Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 135
Filter
1.
Hum Mol Genet ; 33(2): 103-109, 2024 Jan 07.
Article in English | MEDLINE | ID: mdl-37721535

ABSTRACT

Erythromelalgia (EM), is a familial pain syndrome characterized by episodic 'burning' pain, warmth, and erythema. EM is caused by monoallelic variants in SCN9A, which encodes the voltage-gated sodium channel (NaV) NaV1.7. Over 25 different SCN9A mutations attributed to EM have been described to date, all identified in the SCN9A transcript utilizing exon 6N. Here we report a novel SCN9A missense variant identified in seven related individuals with stereotypic episodes of bilateral lower limb pain presenting in childhood. The variant, XM_011511617.3:c.659G>C;p.(Arg220Pro), resides in the exon 6A of SCN9A, an exon previously shown to be selectively incorporated by developmentally regulated alternative splicing. The mutation is located in the voltage-sensing S4 segment of domain I, which is important for regulating channel activation. Functional analysis showed the p.Arg220Pro mutation altered voltage-dependent activation and delayed channel inactivation, consistent with a NaV1.7 gain-of-function molecular phenotype. These results demonstrate that alternatively spliced isoforms of SCN9A should be included in all genomic testing of EM.


Subject(s)
Erythromelalgia , Humans , Erythromelalgia/genetics , Mutation, Missense/genetics , NAV1.7 Voltage-Gated Sodium Channel/genetics , Pain/genetics , Mutation , Exons/genetics
2.
J Gen Physiol ; 155(12)2023 Dec 04.
Article in English | MEDLINE | ID: mdl-37903281

ABSTRACT

Voltage-gated sodium channels in peripheral nerves conduct nociceptive signals from nerve endings to the spinal cord. Mutations in voltage-gated sodium channel NaV1.7 are responsible for a number of severe inherited pain syndromes, including inherited erythromelalgia (IEM). Here, we describe the negative shifts in the voltage dependence of activation in the bacterial sodium channel NaVAb as a result of the incorporation of four different IEM mutations in the voltage sensor, which recapitulate the gain-of-function effects observed with these mutations in human NaV1.7. Crystal structures of NaVAb with these IEM mutations revealed that a mutation in the S1 segment of the voltage sensor facilitated the outward movement of S4 gating charges by widening the pathway for gating charge translocation. In contrast, mutations in the S4 segments modified hydrophobic interactions with surrounding amino acid side chains or membrane phospholipids that would enhance the outward movement of the gating charges. These results provide key structural insights into the mechanisms by which these IEM mutations in the voltage sensors can facilitate outward movements of the gating charges in the S4 segment and cause hyperexcitability and severe pain in IEM. Our work gives new insights into IEM pathogenesis at the near-atomic level and provides a molecular model for mutation-specific therapy of this debilitating disease.


Subject(s)
Erythromelalgia , NAV1.7 Voltage-Gated Sodium Channel , Humans , Erythromelalgia/genetics , Erythromelalgia/metabolism , Erythromelalgia/pathology , Models, Molecular , Mutation , NAV1.7 Voltage-Gated Sodium Channel/genetics , NAV1.7 Voltage-Gated Sodium Channel/chemistry , NAV1.7 Voltage-Gated Sodium Channel/metabolism , Pain/genetics , Pain/metabolism , Pain/pathology
3.
J Physiol ; 601(23): 5341-5366, 2023 Dec.
Article in English | MEDLINE | ID: mdl-37846879

