Skip to main content
Erschienen in: Der Schmerz 4/2015

01.08.2015 | CME Zertifizierte Fortbildung

Sensorisch-autonome Neuropathien und Natriumkanal-assoziierte Schmerzerkrankungen

verfasst von: PD Dr. I. Kurth

Erschienen in: Der Schmerz | Ausgabe 4/2015

Einloggen, um Zugang zu erhalten

Zusammenfassung

Ein Verlust von Sensibilität und Schmerzempfinden kann durch einen Entwicklungsdefekt, die Degeneration nozizeptiver Fasern oder eine veränderte neuronale Erregbarkeit verursacht werden. Eine sensorische Neurodegeneration findet sich insbesondere in der heterogenen Gruppe der hereditären sensorisch-autonomen Neuropathien (HSAN). Wesentliches Merkmal der HSAN ist die Neigung der Patienten zu schweren Verletzungen aufgrund der fehlenden Schutzfunktion von Schmerz. Klinisch ähnlich ist die angeborene Schmerzunempfindlichkeit. Sie wird primär durch eine veränderte Erregbarkeit nozizeptiver Fasern aufgrund von Mutationen in spannungsgesteuerten Natriumkanälen verursacht. Im Gegensatz zu HSAN besteht häufig keine Neurodegeneration. Ziel dieses Weiterbildungsbeitrags ist die Darstellung der Krankheitsbilder der HSAN sowie schmerzassoziierter Natriumkanalerkrankungen mit einem Schwerpunkt auf den zugrunde liegenden Pathomechanismen.
Literatur
1.
Zurück zum Zitat Auer-Grumbach M (2013) Hereditary sensory and autonomic neuropathies. Handb Clin Neurol 115:893–906CrossRefPubMed Auer-Grumbach M (2013) Hereditary sensory and autonomic neuropathies. Handb Clin Neurol 115:893–906CrossRefPubMed
2.
Zurück zum Zitat Rotthier A, Baets J, Timmerman V et al (2012) Mechanisms of disease in hereditary sensory and autonomic neuropathies. Nat Rev Neurol 8:73–85CrossRefPubMed Rotthier A, Baets J, Timmerman V et al (2012) Mechanisms of disease in hereditary sensory and autonomic neuropathies. Nat Rev Neurol 8:73–85CrossRefPubMed
4.
Zurück zum Zitat Guelly C, Zhu PP, Leonardis L et al (2011) Targeted high-throughput sequencing identifies mutations in atlastin-1 as a cause of hereditary sensory neuropathy type I. Am J Hum Genet 88:99–105PubMedCentralCrossRefPubMed Guelly C, Zhu PP, Leonardis L et al (2011) Targeted high-throughput sequencing identifies mutations in atlastin-1 as a cause of hereditary sensory neuropathy type I. Am J Hum Genet 88:99–105PubMedCentralCrossRefPubMed
5.
Zurück zum Zitat Kornak U, Mademan I, Schinke M et al (2014) Sensory neuropathy with bone destruction due to a mutation in the membrane-shaping atlastin GTPase 3. Brain 137(Pt 3):683–692CrossRefPubMed Kornak U, Mademan I, Schinke M et al (2014) Sensory neuropathy with bone destruction due to a mutation in the membrane-shaping atlastin GTPase 3. Brain 137(Pt 3):683–692CrossRefPubMed
6.
Zurück zum Zitat Fischer D, Schabhuttl M, Wieland T et al (2014) A novel missense mutation confirms ATL3 as a gene for hereditary sensory neuropathy type 1. Brain 137:e286CrossRefPubMed Fischer D, Schabhuttl M, Wieland T et al (2014) A novel missense mutation confirms ATL3 as a gene for hereditary sensory neuropathy type 1. Brain 137:e286CrossRefPubMed
7.
Zurück zum Zitat Zhao X, Alvarado D, Rainier S et al (2001) Mutations in a newly identified GTPase gene cause autosomal dominant hereditary spastic paraplegia. Nat Genet 29:326–331CrossRefPubMed Zhao X, Alvarado D, Rainier S et al (2001) Mutations in a newly identified GTPase gene cause autosomal dominant hereditary spastic paraplegia. Nat Genet 29:326–331CrossRefPubMed
