Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Mol Genet Metab ; 117(4): 447-55, 2016 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-26857995

RESUMO

Mucolipidoses II and III (ML II and ML III) are lysosomal disorders in which the mannose 6-phosphate recognition marker is absent from lysosomal hydrolases and other glycoproteins due to mutations in GNPTAB, which encodes two of three subunits of the heterohexameric enzyme, N-acetylglucosamine-1-phosphotransferase. Both disorders are caused by the same gene, but ML II represents the more severe phenotype. Bone manifestations of ML II include hip dysplasia, scoliosis, rickets and osteogenesis imperfecta. In this study, we sought to determine whether a recombinant adeno-associated viral vector (AAV2/8-GNPTAB) could confer high and prolonged gene expression of GNPTAB and thereby influence the pathology in the cartilage and bone tissue of a GNPTAB knock out (KO) mouse model. The results demonstrated significant increases in bone mineral density and content in AAV2/8-GNPTAB-treated as compared to non-treated KO mice. We also showed that IL-6 (interleukin-6) expression in articular cartilage was reduced in AAV2/8-GNPTAB treated ML II mice. Together, these data suggest that AAV-mediated expression of GNPTAB in ML II mice can attenuate bone loss via inhibition of IL-6 production. This study emphasizes the value of the MLII KO mouse to recapitulate the clinical manifestations of the disease and highlights its amenability to therapy.


Assuntos
Desmineralização Patológica Óssea/etiologia , Dependovirus/genética , Expressão Gênica , Vetores Genéticos/genética , Mucolipidoses/genética , Mucolipidoses/patologia , Transdução Genética , Transferases (Outros Grupos de Fosfato Substituídos)/genética , Animais , Desmineralização Patológica Óssea/diagnóstico , Desmineralização Patológica Óssea/terapia , Densidade Óssea , Modelos Animais de Doenças , Ordem dos Genes , Marcação de Genes , Loci Gênicos , Terapia Genética , Vetores Genéticos/administração & dosagem , Genótipo , Humanos , Camundongos , Camundongos Knockout , Mucolipidoses/terapia , Fenótipo
2.
Pediatr Endocrinol Rev ; 11 Suppl 1: 99-109, 2013 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-24380128

RESUMO

Lysosomal storage diseases (LSDs) are a group of single-gene disorders that have proven to be highly informative in revealing the merits of gene transfer as a technology platform. Over the past several years considerable progress has been made in delivering therapeutic genes to peripheral tissues as well as the central nervous system. The current leading vectors for direct genetic modification of target cells in vivo are derived from adeno-associated viruses (AAV) and lentiviruses. These vectors are capable of conferring widespread, robust, and sustained expression of a given gene in several mouse models of LSDs. Here we review recent progress using recombinant AAV and lentiviruses to treat various LSDs and the remaining challenges to translate the results in mice to human patients.


Assuntos
Terapia Genética/métodos , Doenças por Armazenamento dos Lisossomos/terapia , Animais , Técnicas de Transferência de Genes , Humanos , Doenças por Armazenamento dos Lisossomos/genética
3.
J Gene Med ; 13(6): 324-32, 2011 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-21674735

RESUMO

BACKGROUND: The secretory form of acid sphingomyelinase (ASM) is postulated to play a key role in the retention and aggregation of lipoproteins in the subendothelial space of the arterial wall by converting sphingomyelin in lipoproteins into ceramide. The present study aimed to determine whether the level of circulating ASM activity affects lesion development in mouse model of atherosclerosis. METHODS: Apolipoprotein E deficient (ApoE(-/-) ) mice were injected intravenously with a recombinant adeno-associated virus (AAV8-ASM) that constitutively expressed high levels of human ASM in liver and plasma. RESULTS: Plasma sphingomyelin levels were reduced at early but not later time points after the administration of AAV8-ASM despite persistently elevated circulating ASM. No change in serum lipoprotein levels was observed. Thirteen or 17 weeks after the administration of AAV8-ASM, the amount of plaque formation in the aortic sinus was comparable to that of mice treated with a control AAV. CONCLUSIONS: Unexpectedly, the lesion area of the entire aorta was reduced significantly in the AAV8-ASM virus-treated group. Hepatic expression and secretion of ASM into the circulation did not accelerate or exacerbate, but rather decreased, lesion formation in ApoE(-/-) mice. Thus, plasma ASM activity does not appear to be rate limiting for plaque formation during atherogenesis.