ABSTRACT

We show here that hyperpolarization-activated current (Ih ) unexpectedly acts to inhibit the activity of dorsal root ganglion (DRG) neurons expressing WT Nav1.7, the largest inward current and primary driver of DRG neuronal firing, and hyperexcitable DRG neurons expressing a gain-of-function Nav1.7 mutation that causes inherited erythromelalgia (IEM), a human genetic model of neuropathic pain. In this study we created a kinetic model of Ih and used it, in combination with dynamic-clamp, to study Ih function in DRG neurons. We show, for the first time, that Ih increases rheobase and reduces the firing probability in small DRG neurons, and demonstrate that the amplitude of subthreshold oscillations is reduced by Ih . Our results show that Ih , due to slow gating, is not deactivated during action potentials (APs) and has a striking damping action, which reverses from depolarizing to hyperpolarizing, close to the threshold for AP generation. Moreover, we show that Ih reverses the hyperexcitability of DRG neurons expressing a gain-of-function Nav1.7 mutation that causes IEM. In the aggregate, our results show that Ih unexpectedly has strikingly different effects in DRG neurons as compared to previously- and well-studied cardiac cells. Within DRG neurons where Nav1.7 is present, Ih reduces depolarizing sodium current inflow due to enhancement of Nav1.7 channel fast inactivation and creates additional damping action by reversal of Ih direction from depolarizing to hyperpolarizing close to the threshold for AP generation. These actions of Ih limit the firing of DRG neurons expressing WT Nav1.7 and reverse the hyperexcitability of DRG neurons expressing a gain-of-function Nav1.7 mutation that causes IEM. KEY POINTS: Hyperpolarization-activated cyclic nucleotide-gated (HCN) channels, the molecular determinants of hyperpolarization-activated current (Ih ) have been characterized as a 'pain pacemaker', and thus considered to be a potential molecular target for pain therapeutics. Dorsal root ganglion (DRG) neurons express Nav1.7, a channel that is not present in central neurons or cardiac tissue. Gain-of-function mutations (GOF) of Nav1.7 identified in inherited erythromelalgia (IEM), a human genetic model of neuropathic pain, produce DRG neuron hyperexcitability, which in turn produces severe pain. We found that Ih increases rheobase and reduces firing probability in small DRG neurons expressing WT Nav1.7, and demonstrate that the amplitude of subthreshold oscillations is reduced by Ih . We also demonstrate that Ih reverses the hyperexcitability of DRG neurons expressing a GOF Nav1.7 mutation (L858H) that causes IEM. Our results show that, in contrast to cardiac cells and CNS neurons, Ih acts to stabilize DRG neuron excitability and prevents excessive firing.


Subject(s)
Erythromelalgia , Neuralgia , Animals , Humans , Erythromelalgia/genetics , Nociceptors , Rodentia , Ganglia, Spinal/physiology , NAV1.7 Voltage-Gated Sodium Channel/genetics , Neuralgia/genetics , Neurons/physiology , Action Potentials
6.
J Gen Physiol ; 155(9)2023 09 04.
Article in English | MEDLINE | ID: mdl-37531097

ABSTRACT

Voltage-gated sodium channels (Nav) are key players in excitable tissues with the capability to generate and propagate action potentials. Mutations in the genes encoding Navs can lead to severe inherited diseases, and some of these so-called channelopathies show temperature-sensitive phenotypes, for example, paramyotonia congenita, Brugada syndrome, febrile seizure syndromes, and inherited pain syndromes like erythromelalgia (IEM) and paroxysmal extreme pain disorder (PEPD). Nevertheless, most investigations of mutation-induced gating effects have been conducted at room temperature, and thus the role of cooling or warming in channelopathies remains poorly understood. Here, we investigated the temperature sensitivity of four Nav subtypes: Nav1.3, Nav1.5, Nav1.6, and Nav1.7, and two mutations in Nav1.7 causing IEM (Nav1.7/L823R) and PEPD (Nav1.7/I1461T) expressed in cells of the human embryonic kidney cell line using an automated patch clamp system. Our experiments at 15°C, 25°C, and 35°C revealed a shift of the voltage dependence of activation to more hyperpolarized potentials with increasing temperature for all investigated subtypes. Nav1.3 exhibited strongly slowed inactivation kinetics compared with the other subtypes that resulted in enhanced persistent current, especially at 15°C, indicating a possible role in cold-induced hyperexcitability. Impaired fast inactivation of Nav1.7/I1461T was significantly enhanced by a cooling temperature of 15°C. The subtype-specific modulation as well as the intensified mutation-induced gating changes stress the importance to consider temperature as a regulator for channel gating and its impact on cellular excitability as well as disease phenotypes.