8.
Zurück zum Zitat Kurth I, Pamminger T, Hennings JC et al (2009) Mutations in FAM134B, encoding a newly identified Golgi protein, cause severe sensory and autonomic neuropathy. Nat Genet 41:1179–1181CrossRefPubMed Kurth I, Pamminger T, Hennings JC et al (2009) Mutations in FAM134B, encoding a newly identified Golgi protein, cause severe sensory and autonomic neuropathy. Nat Genet 41:1179–1181CrossRefPubMed
9.
Zurück zum Zitat Khaminets A, Heinrich T, Mari M et al (2015) Regulation of endoplasmic reticulum turnover by selective autophagy. Nature 522(7556):354–358CrossRefPubMed Khaminets A, Heinrich T, Mari M et al (2015) Regulation of endoplasmic reticulum turnover by selective autophagy. Nature 522(7556):354–358CrossRefPubMed
10.
Zurück zum Zitat Hubner CA, Kurth I (2014) Membrane-shaping disorders: a common pathway in axon degeneration. Brain 137:3109–3121CrossRefPubMed Hubner CA, Kurth I (2014) Membrane-shaping disorders: a common pathway in axon degeneration. Brain 137:3109–3121CrossRefPubMed
11.
Zurück zum Zitat Bejaoui K, Wu C, Scheffler MD et al (2001) SPTLC1 is mutated in hereditary sensory neuropathy, type 1. Nat Genet 27:261–262CrossRefPubMed Bejaoui K, Wu C, Scheffler MD et al (2001) SPTLC1 is mutated in hereditary sensory neuropathy, type 1. Nat Genet 27:261–262CrossRefPubMed
12.
Zurück zum Zitat Dawkins JL, Hulme DJ, Brahmbhatt SB et al (2001) Mutations in SPTLC1, encoding serine palmitoyltransferase, long chain base subunit-1, cause hereditary sensory neuropathy type I. Nat Genet 27:309–312CrossRefPubMed Dawkins JL, Hulme DJ, Brahmbhatt SB et al (2001) Mutations in SPTLC1, encoding serine palmitoyltransferase, long chain base subunit-1, cause hereditary sensory neuropathy type I. Nat Genet 27:309–312CrossRefPubMed
13.
Zurück zum Zitat Rotthier A, Auer-Grumbach M, Janssens K et al (2010) Mutations in the SPTLC2 subunit of serine palmitoyltransferase cause hereditary sensory and autonomic neuropathy type I. Am J Hum Genet 87:513–522PubMedCentralCrossRefPubMed Rotthier A, Auer-Grumbach M, Janssens K et al (2010) Mutations in the SPTLC2 subunit of serine palmitoyltransferase cause hereditary sensory and autonomic neuropathy type I. Am J Hum Genet 87:513–522PubMedCentralCrossRefPubMed
14.
Zurück zum Zitat Garofalo K, Penno A, Schmidt BP et al (2011) Oral L-serine supplementation reduces production of neurotoxic deoxysphingolipids in mice and humans with hereditary sensory autonomic neuropathy type 1. J Clin Invest 121:4735–4745PubMedCentralCrossRefPubMed Garofalo K, Penno A, Schmidt BP et al (2011) Oral L-serine supplementation reduces production of neurotoxic deoxysphingolipids in mice and humans with hereditary sensory autonomic neuropathy type 1. J Clin Invest 121:4735–4745PubMedCentralCrossRefPubMed
17.
Zurück zum Zitat Einarsdottir E, Carlsson A, Minde J et al (2004) A mutation in the nerve growth factor beta gene (NGFB) causes loss of pain perception. Hum Mol Genet 13:799–805CrossRefPubMed Einarsdottir E, Carlsson A, Minde J et al (2004) A mutation in the nerve growth factor beta gene (NGFB) causes loss of pain perception. Hum Mol Genet 13:799–805CrossRefPubMed
18.
Zurück zum Zitat Indo Y, Tsuruta M, Hayashida Y et al (1996) Mutations in the TRKA/NGF receptor gene in patients with congenital insensitivity to pain with anhidrosis. Nat Genet 13:485–488CrossRefPubMed Indo Y, Tsuruta M, Hayashida Y et al (1996) Mutations in the TRKA/NGF receptor gene in patients with congenital insensitivity to pain with anhidrosis. Nat Genet 13:485–488CrossRefPubMed
