Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 58
Filter
1.
Mol Genet Metab ; 142(4): 108517, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38908075

ABSTRACT

GM2 gangliosidosis is a group of rare lysosomal storage disorders (LSDs) including Tay-Sachs disease (TSD) and Sandhoff disease (SD), caused by deficiency in activity of either ß-hexosaminidase A (HexA) or both ß-hexosaminidase A and ß-hexosaminidase B (HexB). Methods for screening and diagnosis of TSD and SD include measurement and comparison of the activity of these two enzymes. Here we report a novel method for duplex screening of dried blood spots (DBS) for TSD and SD by liquid chromatography-tandem mass spectrometry (LC-MS/MS). The method requires incubation of a single 3 mm DBS punch with the assay cocktail followed by the injection into the LC-MS/MS. The performance of the method was evaluated by comparing the confirmed TSD and SD patient DBS to random healthy newborn DBS which showed easy discrimination between the three cohorts. The method is multiplexable with other LSD MS/MS enzyme assays which is critical to the continued expansion of the NBS panels.


Subject(s)
Dried Blood Spot Testing , Neonatal Screening , Sandhoff Disease , Tandem Mass Spectrometry , Tay-Sachs Disease , Humans , Tay-Sachs Disease/diagnosis , Tay-Sachs Disease/blood , Tay-Sachs Disease/enzymology , Infant, Newborn , Tandem Mass Spectrometry/methods , Neonatal Screening/methods , Dried Blood Spot Testing/methods , Sandhoff Disease/diagnosis , Sandhoff Disease/blood , Chromatography, Liquid/methods , Enzyme Assays/methods , beta-Hexosaminidase alpha Chain/blood , Hexosaminidase A/blood , Hexosaminidase B/blood
2.
Orphanet J Rare Dis ; 18(1): 52, 2023 03 13.
Article in English | MEDLINE | ID: mdl-36907859

ABSTRACT

BACKGROUND: Tay-Sachs disease (TSD), an autosomal recessively inherited neurodegenerative lysosomal storage disease, reported worldwide with a high incidence among population of Eastern European and Ashkenazi Jewish descent. Mutations in the alpha subunit of HEXA that encodes for the ß-hexosaminidase-A lead to deficient enzyme activity and TSD phenotype. This study is the first to highlight the HEXA sequence variations spectrum in a cohort of Egyptian patients with infantile TSD. RESULTS: This study involved 13 Egyptian infant/children patients presented with the infantile form of TSD, ten of the 13 patients were born to consanguineous marriages. ß-hexosaminidase-A enzyme activity was markedly reduced in the 13 patients with a mean activity of 3 µmol/L/h ± 1.56. Sanger sequencing of the HEXA' coding regions and splicing junctions enabled a detection rate of ~ 62% (8/13) in our patients revealing the molecular defects in eight patients; six homozygous-mutant children (five of them were the product of consanguineous marriages) and two patients showed their mutant alleles in heterozygous genotypes, while no disease-causing mutation was identified in the remaining patients. Regulatory intragenic mutations or del/dup may underlie the molecular defect in those patients showing no relevant pathogenic sequencing variants or in the two patients with a heterozygous genotype of the mutant allele. This research identified three novel, likely pathogenic variants in association with the TSD phenotype; two missense, c.920A > C (E307A) and c.952C > G (H318D) in exon 8, and a single base deletion c.484delG causing a frameshift E162Rfs*37 (p.Glu162ArgfsTer37) in exon 5. Three recurrent disease-causing missense mutations; c.1495C > T (R499C), c.1511G > A(R504H), and c.1510C > T(R504C) in exon 13 were identified in five of the eight patients. None of the variants was detected in 50 healthy Egyptians' DNA. Five variants, likely benign or of uncertain significance, S3T, I436V, E506E, and T2T, in exons 1, 11,13, & 1 were detected in our study. CONCLUSIONS: For the proper diagnostics, genetic counseling, and primary prevention, our study stresses the important role of Next Generation Sequencing approaches in delineating the molecular defect in TSD-candidate patients that showed negative Sanger sequencing or a heterozygous mutant allele in their genetic testing results. Interestingly, the three recurrent TSD associated mutations were clustered on chromosome 13 and accounted for 38% of the HEXA mutations detected in this study. This suggested exon 13 as the first candidate for sequencing screening in Egyptian patients with infantile TSD. Larger studies involving our regional population are recommended, hence unique disease associated pathogenic variations could be identified.