Subject(s)
Channelopathies , Erythromelalgia , Humans , NAV1.7 Voltage-Gated Sodium Channel/genetics , Pain , Erythromelalgia/genetics , Erythromelalgia/metabolism , Mutation
7.
J Peripher Nerv Syst ; 28(4): 597-607, 2023 12.
Article in English | MEDLINE | ID: mdl-37555797

ABSTRACT

BACKGROUND AND AIMS: Voltage-gated sodium channel Nav1.7, encoded by the SCN9A gene, has been linked to diverse painful peripheral neuropathies, represented by the inherited erythromelalgia (EM) and paroxysmal extreme pain disorder (PEPD). The aim of this study was to determine the genetic etiology of patients experiencing neuropathic pain, and shed light on the underlying pathogenesis. METHODS: We enrolled eight patients presenting with early-onset painful peripheral neuropathies, consisting of six cases exhibiting EM/EM-like disorders and two cases clinically diagnosed with PEPD. We conducted a gene-panel sequencing targeting 18 genes associated with hereditary sensory and/or autonomic neuropathy. We introduced novel SCN9A mutation (F1624S) into a GFP-2A-Nav1.7rNS plasmid, and the constructs were then transiently transfected into HEK293 cells. We characterized both wild-type and F1624S Nav1.7 channels using an automated high-throughput patch-clamp system. RESULTS: From two patients displaying EM-like/EM phenotypes, we identified two SCN9A mutations, I136V and P1308L. Among two patients diagnosed with PEPD, we found two additional mutations in SCN9A, F1624S (novel) and A1632E. Patch-clamp analysis of Nav1.7-F1624S revealed depolarizing shifts in both steady-state fast inactivation (17.4 mV, p < .001) and slow inactivation (5.5 mV, p < .001), but no effect on channel activation was observed. INTERPRETATION: Clinical features observed in our patients broaden the phenotypic spectrum of SCN9A-related pain disorders, and the electrophysiological analysis enriches the understanding of genotype-phenotype association caused by Nav1.7 gain-of-function mutations.


Subject(s)
Erythromelalgia , Peripheral Nervous System Diseases , Humans , HEK293 Cells , NAV1.7 Voltage-Gated Sodium Channel/genetics , Erythromelalgia/genetics , Erythromelalgia/pathology , Pain , Mutation/genetics
8.
Proc Natl Acad Sci U S A ; 120(14): e2219624120, 2023 04 04.
Article in English | MEDLINE | ID: mdl-36996107

ABSTRACT

Gain-of-function mutations in voltage-gated sodium channel NaV1.7 cause severe inherited pain syndromes, including inherited erythromelalgia (IEM). The structural basis of these disease mutations, however, remains elusive. Here, we focused on three mutations that all substitute threonine residues in the alpha-helical S4-S5 intracellular linker that connects the voltage sensor to the pore: NaV1.7/I234T, NaV1.7/I848T, and NaV1.7/S241T in order of their positions in the amino acid sequence within the S4-S5 linkers. Introduction of these IEM mutations into the ancestral bacterial sodium channel NaVAb recapitulated the pathogenic gain-of-function of these mutants by inducing a negative shift in the voltage dependence of activation and slowing the kinetics of inactivation. Remarkably, our structural analysis reveals a common mechanism of action among the three mutations, in which the mutant threonine residues create new hydrogen bonds between the S4-S5 linker and the pore-lining S5 or S6 segment in the pore module. Because the S4-S5 linkers couple voltage sensor movements to pore opening, these newly formed hydrogen bonds would stabilize the activated state substantially and thereby promote the 8 to 18 mV negative shift in the voltage dependence of activation that is characteristic of the NaV1.7 IEM mutants. Our results provide key structural insights into how IEM mutations in the S4-S5 linkers may cause hyperexcitability of NaV1.7 and lead to severe pain in this debilitating disease.


Subject(s)
Erythromelalgia , Voltage-Gated Sodium Channels , Humans , NAV1.7 Voltage-Gated Sodium Channel/genetics , NAV1.7 Voltage-Gated Sodium Channel/metabolism , Pain/genetics , Pain/metabolism , Mutation , Erythromelalgia/genetics , Erythromelalgia/metabolism , Erythromelalgia/pathology , Voltage-Gated Sodium Channels/genetics , Threonine/genetics
9.
J Dermatol ; 50(7): 938-941, 2023 Jul.
Article in English | MEDLINE | ID: mdl-36815391

