Skip to main content
Top
Published in: Orphanet Journal of Rare Diseases 1/2018

Open Access 01-12-2018 | Review

GNE myopathy: from clinics and genetics to pathology and research strategies

Authors: Oksana Pogoryelova, José Andrés González Coraspe, Nikoletta Nikolenko, Hanns Lochmüller, Andreas Roos

Published in: Orphanet Journal of Rare Diseases | Issue 1/2018

Login to get access

Abstract

GNE myopathy is an ultra-rare autosomal recessive disease, which starts as a distal muscle weakness and ultimately leads to a wheelchair bound state. Molecular research and animal modelling significantly moved forward understanding of GNE myopathy mechanisms and suggested therapeutic interventions to alleviate the symptoms. Multiple therapeutic attempts are being made to supplement sialic acid depleted in GNE myopathy muscle cells. Translational research field provided valuable knowledge through natural history studies, patient registries and clinical trial, which significantly contributed to bringing forward an era of GNE myopathy treatment. In this review, we are summarising current GNE myopathy, scientific trends and open questions, which would be of significant interest for a wide neuromuscular diseases community.
Literature
1.
go back to reference Nonaka I, Sunohara N, Ishiura S, Satoyoshi E. Familial distal myopathy with rimmed vacuole and lamellar (myeloid) body formation. J Neurol Sci. 1981;51:141–55.CrossRefPubMed Nonaka I, Sunohara N, Ishiura S, Satoyoshi E. Familial distal myopathy with rimmed vacuole and lamellar (myeloid) body formation. J Neurol Sci. 1981;51:141–55.CrossRefPubMed
2.
go back to reference Argov Z, Yarom R. "rimmed vacuole myopathy" sparing the quadriceps. A unique disorder in Iranian Jews. J Neurol Sci. 1984;64:33–43.CrossRefPubMed Argov Z, Yarom R. "rimmed vacuole myopathy" sparing the quadriceps. A unique disorder in Iranian Jews. J Neurol Sci. 1984;64:33–43.CrossRefPubMed
3.
4.
go back to reference Mitrani-Rosenbaum S, Argov Z, Blumenfeld A, Seidman CE, Seidman JG. Hereditary inclusion body myopathy maps to chromosome 9p1-q1. Hum Mol Genet. 1996;5:159–63.CrossRefPubMed Mitrani-Rosenbaum S, Argov Z, Blumenfeld A, Seidman CE, Seidman JG. Hereditary inclusion body myopathy maps to chromosome 9p1-q1. Hum Mol Genet. 1996;5:159–63.CrossRefPubMed
5.
go back to reference Eisenberg I, Avidan N, Potikha T, Hochner H, Chen M, Olender T, Barash M, Shemesh M, Sadeh M, Grabov-Nardini G, Shmilevich I, Friedmann A, Karpati G, Bradley WG, Baumbach L, Lancet D, Asher EB, Beckmann JS, Argov Z, Mitrani-Rosenbaum S. The UDP-N-acetylglucosamine 2-epimerase/N-acetylmannosamine kinase gene is mutated in recessive hereditary inclusion body myopathy. Nat Genet. 2001;29:83–7.CrossRefPubMed Eisenberg I, Avidan N, Potikha T, Hochner H, Chen M, Olender T, Barash M, Shemesh M, Sadeh M, Grabov-Nardini G, Shmilevich I, Friedmann A, Karpati G, Bradley WG, Baumbach L, Lancet D, Asher EB, Beckmann JS, Argov Z, Mitrani-Rosenbaum S. The UDP-N-acetylglucosamine 2-epimerase/N-acetylmannosamine kinase gene is mutated in recessive hereditary inclusion body myopathy. Nat Genet. 2001;29:83–7.CrossRefPubMed
6.
go back to reference Nishino I, Noguchi S, Murayama K, Driss A, Sugie K, Oya Y, Nagata T, Chida K, Takahashi T, Takusa Y, Ohi T, Nishimiya J, Sunohara N, Ciafaloni E, Kawai M, Aoki M, Nonaka I. Distal myopathy with rimmed vacuoles is allelic to hereditary inclusion body myopathy. Neurology. 2002;59:1689–93.CrossRefPubMed Nishino I, Noguchi S, Murayama K, Driss A, Sugie K, Oya Y, Nagata T, Chida K, Takahashi T, Takusa Y, Ohi T, Nishimiya J, Sunohara N, Ciafaloni E, Kawai M, Aoki M, Nonaka I. Distal myopathy with rimmed vacuoles is allelic to hereditary inclusion body myopathy. Neurology. 2002;59:1689–93.CrossRefPubMed
7.
go back to reference Huizing M, Carrillo-Carrasco N, Malicdan MC, Noguchi S, Gahl WA, Mitrani-Rosenbaum S, Argov Z, Nishino I. GNE myopathy: new name and new mutation nomenclature. Neuromuscul Disord. 2014;24:387–9.CrossRefPubMedPubMedCentral Huizing M, Carrillo-Carrasco N, Malicdan MC, Noguchi S, Gahl WA, Mitrani-Rosenbaum S, Argov Z, Nishino I. GNE myopathy: new name and new mutation nomenclature. Neuromuscul Disord. 2014;24:387–9.CrossRefPubMedPubMedCentral
8.
go back to reference Nishino I, Carrillo-Carrasco N, Argov Z. GNE myopathy: current update and future therapy. J Neurol Neurosurg Psychiatry. 2015;86:385–92.CrossRefPubMed Nishino I, Carrillo-Carrasco N, Argov Z. GNE myopathy: current update and future therapy. J Neurol Neurosurg Psychiatry. 2015;86:385–92.CrossRefPubMed
9.
go back to reference Preethish-Kumar V, et al. Beevor's sign: a potential clinical marker for GNE myopathy. Eur J Neurol. 2016;23:e46–8.CrossRefPubMed Preethish-Kumar V, et al. Beevor's sign: a potential clinical marker for GNE myopathy. Eur J Neurol. 2016;23:e46–8.CrossRefPubMed
10.
go back to reference Chamova T, et al. GNE myopathy in Roma patients homozygous for the p.I618T founder mutation. Neuromuscul Disord. 2015;25:713–8.CrossRefPubMed Chamova T, et al. GNE myopathy in Roma patients homozygous for the p.I618T founder mutation. Neuromuscul Disord. 2015;25:713–8.CrossRefPubMed
12.
go back to reference Slota C, et al. Patient reported outcomes in GNE myopathy: incorporating a valid assessment of physical function in a rare disease. Disabil Rehabil. 2018;40:1206–13. Slota C, et al. Patient reported outcomes in GNE myopathy: incorporating a valid assessment of physical function in a rare disease. Disabil Rehabil. 2018;40:1206–13.
