Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 6 de 6
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Biomed Pharmacother ; 149: 112808, 2022 May.
Artigo em Inglês | MEDLINE | ID: mdl-35290889

RESUMO

Krabbe disease is a rare, inherited neurodegenerative disease due to impaired lysosomal ß-galactosylceramidase (GALC) activity and formation of neurotoxic ß-galactosylsphingosine ('psychosine'). We investigated substrate reduction therapy with a novel brain-penetrant inhibitor of galactosylceramide biosynthesis, RA 5557, in twitcher mice that lack GALC activity and model Krabbe disease. This thienopyridine derivative selectively inhibits uridine diphosphate-galactose glycosyltransferase 8 (UGT8), the final step in the generation of galactosylceramides which are precursors of sulphatide and, in the pathological lysosome, the immediate source of psychosine. Administration of RA 5557, reduced pathologically elevated psychosine concentrations (72-86%) in the midbrain and cerebral cortex in twitcher mice: the inhibitor decreased galactosylceramides by about 70% in midbrain and cerebral cortex in mutant and wild type animals. Exposure to the inhibitor significantly decreased several characteristic inflammatory response markers without causing apparent toxicity to myelin-producing cells in wild type and mutant mice; transcript abundance of oligodendrocyte markers MBP (myelin basic protein) and murine UGT8 was unchanged. Administration of the inhibitor before conception and during several breeding cycles to mice did not impair fertility and gave rise to healthy offspring. Nevertheless, given the unchanged lifespan, it appears that GALC has critical functions in the nervous system beyond the hydrolysis of galactosylceramide and galactosylsphingosine. Our findings support further therapeutic exploration of orally active UGT8 inhibitors in Krabbe disease and related galactosphingolipid disorders. The potent thienopyridine derivative with effective target engagement here studied appears to have an acceptable safety profile in vivo; judicious dose optimization will be needed to ensure efficacious clinical translation.


Assuntos
Leucodistrofia de Células Globoides , Doenças Neurodegenerativas , Animais , Encéfalo/metabolismo , Modelos Animais de Doenças , Galactosilceramidas/metabolismo , Galactosilceramidas/farmacologia , Leucodistrofia de Células Globoides/tratamento farmacológico , Leucodistrofia de Células Globoides/metabolismo , Leucodistrofia de Células Globoides/patologia , Camundongos , Doenças Neurodegenerativas/patologia , Psicosina/metabolismo , Tienopiridinas
2.
Cells ; 10(11)2021 11 11.
Artigo em Inglês | MEDLINE | ID: mdl-34831346

RESUMO

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.


Assuntos
Arginina/farmacologia , Autofagia , Gangliosidoses GM2/metabolismo , Serina-Treonina Quinases TOR/metabolismo , Autofagossomos/efeitos dos fármacos , Autofagossomos/metabolismo , Autofagossomos/ultraestrutura , Autofagia/efeitos dos fármacos , Catepsinas/metabolismo , Fibroblastos/efeitos dos fármacos , Fibroblastos/metabolismo , Fibroblastos/patologia , Hexosaminidase A/química , Hexosaminidase A/metabolismo , Hexosaminidase B/química , Hexosaminidase B/metabolismo , Humanos , Lisossomos/efeitos dos fármacos , Lisossomos/metabolismo , Mutação/genética , Permeabilidade , Proteínas Proto-Oncogênicas c-akt/metabolismo , Doença de Sandhoff/patologia , Transdução de Sinais/efeitos dos fármacos , Doença de Tay-Sachs/patologia , Transcriptoma/genética
3.
Metabolites ; 11(1)2020 Dec 30.
Artigo em Inglês | MEDLINE | ID: mdl-33396723

RESUMO

Sandhoff disease (SD) is a lysosomal disease caused by mutations in the gene coding for the ß subunit of ß-hexosaminidase, leading to deficiency in the enzymes ß-hexosaminidase (HEX) A and B. SD is characterised by an accumulation of gangliosides and related glycolipids, mainly in the central nervous system, and progressive neurodegeneration. The underlying cellular mechanisms leading to neurodegeneration and the contribution of inflammation in SD remain undefined. The aim of the present study was to measure global changes in metabolism over time that might reveal novel molecular pathways of disease. We used liquid chromatography-mass spectrometry and 1H Nuclear Magnetic Resonance spectroscopy to profile intact lipids and aqueous metabolites, respectively. We examined spinal cord and cerebrum from healthy and Hexb -/- mice, a mouse model of SD, at ages one, two, three and four months. We report decreased concentrations in lipids typical of the myelin sheath, galactosylceramides and plasmalogen-phosphatidylethanolamines, suggesting that reduced synthesis of myelin lipids is an early event in the development of disease pathology. Reduction in neuronal density is progressive, as demonstrated by decreased concentrations of N-acetylaspartate and amino acid neurotransmitters. Finally, microglial activation, indicated by increased amounts of myo-inositol correlates closely with the late symptomatic phases of the disease.

4.
Curr Gene Ther ; 18(2): 68-89, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29618308

RESUMO

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.


