Nav1.7

SCN9A
Available structures
PDBOrtholog search: PDBe RCSB
Identifiers
Aliases SCN9A, ETHA, FEB3B, GEFSP7, HSAN2D, NE-NA, NENA, Nav1.7, PN1, SFNP, sodium voltage-gated channel alpha subunit 9
External IDs MGI: 107636 HomoloGene: 2237 GeneCards: SCN9A
Genetically Related Diseases
dental caries[1]
Targeted by Drug
lacosamide, saxitoxin[2]
Orthologs
Species Human Mouse
Entrez

6335

20274

Ensembl

ENSG00000169432

ENSMUSG00000075316

UniProt

Q15858

Q62205

RefSeq (mRNA)

NM_002977

NM_001290674
NM_001290675
NM_018852

RefSeq (protein)

NP_002968.1

NP_001277603.1

Location (UCSC) Chr 2: 166.2 – 166.38 Mb Chr 2: 66.48 – 66.63 Mb
PubMed search [3] [4]
Wikidata
View/Edit HumanView/Edit Mouse

Nav1.7 is a sodium ion channel that in humans is encoded by the SCN9A gene.[5][6][7] It is usually expressed at high levels in two types of neurons, the nociceptive (pain) neurons at dorsal root ganglion (DRG) and trigeminal ganglion, and sympathetic ganglion neurons, which are part of the autonomic (involuntary) nervous system.[8][9]

Function

Nav1.7 is a voltage-gated sodium channel and plays a critical role in the generation and conduction of action potentials and is thus important for electrical signaling by most excitable cells. Nav1.7 is present at the endings of pain-sensing nerves, the nociceptors, close to the region where the impulse is initiated. Stimulation of the nociceptor nerve endings produces "generator potentials", which are small changes in the voltage across the neuronal membranes. The Nav1.7 channel amplifies these membrane depolarizations, and when the membrane potential difference reaches a specific threshold, the neuron fires. In sensory neurons, multiple voltage-dependent sodium currents can be differentiated by their voltage dependence and by sensitivity to the voltage-gated sodium-channel blocker tetrodotoxin. The Nav1.7 channel produces a rapidly activating and inactivating current which is sensitive to the level of tetrodotoxin.[10] Nav1.7 is important in the early phases of neuronal electrogenesis. Nav1.7 activity consists of a slow transition of the channel into an inactive state when it is depolarized, even to a minor degree. This property allows these channels to remain available for activation with even small or slowly developing depolarizations. Stimulation of the nociceptor nerve endings produces "generator potentials", small changes in the voltage across the neuronal membranes. This brings neurons to a voltage that stimulate Nav1.8, which has a more depolarized activation threshold that produces most of the transmembrane current responsible for the depolarizing phase of action potentials.

Clinical significance

Animal studies

The critical role of Nav1.7 in nociception and pain was originally shown using Cre-Lox recombination tissue specific knockout mice. These transgenic mice specifically lack Nav1.7 in Nav1.8 positive nociceptors and showed reduced behavioural responses, specifically to acute mechanical and inflammatory pain assays. At the same time, behavioural responses to acute thermal and neuropathic pain assays remained intact.[11] However, the expression of Nav1.7 is not restricted to Nav1.8 positive DRG neurons. Further work examining the behavioural response of two other transgenic mouse strains; one lacking Nav1.7 in all DRG neurons and the other lacking Nav1.7 in all DRG neurons as well as all sympathetic neurons, has revealed distinct sets of modality specific peripheral neurons.[12] Therefore, Nav1.7 expressed in Nav1.8 positive DRG neurons is critical for normal responses to acute mechanical and inflammatory pain assays. Whilst Nav1.7 expressed in Nav1.8 negative DRG neurons is critical for normal responses to acute thermal pain assays. Finally, Nav1.7 expressed in sympathetic neurons is critical for normal behavioural responses to neuropathic pain assays.