13.
go back to reference Park YE, et al. Limb-girdle phenotype is frequent in patients with myopathy associated with GNE mutations. J Neurol Sci. 2012;321:77–81.CrossRefPubMed Park YE, et al. Limb-girdle phenotype is frequent in patients with myopathy associated with GNE mutations. J Neurol Sci. 2012;321:77–81.CrossRefPubMed
14.
go back to reference Chaouch A, et al. Two recurrent mutations are associated with GNE myopathy in the north of Britain. J Neurol Neurosurg Psychiatry. 2014;85:1359–65.CrossRefPubMed Chaouch A, et al. Two recurrent mutations are associated with GNE myopathy in the north of Britain. J Neurol Neurosurg Psychiatry. 2014;85:1359–65.CrossRefPubMed
16.
go back to reference Diniz G, et al. GNE myopathy in Turkish sisters with a novel homozygous mutation. Case Rep Neurol Med. 2016; 8647645 Diniz G, et al. GNE myopathy in Turkish sisters with a novel homozygous mutation. Case Rep Neurol Med. 2016; 8647645
17.
go back to reference Das B, et al. Hereditary inclusion body myopathy: a myopathy with unique topography of weakness, yet frequently misdiagnosed: case series and review of literature. Ann Indian Acad Neurol. 2016;19:119–22.CrossRefPubMedPubMedCentral Das B, et al. Hereditary inclusion body myopathy: a myopathy with unique topography of weakness, yet frequently misdiagnosed: case series and review of literature. Ann Indian Acad Neurol. 2016;19:119–22.CrossRefPubMedPubMedCentral
18.
go back to reference Krause S, et al. A novel homozygous missense mutation in the GNE gene of a patient with quadriceps-sparing hereditary inclusion body myopathy associated with muscle inflammation. Case Rep Neurol Med. 2016;2016:8647645. Krause S, et al. A novel homozygous missense mutation in the GNE gene of a patient with quadriceps-sparing hereditary inclusion body myopathy associated with muscle inflammation. Case Rep Neurol Med. 2016;2016:8647645.
19.
go back to reference Choi YA, Park SH, Yi Y, Kim K. Novel mutation of the GNE gene presenting atypical mild clinical feature: a Korean case report. Ann Rehabil Medicine. 2015;39:494–7.CrossRef Choi YA, Park SH, Yi Y, Kim K. Novel mutation of the GNE gene presenting atypical mild clinical feature: a Korean case report. Ann Rehabil Medicine. 2015;39:494–7.CrossRef
20.
go back to reference Haghighi A, et al. Genetics of GNE myopathy in the non-Jewish Persian population. Eur J Hum Genet. 2016;24:243–51.CrossRefPubMed Haghighi A, et al. Genetics of GNE myopathy in the non-Jewish Persian population. Eur J Hum Genet. 2016;24:243–51.CrossRefPubMed
21.
go back to reference Tarnopolsky MA, Hatcher E, Shupak R. Genetic myopathies initially diagnosed and treated as inflammatory myopathy. Can J Neurol Sci. 2016;43(3):381–4.CrossRefPubMed Tarnopolsky MA, Hatcher E, Shupak R. Genetic myopathies initially diagnosed and treated as inflammatory myopathy. Can J Neurol Sci. 2016;43(3):381–4.CrossRefPubMed
22.
go back to reference Mori-Yoshimura M, Oya Y, Yajima H, Yonemoto N, Kobayashi Y, Hayashi YK, Noguchi S, Nishino I, Murata M. GNE myopathy: a prospective natural history study of disease progression. Neuromuscul Disord. 2014;24(5):380–6.CrossRefPubMed Mori-Yoshimura M, Oya Y, Yajima H, Yonemoto N, Kobayashi Y, Hayashi YK, Noguchi S, Nishino I, Murata M. GNE myopathy: a prospective natural history study of disease progression. Neuromuscul Disord. 2014;24(5):380–6.CrossRefPubMed
23.
go back to reference Izumi R, et al. GNE myopathy associated with congenital thrombocytopenia: a report of two siblings. Neuromuscul Disord. 2014;24(12):1068–72.CrossRefPubMed Izumi R, et al. GNE myopathy associated with congenital thrombocytopenia: a report of two siblings. Neuromuscul Disord. 2014;24(12):1068–72.CrossRefPubMed
24.
go back to reference Celeste FV, Vilboux T, Ciccone C, de Dios JK, Malicdan MC, Leoyklang P, et al. Mutation update for GNE gene variants associated with GNE myopathy. Hum Mutat. 2014;35(8):915–26.CrossRefPubMedPubMedCentral Celeste FV, Vilboux T, Ciccone C, de Dios JK, Malicdan MC, Leoyklang P, et al. Mutation update for GNE gene variants associated with GNE myopathy. Hum Mutat. 2014;35(8):915–26.CrossRefPubMedPubMedCentral
25.
go back to reference Argov Z, Eisenberg I, Grabov-Nardini G, Sadeh M, Wirguin I, Soffer D, et al. Hereditary inclusion body myopathy: the middle eastern genetic cluster. Neurology. 2003;60(9):1519–23.CrossRefPubMed Argov Z, Eisenberg I, Grabov-Nardini G, Sadeh M, Wirguin I, Soffer D, et al. Hereditary inclusion body myopathy: the middle eastern genetic cluster. Neurology. 2003;60(9):1519–23.CrossRefPubMed
26.