Subject(s)
Tay-Sachs Disease , beta-Hexosaminidase alpha Chain , Humans , beta-Hexosaminidase alpha Chain/chemistry , beta-Hexosaminidase alpha Chain/genetics , beta-N-Acetylhexosaminidases/genetics , Egypt , Hexosaminidase A/genetics , Mutation , Tay-Sachs Disease/genetics , Infant
3.
J Am Chem Soc ; 144(2): 832-844, 2022 01 19.
Article in English | MEDLINE | ID: mdl-34985906

ABSTRACT

Owing to its roles in human health and disease, the modification of nuclear, cytoplasmic, and mitochondrial proteins with O-linked N-acetylglucosamine residues (O-GlcNAc) has emerged as a topic of great interest. Despite the presence of O-GlcNAc on hundreds of proteins within cells, only two enzymes regulate this modification. One of these enzymes is O-GlcNAcase (OGA), a dimeric glycoside hydrolase that has a deep active site cleft in which diverse substrates are accommodated. Chemical tools to control OGA are emerging as essential resources for helping to decode the biochemical and cellular functions of the O-GlcNAc pathway. Here we describe rationally designed bicyclic thiazolidine inhibitors that exhibit superb selectivity and picomolar inhibition of human OGA. Structures of these inhibitors in complex with human OGA reveal the basis for their exceptional potency and show that they extend out of the enzyme active site cleft. Leveraging this structure, we create a high affinity chemoproteomic probe that enables simple one-step purification of endogenous OGA from brain and targeted proteomic mapping of its post-translational modifications. These data uncover a range of new modifications, including some that are less-known, such as O-ubiquitination and N-formylation. We expect that these inhibitors and chemoproteomics probes will prove useful as fundamental tools to decipher the mechanisms by which OGA is regulated and directed to its diverse cellular substrates. Moreover, the inhibitors and structures described here lay out a blueprint that will enable the creation of chemical probes and tools to interrogate OGA and other carbohydrate active enzymes.


Subject(s)
Antigens, Neoplasm/metabolism , Bridged Bicyclo Compounds/chemistry , Enzyme Inhibitors/chemistry , Histone Acetyltransferases/metabolism , Hyaluronoglucosaminidase/metabolism , Amino Acid Sequence , Brain/metabolism , Bridged Bicyclo Compounds/metabolism , Catalytic Domain , Chromatography, High Pressure Liquid , Enzyme Inhibitors/metabolism , Histone Acetyltransferases/antagonists & inhibitors , Humans , Hyaluronoglucosaminidase/antagonists & inhibitors , Mass Spectrometry , Peptides/analysis , Peptides/chemistry , Protein Processing, Post-Translational , Proteomics/methods , Structure-Activity Relationship , Thiazolidines/chemistry , Thiazolidines/metabolism , beta-Hexosaminidase alpha Chain/antagonists & inhibitors , beta-Hexosaminidase alpha Chain/metabolism
4.
J Mol Neurosci ; 72(3): 555-564, 2022 Mar.
Article in English | MEDLINE | ID: mdl-34554397

ABSTRACT

Lysosomal storage diseases (LSDs) are known as genetic disorders with an overall prevalence of 1 per 7700 live births. Sphingolipidosis, which is a subgroup of LSDs, is resulted from mutations in the coding genes of specific enzymes of sphingolipid hydrolases. The current study aimed to provide additional knowledge on the genotype of sphingolipidoses disease among Iranian patients affected by the disease. In this research, we studied 68 unrelated Iranian patients diagnosed with one kind of sphingolipidoses from 2014 to 2019. Thereafter, genomic DNA was isolated from their peripheral blood leukocytes samples in EDTA in terms of the manufacturer's protocol. All the coding exons and exon-intron boundaries of the related genes were sequenced and then analyzed using the NCBI database. Finally, they were reviewed using some databases such as the Human Gene Mutation Database (HGMD) and ClinVar ( https://www.ncbi.nlm.nih.gov/clinva ). By studying 22 MLD patients, 18 different variations of the ARSA gene were found, one of which was new including, named as c.472 T > G p. (Cys158Gly). Out of 15 Sandhoff disease (SD) patients, 11 different variations of the HEXB gene were found. Correspondingly, the c.1083-2delA was not reported earlier. By investigating 21 Iranian patients with Tay-Sachs disease (TSD), one new variant was found as c.622delG. The study of 10 Niemann-Pick disease A/B (NPDA/B (patients has led to the identification of 9 different SMPD1 gene variations, among which 3 variations were novel mutations. The results of the present study can be expanded to the genotypic spectrum of Iranian patients with MLD, SD, TSD, and NPD diseases and also used to innovate more effective methods for the detection of genetic carriers as well as diagnosing and counseling of Iranian patients affected with these disorders.


Subject(s)
Tay-Sachs Disease , Exons , Genotype , Heterozygote , Humans , Iran , Mutation , Sphingomyelin Phosphodiesterase , Tay-Sachs Disease/genetics , beta-Hexosaminidase alpha Chain , beta-Hexosaminidase beta Chain/genetics
6.
Int J Mol Sci ; 22(13)2021 Jun 23.
Article in English | MEDLINE | ID: mdl-34201771

