1
|
Hosseini K, Fallahi J, Tabei SMB, Razban V. Gene therapy approaches for GM1 gangliosidosis: Focus on animal and cellular studies. Cell Biochem Funct 2023; 41:1093-1105. [PMID: 38018878 DOI: 10.1002/cbf.3887] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2023] [Revised: 11/10/2023] [Accepted: 11/11/2023] [Indexed: 11/30/2023]
Abstract
One of the most important inherited metabolic disorders is GM1 gangliosidosis, which is a progressive neurological disorder. The main cause of this disease is a genetic defect in the enzyme β-galactosidase due to a mutation in the glb1 gene. Lack of this enzyme in cells (especially neurons) leads to the accumulation of ganglioside substrate in nerve tissues, followed by three clinical forms of GM1 disease (neonatal, juvenile, and adult variants). Genetically, many mutations occur in the exons of the glb1 gene, such as exons 2, 6, 15, and 16, so the most common ones reported in scientific studies include missense/nonsense mutations. Therefore, many studies have examined the genotype-phenotype relationships of this disease and subsequently using gene therapy techniques have been able to reduce the complications of the disease and alleviate the signs and symptoms of the disease. In this regard, the present article reviews the general features of GM1 gangliosidosis and its mutations, as well as gene therapy studies and animal and human models of the disease.
Collapse
Affiliation(s)
- Kamran Hosseini
- Student Research Committee, Shiraz University of Medical Sciences, Shiraz, Iran
- Department of Molecular Medicine, Faculty of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Jafar Fallahi
- Department of Molecular Medicine, Faculty of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Seyed M B Tabei
- Department of Medical Genetics, Shiraz University of Medical Sciences, Shiraz, Iran
- Comprehensive Medical Genetic Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Vahid Razban
- Department of Molecular Medicine, Faculty of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran
- Stem Cells Technology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| |
Collapse
|
2
|
Rudinskiy M, Pons-Vizcarra M, Soldà T, Fregno I, Bergmann TJ, Ruano A, Delgado A, Morales S, Barril X, Bellotto M, Cubero E, García-Collazo AM, Pérez-Carmona N, Molinari M. Validation of a highly sensitive HaloTag-based assay to evaluate the potency of a novel class of allosteric β-Galactosidase correctors. PLoS One 2023; 18:e0294437. [PMID: 38019733 PMCID: PMC10686464 DOI: 10.1371/journal.pone.0294437] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Accepted: 10/31/2023] [Indexed: 12/01/2023] Open
Abstract
Site-directed Enzyme Enhancement Therapy (SEE-Tx®) technology is a disease-agnostic drug discovery tool that can be applied to any protein target of interest with a known three-dimensional structure. We used this proprietary technology to identify and characterize the therapeutic potential of structurally targeted allosteric regulators (STARs) of the lysosomal hydrolase β-galactosidase (β-Gal), which is deficient due to gene mutations in galactosidase beta 1 (GLB1)-related lysosomal storage disorders (LSDs). The biochemical HaloTag cleavage assay was used to monitor the delivery of wildtype (WT) β-Gal and four disease-related β-Gal variants (p.Ile51Thr, p.Arg59His, p.Arg201Cys and p.Trp273Leu) in the presence and absence of two identified STAR compounds. In addition, the ability of STARs to reduce toxic substrate was assessed in a canine fibroblast cell model. In contrast to the competitive pharmacological chaperone N-nonyl-deoxygalactonojirimycin (NN-DGJ), the two identified STAR compounds stabilized and substantially enhanced the lysosomal transport of wildtype enzyme and disease-causing β-Gal variants. In addition, the two STAR compounds reduced the intracellular accumulation of exogenous GM1 ganglioside, an effect not observed with the competitive chaperone NN-DGJ. This proof-of-concept study demonstrates that the SEE-Tx® platform is a rapid and cost-effective drug discovery tool for identifying STARs for the treatment of LSDs. In addition, the HaloTag assay developed in our lab has proved valuable in investigating the effect of STARs in promoting enzyme transport and lysosomal delivery. Automatization and upscaling of this assay would be beneficial for screening STARs as part of the drug discovery process.
Collapse
Affiliation(s)
- Mikhail Rudinskiy
- Università Della Svizzera Italiana, Lugano, Switzerland
- Department of Biology, Swiss Federal Institute of Technology, Zurich, Switzerland
- Institute for Research in Biomedicine, Bellinzona, Switzerland
| | - Maria Pons-Vizcarra
- Gain Therapeutics Sucursal en España, Parc Científic de Barcelona, Barcelona, Spain
| | - Tatiana Soldà
- Università Della Svizzera Italiana, Lugano, Switzerland
- Institute for Research in Biomedicine, Bellinzona, Switzerland
| | - Ilaria Fregno
- Università Della Svizzera Italiana, Lugano, Switzerland
- Institute for Research in Biomedicine, Bellinzona, Switzerland
| | - Timothy Jan Bergmann
- Università Della Svizzera Italiana, Lugano, Switzerland
- Institute for Research in Biomedicine, Bellinzona, Switzerland
| | - Ana Ruano
- Gain Therapeutics Sucursal en España, Parc Científic de Barcelona, Barcelona, Spain
| | - Aida Delgado
- Gain Therapeutics Sucursal en España, Parc Científic de Barcelona, Barcelona, Spain
| | - Sara Morales
- Gain Therapeutics Sucursal en España, Parc Científic de Barcelona, Barcelona, Spain
| | - Xavier Barril
- Gain Therapeutics Sucursal en España, Parc Científic de Barcelona, Barcelona, Spain
- Facultat de Farmacia, IBUB & IQTC, Universitat de Barcelona, Barcelona, Spain
- Catalan Institution for Research and Advanced Studies (ICREA), Barcelona, Spain
| | | | - Elena Cubero
- Gain Therapeutics Sucursal en España, Parc Científic de Barcelona, Barcelona, Spain
| | | | | | - Maurizio Molinari
- Università Della Svizzera Italiana, Lugano, Switzerland
- Institute for Research in Biomedicine, Bellinzona, Switzerland
- School of Life Sciences, École Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
| |
Collapse
|
3
|
Kingma SDK, Ceulemans B, Kenis S, Jonckheere AI. Are GMI gangliosidosis and Morquio type B two different disorders or part of one phenotypic spectrum? JIMD Rep 2021; 59:90-103. [PMID: 33977034 PMCID: PMC8100397 DOI: 10.1002/jmd2.12204] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/25/2020] [Revised: 01/12/2021] [Accepted: 01/19/2021] [Indexed: 11/09/2022] Open
Abstract
Monosialotetrahexosylganglioside (GMI) gangliosidosis and Morquio type B (MorB) are two lysosomal storage disorders (LSDs) caused by the same enzyme deficiency, β-galactosidase (βgal). GMI gangliosidosis, associated with GMI ganglioside accumulation, is a neurodegenerative condition characterized by psychomotor regression, visceromegaly, cherry red spot, and facial and skeletal abnormalities. MorB is characterized by prominent and severe skeletal deformities due to keratan sulfate (KS) accumulation. There are only a few reports on intermediate phenotypes between GMI gangliosidosis and MorB. The presentation of two new patients with this rare intermediate phenotype motivated us to review the literature, to study differences and similarities between GMI gangliosidosis and MorB, and to speculate about the possible mechanisms that may contribute to the differences in clinical presentation. In conclusion, we hypothesize that GMI gangliosidosis and MorB are part of one phenotypic spectrum of the same disease and that the classification of LSDs might need to be revised.
Collapse
Affiliation(s)
- Sandra D. K. Kingma
- Centre for Metabolic DiseasesUniversity Hospital Antwerp, University of AntwerpEdegem, AntwerpBelgium
- Department of Pediatric NeurologyAntwerp University Hospital, University of AntwerpEdegem, AntwerpBelgium
| | - Berten Ceulemans
- Department of Pediatric NeurologyAntwerp University Hospital, University of AntwerpEdegem, AntwerpBelgium
| | - Sandra Kenis
- Department of Pediatric NeurologyAntwerp University Hospital, University of AntwerpEdegem, AntwerpBelgium
| | - An I. Jonckheere
- Centre for Metabolic DiseasesUniversity Hospital Antwerp, University of AntwerpEdegem, AntwerpBelgium
- Department of Pediatric NeurologyAntwerp University Hospital, University of AntwerpEdegem, AntwerpBelgium
| |
Collapse
|
4
|
Rha AK, Maguire AS, Martin DR. GM1 Gangliosidosis: Mechanisms and Management. Appl Clin Genet 2021; 14:209-233. [PMID: 33859490 PMCID: PMC8044076 DOI: 10.2147/tacg.s206076] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2020] [Accepted: 01/15/2021] [Indexed: 01/10/2023] Open
Abstract
The lysosomal storage disorder, GM1 gangliosidosis (GM1), is a neurodegenerative condition resulting from deficiency of the enzyme β-galactosidase (β-gal). Mutation of the GLB1 gene, which codes for β-gal, prevents cleavage of the terminal β-1,4-linked galactose residue from GM1 ganglioside. Subsequent accumulation of GM1 ganglioside and other substrates in the lysosome impairs cell physiology and precipitates dysfunction of the nervous system. Beyond palliative and supportive care, no FDA-approved treatments exist for GM1 patients. Researchers are critically evaluating the efficacy of substrate reduction therapy, pharmacological chaperones, enzyme replacement therapy, stem cell transplantation, and gene therapy for GM1. A Phase I/II clinical trial for GM1 children is ongoing to evaluate the safety and efficacy of adeno-associated virus-mediated GLB1 delivery by intravenous injection, providing patients and families with hope for the future.
