Skip to content
Publicly Available Published by De Gruyter October 1, 2014

Exonic mutations in SCN9A (NaV1.7) are found in a minority of patients with erythromelalgia

  • Zhiping Zhang , Martin Schmelz , Märta Segerdahl , Hans Quiding , Carina Centerholt , Anna Juréus , Thomas Hedley Carr , Jessica Whiteley , Hugh Salter , Mari Skylstad Kvernebo , Kristin Ørstavik , Tormod Helås , Inge-Petter Kleggetveit , Lars Kristian Lunden and Ellen Jørum EMAIL logo

Abstract

Background and aim

“Gain-of-function” mutations in voltage-gated sodium channel NaV1.7 have been linked to erythromelalgia (EM), characterized by painful hot and red hands and feet. We investigated the proportion of patients with EM that carry a mutation in NaV1.7 or in other pain-related genes and studied possible clinical differences.

Methods

In this study, 48 patients with EM were screened for mutations in a total of 29 candidate genes, including all sodium channel subunits, transient receptor potential channels (TRPA1, TRPV1, TRPM8), neurotrophic factors (NGF, NGFR, BDNF, GDNF, NTRK1 and WNK1) and other known pain-related genes (CACNG2, KCNS1, COMT, P2RX3, TAC1, TACR1), using a combination of next generation sequencing and classical Sanger sequencing.

Results

In 7/48 patients protein-modifying mutations of NaV1.7 (P187L, I228M, I848T (n = 4) and N1245S) were identified. Patients with the I848T mutation could be identified clinically based on early onset and severity of the disease. In contrast, there were no clinical characteristics that differentiated the other patients with NaV1.7 mutation from those patients without. We also found more than twenty rare protein-modifying genetic variants in the genes coding for sodium channels (NaV1.8, NaV1.9, NaV1.6, NaV1.5, NaV2.1, SCN1B, SCN3B), transient receptor potential channel (TRPA1, TRPV1), and other pain-related targets (WNK1 and NGFR).

Conclusion

We conclude that functionally characterized mutations of NaV1.7 (I848T) are present only in a minority of patient with EM. Albeit the majority of patients (27/48) carried rare protein-modifying mutations the vast majority of those will most probably not be causally linked to their disease.

Implications

The key question remaining to be solved is the possible role of rare variants of NaV1.8, NaV1.9, or beta-subunits in provoking chronic pain conditions or even EM.

1 Introduction

Erythromelalgia (EM) is a condition characterized by episodes of burning pain, warmth and redness in various parts of the body, particularly in the hands and feet. The attacks are periodic and are commonly triggered by heat, pressure, mild activity, exertion, insomnia or stress and local cooling relieves the pain. Ingesting alcohol or spicy foods may also trigger an episode. These symptoms are often symmetric and affect the lower extremities more frequently than the upper extremities. As the disease progresses with age, the affected areas can be constantly red and extend further up [13,53]. Erythromelalgia can occur either as a primary or secondary disorder (i.e. a disorder in and of itself or a symptom of another condition).

Primary erythromelalgia was the first human disorder linked to a mutation in the sodium channel NaV1.7 [11,56]. The sodium NaV1.7 channel alpha subunit is encoded by the gene SCN9A and is highly expressed in primary nociceptors which are involved in transmitting pain signals. Almost all of the mutations in NaV1.7, which were identified in sporadic and familial cases of EM, are missense mutations leading to gain of function [21]. Whole-cell voltage-clamp studies of the mutant NaV1.7 channel in mammalian cells have shown that all identified mutations mainly cause a lowering of the threshold for activation, and increase the ramp response of the channel [15]. As functionally distinct mutations of NaV1.7 result in different clinical entities [15,29], one might expect an exclusive link between EM and gain of function mutations of NaV1.7 leading to reduced activation thresholds.

However, only a minority of sporadic cases of EM are found to harbour mutations in the SCN9A gene [17,33] and even in familial cases of EM coding mutations in the SCN9A gene could be excluded suggesting that either mutations in non-coding regions other targets might cause EM [4,14,17]. Thus, many cases are likely to be caused by other as-yet unidentified genetic defects or to have non-genetic origin. In 6 sporadic and 9 familial cases 1 patient with the I848T mutation was identified, but no mutation of NaV1.8 or NaV1.9 [17]. More recently, one of 13 patients with EM was found with a new variant of SCN9A (Q10K) [33]. We expanded this approach to clarify the proportion of SCN9A mutations among erythromelalgia patients and to find evidence for a potential role of mutations in genes other than SCN9A in EM. We therefore set out to assess mutations of NaV1.7 in a large cohort of erythromelalgia patients and additionally screen all the sodium channels, including a and β subunits, transient receptor potential channels (TRPA1, TRPV1, TRPM8), neurotrophic factors (NGF, NGFR, BDNF, GDNF, NTRK1), and several known pain-related genes (CACNG2, KCNS1, COMT, P2RX3, TAC1, TACR1, WNK1).

2 Materials and methods

2.1 Patients’ inclusion/exclusion criteria

Over a period of five years, 350 patients were referred to the Section of Clinical Neurophysiology, Oslo University Hospital for neurophysiological evaluation of distal pain in their extremities (possible EM). The patients underwent further neurological and basic neurophysiological examinations, including QST, EMG, and neurography. The diagnosis of EM was based on the presence of episodic pain in the extremities, either occuring spontaneously or induced by warming, standing or exercise, warm and red feet during attacks and pain alleviated by cold. Patients were also included as having EM-like symptoms with either ongoing or episodic pain and with either red or warm feet. All patients were above 18 years and were asked about all previous and current diseases, the use of medication as well as characteristics of pain, and investigated clinically. The study was approved by the regional Ethical committee and all patients signed informed consents. Forty-eight patients fulfilled the clinical criteria for classical primary EM or EM-like symptoms. 37 out of 48 patients were from independent families and 11 patients originated from 5 small nuclear families, and were mother-daughter or brother-sister.

2.1.1 EMG/neurography

A keypoint EMG apparatus (Sweden) was employed, measuring motor amplitude, distal latency and conduction velocity of the median, ulnar, peroneal and posterior tibial nerves and sensory amplitude and conduction velocity of the median, ulnar, sural and superficial peroneal nerves bilaterally. EMG of appropriate muscles was performed, both assessing the properties of the motor unit potential as well as searching for possible signs of denervation.

2.2 Quantitative sensory testing

Threshold temperatures for the sensation of warmth, cold, heat pain and cold pain were determined using a computerized Ther-motest (Somedic A/B, Sweden).

2.3 Polymerase chain reaction (PCR) and Sanger-sequencing

After having obtained ethics approval and informed consent, peripheral blood samples (10 ml) were taken from all the individuals and genomic DNA was isolated for genetic analysis. DNA from 96 healthy controls was from Cheshire County in UK.

