Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 11 de 11
1.
Mol Psychiatry ; 28(1): 76-82, 2023 01.
Article En | MEDLINE | ID: mdl-36224259

Pitt Hopkins Syndrome (PTHS) is a rare syndromic form of autism spectrum disorder (ASD) caused by autosomal dominant mutations in the Transcription Factor 4 (TCF4) gene. TCF4 is a basic helix-loop-helix transcription factor that is critical for neurodevelopment and brain function through its binding to cis-regulatory elements of target genes. One potential therapeutic strategy for PTHS is to identify dysregulated target genes and normalize their dysfunction. Here, we propose that SCN10A is an important target gene of TCF4 that is an applicable therapeutic approach for PTHS. Scn10a encodes the voltage-gated sodium channel Nav1.8 and is consistently shown to be upregulated in PTHS mouse models. In this perspective, we review prior literature and present novel data that suggests inhibiting Nav1.8 in PTHS mouse models is effective at normalizing neuron function, brain circuit activity and behavioral abnormalities and posit this therapeutic approach as a treatment for PTHS.


Intellectual Disability , NAV1.8 Voltage-Gated Sodium Channel , Animals , Mice , Autism Spectrum Disorder/drug therapy , Autism Spectrum Disorder/genetics , Autism Spectrum Disorder/metabolism , Facies , Hyperventilation/genetics , Intellectual Disability/drug therapy , Intellectual Disability/genetics , Intellectual Disability/metabolism , Transcription Factor 4/genetics , NAV1.8 Voltage-Gated Sodium Channel/chemistry , NAV1.8 Voltage-Gated Sodium Channel/metabolism
2.
FASEB J ; 35(12): e22025, 2021 12.
Article En | MEDLINE | ID: mdl-34758144

Mepyramine, a first-generation antihistamine targeting the histamine H(1) receptor, was extensively prescribed to patients suffering from allergic reactions and urticaria. Serious adverse effects, especially in case of overdose, were frequently reported, including drowsiness, impaired thinking, convulsion, and coma. Many of these side effects were associated with the blockade of histaminergic or cholinergic receptors. Here we show that mepyramine directly inhibits a variety of voltage-gated sodium channels, including the Tetrodotoxin-sensitive isoforms and the main isoforms (Nav1.7, Nav1.8, and Nav1.9) of nociceptors. Estimated IC50 were within the range of drug concentrations detected in poisoned patients. Mepyramine inhibited sodium channels through fast- or slow-inactivated state preference depending on the isoform. Moreover, mepyramine inhibited the firing responses of C- and Aß-type nerve fibers in ex vivo skin-nerve preparations. Locally applied mepyramine had analgesic effects on the scorpion toxin-induced excruciating pain and produced pain relief in acute, inflammatory, and chronic pain models. Collectively, these data provide evidence that mepyramine has the potential to be developed as a topical analgesic agent.


Arthritis, Experimental/complications , Ganglia, Spinal/drug effects , NAV1.8 Voltage-Gated Sodium Channel/physiology , Nociceptors/drug effects , Pain/drug therapy , Pyrilamine/pharmacology , Sodium Channel Blockers/pharmacology , Action Potentials , Animals , Ganglia, Spinal/metabolism , Ganglia, Spinal/pathology , Histamine H1 Antagonists/pharmacology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , NAV1.8 Voltage-Gated Sodium Channel/chemistry , Nociceptors/metabolism , Nociceptors/pathology , Pain/etiology , Pain/metabolism , Pain/pathology
3.
Biochem Biophys Res Commun ; 533(1): 168-174, 2020 11 26.
Article En | MEDLINE | ID: mdl-32948286

The voltage-gated sodium channels play a key role in the generation and propagation of the cardiac action potential. Emerging data indicate that the Nav1.8 channel, encoded by the SCN10A gene, is a modulator of cardiac conduction and variation in the gene has been associated with arrhythmias such as atrial fibrillation (AF) and Brugada syndrome (BrS). The voltage gated sodium channels contain a calmodulin (CaM)-binding IQ domain involved in channel slow inactivation, we here investigated the role of CaM regulation of Nav1.8 channel function, and showed that CaM enhanced slow inactivation of the Nav1.8 channel and hyperpolarized steady-state inactivation curve of sodium currents. The effects of CaM on the channel gating were disrupted in the Nav1.8 channel truncated IQ domain. We studied Nav1.8 IQ domain mutations associated with AF and BrS, and found that a BrS-linked mutation (R1863Q) reduced the CaM-induced hyperpolarization shift, AF-linked mutations (R1869C and R1869G) disrupted CaM-induced enhanced inactivation, and effects of CaM on both development and recovery from slow inactivation were attenuated in all pathogenic mutations. Our findings indicate a role of CaM in the regulation of Nav1.8 channel function in cardiac arrhythmias.


