Phenotypic Mutation 'Tremord' (pdf version)
AlleleTremord
Mutation Type missense
Chromosome15
Coordinate100,911,385 bp (GRCm39)
Base Change G ⇒ T (forward strand)
Gene Scn8a
Gene Name sodium channel, voltage-gated, type VIII, alpha
Synonym(s) nmf335, nmf58, NMF335, C630029C19Rik, nur14, mnd2, seal, mnd-2, nmf2, med, ataxia 3, NaCh6, Nav1.6, motor end-plate disease
Chromosomal Location 100,767,739-100,943,819 bp (+) (GRCm39)
MGI Phenotype FUNCTION: [Summary is not available for the mouse gene. This summary is for the human ortholog.] This gene encodes a member of the sodium channel alpha subunit gene family. The encoded protein forms the ion pore region of the voltage-gated sodium channel. This protein is essential for the rapid membrane depolarization that occurs during the formation of the action potential in excitable neurons. Mutations in this gene are associated with mental retardation, pancerebellar atrophy and ataxia. Alternate splicing results in multiple transcript variants.[provided by RefSeq, May 2010]
PHENOTYPE: Spontaneous mutant homozygotes have ataxia, dystonia, muscular atrophy, progressive paralysis, Purkinje cell loss, in some cases severe head-tossing and for severe alleles, juvenile lethality. A mild, semidominant ENU allele causes deafness of variable penetrance and severity and mild tremor. [provided by MGI curators]
Accession Number

NCBI RefSeq: NM_001077499; MGI: 103169

MappedYes 
Amino Acid Change Tryptophan changed to Leucine
Institutional SourceBeutler Lab
Gene Model not available
AlphaFold no structure available at present
SMART Domains Protein: ENSMUSP00000080842
Gene: ENSMUSG00000023033
AA Change: W935L

DomainStartEndE-ValueType
Pfam:Ion_trans 131 422 7.4e-82 PFAM
low complexity region 423 452 N/A INTRINSIC
Pfam:Na_trans_cytopl 499 700 3.5e-72 PFAM
low complexity region 701 712 N/A INTRINSIC
Pfam:Ion_trans 750 985 2.2e-57 PFAM
Pfam:Na_trans_assoc 989 1191 2e-59 PFAM
Pfam:Ion_trans 1195 1472 6.2e-69 PFAM
Pfam:Ion_trans 1519 1775 1.2e-56 PFAM
IQ 1892 1914 1.2e-4 SMART
low complexity region 1953 1972 N/A INTRINSIC
Predicted Effect probably damaging

PolyPhen 2 Score 0.999 (Sensitivity: 0.14; Specificity: 0.99)
(Using ENSMUST00000082209)
SMART Domains Protein: ENSMUSP00000104536
Gene: ENSMUSG00000023033
AA Change: W935L

DomainStartEndE-ValueType
Pfam:Ion_trans 72 322 1.9e-76 PFAM
low complexity region 367 378 N/A INTRINSIC
Pfam:Ion_trans 451 640 1.1e-47 PFAM
Pfam:Na_trans_assoc 655 872 1.9e-71 PFAM
Pfam:Ion_trans 898 1127 4.4e-59 PFAM
PDB:1BYY|A 1129 1181 7e-30 PDB
Pfam:Ion_trans 1220 1429 1.9e-51 PFAM
Pfam:PKD_channel 1281 1436 5.6e-7 PFAM
IQ 1558 1580 1.2e-4 SMART
low complexity region 1619 1638 N/A INTRINSIC
Predicted Effect probably damaging

PolyPhen 2 Score 1.000 (Sensitivity: 0.00; Specificity: 1.00)
(Using ENSMUST00000108908)
SMART Domains Protein: ENSMUSP00000104537
Gene: ENSMUSG00000023033
AA Change: W945L

DomainStartEndE-ValueType
Pfam:Ion_trans 72 322 2.2e-76 PFAM
low complexity region 335 364 N/A INTRINSIC
Pfam:DUF3451 390 616 8.7e-70 PFAM
Pfam:Ion_trans 697 886 1.3e-47 PFAM
Pfam:Na_trans_assoc 901 1118 2.3e-71 PFAM
Pfam:Ion_trans 1144 1186 9.7e-10 PFAM
Pfam:Ion_trans 1182 1332 1.7e-31 PFAM
PDB:1BYY|A 1334 1386 2e-29 PDB
Pfam:Ion_trans 1425 1634 2.3e-51 PFAM
Pfam:PKD_channel 1486 1641 6.6e-7 PFAM
IQ 1763 1785 1.2e-4 SMART
low complexity region 1824 1843 N/A INTRINSIC
Predicted Effect
SMART Domains Protein: ENSMUSP00000104538
Gene: ENSMUSG00000023033
AA Change: W935L

DomainStartEndE-ValueType
Pfam:Ion_trans 160 410 2.5e-76 PFAM
low complexity region 423 452 N/A INTRINSIC
Pfam:DUF3451 478 704 9.6e-70 PFAM
Pfam:Ion_trans 785 974 1.4e-47 PFAM
Pfam:Na_trans_assoc 989 1206 2.5e-71 PFAM
Pfam:Ion_trans 1232 1461 5.7e-59 PFAM
PDB:1BYY|A 1463 1515 4e-29 PDB
Pfam:Ion_trans 1554 1763 2.5e-51 PFAM
Pfam:PKD_channel 1615 1770 7.1e-7 PFAM
IQ 1892 1914 1.2e-4 SMART
low complexity region 1953 1972 N/A INTRINSIC
Predicted Effect probably damaging

PolyPhen 2 Score 1.000 (Sensitivity: 0.00; Specificity: 1.00)
(Using ENSMUST00000108910)
SMART Domains Protein: ENSMUSP00000144371
Gene: ENSMUSG00000023033
AA Change: W935L