go back to reference Zhao J, Wang Z, Hong D, Lv H, Zhang W, Chen J, et al. Mutational spectrum and clinical features in 35 unrelated mainland Chinese patients with GNE myopathy. J Neurol Sci. 2015;354(1-2):21–6.CrossRefPubMed Zhao J, Wang Z, Hong D, Lv H, Zhang W, Chen J, et al. Mutational spectrum and clinical features in 35 unrelated mainland Chinese patients with GNE myopathy. J Neurol Sci. 2015;354(1-2):21–6.CrossRefPubMed
27.
go back to reference Cho A, Hayashi YK, Monma K, Oya Y, Noguchi S, Nonaka I, et al. Mutation profile of the GNE gene in Japanese patients with distal myopathy with rimmed vacuoles (GNE myopathy). J Neurol Neurosurg Psychiatry. 2014;85(8):914–7.CrossRefPubMed Cho A, Hayashi YK, Monma K, Oya Y, Noguchi S, Nonaka I, et al. Mutation profile of the GNE gene in Japanese patients with distal myopathy with rimmed vacuoles (GNE myopathy). J Neurol Neurosurg Psychiatry. 2014;85(8):914–7.CrossRefPubMed
28.
go back to reference Bohler HC Jr, Zoeller RT, King JC, Rubin BS, Weber R, Merriam GR. Corticotropin releasing hormone mRNA is elevated on the afternoon of proestrus in the parvocellular paraventricular nuclei of the female rat. Brain Res Mol Brain Res. 1990;8(3):259–62.CrossRefPubMed Bohler HC Jr, Zoeller RT, King JC, Rubin BS, Weber R, Merriam GR. Corticotropin releasing hormone mRNA is elevated on the afternoon of proestrus in the parvocellular paraventricular nuclei of the female rat. Brain Res Mol Brain Res. 1990;8(3):259–62.CrossRefPubMed
29.
go back to reference Zhu W, et al. Missing genetic variations in GNE myopathy: rearrangement hotspots encompassing 5'UTR and founder allele. J Hum Genet. 2017;62(2):159–66.CrossRefPubMed Zhu W, et al. Missing genetic variations in GNE myopathy: rearrangement hotspots encompassing 5'UTR and founder allele. J Hum Genet. 2017;62(2):159–66.CrossRefPubMed
30.
go back to reference Garland J, et al. Identification of an Alu element-mediated deletion in the promoter region of GNE in siblings with GNE myopathy. Mol Genet Genomic Med. 2017;5(4):410–7.CrossRefPubMedPubMedCentral Garland J, et al. Identification of an Alu element-mediated deletion in the promoter region of GNE in siblings with GNE myopathy. Mol Genet Genomic Med. 2017;5(4):410–7.CrossRefPubMedPubMedCentral
31.
go back to reference Hillery CA, Smyth SS, Parise LV. Phosphorylation of human platelet glycoprotein IIIa (GPIIIa). Dissociation from fibrinogen receptor activation and phosphorylation of GPIIIa in vitro. J Biol Chem. 1991;266(22):14663–9.PubMed Hillery CA, Smyth SS, Parise LV. Phosphorylation of human platelet glycoprotein IIIa (GPIIIa). Dissociation from fibrinogen receptor activation and phosphorylation of GPIIIa in vitro. J Biol Chem. 1991;266(22):14663–9.PubMed
32.
go back to reference Noguchi S, Keira Y, Murayama K, Ogawa M, Fujita M, Kawahara G, et al. Reduction of UDP-N-acetylglucosamine 2-epimerase/N-acetylmannosamine kinase activity and sialylation in distal myopathy with rimmed vacuoles. J Biol Chem. 2004;279(12):11402–7.CrossRefPubMed Noguchi S, Keira Y, Murayama K, Ogawa M, Fujita M, Kawahara G, et al. Reduction of UDP-N-acetylglucosamine 2-epimerase/N-acetylmannosamine kinase activity and sialylation in distal myopathy with rimmed vacuoles. J Biol Chem. 2004;279(12):11402–7.CrossRefPubMed
33.
go back to reference Penner J, et al. Influence of UDP-GlcNAc 2-epimerase/ManNAc kinase mutant proteins on hereditary inclusion body myopathy. Biochemistry. 2006;45:2968–77.CrossRefPubMed Penner J, et al. Influence of UDP-GlcNAc 2-epimerase/ManNAc kinase mutant proteins on hereditary inclusion body myopathy. Biochemistry. 2006;45:2968–77.CrossRefPubMed
34.
go back to reference Malicdan MC, Noguchi S, Nishino I. A preclinical trial of sialic acid metabolites on distal myopathy with rimmed vacuoles/hereditary inclusion body myopathy, a sugar-deficient myopathy: a review. Ther Adv Neurol Disord. 2010;3(2):127–35.CrossRefPubMedPubMedCentral Malicdan MC, Noguchi S, Nishino I. A preclinical trial of sialic acid metabolites on distal myopathy with rimmed vacuoles/hereditary inclusion body myopathy, a sugar-deficient myopathy: a review. Ther Adv Neurol Disord. 2010;3(2):127–35.CrossRefPubMedPubMedCentral
35.
go back to reference Ishihara S, Tomimitsu H, Fujigasaki H, Saito F, Mizusawa H. UDP-N-acetylglucosamine 2-epimerase/N-acetylmannosamine kinase in nuclei and rimmed vacuoles of muscle fibers in DMRV (distal myopathy with rimmed vacuoles). J Med Dent Sci. 2008;55(1):181–7.PubMed Ishihara S, Tomimitsu H, Fujigasaki H, Saito F, Mizusawa H. UDP-N-acetylglucosamine 2-epimerase/N-acetylmannosamine kinase in nuclei and rimmed vacuoles of muscle fibers in DMRV (distal myopathy with rimmed vacuoles). J Med Dent Sci. 2008;55(1):181–7.PubMed
36.