Assuntos
Aorta/patologia , Apolipoproteínas E/genética , Dependovirus/metabolismo , Placa Aterosclerótica/enzimologia , Esfingomielina Fosfodiesterase/metabolismo , Análise de Variância , Animais , Técnicas Histológicas , Humanos , Lipoproteínas/sangue , Fígado/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Placa Aterosclerótica/patologia , Esfingomielina Fosfodiesterase/administração & dosagem , Esfingomielina Fosfodiesterase/sangue
4.
J Bone Miner Metab ; 28(5): 510-9, 2010 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-20300790

RESUMO

Studies examining the effects of hypoxia upon osteoclast biology have consistently revealed a stimulatory effect; both osteoclast differentiation and resorption activity have been shown to be enhanced in the presence of hypoxia. In the present study we examined the effects of the hypoxia mimetics dimethyloxallyl glycine (DMOG) and desferrioxamine (DFO) upon osteoclastogenesis. In contrast to hypoxia, our studies revealed a dose-dependent inhibition of osteoclast formation from macrophages treated with DMOG and DFO. Moreover, expression of a constitutively active form of hypoxia-inducible factor 1alpha (HIF-1alpha) did not enhance osteoclastogenesis and actually attenuated the differentiation process. DMOG did not affect cell viability or receptor activator of nuclear factor kappaB ligand (RANKL)-dependent phosphorylation of mitogen-activated protein (MAP) kinases. However, RANKL-dependent transcription of tartrate-resistant acid phosphatase (TRAP) was reduced in the presence of DMOG. Additionally, DMOG promoted transcription of the pro-apoptotic mediator B-Nip3. These studies suggest that a hypoxia-responsive factor other than HIF-1alpha is necessary for enhancing the formation of osteoclasts in hypoxic settings.


Assuntos
Diferenciação Celular/efeitos dos fármacos , Glicina , Osteoclastos , Pró-Colágeno-Prolina Dioxigenase/antagonistas & inibidores , Animais , Linhagem Celular , Desferroxamina/farmacologia , Feminino , Regulação da Expressão Gênica , Glicina/química , Glicina/farmacologia , Hipóxia/metabolismo , Subunidade alfa do Fator 1 Induzível por Hipóxia/genética , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Fator Estimulador de Colônias de Macrófagos/metabolismo , Camundongos , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Osteoclastos/efeitos dos fármacos , Osteoclastos/fisiologia , Ligante RANK/metabolismo , Ratos , Ratos Sprague-Dawley , Sideróforos/farmacologia , Fator A de Crescimento do Endotélio Vascular/metabolismo
5.
Mol Ther ; 16(8): 1400-8, 2008 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-18545223

RESUMO

Peripheral neuropathy is a particularly debilitating complication of both type 1 and type 2 diabetes characterized by sensory and motor neuron damage and decreased circulating levels of insulin-like growth factor 1 (IGF-1). Quite often, an early hyperalgesia is followed by hypoalgesia and muscle weakness. Hypoalgesia can lead to significant morbidity for which there is no current treatment. Hyperglycemic, streptozotocin (STZ)-induced rodent models reproduce these symptoms. We investigated whether increasing systemic IGF-1 could improve neuronal function in hyper- and hypoalgesic STZ-treated mice. Increased circulating levels of IGF-1 were achieved by delivering a plasmid or adeno-associated viral (AAV) vector bearing mouse IGF-1 to the liver. Treating mice in the hyperalgesia stage prevented later hypoalgesia. Treating mice in the hypoalgesia stage reversed existing hypoalgesia. This latter effect could be seen by merely restoring IGF-1 serum levels to normalcy, which was possible to achieve by IGF-1 gene therapy or insulin treatment. Sensory nerve functional correction was seen to be correlated with attenuated Schwann cell vacuolization and demyelination in peripheral sensory nerve fibers. A further increase in serum IGF-1 levels with gene therapy also improved motor function, consistent with the observed prevention of both muscle atrophy and peripheral motor nerve fiber demyelination. These results suggest that the restoration of systemic levels of IGF-1 may prove to be a highly effective therapeutic modality for treating diabetic peripheral neuropathy.