DomainStartEndE-ValueType
Pfam:Ion_trans 131 422 4.1e-80 PFAM
low complexity region 423 452 N/A INTRINSIC
Pfam:Na_trans_cytopl 499 700 2.5e-69 PFAM
low complexity region 701 712 N/A INTRINSIC
Pfam:Ion_trans 750 985 1.2e-55 PFAM
Pfam:Na_trans_assoc 989 1191 9.1e-57 PFAM
Pfam:Ion_trans 1195 1274 7.6e-16 PFAM
Pfam:Ion_trans 1270 1431 2.6e-33 PFAM
Pfam:Ion_trans 1478 1734 6.5e-55 PFAM
IQ 1851 1873 6e-7 SMART
low complexity region 1912 1931 N/A INTRINSIC
Predicted Effect probably damaging

PolyPhen 2 Score 1.000 (Sensitivity: 0.00; Specificity: 1.00)
(Using ENSMUST00000200963)
SMART Domains Protein: ENSMUSP00000143879
Gene: ENSMUSG00000023033

DomainStartEndE-ValueType
Pfam:Ion_trans 131 422 8.8e-81 PFAM
low complexity region 423 452 N/A INTRINSIC
Pfam:Na_trans_cytopl 499 700 6.7e-70 PFAM
low complexity region 701 712 N/A INTRINSIC
Pfam:Ion_trans 750 804 5.3e-9 PFAM
Predicted Effect probably benign
SMART Domains Protein: ENSMUSP00000144013
Gene: ENSMUSG00000023033
AA Change: W935L

DomainStartEndE-ValueType
Pfam:Ion_trans 131 422 7.4e-82 PFAM
low complexity region 423 452 N/A INTRINSIC
Pfam:Na_trans_cytopl 499 700 3.5e-72 PFAM
low complexity region 701 712 N/A INTRINSIC
Pfam:Ion_trans 750 985 2.2e-57 PFAM
Pfam:Na_trans_assoc 989 1191 2e-59 PFAM
Pfam:Ion_trans 1195 1472 6.2e-69 PFAM
Pfam:Ion_trans 1519 1775 1.2e-56 PFAM
IQ 1892 1914 1.2e-4 SMART
low complexity region 1953 1972 N/A INTRINSIC
Predicted Effect probably damaging

PolyPhen 2 Score 0.999 (Sensitivity: 0.14; Specificity: 0.99)
(Using ENSMUST00000201549)
Meta Mutation Damage Score Not available question?
Is this an essential gene? Probably essential (E-score: 0.800) question?
Phenotypic Category Autosomal Semidominant
Candidate Explorer Status loading ...
Single pedigree
Linkage Analysis Data
Penetrance 100% 
Alleles Listed at MGI

All alleles(27) : Targeted(2) Gene trapped(11) Transgenic(1) Spontaneous(5) Chemically induced(8)

