메뉴 건너뛰기




Volumn , Issue , 2012, Pages 63-122

Voltage-Gated Sodium Channels as Therapeutic Targets

Author keywords

Novel, high throughput electrophysiology, for novel drug leads; Protein protein modulation of Nav channels; Sodium (Na+) channels, gated pores conducting Na+ into cell types; Voltage gated sodium channels, as therapeutic targets; Voltage gated sodium channels, therapeutic targets in diseases

Indexed keywords


EID: 84887252097     PISSN: None     EISSN: None     Source Type: Book    
DOI: 10.1002/9781118185537.ch3     Document Type: Chapter
Times cited : (12)

References (367)
  • 1
    • 0026734109 scopus 로고
    • Tracing the roots of ion channels
    • Jan LY, Jan YN. Tracing the roots of ion channels. Cell 69:715-718 (1992).
    • (1992) Cell , vol.69 , pp. 715-718
    • Jan, L.Y.1    Jan, Y.N.2
  • 4
    • 0027497515 scopus 로고
    • Molecular evolution of voltage-sensitive ion channel genes: On the origins of electrical excitability
    • Strong M, Chandy KG, Gutman GA. Molecular evolution of voltage-sensitive ion channel genes: On the origins of electrical excitability. Mol Biol Evol 10:221-242 (1993).
    • (1993) Mol Biol Evol , vol.10 , pp. 221-242
    • Strong, M.1    Chandy, K.G.2    Gutman, G.A.3
  • 6
    • 0026772987 scopus 로고
    • Differential properties of tetrodotoxin-sensitive and tetrodotoxinresistant sodium channels in rat dorsal root ganglion neurons
    • Roy M, Narahashi T. Differential properties of tetrodotoxin-sensitive and tetrodotoxinresistant sodium channels in rat dorsal root ganglion neurons. J Neurosci 12:2104-2111 (1992).
    • (1992) J Neurosci , vol.12 , pp. 2104-2111
    • Roy, M.1    Narahashi, T.2
  • 7
    • 0141529982 scopus 로고    scopus 로고
    • Upregulation of sodium channel Nav1.3 and functional involvement in neuronal hyperexcitability associated with central neuropathic pain after spinal cord injury
    • Hains BC et al. Upregulation of sodium channel Nav1.3 and functional involvement in neuronal hyperexcitability associated with central neuropathic pain after spinal cord injury. J Neurosci 23:8881-8892 (2003).
    • (2003) J Neurosci , vol.23 , pp. 8881-8892
    • Hains, B.C.1
  • 9
    • 0035426149 scopus 로고    scopus 로고
    • Developmental expression of the novel voltage-gated sodium channel auxiliary subunit beta3, in rat CNS
    • Shah BS, Stevens EB, Pinnock RD, Dixon AK, Lee, K. Developmental expression of the novel voltage-gated sodium channel auxiliary subunit beta3, in rat CNS. J Physiol 534:763-776 (2001).
    • (2001) J Physiol , vol.534 , pp. 763-776
    • Shah, B.S.1    Stevens, E.B.2    Pinnock, R.D.3    Dixon, A.K.4    Lee, K.5
  • 10
    • 23044443470 scopus 로고    scopus 로고
    • Distribution and functional characterization of human Nav1.3 splice variants
    • Thimmapaya R et al. Distribution and functional characterization of human Nav1.3 splice variants. Eur J Neurosci 22:1-9 (2005).
    • (2005) Eur J Neurosci , vol.22 , pp. 1-9
    • Thimmapaya, R.1
  • 11
    • 0345034603 scopus 로고    scopus 로고
    • Evolution and diversity of mammalian sodium channel genes
    • Plummer NW, Meisler MH. Evolution and diversity of mammalian sodium channel genes. Genomics 57:323-331 (1999).
    • (1999) Genomics , vol.57 , pp. 323-331
    • Plummer, N.W.1    Meisler, M.H.2
  • 12
    • 0035076298 scopus 로고    scopus 로고
    • Sodium channels and neurological disease: Insights from Scn8a mutations in the mouse
    • Meisler MH, Kearney J, Escayg A, Macdonald BT, Sprunger LK. Sodium channels and neurological disease: Insights from Scn8a mutations in the mouse. Neurosci 7:136-145 (2001).
    • (2001) Neurosci , vol.7 , pp. 136-145
    • Meisler, M.H.1    Kearney, J.2    Escayg, A.3    Macdonald, B.T.4    Sprunger, L.K.5
  • 13
    • 0033694833 scopus 로고    scopus 로고
    • From ionic currents to molecular mechanisms: The structure and function of voltage-gated sodium channels
    • CatterallWA. From ionic currents to molecular mechanisms: The structure and function of voltage-gated sodium channels. Neuron 26:13-25 (2000).
    • (2000) Neuron , vol.26 , pp. 13-25
    • Catterall, W.A.1
  • 14
    • 0041419702 scopus 로고    scopus 로고
    • Sodium channel beta4, a new disulfide-linked auxiliary subunit with similarity to beta2
    • Yu FH et al. Sodium channel beta4, a new disulfide-linked auxiliary subunit with similarity to beta2. J Neurosci 23:7577-7585 (2003).
    • (2003) J Neurosci , vol.23 , pp. 7577-7585
    • Yu, F.H.1
  • 16
    • 2642514674 scopus 로고    scopus 로고
    • Role of auxiliary beta1-, beta2-, and beta3-subunits and their interaction with Na(v)1.8 voltage-gated sodium channel
    • Vijayaragavan K, Powell AJ, Kinghorn IJ, Chahine M. Role of auxiliary beta1-, beta2-, and beta3-subunits and their interaction with Na(v)1.8 voltage-gated sodium channel. Biochem Biophys Res Commun 319:531-540 (2004).
    • (2004) Biochem Biophys Res Commun , vol.319 , pp. 531-540
    • Vijayaragavan, K.1    Powell, A.J.2    Kinghorn, I.J.3    Chahine, M.4
  • 17
    • 51549097064 scopus 로고    scopus 로고
    • An emerging role for voltage-gated Na+ channels in cellular migration: Regulation of central nervous system development and potentiation of invasive cancers
    • Brackenbury WJ, Djamgoz MB, Isom LL. An emerging role for voltage-gated Na+ channels in cellular migration: Regulation of central nervous system development and potentiation of invasive cancers. Neuroscientist 14:571-583 (2008).
    • (2008) Neuroscientist , vol.14 , pp. 571-583
    • Brackenbury, W.J.1    Djamgoz, M.B.2    Isom, L.L.3
  • 18
    • 0345688013 scopus 로고    scopus 로고
    • Molecular cloning and functional expression of the human sodium channel beta1B subunit, a novel splicing variant of the beta1 subunit.
    • Qin N et al. Molecular cloning and functional expression of the human sodium channel beta1B subunit, a novel splicing variant of the beta1 subunit. Eur J Biochem 270:4762- 4770 (2003).
    • (2003) Eur J Biochem , vol.270 , pp. 4762-4770
    • Qin, N.1
  • 19
    • 0027892935 scopus 로고
    • Structure and modulation of Na+ and Ca2+ channels
    • Catterall WA. Structure and modulation of Na+ and Ca2+ channels. Ann NY Acad Sci 707:1-19 (1993).
    • (1993) Ann NY Acad Sci , vol.707 , pp. 1-19
    • Catterall, W.A.1
  • 20
    • 0035931872 scopus 로고    scopus 로고
    • A 3D view of sodium channels.
    • 991
    • Catterall WA. A 3D view of sodium channels. Nature 409:988-989, 991 (2001).
    • (2001) Nature , vol.409 , pp. 988-989
    • Catterall, W.A.1
  • 22
    • 79960621367 scopus 로고    scopus 로고
    • The crystal structure of a voltage-gated sodium channel.
    • Payandeh J, Scheuer T, Zheng N, Catterall WA. The crystal structure of a voltage-gated sodium channel. Nature 475(7356):353-358 (2011).
    • (2011) Nature , vol.475 , Issue.7356 , pp. 353-358
    • Payandeh, J.1    Scheuer, T.2    Zheng, N.3    Catterall, W.A.4
  • 23
    • 0028055161 scopus 로고
    • A structural model of the tetrodotoxin and saxitoxin binding site of the Na+ channel
    • Lipkind GM, Fozzard HA. A structural model of the tetrodotoxin and saxitoxin binding site of the Na+ channel. Biophys J 66:1-13 (1994).
    • (1994) Biophys J , vol.66 , pp. 1-13
    • Lipkind, G.M.1    Fozzard, H.A.2
  • 24
    • 0026698250 scopus 로고
    • Molecular basis for pharmacological differences between brain and cardiac sodium channels
    • Heinemann SH, Terlau H, Imoto K. Molecular basis for pharmacological differences between brain and cardiac sodium channels. Pflugers Arch 422:90-92 (1992).
    • (1992) Pflugers Arch , vol.422 , pp. 90-92
    • Heinemann, S.H.1    Terlau, H.2    Imoto, K.3
  • 25
    • 0026637366 scopus 로고
    • A mutant of TTX-resistant cardiac sodium channels with TTX-sensitive properties
    • Satin J et al. A mutant of TTX-resistant cardiac sodium channels with TTX-sensitive properties. Science 256:1202-1205 (1992).
    • (1992) Science , vol.256 , pp. 1202-1205
    • Satin, J.1
  • 26
    • 0024811092 scopus 로고
    • A single point mutation confers tetrodotoxin and saxitoxin insensitivity on the sodium channel II
    • Noda M, Suzuki H, Numa S, Stuhmer W. A single point mutation confers tetrodotoxin and saxitoxin insensitivity on the sodium channel II. FEBS Lett 259:213-216 (1989).
    • (1989) FEBS Lett , vol.259 , pp. 213-216
    • Noda, M.1    Suzuki, H.2    Numa, S.3    Stuhmer, W.4
  • 27
    • 0026072218 scopus 로고
    • Mapping the site of block by tetrodotoxin and saxitoxin of sodium channel II
    • Terlau H et al. Mapping the site of block by tetrodotoxin and saxitoxin of sodium channel II. FEBS Lett 293:93-96 (1991).
    • (1991) FEBS Lett , vol.293 , pp. 93-96
    • Terlau, H.1
  • 28
    • 0026517122 scopus 로고
    • Calcium channel characteristics conferred on the sodium channel by single mutations
    • Heinemann SH, Terlau H, Stuhmer W, Imoto K, Numa S. Calcium channel characteristics conferred on the sodium channel by single mutations. Nature 356:441-443 (1992).
    • (1992) Nature , vol.356 , pp. 441-443
    • Heinemann, S.H.1    Terlau, H.2    Stuhmer, W.3    Imoto, K.4    Numa, S.5
  • 29
    • 0029754658 scopus 로고    scopus 로고
    • On the structural basis for ionic selectivity among Na+, K+, and Ca2+ in the voltage-gated sodium channel
    • Favre I, Moczydlowski E, Schild L. On the structural basis for ionic selectivity among Na+, K+, and Ca2+ in the voltage-gated sodium channel. Biophys J 71:3110-3125 (1996).
    • (1996) Biophys J , vol.71 , pp. 3110-3125
    • Favre, I.1    Moczydlowski, E.2    Schild, L.3
  • 30
    • 0030062943 scopus 로고    scopus 로고
    • Control of ion flux and selectivity by negatively charged residues in the outer mouth of rat sodium channels.
    • Chiamvimonvat N, Perez-Garcia MT, Tomaselli GF, Marban E. Control of ion flux and selectivity by negatively charged residues in the outer mouth of rat sodium channels. J Physiol 491(Pt 1):51-59 (1996).
    • (1996) J Physiol , vol.491 , Issue.PART 1 , pp. 51-59
    • Chiamvimonvat, N.1    Perez-Garcia, M.T.2    Tomaselli, G.F.3    Marban, E.4
  • 31
    • 77649213717 scopus 로고    scopus 로고
    • The tetrodotoxin binding site is within the outer vestibule of the sodium channel
    • Fozzard HA, Lipkind GM. The tetrodotoxin binding site is within the outer vestibule of the sodium channel. Mar Drugs 8:219-234 (2010).
    • (2010) Mar Drugs , vol.8 , pp. 219-234
    • Fozzard, H.A.1    Lipkind, G.M.2
  • 32
    • 77957223212 scopus 로고    scopus 로고
    • Molecular model of anticonvulsant drug binding to the voltage-gated sodium channel inner pore.
    • Lipkind GM, Fozzard HA. Molecular model of anticonvulsant drug binding to the voltage-gated sodium channel inner pore. Mol Pharmacol 78(4):631-638 (2010).
    • (2010) Mol Pharmacol , vol.78 , Issue.4 , pp. 631-638
    • Lipkind, G.M.1    Fozzard, H.A.2
  • 33
    • 25444498065 scopus 로고    scopus 로고
    • Sodium channel inactivation: Molecular determinants and modulation
    • Ulbricht W. Sodium channel inactivation: Molecular determinants and modulation. Physiol Rev 85:1271-1301 (2005).
    • (2005) Physiol Rev , vol.85 , pp. 1271-1301
    • Ulbricht, W.1
  • 34
    • 0024368695 scopus 로고
    • Structural parts involved in activation and inactivation of the sodium channel
    • Stuhmer Wet al. Structural parts involved in activation and inactivation of the sodium channel. Nature 339:597-603 (1989).
    • (1989) Nature , vol.339 , pp. 597-603
    • Stuhmer, W.1
  • 35
    • 0008453646 scopus 로고
    • Inhibition of inactivation of single sodium channels by a site-directed antibody
    • Vassilev P, Scheuer T, Catterall W. Inhibition of inactivation of single sodium channels by a site-directed antibody. Proc Natl Acad Sci USA 86:8147-8151 (1989).
    • (1989) Proc Natl Acad Sci USA , vol.86 , pp. 8147-8151
    • Vassilev, P.1    Scheuer, T.2    Catterall, W.3
  • 36
    • 0026483172 scopus 로고
    • A cluster of hydrophobic amino acid residues required for fast Na(+)-channel inactivation
    • West JW et al. A cluster of hydrophobic amino acid residues required for fast Na(+)-channel inactivation. Proc Natl Acad Sci USA 89:10910-10914 (1992).
    • (1992) Proc Natl Acad Sci USA , vol.89 , pp. 10910-10914
    • West, J.W.1
  • 37
    • 0031931679 scopus 로고    scopus 로고
    • Kinetic analysis of block of open sodium channels by a peptide containing the isoleucine, phenylalanine, and methionine (IFM) motif from the inactivation gate
    • Eaholtz G, Zagotta WN, Catterall WA. Kinetic analysis of block of open sodium channels by a peptide containing the isoleucine, phenylalanine, and methionine (IFM) motif from the inactivation gate. J Gen Physiol 111:75-82 (1998).
    • (1998) J Gen Physiol , vol.111 , pp. 75-82
    • Eaholtz, G.1    Zagotta, W.N.2    Catterall, W.A.3
  • 38
    • 0039552118 scopus 로고    scopus 로고
    • Solution structure of the sodium channel inactivation gate
    • Rohl CA et al. Solution structure of the sodium channel inactivation gate. Biochemistry 38:855-861 (1998).
    • (1998) Biochemistry , vol.38 , pp. 855-861
    • Rohl, C.A.1
  • 39
    • 0842326191 scopus 로고    scopus 로고
    • The Na+ channel inactivation gate is a molecular complex: A novel role of the COOH-terminal domain
    • Motoike HK et al. The Na+ channel inactivation gate is a molecular complex: A novel role of the COOH-terminal domain. J Gen Physiol 123:155-165 (2004).
    • (2004) J Gen Physiol , vol.123 , pp. 155-165
    • Motoike, H.K.1
  • 40
    • 0031930102 scopus 로고    scopus 로고
    • Slow inactivation does not affect movement of the fast inactivation gate in voltage-gated Na+ channels
    • Vedantham V, Cannon SC. Slow inactivation does not affect movement of the fast inactivation gate in voltage-gated Na+ channels. J Gen Physiol 111:83-93 (1998).
    • (1998) J Gen Physiol , vol.111 , pp. 83-93
    • Vedantham, V.1    Cannon, S.C.2
  • 41
    • 0029730718 scopus 로고    scopus 로고
    • Interaction between fast and slow inactivation in Skm1 sodium channels
    • Featherstone DE, Richmond JE, Ruben PC. Interaction between fast and slow inactivation in Skm1 sodium channels. Biophys J 71:3098-3109 (1996).
    • (1996) Biophys J , vol.71 , pp. 3098-3109
    • Featherstone, D.E.1    Richmond, J.E.2    Ruben, P.C.3
  • 42
    • 0029990996 scopus 로고    scopus 로고
    • Dynamic rearrangement of the outer mouth of a K+ channel during gating
    • Liu Y, Jurman ME, Yellen G. Dynamic rearrangement of the outer mouth of a K+ channel during gating. Neuron 16:859-867 (1996).
    • (1996) Neuron , vol.16 , pp. 859-867
    • Liu, Y.1    Jurman, M.E.2    Yellen, G.3
  • 43
    • 0028297301 scopus 로고
    • An engineered cysteine in the external mouth of a K+ channel allows inactivation to be modulated by metal binding
    • Yellen G, Sodickson D, Chen TY, Jurman ME. An engineered cysteine in the external mouth of a K+ channel allows inactivation to be modulated by metal binding. Biophys J 66:1068-1075 (1994).
    • (1994) Biophys J , vol.66 , pp. 1068-1075
    • Yellen, G.1    Sodickson, D.2    Chen, T.Y.3    Jurman, M.E.4
  • 44
    • 0026049511 scopus 로고
    • Two types of inactivation in Shaker K+ channels: Effects of alterations in the carboxy-terminal region
    • Hoshi T, Zagotta WN, Aldrich RW. Two types of inactivation in Shaker K+ channels: Effects of alterations in the carboxy-terminal region. Neuron 7:547-556 (1991).
    • (1991) Neuron , vol.7 , pp. 547-556
    • Hoshi, T.1    Zagotta, W.N.2    Aldrich, R.W.3
  • 45
    • 34548641160 scopus 로고    scopus 로고
    • Sodium channels: Ionic model of slow inactivation and statedependent drug binding
    • Tikhonov DB, Zhorov BS. Sodium channels: Ionic model of slow inactivation and statedependent drug binding. Biophys J 93:1557-1570 (2007).
    • (2007) Biophys J , vol.93 , pp. 1557-1570
    • Tikhonov, D.B.1    Zhorov, B.S.2
  • 46
    • 0023903168 scopus 로고
    • Slow sodium channel inactivation in mammalian muscle: A possible role in regulating excitability
    • Ruff RL, Simoncini L, Stuhmer W. Slow sodium channel inactivation in mammalian muscle: A possible role in regulating excitability. Muscle Nerve 11:502-510 (1988).
    • (1988) Muscle Nerve , vol.11 , pp. 502-510
    • Ruff, R.L.1    Simoncini, L.2    Stuhmer, W.3
  • 47
    • 0033594335 scopus 로고    scopus 로고
    • Defective slow inactivation of sodium channels contributes to familial periodic paralysis
    • Hayward LJ, Sandoval GM, Cannon SC. Defective slow inactivation of sodium channels contributes to familial periodic paralysis. Neurology 52:1447-1453 (1999).
    • (1999) Neurology , vol.52 , pp. 1447-1453
    • Hayward, L.J.1    Sandoval, G.M.2    Cannon, S.C.3
  • 48
    • 0034954467 scopus 로고    scopus 로고
    • Enhanced inactivation and acceleration of activation of the sodium channel associated with epilepsy in man
    • Alekov AK, Rahman MM, Mitrovic N, Lehmann-Horn F, Lerche H. Enhanced inactivation and acceleration of activation of the sodium channel associated with epilepsy in man. Eur J Neurosci 13:2171-2176 (2001).
    • (2001) Eur J Neurosci , vol.13 , pp. 2171-2176
    • Alekov, A.K.1    Rahman, M.M.2    Mitrovic, N.3    Lehmann-Horn, F.4    Lerche, H.5
  • 49
    • 0034640093 scopus 로고    scopus 로고
    • Two distinct congenital arrhythmias evoked by a multidysfunctional Na(+) channel.