Collapse
Affiliation(s)
- Allisandra K Rha
- Scott-Ritchey Research Center, Auburn University, Auburn, AL, 36849, USA
| | - Anne S Maguire
- Scott-Ritchey Research Center, Auburn University, Auburn, AL, 36849, USA
- Department of Anatomy, Physiology, and Pharmacology, Auburn University College of Veterinary Medicine, Auburn, AL, 36849, USA
| | - Douglas R Martin
- Scott-Ritchey Research Center, Auburn University, Auburn, AL, 36849, USA
- Department of Anatomy, Physiology, and Pharmacology, Auburn University College of Veterinary Medicine, Auburn, AL, 36849, USA
| |
Collapse
|
5
|
Tebani A, Sudrié-Arnaud B, Dabaj I, Torre S, Domitille L, Snanoudj S, Heron B, Levade T, Caillaud C, Vergnaud S, Saugier-Veber P, Coutant S, Dranguet H, Froissart R, Al Khouri M, Alembik Y, Baruteau J, Arnoux JB, Brassier A, Brehin AC, Busa T, Cano A, Chabrol B, Coubes C, Desguerre I, Doco-Fenzy M, Drenou B, Elcioglu NH, Elsayed S, Fouilhoux A, Poirsier C, Goldenberg A, Jouvencel P, Kuster A, Labarthe F, Lazaro L, Pichard S, Rivera S, Roche S, Roggerone S, Roubertie A, Sigaudy S, Spodenkiewicz M, Tardieu M, Vanhulle C, Marret S, Bekri S. Disentangling molecular and clinical stratification patterns in beta-galactosidase deficiency. J Med Genet 2021; 59:377-384. [PMID: 33737400 DOI: 10.1136/jmedgenet-2020-107510] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2020] [Revised: 01/16/2021] [Accepted: 01/19/2021] [Indexed: 11/04/2022]
Abstract
INTRODUCTION This study aims to define the phenotypic and molecular spectrum of the two clinical forms of β-galactosidase (β-GAL) deficiency, GM1-gangliosidosis and mucopolysaccharidosis IVB (Morquio disease type B, MPSIVB). METHODS Clinical and genetic data of 52 probands, 47 patients with GM1-gangliosidosis and 5 patients with MPSIVB were analysed. RESULTS The clinical presentations in patients with GM1-gangliosidosis are consistent with a phenotypic continuum ranging from a severe antenatal form with hydrops fetalis to an adult form with an extrapyramidal syndrome. Molecular studies evidenced 47 variants located throughout the sequence of the GLB1 gene, in all exons except 7, 11 and 12. Eighteen novel variants (15 substitutions and 3 deletions) were identified. Several variants were linked specifically to early-onset GM1-gangliosidosis, late-onset GM1-gangliosidosis or MPSIVB phenotypes. This integrative molecular and clinical stratification suggests a variant-driven patient assignment to a given clinical and severity group. CONCLUSION This study reports one of the largest series of b-GAL deficiency with an integrative patient stratification combining molecular and clinical features. This work contributes to expand the community knowledge regarding the molecular and clinical landscapes of b-GAL deficiency for a better patient management.
Collapse
Affiliation(s)
- Abdellah Tebani
- Department of Metabolic Biochemistry, Rouen University Hospital, Rouen, France.,Normandie Univ, UNIROUEN, CHU Rouen, INSERM U1245, 76000 Rouen, Normandy Center for Genomic and Personalized Medicine, Rouen, France
| | | | - Ivana Dabaj
- Normandie Univ, UNIROUEN, CHU Rouen, INSERM U1245, 76000 Rouen, Normandy Center for Genomic and Personalized Medicine, Rouen, France.,Department of Neonatal Pediatrics, Intensive Care and Neuropediatrics, Rouen University Hospital, Rouen, France
| | - Stéphanie Torre
- Department of Neonatal Pediatrics, Intensive Care and Neuropediatrics, Rouen University Hospital, Rouen, France
| | - Laur Domitille
- Pediatric Neurology Department, Robert Debré Hospital, Public Hospital Network of Paris, Paris, France
| | - Sarah Snanoudj
- Department of Metabolic Biochemistry, Rouen University Hospital, Rouen, France.,Normandie Univ, UNIROUEN, CHU Rouen, INSERM U1245, 76000 Rouen, Normandy Center for Genomic and Personalized Medicine, Rouen, France
| | - Benedicte Heron
- Reference Center for Lysosomal Diseases, Pediatric Neurology Department, UH Armand Trousseau-La Roche Guyon, APHP, GUEP, Paris, France
| | - Thierry Levade
- Laboratoire de Biochimie Métabolique, Institut Fédératif de Biologie, CHU Purpan, Toulouse, France.,Cancer Research Center, INSERM UMR1037 CRCT, Toulouse, France
| | - Catherine Caillaud
- Biochemistry, Metabolomic and Proteomic Department, Necker Enfants Malades University Hospital, Assistance Publique Hôpitaux de Paris, UMRS 1151, INSERM, Institute Necker Enfants Malades, Paris Descartes University, Paris, France
| | - Sabrina Vergnaud
- UF Maladies Héréditaires Enzymatiques Rares-CGD, Institut de Biologie et de Pathologies, CHU de Grenoble Alpes, Grenoble, France
| | - Pascale Saugier-Veber
- Normandie Univ, UNIROUEN, CHU Rouen, INSERM U1245, 76000 Rouen, Normandy Center for Genomic and Personalized Medicine, Rouen, France
| | - Sophie Coutant
- Department of Genetics, Normandie Univ, UNIROUEN, Inserm U1245 and Rouen University Hospital, F76000, Normandy Centre for Genomic and Personalized Medicine, ROUEN, France
| | - Hélène Dranguet
- Department of Metabolic Biochemistry, Rouen University Hospital, Rouen, France
| | - Roseline Froissart
- Biochemical and Molecular Biology Department, Centre de Biologie et de Pathologie Est Hospices Civils de Lyon, Lyon, France
| | - Majed Al Khouri
- Department of Pediatric Gastroenterology, hepatology and Nutrition, University hospital of Montpellier, Montpellier, France
| | - Yves Alembik
- Department of Clinical Genetic, Hôpitaux Universitaires de Strasbourg, Strasbourg, France
| | - Julien Baruteau
- Metabolic Medicine Department, Great Ormond Street Hospital for Children NHS Foundation Trust, Great Ormond Street Institute of Child Health, University College London, London, UK
| | - Jean-Baptiste Arnoux
- Department of Inherited Metabolic Disease, Necker-Enfants Malades University Hospital, AP-HP, Paris, France
| | - Anais Brassier
- Reference Center of Inherited Metabolic Diseases, Necker Enfants Malades Hospital, Imagine Institute, University Paris Descartes, Paris, France
| | - Anne-Claire Brehin
- Normandie Univ, UNIROUEN, CHU Rouen, INSERM U1245, 76000 Rouen, Normandy Center for Genomic and Personalized Medicine, Rouen, France
| | - Tiffany Busa
- Département de Génétique Médicale, Hôpital Timone Enfant, Marseille, France
| | - Aline Cano
- Centre de Référence des Maladies Héréditaires du Métabolisme, Service de Neuropédiatrie, CHU La Timone Enfants, APHM, Marseille, France
| | - Brigitte Chabrol
- Centre de Référence des Maladies Héréditaires du Métabolisme, Service de Neuropédiatrie, CHU La Timone Enfants, APHM, Marseille, France
| | - Christine Coubes
- Genetic Services, A. de Villeneuve Hospital, Montpellier, France
| | - Isabelle Desguerre
- Department of Paediatric Neurology, Hopital universitaire Necker-Enfants malades Service de Pediatrie generale, Paris, Île-de-France, France
| | - Martine Doco-Fenzy
- Service de génétique, CHRU Reims, Reims, France.,EA3801, UFR médecine, France
| | - Bernard Drenou
- Department of Hematolog, Hôpital Emile Muller - CH de Mulhouse, Mulhouse, France
| | - Nursel H Elcioglu
- Pediatric Genetics, Marmara University Medical School, Istanbul, Turkey
| | - Solaf Elsayed
- Genetics, Children's Hospital, Ain Shams University, Cairo, Egypt
| | - Alain Fouilhoux
- Department of Pediatric Metabolism, Reference Center of Inherited Metabolic Disorders, Femme Mère Enfant Hospital, Lyon, France
| | - Céline Poirsier
- Genetic department, CHU-Reims, EA3801, SFR CAP santé, Reims, France
| | - Alice Goldenberg
- Department of Genetics, Normandie Univ, UNIROUEN, Inserm U1245 and Rouen University Hospital, F76000, Normandy Centre for Genomic and Personalized Medicine, ROUEN, France
| | - Philippe Jouvencel
- Department of Neonatology and Paediatrics, Centre Hospitalier de la Côte Basque, Bayonne, France
| | - Alice Kuster
- Pediatric Critical Care Unit, Femme-Enfants-Adolescents Hospital, Nantes University, Nantes, France
| | | | - Leila Lazaro
- Department of Neonatology and Paediatrics, Centre Hospitalier de la Côte Basque, Bayonne, France
| | - Samia Pichard
- Reference Centre for Inborn Errors of Metabolism, Robert-Debré University Hospital, APHP, Paris, France
| | - Serge Rivera
- Department of Neonatology and Paediatrics, Centre Hospitalier de la Côte Basque, Bayonne, France
| | - Sandrine Roche
- Department of Pediatrics, Bordeaux University Hospital, Bordeaux, France
| | | | - Agathe Roubertie
- INSERM U 1051, Institut des Neurosciences de Montpellier, Montpellier, Hérault, France.,Département de Neuropédiatrie, CHU Gui de Chauliac, Montpellier, France
| | - Sabine Sigaudy
- Genetics, Hôpital d'Enfants de la Timone, Marseille, France
| | | | - Marine Tardieu
- Department of Pediatrics, Reference Center of Inherited Metabolic Disorders, Clocheville Hospital, Tours, France
| | - Catherine Vanhulle
- Department of Neonatal Pediatrics, Intensive Care and Neuropediatrics, Rouen University Hospital, Rouen, France
| | - Stéphane Marret
- Normandie Univ, UNIROUEN, CHU Rouen, INSERM U1245, 76000 Rouen, Normandy Center for Genomic and Personalized Medicine, Rouen, France.,Department of Neonatal Pediatrics, Intensive Care and Neuropediatrics, Rouen University Hospital, Rouen, France
| | - Soumeya Bekri
- Normandie Univ, UNIROUEN, CHU Rouen, INSERM U1245, 76000 Rouen, Normandy Center for Genomic and Personalized Medicine, Rouen, France .,Department of Metabolic Biochemistry, University Hospital Centre Rouen, Rouen, Normandie, France
| |
Collapse
|
6
|
Percival BC, Latour YL, Tifft CJ, Grootveld M. Rapid Identification of New Biomarkers for the Classification of GM1 Type 2 Gangliosidosis Using an Unbiased 1H NMR-Linked Metabolomics Strategy. Cells 2021; 10:572. [PMID: 33807817 PMCID: PMC7998791 DOI: 10.3390/cells10030572] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2020] [Revised: 02/16/2021] [Accepted: 02/17/2021] [Indexed: 01/04/2023] Open
Abstract
Biomarkers currently available for the diagnosis, prognosis, and therapeutic monitoring of GM1 gangliosidosis type 2 (GM1T2) disease are mainly limited to those discovered in targeted proteomic-based studies. In order to identify and establish new, predominantly low-molecular-mass biomarkers for this disorder, we employed an untargeted, multi-analyte approach involving high-resolution 1H NMR analysis coupled to a range of multivariate analysis and computational intelligence technique (CIT) strategies to explore biomolecular distinctions between blood plasma samples collected from GM1T2 and healthy control (HC) participants (n = 10 and 28, respectively). The relationship of these differences to metabolic mechanisms underlying the pathogenesis of GM1T2 disorder was also investigated. 1H NMR-linked metabolomics analyses revealed significant GM1T2-mediated dysregulations in ≥13 blood plasma metabolites (corrected p < 0.04), and these included significant upregulations in 7 amino acids, and downregulations in lipoprotein-associated triacylglycerols and alanine. Indeed, results acquired demonstrated a profound distinctiveness between the GM1T2 and HC profiles. Additionally, employment of a genome-scale network model of human metabolism provided evidence that perturbations to propanoate, ethanol, amino-sugar, aspartate, seleno-amino acid, glutathione and alanine metabolism, fatty acid biosynthesis, and most especially branched-chain amino acid degradation (p = 10-12-10-5) were the most important topologically-highlighted dysregulated pathways contributing towards GM1T2 disease pathology. Quantitative metabolite set enrichment analysis revealed that pathological locations associated with these dysfunctions were in the order fibroblasts > Golgi apparatus > mitochondria > spleen ≈ skeletal muscle ≈ muscle in general. In conclusion, results acquired demonstrated marked metabolic imbalances and alterations to energy demand, which are consistent with GM1T2 disease pathogenesis mechanisms.