Lab Alleles
AlleleSourceChrCoordTypePredicted EffectPPH Score
IGL00230:Scn8a APN 15 100853413 unclassified probably benign
IGL00979:Scn8a APN 15 100853287 unclassified probably benign
IGL01339:Scn8a APN 15 100930082 missense probably benign
IGL01992:Scn8a APN 15 100866938 missense probably damaging 1.00
IGL02215:Scn8a APN 15 100927453 splice site probably null
IGL02311:Scn8a APN 15 100911164 missense probably damaging 0.97
IGL02404:Scn8a APN 15 100937611 missense probably damaging 1.00
IGL02652:Scn8a APN 15 100911357 missense probably damaging 0.98
IGL02690:Scn8a APN 15 100868135 missense probably damaging 1.00
IGL02704:Scn8a APN 15 100905943 missense possibly damaging 0.94
IGL03084:Scn8a APN 15 100915053 missense probably damaging 1.00
IGL03108:Scn8a APN 15 100872496 missense probably benign
IGL03224:Scn8a APN 15 100933520 missense probably damaging 1.00
dan UTSW 15 100933505 nonsense probably null
nymph UTSW 15 100933527 missense probably damaging 1.00
3-1:Scn8a UTSW 15 100937820 missense probably benign 0.04
PIT4280001:Scn8a UTSW 15 100855370 missense probably damaging 1.00
PIT4508001:Scn8a UTSW 15 100927573 missense probably damaging 0.98
R0010:Scn8a UTSW 15 100911454 missense probably damaging 1.00
R0010:Scn8a UTSW 15 100911454 missense probably damaging 1.00
R0254:Scn8a UTSW 15 100916245 missense probably damaging 1.00
R0412:Scn8a UTSW 15 100906187 splice site probably benign
R0538:Scn8a UTSW 15 100933505 nonsense probably null
R0539:Scn8a UTSW 15 100914449 missense probably damaging 1.00
R0631:Scn8a UTSW 15 100933418 missense probably damaging 1.00
R0726:Scn8a UTSW 15 100870711 missense probably damaging 1.00
R0945:Scn8a UTSW 15 100913668 missense possibly damaging 0.54
R0967:Scn8a UTSW 15 100933527 missense probably damaging 1.00
R1164:Scn8a UTSW 15 100938043 missense probably benign 0.06
R1283:Scn8a UTSW 15 100867052 missense possibly damaging 0.82
R1368:Scn8a UTSW 15 100933422 missense probably damaging 1.00
R1633:Scn8a UTSW 15 100927696 missense probably benign 0.01
R1669:Scn8a UTSW 15 100909001 missense probably damaging 1.00
R1694:Scn8a UTSW 15 100853409 nonsense probably null
R1735:Scn8a UTSW 15 100913742 missense possibly damaging 0.94
R1773:Scn8a UTSW 15 100937496 missense probably damaging 0.97
R1940:Scn8a UTSW 15 100868085 missense probably benign 0.22
R1996:Scn8a UTSW 15 100922260 missense probably damaging 1.00
R2107:Scn8a UTSW 15 100916244 missense probably damaging 0.99
R2251:Scn8a UTSW 15 100914987 missense probably benign 0.02
R2516:Scn8a UTSW 15 100867043 missense probably benign 0.05
R2917:Scn8a UTSW 15 100937613 missense probably damaging 1.00
R3417:Scn8a UTSW 15 100869549 splice site probably benign
R3896:Scn8a UTSW 15 100933379 missense probably benign
R4024:Scn8a UTSW 15 100937674 missense probably damaging 1.00
R4050:Scn8a UTSW 15 100911294 nonsense probably null
R4193:Scn8a UTSW 15 100869484 missense probably damaging 1.00
R4212:Scn8a UTSW 15 100854954 missense possibly damaging 0.88
R4358:Scn8a UTSW 15 100838014 missense probably benign 0.00
R4396:Scn8a UTSW 15 100870711 missense probably damaging 1.00
R4428:Scn8a UTSW 15 100881784 missense probably damaging 1.00
R4452:Scn8a UTSW 15 100854972 missense possibly damaging 0.95
R4631:Scn8a UTSW 15 100914384 nonsense probably null
R4693:Scn8a UTSW 15 100913572 missense probably damaging 1.00
R4765:Scn8a UTSW 15 100938352 missense probably benign 0.07
R4777:Scn8a UTSW 15 100913832 missense probably damaging 1.00
R4949:Scn8a UTSW 15 100927663 missense probably damaging 1.00
R4997:Scn8a UTSW 15 100854935 missense probably damaging 1.00
R5246:Scn8a UTSW 15 100908938 missense probably damaging 1.00
R5566:Scn8a UTSW 15 100872415 missense probably damaging 1.00
R5875:Scn8a UTSW 15 100870703 nonsense probably null
R6031:Scn8a UTSW 15 100881865 missense probably damaging 1.00
R6031:Scn8a UTSW 15 100881865 missense probably damaging 1.00
R6057:Scn8a UTSW 15 100872548 missense possibly damaging 0.94
R6114:Scn8a UTSW 15 100938477 missense probably damaging 0.99
R6362:Scn8a UTSW 15 100837996 splice site probably null
R6535:Scn8a UTSW 15 100857588 intron probably benign
R6677:Scn8a UTSW 15 100866953 missense probably damaging 1.00
R6687:Scn8a UTSW 15 100872508 missense probably benign 0.12
R6701:Scn8a UTSW 15 100937977 missense probably damaging 1.00
R6719:Scn8a UTSW 15 100908896 critical splice acceptor site probably null
R6739:Scn8a UTSW 15 100913836 missense possibly damaging 0.82
R6769:Scn8a UTSW 15 100933445 missense probably benign
R6786:Scn8a UTSW 15 100930096 missense probably benign 0.00
R6849:Scn8a UTSW 15 100853468 splice site probably null
R7108:Scn8a UTSW 15 100937659 missense probably benign 0.01
R7215:Scn8a UTSW 15 100927711 missense possibly damaging 0.80
R7217:Scn8a UTSW 15 100868108 missense probably benign 0.00
R7219:Scn8a UTSW 15 100866984 missense probably damaging 1.00
R7356:Scn8a UTSW 15 100855460 missense probably damaging 1.00
R7479:Scn8a UTSW 15 100853358 missense probably damaging 0.99
R7816:Scn8a UTSW 15 100908917 missense possibly damaging 0.63
R7985:Scn8a UTSW 15 100914843 splice site probably null
R8112:Scn8a UTSW 15 100927718 missense probably benign 0.27
R8263:Scn8a UTSW 15 100881736 missense probably damaging 1.00
R8305:Scn8a UTSW 15 100938387 missense probably benign 0.01
R8489:Scn8a UTSW 15 100867014 missense probably damaging 1.00
R8983:Scn8a UTSW 15 100900030 missense possibly damaging 0.81
R9034:Scn8a UTSW 15 100927642 missense probably damaging 0.98
R9050:Scn8a UTSW 15 100906161 missense possibly damaging 0.80
R9240:Scn8a UTSW 15 100915068 nonsense probably null
R9249:Scn8a UTSW 15 100914456 missense probably benign 0.00
R9462:Scn8a UTSW 15 100930159 missense
R9599:Scn8a UTSW 15 100911172 missense probably damaging 1.00
R9609:Scn8a UTSW 15 100834407 missense possibly damaging 0.91
R9653:Scn8a UTSW 15 100937947 missense probably damaging 1.00
R9794:Scn8a UTSW 15 100933332 missense probably benign 0.00
X0066:Scn8a UTSW 15 100937962 missense probably damaging 1.00
X0066:Scn8a UTSW 15 100937961 missense probably damaging 1.00
Z1176:Scn8a UTSW 15 100931399 missense probably damaging 1.00
Z1177:Scn8a UTSW 15 100938103 missense probably benign 0.00
Mode of Inheritance Autosomal Semidominant
Local Stock Embryos
MMRRC Submission 030742-UCD
Last Updated 2018-05-22 9:44 AM by Anne Murray
Record Created unknown
Record Posted 2008-10-07
Phenotypic Description

TremorD was identified as a visible mutant amongst N-ethyl-N-nitrosourea (ENU)-mutagenized G3 mice, and is a recessive or weakly semidominant phenotype characterized by a tremor and a gait disturbance, both of varying severity.

Nature of Mutation
The TremorD mutation was mapped to Chromosome 15, and corresponds to a G to T transversion at position 2919 of the Scn8a transcript, in exon 16 of 27 total exons.
 