    • Veldkamp MW et al. Two distinct congenital arrhythmias evoked by a multidysfunctional Na(+) channel. Circ Res 86:E91-E97 (2000).
    • (2000) Circ Res , vol.86
    • Veldkamp, M.W.1
  • 50
    • 0037154288 scopus 로고    scopus 로고
    • Clinical, genetic, and biophysical characterization of SCN5A mutations associated with atrioventricular conduction block
    • Wang DW, Viswanathan PC, Balser JR, George AL, Jr, Benson DW. Clinical, genetic, and biophysical characterization of SCN5A mutations associated with atrioventricular conduction block. Circulation 105:341-346 (2002).
    • (2002) Circulation , vol.105 , pp. 341-346
    • Wang, D.W.1    Viswanathan, P.C.2    Balser, J.R.3    George Jr., A.L.4    Benson, D.W.5
  • 51
    • 0345039873 scopus 로고    scopus 로고
    • A novel LQT3 mutation implicates the human cardiac sodium channel domain IVS6 in inactivation kinetics
    • Groenewegen WA et al. A novel LQT3 mutation implicates the human cardiac sodium channel domain IVS6 in inactivation kinetics. Cardiovasc Res 57:1072-1078 (2003).
    • (2003) Cardiovasc Res , vol.57 , pp. 1072-1078
    • Groenewegen, W.A.1
  • 53
    • 0342547301 scopus 로고    scopus 로고
    • A mutation in segment I-S6 alters slow inactivation of sodium channels
    • Wang SY, Wang GK. A mutation in segment I-S6 alters slow inactivation of sodium channels. Biophys J 72:1633-1640 (1997).
    • (1997) Biophys J , vol.72 , pp. 1633-1640
    • Wang, S.Y.1    Wang, G.K.2
  • 54
    • 0032819357 scopus 로고    scopus 로고
    • Structural determinants of slow inactivation in human cardiac and skeletal muscle sodium channels
    • Vilin YY, Makita N, George AL, Jr, Ruben PC. Structural determinants of slow inactivation in human cardiac and skeletal muscle sodium channels. Biophys J 77:1384-1393 (1999).
    • (1999) Biophys J , vol.77 , pp. 1384-1393
    • Vilin, Y.Y.1    Makita, N.2    George Jr., A.L.3    Ruben, P.C.4
  • 55
    • 33750402366 scopus 로고    scopus 로고
    • A conserved ring of charge in mammalian Na+ channels: A molecular regulator of the outer pore conformation during slow inactivation.
    • Xiong W. et al. A conserved ring of charge in mammalian Na+ channels: A molecular regulator of the outer pore conformation during slow inactivation. J Physiol 576:739- 754 (2006).
    • (2006) J Physiol , vol.576 , pp. 739-754
    • Xiong, W.1
  • 57
    • 0019596088 scopus 로고
    • Sodium channels and gating currents
    • Armstrong CM. Sodium channels and gating currents. Physiol Rev 61:644-683 (1981).
    • (1981) Physiol Rev , vol.61 , pp. 644-683
    • Armstrong, C.M.1
  • 58
    • 0022658001 scopus 로고
    • Voltage-dependent gating of sodium channels: Correlating structure and function
    • Catterall W. Voltage-dependent gating of sodium channels: Correlating structure and function. Trends Neurosci 9:7-10 (1986).
    • (1986) Trends Neurosci , vol.9 , pp. 7-10
    • Catterall, W.1
  • 60
    • 54449100445 scopus 로고    scopus 로고
    • Disulfide locking a sodium channel voltage sensor reveals ion pair formation during activation
    • DeCaen PG, Yarov-Yarovoy V, Zhao Y, Scheuer T, Catterall WA. Disulfide locking a sodium channel voltage sensor reveals ion pair formation during activation. Proc Natl Acad Sci USA 105:15142-15147 (2008).
    • (2008) Proc Natl Acad Sci USA , vol.105 , pp. 15142-15147
    • DeCaen, P.G.1    Yarov-Yarovoy, V.2    Zhao, Y.3    Scheuer, T.4    Catterall, W.A.5
  • 61
    • 0034017867 scopus 로고    scopus 로고
    • The voltage sensor in voltage-dependent ion channels.
    • Bezanilla F. The voltage sensor in voltage-dependent ion channels. Physiol Rev 80:555- 592 (2000).
    • (2000) Physiol Rev , vol.80 , pp. 555-592
    • Bezanilla, F.1
  • 62
    • 63049120380 scopus 로고    scopus 로고
    • Protein-protein interactions involving voltage-gated sodium channels: Post-translational regulation, intracellular trafficking and functional expression
    • Shao D, Okuse K, Djamgoz MB. Protein-protein interactions involving voltage-gated sodium channels: Post-translational regulation, intracellular trafficking and functional expression. Int J Biochem Cell Biol 41:1471-1481 (2009).
    • (2009) Int J Biochem Cell Biol , vol.41 , pp. 1471-1481
    • Shao, D.1    Okuse, K.2    Djamgoz, M.B.3
  • 63
    • 0037030729 scopus 로고    scopus 로고
    • Annexin II light chain regulates sensory neuron-specific sodium channel expression
    • Okuse K et al. Annexin II light chain regulates sensory neuron-specific sodium channel expression. Nature 417:653-656 (2002).
    • (2002) Nature , vol.417 , pp. 653-656
    • Okuse, K.1
  • 64
    • 63849342117 scopus 로고    scopus 로고
    • The ataxia3 mutation in the N-terminal cytoplasmic domain of sodium channel Nav1.6 disrupts intracellular trafficking
    • SharkeyLM et al. The ataxia3 mutation in the N-terminal cytoplasmic domain of sodium channel Nav1.6 disrupts intracellular trafficking. J Neurosci 29:2733-2741 (2009).
    • (2009) J Neurosci , vol.29 , pp. 2733-2741
    • Sharkey, L.M.1
  • 65
    • 67649342668 scopus 로고    scopus 로고
    • Role of the terminal domains in sodium channel localization
    • Lee A, Goldin AL. Role of the terminal domains in sodium channel localization. Channels (Austin) 3:171-180 (2009).
    • (2009) Channels (Austin) , vol.3 , pp. 171-180
    • Lee, A.1    Goldin, A.L.2
  • 66
    • 0035910013 scopus 로고    scopus 로고
    • Role of the C-terminal domain in inactivation of brain and cardiac sodium channels
    • Mantegazza M, Yu FH, Catterall WA, Scheuer T. Role of the C-terminal domain in inactivation of brain and cardiac sodium channels. Proc Natl Acad Sci USA 98:15348-15353 (2001).
    • (2001) Proc Natl Acad Sci USA , vol.98 , pp. 15348-15353
    • Mantegazza, M.1    Yu, F.H.2    Catterall, W.A.3    Scheuer, T.4
  • 67
    • 77950556594 scopus 로고    scopus 로고
    • Sodium channel carboxyl-terminal residue regulates fast inactivation
    • Nguyen HM, Goldin AL. Sodium channel carboxyl-terminal residue regulates fast inactivation. J. Biol Chem 285:9077-9089 (2010).
    • (2010) J. Biol Chem , vol.285 , pp. 9077-9089
    • Nguyen, H.M.1    Goldin, A.L.2
  • 68
    • 0141518276 scopus 로고    scopus 로고
    • Unified mechanisms of Ca2+ regulation across the Ca2+ channel family
    • Liang H et al. Unified mechanisms of Ca2+ regulation across the Ca2+ channel family. Neuron 39:951-960 (2003).
    • (2003) Neuron , vol.39 , pp. 951-960
    • Liang, H.1
  • 69
    • 0141856484 scopus 로고    scopus 로고
    • Calmodulin binds to the C terminus of sodium channels Nav1.4 and Nav1.6 and differentially modulates their functional properties
    • Herzog RI, Liu C, Waxman SG, Cummins TR. Calmodulin binds to the C terminus of sodium channels Nav1.4 and Nav1.6 and differentially modulates their functional properties. J Neurosci 23:8261-8270 (2003).
    • (2003) J Neurosci , vol.23 , pp. 8261-8270
    • Herzog, R.I.1    Liu, C.2    Waxman, S.G.3    Cummins, T.R.4
  • 70
    • 21044453817 scopus 로고    scopus 로고
    • Modulation of skeletal and cardiac voltage-gated sodium channels by calmodulin
    • Young KA, Caldwell JH. Modulation of skeletal and cardiac voltage-gated sodium channels by calmodulin. J Physiol 565:349-370 (2005).
    • (2005) J Physiol , vol.565 , pp. 349-370
    • Young, K.A.1    Caldwell, J.H.2
  • 71
    • 65249153083 scopus 로고    scopus 로고
    • Solution structure of the NaV1.2 C-terminal EF-hand domain
    • Miloushev VZ et al. Solution structure of the NaV1.2 C-terminal EF-hand domain. J Biol Chem 284:6446-6454 (2009).
    • (2009) J Biol Chem , vol.284 , pp. 6446-6454
    • Miloushev, V.Z.1
  • 72
    • 0041345748 scopus 로고    scopus 로고
    • Identification of a conserved ankyrin-binding motif in the family of sodium channel alpha subunits
    • Lemaillet G, Walker B, Lambert S. Identification of a conserved ankyrin-binding motif in the family of sodium channel alpha subunits. J Biol Chem 278:27333-27339 (2003).
    • (2003) J Biol Chem , vol.278 , pp. 27333-27339
    • Lemaillet, G.1    Walker, B.2    Lambert, S.3
  • 73
    • 0038270765 scopus 로고    scopus 로고
    • A targeting motif involved in sodium channel clustering at the axonal initial segment
    • Garrido JJ et al. A targeting motif involved in sodium channel clustering at the axonal initial segment. Science 300:2091-2094 (2003).
    • (2003) Science , vol.300 , pp. 2091-2094
    • Garrido, J.J.1
  • 74
    • 18944397275 scopus 로고    scopus 로고
    • Ankyrin-based cardiac arrhythmias: A new class of channelopathies due to loss of cellular targeting
    • Mohler PJ, Bennett V. Ankyrin-based cardiac arrhythmias: A new class of channelopathies due to loss of cellular targeting. Curr Opin Cardiol 20:189-193 (2005).
    • (2005) Curr Opin Cardiol , vol.20 , pp. 189-193
    • Mohler, P.J.1    Bennett, V.2
  • 75
    • 0035378383 scopus 로고    scopus 로고
    • Neuromodulation of Na+ channels: An unexpected form of cellular plasticity
    • Cantrell AR, Catterall WA. Neuromodulation of Na+ channels: An unexpected form of cellular plasticity. Nat Rev Neurosci 2:397-407 (2001).
    • (2001) Nat Rev Neurosci , vol.2 , pp. 397-407
    • Cantrell, A.R.1    Catterall, W.A.2
  • 76
    • 0027717957 scopus 로고
    • Identification of the sites of selective phosphorylation and dephosphorylation of the rat brain Na+ channel alpha subunit by cAMP-dependent protein kinase and phosphoprotein phosphatases
    • Murphy BJ, Rossie S, De Jongh KS, CatterallWA. Identification of the sites of selective phosphorylation and dephosphorylation of the rat brain Na+ channel alpha subunit by cAMP-dependent protein kinase and phosphoprotein phosphatases. J Biol Chem 268:27355-27362 (1993).
    • (1993) J Biol Chem , vol.268 , pp. 27355-27362
    • Murphy, B.J.1    Rossie, S.2    De Jongh, K.S.3    Catterall, W.A.4
  • 77
    • 0026341651 scopus 로고
    • A phosphorylation site in the Na+ channel required for modulation by protein kinase C
    • West JW, Numann R, Murphy BJ, Scheuer T, Catterall WA. A phosphorylation site in the Na+ channel required for modulation by protein kinase C. Science 254:866-868 (1991).
    • (1991) Science , vol.254 , pp. 866-868
    • West, J.W.1    Numann, R.2    Murphy, B.J.3    Scheuer, T.4    Catterall, W.A.5
  • 78
    • 0033777011 scopus 로고    scopus 로고
    • ERM proteins: From cellular architecture to cell signaling
    • Louvet-Vallee S. ERM proteins: From cellular architecture to cell signaling. Biol Cell 92:305-316 (2000).
    • (2000) Biol Cell , vol.92 , pp. 305-316
    • Louvet-Vallee, S.1
  • 79
    • 0037456077 scopus 로고    scopus 로고
    • Sensory neuron proteins interact with the intracellular domains of sodium channel NaV1.8
    • Malik-Hall M, Poon WY, Baker MD, Wood JN, Okuse K. Sensory neuron proteins interact with the intracellular domains of sodium channel NaV1.8. Brain Res Mol Brain Res 110:298-304 (2003).
    • (2003) Brain Res Mol Brain Res , vol.110 , pp. 298-304
    • Malik-Hall, M.1    Poon, W.Y.2    Baker, M.D.3    Wood, J.N.4    Okuse, K.5
  • 80
    • 0031974345 scopus 로고    scopus 로고
    • Interaction of muscle and brain sodium channels with multiple members of the syntrophin family of dystrophin-associated proteins
    • Gee SH et al. Interaction of muscle and brain sodium channels with multiple members of the syntrophin family of dystrophin-associated proteins. J Neurosci 18:128-137 (1998).
    • (1998) J Neurosci , vol.18 , pp. 128-137
    • Gee, S.H.1
  • 81
    • 0031985105 scopus 로고    scopus 로고
    • Specific interactions between the syntrophin PDZ domain and voltagegated sodium channels
    • Schultz J et al. Specific interactions between the syntrophin PDZ domain and voltagegated sodium channels. Nat Struct Biol 5:19-24 (1998).
    • (1998) Nat Struct Biol , vol.5 , pp. 19-24
    • Schultz, J.1
  • 82
    • 33747425933 scopus 로고    scopus 로고
    • Cardiac sodium channel Nav1. 5 is regulated by a multiprotein complex composed of syntrophins and dystrophin.
    • Gavillet B et al. Cardiac sodium channel Nav1. 5 is regulated by a multiprotein complex composed of syntrophins and dystrophin. Circ Res 99, 407-414 (2006).
    • (2006) Circ Res , vol.99 , pp. 407-414
    • Gavillet, B.1
  • 83
    • 51549096558 scopus 로고    scopus 로고
    • Voltage-gated Na+ channels: Potential for beta subunits as therapeutic targets
    • Brackenbury WJ, Isom LL. Voltage-gated Na+ channels: Potential for beta subunits as therapeutic targets. Expert Opin Ther Targets 12:1191-11203 (2008).
    • (2008) Expert Opin Ther Targets , vol.12 , pp. 1191-11203
    • Brackenbury, W.J.1    Isom, L.L.2
  • 84
    • 78049361936 scopus 로고    scopus 로고
    • Electrophysiology and beyond: Multiple roles of Na+ channel [beta] subunits in development and disease.
    • Patino GA, Isom LL. Electrophysiology and beyond: Multiple roles of Na+ channel [beta] subunits in development and disease. Neurosci Lett 486:53-59.
    • Neurosci Lett , vol.486 , pp. 53-59
    • Patino, G.A.1    Isom, L.L.2
  • 85
    • 12944260700 scopus 로고    scopus 로고
    • beta3: An additional auxiliary subunit of the voltage-sensitive sodium channel that modulates channel gating with distinct kinetics
    • Morgan, K et al. beta3: An additional auxiliary subunit of the voltage-sensitive sodium channel that modulates channel gating with distinct kinetics. Proc Natl Acad Sci USA 97:2308-2313 (2000).
    • (2000) Proc Natl Acad Sci USA , vol.97 , pp. 2308-2313
    • Morgan, K.1
  • 86
    • 0028972374 scopus 로고
    • Structure and function of the [beta]2 subunit of brain sodium channels, a transmembrane glycoprotein with a CAM motif
    • Isom LL et al. Structure and function of the [beta]2 subunit of brain sodium channels, a transmembrane glycoprotein with a CAM motif. Cell 83:433-442 (1995).
    • (1995) Cell , vol.83 , pp. 433-442
    • Isom, L.L.1
  • 87
    • 0026551357 scopus 로고
    • Primary structure and functional expression of the beta 1 subunit of the rat brain sodium channel
    • Isom L et al. Primary structure and functional expression of the beta 1 subunit of the rat brain sodium channel. Science 256:839-842 (1992).
    • (1992) Science , vol.256 , pp. 839-842
    • Isom, L.1
  • 88
    • 20744458852 scopus 로고    scopus 로고
    • Presenilin/gammasecretase- mediated cleavage of the voltage-gated sodium channel beta2-subunit regulates cell adhesion and migration
    • Kim DY, Ingano LA, Carey BW, Pettingell WH, Kovacs DM. Presenilin/gammasecretase- mediated cleavage of the voltage-gated sodium channel beta2-subunit regulates cell adhesion and migration. J Biol Chem 280:23251-23261 (2005).
    • (2005) J Biol Chem , vol.280 , pp. 23251-23261
    • Kim, D.Y.1    Ingano, L.A.2    Carey, B.W.3    Pettingell, W.H.4    Kovacs, D.M.5
  • 89
    • 10944261324 scopus 로고    scopus 로고
    • Sodium channel beta1 subunits promote neurite outgrowth in cerebellar granule neurons
    • Davis TH, Chen C, Isom LL. Sodium channel beta1 subunits promote neurite outgrowth in cerebellar granule neurons. J Biol Chem 279:51424-51432 (2004).
    • (2004) J Biol Chem , vol.279 , pp. 51424-51432
    • Davis, T.H.1    Chen, C.2    Isom, L.L.3
  • 91
    • 45749090058 scopus 로고    scopus 로고
    • Sodiumchannel beta1 subunit mutations associated with Brugada syndrome and cardiac conduction disease in humans.
    • WatanabeHet al.Sodiumchannel beta1 subunit mutations associated with Brugada syndrome and cardiac conduction disease in humans. J. Clin. Investig. 118:2260-2268 (2008).
    • (2008) J. Clin. Investig. , vol.118 , pp. 2260-2268
    • Watanabe, H.1
  • 92
    • 33845887196 scopus 로고    scopus 로고
    • Temporal lobe epilepsy and GEFS+ phenotypes associated with SCN1B mutations
    • Scheffer IE et al. Temporal lobe epilepsy and GEFS+ phenotypes associated with SCN1B mutations. Brain 130:100-109 (2007).
    • (2007) Brain , vol.130 , pp. 100-109
    • Scheffer, I.E.1
  • 93
    • 17344367657 scopus 로고    scopus 로고
    • Febrile seizures and generalized epilepsy associated with a mutation in the Na+-channel beta1 subunit gene SCN1B
    • Wallace R et al. Febrile seizures and generalized epilepsy associated with a mutation in the Na+-channel beta1 subunit gene SCN1B. Nat Genet 19:366-370 (1998).
    • (1998) Nat Genet , vol.19 , pp. 366-370
    • Wallace, R.1
  • 94
    • 73949096000 scopus 로고    scopus 로고
    • Voltage-gated sodium channels: Therapeutic targets for pain
    • Dib-Hajj SD, Black JA, Waxman SG. Voltage-gated sodium channels: Therapeutic targets for pain. Pain Med 10:1260-1269 (2009).
    • (2009) Pain Med , vol.10 , pp. 1260-1269
    • Dib-Hajj, S.D.1    Black, J.A.2    Waxman, S.G.3
  • 95
    • 40549135991 scopus 로고    scopus 로고
    • Mechanisms of disease: Sodium channels and neuroprotection in multiple sclerosis-current status
    • Waxman SG. Mechanisms of disease: Sodium channels and neuroprotection in multiple sclerosis-current status. Nat Clin Pract Neurol 4:159-169 (2008).
    • (2008) Nat Clin Pract Neurol , vol.4 , pp. 159-169
    • Waxman, S.G.1
  • 97
    • 78049435050 scopus 로고    scopus 로고
    • Voltage-gated sodium channel organization in neurons: Protein interactions and trafficking pathways.
