Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 131
1.
J Inherit Metab Dis ; 47(2): 327-339, 2024 Mar.
Article En | MEDLINE | ID: mdl-38112342

Cerebellar atrophy is a characteristic sign of late-onset Tay-Sachs disease (LOTS). Other structural neuroimaging abnormalities are inconsistently reported. Our study aimed to perform a detailed whole-brain analysis and quantitatively characterize morphometric changes in LOTS patients. Fourteen patients (8 M/6F) with LOTS from three centers were included in this retrospective study. For morphometric brain analyses, we used deformation-based morphometry, voxel-based morphometry, surface-based morphometry, and spatially unbiased cerebellar atlas template. The quantitative whole-brain morphometric analysis confirmed the finding of profound pontocerebellar atrophy with most affected cerebellar lobules V and VI in LOTS patients. Additionally, the atrophy of structures mainly involved in motor control, including bilateral ventral and lateral thalamic nuclei, primary motor and sensory cortex, supplementary motor area, and white matter regions containing corticospinal tract, was present. The atrophy of the right amygdala, hippocampus, and regions of occipital, parietal and temporal white matter was also observed in LOTS patients in contrast with controls (p < 0.05, FWE corrected). Patients with dysarthria and those initially presenting with ataxia had more severe cerebellar atrophy. Our results show predominant impairment of cerebellar regions responsible for speech and hand motor function in LOTS patients. Widespread morphological changes of motor cortical and subcortical regions and tracts in white matter indicate abnormalities in central motor circuits likely coresponsible for impaired speech and motor function.


Tay-Sachs Disease , White Matter , Humans , Tay-Sachs Disease/pathology , White Matter/diagnostic imaging , Retrospective Studies , Magnetic Resonance Imaging , Brain/pathology , Atrophy/pathology
2.
PLoS One ; 18(3): e0280650, 2023.
Article En | MEDLINE | ID: mdl-36928510

Tay-Sachs disease is a lethal lysosomal storage disorder caused by mutations in the HexA gene encoding the α subunit of the lysosomal ß-hexosaminidase enzyme (HEXA). Abnormal GM2 ganglioside accumulation causes progressive deterioration in the central nervous system in Tay-Sachs patients. Hexa-/- mouse model failed to display abnormal phenotype. Recently, our group generated Hexa-/-Neu3-/- mouse showed severe neuropathological indications similar to Tay-Sachs patients. Despite excessive GM2 ganglioside accumulation in the brain and visceral organs, the regulation of autophagy has not been clarified yet in the Tay-Sachs disease mouse model. Therefore, we investigated distinct steps of autophagic flux using markers including LC3 and p62 in four different brain regions from the Hexa-/-Neu3-/- mice model of Tay-Sachs disease. Our data revealed accumulated autophagosomes and autophagolysosomes indicating impairment in autophagic flux in the brain. We suggest that autophagy might be a new therapeutic target for the treatment of devastating Tay-Sachs disease.


Autophagy , Tay-Sachs Disease , Animals , Mice , Autophagy/physiology , beta-N-Acetylhexosaminidases/genetics , beta-N-Acetylhexosaminidases/metabolism , Brain/metabolism , Brain/pathology , G(M2) Ganglioside/therapeutic use , Hexosaminidase A/metabolism , Tay-Sachs Disease/metabolism , Tay-Sachs Disease/pathology , Disease Models, Animal
3.
J Inherit Metab Dis ; 46(4): 687-694, 2023 07.
Article En | MEDLINE | ID: mdl-36700853

Treatment of monogenic disorders has historically relied on symptomatic management with limited ability to target primary molecular deficits. However, recent advances in gene therapy and related technologies aim to correct these underlying deficiencies, raising the possibility of disease management or even prevention for diseases that can be treated pre-symptomatically. Tay-Sachs disease (TSD) would be one such candidate, however very little is known about the presymptomatic stage of TSD. To better understand the effects of TSD on brain development, we evaluated the transcriptomes of human fetal brain samples with biallelic pathogenic variants in HEXA. We identified dramatic changes in the transcriptome, suggesting a perturbation of normal development. We also observed a shift in the expression of the sphingolipid metabolic pathway away from production of the HEXA substrate, GM2 ganglioside, presumptively to compensate for dysfunction of the enzyme. However, we do not observe transcriptomic signatures of end-stage disease, suggesting that developmental perturbations precede neurodegeneration. To our knowledge, this is the first report of the relationship between fetal disease pathology in juvenile onset TSD and the analysis of gene expression in fetal TSD tissues. This study highlights the need to better understand the "pre-symptomatic" stage of disease to set realistic expectations for patients receiving early therapeutic intervention.


