Channelpedia

Nav1.8

Description: sodium channel, voltage-gated, type X, alpha subunit
Gene: Scn10a
Alias: nav1.8, scn10a, PN3, SNS, hPN3

Edit - History

Introduction

Nav1.8 (also known as PN3; SNS; hPN3), encoded by the gene scn10a, is a sodium, voltage-gated, type 10, alpha subunit channel. Nav1.8 is predominantly expressed in the PNS. It is involved in the action potential initiation and nociception. Mutations to the channel are often the cause of chronic pain and inflammation disorders.


Edit - History

Gene

scn10a is the coding gene for Nav1.8. In humans, it is located on chromosome 3 (3p22.2) and is made of 28 exons, 27 of which are coding and exon 1 being non-coding. scn10a is found on the same gene cluster as scn5a (Nav1.5) and scn11a (Nav1.9). All 3 genes contain an extra exon (17b) between exons 17 and 18, which corresponds to a section on the loop between domain II and domain III.

Species NCBI gene ID Chromosome Position
Human 6336 3 119410
Mouse 20264 9 110866
Rat 29571 8 112159

Edit - History

Transcript

The scn10a transcript has a length of 6.5 kilobase [859]

Species NCBI accession Length (nt)
Human NM_006514.4 6626
Mouse NM_001205321.1 6416
Rat NM_017247.2 6507

Edit - History

Protein Isoforms

Human Nav1.8 is made up of 1,956 amino acids (aa) and has a molecular weight of 220 Kda.

Species Uniprot ID Length (aa)
Human Q9Y5Y9 1956
Mouse Q6QIY3 1958
Rat Q62968 1956

Isoforms

Transcript
Length (nt)
Protein
Length (aa)
Variant
Isoform

Edit - History

Post-Translational Modifications

Nav1.8 contains a number of glycosylation sites, indicating that the protein is likely glycosylated. However, the exact timing of glycosylation and its impacts on channel kinetics or expression have not been extensively studied [860].

Nav1.8 is subject to phosphorylation by various enzymes:

  • PKA was shown to modulate Nav1.8 on phosphorylation sites, notably some located in the intracellular loop between DI-DII. Removal of these sites via mutagenesis lead to a right shift in activation and a slowing of current inactivation, indicating that phosphorylation enhances the channel’s activity [2221].
  • p38 MAPK phosphorylates Nav1.8 in loop 1 (L1) of the channel, leading to increases in current density but does not alter gating properties [2222].
  • A number of cAMP-dependent phosphorylation sites were also identified in Nav1.8: one in domain II between S3 and S4 and another in the interdomain II-III [860].

These phosphorylation events do not only change the activity and gating of the protein but also alter its expression. Often times, phosphorylation of the protein increases its trafficking to the cell membrane, in turn making the neuron more excitable. [2128]

Nav1.8 is subject to ubiquitination as it possesses a PY motif and was shown to be negatively regulated by Nedd4-2. This regulation was further demonstrated by experiments that over- and under-expressed Nedd4-2, leading to respective increase and decrease densities of Nav1.8 and neuron excitability. [2128]

PTM
Position
Type

Edit - History

Structure

Nav1.8
Visual Representation of Nav1.8 Structure
Methodology for visual representation of structure available here

Like all voltage gated sodium channels, Nav1.8 is made up of a single protein comprised of 4 homologous domains (DI-DIV). Each domain is made up of 6 transmembrane subunits (S1-S6). S1-4 form the voltage sensing domain (VSD) whereas the S5-6 form the pore module (PM). The S4 subunit of each domain contains a series of positively charged residues. When membrane depolarization occurs, these charged residues cause the movement of the S4 subunit, inducing a conformational change in S5-S6, opening of the channel and allowing the entry of sodium ions into the cell. Soon after opening, rapid inactivation of Nav1.8 is instigated by the binding of the IFM motif, found in the loop between D3 and D4, to a hydrophobic receptor site next to the S6 in D4. This binding causes the shift of S6, allosterically closing the channel, thus deactivating the channel. Nav1.8 then returns to its resting state following the hyperpolarization of the cell membrane [2115].

