Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Mol Ther ; 27(8): 1495-1506, 2019 08 07.
Artigo em Inglês | MEDLINE | ID: mdl-31208914

RESUMO

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.


Assuntos
Carbamatos/farmacologia , Inibidores Enzimáticos/farmacologia , Glucosiltransferases/antagonistas & inibidores , Quinuclidinas/farmacologia , Doença de Sandhoff/enzimologia , Animais , Encéfalo/efeitos dos fármacos , Encéfalo/metabolismo , Encéfalo/patologia , Modelos Animais de Doenças , Feminino , Glucosiltransferases/genética , Glucosiltransferases/metabolismo , Ligantes , Fígado/efeitos dos fármacos , Fígado/metabolismo , Fígado/patologia , Masculino , Espectrometria de Massas , Camundongos , Camundongos Knockout , Imagem Molecular , Receptores de GABA/metabolismo , Doença de Sandhoff/diagnóstico , Doença de Sandhoff/genética , Doença de Sandhoff/terapia , Esfingolipídeos/metabolismo , Cadeia beta da beta-Hexosaminidase/genética , Cadeia beta da beta-Hexosaminidase/metabolismo
2.
Mol Ther ; 24(6): 1019-1029, 2016 06.
Artigo em Inglês | MEDLINE | ID: mdl-26948439

RESUMO

Gaucher disease (GD) is caused by a deficiency of glucocerebrosidase and the consequent lysosomal accumulation of unmetabolized glycolipid substrates. Enzyme-replacement therapy adequately manages the visceral manifestations of nonneuronopathic type-1 Gaucher patients, but not the brain disease in neuronopathic types 2 and 3 GD. Substrate reduction therapy through inhibition of glucosylceramide synthase (GCS) has also been shown to effectively treat the visceral disease. Here, we evaluated the efficacy of a novel small molecule inhibitor of GCS with central nervous system (CNS) access (Genz-682452) to treat the brain disease. Treatment of the conduritol ß epoxide-induced mouse model of neuronopathic GD with Genz-682452 reduced the accumulation of liver and brain glycolipids (>70% and >20% respectively), extent of gliosis, and severity of ataxia. In the genetic 4L;C* mouse model, Genz-682452 reduced the levels of substrate in the brain by >40%, the extent of gliosis, and paresis. Importantly, Genz-682452-treated 4L;C* mice also exhibited an ~30% increase in lifespan. Together, these data indicate that an orally available antagonist of GCS that has CNS access is effective at attenuating several of the neuropathologic and behavioral manifestations associated with mouse models of neuronopathic GD. Therefore, Genz-682452 holds promise as a potential therapeutic approach for patients with type-3 GD.


Assuntos
Carbamatos/administração & dosagem , Sistema Nervoso Central/metabolismo , Inibidores Enzimáticos/administração & dosagem , Doença de Gaucher/tratamento farmacológico , Glucosiltransferases/antagonistas & inibidores , Glicolipídeos/metabolismo , Quinuclidinas/administração & dosagem , Administração Oral , Animais , Carbamatos/farmacologia , Modelos Animais de Doenças , Inibidores Enzimáticos/farmacologia , Doença de Gaucher/induzido quimicamente , Doença de Gaucher/metabolismo , Humanos , Inositol/análogos & derivados , Fígado/metabolismo , Lisossomos/efeitos dos fármacos , Lisossomos/enzimologia , Camundongos , Quinuclidinas/farmacologia , Distribuição Tecidual , Resultado do Tratamento
3.
Mol Med ; 21: 389-99, 2015 Apr 30.
Artigo em Inglês | MEDLINE | ID: mdl-25938659

RESUMO

Fabry disease, an X-linked glycosphingolipid storage disorder, is caused by the deficient activity of α-galactosidase A (α-Gal A). This results in the lysosomal accumulation in various cell types of its glycolipid substrates, including globotriaosylceramide (GL-3) and lysoglobotriaosylceramide (globotriaosyl lysosphingolipid, lyso-GL-3), leading to kidney, heart, and cerebrovascular disease. To complement and potentially augment the current standard of care, biweekly infusions of recombinant α-Gal A, the merits of substrate reduction therapy (SRT) by selectively inhibiting glucosylceramide synthase (GCS) were examined. Here, we report the development of a novel, orally available GCS inhibitor (Genz-682452) with pharmacological and safety profiles that have potential for treating Fabry disease. Treating Fabry mice with Genz-682452 resulted in reduced tissue levels of GL-3 and lyso-GL-3 and a delayed loss of the thermal nociceptive response. Greatest improvements were realized when the therapeutic intervention was administered to younger mice before they developed overt pathology. Importantly, as the pharmacologic profiles of α-Gal A and Genz-682452 are different, treating animals with both drugs conferred the greatest efficacy. For example, because Genz-682452, but not α-Gal A, can traverse the blood-brain barrier, levels of accumulated glycosphingolipids were reduced in the brain of Genz-682452-treated but not α-Gal A-treated mice. These results suggest that combining substrate reduction and enzyme replacement may confer both complementary and additive therapeutic benefits in Fabry disease.