Calmodulin/metabolism , NAV1.8 Voltage-Gated Sodium Channel/genetics , Point Mutation , Amino Acid Sequence , Animals , Atrial Fibrillation/genetics , Atrial Fibrillation/metabolism , Brugada Syndrome/genetics , Brugada Syndrome/metabolism , Cell Line , Humans , Mice , NAV1.8 Voltage-Gated Sodium Channel/chemistry , NAV1.8 Voltage-Gated Sodium Channel/metabolism , Protein Binding , Protein Interaction Domains and Motifs , Rats
4.
Sci Rep ; 10(1): 14791, 2020 09 09.
Article En | MEDLINE | ID: mdl-32908170

The voltage-gated sodium channel isoform NaV1.7 is highly expressed in dorsal root ganglion neurons and is obligatory for nociceptive signal transmission. Genetic gain-of-function and loss-of-function NaV1.7 mutations have been identified in select individuals, and are associated with episodic extreme pain disorders and insensitivity to pain, respectively. These findings implicate NaV1.7 as a key pharmacotherapeutic target for the treatment of pain. While several small molecules targeting NaV1.7 have been advanced to clinical development, no NaV1.7-selective compound has shown convincing efficacy in clinical pain applications. Here we describe the discovery and characterization of ST-2262, a NaV1.7 inhibitor that blocks the extracellular vestibule of the channel with an IC50 of 72 nM and greater than 200-fold selectivity over off-target sodium channel isoforms, NaV1.1-1.6 and NaV1.8. In contrast to other NaV1.7 inhibitors that preferentially inhibit the inactivated state of the channel, ST-2262 is equipotent in a protocol that favors the resting state of the channel, a protocol that favors the inactivated state, and a high frequency protocol. In a non-human primate study, animals treated with ST-2262 exhibited reduced sensitivity to noxious heat. These findings establish the extracellular vestibule of the sodium channel as a viable receptor site for the design of selective ligands targeting NaV1.7.


Guanidine/chemistry , NAV1.7 Voltage-Gated Sodium Channel/chemistry , Sodium Channel Blockers/chemistry , Sodium Channel Blockers/pharmacology , Animals , Drug Discovery , Ganglia, Spinal/metabolism , Humans , NAV1.1 Voltage-Gated Sodium Channel/chemistry , NAV1.2 Voltage-Gated Sodium Channel/chemistry , NAV1.3 Voltage-Gated Sodium Channel/chemistry , NAV1.4 Voltage-Gated Sodium Channel/chemistry , NAV1.5 Voltage-Gated Sodium Channel/chemistry , NAV1.6 Voltage-Gated Sodium Channel/chemistry , NAV1.8 Voltage-Gated Sodium Channel/chemistry , Protein Structure, Secondary
5.
Sci Rep ; 10(1): 2326, 2020 02 11.
Article En | MEDLINE | ID: mdl-32047194

Acute pruritus occurs in various disorders. Despite severe repercussions on quality of life treatment options remain limited. Voltage-gated sodium channels (NaV) are indispensable for transformation and propagation of sensory signals implicating them as drug targets. Here, NaV1.7, 1.8 and 1.9 were compared for their contribution to itch by analysing NaV-specific knockout mice. Acute pruritus was induced by a comprehensive panel of pruritogens (C48/80, endothelin, 5-HT, chloroquine, histamine, lysophosphatidic acid, trypsin, SLIGRL, ß-alanine, BAM8-22), and scratching was assessed using a magnet-based recording technology. We report an unexpected stimulus-dependent diversity in NaV channel-mediated itch signalling. NaV1.7-/- showed substantial scratch reduction mainly towards strong pruritogens. NaV1.8-/- impaired histamine and 5-HT-induced scratching while NaV1.9 was involved in itch signalling towards 5-HT, C48/80 and SLIGRL. Furthermore, similar microfluorimetric calcium responses of sensory neurons and expression of itch-related TRP channels suggest no change in sensory transduction but in action potential transformation and conduction. The cumulative sum of scratching over all pruritogens confirmed a leading role of NaV1.7 and indicated an overall contribution of NaV1.9. Beside the proposed general role of NaV1.7 and 1.9 in itch signalling, scrutiny of time courses suggested NaV1.8 to sustain prolonged itching. Therefore, NaV1.7 and 1.9 may represent targets in pruritus therapy.