go back to reference Krause S, Aleo A, Hinderlich S, Merlini L, Tournev I, Walter MC, Argov Z, Mitrani-Rosenbaum S, Lochmüller H. GNE protein expression and subcellular distribution are unaltered in HIBM. Neurology. 2007;69(7):655–9.CrossRefPubMed Krause S, Aleo A, Hinderlich S, Merlini L, Tournev I, Walter MC, Argov Z, Mitrani-Rosenbaum S, Lochmüller H. GNE protein expression and subcellular distribution are unaltered in HIBM. Neurology. 2007;69(7):655–9.CrossRefPubMed
37.
go back to reference Saito F, Tomimitsu H, Arai K, Nakai S, Kanda T, Shimizu T, Mizusawa H, Matsumura K. A Japanese patient with distal myopathy with rimmed vacuoles: missense mutations in the epimerase domain of the UDP-N-acetylglucosamine 2-epimerase/N-acetylmannosamine kinase (GNE) gene accompanied by hyposialylation of skeletal muscle glycoproteins. Neuromuscul Disord. 2004;14(2):158–61.CrossRefPubMed Saito F, Tomimitsu H, Arai K, Nakai S, Kanda T, Shimizu T, Mizusawa H, Matsumura K. A Japanese patient with distal myopathy with rimmed vacuoles: missense mutations in the epimerase domain of the UDP-N-acetylglucosamine 2-epimerase/N-acetylmannosamine kinase (GNE) gene accompanied by hyposialylation of skeletal muscle glycoproteins. Neuromuscul Disord. 2004;14(2):158–61.CrossRefPubMed
38.
go back to reference Voermans NC, Guillard M, Doedée R, Lammens M, Huizing M, Padberg GW, Wevers RA, van Engelen BG, Lefeber DJ. Clinical features, lectin staining, and a novel GNE frameshift mutation in hereditary inclusion body myopathy. Clin Neuropathol. 2010;29(2):71–7.PubMedPubMedCentral Voermans NC, Guillard M, Doedée R, Lammens M, Huizing M, Padberg GW, Wevers RA, van Engelen BG, Lefeber DJ. Clinical features, lectin staining, and a novel GNE frameshift mutation in hereditary inclusion body myopathy. Clin Neuropathol. 2010;29(2):71–7.PubMedPubMedCentral
39.
go back to reference Leoyklang P, Malicdan MC, Yardeni T, Celeste F, Ciccone C, Li X, Jiang R, Gahl WA, Carrillo-Carrasco N, He M, Huizing M. Sialylation of Thomsen-Friedenreich antigen is a noninvasive blood-based biomarker for GNE myopathy. Biomark Med. 2014;8(5):641–52.CrossRefPubMedPubMedCentral Leoyklang P, Malicdan MC, Yardeni T, Celeste F, Ciccone C, Li X, Jiang R, Gahl WA, Carrillo-Carrasco N, He M, Huizing M. Sialylation of Thomsen-Friedenreich antigen is a noninvasive blood-based biomarker for GNE myopathy. Biomark Med. 2014;8(5):641–52.CrossRefPubMedPubMedCentral
40.
go back to reference Huizing M, Rakocevic G, Sparks SE, Mamali I, Shatunov A, Goldfarb L, Krasnewich D, Gahl WA, Dalakas MC. Hypoglycosylation of alpha-dystroglycan in patients with hereditary IBM due to GNE mutations. Mol Genet Metab. 2004;81(3):196–202.CrossRefPubMed Huizing M, Rakocevic G, Sparks SE, Mamali I, Shatunov A, Goldfarb L, Krasnewich D, Gahl WA, Dalakas MC. Hypoglycosylation of alpha-dystroglycan in patients with hereditary IBM due to GNE mutations. Mol Genet Metab. 2004;81(3):196–202.CrossRefPubMed
41.
go back to reference Broccolini A, Gliubizzi C, Pavoni E, Gidaro T, Morosetti R, Sciandra F, Giardina B, Tonali P, Ricci E, Brancaccio A, Mirabella M. Alpha-Dystroglycan does not play a major pathogenic role in autosomal recessive hereditary inclusion-body myopathy. Neuromuscul Disord. 2005;15(2):177–84.CrossRefPubMed Broccolini A, Gliubizzi C, Pavoni E, Gidaro T, Morosetti R, Sciandra F, Giardina B, Tonali P, Ricci E, Brancaccio A, Mirabella M. Alpha-Dystroglycan does not play a major pathogenic role in autosomal recessive hereditary inclusion-body myopathy. Neuromuscul Disord. 2005;15(2):177–84.CrossRefPubMed
42.
go back to reference Sela I, Milman Krentsis I, Shlomai Z, Sadeh M, Dabby R, Argov Z, Ben-Bassat H, Mitrani-Rosenbaum S. The proteomic profile of hereditary inclusion body myopathy. PLoS One. 2011;6(1):e16334.CrossRefPubMedPubMedCentral Sela I, Milman Krentsis I, Shlomai Z, Sadeh M, Dabby R, Argov Z, Ben-Bassat H, Mitrani-Rosenbaum S. The proteomic profile of hereditary inclusion body myopathy. PLoS One. 2011;6(1):e16334.CrossRefPubMedPubMedCentral
43.
go back to reference Eisenberg I, Novershtern N, Itzhaki Z, Becker-Cohen M, Sadeh M, Willems PH, Friedman N, Koopman WJ, Mitrani-Rosenbaum S. Mitochondrial processes are impaired in hereditary inclusion body myopathy. Hum Mol Genet. 2008;17(23):3663–74.CrossRefPubMed Eisenberg I, Novershtern N, Itzhaki Z, Becker-Cohen M, Sadeh M, Willems PH, Friedman N, Koopman WJ, Mitrani-Rosenbaum S. Mitochondrial processes are impaired in hereditary inclusion body myopathy. Hum Mol Genet. 2008;17(23):3663–74.CrossRefPubMed
44.
go back to reference Nogalska A, D'Agostino C, Engel WK, Cacciottolo M, Asada S, Mori K, Askanas V. Activation of the unfolded protein response in sporadic inclusion-body myositis but not in hereditary GNE inclusion-body myopathy. J Neuropathol Exp Neurol. 2015;74(6):538–46.CrossRefPubMedPubMedCentral Nogalska A, D'Agostino C, Engel WK, Cacciottolo M, Asada S, Mori K, Askanas V. Activation of the unfolded protein response in sporadic inclusion-body myositis but not in hereditary GNE inclusion-body myopathy. J Neuropathol Exp Neurol. 2015;74(6):538–46.CrossRefPubMedPubMedCentral
45.