Assuntos
Transplante de Medula Óssea , Terapia de Reposição de Enzimas , Terapia Genética , Doença de Sandhoff/genética , Doença de Sandhoff/terapia , Doença de Tay-Sachs/genética , Doença de Tay-Sachs/terapia , Animais , Modelos Animais de Doenças , Glicoesfingolipídeos/metabolismo , Humanos , Lactente , Lisossomos/enzimologia , Lisossomos/genética , Lisossomos/patologia , Camundongos , Mutação , Doenças Raras , Doença de Sandhoff/enzimologia , Doença de Sandhoff/patologia , Doença de Tay-Sachs/enzimologia , Doença de Tay-Sachs/patologia , beta-N-Acetil-Hexosaminidases/genética
5.
Hum Mol Genet ; 23(3): 730-48, 2014 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-24057669

RESUMO

The GM2 gangliosidoses are progressive neurodegenerative disorders due to defects in the lysosomal ß-N-acetylhexosaminidase system. Accumulation of ß-hexosaminidases A and B substrates is presumed to cause this fatal condition. An authentic mouse model of Sandhoff disease (SD) with pathological characteristics resembling those noted in infantile GM2 gangliosidosis has been described. We have shown that expression of ß-hexosaminidase by intracranial delivery of recombinant adeno-associated viral vectors to young adult SD mice can prevent many features of the disease and extends lifespan. To investigate the nature of the neurological injury in GM2 gangliosidosis and the extent of its reversibility, we have examined the evolution of disease in the SD mouse; we have moreover explored the effects of gene transfer delivered at key times during the course of the illness. Here we report greatly increased survival only when the therapeutic genes are expressed either before the disease is apparent or during its early manifestations. However, irrespective of when treatment was administered, widespread and abundant expression of ß-hexosaminidase with consequent clearance of glycoconjugates, α-synuclein and ubiquitinated proteins, and abrogation of inflammatory responses and neuronal loss was observed. We also show that defects in myelination occur in early life and cannot be easily resolved when treatment is given to the adult brain. These results indicate that there is a limited temporal opportunity in which function and survival can be improved-but regardless of resolution of the cardinal pathological features of GM2 gangliosidosis, a point is reached when functional deterioration and death cannot be prevented.


Assuntos
Encéfalo/enzimologia , Vetores Genéticos/farmacologia , Doença de Sandhoff/genética , Doença de Sandhoff/patologia , Doença de Sandhoff/terapia , Doença de Tay-Sachs/patologia , beta-N-Acetil-Hexosaminidases/genética , Animais , Encéfalo/efeitos dos fármacos , Dependovirus/genética , Modelos Animais de Doenças , Gangliosídeo G(M2)/genética , Gangliosídeo G(M2)/metabolismo , Terapia Genética/métodos , Vetores Genéticos/administração & dosagem , Humanos , Injeções Intralesionais , Camundongos , Camundongos Knockout , Camundongos Transgênicos , Doença de Sandhoff/mortalidade , Ubiquitina/metabolismo , alfa-Sinucleína/metabolismo , beta-N-Acetil-Hexosaminidases/metabolismo
6.
Hum Mol Genet ; 20(22): 4371-80, 2011 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-21852247

RESUMO

Sandhoff disease, a GM2 gangliosidosis caused by a deficiency in ß-hexosaminidase, is characterized by progressive neurodegeneration. Although loss of neurons in association with lysosomal storage of glycosphingolipids occurs in patients with this disease, the molecular pathways that lead to the accompanying neurological defects are unclear. Using an authentic murine model of GM2 gangliosidosis, we examined the pattern of neuronal loss in the central nervous system and investigated the effects of gene transfer using recombinant adeno-associated viral vectors expressing ß-hexosaminidase subunits (rAAV2/1-Hex). In 4-month-old Sandhoff mice with neurological deficits, cells staining positively for the apoptotic signature in the TUNEL reaction were found in the ventroposterior medial and ventroposterior lateral (VPM/VPL) nuclei of the thalamus. There was progressive loss of neuronal density in this region with age. Comparable loss of neuronal density was identified in the lateral vestibular nucleus of the brainstem and a small but statistically significant loss was present in the ventral spinal cord. Loss of neurons was not detected in other regions that were analysed. Administration of rAAV2/1-Hex into the brain of Sandhoff mice prevented the decline in neuronal density in the VPM/VPL. Preservation of neurons in the VPM/VPL was variable at the humane endpoint in treated animals, but correlated directly with increased lifespan. Loss of neurons was localized to only a few regions in the Sandhoff brain and was prevented by rAAV-mediated transfer of ß-hexosaminidase gene function at considerable distances from the site of vector administration.


Assuntos
Encéfalo/metabolismo , Encéfalo/patologia , Dependovirus/genética , Vetores Genéticos/genética , Neurônios/patologia , Doença de Sandhoff/terapia , beta-N-Acetil-Hexosaminidases/metabolismo , Animais , Apoptose/genética , Apoptose/fisiologia , Marcação In Situ das Extremidades Cortadas , Camundongos , Neurônios/metabolismo , Doença de Sandhoff/enzimologia , Doença de Sandhoff/metabolismo , beta-N-Acetil-Hexosaminidases/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...