Histamine/toxicity , NAV1.7 Voltage-Gated Sodium Channel/physiology , NAV1.8 Voltage-Gated Sodium Channel/physiology , NAV1.9 Voltage-Gated Sodium Channel/physiology , Pruritus/prevention & control , Animals , Mice , Mice, Knockout , NAV1.7 Voltage-Gated Sodium Channel/chemistry , NAV1.8 Voltage-Gated Sodium Channel/chemistry , NAV1.9 Voltage-Gated Sodium Channel/chemistry , Pruritus/chemically induced , Pruritus/pathology , Signal Transduction
7.
Dokl Biochem Biophys ; 473(1): 137-140, 2017 Mar.
Article En | MEDLINE | ID: mdl-28510137

The use of the mathematical model of rat nociceptive neuron membrane allowed us to predict a new mechanism of suppression of ectopic bursting discharges, which arise in neurons of dorsal root ganglia and are one of the causes of neuropathic pain. The treatment with comenic acid leads to switching off the ectopic bursting discharges due to a decrease in the effective charge transferring via the activation gating structure of the slow sodium channels (Na V1.8a). Comenic acid is a drug substance of a new non-opioid analgesic [1] Thus, this analgesic not only reduces the frequency of rhythmic discharges of nociceptive neuron membrane [2] but also it suppresses its ectopic bursting discharges.


Models, Neurological , Nociceptors/cytology , Carboxylic Acids/pharmacology , Ion Channel Gating/drug effects , NAV1.8 Voltage-Gated Sodium Channel/chemistry , NAV1.8 Voltage-Gated Sodium Channel/metabolism , Nociceptors/drug effects , Nociceptors/metabolism , Pyrones/pharmacology , Tetrodotoxin/pharmacology
8.
J Biol Chem ; 291(22): 11829-42, 2016 May 27.
Article En | MEDLINE | ID: mdl-27026701

The µO-conotoxins MrVIA, MrVIB, and MfVIA inhibit the voltage-gated sodium channel NaV1.8, a well described target for the treatment of pain; however, little is known about the residues or structural elements that define this activity. In this study, we determined the three-dimensional structure of MfVIA, examined its membrane binding properties, performed alanine-scanning mutagenesis, and identified residues important for its activity at human NaV1.8. A second round of mutations resulted in (E5K,E8K)MfVIA, a double mutant with greater positive surface charge and greater affinity for lipid membranes compared with MfVIA. This analogue had increased potency at NaV1.8 and was analgesic in the mouse formalin assay.


Analgesics/pharmacology , Cell Membrane/metabolism , Conotoxins/pharmacology , NAV1.8 Voltage-Gated Sodium Channel/metabolism , Pain/prevention & control , Amino Acid Sequence , Animals , Behavior, Animal/drug effects , Crystallography, X-Ray , Electrophysiology , HEK293 Cells , Humans , Liposomes , Magnetic Resonance Spectroscopy , Male , Mice , Mice, Inbred C57BL , Molecular Sequence Data , NAV1.8 Voltage-Gated Sodium Channel/chemistry , NAV1.8 Voltage-Gated Sodium Channel/genetics , Pain/chemically induced , Protein Conformation , Sequence Homology, Amino Acid
9.
J Neurosci ; 34(21): 7190-7, 2014 May 21.
Article En | MEDLINE | ID: mdl-24849353

Resurgent sodium currents contribute to the regeneration of action potentials and enhanced neuronal excitability. Tetrodotoxin-sensitive (TTX-S) resurgent currents have been described in many different neuron populations, including cerebellar and dorsal root ganglia (DRG) neurons. In most cases, sodium channel Nav1.6 is the major contributor to these TTX-S resurgent currents. Here we report a novel TTX-resistant (TTX-R) resurgent current recorded from rat DRG neurons. The TTX-R resurgent currents are similar to classic TTX-S resurgent currents in many respects, but not all. As with TTX-S resurgent currents, they are activated by membrane repolarization, inhibited by lidocaine, and enhanced by a peptide-mimetic of the ß4 sodium channel subunit intracellular domain. However, the TTX-R resurgent currents exhibit much slower kinetics, occur at more depolarized voltages, and are sensitive to the Nav1.8 blocker A803467. Moreover, coimmunoprecipitation experiments from rat DRG lysates indicate the endogenous sodium channel ß4 subunits associate with Nav1.8 in DRG neurons. These results suggest that slow TTX-R resurgent currents in DRG neurons are mediated by Nav1.8 and are generated by the same mechanism underlying TTX-S resurgent currents. We also show that both TTX-S and TTX-R resurgent currents in DRG neurons are enhanced by inflammatory mediators. Furthermore, the ß4 peptide increased excitability of small DRG neurons in the presence of TTX. We propose that these slow TTX-R resurgent currents contribute to the membrane excitability of nociceptive DRG neurons under normal conditions and that enhancement of both types of resurgent currents by inflammatory mediators could contribute to sensory neuronal hyperexcitability associated with inflammatory pain.