go back to reference Li H, Chen Q, Liu F, Zhang X, Li W, Liu S, Zhao Y, Gong Y, Yan C. Unfolded protein response and activated degradative pathways regulation in GNE myopathy. PLoS One. 2013;8(3):e58116.CrossRefPubMedPubMedCentral Li H, Chen Q, Liu F, Zhang X, Li W, Liu S, Zhao Y, Gong Y, Yan C. Unfolded protein response and activated degradative pathways regulation in GNE myopathy. PLoS One. 2013;8(3):e58116.CrossRefPubMedPubMedCentral
46.
go back to reference Fischer C, Kleinschnitz K, Wrede A, Muth I, Kruse N, Nishino I, et al. Cell stress molecules in the skeletal muscle of GNE myopathy. BMC Neurol. 2013;12:13–24. Fischer C, Kleinschnitz K, Wrede A, Muth I, Kruse N, Nishino I, et al. Cell stress molecules in the skeletal muscle of GNE myopathy. BMC Neurol. 2013;12:13–24.
47.
go back to reference Nakamura K, Tsukamoto Y, Hijiya N, Higuchi Y, Yano S, Yokoyama S, Kumamoto T, Moriyama M. Induction of GNE in myofibers after muscle injury. Pathobiology. 2010;77(4):191–9.CrossRefPubMed Nakamura K, Tsukamoto Y, Hijiya N, Higuchi Y, Yano S, Yokoyama S, Kumamoto T, Moriyama M. Induction of GNE in myofibers after muscle injury. Pathobiology. 2010;77(4):191–9.CrossRefPubMed
48.
go back to reference Salama I, Hinderlich S, Shlomai Z, Eisenberg I, Krause S, Yarema K, Argov Z, Lochmuller H, Reutter W, Dabby R, Sadeh M, Ben-Bassat H, Mitrani-Rosenbaum S. No overall hyposialylation in hereditary inclusion body myopathy myoblasts carrying the homozygous M712T GNE mutation. Biochem Biophys Res Commun. 2005;328(1):221–6.CrossRefPubMed Salama I, Hinderlich S, Shlomai Z, Eisenberg I, Krause S, Yarema K, Argov Z, Lochmuller H, Reutter W, Dabby R, Sadeh M, Ben-Bassat H, Mitrani-Rosenbaum S. No overall hyposialylation in hereditary inclusion body myopathy myoblasts carrying the homozygous M712T GNE mutation. Biochem Biophys Res Commun. 2005;328(1):221–6.CrossRefPubMed
49.
go back to reference Bosch-Morató M, Iriondo C, Guivernau B, Valls-Comamala V, Vidal N, Olivé M, Querfurth H, Muñoz FJ. Increased amyloid β-peptide uptake in skeletal muscle is induced by hyposialylation and may account for apoptosis in GNE myopathy. Oncotarget. 2016;7(12):13354–71.CrossRefPubMedPubMedCentral Bosch-Morató M, Iriondo C, Guivernau B, Valls-Comamala V, Vidal N, Olivé M, Querfurth H, Muñoz FJ. Increased amyloid β-peptide uptake in skeletal muscle is induced by hyposialylation and may account for apoptosis in GNE myopathy. Oncotarget. 2016;7(12):13354–71.CrossRefPubMedPubMedCentral
50.
go back to reference Singh R, Arya R. GNE myopathy and cell apoptosis: a comparative mutation analysis. Mol Neurobiol. 2016;53(5):3088–101.CrossRefPubMed Singh R, Arya R. GNE myopathy and cell apoptosis: a comparative mutation analysis. Mol Neurobiol. 2016;53(5):3088–101.CrossRefPubMed
51.
go back to reference Grover S, Arya R. Role of UDP-N-acetylglucosamine2-epimerase/N-acetylmannosamine kinase (GNE) in β1-integrin-mediated cell adhesion. Mol Neurobiol. 2014;50(2):257–73.CrossRefPubMed Grover S, Arya R. Role of UDP-N-acetylglucosamine2-epimerase/N-acetylmannosamine kinase (GNE) in β1-integrin-mediated cell adhesion. Mol Neurobiol. 2014;50(2):257–73.CrossRefPubMed
52.
go back to reference Patzel KA, Yardeni T, Le Poëc-Celic E, Leoyklang P, Dorward H, Alonzi DS, Kukushkin NV, Xu B, Zhang Y, Sollogoub M, Blériot Y, Gahl WA, Huizing M, Butters TD. Non-specific accumulation of glycosphingolipids in GNE myopathy. J Inherit Metab Dis. 2014;37(2):297–308.CrossRefPubMed Patzel KA, Yardeni T, Le Poëc-Celic E, Leoyklang P, Dorward H, Alonzi DS, Kukushkin NV, Xu B, Zhang Y, Sollogoub M, Blériot Y, Gahl WA, Huizing M, Butters TD. Non-specific accumulation of glycosphingolipids in GNE myopathy. J Inherit Metab Dis. 2014;37(2):297–308.CrossRefPubMed
53.
go back to reference Amsili S, Shlomai Z, Levitzki R, Krause S, Lochmuller H, Ben-Bassat H, Mitrani-Rosenbaum S. Characterization of hereditary inclusion body myopathy myoblasts: possible primary impairment of apoptotic events. Cell Death Differ. 2007;14(11):1916–24.CrossRefPubMed Amsili S, Shlomai Z, Levitzki R, Krause S, Lochmuller H, Ben-Bassat H, Mitrani-Rosenbaum S. Characterization of hereditary inclusion body myopathy myoblasts: possible primary impairment of apoptotic events. Cell Death Differ. 2007;14(11):1916–24.CrossRefPubMed
54.