Collapse
Affiliation(s)
- Benita C. Percival
- Leicester School of Pharmacy, De Montfort University, The Gateway, Leicester LE1 9BH, UK;
| | - Yvonne L. Latour
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University, Nashville, TN 37232-0252, USA;
| | - Cynthia J. Tifft
- Deputy Clinical Director, National Human Genome Research Institute, Director, National Institutes of Health, Bethesda, MD 20892-1205, USA;
| | - Martin Grootveld
- Leicester School of Pharmacy, De Montfort University, The Gateway, Leicester LE1 9BH, UK;
| |
Collapse
|
7
|
Caciotti A, Cellai L, Tonin R, Mei D, Procopio E, Di Rocco M, Andaloro A, Antuzzi D, Rampazzo A, Rigoldi M, Forni G, la Marca G, Guerrini R, Morrone A. Morquio B disease: From pathophysiology towards diagnosis. Mol Genet Metab 2021; 132:180-188. [PMID: 33558080 DOI: 10.1016/j.ymgme.2021.01.008] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/12/2020] [Revised: 01/20/2021] [Accepted: 01/20/2021] [Indexed: 11/25/2022]
Abstract
Morquio B disease is an attenuated phenotype within the spectrum of beta galactosidase (GLB1) deficiencies. It is characterised by dysostosis multiplex, ligament laxity, mildly coarse facies and heart valve defects due to keratan sulphate accumulation, predominantly in the cartilage. Morquio B patients have normal neurological development, setting them apart from those with the more severe GM1 gangliosidosis. Morquio B disease, with an incidence of 1:250.000 to 1:1.000.000 live births, is very rare. Here we report the clinical-biochemical data of nine patients. High amounts of keratan sulfate were detected using LC-MS/MS in the patients' urinary samples, while electrophoresis, the standard procedure of qualitative glycosaminoglycans analysis, failed to identify this metabolite in any of the patients' samples. We performed molecular analyses at gene, gene expression and protein expression levels, for both isoforms of the GLB1 gene, lysosomal GLB1, and the cell-surface expressed Elastin Binding Protein. We characterised three novel GLB1 mutations [c.75 + 2 T > G, c.575A > G (p.Tyr192Cys) and c.2030 T > G (p.Val677Gly)] identified in three heterozygous patients. We also set up a copy number variation assay by quantitative PCR to evaluate the presence of deletions/ insertions in the GLB1 gene. We propose a diagnostic plan, setting out the specific clinical- biochemical and molecular features of Morquio B, in order to avoid misdiagnoses and improve patients' management.
Collapse
Affiliation(s)
- Anna Caciotti
- Molecular and Cell Biology Laboratory, Paediatric Neurology Unit and Laboratories, Neuroscience Department, A. Meyer Children's Hospital, Florence, Italy
| | - Lucrezia Cellai
- Molecular and Cell Biology Laboratory, Paediatric Neurology Unit and Laboratories, Neuroscience Department, A. Meyer Children's Hospital, Florence, Italy
| | - Rodolfo Tonin
- Molecular and Cell Biology Laboratory, Paediatric Neurology Unit and Laboratories, Neuroscience Department, A. Meyer Children's Hospital, Florence, Italy
| | - Davide Mei
- Neurogenetics, Paediatric Neurology Unit and Laboratories, Neuroscience Department, A. Meyer Children's Hospital, Florence, Italy
| | - Elena Procopio
- Metabolic and Muscular Unit, A. Meyer Children's Hospital, Florence, Italy
| | - Maja Di Rocco
- Unit of Rare Diseases, Dept of Pediatrics, IRCCS G. Gaslini, Genoa, Italy
| | - Antonio Andaloro
- Unit of Rare Diseases, Dept of Pediatrics, IRCCS G. Gaslini, Genoa, Italy
| | - Daniela Antuzzi
- Pediatric Clinic, Catholic University of "Sacro Cuore", Policlinico "Gemelli", Rome, Italy
| | | | - Miriam Rigoldi
- Mario Negri Institute for Pharmacological Research, IRCCS, Clinical Research Center for Rare Diseases "Aldo e Cele Daccò", Bergamo, Italy
| | - Giulia Forni
- Newborn Screening, Biochemistry and Pharmacology Laboratory, A. Meyer Children's Hospital, Florence, Italy
| | - Giancarlo la Marca
- Newborn Screening, Biochemistry and Pharmacology Laboratory, A. Meyer Children's Hospital, Florence, Italy; Department of Experimental and Clinical Biomedical Sciences, University of Florence, Italy
| | - Renzo Guerrini
- Molecular and Cell Biology Laboratory, Paediatric Neurology Unit and Laboratories, Neuroscience Department, A. Meyer Children's Hospital, Florence, Italy; Department of Neurosciences, Psychology, Pharmacology and Child Health, University of Florence, Italy
| | - Amelia Morrone
- Molecular and Cell Biology Laboratory, Paediatric Neurology Unit and Laboratories, Neuroscience Department, A. Meyer Children's Hospital, Florence, Italy; Department of Neurosciences, Psychology, Pharmacology and Child Health, University of Florence, Italy.
| |
Collapse
|
8
|
Lang FM, Korner P, Harnett M, Karunakara A, Tifft CJ. The natural history of Type 1 infantile GM1 gangliosidosis: A literature-based meta-analysis. Mol Genet Metab 2020; 129:228-235. [PMID: 31937438 PMCID: PMC7093236 DOI: 10.1016/j.ymgme.2019.12.012] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/25/2019] [Revised: 12/26/2019] [Accepted: 12/29/2019] [Indexed: 10/25/2022]
Abstract
INTRODUCTION Type 1 GM1 gangliosidosis is an ultra-rare, rapidly fatal lysosomal storage disorder, with life expectancy of <3 years of age. To date, only one prospective natural history study of limited size has been reported. Thus, there is a need for additional research to provide a better understanding of the progression of this disease. We have leveraged the past two decades of medical literature to conduct the first comprehensive retrospective study characterizing the natural history of Type 1 GM1 gangliosidosis. OBJECTIVES The objectives of this study were to establish a large sample of patients from the literature in order to identify: 1) clinically distinguishing factors between Type 1 and Type 2 GM1 gangliosidosis, 2) age at first symptom onset, first hospital admission, diagnosis, and death, 3) time to onset of common clinical findings, and 4) timing of developmental milestone loss. METHODS PubMed was searched with the keyword "GM1 Gangliosidosis" and for articles from the year 2000 onwards. A preliminary review of these results was conducted to establish subtype classification criteria for inclusion of only Type 1 patients, resulting in 44 articles being selected to generate the literature dataset of 154 Type 1 GM1 gangliosidosis patients. Key clinical events of these patient cases were recorded from the articles. RESULTS Comprehensive subtyping criteria for Type 1 GM1 gangliosidosis were created, and clinical events, including onset, diagnosis, death, and symptomology, were mapped over time. In this dataset, average age of diagnosis was 8.7 months, and average age of death was 18.9 months. DISCUSSION This analysis demonstrates the predictable clinical course of this disease, as almost all patients experienced significant multi-organ system dysfunction and neurodevelopmental regression, particularly in the 6- to 18-month age range. Patients were diagnosed at a late age relative to disease progression, indicating the need for improved public awareness and screening. CONCLUSION This study highlights the significant burden of illness in this disease and provides critical natural history data to drive earlier diagnosis, inform clinical trial design, and facilitate family counseling.
Collapse
Affiliation(s)
- Frederick M Lang
- Axovant Sciences, a subsidiary of Axovant Gene Therapies (Axovant), United States of America
| | - Paul Korner
- Axovant Sciences, a subsidiary of Axovant Gene Therapies (Axovant), United States of America
| | - Mark Harnett
- Axovant Sciences, a subsidiary of Axovant Gene Therapies (Axovant), United States of America
| | - Ajith Karunakara
- Axovant Sciences, a subsidiary of Axovant Gene Therapies (Axovant), United States of America
| | - Cynthia J Tifft
- Office of the Clinical Director & Medical Genetics Branch, National Human Genome Research Institute, National Institutes of Health (NHGRI), United States of America.
| |
Collapse
|
9
|
Arash-Kaps L, Komlosi K, Seegräber M, Diederich S, Paschke E, Amraoui Y, Beblo S, Dieckmann A, Smitka M, Hennermann JB. The Clinical and Molecular Spectrum of GM1 Gangliosidosis. J Pediatr 2019; 215:152-157.e3. [PMID: 31761138 DOI: 10.1016/j.jpeds.2019.08.016] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/15/2019] [Revised: 07/23/2019] [Accepted: 08/07/2019] [Indexed: 01/10/2023]
Abstract
OBJECTIVE To evaluate the clinical presentation of patients with GM1 gangliosidosis and to determine whether specific clinical or biochemical signs could lead to a prompt diagnosis. STUDY DESIGN We retrospectively analyzed clinical, biochemical, and genetic data of 22 patients with GM1 gangliosidosis from 5 metabolic centers in Germany and Austria. RESULTS Eight patients were classified as infantile, 11 as late-infantile, and 3 as juvenile form. Delay of diagnosis was 6 ± 2.6 months in the infantile, 2.6 ± 3.79 years in the late-infantile, and 14 ± 3.48 years in the juvenile form. Coarse facial features, cherry red spots, and visceromegaly occurred only in patients with the infantile form. Patients with the late-infantile and juvenile forms presented with variable neurologic symptoms. Seventeen patients presented with dystonia and 14 with dysphagia. Laboratory analysis revealed an increased ASAT concentration (13/20), chitotriosidase activity (12/15), and pathologic urinary oligosaccharides (10/19). Genotype analyses revealed 23 causative or likely causative mutations in 19 patients, 7 of them being novel variants. In the majority, a clear genotype-phenotype correlation was found. CONCLUSIONS Diagnosis of GM1 gangliosidosis often is delayed, especially in patients with milder forms of the disease. GM1 gangliosidosis should be considered in patients with progressive neurodegeneration and spastic-dystonic movement disorders, even in the absence of visceral symptoms or cherry red spots. ASAT serum concentrations and chitotriosidase activity may be of value in screening for GM1 gangliosidosis.
