Skip to main content
Top
Published in: Molecular Pain 1/2008

Open Access 01-12-2008 | Research

Paroxysmal extreme pain disorder M1627K mutation in human Nav1.7 renders DRG neurons hyperexcitable

Authors: Sulayman D Dib-Hajj, Mark Estacion, Brian W Jarecki, Lynda Tyrrell, Tanya Z Fischer, Mark Lawden, Theodore R Cummins, Stephen G Waxman

Published in: Molecular Pain | Issue 1/2008

Login to get access

Abstract

Background

Paroxysmal extreme pain disorder (PEPD) is an autosomal dominant painful neuropathy with many, but not all, cases linked to gain-of-function mutations in SCN9A which encodes voltage-gated sodium channel Nav1.7. Severe pain episodes and skin flushing start in infancy and are induced by perianal probing or bowl movement, and pain progresses to ocular and mandibular areas with age. Carbamazepine has been effective in relieving symptoms, while other drugs including other anti-epileptics are less effective.

Results

Sequencing of SCN9A coding exons from an English patient, diagnosed with PEPD, has identified a methionine 1627 to lysine (M1627K) substitution in the linker joining segments S4 and S5 in domain IV. We confirm that M1627K depolarizes the voltage-dependence of fast-inactivation without substantially altering activation or slow-inactivation, and inactivates from the open state with slower kinetics. We show here that M1627K does not alter development of closed-state inactivation, and that M1627K channels recover from fast-inactivation faster than wild type channels, and produce larger currents in response to a slow ramp stimulus. Using current-clamp recordings, we also show that the M1627K mutant channel reduces the threshold for single action potentials in DRG neurons and increases the number of action potentials in response to graded stimuli.

Conclusion

M1627K mutation was previously identified in a sporadic case of PEPD from France, and we now report it in an English family. We confirm the initial characterization of mutant M1627K effect on fast-inactivation of Nav1.7 and extend the analysis to other gating properties of the channel. We also show that M1627K mutant channels render DRG neurons hyperexcitable. Our new data provide a link between altered channel biophysics and pain in PEPD patients.
Appendix
Available only for authorised users
Literature
1.
go back to reference Dib-Hajj SD, Cummins TR, Black JA, Waxman SG: From genes to pain: Nav1.7 and human pain disorders. Trends Neurosci 2007, 30(11):555–563. 10.1016/j.tins.2007.08.004CrossRefPubMed Dib-Hajj SD, Cummins TR, Black JA, Waxman SG: From genes to pain: Nav1.7 and human pain disorders. Trends Neurosci 2007, 30(11):555–563. 10.1016/j.tins.2007.08.004CrossRefPubMed
2.
go back to reference Waxman SG: Channel, neuronal and clinical function in sodium channelopathies: from genotype to phenotype. Nat Neurosci 2007, 10(4):405–409. 10.1038/nn1857CrossRefPubMed Waxman SG: Channel, neuronal and clinical function in sodium channelopathies: from genotype to phenotype. Nat Neurosci 2007, 10(4):405–409. 10.1038/nn1857CrossRefPubMed
3.
go back to reference Black JA, Dib-Hajj S, McNabola K, Jeste S, Rizzo MA, Kocsis JD, Waxman SG: Spinal sensory neurons express multiple sodium channel alpha-subunit mRNAs. Mol Brain Res 1996, 43(1–2):117–131. 10.1016/S0169-328X(96)00163-5CrossRefPubMed Black JA, Dib-Hajj S, McNabola K, Jeste S, Rizzo MA, Kocsis JD, Waxman SG: Spinal sensory neurons express multiple sodium channel alpha-subunit mRNAs. Mol Brain Res 1996, 43(1–2):117–131. 10.1016/S0169-328X(96)00163-5CrossRefPubMed
4.
go back to reference Felts PA, Yokoyama S, Dib-Hajj S, Black JA, Waxman SG: Sodium channel alpha-subunit mRNAs I, II, III, NaG, Na6 and HNE (PN1) – different expression patterns in developing rat nervous system. Mol Brain Res 1997, 45(1):71–82. 10.1016/S0169-328X(96)00241-0CrossRefPubMed Felts PA, Yokoyama S, Dib-Hajj S, Black JA, Waxman SG: Sodium channel alpha-subunit mRNAs I, II, III, NaG, Na6 and HNE (PN1) – different expression patterns in developing rat nervous system. Mol Brain Res 1997, 45(1):71–82. 10.1016/S0169-328X(96)00241-0CrossRefPubMed
5.
