrins/genetics , HEK293 Cells , Humans , Mice, Inbred C57BL , Models, Biological , Mutation/genetics , Protein Subunits/metabolism , Synapses/metabolism
17.
Nat Neurosci ; 22(10): 1659-1668, 2019 10.
Article in English | MEDLINE | ID: mdl-31501573

ABSTRACT

Neuropathic pain can be a debilitating condition with both sensory and affective components, the underlying brain circuitry of which remains poorly understood. In the present study, a basolateral amygdala (BLA)-prefrontal cortex (PFC)-periaqueductal gray (PAG)-spinal cord pathway was identified that is critical for the development of mechanical and thermal hypersensitivity after peripheral nerve injury. It was shown that nerve injury strengthens synaptic input from the BLA onto inhibitory interneurons located in the prelimbic medial PFC, by virtue of reduced endocannabinoid modulation. These augmented synaptic connections mediate a feedforward inhibition of projections from the PFC to the ventrolateral PAG region and its downstream targets. Optogenetic approaches combined with in vivo pharmacology reveal that these BLA-PFC-PAG connections alter pain behaviors by reducing descending noradrenergic and serotoninergic modulation of spinal pain signals. Thus, a long-range brain circuit was identified that is crucial for pain processing and that can potentially be exploited toward targeting neuropathic pain.


Subject(s)
Neural Pathways/pathology , Neuralgia/pathology , Neurons/pathology , Amygdala/pathology , Animals , Behavior, Animal , Hot Temperature , Male , Mice , Mice, Inbred C57BL , Motor Activity , Neuralgia/psychology , Optogenetics , Periaqueductal Gray/pathology , Peripheral Nerve Injuries/pathology , Peripheral Nerve Injuries/psychology , Physical Stimulation , Prefrontal Cortex/pathology , Spinal Cord/pathology , Synapses/pathology
18.
Mol Brain ; 12(1): 62, 2019 06 28.
Article in English | MEDLINE | ID: mdl-31253182

ABSTRACT

This study describes the interaction between Cav2 calcium channels and Rabconnectin-3, a di-subunit protein that is associated with synaptic vesicles. Immunostaining reveals that both Rabconnectin-3α (RB-3α) and Rabconnectin-3ß (RB-3ß) are colocalized in mouse hippocampal neurons. Co-immunoprecipitations from brain tissue is consistent with the formation of a protein complex between RB-3α and RB-3ß and both Cav2.2 and the related Cav2.1 calcium channel. The coexpression of either RB-3α or RB-3ß with Cav2.2 calcium channels in tsA-201 cells led to a reduction in Cav2.2 current density without any effects on the voltage-dependence of activation or inactivation. Coexpression of both Rabconnectin-3 subunits did not cause an additive effect on current densities. Finally, the presence of Rabconnectin-3 did not interfere with µ-opioid receptor mediated Gßγ modulation of Cav2.2 channels. Altogether, our findings show that Rabconnectin-3 has the propensity to regulate calcium entry mediated by Cav2.2 channels.


Subject(s)
Caveolin 2/metabolism , Nerve Tissue Proteins/metabolism , Animals , GTP-Binding Proteins/metabolism , Hippocampus/cytology , Humans , Mice , Neurons/metabolism , Protein Binding , Rats
20.
Mol Brain ; 12(1): 12, 2019 02 08.
Article in English | MEDLINE | ID: mdl-30736831

ABSTRACT

This study describes the functional interaction between Cav3.2 calcium channels and the Epithelial Sodium Channel (ENaC). ß-ENaC subunits showed overlapping expression with endogenous Cav3.2 calcium channels in the thalamus and hypothalamus as detected by immunostaining. Moreover, ß- and γ-ENaC subunits could be co-immunoprecipitated with Cav3.2 calcium channels from brain lysates, dorsal horn and lumbar dorsal root ganglia. Mutation of a cluster of lysines present in the intracellular N-terminus region of ß-ENaC (K4R/ K5R/ K9R/ K16R/ K23R) reduced interactions with Cav3.2 calcium channels. Αßγ-ENaC channels enhanced Cav3.2 calcium channel trafficking to the plasma membrane in tsA-201 cells. This effect was reciprocal such that Cav3.2 channel expression also enhanced ß-ENaC trafficking to the cell surface. T-type current density was increased when fully assembled αßγ-ENaC channels were transiently expressed in CAD cells, a neuronal derived cell line. Altogether, these findings reveal ENaC as an interactor and potential regulator of Cav3.2 calcium channels expressed in neuronal tissues.


Subject(s)
Calcium Channels, T-Type/metabolism , Epithelial Sodium Channels/metabolism , Animals , Brain/metabolism , Cell Membrane/metabolism , Epithelial Sodium Channels/chemistry , Ion Channel Gating , Mice, Inbred C57BL , Protein Binding , Protein Subunits/metabolism , Protein Transport , Rats
SELECTION OF CITATIONS
SEARCH DETAIL
...