19.
Zurück zum Zitat Zhang K, Fishel Ben Kenan R, Osakada Y et al (2013) Defective axonal transport of Rab7 GTPase results in dysregulated trophic signaling. J Neurosci 33:7451–7462PubMedCentralCrossRefPubMed Zhang K, Fishel Ben Kenan R, Osakada Y et al (2013) Defective axonal transport of Rab7 GTPase results in dysregulated trophic signaling. J Neurosci 33:7451–7462PubMedCentralCrossRefPubMed
20.
Zurück zum Zitat Riviere JB, Ramalingam S, Lavastre V et al (2011) KIF1A, an axonal transporter of synaptic vesicles, is mutated in hereditary sensory and autonomic neuropathy type 2. Am J Hum Genet 89:219–230PubMedCentralCrossRefPubMed Riviere JB, Ramalingam S, Lavastre V et al (2011) KIF1A, an axonal transporter of synaptic vesicles, is mutated in hereditary sensory and autonomic neuropathy type 2. Am J Hum Genet 89:219–230PubMedCentralCrossRefPubMed
21.
Zurück zum Zitat Lee JR, Srour M, Kim D et al (2015) De novo mutations in the motor domain of KIF1A cause cognitive impairment, spastic paraparesis, axonal neuropathy, and cerebellar atrophy. Hum Mutat 36(1):69–78CrossRefPubMed Lee JR, Srour M, Kim D et al (2015) De novo mutations in the motor domain of KIF1A cause cognitive impairment, spastic paraparesis, axonal neuropathy, and cerebellar atrophy. Hum Mutat 36(1):69–78CrossRefPubMed
22.
Zurück zum Zitat Lafreniere RG, MacDonald ML, Dube MP et al (2004) Identification of a novel gene (HSN2) causing hereditary sensory and autonomic neuropathy type II through the Study of Canadian Genetic Isolates. Am J Hum Genet 74:1064–1073PubMedCentralCrossRefPubMed Lafreniere RG, MacDonald ML, Dube MP et al (2004) Identification of a novel gene (HSN2) causing hereditary sensory and autonomic neuropathy type II through the Study of Canadian Genetic Isolates. Am J Hum Genet 74:1064–1073PubMedCentralCrossRefPubMed
23.
Zurück zum Zitat Shekarabi M, Girard N, Riviere JB et al (2008) Mutations in the nervous system – specific HSN2 exon of WNK1 cause hereditary sensory neuropathy type II. J Clin Invest 118:2496–2505PubMedCentralPubMed Shekarabi M, Girard N, Riviere JB et al (2008) Mutations in the nervous system – specific HSN2 exon of WNK1 cause hereditary sensory neuropathy type II. J Clin Invest 118:2496–2505PubMedCentralPubMed
24.
Zurück zum Zitat Loggia ML, Bushnell MC, Tetreault M et al (2009) Carriers of recessive WNK1/HSN2 mutations for hereditary sensory and autonomic neuropathy type 2 (HSAN2) are more sensitive to thermal stimuli. J Neurosci 29:2162–2166PubMedCentralCrossRefPubMed Loggia ML, Bushnell MC, Tetreault M et al (2009) Carriers of recessive WNK1/HSN2 mutations for hereditary sensory and autonomic neuropathy type 2 (HSAN2) are more sensitive to thermal stimuli. J Neurosci 29:2162–2166PubMedCentralCrossRefPubMed
26.
Zurück zum Zitat Slaugenhaupt SA, Blumenfeld A, Gill SP et al (2001) Tissue-specific expression of a splicing mutation in the IKBKAP gene causes familial dysautonomia. Am J Hum Genet 68:598–605PubMedCentralCrossRefPubMed Slaugenhaupt SA, Blumenfeld A, Gill SP et al (2001) Tissue-specific expression of a splicing mutation in the IKBKAP gene causes familial dysautonomia. Am J Hum Genet 68:598–605PubMedCentralCrossRefPubMed
27.