Gangliosidoses, GM2 , Tay-Sachs Disease , Humans , Tay-Sachs Disease/genetics , Tay-Sachs Disease/metabolism , Tay-Sachs Disease/pathology , Gangliosidoses, GM2/genetics , Gangliosidoses, GM2/metabolism , Brain/pathology , Gene Expression
4.
Cells ; 10(11)2021 11 11.
Article En | MEDLINE | ID: mdl-34831346

AIMS: Tay-Sachs and Sandhoff diseases (GM2 gangliosidosis) are autosomal recessive disorders of lysosomal function that cause progressive neurodegeneration in infants and young children. Impaired hydrolysis catalysed by ß-hexosaminidase A (HexA) leads to the accumulation of GM2 ganglioside in neuronal lysosomes. Despite the storage phenotype, the role of autophagy and its regulation by mTOR has yet to be explored in the neuropathogenesis. Accordingly, we investigated the effects on autophagy and lysosomal integrity using skin fibroblasts obtained from patients with Tay-Sachs and Sandhoff diseases. RESULTS: Pathological autophagosomes with impaired autophagic flux, an abnormality confirmed by electron microscopy and biochemical studies revealing the accelerated release of mature cathepsins and HexA into the cytosol, indicating increased lysosomal permeability. GM2 fibroblasts showed diminished mTOR signalling with reduced basal mTOR activity. Accordingly, provision of a positive nutrient signal by L-arginine supplementation partially restored mTOR activity and ameliorated the cytopathological abnormalities. INNOVATION: Our data provide a novel molecular mechanism underlying GM2 gangliosidosis. Impaired autophagy caused by insufficient lysosomal function might represent a new therapeutic target for these diseases. CONCLUSIONS: We contend that the expression of autophagy/lysosome/mTOR-associated molecules may prove useful peripheral biomarkers for facile monitoring of treatment of GM2 gangliosidosis and neurodegenerative disorders that affect the lysosomal function and disrupt autophagy.


Arginine/pharmacology , Autophagy , Gangliosidoses, GM2/metabolism , TOR Serine-Threonine Kinases/metabolism , Autophagosomes/drug effects , Autophagosomes/metabolism , Autophagosomes/ultrastructure , Autophagy/drug effects , Cathepsins/metabolism , Fibroblasts/drug effects , Fibroblasts/metabolism , Fibroblasts/pathology , Hexosaminidase A/chemistry , Hexosaminidase A/metabolism , Hexosaminidase B/chemistry , Hexosaminidase B/metabolism , Humans , Lysosomes/drug effects , Lysosomes/metabolism , Mutation/genetics , Permeability , Proto-Oncogene Proteins c-akt/metabolism , Sandhoff Disease/pathology , Signal Transduction/drug effects , Tay-Sachs Disease/pathology , Transcriptome/genetics
5.
Proteins ; 89(11): 1587-1601, 2021 11.
Article En | MEDLINE | ID: mdl-34288098

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.