go back to reference Sangameswaran L, Fish LM, Koch BD, Rabert DK, Delgado SG, Ilnicka M, Jakeman LB, Novakovic S, Wong K, Sze P, et al.: A novel tetrodotoxin-sensitive, voltage-gated sodium channel expressed in rat and human dorsal root ganglia. J Biol Chem 1997, 272(23):14805–14809. 10.1074/jbc.272.23.14805CrossRefPubMed Sangameswaran L, Fish LM, Koch BD, Rabert DK, Delgado SG, Ilnicka M, Jakeman LB, Novakovic S, Wong K, Sze P, et al.: A novel tetrodotoxin-sensitive, voltage-gated sodium channel expressed in rat and human dorsal root ganglia. J Biol Chem 1997, 272(23):14805–14809. 10.1074/jbc.272.23.14805CrossRefPubMed
6.
go back to reference Toledo-Aral JJ, Moss BL, He ZJ, Koszowski AG, Whisenand T, Levinson SR, Wolf JJ, Silossantiago I, Halegoua S, Mandel G: Identification of PN1, a predominant voltage-dependent sodium channel expressed principally in peripheral neurons. Proc Natl Acad Sci (USA) 1997, 94(4):1527–1532. 10.1073/pnas.94.4.1527CrossRef Toledo-Aral JJ, Moss BL, He ZJ, Koszowski AG, Whisenand T, Levinson SR, Wolf JJ, Silossantiago I, Halegoua S, Mandel G: Identification of PN1, a predominant voltage-dependent sodium channel expressed principally in peripheral neurons. Proc Natl Acad Sci (USA) 1997, 94(4):1527–1532. 10.1073/pnas.94.4.1527CrossRef
7.
go back to reference Djouhri L, Newton R, Levinson SR, Berry CM, Carruthers B, Lawson SN: Sensory and electrophysiological properties of guinea-pig sensory neurones expressing Nav1.7 (PN1) Na+channel alpha-subunit protein. J Physiol (Lond) 2003, 546(Pt 2):565–576. 10.1113/jphysiol.2002.026559CrossRef Djouhri L, Newton R, Levinson SR, Berry CM, Carruthers B, Lawson SN: Sensory and electrophysiological properties of guinea-pig sensory neurones expressing Nav1.7 (PN1) Na+channel alpha-subunit protein. J Physiol (Lond) 2003, 546(Pt 2):565–576. 10.1113/jphysiol.2002.026559CrossRef
8.
go back to reference Nassar MA, Stirling LC, Forlani G, Baker MD, Matthews EA, Dickenson AH, Wood JN: Nociceptor-specific gene deletion reveals a major role for Nav1.7 (PN1) in acute and inflammatory pain. Proc Natl Acad Sci USA 2004, 101(34):12706–12711. 10.1073/pnas.0404915101PubMedCentralCrossRefPubMed Nassar MA, Stirling LC, Forlani G, Baker MD, Matthews EA, Dickenson AH, Wood JN: Nociceptor-specific gene deletion reveals a major role for Nav1.7 (PN1) in acute and inflammatory pain. Proc Natl Acad Sci USA 2004, 101(34):12706–12711. 10.1073/pnas.0404915101PubMedCentralCrossRefPubMed
9.
go back to reference Ahmad S, Dahllund L, Eriksson AB, Hellgren D, Karlsson U, Lund PE, Meijer IA, Meury L, Mills T, Moody A, et al.: A stop codon mutation in SCN9A causes lack of pain sensation. Hum Mol Genet 2007, 16(17):2114–2121. 10.1093/hmg/ddm160CrossRefPubMed Ahmad S, Dahllund L, Eriksson AB, Hellgren D, Karlsson U, Lund PE, Meijer IA, Meury L, Mills T, Moody A, et al.: A stop codon mutation in SCN9A causes lack of pain sensation. Hum Mol Genet 2007, 16(17):2114–2121. 10.1093/hmg/ddm160CrossRefPubMed
10.