PCR primers were designed using Oligo 5.0 software and amplified PCR fragments ranged from 300 to 900 bp in length covering the coding exons including the known splicing variants and at least 50 bp of exon-intron boundaries. Sanger sequencing data was collected using standard methods on an ABI3130xl sequencer, and sequence traces were analyzed after assembly using DNA Star v5.

2.4 Genomic capture and high throughput sequencing (next generation sequencing)

Sequencing of selected gene regions (Table 1) was performed using a custom SureSelect XT solution phase target enrichment kit (Agilent Technologies, Inc.), followed by sequencing via a HiSeq2000 (Illumina, Inc.). Approximately three micrograms of high MW DNA from each sample was prepared, captured and enriched according to the manufacturer’s protocols. Selected material was amplified to incorporate relevant Illumina adapter and index sequences and samples pooled and run as sets of 12 per HiSeq2000 channel in 100bp paired end mode and the resulting data demultiplexed by index into sample-specific read sets (FastQ files). All DNA capture, preparation and sequencing steps were performed by a 3rd party sequence service provider (Source: Bioscience, Nottingham, UK). Data analysis (mapping and variant calling) was primarily performed using CLCBio Genomics Workbench v4.6 (CLCBio, Aarhus, Denmark). Additional assessment of read set quality was performed using FastQC.

Table 1

List of genes analyzed in this study.

Gene Synonym Description Ensemble gene IDs[a]
SCN1A NaV1.1 Sodium channel, voltage-gated, type I, alpha subunit ENSG00000144285
SCN2A NaV1.2 Sodium channel, voltage-gated, type II, alpha subunit ENSG00000136531
SCN3A NaV1.3 Sodium channel, voltage-gated, type III, alpha subunit ENSG00000153253
SCN4A NaV1.4 Sodium channel, voltage-gated, type IV, alpha subunit ENSG00000007314
SCN5A NaV1.5 Sodium channel, voltage-gated, type V, alpha subunit ENSG00000183873
SCN8A NaV1.6 Sodium channel protein type VIII subunit alpha isoform 1 ENSG00000196876
SCN9A NaV1.7 Sodium channel, voltage-gated, type IX, alpha subunit ENSG00000169432
SCN10A NaV1.8 Sodium channel, voltage-gated, type X, alpha subunit ENSG00000185313
SCN11A NaV1.9 Sodium channel, voltage-gated, type XI, alpha subunit ENSG00000168356
SCN7A NaV2.1 Sodium channel, voltage-gated, type VII, alpha subunit ENSG00000136546
SCN1B NaV1B Sodium channel, voltage-gated, type I, beta subunit ENSG00000105711
SCN2B NaV2B Sodium channel, voltage-gated, type II, beta subunit ENSG00000149575
SCN3B Nav3B Sodium channel, voltage-gated, type III, beta subunit ENSG00000166275
SCN4B Nav4B Sodium channel, voltage-gated, type IV, beta subunit ENSG00000177098
TRPA1 ANKTM1 Transient receptor potential cation channel subfamily A member 1 ENSG00000104321
TRPV1 VR1 Transient receptor potential cation channel subfamily V member 1 ENSG00000196689
TRPM8 Transient receptor potential cation channel subfamily M member 8 ENSG00000144481
WNK1 HSN2 WNK lysine deficient protein kinase 1 ENSG00000060237
NGF NGFB Nerve growth factor (beta polypeptide) ENSG00000134259
NGFR p75 (NTR) Nerve growth factor receptor ENSG00000064300
BDNF Brain-derived neurotrophic factor ENSG00000176697
GDNF Glial cell derived neurotrophic factor ENSG00000168621
NTRK1 TRKA Neurotrophic tyrosine kinase, receptor, type 1 ENSG00000198400
COMT Catechol O-methyltransferase isoform ENSG00000093010
CACNG2 CACGN2 Calcium channel, voltage-dependent, gamma subunit 2 ENSG00000166862
P2RX3 P2X3 Purinergic receptor P2X, ligand-gated ion channel, 3 ENSG00000109991
TAC1 NK2 Tachykinin, precursor 1 ENSG00000006128
TACR1 NK1R Tachykinin receptor 1 ENSG00000115353
KCNS1 KV9.1 K+ voltage-gated channel, delayed-rectifier, subfamily S, member 1 ENSG00000124134

3 Results

3.1 Genetics

In total, 29 genes were screened for mutation in this study including all the a subunits (SCN1A, 2A, 3A, 4A, 5A, 7A, 8A, 9A, 10A and 11A) and β subunits (SCN1B, 2B, 3B and 4B) of sodium channels, transient receptor potential channels (TRPA1, TRPV1, TRPM8), neurotrophic factors (NGF, NGFR, BDNF, GDNF, NTRK1) and other known pain-related genes (CACNG2, KCNS1, COMT, P2RX3, TAC1, TACR1 and WNK1) using next generation sequencing technique combined with classical Sanger sequencing (Table 1). Beside the mutations in NaV 1.7, more than twenty rare genetic variants were identified in the genes coding for sodium channels (NaV1.8, NaV1.9, NaV1.6, NaV1.5, NaV2.1, SCN1B, SCN3B), transient receptor potential channel (TRPA1, TRPV1), and for WNK1 and NGFR (Table 2A). No mutation was found in the genes coding for NaV1.1, NaV1.2, NaV1.3, NaV1.4, SCN2B, SCN4B, NGF, BDNF, GDNF, NTRK1, TRPM8, P2RX3, CACNG2, KCNS1, COMT, TAC1 and TACR1. All of the rare genetic variants identified in this study were further confirmed individually by classical Sanger sequencing and additionally, 96 healthy controls were screened routinely for all the variants.

Table 2A

A Rare protein modifying genetic variants in patients with erythromelalgia-like symptoms are described. Novel variants are indicated with bold print.

NaV1.7 NaV1.8 NaV1.9 NaV1.5 NaV1.6 SCN1B SCN3B NaV2.1 TRPA1 TRPV1
P187L E285K N1169S T220I I1583T R125C L10P V133I N460S F305S
I228M M650K I1293V P648L Q236P R343C (2) Q498X
I848T (4) L741V S1103Y D803V C608X
N1245S V7631 P2006Y S1154Y
11599V

The locations of the mutations in the various proteins are shown in Fig. 1. Among the α-subunits of voltage dependent sodium channels we found four mutations (P187L, I228M, I848T, N1245S) in NaV1.7 (SCN9A) in 7 out of 48 patients with EM-like symptoms (Table 2B). The most common mutation associated with EM, I848T, is also common in our patients. Four patients who are from two independent families (mother and daughter) are heterozygous for the mutation. P187L is located in the extracellular loop between DIS1 and DIS2 and N1245S in the cytoplasmic loop between DIIIS2 and DIIIS3 (Fig. 1).

Table 2B

Patients characteristics.