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
6.
Mol Genet Metab ; 133(3): 297-306, 2021 07.
Article En | MEDLINE | ID: mdl-34119419

Gangliosidoses are inherited lysosomal storage disorders caused by reduced or absent activity of either a lysosomal enzyme involved in ganglioside catabolism, or an activator protein required for the proper activity of a ganglioside hydrolase, which results in the intra-lysosomal accumulation of undegraded metabolites. We hereby describe morphological, ultrastructural, biochemical and genetic features of GM2 gangliosidosis in three captive bred wild boar littermates. The piglets were kept in a partially-free range farm and presented progressive neurological signs, starting at 6 months of age. Animals were euthanized at approximately one year of age due to their poor conditions. Neuropathogens were excluded as a possible cause of the signs. Gross examination showed a reduction of cerebral and cerebellar consistency. Central (CNS) and peripheral (PNS) nervous system neurons were enlarged and foamy, with severe and diffuse cytoplasmic vacuolization. Transmission electron microscopy (TEM) of CNS neurons demonstrated numerous lysosomes, filled by parallel or concentric layers of membranous electron-dense material, defined as membranous cytoplasmic bodies (MCB). Biochemical composition of gangliosides analysis from CNS revealed accumulation of GM2 ganglioside; furthermore, Hex A enzyme activity was less than 1% compared to control animals. These data confirmed the diagnosis of GM2 gangliosidosis. Genetic analysis identified, at a homozygous level, the presence of a missense nucleotide variant c.1495C > T (p Arg499Cys) in the hexosaminidase subunit alpha gene (HEXA), located within the GH20 hexosaminidase superfamily domain of the encoded protein. This specific HEXA variant is known to be pathogenic and associated with Tay-Sachs disease in humans, but has never been identified in other animal species. This is the first report of a HEXA gene associated Tay-Sachs disease in wild boars and provides a comprehensive description of a novel spontaneous animal model for this lysosomal storage disease.


Genetic Variation , Hexosaminidase A/genetics , Mutation, Missense , Sus scrofa/genetics , Tay-Sachs Disease/genetics , Tay-Sachs Disease/physiopathology , Animals , Cerebellum/pathology , Disease Models, Animal , Female , Gangliosidoses, GM2/metabolism , Hexosaminidase A/metabolism , Male , Tay-Sachs Disease/pathology , Whole Genome Sequencing
7.
Int J Mol Sci ; 21(18)2020 Sep 19.
Article En | MEDLINE | ID: mdl-32961778

Glycosphingolipids (GSLs) are a specialized class of membrane lipids composed of a ceramide backbone and a carbohydrate-rich head group. GSLs populate lipid rafts of the cell membrane of eukaryotic cells, and serve important cellular functions including control of cell-cell signaling, signal transduction and cell recognition. Of the hundreds of unique GSL structures, anionic gangliosides are the most heavily implicated in the pathogenesis of lysosomal storage diseases (LSDs) such as Tay-Sachs and Sandhoff disease. Each LSD is characterized by the accumulation of GSLs in the lysosomes of neurons, which negatively interact with other intracellular molecules to culminate in cell death. In this review, we summarize the biosynthesis and degradation pathways of GSLs, discuss how aberrant GSL metabolism contributes to key features of LSD pathophysiology, draw parallels between LSDs and neurodegenerative proteinopathies such as Alzheimer's and Parkinson's disease and lastly, discuss possible therapies for patients.


Gangliosides/metabolism , Glycosphingolipids/metabolism , Lysosomes/metabolism , Sandhoff Disease/metabolism , Tay-Sachs Disease/metabolism , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Alzheimer Disease/therapy , Animals , Humans , Lysosomes/pathology , Parkinson Disease/metabolism , Parkinson Disease/pathology , Parkinson Disease/therapy , Sandhoff Disease/pathology , Sandhoff Disease/therapy , Tay-Sachs Disease/pathology , Tay-Sachs Disease/therapy
8.
J Neuroinflammation ; 17(1): 277, 2020 Sep 20.
Article En | MEDLINE | ID: mdl-32951593