    • Leterrier C, Brachet A, Fache MP, Dargent B. Voltage-gated sodium channel organization in neurons: Protein interactions and trafficking pathways. Neurosci Lett 486(2):92- 100 (2010).
    • (2010) Neurosci Lett , vol.486 , Issue.2 , pp. 92-100
    • Leterrier, C.1    Brachet, A.2    Fache, M.P.3    Dargent, B.4
  • 98
    • 23044441734 scopus 로고    scopus 로고
    • Voltage-gated sodium channel expression and potentiation of human breast cancer metastasis
    • Fraser SP et al. Voltage-gated sodium channel expression and potentiation of human breast cancer metastasis. Clin Cancer Res 11:5381-5389 (2005).
    • (2005) Clin Cancer Res , vol.11 , pp. 5381-5389
    • Fraser, S.P.1
  • 99
    • 0030608445 scopus 로고    scopus 로고
    • Spinal sensory neurons express multiple sodium channel alpha-subunit mRNAs
    • Black JA et al. Spinal sensory neurons express multiple sodium channel alpha-subunit mRNAs. Brain Res Mol Brain Res 43:117-131 (1996).
    • (1996) Brain Res Mol Brain Res , vol.43 , pp. 117-131
    • Black, J.A.1
  • 100
    • 0037109221 scopus 로고    scopus 로고
    • Na(v)1.5 underlies the "third TTX-R sodium current" in rat small DRG neurons.
    • Renganathan M, Dib-Hajj S, Waxman SG Na(v)1.5 underlies the "third TTX-R sodium current" in rat small DRG neurons. Brain Res Mol Brain Res 106:70-82 (2002).
    • (2002) Brain Res Mol Brain Res , vol.106 , pp. 70-82
    • Renganathan, M.1    Dib-Hajj, S.2    Waxman, S.G.3
  • 101
    • 0027933696 scopus 로고
    • Type III sodium channel mRNA is expressed in embryonic but not adult spinal sensory neurons, and is reexpressed following axotomy
    • Waxman SG, Kocsis JD, Black JA. Type III sodium channel mRNA is expressed in embryonic but not adult spinal sensory neurons, and is reexpressed following axotomy. J Neurophysiol 72:466-470 (1994).
    • (1994) J Neurophysiol , vol.72 , pp. 466-470
    • Waxman, S.G.1    Kocsis, J.D.2    Black, J.A.3
  • 102
    • 0035500407 scopus 로고    scopus 로고
    • The changes in expression of three subtypes of TTX sensitive sodium channels in sensory neurons after spinal nerve ligation
    • Kim CH, Oh, Y, Chung JM, Chung K. The changes in expression of three subtypes of TTX sensitive sodium channels in sensory neurons after spinal nerve ligation. Brain Res Mol Brain Res 95:153-161 (2001).
    • (2001) Brain Res Mol Brain Res , vol.95 , pp. 153-161
    • Kim, C.H.1    Oh, Y.2    Chung, J.M.3    Chung, K.4
  • 103
    • 0035882277 scopus 로고    scopus 로고
    • Nav1.3 sodium channels: Rapid repriming and slow closed-state inactivation display quantitative differences after expression in a mammalian cell line and in spinal sensory neurons.
    • Cummins TR et al. Nav1.3 sodium channels: Rapid repriming and slow closed-state inactivation display quantitative differences after expression in a mammalian cell line and in spinal sensory neurons. J Neurosci 21, 5952-5961 (2001).
    • (2001) J Neurosci , vol.21 , pp. 5952-5961
    • Cummins, T.R.1
  • 104
    • 0029907091 scopus 로고    scopus 로고
    • Down-regulation of transcripts for Na channel alpha-SNS in spinal sensory neurons following axotomy
    • Dib-Hajj S, Black JA, Felts P, Waxman SG. Down-regulation of transcripts for Na channel alpha-SNS in spinal sensory neurons following axotomy. Proc Natl Acad Sci USA 93:14950-14954 (1996).
    • (1996) Proc Natl Acad Sci USA , vol.93 , pp. 14950-14954
    • Dib-Hajj, S.1    Black, J.A.2    Felts, P.3    Waxman, S.G.4
  • 105
    • 2442651551 scopus 로고    scopus 로고
    • Altered sodium channel expression in second-order spinal sensory neurons contributes to pain after peripheral nerve injury
    • Hains BC, Saab CY, Klein JP, Craner MJ, Waxman SG. Altered sodium channel expression in second-order spinal sensory neurons contributes to pain after peripheral nerve injury. J Neurosci 24:4832-4839 (2004).
    • (2004) J Neurosci , vol.24 , pp. 4832-4839
    • Hains, B.C.1    Saab, C.Y.2    Klein, J.P.3    Craner, M.J.4    Waxman, S.G.5
  • 106
    • 26044434843 scopus 로고    scopus 로고
    • Changes in electrophysiological properties and sodium channel Nav1.3 expression in thalamic neurons after spinal cord injury.
    • Hains BC, Saab CY, Waxman SG. Changes in electrophysiological properties and sodium channel Nav1.3 expression in thalamic neurons after spinal cord injury. Brain 128, 2359-2371 (2005).
    • (2005) Brain , vol.128 , pp. 2359-2371
    • Hains, B.C.1    Saab, C.Y.2    Waxman, S.G.3
  • 107
    • 33751014049 scopus 로고    scopus 로고
    • Nerve injury induces robust allodynia and ectopic discharges in Nav1.3 null mutant mice.
    • Nassar MA. et al. Nerve injury induces robust allodynia and ectopic discharges in Nav1.3 null mutant mice. Mol Pain 2:33 (2006).
    • (2006) Mol Pain , vol.2 , pp. 33
    • Nassar, M.A.1
  • 108
    • 28844448678 scopus 로고    scopus 로고
    • Relationship between sodium channel NaV1.3 expression and neuropathic pain behavior in rats
    • Lindia JA, Kohler MG, Martin WJ, Abbadie, C. Relationship between sodium channel NaV1.3 expression and neuropathic pain behavior in rats. Pain 117:145-153 (2005).
    • (2005) Pain , vol.117 , pp. 145-153
    • Lindia, J.A.1    Kohler, M.G.2    Martin, W.J.3    Abbadie, C.4
  • 109
    • 63449107353 scopus 로고    scopus 로고
    • Voltage-gated sodium channels in pain states: Role in pathophysiology and targets for treatment
    • Dib-Hajj SD et al. Voltage-gated sodium channels in pain states: Role in pathophysiology and targets for treatment. Brain Res Rev 60:65-83 (2009).
    • (2009) Brain Res Rev , vol.60 , pp. 65-83
    • Dib-Hajj, S.D.1
  • 110
    • 12644288297 scopus 로고    scopus 로고
    • Identification of PN1, a predominant voltage-dependent sodium channel expressed principally in peripheral neurons.
    • Toledo-Aral JJ et al. Identification of PN1, a predominant voltage-dependent sodium channel expressed principally in peripheral neurons. Proc Natl Acad Sci USA 94:1527- 1532 (1997).
    • (1997) Proc Natl Acad Sci USA , vol.94 , pp. 1527-1532
    • Toledo-Aral, J.J.1
  • 111
    • 0031127498 scopus 로고    scopus 로고
    • Sodium channel alphasubunit mRNAs I, II, III, NaG, Na6 and hNE (PN1): Different expression patterns in developing rat nervous system
    • Felts PA, Yokoyama S, Dib-Hajj S, Black JA, Waxman SG. Sodium channel alphasubunit mRNAs I, II, III, NaG, Na6 and hNE (PN1): Different expression patterns in developing rat nervous system. Brain Res Mol Brain Res 45:71-82 (1997).
    • (1997) Brain Res Mol Brain Res , vol.45 , pp. 71-82
    • Felts, P.A.1    Yokoyama, S.2    Dib-Hajj, S.3    Black, J.A.4    Waxman, S.G.5
  • 112
    • 0028985863 scopus 로고
    • Structure and functional expression of a new member of the tetrodotoxin-sensitive voltage-activated sodium channel family from human neuroendocrine cells
    • 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 14:1084-1090 (1995).
    • (1995) EMBO J , vol.14 , pp. 1084-1090
    • Klugbauer, N.1    Lacinova, L.2    Flockerzi, V.3    Hofmann, F.4
  • 113
    • 12644256626 scopus 로고    scopus 로고
    • A novel tetrodotoxin-sensitive, voltage-gated sodium channel expressed in rat and human dorsal root ganglia
    • Sangameswaran L et al. A novel tetrodotoxin-sensitive, voltage-gated sodium channel expressed in rat and human dorsal root ganglia. J Biol Chem 272:14805-14809 (1997).
    • (1997) J Biol Chem , vol.272 , pp. 14805-14809
    • Sangameswaran, L.1
  • 114
    • 0032400799 scopus 로고    scopus 로고
    • Slow closed-state inactivation: A novel mechanism underlying ramp currents in cells expressing the hNE/PN1 sodium channel
    • 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 18:9607-9619 (1998).
    • (1998) J Neurosci , vol.18 , pp. 9607-9619
    • Cummins, T.R.1    Howe, J.R.2    Waxman, S.G.3
  • 115
    • 34548476521 scopus 로고    scopus 로고
    • The roles of sodium channels in nociception: Implications for mechanisms of pain
    • Cummins TR, Sheets PL, Waxman SG. The roles of sodium channels in nociception: Implications for mechanisms of pain. Pain 131:243-257 (2007).
    • (2007) Pain , vol.131 , pp. 243-257
    • Cummins, T.R.1    Sheets, P.L.2    Waxman, S.G.3
  • 116
    • 0141646635 scopus 로고    scopus 로고
    • Distinct repriming and closed-state inactivation kinetics of Nav1.6 and Nav1.7 sodium channels in mouse spinal sensory neurons
    • 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 551:741-750 (2003).
    • (2003) J Physiol , vol.551 , pp. 741-750
    • Herzog, R.I.1    Cummins, T.R.2    Ghassemi, F.3    Dib-Hajj, S.D.4    Waxman, S.G.5
  • 117
    • 33847168937 scopus 로고    scopus 로고
    • SCN9A mutations in paroxysmal extreme pain disorder: Allelic variants underlie distinct channel defects and phenotypes
    • Fertleman CR et al. SCN9A mutations in paroxysmal extreme pain disorder: Allelic variants underlie distinct channel defects and phenotypes. Neuron 52:767-774 (2006).
    • (2006) Neuron , vol.52 , pp. 767-774
    • Fertleman, C.R.1
  • 118
    • 20444408342 scopus 로고    scopus 로고
    • Erythromelalgia: A hereditary pain syndrome enters the molecular era
    • Waxman SG, Dib-Hajj SD. Erythromelalgia: A hereditary pain syndrome enters the molecular era. Ann Neurol 57:785-788 (2005).
    • (2005) Ann Neurol , vol.57 , pp. 785-788
    • Waxman, S.G.1    Dib-Hajj, S.D.2
  • 119
    • 33845901486 scopus 로고    scopus 로고
    • An SCN9A channelopathy causes congenital inability to experience pain
    • Cox JJ et al. An SCN9A channelopathy causes congenital inability to experience pain. Nature 444:894-898 (2006).
    • (2006) Nature , vol.444 , pp. 894-898
    • Cox, J.J.1
  • 120
    • 34247874778 scopus 로고    scopus 로고
    • Loss-of-function mutations in the Nav1.7 gene underlie congenital indifference to pain in multiple human populations
    • Goldberg YP et al. Loss-of-function mutations in the Nav1.7 gene underlie congenital indifference to pain in multiple human populations. Clin Genet 71:311-319 (2007).
    • (2007) Clin Genet , vol.71 , pp. 311-319
    • Goldberg, Y.P.1
  • 121
    • 77949896163 scopus 로고    scopus 로고
    • Familial pain syndromes from mutations of the NaV1.7 sodium channel
    • Fischer TZ, Waxman SG. Familial pain syndromes from mutations of the NaV1.7 sodium channel. Ann NY Acad Sci 1184:196-207 (2010).
    • (2010) Ann NY Acad Sci , vol.1184 , pp. 196-207
    • Fischer, T.Z.1    Waxman, S.G.2
  • 122
    • 1542498432 scopus 로고    scopus 로고
    • Changes in the expression of tetrodotoxin-sensitive sodium channels within dorsal root ganglia neurons in inflammatory pain
    • Black JA, Liu S, Tanaka M, Cummins TR, Waxman SG. Changes in the expression of tetrodotoxin-sensitive sodium channels within dorsal root ganglia neurons in inflammatory pain. Pain 108:237-247 (2004).
    • (2004) Pain , vol.108 , pp. 237-247
    • Black, J.A.1    Liu, S.2    Tanaka, M.3    Cummins, T.R.4    Waxman, S.G.5
  • 123
    • 0028947293 scopus 로고
    • A single pulse of nerve growth factor triggers long-term neuronal excitability through sodium channel gene induction
    • Toledo-Aral JJ, Brehm P, Halegoua S, Mandel G. A single pulse of nerve growth factor triggers long-term neuronal excitability through sodium channel gene induction. Neuron 14:607-611 (1995).
    • (1995) Neuron , vol.14 , pp. 607-611
    • Toledo-Aral, J.J.1    Brehm, P.2    Halegoua, S.3    Mandel, G.4
  • 124
    • 0034900781 scopus 로고    scopus 로고
    • Contribution of Na(v)1.8 sodium channels to action potential electrogenesis in DRG neurons
    • Renganathan M, Cummins TR, Waxman SG. Contribution of Na(v)1.8 sodium channels to action potential electrogenesis in DRG neurons. J Neurophysiol 86:629-640 (2001).
    • (2001) J Neurophysiol , vol.86 , pp. 629-640
    • Renganathan, M.1    Cummins, T.R.2    Waxman, S.G.3
  • 125
    • 0036895970 scopus 로고    scopus 로고
    • Roles of tetrodotoxin (TTX)-sensitive Na+ current, TTX-resistant Na+ current, and Ca2+ current in the action potentials of nociceptive sensory neurons
    • Blair NT, Bean BP. Roles of tetrodotoxin (TTX)-sensitive Na+ current, TTX-resistant Na+ current, and Ca2+ current in the action potentials of nociceptive sensory neurons. J Neurosci 22:10277-10290 (2002).
    • (2002) J Neurosci , vol.22 , pp. 10277-10290
    • Blair, N.T.1    Bean, B.P.2
  • 126
    • 13344294399 scopus 로고    scopus 로고
    • Structure and function of a novel voltage-gated, tetrodotoxinresistant sodium channel specific to sensory neurons
    • Sangameswaran L et al. Structure and function of a novel voltage-gated, tetrodotoxinresistant sodium channel specific to sensory neurons. J Biol Chem 271:5953-5956 (1996).
    • (1996) J Biol Chem , vol.271 , pp. 5953-5956
    • Sangameswaran, L.1
  • 127
    • 0030041548 scopus 로고    scopus 로고
    • A tetrodotoxin-resistant voltage-gated sodium channel expressed by sensory neurons
    • Akopian AN, Sivilotti L, Wood JN. A tetrodotoxin-resistant voltage-gated sodium channel expressed by sensory neurons. Nature 379:257-262 (1996).
    • (1996) Nature , vol.379 , pp. 257-262
    • Akopian, A.N.1    Sivilotti, L.2    Wood, J.N.3
  • 128
    • 0028088712 scopus 로고
    • Slow sodium conductances of dorsal root ganglion neurons: intraneuronal homogeneity and interneuronal heterogeneity
    • Rizzo MA, Kocsis JD, Waxman SG. Slow sodium conductances of dorsal root ganglion neurons: intraneuronal homogeneity and interneuronal heterogeneity. J Neurophysiol 72:2796-2815 (1994).
    • (1994) J Neurophysiol , vol.72 , pp. 2796-2815
    • Rizzo, M.A.1    Kocsis, J.D.2    Waxman, S.G.3
  • 129
    • 0026772987 scopus 로고
    • Differential properties of tetrodotoxin-sensitive and tetrodotoxinresistant sodium channels in rat dorsal root ganglion neurons
    • Roy ML, Narahashi T. Differential properties of tetrodotoxin-sensitive and tetrodotoxinresistant sodium channels in rat dorsal root ganglion neurons. J Neurosci 12:2104-2111 (1992).
    • (1992) J Neurosci , vol.12 , pp. 2104-2111
    • Roy, M.L.1    Narahashi, T.2
  • 130
    • 33846915664 scopus 로고    scopus 로고
    • Differential slow inactivation and use-dependent inhibition of Nav1.8 channels contribute to distinct firing properties in IB4+ and IB4- DRG neurons
    • Choi JS, Dib-Hajj SD, Waxman SG. Differential slow inactivation and use-dependent inhibition of Nav1.8 channels contribute to distinct firing properties in IB4+ and IB4- DRG neurons. J Neurophysiol 97:1258-1265 (2007).
    • (2007) J Neurophysiol , vol.97 , pp. 1258-1265
    • Choi, J.S.1    Dib-Hajj, S.D.2    Waxman, S.G.3
  • 131
    • 33744454923 scopus 로고    scopus 로고
    • A single sodium channel mutation produces hyper- or hypoexcitability in different types of neurons
    • RushAM et al. A single sodium channel mutation produces hyper- or hypoexcitability in different types of neurons. Proc Natl Acad Sci USA 103:8245-8250 (2006).
    • (2006) Proc Natl Acad Sci USA , vol.103 , pp. 8245-8250
    • Rush, A.M.1
  • 132
    • 0036712354 scopus 로고    scopus 로고
    • Prostaglandin E(2) modulates TTX-R I(Na) in rat colonic sensory neurons
    • Gold MS, Zhang L, Wrigley DL, Traub RJ. Prostaglandin E(2) modulates TTX-R I(Na) in rat colonic sensory neurons. J Neurophysiol 88:1512-1522 (2002).
    • (2002) J Neurophysiol , vol.88 , pp. 1512-1522
    • Gold, M.S.1    Zhang, L.2    Wrigley, D.L.3    Traub, R.J.4
  • 133
    • 0030581103 scopus 로고    scopus 로고
    • DAMGO inhibits prostaglandin E2-induced potentiation of a TTX-resistant Na+ current in rat sensory neurons in vitro
    • Gold MS, Levine JD. DAMGO inhibits prostaglandin E2-induced potentiation of a TTX-resistant Na+ current in rat sensory neurons in vitro. Neurosci Lett 212:83-86 (1996).
    • (1996) Neurosci Lett , vol.212 , pp. 83-86
    • Gold, M.S.1    Levine, J.D.2
  • 135
    • 33748888226 scopus 로고    scopus 로고
    • Dissecting the role of sodium currents in visceral sensory neurons in a model of chronic hyperexcitability using Nav1.8 and Nav1.9 null mice
    • Hillsley K et al. Dissecting the role of sodium currents in visceral sensory neurons in a model of chronic hyperexcitability using Nav1.8 and Nav1.9 null mice. J Physiol 576:257-267 (2006).
    • (2006) J Physiol , vol.576 , pp. 257-267
    • Hillsley, K.1
  • 136
    • 0036813714 scopus 로고    scopus 로고
    • Deficits in visceral pain and referred hyperalgesia in Nav1.8 (SNS/PN3)-null mice
    • Laird JM, Souslova V, Wood JN, Cervero F. Deficits in visceral pain and referred hyperalgesia in Nav1.8 (SNS/PN3)-null mice. J Neurosci 22:8352-8356 (2002).
    • (2002) J Neurosci , vol.22 , pp. 8352-8356
    • Laird, J.M.1    Souslova, V.2    Wood, J.N.3    Cervero, F.4
  • 137
    • 0037107437 scopus 로고    scopus 로고
    • Ceramide, a putative second messenger for nerve growth factor, modulates the TTX-resistant Na(+) current and delayed rectifier K(+) current in rat sensory neurons
    • Zhang YH, Vasko MR, Nicol GD. Ceramide, a putative second messenger for nerve growth factor, modulates the TTX-resistant Na(+) current and delayed rectifier K(+) current in rat sensory neurons. J Physiol 544:385-402 (2002).