go back to reference Bennmann D, Weidemann W, Thate A, Kreuzmann D, Horstkorte R. Aberrant O-GlcNAcylation disrupts GNE enzyme activity in GNE myopathy. FEBS J. 2016;283(12):2285–94.CrossRefPubMed Bennmann D, Weidemann W, Thate A, Kreuzmann D, Horstkorte R. Aberrant O-GlcNAcylation disrupts GNE enzyme activity in GNE myopathy. FEBS J. 2016;283(12):2285–94.CrossRefPubMed
55.
go back to reference Amsili S, Zer H, Hinderlich S, Krause S, Becker-Cohen M, MacArthur DG, North KN, Mitrani-Rosenbaum S. UDP-N-acetylglucosamine 2-epimerase/N-acetylmannosamine kinase (GNE) binds to alpha-actinin 1: novel pathways in skeletal muscle? PLoS One. 2008;3(6):e2477.CrossRefPubMedPubMedCentral Amsili S, Zer H, Hinderlich S, Krause S, Becker-Cohen M, MacArthur DG, North KN, Mitrani-Rosenbaum S. UDP-N-acetylglucosamine 2-epimerase/N-acetylmannosamine kinase (GNE) binds to alpha-actinin 1: novel pathways in skeletal muscle? PLoS One. 2008;3(6):e2477.CrossRefPubMedPubMedCentral
56.
go back to reference Harazi A, Becker-Cohen M, Zer H, Moshel O, Hinderlich S, Mitrani-Rosenbaum S. The interaction of UDP-N-Acetylglucosamine 2-epimerase/N-Acetylmannosamine kinase (GNE) and alpha-Actinin 2 is altered in GNE myopathy M743T mutant. Mol Neurobiol. 2017;54(4):2928–38.CrossRefPubMed Harazi A, Becker-Cohen M, Zer H, Moshel O, Hinderlich S, Mitrani-Rosenbaum S. The interaction of UDP-N-Acetylglucosamine 2-epimerase/N-Acetylmannosamine kinase (GNE) and alpha-Actinin 2 is altered in GNE myopathy M743T mutant. Mol Neurobiol. 2017;54(4):2928–38.CrossRefPubMed
57.
go back to reference Harazi A, Chaouat M, Shlomai Z, Levitzki R, Becker-Cohen M, Sadeh M, Dabby R, Ben-Bassat H, Mitrani-Rosenbaum S. Survival-apoptosis associated signaling in GNE myopathy-cultured myoblasts. J Recept Signal Transduct Res. 2015;35(4):249–57.CrossRefPubMed Harazi A, Chaouat M, Shlomai Z, Levitzki R, Becker-Cohen M, Sadeh M, Dabby R, Ben-Bassat H, Mitrani-Rosenbaum S. Survival-apoptosis associated signaling in GNE myopathy-cultured myoblasts. J Recept Signal Transduct Res. 2015;35(4):249–57.CrossRefPubMed
58.
go back to reference Pham ND, Pang PC, Krishnamurthy S, Wands AM, Grassi P, Dell A, Haslam SM, Kohler JJ. Effects of altered sialic acid biosynthesis on N-linked glycan branching and cell surface interactions. J Biol Chem. 2017;292(23):9637–51.CrossRefPubMedPubMedCentral Pham ND, Pang PC, Krishnamurthy S, Wands AM, Grassi P, Dell A, Haslam SM, Kohler JJ. Effects of altered sialic acid biosynthesis on N-linked glycan branching and cell surface interactions. J Biol Chem. 2017;292(23):9637–51.CrossRefPubMedPubMedCentral
59.
go back to reference Grover S, Aslam S, Sharma V, Arya R. Expression and secretion of wild type and mutant GNE proteins in Dictyostelium discoideum. CNS Neurol Disord Drug Targets. 2014;13(7):1263–72.CrossRefPubMed Grover S, Aslam S, Sharma V, Arya R. Expression and secretion of wild type and mutant GNE proteins in Dictyostelium discoideum. CNS Neurol Disord Drug Targets. 2014;13(7):1263–72.CrossRefPubMed
60.
go back to reference Malicdan MC, Noguchi S, Nonaka I, Hayashi YK, Nishino I. A GNE knockout mouse expressing human GNE D176V mutation develops features similar to distal myopathy with rimmed vacuoles or hereditary inclusion body myopathy. Hum Mol Genet. 2007;16(22):2669–82.CrossRefPubMed Malicdan MC, Noguchi S, Nonaka I, Hayashi YK, Nishino I. A GNE knockout mouse expressing human GNE D176V mutation develops features similar to distal myopathy with rimmed vacuoles or hereditary inclusion body myopathy. Hum Mol Genet. 2007;16(22):2669–82.CrossRefPubMed
61.
go back to reference Malicdan MC, Noguchi S, Hayashi YK, Nonaka I, Nishino I. Prophylactic treatment with sialic acid metabolites precludes the development of the myopathic phenotype in the DMRV-hIBM mouse model. Nat Med. 2009;15(6):690–5.CrossRefPubMed Malicdan MC, Noguchi S, Hayashi YK, Nonaka I, Nishino I. Prophylactic treatment with sialic acid metabolites precludes the development of the myopathic phenotype in the DMRV-hIBM mouse model. Nat Med. 2009;15(6):690–5.CrossRefPubMed
62.
go back to reference Malicdan MC, Noguchi S, Tokutomi T, Goto Y, Nonaka I, Hayashi YK, et al. Peracetylated N-acetylmannosamine, a synthetic sugar molecule, efficiently rescues muscle phenotype and biochemical defects in mouse model of sialic acid-deficient myopathy. J Biol Chem. 2012;287(4):2689–705.CrossRefPubMed Malicdan MC, Noguchi S, Tokutomi T, Goto Y, Nonaka I, Hayashi YK, et al. Peracetylated N-acetylmannosamine, a synthetic sugar molecule, efficiently rescues muscle phenotype and biochemical defects in mouse model of sialic acid-deficient myopathy. J Biol Chem. 2012;287(4):2689–705.CrossRefPubMed
63.