Collapse
Affiliation(s)
- Laila Arash-Kaps
- Villa Metabolica, Department of Pediatric and University Medical Center Mainz, Germany
| | - Katalin Komlosi
- Adolescent Medicine, and Institute of Human Genetics, University Medical Center Mainz, Germany
| | - Marlene Seegräber
- Villa Metabolica, Department of Pediatric and University Medical Center Mainz, Germany
| | - Stefan Diederich
- Adolescent Medicine, and Institute of Human Genetics, University Medical Center Mainz, Germany
| | | | - Yasmina Amraoui
- Villa Metabolica, Department of Pediatric and University Medical Center Mainz, Germany
| | - Skadi Beblo
- Department of Women and Child Health, Hospital for Children and Adolescents, Centre for Paediatric Research Leipzig (CPL), University Hospitals, University of Leipzig, Leipzig
| | - Andrea Dieckmann
- Center for Inborn Metabolic Disorders, Department of Neuropediatrics, Jena University Hospital, Jena
| | - Martin Smitka
- Neuropediatric Department, Carl Gustav Carus University Children's Hospital Dresden, Germany
| | - Julia B Hennermann
- Villa Metabolica, Department of Pediatric and University Medical Center Mainz, Germany
| |
Collapse
|
10
|
Deodato F, Procopio E, Rampazzo A, Taurisano R, Donati MA, Dionisi-Vici C, Caciotti A, Morrone A, Scarpa M. The treatment of juvenile/adult GM1-gangliosidosis with Miglustat may reverse disease progression. Metab Brain Dis 2017; 32:1529-1536. [PMID: 28577204 DOI: 10.1007/s11011-017-0044-y] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/04/2017] [Accepted: 05/24/2017] [Indexed: 10/19/2022]
Abstract
Juvenile and adult GM1-gangliosidosis are invariably characterized by progressive neurological deterioration. To date only symptomatic therapies are available. We report for the first time the positive results of Miglustat (OGT 918, N-butyl-deoxynojirimycin) treatment on three Italian GM1-gangliosidosis patients. The first two patients had a juvenile form (enzyme activity ≤5%, GLB1 genotype p.R201H/c.1068 + 1G > T; p.R201H/p.I51N), while the third patient had an adult form (enzyme activity about 7%, p.T329A/p.R442Q). Treatment with Miglustat at the dose of 600 mg/day was started at the age of 10, 17 and 28 years; age at last evaluation was 21, 20 and 38 respectively. Response to treatment was evaluated using neurological examinations in all three patients every 4-6 months, the assessment of Movement Disorder-Childhood Rating Scale (MD-CRS) in the second patient, and the 6-Minute Walking Test (6-MWT) in the third patient. The baseline neurological status was severely impaired, with loss of autonomous ambulation and speech in the first two patients, and gait and language difficulties in the third patient. All three patients showed gradual improvement while being treated; both juvenile patients regained the ability to walk without assistance for few meters, and increased alertness and vocalization. The MD-CRS class score in the second patient decreased from 4 to 2. The third patient improved in movement and speech control, the distance covered during the 6-MWT increased from 338 to 475 m. These results suggest that Miglustat may help slow down or reverse the disease progression in juvenile/adult GM1-gangliosidosis.
Collapse
Affiliation(s)
- Federica Deodato
- Division of Metabolic Disease, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - Elena Procopio
- Metabolic and Neuromuscular Unit, Neuroscience Department, A. Meyer Children's Hospital, Florence, Italy
| | - Angelica Rampazzo
- Department of Pediatrics, University Children's Hospital, Padua, Italy
| | - Roberta Taurisano
- Division of Metabolic Disease, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - Maria Alice Donati
- Metabolic and Neuromuscular Unit, Neuroscience Department, A. Meyer Children's Hospital, Florence, Italy
| | - Carlo Dionisi-Vici
- Division of Metabolic Disease, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - Anna Caciotti
- Molecular and Cell Biology Laboratory, Pediatric Neurology Unit and Laboratories, Neuroscience Department, A. Meyer Children's Hospital, Florence, Italy.
| | - Amelia Morrone
- Molecular and Cell Biology Laboratory, Pediatric Neurology Unit and Laboratories, Neuroscience Department, A. Meyer Children's Hospital, Florence, Italy
- Department of Neurosciences, Psychology, Pharmacology and Child Health, University of Florence, Florence, Italy
| | - Maurizio Scarpa
- Department of Pediatrics, University Children's Hospital, Padua, Italy
- Center for Rare Diseases, HELIOS Horst Schmidt Klinik, Wiesbaden, DE, Germany
| |
Collapse
|
11
|
Karimzadeh P, Naderi S, Modarresi F, Dastsooz H, Nemati H, Farokhashtiani T, Shamsian BS, Inaloo S, Faghihi MA. Case reports of juvenile GM1 gangliosidosisis type II caused by mutation in GLB1 gene. BMC MEDICAL GENETICS 2017; 18:73. [PMID: 28716012 PMCID: PMC5513107 DOI: 10.1186/s12881-017-0417-4] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/03/2016] [Accepted: 05/06/2017] [Indexed: 01/02/2023]
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.
Collapse
Affiliation(s)
- Parvaneh Karimzadeh
- Pediatric Neurology Department, Pediatric Neurology Research Center, Shahid Beheshti University of Medical Sciences (SBMU), Tehran, Iran
| | - Samaneh Naderi
- Diagnostic Laboratory Sciences and Technology Research Center, School of Paramedical Sciences, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Farzaneh Modarresi
- Center for Therapeutic Innovation, Department of Psychiatry and Behavioral Sciences, University of Miami Miller School of Medicine, Miami, USA
| | - Hassan Dastsooz
- Comprehensive Medical Genetic Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Hamid Nemati
- Shiraz Neuroscience Research Center, Shiraz University of Medical Sciences, Shiraz, Iran.
| | - Tayebeh Farokhashtiani
- Pediatric Neurology Department, Pediatric Neurology Research Center, Shahid Beheshti University of Medical Sciences (SBMU), Tehran, Iran
| | - Bibi Shahin Shamsian
- Pediatric Congenital Hematologic Disorders Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Soroor Inaloo
- Neonatal Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Mohammad Ali Faghihi
- Center for Therapeutic Innovation, Department of Psychiatry and Behavioral Sciences, University of Miami Miller School of Medicine, Miami, USA.
| |
Collapse
|
12
|
Khan SA, Peracha H, Ballhausen D, Wiesbauer A, Rohrbach M, Gautschi M, Mason RW, Giugliani R, Suzuki Y, Orii KE, Orii T, Tomatsu S. Epidemiology of mucopolysaccharidoses. Mol Genet Metab 2017; 121:227-240. [PMID: 28595941 PMCID: PMC5653283 DOI: 10.1016/j.ymgme.2017.05.016] [Citation(s) in RCA: 158] [Impact Index Per Article: 22.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/21/2017] [Revised: 05/22/2017] [Accepted: 05/22/2017] [Indexed: 12/15/2022]
Abstract
The aim of this study was to obtain data about the epidemiology of the different types of mucopolysaccharidoses in Japan and Switzerland and to compare with similar data from other countries. Data for Japan was collected between 1982 and 2009, and 467 cases with MPS were identified. The combined birth prevalence was 1.53 per 100,000 live births. The highest birth prevalence was 0.84 for MPS II, accounting for 55% of all MPS. MPS I, III, and IV accounted for 15, 16, and 10%, respectively. MPS VI and VII were more rare and accounted for 1.7 and 1.3%, respectively. A retrospective epidemiological data collection was performed in Switzerland between 1975 and 2008 (34years), and 41 living MPS patients were identified. The combined birth prevalence was 1.56 per 100,000 live births. The highest birth prevalence was 0.46 for MPS II, accounting for 29% of all MPS. MPS I, III, and IV accounted for 12, 24, and 24%, respectively. As seen in the Japanese population, MPS VI and VII were more rare and accounted for 7.3 and 2.4%, respectively. The high birth prevalence of MPS II in Japan was comparable to that seen in other East Asian countries where this MPS accounted for approximately 50% of all forms of MPS. Birth prevalence was also similar in some European countries (Germany, Northern Ireland, Portugal and the Netherlands) although the prevalence of other forms of MPS is also reported to be higher in these countries. Birth prevalence of MPS II in Switzerland and other European countries is comparatively lower. The birth prevalence of MPS III and IV in Switzerland is higher than in Japan but comparable to that in most other European countries. Moreover, the birth prevalence of MPS VI and VII was very low in both, Switzerland and Japan. Overall, the frequency of MPS varies for each population due to differences in ethnic backgrounds and/or founder effects that affect the birth prevalence of each type of MPS, as seen for other rare genetic diseases. Methods for identification of MPS patients are not uniform across all countries, and consequently, if patients are not identified, recorded prevalence rates will be aberrantly low.
Collapse
Affiliation(s)
- Shaukat A Khan
- Nemours/Alfred I. duPont Hospital for Children, Wilmington, DE, United States
| | - Hira Peracha
- Nemours/Alfred I. duPont Hospital for Children, Wilmington, DE, United States
| | - Diana Ballhausen
- Centre for Molecular Diseases, Service for Genetic Medicine, University Hospital Lausanne, Switzerland
| | - Alfred Wiesbauer
- Institute of Social and Preventive Medicine, University of Bern, Switzerland
| | - Marianne Rohrbach
- Division of Metabolism and Children's Research Centre (CRC), University Children's Hospital, Zurich, Switzerland
| | - Matthias Gautschi
- Division of Endocrinology, Diabetology and Metabolism, University Children's Hospital, University Institute of Clinical Chemistry, Inselspital, University of Bern, Bern, Switzerland
| | - Robert W Mason
- Nemours/Alfred I. duPont Hospital for Children, Wilmington, DE, United States
| | - Roberto Giugliani
- Medical Genetics Service, HCPA, Dep. Genetics, UFRGS, and INAGEMP, Porto Alegre, Brazil
| | | | - Kenji E Orii
- Department of Pediatrics, Gifu University, Gifu, Japan
| | - Tadao Orii
- Department of Pediatrics, Gifu University, Gifu, Japan
| | - Shunji Tomatsu
- Nemours/Alfred I. duPont Hospital for Children, Wilmington, DE, United States; Department of Pediatrics, Gifu University, Gifu, Japan; Department of Pediatrics, Thomas Jefferson University, Philadelphia, PA, United States.
| |
Collapse
|
13
|
Bidchol AM, Dalal A, Trivedi R, Shukla A, Nampoothiri S, Sankar VH, Danda S, Gupta N, Kabra M, Hebbar SA, Bhat RY, Matta D, Ekbote AV, Puri RD, Phadke SR, Gowrishankar K, Aggarwal S, Ranganath P, Sharda S, Kamate M, Datar CA, Bhat K, Kamath N, Shah H, Krishna S, Gopinath PM, Verma IC, Nagarajaram HA, Satyamoorthy K, Girisha KM. Recurrent and novel GLB1 mutations in India. Gene 2015; 567:173-81. [PMID: 25936995 DOI: 10.1016/j.gene.2015.04.078] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2015] [Revised: 04/27/2015] [Accepted: 04/28/2015] [Indexed: 02/08/2023]
Abstract
GM1 gangliosidosis is a lysosomal storage disorder caused by mutations in the GLB1 gene, leading to the deficiency of the enzyme β-d-galactosidase. In this study, we report molecular findings in 50 Asian Indian families with GM1 gangliosidosis. We sequenced all the exons and flanking intronic sequences of GLB1 gene. We identified 33 different mutations (20 novel and 13 previously reported). The novel mutations include 12 missense (p.M1?, p.E129Q, p.G134R, p.L236P, p.G262E, p.L297F, p.Y331C, p.G414V, p.K493N, p.L514P, p.P597L, p.T600I), four splicing (c.246-2A>G, c.397-2A>G, c.552+1G>T, c.956-2A>G), three indels (p.R22Qfs*8, p.L24Cfs*47, p.I489Qfs*4) and one nonsense mutation (p.Q452*). Most common mutations identified in this study were c.75+2InsT (14%) and p.L337P (10%). Known mutations accounted for 67% of allele frequency in our cohort of patients, suggesting that these mutations in GLB1 are recurrent across different populations. Twenty three mutations were localized in the TIM barrel domain, β-domain 1 and β-domain 2. In silico sequence and structure analysis of GLB1 reveal that all the novel mutations affect the function and structure of the protein. We hereby report on the largest series of patients with GM1 gangliosidosis and the first from India.