go back to reference Cox JJ, Reimann F, Nicholas AK, Thornton G, Roberts E, Springell K, Karbani G, Jafri H, Mannan J, Raashid Y, et al.: An SCN9A channelopathy causes congenital inability to experience pain. Nature 2006, 444(7121):894–898. 10.1038/nature05413CrossRefPubMed Cox JJ, Reimann F, Nicholas AK, Thornton G, Roberts E, Springell K, Karbani G, Jafri H, Mannan J, Raashid Y, et al.: An SCN9A channelopathy causes congenital inability to experience pain. Nature 2006, 444(7121):894–898. 10.1038/nature05413CrossRefPubMed
11.
go back to reference Goldberg Y, Macfarlane J, Macdonald M, Thompson J, Dube MP, Mattice M, Fraser R, Young C, Hossain S, Pape T, et al.: Loss-of-function mutations in the Nav1.7 gene underlie congenital indifference to pain in multiple human populations. Clin Genet 2007, 71(4):311–319. 10.1111/j.1399-0004.2007.00790.xCrossRefPubMed Goldberg Y, Macfarlane J, Macdonald M, Thompson J, Dube MP, Mattice M, Fraser R, Young C, Hossain S, Pape T, et al.: Loss-of-function mutations in the Nav1.7 gene underlie congenital indifference to pain in multiple human populations. Clin Genet 2007, 71(4):311–319. 10.1111/j.1399-0004.2007.00790.xCrossRefPubMed
12.
go back to reference Yang Y, Wang Y, Li S, Xu Z, Li H, Ma L, Fan J, Bu D, Liu B, Fan Z, et al.: Mutations in SCN9A, encoding a sodium channel alpha subunit, in patients with primary erythermalgia. J Med Genet 2004, 41(3):171–174. 10.1136/jmg.2003.012153PubMedCentralCrossRefPubMed Yang Y, Wang Y, Li S, Xu Z, Li H, Ma L, Fan J, Bu D, Liu B, Fan Z, et al.: Mutations in SCN9A, encoding a sodium channel alpha subunit, in patients with primary erythermalgia. J Med Genet 2004, 41(3):171–174. 10.1136/jmg.2003.012153PubMedCentralCrossRefPubMed
13.
go back to reference Dib-Hajj SD, Rush AM, Cummins TR, Hisama FM, Novella S, Tyrrell L, Marshall L, Waxman SG: Gain-of-function mutation in Nav1.7 in familial erythromelalgia induces bursting of sensory neurons. Brain 2005, 128(Pt 8):1847–1854. 10.1093/brain/awh514CrossRefPubMed Dib-Hajj SD, Rush AM, Cummins TR, Hisama FM, Novella S, Tyrrell L, Marshall L, Waxman SG: Gain-of-function mutation in Nav1.7 in familial erythromelalgia induces bursting of sensory neurons. Brain 2005, 128(Pt 8):1847–1854. 10.1093/brain/awh514CrossRefPubMed
14.
go back to reference Drenth JP, Te Morsche RH, Guillet G, Taieb A, Kirby RL, Jansen JB: SCN9A mutations define primary erythermalgia as a neuropathic disorder of voltage gated sodium channels. J Invest Dermatol 2005, 124(6):1333–1338. 10.1111/j.0022-202X.2005.23737.xCrossRefPubMed Drenth JP, Te Morsche RH, Guillet G, Taieb A, Kirby RL, Jansen JB: SCN9A mutations define primary erythermalgia as a neuropathic disorder of voltage gated sodium channels. J Invest Dermatol 2005, 124(6):1333–1338. 10.1111/j.0022-202X.2005.23737.xCrossRefPubMed
15.
go back to reference Michiels JJ, te Morsche RH, Jansen JB, Drenth JP: Autosomal dominant erythermalgia associated with a novel mutation in the voltage-gated sodium channel alpha subunit Nav1.7. Arch Neurol 2005, 62(10):1587–1590. 10.1001/archneur.62.10.1587CrossRefPubMed Michiels JJ, te Morsche RH, Jansen JB, Drenth JP: Autosomal dominant erythermalgia associated with a novel mutation in the voltage-gated sodium channel alpha subunit Nav1.7. Arch Neurol 2005, 62(10):1587–1590. 10.1001/archneur.62.10.1587CrossRefPubMed
16.
go back to reference 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: 553–558. 10.1002/ana.20776CrossRefPubMed 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: 553–558. 10.1002/ana.20776CrossRefPubMed
17.