BACKGROUND: Tay-Sachs disease (TSD), a type of GM2-gangliosidosis, is a progressive neurodegenerative lysosomal storage disorder caused by mutations in the α subunit of the lysosomal ß-hexosaminidase enzyme. This disease is characterized by excessive accumulation of GM2 ganglioside, predominantly in the central nervous system. Although Tay-Sachs patients appear normal at birth, the progressive accumulation of undegraded GM2 gangliosides in neurons leads to death. Recently, an early onset Tay-Sachs disease mouse model, with genotype Hexa-/-Neu3-/-, was generated. Progressive accumulation of GM2 led to premature death of the double KO mice. Importantly, this double-deficient mouse model displays typical features of Tay-Sachs patients, such as cytoplasmic vacuolization of nerve cells, deterioration of Purkinje cells, neuronal death, deceleration in movement, ataxia, and tremors. GM2-gangliosidosis is characterized by acute neurodegeneration preceded by activated microglia expansion, macrophage, and astrocyte activation, along with the production of inflammatory mediators. However, the mechanism of disease progression in Hexa-/-Neu3-/- mice, relevant to neuroinflammation is poorly understood. METHOD: In this study, we investigated the onset and progression of neuroinflammatory changes in the cortex, cerebellum, and retina of Hexa-/-Neu3-/- mice and control littermates by using a combination of molecular genetics and immunochemical procedures. RESULTS: We found elevated levels of pro-inflammatory cytokine and chemokine transcripts, such as Ccl2, Ccl3, Ccl4, and Cxcl10 and also extensive microglial and astrocyte activation and proliferation, accompanied by peripheral blood mononuclear cell infiltration in the vicinity of neurons and oligodendrocytes. Behavioral tests demonstrated a high level of anxiety, and age-dependent loss in both spatial learning and fear memory in Hexa-/-Neu3-/- mice compared with that in the controls. CONCLUSION: Altogether, our data suggest that Hexa-/-Neu3-/- mice display a phenotype similar to Tay-Sachs patients suffering from chronic neuroinflammation triggered by GM2 accumulation. Furthermore, our work contributes to better understanding of the neuropathology in a mouse model of early onset Tay-Sachs disease.


Brain/metabolism , Disease Models, Animal , G(M2) Ganglioside/metabolism , Inflammation Mediators/metabolism , Retina/metabolism , Tay-Sachs Disease/metabolism , Animals , Brain/pathology , G(M2) Ganglioside/genetics , Leukocytes, Mononuclear/metabolism , Leukocytes, Mononuclear/pathology , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Knockout , Neurons/metabolism , Neurons/pathology , Retina/pathology , Tay-Sachs Disease/genetics , Tay-Sachs Disease/pathology
9.
J Gene Med ; 22(9): e3205, 2020 09.
Article En | MEDLINE | ID: mdl-32335981

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.


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.
Curr Gene Ther ; 18(2): 68-89, 2018.
Article En | MEDLINE | ID: mdl-29618308

Tay-Sachs disease, caused by impaired ß-N-acetylhexosaminidase activity, was the first GM2 gangliosidosis to be studied and one of the most severe and earliest lysosomal diseases to be described. The condition, associated with the pathological build-up of GM2 ganglioside, has acquired almost iconic status and serves as a paradigm in the study of lysosomal storage diseases. Inherited as a classical autosomal recessive disorder, this global disease of the nervous system induces developmental arrest with regression of attained milestones; neurodegeneration progresses rapidly to cause premature death in young children. There is no effective treatment beyond palliative care, and while the genetic basis of GM2 gangliosidosis is well established, the molecular and cellular events, from diseasecausing mutations and glycosphingolipid storage to disease manifestations, remain to be fully delineated. Several therapeutic approaches have been attempted in patients, including enzymatic augmentation, bone marrow transplantation, enzyme enhancement, and substrate reduction therapy. Hitherto, none of these stratagems has materially altered the course of the disease. Authentic animal models of GM2 gangliodidosis have facilitated in-depth evaluation of innovative applications such as gene transfer, which in contrast to other interventions, shows great promise. This review outlines current knowledge pertaining the pathobiology as well as potential innovative treatments for the GM2 gangliosidoses.