go back to reference Malicdan MC, Noguchi S, Hayashi YK, Nishino I. Muscle weakness correlates with muscle atrophy and precedes the development of inclusion body or rimmed vacuoles in the mouse model of DMRV/hIBM. Physiol Genomics. 2008;35(1):106–15.CrossRefPubMed Malicdan MC, Noguchi S, Hayashi YK, Nishino I. Muscle weakness correlates with muscle atrophy and precedes the development of inclusion body or rimmed vacuoles in the mouse model of DMRV/hIBM. Physiol Genomics. 2008;35(1):106–15.CrossRefPubMed
64.
go back to reference Yonekawa T, Malicdan MC, Cho A, Hayashi YK, Nonaka I, Mine T, Yamamoto T, Nishino I, Noguchi S. Sialyllactose ameliorates myopathic phenotypes in symptomatic GNE myopathy model mice. Brain. 2014;137(Pt 10):2670–9.CrossRefPubMedPubMedCentral Yonekawa T, Malicdan MC, Cho A, Hayashi YK, Nonaka I, Mine T, Yamamoto T, Nishino I, Noguchi S. Sialyllactose ameliorates myopathic phenotypes in symptomatic GNE myopathy model mice. Brain. 2014;137(Pt 10):2670–9.CrossRefPubMedPubMedCentral
65.
go back to reference Malicdan MC, Noguchi S, Nonaka I, Hayashi YK, Nishino I. A Gne knockout mouse expressing human V572L mutation develops features similar to distal myopathy with rimmed vacuoles or hereditary inclusion body myopathy. Hum Mol Genet. 2007;16(2):115–28.CrossRefPubMed Malicdan MC, Noguchi S, Nonaka I, Hayashi YK, Nishino I. A Gne knockout mouse expressing human V572L mutation develops features similar to distal myopathy with rimmed vacuoles or hereditary inclusion body myopathy. Hum Mol Genet. 2007;16(2):115–28.CrossRefPubMed
66.
go back to reference Malicdan MC, Noguchi S, Nishino I. Autophagy in a mouse model of distal myopathy with rimmed vacuoles or hereditary inclusion body myopathy. Autophagy. 2007;3(4):396–8.CrossRefPubMed Malicdan MC, Noguchi S, Nishino I. Autophagy in a mouse model of distal myopathy with rimmed vacuoles or hereditary inclusion body myopathy. Autophagy. 2007;3(4):396–8.CrossRefPubMed
67.
go back to reference Ito M, Sugihara K, Asaka T, Toyama T, Yoshihara T, Furuichi K, Wada T, Asano M. Glycoprotein hyposialylation gives rise to a nephrotic-like syndrome that is prevented by sialic acid administration in GNE V572L point-mutant mice. PLoS One. 2012;7(1):e29873.CrossRefPubMedPubMedCentral Ito M, Sugihara K, Asaka T, Toyama T, Yoshihara T, Furuichi K, Wada T, Asano M. Glycoprotein hyposialylation gives rise to a nephrotic-like syndrome that is prevented by sialic acid administration in GNE V572L point-mutant mice. PLoS One. 2012;7(1):e29873.CrossRefPubMedPubMedCentral
68.
go back to reference Cho A, Christine M, Malicdan V, Miyakawa M, Nonaka I, Nishino I, Noguchi S. Sialic acid deficiency is associated with oxidative stress leading to muscle atrophy and weakness in GNE myopathy. Hum Mol Genet. 2017;26(16):3081–93.CrossRefPubMed Cho A, Christine M, Malicdan V, Miyakawa M, Nonaka I, Nishino I, Noguchi S. Sialic acid deficiency is associated with oxidative stress leading to muscle atrophy and weakness in GNE myopathy. Hum Mol Genet. 2017;26(16):3081–93.CrossRefPubMed
69.
go back to reference Sela I, Yakovlev L, Becker Cohen M, Elbaz M, Yanay N, Ben Shlomo U, Yotvat H, Fellig Y, Argov Z, Mitrani-Rosenbaum S. Variable phenotypes of knockin mice carrying the M712T Gne mutation. NeuroMolecular Med. 2013;15(1):180–91.CrossRefPubMed Sela I, Yakovlev L, Becker Cohen M, Elbaz M, Yanay N, Ben Shlomo U, Yotvat H, Fellig Y, Argov Z, Mitrani-Rosenbaum S. Variable phenotypes of knockin mice carrying the M712T Gne mutation. NeuroMolecular Med. 2013;15(1):180–91.CrossRefPubMed
70.
go back to reference Paccalet T, Coulombe Z, Tremblay JP. Ganglioside GM3 levels are altered in a mouse model of HIBM: GM3 as a cellular marker of the disease. PLoS One. 2010;5(4):e10055.CrossRefPubMedPubMedCentral Paccalet T, Coulombe Z, Tremblay JP. Ganglioside GM3 levels are altered in a mouse model of HIBM: GM3 as a cellular marker of the disease. PLoS One. 2010;5(4):e10055.CrossRefPubMedPubMedCentral
71.
go back to reference Niethamer TK, Yardeni T, Leoyklang P, Ciccone C, Astiz-Martinez A, Jacobs K, Dorward HM, Zerfas PM, Gahl WA, Huizing M. Oral monosaccharide therapies to reverse renal and muscle hyposialylation in a mouse model of GNE myopathy. Mol Genet Metab. 2012;107(4):748–55.CrossRefPubMedPubMedCentral Niethamer TK, Yardeni T, Leoyklang P, Ciccone C, Astiz-Martinez A, Jacobs K, Dorward HM, Zerfas PM, Gahl WA, Huizing M. Oral monosaccharide therapies to reverse renal and muscle hyposialylation in a mouse model of GNE myopathy. Mol Genet Metab. 2012;107(4):748–55.CrossRefPubMedPubMedCentral
72.
go back to reference Mitrani-Rosenbaum S, Yakovlev L, Becker Cohen M, Telem M, Elbaz M, Yanay N, Yotvat H, Ben Shlomo U, Harazi A, Fellig Y, Argov Z, Sela I. Sustained expression and safety of human GNE in normal mice after gene transfer based on AAV8 systemic delivery. Neuromuscul Disord. 2012;22(11):1015–24.CrossRefPubMed Mitrani-Rosenbaum S, Yakovlev L, Becker Cohen M, Telem M, Elbaz M, Yanay N, Yotvat H, Ben Shlomo U, Harazi A, Fellig Y, Argov Z, Sela I. Sustained expression and safety of human GNE in normal mice after gene transfer based on AAV8 systemic delivery. Neuromuscul Disord. 2012;22(11):1015–24.CrossRefPubMed
73.