2903 GTGCTGTGTGGGGAGTGGATCGAGACCATGTGG
930  -V--L--C--G--E--W--I--E--T--M--W-
 
The mutated nucleotide is indicated in red lettering, and causes a tryptophan to leucine substitution at residue 935 of the SCN8A sodium channel.
Illustration of Mutations in
Gene & Protein
Protein Prediction

The voltage-gated sodium channels form part of the ion channel superfamily that also includes voltage-gated potassium and calcium channels.  The 1978 amino acid protein SCN8A (also known as Nav1.6) is a voltage-gated sodium channel α subunit.  The α subunit, approximately 220-260 kDa in size, is the pore-forming entity of sodium channels, and may be joined by one or more auxiliary β subunits that modify the kinetics and voltage dependence of channel gating, and regulate channel localization and interaction with cell adhesion molecules, extracellular matrix and cytoskeletal components (1-3).  Nine proteins comprise the mammalian sodium channel family, and all are greater than 50% identical in the transmembrane and extracellular domains, although the intracellular domains are not highly conserved and they display distinct, voltage-dependence as well as unique kinetic and pharmacological properties [reviewed in (4)].  The nomenclature of the channel is determined by the principal permeating ion (Na), the principal physiological regulator (voltage, indicated as the abbreviated subscript, v), the gene subfamily number (currently only 1), and a specific channel number (1-9, assigned based on the order in which they were identified).  Splice variants are identified by lowercase letters following the channel number (see record for Possum).

 
 

Figure 1. A, Domain structure of SCN8A. B, Transmembrane organization of sodium channel subunits. Cylinders represent probable α-helical transmembrane segments (numbered 1-6 for each domain) with green cylinders indicating the pore-lining segments and yellow cylinders indicating the S4 voltage sensors (positively charged residues are represented by the + signs). The extracellular immunoglobulin-like domains of the β1 and β2 subunits are shown and a site of interaction between the  α and β1 subunit is indicated by the wavy, gray lines. Consensus phosphorylation sites are indicated by the orange circles. Purple circles represent the outer (EEDD) and inner (DEKA) rings of amino acid residues forming the ion selectivity filter and the TTX binding site. White circles indicate residues critical for fast inactivation. Binding sites for α- and β-scorpion toxins and anesthetic drugs are also shown. The residue affected by the TremorD mutation is located in the pore loop between the S5 and S6 helices in domain II, and is adjacent to the glutamic acid (E) that forms part of the inner ring of the ion selectivity filter. Image is interactive; click to see another Scn8a​ mutation.

Nav1.6, like the other sodium channel α subunits, contains four homologous domains (I-IV) each consisting of six transmembrane α helices (S1-S6) (see Figure 1) (5;6).  A membrane-reentrant extracellular loop called the “pore loop” is located between the S5 and S6 helices in each of the four domains [see (4;7) and references therein for detailed information on sodium channel structure].  The pore loops line the outer, narrow entry to the pore, and each loop contributes one amino acid to both the outer and inner rings serving as the ion selectivity filter in the outer pore of the channel.  The outer ring of acidic residues is EEDD, while the inner ring is DEKA.  A single amino acid residue in the pore loop of domain I determines the affinity of different sodium channels for the pore blocker tetrodotoxin (TTX).   For example, rat cardiac sodium channels bind TTX with 200-fold lower affinity than brain or skeletal sodium channels because of the substitution of an aromatic tyrosine or phenylalanine residue with cysteine (8;9).  Nav1.6 is highly sensitive to TTX (10).  The S4 helices constitute the voltage sensors of the channel, and contain positively charged amino acid residues (arginine and/or lysine) at every third position.  Depolarization of the membrane allows the S4 segments to move outward and initiate a conformational change that opens the pore.  This gating movement is targeted by the β-scorpion toxins, which enhance sodium channel activation by shifting voltage dependence to more negative membrane potentials (11).  The short, intracellular loop between domain III, S6 and domain IV, S1 functions as the inactivation gate, which folds into the channel and blocks the pore from the inside during sustained depolarization of the membrane.  A hydrophobic triad of isoleucine, phenylalanine and methionine (IFM) within this loop is critical for fast inactivation (see Background), and glycine and proline residues flanking the IFM motif serve as hinges to allow closure of the inactivation gate.  The S5 and S6 helices line the inner, wider exit from the pore, and local anesthetics (e.g. procaine, etidocaine, lidocaine) and related antiepileptic (e.g. phenytoin) and antiarrhythmic drugs (e.g. lidocaine, quinidine) all bind from inside the cell to overlapping sites within this pore to block ionic current through the channel.  Amino acids in the S6 segments from at least three of the four domains contribute to drug binding, with residues from the S6 helix of domain IV (F1752 and Y1759) being the most important (12;13)

The most abundant mouse Nav1.6 isoform is 1978 amino acids long (10).  Human SCN8A encodes a 1980-amino acid protein that shares close to 99% sequence identity with the mouse protein.  It contains several consensus sites for phosphorylation of serine and threonine residues that are also conserved in other sodium channel family members.  Nav1.6 also contains a consensus site for tyrosine phosphorylation at residue 1151 (14).  The C-terminus of Nav1.6 has been shown to bind to calmodulin (CaM), an interaction that is important for its functional activity and inactivation kinetics (15).  Two pairs of alternatively spliced exons have been identified in Scn8A/SCN8A consistent with related sodium channels, exons 5N and 5A and exons 18N and 18A (14;16).  Exons 5N and 5A encode part of transmembrane segment S3 and all of S4 in domain I and differ by a single amino acid at residue 212 (Asp in 5A and Asn in 5N) (14).  Exons 18A and 18N encode part of transmembrane segment S3 and all of S4 in domain III, but exon 18N has a conserved in-frame stop codon that predicts the synthesis of a truncated, two-domain protein. A cDNA transcript that deletes exon 18 but maintains an open reading frame has also been discovered (16).  Also observed are alternatively spliced forms of the linker region between SI and SII in domain I.  The major form of the transcript lacks 10 amino acids (11 in humans) following residue 664 in the I-II linker (10;14)

The TremorD mutation is located in the pore loop between the S5 and S6 helices in domain II, and is adjacent to the glutamic acid (E) that forms part of the inner ring of the ion selectivity filter.

