Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 48
Filter
Add more filters

Publication year range
1.
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
2.
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
3.
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
4.
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
5.
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
6.
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
7.
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
8.
Biochim Biophys Acta ; 1849(8): 1104-15, 2015 Aug.
Article in English | MEDLINE | ID: mdl-25934542

ABSTRACT

Nonsense-mediated mRNA decay (NMD), an mRNA surveillance mechanism, eliminates premature termination codon-containing (PTC⁺) transcripts. For instance, it maintains the homeostasis of splicing factors and degrades aberrant transcripts of human genetic disease genes. Here we examine the inhibitory effect on the NMD pathway and consequent increase of PTC+ transcripts by the dietary compound curcumin. We have found that several PTC⁺ transcripts including that of serine/arginine-rich splicing factor 1 (SRSF1) were specifically increased in cells by curcumin. We also observed a similar curcumin effect on the PTC⁺ mutant transcript from a Tay-Sachs-causing HEXA allele or from a beta-globin reporter gene. The curcumin effect was accompanied by significantly reduced expression of the NMD factors UPF1, 2, 3A and 3B. Consistently, in chromatin immunoprecipitation assays, curcumin specifically reduced the occupancy of acetyl-histone H3 and RNA polymerase II at the promoter region (-376 to -247nt) of human UPF1, in a time- and dosage-dependent way. Importantly, knocking down UPF1 abolished or substantially reduced the difference of PTC(+) transcript levels between control and curcumin-treated cells. The disrupted curcumin effect was efficiently rescued by expression of exogenous Myc-UPF1 in the knockdown cells. Together, our data demonstrate that a group of PTC⁺ transcripts are stabilized by a dietary compound curcumin through the inhibition of UPF factor expression and the NMD pathway.


Subject(s)
Codon, Nonsense/genetics , Curcumin/pharmacology , Nonsense Mediated mRNA Decay/drug effects , RNA, Messenger/metabolism , Transcription Termination, Genetic/drug effects , Cells, Cultured , Fibroblasts/drug effects , Fibroblasts/physiology , Humans , Metabolic Networks and Pathways/drug effects , Metabolic Networks and Pathways/genetics , Nuclear Proteins/genetics , RNA, Messenger/genetics , RNA-Binding Proteins/genetics , Serine-Arginine Splicing Factors , Tay-Sachs Disease/genetics , Tay-Sachs Disease/metabolism , beta-Hexosaminidase alpha Chain/genetics , beta-Hexosaminidase alpha Chain/metabolism
9.
Muscle Nerve ; 49(5): 768-71, 2014 May.
Article in English | MEDLINE | ID: mdl-24327357

ABSTRACT

INTRODUCTION: Late-onset Tay-Sachs disease (LOTS) is a lysosomal storage disease caused by deficient Beta-hexosaminidase A activity. METHODS: We describe a 53-year-old woman who presented with adult-onset leg weakness, and whose initial diagnosis was progressive muscular atrophy without identifiable etiology. Development of cerebellar ataxia in mid-life prompted reassessment. RESULTS: Beta-hexosaminidase A quantification assay demonstrated absence of the isozyme. Genetic testing identified compound heterozygous mutations in the HEXA gene, confirming the diagnosis of LOTS. CONCLUSIONS: The phenotypic spectrum of LOTS includes motor neuronopathy, ataxia, choreoathetosis, neuropathy, and psychiatric symptoms in various combinations. This patient highlights the emergence of different clinical features over many years and emphasizes the need to consider LOTS in the differential diagnosis of progressive muscular atrophy.


Subject(s)
Electrodiagnosis , Muscular Atrophy, Spinal/diagnosis , Tay-Sachs Disease/diagnosis , Diagnosis, Differential , Female , Heterozygote , Humans , Middle Aged , Phenotype , Tay-Sachs Disease/genetics , beta-Hexosaminidase alpha Chain/blood , beta-Hexosaminidase alpha Chain/genetics
10.
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
11.
Biochem Biophys Res Commun ; 426(2): 286-8, 2012 Sep 21.
Article in English | MEDLINE | ID: mdl-22943850

ABSTRACT

Mutations of the glycogen branching enzyme gene, GBE1, result in glycogen storage disease (GSD) type IV, an autosomal recessive disorder having multiple clinical forms. One mutant allele of this gene, GBE1 c.1076A>C, has been reported in Ashkenazi Jewish cases of an adult-onset form of GSD type IV, adult polyglucosan body disease (APBD), but no epidemiological analyses of this mutation have been performed. We report here the first epidemiological study of this mutation in persons of Ashkenazi Jewish background and find that this mutation has a gene frequency of 1 in 34.5 (95% CI: 0.0145-0.0512), similar to the frequency of the common mutation causing Tay-Sachs disease among Ashkenazi Jews. This finding reveals APBD to be another monogenic disorder that occurs with increased frequency in persons of Ashkenazi Jewish ancestry.