Bone Marrow Transplantation , Enzyme Replacement Therapy , Genetic Therapy , Sandhoff Disease/genetics , Sandhoff Disease/therapy , Tay-Sachs Disease/genetics , Tay-Sachs Disease/therapy , Animals , Disease Models, Animal , Glycosphingolipids/metabolism , Humans , Infant , Lysosomes/enzymology , Lysosomes/genetics , Lysosomes/pathology , Mice , Mutation , Rare Diseases , Sandhoff Disease/enzymology , Sandhoff Disease/pathology , Tay-Sachs Disease/enzymology , Tay-Sachs Disease/pathology , beta-N-Acetylhexosaminidases/genetics
12.
Exp Neurol ; 299(Pt A): 26-41, 2018 01.
Article En | MEDLINE | ID: mdl-28974375

Tay-Sachs disease is a severe lysosomal storage disorder caused by mutations in Hexa, the gene that encodes for the α subunit of lysosomal ß-hexosaminidase A (HEXA), which converts GM2 to GM3 ganglioside. Unexpectedly, Hexa-/- mice have a normal lifespan and show no obvious neurological impairment until at least one year of age. These mice catabolize stored GM2 ganglioside using sialidase(s) to remove sialic acid and form the glycolipid GA2, which is further processed by ß-hexosaminidase B. Therefore, the presence of the sialidase (s) allows the consequences of the Hexa defect to be bypassed. To determine if the sialidase NEU3 contributes to GM2 ganglioside degradation, we generated a mouse model with combined deficiencies of HEXA and NEU3. The Hexa-/-Neu3-/- mice were healthy at birth, but died at 1.5 to 4.5months of age. Thin-layer chromatography and mass spectrometric analysis of the brains of Hexa-/-Neu3-/- mice revealed the abnormal accumulation of GM2 ganglioside. Histological and immunohistochemical analysis demonstrated cytoplasmic vacuolation in the neurons. Electron microscopic examination of the brain, kidneys and testes revealed pleomorphic inclusions of many small vesicles and complex lamellar structures. The Hexa-/-Neu3-/- mice exhibited progressive neurodegeneration with neuronal loss, Purkinje cell depletion, and astrogliosis. Slow movement, ataxia, and tremors were the prominent neurological abnormalities observed in these mice. Furthermore, radiographs revealed abnormalities in the skeletal bones of the Hexa-/-Neu3-/- mice. Thus, the Hexa-/-Neu3-/- mice mimic the neuropathological and clinical abnormalities of the classical early-onset Tay-Sachs patients, and provide a suitable model for the future pre-clinical testing of potential treatments for this condition.


Gangliosidoses, GM2/genetics , Hexosaminidase B/genetics , Neuraminidase/genetics , Tay-Sachs Disease/genetics , Animals , Brain Chemistry/genetics , Cytoplasmic Vesicles/pathology , Gangliosidoses, GM2/metabolism , Gliosis/genetics , Gliosis/pathology , Glycosphingolipids/metabolism , Lameness, Animal/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Neuraminidase/deficiency , Neurons/pathology , Purkinje Cells/pathology , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization , Tay-Sachs Disease/pathology
15.
Anal Chem ; 89(4): 2622-2627, 2017 02 21.
Article En | MEDLINE | ID: mdl-28194950

Single nucleotide polymorphisms (SNPs) are the most fundamental internal causes for many genetic diseases. However, the location information on SNPs in a specific DNA sequence is not well acquired through current SNPs detection methods, except for accurate DNA sequencing. Here we report a fluorescence enhancement phenomenon in the process of two silver nanoclusters (AgNCs) approaching closely to form a nanocluster dimer (NCD). The fluorescence intensity is sensitive to the distance between two AgNCs; therefore, the NCD lights into different fluorescence intensities upon binding SNPs targets with mismatched bases at different positions. Interestingly, the fluorescence intensities of the NCD decrease linearly when the position of single mismatched base moves gradually from the middle point to the end of the target DNA. The NCD is a single probe acting as a universal platform to pinpoint various SNP positions. With this single probe, we cannot only identify the existence of SNPs but also pinpoint the location of a specific single mismatched base in the adjacent positions. This strategy is feasible to detect specific gene point mutations in clinical samples.


Metal Nanoparticles/chemistry , Polymorphism, Single Nucleotide , Spectrometry, Fluorescence , Base Pair Mismatch , DNA/blood , DNA/metabolism , DNA Probes/metabolism , Dimerization , Fluorescent Dyes/chemistry , Humans , Kinetics , Silver/chemistry , Tay-Sachs Disease/genetics , Tay-Sachs Disease/pathology
16.
Stem Cell Res ; 17(2): 289-291, 2016 09.
Article En | MEDLINE | ID: mdl-27879213

Human iPSC line TSD-01-hiPSC was generated from fibroblasts of a patient with infantile Tay-Sachs disease (TSD). The patient is compound heterozygous at the HEXA gene by carrying a 1278insTATC allele and an IVS12+1G>C allele. STEMCCA lentivirus, which expresses OCT4, SOX2, KLF4, and c-MYC from a polycistronic transcript, were used for reprogramming. TSD-01-hiPSC express pluripotency markers such as OCT4, SOX2, NANOG, Tra-1-60, and alkaline phosphatase, and can differentiate into tissues from all the three embryonic germ layers. This TSD patient-derived hiPSC line may serve as a valuable in vitro tool for disease modeling and drug test.


Cellular Reprogramming , Induced Pluripotent Stem Cells/cytology , Tay-Sachs Disease/pathology , beta-Hexosaminidase alpha Chain/genetics , Alleles , Animals , Base Sequence , Cell Differentiation , Cell Line , DNA Mutational Analysis , Fibroblasts/cytology , Fibroblasts/metabolism , Genotype , Heterozygote , Humans , Induced Pluripotent Stem Cells/metabolism , Induced Pluripotent Stem Cells/transplantation , Karyotype , Kruppel-Like Factor 4 , Male , Mice , Mice, Inbred NOD , Mutagenesis, Insertional , Tay-Sachs Disease/genetics , Tay-Sachs Disease/metabolism , Teratoma/pathology , Transcription Factors/genetics , Transcription Factors/metabolism , Transplantation, Heterologous
17.
Genet Med ; 18(12): 1214-1217, 2016 12.
Article En | MEDLINE | ID: mdl-27054707

PURPOSE: The Center for Jewish Genetics provides genetic education and carrier screening to individuals of Jewish descent. Carrier screening has traditionally been performed by targeted mutation analysis for founder mutations with an enzyme assay for Tay-Sachs carrier detection. The development of next-generation sequencing (NGS) allows for higher detection rates regardless of ethnicity. Here, we explore differences in carrier detection rates between genotyping and NGS in a primarily Jewish population. METHODS: Peripheral blood samples or saliva samples were obtained from 506 individuals. All samples were analyzed by sequencing, targeted genotyping, triplet-repeat detection, and copy-number analysis; the analyses were carried out at Counsyl. RESULTS: Of 506 individuals screened, 288 were identified as carriers of at least 1 condition and 8 couples were carriers for the same disorder. A total of 434 pathogenic variants were identified. Three hundred twelve variants would have been detected via genotyping alone. Although no additional mutations were detected by NGS in diseases routinely screened for in the Ashkenazi Jewish population, 26.5% of carrier results and 2 carrier couples would have been missed without NGS in the larger panel. CONCLUSION: In a primarily Jewish population, NGS reveals a larger number of pathogenic variants and provides individuals with valuable information for family planning.Genet Med 18 12, 1214-1217.


Genetic Carrier Screening , Genetic Testing , Tay-Sachs Disease/blood , Tay-Sachs Disease/genetics , Adult , Aged , Female , Heterozygote , High-Throughput Nucleotide Sequencing , Humans , Jews/genetics , Male , Middle Aged , Saliva/metabolism , Tay-Sachs Disease/pathology
19.
J Lipid Res ; 56(9): 1747-61, 2015 Sep.
Article En | MEDLINE | ID: mdl-26175473

Ganglioside GM2 is the major lysosomal storage compound of Tay-Sachs disease. It also accumulates in Niemann-Pick disease types A and B with primary storage of SM and with cholesterol in type C. Reconstitution of GM2 catabolism with ß-hexosaminidase A and GM2 activator protein (GM2AP) at uncharged liposomal surfaces carrying GM2 as substrate generated only a physiologically irrelevant catabolic rate, even at pH 4.2. However, incorporation of anionic phospholipids into the GM2 carrying liposomes stimulated GM2 hydrolysis more than 10-fold, while the incorporation of plasma membrane stabilizing lipids (SM and cholesterol) generated a strong inhibition of GM2 hydrolysis, even in the presence of anionic phospholipids. Mobilization of membrane lipids by GM2AP was also inhibited in the presence of cholesterol or SM, as revealed by surface plasmon resonance studies. These lipids also reduced the interliposomal transfer rate of 2-NBD-GM1 by GM2AP, as observed in assays using Förster resonance energy transfer. Our data raise major concerns about the usage of recombinant His-tagged GM2AP compared with untagged protein. The former binds more strongly to anionic GM2-carrying liposomal surfaces, increases GM2 hydrolysis, and accelerates intermembrane transfer of 2-NBD-GM1, but does not mobilize membrane lipids.


G(M2) Activator Protein/metabolism , G(M2) Ganglioside/metabolism , Liposomes/metabolism , Membrane Lipids/metabolism , Ceramides/metabolism , Cholesterol/genetics , Cholesterol/metabolism , Fluorescence Resonance Energy Transfer , G(M2) Activator Protein/genetics , HEK293 Cells , Humans , Hydrolysis/drug effects , Lysophospholipids/administration & dosage , Membrane Lipids/genetics , Monoglycerides/administration & dosage , Niemann-Pick Diseases/genetics , Niemann-Pick Diseases/metabolism , Niemann-Pick Diseases/pathology , Sphingomyelins/metabolism , Surface Plasmon Resonance , Tay-Sachs Disease/genetics , Tay-Sachs Disease/metabolism , Tay-Sachs Disease/pathology , beta-Hexosaminidase alpha Chain/metabolism
20.
Curr Mol Med ; 15(2): 138-45, 2015.
Article En | MEDLINE | ID: mdl-25732146

Methodologies for generating functional neuronal cells directly from human fibroblasts [induced neuronal (iN) cells] have been recently developed, but the research so far has only focused on technical refinements or recapitulation of known pathological phenotypes. A critical question is whether this novel technology will contribute to elucidation of novel disease mechanisms or evaluation of therapeutic strategies. Here we have addressed this question by studying Tay-Sachs disease, a representative lysosomal storage disease, and Dravet syndrome, a form of severe myoclonic epilepsy in infancy, using human iN cells with feature of immature postmitotic glutamatergic neuronal cells. In Tay-Sachs disease, we have successfully characterized canonical neuronal pathology, massive accumulation of GM2 ganglioside, and demonstrated the suitability of this novel cell culture for future drug screening. In Dravet syndrome, we have identified a novel functional phenotype that was not suggested by studies of classical mouse models and human autopsied brains. Taken together, the present study demonstrates that human iN cells are useful for translational neuroscience research to explore novel disease mechanisms and evaluate therapeutic compounds. In the future, research using human iN cells with well-characterized genomic landscape can be integrated into multidisciplinary patient-oriented research on neuropsychiatric disorders to address novel disease mechanisms and evaluate therapeutic strategies.


Epilepsies, Myoclonic/metabolism , Fibroblasts/metabolism , G(M2) Ganglioside/metabolism , Neurons/metabolism , Tay-Sachs Disease/metabolism , 1-Deoxynojirimycin/analogs & derivatives , 1-Deoxynojirimycin/pharmacology , Action Potentials/drug effects , Basic Helix-Loop-Helix Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/metabolism , Cell Differentiation , Epilepsies, Myoclonic/pathology , Fibroblasts/drug effects , Fibroblasts/pathology , Gene Expression , Genetic Vectors/chemistry , Genetic Vectors/metabolism , Glycoside Hydrolase Inhibitors/pharmacology , Humans , Lentivirus/genetics , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Neurons/drug effects , Neurons/pathology , Plasmids/chemistry , Plasmids/metabolism , Primary Cell Culture , Tay-Sachs Disease/pathology , Transcription Factors/genetics , Transcription Factors/metabolism , Transduction, Genetic , Transgenes
...