Sex Age Family Pain attacks Ongoing pain Character of pain Red feet Warm Relief by feet Eliciting/worsening cooling Concomitant factors Mutated diseases Mutation protein
1 m 67 Yes x B, A, S x x x Warmth Phys. Act Connective tissue disease NaV1.7 N1245S
2 f 35 Yes x x B, Cut x x x Walking, stress Red wine No NaV1.7 I848T
3 f 57 Yes x x B x x x Warmth No NaV1.7 I848T
4 f 45 Yes x B, A x x x Warmth, stress Phys. Act., Alc No NaV1.7 I848T
5 f 21 Yes x x B, S, A x x x Phys. Act No NaV1.7 I848T
6 f 71 Yes x B, A x x x Warmth No NaV1.7 P187L
7 f 62 No x B, S x x ? Phys. Act Borreliosis (?) NaV1.7 I228M
8 f 63 Yes x B, P, S, I x x Warmth No Na V 2.1 Q236P
9 f 55 Yes x B, S, I x x x Warmth, stress Alcohol Fibromyalgia Joint pain NaV2.1 NaV1.8 V133I V763I
10f 55 No x x x x None No NdV2.1 D803V
11 f 42 Yes x B x Warmth Conn.tissue dis (?) Na V 2.1 I1599V
12 f 60 Yes x B, S, Sore x x x Phys. Act No Na V 2.1 TRPA1 WNKl S1154Y N460S S2208N
13 f 45 No x B, S, Sore x x x Warmth, stress Phys. Act No NaV1.6 I1583T
14 f 67 Yes x A x x None Fibromyalgia (?) NaVl.5 T220I/
15 f 25 No x B x x x Warmth Pressure Polyc. ovar. syndr.(?) NaV1.5 P648L
16 f 64 No x x B x x x Warmth Phys. Act No NaV1.5 S1103Y
17 f 47 No x B, Sore x x No NaV1.5 P2006A
18 f 40 Yes x B, S, P x Warmth, stress Wine no SCN1B R125C
19 f 40 No x B x x x Phys. Act No SCN3B L10P
20 f 35 Yes x B x Warmth, Alc. Phys. Act No TRPA1 R343C
21 f 61 Yes x B x Alcohol No TRPA1 R343C
22 f 38 No x B x x x None No TRPA1 TRPV1 C608X Q498X
23 f 52 Yes x B x x Warmth Phys. Act No NaV1.8 M650K
24 f 54 ? x C, A, shoot x x x ? NaV1.8 TRPV1 L741V F305S
25 f 79 Yes x B, Sore x x x Pressure No NaV1.8 WNK1 E285K T764A
26 f 63 No x B, P, S x x x Phys. Act No NaV1.9 N1169S
27 f 63 No x B x h x h x None No NaV1.9 I1293V
28 m 74 No x A, I x x x None Vasculitis (?) None
29 f 74 Yes x x B x x x Warmth Walking Arthritis (?) None
30 m 33 No x B x Warmth, Cold Pressure Raynaud SLE (?) None
31 f 42 Yes x B x None Raynaud Rheumat. Dis. (?) None
32 m 45 No x cramp-like x Phys. Act None
33 f 49 Yes x B x x None
34 f 65 Yes x B x x Warmth No None
35 f 28 Yes x B, shoot ? Walking Sitting No None
36 f 55 No x B, S, P, A, T, I x x Warmth Sjogren’s disease RA None
37 f 46 No x B, I x x x Pressure Fibromyalgia (?) None
38 f 61 No x B, P x x Phys. Act. Alcohol No None
39 m 71 Yes x B, P, S, T x x x Warmth Red wine No None
40 m 68 No x B x x x Phys. Act. Alcohol No None
41 f 57 ? x B, sore x x x Warmth No None
42 f 66 No x B, T x x x Warmth Walking No None
43 f 60 Yes x T x x x Warmth Rheumat. Dis. (?) None
44 m 22 Yes x B, S x x Warmth Rheumat. Dis. (?) None
45 m 58 No x B x Pressure Kidney-disease None
46 m 64 No x B, Sore x x ? No None
47 f 42 Yes x B x No None
48 f 30 No x B x x SLE (?) None
  1. Summary of patients characteristics including family history (“family”), presence of distinct pain attacks and ongoing pain, pain characteristics, factors eliciting or worsening pain and the nature of the mutation

  2. Burning (b), aching (a), pricking (p), stinging (s), tightening (t), itching (i), shooting (shoot), physical activity (Phys. Act.), rheumatological disease (Rheumatol. Dis.), rheumatoid arthritis (RA), hands (h), systemic lupus erythematosus (SLE)

Fig. 1 
              Rare genetic variants in the genes identified in the patients. Locations of the mutations are marked with black dots.
Fig. 1

Rare genetic variants in the genes identified in the patients. Locations of the mutations are marked with black dots.

Four rare genetic variants within the NaV1.8 gene (E285K, M650K, L741V and V763I) were identified (Table 2A). E285K is located in the loop between DIS5 and D1S6, while the M650K is located in the cytoplasmic loop between domains I and II (Fig. 1).

L741V and V763I are situated in the transmembrane segments DIIS3 and DIIS4 which are extremely conserved (100%) between human, dog, mouse and rat (Figs. 1 and 2). The voltage sensors are formed by S1 to S4 in each domain which are crucial for sensing changes in membrane potential.

Fig. 2 
              Comparison of amino acid sequence between different species in the region where rare variants were found in NaV1.8 and 1.9 (position highlighted in red).
Fig. 2

Comparison of amino acid sequence between different species in the region where rare variants were found in NaV1.8 and 1.9 (position highlighted in red).

Two rare genetic variants within the NaV1.9 gene (N1169S, I1293V) were found (Table 1). N1169S is located in the small loop between DIIIS4 and DIIIS5 which is supposed to be important for the regulation of channel activity (Fig. 1). I1293V is in the transmembrane segment DIIIS6, a region for ion pore formation that is highly conserved between different species (human, dog, mouse and rat) and between all sodium channels (Fig. 2).

In total, four rare genetic variants, T220I, P648L, S1103Y and P2006A were found in NaV1.5 with P648L and P2006A being de novo (Table 1, Fig. 1). Patients with mutations did not show any signs of cardiac defects.

Five rare genetic variants in the NaV2.1 (V133I, Q236P, D803V, S1154Y and I1599V) were identified in this study and all these variants are neither present in public domain datasets nor in our 96 healthy controls (Table 2, Fig. 1).

Two rare variants, R125C in SCN1B and L10P in SCN3B, were found in two patients respectively (Table 2). The variant, R125C, may create a new position for the disulfide bridge and alter the secondary structure of the extracellular domain of SCN1B (Fig. 1). Mutation L10P is located at a highly conserved residue in the extracellular domain. This variant was also detected when 96 healthy controls were screened, but with an extremely low allele frequency (0.005, 1/192 chromosomes). No mutation within SCN2B and SCN4B was detected in this study.

3.1.1 Transient receptor potential channels (TRPA1, TRPV1, TRPM8)

Two rare genetic variants and a nonsense mutation of TRPA1 were identified. R343C, N460S and C608X are located at the 9th, 11th and 15th ankyrin repeats respectively (Table 2, Fig. 1). R343C was also traced back to the mother who is affected, too, but her affected sister carries R125C mutation in SCN1B (Table 2). Unfortunately, unaffected members of this family were not available for genetic analysis.