ABSTRACT

Primary erythromelalgia (PEM) is a rare condition characterized by severe burning pain, erythema, and increased temperature in the extremeties. Mutations in the Nav1.7 sodium channel encoded by the SCN9A are responsible for PEM. The pathophysiology of PEM is unclear, but the involvement of neurogenic and vasogenic mechanisms has been suggested. Here we report a case of severe PEM in a 9-year-old child with a novel SCN9A mutation and examine the distribution of nerve fibers and expression of neuropeptides in the affected skin. Gene mutation analysis revealed a novel mutation p.L951I (c.2851C>A) in the heterozygous form of the SCN9A. An immunofluorescence study showed that intraepidermal nerve fibers were decreased in the affected leg, suggesting small fiber neuropathy. There was no increase in the expression of substance P (SP) or calcitonin gene-related peptide (CGRP) in the lesional skin tissue. These findings suggest SP and CGRP do not play a major role in the pathophysiology of primary erythromelalgia.


Subject(s)
Erythromelalgia , Small Fiber Neuropathy , Child , Humans , Erythromelalgia/diagnosis , Erythromelalgia/genetics , Erythromelalgia/metabolism , NAV1.7 Voltage-Gated Sodium Channel/genetics , NAV1.7 Voltage-Gated Sodium Channel/chemistry , NAV1.7 Voltage-Gated Sodium Channel/metabolism , Small Fiber Neuropathy/diagnosis , Small Fiber Neuropathy/genetics , Calcitonin Gene-Related Peptide/genetics , Calcitonin Gene-Related Peptide/metabolism , Pain , Mutation
10.
Brain ; 146(1): 359-371, 2023 01 05.
Article in English | MEDLINE | ID: mdl-35088838

ABSTRACT

Effective treatment of pain remains an unmet healthcare need that requires new and effective therapeutic approaches. NaV1.7 has been genetically and functionally validated as a mediator of pain. Preclinical studies of NaV1.7-selective blockers have shown limited success and translation to clinical studies has been limited. The degree of NaV1.7 channel blockade necessary to attenuate neuronal excitability and ameliorate pain is an unanswered question important for drug discovery. Here, we utilize dynamic clamp electrophysiology and induced pluripotent stem cell-derived sensory neurons (iPSC-SNs) to answer this question for inherited erythromelalgia, a pain disorder caused by gain-of-function mutations in Nav1.7. We show that dynamic clamp can produce hyperexcitability in iPSC-SNs associated with two different inherited erythromelalgia mutations, NaV1.7-S241T and NaV1.7-I848T. We further show that blockade of approximately 50% of NaV1.7 currents can reverse neuronal hyperexcitability to baseline levels.


Subject(s)
Erythromelalgia , Humans , Erythromelalgia/genetics , Erythromelalgia/drug therapy , NAV1.7 Voltage-Gated Sodium Channel/genetics , Mutation/genetics , Pain , Sensory Receptor Cells , Ganglia, Spinal
11.
Pediatr Dermatol ; 39(1): 135-136, 2022 Jan.
Article in English | MEDLINE | ID: mdl-34888934

ABSTRACT

Primary erythromelalgia is a rare autosomal-dominant condition due to pathogenic variant in the SCN9A gene, characterized by childhood onset of excruciating pain, redness, and warmth of acral sites. Patients often resort to ice water baths and other cooling measures to manage the discomfort. Hypothermia is a rare complication, reported only twice previously. We report a child with primary erythromelalgia due to a confirmed pathogenic variant admitted with life-threatening hypothermia. Although the overuse of cooling mechanisms may have contributed, we postulate that the SCN9A mutation may lead to thermodysregulation and make patients with primary erythromelalgia particularly susceptible to this complication.


Subject(s)
Erythromelalgia , Hypothermia , Child , Erythromelalgia/diagnosis , Erythromelalgia/genetics , Erythromelalgia/therapy , Humans , Mutation , NAV1.7 Voltage-Gated Sodium Channel/genetics , Pain
12.
J Biol Chem ; 296: 100227, 2021.
Article in English | MEDLINE | ID: mdl-33361158