ABSTRACT

GM2 gangliosidosis disorders are a group of neurodegenerative diseases that result from a functional deficiency of the enzyme ß-hexosaminidase A (HexA). HexA consists of an α- and ß-subunit; a deficiency in either subunit results in Tay-Sachs Disease (TSD) or Sandhoff Disease (SD), respectively. Viral vector gene transfer is viewed as a potential method of treating these diseases. A recently constructed isoenzyme to HexA, called HexM, has the ability to effectively catabolize GM2 gangliosides in vivo. Previous gene transfer studies have revealed that the scAAV9-HEXM treatment can improve survival in the murine SD model. However, it is speculated that this treatment could elicit an immune response to the carrier capsid and "non-self"-expressed transgene. This study was designed to assess the immunocompetence of TSD and SD mice, and test the immune response to the scAAV9-HEXM gene transfer. HexM vector-treated mice developed a significant anti-HexM T cell response and antibody response. This study confirms that TSD and SD mouse models are immunocompetent, and that gene transfer expression can create an immune response in these mice. These mouse models could be utilized for investigating methods of mitigating immune responses to gene transfer-expressed "non-self" proteins, and potentially improve treatment efficacy.


Subject(s)
Dependovirus/genetics , G(M2) Ganglioside/metabolism , Genetic Vectors/administration & dosage , Immunity/immunology , Sandhoff Disease/immunology , Tay-Sachs Disease/immunology , beta-Hexosaminidase alpha Chain/genetics , Animals , Disease Models, Animal , Female , Genetic Therapy , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Sandhoff Disease/genetics , Sandhoff Disease/therapy , Tay-Sachs Disease/genetics , Tay-Sachs Disease/therapy
7.
Proteins ; 89(11): 1587-1601, 2021 11.
Article in English | MEDLINE | ID: mdl-34288098

ABSTRACT

ß-hexosaminidase A (HexA) protein is responsible for the degradation of GM2 gangliosides in the central and peripheral nervous systems. Tay-Sachs disease occurs when HexA within Hexosaminidase does not properly function and harmful GM2 gangliosides begin to build up within the neurons. In this study, in silico methods such as SIFT, PolyPhen-2, PhD-SNP, and MutPred were utilized to analyze the effects of nonsynonymous single nucleotide polymorphisms (nsSNPs) on HexA in order to identify possible pathogenetic and deleterious variants. Molecular dynamics (MD) simulations showed that two mutants, P25S and W485R, experienced an increase in structural flexibility compared to the native protein. Particularly, there was a decrease in the overall number and frequencies of hydrogen bonds for the mutants compared to the wildtype. MM/GBSA calculations were performed to help assess the change in binding affinity between the wildtype and mutant structures and a mechanism-based inhibitor, NGT, which is known to help increase the residual activity of HexA. Both of the mutants experienced a decrease in the binding affinity from -23.8 kcal/mol in wildtype to -20.9 and -18.7 kcal/mol for the P25S and W485R variants of HexA, respectively.


Subject(s)
G(M2) Ganglioside/chemistry , Molecular Dynamics Simulation , Point Mutation , Polymorphism, Single Nucleotide , Tay-Sachs Disease/genetics , beta-Hexosaminidase alpha Chain/chemistry , Acetylglucosamine/analogs & derivatives , Acetylglucosamine/chemistry , Acetylglucosamine/pharmacology , Binding Sites , Central Nervous System/enzymology , Central Nervous System/pathology , G(M2) Ganglioside/metabolism , Gene Expression , Humans , Hydrogen Bonding , Neurons/enzymology , Neurons/pathology , Peripheral Nervous System/enzymology , Peripheral Nervous System/pathology , Protein Binding , Protein Conformation, alpha-Helical , Protein Conformation, beta-Strand , Protein Interaction Domains and Motifs , Tay-Sachs Disease/enzymology , Tay-Sachs Disease/pathology , Thermodynamics , Thiazoles/chemistry , Thiazoles/pharmacology , beta-Hexosaminidase alpha Chain/genetics , beta-Hexosaminidase alpha Chain/metabolism
8.
Mol Genet Genomic Med ; 9(6): e1677, 2021 06.
Article in English | MEDLINE | ID: mdl-33811753

ABSTRACT

BACKGROUND: Tay-Sachs disease (TSD) is a lysosomal storage disease caused by mutations in the HEXA gene that encodes the HexosaminidaseA (HEXA) enzyme. As HEXA normally functions to degrade the protein GM2-ganglioside in lysosomes, decreased levels of HEXAcauses an accumulation of the protein and leads to neurological toxicity. Typical clinical manifestations of TSD include neurodevelopmental regression, muscle weakness, hypotonia, hyperreflexia, ataxia, seizures, and other neurological symptoms. It is quite rare in Asian populations, wherein only two cases have been reported in Korea to date. METHODS: Clinical records, radiological assessments, and laboratory findings, such as plasma hexosaminidase assay and HEXA analysis, were extracted from the medical records of three (1 male and 2 female) independent Korean children with infantile form of Tay-Sachs disease. RESULTS: All three children presented with neurodevelopmental regression and strabismus at around 8 months of age. Presence of cherry-red spots in the macula led to conduction of biochemical and genetic studies for TSD confirmation. The plasma hexosaminidase assay revealed decreased HEXA activity and low to normal total hexosaminidase activity. Similarly, genetic analysis revealed 4 variants from 6 alleles, including 2 previously reported and 2 novel variants, in the HEXA gene. CONCLUSION: We presented three Korean children, who were recently diagnosed with infantile-type TSDvia enzyme assay and genetic analysis. Furthermore, results showed that fundus examination can be helpful for early diagnosis of children with neurodevelopmental regression.