One rare genetic variant (F305S) and a nonsense mutation (Q498X) within TRPV1 were detected. F305S is located at the connection between ankyrin binding domain 2 and 3 and the nonsense mutation Q498X in the cytoplasmic loop between transmembrane segment 2 and 3 (Table 2 and Fig. 1).Itisworth noting that the same patient carrying nonsense mutation Q498X in TRPV1 also carries another nonsense mutation, C608X in TRPA1; thus, this patient has only one normal copy of each of TRPA1 and TRPV1.

No mutation within TRPM8 was detected in the patients with Erythromelalgia-like symptoms in this study.

3.1.2 Neurotrophic factor (NGF, NGFR, BDNF, GDNF, NTRK1) and other pain related genes

Rare genetic variants, T764A and S2208N in WNK1 and S132L in NGFR, were detected (Fig. 1). However, these variants are not located in the critical region of proteins and a polymorphism (rs72650768, S2208R) affecting the same codon as the S2208N in WNK1 has been observed in the 1000 genomes study (0.2%). Variants in WNK1 and NGFR identified in this study may not impact normal function of the proteinsignificantly. Except forknownpoly-morphisms already in the public domain, nomutation wasdetected in pain-related genes coding for COMT, CACNG2, P2RX3, KCNS1, TAC1 and TACR1 in our patients.

3.2 Clinical characterization

We separated our patients according to results of the genetic analysis into those with mutations that have been proven to be linked to EM (marked in grey; n =4), mutations with a known gain of function in NaV1.7 (marked in light grey; n =1), mutations in genes that have a close link to axonal excitability of nociceptors but for which functional characterizationis lacking (NaV1.7, NaV1.8, NaV1.9, SCN1B, SCN3B; n =10), mutationsin genes that have aclose link to nociceptors (NaV1.6, TRPA1, TRPV1; n =5), mutations in genes that have no close link to nociceptors (italics, NaV1.5, NaV2.1; n =7), and patients that show ednomutations in the analyzed genes (n =21) (Table 2B). The percentage of patients that reported incidence of similar symptoms among their relatives was higher in the I848T mutation patients (4/4) as compared to patients without evidence for mutations (9/21). The other groups of patients with mutations did not differ significantly from those without evidence for mutations in their family history (Table 2B).

While the age of onset was low (3–5 years) in the patients with the I848T mutations, no significant difference in age of pain debut was found in the other groups. Similarly, maximal intensity of pain (NRS-value) was highest in the I848T group, but did not differ significantly in the other patient groups. Pain descriptors did not contribute to differentiate the groups. In all groups, warmth and physical activity were the most prominent eliciting or worsening factors, without significant differences between the groups (see also Table 2).

3.2.1 Pain profile

Pain attacks were reported by the majority of patients in the different mutations groups (4/4, 5/10, 4/5, 6/7), but significantly less frequent in patients without evidence for mutations (2/20; Chi square<0.01) (Table 2). When only EM patients with the classical combination of red hot painful feet and pain relief by cooling, pain attacks were reported by 4/4, 3/7, 2/3 and 3/3 patients in the patients with mutations or rare variants and 2/7 in group without evidence for mutations.

3.3 Results of clinical examination

Two patients (7.4%) with mutations showed clinical signs of sen-sory neuropathy (reduced sensibility to light touch in distal parts of legs), but with normal findings on neurography. A total of seven patients (36.8%) without mutations displayed signsof sensory neuropathy (verified by sensory neurography in two of the patients, one more pronounced with sensory conduction velocities of 33 and 36 m/s, sural nerve bilaterally). Allodynia to light touch distally in the lower extremities was present in one of the patients in each group, whereas pin-prick hyperalgesia was demonstrated in seven patients with mutations and in four patients without mutations.

None of the patients displayed increased skin temperature at the time of investigation. The skin temperature in the dorsum of the feet did not differ significantly between the two groups (in the group with mutation: median 28.6°C, (27.7–30.4 (quartiles)) (in the group without mutation: median 29.6°C (28.0–30.7) (p = 0.34, Mann–Whitney U-test).

4 Discussion

Since Yang et al. [56] mapped the primary EM to chromosome 2q and identified mutations of SCN9A in patients with EM, genetic studies on EM have identified a variety of mutations of SCN9A that increase neuronal excitability by a shift in voltage-dependence of NaV1.7 in a hyperpolarized direction and amplify rampcurrent[15]. NaV1.7 is considered to be an important player in the sensation of peripheral pain due to its gating properties. “Gain-of-function” mutations in NaV1.7 were found in patients with EM, paroxysmal extreme pain disorder (PEPD) and small fibre neuropathy (SFN), and epilepsies [18,23,57], while “loss-of-function” mutations were detected in almost all the patients with insensitivity to pain (CIP, [9]) and anosmia [54]. As an interesting observation, whole-cell patch-clamp studies suggested that changes in biophysical properties of mutated NaV1.7 in PE, PEPD, SFN and epilepsies [16,46] are different [21].

4.1 Variants of targets with proven link to EM

The degree of neuronal hyperexcitability induced by NaV1.7 mutations correlates with earlier onset and more severe symptoms [7,10,26]. In addition to familiar cases of EM also de-novo mutations of NaV1.7 have been identified in patients without family history [27,28]. In our study a larger cohort of patients diagnosed with primary erythromelalgia has been genotyped providing an estimate of the prevalence of NaV1.7 mutations among EM patients. Among the 48 patients there were 5 with a gain of function mutation of NaV1.7 that has already been functionally characterized. Four patients carried the I848T mutation known tocause a massive shift of activation threshold and increased ramp current. The proportion of 4 in 48 is close to 1 in 15 [17] or 1 in 13 [33] reported earlier. In one patient the I228M variant of NaV1.7 was identified that has been found in healthy controls and epilepsy patients [46], but recently was also linked to small fibre neuropathy and neuropathic pain [21,23,42]. The occurrence of small fibre neuropathy in EM has already been described [40]. Basically, small fibre neuropathy might imitate symptoms of EM or represent an independent pathophysiological endpoint of hyperexcitability leading to EM and, in certain mutations also to neuropathy. The gain of function mutations in NaV1.7 induced by the I228M mutation suggests overlapping pathophysiologic mechanism between EM and small fibre neuropathy, i.e. increased excitability underlies pain and increased sodium influx per action potential might – after sodium-calcium exchange – cause axonal degeneration [42]. The clinical picture concerning severity of small fibre neuropathy vs. intensity of chronic pain varies between carriers of I228M variant with either dominance of pain, dominance of neuropathy or even absence of clinical symptoms [21]. Our patient carrying the I228M variant had typical painful, red and hot feet, but thepain relief upon cooling was mild and the pain was reported as constant rather than in attacks – this ambiguity of clinical symptoms might reflect the “in-between” type of hyperexcitability generated by this variant. Interestingly, a similar overlap of clinical symptoms and sodium channel characteristics of the mutated channel has also been described between EM and PEPD [19,20].