Primary erythromelalgia

Mutation in Nav1.7 may result in primary erythromelalgia (PE), an autosomal dominant, inherited disorder which is characterized by attacks or episodes of symmetrical burning pain of the feet, lower legs, and sometimes hands, elevated skin temperature of affected areas, and reddened extremities. The mutation causes excessive channel activity which suggests that Nav1.7 sets the gain on pain signaling in humans. It was observed that a missense mutation in the SCN9A gene affected conserved residues in the pore-forming α subunit of the Nav1.7 channel. Many studies have found a dozen SCN9A mutations in multiple families as causing erythromelagia. All of the observed erythromelalgia mutations that are observed are missense mutations that change important and highly conserved amino acid residues of the Nav1.7 protein. The majority of mutations that cause PE are located in cytoplasmic linkers of the Nav1.7 channel, however some mutations are present in transmembrane domains of the channel. The PE mutations cause a hyperpolarizing shift in the voltage dependence of channel activation, which allows the channel to be activated by smaller than normal depolarizations, thus enhancing the activity of Nav1.7. Moreover, the majority of the PE mutations also slow deactivation, thus keeping the channel open longer once it is activated.[13] In addition, in response to a slow, depolarizing stimulus, most mutant channels will generate a larger than normal sodium current. Each of these alterations in activation and deactivation can contribute to the hyperexcitability of pain-signaling DRG neurons expressing these mutant channels, thus causing extreme sensitivity to pain (hyperalgesia). While the expression of PE Nav1.7 mutations produces hyperexcitability in DRG neurons, studies on cultured rat in sympathetic ganglion neurons indicate that expression of these same PE mutations results in reduction of excitability of these cells. This occurs because Nav1.8 channels, which are selectively expressed in addition to Nav1.7 in DRG neurons, are not present within sympathetic ganglion neurons. Thus lack of Nav1.7 results in inactivation of the sodium channels results in reduced excitability. Thus physiological interaction of Nav1.7 and Nav1.8 can explain the reason that PE presents with pain due to hyperexcitability of nociceptors and with sympathetic dysfunction that is most likely due to hypoexcitability of sympathetic ganglion neurons.[9] Recent studies have associated a defect in SCN9A with congenital insensitivity to pain.[14]

Paroxysmal extreme pain disorder

Paroxysmal extreme pain disorder (PEPD) is another rare, extreme pain disorder.[15][16] Like primary erythromelalgia, PEPD is similarly the result of a gain-of-function mutation in the gene encoding the Nav1.7 channel.[15][16]

Congenital insensitivity to pain

Individuals with congenital insensitivity to pain have painless injuries beginning in infancy but otherwise normal sensory responses upon examination. Patients frequently have bruises and cuts, and are often only diagnosed because of limping or lack of use of a limb. Individuals have been reported to be able to walk over burning coals and to insert knives and drive spikes through their arms. It has been observed that the insensitivity to pain does not appear to be due to axonal degeneration.

A mutation that causes loss of Nav1.7 function has been detected in three consanguineous families from northern Pakistan. All mutations observed were nonsense mutation, with the majority of affected patients having a homozygous mutation in the SCN9A gene. This discovery linked loss of Nav1.7 function with the inability to experience pain. This is in contrast with the genetic basis of primary erythromelalgia in which the disorder results from gain-of-function mutations.[14]

Clinical analgesics

Local anesthetics such as lidocaine mediate their analgesic effects by non-selectively blocking voltage-gated sodium channels.[17][18] Nav1.7, as well as Nav1.3, Nav1.8, and Nav1.9, are the specific channels that have been implicated in pain signaling.[17][19] Thus, the blockade of these specific channels is likely to underlie the analgesia of local anesthetics.[17] In addition, inhibition of these channels is also likely responsible for the analgesic efficacy of certain anticonvulsants, as well as, in part, that of certain tricyclic antidepressants, and of mexiletine.[20][21]