Subject(s)
Tay-Sachs Disease/genetics , beta-Hexosaminidase alpha Chain/genetics , Child, Preschool , Disease Progression , Early Diagnosis , Female , Fundus Oculi , Humans , Infant , Male , Mutation , Republic of Korea , Tay-Sachs Disease/diagnosis , beta-Hexosaminidase alpha Chain/blood
9.
J Gene Med ; 22(9): e3205, 2020 09.
Article in English | MEDLINE | ID: mdl-32335981

ABSTRACT

BACKGROUND: Tay-Sachs and Sandhoff disease are debilitating genetic diseases that affect the central nervous system leading to neurodegeneration through the accumulation of GM2 gangliosides. There are no cures for these diseases and treatments do not alleviate all symptoms. Hematopoietic stem cell gene therapy offers a promising treatment strategy for delivering wild-type enzymes to affected cells. By genetically modifying hematopoietic stem cells to express wild-type HexA and HexB, systemic delivery of functional enzyme can be achieved. METHODS: Primary human hematopoietic stem/progenitor cells and Tay-Sachs affected cells were used to evaluate the functionality of the vector. An immunodeficient and humanized mouse model of Sandhoff disease was used to evaluate whether the HexA/HexB lentiviral vector transduced cells were able to improve the phenotypes associated with Sandhoff disease. An immunodeficient NOD-RAG1-/-IL2-/- (NRG) mouse model was used to evaluate whether the HexA/HexB vector transduced human CD34+ cells were able to engraft and undergo normal multilineage hematopoiesis. RESULTS: HexA/HexB lentiviral vector transduced cells demonstrated strong expression of HexA and HexB and restored enzyme activity in Tay-Sachs affected cells. Upon transplantation into a humanized Sandhoff disease mouse model, improved motor and behavioral skills were observed. Decreased GM2 gangliosides were observed in the brains of HexA/HexB vector transduced cell transplanted mice. Increased peripheral blood levels of HexB was also observed in transplanted mice. Normal hematopoiesis in the peripheral blood and various lymphoid organs was also observed in transplanted NRG mice. CONCLUSIONS: These results highlight the potential use of stem cell gene therapy as a treatment strategy for Tay-Sachs and Sandhoff disease.


Subject(s)
Antigens, CD34/genetics , Motor Activity/genetics , Sandhoff Disease/genetics , Tay-Sachs Disease/genetics , Animals , Behavior, Animal/physiology , Disease Models, Animal , Genetic Vectors/pharmacology , Hematopoietic Stem Cell Transplantation , Hematopoietic Stem Cells/metabolism , Homeodomain Proteins/genetics , Humans , Interleukin-2/genetics , Lentivirus/genetics , Mice , Mice, Inbred NOD , Sandhoff Disease/pathology , Sandhoff Disease/therapy , Tay-Sachs Disease/pathology , Tay-Sachs Disease/therapy , beta-Hexosaminidase alpha Chain/genetics , beta-Hexosaminidase beta Chain/genetics
10.
Neurobiol Dis ; 134: 104667, 2020 02.
Article in English | MEDLINE | ID: mdl-31682993

ABSTRACT

The favorable outcome of in vivo and ex vivo gene therapy approaches in several Lysosomal Storage Diseases suggests that these treatment strategies might equally benefit GM2 gangliosidosis. Tay-Sachs and Sandhoff disease (the main forms of GM2 gangliosidosis) result from mutations in either the HEXA or HEXB genes encoding, respectively, the α- or ß-subunits of the lysosomal ß-Hexosaminidase enzyme. In physiological conditions, α- and ß-subunits combine to generate ß-Hexosaminidase A (HexA, αß) and ß-Hexosaminidase B (HexB, ßß). A major impairment to establishing in vivo or ex vivo gene therapy for GM2 gangliosidosis is the need to synthesize the α- and ß-subunits at high levels and with the correct stoichiometric ratio, and to safely deliver the therapeutic products to all affected tissues/organs. Here, we report the generation and in vitro validation of novel bicistronic lentiviral vectors (LVs) encoding for both the murine and human codon optimized Hexa and Hexb genes. We show that these LVs drive the safe and coordinate expression of the α- and ß-subunits, leading to supranormal levels of ß-Hexosaminidase activity with prevalent formation of a functional HexA in SD murine neurons and glia, murine bone marrow-derived hematopoietic stem/progenitor cells (HSPCs), and human SD fibroblasts. The restoration/overexpression of ß-Hexosaminidase leads to the reduction of intracellular GM2 ganglioside storage in transduced and in cross-corrected SD murine neural progeny, indicating that the transgenic enzyme is secreted and functional. Importantly, bicistronic LVs safely and efficiently transduce human neurons/glia and CD34+ HSPCs, which are target and effector cells, respectively, in prospective in vivo and ex vivo GT approaches. We anticipate that these bicistronic LVs may overcome the current requirement of two vectors co-delivering the α- or ß-subunits genes. Careful assessment of the safety and therapeutic potential of these bicistronic LVs in the SD murine model will pave the way to the clinical development of LV-based gene therapy for GM2 gangliosidosis.