    • (2002) J Physiol , vol.544 , pp. 385-402
    • Zhang, Y.H.1    Vasko, M.R.2    Nicol, G.D.3
  • 138
    • 33845187874 scopus 로고    scopus 로고
    • Neuropathic pain develops normally in mice lacking both Na(v)1.7 and Na(v)1.8.
    • Nassar MA, Levato A, Stirling LC, Wood JN. Neuropathic pain develops normally in mice lacking both Na(v)1.7 and Na(v)1.8. Mol Pain 1:24 (2005).
    • (2005) Mol Pain , vol.1 , pp. 24
    • Nassar, M.A.1    Levato, A.2    Stirling, L.C.3    Wood, J.N.4
  • 139
    • 34250306409 scopus 로고    scopus 로고
    • Sensory neuron sodium channel Nav1.8 is essential for pain at low temperatures
    • Zimmermann K et al. Sensory neuron sodium channel Nav1.8 is essential for pain at low temperatures. Nature 447:855-858 (2007).
    • (2007) Nature , vol.447 , pp. 855-858
    • Zimmermann, K.1
  • 140
    • 48749123103 scopus 로고    scopus 로고
    • The cell and molecular basis of mechanical, cold, and inflammatory pain
    • Abrahamsen B et al. The cell and molecular basis of mechanical, cold, and inflammatory pain. Science 321:702-705 (2008).
    • (2008) Science , vol.321 , pp. 702-705
    • Abrahamsen, B.1
  • 141
    • 0032555278 scopus 로고    scopus 로고
    • NaN, a novel voltage-gated Na channel, is expressed preferentially in peripheral sensory neurons and down-regulated after axotomy
    • Dib-Hajj SD, Tyrrell, L, Black JA,Waxman SG. NaN, a novel voltage-gated Na channel, is expressed preferentially in peripheral sensory neurons and down-regulated after axotomy. Proc Natl Acad Sci USA 95:8963-8968 (1998).
    • (1998) Proc Natl Acad Sci USA , vol.95 , pp. 8963-8968
    • Dib-Hajj, S.D.1    Tyrrell, L.2    Black, J.A.3    Waxman, S.G.4
  • 142
    • 0033572137 scopus 로고    scopus 로고
    • A novel persistent tetrodotoxin-resistant sodium current in SNS-null and wild-type small primary sensory neurons.
    • Cummins TR et al. A novel persistent tetrodotoxin-resistant sodium current in SNS-null and wild-type small primary sensory neurons. J Neurosci 19:RC43 (1999).
    • (1999) J Neurosci , vol.19
    • Cummins, T.R.1
  • 143
    • 21544455502 scopus 로고    scopus 로고
    • Contribution of the tetrodotoxin-resistant voltage-gated sodium channel NaV1.9 to sensory transmission and nociceptive behavior
    • Priest BT et al. Contribution of the tetrodotoxin-resistant voltage-gated sodium channel NaV1.9 to sensory transmission and nociceptive behavior. Proc Natl Acad Sci USA 102:9382-9387 (2005).
    • (2005) Proc Natl Acad Sci USA , vol.102 , pp. 9382-9387
    • Priest, B.T.1
  • 144
    • 77951911566 scopus 로고    scopus 로고
    • Exploring the role of nociceptor-specific sodium channels in pain transmission using Nav1.8 and Nav1.9 knockout mice
    • Leo S, D'Hooge, R, Meert, T. Exploring the role of nociceptor-specific sodium channels in pain transmission using Nav1.8 and Nav1.9 knockout mice. Behav Brain Res 208:149-157 (2009).
    • (2009) Behav Brain Res , vol.208 , pp. 149-157
    • Leo, S.1    D'Hooge, R.2    Meert, T.3
  • 145
    • 70349667578 scopus 로고    scopus 로고
    • Skeletal muscle channelopathies: New insights into the periodic paralyses and nondystrophic myotonias
    • Platt D, Griggs R. Skeletal muscle channelopathies: New insights into the periodic paralyses and nondystrophic myotonias. Curr Opin Neurol 22:524-531 (2009).
    • (2009) Curr Opin Neurol , vol.22 , pp. 524-531
    • Platt, D.1    Griggs, R.2
  • 146
    • 58249084640 scopus 로고    scopus 로고
    • Therapeutic approaches to ion channel diseases
    • Camerino DC et al. Therapeutic approaches to ion channel diseases. Advances in Genetics 64:81-145 (2008).
    • (2008) Advances in Genetics , vol.64 , pp. 81-145
    • Camerino, D.C.1
  • 147
    • 0033374373 scopus 로고    scopus 로고
    • Neurological channelopathies: Diagnosis and therapy in the new millennium
    • Davies NP, Hanna MG. Neurological channelopathies: Diagnosis and therapy in the new millennium. Ann Med 31:406-420 (1999).
    • (1999) Ann Med , vol.31 , pp. 406-420
    • Davies, N.P.1    Hanna, M.G.2
  • 149
    • 0029116230 scopus 로고
    • Cardiac sodium channel mutations in patients with long QT syndrome, an inherited cardiac arrhythmia
    • Wang Q et al. Cardiac sodium channel mutations in patients with long QT syndrome, an inherited cardiac arrhythmia. Hum Mol Genet 4:1603-1607 (1995).
    • (1995) Hum Mol Genet , vol.4 , pp. 1603-1607
    • Wang, Q.1
  • 150
    • 67649547603 scopus 로고    scopus 로고
    • Sodium channel mutations and arrhythmias
    • Ruan Y, Liu N, Priori SG. Sodium channel mutations and arrhythmias. Nat Rev Cardiol 6:337-348 (2009).
    • (2009) Nat Rev Cardiol , vol.6 , pp. 337-348
    • Ruan, Y.1    Liu, N.2    Priori, S.G.3
  • 151
    • 61849154781 scopus 로고    scopus 로고
    • Genetic Na+ channelopathies and sinus node dysfunction
    • Lei M, Huang CL, Zhang Y. Genetic Na+ channelopathies and sinus node dysfunction. Prog Biophys Mol Biol 98:171-178 (2008).
    • (2008) Prog Biophys Mol Biol , vol.98 , pp. 171-178
    • Lei, M.1    Huang, C.L.2    Zhang, Y.3
  • 152
    • 0032546384 scopus 로고    scopus 로고
    • Genetic basis and molecular mechanism for idiopathic ventricular fibrillation
    • Chen Q et al. Genetic basis and molecular mechanism for idiopathic ventricular fibrillation. Nature 392:293-296 (1998).
    • (1998) Nature , vol.392 , pp. 293-296
    • Chen, Q.1
  • 153
    • 0345308446 scopus 로고    scopus 로고
    • Inherited arrhythmia syndromes: Applying the molecular biology and genetic to the clinical management
    • Priori SG, Napolitano C, Vicentini A. Inherited arrhythmia syndromes: Applying the molecular biology and genetic to the clinical management. J Interv Card Electrophysiol 9:93-101 (2003).
    • (2003) J Interv Card Electrophysiol , vol.9 , pp. 93-101
    • Priori, S.G.1    Napolitano, C.2    Vicentini, A.3
  • 154
    • 0021363029 scopus 로고
    • Class 1 antiarrhythmic drugs-characteristic electrocardiograph differences when assessed by atrial and ventricular pacing
    • Milne JR et al. Class 1 antiarrhythmic drugs-characteristic electrocardiograph differences when assessed by atrial and ventricular pacing. Eur Heart J 5:99-107 (1984).
    • (1984) Eur Heart J , vol.5 , pp. 99-107
    • Milne, J.R.1
  • 156
  • 157
    • 58249130592 scopus 로고    scopus 로고
    • A catalog of SCN1A variants
    • Lossin C. A catalog of SCN1A variants. Brain Dev 31:114-130 (2009).
    • (2009) Brain Dev , vol.31 , pp. 114-130
    • Lossin, C.1
  • 158
    • 33947123754 scopus 로고    scopus 로고
    • The spectrum of SCN1A-related infantile epileptic encephalopathies
    • Harkin LA et al. The spectrum of SCN1A-related infantile epileptic encephalopathies. Brain 130:843-852 (2007).
    • (2007) Brain , vol.130 , pp. 843-852
    • Harkin, L.A.1
  • 159
    • 39149136856 scopus 로고    scopus 로고
    • Mutation of sodium channel SCN3A in a patient with cryptogenic pediatric partial epilepsy
    • Holland KD et al. Mutation of sodium channel SCN3A in a patient with cryptogenic pediatric partial epilepsy. Neurosci Lett 433:65-70 (2008).
    • (2008) Neurosci Lett , vol.433 , pp. 65-70
    • Holland, K.D.1
  • 160
    • 77954658120 scopus 로고    scopus 로고
    • A sodium channel mutation linked to epilepsy increases ramp and persistent current of Nav1.3 and induces hyperexcitability in hippocampal neurons
    • Estacion M, Gasser A, Dib-Hajj SD,Waxman SG. A sodium channel mutation linked to epilepsy increases ramp and persistent current of Nav1.3 and induces hyperexcitability in hippocampal neurons. Exp. Neuro. 224:362-368 (2010).
    • (2010) Exp. Neuro. , vol.224 , pp. 362-368
    • Estacion, M.1    Gasser, A.2    Dib-Hajj, S.D.3    Waxman, S.G.4
  • 161
    • 58149215915 scopus 로고    scopus 로고
    • Role of hippocampal sodium channel Nav1.6 in kindling epileptogenesis
    • Blumenfeld H et al. Role of hippocampal sodium channel Nav1.6 in kindling epileptogenesis. Epilepsia 50:44-55 (2009).
    • (2009) Epilepsia , vol.50 , pp. 44-55
    • Blumenfeld, H.1
  • 162
    • 77954514571 scopus 로고    scopus 로고
    • Sodium channel gene family: Epilepsy mutations, gene interactions and modifier effects
    • Meisler MH, O'Brien JE, Sharkey LM. Sodium channel gene family: Epilepsy mutations, gene interactions and modifier effects. J Physiol 588:1841-1848 (2010).
    • (2010) J Physiol , vol.588 , pp. 1841-1848
    • Meisler, M.H.1    O'Brien, J.E.2    Sharkey, L.M.3
  • 163
    • 60849124532 scopus 로고    scopus 로고
    • Regulation of persistent Na current by interactions between {beta} subunits of voltage-gated Na channels
    • Aman TK et al. Regulation of persistent Na current by interactions between {beta} subunits of voltage-gated Na channels. J Neurosci 29:2027-2042 (2009).
    • (2009) J Neurosci , vol.29 , pp. 2027-2042
    • Aman, T.K.1
  • 164
    • 63849160970 scopus 로고    scopus 로고
    • Hippocampal Navb3 expression in patients with temporal lobe epilepsy
    • Van Gassen KLI et al. Hippocampal Navb3 expression in patients with temporal lobe epilepsy. Epilepsia 50:957-962 (2009).
    • (2009) Epilepsia , vol.50 , pp. 957-962
    • Van Gassen, K.L.I.1
  • 165
    • 45649085478 scopus 로고    scopus 로고
    • How do mutant Nav1.1 sodium channels cause epilepsy?
    • Ragsdale DS. How do mutant Nav1.1 sodium channels cause epilepsy? Brain Res Rev 58:149-159 (2008).
    • (2008) Brain Res Rev , vol.58 , pp. 149-159
    • Ragsdale, D.S.1
  • 167
    • 37349043190 scopus 로고    scopus 로고
    • Voltage-gated sodium channel Nav1.1, Nav1.3 and beta1 subunit were upregulated in the hippocampus of spontaneously epileptic rat
    • Guo F et al. Voltage-gated sodium channel Nav1.1, Nav1.3 and beta1 subunit were upregulated in the hippocampus of spontaneously epileptic rat. Brain Res Bull 75:179-187 (2008).
    • (2008) Brain Res Bull , vol.75 , pp. 179-187
    • Guo, F.1
  • 168
    • 3142774111 scopus 로고    scopus 로고
    • The neurobiology of antiepileptic drugs for the treatment of nonepileptic conditions
    • Rogawski MA Loscher W. The neurobiology of antiepileptic drugs for the treatment of nonepileptic conditions. Nat Med 10:685-692 (2004).
    • (2004) Nat Med , vol.10 , pp. 685-692
    • Rogawski, M.A.1    Loscher, W.2
  • 169
    • 3042798891 scopus 로고    scopus 로고
    • The neurobiology of antiepileptic drugs
    • Rogawski MA, Loscher W. The neurobiology of antiepileptic drugs. Nat Rev Neurosci 5:553-564 (2004).
    • (2004) Nat Rev Neurosci , vol.5 , pp. 553-564
    • Rogawski, M.A.1    Loscher, W.2
  • 170
    • 0029787945 scopus 로고    scopus 로고
    • Common molecular determinants of local anesthetic, antiarrhythmic, and anticonvulsant block of voltage-gated Na+ channels
    • Ragsdale DS, McPhee JC, Scheuer T, CatterallWA. Common molecular determinants of local anesthetic, antiarrhythmic, and anticonvulsant block of voltage-gated Na+ channels. Proc Natl Acad Sci USA 93:9270-9275 (1996).
    • (1996) Proc Natl Acad Sci USA , vol.93 , pp. 9270-9275
    • Ragsdale, D.S.1    McPhee, J.C.2    Scheuer, T.3    Catterall, W.A.4
  • 172
    • 16044370232 scopus 로고    scopus 로고
    • Familial hemiplegic migraine and episodic ataxia type-2 are caused by mutations in the Ca2+ channel gene CACNL1A4
    • Ophoff RA et al. Familial hemiplegic migraine and episodic ataxia type-2 are caused by mutations in the Ca2+ channel gene CACNL1A4. Cell 87:543-552 (1996).
    • (1996) Cell , vol.87 , pp. 543-552
    • Ophoff, R.A.1
  • 173
    • 0037312922 scopus 로고    scopus 로고
    • Haploinsufficiency of ATP1A2 encoding the Na+/K+ pump [alpha]2 subunit associated with familial hemiplegic migraine type 2
    • Fusco MD et al. Haploinsufficiency of ATP1A2 encoding the Na+/K+ pump [alpha]2 subunit associated with familial hemiplegic migraine type 2. Nat Genet 33:192-196 (2003).
    • (2003) Nat Genet , vol.33 , pp. 192-196
    • Fusco, M.D.1
  • 174
    • 23044459961 scopus 로고    scopus 로고
    • Mutation in the neuronal voltage-gated sodium channel SCN1A in familial hemiplegic migraine
    • Martin D et al. Mutation in the neuronal voltage-gated sodium channel SCN1A in familial hemiplegic migraine. Lancet 366:371-377 (2005).
    • (2005) Lancet , vol.366 , pp. 371-377
    • Martin, D.1
  • 175
    • 47749104145 scopus 로고    scopus 로고
    • Divergent sodium channel defects in familial hemiplegic migraine
    • Kahlig KM et al. Divergent sodium channel defects in familial hemiplegic migraine. Proc Natl Acad Sci USA 105:9799-9804 (2008).
    • (2008) Proc Natl Acad Sci USA , vol.105 , pp. 9799-9804
    • Kahlig, K.M.1
  • 176
    • 0034069651 scopus 로고    scopus 로고
    • Mutations of SCN1A, encoding a neuronal sodium channel, in two families with GEFS+2
    • Escayg A et al. Mutations of SCN1A, encoding a neuronal sodium channel, in two families with GEFS+2. Nat Genet 24:343-345 (2000).
    • (2000) Nat Genet , vol.24 , pp. 343-345
    • Escayg, A.1
  • 177
    • 0347479237 scopus 로고    scopus 로고
    • Epilepsy-associated dysfunction in the voltage-gated neuronal sodium channel SCN1A
    • Lossin C et al. Epilepsy-associated dysfunction in the voltage-gated neuronal sodium channel SCN1A. J Neurosci 23:11289-11295 (2003).
    • (2003) J Neurosci , vol.23 , pp. 11289-11295
    • Lossin, C.1
  • 178
    • 3342929286 scopus 로고    scopus 로고
    • Noninactivating voltagegated sodium channels in severe myoclonic epilepsy of infancy
    • Rhodes TH, Lossin C, Vanoye CG, Wang DW, George AL. Noninactivating voltagegated sodium channels in severe myoclonic epilepsy of infancy. Proc Natl Acad Sci USA 101:11147-11152 (2004).
    • (2004) Proc Natl Acad Sci USA , vol.101 , pp. 11147-11152
    • Rhodes, T.H.1    Lossin, C.2    Vanoye, C.G.3    Wang, D.W.4    George, A.L.5
  • 179
    • 59149098688 scopus 로고    scopus 로고
    • First mutation in the voltage-gated NaV1.1 subunit gene SCN1Awith cooccurring familial hemiplegic migraine and epilepsy
    • Castro MJ et al. First mutation in the voltage-gated NaV1.1 subunit gene SCN1Awith cooccurring familial hemiplegic migraine and epilepsy. Cephalalgia 29:308-313 (2009).
    • (2009) Cephalalgia , vol.29 , pp. 308-313
    • Castro, M.J.1
  • 180
    • 0032512628 scopus 로고    scopus 로고
    • Sodium channel protein expression enhances the invasiveness of rat and human prostate cancer cells
    • Smith P et al. Sodium channel protein expression enhances the invasiveness of rat and human prostate cancer cells. FEBS Lett 423:19-24 (1998).
    • (1998) FEBS Lett , vol.423 , pp. 19-24
    • Smith, P.1
  • 181
    • 0030896235 scopus 로고    scopus 로고
    • Expression and functional analysis of voltage-activated Na+ channels in human prostate cancer cell lines and their contribution to invasion in vitro
    • Laniado ME et al. Expression and functional analysis of voltage-activated Na+ channels in human prostate cancer cell lines and their contribution to invasion in vitro. Am J Pathol 150:1213-1221 (1997).
    • (1997) Am J Pathol , vol.150 , pp. 1213-1221
    • Laniado, M.E.1
  • 182
    • 0032079293 scopus 로고    scopus 로고
    • Expression of skeletal muscle-type voltage-gated Na+ channel in rat and human prostate cancer cell lines
    • Diss JK et al. Expression of skeletal muscle-type voltage-gated Na+ channel in rat and human prostate cancer cell lines. FEBS Lett 427:5-10 (1998).
    • (1998) FEBS Lett , vol.427 , pp. 5-10
    • Diss, J.K.1
  • 183
    • 0034908909 scopus 로고    scopus 로고
    • Expression profiles of voltagegated Na(+) channel alpha-subunit genes in rat and human prostate cancer cell lines
    • Diss JK, Archer SN, Hirano J, Fraser SP, Djamgoz MB. Expression profiles of voltagegated Na(+) channel alpha-subunit genes in rat and human prostate cancer cell lines. Prostate 48:165-178 (2001).
    • (2001) Prostate , vol.48 , pp. 165-178
    • Diss, J.K.1    Archer, S.N.2    Hirano, J.3    Fraser, S.P.4    Djamgoz, M.B.5
  • 184
    • 1642375706 scopus 로고    scopus 로고
    • Voltage-gated Na+ channels confer invasive properties on human prostate cancer cells
    • Bennett ES, Smith BA, Harper JM. Voltage-gated Na+ channels confer invasive properties on human prostate cancer cells. Pflugers Arch 447:908-914 (2004).
    • (2004) Pflugers Arch , vol.447 , pp. 908-914
    • Bennett, E.S.1    Smith, B.A.2    Harper, J.M.3
  • 185
    • 27944442711 scopus 로고    scopus 로고
    • A potential novel marker for human prostate cancer: Voltage-gated sodium channel expression in vivo
    • Diss JK et al. A potential novel marker for human prostate cancer: Voltage-gated sodium channel expression in vivo. Prostate Cancer Prostatic Dis 8:266-273 (2005).
    • (2005) Prostate Cancer Prostatic Dis , vol.8 , pp. 266-273
    • Diss, J.K.1
  • 186
    • 3042608228 scopus 로고    scopus 로고
    • Voltage-gated Na+ channels: Multiplicity of expression, plasticity, functional implications and pathophysiological aspects
    • Diss JK, Fraser SP, Djamgoz MB. Voltage-gated Na+ channels: Multiplicity of expression, plasticity, functional implications and pathophysiological aspects. Eur Biophys J 33:180-193 (2004).