go back to reference Harty TP, Dib-Hajj SD, Tyrrell L, Blackman R, Hisama FM, Rose JB, Waxman SG: NaV1.7 mutant A863P in erythromelalgia: effects of altered activation and steady-state inactivation on excitability of nociceptive dorsal root ganglion neurons. J Neurosci 2006, 26(48):12566–12575. 10.1523/JNEUROSCI.3424-06.2006CrossRefPubMed Harty TP, Dib-Hajj SD, Tyrrell L, Blackman R, Hisama FM, Rose JB, Waxman SG: NaV1.7 mutant A863P in erythromelalgia: effects of altered activation and steady-state inactivation on excitability of nociceptive dorsal root ganglion neurons. J Neurosci 2006, 26(48):12566–12575. 10.1523/JNEUROSCI.3424-06.2006CrossRefPubMed
18.
go back to reference Lee MJ, Yu HS, Hsieh ST, Stephenson DA, Lu CJ, Yang CC: Characterization of a familial case with primary erythromelalgia from Taiwan. J Neurol 2007, 254(2):210–214. 10.1007/s00415-006-0328-3CrossRefPubMed Lee MJ, Yu HS, Hsieh ST, Stephenson DA, Lu CJ, Yang CC: Characterization of a familial case with primary erythromelalgia from Taiwan. J Neurol 2007, 254(2):210–214. 10.1007/s00415-006-0328-3CrossRefPubMed
19.
go back to reference Fertleman CR, Baker MD, Parker KA, Moffatt S, Elmslie FV, Abrahamsen B, Ostman J, Klugbauer N, Wood JN, Gardiner RM, et al.: SCN9A mutations in paroxysmal extreme pain disorder: allelic variants underlie distinct channel defects and phenotypes. Neuron 2006, 52(5):767–774. 10.1016/j.neuron.2006.10.006CrossRefPubMed Fertleman CR, Baker MD, Parker KA, Moffatt S, Elmslie FV, Abrahamsen B, Ostman J, Klugbauer N, Wood JN, Gardiner RM, et al.: SCN9A mutations in paroxysmal extreme pain disorder: allelic variants underlie distinct channel defects and phenotypes. Neuron 2006, 52(5):767–774. 10.1016/j.neuron.2006.10.006CrossRefPubMed
20.
go back to reference Fertleman CR, Ferrie CD, Aicardi J, Bednarek NA, Eeg-Olofsson O, Elmslie FV, Griesemer DA, Goutieres F, Kirkpatrick M, Malmros IN, et al.: Paroxysmal extreme pain disorder (previously familial rectal pain syndrome). Neurology 2007, 69(6):586–595. 10.1212/01.wnl.0000268065.16865.5fCrossRefPubMed Fertleman CR, Ferrie CD, Aicardi J, Bednarek NA, Eeg-Olofsson O, Elmslie FV, Griesemer DA, Goutieres F, Kirkpatrick M, Malmros IN, et al.: Paroxysmal extreme pain disorder (previously familial rectal pain syndrome). Neurology 2007, 69(6):586–595. 10.1212/01.wnl.0000268065.16865.5fCrossRefPubMed
21.
go back to reference Choi JS, Dib-Hajj SD, Waxman SG: Inherited erythermalgia. Limb pain from an S4 charge-neutral Na channelopathy. Neurology 2006, 67(9):1563–1567. 10.1212/01.wnl.0000231514.33603.1eCrossRefPubMed Choi JS, Dib-Hajj SD, Waxman SG: Inherited erythermalgia. Limb pain from an S4 charge-neutral Na channelopathy. Neurology 2006, 67(9):1563–1567. 10.1212/01.wnl.0000231514.33603.1eCrossRefPubMed
22.
go back to reference Cummins TR, Dib-Hajj SD, Waxman SG: Electrophysiological properties of mutant Nav1.7 sodium channels in a painful inherited neuropathy. J Neurosci 2004, 24(38):8232–8236. 10.1523/JNEUROSCI.2695-04.2004CrossRefPubMed Cummins TR, Dib-Hajj SD, Waxman SG: Electrophysiological properties of mutant Nav1.7 sodium channels in a painful inherited neuropathy. J Neurosci 2004, 24(38):8232–8236. 10.1523/JNEUROSCI.2695-04.2004CrossRefPubMed
23.
go back to reference Lampert A, Dib-Hajj SD, Tyrrell L, Waxman SG: Size matters: Erythromelalgia mutation S241T in Nav1.7 alters channel gating. J Biol Chem 2006, 281(47):36029–36035. 10.1074/jbc.M607637200CrossRefPubMed Lampert A, Dib-Hajj SD, Tyrrell L, Waxman SG: Size matters: Erythromelalgia mutation S241T in Nav1.7 alters channel gating. J Biol Chem 2006, 281(47):36029–36035. 10.1074/jbc.M607637200CrossRefPubMed
24.