ABSTRACT

Mutations in voltage-gated sodium channels (Navs) can cause alterations in pain sensation, such as chronic pain diseases like inherited erythromelalgia. The mutation causing inherited erythromelalgia, Nav1.7 p.I848T, is known to induce a hyperpolarized shift in the voltage dependence of activation in Nav1.7. So far, however, the mechanism to explain this increase in voltage sensitivity remains unknown. In the present study, we show that phosphorylation of the newly introduced Thr residue explains the functional change. We expressed wildtype human Nav1.7, the I848T mutant, or other mutations in HEK293T cells and performed whole-cell patch-clamp electrophysiology. As the insertion of a Thr residue potentially creates a novel phosphorylation site for Ser/Thr kinases and because Nav1.7 had been shown in Xenopus oocytes to be affected by protein kinases C and A, we used different nonselective and selective kinase inhibitors and activators to test the effect of phosphorylation on Nav1.7 in a human system. We identify protein kinase C, but not protein kinase A, to be responsible for the phosphorylation of T848 and thereby for the shift in voltage sensitivity. Introducing a negatively charged amino acid instead of the putative phosphorylation site mimics the effect on voltage gating to a lesser extent. 3D modeling using the published cryo-EM structure of human Nav1.7 showed that introduction of this negatively charged site seems to alter the interaction of this residue with the surrounding amino acids and thus to influence channel function. These results could provide new opportunities for the development of novel treatment options for patients with chronic pain.


Subject(s)
Membrane Potentials/physiology , NAV1.7 Voltage-Gated Sodium Channel/chemistry , Protein Kinase C/metabolism , Protein Processing, Post-Translational , Threonine/chemistry , Amino Acid Substitution , Binding Sites , Chronic Pain/genetics , Chronic Pain/metabolism , Chronic Pain/physiopathology , Erythromelalgia/genetics , Erythromelalgia/metabolism , Erythromelalgia/physiopathology , Gene Expression , HEK293 Cells , Humans , Ion Channel Gating/physiology , Isoleucine/chemistry , Isoleucine/metabolism , Models, Molecular , Mutation , NAV1.7 Voltage-Gated Sodium Channel/genetics , NAV1.7 Voltage-Gated Sodium Channel/metabolism , Patch-Clamp Techniques , Phosphorylation/drug effects , Protein Binding , Protein Conformation, alpha-Helical , Protein Conformation, beta-Strand , Protein Interaction Domains and Motifs , Protein Kinase C/antagonists & inhibitors , Protein Kinase C/genetics , Protein Kinase Inhibitors/pharmacology , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Staurosporine/pharmacology , Threonine/metabolism
13.
Schmerz ; 33(5): 475-490, 2019 Oct.
Article in German | MEDLINE | ID: mdl-31485751

ABSTRACT

Erythromelalgia is a rare disease that is associated with hemato-oncological diseases or after taking certain drugs and toxins, but it can also occur as an independent clinical picture, for example, due to mutations in the sodium channel NaV1.7. Clinically, there is a characteristic triad of attack-like burning pain and skin redness in the area of the distal extremities, which can be alleviated by excessive cooling. The attacks are triggered by heat, exertion, and stress. The diagnosis is primarily made clinically and can be confirmed by genetic testing if a sodium channel NaV1.7 mutation is present. Important differential diagnoses are complex regional pain syndrome, the non-freezing cold injury, and small fiber neuropathies. Therapy is multidisciplinary and has to be planned individually and include physical therapy and psychotherapy as well as drug therapy as integral components.


Subject(s)
Erythromelalgia , Pain , Erythromelalgia/diagnosis , Erythromelalgia/genetics , Erythromelalgia/pathology , Erythromelalgia/therapy , Humans , Mutation , NAV1.7 Voltage-Gated Sodium Channel/genetics , Pain/etiology , Skin/pathology
14.
Sci Rep ; 9(1): 12251, 2019 08 22.
Article in English | MEDLINE | ID: mdl-31439884

ABSTRACT

Inherited erythromelalgia (IEM), caused by mutations in Nav1.7 channel is characterized by episodic neuropathic pain triggered especially by warm temperature. However, the mechanism underlying the temperature-dependent episodic attacks of IEM remains elusive. We investigated the electrophysiological effect of temperature changes on Nav1.7 channels with three different mutations, p.I136V, p. I848T, and p.V1316A, both in vitro and in vivo. In vitro biophysical studies of the mutant channels show consistent temperature-dependent enhancement of the relative resurgent currents if normalized to the transient currents, as well as temperature-dependent changes in the time to peak and the kinetics of decay of the resurgent currents, but no congruent temperature-dependent changes in steady-state parameters such as shift of activation/inactivation curves and changes of the absolute size of the window or resurgent currents. In vivo nerve excitability tests (NET) in IEM patients reveal the essentially normal indices of NET at a single stimulus. However, there are evident abnormalities if assessed with preconditioning pulses, such as the decrease of threshold elevation in hyperpolarizing threshold electrotonus (50-100 ms), the increase of inward rectification in current-voltage curve, and the increase of refractoriness at the interpulse interval of 2-6 ms in recovery cycle, probably also implicating derangements in temperature dependence of inactivation and of recovery from inactivation in the mutant channels. The pathogenesis of heat-enhanced pain in IEM could be attributed to deranged temperature dependence of Nav1.7 channels responsible for the genesis of resurgent currents.