Inflammation Mediators/pharmacology , Membrane Potentials/drug effects , NAV1.8 Voltage-Gated Sodium Channel/metabolism , Sensory Receptor Cells/drug effects , Sensory Receptor Cells/physiology , Sodium Channel Blockers/pharmacology , Tetrodotoxin/pharmacology , Aniline Compounds/pharmacology , Animals , Biophysics , Cells, Cultured , Electric Stimulation , Furans/pharmacology , Ganglia, Spinal/cytology , Immunoprecipitation , Lidocaine/pharmacology , NAV1.8 Voltage-Gated Sodium Channel/chemistry , Patch-Clamp Techniques , Peptides/pharmacology , Protein Subunits/metabolism , Rats , Rats, Sprague-Dawley
10.
Channels (Austin) ; 8(3): 210-5, 2014.
Article En | MEDLINE | ID: mdl-24763188

The voltage-gated sodium channel Na(V)1.8 (encoded by SCN10A) is predominantly expressed in dorsal root ganglia(DRG) and plays a critical role in pain perception. We analyzed SCN10A transcripts isolated from human DRGs using deep sequencing and found a novel splice variant lacking exon 11, which codes for 98 amino acids of the domain I/II linker. Quantitative PCR analysis revealed an abundance of this variant of up to 5­10% in human, while no such variants were detected in mouse or rat. Since no obvious functional differences between channels with and without the exon-11 sequence were detected, it is suggested that SCN10A exon 11 skipping in humans is a tolerated event.


Alternative Splicing , Exons , Ganglia, Spinal/metabolism , NAV1.8 Voltage-Gated Sodium Channel/genetics , Amino Acid Sequence , Animals , Humans , Mice , Mice, Inbred C57BL , Molecular Sequence Data , NAV1.8 Voltage-Gated Sodium Channel/chemistry , NAV1.8 Voltage-Gated Sodium Channel/metabolism , Rats , Rats, Wistar , Sequence Alignment
11.
Science ; 342(6157): 441-446, 2013 Oct 25.
Article En | MEDLINE | ID: mdl-24159039

Painful venoms are used to deter predators. Pain itself, however, can signal damage and thus serves an important adaptive function. Evolution to reduce general pain responses, although valuable for preying on venomous species, is rare, likely because it comes with the risk of reduced response to tissue damage. Bark scorpions capitalize on the protective pain pathway of predators by inflicting intensely painful stings. However, grasshopper mice regularly attack and consume bark scorpions, grooming only briefly when stung. Bark scorpion venom induces pain in many mammals (house mice, rats, humans) by activating the voltage-gated Na(+) channel Nav1.7, but has no effect on Nav1.8. Grasshopper mice Nav1.8 has amino acid variants that bind bark scorpion toxins and inhibit Na(+) currents, blocking action potential propagation and inducing analgesia. Thus, grasshopper mice have solved the predator-pain problem by using a toxin bound to a nontarget channel to block transmission of the pain signals the venom itself is initiating.


Arvicolinae/metabolism , Food Chain , NAV1.7 Voltage-Gated Sodium Channel/metabolism , NAV1.8 Voltage-Gated Sodium Channel/metabolism , Pain/metabolism , Predatory Behavior , Action Potentials/drug effects , Action Potentials/physiology , Amino Acid Sequence , Animals , Formaldehyde/pharmacology , Mice , Molecular Sequence Data , NAV1.7 Voltage-Gated Sodium Channel/chemistry , NAV1.7 Voltage-Gated Sodium Channel/genetics , NAV1.8 Voltage-Gated Sodium Channel/chemistry , NAV1.8 Voltage-Gated Sodium Channel/genetics , Pain/chemically induced , Protein Structure, Tertiary , Scorpion Venoms
...