go back to reference Sheets PL, Jackson Ii JO, Waxman SG, Dib-Hajj S, Cummins TR: A Nav1.7 channel mutation associated with hereditary erythromelalgia contributes to neuronal hyperexcitability and displays reduced lidocaine sensitivity. J Physiol (Lond) 2007, 581: 1019–1031. 10.1113/jphysiol.2006.127027CrossRef Sheets PL, Jackson Ii JO, Waxman SG, Dib-Hajj S, Cummins TR: A Nav1.7 channel mutation associated with hereditary erythromelalgia contributes to neuronal hyperexcitability and displays reduced lidocaine sensitivity. J Physiol (Lond) 2007, 581: 1019–1031. 10.1113/jphysiol.2006.127027CrossRef
25.
go back to reference Cheng X, Dib-Hajj SD, Tyrrell L, Waxman SG: Mutation I136V alters electrophysiological properties of the NaV1.7 channel in a family with onset of erythromelalgia in the second decade. Mol Pain 2008, 4(1):1. 10.1186/1744-8069-4-1PubMedCentralCrossRefPubMed Cheng X, Dib-Hajj SD, Tyrrell L, Waxman SG: Mutation I136V alters electrophysiological properties of the NaV1.7 channel in a family with onset of erythromelalgia in the second decade. Mol Pain 2008, 4(1):1. 10.1186/1744-8069-4-1PubMedCentralCrossRefPubMed
26.
27.
go back to reference Klugbauer N, Lacinova L, Flockerzi V, Hofmann F: Structure and functional expression of a new member of the tetrodotoxin-sensitive voltage-activated sodium channel family from human neuroendocrine cells. EMBO J 1995, 14(6):1084–1090.PubMedCentralPubMed Klugbauer N, Lacinova L, Flockerzi V, Hofmann F: Structure and functional expression of a new member of the tetrodotoxin-sensitive voltage-activated sodium channel family from human neuroendocrine cells. EMBO J 1995, 14(6):1084–1090.PubMedCentralPubMed
28.
go back to reference Cummins TR, Howe JR, Waxman SG: Slow closed-state inactivation: a novel mechanism underlying ramp currents in cells expressing the hNE/PN1 sodium channel. J Neurosci 1998, 18(23):9607–9619.PubMed Cummins TR, Howe JR, Waxman SG: Slow closed-state inactivation: a novel mechanism underlying ramp currents in cells expressing the hNE/PN1 sodium channel. J Neurosci 1998, 18(23):9607–9619.PubMed
29.
go back to reference Jarecki BW, Sheets PL, Jackson Ii JO, Cummins TR: Paroxysmal Extreme Pain Disorder mutations within the D3/S4–S5 linker of Nav1.7 cause moderate destabilization of fast-inactivation. J Physiol (Lond) 2008, 586(Pt 17):4137–4153. 10.1113/jphysiol.2008.154906CrossRef Jarecki BW, Sheets PL, Jackson Ii JO, Cummins TR: Paroxysmal Extreme Pain Disorder mutations within the D3/S4–S5 linker of Nav1.7 cause moderate destabilization of fast-inactivation. J Physiol (Lond) 2008, 586(Pt 17):4137–4153. 10.1113/jphysiol.2008.154906CrossRef
30.
go back to reference 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(3):320–324. 10.1001/archderm.144.3.320CrossRefPubMed 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(3):320–324. 10.1001/archderm.144.3.320CrossRefPubMed
31.
go back to reference Burns TM, Te Morsche RH, Jansen JB, JP HD: Genetic heterogeneity and exclusion of a modifying locus at 2q in a family with autosomal dominant primary erythermalgia. Br J Dermatol 2005, 153(1):174–177. 10.1111/j.1365-2133.2005.06441.xCrossRefPubMed Burns TM, Te Morsche RH, Jansen JB, JP HD: Genetic heterogeneity and exclusion of a modifying locus at 2q in a family with autosomal dominant primary erythermalgia. Br J Dermatol 2005, 153(1):174–177. 10.1111/j.1365-2133.2005.06441.xCrossRefPubMed
32.