Assuntos
Neuropatias Diabéticas/terapia , Terapia Genética/métodos , Hiperalgesia/terapia , Fator de Crescimento Insulin-Like I/fisiologia , Animais , Peso Corporal/fisiologia , Dependovirus/genética , Diabetes Mellitus Experimental/complicações , Diabetes Mellitus Experimental/fisiopatologia , Neuropatias Diabéticas/etiologia , Neuropatias Diabéticas/fisiopatologia , Modelos Animais de Doenças , Vetores Genéticos/genética , Hiperalgesia/induzido quimicamente , Hiperalgesia/fisiopatologia , Fator de Crescimento Insulin-Like I/genética , Fator de Crescimento Insulin-Like I/metabolismo , Fígado/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Neurônios Motores/citologia , Neurônios Motores/metabolismo , Neurônios Motores/fisiologia
6.
Mol Ther ; 12(2): 264-73, 2005 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-15946902

RESUMO

Hepatocytes are an effective depot for protein production from gene therapy vectors. However, when gene transfer vectors or their delivery induces hepatic inflammation, adaptive immune responses against the transgene product can ensue. In BALB/c mice, hydrodynamic delivery of a CMV-driven plasmid DNA (pDNA) bearing human alpha-galactosidase A (alphagal) to the liver generated antibodies against alphagal. This humoral immune response was more robust in a transgenic knockout for alphagal, the Fabry mouse. The antibody response could be attenuated in both mouse strains by using a promoter more restricted to hepatocytes. In an attempt to reduce further the humoral responses to alphagal, expression from the transgene was attenuated by using siRNA during the period of initial delivery-associated liver inflammation. In both mouse models and with both promoters, codelivering an alphagal siRNA resulted in a 2 log decrease in initial expression that then increased over the next few weeks to levels generated by the pDNA alone. This strategy led to both attenuated antibodies and an immune status approximating "tolerance" to alphagal. Importantly, in the Fabry mouse, an alphagal siRNA together with a hepatocyte-restricted promoter gave minimal anti-alphagal antibodies and profound tolerance, suggesting that such an approach might have clinical utility for genetic diseases.


Assuntos
Hepatócitos/metabolismo , Tolerância Imunológica/efeitos dos fármacos , RNA Interferente Pequeno/farmacologia , alfa-Galactosidase/imunologia , Animais , Comunicação Celular , Modelos Animais de Doenças , Doença de Fabry , Expressão Gênica/efeitos dos fármacos , Técnicas de Transferência de Genes , Terapia Genética , Vetores Genéticos , Hepatócitos/imunologia , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Knockout , Plasmídeos/genética , Regiões Promotoras Genéticas , RNA Interferente Pequeno/uso terapêutico , Transfecção , alfa-Galactosidase/genética , alfa-Galactosidase/metabolismo
7.
J Gene Med ; 6(1): 85-92, 2004 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-14716680

RESUMO

BACKGROUND: Fabry disease is a recessive, X-linked disorder caused by a deficiency of the lysosomal enzyme alpha-galactosidase A, leading to an accumulation of the glycosphingolipid globotriaosylceramide (GL-3) in most tissues of the body. The goal of this study was to determine if systemic delivery of a nonviral vector could correct the enzyme deficiency and reduce the levels of GL-3 in different tissues of a transgenic knockout mouse model of the disease. METHODS: Cationic lipid was complexed with a CpG-depleted plasmid DNA vector and then injected intravenously into Fabry mice. The levels of alpha-galactosidase A and GL-3 in different tissues were assayed at various time points after injection. RESULTS: Expression of alpha-galactosidase A was detected in the different tissues of Fabry mice for up to 3 months after complex administration, but resulted in minimal reductions in GL-3 levels. However, the use of the anti-inflammatory drug dexamethasone and multiple dosing increased alpha-galactosidase A expression and resulted in significant reductions of GL-3 in all the organs with the exception of the kidney. In addition, injecting complex into young Fabry mice partially prevented the normal accumulation of GL-3 in the heart, lung, and liver. CONCLUSIONS: Systemic delivery of a cationic lipid-pDNA complex partially corrected the enzyme deficiency and reduced glycolipid storage in a mouse model of Fabry disease. The results are one of the few demonstrations of long-term efficacy in a genetic disease model using nonviral vectors. However, substantial improvements in expression, especially in critical organs such as the kidney, are required before these vectors can become a viable approach to treat Fabry disease and other lysosomal storage disorders.