Itch

Mutations of Nav1.7 have been linked to itching (pruritus),[22][23] and genetic knockouts of Nav1.7[24] and an antibody that inhibits Nav1.7 also appear to inhibit itching.[25][26][27]

Future prospects

As the Nav1.7 channel appears to be a highly important component in nociception, with null activity conferring total analgesia,[16] there has been immense interest in developing selective Nav1.7 channel blockers as potential novel analgesics.[28] Since Nav1.7 is not present in heart tissue or the central nervous system, selective blockers of Nav1.7, unlike non-selective blockers such as local anesthetics, could be safely used systemically for pain relief. Moreover, selective Nav1.7 blockers may prove to be far more effective analgesics, and with fewer undesirable effects, relative to current pharmacotherapies.[28][29][30]

A number of selective Nav1.7 (and/or Nav1.8) blockers are in clinical development, including funapide (formerly TV-45070, XEN402), raxatrigine (formerly CNV1014802, GSK-1014802), PF-05089771, PF-04531083, DSP-2230, AZD-3161, NKTR-171, GDC-0276, and RG7893 (formerly GDC-0287).[31][32][33] Ralfinamide (formerly NW-1029, FCE-26742A, PNU-0154339E) is a multimodal, non-selective Nav channel blocker which is under development for the treatment of pain.[34]

Surprisingly, many potent Nav1.7 blockers have been found to be clinically effective but only relatively weak analgesics.[35] Recently, it has been elucidated that congenital loss of Navv1.7 results in a dramatic increase in the levels of endogenous enkephalins, and it was found that blocking these opioids with the opioid antagonist naloxone allowed for pain sensitivity both in Navv1.7 null mice and in a woman with a defective Navv1.7 gene and associated congenital insensitivity to pain.[35] Apparently complete inactivation of Nav 1.7-mediated sodium efflux is necessary to upregulate enkephalin expression enough to achieve complete analgesia such that "even potent [Nav 1.7] antagonists are likely to fail" in regards to achieving the same degree of analgesia as congenital Nav 1.7 inactivity.[35] As such, the analgesic effects of Nav1.7 blockers may be greatly potentiated by the co-administration of exogenous opioids or enkephalinase inhibitors.[35] Supporting this idea, a strong analgesic synergy between local anesthetics and topical opioids has already been observed in clinical research.[35]

An additionitional implication of the aforementioned findings is that congenital insensitivity to pain may be clinically treatable with opioid antagonists.[35]