go back to reference Ragsdale DS, Avoli M: Sodium channels as molecular targets for antiepileptic drugs. Brain Res Rev 1998, 26(1):16–28. 10.1016/S0165-0173(97)00054-4CrossRefPubMed Ragsdale DS, Avoli M: Sodium channels as molecular targets for antiepileptic drugs. Brain Res Rev 1998, 26(1):16–28. 10.1016/S0165-0173(97)00054-4CrossRefPubMed
33.
go back to reference Rush AM, Elliott JR: Phenytoin and Carbamazepine – Differential Inhibition Of Sodium Currents In Small Cells From Adult Rat Dorsal Root Ganglia. Neuroscience Letters 1997, 226(2):95–98. 10.1016/S0304-3940(97)00258-9CrossRefPubMed Rush AM, Elliott JR: Phenytoin and Carbamazepine – Differential Inhibition Of Sodium Currents In Small Cells From Adult Rat Dorsal Root Ganglia. Neuroscience Letters 1997, 226(2):95–98. 10.1016/S0304-3940(97)00258-9CrossRefPubMed
34.
go back to reference Sheets PL, Heers C, Stoehr T, Cummins TR: Differential block of sensory neuronal voltage-gated sodium channels by lacosamide [(2R)-2-(acetylamino)-N-benzyl-3-methoxypropanamide], lidocaine, and carbamazepine. J Pharmacol Exp Ther 2008, 326(1):89–99. 10.1124/jpet.107.133413CrossRefPubMed Sheets PL, Heers C, Stoehr T, Cummins TR: Differential block of sensory neuronal voltage-gated sodium channels by lacosamide [(2R)-2-(acetylamino)-N-benzyl-3-methoxypropanamide], lidocaine, and carbamazepine. J Pharmacol Exp Ther 2008, 326(1):89–99. 10.1124/jpet.107.133413CrossRefPubMed
35.
go back to reference Herzog RI, Cummins TR, Ghassemi F, Dib-Hajj SD, Waxman SG: Distinct repriming and closed-state inactivation kinetics of Nav1.6 and Nav1.7 sodium channels in mouse spinal sensory neurons. J Physiol (Lond) 2003, 551(Pt 3):741–750. 10.1113/jphysiol.2003.047357CrossRef Herzog RI, Cummins TR, Ghassemi F, Dib-Hajj SD, Waxman SG: Distinct repriming and closed-state inactivation kinetics of Nav1.6 and Nav1.7 sodium channels in mouse spinal sensory neurons. J Physiol (Lond) 2003, 551(Pt 3):741–750. 10.1113/jphysiol.2003.047357CrossRef
36.
go back to reference Rush AM, Cummins TR, Waxman SG: Multiple sodium channels and their roles in electrogenesis within dorsal root ganglion neurons. J Physiol (Lond) 2007, 579(Pt 1):1–14. 10.1113/jphysiol.2006.121483CrossRef Rush AM, Cummins TR, Waxman SG: Multiple sodium channels and their roles in electrogenesis within dorsal root ganglion neurons. J Physiol (Lond) 2007, 579(Pt 1):1–14. 10.1113/jphysiol.2006.121483CrossRef
37.
go back to reference Cummins TR, Sheets PL, Waxman SG: The roles of sodium channels in nociception: Implications for mechanisms of pain. Pain 2007. Cummins TR, Sheets PL, Waxman SG: The roles of sodium channels in nociception: Implications for mechanisms of pain. Pain 2007.
38.
go back to reference Mitrovic N, Lerche H, Heine R, Fleischhauer R, Pika-Hartlaub U, Hartlaub U, George AL Jr, Lehmann-Horn F: Role in fast inactivation of conserved amino acids in the IV/S4–S5 loop of the human muscle Na+ channel. Neuroscience Letters 1996, 214(1):9–12.PubMed Mitrovic N, Lerche H, Heine R, Fleischhauer R, Pika-Hartlaub U, Hartlaub U, George AL Jr, Lehmann-Horn F: Role in fast inactivation of conserved amino acids in the IV/S4–S5 loop of the human muscle Na+ channel. Neuroscience Letters 1996, 214(1):9–12.PubMed
39.