Zurück zum Zitat Edvardson S, Cinnamon Y, Jalas C et al (2012) Hereditary sensory autonomic neuropathy caused by a mutation in dystonin. Ann Neurol 71:569–572CrossRefPubMed Edvardson S, Cinnamon Y, Jalas C et al (2012) Hereditary sensory autonomic neuropathy caused by a mutation in dystonin. Ann Neurol 71:569–572CrossRefPubMed
28.
Zurück zum Zitat Klein CJ, Botuyan MV, Wu Y et al (2011) Mutations in DNMT1 cause hereditary sensory neuropathy with dementia and hearing loss. Nat Genet 43:595–600PubMedCentralCrossRefPubMed Klein CJ, Botuyan MV, Wu Y et al (2011) Mutations in DNMT1 cause hereditary sensory neuropathy with dementia and hearing loss. Nat Genet 43:595–600PubMedCentralCrossRefPubMed
29.
Zurück zum Zitat Winkelmann J, Lin L, Schormair B et al (2012) Mutations in DNMT1 cause autosomal dominant cerebellar ataxia, deafness and narcolepsy. Hum Mol Genet 21:2205–2210PubMedCentralCrossRefPubMed Winkelmann J, Lin L, Schormair B et al (2012) Mutations in DNMT1 cause autosomal dominant cerebellar ataxia, deafness and narcolepsy. Hum Mol Genet 21:2205–2210PubMedCentralCrossRefPubMed
30.
Zurück zum Zitat Chen YC, Auer-Grumbach M, Matsukawa S et al (2015) Transcriptional regulator PRDM12 is essential for human pain perception. Nat Genet 47:803–808CrossRefPubMed Chen YC, Auer-Grumbach M, Matsukawa S et al (2015) Transcriptional regulator PRDM12 is essential for human pain perception. Nat Genet 47:803–808CrossRefPubMed
31.
Zurück zum Zitat Cox JJ, Reimann F, Nicholas AK et al (2006) An SCN9A channelopathy causes congenital inability to experience pain. Nature 444:894–898CrossRefPubMed Cox JJ, Reimann F, Nicholas AK et al (2006) An SCN9A channelopathy causes congenital inability to experience pain. Nature 444:894–898CrossRefPubMed
32.
Zurück zum Zitat Goldberg YP, MacFarlane J, MacDonald ML et al (2007) Loss-of-function mutations in the Nav1.7 gene underlie congenital indifference to pain in multiple human populations. Clin Genet 71:311–319CrossRefPubMed Goldberg YP, MacFarlane J, MacDonald ML et al (2007) Loss-of-function mutations in the Nav1.7 gene underlie congenital indifference to pain in multiple human populations. Clin Genet 71:311–319CrossRefPubMed
34.
Zurück zum Zitat Goldberg YP, Pimstone SN, Namdari R et al (2012) Human Mendelian pain disorders: a key to discovery and validation of novel analgesics. Clin Genet 82:367–373CrossRefPubMed Goldberg YP, Pimstone SN, Namdari R et al (2012) Human Mendelian pain disorders: a key to discovery and validation of novel analgesics. Clin Genet 82:367–373CrossRefPubMed
35.
Zurück zum Zitat Goldberg YP, Price N, Namdari R et al (2012) Treatment of Na(v)1.7-mediated pain in inherited erythromelalgia using a novel sodium channel blocker. Pain 153:80–85CrossRefPubMed Goldberg YP, Price N, Namdari R et al (2012) Treatment of Na(v)1.7-mediated pain in inherited erythromelalgia using a novel sodium channel blocker. Pain 153:80–85CrossRefPubMed
36.
Zurück zum Zitat Yuan J, Matsuura E, Higuchi Y et al (2013) Hereditary sensory and autonomic neuropathy type IID caused by an SCN9A mutation. Neurology 80:1641–1649CrossRefPubMed Yuan J, Matsuura E, Higuchi Y et al (2013) Hereditary sensory and autonomic neuropathy type IID caused by an SCN9A mutation. Neurology 80:1641–1649CrossRefPubMed
37.
Zurück zum Zitat Leipold E, Liebmann L, Korenke GC et al (2013) A de novo gain-of-function mutation in SCN11A causes loss of pain perception. Nat Genet 45:1399–1404CrossRefPubMed Leipold E, Liebmann L, Korenke GC et al (2013) A de novo gain-of-function mutation in SCN11A causes loss of pain perception. Nat Genet 45:1399–1404CrossRefPubMed