Assuntos
Carbamatos/administração & dosagem , Doença de Fabry/tratamento farmacológico , Glucosiltransferases/metabolismo , Glicolipídeos/metabolismo , Quinuclidinas/administração & dosagem , Esfingolipídeos/metabolismo , Triexosilceramidas/metabolismo , Animais , Barreira Hematoencefálica/efeitos dos fármacos , Modelos Animais de Doenças , Doença de Fabry/metabolismo , Doença de Fabry/patologia , Glucosiltransferases/antagonistas & inibidores , Humanos , Camundongos , alfa-Galactosidase/administração & dosagem , alfa-Galactosidase/metabolismo
4.
Mol Genet Metab ; 105(4): 621-8, 2012 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-22366055

RESUMO

Niemann Pick type C (NPC) disease is a progressive neurodegenerative disease caused by mutations in NPC1 or NPC2, the gene products of which are involved in cholesterol transport in late endosomes. NPC is characterized by an accumulation of cholesterol, sphingomyelin and glycosphingolipids in the visceral organs, primarily the liver and spleen. In the brain, there is a redistribution of unesterified cholesterol and a concomitant accumulation of glycosphingolipids. It has been suggested that reducing the aberrant lysosomal storage of glycosphingolipids in the brain by a substrate reduction therapy (SRT) approach may prove beneficial. Inhibiting glucosylceramide synthase (GCS) using the iminosugar-based inhibitor miglustat (NB-DNJ) has been reported to increase the survival of NPC mice. Here, we tested the effects of Genz-529468, a more potent iminosugar-based inhibitor of GCS, in the NPC mouse. Oral administration of Genz-529468 or NB-DNJ to NPC mice improved their motor function, reduced CNS inflammation, and increased their longevity. However, Genz-529468 offered a wider therapeutic window and better therapeutic index than NB-DNJ. Analysis of the glycolipids in the CNS of the iminosugar-treated NPC mouse revealed that the glucosylceramide (GL1) but not the ganglioside levels were highly elevated. This increase in GL1 was likely caused by the off-target inhibition of the murine non-lysosomal glucosylceramidase, Gba2. Hence, the basis for the observed effects of these inhibitors in NPC mice might be related to their inhibition of Gba2 or another unintended target rather than a result of substrate reduction.


Assuntos
Encéfalo/metabolismo , Inibidores Enzimáticos/uso terapêutico , Glucosiltransferases/antagonistas & inibidores , Imino Açúcares/uso terapêutico , Doença de Niemann-Pick Tipo C/tratamento farmacológico , Doença de Niemann-Pick Tipo C/mortalidade , Animais , Encéfalo/citologia , Encéfalo/efeitos dos fármacos , Modelos Animais de Doenças , Sinergismo Farmacológico , Glucosilceramidas/metabolismo , Glicoesfingolipídeos/metabolismo , Fígado/citologia , Fígado/efeitos dos fármacos , Fígado/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Knockout , Doença de Niemann-Pick Tipo C/enzimologia , Taxa de Sobrevida
5.
PLoS One ; 6(6): e21758, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21738789

RESUMO

The neuropathic glycosphingolipidoses are a subgroup of lysosomal storage disorders for which there are no effective therapies. A potential approach is substrate reduction therapy using inhibitors of glucosylceramide synthase (GCS) to decrease the synthesis of glucosylceramide and related glycosphingolipids that accumulate in the lysosomes. Genz-529468, a blood-brain barrier-permeant iminosugar-based GCS inhibitor, was used to evaluate this concept in a mouse model of Sandhoff disease, which accumulates the glycosphingolipid GM2 in the visceral organs and CNS. As expected, oral administration of the drug inhibited hepatic GM2 accumulation. Paradoxically, in the brain, treatment resulted in a slight increase in GM2 levels and a 20-fold increase in glucosylceramide levels. The increase in brain glucosylceramide levels might be due to concurrent inhibition of the non-lysosomal glucosylceramidase, Gba2. Similar results were observed with NB-DNJ, another iminosugar-based GCS inhibitor. Despite these unanticipated increases in glycosphingolipids in the CNS, treatment nevertheless delayed the loss of motor function and coordination and extended the lifespan of the Sandhoff mice. These results suggest that the CNS benefits observed in the Sandhoff mice might not necessarily be due to substrate reduction therapy but rather to off-target effects.