go back to reference Claes L, Ceulemans B, Audenaert D, Smets K, Lofgren A, Del-Favero J, Ala-Mello S, Basel-Vanagaite L, Plecko B, Raskin S, et al.: De novo SCN1A mutations are a major cause of severe myoclonic epilepsy of infancy. Hum Mutat 2003, 21(6):615–621. 10.1002/humu.10217CrossRefPubMed Claes L, Ceulemans B, Audenaert D, Smets K, Lofgren A, Del-Favero J, Ala-Mello S, Basel-Vanagaite L, Plecko B, Raskin S, et al.: De novo SCN1A mutations are a major cause of severe myoclonic epilepsy of infancy. Hum Mutat 2003, 21(6):615–621. 10.1002/humu.10217CrossRefPubMed
40.
go back to reference Nabbout R, Gennaro E, Dalla Bernardina B, Dulac O, Madia F, Bertini E, Capovilla G, Chiron C, Cristofori G, Elia M, et al.: Spectrum of SCN1A mutations in severe myoclonic epilepsy of infancy. Neurology 2003, 60(12):1961–1967.CrossRefPubMed Nabbout R, Gennaro E, Dalla Bernardina B, Dulac O, Madia F, Bertini E, Capovilla G, Chiron C, Cristofori G, Elia M, et al.: Spectrum of SCN1A mutations in severe myoclonic epilepsy of infancy. Neurology 2003, 60(12):1961–1967.CrossRefPubMed
41.
go back to reference Fleischhauer R, Mitrovic N, Deymeer F, Lehmann-Horn F, Lerche H: Effects of temperature and mexiletine on the F1473S Na+ channel mutation causing paramyotonia congenita. Pflugers Arch 1998, 436(5):757–765. 10.1007/s004240050699CrossRefPubMed Fleischhauer R, Mitrovic N, Deymeer F, Lehmann-Horn F, Lerche H: Effects of temperature and mexiletine on the F1473S Na+ channel mutation causing paramyotonia congenita. Pflugers Arch 1998, 436(5):757–765. 10.1007/s004240050699CrossRefPubMed
42.
go back to reference Rossignol E, Mathieu J, Thiffault I, Tetreault M, Dicaire MJ, Chrestian N, Dupre N, Puymirat J, Brais B: A novel founder SCN4A mutation causes painful cold-induced myotonia in French-Canadians. Neurology 2007, 69(20):1937–1941. 10.1212/01.wnl.0000290831.08585.2cCrossRefPubMed Rossignol E, Mathieu J, Thiffault I, Tetreault M, Dicaire MJ, Chrestian N, Dupre N, Puymirat J, Brais B: A novel founder SCN4A mutation causes painful cold-induced myotonia in French-Canadians. Neurology 2007, 69(20):1937–1941. 10.1212/01.wnl.0000290831.08585.2cCrossRefPubMed
43.
go back to reference Schoser BG, Schroder JM, Grimm T, Sternberg D, Kress W: A large german kindred with cold-aggravated myotonia and a heterozygous A1481D mutation in the SCN4A gene. Muscle Nerve 2007, 35: 599–606. 10.1002/mus.20733CrossRefPubMed Schoser BG, Schroder JM, Grimm T, Sternberg D, Kress W: A large german kindred with cold-aggravated myotonia and a heterozygous A1481D mutation in the SCN4A gene. Muscle Nerve 2007, 35: 599–606. 10.1002/mus.20733CrossRefPubMed
44.
go back to reference Ruan Y, Liu N, Bloise R, Napolitano C, Priori SG: Gating properties of SCN5A mutations and the response to mexiletine in long-QT syndrome type 3 patients. Circulation 2007, 116(10):1137–1144. 10.1161/CIRCULATIONAHA.107.707877CrossRefPubMed Ruan Y, Liu N, Bloise R, Napolitano C, Priori SG: Gating properties of SCN5A mutations and the response to mexiletine in long-QT syndrome type 3 patients. Circulation 2007, 116(10):1137–1144. 10.1161/CIRCULATIONAHA.107.707877CrossRefPubMed
46.
go back to reference McPhee JC, Ragsdale DS, Scheuer T, Catterall WA: A critical role for the S4–S5 intracellular loop in domain IV of the sodium channel alpha-subunit in fast inactivation. J Biol Chem 1998, 273(2):1121–1129. 10.1074/jbc.273.2.1121CrossRefPubMed McPhee JC, Ragsdale DS, Scheuer T, Catterall WA: A critical role for the S4–S5 intracellular loop in domain IV of the sodium channel alpha-subunit in fast inactivation. J Biol Chem 1998, 273(2):1121–1129. 10.1074/jbc.273.2.1121CrossRefPubMed
47.