Subject(s)
Glycogen Debranching Enzyme System/genetics , Glycogen Storage Disease/genetics , Jews/genetics , Nervous System Diseases/genetics , Gene Frequency , Humans , Mutation , beta-Hexosaminidase alpha Chain/genetics
12.
Mol Cell Biochem ; 363(1-2): 109-18, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22147196

ABSTRACT

Lysosomes are not only degrading organelles but also involved in other critical cellular processes. In addition, active lysosomal glycohydrolases have been detected in an extra-lysosomal compartment: the presence of glycohydrolases on the plasma membrane (PM) has been widely demonstrated, and a possible role on the modification of the cell surface glycosphingolipids (GSL) participating in the modulation of cell functions such as cell-to-cell interactions and signal transduction pathways has been proposed. On this basis, the coordinated expression of lysosomal glycohydrolases and their translocation to the PM appear to be crucial for many cellular events. In this paper, we report evidence for the existence of a coordinated mechanism regulating the expression/activity of both lysosomal and PM-associated glycohydrolases. We show that the over-expression of the acidic glycohydrolase ß-hexosaminidase α-subunit in mouse NIH/3T3 fibroblasts induces the increased expression of the Hex ß-subunit necessary to form the active isoenzyme dimers as well as of other glycohydrolases participating in the GSL catabolism, such as ß-galactosidase and ß-glucocerebrosidase. More interestingly, this regulatory effect was also extended to the PM-associated hydrolases. In addition, transfected cells displayed a rearrangement of the GSL expression pattern that cannot be simply explained by the increased activity of a single enzyme. These observations clearly indicate that the expression level of metabolically related glycohydrolases is regulated in a coordinated manner and this regulation mechanism also involves the PM-associated isoforms.


Subject(s)
Cell Membrane/enzymology , Fibroblasts/enzymology , Glycoside Hydrolases/metabolism , Glycosphingolipids/metabolism , Lysosomes/enzymology , beta-Hexosaminidase alpha Chain/metabolism , Animals , Exocytosis , Humans , Mice , NIH 3T3 Cells , Transfection , beta-Hexosaminidase alpha Chain/genetics
13.
Cell Biochem Funct ; 30(1): 61-8, 2012 Jan.
Article in English | MEDLINE | ID: mdl-21997228

ABSTRACT

In an attempt to investigate whether the genetic defect in the HEXA and HEXB genes (which causes the absence of the lysosomal ß-N-acetyl-hexosaminidase), are related to the wide inflammation in GM2 gangliosidoses (Tay-Sachs and Sandhoff disease), we have chosen the dendritic cells (DCs) as a study model. Using the RNA interference approach, we generated an in vitro model of HEXs knock-down immunogenic DCs (i-DCs) from CD34(+)-haemopoietic stem cells (CD34(+)-HSCs), thus mimicking the Tay-Sachs (HEXA-/-) and Sandhoff (HEXB-/-) cells. We showed that the absence of ß-N-acetyl-hexosaminidase activity does not alter the differentiation of i-DCs from HSCs, but it is critical for the activation of CD4(+)T cells because knock-down of HEXA or HEXB gene causes a loss of function of i-DCs. Notably, the silencing of the HEXA gene had a stronger immune inhibitory effect, thereby indicating a major involvement of ß-N-acetyl-hexosaminidase A isoenzyme within this mechanism.


Subject(s)
Dendritic Cells/immunology , Stem Cells/immunology , beta-Hexosaminidase alpha Chain/genetics , beta-Hexosaminidase beta Chain/genetics , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , Cells, Cultured , Dendritic Cells/cytology , Dendritic Cells/metabolism , Gangliosidoses, GM2/immunology , Gangliosidoses, GM2/metabolism , Gene Knockdown Techniques , Humans , Inflammation/immunology , Inflammation/metabolism , Stem Cells/cytology , Stem Cells/metabolism , beta-Hexosaminidase alpha Chain/metabolism , beta-Hexosaminidase beta Chain/metabolism
14.
Mol Genet Metab ; 104(4): 700-2, 2011 Dec.
Article in English | MEDLINE | ID: mdl-21967858

ABSTRACT

Tay-Sachs disease (TSD), a pan-ethnic, autosomal recessive, neurodegenerative, lysosomal disease, results from deficient ß-hexosaminidase A activity due to ß-hexosaminidase α-subunit (HEXA) mutations. Prenatal/premarital carrier screening programs in the Ashkenazi Jewish community have markedly reduced disease occurrence. We report the first Jordanian Arab TSD patient diagnosed by deficient ß-hexosaminidase A activity. HEXA mutation analysis revealed homozygosity for a nonsense mutation, c.78G>A (p.W26X). Previously reported in Arab patients, this mutation is a candidate for TSD screening in Arab populations.