Expression/Localization

Seven of the ten channel isoforms (i.e. Nav1.1, Nav1.2, Nav1.3, Nav1.6, Nav1.7, Nav1.8 and Nav1.9) are expressed during some phase of development in neurons (2;3;17;18); three of the channels (Nav1.7, Nav1.8 and Nav1.9) are expressed primarily in the PNS (2).  Nav1.6 is one of the most abundant voltage-gated sodium channels (VGSC) in the brain (6).  RT-PCR, in situ hybridization studies and immunohistochemistry in rats, mice and humans have found Nav1.6 expression in the dendrites of output neurons of the cerebellum, cerebral cortex and hippocampus, and also in cerebellar Purkinje cells and granule cells, motor neurons of the brain stem, astrocytes, Schwann cells, and dorsal root ganglion (DRG) (6;19-22).  Nav1.6 was identified as the major sodium channel present at the nodes of Ranvier (gaps in the myelin sheath) in mature myelinated sensory, motor and central nervous system (CNS) axons (23).  Nav1.6 is also expressed in nonmyelinated axons (21).  Subcellular localization in the CNS includes dendrites, presynaptic and postsynaptic membranes (2;20;23;24)In situ hybridization has also localized Scn8a mRNA expression to the ganglion and inner nuclear layers of the adult retina (25).

Nav1.6 is not expressed in embryonic brain or spinal cord, but begins to be expressed shortly after birth (20).  The cDNA transcript expressing the alternatively spliced forms of exons 5 and 18 (5N and 18N) predominate in fetal and neonatal brain, whereas in adult brain the predominant transcript contains exons 5A and 18A.  Exon 18N is also expressed in non-neuronal tissues (16).      

Although Nav1.6 is considered to be a neuronal sodium channel, expression has also been found elsewhere including the heart and certain immune cells.  In the heart, Nav1.6 is restricted to the Z lines, regularly spaced lines that divide the myofibrils into sarcomeres (26).  Cultured microglia were shown to express the tetrodotoxin (TTX)-sensitive Nav1.1 and Nav1.6 channels as well as the TTX-resistant Nav1.5  channel; Nav1.2, Nav1.3 , Nav1.7, Nav1.8, and Nav1.9 were not detectable (2).  Nav1.6 is highly expressed in PNS and CNS neurons and is the predominant isoform in the cultured microglia (2).  Nav1.6 is also expressed in microglia and macrophages in the CNS (27).  In primary human macrophages, Nav1.6 expression occurs in a filamentous pattern that is independent of the activation state of the cell and is associated with cytoskeletal structures (28).

The Scn8a gene undergoes tissue-specific splicing by the neuronal splice factors Fox-1 and Fox-2 (24;29).  Transcripts that contain the alternative exon, 18A, encode the full-length channel and are restricted to the brain and nervous system (24).  Alternatively, transcripts that encode the other alternative exon, 18N, have an in-frame stop codon in exon 18N that encodes a truncated, inactive 2-domain protein (29).  Studies have determined that the full-length Nav1.6 protein is expressed in mouse ventricles, localized mainly in the t tubules of the myocyte (29).

Background
Figure 2. α-subunit of voltage-gated sodium channels. A, Structure of the Nav channel. Domains are numbered I through IV. The four domains form an ion selective pore in the membrane. The inactivation gate is between domains III and IV.  In a resting state, the interior of the cell is negatively charged, while the exterior is positively charged. B, Deactivated, activated, and inactivated states of channel in cross-section. Positively charged residues in the voltage sensor S4 helices are indicated by the + signs. See text for details.
VGSCs are essential for the initiation and propagation of action potentials in neurons and excitable cells by mediating the rapid influx of sodium [reviewed in (7)] (Figure 2).  Three key features characterize sodium channels: voltage-dependent activation, rapid inactivation, and selective ion conductance (30).  VGSCs typically open (“activate”) within a millisecond in response to membrane depolarization, often leading to a regenerative all or none depolarization typical of action potentials in neurons.  Once activated, VGSCs do not remain open for long, and current through VGSCs can be terminated within approximately one millisecond by a process known as “fast inactivation” mediated by the intracellular inactivation gate folding into the channel and blocking the pore (see Protein Prediction).  Recovery from inactivation occurs after the cell membrane potential is repolarized to negative membrane potentials (mediated by other ion channels), allowing sodium channels to return to the resting or closed state in which they are again ready to open in response to membrane depolarization.

Microglia are immune cells in the brain and spinal cord and are activated following an injury, a pathogenic challenge, or from signaling within the central nervous system parenchyma (2).  Microglia express several cell surface and nuclear receptors as well as ion channels that respond to bacterial products, steroids, immunoglobins, cytokines and chemokines (2). The activation of microglia results in proliferation, migration, promotion of tissue repair and phagocytosis as well as the secretion of cytokines/chemokines, reactive species and proteases (2). The chloride, potassium, hydrogen and calcium channels expressed by microglia maintain membrane potential and intracellular ion concentrations (2).  In addition to the above-mentioned ion channels, microglia also express voltage-gated sodium channels (2;27).  Sodium channels in neurons function to generate and transmit action potentials.  In contrast, their function in non-excitable cells (e.g. lymphocytes (31), astrocytes (32), metastatic cancer cells (33), osteoblasts (34), odontoblasts (35), vein endothelial cells (36), chondrocytes (37), and keratinocytes(38)) is not fully known.  Sodium channels are present in and regulate the activity of microglia and macrophages in the CNS (27).  Both of these immune cells are activated during MS and contribute to axonal degeneration [reviewed in (39)]. Nav1.6 expression is upregulated in microglia and macrophages in experimental models of MS and in acute lesions of MS patients (27).  Na+ channel blockade attenuates the inflammatory activity of these cells and reduces phagocytic activity in vitro.  Moreover, microglia from animals with mutations in Scn8a showed a significant reduction in phagocytic capacity compared to wild-type microglia.  These data suggest that Nav1.6 plays an important role in microglia activation and that Nav1.6 activity may contribute to axonal damage in MS both directly and indirectly.  Despite these data, bacterial phagocytosis by macrophages depends on Nav1.5 not Nav1.6, although colocalization of Nav1.6 with F-actin and vimentin suggests possible regulation of cytoskeleton, which is also important for macrophage function (23).  Mice with mutations in Scn8a have been reported to have abnormal ratios of T cell subsets along with reduced suppressor cell function and precocious maturation of the cytotoxic response to allogeneic cells, but it is unknown whether these defects are secondary to the neurological abnormalities present in these animals (40;41).