References

  1. "Diseases that are genetically associated with SCN9A view/edit references on wikidata".
  2. "Drugs that physically interact with Sodium channel protein type 9 subunit alpha view/edit references on wikidata".
  3. "Human PubMed Reference:".
  4. "Mouse PubMed Reference:".
  5. Klugbauer N, Lacinova L, Flockerzi V, Hofmann F (March 1995). "Structure and functional expression of a new member of the tetrodotoxin-sensitive voltage-activated sodium channel family from human neuroendocrine cells". The EMBO Journal. 14 (6): 1084–90. PMC 398185Freely accessible. PMID 7720699.
  6. Plummer NW, Meisler MH (April 1999). "Evolution and diversity of mammalian sodium channel genes". Genomics. 57 (2): 323–31. doi:10.1006/geno.1998.5735. PMID 10198179.
  7. Catterall WA, Goldin AL, Waxman SG (December 2005). "International Union of Pharmacology. XLVII. Nomenclature and structure-function relationships of voltage-gated sodium channels". Pharmacological Reviews. 57 (4): 397–409. doi:10.1124/pr.57.4.4. PMID 16382098.
  8. Raymond CK, Castle J, Garrett-Engele P, Armour CD, Kan Z, Tsinoremas N, Johnson JM (October 2004). "Expression of alternatively spliced sodium channel alpha-subunit genes. Unique splicing patterns are observed in dorsal root ganglia". The Journal of Biological Chemistry. 279 (44): 46234–41. doi:10.1074/jbc.M406387200. PMID 15302875.
  9. 1 2 Rush AM, Dib-Hajj SD, Liu S, Cummins TR, Black JA, Waxman SG (May 2006). "A single sodium channel mutation produces hyper- or hypoexcitability in different types of neurons". Proceedings of the National Academy of Sciences of the United States of America. 103 (21): 8245–50. doi:10.1073/pnas.0602813103. PMC 1472458Freely accessible. PMID 16702558.
  10. Catterall WA (2000). "Structure and regulation of voltage-gated Ca2+ channels". Annual Review of Cell and Developmental Biology. 16: 521–55. doi:10.1146/annurev.cellbio.16.1.521. PMID 11031246.
  11. Nassar MA, Stirling LC, Forlani G, Baker MD, Matthews EA, Dickenson AH, Wood JN (August 2004). "Nociceptor-specific gene deletion reveals a major role for Nav1.7 (PN1) in acute and inflammatory pain". Proceedings of the National Academy of Sciences of the United States of America. 101 (34): 12706–11. doi:10.1073/pnas.0404915101. PMC 515119Freely accessible. PMID 15314237.
  12. Minett MS, Nassar MA, Clark AK, Passmore G, Dickenson AH, Wang F, Malcangio M, Wood JN (April 2012). "Distinct nav1.7-dependent pain sensations require different sets of sensory and sympathetic neurons". Nature Communications. 3 (4): 791–799. doi:10.1038/ncomms1795. PMC 3337979Freely accessible. PMID 22531176.
  13. Drenth JP, Michiels JJ (June 1994). "Erythromelalgia and erythermalgia: diagnostic differentiation". International Journal of Dermatology. 33 (6): 393–7. doi:10.1111/j.1365-4362.1994.tb04037.x. PMID 8056469.
  14. 1 2 Cox JJ, Reimann F, Nicholas AK, Thornton G, Roberts E, Springell K, Karbani G, Jafri H, Mannan J, Raashid Y, Al-Gazali L, Hamamy H, Valente EM, Gorman S, Williams R, McHale DP, Wood JN, Gribble FM, Woods CG (2006). "An SCN9A channelopathy causes congenital inability to experience pain". Nature. 444 (7121): 894–8. doi:10.1038/nature05413. PMID 17167479.
  15. 1 2 Allerton C, Fox D (2013). Pain Therapeutics: Current and Future Treatment Paradigms. Royal Society of Chemistry. pp. 146–148. ISBN 978-1-84973-645-9.
  16. 1 2 3 Kew JN, Davies CH (2010). Ion Channels: From Structure to Function. Oxford University Press. pp. 153–154. ISBN 978-0-19-929675-0.
  17. 1 2 3 Mashour GA, Lydic R (7 September 2011). Neuroscientific Foundations of Anesthesiology. Oxford University Press. p. 154. ISBN 978-0-19-987546-7.