    • (2004) Eur Biophys J , vol.33 , pp. 180-193
    • Diss, J.K.1    Fraser, S.P.2    Djamgoz, M.B.3
  • 187
    • 1642326838 scopus 로고    scopus 로고
    • Involvement of a novel fast inward sodium current in the invasion capacity of a breast cancer cell line
    • Roger S, Besson P, Le Guennec JY. Involvement of a novel fast inward sodium current in the invasion capacity of a breast cancer cell line. Biochim Biophys Acta 1616:107-111 (2003).
    • (2003) Biochim Biophys Acta , vol.1616 , pp. 107-111
    • Roger, S.1    Besson, P.2    Le Guennec, J.Y.3
  • 189
    • 0017342059 scopus 로고
    • Density of sodium channels in mammalian myelinated nerve fibers and nature of the axonal membrane under the myelin sheath
    • Ritchie JM, Rogart RB. Density of sodium channels in mammalian myelinated nerve fibers and nature of the axonal membrane under the myelin sheath. Proc Natl Acad Sci USA 74:211-215 (1977).
    • (1977) Proc Natl Acad Sci USA , vol.74 , pp. 211-215
    • Ritchie, J.M.1    Rogart, R.B.2
  • 190
    • 33947546463 scopus 로고    scopus 로고
    • Sodium channels and multiple sclerosis: Roles in symptom production, damage and therapy
    • Smith KJ. Sodium channels and multiple sclerosis: Roles in symptom production, damage and therapy. Brain Pathol 17:230-242 (2007).
    • (2007) Brain Pathol , vol.17 , pp. 230-242
    • Smith, K.J.1
  • 191
    • 34249092704 scopus 로고    scopus 로고
    • Calcium and neurodegeneration
    • Mattson MP. Calcium and neurodegeneration. Aging Cell 6:337-350 (2007).
    • (2007) Aging Cell , vol.6 , pp. 337-350
    • Mattson, M.P.1
  • 193
    • 0026556810 scopus 로고
    • Ionic mechanisms of anoxic injury in mammalian CNS white matter: Role of Na+ channels and Na(+)-Ca2+ exchanger
    • Stys PK, Waxman SG, Ransom BR. Ionic mechanisms of anoxic injury in mammalian CNS white matter: Role of Na+ channels and Na(+)-Ca2+ exchanger. J Neurosci 12:430-439 (1992).
    • (1992) J Neurosci , vol.12 , pp. 430-439
    • Stys, P.K.1    Waxman, S.G.2    Ransom, B.R.3
  • 194
    • 0037441439 scopus 로고    scopus 로고
    • Hypoxia-like tissue injury as a component of multiple sclerosis lesions
    • Lassmann H. Hypoxia-like tissue injury as a component of multiple sclerosis lesions. J Neurol Sci 206:187-191 (2003).
    • (2003) J Neurol Sci , vol.206 , pp. 187-191
    • Lassmann, H.1
  • 195
    • 0031002545 scopus 로고    scopus 로고
    • Axonal damage in acute multiple sclerosis lesions.
    • Ferguson B, Matyszak MK, Esiri MM, Perry VH. Axonal damage in acute multiple sclerosis lesions. Brain 120 (Pt 3):393-399 (1997).
    • (1997) Brain , vol.120 , Issue.PART 3 , pp. 393-399
    • Ferguson, B.1    Matyszak, M.K.2    Esiri, M.M.3    Perry, V.H.4
  • 196
    • 0013292906 scopus 로고    scopus 로고
    • The role of nitric oxide in multiple sclerosis
    • Smith KJ, Lassmann H. The role of nitric oxide in multiple sclerosis. Lancet Neurol 1:232-241 (2002).
    • (2002) Lancet Neurol , vol.1 , pp. 232-241
    • Smith, K.J.1    Lassmann, H.2
  • 197
    • 0036889794 scopus 로고    scopus 로고
    • Nitric oxide inhibition of mitochondrial respiration and its role in cell death
    • Brown GC, Borutaite V. Nitric oxide inhibition of mitochondrial respiration and its role in cell death. Free Radic Biol Med 33:1440-1450 (2002).
    • (2002) Free Radic Biol Med , vol.33 , pp. 1440-1450
    • Brown, G.C.1    Borutaite, V.2
  • 199
    • 0037309442 scopus 로고    scopus 로고
    • Blockers of sodium and calcium entry protect axons from nitric oxide-mediated degeneration
    • Kapoor R, Davies M, Blaker PA, Hall SM, Smith KJ. Blockers of sodium and calcium entry protect axons from nitric oxide-mediated degeneration. Ann Neurol 53:174-180 (2003).
    • (2003) Ann Neurol , vol.53 , pp. 174-180
    • Kapoor, R.1    Davies, M.2    Blaker, P.A.3    Hall, S.M.4    Smith, K.J.5
  • 200
    • 0026599820 scopus 로고
    • Tertiary and quaternary local anesthetics protect CNS white matter from anoxic injury at concentrations that do not block excitability
    • Stys PK, Ransom BR, Waxman SG. Tertiary and quaternary local anesthetics protect CNS white matter from anoxic injury at concentrations that do not block excitability. J Neurophysiol 67:236-240 (1992).
    • (1992) J Neurophysiol , vol.67 , pp. 236-240
    • Stys, P.K.1    Ransom, B.R.2    Waxman, S.G.3
  • 201
    • 0030042661 scopus 로고    scopus 로고
    • Correlation between electrophysiological effects of mexiletine and ischemic protection in central nervous system white matter
    • Stys PK, Lesiuk H. Correlation between electrophysiological effects of mexiletine and ischemic protection in central nervous system white matter. Neuroscience 71:27-36 (1996).
    • (1996) Neuroscience , vol.71 , pp. 27-36
    • Stys, P.K.1    Lesiuk, H.2
  • 202
    • 0027874438 scopus 로고
    • Pharmacological protection of CNS white matter during anoxia: Actions of phenytoin, carbamazepine and diazepam
    • Fern R, Ransom BR, Stys PK, Waxman SG. Pharmacological protection of CNS white matter during anoxia: Actions of phenytoin, carbamazepine and diazepam. J Pharmacol Exp Ther 266:1549-1555 (1993).
    • (1993) J Pharmacol Exp Ther , vol.266 , pp. 1549-1555
    • Fern, R.1    Ransom, B.R.2    Stys, P.K.3    Waxman, S.G.4
  • 203
    • 0242508548 scopus 로고    scopus 로고
    • Phenytoin protects spinal cord axons and preserves axonal conduction and neurological function in a model of neuroinflammation in vivo
    • Lo AC, Saab CY, Black JA, Waxman SG. Phenytoin protects spinal cord axons and preserves axonal conduction and neurological function in a model of neuroinflammation in vivo. J Neurophysiol 90:3566-3571 (2003).
    • (2003) J Neurophysiol , vol.90 , pp. 3566-3571
    • Lo, A.C.1    Saab, C.Y.2    Black, J.A.3    Waxman, S.G.4
  • 204
    • 2142760098 scopus 로고    scopus 로고
    • Axonal protection using flecainide in experimental autoimmune encephalomyelitis
    • Bechtold DA, Kapoor R, Smith KJ. Axonal protection using flecainide in experimental autoimmune encephalomyelitis. Ann Neurol 55:607-616 (2004).
    • (2004) Ann Neurol , vol.55 , pp. 607-616
    • Bechtold, D.A.1    Kapoor, R.2    Smith, K.J.3
  • 205
    • 33744489487 scopus 로고    scopus 로고
    • Axonal protection achieved in a model of multiple sclerosis using lamotrigine
    • Bechtold DA et al. Axonal protection achieved in a model of multiple sclerosis using lamotrigine. J Neurol 253:1542-1551 (2006).
    • (2006) J Neurol , vol.253 , pp. 1542-1551
    • Bechtold, D.A.1
  • 206
    • 0025884390 scopus 로고
    • Ion channels in leukocytes
    • Gallin EK. Ion channels in leukocytes. Physiol Rev 71:775-811 (1991).
    • (1991) Physiol Rev , vol.71 , pp. 775-811
    • Gallin, E.K.1
  • 207
  • 208
    • 69449105146 scopus 로고    scopus 로고
    • Sodium channel activity modulates multiple functions in microglia
    • Black JA, Liu S, Waxman SG. Sodium channel activity modulates multiple functions in microglia. Glia 57:1072-1081 (2009).
    • (2009) Glia , vol.57 , pp. 1072-1081
    • Black, J.A.1    Liu, S.2    Waxman, S.G.3
  • 209
    • 13244258365 scopus 로고    scopus 로고
    • Sodium channels contribute to microglia/macrophage activation and function in EAE and MS
    • Craner MJ et al. Sodium channels contribute to microglia/macrophage activation and function in EAE and MS. Glia 49:220-229 (2005).
    • (2005) Glia , vol.49 , pp. 220-229
    • Craner, M.J.1
  • 210
    • 34547773583 scopus 로고    scopus 로고
    • Exacerbation of experimental autoimmune encephalomyelitis after withdrawal of phenytoin and carbamazepine
    • Black JA, Liu S, Carrithers M, Carrithers LM, Waxman SG. Exacerbation of experimental autoimmune encephalomyelitis after withdrawal of phenytoin and carbamazepine. Ann Neurol 62:21-33 (2007).
    • (2007) Ann Neurol , vol.62 , pp. 21-33
    • Black, J.A.1    Liu, S.2    Carrithers, M.3    Carrithers, L.M.4    Waxman, S.G.5
  • 211
    • 0018873513 scopus 로고
    • Neurotoxins that act on voltage-sensitive sodium channels in excitable membranes
    • Catterall WA. Neurotoxins that act on voltage-sensitive sodium channels in excitable membranes. Annu Rev Pharmacol Toxicol 20:15-43 (1980).
    • (1980) Annu Rev Pharmacol Toxicol , vol.20 , pp. 15-43
    • Catterall, W.A.1
  • 212
    • 0019387751 scopus 로고
    • Toxin T4(6) from Ptychodiscus brevis (formerly Gymnodinium breve) enhances activation of voltage-sensitive sodium channels by veratridine
    • Catterall WA, Risk M. Toxin T4(6) from Ptychodiscus brevis (formerly Gymnodinium breve) enhances activation of voltage-sensitive sodium channels by veratridine. Mol Pharmacol 19:345-348 (1981).
    • (1981) Mol Pharmacol , vol.19 , pp. 345-348
    • Catterall, W.A.1    Risk, M.2
  • 213
    • 0022901519 scopus 로고
    • Brevetoxins, unique activators of voltage-sensitive sodium channels, bind to specific sites in rat brain synaptosomes
    • Poli M, Mende T, Baden D. Brevetoxins, unique activators of voltage-sensitive sodium channels, bind to specific sites in rat brain synaptosomes. Mol Pharmacol 30:129-135 (1986).
    • (1986) Mol Pharmacol , vol.30 , pp. 129-135
    • Poli, M.1    Mende, T.2    Baden, D.3
  • 214
    • 0028049479 scopus 로고
    • A new neurotoxin receptor site on sodium channels is identified by a conotoxin that affects sodium channel inactivation in molluscs and acts as an antagonist in rat brain.
    • Fainzilber M, Kofman O, Zlotkin E, Gordon D. A new neurotoxin receptor site on sodium channels is identified by a conotoxin that affects sodium channel inactivation in molluscs and acts as an antagonist in rat brain. J Biol Chem269:2574-2580 (1994).
    • (1994) J Biol Chem , vol.269 , pp. 2574-2580
    • Fainzilber, M.1    Kofman, O.2    Zlotkin, E.3    Gordon, D.4
  • 215
    • 0019276817 scopus 로고
    • Interaction of polypeptide neurotoxins with a receptor site associated with voltage-sensitive sodium channels
    • Catterall WA, Beneski DA. Interaction of polypeptide neurotoxins with a receptor site associated with voltage-sensitive sodium channels. J Supramol Struct 14:295-303 (1980).
    • (1980) J Supramol Struct , vol.14 , pp. 295-303
    • Catterall, W.A.1    Beneski, D.A.2
  • 216
    • 0033731909 scopus 로고    scopus 로고
    • Molecular mechanisms of neurotoxin action on voltage-gated sodium channels
    • Cestele S, Catterall WA. Molecular mechanisms of neurotoxin action on voltage-gated sodium channels. Biochimie 82:883-892 (2000).
    • (2000) Biochimie , vol.82 , pp. 883-892
    • Cestele, S.1    Catterall, W.A.2
  • 217
    • 0016782794 scopus 로고
    • The receptor for tetrodotoxin and saxitoxin
    • Hille B. The receptor for tetrodotoxin and saxitoxin. A structural hypothesis. Biophys J 15:615-619 (1975).
    • (1975) A structural hypothesis. Biophys J , vol.15 , pp. 615-619
    • Hille, B.1
  • 218
    • 0022931513 scopus 로고
    • Specific inhibition of [3H] saxitoxin binding to skeletal muscle sodium channels by geographutoxin II, a polypeptide channel blocker
    • Ohizumi Y, Nakamura H, Kobayashi J, Catterall WA. Specific inhibition of [3H] saxitoxin binding to skeletal muscle sodium channels by geographutoxin II, a polypeptide channel blocker. J Biol Chem 261:6149-6152 (1986).
    • (1986) J Biol Chem , vol.261 , pp. 6149-6152
    • Ohizumi, Y.1    Nakamura, H.2    Kobayashi, J.3    Catterall, W.A.4
  • 219
    • 0022559685 scopus 로고
    • Blockade of [3H]lysine-tetrodotoxin binding to sodium channel proteins by conotoxin GIII
    • Yanagawa Y, Abe T, Satake M. Blockade of [3H]lysine-tetrodotoxin binding to sodium channel proteins by conotoxin GIII. Neurosci Lett 64:7-12 (1986).
    • (1986) Neurosci Lett , vol.64 , pp. 7-12
    • Yanagawa, Y.1    Abe, T.2    Satake, M.3
  • 220
    • 0028970541 scopus 로고
    • A mu-conotoxin-insensitive Na+ channel mutant: Possible localization of a binding site at the outer vestibule
    • Dudley SC, Jr, Todt H, Lipkind G, Fozzard HA. A mu-conotoxin-insensitive Na+ channel mutant: Possible localization of a binding site at the outer vestibule. Biophys J 69:1657-1665 (1995).
    • (1995) Biophys J , vol.69 , pp. 1657-1665
    • Dudley Jr., S.C.1    Todt, H.2    Lipkind, G.3    Fozzard, H.A.4
  • 221
    • 0025955695 scopus 로고
    • Active site of mu-conotoxin GIIIA, a peptide blocker of muscle sodium channels
    • Sato K et al. Active site of mu-conotoxin GIIIA, a peptide blocker of muscle sodium channels. J Biol Chem 266:16989-16991 (1991).
    • (1991) J Biol Chem , vol.266 , pp. 16989-16991
    • Sato, K.1
  • 223
    • 0017661782 scopus 로고
    • Activation of the action potential Na+ ionophore by neurotoxins
    • Catterall WA. Activation of the action potential Na+ ionophore by neurotoxins. An allosteric model. J Biol Chem 252:8669-8676 (1977).
    • (1977) An allosteric model. J Biol Chem , vol.252 , pp. 8669-8676
    • Catterall, W.A.1
  • 224
    • 0033988693 scopus 로고    scopus 로고
    • On site of action of grayanotoxin in domain 4 segment 6 of rat skeletal muscle sodium channel
    • Kimura T et al. On site of action of grayanotoxin in domain 4 segment 6 of rat skeletal muscle sodium channel. FEBS Lett 465:18-22 (2000).
    • (2000) FEBS Lett , vol.465 , pp. 18-22
    • Kimura, T.1
  • 225
    • 0032506024 scopus 로고    scopus 로고
    • Interaction of batrachotoxin with the local anesthetic receptor site in transmembrane segment IVS6 of the voltagegated sodium channel
    • Linford NJ, Cantrell AR, Qu Y, Scheuer T, Catterall WA. Interaction of batrachotoxin with the local anesthetic receptor site in transmembrane segment IVS6 of the voltagegated sodium channel. Proc Natl Acad Sci USA 95:13947-13952 (1998).
    • (1998) Proc Natl Acad Sci USA , vol.95 , pp. 13947-13952
    • Linford, N.J.1    Cantrell, A.R.2    Qu, Y.3    Scheuer, T.4    Catterall, W.A.5
  • 226
    • 0021903373 scopus 로고
    • Batrachotoxin as a tool to study voltage-sensitive sodium channels of excitable membranes
    • Khodorov BI. Batrachotoxin as a tool to study voltage-sensitive sodium channels of excitable membranes. Prog Biophys Mol Biol 45:57-148 (1985).
    • (1985) Prog Biophys Mol Biol , vol.45 , pp. 57-148
    • Khodorov, B.I.1
  • 227
    • 0022643902 scopus 로고
    • Kinetics of veratridine action on Na channels of skeletal muscle
    • Sutro JB. Kinetics of veratridine action on Na channels of skeletal muscle. J Gen Physiol 87:1-24 (1986).
    • (1986) J Gen Physiol , vol.87 , pp. 1-24
    • Sutro, J.B.1
  • 228
    • 15844371139 scopus 로고    scopus 로고
    • Site of covalent labeling by a photoreactive batrachotoxin derivative near transmembrane segment IS6 of the sodium channel alpha subunit
    • Trainer VL, Brown GB, Catterall WA. Site of covalent labeling by a photoreactive batrachotoxin derivative near transmembrane segment IS6 of the sodium channel alpha subunit. J Biol Chem 271:11261-11267 (1996).
    • (1996) J Biol Chem , vol.271 , pp. 11261-11267
    • Trainer, V.L.1    Brown, G.B.2    Catterall, W.A.3
  • 229
    • 33644844639 scopus 로고    scopus 로고
    • How batrachotoxin modifies the sodium channel permeation pathway: Computer modeling and site-directed mutagenesis
    • Wang S-Y, Mitchell J, Tikhonov DB, Zhorov BS,Wang GK. How batrachotoxin modifies the sodium channel permeation pathway: Computer modeling and site-directed mutagenesis. Mol Pharmacol 69:788-795 (2006).
    • (2006) Mol Pharmacol , vol.69 , pp. 788-795
    • Wang, S.-Y.1    Mitchell, J.2    Tikhonov, D.B.3    Zhorov, B.S.4    Wang, G.K.5
  • 230
    • 0023666436 scopus 로고
    • Ciguatoxin and brevetoxins share a common receptor site on the neuronal voltage-dependent Na+ channel
    • Lombet A, Bidard JN, Lazdunski M. Ciguatoxin and brevetoxins share a common receptor site on the neuronal voltage-dependent Na+ channel. FEBS Lett 219:355-359 (1987).
    • (1987) FEBS Lett , vol.219 , pp. 355-359
    • Lombet, A.1    Bidard, J.N.2    Lazdunski, M.3
  • 231
    • 0022655450 scopus 로고
    • Effects of ciguatoxin on current and voltage clamped frog myelinated nerve fibre
    • Benoit E, Legrand AM, Dubois JM. Effects of ciguatoxin on current and voltage clamped frog myelinated nerve fibre. Toxicon 24:357-364 (1986).
    • (1986) Toxicon , vol.24 , pp. 357-364
    • Benoit, E.1    Legrand, A.M.2    Dubois, J.M.3
  • 232
    • 0021324876 scopus 로고
    • Depolarizing action of a red-tide dinoflagellate brevetoxin on axonal membranes
    • Huang JM, Wu CH, Baden DG. Depolarizing action of a red-tide dinoflagellate brevetoxin on axonal membranes. J Pharmacol Exp Ther 229:615-621 (1984).
    • (1984) J Pharmacol Exp Ther , vol.229 , pp. 615-621
    • Huang, J.M.1    Wu, C.H.2    Baden, D.G.3
  • 233
    • 0027991485 scopus 로고
    • Identification of peptide components of the brevetoxin receptor site of rat brain sodium channels
    • Trainer VL, Baden DG, Catterall WA. Identification of peptide components of the brevetoxin receptor site of rat brain sodium channels. J Biol Chem 269:19904-19909 (1994).