Following injury or infection to the CNS, activated microglia have a prominent role in phagocytosis.  To test whether Nav1.6 channels are involved in LPS-stimulated phagocytosis, the microglia from med mice (lack functional Nav1.6 channels (42)) were tested for their ability to phagocytose latex beads after LPS activation (2;27).  Following LPS stimulation, phagocytosis was reduced by 65% in microglia in the med mice (compared to wild-type cohorts) (2).  In contrast, previous reports with human macrophages and human monocytic-derived THP-1 cells  were dissimilar to the rodent studies indicating that there may be a cell and/or species specific role for sodium channels in phagocytosis (2).

LPS stimulation results in a stark increase in the secretion of cytokines from activated microglia.  Microglial cultures treated with sodium channel blockers phenytoin and TTX were assayed for changes in the secretion of cytokines after LPS stimulation.  Treatment of the cultures with phenytoin led to a 50-60% decrease in the secretion of cytokines (i.e. IL-1α, IL-1β, and TNF-α) from the LPS-stimulated microglia (2).  The levels of IL-6 and IL-10 were not significantly changed upon phenytoin treatment.  TTX treatment (provides blockade of Nav1.1 and Nav1.6, but only partial blockade of Nav1.5) led to a decrease in IL-1α, IL-1β, and TNF-α as well (2).  Taken together, these results indicate that the activity of sodium channels in microglia is essential for the release of some, but not all, cytokines after LPS stimulation.

Microglia were also examined to determine if Nav1.6 were necessary for microglial migration.   A trans-well migration assay comparing microglia cultured from wild-type and med mice found that the med microglia, unlike the wild-type, did not have an increase in the migration of activated cells (2).  These results indicate that in addition to the other roles for sodium channels in microglia function, as discussed above, the Nav1.6 channels also function in the migration of activated microglia (2).  The role of Nav1.6 in cell invasion has also been demonstrated in other cell types.  For example, the upregulation of the channel led to potentiation of metastasis of several cancer cell lines including prostate, breast, lymphoma, and cervical (2;43-45). As in other eukaryotic cells, the levels of free calcium are tightly regulated are essential for signaling in microglia (2).  The Nav1.6 channel also produces a persistent sodium current that can reverse the operation of the sodium-calcium exchanger (NCX), leading to importation of calcium into the cytoplasm (2).  It is proposed that Nav1.6 activity contributes to microglial migration by regulating the activity of the NCX (2).

In the heart, the sodium channels, including Nav1.6 and Nav1.1 are proposed to be involved in (a) excitation-contraction coupling by synchronizing the action potential in the sarcolemma of the t tubules and/or (b) the automaticity of sinoatrial nodal cells of the mouse heart (29).   After heart failure, there is a down-regulation of Nav1.1 and Nav1.6 (29).

In neurons (e.g. cerebellar Purkinje cells, dorsal root ganglia C-fibers, spinal sensory neurons, trigeminal neurons, subthalamic neurons, retinal ganglion cells, pre-frontal cortical pyramidal neurons, hippocampal CA1 neurons, globus pallidus neurons, and cerebellar granule cells), mutations of Nav1.6 lead to reductions in repetitive firing and persistent currents (21;24;46).

Studies have implicated a change in Nav1.6 expression in the dorsal root ganglia as a factor in type 2-associated diabetic neuropathic pain (47).  It is abnormal, spontaneous activity in the DRG neurons that is considered the instigator of neuropathic pain (47).  Type 2 diabetes causes a significant increase in the expression of both the Nav1.6 protein and mRNA in the DRG (47).  The role of Nav1.3 and Nav1.8 in the development of neuropathic pain has been studied, through the findings are controversial (47).

Several mouse strains containing mutations in the Scn8a gene have been identified and provide insights into the role Nav1.6 plays in neuronal function [reviewed in (48)].  Functionally null mutations include two alleles (Scn8amed and Scn8atg) that respectively result in early protein truncation and a large internal deletion as well as targeted Scn8a mutations (5;42;49).  Homozygotes for these null mutations are characterized by tremor, ataxia, early-onset progressive paralysis of the hind limbs, severe muscle atrophy, degeneration of Purkinje cells, and juvenile lethality (29).  The age of lethality at three weeks corresponds to the final stage of developmental replacement of sodium channel Nav1.2 at immature nodes of Ranvier by Nav1.6 (50).  Less severe mutations of Scn8a (Scn8amed-J and jolting or Scn8amed-jo) cause ataxia, tremor, muscle weakness, and dystonia.  The Scn8amed-J mutation is a 4bp deletion in the splice donor site of intron 3 that results in skipping of exons 2 and 3 and resulting in protein truncation in domain 1 (42).  However 12% of the transcripts are correctly spliced leading to a hypomorphic phenotype.  Homozygotes have severe muscle weakness and dystonia.  In C57BL/6J mice, this allele causes juvenile lethality because these animals are also homozygous for a mutation in the sodium channel modifier 1 (SCNM1), a zinc finger protein and a putative splice factor that causes reduction of correctly spliced Scn8a transcripts (51;52).  The Scn8amed-jo mutation occurs in the linker region between transmembrane helices S4 and S5 in domain III of Nav1.6 (53).  Homozygous animals carrying this mutation have chronic ataxia with an altered gait, tremor of the head and neck induced by attempted movement, as well as progressive loss of Purkinje cells that are known to be important for motor coordination (54).  To test the importance of cerebellar Nav1.6 function on motor coordination, Scn8a was specifically deleted in Purkinje and granule neurons.  Deletion of Scn8a in Purkinje cells resulted in mice that exhibited mild ataxia, tremor, and impaired coordination, while deletion in granule cells resulted in only mild behavioral deficits (55).  Phenotypes were exacerbated in double mutants lacking Scn8a in both Purkinje and granule cells.