38.
Zurück zum Zitat Woods CG, Babiker MO, Horrocks I et al (2014) The phenotype of congenital insensitivity to pain due to the Na1.9 variant p.L811P. Eur J Hum Genet 23:561–563CrossRefPubMed Woods CG, Babiker MO, Horrocks I et al (2014) The phenotype of congenital insensitivity to pain due to the Na1.9 variant p.L811P. Eur J Hum Genet 23:561–563CrossRefPubMed
40.
Zurück zum Zitat Fertleman CR, Baker MD, Parker KA et al (2006) SCN9A mutations in paroxysmal extreme pain disorder: allelic variants underlie distinct channel defects and phenotypes. Neuron 52:767–774CrossRefPubMed Fertleman CR, Baker MD, Parker KA et al (2006) SCN9A mutations in paroxysmal extreme pain disorder: allelic variants underlie distinct channel defects and phenotypes. Neuron 52:767–774CrossRefPubMed
41.
Zurück zum Zitat Yang Y, Wang Y, Li S et al (2004) Mutations in SCN9A, encoding a sodium channel alpha subunit, in patients with primary erythermalgia. J Med Genet 41:171–174PubMedCentralCrossRefPubMed Yang Y, Wang Y, Li S et al (2004) Mutations in SCN9A, encoding a sodium channel alpha subunit, in patients with primary erythermalgia. J Med Genet 41:171–174PubMedCentralCrossRefPubMed
42.
Zurück zum Zitat Dib-Hajj SD, Cummins TR, Black JA et al (2010) Sodium channels in normal and pathological pain. Annu Rev Neurosci 33:325–347CrossRefPubMed Dib-Hajj SD, Cummins TR, Black JA et al (2010) Sodium channels in normal and pathological pain. Annu Rev Neurosci 33:325–347CrossRefPubMed
43.
Zurück zum Zitat Faber CG, Hoeijmakers JG, Ahn HS et al (2012) Gain of function NaV1.7 mutations in idiopathic small fiber neuropathy. Ann Neurol 71:26–39CrossRefPubMed Faber CG, Hoeijmakers JG, Ahn HS et al (2012) Gain of function NaV1.7 mutations in idiopathic small fiber neuropathy. Ann Neurol 71:26–39CrossRefPubMed
44.
Zurück zum Zitat Faber CG, Lauria G, Merkies IS et al (2012) Gain-of-function Nav1.8 mutations in painful neuropathy. Proc Natl Acad Sci U S A 109:19444–19449PubMedCentralCrossRefPubMed Faber CG, Lauria G, Merkies IS et al (2012) Gain-of-function Nav1.8 mutations in painful neuropathy. Proc Natl Acad Sci U S A 109:19444–19449PubMedCentralCrossRefPubMed
45.
Zurück zum Zitat Huang J, Han C, Estacion M et al (2014) Gain-of-function mutations in sodium channel Na(v)1.9 in painful neuropathy. Brain 137:1627–1642CrossRefPubMed Huang J, Han C, Estacion M et al (2014) Gain-of-function mutations in sodium channel Na(v)1.9 in painful neuropathy. Brain 137:1627–1642CrossRefPubMed
46.
Zurück zum Zitat Kremeyer B, Lopera F, Cox JJ et al (2010) A gain-of-function mutation in TRPA1 causes familial episodic pain syndrome. Neuron 66:671–680CrossRefPubMed Kremeyer B, Lopera F, Cox JJ et al (2010) A gain-of-function mutation in TRPA1 causes familial episodic pain syndrome. Neuron 66:671–680CrossRefPubMed
Metadaten
Titel
Sensorisch-autonome Neuropathien und Natriumkanal-assoziierte Schmerzerkrankungen
verfasst von
PD Dr. I. Kurth
Publikationsdatum
01.08.2015
Verlag
Springer Berlin Heidelberg
Erschienen in
Der Schmerz / Ausgabe 4/2015
Print ISSN: 0932-433X
Elektronische ISSN: 1432-2129
DOI
https://doi.org/10.1007/s00482-015-0024-2

Weitere Artikel der Ausgabe 4/2015

Der Schmerz 4/2015 Zur Ausgabe

Mitteilungen der SGSS

Mitteilungen der SGSS

Update AINS

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.