Subject(s)
Gangliosidoses, GM2/metabolism , Genetic Therapy/methods , Hematopoietic Stem Cells/metabolism , Neural Stem Cells/metabolism , beta-Hexosaminidase alpha Chain/metabolism , beta-Hexosaminidase beta Chain/metabolism , Animals , Gangliosidoses, GM2/genetics , Genetic Vectors , Humans , Lentivirus , Mice , beta-Hexosaminidase alpha Chain/genetics , beta-Hexosaminidase beta Chain/genetics
11.
J Cereb Blood Flow Metab ; 40(6): 1338-1350, 2020 06.
Article in English | MEDLINE | ID: mdl-31357902

ABSTRACT

In Tay-Sachs and Sandhoff disease, a deficiency of the lysosomal enzyme ß-hexosaminidase causes GM2 and other gangliosides to accumulate in neurons and triggers neurodegeneration. Although the pathology centers on neurons, ß-hexosaminidase is mainly expressed outside of neurons, suggesting that gene therapy of these diseases should target non-neuronal cells to reconstitute physiological conditions. Here, we tested in Hexb-/- mice, a model of Sandhoff disease, to determine whether endothelial expression of the genes for human ß-hexosaminidase subunit A and B (HEXA, HEXB) is able to reduce disease symptoms and prolong survival of the affected mice. The brain endothelial selective vectors AAV-BR1-CAG-HEXA and AAV-BR1-CAG-HEXB transduced brain endothelial cells, which subsequently released ß-hexosaminidase enzyme. In vivo intravenous administration of the gene vectors to adult and neonatal mice prolonged survival. They improved neurological function and reduced accumulation of the ganglioside GM2 and the glycolipid GA2 as well as astrocytic activation. Overall, the data demonstrate that endothelial cells are a suitable target for intravenous gene therapy of GM2 gangliosidoses and possibly other lysosomal storage disorders.


Subject(s)
Endothelial Cells , Genetic Therapy/methods , Sandhoff Disease , beta-Hexosaminidase alpha Chain/administration & dosage , beta-Hexosaminidase beta Chain/administration & dosage , Animals , Brain , Dependovirus , Disease Models, Animal , Genetic Vectors , Humans , Mice , Mice, Knockout , Transduction, Genetic , beta-Hexosaminidase alpha Chain/genetics , beta-Hexosaminidase beta Chain/genetics
12.
Sci Rep ; 9(1): 14241, 2019 10 02.
Article in English | MEDLINE | ID: mdl-31578452

ABSTRACT

GRP94 is an ATP-dependent chaperone able to regulate pro-oncogenic signaling pathways. Previous studies have shown a critical role of GRP94 in brain metastasis (BrM) pathogenesis and progression. In this work, an untargeted lipidomic analysis revealed that some lipid species were altered in GRP94-deficient cells, specially GM2 and GM3 gangliosides. The catalytic pathway of GM2 is affected by the low enzymatic activity of ß-Hexosaminidase (HexA), responsible for the hydrolysis of GM2 to GM3. Moreover, a deficiency of the GM2-activator protein (GM2-AP), the cofactor of HexA, is observed without alteration of gene expression, indicating a post-transcriptional alteration of GM2-AP in the GRP94-ablated cells. One plausible explanation of these observations is that GM2-AP is a client of GRP94, resulting in defective GM2 catabolic processing and lysosomal accumulation of GM2 in GRP94-ablated cells. Overall, given the role of gangliosides in cell surface dynamics and signaling, their imbalance might be linked to modifications of cell behaviour acquired in BrM progression. This work indicates that GM2-AP could be an important factor in ganglioside balance maintenance. These findings highlight the relevance of GM3 and GM2 gangliosides in BrM and reveal GM2-AP as a promising diagnosis and therapeutic target in BrM research.


Subject(s)
Brain Neoplasms/secondary , Carcinoma/secondary , G(M2) Activator Protein/biosynthesis , G(M2) Ganglioside/analysis , G(M3) Ganglioside/analysis , Membrane Glycoproteins/physiology , Neoplasm Proteins/physiology , Animals , Brain Neoplasms/metabolism , Carcinoma/metabolism , Cell Line, Tumor , Culture Media, Conditioned/chemistry , Down-Regulation , Female , G(M2) Activator Protein/genetics , Gene Expression Regulation, Neoplastic , Genes, Reporter , Humans , Lipidomics , Lysosomes/metabolism , Membrane Glycoproteins/antagonists & inhibitors , Membrane Glycoproteins/genetics , Mice , Neoplasm Proteins/antagonists & inhibitors , Neoplasm Proteins/biosynthesis , Neoplasm Proteins/genetics , RNA Interference , RNA, Small Interfering/genetics , Triple Negative Breast Neoplasms/pathology , beta-Hexosaminidase alpha Chain/biosynthesis , beta-Hexosaminidase alpha Chain/genetics
13.
BMJ Case Rep ; 12(9)2019 Sep 12.
Article in English | MEDLINE | ID: mdl-31519716