Electrophysiological studies of these mutants (see Putative Mechanism) and of Nav1.6 in in vitro systems suggest that Nav1.6 is required for persistent (non-inactivating) sodium current in several classes of neurons, including some with pacemaker roles (2;10;56).  In Purkinje cells, SCN8A deficiency not only affects persistent current but also reduces bursting activity and resurgent current, an unusual current that is found in some neurons and occurs when the cells are repolarized to intermediate potentials following a strong depolarization that completely inactivates transient sodium current (46).

In humans, only one SCN8A mutation has been found to be linked with disease. In a patient with mental retardation, pancerebellar atrophy, and ataxia, a 2-bp deletion in exon 24 of SCN8A was discovered (57).  Three additional family members, who were heterozygous for this mutation, exhibited milder cognitive behavioral deficits including attention deficit hyperactivity disorder (ADHD; OMIM #143465).  In addition, preferential transmission of one allele of a single-nucleotide polymorphism in intron 21 was found in families with histories of attempted suicide (58).  These data suggest that Nav1.6 may have an important role in human behavior, and is supported by behavioral studies of mice heterozygous for a null mutation of Scn8a.  These mice exhibit greater conditioned freezing in the Pavlovian fear conditioning paradigm, more pronounced avoidance of well-lit, open environments, and more stress-induced coping behavior, but showed normal learning and memory (59).  Although mutations of SCN8A have not been associated with epilepsy, Nav1.6 may play a modifying role in this condition (60).  Mutations in the sodium channels SCN1A and SCN2A, encoding Nav1.1 and Nav1.2, are known to cause several types of epilepsy in humans (OMIM #604233, #607745, #607208), and animals heterozygous for a targeted disruption of Scn1a exhibit spontaneous seizures (61).  By contrast, mice heterozygous for Scn8a mutations are more resistant to chemically induced seizures and have ameliorated seizure severity in a Scn1a heterozygous background (60).

Increasing evidence suggests that Nav1.6 also plays a role in the disease process of multiple sclerosis (MS; OMIM #126200) [reviewed in (39)].  In MS, myelinated axons are denuded of myelin and/or degenerate.  The higher density of sodium channels, primarily Nav1.6, in the nodes of Ranvier in myelinated axons supports saltatory conduction, a process by which the action potential leaps discontinuously and rapidly from one node of Ranvier to the next.  Although the myelin lost in MS is not usually replaced in full, remissions can occur due to the redistribution of sodium channels, including Nav1.6, along the axon, allowing the restoration of action potential conductance (62;63).  In addition to becoming demyelinated, some axons degenerate in MS.  Injury and inflammation leading to increased concentrations of nitric oxide (NO) in affected neurons has a deleterious effect on mitochondria and ATP production.  This causes a failure of the Na+/K+ ATPase, which pumps Na+ out of the cytoplasm.  Sustained Na+ influx drives reverse operation of the Na+-Ca2+ exchanger, allowing deleterious amounts of Ca2+ into the axon.  Ca2+ influx triggers a host of secondary cascades, such as activation of proteases and lipases, leading to axonal injury.  Axonal degeneration may be exacerbated by the increased distribution of Nav1.6 along the axon because Nav1.6 produces a large, persistent Na+ current that could trigger the Na+-Ca2+ exchanger (64).  Sodium channel blockers such as phenytoin have a protective effect in an experimental model of MS (39).  Mice with an induced form of an inflammatory/demyelinating form of multiple sclerosis (MS) (i.e. experimental autoimmune encephalomyelitis (EAE)) have a significant upregulation of Nav1.6 within activated microglia, corresponding to an upregulation of CD11b/c and CD45 in these cells (2;27).  In these mice, there is a correlation between the severity of the EAE and amount of Nav1.6 expressed as well as the morphology of the microglia to an amoeboid-like appearance (2).  In addition, there is an upregulation in Nav1.6 in microglia within MS lesions (27).  Treatment of mice inoculated with myelin oligodendrocyte glycoprotein to induce EAE with a sodium channel blocker (i.e. phenytoin) led to a decrease in the density of CD45/CD11b/c-positive microglia, indicating that sodium channels are involved in the activation of microglia during inflammation and demyelination (2).

Putative Mechanism

Allelic mutations of Scn8a in the mouse have revealed a range of neurological disorders that can be explained by the wide distribution and unique properties of the Nav1.6 sodium channel.  In null mutations, the sciatic nerve between 17 and 23 days of age exhibits reduced conduction velocity, prolonged refractory period and widening of the non-myelinated gaps at the nodes of Ranvier (51).  These changes are accompanied by failure of transmission at the neuromuscular junction and muscle denervation (65;66).  The paralysis and death in mice with null Scn8a mutations is likely due to motor neuron dysfunction.  In mature motor neurons (as in the CNS), Nav1.6 is the major channel present at the nodes of Ranvier and is required for normal saltatory conductance.  The total sodium current of these neurons increases three fold during the first week of life in normal mice, is dependent upon Nav1.6 function, and corresponds to the increasing expression of Nav1.6 in mature nodes of Ranvier (50;67).  Death in these animals may be due to the failure of the motor neurons innervating the diaphragm muscle. 