The structure of human Nav1.8, in complex with its selective blocker A-803467, was resolved via cryo-electron microscopy, giving us a detailed insight to the specific structural features of Nav1.8. Structural resolution of the ion channel showed that only the extracellular interface of Domain I and Domain II-S5 have Nav1.8 unique residues that may be the main contributors to the channel’s unique kinetics (see Kinetics section) [2223].
Nav1.8’s resistance to tetrodotoxin is conferred by specific residues in the S5-S6 linker. Indeed, aromatic amino acid substitutions in this area make the channel TTX-sensitive. [2128] [860].

Nav1.8 predicted AlphaFold size

Species Area (Å2) Reference
Human 10685.97 source
Mouse 11363.14 source
Rat 10753.58 source

Methodology for AlphaFold size prediction and disclaimer are available here


Edit - History

Kinetics

The electrophysiological activity of Nav1.8 is somewhat similar to that of other tetrodotoxin-resistant sodium channels [859].
However, the channel has distinctively slow gating kinetics. Nav1.8 exhibits slow-inactivating kinetics with a high threshold for activation, which generates a large inward current [1430]. These kinetics properties make Nav1.8 is a major contributor to the rising phase on the action potential [2224].


Edit - History

Biophysics

Single Channel Unitary Conductance

Single channel unitary conductance is determined experimentally.
For Nav1.8, single channel unitary conductance has yet to be determined experimentally.

Model

A single kinetic model for all human voltage-gated sodium channels (Balbi et al, 2017)
https://modeldb.science/230137
Species : Human   |   Gene: scn10a
Host cell: DRG neuron   |   Temperature: RT (to 25 C by Q10)
Formalism: Markov   |   States: C1, C2, O1, O2, I1, I2
Implementation: NEURON   |   Simulation: Nav18_a.mod
Nav1.8 Balbi 2017


Edit - History

Expression and Distribution

Tissue and Cellular

Nav1.8 is predominantly expressed in sensory neurons of the peripheral nervous system (PNS). More specifically, it is almost exclusively located in most peptidergic and non peptidergic small dorsal root ganglia (DRG). In situ hybridization using Nav1.8-specific oligonucleotide probes showed, that in the PNS, approximately 76% of the small neuronal cell population (400-1000 μm2) and 33% of the large cell population (1400-2000 μm2) were hybridized with probes for Nav1.8 [860]. It has also been detected, to a much lesser extent in the sciatic nerve, trigeminal ganglion, and nodose ganglia. [2225] [860] [2226].

Developmental

Nav1.8 is primarily expressed in adult cells. However, the channel’s expression starts in the embryo, at day 15, with expression then increasing to hit adult levels by postnatal day 7 [2124].


Edit - History

CNS Sub-cellular Distribution

Nav1.8 is present at the unmyelinated nerve endings within sensory neurons [2227].


Edit - History

Function

Action potential initiation

Given the channel’s kinetic properties, Nav1.8 is the major contributor to the upstroke of the action potential (AP) in sensory neurons, accounting for 80–90% of the inward current flow during the AP upstroke.

Nociception

Given its location, Nav1.8 is essential for AP firing in sensory neurons, allowing for the transmission of sensory information, particularly in sensitized nociceptors [1434]. Mouse models showed that Nav1.8-null mice had weak pain responses compared to their wild-type littermates, when subjected to stimulations that triggered a response from sensitized nociceptors [2228] [1436].

