Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 114
1.
Yakugaku Zasshi ; 143(1): 65-75, 2023.
Article Ja | MEDLINE | ID: mdl-36596541

Sandhoff disease (SD) is a glycosphingolipid storage disease resulting from a genetic mutation in HEXB and associated deficiency in ß-hexosaminidase activity. This defect causes abnormal accumulation of ganglioside GM2 and related glycolipids in lysosomes, resulting in progressive deterioration of the central nervous system. Hexb-knockout (Hexb-/-) mice, an established animal model, show abnormalities similar to the severe phenotype seen in human infants. We used iPS cells derived from this mouse model (SD-iPSCs) to examine abnormal neuronal lineage differentiation and development in vitro during the asymptomatic phase of SD. Differentiation ability along the time axis appears to be altered in SD-iPSCs in which the differentiation ability of neural stem cells is promoted and differentiation into neurons is completed earlier, while the timing of differentiation into astrocytes is accelerated. This abnormal differentiation was suppressed by introducing the Hexb gene. These results indicate that the abnormal differentiation of SD-iPSCs into the nervous system reflects the pathogenesis of SD. Analysis using Hexb-/- mice revealed that activated microglia causes astrogliosis at the early stage of development that can be ameliorated via immunosuppression. Furthermore, reactive astrocytes in the cortex of Hexb-/- mice express adenosine A2A receptors in the late inflammatory phase. Inhibition of this receptor resulted in a decrease in activated microglial cells and inflammatory cytokines/chemokines. These results suggest that the astrocyte A2A receptor is important as a sensor that regulates microglial activation in the late inflammatory phase. Thus, our results provide new insights into the complex pathogenesis of SD.


Neural Stem Cells , Sandhoff Disease , Humans , Mice , Animals , Sandhoff Disease/genetics , Sandhoff Disease/pathology , Mice, Knockout , Neurons/pathology , Astrocytes/pathology , Disease Models, Animal
2.
Genes (Basel) ; 13(11)2022 11 03.
Article En | MEDLINE | ID: mdl-36360256

Sandhoff disease (SD) is a fatal neurodegenerative disorder belonging to the family of diseases called GM2 Gangliosidosis. There is no curative treatment of SD. The molecular pathogenesis of SD is still unclear though it is clear that the pathology initiates with the build-up of ganglioside followed by microglial activation, inflammation, demyelination and apoptosis, leading to massive neuronal loss. In this article, we explored the expression profile of selected immune and myelination associated transcripts (Wfdc17, Ccl3, Lyz2, Fa2h, Mog and Ugt8a) at 5-, 10- and 16-weeks, representing young, pre-symptomatic and late stages of the SD mice. We found that immune system related genes (Wfdc17, Ccl3, Lyz2) are significantly upregulated by several fold at all ages in Hexb-KO mice relative to Hexb-het mice, while the difference in the expression levels of myelination related genes is not statistically significant. There is an age-dependent significant increase in expression of microglial/pro-inflammatory genes, from 5-weeks to the near humane end-point, i.e., 16-week time point; while the expression of those genes involved in myelination decreases slightly or remains unchanged. Future studies warrant use of new high-throughput gene expression modalities (such as 10X genomics) to delineate the underlying pathogenesis in SD by detecting gene expression changes in specific neuronal cell types and thus, paving the way for rational and precise therapeutic modalities.


Sandhoff Disease , Transcriptome , Animals , Mice , Transcriptome/genetics , Disease Models, Animal , Sandhoff Disease/genetics , Sandhoff Disease/metabolism , Sandhoff Disease/pathology , Microglia/metabolism , Brain/metabolism
3.
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
4.
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
5.
J Orthop Res ; 38(12): 2580-2591, 2020 12.
Article En | MEDLINE | ID: mdl-32678923