Assuntos
Encéfalo/efeitos dos fármacos , Encéfalo/metabolismo , Inibidores Enzimáticos/uso terapêutico , Glucosiltransferases/antagonistas & inibidores , Glicoesfingolipídeos/metabolismo , Imino Açúcares/química , Doença de Sandhoff/tratamento farmacológico , Doença de Sandhoff/metabolismo , Animais , Inibidores Enzimáticos/química , Imuno-Histoquímica , Camundongos
6.
Mol Ther ; 19(11): 1999-2011, 2011 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-21712814

RESUMO

In mice, liver-restricted expression of lysosomal enzymes from adeno-associated viral serotype 8 (AAV8) vectors results in reduced antibodies to the expressed proteins. To ask whether this result might translate to patients, nonhuman primates (NHPs) were injected systemically with AAV8 encoding α-galactosidase A (α-gal). As in mice, sustained expression in monkeys attenuated antibody responses to α-gal. However, this effect was not robust, and sustained α-gal levels were 1-2 logs lower than those achieved in male mice at the same vector dose. Because our mouse studies had shown that antibody levels were directly related to expression levels, several strategies were evaluated to increase expression in monkeys. Unlike mice, expression in monkeys did not respond to androgens. Local delivery to the liver, immune suppression, a self-complementary vector and pharmacologic approaches similarly failed to increase expression. While equivalent vector copies reached mouse and primate liver and there were no apparent differences in vector form, methylation or deamination, transgene expression was limited at the mRNA level in monkeys. These results suggest that compared to mice, transcription from an AAV8 vector in monkeys can be significantly reduced. They also suggest some current limits on achieving clinically useful antibody reduction and therapeutic benefit for lysosomal storage diseases using a systemic AAV8-based approach.


Assuntos
Dependovirus/genética , Vetores Genéticos/administração & dosagem , Tolerância Imunológica , Imunidade Humoral , Fígado/metabolismo , alfa-Galactosidase/genética , Androgênios/farmacologia , Animais , Metilação de DNA , Desaminação , Dependovirus/imunologia , Dosagem de Genes , Regulação da Expressão Gênica/efeitos dos fármacos , Vetores Genéticos/imunologia , Humanos , Injeções , Macaca mulatta , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Transcrição Gênica , alfa-Galactosidase/imunologia , alfa-Galactosidase/metabolismo
7.
Mol Ther ; 18(11): 1983-94, 2010 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-20736932

RESUMO

Liver-directed gene therapy with adeno-associated virus (AAV) vectors effectively treats mouse models of lysosomal storage diseases (LSDs). We asked whether these results were likely to translate to patients. To understand to what extent preexisting anti-AAV8 antibodies could impede AAV8-mediated liver transduction in primates, commonly preexposed to AAV, we quantified the effects of preexisting antibodies on liver transduction and subsequent transgene expression in mouse and nonhuman primate (NHP) models. Using the highest viral dose previously reported in a clinical trial, passive transfer of NHP sera containing relatively low anti-AAV8 titers into mice blocked liver transduction, which could be partially overcome by increasing vector dose tenfold. Based on this and a survey of anti-AAV8 titers in 112 humans, we predict that high-dose systemic gene therapy would successfully transduce liver in >50% of human patients. However, although high-dose AAV8 administration to mice and monkeys with equivalent anti-AAV8 titers led to comparable liver vector copy numbers, the resulting transgene expression in primates was ~1.5-logs lower than mice. This suggests vector fate differs in these species and that strategies focused solely on overcoming preexisting vector-specific antibodies may be insufficient to achieve clinically meaningful expression levels of LSD genes using a liver-directed gene therapy approach in patients.