Collapse
Affiliation(s)
- Abdul Mueed Bidchol
- Department of Medical Genetics, Kasturba Medical College, Manipal University, Manipal, Karnataka, India
| | - Ashwin Dalal
- Diagnostics Division, Centre for DNA Fingerprinting and Diagnostics, Hyderabad, Telangana, India
| | - Rakesh Trivedi
- Laboratory of Computational Biology & Bioinformatics Facility, Centre for DNA Fingerprinting and Diagnostics, Hyderabad, Telangana, India; Graduate Studies, Manipal University, Manipal, Karnataka, India
| | - Anju Shukla
- Department of Medical Genetics, Kasturba Medical College, Manipal University, Manipal, Karnataka, India
| | - Sheela Nampoothiri
- Department of Pediatric Genetics, Amrita Institute of Medical Sciences and Research Center, Kochi, AIMS Ponekkara, Kerala, India
| | - V H Sankar
- Genetic Clinic, Department of Pediatrics, SAT Hospital, Government Medical College, Thiruvananthapuram, Kerala, India
| | - Sumita Danda
- Department of Clinical Genetics, Christian Medical College and Hospital, Vellore, Tamil Nadu, India
| | - Neerja Gupta
- Genetics Unit, Department of Pediatrics, All India Institute of Medical Sciences, New Delhi, India
| | - Madhulika Kabra
- Genetics Unit, Department of Pediatrics, All India Institute of Medical Sciences, New Delhi, India
| | - Shrikiran A Hebbar
- Department of Pediatrics, Kasturba Medical College, Manipal University, Manipal, Karnataka, India
| | - Ramesh Y Bhat
- Department of Pediatrics, Kasturba Medical College, Manipal University, Manipal, Karnataka, India
| | - Divya Matta
- Diagnostics Division, Centre for DNA Fingerprinting and Diagnostics, Hyderabad, Telangana, India
| | - Alka V Ekbote
- Department of Clinical Genetics, Christian Medical College and Hospital, Vellore, Tamil Nadu, India
| | - Ratna Dua Puri
- Center of Medical Genetics, Sir Ganga Ram Hospital, New Delhi, India
| | - Shubha R Phadke
- Department of Medical Genetics, Sanjay Gandhi Postgraduate Institute of Medical Sciences, Lucknow, Uttar Pradesh, India
| | - Kalpana Gowrishankar
- Department of Medical Genetics, Childs Trust Medical Research Foundation, Kanchi Kamakoti Childs Trust Hospital, Chennai, Tamil Nadu, India
| | - Shagun Aggarwal
- Diagnostics Division, Centre for DNA Fingerprinting and Diagnostics, Hyderabad, Telangana, India; Department of Medical Genetics, Nizam's Institute of Medical Sciences, Hyderabad, Telangana, India
| | - Prajnya Ranganath
- Diagnostics Division, Centre for DNA Fingerprinting and Diagnostics, Hyderabad, Telangana, India; Department of Medical Genetics, Nizam's Institute of Medical Sciences, Hyderabad, Telangana, India
| | - Sheetal Sharda
- Post Graduate Institute of Medical Education and Research, Chandigarh, Punjab, India
| | - Mahesh Kamate
- Pediatric Neurology, KLE University's J N Medical College, Belgaum, Karnataka, India
| | - Chaitanya A Datar
- Rare Genetic Disorder Clinic, Sahyadri Hospital, Pune, Maharashtra, India
| | - Kamalakshi Bhat
- Department of Pediatrics, Kasturba Medical College, Mangalore, Manipal University, Karnataka, India
| | - Nutan Kamath
- Department of Pediatrics, Kasturba Medical College, Mangalore, Manipal University, Karnataka, India
| | - Hitesh Shah
- Pediatric Orthopedics Services, Department of Orthopedics, Kasturba Medical College, Manipal, Manipal University, Karnataka, India
| | - Shuba Krishna
- Strand Life Sciences Pvt Ltd, Bengaluru, Karnataka, India
| | - Puthiya Mundyat Gopinath
- Division of Biotechnology, School of Life Sciences, Manipal University, Manipal, Karnataka, India
| | - Ishwar C Verma
- Center of Medical Genetics, Sir Ganga Ram Hospital, New Delhi, India
| | - H A Nagarajaram
- Laboratory of Computational Biology & Bioinformatics Facility, Centre for DNA Fingerprinting and Diagnostics, Hyderabad, Telangana, India
| | - Kapaettu Satyamoorthy
- Division of Biotechnology, School of Life Sciences, Manipal University, Manipal, Karnataka, India
| | - Katta Mohan Girisha
- Department of Medical Genetics, Kasturba Medical College, Manipal University, Manipal, Karnataka, India.
| |
Collapse
|
14
|
Morrone A, Caciotti A, Atwood R, Davidson K, Du C, Francis-Lyon P, Harmatz P, Mealiffe M, Mooney S, Oron TR, Ryles A, Zawadzki KA, Miller N. Morquio A syndrome-associated mutations: a review of alterations in the GALNS gene and a new locus-specific database. Hum Mutat 2014; 35:1271-9. [PMID: 25137622 PMCID: PMC4238747 DOI: 10.1002/humu.22635] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2014] [Accepted: 08/06/2014] [Indexed: 11/09/2022]
Abstract
Morquio A syndrome (mucopolysaccharidosis IVA) is an autosomal recessive disorder that results from deficient activity of the enzyme N-acetylgalactosamine-6-sulfatase (GALNS) due to alterations in the GALNS gene, which causes major skeletal and connective tissue abnormalities and effects on multiple organ systems. The GALNS alterations associated with Morquio A are numerous and heterogeneous, and new alterations are continuously identified. To aid detection and interpretation of GALNS alterations, from previously published research, we provide a comprehensive and up-to-date listing of 277 unique GALNS alterations associated with Morquio A identified from 1,091 published GALNS alleles. In agreement with previous findings, most reported GALNS alterations are missense changes and even the most frequent alterations are relatively uncommon. We found that 48% of patients are assessed as homozygous for a GALNS alteration, 39% are assessed as heterozygous for two identified GALNS alterations, and in 13% of patients only one GALNS alteration is detected. We report here the creation of a locus-specific database for the GALNS gene (http://galns.mutdb.org/) that catalogs all reported alterations in GALNS to date. We highlight the challenges both in alteration detection and genotype-phenotype interpretation caused in part by the heterogeneity of GALNS alterations and provide recommendations for molecular testing of GALNS.
Collapse
Affiliation(s)
- Amelia Morrone
- Molecular and Cell Biology Laboratory, Pediatric Neurology Unit and Laboratories, Meyer Children's Hospital, Florence, Italy; Department of Neurosciences, Psychology, Pharmacology and Child Health, University of Florence, Florence, Italy
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
15
|
Morrone A, Tylee KL, Al-Sayed M, Brusius-Facchin AC, Caciotti A, Church HJ, Coll MJ, Davidson K, Fietz MJ, Gort L, Hegde M, Kubaski F, Lacerda L, Laranjeira F, Leistner-Segal S, Mooney S, Pajares S, Pollard L, Ribeiro I, Wang RY, Miller N. Molecular testing of 163 patients with Morquio A (Mucopolysaccharidosis IVA) identifies 39 novel GALNS mutations. Mol Genet Metab 2014; 112:160-70. [PMID: 24726177 PMCID: PMC4203673 DOI: 10.1016/j.ymgme.2014.03.004] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/18/2014] [Revised: 03/11/2014] [Accepted: 03/12/2014] [Indexed: 01/09/2023]
Abstract
Morquio A (Mucopolysaccharidosis IVA; MPS IVA) is an autosomal recessive lysosomal storage disorder caused by partial or total deficiency of the enzyme galactosamine-6-sulfate sulfatase (GALNS; also known as N-acetylgalactosamine-6-sulfate sulfatase) encoded by the GALNS gene. Patients who inherit two mutated GALNS gene alleles have a decreased ability to degrade the glycosaminoglycans (GAGs) keratan sulfate and chondroitin 6-sulfate, thereby causing GAG accumulation within lysosomes and consequently pleiotropic disease. GALNS mutations occur throughout the gene and many mutations are identified only in single patients or families, causing difficulties both in mutation detection and interpretation. In this study, molecular analysis of 163 patients with Morquio A identified 99 unique mutations in the GALNS gene believed to negatively impact GALNS protein function, of which 39 are previously unpublished, together with 26 single-nucleotide polymorphisms. Recommendations for the molecular testing of patients, clear reporting of sequence findings, and interpretation of sequencing data are provided.