Sandhoff disease (SD) is caused by decreased function of the enzyme ß-N-acetylhexosaminidase, resulting in accumulation of GM2 ganglioside in tissues. Neural tissue is primarily affected and individuals with the infantile form of the disease generally do not survive beyond 4 years of age. Current treatments address neurometabolic deficits to improve lifespan, however, this extended lifespan allows clinical disease to become manifest in other tissues, including the musculoskeletal system. The impact of SD on bone and joint tissues has yet to be fully determined. In a feline model of infantile SD, animals were treated by intracranial injection of adeno-associated virus vectors to supply the central nervous system with corrective levels of hexosaminidase, resulting in a twofold to threefold increase in lifespan. As treated animals aged, signs of musculoskeletal disease were identified. The present study characterized bone and joint lesions from affected cats using micro-computed tomography and histology. All affected cats had similar lesions, whether or not they were treated. SD cats displayed a significant reduction in metaphyseal trabecular bone and markedly abnormal size and shape of epiphyses. Abnormalities increased in severity with age and appear to be due to alteration in the function of chondrocytes within epiphyseal cartilage, particularly the articular-epiphyseal complex. Older cats developed secondary osteoarthritic changes. The changes identified are similar to those seen in humans with mucopolysaccharidoses. Statement of clinical significance: the lesions identified will have significant implications on the quality of life of individuals whose lifespans are extended due to treatments for the primary neurological effects of SD.


Growth Plate/physiopathology , Sandhoff Disease/physiopathology , Animals , Cats , Disease Models, Animal , Genetic Therapy , Growth Plate/diagnostic imaging , Growth Plate/growth & development , Growth Plate/pathology , Sandhoff Disease/diagnostic imaging , Sandhoff Disease/pathology , Sandhoff Disease/therapy , X-Ray Microtomography
6.
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
7.
J Mol Neurosci ; 70(4): 481-487, 2020 Apr.
Article En | MEDLINE | ID: mdl-31919734

Infantile Sandhoff disease is an autosomal recessive inherited disease primarily characterized by cherry red spots in the retina, muscle weakness, seizure, truncal hypotonia, hyperacusis, developmental delay and regression. The pathogenic genetic defects of the HEXB gene, which encodes the ß subunit of the hexosaminidase A (ɑß) and hexosaminidase B (ßß) enzymes, cause deficiency of both the Hex A and Hex B enzymes, resulting in the deposition of GM2 ganglion glycerides in the lysosomes of the central nervous system and somatic cells. The aim of this study was to discover disease-causing variants of the HEXB gene in two Chinese families through the use of exome sequencing. By characterizing three novel variants by molecular genetics, bioinformatics analysis, and three-dimensional structure modeling, we showed that all these novel variants influenced the protein structure. The results broaden the variant spectrum of HEXB in different ethnic groups. Furthermore, not all patients diagnosed with infantile Sandhoff disease had characteristic cranial imaging findings, which can only be used as supplementary information for diagnosis. The results of this study may contribute to clinical management, genetic counseling, and gene-targeted treatments for Sandhoff disease.


Sandhoff Disease/genetics , beta-Hexosaminidase beta Chain/genetics , Brain/diagnostic imaging , Female , Fundus Oculi , Humans , Infant , Mutation , Protein Domains , Sandhoff Disease/pathology , beta-Hexosaminidase beta Chain/chemistry
8.
Mol Genet Metab ; 126(2): 151-156, 2019 02.
Article En | MEDLINE | ID: mdl-30236619

Sandhoff disease (SD) results from mutations in the HEXB gene, subsequent deficiency of N-acetyl-ß-hexosaminidase (Hex) and accumulation of GM2 gangliosides. SD leads to progressive neurodegeneration and early death. However, there is a lack of established SD biomarkers, while the pathogenesis etiology remains to be elucidated. To identify potential biomarkers and unveil the pathogenic mechanisms, metabolomics analysis with reverse phase liquid chromatography (RPLC) was conducted. A total of 177, 112 and 119 metabolites were found to be significantly dysregulated in mouse liver, mouse brain and human hippocampus samples, respectively (p < .05, ID score > 0.5). Principal component analysis (PCA) analysis of the metabolites showed clear separation of metabolomics profiles between normal and diseased individuals. Among these metabolites, dipeptides, amino acids and derivatives were elevated, indicating a robust protein catabolism. Through pathway enrichment analysis, we also found alterations in metabolites associated with neurotransmission, lipid metabolism, oxidative stress and inflammation. In addition, N-acetylgalactosamine 4-sulphate, key component of glycosaminoglycans (GAG) was significantly elevated, which was also confirmed by biochemical assays. Collectively, these results indicated major shifts of energy utilization and profound metabolic impairments, contributing to the pathogenesis mechanisms of SD. Global metabolomics profiling may provide an innovative tool for better understanding the disease mechanisms, and identifying potential diagnostic biomarkers for SD.