Two more variants of NaV1.7, N1245S and P187L, were identified in our patients. N1245S has been reported before as rare variant (minor allele frequency: 0.0041; NCBI, SNP database) whereas the P187L mutation has not been described yet. In the absence of functional characterization, the clinical relevance of these NaV1.7 variants remains unclear.

4.2 Variants of targets with probable link to axonal excitability

In contrast to earlier attempts [17] we found rare variants of NaV1.8 and NaV1.9 in our EM patients. NaV1.8 is found in small-diameter C-fibre associated sensory neurons [8], but is also expressed in non-nociceptive primary afferent fibres [45] and in intracardiac neurons [25,48]. NaV1.8 has been shown to have a specific role in the detection of noxious thermal, mechanical and inflammatory stimuli [1]. Recently, several reports indicate that SNP rs6795970 (A1073V) within the DII/III intracellular loop of, is associated with altered electrophysiological properties and abnormalities of cardiac conduction [30]. Most interestingly, among 104 patients with painful small fibre neuropathy nine were identified carrying mutations of NaV1.8 with 3 of those mutations (Leu554Pro, Ala1304Thr, Cys1523Tyr) proven to result inagain offunction [24]. One of those patients also showed paroxysmal reddening of the soles; however, some pain relief was achieved by warming. Thus, none of these patients would have been diagnosed as EM.

We identified the rare variants L741V, M650K, and E285K of NaV1.8inour patients. However, all of them have reported before as rarevariant (unknown minor allele frequency; NCBI, SNP database). Therefore, the variants found in our patient could be either unas-sociated polymorphisms or the disease penetration of the variants is low.

NaV1.9 contributes to the persistent tetrodotoxin-resistant current in small diameter DRG neurons and to the persistent thermal hypersensitivity and spontaneous pain behaviour following the exposure to inflammatory mediators (inflammation-induced hyperalgesia). In our patients the rare variants N1169S and I1293V were identified, with N1169S, but not I1293V having been reported as SNP before (unknown minor allele frequency; NCBI, SNP database).

The sodium channel a subunits determine the basic structure of the channel, while β-subunits modulate its properties. Genetic defects in the β subunits were reported to be associated with generalized epilepsy, with febrile seizures plus (SCN1B, [50]), cardiac conduction defect and Brugada syndrome 5 (SCN1B, [52]), Bru-gada syndrome 7 (SCN3B, [31]) and long QT syndrome 10 (SCN4B, [39] ). The R125C variant in the β1 subunit has been associated with epilepsy [41]. Albeit the β1 subunit can modify gating properties of NaV1.7 and NaV1.8 [38,49] its clinical role appears to be more obvious for epilepsy and cardiac arrhythmia [6]. In contrast, the β3 subunit is expressed mainly in small and medium size sensory neurons [6] and neuropathic pain models lead to its upregulation [12,44]. Despite these links to nociceptive fibres there are yet only reports of mutated β3 subunits leading to ventricular fibrillation via interaction with NaV1.5 [32], but none that would be linked to chronic pain states.

4.3 Variants of targets with possible links to axonal hyperexcitability

NaV1.6 is mainly expressed in myelinated afferents and has been shown to be up-regulated in neuropathic pain models in rodents [43]. Moreover, siRNA knock down of NaV1.6 reduces mechanical hypersensitivity and bursting of primary afferents following inflammation of the dorsal root ganglion [55]. Mechanistically, resurgent currents leading to repetitive firing have been suggested to mediate amplification of afferent information including pain [34,47]. The rare variant I1583T of NaV1.6 we found in our patient has not been reported as SNP before (NCBI, SNP database), however there is no information about potential functional consequences.

The transient receptor potential (TRP) channel family has been shown to play a dominant role with regard to thermo-and chemo-sensitivity [2]. Transient receptor potential A1 (TRPA1) is expressed by primary afferent sensory neurons of the pain pathway where it functions as a sensor for environmental and endogenous chemical irritants and contributes to inflammatory pain [2]. E179K (rs920829), a common polymorphism within the 4th ankyrin repeat of TRPA1, was shown to be associated with the somatosen-sory function in neuropathic pain patients [3], and a gain of function mutation N855S in the 4th transmembrane segment was confirmed to be the gene responsible for family episodic pain syndrome [35]. In our patients the variants N40S, R434C, and C608X were not reported as polymorphism (NCBI, SNP database), but there is also no functional data available to further judge their potential clinical relevance.

Transient receptor potential cation channel, subfamily V, member 1 (TRPV1) is a nonselective cation channel directly activated by harmful heat, extracellular protons, and vanilloid compounds. Elec-trophysiological results in TRPV1-nullmice (TRPV1–/–) indicate that TRPV1 is essential for selective modalities of pain sensation and for tissue injury-induced thermal hyperalgesia (temperature by generating inward membrane currents [5]). The variants found in our patients Q498X and F305S were not reported as polymorphism (NCBI, SNP database), but again no functional data are available.

4.4 Variants of targets with possible links to axonal hyperexcitability

NaV1.5 is expressed exclusively in human heart and is responsible for the initial upstroke of action potential. Mutations in the SCN5A cause susceptibility to cardiac arrhythmias and sudden death in the long QT syndrome (LQT3, [51]). High incidence of abdominal pain in families with SCN5 A related cardiac channelopathy was observed and has led to the hypothesis that NaV1.5 may contribute to the pathogenesis of functional bowel disorder [36]. Thus, the variants of NaV1.5 found in our patients are most probably not related to their EM pain phenotype.

Sodium channel NaV2.1 coded for by SCN6A is widely expressed and can bedetected in lung, heart, dorsal root ganglia and Schwann cells in the peripheral nervous system as well as in CNS. No mutation in NaV2.1 was reported to be associated with human disease phenotype. Serine/threonine-protein kinase WNK1 plays an important role in regulating electrolyte homeostasis. In particular, it modulates sodium- and potassium-coupled chloride cotrans-porters. Mutations in WNK1 have been linked to higher sensitivity to thermal stimuli including cold pain [37], but not yet to human disease. Overall, the variants of NaV2.1 and WNK1 therefore most probably represent polymorphisms without direct link to the pain phenotype of our patients.

5 Conclusions

Only the patients carrying the I848T mutation of NaV1.7 that has been functionally validated as gain-of-function mutation could be clinically identified by their early debut of pain and their high pain levels. Among our patients there were 4 cases with this mutation and one additional with the functionally characterized I228M mutation. Thus, our analysis clarified the genetic cause in 5 of 48 patients of the EM patients which is close to the combined 2 in 28 reported before [17,33]. This percentage represents the lowest estimate as only mutations in the coding sequence were assessed and EM might also be caused by mutations in the regulating intron regions. Moreover, the two rare variantsof NaV1.7 (N1245S, P187L) might alsobecausally linked to EM even though one was also found in healthy controls. Our data confirm the absence of SCN9A mutations in families with a strong pattern of autosomal dominant EM [4] leading to the obvious question whether mutations other than SCN9A could play be involved. In our EM patients’ rare variants of NaV1.8, NaV1.9, or beta subunits were found in 8 of 48 patients. Even though we have no data to support a functional role for these rare variants for EM, still they do not appear to be rare in our sample of EM patients. Similar to polymorphisms of SCN9A that do not cause the disease itself, but have been found to increase the risk of developing chronic painful conditions [22], also the rare variants found in these patients might increase the risk of developing EM.