Subject(s)
Erythromelalgia/genetics , NAV1.7 Voltage-Gated Sodium Channel/genetics , Neuralgia/metabolism , Sodium/metabolism , Erythromelalgia/congenital , Erythromelalgia/metabolism , Female , Hot Temperature , Humans , Male , Mutation, Missense , Neuralgia/congenital , Neuralgia/genetics , Patch-Clamp Techniques
15.
Exp Neurol ; 320: 112958, 2019 10.
Article in English | MEDLINE | ID: mdl-31132363

ABSTRACT

We identified a homozygous missense mutation in the gene encoding NAD synthesizing enzyme NMNAT2 in two siblings with childhood onset polyneuropathy with erythromelalgia. No additional homozygotes for this rare allele, which leads to amino acid substitution T94M, were present among the unaffected relatives tested or in the 60,000 exomes of the ExAC database. For axons to survive, axonal NMNAT2 activity has to be maintained above a threshold level but the T94M mutation confers a partial loss of function both in the ability of NMNAT2 to support axon survival and in its enzymatic properties. Electrophysiological tests and histological analysis of sural nerve biopsies in the patients were consistent with loss of distal sensory and motor axons. Thus, it is likely that NMNAT2 mutation causes this pain and axon loss phenotype making this the first disorder associated with mutation of a key regulator of Wallerian-like axon degeneration in humans. This supports indications from numerous animal studies that the Wallerian degeneration pathway is important in human disease and raises important questions about which other human phenotypes could be linked to this gene.


Subject(s)
Erythromelalgia/genetics , Nicotinamide-Nucleotide Adenylyltransferase/genetics , Polyneuropathies/genetics , Female , Homozygote , Humans , Mutation, Missense , Pedigree , Siblings , Wallerian Degeneration/genetics
16.
Pain ; 160(6): 1327-1341, 2019 06.
Article in English | MEDLINE | ID: mdl-30720580

ABSTRACT

The chronic pain syndrome inherited erythromelalgia (IEM) is attributed to mutations in the voltage-gated sodium channel (NaV) 1.7. Still, recent studies targeting NaV1.7 in clinical trials have provided conflicting results. Here, we differentiated induced pluripotent stem cells from IEM patients with the NaV1.7/I848T mutation into sensory nociceptors. Action potentials in these IEM nociceptors displayed a decreased firing threshold, an enhanced upstroke, and afterhyperpolarization, all of which may explain the increased pain experienced by patients. Subsequently, we investigated the voltage dependence of the tetrodotoxin-sensitive NaV activation in these human sensory neurons using a specific prepulse voltage protocol. The IEM mutation induced a hyperpolarizing shift of NaV activation, which leads to activation of NaV1.7 at more negative potentials. Our results indicate that NaV1.7 is not active during subthreshold depolarizations, but that its activity defines the action potential threshold and contributes significantly to the action potential upstroke. Thus, our model system with induced pluripotent stem cell-derived sensory neurons provides a new rationale for NaV1.7 function and promises to be valuable as a translational tool to profile and develop more efficacious clinical analgesics.


Subject(s)
Erythromelalgia/physiopathology , Induced Pluripotent Stem Cells/cytology , NAV1.7 Voltage-Gated Sodium Channel/metabolism , Sensory Receptor Cells/metabolism , Action Potentials/drug effects , Electric Stimulation/methods , Erythromelalgia/genetics , Ganglia, Spinal/cytology , Humans , Membrane Potentials/drug effects , NAV1.7 Voltage-Gated Sodium Channel/genetics , Nociceptors/physiology , Pain/diagnosis , Pain/genetics , Patch-Clamp Techniques/methods , Tetrodotoxin/pharmacology
17.
Clin Exp Dermatol ; 44(5): 477-482, 2019 Jul.
Article in English | MEDLINE | ID: mdl-30609105

ABSTRACT

Erythromelalgia is a condition characterized by episodic pain, erythema and temperature of the extremities, which is relieved by cooling and aggravated by warming. It is useful to review this topic in light of recent discoveries of the genetic mutations that now define primary erythromelalgia, as opposed to secondary erythromelalgia, which is often associated with underlying medical disorders.