Unlike the null alleles, Scn8amed-jo mice have normal neuromuscular junctions (66), and the primary abnormality is the inability of Purkinje cells to spontaneously generate a repetitive Na+-dependent action potential.  The Scn8amed-jo mutation occurs in the S4-S5 linker region of domain III, which is predicted to play a role in inactivation.  However, in vitro analysis suggests that the mutation alters the voltage dependence of the channel, resulting in a depolarizing shift.  The mutant channel is more stable in the closed state than the wild-type channel and requires a stronger depolarization across the cell to elicit opening (53).  This defect results in reduction in Purkinje cell firing.  As the effects of Purkinje cell discharge are inhibitory, a decrease in the inhibitory signal from the cerebellum results in loss of control of rapid movements and poor correlation between posture and movement.  The results of targeted deletions of Nav1.6 in Purkinje cells support this hypothesis as the loss of Nav1.6 specifically from these cells results in decreased rates of Purkinje cell firing (55).  The differences in phenotypes between null Scn8a alleles and hypomorphic alleles suggest that in motor neurons Nav1.6 functions primarily in the propagation of action potentials, whereas in the cerebellum it is also required for initiation of action potentials.  Thus, the motor neurons in jolting mice are less affected by the small change in voltage dependence of Nav1.6, whereas the higher-voltage activation threshold of the mutant channel may reduce the chances of action potential initiation in Purkinje cells in response to low-amplitude inputs from the mossy fiber/granule cell system.

Alleles of Scn8a are considered recessive although mild phenotypes are occasionally reported in heterozygotes (40).  Haploinsufficiency of Scn8a is generally thought to be the mechanism behind the occasional phenotypes seen in heterozygotes.  These data are consistent with the phenotypes of TremorD mice, which are similar to the phenotypes seen in Scn8amed-jo homozygotes.  As the Scn8amed-jo mutation affects the voltage gating of the channel, TremorD may similarly shift the voltage dependence of the sodium channel.  Indeed, the TremorD mutation occurs in the pore loop, which is known to be essential both for ion selectivity and voltage gating (68).  The importance of this region in the function of Nav1.6 is supported by the discovery of three ENU missense mutations occurring in the pore loop of domain 3.  Two of these mutations cause phenotypes similar to those seen in null alleles of Scn8a, while the third causes a less severe movement disorder and sustained dystonic postures (69).  The two mutations causing the more severe phenotypes are located in the extracellular region of the loop and contribute to the structure of the outer pore, while the non-lethal mutation is located within the membrane.  These data suggest that the pore loop residues present within the membrane portions have less effect on the function of the sodium channel, and support evidence that regions located immediately outside the membrane are essential for maintaining overall pore structure (70).  The residue altered by the TremorD is adjacent to the glutamic acid (E) that forms part of the inner ring of the ion selectivity filter (see Protein Prediction) (71).

Primers Primers cannot be located by automatic search.
Genotyping
TremorD genotyping is performed by amplifying the region containing the mutation using PCR, followed by sequencing of the amplified region to detect the single nucleotide change.  
 
Primers for PCR amplification
TremD(F): 5’- ACAGTCTGCGACACGATCAGGAAG -3’
TremD(R): 5’- CCTGCTCTCAGGAGCACAAGTTAC -3’
 
PCR program (use SIGMA JumpStart REDTaq)
1) 94°C             2:00
2) 94°C             0:30
3) 56°C             0:30
4) 72°C             1:00
5) repeat steps (2-4) 29X
6) 72°C             7:00
7) 4°C               ∞
 
Primers for sequencing
TremD_seq(F): 5’- CAAGATCATTGGGAACTCCGTTG -3’
TremD_seq(R): 5’- GGAGCACAAGTTACCCACTG -3’
 
The following sequence of 673 nucleotides (from Genbank genomic region NC_000081 for linear DNA sequence of Scn8a) is amplified:
 
142405                           acagtc tgcgacacga tcaggaagtt ctgagaattt
142441 cctaaggatg gatgataaat aaatttaagc caatctttct aattctgcat ttttttttat
142501 ttcagcaatg tggggtagca gattatcatt tcatatttta ttatttgtct ccttagctcc
142561 gagtcttcaa attggccaag tcctggccca ccctgaacat gctgatcaag atcattggga
142621 actccgttgg cgccctgggc aacctgaccc tggtgctggc catcattgtc ttcatctttg
142681 ccgtggtggg gatgcagctc tttggaaaga gctacaagga gtgcgtctgt aagatcagcc
142741 aggagtgcaa gctcccgcgc tggcacatga acgacttctt ccactccttc ctcatcgtct
142801 tccgagtgct gtgtggggag tggatcgaga ccatgtggga ttgcatggaa gtggccggcc
142861 aggccatgtg ccttatcgtg ttcatgatgg tcatggtcat cggcaacctg gtggttagta
142921 ccaacgtctt cttctccacc accttctctg gtgaactctt tcctggccca gatctgtgtt
142981 aaggcccatc agctaccatc tcccaaatct caatgaccag aaatgtccgc agactttgcc
143041 acgtgttcag tgggtaactt gtgctcctga gagcagg
 
PCR primer binding sites are underlined; sequencing primer binding sites are highlighted in gray; the mutated G is shown in red text.
References
Science Writers Nora G. Smart, Anne Murray
Illustrators Diantha La Vine
AuthorsHolly Timms, Bruce Beutler
Edit History
2011-01-07 9:56 AM (current)
2010-10-12 11:28 AM
2010-10-12 11:26 AM
2010-02-03 10:44 AM