    • (1994) J Biol Chem , vol.269 , pp. 19904-19909
    • Trainer, V.L.1    Baden, D.G.2    Catterall, W.A.3
  • 234
    • 0024240855 scopus 로고
    • Site of covalent attachment of alpha-scorpion toxin derivatives in domain I of the sodium channel alpha subunit
    • Tejedor FJ, Catterall WA. Site of covalent attachment of alpha-scorpion toxin derivatives in domain I of the sodium channel alpha subunit. Proc Natl Acad Sci USA 85:8742-8746 (1988).
    • (1988) Proc Natl Acad Sci USA , vol.85 , pp. 8742-8746
    • Tejedor, F.J.1    Catterall, W.A.2
  • 235
    • 0032191111 scopus 로고    scopus 로고
    • Voltage sensor-trapping: Enhanced activation of sodium channels by beta-scorpion toxin bound to the S3-S4 loop in domain II
    • Cestele S et al. Voltage sensor-trapping: Enhanced activation of sodium channels by beta-scorpion toxin bound to the S3-S4 loop in domain II. Neuron 21:919-931 (1998).
    • (1998) Neuron , vol.21 , pp. 919-931
    • Cestele, S.1
  • 236
    • 49549134070 scopus 로고
    • Decreased rate of sodium conductance inactivation in the node of Ranvier induced by a polypeptide toxin from sea anemone
    • Bergman C, Dubois JM, Rojas E, Rathmayer W. Decreased rate of sodium conductance inactivation in the node of Ranvier induced by a polypeptide toxin from sea anemone. Biochim Biophys Acta 455:173-184 (1976).
    • (1976) Biochim Biophys Acta , vol.455 , pp. 173-184
    • Bergman, C.1    Dubois, J.M.2    Rojas, E.3    Rathmayer, W.4
  • 238
    • 0031978687 scopus 로고    scopus 로고
    • Characterisation of the effects of robustoxin, the lethal neurotoxin from the Sydney funnel-web spider Atrax robustus, on sodium channel activation and inactivation
    • Nicholson GM, Walsh R, Little MJ, Tyler MI. Characterisation of the effects of robustoxin, the lethal neurotoxin from the Sydney funnel-web spider Atrax robustus, on sodium channel activation and inactivation. Pflugers Arch 436:117-126 (1998).
    • (1998) Pflugers Arch , vol.436 , pp. 117-126
    • Nicholson, G.M.1    Walsh, R.2    Little, M.J.3    Tyler, M.I.4
  • 239
    • 0028111357 scopus 로고
    • Modification of sodium channel gating and kinetics by versutoxin from the Australian funnel-web spider Hadronyche versuta
    • Nicholson GM, Willow M, Howden ME, Narahashi T. Modification of sodium channel gating and kinetics by versutoxin from the Australian funnel-web spider Hadronyche versuta. Pflugers Arch 428:400-409 (1994).
    • (1994) Pflugers Arch , vol.428 , pp. 400-409
    • Nicholson, G.M.1    Willow, M.2    Howden, M.E.3    Narahashi, T.4
  • 240
    • 0022346793 scopus 로고
    • The voltage-dependent Na+ channel of insect nervous system identified by receptor sites for tetrodotoxin, and scorpion and sea anemone toxins
    • Pauron D, Barhanin J, Lazdunski M. The voltage-dependent Na+ channel of insect nervous system identified by receptor sites for tetrodotoxin, and scorpion and sea anemone toxins. Biochem Biophys Res Commun 131:1226-1233 (1985).
    • (1985) Biochem Biophys Res Commun , vol.131 , pp. 1226-1233
    • Pauron, D.1    Barhanin, J.2    Lazdunski, M.3
  • 241
    • 0030037704 scopus 로고    scopus 로고
    • Molecular determinants of high affinity binding of alpha-scorpion toxin and sea anemone toxin in the S3-S4 extracellular loop in domain IV of the Na+ channel alpha subunit
    • Rogers JC, Qu Y, Tanada TN, Scheuer T, Catterall WA. Molecular determinants of high affinity binding of alpha-scorpion toxin and sea anemone toxin in the S3-S4 extracellular loop in domain IV of the Na+ channel alpha subunit. J Biol Chem 271:15950-15962 (1996).
    • (1996) J Biol Chem , vol.271 , pp. 15950-15962
    • Rogers, J.C.1    Qu, Y.2    Tanada, T.N.3    Scheuer, T.4    Catterall, W.A.5
  • 242
    • 0031972937 scopus 로고    scopus 로고
    • A specific interaction between the cardiac sodium channel and site-3 toxin anthopleurin B
    • Benzinger GR, Kyle JW, Blumenthal KM, Hanck DA. A specific interaction between the cardiac sodium channel and site-3 toxin anthopleurin B. J Biol Chem 273:80-84 (1998).
    • (1998) J Biol Chem , vol.273 , pp. 80-84
    • Benzinger, G.R.1    Kyle, J.W.2    Blumenthal, K.M.3    Hanck, D.A.4
  • 243
    • 0016424652 scopus 로고
    • Modification of sodium channel gating in frog myelinated nerve fibres by Centruroides sculpturatus scorpion venom
    • Cahalan MD. Modification of sodium channel gating in frog myelinated nerve fibres by Centruroides sculpturatus scorpion venom. J Physiol 244:511-534 (1975).
    • (1975) J Physiol , vol.244 , pp. 511-534
    • Cahalan, M.D.1
  • 244
    • 0022552962 scopus 로고
    • Toxin gamma of the scorpion Tityus serrulatus modifies both activation and inactivation of sodium permeability of nerve membrane
    • Jonas P, VogelW, Arantes EC, Giglio JR. Toxin gamma of the scorpion Tityus serrulatus modifies both activation and inactivation of sodium permeability of nerve membrane. Pflugers Arch 407:92-99 (1986).
    • (1986) Pflugers Arch , vol.407 , pp. 92-99
    • Jonas, P.1    Vogel, W.2    Arantes, E.C.3    Giglio, J.R.4
  • 245
    • 0020641535 scopus 로고
    • Purification and physiological characterization of neurotoxins from venoms of the scorpions centruroides sculpturatus and leiurus quinquestriatus
    • Wang GK, Strichartz GR. Purification and physiological characterization of neurotoxins from venoms of the scorpions centruroides sculpturatus and leiurus quinquestriatus. Mol Pharmacol 23:519-533 (1983).
    • (1983) Mol Pharmacol , vol.23 , pp. 519-533
    • Wang, G.K.1    Strichartz, G.R.2
  • 246
    • 0031057891 scopus 로고    scopus 로고
    • Effects of Tityus serrulatus scorpion toxin gamma on voltage-gated Na+ channels
    • Marcotte P, Chen LQ, Kallen RG, Chahine M. Effects of Tityus serrulatus scorpion toxin gamma on voltage-gated Na+ channels. Circ Res 80:363-369 (1997).
    • (1997) Circ Res , vol.80 , pp. 363-369
    • Marcotte, P.1    Chen, L.Q.2    Kallen, R.G.3    Chahine, M.4
  • 247
    • 33745228127 scopus 로고    scopus 로고
    • Subtype specificity of scorpion betatoxin Tz1 interaction with voltage-gated sodium channels is determined by the pore loop of domain 3
    • Leipold E, Hansel A, Borges A, Heinemann SH. Subtype specificity of scorpion betatoxin Tz1 interaction with voltage-gated sodium channels is determined by the pore loop of domain 3. Mol Pharmacol 70:340-347 (2006).
    • (2006) Mol Pharmacol , vol.70 , pp. 340-347
    • Leipold, E.1    Hansel, A.2    Borges, A.3    Heinemann, S.H.4
  • 248
    • 0027479426 scopus 로고
    • Alteration of sodium currents by new peptide toxins from the venom of a molluscivorous Conus snail
    • Hasson A, Fainzilber M, Gordon D, Zlotkin E, Spira ME. Alteration of sodium currents by new peptide toxins from the venom of a molluscivorous Conus snail. Eur J Neurosci 5:56-64 (1993).
    • (1993) Eur J Neurosci , vol.5 , pp. 56-64
    • Hasson, A.1    Fainzilber, M.2    Gordon, D.3    Zlotkin, E.4    Spira, M.E.5
  • 249
    • 21844466444 scopus 로고    scopus 로고
    • Molecular interaction of delta-conotoxins with voltage-gated sodium channels
    • Leipold E, Hansel A, Olivera BM., Terlau, H, Heinemann SH. Molecular interaction of delta-conotoxins with voltage-gated sodium channels. FEBS Lett 579:3881-3884 (2005).
    • (2005) FEBS Lett , vol.579 , pp. 3881-3884
    • Leipold, E.1    Hansel, A.2    Olivera, B.M.3    Terlau, H.4    Heinemann, S.H.5
  • 250
    • 77949300922 scopus 로고    scopus 로고
    • Voltage-gated sodium channels as therapeutic targets in epilepsy and other neurological disorders
    • Mantegazza M, Curia G, Biagini G, Ragsdale DS, Avoli M. Voltage-gated sodium channels as therapeutic targets in epilepsy and other neurological disorders. Lancet Neurol 9:413-424 (2010).
    • (2010) Lancet Neurol , vol.9 , pp. 413-424
    • Mantegazza, M.1    Curia, G.2    Biagini, G.3    Ragsdale, D.S.4    Avoli, M.5
  • 251
    • 34248396038 scopus 로고    scopus 로고
    • Blockers of voltage-gated sodium channels for the treatment of central nervous systemdiseases.
    • Tarnawa I, Bolcskei H, Kocsis P. Blockers of voltage-gated sodium channels for the treatment of central nervous systemdiseases.Recent PatCNSDrugDiscov 2:57-78 (2007).
    • (2007) Recent PatCNSDrugDiscov , vol.2 , pp. 57-78
    • Tarnawa, I.1    Bolcskei, H.2    Kocsis, P.3
  • 252
    • 66449110564 scopus 로고    scopus 로고
    • Recent advances in the medicinal chemistry of sodium channel blockers and their therapeutic potential
    • Zuliani V, Patel MK, Fantini M, Rivara M. Recent advances in the medicinal chemistry of sodium channel blockers and their therapeutic potential. Curr Top Med Chem 9:396-415 (2009).
    • (2009) Curr Top Med Chem , vol.9 , pp. 396-415
    • Zuliani, V.1    Patel, M.K.2    Fantini, M.3    Rivara, M.4
  • 253
    • 59149087853 scopus 로고    scopus 로고
    • Role of antiepileptic drugs in the management of eating disorders
    • McElroy SL et al. Role of antiepileptic drugs in the management of eating disorders. CNS Drugs 23:139-156 (2009).
    • (2009) CNS Drugs , vol.23 , pp. 139-156
    • McElroy, S.L.1
  • 254
    • 78650921573 scopus 로고    scopus 로고
    • Classification of drugs based on properties of sodiumchannel inhibition: A comparative automated patch-clamp study.
    • LenkeyNet al.Classification of drugs based on properties of sodiumchannel inhibition: A comparative automated patch-clamp study. PLoS One 5(12):e15568 (2010).
    • (2010) PLoS One , vol.5 , Issue.12
    • Lenkey, N.1
  • 255
    • 28844507943 scopus 로고    scopus 로고
    • Analysis of human Nav1.8 expressed in SH-SY5Y neuroblastoma cells
    • Dekker LVet al. Analysis of human Nav1.8 expressed in SH-SY5Y neuroblastoma cells. Eur J Pharmacol 528:52-58 (2005).
    • (2005) Eur J Pharmacol , vol.528 , pp. 52-58
    • Dekker, L.V.1
  • 256
    • 67449089458 scopus 로고    scopus 로고
    • Structural determinants of drugs acting on the Nav1.8 channel
    • Browne LE, Blaney FE, Yusaf SP, Clare JJ, Wray D. Structural determinants of drugs acting on the Nav1.8 channel. J Biol Chem 284:10523-10536 (2009).
    • (2009) J Biol Chem , vol.284 , pp. 10523-10536
    • Browne, L.E.1    Blaney, F.E.2    Yusaf, S.P.3    Clare, J.J.4    Wray, D.5
  • 257
    • 63249113812 scopus 로고    scopus 로고
    • Functional and pharmacological properties of human and rat NaV1.8 channels
    • Browne LE, Clare JJ, Wray D. Functional and pharmacological properties of human and rat NaV1.8 channels. Neuropharmacology 56:905-914 (2009).
    • (2009) Neuropharmacology , vol.56 , pp. 905-914
    • Browne, L.E.1    Clare, J.J.2    Wray, D.3
  • 259
    • 0033694833 scopus 로고    scopus 로고
    • From ionic currents to molecular mechanisms: The structure and function of voltage-gated sodium channels
    • CatterallW. From ionic currents to molecular mechanisms: The structure and function of voltage-gated sodium channels. Neuron 26:13-25 (2000).
    • (2000) Neuron , vol.26 , pp. 13-25
    • Catterall, W.1
  • 260
    • 77958471998 scopus 로고    scopus 로고
    • The sodium channel {beta}3-subunit induces multiphasic gating in NaV1.3 and affects fast inactivation via distinct intracellular regions.
    • Cusdin FS et al. The sodium channel {beta}3-subunit induces multiphasic gating in NaV1.3 and affects fast inactivation via distinct intracellular regions. J Biol Chem 285:33404-33412.
    • J Biol Chem , vol.285 , pp. 33404-33412
    • Cusdin, F.S.1
  • 261
    • 0141644287 scopus 로고    scopus 로고
    • Expression of auxiliary beta subunits of sodium channels in primary afferent neurons and the effect of nerve injury
    • Takahashi N et al. Expression of auxiliary beta subunits of sodium channels in primary afferent neurons and the effect of nerve injury. Neuroscience 121:441-450 (2003).
    • (2003) Neuroscience , vol.121 , pp. 441-450
    • Takahashi, N.1
  • 262
    • 40449127990 scopus 로고    scopus 로고
    • Toxins from cone snails: Properties, applications and biotechnological production
    • Becker S, Terlau H. Toxins from cone snails: Properties, applications and biotechnological production. Appl Microbiol Biotechnol 79:1-9 (2008).
    • (2008) Appl Microbiol Biotechnol , vol.79 , pp. 1-9
    • Becker, S.1    Terlau, H.2
  • 263
    • 42149193214 scopus 로고    scopus 로고
    • High-throughput electrophysiology: An emerging paradigm for ion-channel screening and physiology
    • Dunlop J, Bowlby M, Peri R, Vasilyev D, Arias R. High-throughput electrophysiology: An emerging paradigm for ion-channel screening and physiology. Nat Rev Drug Discov 7:358-368 (2008).
    • (2008) Nat Rev Drug Discov , vol.7 , pp. 358-368
    • Dunlop, J.1    Bowlby, M.2    Peri, R.3    Vasilyev, D.4    Arias, R.5
  • 264
    • 0035892592 scopus 로고    scopus 로고
    • Ion-channel assay technologies: Quo vadis?
    • Xu J et al. Ion-channel assay technologies: Quo vadis? Drug Discov Today 6:1278-1287 (2001).
    • (2001) Drug Discov Today , vol.6 , pp. 1278-1287
    • Xu, J.1
  • 265
    • 62649139727 scopus 로고    scopus 로고
    • Sodium channel inhibitor drug discovery using automated high throughput electrophysiology platforms
    • Castle N et al. Sodium channel inhibitor drug discovery using automated high throughput electrophysiology platforms. Comb Chem High Throughput Screen 12:107-122 (2009).
    • (2009) Comb Chem High Throughput Screen , vol.12 , pp. 107-122
    • Castle, N.1
  • 266
    • 77957939445 scopus 로고    scopus 로고
    • Recent advances in ion channel screening technologies.
    • In Fermini B, Priest B. eds.
    • Fermini B. Recent advances in ion channel screening technologies. In Fermini B, Priest B. eds. Ion Channels. Springer, 2008, pp. 1-26.
    • (2008) Ion Channels. Springer , pp. 1-26
    • Fermini, B.1
  • 268
    • 33747102760 scopus 로고    scopus 로고
    • Population patch clamp improves data consistency and success rates in the measurement of ionic currents
    • Finkel A et al. Population patch clamp improves data consistency and success rates in the measurement of ionic currents. J Biomol Screen 11:488-496 (2006).
    • (2006) J Biomol Screen , vol.11 , pp. 488-496
    • Finkel, A.1
  • 269
    • 33645780149 scopus 로고    scopus 로고
    • QPatch: The past, present and future of automated patch clamp
    • Mathes C. QPatch: The past, present and future of automated patch clamp. Expert Opin Ther Targets 10:319-327 (2006).
    • (2006) Expert Opin Ther Targets , vol.10 , pp. 319-327
    • Mathes, C.1
  • 271
    • 58149216783 scopus 로고    scopus 로고
    • Automated voltage-clamp technique.
    • Ghetti A, Guia A, Xu J. Automated voltage-clamp technique. Methods Mol Biol 403:59- 69 (2007).
    • (2007) Methods Mol Biol , vol.403 , pp. 59-69
    • Ghetti, A.1    Guia, A.2    Xu, J.3
  • 272
    • 22444443481 scopus 로고    scopus 로고
    • Automated electrophysiology in the preclinical evaluation of drugs for potential QT prolongation
    • Guo L, Guthrie H. Automated electrophysiology in the preclinical evaluation of drugs for potential QT prolongation. J Pharmacol Toxicol Methods 52:123-135 (2005).
    • (2005) J Pharmacol Toxicol Methods , vol.52 , pp. 123-135
    • Guo, L.1    Guthrie, H.2
  • 273
    • 34548856200 scopus 로고    scopus 로고
    • Population patch clamp electrophysiology: A breakthrough technology for ion channel screening
    • Dale TJ, Townsend C, Hollands EC, Trezise DJ. Population patch clamp electrophysiology: A breakthrough technology for ion channel screening. Mol Biosyst 3:714-722 (2007).
    • (2007) Mol Biosyst , vol.3 , pp. 714-722
    • Dale, T.J.1    Townsend, C.2    Hollands, E.C.3    Trezise, D.J.4
  • 274
    • 77956064278 scopus 로고    scopus 로고
    • Assessing use-dependent inhibition of the cardiac Na(+/-) current (I(Na)) in the PatchXpress automated patch clamp.
    • Penniman JR, Kim DC, Salata JJ, Imredy JP. Assessing use-dependent inhibition of the cardiac Na(+/-) current (I(Na)) in the PatchXpress automated patch clamp. J Pharmacol Toxicol Methods 62:107-118.
    • J Pharmacol Toxicol Methods , vol.62 , pp. 107-118
    • Penniman, J.R.1    Kim, D.C.2    Salata, J.J.3    Imredy, J.P.4
  • 275
    • 1942433697 scopus 로고    scopus 로고
    • A benchmark study with sealchip planar patch-clamp technology
    • Xu J et al. A benchmark study with sealchip planar patch-clamp technology. Assay Drug Dev Technol 1:675-684 (2003).
    • (2003) Assay Drug Dev Technol , vol.1 , pp. 675-684
    • Xu, J.1
  • 276
    • 63749092193 scopus 로고    scopus 로고
    • Optimization of Ca(v)1.2 screening with an automated planar patch clamp platform
    • Balasubramanian B et al. Optimization of Ca(v)1.2 screening with an automated planar patch clamp platform. J Pharmacol Toxicol Methods 59:62-72 (2009).
    • (2009) J Pharmacol Toxicol Methods , vol.59 , pp. 62-72
    • Balasubramanian, B.1
  • 277
    • 20144389310 scopus 로고    scopus 로고
    • Identifying modulators of hERG channel activity using the PatchXpress planar patch clamp
    • Dubin AE et al. Identifying modulators of hERG channel activity using the PatchXpress planar patch clamp. J Biomol Screen 10:168-181 (2005).