Metabolome , Sandhoff Disease/metabolism , Sandhoff Disease/pathology , Animals , Biomarkers/analysis , Chromatography, Liquid , Disease Models, Animal , Hippocampus/metabolism , Humans , Inflammation , Lipid Metabolism , Male , Mice , Oxidative Stress , Synaptic Transmission
10.
Orphanet J Rare Dis ; 13(1): 130, 2018 08 03.
Article En | MEDLINE | ID: mdl-30075786

BACKGROUND: Infantile Sandhoff disease (ISD) is a GM2 gangliosidosis that is classified as a lysosomal storage disorder. The most common symptoms of affected individuals at presentation are neurologic involvement. Here we report clinical course and demographic features in a case series of infantile Sandhoff disease. Enzymatically and some genetically proven cases of ISD were extracted from the Iranian Neurometabolic Registry (INMR) in Children's Medical Center, Iran, Tehran from December 2010 to December 2016. RESULT: Twenty five cases of infantile SD (13 female, 12 male) were included in this study. The age range of patients was 9-24 months with a mean of 15.8 months. The consanguinity rate of parents affected families was about 80%. The mean age of patients at disease onset was 6.4 months and the mean age at diagnosis was 14 months. Patients were diagnosed with a mean delay of 7.8 months. Eleven of patients died due to aspiration pneumonia and intractable seizure. The most common features at presentation (92%) were developmental delay or regression in speech and cognitive domains. Cherry red spots were detected in 17 patients (68%). Organomegaly was detected only in two patients. Enzyme studies showed marked reductions of both Hexosaminidase A and B in all patients. HEXB gene mutation studies performed in eight patients identified 6 different mutations, which five of them were novel. CONCLUSION: Infantile SD should be considered for each child presented with neurologic symptoms such as developmental delay and regression and cherry red spots in ophthalmic examination. Organomegaly is not a frequent clinical finding in infantile SD. Additionally; there are a genetic heterogenisity among Iranian patients.


Mutation/genetics , Sandhoff Disease/genetics , Sandhoff Disease/pathology , Child, Preschool , Female , Hexosaminidase A/genetics , Humans , Infant , Iran , Male , beta-Hexosaminidase beta Chain/genetics
11.
J Zoo Wildl Med ; 49(2): 335-344, 2018 Jun.
Article En | MEDLINE | ID: mdl-29900785

This study reports the occurrence of the lysosomal storage disease GM2 gangliosidosis (Sandhoff disease) in two 11-mo-old captive-bred, male and female mongoose siblings ( Mungos mungo). The clinical signs and the pathological findings reported here were similar to those reported in other mammalian species. Light microscopy revealed an accumulation of stored material in neurons and macrophages accompanied by a significant neuronal degeneration (swelling of neuronal soma, loss of Nissl substance, and neuronal loss) and gliosis. Electron microscopy of brain tissue identified the stored material as membrane-bound multilamellar bodies. An almost complete lack of total hexosaminidase activity in serum suggested a defect in the HEXB gene (Sandhoff disease in humans). High-performance thin-layer chromatography and mass spectrometry confirmed the accumulation of GM2 ganglioside in brain and kidney tissue, and the lectin staining pattern of the brain tissue further corroborated the diagnosis of a Sandhoff-type lysosomal storage disease.