Indeed, Nav1.8’s role in nociception is further highlighted by its increased excitability to cold temperatures. Contrary to other sodium channels, whose activity decreases with lower temperatures, Nav1.8’s inactivation properties are cold-resistant, enabling the channel to fire at low temperatures and relay the information to the central nervous system. This property of Nav1.8 represents an important means of protection, in warm blooded animals, to help them detect and avoid tissue damaging at low temperatures [1430]

Pain & other channelopathies

However, as Nav1.8 is involved in nociception, deregulation of the channel often leads to issues with chronic pain and inflammation.

Several studies suggest that Nav1.8 plays an important role in inflammatory hyperexcitability as the channel’s expression is upregulated following certain noxious stimuli (intracolonic capsaicin or mustard oil). Nav1.8 knockout mice do not present the same response to these inflammatory compounds [1434] Intraplantar injection of complete Freund's adjuvant (CFA) experiments in rats also showed an increase in Nav1.8 expression and its accumulation at the sites of nerve injury in human patients with chronic neuropathic pain and chronic local hyperalgesia [2229]

Hyperexcitability of the channel is responsible for increased pain in conditions such as painful small fiber neuropathy (SFN). Two functional variants of Nav1.8, one that enhances ramp current and another shifting activation in a hyperpolarizing direction, render DRG neurons hyperexcitable in idiopathic SFN patients [2212] [2224]

Mutant Nav1.8 channels are also responsible for a number of other channelopathies:

  • Human hypertrophied and failing ventricles [2230]
  • Pitt-Hopkins Syndrome [2231]
  • Brugada Syndrome [[[2232]]

Edit - History

Interaction

Nav1.8 channels interact with various commercially available compounds such as menthol, lidocaine, tetracaine, vinpocetine, ambroxol, lamotrigine, mexilitine, veratridine, and A-803467, whereas very few animal toxins have been shown to be capable of reshaping Nav1.8 currents [1429]. For example, lidocaine, suppresses Na+ currents by binding not only to DIV–S6 but also to S6 of DI and DII, blocking the channels in a use-dependent (frequency-dependent) and voltage-dependent manner [856], [857]. Lidocaine enhanced current decrease in a frequency-dependent manner. Steady-state inactivation of Nav1.8 and Nav1.7 channels was also affected by lidocaine, Nav1.7 being the most sensitive. Only the steady-state activation of Nav1.8 was affected while the entry of both channels into slow inactivation was affected by lidocaine, Nav1.8 being affected to a larger degree. [217]

Beta subunits [220]

Nav1.8 is likely to be co-expressed with the b1, b2, and b3-subunits in C-fiber neurons from the DRG, and these subunits interact and modulate the channel.

  • b1-subunit alone, or in combination with other b-subunits, accelerates the time constant of inactivation and shifts the voltage-dependence of activation to more hyperpolarized potentials. Beta-1 also increases levels of Nav1.8 channel functional expression, either by increasing channel density at the oocyte membrane or by increasing individual channel conductance.

  • b3-subunit shifts the voltage-dependence of inactivation to more positive potentials and do not influence the time constant of current decay, steadystate activation or expression of Nav1.8. Co-expression of b1 and b3-subunits with Nav1.8 resulted in little change in availability compared to channel alone.

  • b2-subunit do not alter the kinetic properties or current amplitude of However, the co-expression of b1 and b2 shift the Nav1.8 channel availability to more depolarized potentials in comparison to when channels are expressed with either b1 or b2 alone.

Other proteins that interact with Nav1.8 [1431]

p11 directly binds only to NaV1.8 and translocate it to the plasma membrane.

PDZD2 and syntrophin-associated serine/threonine kinase (SASTK) are linked to the functional expression of NaV1.8 on the plasma membrane.

Contactin regulates the surface expression of Nav1.8.

CAP-1A binds to a conserved motif present in NaV1.8 linking voltage-gated sodium channels to clathrin, which is involved in coated vesicle assembly.

Tetrodotoxin

Nav1.8 is TTX resistant [1376]


  • Known and predicted drug interactions with Nav1.8
  • Known and predicted animal toxin interactions with Nav1.8

References

219

Browne LE et al. Functional and pharmacological properties of human and rat NaV1.8 channels.
Neuropharmacology, 2009 Apr , 56 (905-14).