    • (2005) J Biomol Screen , vol.10 , pp. 168-181
    • Dubin, A.E.1
  • 278
    • 34548683936 scopus 로고    scopus 로고
    • In vitro screening strategies for nicotinic receptor ligands
    • Dunlop J et al. In vitro screening strategies for nicotinic receptor ligands. Biochem Pharmacol 74:1172-1181 (2007).
    • (2007) Biochem Pharmacol , vol.74 , pp. 1172-1181
    • Dunlop, J.1
  • 279
    • 68749093821 scopus 로고    scopus 로고
    • Effect of compound plate composition on measurement of hERG current IC(50) using PatchXpress
    • Mo ZL et al. Effect of compound plate composition on measurement of hERG current IC(50) using PatchXpress. J Pharmacol Toxicol Methods 60:39-44 (2009).
    • (2009) J Pharmacol Toxicol Methods , vol.60 , pp. 39-44
    • Mo, Z.L.1
  • 280
    • 35348959556 scopus 로고    scopus 로고
    • Application of PatchXpress planar patch clamp technology to the screening of new drug candidates for cardiac KCNQ1/KCNE1 (I Ks) activity
    • Trepakova ES et al. Application of PatchXpress planar patch clamp technology to the screening of new drug candidates for cardiac KCNQ1/KCNE1 (I Ks) activity. Assay Drug Dev Technol 5:617-627 (2007).
    • (2007) Assay Drug Dev Technol , vol.5 , pp. 617-627
    • Trepakova, E.S.1
  • 281
    • 57849127303 scopus 로고    scopus 로고
    • Characterization of compounds on nicotinic acetylcholine receptor alpha7 channels using higher throughput electrophysiology
    • Friis S, Mathes C, Sunesen M, Bowlby MR, Dunlop J. Characterization of compounds on nicotinic acetylcholine receptor alpha7 channels using higher throughput electrophysiology. J Neurosci Methods 177:142-148 (2009).
    • (2009) J Neurosci Methods , vol.177 , pp. 142-148
    • Friis, S.1    Mathes, C.2    Sunesen, M.3    Bowlby, M.R.4    Dunlop, J.5
  • 282
    • 62649141502 scopus 로고    scopus 로고
    • Automated planar electrode electrophysiology in drug discovery: Examples of the use of QPatch in basic characterization and high content screening on Na(v), K(Ca)2.3, and K(v)11.1 channels
    • Korsgaard MP, Strobaek D, Christophersen P. Automated planar electrode electrophysiology in drug discovery: Examples of the use of QPatch in basic characterization and high content screening on Na(v), K(Ca)2.3, and K(v)11.1 channels. Comb Chem High Throughput Screen 12:51-63 (2009).
    • (2009) Comb Chem High Throughput Screen , vol.12 , pp. 51-63
    • Korsgaard, M.P.1    Strobaek, D.2    Christophersen, P.3
  • 283
    • 1942529810 scopus 로고    scopus 로고
    • Characterization of potassium channel modulators with QPatch automated patch-clamp technology: System characteristics and performance
    • Kutchinsky J et al. Characterization of potassium channel modulators with QPatch automated patch-clamp technology: System characteristics and performance. Assay Drug Dev Technol 1:685-693 (2003).
    • (2003) Assay Drug Dev Technol , vol.1 , pp. 685-693
    • Kutchinsky, J.1
  • 285
    • 1942465537 scopus 로고    scopus 로고
    • High quality ion channel analysis on a chip with the NPC technology
    • Bruggemann A et al. High quality ion channel analysis on a chip with the NPC technology. Assay Drug Dev Technol 1:665-673 (2003).
    • (2003) Assay Drug Dev Technol , vol.1 , pp. 665-673
    • Bruggemann, A.1
  • 286
    • 62649105798 scopus 로고    scopus 로고
    • Port-a-patch and patchliner: High fidelity electrophysiology for secondary screening and safety pharmacology
    • Farre C et al. Port-a-patch and patchliner: High fidelity electrophysiology for secondary screening and safety pharmacology. Comb Chem High Throughput Screen 12:24-37 (2009).
    • (2009) Comb Chem High Throughput Screen , vol.12 , pp. 24-37
    • Farre, C.1
  • 287
    • 33748129547 scopus 로고    scopus 로고
    • Optimisation and validation of a medium-throughput electrophysiology-based hERG assay using IonWorks HT
    • Bridgland-Taylor MH et al. Optimisation and validation of a medium-throughput electrophysiology-based hERG assay using IonWorks HT. J Pharmacol Toxicol Methods 54:189-199 (2006).
    • (2006) J Pharmacol Toxicol Methods , vol.54 , pp. 189-199
    • Bridgland-Taylor, M.H.1
  • 288
    • 0038623824 scopus 로고    scopus 로고
    • Measuring intracellular calcium fluxes in high throughput mode
    • Chambers C et al. Measuring intracellular calcium fluxes in high throughput mode. Comb Chem High Throughput Screen 6:355-362 (2003).
    • (2003) Comb Chem High Throughput Screen , vol.6 , pp. 355-362
    • Chambers, C.1
  • 290
    • 43449092864 scopus 로고    scopus 로고
    • Cellular HTS assays for pharmacological characterization of Na(V)1.7 modulators
    • Trivedi S et al. Cellular HTS assays for pharmacological characterization of Na(V)1.7 modulators. Assay Drug Dev Technol 6:167-179 (2008).
    • (2008) Assay Drug Dev Technol , vol.6 , pp. 167-179
    • Trivedi, S.1
  • 291
    • 32844466512 scopus 로고    scopus 로고
    • State-dependent compound inhibition of Nav1.2 sodium channels using the FLIPR Vm dye: On-target and off-target effects of diverse pharmacological agents
    • Benjamin ER et al. State-dependent compound inhibition of Nav1.2 sodium channels using the FLIPR Vm dye: On-target and off-target effects of diverse pharmacological agents. J Biomol Screen 11:29-39 (2006).
    • (2006) J Biomol Screen , vol.11 , pp. 29-39
    • Benjamin, E.R.1
  • 292
    • 0036671206 scopus 로고    scopus 로고
    • A flexible technology platform to explore valuable drug targets
    • Connick JH. A flexible technology platform to explore valuable drug targets. Biochem Soc Trans 30:786-788 (2002).
    • (2002) Biochem Soc Trans , vol.30 , pp. 786-788
    • Connick, J.H.1
  • 293
    • 33645733252 scopus 로고    scopus 로고
    • Characterization of voltage-gated sodium-channel blockers by electrical stimulation and fluorescence detection of membrane potential.
    • Huang CJ et al. Characterization of voltage-gated sodium-channel blockers by electrical stimulation and fluorescence detection of membrane potential. Nat Biotechnol 24:439- 446 (2006).
    • (2006) Nat Biotechnol , vol.24 , pp. 439-446
    • Huang, C.J.1
  • 294
    • 84859494509 scopus 로고    scopus 로고
    • Assay dependent activity of the sodium channel gating modifier protoxin-I: Implications for sodium channel drug discovery.
    • Bhattacharya A, Wang Q, Wu N, Wickenden AD. Assay dependent activity of the sodium channel gating modifier protoxin-I: Implications for sodium channel drug discovery. FASEB J 23 (Suppl.):998.31 (2009).
    • (2009) FASEB J , vol.23 , Issue.SUPPL
    • Bhattacharya, A.1    Wang, Q.2    Wu, N.3    Wickenden, A.D.4
  • 295
    • 0029269869 scopus 로고
    • Detection of sodium channel toxins: Directed cytotoxicity assays of purified ciguatoxins, brevetoxins, saxitoxins, and seafood extracts
    • Manger RL et al. Detection of sodium channel toxins: Directed cytotoxicity assays of purified ciguatoxins, brevetoxins, saxitoxins, and seafood extracts. J Assoc Official Anal Chem Int 78:521-527 (1995).
    • (1995) J Assoc Official Anal Chem Int , vol.78 , pp. 521-527
    • Manger, R.L.1
  • 296
    • 0026549496 scopus 로고
    • A tissue culture assay for direct detection of sodium channel blocking toxins in bacterial culture supernates
    • Gallacher S, Birkbeck TH. A tissue culture assay for direct detection of sodium channel blocking toxins in bacterial culture supernates. FEMS Microbiol Lett 71:101-107 (1992).
    • (1992) FEMS Microbiol Lett , vol.71 , pp. 101-107
    • Gallacher, S.1    Birkbeck, T.H.2
  • 297
    • 33750546736 scopus 로고    scopus 로고
    • The troublewithmaking combination drugs.
    • FrantzS.The troublewithmaking combination drugs. Nat Rev Drug Discov 5:881-882 (2006).
    • (2006) Nat Rev Drug Discov , vol.5 , pp. 881-882
    • Frantz, S.1
  • 298
    • 66949176303 scopus 로고    scopus 로고
    • Venomics as a drug discovery platform
    • Escoubas P, King GF. Venomics as a drug discovery platform. Expert Rev. Proteom 6:221-224 (2009).
    • (2009) Expert Rev. Proteom , vol.6 , pp. 221-224
    • Escoubas, P.1    King, G.F.2
  • 299
    • 8344266043 scopus 로고    scopus 로고
    • The economics of follow-on drug research and development: Trends in entry rates and the timing of development
    • DiMasi JA, Paquette C. The economics of follow-on drug research and development: Trends in entry rates and the timing of development. Pharmacoeconomics 22:1-14 (2004).
    • (2004) Pharmacoeconomics , vol.22 , pp. 1-14
    • DiMasi, J.A.1    Paquette, C.2
  • 300
    • 34247109045 scopus 로고    scopus 로고
    • Natural products as sources of new drugs over the last 25 years
    • Newman DJ, Cragg GM. Natural products as sources of new drugs over the last 25 years. J Nat Prod 70:461-477 (2007).
    • (2007) J Nat Prod , vol.70 , pp. 461-477
    • Newman, D.J.1    Cragg, G.M.2
  • 301
    • 0242331757 scopus 로고    scopus 로고
    • Therapeutic potential of venom peptides
    • Lewis RJ, Garcia ML. Therapeutic potential of venom peptides. Nat Rev Drug Discov 2:790-802 (2003).
    • (2003) Nat Rev Drug Discov , vol.2 , pp. 790-802
    • Lewis, R.J.1    Garcia, M.L.2
  • 302
    • 77951558539 scopus 로고    scopus 로고
    • Use of venom peptides to probe lon channel structure and function
    • Dutertre S, Lewis RJ. Use of venom peptides to probe lon channel structure and function. J. Biol. Chem. 285:13315-13320 (2010).
    • (2010) J. Biol. Chem. , vol.285 , pp. 13315-13320
    • Dutertre, S.1    Lewis, R.J.2
  • 303
    • 52949131528 scopus 로고    scopus 로고
    • Animal peptides targeting voltage-activated sodium channels
    • Billen B, Bosmans F, Tytgat J. Animal peptides targeting voltage-activated sodium channels. Curr Pharm Design 14:2492-2502 (2008).
    • (2008) Curr Pharm Design , vol.14 , pp. 2492-2502
    • Billen, B.1    Bosmans, F.2    Tytgat, J.3
  • 304
    • 0024972502 scopus 로고
    • Substantial increase of protein stability by multiple disulphide bonds
    • Matsumura M, Signor G, Matthews BW. Substantial increase of protein stability by multiple disulphide bonds. Nature 342:291-293 (1989).
    • (1989) Nature , vol.342 , pp. 291-293
    • Matsumura, M.1    Signor, G.2    Matthews, B.W.3
  • 305
    • 33845992556 scopus 로고    scopus 로고
    • Conus peptides: Biodiversity-based discovery and exogenomics
    • Olivera BM. Conus peptides: Biodiversity-based discovery and exogenomics. J Biol Chem 281:31173-31177 (2006).
    • (2006) J Biol Chem , vol.281 , pp. 31173-31177
    • Olivera, B.M.1
  • 307
    • 28444442251 scopus 로고    scopus 로고
    • Overview of scorpion toxins specific for Na+ channels and related peptides: Biodiversity, structure-function relationships and evolution
    • Rodriguez de la Vega RC, Possani LD. Overview of scorpion toxins specific for Na+ channels and related peptides: Biodiversity, structure-function relationships and evolution. Toxicon 46:831-844 (2005).
    • (2005) Toxicon , vol.46 , pp. 831-844
    • Rodriguez de la Vega, R.C.1    Possani, L.D.2
  • 309
    • 33846474874 scopus 로고    scopus 로고
    • Voltage-gated sodium channel modulation by scorpion alphatoxins
    • Bosmans F, Tytgat J. Voltage-gated sodium channel modulation by scorpion alphatoxins. Toxicon 49:142-158 (2007).
    • (2007) Toxicon , vol.49 , pp. 142-158
    • Bosmans, F.1    Tytgat, J.2
  • 310
    • 77955564975 scopus 로고    scopus 로고
    • The world spider catalog, version 12.5.
    • DOI: 10.5531/db.iz.0001.
    • Platnick NI. The world spider catalog, version 12.5. American Museum of Natural History, 2012, online at http://research.amnh.org/iz/spiders/catalog. DOI: 10.5531/db.iz.0001.
    • (2012) American Museum of Natural History
    • Platnick, N.I.1
  • 311
    • 33646133193 scopus 로고    scopus 로고
    • Venomlandscapes:Mining the complexity of spider venoms via a combined cDNA and mass spectrometric approach
    • Escoubas P, Sollod B, King GF. Venomlandscapes:Mining the complexity of spider venoms via a combined cDNA and mass spectrometric approach. Toxicon 47:650-663 (2006).
    • (2006) Toxicon , vol.47 , pp. 650-663
    • Escoubas, P.1    Sollod, B.2    King, G.F.3
  • 312
    • 11244285099 scopus 로고    scopus 로고
    • Electrospray ionization quadrupole time-of-flight and matrixassisted laser desorption/ionization tandem time-of-flight mass spectrometric analyses to solvemicro-heterogeneity in post-translationally modified peptides from Phoneutria nigriventer (Aranea, Ctenidae) venom
    • Pimenta AM et al. Electrospray ionization quadrupole time-of-flight and matrixassisted laser desorption/ionization tandem time-of-flight mass spectrometric analyses to solvemicro-heterogeneity in post-translationally modified peptides from Phoneutria nigriventer (Aranea, Ctenidae) venom. Rapid Commun Mass Spectrom 19:31-37 (2005).
    • (2005) Rapid Commun Mass Spectrom , vol.19 , pp. 31-37
    • Pimenta, A.M.1
  • 313
    • 0031796848 scopus 로고    scopus 로고
    • The cystine knot structure of ion channel toxins and related polypeptides
    • Norton RS, Pallaghy PK. The cystine knot structure of ion channel toxins and related polypeptides. Toxicon 36:1573-1583 (1998).
    • (1998) Toxicon , vol.36 , pp. 1573-1583
    • Norton, R.S.1    Pallaghy, P.K.2
  • 314
    • 0037033085 scopus 로고    scopus 로고
    • Function and solution structure of huwentoxin-IV, a potent neuronal tetrodotoxin (TTX)-sensitive sodium channel antagonist from Chinese bird spider Selenocosmia huwena
    • Peng K, Shu Q, Liu Z, Liang S. Function and solution structure of huwentoxin-IV, a potent neuronal tetrodotoxin (TTX)-sensitive sodium channel antagonist from Chinese bird spider Selenocosmia huwena. J Biol Chem 277:47564-47571 (2002).
    • (2002) J Biol Chem , vol.277 , pp. 47564-47571
    • Peng, K.1    Shu, Q.2    Liu, Z.3    Liang, S.4
  • 315
    • 0347816398 scopus 로고    scopus 로고
    • Inhibition of neuronal tetrodotoxin-sensitive Na+ channels by two spider toxins: Hainantoxin-III and hainantoxin-IV
    • Xiao Y, Liang S. Inhibition of neuronal tetrodotoxin-sensitive Na+ channels by two spider toxins: Hainantoxin-III and hainantoxin-IV. Eur J Pharmacol 477:1-7 (2003).
    • (2003) Eur J Pharmacol , vol.477 , pp. 1-7
    • Xiao, Y.1    Liang, S.2
  • 316
    • 0037408077 scopus 로고    scopus 로고
    • Purification and characterization of Hainantoxin-V, a tetrodotoxinsensitive sodium channel inhibitor from the venom of the spider Selenocosmia hainana
    • Xiao YC, Liang SP. Purification and characterization of Hainantoxin-V, a tetrodotoxinsensitive sodium channel inhibitor from the venom of the spider Selenocosmia hainana. Toxicon 41:643-650 (2003).
    • (2003) Toxicon , vol.41 , pp. 643-650
    • Xiao, Y.C.1    Liang, S.P.2
  • 317
    • 34548421038 scopus 로고    scopus 로고
    • Characterization of the excitatory mechanism induced by Jingzhaotoxin-I inhibiting sodium channel inactivation
    • Xiao Y, Li J, Deng M, Dai C, Liang S. Characterization of the excitatory mechanism induced by Jingzhaotoxin-I inhibiting sodium channel inactivation. Toxicon 50:507-517 (2007).
    • (2007) Toxicon , vol.50 , pp. 507-517
    • Xiao, Y.1    Li, J.2    Deng, M.3    Dai, C.4    Liang, S.5
  • 318
    • 16844367468 scopus 로고    scopus 로고
    • Jingzhaotoxin-I, a novel spider neurotoxin preferentially inhibiting cardiac sodium channel inactivation
    • Xiao Yet al. Jingzhaotoxin-I, a novel spider neurotoxin preferentially inhibiting cardiac sodium channel inactivation. J Biol Chem 280:12069-12076 (2005).
    • (2005) J Biol Chem , vol.280 , pp. 12069-12076
    • Xiao, Y.1
  • 319
    • 1942438958 scopus 로고    scopus 로고
    • Structure and function of [delta]- atracotoxins: Lethal neurotoxins targeting the voltage-gated sodium channel
    • Nicholson GM, Little MJ, Birinyi-Strachan LC. Structure and function of [delta]- atracotoxins: Lethal neurotoxins targeting the voltage-gated sodium channel. Toxicon 43:587-599 (2004).
    • (2004) Toxicon , vol.43 , pp. 587-599
    • Nicholson, G.M.1    Little, M.J.2    Birinyi-Strachan, L.C.3
  • 320
    • 0031761564 scopus 로고    scopus 로고
    • [delta]-Atracotoxins from Australian funnel-web spiders compete with scorpion [alpha]-toxin binding on both rat brain and insect sodium channels
    • Little MJ et al. [delta]-Atracotoxins from Australian funnel-web spiders compete with scorpion [alpha]-toxin binding on both rat brain and insect sodium channels. FEBS Lett 439:246-252 (1998).
    • (1998) FEBS Lett , vol.439 , pp. 246-252
    • Little, M.J.1
  • 321
    • 0037408173 scopus 로고    scopus 로고
    • Purification and characterization of raventoxin-I and raventoxin-III, two neurotoxic peptides from the venom of the spider Macrothele raveni
    • Zeng X-Z, Xiao Q-B, Liang S-P. Purification and characterization of raventoxin-I and raventoxin-III, two neurotoxic peptides from the venom of the spider Macrothele raveni. Toxicon 41:651-656 (2003).
    • (2003) Toxicon , vol.41 , pp. 651-656
    • Zeng, X.-Z.1    Xiao, Q.-B.2    Liang, S.-P.3
  • 322
    • 0038304846 scopus 로고    scopus 로고
    • Distinct primary structures of the major peptide toxins from the venom of the spider Macrothele gigas that bind to sites 3 and 4 in the sodium channel
    • Corzo G et al. Distinct primary structures of the major peptide toxins from the venom of the spider Macrothele gigas that bind to sites 3 and 4 in the sodium channel. FEBS Lett 547:43-50 (2003).
    • (2003) FEBS Lett , vol.547 , pp. 43-50
    • Corzo, G.1
  • 323
    • 2942718770 scopus 로고    scopus 로고
    • Jingzhaotoxin-III, a novel spider toxin inhibiting activation of voltage-gated sodium channel in rat cardiac myocytes
    • Xiao Yet al. Jingzhaotoxin-III, a novel spider toxin inhibiting activation of voltage-gated sodium channel in rat cardiac myocytes. J Biol Chem 279:26220-26226 (2004).