Collapse
Affiliation(s)
- A Morrone
- Molecular and Cell Biology Laboratory, Pediatric Neurology Unit and Laboratories, Meyer Children's Hospital, Florence, Italy; Department of Neurosciences, Psychology, Pharmacology and Child Health, University of Florence, Florence Italy
| | - K L Tylee
- Willink Biochemical Genetics, Central Manchester University Hospitals NHS Foundation Trust, Saint Mary's Hospital Oxford Road, Manchester, UK
| | - M Al-Sayed
- Department of Medical Genetics, King Faisal Specialist Hospital and Research Center, Riyadh, Saudi Arabia
| | - A C Brusius-Facchin
- Laboratório de Genética Molecular, Serviço de Genética Médica, Hospital de Clínicas de Porto Alegre (HCPA), Porto Alegre, Brazil
| | - A Caciotti
- Molecular and Cell Biology Laboratory, Pediatric Neurology Unit and Laboratories, Meyer Children's Hospital, Florence, Italy
| | - H J Church
- Willink Biochemical Genetics, Central Manchester University Hospitals NHS Foundation Trust, Saint Mary's Hospital Oxford Road, Manchester, UK
| | - M J Coll
- Sección de Errores Congénitos del Metabolismo-IBC, Servicio de Bioquímica y Genética Molecular, Hospital Clínic, CIBERER, IDIBAPS, Barcelona, Spain
| | - K Davidson
- BioMarin Pharmaceutical Inc., Novato, CA, USA
| | - M J Fietz
- SA Pathology, Women's and Children's Hospital, North Adelaide, SA, Australia
| | - L Gort
- Sección de Errores Congénitos del Metabolismo-IBC, Servicio de Bioquímica y Genética Molecular, Hospital Clínic, CIBERER, IDIBAPS, Barcelona, Spain
| | - M Hegde
- Emory Genetics Laboratory, Emory University School of Medicine, Atlanta, GA, USA
| | - F Kubaski
- Laboratório de Genética Molecular, Serviço de Genética Médica, Hospital de Clínicas de Porto Alegre (HCPA), Porto Alegre, Brazil
| | - L Lacerda
- Unidade de Bioquímica Genética, Centro de Genética Médica Jacinto Magalhães (CGMJM) do Centro Hospitalar do Porto (CHP), Porto, Portugal
| | - F Laranjeira
- Unidade de Bioquímica Genética, Centro de Genética Médica Jacinto Magalhães (CGMJM) do Centro Hospitalar do Porto (CHP), Porto, Portugal
| | - S Leistner-Segal
- Laboratório de Genética Molecular, Serviço de Genética Médica, Hospital de Clínicas de Porto Alegre (HCPA), Porto Alegre, Brazil
| | - S Mooney
- The Buck Institute for Research on Aging, Novato, CA, USA
| | - S Pajares
- Sección de Errores Congénitos del Metabolismo-IBC, Servicio de Bioquímica y Genética Molecular, Hospital Clínic, CIBERER, IDIBAPS, Barcelona, Spain
| | - L Pollard
- Biochemical Genetics Laboratory, Greenwood Genetic Center, Greenwood, SC, USA
| | - I Ribeiro
- Unidade de Bioquímica Genética, Centro de Genética Médica Jacinto Magalhães (CGMJM) do Centro Hospitalar do Porto (CHP), Porto, Portugal
| | - R Y Wang
- Children's Hospital of Orange County, Orange, CA, USA
| | - N Miller
- BioMarin Pharmaceutical Inc., Novato, CA, USA.
| |
Collapse
|
16
|
Cooper DN, Krawczak M, Polychronakos C, Tyler-Smith C, Kehrer-Sawatzki H. Where genotype is not predictive of phenotype: towards an understanding of the molecular basis of reduced penetrance in human inherited disease. Hum Genet 2013; 132:1077-130. [PMID: 23820649 PMCID: PMC3778950 DOI: 10.1007/s00439-013-1331-2] [Citation(s) in RCA: 423] [Impact Index Per Article: 38.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2013] [Accepted: 06/15/2013] [Indexed: 02/06/2023]
Abstract
Some individuals with a particular disease-causing mutation or genotype fail to express most if not all features of the disease in question, a phenomenon that is known as 'reduced (or incomplete) penetrance'. Reduced penetrance is not uncommon; indeed, there are many known examples of 'disease-causing mutations' that fail to cause disease in at least a proportion of the individuals who carry them. Reduced penetrance may therefore explain not only why genetic diseases are occasionally transmitted through unaffected parents, but also why healthy individuals can harbour quite large numbers of potentially disadvantageous variants in their genomes without suffering any obvious ill effects. Reduced penetrance can be a function of the specific mutation(s) involved or of allele dosage. It may also result from differential allelic expression, copy number variation or the modulating influence of additional genetic variants in cis or in trans. The penetrance of some pathogenic genotypes is known to be age- and/or sex-dependent. Variable penetrance may also reflect the action of unlinked modifier genes, epigenetic changes or environmental factors. At least in some cases, complete penetrance appears to require the presence of one or more genetic variants at other loci. In this review, we summarize the evidence for reduced penetrance being a widespread phenomenon in human genetics and explore some of the molecular mechanisms that may help to explain this enigmatic characteristic of human inherited disease.
Collapse
Affiliation(s)
- David N. Cooper
- Institute of Medical Genetics, School of Medicine, Cardiff University, Heath Park, Cardiff, CF14 4XN UK
| | - Michael Krawczak
- Institute of Medical Informatics and Statistics, Christian-Albrechts University, 24105 Kiel, Germany
| | | | - Chris Tyler-Smith
- The Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton, Cambridge, CB10 1SA UK
| | | |
Collapse
|
17
|
Abstract
Lysosomal storage disorders (LSDs) are a large group of more than 50 different inherited metabolic diseases which, in the great majority of cases, result from the defective function of specific lysosomal enzymes and, in cases, of non-enzymatic lysosomal proteins or non-lysosomal proteins involved in lysosomal biogenesis. The progressive lysosomal accumulation of undegraded metabolites results in generalised cell and tissue dysfunction, and, therefore, multi-systemic pathology. Storage may begin during early embryonic development, and the clinical presentation for LSDs can vary from an early and severe phenotype to late-onset mild disease. The diagnosis of most LSDs--after accurate clinical/paraclinical evaluation, including the analysis of some urinary metabolites--is based mainly on the detection of a specific enzymatic deficiency. In these cases, molecular genetic testing (MGT) can refine the enzymatic diagnosis. Once the genotype of an individual LSD patient has been ascertained, genetic counselling should include prediction of the possible phenotype and the identification of carriers in the family at risk. MGT is essential for the identification of genetic disorders resulting from non-enzymatic lysosomal protein defects and is complementary to biochemical genetic testing (BGT) in complex situations, such as in cases of enzymatic pseudodeficiencies. Prenatal diagnosis is performed on the most appropriate samples, which include fresh or cultured chorionic villus sampling or cultured amniotic fluid. The choice of the test--enzymatic and/or molecular--is based on the characteristics of the defect to be investigated. For prenatal MGT, the genotype of the family index case must be known. The availability of both tests, enzymatic and molecular, enormously increases the reliability of the entire prenatal diagnostic procedure. To conclude, BGT and MGT are mostly complementary for post- and prenatal diagnosis of LSDs. Whenever genotype/phenotype correlations are available, they can be helpful in predicting prognosis and in making decisions about therapy.
Collapse
Affiliation(s)
- Mirella Filocamo
- S.S.D. Lab. Diagnosi Pre-Postnatale Malattie Metaboliche, Dipartimento di Neuroscienze, IRCCS G. Gaslini, Largo G. Gaslini 5, Genova, Italy.
| | | |
Collapse
|
18
|
Ferri L, Guido C, la Marca G, Malvagia S, Cavicchi C, Fiumara A, Barone R, Parini R, Antuzzi D, Feliciani C, Zampetti A, Manna R, Giglio S, Della Valle CM, Wu X, Valenzano KJ, Benjamin R, Donati MA, Guerrini R, Genuardi M, Morrone A. Fabry disease: polymorphic haplotypes and a novel missense mutation in the GLA gene. Clin Genet 2011; 81:224-33. [PMID: 21517827 DOI: 10.1111/j.1399-0004.2011.01689.x] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Fabry disease: polymorphic haplotypes and a novel missense mutation in the GLA gene. Fabry disease (FD) is an X-linked lysosomal storage disorder with a heterogeneous spectrum of clinical manifestations that are caused by the deficiency of α-galactosidase A (α-Gal-A) activity. Although useful for diagnosis in males, enzyme activity is not a reliable biochemical marker in heterozygous females due to random X-chromosome inactivation, thus rendering DNA sequencing of the α-Gal-A gene, alpha-galactosidase gene (GLA), the most reliable test for the confirmation of diagnosis in females. The spectrum of GLA mutations is highly heterogeneous. Many polymorphic GLA variants have been described, but it is unclear if haplotypes formed by combinations of such variants correlate with FD, thus complicating molecular diagnosis in females with normal α-Gal-A activity. We tested 67 female probands with clinical manifestations that may be associated with FD and 110 control males with normal α-Gal-A activity. Five different combinations of GLA polymorphic variants were identified in 14 of the 67 females, whereas clearcut pathogenetic alterations, p.Met51Ile and p.Met290Leu, were identified in two cases. The latter has not been reported so far, and both mutant forms were found to be responsive to the pharmacological chaperone deoxygalactonojirimycin (DGJ; migalastat hydrochloride). Analysis of the male control population, as well as male relatives of a suspected FD female proband, permitted the identification of seven different GLA gene haplotypes in strong linkage disequilibrium. The identification of haplotypes in control males provides evidence against their involvement in the development of FD phenotypic manifestations.
Collapse
Affiliation(s)
- L Ferri
- Department of Sciences for Woman and Child's Health, University of Florence, Florence, Italy
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
19
|
GM1 gangliosidosis and Morquio B disease: an update on genetic alterations and clinical findings. Biochim Biophys Acta Mol Basis Dis 2011; 1812:782-90. [PMID: 21497194 DOI: 10.1016/j.bbadis.2011.03.018] [Citation(s) in RCA: 94] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2010] [Revised: 03/18/2011] [Accepted: 03/30/2011] [Indexed: 11/20/2022]
Abstract
GM1 gangliosidosis and Morquio B syndrome, both arising from beta-galactosidase (GLB1) deficiency, are very rare lysosomal storage diseases with an incidence of about 1:100,000-1:200,000 live births worldwide. Here we report the beta-galactosidase gene (GLB1) mutation analysis of 21 unrelated GM1 gangliosidosis patients, and of 4 Morquio B patients, of whom two are brothers. Clinical features of the patients were collected and compared with those in literature. In silico analyses were performed by standard alignments tools and by an improved version of GLB1 three-dimensional models. The analysed cohort includes remarkable cases. One patient with GM1 gangliosidosis had a triple X syndrome. One patient with juvenile GM1 gangliosidosis was homozygous for a mutation previously identified in Morquio type B. A patient with infantile GM1 gangliosidosis carried a complex GLB1 allele harbouring two genetic variants leading to p.R68W and p.R109W amino acid changes, in trans with the known p.R148C mutation. Molecular analysis showed 27 mutations, 9 of which are new: 5 missense, 3 microdeletions and a nonsense mutation. We also identified four new genetic variants with a predicted polymorphic nature that was further investigated by in silico analyses. Three-dimensional structural analysis of GLB1 homology models including the new missense mutations and the p.R68W and p.R109W amino acid changes showed that all the amino acid replacements affected the resulting protein structures in different ways, from changes in polarity to folding alterations. Genetic and clinical associations led us to undertake a critical review of the classifications of late-onset GM1 gangliosidosis and Morquio B disease.