Subject(s)
Codon, Nonsense , Tay-Sachs Disease/diagnosis , beta-Hexosaminidase alpha Chain/genetics , Consanguinity , DNA Mutational Analysis , Genetic Association Studies , Homozygote , Humans , Infant , Jordan , Male , Tay-Sachs Disease/blood , Tay-Sachs Disease/genetics , beta-Hexosaminidase alpha Chain/blood
15.
J Hum Genet ; 56(9): 682-4, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21796138

ABSTRACT

The ß-hexosaminidase A (HEXA) mutations in the first reported cases of infantile Tay-Sachs disease in the Persian population were identified in two unrelated consanguineous families. The clinical diagnoses of the affected infants were confirmed by their markedly deficient levels of HEXA activity in plasma or peripheral leukocytes. The specific causative mutation in each family was determined by sequencing the HEXA alleles in both sets of related parents. Two mutations were identified: c.1A>G (p.MIV), which obliterated the initiating methionine in codon 1, and c.1177C>T (p.R393X), which predicted a termination codon or nonsense mutation.


Subject(s)
Codon, Nonsense , Genetic Predisposition to Disease/genetics , Mutation, Missense , Tay-Sachs Disease/genetics , beta-Hexosaminidase alpha Chain/genetics , Adult , Alleles , Child, Preschool , Codon/genetics , Consanguinity , DNA Mutational Analysis , Female , Humans , Infant , Iran , Male , Polymerase Chain Reaction
16.
Am J Med Genet A ; 155A(6): 1281-4, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21567908

ABSTRACT

The GM2 gangliosidose, Tay-Sachs and Sandhoff diseases, are a class of lysosomal storage diseases in which relentless neurodegeneration results in devastating neurological disability and premature death. Primary prevention is the most effective intervention since no effective therapy is currently available. An extremely successful model for the prevention of GM2 gangliosidosis in the Ashkenazi Jewish community is largely attributable to the very limited number of founder mutations in that population. Consistent with our previous observation of allelic heterogeneity in consanguineous populations, we show here that these diseases are largely caused by private mutations which present a major obstacle in replicating the Ashkenazi success story. Alternative solutions are proposed which can also be implemented for other autosomal recessive diseases in our population.


Subject(s)
Gangliosidoses, GM2/genetics , Genetic Testing/methods , Mutation/genetics , beta-Hexosaminidase alpha Chain/genetics , beta-Hexosaminidase beta Chain/genetics , DNA Mutational Analysis , Gangliosidoses, GM2/prevention & control , Humans , Saudi Arabia
17.
J Inherit Metab Dis ; 34(5): 1061-8, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21598013

ABSTRACT

Sandhoff disease (SD) is a lysosomal disease caused by a mutation of the HEXB gene associated with excessive accumulation of GM2 ganglioside (GM2) in lysosomes and neurological manifestations. Production of autoantibodies against the accumulated gangliosides has been reported to be involved in the progressive pathogenesis of GM2 gangliosidosis, although the underlying mechanism has not been fully elucidated. The thymus is the key organ in the acquired immune system including the development of autoantibodies. We showed here that thymic involution and an increase in cell death in the organ occur in SD model mice at a late stage of the pathogenesis. Dramatic increases in the populations of Annexin-V(+) cells and terminal deoxynucletidyl transferase dUTP nick end labeling (TUNEL) (+) cells were observed throughout the thymuses of 15-week old SD mice. Enhanced caspase-3/7 activation, but not that of caspase-1/4, -6 ,-8, or -9, was also demonstrated. Furthermore, the serum level of corticosterone, a potent inducer of apoptosis of thymocytes, was elevated during the same period of apoptosis. Our studies suggested that an increase in endocrine corticosterone may be one of the causes that accelerate the apoptosis of thymocytes leading to thymic involution in GM2 gangliosidosis, and thus can be used as a disease marker for evaluation of the thymic condition and disease progression.