Assuntos
Dependovirus/genética , Terapia Genética , Hepatócitos/imunologia , Doenças por Armazenamento dos Lisossomos/terapia , Transgenes/fisiologia , alfa-Galactosidase/sangue , Animais , Anticorpos Neutralizantes/imunologia , Western Blotting , Vetores Genéticos/administração & dosagem , Células HeLa , Hepatócitos/metabolismo , Humanos , Doenças por Armazenamento dos Lisossomos/genética , Doenças por Armazenamento dos Lisossomos/imunologia , Macaca fascicularis , Macaca mulatta , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Plasmaferese , Biossíntese de Proteínas , RNA Mensageiro/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , alfa-Galactosidase/genética
8.
J Gene Med ; 8(6): 719-29, 2006 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-16528760

RESUMO

BACKGROUND: Gaucher disease is the most common of the lysosomal storage disorders. The primary manifestation is the accumulation of glucosylceramide (GL-1) in the macrophages of liver and spleen (Gaucher cells), due to a deficiency in the lysosomal hydrolase glucocerebrosidase (GC). A Gaucher mouse model (D409V/null) exhibiting reduced GC activity and accumulation of GL-1 was used to evaluate adeno-associated viral (AAV)-mediated gene therapy. METHODS: A recombinant AAV8 serotype vector bearing human GC (hGC) was administered intravenously to the mice. The levels of hGC in blood and tissues were determined, as were the effects of gene transfer on the levels of GL-1. Histopathological evaluation was performed on liver, spleen and lungs. RESULTS: Vector administration to pre-symptomatic Gaucher mice resulted in sustained hepatic secretion of hGC at levels that prevented GL-1 accumulation and the appearance of Gaucher cells in the liver, spleen and lungs. AAV administration to older mice with established disease resulted in normalization of GL-1 levels in the spleen and liver and partially reduced that in the lung. Analysis of the bronchoalveolar lavage fluid (BALF) from treated mice showed significant correction of the abnormal cellularity and cell differentials. No antibodies to the expressed hGC were detected following a challenge with recombinant enzyme suggesting the animals were tolerized to human enzyme. CONCLUSIONS: These data demonstrate the effectiveness of AAV-mediated gene therapy at preventing and correcting the biochemical and pathological abnormalities in a Gaucher mouse model, and thus support the continued consideration of this vector as an alternative approach to treating Gaucher disease.


Assuntos
Dependovirus/genética , Doença de Gaucher/enzimologia , Doença de Gaucher/patologia , Terapia Genética , Glucosilceramidase/genética , Glucosilceramidase/metabolismo , Vísceras/patologia , Animais , Líquido da Lavagem Broncoalveolar/citologia , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Doença de Gaucher/genética , Doença de Gaucher/terapia , Regulação Enzimológica da Expressão Gênica , Humanos , Tolerância Imunológica/imunologia , Fígado/citologia , Fígado/patologia , Pulmão/citologia , Pulmão/patologia , Macrófagos/citologia , Macrófagos/patologia , Camundongos , Camundongos Endogâmicos C57BL , Baço/citologia , Baço/patologia , Resultado do Tratamento
9.
Mol Ther ; 6(2): 179-89, 2002 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-12161184

RESUMO

Progress towards developing gene therapy for Gaucher disease has been hindered by the lack of an animal model. Here we describe a mouse model of Gaucher disease which has a chemically induced deficiency of glucocerebrosidase and that accumulates elevated levels of glucosylceramide (GL-1) in the lysosomes of Kupffer cells. Administration of mannose-terminated glucocerebrosidase (Cerezyme) resulted in the reduction of GL-1 levels in the livers of these animals. Gene transduction of hepatocytes with a plasmid DNA vector encoding human glucocerebrosidase (pGZB-GC) generated high-level expression and secretion of the enzyme into systemic circulation with consequent normalization of Kupffer cell GL-1 levels. This suggested that the de novo synthesized and unmodified enzyme produced by hepatocyte transduction was also capable of being delivered to the cells that are primarily affected in Gaucher disease. Immunolocalization studies also revealed that preferential transduction and expression of human glucocerebrosidase in the Kupffer cells with subsequent reduction in the GL-1 levels could be attained with a low dose of a recombinant adenoviral vector encoding the human enzyme (Ad2/CMV-GC). This observation raises the possibility of gene therapy for Gaucher disease that involves directly transducing the affected histiocytes using recombinant adenoviral vectors. Together, these data demonstrate the potential for use of in vivo gene therapy vectors for treating Gaucher disease.


Assuntos
Doença de Gaucher/terapia , Terapia Genética/métodos , Adenoviridae/genética , Animais , Modelos Animais de Doenças , Feminino , Doença de Gaucher/induzido quimicamente , Doença de Gaucher/enzimologia , Doença de Gaucher/genética , Expressão Gênica , Vetores Genéticos , Glucosilceramidase/deficiência , Glucosilceramidase/genética , Glucosilceramidase/metabolismo , Glucosilceramidase/uso terapêutico , Glucosilceramidas/metabolismo , Humanos , Células de Kupffer/metabolismo , Lisossomos/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Ratos , Ratos Endogâmicos F344
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...