6 Implications

Irrespective of the pain phenotype, gain of function mutations of NaV1.7 and NaV1.8 have been linked to small fibre neuropathy most probably via increased sodium load. While obviously mutations of both, NaV1.7 and NaV1.8 can underlie small fibre neuropathy a similar overlap for EM has not yet been shown. As we found rare variants of NaV1.8, NaV1.9, or beta-subunits in 8/48 patients, the question whether there might be a functional role of these rare variants to provoke chronic pain conditions or even EM appears to be appropriate.

Highlights

  • Protein modifying mutations of NaV1.7 in 7/48 patients with erythromelalgia.

  • Unidentified cause in majority of erythromelalgia patients.

  • Rare variants of NaV1.8, NaV1.9, orbeta-subunits in eight patients with unclear functional link.


DOI of refers to article: http://dx.doi.org/10.1016/j.sjpain.2014.09.001.



Section of Clinical Neurophysiology, Department of Neurology, Oslo University Hospital–Rikshospitalet, Box 4950, Nydalen, 0424 Oslo, Norway. Tel.: +47 2307 0834; fax: +47 2307 3578
1

These authors contributed equally to this work.


  1. Conflict of interest: The authors declare no conflicts of interest.

Acknowledgements

We acknowledge John E. N. Morten for his scientific advice during sequencing data interpretation, Amanda J. Gladwin for her logistical support in DNA sample handling and Charlotta Björk for her operational leadership in the project.

This work was supported by AstraZeneca and the Kompe-tenzzentrum Schmerz State Baden-Württemberg [MS].

References

1 Abrahamsen B, Zhao J, Asante CO, Cendan CM, Marsh S, Martinez-Barbera JP, Nassar MA, Dickenson AH, Wood JN. The cell and molecular basis of mechanical, cold, and inflammatory pain. Science 2008;321:702–5.Search in Google Scholar

2 Bautista DM, Jordt SE, Nikai T, Tsuruda PR, Read AJ, Poblete J, Yamoah EN, Basbaum AI, Julius D. TRPA1 mediates the inflammatory actions of environmental irritants and proalgesic agents. Cell 2006;124:1269–82.Search in Google Scholar

3 Binder A, May D, Baron R, Maier C, Tolle TR, Treede RD, Berthele A, Faltraco F, Flor H, Gierthmuhlen J, Haenisch S, Huge V, Magerl W, Maihofner C, Richter H, Rolke R, Scherens A, Uceyler N, Ufer M, Wasner G, Zhu J, Cascorbi I. Transient receptor potential channel polymorphisms are associated with the somatosensory function in neuropathic pain patients. PLo S ONE 2011;6:e17387.Search in Google Scholar

4 Burns TM, te Morsche RH, Jansen JB, Drenth JP. Genetic heterogeneity and exclusion of a modifying locus at 2q in a family with autosomal dominant primary erythermalgia. Br J Dermatol 2005;153:174–7.Search in Google Scholar

5 Caterina MJ, Leffler A, Malmberg AB, Martin WJ, Trafton J, Petersen-Zeitz KR, Koltzenburg M, Basbaum AI, Julius D. Impaired nociception and pain sensation in mice lacking the capsaicin receptor. Science 2000;288:306–13.Search in Google Scholar

6 Chahine M, O’Leary ME. Regulatory role of voltage-gated Na channel beta sub-units in sensory neurons. Front Pharmacol 2011;2:70.Search in Google Scholar

7 Cheng X, Dib-Hajj SD, Tyrrell L, Waxman SG. Mutation I136V alters electro-physiological properties of the Na (v) 1. 7 channel in a family with onset of erythromelalgia in the second decade. Mol Pain 2008;4:1.Search in Google Scholar

8 Choi JS, Hudmon A, Waxman SG, Dib-Hajj SD. Calmodulin regulates current density and frequency-dependent inhibition of sodium channel Nav1. 8 in DRG neurons. J Neurophysiol 2006;96:97–108.Search in Google Scholar

9 Cox JJ, Reimann F, Nicholas AK, Thornton G, Roberts E, Springell K, Karbani G, Jafri H, Mannan J, Raashid Y, Al Gazali L, Hamamy H, Valente EM, Gorman S, Williams R, McHale DP, Wood JN, Gribble FM, Woods CG. An SCN9A channelopathy causes congenital inability to experience pain. Nature 2006;444:894–8.Search in Google Scholar

10 Cregg R, Laguda B, Werdehausen R, Cox JJ, Linley JE, Ramirez JD, Bodi I, Markiewicz M, Howell KJ, Chen YC, Agnew K, Houlden H, Lunn MP, Bennett DL, Wood JN, Kinali M. Novel mutations mapping tothe fourth sodium channel domain of Nav1. 7 result in variable clinical manifestations of primary ery-thromelalgia. Neuromolecular Med 2013;15:265–78.Search in Google Scholar

11 Cummins TR, Dib-Hajj SD, Waxman SG. Electrophysiological properties of mutant Nav1. 7 sodium channels in a painful inherited neuropathy. J Neurosci 2004;24:8232–6.Search in Google Scholar

12 Cummins TR, Waxman SG. Downregulation of tetrodotoxin-resistant sodium currents and upregulation of a rapidly repriming tetrodotoxin-sensitive sodium current in small spinal sensory neurons after nerve injury. J Neurosci 1997;17:3503–14.Search in Google Scholar

13 Davis MD, O’Fallon WM, Rogers III RS, Rooke TW. Natural history of ery-thromelalgia: presentation and outcome in 168 patients. Arch Dermatol 2000;136:330–6.Search in Google Scholar

14 Dib-Hajj SD, Cummins TR, Black JA, Waxman SG. From genes to pain: Nav1. 7 and human pain disorders. Trends Neurosci 2007;30:555–63.Search in Google Scholar

15 Dib-Hajj SD, Yang Y, Black JA, Waxman SG. The Na (V) 1. 7 sodium channel: from molecule to man. Nat Rev Neurosci 2013;14:49–62.Search in Google Scholar

16 Doty CN. SCN9A: another sodium channel excited to play a role in human epilepsies. Clin Genet 2010;77:326–8.Search in Google Scholar

17 Drenth JP, te Morsche RH, Mansour S, Mortimer PS. Primary erythermalgia as a sodium channelopathy: screening for SCN9A mutations: exclusion of a causal role of SCN10A and SCN11A. Arch Dermatol 2008;144:320–4.Search in Google Scholar