Herpestidae , Sandhoff Disease/veterinary , Animals , Animals, Zoo , Female , Male , Sandhoff Disease/diagnosis , Sandhoff Disease/pathology , Sandhoff Disease/physiopathology
12.
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
13.
Hum Mol Genet ; 27(6): 954-968, 2018 03 15.
Article En | MEDLINE | ID: mdl-29325092

Sandhoff disease (SD) is a rare inherited disorder caused by a deficiency of ß-hexosaminidase activity which is fatal because no effective treatment is available. A mouse model of Hexb deficiency reproduces the key pathognomonic features of SD patients with severe ubiquitous lysosomal dysfunction, GM2 accumulation, neuroinflammation and neurodegeneration, culminating in death at 4 months. Here, we show that a single intravenous neonatal administration of a self-complementary adeno-associated virus 9 vector (scAAV9) expressing the Hexb cDNA in SD mice is safe and sufficient to prevent disease development. Importantly, we demonstrate for the first time that this treatment results in a normal lifespan (over 700 days) and normalizes motor function assessed by a battery of behavioral tests, with scAAV9-treated SD mice being indistinguishable from wild-type littermates. Biochemical analyses in multiple tissues showed a significant increase in hexosaminidase A activity, which reached 10-15% of normal levels. AAV9 treatment was sufficient to prevent GM2 and GA2 storage almost completely in the cerebrum (less so in the cerebellum), as well as thalamic reactive gliosis and thalamocortical neuron loss in treated Hexb-/- mice. In summary, this study demonstrated a widespread protective effect throughout the entire CNS after a single intravenous administration of the scAAV9-Hexb vector to neonatal SD mice.


Hexosaminidase B/pharmacology , Sandhoff Disease/drug therapy , Sandhoff Disease/pathology , Administration, Intravenous , Animals , Animals, Newborn , Brain/metabolism , Disease Models, Animal , Female , G(M2) Ganglioside/metabolism , Gangliosides/metabolism , Hexosaminidase B/genetics , Hexosaminidase B/metabolism , Male , Mice , Mice, Inbred C57BL , Sandhoff Disease/metabolism
14.
J Lipid Res ; 59(3): 550-563, 2018 03.
Article En | MEDLINE | ID: mdl-29358305

Sandhoff disease, one of the GM2 gangliosidoses, is a lysosomal storage disorder characterized by the absence of ß-hexosaminidase A and B activity and the concomitant lysosomal accumulation of its substrate, GM2 ganglioside. It features catastrophic neurodegeneration and death in early childhood. How the lysosomal accumulation of ganglioside might affect the early development of the nervous system is not understood. Recently, cerebral organoids derived from induced pluripotent stem (iPS) cells have illuminated early developmental events altered by disease processes. To develop an early neurodevelopmental model of Sandhoff disease, we first generated iPS cells from the fibroblasts of an infantile Sandhoff disease patient, then corrected one of the mutant HEXB alleles in those iPS cells using CRISPR/Cas9 genome-editing technology, thereby creating isogenic controls. Next, we used the parental Sandhoff disease iPS cells and isogenic HEXB-corrected iPS cell clones to generate cerebral organoids that modeled the first trimester of neurodevelopment. The Sandhoff disease organoids, but not the HEXB-corrected organoids, accumulated GM2 ganglioside and exhibited increased size and cellular proliferation compared with the HEXB-corrected organoids. Whole-transcriptome analysis demonstrated that development was impaired in the Sandhoff disease organoids, suggesting that alterations in neuronal differentiation may occur during early development in the GM2 gangliosidoses.


Cell Differentiation , Cerebral Cortex/pathology , Induced Pluripotent Stem Cells/pathology , Neurons/pathology , Organoids/pathology , Sandhoff Disease/pathology , Cell Proliferation , Cells, Cultured , Humans , Lysosomes/metabolism , beta-N-Acetylhexosaminidases/deficiency , beta-N-Acetylhexosaminidases/metabolism
15.
PLoS One ; 12(6): e0178978, 2017.
Article En | MEDLINE | ID: mdl-28575132

