Skip to main content
Top
Published in: BMC Medical Genetics 1/2017

Open Access 01-12-2017 | Case report

Case reports of juvenile GM1 gangliosidosisis type II caused by mutation in GLB1 gene

Authors: Parvaneh Karimzadeh, Samaneh Naderi, Farzaneh Modarresi, Hassan Dastsooz, Hamid Nemati, Tayebeh Farokhashtiani, Bibi Shahin Shamsian, Soroor Inaloo, Mohammad Ali Faghihi

Published in: BMC Medical Genetics | Issue 1/2017

Login to get access

Abstract

Background

Type II or juvenile GM1-gangliosidosis is an autosomal recessive lysosomal storage disorder, which is clinically distinct from infantile form of the disease by the lack of characteristic cherry-red spot and hepatosplenomegaly. The disease is characterized by slowly progressive neurodegeneration and mild skeletal changes. Due to the later age of onset and uncharacteristic presentation, diagnosis is frequently puzzled with other ataxic and purely neurological disorders. Up to now, 3–4 types of GM1-gangliosidosis have been reported and among them type I is the most common phenotype with the age of onset around 6 months. Various forms of GM1-gangliosidosis are caused by GLB1 gene mutations but severity of the disease and age of onset are directly related to the position and the nature of deleterious mutations. However, due to its unique genetic cause and overlapping clinical features, some researchers believe that GM1 gangliosidosis represents an overlapped disease spectrum instead of four distinct types.

Case presentation

Here, we report a less frequent type of autosomal recessive GM1 gangliosidosis with perplexing clinical presentation in three families in the southwest part of Iran, who are unrelated but all from “Lurs” ethnic background. To identify disease-causing mutations, Whole Exome Sequencing (WES) utilizing next generation sequencing was performed. Four patients from three families were investigated with the age of onset around 3 years old. Clinical presentations were ataxia, gate disturbances and dystonia leading to wheelchair-dependent disability, regression of intellectual abilities, and general developmental regression. They all were born in consanguineous families with no previous documented similar disease in their parents. A homozygote missense mutation in GLB1 gene (c. 601 G > A, p.R201C) was found in all patients. Using Sanger sequencing this identified mutation was confirmed in the proband, their parents, grandparents, and extended family members, confirming its autosomal recessive pattern of inheritance.