ABSTRACT

Tay-Sachs disease (TSD) is a type 1 gangliosidosis (GM2) and caused by hexosaminidase A deficiency resulting in abnormal sphingolipid metabolism and deposition of precursors in different organs. It is a progressive neurodegenerative disorder transmitted in an autosomal-recessive manner. There is an accumulation of GM2 in neurocytes and retinal ganglions which result in progressive loss of neurological function and formation of the cherry-red spot which is the hallmark of TSD. We report the first case of juvenile TSD from Pakistan in a child with death of an older sibling without the diagnosis.


Subject(s)
Tay-Sachs Disease/diagnosis , Tay-Sachs Disease/genetics , beta-Hexosaminidase alpha Chain/genetics , Child, Preschool , Gait Disorders, Neurologic/diagnosis , Gait Disorders, Neurologic/etiology , Humans , Male , Mutation/genetics , Pakistan/epidemiology , Palliative Care/methods , Tay-Sachs Disease/physiopathology , Exome Sequencing/methods
14.
J Hum Genet ; 64(10): 985-994, 2019 Oct.
Article in English | MEDLINE | ID: mdl-31388111

ABSTRACT

Tay-Sachs disease (TSD) (OMIM) is a neurodegenerative lysosomal storage disorder caused due to mutations in the HEXA gene. To date, nearly 190 mutations have been reported in HEXA gene. Here, we have characterized 34 enzymatically confirmed TSD families to investigate the presence of novel as well as known variants in HEXA gene. Overall study detected 25 variants belonging to 31 affected TSD patients and 3 carrier couples confirmed by enzyme study. Of these 17 patients harbors 15 novel variants, including seven missense variants [p.V206L, p.Y213H, p.R252C, p.F257S, p.C328G, p.G454R, and p.P475R], four nonsense variant [p.S9X, p.E91X, p.W420X, and p.W482X], two splice site variants [c.347-1G>A and c.460-1G>A], and two small deletion [c.1349delC (p.A450VfsX3) and c.52delG (p.G18Dfs*82)]. While remaining 17 patients harbors 10 previously reported variants that includes six missense variants [p.M1T, p.R170Q, p.D322Y, p.D322N, p.E462V, and p.R499C], one nonsense variant [p.Q106X], two splice site variants [c.1073+1G>A and c.459+4A>G] and one 4 bp insertion [c.1278insTATC (p.Y427IfsX5)]. In conclusion, Indian infantile TSD patients provide newer insight into the molecular heterogeneity of the TSD. Combining present study and our earlier studies, we have observed that 67% genotypes found in Indian TSD patients are novel, which are associated with severe infantile phenotypes, while rest 33% genotypes found in our cohort were previously reported in various populations. In addition, higher frequency of the p.E462V and c.1278insTATC mutations in the present study further support and suggest the prevalence of p.E462V mutation in the Indian population.


Subject(s)
Tay-Sachs Disease/genetics , beta-Hexosaminidase alpha Chain/genetics , Alleles , Child, Preschool , Codon, Nonsense , Demography , Female , Genetic Association Studies , Humans , India , Infant , Male , Mutation, Missense , Sequence Deletion , Tay-Sachs Disease/enzymology , Tay-Sachs Disease/physiopathology , beta-Hexosaminidase alpha Chain/chemistry
15.
Mol Genet Genomic Med ; 7(8): e836, 2019 08.
Article in English | MEDLINE | ID: mdl-31293106

ABSTRACT

BACKGROUND: Pathogenic variants in HEXA that impair ß-hexosaminidase A (Hex A) enzyme activity cause Tay-Sachs Disease (TSD), a severe autosomal-recessive neurodegenerative disorder. Hex A enzyme analysis demonstrates near-zero activity in patients affected with TSD and can also identify carriers, whose single functional copy of HEXA results in reduced enzyme activity relative to noncarriers. Although enzyme testing has been optimized and widely used for carrier screening in Ashkenazi Jewish (AJ) individuals, it has unproven sensitivity and specificity in a pan-ethnic population. The ability to detect HEXA variants via DNA analysis has evolved from limited targeting of a few ethnicity-specific variants to next-generation sequencing (NGS) of the entire coding region coupled with interpretation of any discovered novel variants. METHODS: We combined results of enzyme testing, retrospective computational analysis, and variant reclassification to estimate the respective clinical performance of TSD screening via enzyme analysis and NGS. We maximized NGS accuracy by reclassifying variants of uncertain significance and compared to the maximum performance of enzyme analysis estimated by calculating ethnicity-specific frequencies of variants known to yield false-positive or false-negative enzyme results (e.g., pseudodeficiency and B1 alleles). RESULTS: In both AJ and non-AJ populations, the estimated clinical sensitivity, specificity, and positive predictive value were higher by NGS than by enzyme testing. The differences were significant for all comparisons except for AJ clinical sensitivity, where NGS exceeded enzyme testing, but not significantly. CONCLUSIONS: Our results suggest that performance of an NGS-based TSD carrier screen that interrogates the entire coding region and employs novel variant interpretation exceeds that of Hex A enzyme testing, warranting a reconsideration of existing guidelines.