18 Drenth JP, Waxman SG. Mutations in sodium-channel gene SCN9A cause a spectrum of human genetic pain disorders. J Clin Invest 2007;117:3603–9.Search in Google Scholar

19 Eberhardt M, Nakajima J, Klinger AB, Neacsu C, Huhne K, O’Reilly AO, Kist AM, Lampe AK, Fischer K, Gibson J, Nau C, Winterpacht A, Lampert A. Inherited pain: sodium channel Nav1. 7 A1632T mutation causes erythromelalgia dueto ashift of fast inactivation. J Biol Chem 2014;289:1971–80.Search in Google Scholar

20 Estacion M, Dib-Hajj SD, Benke PJ, te Morsche RH, Eastman EM, Macala LJ, Drenth JP, Waxman SG. NaV1. 7 gain-of-function mutations as a continuum: A1632E displays physiological changes associated with erythromelalgia and paroxysmal extreme pain disorder mutations and produces symptoms of both disorders. J Neurosci 2008;28:11079–88.Search in Google Scholar

21 Estacion M, Han C, Choi JS, Hoeijmakers JG, Lauria G, Drenth JP, Gerrits MM, Dib-Hajj SD, Faber CG, Merkies IS, Waxman SG. Intra- and interfamily pheno-typic diversity in pain syndromes associated with a gain-of-function variant of NaV1. 7. Mol Pain 2011;7:92.Search in Google Scholar

22 Estacion M, Harty TP, Choi JS, Tyrrell L, Dib-Hajj SD, Waxman SG. A sodium channel gene SCN9A polymorphism that increases nociceptor excitability. Ann Neurol 2009;66:862–6.Search in Google Scholar

23 Faber CG, Hoeijmakers JG, Ahn HS, Cheng X, Han C, Choi JS, Estacion M, Lauria G, Vanhoutte EK, Gerrits MM, Dib-Hajj S, Drenth JP, Waxman SG, Merkies IS. Gain of function Na (V) 1. 7 mutations in idiopathic small fiber neuropathy. Ann Neurol 2012;71:26–39.Search in Google Scholar

24 Faber CG, Lauria G, Merkies IS, Cheng X, Han C, Ahn HS, Persson AK, Hoeijmakers JG, Gerrits MM, Pierro T, Lombardi R, Kapetis D, Dib-Hajj SD, Waxman SG. Gain-of-function Nav1. 8 mutations in painful neuropathy. Proc Natl Acad Sci U S A 2012;109:19444–9.Search in Google Scholar

25 Facer P, Punjabi PP, Abrari A, Kaba RA, Severs NJ, Chambers J, Kooner JS, Anand P. Localisation of SCN10A gene product Na (v) 1. 8 and novel pain-related ion channels in human heart. Int Heart J 2011;52:146–52.Search in Google Scholar

26 Han C, Dib-Hajj SD, Lin Z, Li Y, Eastman EM, Tyrrell L, Cao X, Yang Y, Waxman SG. Early- and late-onset inherited erythromelalgia: genotype–phenotype correlation. Brain 2009;132:1711–22.Search in Google Scholar

27 Han C, Rush AM, Dib-Hajj SD, Li S, Xu Z, Wang Y, Tyrrell L, Wang X, Yang Y, Waxman SG. Sporadic onset of erythermalgia: a gain-of-function mutation in Nav1. 7. Ann Neurol 2006;59:553–8.Search in Google Scholar

28 Harty TP, Dib-Hajj SD, Tyrrell L, Blackman R, Hisama FM, Rose JB, Waxman SG. Na (V) 1. 7 mutant A863P in erythromelalgia: effects of altered activation and steady-state inactivation onexcitability of nociceptive dorsal root ganglion neurons. J Neurosci 2006;26:12566–75.Search in Google Scholar

29 Hoeijmakers JG, Han C, Merkies IS, Macala LJ, Lauria G, Gerrits MM, Dib-Hajj SD, Faber CG, Waxman SG. Small nerve fibres, small hands and small feet: a new syndrome of pain, dysautonomia and acromesomelia in a kindred with a novel NaV1. 7 mutation. Brain 2012;135:345–58.Search in Google Scholar

30 Holm H, Gudbjartsson DF, Arnar DO, Thorleifsson G, Thorgeirsson G, Stefansdottir H, Gudjonsson SA, Jonasdottir A, Mathiesen EB, Njolstad I, Nyrnes A, Wilsgaard T, Hald EM, Hveem K, Stoltenberg C, Lochen ML, Kong A, Thorsteins-dottir U, Stefansson K. Several common variants modulate heart rate, PR interval and QRS duration. Nat Genet 2010;42:117–22.Search in Google Scholar

31 Hu D, Barajas-Martinez H, Burashnikov E, Springer M, Wu Y, Varro A, Pfeiffer R, Koopmann TT, Cordeiro JM, Guerchicoff A, Pollevick GD, Antzelevitch C. A mutation in the beta 3 subunit of the cardiac sodium channel associated with Brugada ECG phenotype. Circ Cardiovasc Genet 2009;2: 270–8.Search in Google Scholar

32 Ishikawa T, Takahashi N, Ohno S, Sakurada H, Nakamura K, On YK, Park JE, Makiyama T, Horie M, Arimura T, Makita N, Kimura A. Novel SCN3B mutation associated with Brugada syndrome affects intracellular trafficking and function of Nav1. 5. Circ J 2013;77:959–67.Search in Google Scholar

33 Klein CJ, Wu Y, Kilfoyle DH, Sandroni P, Davis MD, Gavrilova RH, Low PA, Dyck PJ. Infrequent SCN9A mutations in congenital insensitivity to pain and erythromelalgia. J Neurol Neurosurg Psychiatry 2013;84:386–91.Search in Google Scholar

34 Klinger AB, Eberhardt M, Link AS, Namer B, Kutsche LK, Schuy ET, Sittl R, Hoffmann T, Alzheimer C, Huth T, Carr RW, Lampert A. Sea-anemone toxin ATX-II elicits A-fiber-dependent pain and enhances resurgent and persistent sodium currents in large sensory neurons. Mol Pain 2012;8:69.Search in Google Scholar

35 Kremeyer B, Lopera F, Cox JJ, Momin A, Rugiero F, Marsh S, Woods CG, Jones NG, Paterson KJ, Fricker FR, Villegas A, Acosta N, Pineda-Trujillo NG, Ramirez JD, Zea J, Burley MW, Bedoya G, Bennett DL, Wood JN, Ruiz-Linares A. A gain-of-function mutationin TRPA1 causes familial episodic pain syndrome. Neuron 2010;66:671–80.Search in Google Scholar

36 Locke III GR, Ackerman MJ, Zinsmeister AR, Thapa P, Farrugia G. Gastrointestinal symptoms in families of patients with an SCN5A-encoded cardiac channelopathy: evidence of an intestinal channelopathy. Am J Gastroenterol 2006;101:1299–304.Search in Google Scholar