220

Vijayaragavan K et al. Role of auxiliary beta1-, beta2-, and beta3-subunits and their interaction with Na(v)1.8 voltage-gated sodium channel.
Biochem. Biophys. Res. Commun., 2004 Jun 25 , 319 (531-40).

855

Vijayaragavan K et al. Gating properties of Na(v)1.7 and Na(v)1.8 peripheral nerve sodium channels.
J. Neurosci., 2001 Oct 15 , 21 (7909-18).

856

Ragsdale DS et al. Molecular determinants of state-dependent block of Na+ channels by local anesthetics.
Science, 1994 Sep 16 , 265 (1724-8).

858

859

Akopian AN et al. A tetrodotoxin-resistant voltage-gated sodium channel expressed by sensory neurons.
Nature, 1996 Jan 18 , 379 (257-62).

865

Renganathan M et al. Contribution of Na(v)1.8 sodium channels to action potential electrogenesis in DRG neurons.
J. Neurophysiol., 2001 Aug , 86 (629-40).

Nassar MA et al. Nociceptor-specific gene deletion reveals a major role for Nav1.7 (PN1) in acute and inflammatory pain.
Proc. Natl. Acad. Sci. U.S.A., 2004 Aug 24 , 101 (12706-11).

Gilchrist J et al. Animal toxins can alter the function of Nav1.8 and Nav1.9.
Toxins (Basel), 2012 Aug , 4 (620-32).

Zimmermann K et al. Sensory neuron sodium channel Nav1.8 is essential for pain at low temperatures.
Nature, 2007 Jun 14 , 447 (855-8).

Swanwick RS et al. The trafficking of Na(V)1.8.
Neurosci. Lett., 2010 Dec 10 , 486 (78-83).

Faber CG et al. Gain-of-function Nav1.8 mutations in painful neuropathy.
Proc. Natl. Acad. Sci. U.S.A., 2012 Nov 20 , 109 (19444-9).

Abrahamsen B et al. The cell and molecular basis of mechanical, cold, and inflammatory pain.
Science, 2008 Aug 1 , 321 (702-5).

Black JA et al. Molecular identities of two tetrodotoxin-resistant sodium channels in corneal axons.
Exp. Eye Res., 2002 Aug , 75 (193-9).

Widmark J et al. Differential evolution of voltage-gated sodium channels in tetrapods and teleost fishes.
Mol. Biol. Evol., 2011 Jan , 28 (859-71).

Fitzgerald EM et al. cAMP-dependent phosphorylation of the tetrodotoxin-resistant voltage-dependent sodium channel SNS.
J. Physiol. (Lond.), 1999 Apr 15 , 516 ( Pt 2) (433-46).

Huang X et al. Structural basis for high-voltage activation and subtype-specific inhibition of human Nav1.8.
Proc Natl Acad Sci U S A, 2022Jul26, 119 (e2208211119).

Zerres K et al. Spinal muscular atrophy--clinical and genetic correlations.
Neuromuscul Disord, 1997May, 7 (202-7).

Fukuyama M et al. Novel SCN10A variants associated with Brugada syndrome.
Europace, 2016Jun, 18 (905-11).

Brock JA et al. Tetrodotoxin-resistant impulses in single nociceptor nerve terminals in guinea-pig cornea.
J. Physiol. (Lond.), 1998 Oct 1 , 512 ( Pt 1) (211-7).

Laird JM et al. Deficits in visceral pain and referred hyperalgesia in Nav1.8 (SNS/PN3)-null mice.
J. Neurosci., 2002 Oct 1 , 22 (8352-6).


Edit

Credits

To cite this page: [Contributors] Channelpedia https://channelpedia.epfl.ch/wikipages/127/ , accessed on 2024 Apr 27