go back to reference Lerche H, Peter W, Fleischhauer R, Pikahartlaub U, Malina T, Mitrovic N, Lehmann-horn F: Role in fast inactivation of the IV/S4–S5 loop of the human muscle Na+ channel probed by cysteine mutagenesis. J Physiol (Lond) 1997, 505(2):345–352. 10.1111/j.1469-7793.1997.345bb.xCrossRef Lerche H, Peter W, Fleischhauer R, Pikahartlaub U, Malina T, Mitrovic N, Lehmann-horn F: Role in fast inactivation of the IV/S4–S5 loop of the human muscle Na+ channel probed by cysteine mutagenesis. J Physiol (Lond) 1997, 505(2):345–352. 10.1111/j.1469-7793.1997.345bb.xCrossRef
48.
go back to reference Miyamoto K, Nakagawa T, Kuroda Y: Solution structures of the cytoplasmic linkers between segments S4 and S5 (S4–S5) in domains III and IV of human brain sodium channels in SDS micelles. J Pept Res 2001, 58(3):193–203. 10.1034/j.1399-3011.2001.00912.xCrossRefPubMed Miyamoto K, Nakagawa T, Kuroda Y: Solution structures of the cytoplasmic linkers between segments S4 and S5 (S4–S5) in domains III and IV of human brain sodium channels in SDS micelles. J Pept Res 2001, 58(3):193–203. 10.1034/j.1399-3011.2001.00912.xCrossRefPubMed
49.
go back to reference Kellenberger S, Scheuer T, Catterall WA: Movement of the Na+ channel inactivation gate during inactivation. J Biol Chem 1996, 271(48):30971–30979. 10.1074/jbc.271.48.30971CrossRefPubMed Kellenberger S, Scheuer T, Catterall WA: Movement of the Na+ channel inactivation gate during inactivation. J Biol Chem 1996, 271(48):30971–30979. 10.1074/jbc.271.48.30971CrossRefPubMed
50.
go back to reference Filatov GN, Nguyen TP, Kraner SD, Barchi RL: Inactivation and secondary structure in the D4/S4–5 region of the SkM1 sodium channel. J Gen Physiol 1998, 111(6):703–715. 10.1085/jgp.111.6.703PubMedCentralCrossRefPubMed Filatov GN, Nguyen TP, Kraner SD, Barchi RL: Inactivation and secondary structure in the D4/S4–5 region of the SkM1 sodium channel. J Gen Physiol 1998, 111(6):703–715. 10.1085/jgp.111.6.703PubMedCentralCrossRefPubMed
51.
go back to reference Ahern CA, Zhang JF, Wookalis MJ, Horn R: Modulation of the cardiac sodium channel NaV1.5 by Fyn, a Src family tyrosine kinase. Circ Res 2005, 96: 991–998. 10.1161/01.RES.0000166324.00524.ddCrossRefPubMed Ahern CA, Zhang JF, Wookalis MJ, Horn R: Modulation of the cardiac sodium channel NaV1.5 by Fyn, a Src family tyrosine kinase. Circ Res 2005, 96: 991–998. 10.1161/01.RES.0000166324.00524.ddCrossRefPubMed
52.
go back to reference Rizzo MA, Kocsis JD, Waxman SG: Slow sodium conductances of dorsal root ganglion neurons: intraneuronal homogeneity and interneuronal heterogeneity. J Neurophysiol 1994, 72(6):2796–2815.PubMedCentralPubMed Rizzo MA, Kocsis JD, Waxman SG: Slow sodium conductances of dorsal root ganglion neurons: intraneuronal homogeneity and interneuronal heterogeneity. J Neurophysiol 1994, 72(6):2796–2815.PubMedCentralPubMed
Metadata
Title
Paroxysmal extreme pain disorder M1627K mutation in human Nav1.7 renders DRG neurons hyperexcitable
Authors
Sulayman D Dib-Hajj
Mark Estacion
Brian W Jarecki
Lynda Tyrrell
Tanya Z Fischer
Mark Lawden
Theodore R Cummins
Stephen G Waxman
Publication date
01-12-2008
Publisher
BioMed Central
Published in
Molecular Pain / Issue 1/2008
Electronic ISSN: 1744-8069
DOI
https://doi.org/10.1186/1744-8069-4-37

Other articles of this Issue 1/2008

Molecular Pain 1/2008 Go to the issue