  18. Mohamed Chahine. Recent advances in voltage-gated sodium channels, their pharmacology and related diseases. Frontiers E-books. p. 90. ISBN 978-2-88919-128-4.
  19. Lamberth C, Dinges J (9 August 2012). Bioactive Heterocyclic Compound Classes: Pharmaceuticals. John Wiley & Sons. p. 127. ISBN 978-3-527-66448-1.
  20. Cairns BE (1 September 2009). Peripheral Receptor Targets for Analgesia: Novel Approaches to Pain Management. John Wiley & Sons. pp. 66–68. ISBN 978-0-470-52221-9.
  21. William D. James; Timothy Berger; Dirk Elston (12 April 2015). Andrews' Diseases of the Skin: Clinical Dermatology. Elsevier Health Sciences. pp. 810–. ISBN 978-0-323-31969-0.
  22. Grazia Devigilia, Roberto Eleopraa, Tiziana Pierrob, Raffaella Lombardib, Sara Rinaldoa, Christian Lettieria, Catharina G. Faberc, Ingemar S.J. Merkiesc, d, Stephen G. Waxmane, Giuseppe Lauriab (September 2014). "Paroxysmal itch caused by gain-of-function Nav1.7 mutation". Pain.
  23. "An SCN9A variant, known to cause pain, is now found to cause itch" (PDF). Pain.
  24. Jacinthe Gingras; Sarah Smith; David J. Matson; Danielle Johnson; Kim Nye; Lauren Couture; Elma Feric; Ruoyuan Yin; Bryan D. Moyer; Matthew L. Peterson; James B. Rottman; Rudolph J. Beiler; Annika B. Malmberg; Stefan I. McDonough (September 4, 2014). "Global Nav1.7 Knockout Mice Recapitulate the Phenotype of Human Congenital Indifference to Pain". PLOS ONE.
  25. Jun-Ho Lee; Chul-Kyu Park; Gang Chen; Qingjian Han; Rou-Gang Xie; Tong Liu; Ru-Rong Ji; Seok-Yong Lee (5 June 2014). "A monoclonal antibody that targets a NaV1.7 channel voltage sensor for pain and itch relief".
  26. Lauren Martz. "Nav-i-gating antibodies for pain". SciBX.
  27. Sheila Yong (May 22, 2014). "One Molecule To Block Both Pain And Itch".
  28. 1 2 Altman RB, Flockhart D, Goldstein DB (23 January 2012). Principles of Pharmacogenetics and Pharmacogenomics. Cambridge University Press. p. 224. ISBN 978-1-107-37747-9.
  29. Waxman SG (December 2006). "Neurobiology: a channel sets the gain on pain". Nature. 444 (7121): 831–2. doi:10.1038/444831a. PMID 17167466.
  30. Dib-Hajj SD, Cummins TR, Black JA, Waxman SG (November 2007). "From genes to pain: Nav1.7 and human pain disorders". Trends in Neurosciences. 30 (11): 555–63. doi:10.1016/j.tins.2007.08.004. PMID 17950472.
  31. Bagal SK, Chapman ML, Marron BE, Prime R, Storer RI, Swain NA (2014). "Recent progress in sodium channel modulators for pain". Bioorganic & Medicinal Chemistry Letters. 24: 3690–9. doi:10.1016/j.bmcl.2014.06.038. PMID 25060923.
  32. Martz L (2014). "Nav-i-gating antibodies for pain". Science-Business eXchange. 7 (23). doi:10.1038/scibx.2014.662. ISSN 1945-3477.
  33. McMahon S, Koltzenburg M, Tracey I, Turk DC (1 March 2013). Wall & Melzack's Textbook of Pain: Expert Consult - Online. Elsevier Health Sciences. p. 508. ISBN 0-7020-5374-0.
  34. Simpson DM, McArthur JC, Dworkin RH (21 June 2012). Neuropathic Pain: Mechanisms, Diagnosis and Treatment. Oxford University Press. pp. 40–. ISBN 978-0-19-539470-2.
  35. 1 2 3 4 5 6 Minett MS, Pereira V, Sikandar S, Matsuyama A, Lolignier S, Kanellopoulos AH, Mancini F, Iannetti GD, Bogdanov YD, Santana-Varela S, Millet Q, Baskozos G, MacAllister R, Cox JJ, Zhao J, Wood JN (2015). "Endogenous opioids contribute to insensitivity to pain in humans and mice lacking sodium channel Nav1.7". Nature Communications. 6: 8967. doi:10.1038/ncomms9967. PMC 4686868Freely accessible. PMID 26634308.

Further reading

This article is issued from Wikipedia - version of the 10/4/2016. The text is available under the Creative Commons Attribution/Share Alike but additional terms may apply for the media files.