Conclusions

Our study identified a rare pathogenic missense mutation in GLB1 gene in patients with complex neurodevelopmental findings, which can extend the list of differential diagnoses for childhood ataxia in Iranian patients.
Literature
1.
go back to reference Sanchez-Fernandez EM, Garcia Fernandez JM, Mellet CO. Glycomimetic-based pharmacological chaperones for lysosomal storage disorders: lessons from Gaucher, GM1-gangliosidosis and Fabry diseases. Chem Commun (Camb). 2016;52(32):5497–515.CrossRef Sanchez-Fernandez EM, Garcia Fernandez JM, Mellet CO. Glycomimetic-based pharmacological chaperones for lysosomal storage disorders: lessons from Gaucher, GM1-gangliosidosis and Fabry diseases. Chem Commun (Camb). 2016;52(32):5497–515.CrossRef
2.
go back to reference Kannebley JS, Silveira-Moriyama L, Bastos LO, Steiner CE. Clinical Findings and Natural History in Ten Unrelated Families with Juvenile and Adult GM1 Gangliosidosis. JIMD Rep. 2015;24:115–22.CrossRefPubMedPubMedCentral Kannebley JS, Silveira-Moriyama L, Bastos LO, Steiner CE. Clinical Findings and Natural History in Ten Unrelated Families with Juvenile and Adult GM1 Gangliosidosis. JIMD Rep. 2015;24:115–22.CrossRefPubMedPubMedCentral
3.
go back to reference Sandhoff K, Harzer K. Gangliosides and gangliosidoses: principles of molecular and metabolic pathogenesis. J Neurosci. 2013;33(25):10195–208.CrossRefPubMed Sandhoff K, Harzer K. Gangliosides and gangliosidoses: principles of molecular and metabolic pathogenesis. J Neurosci. 2013;33(25):10195–208.CrossRefPubMed
4.
go back to reference Jeyakumar M, Thomas R, Elliot‐Smith E, Smith D, Van Der Spoel A, d’Azzo A, Perry VH, Butters T, Dwek R, Platt F. Central nervous system inflammation is a hallmark of pathogenesis in mouse models of GM1 and GM2 gangliosidosis. Brain. 2003;126(4):974–87.CrossRefPubMed Jeyakumar M, Thomas R, Elliot‐Smith E, Smith D, Van Der Spoel A, d’Azzo A, Perry VH, Butters T, Dwek R, Platt F. Central nervous system inflammation is a hallmark of pathogenesis in mouse models of GM1 and GM2 gangliosidosis. Brain. 2003;126(4):974–87.CrossRefPubMed
5.
go back to reference Armstrong-Javors A, Chu CJ. Child neurology: Exaggerated dermal melanocytosis in a hypotonic infant: a harbinger of GM1 gangliosidosis. Neurology. 2014;83(17):e166–8.CrossRefPubMedPubMedCentral Armstrong-Javors A, Chu CJ. Child neurology: Exaggerated dermal melanocytosis in a hypotonic infant: a harbinger of GM1 gangliosidosis. Neurology. 2014;83(17):e166–8.CrossRefPubMedPubMedCentral
6.
go back to reference Callahan JW. Molecular basis of GM1 gangliosidosis and Morquio disease, type B. Structure–function studies of lysosomal β-galactosidase and the non-lysosomal β-galactosidase-like protein. Biochim Biophys Acta (BBA) - Mol Basis Dis. 1999;1455(2):85–103.CrossRef Callahan JW. Molecular basis of GM1 gangliosidosis and Morquio disease, type B. Structure–function studies of lysosomal β-galactosidase and the non-lysosomal β-galactosidase-like protein. Biochim Biophys Acta (BBA) - Mol Basis Dis. 1999;1455(2):85–103.CrossRef
7.
go back to reference Regier DS, Kwon HJ, Johnston J, Golas G, Yang S, Wiggs E, Latour Y, Thomas S, Portner C, Adams D, et al. MRI/MRS as a surrogate marker for clinical progression in GM1 gangliosidosis. Am J Med Genet A. 2016;170(3):634–44.CrossRefPubMed Regier DS, Kwon HJ, Johnston J, Golas G, Yang S, Wiggs E, Latour Y, Thomas S, Portner C, Adams D, et al. MRI/MRS as a surrogate marker for clinical progression in GM1 gangliosidosis. Am J Med Genet A. 2016;170(3):634–44.CrossRefPubMed
8.
go back to reference Giugliani R, Dutra JC, Pereira MLS, Rotta N, de Drachler ML, Ohlweiller L, de Pina Neto JM, Pinheiro CE, Breda DJ. GM1 gangliosidosis: clinical and laboratory findings in eight families. Hum Genet. 1985;70(4):347–54.CrossRefPubMed Giugliani R, Dutra JC, Pereira MLS, Rotta N, de Drachler ML, Ohlweiller L, de Pina Neto JM, Pinheiro CE, Breda DJ. GM1 gangliosidosis: clinical and laboratory findings in eight families. Hum Genet. 1985;70(4):347–54.CrossRefPubMed
9.
go back to reference Wolfe LS, Senior RG, Kin NNY. The structures of oligosaccharides accumulating in the liver of GM1-gangliosidosis, type I. J Biol Chem. 1974;249(6):1828–38.PubMed Wolfe LS, Senior RG, Kin NNY. The structures of oligosaccharides accumulating in the liver of GM1-gangliosidosis, type I. J Biol Chem. 1974;249(6):1828–38.PubMed
10.
go back to reference Suzuki Y, Oshima A. A beta-galactosidase gene mutation identified in both Morquio B disease and infantile GM1 gangliosidosis. Hum Genet. 1993;91(4):407.CrossRefPubMed Suzuki Y, Oshima A. A beta-galactosidase gene mutation identified in both Morquio B disease and infantile GM1 gangliosidosis. Hum Genet. 1993;91(4):407.CrossRefPubMed