Subject(s)
Corticosterone/blood , Disease Models, Animal , Mice, Knockout , Sandhoff Disease/blood , Sandhoff Disease/pathology , Thymus Gland/pathology , Age Factors , Animals , Apoptosis/physiology , Atrophy/genetics , Caspases/metabolism , Disease Progression , G(M2) Ganglioside/metabolism , Mice , Mice, Inbred C57BL , Phenotype , Sandhoff Disease/etiology , Sandhoff Disease/genetics , beta-Hexosaminidase alpha Chain/genetics
18.
Mol Ther ; 18(8): 1519-26, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20571546

ABSTRACT

Human lysosomal beta-hexosaminidase A is a heterodimer composed of alpha- and beta-subunits encoded by HEXA and HEXB, respectively. We genetically introduced an additional N-glycosylation sequon into HEXA, which caused amino acid substitutions (S51 to N and A53 to T) at homologous positions to N84 and T86 in the beta-subunit. The mutant HexA (NgHexA) obtained from a Chinese hamster ovary (CHO) cell line co-expressing the mutated HEXA and wild-type HEXB complementary DNAs was demonstrated to contain an additional mannose-6-phosphate (M6P)-type-N-glycan. NgHexA was more efficiently taken up than the wild-type HexA and delivered to lysosomes, where it degraded accumulated substrates including GM2 ganglioside (GM2) when administered to cultured fibroblasts derived from a Sandhoff disease (SD) patient. On intracerebroventricular (i.c.v.) administration of NgHexA to SD model mice, NgHexA more efficiently restored the HexA activity and reduced the GM2 and GA2 (asialoGM2) accumulated in neural cells of the brain parenchyma than the wild-type HexA. These findings indicate that i.c.v. administration of the modified human HexA with an additional M6P-type N-glycan is applicable for enzyme replacement therapy (ERT) involving an M6P-receptor as a molecular target for HexA deficiencies including Tay-Sachs disease and SD.


Subject(s)
Polysaccharides/metabolism , Sandhoff Disease/metabolism , beta-Hexosaminidase alpha Chain/metabolism , beta-N-Acetylhexosaminidases/chemistry , beta-N-Acetylhexosaminidases/therapeutic use , Animals , CHO Cells , Cells, Cultured , Chromatography, Thin Layer , Cricetinae , Cricetulus , G(M2) Ganglioside/metabolism , Glycosylation , Humans , Immunoblotting , Mice , Polysaccharides/chemistry , Sandhoff Disease/drug therapy , Sandhoff Disease/genetics , beta-Hexosaminidase alpha Chain/chemistry , beta-Hexosaminidase alpha Chain/genetics , beta-Hexosaminidase alpha Chain/therapeutic use , beta-Hexosaminidase beta Chain/genetics , beta-Hexosaminidase beta Chain/metabolism , beta-N-Acetylhexosaminidases/genetics , beta-N-Acetylhexosaminidases/metabolism
19.
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
20.
Biochem Biophys Res Commun ; 392(4): 599-602, 2010 Feb 19.
Article in English | MEDLINE | ID: mdl-20100466

ABSTRACT

Tay-Sachs disease (TSD) is a recessively inherited neurodegenerative disorder due to mutations in the HEXA gene resulting in a beta-hexosaminidase A (Hex A) deficiency. The purpose of this study was to characterize the molecular abnormalities in patients with infantile or later-onset forms of the disease. The complete sequencing of the 14 exons and flanking regions of the HEXA gene was performed with a unique technical condition in 10 unrelated TSD patients. Eleven mutations were identified, including five splice mutations, one insertion, two deletions and three single-base substitutions. Four mutations were novel: two splice mutations (IVS8+5G>A, IVS2+4delAGTA), one missense mutation in exon 6 (c.621T>G (p.D207E)) and one small deletion (c.1211-1212delTG) in exon 11 resulting in a premature stop codon at residue 429. The c.621T>G missense mutation was found in a patient presenting an infantile form. Its putative role in the pathogenesis of TSD is suspected as residue 207 is highly conserved in human, mouse and rat. Moreover, structural modelling predicted changes likely to affect substrate binding and catalytic activity of the enzyme. The time-saving procedure reported here could be useful for the characterization of Tay-Sachs-causing mutations, in particular in non-Ashkenazi patients mainly exhibiting rare mutations.


Subject(s)
DNA Mutational Analysis/methods , Polymerase Chain Reaction/methods , Tay-Sachs Disease/diagnosis , beta-Hexosaminidase alpha Chain/genetics , Adult , Animals , Humans , Infant , Mice , Models, Molecular , Mutation, Missense , Protein Conformation , Rats , beta-Hexosaminidase alpha Chain/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL