Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 57
Filter
Add more filters

Publication year range
1.
Nat Immunol ; 21(7): 802-815, 2020 07.
Article in English | MEDLINE | ID: mdl-32541832

ABSTRACT

Microglia and central nervous system (CNS)-associated macrophages (CAMs), such as perivascular and meningeal macrophages, are implicated in virtually all diseases of the CNS. However, little is known about their cell-type-specific roles in the absence of suitable tools that would allow for functional discrimination between the ontogenetically closely related microglia and CAMs. To develop a new microglia gene targeting model, we first applied massively parallel single-cell analyses to compare microglia and CAM signatures during homeostasis and disease and identified hexosaminidase subunit beta (Hexb) as a stably expressed microglia core gene, whereas other microglia core genes were substantially downregulated during pathologies. Next, we generated HexbtdTomato mice to stably monitor microglia behavior in vivo. Finally, the Hexb locus was employed for tamoxifen-inducible Cre-mediated gene manipulation in microglia and for fate mapping of microglia but not CAMs. In sum, we provide valuable new genetic tools to specifically study microglia functions in the CNS.


Subject(s)
Brain/pathology , Encephalomyelitis, Autoimmune, Experimental/pathology , Facial Nerve Injuries/pathology , Microglia/metabolism , beta-Hexosaminidase beta Chain/metabolism , Animals , Brain/cytology , Brain/immunology , CRISPR-Cas Systems/genetics , Encephalomyelitis, Autoimmune, Experimental/immunology , Facial Nerve Injuries/immunology , Gene Knock-In Techniques , Genes, Reporter/genetics , Genetic Loci/genetics , Humans , Intravital Microscopy , Luminescent Agents/chemistry , Luminescent Proteins/chemistry , Luminescent Proteins/genetics , Macrophages/immunology , Macrophages/metabolism , Mice , Microglia/immunology , NIH 3T3 Cells , RNA-Seq , Single-Cell Analysis , Transfection , beta-Hexosaminidase beta Chain/genetics , Red Fluorescent Protein
2.
BMC Neurol ; 23(1): 240, 2023 Jun 21.
Article in English | MEDLINE | ID: mdl-37344817

ABSTRACT

BACKGROUND: Sandhoff disease (SD) is a rare neurological disease with high clinical heterogeneity. SD in juvenile form is much rarer and it is often misdiagnosed in clinics. Therein, it is necessary to provide more cases and review the literature on juvenile onset SD. CASE PRESENTATION: A 14 years-old boy with eight years of walking difficulties, and was ever misdiagnosed as spinocerebellar ataxia. We found this patient after genetic testing carried rs201580118 and a novel gross deletion in HEXB (g.74012742_74052694del). Through review the literature, we found that was the first gross deletion identified at the 3'end of HEXB, associated with juvenile onset SD from China. CONCLUSION: This case expanded our knowledge about the genotype and phenotype correlations in SD. Comprehensive genetic testing is important for the diagnosis of unexplained ataxia.


Subject(s)
Sandhoff Disease , Humans , Sandhoff Disease/diagnosis , Sandhoff Disease/genetics , beta-Hexosaminidase beta Chain/genetics , Genetic Testing , Genotype , Phenotype , Mutation
3.
Metab Brain Dis ; 37(8): 2669-2675, 2022 12.
Article in English | MEDLINE | ID: mdl-36190588

ABSTRACT

Sandhoff disease is a rare neurodegenerative and autosomal recessive disorder, which is characterized by a defect in ganglioside metabolism. Also, it is caused by mutations in the HEXB gene for the ß-subunit isoform 1 of ß-N-acetyl hexosaminidase. In the present study, an Iranian 14- month -old girl with 8- month history of unsteady walking and involuntary movements was described. In this regard, biochemical testing showed some defects in the normal activity of beta-hexosaminidase protein. Following sequencing of HEXB gene, a homozygous c.833C > T mutation was identified in the patient's genome. After recognition of p.A278V, several different in silico methods were used to assess the mutant protein stability, ranging from mutation prediction methods to ligand docking. The p.A278V mutation might be disruptive because of changing the three-dimensional folding at the end of the 5th alpha helix. According to the medical prognosis, in silico and structural analyses, it was predicted to be disease cause.


Subject(s)
Sandhoff Disease , Female , Humans , Sandhoff Disease/genetics , Sandhoff Disease/metabolism , Iran , Mutation , Homozygote , beta-Hexosaminidase beta Chain/genetics
4.
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
5.
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
6.
Mol Ther ; 27(8): 1495-1506, 2019 08 07.
Article in English | MEDLINE | ID: mdl-31208914

ABSTRACT

Neuronopathic glycosphingolipidoses are a sub-group of lysosomal storage disorders for which there are presently no effective therapies. Here, we evaluated the potential of substrate reduction therapy (SRT) using an inhibitor of glucosylceramide synthase (GCS) to decrease the synthesis of glucosylceramide (GL1) and related glycosphingolipids. The substrates that accumulate in Sandhoff disease (e.g., ganglioside GM2 and its nonacylated derivative, lyso-GM2) are distal to the drug target, GCS. Treatment of Sandhoff mice with a GCS inhibitor that has demonstrated CNS access (Genz-682452) reduced the accumulation of GL1 and GM2, as well as a variety of disease-associated substrates in the liver and brain. Concomitant with these effects was a significant decrease in the expression of CD68 and glycoprotein non-metastatic melanoma B protein (Gpnmb) in the brain, indicating a reduction in microgliosis in the treated mice. Moreover, using in vivo imaging, we showed that the monocytic biomarker translocator protein (TSPO), which was elevated in Sandhoff mice, was normalized following Genz-682452 treatment. These positive effects translated in turn into a delay (∼28 days) in loss of motor function and coordination, as measured by rotarod latency, and a significant increase in longevity (∼17.5%). Together, these results support the development of SRT for the treatment of gangliosidoses, particularly in patients with residual enzyme activity.


Subject(s)
Carbamates/pharmacology , Enzyme Inhibitors/pharmacology , Glucosyltransferases/antagonists & inhibitors , Quinuclidines/pharmacology , Sandhoff Disease/enzymology , Animals , Brain/drug effects , Brain/metabolism , Brain/pathology , Disease Models, Animal , Female , Glucosyltransferases/genetics , Glucosyltransferases/metabolism , Ligands , Liver/drug effects , Liver/metabolism , Liver/pathology , Male , Mass Spectrometry , Mice , Mice, Knockout , Molecular Imaging , Receptors, GABA/metabolism , Sandhoff Disease/diagnosis , Sandhoff Disease/genetics , Sandhoff Disease/therapy , Sphingolipids/metabolism , beta-Hexosaminidase beta Chain/genetics , beta-Hexosaminidase beta Chain/metabolism
7.
Glia ; 67(9): 1705-1718, 2019 09.
Article in English | MEDLINE | ID: mdl-31140649

ABSTRACT

Sphingolipidoses are severe, mostly infantile lysosomal storage disorders (LSDs) caused by defective glycosphingolipid degradation. Two of these sphingolipidoses, Tay Sachs and Sandhoff diseases, are caused by ß-Hexosaminidase (HEXB) enzyme deficiency, resulting in ganglioside (GM2) accumulation and neuronal loss. The precise sequence of cellular events preceding, and leading to, neuropathology remains unclear, but likely involves inflammation and lysosomal accumulation of GM2 in multiple cell types. We aimed to determine the consequences of Hexb activity loss for different brain cell types using zebrafish. Hexb deficient zebrafish (hexb-/- ) showed lysosomal abnormalities already early in development both in radial glia, which are the neuronal and glial progenitors, and in microglia. Additionally, at 5 days postfertilization, hexb-/- zebrafish showed reduced locomotor activity. Although specific oligosaccharides accumulate in the adult brain, hexb-/- ) zebrafish are viable and apparently resistant to Hexb deficiency. In all, we identified cellular consequences of loss of Hexb enzyme activity during embryonic brain development, showing early effects on glia, which possibly underlie the behavioral aberrations. Hereby, we identified clues into the contribution of non-neuronal lysosomal abnormalities in LSDs affecting the brain and provide a tool to further study what underlies the relative resistance to Hexb deficiency in vivo.


Subject(s)
Brain/enzymology , Brain/growth & development , Lysosomes/enzymology , Neuroglia/enzymology , beta-Hexosaminidase beta Chain/genetics , Animals , Animals, Genetically Modified , Apoptosis/physiology , Brain/pathology , Disease Models, Animal , Lysosomes/pathology , Motor Activity/physiology , Neuroglia/pathology , Sphingolipidoses/enzymology , Zebrafish
8.
Hum Mol Genet ; 26(4): 661-673, 2017 02 15.
Article in English | MEDLINE | ID: mdl-28007910

ABSTRACT

GM2 gangliosidoses are a group of lysosomal storage disorders which include Sandhoff disease and Tay-Sachs disease. Dysregulation of glutamate receptors has been recently postulated in the pathology of Sandhoff disease. Glutamate receptor association with neuronal pentraxins 1 and 2, and the neuronal pentraxin receptor facilitates receptor potentiation and synaptic shaping. In this study, we have observed an upregulation of a novel form of neuronal pentraxin 1 (NP1-38) in the brains of a mouse model of Sandhoff disease and Tay-Sachs disease. In order to determine the impact of NP1 on the pathophysiology of Sandhoff disease mouse models, we have generated an Np1-/-Hexb-/- double knockout mouse, and observed extended lifespan, improved righting reflex and enhanced body condition relative to Hexb-/- mice, with no effect on gliosis or apoptotic markers in the CNS. Sandhoff mouse brain slices reveals a reduction in AMPA receptor-mediated currents, and increased variability in total glutamate currents in the CA1 region of the hippocampus; Np1-/-Hexb-/- mice show a correction of this phenotype, suggesting NP1-38 may be interfering with glutamate receptor function. Indeed, some of the psychiatric aspects of Sandhoff and Tay-Sachs disease (particularly late onset) may be attributed to a dysfunctional hippocampal glutamatergic system. Our work highlights a potential role for synaptic proteins, such as NP1 and glutamate receptors in lysosomal storage diseases.


Subject(s)
C-Reactive Protein/biosynthesis , CA1 Region, Hippocampal/metabolism , Nerve Tissue Proteins/biosynthesis , Sandhoff Disease/metabolism , Up-Regulation , beta-Hexosaminidase beta Chain/biosynthesis , Animals , C-Reactive Protein/genetics , CA1 Region, Hippocampal/pathology , Humans , Mice , Mice, Knockout , Nerve Tissue Proteins/genetics , Sandhoff Disease/pathology , beta-Hexosaminidase beta Chain/genetics
9.
BMC Med Genet ; 20(1): 199, 2019 12 18.
Article in English | MEDLINE | ID: mdl-31852446

ABSTRACT

BACKGROUND: Neurological disorders are a common cause of morbidity and mortality within Pakistani populations. It is one of the most important challenges in healthcare, with significant life-long socio-economic burden. METHODS: We investigated the cause of disease in three Pakistani families in individuals with unexplained autosomal recessive neurological conditions, using both genome-wide SNP mapping and whole exome sequencing (WES) of affected individuals. RESULTS: We identified a homozygous splice site variant (NM_000521:c.445 + 1G > T) in the hexosaminidase B (HEXB) gene confirming a diagnosis of Sandhoff disease (SD; type II GM2-gangliosidosis), an autosomal recessive lysosomal storage disorder caused by deficiency of hexosaminidases in a single family. In two further unrelated families, we identified a homozygous frameshift variant (NM_024298.3:c.758_778del; p.Glu253_Ala259del) in membrane-bound O-acyltransferase family member 7 (MBOAT7) as the likely cause of disease. MBOAT7 gene variants have recently been identified as a cause of intellectual disability (ID), seizures and autistic features. CONCLUSIONS: We identified two metabolic disorders of lipid biosynthesis within three Pakistani families presenting with undiagnosed neurodevelopmental conditions. These findings enabled an accurate neurological disease diagnosis to be provided for these families, facilitating disease management and genetic counselling within this population. This study consolidates variation within MBOAT7 as a cause of neurodevelopmental disorder, broadens knowledge of the clinical outcomes associated with MBOAT7-related disorder, and confirms the likely presence of a regionally prevalent founder variant (c.758_778del; p.Glu253_Ala259del) in Pakistan.


Subject(s)
Acyltransferases/genetics , Homozygote , Membrane Proteins/genetics , Nervous System Diseases/genetics , beta-Hexosaminidase beta Chain/genetics , Consanguinity , Electroencephalography , Female , Genes, Recessive , Humans , Infant , Magnetic Resonance Imaging , Male , Mutation , Nervous System Diseases/diagnostic imaging , Nervous System Diseases/physiopathology , Pakistan , Polymorphism, Single Nucleotide , Exome Sequencing
10.
Zhonghua Yi Xue Yi Chuan Xue Za Zhi ; 36(9): 930-934, 2019 Sep 10.
Article in Zh | MEDLINE | ID: mdl-31515793

ABSTRACT

OBJECTIVE: To detect potential mutations of HEXB gene in an infant with Sandhoff disease (SD). METHODS: Genomic DNA was extracted from peripheral blood sample of the infant. All coding exons (exons 1 to 14) and splicing sites of the HEXB gene were subjected to PCR amplification and direct sequencing.PubMed Protein BLAST system was employed to analyze cross-species conservation of the mutant amino acid. PubMed BLAST CD-search was performed to identify functional domains destroyed by thecandidate mutations. Impact of the mutations was analyzed with software including PolyPhen-2, Mutation Taster and SIFT. Whole-exome sequencing was carried out to identify additional mutations. RESULTS: The infant was found to carry compound heterozygous mutations c.1652G>A(p.Cys551Tyr) and c.1389C>G (p.Tyr463*) of the HEXB gene. The c.1389C>G (p.Tyr463*) mutation may lead to destruction of two functional domains in ß subunit of the Hex protein. The c.1652G>A(p.Cys551Tyr) mutation, unreported previously,was predicted to be probably damaging by Bioinformatic analysis. CONCLUSION: Compound heterozygous mutations c.1652G>A(p.Cys551Tyr) and c.1389C>G (p.Tyr463*) in the HEXB gene probably underlie the disease in this patient.


Subject(s)
Sandhoff Disease/genetics , beta-Hexosaminidase beta Chain/genetics , DNA Mutational Analysis , Exons , Heterozygote , Humans , Infant , Mutation , Polymerase Chain Reaction
11.
J Hum Genet ; 61(2): 163-6, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26582265

ABSTRACT

Sandhoff disease (SD) is an autosomal recessive neurodegenerative lysosomal storage disorder caused by mutations in HEXB gene. Molecular pathology is unknown in Indian patients with SD. The present study is aimed to determine mutations spectrum and molecular pathology leading to SD in 22 unrelated patients confirmed by the deficiency of ß-hexosaminidase-A and total-hexosaminidase in leukocytes. To date, nearly 86 mutations of HEXB have been described, including five large deletions. Over all we have identified 13 mutations in 19 patients, eight of which were novel, including two missense mutations [c.611G>A (p.G204E), c. 634A>T (p.H212Y)], two nonsense mutations [c.333G>A (p.W111X), c.298C>T (p.R100X)], one splice site mutation c.1082+5 G>T, two small in-frame deletions [c.534_541delAGTTTATC (p.V179RfsX10), c.1563_1573delTATGGATGACG (p.M522LfsX2)] and one insertion c.1553_1554insAAGA (p.D518EfsX8). We have also identified previously known, five sequence variations leading to amino acid changes [c.926G>A (p.C309Y), c.1597C>T (p.R533C)], one nonsense mutation c.850 C>T (p.R284X), one splice site mutation c.1417+1 G-A and one insertion c.1591_1592insC (p.R531TfsX22). Mutation was not identified in three patients. We observed from this study that mutation c.850C>T (p.R284X) was identified in 4/19 (21%) patients which is likely to be the most common mutation in the country. This is the first study providing insight into the molecular basis of SD in India.


Subject(s)
Mutation , Sandhoff Disease/genetics , beta-Hexosaminidase beta Chain/genetics , Child, Preschool , Humans , Infant , Sandhoff Disease/diagnosis , Sandhoff Disease/enzymology , beta-Hexosaminidase beta Chain/metabolism
12.
Mol Ther ; 23(3): 414-22, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25515709

ABSTRACT

G(M2) gangliosidoses are severe neurodegenerative disorders resulting from a deficiency in ß-hexosaminidase A activity and lacking effective therapies. Using a Sandhoff disease (SD) mouse model (Hexb(-/-)) of the G(M2) gangliosidoses, we tested the potential of systemically delivered adeno-associated virus 9 (AAV9) expressing Hexb cDNA to correct the neurological phenotype. Neonatal or adult SD and normal mice were intravenously injected with AAV9-HexB or -LacZ and monitored for serum ß-hexosaminidase activity, motor function, and survival. Brain G(M2) ganglioside, ß-hexosaminidase activity, and inflammation were assessed at experimental week 43, or an earlier humane end point. SD mice injected with AAV9-LacZ died by 17 weeks of age, whereas all neonatal AAV9-HexB-treated SD mice survived until 43 weeks (P < 0.0001) with only three exhibiting neurological dysfunction. SD mice treated as adults with AAV9-HexB died between 17 and 35 weeks. Neonatal SD-HexB-treated mice had a significant increase in brain ß-hexosaminidase activity, and a reduction in G(M2) ganglioside storage and neuroinflammation compared to adult SD-HexB- and SD-LacZ-treated groups. However, at 43 weeks, 8 of 10 neonatal-HexB injected control and SD mice exhibited liver or lung tumors. This study demonstrates the potential for long-term correction of SD and other G(M2) gangliosidoses through early rAAV9 based systemic gene therapy.


Subject(s)
Dependovirus/genetics , G(M2) Ganglioside/metabolism , Genetic Therapy/methods , Genetic Vectors/administration & dosage , Sandhoff Disease/therapy , beta-Hexosaminidase beta Chain/genetics , Age Factors , Animals , Animals, Newborn , Brain/enzymology , Brain/pathology , Disease Models, Animal , Female , Gene Expression , Genetic Vectors/adverse effects , Inflammation/genetics , Inflammation/mortality , Inflammation/pathology , Inflammation/therapy , Injections, Intravenous , Lac Operon , Liver Neoplasms/etiology , Liver Neoplasms/pathology , Lung Neoplasms/etiology , Lung Neoplasms/pathology , Lysosomes/enzymology , Lysosomes/pathology , Male , Mice , Mice, Knockout , Motor Activity/genetics , Sandhoff Disease/genetics , Sandhoff Disease/mortality , Sandhoff Disease/pathology , Survival Analysis , beta-Hexosaminidase beta Chain/metabolism
13.
Metab Brain Dis ; 31(4): 861-7, 2016 08.
Article in English | MEDLINE | ID: mdl-27021291

ABSTRACT

Sandhoff disease (SD) is a rare autosomal recessive lysosomal storage disorder of sphingolipid metabolism resulting from the deficiency of ß-hexosaminidase (HEX). Mutations of the HEXB gene cause Sandhoff disease. In order to improve the diagnosis and expand the knowledge of the disease, we collected and analyzed relevant data of clinical diagnosis, biochemical investigation, and molecular mutational analysis in five Chinese patients with SD. The patients presented with heterogenous symptoms of neurologic deterioration. HEX activity in leukocytes was severely deficient. We identified seven different mutations, including three known mutations: IVS12-26G > A, p.T209I, p.I207V, and four novel mutations: p.P468PfsX62, p.L223P, p.Y463X, p.G549R. We also detected two different heterozygous mutations c.-122delC and c.-126C > T in the promoter which were suspected to be deleterious mutations. We attempted to correlate these mutations with the clinical presentation of the patients. Our study indicates that the mutation p.T209I and p.P468PfsX62 may link to the infantile form of SD. Our study expands the spectrum of genotype of SD in China, provides new insights into the molecular mechanism of SD and helps to the diagnosis and treatment of this disease.


Subject(s)
Mutation , Sandhoff Disease/diagnosis , beta-Hexosaminidase beta Chain/genetics , Child , DNA Mutational Analysis , Female , Humans , Infant , Male , Pedigree , Promoter Regions, Genetic , Sandhoff Disease/genetics , Sandhoff Disease/metabolism , Symptom Assessment
14.
Mol Genet Metab ; 111(3): 382-389, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24461908

ABSTRACT

Sandhoff disease is a rare progressive neurodegenerative genetic disorder with a high incidence among certain isolated communities and ethnic groups around the world. Previous reports have shown a high occurrence of Sandhoff disease in northern Saskatchewan. Newborn screening cards from northern Saskatchewan were retrospectively screened in order to investigate the incidence and determine the carrier frequency of Sandhoff disease in these communities. PCR-based screening was conducted for the c.115delG (p.(Val39fs)) variant in the HEXB gene that was previously found in 4 Sandhoff disease patients from this area. The carrier frequency for this allele was estimated to be ~1:27. MS/MS-based screening of hexosaminidase activity along with genetic sequencing allowed for the identification of additional variants based on low total hexosaminidase activity and high % hexosaminidase A activity relative to c.115delG carriers. In total 4 pathogenic variants were discovered in the population (c.115delG, c.619A>G, c.1601G>T, and c.1652G>A) of which two are previously unreported (c.1601G>T and c.1652G>A). The combined carrier frequency of these alleles in the study area was estimated at ~1:15. Based on the number of cases of Sandhoff disease from this area we estimate the incidence to be ~1:390 corresponding to a child being born with the disease every 1-2 years on average. The results from our study were then compared with variants in the HEXB gene from the genomes available from the 1000 Genomes project. A total of 19 HEXB variants were found in the 1092 genomes of which 5 are suspected of having a deleterious effect on hexosaminidase activity. The estimated carrier frequency of Sandhoff disease in Saskatchewan at 1:15 is more than 3 times higher than the carrier frequency in the global sample provided by the 1000 Genomes project at 1:57.


Subject(s)
Heterozygote , Neonatal Screening , Sandhoff Disease/genetics , beta-Hexosaminidase beta Chain/genetics , Child , Female , Humans , Infant, Newborn , Male , Molecular Biology/methods , Mutation , Retrospective Studies , Sandhoff Disease/diagnosis , Sandhoff Disease/epidemiology , Saskatchewan , Substrate Specificity , Tandem Mass Spectrometry
15.
Mol Cell Probes ; 27(1): 32-7, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23010210

ABSTRACT

Multiplex ligation dependent probe amplification (MLPA) assays were designed for the genes HEXB (OMIM: 606873), GM2A (OMIM: 613109) and SMARCAL1 (OMIM: 606622) of humans. Two sets of synthetic MLPA probes for these coding exons were tested. Changes in copy numbers were detected as well as single nucleotide polymorphisms (SNPs) by complementary DNA sequence analyses. The MLPA method was shown to be reliable for mutation detection and identified five published and 12 new mutations. In all cases from a Morbus Sandhoff cohort of patients, exclusively one variation in copy number was observed and linked to a nucleotide alteration called c.1614-14C>A. This deletion comprised exons 1-5. One of these cases is described in detail. Deletions were neither detected in the GM2A nor the SMARCAL1 genes. The MLPA assays complement routine diagnostics for M. Sandhoff (OMIM: 268800), M. Tay-Sachs variant AB (OMIM: 272750) and Schimke immuno-osseous dysplasia (OMIM: 242900).


Subject(s)
Arteriosclerosis/genetics , Immunologic Deficiency Syndromes/genetics , Nephrotic Syndrome/genetics , Osteochondrodysplasias/genetics , Pulmonary Embolism/genetics , Sandhoff Disease/genetics , Tay-Sachs Disease, AB Variant/genetics , Arteriosclerosis/diagnosis , Base Sequence , DNA Copy Number Variations , DNA Helicases/genetics , G(M2) Activator Protein/genetics , Humans , Immunologic Deficiency Syndromes/diagnosis , Multiplex Polymerase Chain Reaction , Mutation , Nephrotic Syndrome/diagnosis , Osteochondrodysplasias/diagnosis , Polymorphism, Single Nucleotide , Primary Immunodeficiency Diseases , Pulmonary Embolism/diagnosis , Sandhoff Disease/diagnosis , Sequence Analysis, DNA , Sequence Deletion , Tay-Sachs Disease, AB Variant/diagnosis , beta-Hexosaminidase beta Chain/genetics
16.
Zhejiang Da Xue Xue Bao Yi Xue Ban ; 42(4): 403-10, 2013 07.
Article in Zh | MEDLINE | ID: mdl-24022928

ABSTRACT

OBJECTIVE: To investigate the phenotype and genotype of a Chinese boy and his family affected by infantile Sandhoff disease. METHODS: The proband, a boy, was the first child born to a non-consanguineous couple. He showed startle reaction after birth and progressive psychomotor regression from the age of 8 months. From the age of 16 months, he presented seizures. When he was admitted at 17 months old, severe mental retardation and weakness were observed. Fundus examination revealed bilateral cherry-red spots in the macula and optic atrophy. Cranial MRI revealed abnormal signals in the thalamus, basal ganglia and white matter. Enzymatic assay and genetic testing were performed for the diagnosis. His mother visited us at 18 weeks of pregnancy seeking for prenatal diagnosis. HEXB gene diagnosis to the fetus was performed by direct sequencing. RESULTS: Significant deficient total ß-hexosaminidase (A and B) activity in peripheral leucocytes of the patient (0.0 nmol/h/mg compared with normal control, 41.9 to 135.1 nmol/h/mg) supported the diagnosis of Sandhoff disease. On his HEXB gene, two mutations were found. c.1645G-A (p.G549R) was novel. c.IVS7-48T was a reported mutation. Now, the patient was 2 years and 3 months old, with progressive general failure, severe epilepsy, blindness and hypermyotonia. Subsequently, the mother visited us at 18 weeks of pregnancy seeking for prenatal diagnosis. HEXB gene analysis of the amniocytes was performed by direct sequencing. Both of the two mutations were not detected from cultured amniocytes. The result revealed that the fetus was not affected by Sandhoff disease. A healthy girl, the sibling of the proband, was born in term. Postnatal enzyme analysis and genetic analysis of the cord blood cells confirmed the prenatal diagnosis. CONCLUSION: One novel mutation on HEXB gene was identified. Prenatal diagnosis to the fetus of this family was performed by amniocytes gene analysis.


Subject(s)
Prenatal Diagnosis , Sandhoff Disease/diagnosis , Sandhoff Disease/genetics , beta-Hexosaminidase beta Chain/genetics , Adult , Amniotic Fluid/cytology , Child, Preschool , DNA Mutational Analysis , Female , Genetic Testing , Humans , Male , Mutation , Pregnancy
17.
Medicine (Baltimore) ; 102(24): e33890, 2023 Jun 16.
Article in English | MEDLINE | ID: mdl-37327298

ABSTRACT

BACKGROUND: Sandhoff disease (SD, Online Mendelian Inheritance in Man: 268800) is an autosomal recessive lysosomal storage disorder caused by variants of the ß-hexosaminidase B (HEXB) gene (Online Mendelian Inheritance in Man: 606873). The HEXB gene has been mapped to chromosome 5q13 and contains 14 exons. The symptoms of SD include progressive weakness, intellectual disability, visual and hearing impairment, exaggerated startle response, and seizures; the patients usually die before the age of 3 years.[1]. CASE SUMMARY: We present a case of SD caused by a homozygous frameshift mutation in the HEXB gene, c.118delG (p.A40fs*24). The male child, aged 2 years 7 months, showed movement retrogression with orbital hypertelorism at age 2 years, accompanied by seizures. Magnetic resonance imaging of the head showed cerebral atrophy and delayed myelination of the white matter of the brain. CONCLUSION: A novel homozygous frameshift c.118delG (p.A40fs*24) variant of HEXB has caused SD in the child. The major symptoms are intellectual disability, visual and hearing impairment, and seizures. Investigation will be continued in the future to comprehensively describe the genotype/phenotype and gain information on other associated features to understand the variable expressivity of this condition.


Subject(s)
Intellectual Disability , Sandhoff Disease , Humans , Male , beta-Hexosaminidase beta Chain/genetics , beta-N-Acetylhexosaminidases/genetics , Frameshift Mutation , Hexosaminidase B/genetics , Mutation , Sandhoff Disease/diagnosis , Sandhoff Disease/genetics , Seizures , Child, Preschool
18.
Mol Ther ; 19(6): 1017-24, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21487393

ABSTRACT

To develop a novel enzyme replacement therapy for neurodegenerative Tay-Sachs disease (TSD) and Sandhoff disease (SD), which are caused by deficiency of ß-hexosaminidase (Hex) A, we designed a genetically engineered HEXB encoding the chimeric human ß-subunit containing partial amino acid sequence of the α-subunit by structure-based homology modeling. We succeeded in producing the modified HexB by a Chinese hamster ovary (CHO) cell line stably expressing the chimeric HEXB, which can degrade artificial anionic substrates and GM2 ganglioside in vitro, and also retain the wild-type (WT) HexB-like thermostability in the presence of plasma. The modified HexB was efficiently incorporated via cation-independent mannose 6-phosphate receptor into fibroblasts derived from Tay-Sachs patients, and reduced the GM2 ganglioside accumulated in the cultured cells. Furthermore, intracerebroventricular administration of the modified HexB to Sandhoff mode mice restored the Hex activity in the brains, and reduced the GM2 ganglioside storage in the parenchyma. These results suggest that the intracerebroventricular enzyme replacement therapy involving the modified HexB should be more effective for Tay-Sachs and Sandhoff than that utilizing the HexA, especially as a low-antigenic enzyme replacement therapy for Tay-Sachs patients who have endogenous WT HexB.


Subject(s)
G(M2) Ganglioside/metabolism , beta-Hexosaminidase beta Chain/chemistry , beta-Hexosaminidase beta Chain/metabolism , Animals , CHO Cells , Cells, Cultured , Cricetinae , Cricetulus , Humans , Immunoblotting , Mice , Models, Molecular , Protein Structure, Secondary , Sandhoff Disease/drug therapy , Tay-Sachs Disease/drug therapy , beta-Hexosaminidase beta Chain/genetics , beta-Hexosaminidase beta Chain/therapeutic use
19.
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
20.
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
SELECTION OF CITATIONS
SEARCH DETAIL