11.
go back to reference Wenger DA, Goodman SI, Myers GG. Letter: Beta-galactosidase deficiency in young adults. Lancet. 1974;2(7892):1319–20.CrossRefPubMed Wenger DA, Goodman SI, Myers GG. Letter: Beta-galactosidase deficiency in young adults. Lancet. 1974;2(7892):1319–20.CrossRefPubMed
12.
go back to reference Suzuki Y, Nakamura N, Fukuoka K, Shimada Y, Uono M. beta-Galactosidase deficiency in juvenile and adult patients. Report of six Japanese cases and review of literature. Hum Genet. 1977;36(2):219–29.CrossRefPubMed Suzuki Y, Nakamura N, Fukuoka K, Shimada Y, Uono M. beta-Galactosidase deficiency in juvenile and adult patients. Report of six Japanese cases and review of literature. Hum Genet. 1977;36(2):219–29.CrossRefPubMed
13.
go back to reference Quakenbush A. Analysis of the Ovine NEU1 Gene in a Unique Model of GM1 Gangliosidosis. 2015. Quakenbush A. Analysis of the Ovine NEU1 Gene in a Unique Model of GM1 Gangliosidosis. 2015.
14.
go back to reference Caciotti A, Bardelli T, Cunningham J, D’Azzo A, Zammarchi E, Morrone A. Modulating action of the new polymorphism L436F detected in the GLB1 gene of a type-II GM1 gangliosidosis patient. Hum Genet. 2003;113(1):44–50.PubMed Caciotti A, Bardelli T, Cunningham J, D’Azzo A, Zammarchi E, Morrone A. Modulating action of the new polymorphism L436F detected in the GLB1 gene of a type-II GM1 gangliosidosis patient. Hum Genet. 2003;113(1):44–50.PubMed
15.
go back to reference Brunetti-Pierri N, Scaglia F. GM 1 gangliosidosis: Review of clinical, molecular, and therapeutic aspects. Mol Genet Metab. 2008;94(4):391–6.CrossRefPubMed Brunetti-Pierri N, Scaglia F. GM 1 gangliosidosis: Review of clinical, molecular, and therapeutic aspects. Mol Genet Metab. 2008;94(4):391–6.CrossRefPubMed
16.
go back to reference Okada S, O’Brien JS. Generalized gangliosidosis: beta-galactosidase deficiency. Science. 1968;160(3831):1002–4.CrossRefPubMed Okada S, O’Brien JS. Generalized gangliosidosis: beta-galactosidase deficiency. Science. 1968;160(3831):1002–4.CrossRefPubMed
17.
go back to reference Filocamo M, Morrone A. Lysosomal storage disorders: molecular basis and laboratory testing. Hum Genomics. 2011;5(3):1.CrossRef Filocamo M, Morrone A. Lysosomal storage disorders: molecular basis and laboratory testing. Hum Genomics. 2011;5(3):1.CrossRef
18.
go back to reference Yoshida K, Oshima A, Shimmoto M, Fukuhara Y, Sakuraba H, Yanagisawa N, Suzuki Y. Human beta-galactosidase gene mutations in GM1-gangliosidosis: a common mutation among Japanese adult/chronic cases. Am J Hum Genet. 1991;49(2):435–42.PubMedPubMedCentral Yoshida K, Oshima A, Shimmoto M, Fukuhara Y, Sakuraba H, Yanagisawa N, Suzuki Y. Human beta-galactosidase gene mutations in GM1-gangliosidosis: a common mutation among Japanese adult/chronic cases. Am J Hum Genet. 1991;49(2):435–42.PubMedPubMedCentral
19.
go back to reference Acosta W, Martin R, Radin DN, Cramer CL. High-throughput imaging method for direct assessment of GM1 ganglioside levels in mammalian cells. Data Brief. 2016;6:1016–22.CrossRefPubMedPubMedCentral Acosta W, Martin R, Radin DN, Cramer CL. High-throughput imaging method for direct assessment of GM1 ganglioside levels in mammalian cells. Data Brief. 2016;6:1016–22.CrossRefPubMedPubMedCentral
20.
go back to reference Georgiou T, Stylianidou G, Anastasiadou V, Caciotti A, Campos Y, Zammarchi E, Morrone A, D’azzo A, Drousiotou A. The Arg482His mutation in the β-galactosidase gene is responsible for a high frequency of GM1 gangliosidosis carriers in a Cypriot village. Genet Test. 2005;9(2):126–32.CrossRefPubMed Georgiou T, Stylianidou G, Anastasiadou V, Caciotti A, Campos Y, Zammarchi E, Morrone A, D’azzo A, Drousiotou A. The Arg482His mutation in the β-galactosidase gene is responsible for a high frequency of GM1 gangliosidosis carriers in a Cypriot village. Genet Test. 2005;9(2):126–32.CrossRefPubMed
Metadata
Title
Case reports of juvenile GM1 gangliosidosisis type II caused by mutation in GLB1 gene
Authors
Parvaneh Karimzadeh
Samaneh Naderi
Farzaneh Modarresi
Hassan Dastsooz
Hamid Nemati
Tayebeh Farokhashtiani
Bibi Shahin Shamsian
Soroor Inaloo
Mohammad Ali Faghihi
Publication date
01-12-2017
Publisher
BioMed Central
Published in
BMC Medical Genetics / Issue 1/2017
Electronic ISSN: 1471-2350
DOI
https://doi.org/10.1186/s12881-017-0417-4

Other articles of this Issue 1/2017

BMC Medical Genetics 1/2017 Go to the issue