Subject(s)
Enzyme Assays/standards , Genetic Carrier Screening/methods , High-Throughput Nucleotide Sequencing/standards , Tay-Sachs Disease/diagnosis , beta-Hexosaminidase alpha Chain/genetics , Cohort Studies , Ethnicity/genetics , False Negative Reactions , False Positive Reactions , Genetic Carrier Screening/standards , Genetic Counseling/methods , Genetic Counseling/standards , Heterozygote , Humans , Mutation , Polymorphism, Single Nucleotide , Practice Guidelines as Topic , Retrospective Studies , Sensitivity and Specificity , Tay-Sachs Disease/genetics
16.
Methods Mol Biol ; 1885: 233-250, 2019.
Article in English | MEDLINE | ID: mdl-30506202

ABSTRACT

Tay-Sachs disease (TSD) is an autosomal recessive lysosomal storage disorder caused by mutations of the HEXA gene resulting in the deficiency of hexosaminidase A (Hex A) and subsequent neuronal accumulation of GM2 gangliosides. Infantile TSD is a devastating and fetal neurodegenerative disease with death before the age of 3-5 years. A small proportion of TSD patients carry milder mutations and may present juvenile or adult onset milder disease. TSD is more prevalent among Ashkenazi Jewish (AJ) individuals and some other genetically isolated populations with carrier frequencies of approximately ~1:27 which is much higher than that of 1:300 in the general population. Carrier screening and prenatal testing for TSD are effective in preventing the birth of affected fetuses greatly diminishing the incidence of TSD. Testing of targeted HEXA mutations by genotyping or sequencing can detect 98% of carriers in AJ individuals; however, the detection rate is much lower for most other ethnic groups. When combined with enzyme analysis, above 98% of carriers can be reliably identified regardless of ethnic background. Multiplex PCR followed by allele-specific primer extension is one method to test for known and common mutations. Sanger sequencing or other sequencing methods are useful to identify private mutations. For prenatal testing, only predefined parental mutations need to be tested. In the event of unknown mutational status or the presence of variants of unknown significance (VUS), enzyme analysis must be performed in conjunction with DNA-based assays to enhance the diagnostic accuracy. Enzymatic assays involve the use of synthetic substrates 4-methylumbelliferyl-N-acetyl-ß-glucosamine (4-MUG) and 4-methylumbelliferyl-6-sulfo-2-acetamido-2-deoxy-ß-D-glucopyranoside (4-MUGS) to measure the percentage Hex A activity (Hex A%) and specific Hex A activity respectively. These biochemical and molecular tests can be performed in both direct specimens and cultured cells from chorionic villi sampling or amniocentesis.


Subject(s)
Genetic Testing , Prenatal Diagnosis/methods , Tay-Sachs Disease/diagnosis , Tay-Sachs Disease/genetics , Alleles , DNA Contamination , DNA Mutational Analysis , Electrophoresis, Capillary , Genetic Testing/methods , Genetic Testing/standards , Genotype , Humans , Mutation , Polymerase Chain Reaction , Prenatal Diagnosis/standards , Tay-Sachs Disease/metabolism , beta-Hexosaminidase alpha Chain/genetics , beta-Hexosaminidase alpha Chain/metabolism
17.
BMC Med Genet ; 19(1): 162, 2018 09 12.
Article in English | MEDLINE | ID: mdl-30208878

ABSTRACT

BACKGROUND: Mucolipidosis alpha/beta is an inborn error of metabolism characterized by deficiency of GlcNAc-1-phosphotransferase, in which essential alpha/beta subunits are encoded by the GNPTAB gene. The autosomal recessive condition is due to disruptions of hydrolase mannose 6-phosphate marker generation, defective lysosomal targeting and subsequent intracellular accumulation of non-degraded material. Clinical severity depends on residual GlcNAc-1-phosphotransferase activity, which distinguishes between the milder type III disease and the severe, neonatal onset type II disease. CASE PRESENTATION: We report the clinical, biochemical and genetic diagnosis of mucolipidosis III alpha/beta in a two-year-old Chinese boy who initially presented with poor weight gain, microcephaly and increased tone. He was confirmed to harbor the common splice site mutation c.2715 + 1G > A and the nonsense variant c.2404C > T (p.Q802*). Clinically, the patient had multiple phenotypic features typical of mucopolysaccharidosis including joint contractures, coarse facial features, kypho-lordosis, pectus carinatum and umbilical hernia. However, the relatively mild developmental delay compared to severe type I and type II mucopolysaccharidosis and the absence of macrocephaly raised the possibility of the less commonly diagnosed mucolipidosis alpha/beta. Critical roles of lysosomal enzyme activity assay, which showed elevated α-iduronidase, iduronate sulfatase, galactose-6-sulphate sulphatase, arylsulfatase B and α-hexosaminidase activities; and genetic study, which confirmed the parental origin of both mutations, were highlighted. CONCLUSIONS: The recently reported nonsense variant c.2404C > T in the GNPTAB gene is further recognized and this contributes to the genotype-phenotype spectrum of mucolipidosis alpha/beta.