In Sandhoff disease (SD), the activity of the lysosomal hydrolytic enzyme, ß-hexosaminidase (Hex), is lost due to a Hexb gene defect, which results in the abnormal accumulation of the substrate, GM2 ganglioside (GM2), in neuronal cells, causing neuronal loss, microglial activation, and astrogliosis. We established induced pluripotent stem cells from the cells of SD mice (SD-iPSCs). In the present study, we investigated the occurrence of abnormal differentiation and development of a neural lineage in the asymptomatic phase of SD in vitro using SD mouse fetus-derived neural stem cells (NSCs) and SD-iPSCs. It was assumed that the number of SD mouse fetal brain-derived NSCs was reduced and differentiation was promoted, resulting in the inhibition of differentiation into neurons and enhancement of differentiation into astrocytes. The number of SD-iPSC-derived NSCs was also reduced, suggesting that the differentiation of NSCs was promoted, resulting in the inhibition of differentiation into neurons and enhancement of that into astrocytes. This abnormal differentiation of SD-iPSCs toward a neural lineage was reduced by the glucosylceramide synthase inhibitor, miglustat. Furthermore, abnormal differentiation toward a neural lineage was reduced in SD-iPSCs with Hexb gene transfection. Therefore, differentiation ability along the time axis appears to be altered in SD mice in which the differentiation ability of NSCs is promoted and differentiation into neurons is completed earlier, while the timing of differentiation into astrocytes is accelerated. These results clarified that the abnormal differentiation of SD-iPSCs toward a neural lineage in vitro was shown to reflect the pathology of SD.


Astrocytes/pathology , Induced Pluripotent Stem Cells/pathology , Neural Stem Cells/pathology , Neurogenesis , Neurons/pathology , Sandhoff Disease/pathology , Animals , Brain/pathology , Cell Count , Cell Proliferation , Cells, Cultured , Disease Models, Animal , Mice , Mice, Inbred C57BL
16.
J Lipid Res ; 58(7): 1306-1314, 2017 07.
Article En | MEDLINE | ID: mdl-28377426

Drug-induced phospholipidosis (DIPL) is characterized by an increase in the phospholipid content of the cell and the accumulation of drugs and lipids inside the lysosomes of affected tissues, including in the liver. Although of uncertain pathological significance for patients, the condition remains a major impediment for the clinical development of new drugs. Human Sandhoff disease (SD) is caused by inherited defects of the ß subunit of lysosomal ß-hexosaminidases (Hex) A and B, leading to a large array of symptoms, including neurodegeneration and ultimately death by the age of 4 in its most common form. The substrates of Hex A and B, gangliosides GM2 and GA2, accumulate inside the lysosomes of the CNS and in peripheral organs. Given that both DIPL and SD are associated with lysosomes and lipid metabolism in general, we measured the hepatic lipid profiles in rodent models of these two conditions using untargeted LC/MS to examine potential commonalities. Both model systems shared a number of perturbed lipid pathways, notably those involving metabolism of cholesteryl esters, lysophosphatidylcholines, bis(monoacylglycero)phosphates, and ceramides. We report here profound alterations in lipid metabolism in the SD liver. In addition, DIPL induced a wide range of lipid changes not previously observed in the liver, highlighting similarities with those detected in the model of SD and raising concerns that these lipid changes may be associated with underlying pathology associated with lysosomal storage disorders.


Liver/drug effects , Liver/metabolism , Lysosomes/metabolism , Phospholipids/metabolism , Sandhoff Disease/chemically induced , Sandhoff Disease/metabolism , Animals , Disease Models, Animal , Liver/pathology , Lysosomes/drug effects , Male , Rats , Rats, Sprague-Dawley , Sandhoff Disease/pathology
18.
Sci Rep ; 7: 40518, 2017 01 13.
Article En | MEDLINE | ID: mdl-28084424