Subject(s)
Erythromelalgia/diagnosis , Capsaicin/therapeutic use , Erythromelalgia/complications , Erythromelalgia/genetics , Erythromelalgia/therapy , Humans , Mass Screening , Mexiletine/therapeutic use , Myeloproliferative Disorders/complications , Myeloproliferative Disorders/diagnosis , NAV1.7 Voltage-Gated Sodium Channel/genetics , Ranolazine/therapeutic use , Sensory System Agents/therapeutic use , Sodium Channel Blockers/therapeutic use , Sympathectomy
18.
J Pediatr ; 206: 217-224.e9, 2019 03.
Article in English | MEDLINE | ID: mdl-30416015

ABSTRACT

OBJECTIVES: To evaluate the clinical features of erythromelalgia in childhood associated with gain-of-function SCN9A mutations that increase activity of the Nav1.7 voltage-gated sodium channel, we conducted a systematic review of pediatric presentations of erythromelalgia related to SCN9A mutations, and compared pediatric clinical presentations of symptomatic erythromelalgia, with or without SCN9A mutations. STUDY DESIGN: PubMed, Embase, and PsycINFO Databases were searched for reports of inherited erythromelalgia in childhood. Clinical features, management, and genotype were extracted. Case notes of pediatric patients with erythromelalgia from the Great Ormond Street Hospital Pain Service were reviewed for clinical features, patient-reported outcomes, and treatments. Children aged over 10 years were recruited for quantitative sensory testing. RESULTS: Twenty-eight publications described erythromelalgia associated with 15 different SCN9A gene variants in 25 children. Pain was severe and often refractory to multiple treatments, including nonspecific sodium channel blockers. Skin damage or other complications of cold immersion for symptomatic relief were common (60%). SCN9A mutations resulting in greater hyperpolarizing shifts in Nav1.7 sodium channels correlated with symptom onset at younger ages (P = .016). Variability in reporting, and potential publication bias toward severe cases, limit any estimations of overall prevalence. In our case series, symptoms were similar but comorbidities were more common in children with SCN9A mutations. Quantitative sensory testing revealed marked dynamic warm allodynia. CONCLUSIONS: Inherited erythromelalgia in children is associated with difficult-to-manage pain and significant morbidity. Standardized reporting of outcome and management in larger series will strengthen identification of genotype-phenotype relationships. More effective long-term therapies are a significant unmet clinical need.


Subject(s)
Erythromelalgia/complications , Erythromelalgia/genetics , Mutation/genetics , NAV1.7 Voltage-Gated Sodium Channel/genetics , Pain/etiology , Adolescent , Child , Female , Humans , Male , Retrospective Studies , Symptom Assessment
19.
J Neurosci ; 39(3): 382-392, 2019 01 16.
Article in English | MEDLINE | ID: mdl-30459225

ABSTRACT

Pain is a complex process that involves both detection in the peripheral nervous system and perception in the CNS. Individual-to-individual differences in pain are well documented, but not well understood. Here we capitalized on inherited erythromelalgia (IEM), a well characterized human genetic model of chronic pain, and studied a unique family containing related IEM subjects with the same disease-causing NaV1.7 mutation, which is known to make dorsal root ganglion (DRG) neurons hyperexcitable, but different pain profiles (affected son with severe pain, affected mother with moderate pain, and an unaffected father). We show, first, that, at least in some cases, relative sensitivity to pain can be modeled in subject-specific induced pluripotent stem cell (iPSC)-derived sensory neurons in vitro; second, that, in some cases, mechanisms operating in peripheral sensory neurons contribute to interindividual differences in pain; and third, using whole exome sequencing (WES) and dynamic clamp, we show that it is possible to pinpoint a specific variant of another gene, KCNQ in this particular kindred, that modulates the excitability of iPSC-derived sensory neurons in this family. While different gene variants may modulate DRG neuron excitability and thereby contribute to interindividual differences in pain in other families, this study shows that subject-specific iPSCs can be used to model interindividual differences in pain. We further provide proof-of-principle that iPSCs, WES, and dynamic clamp can be used to investigate peripheral mechanisms and pinpoint specific gene variants that modulate pain signaling and contribute to interindividual differences in pain.SIGNIFICANCE STATEMENT Individual-to-individual differences in pain are well documented, but not well understood. In this study, we show, first, that, at least in some cases, relative sensitivity to pain can be modeled in subject-specific induced pluripotent stem cell-derived sensory neurons in vitro; second, that, in some cases, mechanisms operating in peripheral sensory neurons contribute to interindividual differences in pain; and third, using whole exome sequencing and dynamic clamp, we show that it is possible to pinpoint a specific gene variant that modulates pain signaling and contributes to interindividual differences in pain.