go back to reference Daya A, Vatine GD, Becker-Cohen M, Tal-Goldberg T, Friedmann A, Gothilf Y, Du SJ, Mitrani-Rosenbaum S. Gne depletion during zebrafish development impairs skeletal muscle structure and function. Hum Mol Genet. 2014;23(13):3349–61.CrossRefPubMed Daya A, Vatine GD, Becker-Cohen M, Tal-Goldberg T, Friedmann A, Gothilf Y, Du SJ, Mitrani-Rosenbaum S. Gne depletion during zebrafish development impairs skeletal muscle structure and function. Hum Mol Genet. 2014;23(13):3349–61.CrossRefPubMed
74.
go back to reference Valles-Ayoub Y, Esfandiarifard S, Sinai P, Carbajo R, Khokher Z, No D, Pietruszka M, Darvish B, Kakkis E, Darvish D. Serum neural cell adhesion molecule is hyposialylated in hereditary inclusion body myopathy. Genet Test Mol Biomarkers. 2012;16(5):313–7.CrossRefPubMed Valles-Ayoub Y, Esfandiarifard S, Sinai P, Carbajo R, Khokher Z, No D, Pietruszka M, Darvish B, Kakkis E, Darvish D. Serum neural cell adhesion molecule is hyposialylated in hereditary inclusion body myopathy. Genet Test Mol Biomarkers. 2012;16(5):313–7.CrossRefPubMed
75.
go back to reference Chipman PH, Franz CK, Nelson A, Schachner M, Rafuse VF. Neural cell adhesion molecule is required for stability of reinnervated neuromuscular junctions. Eur J Neurosci. 2010;31(2):238–49.CrossRefPubMed Chipman PH, Franz CK, Nelson A, Schachner M, Rafuse VF. Neural cell adhesion molecule is required for stability of reinnervated neuromuscular junctions. Eur J Neurosci. 2010;31(2):238–49.CrossRefPubMed
76.
go back to reference Zygmunt DA, Crowe KE, Flanigan KM, Martin PT. Comparison of serum rAAV serotype-specific antibodies in patients with Duchenne muscular dystrophy, Becker muscular dystrophy, inclusion body myositis, or GNE myopathy. Hum Gene Ther. 2017;28(9):737–46.CrossRefPubMed Zygmunt DA, Crowe KE, Flanigan KM, Martin PT. Comparison of serum rAAV serotype-specific antibodies in patients with Duchenne muscular dystrophy, Becker muscular dystrophy, inclusion body myositis, or GNE myopathy. Hum Gene Ther. 2017;28(9):737–46.CrossRefPubMed
77.
go back to reference Burch PM, Pogoryelova O, Palandra J, Goldstein R, Bennett D, Fitz L, Guglieri M, Bettolo CM, Straub V, Evangelista T, Neubert H, Lochmüller H, Morris C. Reduced serum myostatin concentrations associated with genetic muscle disease progression. J Neurol. 2017;264(3):541–53.CrossRefPubMed Burch PM, Pogoryelova O, Palandra J, Goldstein R, Bennett D, Fitz L, Guglieri M, Bettolo CM, Straub V, Evangelista T, Neubert H, Lochmüller H, Morris C. Reduced serum myostatin concentrations associated with genetic muscle disease progression. J Neurol. 2017;264(3):541–53.CrossRefPubMed
78.
go back to reference Nemunaitis G, et al. Hereditary inclusion body myopathy: single patient response to intravenous dosing of GNE gene lipoplex. Hum Gene Ther. 2011;22:1331–41.CrossRefPubMedPubMedCentral Nemunaitis G, et al. Hereditary inclusion body myopathy: single patient response to intravenous dosing of GNE gene lipoplex. Hum Gene Ther. 2011;22:1331–41.CrossRefPubMedPubMedCentral
79.
go back to reference Argov Z, et al. Aceneuramic acid extended release administration maintains upper limb muscle strength in a 48-week study of subjects with GNE myopathy: results from a phase 2, randomized, controlled study. J Neuromuscul Dis. 2016;3:49–66.CrossRefPubMedPubMedCentral Argov Z, et al. Aceneuramic acid extended release administration maintains upper limb muscle strength in a 48-week study of subjects with GNE myopathy: results from a phase 2, randomized, controlled study. J Neuromuscul Dis. 2016;3:49–66.CrossRefPubMedPubMedCentral
80.
go back to reference Dawes H, Korpershoek N, Freebody J, Elsworth C, van Tintelen N, Wade DT, Izadi H, Jones DH. A pilot randomised controlled trial of a home-based exercise programme aimed at improving endurance and function in adults with neuromuscular disorders. J Neurol Neurosurg Psychiatry. 2006;77(8):959–62.CrossRefPubMedPubMedCentral Dawes H, Korpershoek N, Freebody J, Elsworth C, van Tintelen N, Wade DT, Izadi H, Jones DH. A pilot randomised controlled trial of a home-based exercise programme aimed at improving endurance and function in adults with neuromuscular disorders. J Neurol Neurosurg Psychiatry. 2006;77(8):959–62.CrossRefPubMedPubMedCentral
Metadata
Title
GNE myopathy: from clinics and genetics to pathology and research strategies
Authors
Oksana Pogoryelova
José Andrés González Coraspe
Nikoletta Nikolenko
Hanns Lochmüller
Andreas Roos
Publication date
01-12-2018
Publisher
BioMed Central
Published in
Orphanet Journal of Rare Diseases / Issue 1/2018
Electronic ISSN: 1750-1172
DOI
https://doi.org/10.1186/s13023-018-0802-x

Other articles of this Issue 1/2018

Orphanet Journal of Rare Diseases 1/2018 Go to the issue