Collapse
|
20
|
Coutinho MF, Lacerda L, Macedo-Ribeiro S, Baptista E, Ribeiro H, Prata MJ, Alves S. Lysosomal multienzymatic complex-related diseases: a genetic study among Portuguese patients. Clin Genet 2011; 81:379-93. [PMID: 21214877 DOI: 10.1111/j.1399-0004.2011.01625.x] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
The functional activity of lysosomal enzymes sialidase, β-galactosidase and N-acetylaminogalacto-6-sulfate-sulfatase in the cell depends on their association in a multienzyme complex with cathepsin A. Mutations in any of the components of this complex result in functional deficiency thereby causing severe lysosomal storage disorders. Here, we report the molecular defects underlying sialidosis (mutations in sialidase; gene NEU1), galactosialidosis (mutations in cathepsin A; gene PPGB) and GM1 gangliosidosis (mutations in β-galactosidase; gene GLB1) in Portuguese patients. We performed molecular studies of the PPGB, NEU1 and GLB1 genes in biochemically diagnosed Portuguese patients. Gene expression was determined and the effect of each mutation predicted at protein levels. In the NEU1 gene, we found three novel missense mutations (p.P200L, p.D234N and p.Q282H) and one nonsense mutation (p.R341X). In the PPGB gene, we identified two missense mutations, one novel (p.G86V) and one already described (p.V104M), as well as two new deletions (c.230delC and c.991-992delT) that give rise to non-functional proteins. We also present the first molecular evidence of a causal missense mutation localized to the cathepsin A active site. Finally, in the GLB1 gene, we found six different mutations, all of them previously described (p.R59H, p.R201H, p.H281Y, p.W527X, c.1572-1577InsG and c.845-846delC). Seven novel mutations are reported here, contributing to our knowledge of the mutational spectrum of these diseases and to a better understanding of the genetics of the lysosomal multienzymatic complex. The results of this study will allow carrier detection in affected families and prenatal molecular diagnosis, leading to the improvement of genetic counseling.
Collapse
Affiliation(s)
- M F Coutinho
- Centro de Genética Médica Doutor Jacinto de Magalhães, INSA, I.P., Praça Pedro Nunes 88, Porto, Portugal.
| | | | | | | | | | | | | |
Collapse
|
21
|
Caciotti A, Donati MA, d'Azzo A, Salvioli R, Guerrini R, Zammarchi E, Morrone A. The potential action of galactose as a "chemical chaperone": increase of beta galactosidase activity in fibroblasts from an adult GM1-gangliosidosis patient. Eur J Paediatr Neurol 2009; 13:160-4. [PMID: 18571950 DOI: 10.1016/j.ejpn.2008.03.004] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/06/2007] [Revised: 03/26/2008] [Accepted: 03/29/2008] [Indexed: 11/28/2022]
Abstract
BACKGROUND The glycosphingolipid storage disorder GM1-gangliosidosis is a severe neurodegenerative condition for which no therapy is currently available. Protein misfolding in lysosomal defects may have the potential to be corrected by chemical chaperones: in vitro and clinical approaches are being investigated. AIMS We investigated the in vitro effect of galactose on some lysosomal hydrolases, and its in vitro efficacy as a chemical chaperone in GM1-gangliosidosis. METHODS Galactose was added to the culture medium of fibroblasts from patients, controls and transfected COS-1 cells. Enzyme assays of lysosomal hydrolases, beta galactosidase in particular, were performed. RESULTS Our data show that galactose alters selectively alpha and beta galactosidases. A significant increase (2,5 fold) in beta galactosidase activity occurred when galactose was added to the cultured fibroblasts of an adult patient. Chemical chaperone therapy requires the presence of residual enzyme activity. The adult patient here reported is heterozygous for the p.T329A mutation that showed no beta galactosidase activity, and for the p.R442Q mutation with residual enzyme activity. The p.R442Q mutation was therefore selected as a potential target for the galactose chaperone; after the addition of galactose, COS-1 cells transfected with this mutation showed an increase in beta galactosidase activity from 6.9% to 12% of control values. CONCLUSIONS These results suggest that galactose or its derivatives with potential chaperone properties could be used in the development of non-invasive therapies for GM1-gangliosidosis.
Collapse
Affiliation(s)
- Anna Caciotti
- Metabolic and Muscular Unit, Clinic of Pediatric Neurology, AOU Meyer, Florence, Italy
| | | | | | | | | | | | | |
Collapse
|
22
|
Brunetti-Pierri N, Scaglia F. GM1 gangliosidosis: review of clinical, molecular, and therapeutic aspects. Mol Genet Metab 2008; 94:391-396. [PMID: 18524657 DOI: 10.1016/j.ymgme.2008.04.012] [Citation(s) in RCA: 174] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/16/2008] [Revised: 04/12/2008] [Accepted: 04/12/2008] [Indexed: 10/22/2022]
Abstract
GM(1) gangliosidosis is a lysosomal storage disorder due to deficiency of the beta-galactosidase enzyme. This deficiency results in accumulation of GM(1) gangliosides and related glycoconjugates in the lysosomes leading to lysosomal swelling, cellular damage, and organ dysfunction. The disease is lethal in the infantile and juvenile forms. To date, up to 102 mutations distributed along the beta-galactosidase gene (GLB1) have been reported. This review gives an overview of the clinical and molecular findings in patients with GM(1) gangliosidosis. Furthermore, it describes therapeutic approaches which are currently under investigation in animal models of the disease.
Collapse
Affiliation(s)
- Nicola Brunetti-Pierri
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Fernando Scaglia
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA; Texas Children's Hospital, Houston, TX 77030, USA
| |
Collapse
|
23
|
Kreutzer R, Kreutzer M, Pröpsting MJ, Sewell AC, Leeb T, Naim HY, Baumgärtner W. Insights into post-translational processing of beta-galactosidase in an animal model resembling late infantile human G-gangliosidosis. J Cell Mol Med 2007; 12:1661-71. [PMID: 18088383 PMCID: PMC3918082 DOI: 10.1111/j.1582-4934.2007.00204.x] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022] Open
Abstract
GM1-gangliosidosis is a lysosomal storage disorder caused by a deficiency of ß-galactosidase activity. Human GM1-gangliosidosis has been classified into three forms according to the age of clinical onset and specific biochemical parameters. In the present study, a canine model for type II late infantile human GM1-gangliosidosis was investigated ‘in vitro’ in detail. For a better understanding of the molecular pathogenesis underlying GM1-gangliosidosis the study focused on the analysis of the molecular events and subsequent intracellular protein trafficking of β-galactosidase. In the canine model the genetic defect results in exclusion or inclusion of exon 15 in the mRNA transcripts and to translation of two mutant precursor proteins. Intracellular localization, processing and enzymatic activity of these mutant proteins were investigated. The obtained results suggested that the β-galactosidase C-terminus encoded by exons 15 and 16 is necessary for correct C-terminal proteolytic processing and enzyme activity but does not affect the correct routing to the lysosomes. Both mutant protein precursors are enzymatically inactive, but are transported to the lysosomes clearly indicating that the amino acid sequences encoded by exons 15 and 16 are necessary for correct folding and association with protective protein/cathepsin A, whereas the routing to the lysosomes is not influenced. Thus, the investigated canine model is an appropriate animal model for the human late infantile form and represents a versatile system to test gene therapeutic approaches for human and canine GM1-gangliosidosis.
Collapse
Affiliation(s)
- R Kreutzer
- Department of Pathology, University of Veterinary Medicine, Hannover, Germany.
| | | | | | | | | | | | | |
Collapse
|
24
|
Gort L, Santamaria R, Grinberg D, Vilageliu L, Chabás A. Identification of a novel pseudodeficiency allele in the GLB1 gene in a carrier of GM1 gangliosidosis. Clin Genet 2007; 72:109-11. [PMID: 17661814 DOI: 10.1111/j.1399-0004.2007.00843.x] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
Abstract
The term 'pseudodeficiency' is used in lysosomal storage diseases to denote the situation in which individuals show greatly reduced enzyme activity but remain clinically healthy. Pseudodeficiencies have been reported for several lysosomal hydrolases. GM1 gangliosidosis is a rare autosomal recessive lysosomal storage disorder caused by beta-galactosidase hydrolase deficiency as a result of mutations in the GLB1 gene. Until now, two variants altering the beta-galactosidase activity have been described, p.Arg521Cys and p.Ser532Gly. Here we report the new variant p.Arg595Trp in the GLB1 gene, which markedly reduces beta-galactosidase activity when expressed in COS-1 cells. The variant was identified in the healthy father of a girl with GM1 gangliosidosis. He was a heterozygous compound with p.Arg595Trp in trans with one of the disease-causing mutations identified in his daughter; in leukocytes and plasma he showed lower beta-galactosidase activity than that observed in GM1 gangliosidosis carriers. As this family originated from the Basque Country in the north of Spain, we decided to analyse individuals of Basque and non-Basque origin, finding the p.Arg595Trp allele in 3.2% of Basque and in 0.8% of non-Basque alleles. The detection of the presence of alterations resulting in pseudodeficient activity in leukocytes and plasma is important for the correct diagnosis of GM1 gangliosidosis.
Collapse
Affiliation(s)
- L Gort
- Institut de Bioquímica Clínica, Servei de Bioquimica i Genètica Molecular, Hospital Clínic, Barcelona, Spain
| | | | | | | | | |
Collapse
|
25
|
Santamaria R, Chabás A, Callahan JW, Grinberg D, Vilageliu L. Expression and characterization of 14 GLB1 mutant alleles found in GM1-gangliosidosis and Morquio B patients. J Lipid Res 2007; 48:2275-82. [PMID: 17664528 DOI: 10.1194/jlr.m700308-jlr200] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
GM1-gangliosidosis and Morquio B disease are lysosomal storage disorders caused by beta-galactosidase deficiency attributable to mutations in the GLB1 gene. On reaching the endosomal-lysosomal compartment, the beta-galactosidase protein associates with the protective protein/cathepsin A (PPCA) and neuraminidase proteins to form the lysosomal multienzyme complex (LMC). The correct interaction of these proteins in the complex is essential for their activity. More than 100 mutations have been described in GM1-gangliosidosis and Morquio B patients, but few have been further characterized. We expressed 12 mutations suspected to be pathogenic, one known polymorphic change (p.S532G), and a variant described as either a pathogenic or a polymorphic change (p.R521C). Ten of them had not been expressed before. The expression analysis confirmed the pathogenicity of the 12 mutations, whereas the relatively high activity of p.S532G is consistent with its definition as a polymorphism. The results for p.R521C suggest that this change is a low-penetrant disease-causing allele. Furthermore, the effect of these beta-galactosidase changes on the LMC was also studied by coimmunoprecipitations and Western blotting. The alteration of neuraminidase and PPCA patterns in several of the Western blotting analyses performed on patient protein extracts indicated that the LMC is affected in at least some GM1-gangliosidosis and Morquio B patients.