    • (2004) J Biol Chem , vol.279 , pp. 26220-26226
    • Xiao, Y.1
  • 324
    • 0030729069 scopus 로고    scopus 로고
    • Conus venom peptides, receptor and ion channel targets, and drug design: 50 million years of neuropharmacology.
    • Just Lecture 1996
    • Olivera BM. E.E. Just Lecture, 1996. Conus venom peptides, receptor and ion channel targets, and drug design: 50 million years of neuropharmacology. Mol Biol Cell 8:2101- 2109 (1997).
    • (1997) Mol Biol Cell , vol.8 , pp. 2101-2109
    • Olivera, B.M.1
  • 325
    • 0029767552 scopus 로고    scopus 로고
    • Strategy for rapid immobilization of prey by a fish-hunting marine snail
    • Terlau H et al. Strategy for rapid immobilization of prey by a fish-hunting marine snail. Nature 381:148-151 (1996).
    • (1996) Nature , vol.381 , pp. 148-151
    • Terlau, H.1
  • 326
    • 0036930324 scopus 로고    scopus 로고
    • Conus venom peptides: Reflections from the biology of clades and species.
    • Olivera BM Conus venom peptides: Reflections from the biology of clades and species. Annu Rev Ecol Systematics 33:25-47 (2002).
    • (2002) Annu Rev Ecol Systematics , vol.33 , pp. 25-47
    • Olivera, B.M.1
  • 327
    • 0347989461 scopus 로고    scopus 로고
    • Conus venoms: A rich source of novel ion channel-targeted peptides
    • Terlau H, Olivera BM. Conus venoms: A rich source of novel ion channel-targeted peptides. Physiol Rev 84:41-68 (2004).
    • (2004) Physiol Rev , vol.84 , pp. 41-68
    • Terlau, H.1    Olivera, B.M.2
  • 328
    • 35349014163 scopus 로고    scopus 로고
    • Diversity of the neurotoxic Conus peptides: A model for concerted pharmacological discovery
    • Olivera BM, Teichert RW. Diversity of the neurotoxic Conus peptides: A model for concerted pharmacological discovery. Mol Interv 7:251-260 (2007).
    • (2007) Mol Interv , vol.7 , pp. 251-260
    • Olivera, B.M.1    Teichert, R.W.2
  • 329
    • 0025300052 scopus 로고
    • Diversity of Conus neuropeptides
    • Olivera BM et al. Diversity of Conus neuropeptides. Science 249:257-263 (1990).
    • (1990) Science , vol.249 , pp. 257-263
    • Olivera, B.M.1
  • 330
    • 67349175001 scopus 로고    scopus 로고
    • Remarkable inter- and intra-species complexity of conotoxins revealed by LC/MS
    • Davis J, Jones A, Lewis RJ. Remarkable inter- and intra-species complexity of conotoxins revealed by LC/MS. Peptides 30:1222-1227 (2009).
    • (2009) Peptides , vol.30 , pp. 1222-1227
    • Davis, J.1    Jones, A.2    Lewis, R.J.3
  • 331
    • 33847158419 scopus 로고    scopus 로고
    • Isolation and structure-activity of m-conotoxin TIIIA, a potent inhibitor of tetrodotoxin-sensitive voltage-gated sodium channels
    • Lewis RJ et al. Isolation and structure-activity of m-conotoxin TIIIA, a potent inhibitor of tetrodotoxin-sensitive voltage-gated sodium channels. Mol Pharmacol 71:676-685 (2007).
    • (2007) Mol Pharmacol , vol.71 , pp. 676-685
    • Lewis, R.J.1
  • 332
    • 33645237703 scopus 로고    scopus 로고
    • Structural and functional diversities among mu-conotoxins targeting TTX-resistant sodium channels
    • Zhang MM et al. Structural and functional diversities among mu-conotoxins targeting TTX-resistant sodium channels. Biochemistry 45:3723-3732 (2006).
    • (2006) Biochemistry , vol.45 , pp. 3723-3732
    • Zhang, M.M.1
  • 333
    • 48249132363 scopus 로고    scopus 로고
    • Conotoxin modulation of voltage-gated sodium channels
    • Ekberg J, Craik DJ, Adams DJ. Conotoxin modulation of voltage-gated sodium channels. Int J Biochem Cell Biol 40:2363-2368 (2008).
    • (2008) Int J Biochem Cell Biol , vol.40 , pp. 2363-2368
    • Ekberg, J.1    Craik, D.J.2    Adams, D.J.3
  • 334
    • 0029658417 scopus 로고    scopus 로고
    • MicroO-conotoxin MrVIA inhibits mammalian sodium channels, but not through site I
    • Terlau H, Stocker M, Shon KJ, McIntosh JM, Olivera BM. MicroO-conotoxin MrVIA inhibits mammalian sodium channels, but not through site I. J Neurophysiol 76:1423-1429 (1996).
    • (1996) J Neurophysiol , vol.76 , pp. 1423-1429
    • Terlau, H.1    Stocker, M.2    Shon, K.J.3    McIntosh, J.M.4    Olivera, B.M.5
  • 335
    • 32344433189 scopus 로고    scopus 로고
    • The [mu]O-conotoxin MrVIA inhibits voltage-gated sodium channels by associating with domain-3
    • Zorn S et al. The [mu]O-conotoxin MrVIA inhibits voltage-gated sodium channels by associating with domain-3. FEBS Lett 580:1360-1364 (2006).
    • (2006) FEBS Lett , vol.580 , pp. 1360-1364
    • Zorn, S.1
  • 336
    • 33750941949 scopus 로고    scopus 로고
    • muO-conotoxin MrVIB selectively blocks Nav1.8 sensory neuron specific sodium channels and chronic pain behavior without motor deficits
    • Ekberg J et al. muO-conotoxin MrVIB selectively blocks Nav1.8 sensory neuron specific sodium channels and chronic pain behavior without motor deficits. Proc Natl Acad Sci USA 103:17030-17035 (2006).
    • (2006) Proc Natl Acad Sci USA , vol.103 , pp. 17030-17035
    • Ekberg, J.1
  • 337
    • 33745034230 scopus 로고    scopus 로고
    • Synthetic muO-conotoxin MrVIB blocks TTX-resistant sodium channel NaV1.8 and has a long-lasting analgesic activity
    • Bulaj G et al. Synthetic muO-conotoxin MrVIB blocks TTX-resistant sodium channel NaV1.8 and has a long-lasting analgesic activity. Biochemistry 45:7404-7414 (2006).
    • (2006) Biochemistry , vol.45 , pp. 7404-7414
    • Bulaj, G.1
  • 338
    • 0035818423 scopus 로고    scopus 로고
    • Delta-conotoxin structure/function through a cladistic analysis
    • Bulaj G et al. Delta-conotoxin structure/function through a cladistic analysis. Biochemistry 40:13201-13208 (2001).
    • (2001) Biochemistry , vol.40 , pp. 13201-13208
    • Bulaj, G.1
  • 339
    • 0038335917 scopus 로고    scopus 로고
    • Novel excitatory Conus peptides define a new conotoxin superfamily
    • Jimenez EC et al. Novel excitatory Conus peptides define a new conotoxin superfamily. J Neurochem 85:610-621 (2003).
    • (2003) J Neurochem , vol.85 , pp. 610-621
    • Jimenez, E.C.1
  • 340
    • 70349623944 scopus 로고    scopus 로고
    • Identification of a novel M-superfamily conotoxin with the ability to enhance tetrodotoxin sensitive sodium currents
    • Wang L et al. Identification of a novel M-superfamily conotoxin with the ability to enhance tetrodotoxin sensitive sodium currents. Arch Toxicol 83:925-932 (2009).
    • (2009) Arch Toxicol , vol.83 , pp. 925-932
    • Wang, L.1
  • 341
    • 36248939803 scopus 로고    scopus 로고
    • Isolation and characterization of aT-superfamily conotoxin fromConus litteratus with targeting tetrodotoxin-sensitive sodium channels
    • Liu J et al. Isolation and characterization of aT-superfamily conotoxin fromConus litteratus with targeting tetrodotoxin-sensitive sodium channels. Peptides 28:2313-2319 (2007).
    • (2007) Peptides , vol.28 , pp. 2313-2319
    • Liu, J.1
  • 342
    • 33644681622 scopus 로고    scopus 로고
    • A biologically active hydrophobic T-1-conotoxin from the venom of Conus spurius
    • Aguilar MB et al. A biologically active hydrophobic T-1-conotoxin from the venom of Conus spurius. Peptides 27:500-505 (2006).
    • (2006) Peptides , vol.27 , pp. 500-505
    • Aguilar, M.B.1
  • 343
    • 35349014163 scopus 로고    scopus 로고
    • Diversity of the neurotoxic conus peptides: A model for concerted pharmacological discovery
    • Olivera BM, Teichert RW. Diversity of the neurotoxic conus peptides: A model for concerted pharmacological discovery. Mol Interv 7:251-260 (2007).
    • (2007) Mol Interv , vol.7 , pp. 251-260
    • Olivera, B.M.1    Teichert, R.W.2
  • 344
    • 11144319833 scopus 로고    scopus 로고
    • Were arachnids the first to use combinatorial peptide libraries?
    • Sollod BL et al.Were arachnids the first to use combinatorial peptide libraries? Peptides 26:131-139 (2005).
    • (2005) Peptides , vol.26 , pp. 131-139
    • Sollod, B.L.1
  • 345
    • 1942438962 scopus 로고    scopus 로고
    • Tarantulas: Eight-legged pharmacists and combinatorial chemists
    • Escoubas P, Rash L. Tarantulas: Eight-legged pharmacists and combinatorial chemists. Toxicon 43:555-574 (2004).
    • (2004) Toxicon , vol.43 , pp. 555-574
    • Escoubas, P.1    Rash, L.2
  • 346
    • 65549094126 scopus 로고    scopus 로고
    • Clinical spectrum of SCN1A mutations.
    • Gambardella A, Marini C. Clinical spectrum of SCN1A mutations. Epilepsia 50 (Suppl 5):20-23 (2009).
    • (2009) Epilepsia , vol.50 , Issue.SUPPL 5 , pp. 20-23
    • Gambardella, A.1    Marini, C.2
  • 347
    • 0026767259 scopus 로고
    • Primary structure, chromosomal localization, and functional expression of a voltage-gated sodium channel from human brain
    • Ahmed CM et al. Primary structure, chromosomal localization, and functional expression of a voltage-gated sodium channel from human brain. Proc Natl Acad Sci USA 89:8220-8224 (1992).
    • (1992) Proc Natl Acad Sci USA , vol.89 , pp. 8220-8224
    • Ahmed, C.M.1
  • 348
    • 48649086182 scopus 로고    scopus 로고
    • Nav1.4 deregulation in dystrophic skeletal muscle leads to Na+overload and enhanced cell death
    • Hirn C, Shapovalov G, Petermann O, Roulet E, Ruegg UT. Nav1.4 deregulation in dystrophic skeletal muscle leads to Na+overload and enhanced cell death. J Gen Physiol 132:199-208 (2008).
    • (2008) J Gen Physiol , vol.132 , pp. 199-208
    • Hirn, C.1    Shapovalov, G.2    Petermann, O.3    Roulet, E.4    Ruegg, U.T.5
  • 349
    • 67649397890 scopus 로고    scopus 로고
    • Voltage sensor charge loss accounts for most cases of hypokalemic periodic paralysis
    • Matthews E et al. Voltage sensor charge loss accounts for most cases of hypokalemic periodic paralysis. Neurology 72:1544-1547 (2009).
    • (2009) Neurology , vol.72 , pp. 1544-1547
    • Matthews, E.1
  • 350
    • 77949494524 scopus 로고    scopus 로고
    • Persistent Nav1.6 current at axon initial segments tunes spike timing of cerebellar granule cells.
    • Osorio N et al. Persistent Nav1.6 current at axon initial segments tunes spike timing of cerebellar granule cells. J Physiol 588(Pt 4): 651-670 (2010).
    • (2010) J Physiol , vol.588 , Issue.PART 4 , pp. 651-670
    • Osorio, N.1
  • 351
    • 64949083602 scopus 로고    scopus 로고
    • Increased peripheral nerve excitability and local NaV1.8 mRNA upregulation in painful neuropathy.
    • Thakor DK et al. Increased peripheral nerve excitability and local NaV1.8 mRNA upregulation in painful neuropathy. Mol Pain 5:14 (2009).
    • (2009) Mol Pain , vol.5 , pp. 14
    • Thakor, D.K.1
  • 352
    • 0027507511 scopus 로고
    • Lamotrigine, phenytoin and carbamazepine interactions on the sodium current present in N4TG1 mouse neuroblastoma cells
    • Lang DG, Wang CM, Cooper BR. Lamotrigine, phenytoin and carbamazepine interactions on the sodium current present in N4TG1 mouse neuroblastoma cells. J Pharmacol Exp Ther 266:829-835 (1993).
    • (1993) J Pharmacol Exp Ther , vol.266 , pp. 829-835
    • Lang, D.G.1    Wang, C.M.2    Cooper, B.R.3
  • 353
    • 0033023323 scopus 로고    scopus 로고
    • Inhibition of transient and persistent Na+ current fractions by the new anticonvulsant topiramate
    • Taverna S, Sancini G, Mantegazza M, Franceschetti S,Avanzini G. Inhibition of transient and persistent Na+ current fractions by the new anticonvulsant topiramate. J Pharmacol Exp Ther 288:960-968 (1999).
    • (1999) J Pharmacol Exp Ther , vol.288 , pp. 960-968
    • Taverna, S.1    Sancini, G.2    Mantegazza, M.3    Franceschetti, S.4    Avanzini, G.5
  • 354
    • 37349071555 scopus 로고    scopus 로고
    • The investigational anticonvulsant lacosamide selectively enhances slow inactivation of voltage-gated sodium channels
    • Errington AC, Stohr T, Heers C, Lees G. The investigational anticonvulsant lacosamide selectively enhances slow inactivation of voltage-gated sodium channels. Mol Pharmacol 73:157-169 (2008).
    • (2008) Mol Pharmacol , vol.73 , pp. 157-169
    • Errington, A.C.1    Stohr, T.2    Heers, C.3    Lees, G.4
  • 355
    • 0034637353 scopus 로고    scopus 로고
    • Amitriptyline modulation of Na+ channels in rat dorsal root ganglion neurons
    • Song J-H, Ham S-S, Shin Y-K, Lee C-S. Amitriptyline modulation of Na+ channels in rat dorsal root ganglion neurons. Eur J Pharmacol 401:297-305 (2000).
    • (2000) Eur J Pharmacol , vol.401 , pp. 297-305
    • Song, J.-H.1    Ham, S.-S.2    Shin, Y.-K.3    Lee, C.-S.4
  • 356
    • 35348997034 scopus 로고    scopus 로고
    • Effects of ralfinamide, a Na+ channel blocker, on firing properties of nociceptive dorsal root ganglion neurons of adult rats
    • Yamane H, de Groat WC, Sculptoreanu A. Effects of ralfinamide, a Na+ channel blocker, on firing properties of nociceptive dorsal root ganglion neurons of adult rats. Exp Neurol 208:63-72 (2007).
    • (2007) Exp Neurol , vol.208 , pp. 63-72
    • Yamane, H.1    de Groat, W.C.2    Sculptoreanu, A.3
  • 357
    • 34347213786 scopus 로고    scopus 로고
    • A-803467, a potent and selective Nav1.8 sodium channel blocker, attenuates neuropathic and inflammatory pain in the rat
    • Jarvis MF et al. A-803467, a potent and selective Nav1.8 sodium channel blocker, attenuates neuropathic and inflammatory pain in the rat. Proc Natl Acad Sci USA 104:8520-8525 (2007).
    • (2007) Proc Natl Acad Sci USA , vol.104 , pp. 8520-8525
    • Jarvis, M.F.1
  • 358
    • 0025740422 scopus 로고
    • Neuropathic pain can be relieved by drugs that are usedependent sodium channel blockers: Lidocaine, carbamazepine, and mexiletine
    • Tanelian DL, Brose WG. Neuropathic pain can be relieved by drugs that are usedependent sodium channel blockers: Lidocaine, carbamazepine, and mexiletine. Anesthesiology 74:949-951 (1991).
    • (1991) Anesthesiology , vol.74 , pp. 949-951
    • Tanelian, D.L.1    Brose, W.G.2
  • 360
    • 64149085800 scopus 로고    scopus 로고
    • Flecainide prevents catecholaminergic polymorphic ventricular tachycardia in mice and humans
    • Watanabe H et al. Flecainide prevents catecholaminergic polymorphic ventricular tachycardia in mice and humans. Nat Med 15:380-383 (2009).
    • (2009) Nat Med , vol.15 , pp. 380-383
    • Watanabe, H.1
  • 361
    • 78049410367 scopus 로고    scopus 로고
    • Ranolazine selectively blocks persistent current evoked by epilepsy-associated NaV1.1 mutations
    • Kahlig KM, Lepist I, Leung K, Rajamani S, George AL. Ranolazine selectively blocks persistent current evoked by epilepsy-associated NaV1.1 mutations. Br J Pharmacol 161:1414-1426 (2010).
    • (2010) Br J Pharmacol , vol.161 , pp. 1414-1426
    • Kahlig, K.M.1    Lepist, I.2    Leung, K.3    Rajamani, S.4    George, A.L.5
  • 362
    • 0019169574 scopus 로고
    • Differential sensitivities of mammalian nerve fibers to local anesthetic agents
    • Gissen AJ, Covino BG, Gregus J. Differential sensitivities of mammalian nerve fibers to local anesthetic agents. Anesthesiology 53:467-474 (1980).
    • (1980) Anesthesiology , vol.53 , pp. 467-474
    • Gissen, A.J.1    Covino, B.G.2    Gregus, J.3
  • 363
    • 0021359561 scopus 로고
    • Voltage-dependent block by saxitoxin of sodium channels incorporated into planar lipid bilayers
    • French RJ,Worley, JF 3rd Krueger BK. Voltage-dependent block by saxitoxin of sodium channels incorporated into planar lipid bilayers. Biophysical J 45:301-310 (1984).
    • (1984) Biophysical J , vol.45 , pp. 301-310
    • French, R.J.1    Worley, J.F.2    Krueger, B.K.3
  • 365
    • 0032940658 scopus 로고    scopus 로고
    • Differential actions of pacific ciguatoxin-1 on sodium channel subtypes in mammalian sensory neurons
    • Strachan LC, Lewis RJ, Nicholson GM. Differential actions of pacific ciguatoxin-1 on sodium channel subtypes in mammalian sensory neurons. J Pharmacol Exp Ther 288:379-388 (1999).
    • (1999) J Pharmacol Exp Ther , vol.288 , pp. 379-388
    • Strachan, L.C.1    Lewis, R.J.2    Nicholson, G.M.3
  • 366
    • 70349959675 scopus 로고    scopus 로고
    • Brevetoxins 2 3, 6, and 9 show variability in potency and cause significant induction of DNA damage and apoptosis in Jurkat E6-1 cells
    • Murrell R, Gibson J. Brevetoxins 2 3, 6, and 9 show variability in potency and cause significant induction of DNA damage and apoptosis in Jurkat E6-1 cells. Arch Toxicology 83:1009-1019 (2009).
    • (2009) Arch Toxicology , vol.83 , pp. 1009-1019
    • Murrell, R.1    Gibson, J.2
  • 367
    • 0037648783 scopus 로고    scopus 로고
    • Veratridine block of rat skeletal muscle Nav1.4 sodium channels in the inner vestibule.
    • Wang GK,Wang SY. Veratridine block of rat skeletal muscle Nav1.4 sodium channels in the inner vestibule. J Physiol 548:667-675 (2003).
    • (2003) J Physiol , vol.548 , pp. 667-675
    • Wang, G.K.1    Wang, S.Y.2


* 이 정보는 Elsevier사의 SCOPUS DB에서 KISTI가 분석하여 추출한 것입니다.