Subject(s)
Chronic Pain/genetics , Induced Pluripotent Stem Cells , Resilience, Psychological , Adult , Child , Chronic Pain/physiopathology , Erythromelalgia/genetics , Erythromelalgia/physiopathology , Excitatory Postsynaptic Potentials , Exome/genetics , Female , Ganglia, Spinal/cytology , Ganglia, Spinal/physiopathology , Humans , Immunohistochemistry , Individuality , KCNQ Potassium Channels/genetics , KCNQ Potassium Channels/metabolism , Male , Membrane Potentials , NAV1.7 Voltage-Gated Sodium Channel/genetics , Pain Measurement , Patch-Clamp Techniques , Sensory Receptor Cells
20.
J Vasc Surg ; 68(6): 1897-1905, 2018 12.
Article in English | MEDLINE | ID: mdl-30126782

ABSTRACT

OBJECTIVE: Erythromelalgia is highly disabling and treatment is often very challenging. There have been solitary case reports that it might benefit from sympathectomy. This study sought to evaluate the short-term and long-term efficacy of chemical lumbar sympathectomy (CLS) for treatment of recalcitrant erythromelalgia and try to identify a CLS-responsive subset. METHODS: Patients with recalcitrant erythromelalgia were recruited from a tertiary hospital over a 10-year period. L3 to L4 CLS was performed using 5% phenol. The pain intensity score (visual analog scale [VAS] 0-10) was assessed before CLS and at 1 day, 1 week, 3 months, 6 months, 1 year, and 2 years after CLS. A VAS decrease of 90%-100% is defined as complete response, 60%-89% as major partial response. Relapse was defined by a return of a VAS score of 5 or higher. SCN9A gene mutations were screened. RESULTS: Thirteen patients were enrolled, with a median age of 15 years. The mean follow-up was 6.2 ± 3.8 years. SCN9A gene mutation was identified in five patients having family histories. The VAS was 8.2 ± 2.0 at baseline; it decreased to 4.9 ± 2.7 at 1 day and 1.9 ± 3.0 at 1 week after CLS. Nine patients (69.2%) achieved complete response at 1 week after CLS, including three patients with SCN9A gene mutation. Among the three complete response patients having the gene mutation, two reverted to major partial response and one relapsed at 2 years after CLS. Among the six complete response patients without mutation, five maintained complete response and one relapsed. Among the four patients who did not achieve complete response, one patient died at 3.5 months and one patient had an amputation performed at 4 months after CLS. CONCLUSIONS: CLS provides a valid option for the treatment of recalcitrant erythromelalgia. It takes about 1 week to achieve full efficacy. Relapse may occur, especially in patients with an SCN9A gene mutation.


Subject(s)
Erythromelalgia/therapy , Lumbar Vertebrae/innervation , Sympathectomy, Chemical/methods , Adolescent , Amputation, Surgical , Child , DNA Mutational Analysis , Erythromelalgia/diagnosis , Erythromelalgia/genetics , Erythromelalgia/physiopathology , Female , Humans , Male , Middle Aged , Mutation , NAV1.7 Voltage-Gated Sodium Channel/genetics , Pain Measurement , Prospective Studies , Recurrence , Remission Induction , Sympathectomy, Chemical/adverse effects , Time Factors , Treatment Outcome , Young Adult
SELECTION OF CITATIONS
SEARCH DETAIL
...