Sandhoff disease (SD) is caused by the loss of ß-hexosaminidase (Hex) enzymatic activity in lysosomes resulting from Hexb mutations. In SD patients, the Hex substrate GM2 ganglioside accumulates abnormally in neuronal cells, resulting in neuronal loss, microglial activation, and astrogliosis. Hexb-/- mice, which manifest a phenotype similar to SD, serve as animal models for examining the pathophysiology of SD. Hexb-/- mice reach ~8 weeks without obvious neurological defects; however, trembling begins at 12 weeks and is accompanied by startle reactions and increased limb tone. These symptoms gradually become severe by 16-18 weeks. Immune reactions caused by autoantibodies have been recently associated with the pathology of SD. The inhibition of immune activation may represent a novel therapeutic target for SD. Herein, SD mice (Hexb-/-) were crossed to mice lacking an activating immune receptor (FcRγ-/-) to elucidate the potential relationship between immune responses activated through SD autoantibodies and astrogliosis. Microglial activation and astrogliosis were observed in cortices of Hexb-/- mice during the asymptomatic phase, and were inhibited in Hexb-/- FcRγ-/- mice. Moreover, early astrogliosis and impaired motor coordination in Hexb-/- mice could be ameliorated by immunosuppressants, such as FTY720. Our findings demonstrate the importance of early treatment and the therapeutic effectiveness of immunosuppression in SD.


Astrocytes/immunology , Astrocytes/pathology , Gliosis/immunology , Gliosis/pathology , Immunity , Receptors, Fc/metabolism , Sandhoff Disease/immunology , Sandhoff Disease/pathology , Animals , Astrocytes/drug effects , Astrocytes/metabolism , Cerebral Cortex/pathology , Disease Models, Animal , Fingolimod Hydrochloride/pharmacology , G(M2) Ganglioside/metabolism , Heterozygote , Immunity/drug effects , Immunosuppressive Agents/pharmacology , Mice, Inbred C57BL , Motor Activity/drug effects , Phenotype , Receptors, Fc/deficiency , Sandhoff Disease/physiopathology , Up-Regulation/drug effects , Walking , beta-Hexosaminidase beta Chain/metabolism
19.
Hum Mol Genet ; 26(4): 661-673, 2017 02 15.
Article En | MEDLINE | ID: mdl-28007910

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.


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
20.
Neuroscience ; 340: 117-125, 2017 01 06.
Article En | MEDLINE | ID: mdl-27793778

Sandhoff disease (SD) is a lysosomal storage disorder characterized by the absence of hydrolytic enzyme ß-N-acetylhexosaminidase (Hex), which results in storage of GM2 ganglioside in neurons and unremitting neurodegeneration. Neuron loss initially affects fine motor skills, but rapidly progresses to loss of all body faculties, a vegetative state, and death by five years of age in humans. A well-established feline model of SD allows characterization of the disease in a large animal model and provides a means to test the safety and efficacy of therapeutic interventions before initiating clinical trials. In this study, we demonstrate a robust central nervous system (CNS) inflammatory response in feline SD, primarily marked by expansion and activation of the microglial cell population. Quantification of major histocompatibility complex II (MHC-II) labeling revealed significant up-regulation throughout the CNS with areas rich in white matter most severely affected. Expression of the leukocyte chemokine macrophage inflammatory protein-1 alpha (MIP-1α) was also up-regulated in the brain. SD cats were treated with intracranial delivery of adeno-associated viral (AAV) vectors expressing feline Hex, with a study endpoint 16weeks post treatment. AAV-mediated gene delivery repressed the expansion and activation of microglia and normalized MHC-II and MIP-1α levels. These data reiterate the profound inflammatory response in SD and show that neuroinflammation is abrogated after AAV-mediated restoration of enzymatic activity.


Brain/immunology , Genetic Therapy , Sandhoff Disease/immunology , Sandhoff Disease/therapy , Adaptor Proteins, Signal Transducing/metabolism , Animals , Astrocytes/immunology , Astrocytes/pathology , Brain/pathology , Cats , Dependovirus/genetics , Disease Models, Animal , Genes, MHC Class II/physiology , Genetic Vectors , Gliosis/immunology , Gliosis/pathology , Gliosis/therapy , Immunohistochemistry , Microglia/immunology , Microglia/pathology , Neurons/immunology , Neurons/pathology , Polymerase Chain Reaction , Sandhoff Disease/pathology , Transcription Factors/genetics , Transcription Factors/metabolism
...