37 Loggia ML, Bushnell MC, Tetreault M, Thiffault I, Bherer C, Mohammed NK, Kuchinad AA, Laferriere A, Dicaire MJ, Loisel L, Mogil JS, Brais B. Carriers of recessive WNK1/HSN2 mutations for hereditary sensory and autonomic neuropathy type 2 (HSAN2) are more sensitive to thermal stimuli. J Neurosci 2009;29:2162–6.Search in Google Scholar

38 Lopez-Santiago LF, Brackenbury WJ, Chen C, Isom LL. Na+ channel Scn1b gene regulates dorsal root ganglion nociceptor excitability in vivo. J Biol Chem 2011;286:22913–23.Search in Google Scholar

39 Medeiros-Domingo A, Kaku T, Tester DJ, Iturralde-Torres P, Itty A, Ye B, Valdivia C, Ueda K, Canizales-Quinteros S, Tusie-Luna MT, Makielski JC, Ackerman MJ. SCN4B-encoded sodium channel beta4 subunit in congenital long-QT syndrome. Circulation 2007;116:134–42.Search in Google Scholar

40 Orstavik K, Mork C, Kvernebo K, Jorum E. Pain in primary erythromelalgia – a neuropathic component. Pain 2004;110:531–8.Search in Google Scholar

41 Patino GA, Brackenbury WJ, Bao Y, Lopez-Santiago LF, O’Malley HA, Chen C, Calhoun JD, Lafreniere RG, Cossette P, Rouleau GA, Isom LL. Voltage-gated Na+ channel beta1B:a secreted cell adhesion molecule involved in human epilepsy. J Neurosci 2011;31:14577–91.Search in Google Scholar

42 Persson AK, Liu S, Faber CG, Merkies IS, Black JA, Waxman SG. Neuropathy-associated Na (V) 1. 7 variant I228M impairs integrity of dorsal root ganglion neuron axons. Ann Neurol 2012:10.Search in Google Scholar

43 Ren YS, Qian NS, Tang Y, Liao YH, Yang YL, Dou KF, Toi M. Sodium channel Nav1. 6 is up-regulated in the dorsal root ganglia in a mouse model of type 2 diabetes. Brain Res Bull 2012;87:244–9.Search in Google Scholar

44 Shah BS, Stevens EB, Gonzalez MI, Bramwell S, Pinnock RD, Lee K, Dixon AK. beta3, a novel auxiliary subunit for the voltage-gated sodium channel, is expressed preferentially in sensory neurons and is upregulated in the chronic constriction injury model of neuropathic pain. Eur J Neurosci 2000;12:3985–90.Search in Google Scholar

45 Shields SD, Ahn HS, Yang Y, Han C, Seal RP, Wood JN, Waxman SG, Dib-Hajj SD. Nav1. 8 expression is not restricted to nociceptors in mouse peripheral nervous system. Pain 2012;153:2017–30.Search in Google Scholar

46 Singh NA, Pappas C, Dahle EJ, Claes LR, Pruess TH, De JP, Thompson J, Dixon M, Gurnett C, Peiffer A, White HS, Filloux F, Leppert MF. A role of SCN9A in human epilepsies, as a cause of febrile seizures and as a potential modifier of Dravet syndrome. PLoS Genet 2009;5:e1000649.Search in Google Scholar

47 Sittl R, Lampert A, Huth T, Schuy ET, Link AS, Fleckenstein J, Alzheimer C, Grafe P, Carr RW. Anticancer drug oxaliplatin induces acute cooling-aggravated neuropathy via sodium channel subtype Na (V) 1. 6-resurgent and persistent current. Proc Natl Acad Sci USA 2012;109:6704–9.Search in Google Scholar

48 Verkerk AO, Remme CA, Schumacher CA, Scicluna BP, Wolswinkel R, de JB, Bezzina CR, Veldkamp MW. Functional Nav1. 8 channels in intracardiac neurons: the link between SCN10A and cardiac electrophysiology. Circ Res 2012;111:333–43.Search in Google Scholar

49 Vijayaragavan K, O’Leary ME, Chahine M. Gating properties of Na (v) 1.7 and Na (v) 1.8 peripheral nerve sodium channels. J Neurosci 2001;21:7909–18.Search in Google Scholar

50 Wallace RH, Wang DW, Singh R, Scheffer IE, George Jr AL, Phillips HA, Saar K, Reis A, Johnson EW, Sutherland GR, Berkovic SF, Mulley JC. Febrile seizures and generalized epilepsy associated with a mutation in the Na+-channel beta1 subunit gene SCN1B. Nat Genet 1998;19:366–70.Search in Google Scholar

51 Wang Q, Shen J, Splawski I, Atkinson D, Li Z, Robinson JL, Moss AJ, Towbin JA, Keating MT. SCN5A mutations associated with an inherited cardiac arrhythmia, long QT syndrome. Cell 1995;80:805–11.Search in Google Scholar

52 Watanabe H, Koopmann TT, Le SS, Yang T, Ingram CR, Schott JJ, Demolombe S, Probst V, Anselme F, Escande D, Wiesfeld AC, Pfeufer A, Kaab S, Wichmann HE, Hasdemir C, Aizawa Y, Wilde AA, Roden DM, Bezzina CR. Sodium channel beta1 subunit mutations associated with Brugada syndrome and cardiac conduction disease in humans. J Clin Invest 2008;118:2260–8.Search in Google Scholar

53 Waxman SG, Dib-Hajj SD. Erythromelalgia: a hereditary pain syndrome enters the molecular era. Ann Neurol 2005;57:785–8.Search in Google Scholar

54 Weiss J, Pyrski M, Jacobi E, Bufe B, Willnecker V, Schick B, Zizzari P, Gossage SJ, Greer CA, Leinders-Zufall T, Woods CG, Wood JN, Zufall F. Loss-of-function mutations in sodium channel Nav1. 7 cause anosmia. Nature 2011;472:186–90.Search in Google Scholar

55 Xie W, Strong JA, Ye L, Mao JX, Zhang JM. Knockdown of sodium channel Na1. 6 blocks mechanical pain and abnormal bursting activity of afferent neurons in inflamed sensory ganglia. Pain 2013:10.Search in Google Scholar

56 Yang Y, Wang Y, Li S, Xu Z, Li H, Ma L, Fan J, Bu D, Liu B, Fan Z, Wu G, Jin J, Ding B, Zhu X, Shen Y. Mutations in SCN9A, encoding a sodium channel alpha subunit, in patients with primary erythermalgia. J Med Genet 2004;41:171–4.Search in Google Scholar

57 Young FB. When adaptive processes go awry: gain-of-function in SCN9A. Clin Genet 2008;73:34–6.Search in Google Scholar

Received: 2014-08-29
Accepted: 2014-09-03
Published Online: 2014-10-01
Published in Print: 2014-10-01

© 2014 Scandinavian Association for the Study of Pain

Downloaded on 25.4.2024 from https://www.degruyter.com/document/doi/10.1016/j.sjpain.2014.09.002/html
Scroll to top button