Subject(s)
Codon, Nonsense , Mucolipidoses/genetics , Transferases (Other Substituted Phosphate Groups)/genetics , Child, Preschool , Chondroitinsulfatases/genetics , Chondroitinsulfatases/metabolism , Gene Expression Regulation , Genes, Recessive , Humans , Iduronate Sulfatase/genetics , Iduronate Sulfatase/metabolism , Iduronidase/genetics , Iduronidase/metabolism , Lysosomes/enzymology , Lysosomes/pathology , Male , Mucolipidoses/diagnosis , Mucolipidoses/enzymology , Mucolipidoses/pathology , N-Acetylgalactosamine-4-Sulfatase/genetics , N-Acetylgalactosamine-4-Sulfatase/metabolism , Pedigree , Transferases (Other Substituted Phosphate Groups)/deficiency , beta-Hexosaminidase alpha Chain/genetics , beta-Hexosaminidase alpha Chain/metabolism
18.
BMC Med Genet ; 19(1): 109, 2018 07 04.
Article in English | MEDLINE | ID: mdl-29973161

ABSTRACT

BACKGROUND: Tay-Sachs disease (TSD) is a sphingolipid storage disorder caused by mutations in the HEXA gene. To date, nearly 170 mutations of HEXA have been described, including only one 7.6 kb large deletion. METHODS: Multiplex Ligation-dependent Probe Amplification (MLPA) study was carried out in 5 unrelated patients for copy number changes where heterozygous and/or homozygous disease causing mutation/s could not be identified in the coding region by sequencing of HEXA gene. RESULTS: The study has identified the presence of a homozygous deletion of exon-2 and exon-3 in two patients, two patient showed compound heterozygosity with exon 1 deletion combined with missense mutation p.E462V and one patient was identified with duplication of exon-1 with novel variants c.1527-2A > T as a second allele. CONCLUSION: This is the first report of deletion/duplication in HEXA gene providing a new insight into the molecular basis of TSD and use of MLPA assay for detecting large copy number changes in the HEXA gene.


Subject(s)
Sequence Deletion/genetics , Tay-Sachs Disease/genetics , beta-Hexosaminidase alpha Chain/genetics , Exons/genetics , Female , Heterozygote , Homozygote , Humans , India , Infant , Male , Mutation, Missense/genetics
20.
Hum Gene Ther ; 29(3): 312-326, 2018 03.
Article in English | MEDLINE | ID: mdl-28922945

ABSTRACT

Tay-Sachs disease (TSD) is a fatal neurodegenerative disorder caused by a deficiency of the enzyme hexosaminidase A (HexA). TSD also occurs in sheep, the only experimental model of TSD that has clinical signs of disease. The natural history of sheep TSD was characterized using serial neurological evaluations, 7 Tesla magnetic resonance imaging, echocardiograms, electrodiagnostics, and cerebrospinal fluid biomarkers. Intracranial gene therapy was also tested using AAVrh8 monocistronic vectors encoding the α-subunit of Hex (TSD α) or a mixture of two vectors encoding both the α and ß subunits separately (TSD α + ß) injected at high (1.3 × 1013 vector genomes) or low (4.2 × 1012 vector genomes) dose. Delay of symptom onset and/or reduction of acquired symptoms were noted in all adeno-associated virus-treated sheep. Postmortem evaluation showed superior HexA and vector genome distribution in the brain of TSD α + ß sheep compared to TSD α sheep, but spinal cord distribution was low in all groups. Isozyme analysis showed superior HexA formation after treatment with both vectors (TSD α + ß), and ganglioside clearance was most widespread in the TSD α + ß high-dose sheep. Microglial activation and proliferation in TSD sheep-most prominent in the cerebrum-were attenuated after gene therapy. This report demonstrates therapeutic efficacy for TSD in the sheep brain, which is on the same order of magnitude as a child's brain.


Subject(s)
Dependovirus , Genetic Therapy , Tay-Sachs Disease/therapy , beta-Hexosaminidase alpha Chain/biosynthesis , beta-Hexosaminidase beta Chain/biosynthesis , Animals , Brain/diagnostic imaging , Brain/enzymology , Disease Models, Animal , Echocardiography , Humans , Magnetic Resonance Imaging , Microglia/enzymology , Sheep , Tay-Sachs Disease/diagnostic imaging , Tay-Sachs Disease/enzymology , Tay-Sachs Disease/genetics , beta-Hexosaminidase alpha Chain/genetics , beta-Hexosaminidase beta Chain/genetics
SELECTION OF CITATIONS
SEARCH DETAIL