Collapse
Affiliation(s)
- Raül Santamaria
- Departament de Genètica, Universitat de Barcelona, Barcelona, Spain
| | | | | | | | | |
Collapse
|
26
|
Santamaria R, Blanco M, Chabás A, Grinberg D, Vilageliu L. Identification of 14 novel GLB1 mutations, including five deletions, in 19 patients with GM1 gangliosidosis from South America. Clin Genet 2007; 71:273-9. [PMID: 17309651 DOI: 10.1111/j.1399-0004.2007.00767.x] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
GM1 gangliosidosis is a lysosomal storage disorder caused by the absence or reduction of lysosomal beta-galactosidase activity because of mutations in the GLB1 gene. Three major clinical forms have been established: type I (infantile), type II (late infantile/juvenile) and type III (adult). A mutational analysis was performed in 19 patients with GM1 gangliosidosis from South America, mainly from Argentina. Two of them were of Gypsy origin. Main clinical findings of the patients are presented. All 38 mutant alleles were identified: of the 22 different mutations found, 14 mutations are described here for the first time. Among the novel mutations, five deletions were found. Four of them are relatively small (c.435_440delTCT, c.845_846delC, c.1131_1145del15 and c.1706_1707delC), while the other one is a deletion of 1529 nucleotides that includes exon 5 and is caused by an unequal crossover between intronic Alu sequences. All the described patients with GM1 gangliosidosis were affected by the infantile form, except for four unrelated patients classified as type II, III, and II/III (two cases). The two type II/III patients bore the previously described p.R201H mutation, while the adult patient bore the new p.L155R. The juvenile patient bore two novel mutations: p.S434L and p.G554E. The two Gypsy patients are homozygous for the p.R59H mutation as are all Gypsy patients previously genotyped.
Collapse
Affiliation(s)
- R Santamaria
- Departament de Genètica, Facultat de Biologia, Universitat de Barcelona, Barcelona, Spain
| | | | | | | | | |
Collapse
|
27
|
Georgiou T, Stylianidou G, Anastasiadou V, Caciotti A, Campos Y, Zammarchi E, Morrone A, D'azzo A, Drousiotou A. The Arg482His mutation in the beta-galactosidase gene is responsible for a high frequency of GM1 gangliosidosis carriers in a Cypriot village. ACTA ACUST UNITED AC 2006; 9:126-32. [PMID: 15943552 DOI: 10.1089/gte.2005.9.126] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
GM1 gangliosidosis is a lysosomal storage disorder caused by deficiency of beta-galactosidase. It is mainly characterized by progressive neurodegeneration, and in its most severe infantile form, it leads to death before the age of 4. The GLB1 gene gives rise to two alternatively spliced mRNAs that encode the beta-galactosidase and the elastin binding protein (EBP). The diagnosis of two patients with the infantile form of GM1 gangliosidosis and 11 carriers in a small mountainous village in Cyprus prompted us to carry out a study in order to establish the frequency of carriers in the village and identify the mutations involved. Carrier detection was initially based on the measurement of beta-galactosidase activity in leucocytes. Among 85 random samples from the village, 10 were classified as carriers. Sequencing of the GLB1 gene in a Cypriot patient identified the missense mutation c.1445G>A (p.Arg482His) in the homozygous state. Seven of the 10 carriers identified using the enzyme assay were found to carry the same mutation by NspI restriction enzyme analysis. The three individuals who were negative for the c.1445G>A had borderline enzyme results and were probably wrongly classified as carriers. The frequency of GM1 gangliosidosis carriers in this village is approximately 8% (1:12). Western blot analysis showed a marked decrease of the 64-kDa mature form of the enzyme protein and a similar reduction of the 67-kDa EBP. Our results indicate that the c.1445G>A mutation, which appears to be responsible for all GM1 gangliosidosis alleles in this Cypriot village, affects protein conformation.
Collapse
Affiliation(s)
- Theodoros Georgiou
- Department of Biochemical Genetics, Cyprus Institute of Neurology and Genetics, 1683 Nicosia, Cyprus
| | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Caciotti A, Donati MA, Bardelli T, d'Azzo A, Massai G, Luciani L, Zammarchi E, Morrone A. Primary and secondary elastin-binding protein defect leads to impaired elastogenesis in fibroblasts from GM1-gangliosidosis patients. THE AMERICAN JOURNAL OF PATHOLOGY 2006; 167:1689-98. [PMID: 16314480 PMCID: PMC1613190 DOI: 10.1016/s0002-9440(10)61251-5] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
G(M1)-gangliosidosis is a lysosomal storage disorder caused by acid beta-galactosidase deficiency. Aside from the lysosomal beta-galactosidase enzyme, the beta-galactosidase gene also encodes the elastin-binding protein (EBP), deficiency in which impairs elastogenesis. Using expression studies and Western blots of COS-1 cells, we identified and characterized four new and two known beta-galactosidase gene mutations detected in G(M1)-gangliosidosis patients with infantile, juvenile, or adult forms of disease. We then focused on impaired elastogenesis detected in fibroblasts from patients with infantile and juvenile disease. The juvenile patient showed connective-tissue abnormalities, unusual urinary keratan sulfate excretion, and an EBP reduction, despite mutations affecting only beta-galactosidase. Because galactosugar-bearing moieties may alter EBP function and impair elastogenesis, we assessed infantile and juvenile patients for the source of altered elastogenesis. We confirmed that the infantile patient's impaired elastogenesis arose from a primary EBP defect, according to molecular analysis. We examined the juvenile's fibroblasts by immunohistochemistry, addition of keratanase, soluble/insoluble elastin assay, and radiolabeling of tropoelastin. These experiments revealed that the juvenile's impaired elastogenesis likely arose from secondary EBP deficiency caused by keratan sulfate accumulation. Thus, impaired elastogenesis in G(M1)-gangliosidosis can arise from primary or secondary EBP defects in fibroblasts from infantile and juvenile patients, respectively.
Collapse
Affiliation(s)
- Anna Caciotti
- Department of Pediatrics, Meyer Hospital, Florence, Italy
| | | | | | | | | | | | | | | |
Collapse
|
29
|
Clain J, Lehmann-Che J, Girodon E, Lipecka J, Edelman A, Goossens M, Fanen P. A neutral variant involved in a complex CFTR allele contributes to a severe cystic fibrosis phenotype. Hum Genet 2005; 116:454-60. [PMID: 15744523 DOI: 10.1007/s00439-004-1246-z] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2004] [Accepted: 12/12/2004] [Indexed: 11/26/2022]
Abstract
In order to further elucidate the contribution of complex alleles to the wide phenotypic variability of cystic fibrosis (CF), we investigated the structure-function relationships of a severe CF-associated complex allele [p.S912L;p.G1244V]. To evaluate the contribution of each mutation to the phenotype, cystic fibrosis transmembrane conductance regulator (CFTR) mutants were expressed in HeLa cells and analysed for protein processing and Cl- channel activity. Both p.G1244V and [p.S912L;p.G1244V] mutants had normal protein processing but markedly decreased Cl- channel activity compared with wild-type. Notably, the double mutant displayed a dramatic decrease in Cl- channel activity compared with p.G1244V (P<0.001). p.S912L had normal protein processing and no detectable impact on CFTR function. In other respects, the p.S912L variation was identified in compound heterozygosity with p.R709X in a healthy fertile man. Together, these data strongly support the view that p.S912L in isolation should be considered as a neutral variant but one that might significantly impair CFTR function when inherited in cis with another CFTR mutation. Our data also further document the contribution of complex alleles to the wide phenotypic variability of CF. The results of functional studies of such complex alleles in other genetic diseases are discussed.
Collapse
Affiliation(s)
- Jérôme Clain
- Service de Biochimie et Génétique, Hôpital Henri Mondor, Institut National de la Santé et de la Recherche Médicale U.468, AP-HP, 94010, Créteil, France
| | | | | | | | | | | | | |
Collapse
|
30
|
Caciotti A, Donati MA, Boneh A, d'Azzo A, Federico A, Parini R, Antuzzi D, Bardelli T, Nosi D, Kimonis V, Zammarchi E, Morrone A. Role of ?-galactosidase and elastin binding protein in lysosomal and nonlysosomal complexes of patients with GM1-gangliosidosis. Hum Mutat 2005; 25:285-92. [PMID: 15714521 DOI: 10.1002/humu.20147] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
G(M1)-gangliosidosis is a lysosomal storage disorder caused by a deficiency of beta-galactosidase (GLB1). The GLB1 gene gives rise to the GLB1 lysosomal enzyme and to the elastin binding protein (EBP), involved in elastic fiber deposition. GLB1 forms a complex with protective protein cathepsin A (PPCA), alpha neuraminidase (NEU1), and galactosamine 6-sulphate sulfatase (GALNS) inside lysosomes, while EBP binds to PPCA and NEU1 on the cell surface. We investigated the function of the GLB1 and EBP mutated proteins by analyzing the clinical, genetic, and cellular data of 11 G(M1)-gangliosidosis patients. Their molecular analysis, followed by expression studies, lead to the identification of four new and 10 known GLB1 mutations. Some common amino acid substitutions [c.1445G>A (p.Arg482H), c.622C>T (p.Arg208His), c.175C>T (p.Arg59Cys) and c.176G>A (p.Arg59His)] were present in the GLB1 enzyme of several patients, all of Mediterranean origin, suggesting a common origin. Western blotting analyses against GLB1, EBP, and PPCA proteins showed that the identified mutations affect GLB1 enzyme activity and/or stability. The c.1445G>A (p.Arg482His), c.175C>T (p.Arg59Cys), c.733+2T>C, c.1736G>A (p.Gly579Asp), and c.1051C>T (p.Arg351X) GLB1 mutations, affect the stabilization of PPCA probably because they hamper the interaction between GLB1/EBP and PPCA within the multiprotein complex. The amount of EBP was normal, but the detection of impaired elastogenesis in such patients suggests an alteration in its function. We conclude that the presence of genetic lesions in both GLB1 and EBP coding region does not directly predict impaired elastogenesis and that elastic fiber assembly has to be evaluated specifically in each case. Nevertheless, the degree of EBP involvement may be linked to specific clinical findings.
Collapse
MESH Headings
- Adult
- Alternative Splicing
- Amino Acid Sequence
- Amino Acid Substitution
- Animals
- COS Cells
- Cathepsin A/chemistry
- Cells, Cultured/metabolism
- Chlorocebus aethiops
- Elastic Tissue/ultrastructure
- Female
- Fibroblasts/metabolism
- Gangliosidosis, GM1/classification
- Gangliosidosis, GM1/genetics
- Gangliosidosis, GM1/pathology
- Humans
- Infant
- Infant, Newborn
- Lysosomes/enzymology
- Male
- Molecular Sequence Data
- Multiprotein Complexes
- Mutation, Missense
- Phenotype
- Protein Binding
- Protein Folding
- Protein Interaction Mapping
- Receptors, Cell Surface/chemistry
- Receptors, Cell Surface/genetics
- Receptors, Cell Surface/physiology
- Sequence Alignment
- Sequence Homology, Amino Acid
- Species Specificity
- Transfection
- beta-Galactosidase/chemistry
- beta-Galactosidase/genetics
- beta-Galactosidase/physiology
Collapse
Affiliation(s)
- Anna Caciotti
- Department of Pediatrics, University of Florence, Meyer Hospital, Florence, Italy
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|