Assuntos
Doença de Fabry/genética , Doença de Fabry/terapia , Terapia Genética , Vetores Genéticos , Triexosilceramidas/metabolismo , alfa-Galactosidase/genética , Animais , Modelos Animais de Doenças , Feminino , Expressão Gênica , Lipídeos , Camundongos , Camundongos Endogâmicos BALB C , Plasmídeos/genética , alfa-Galactosidase/biossíntese
8.
Hum Gene Ther ; 14(3): 203-14, 2003 Feb 10.
Artigo em Inglês | MEDLINE | ID: mdl-12639301

RESUMO

Systemic delivery of synthetic gene transfer vectors such as cationic lipid:plasmid DNA (pDNA) complexes elicits a range of acute physiologic responses, which in the context of therapeutic gene delivery represent dose-limiting toxicities. The most prominent responses are transient leukopenia, thrombocytopenia, serum transaminase elevations, and elevations of proinflammatory cytokines such as interferon-gamma (IFN-gamma), interleukin-12 (IL-12), and tumor necrosis factor-alpha (TNF-alpha). The unmethylated CpG sequences present in plasmid DNA have been implicated as a major cause of the robust cytokine response that follows systemic administration of cationic lipid:pDNA complexes. However, the factors causing the additional significant toxicities (leukopenia, thrombocytopenia, and serum transaminase elevations) recently shown to be associated with vector administration have not been defined. We show here that DNA sequences, such as immune stimulatory CpG sequences, play a significant role in inducing the additional acute toxicities associated with cationic lipid:pDNA complex administration. Importantly, while methylating these CpG sequences results in greatly reduced cytokine levels, this modification does not eliminate their ability to generate the other systemic toxicities. Examples of non-CpG DNA sequences that induce distinct toxicity profiles when administered systemically in the form of cationic lipid:DNA complexes are also identified. Taken together, these results imply that specific DNA sequences are responsible for a significant portion of the systemic toxicities observed after administration of cationic lipid:pDNA complexes.


Assuntos
DNA/metabolismo , Metabolismo dos Lipídeos , Animais , Ilhas de CpG , Citocinas/metabolismo , DNA/administração & dosagem , DNA/toxicidade , Metilação de DNA , Interleucina-12/metabolismo , Lipídeos/administração & dosagem , Lipídeos/toxicidade , Camundongos , Camundongos Endogâmicos BALB C
9.
J Thorac Cardiovasc Surg ; 124(2): 259-69, 2002 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-12167785

RESUMO

OBJECTIVE: Multiple gene transfer might permit modulation of concurrent biochemical pathways involved in lung graft ischemia-reperfusion injury. In this study we analyzed whether recipient intramuscular naked plasmid cotransfection of transforming growth factor beta(1) and interleukin 10 would result in amelioration of lung graft ischemia-reperfusion injury. METHODS: Forty-eight hours before transplantation, 6 groups (n = 6) of F344 rats received intramuscular injection of naked plasmid encoding chloramphenicol acetyltransferase, chloramphenicol acetyltransferase plus beta-galactosidase, transforming growth factor beta(1), interleukin 10, or transforming growth factor beta(1) plus interleukin 10 or were not treated. Donor lungs were flushed and stored for 18 hours at 4 degrees C before transplantation. Twenty-four hours later, grafts were assessed immediately before the animals were killed. Arterial oxygenation, wet/dry ratio, myeloperoxidase, and proinflammatory cytokines (interleukin 1, tumor necrosis factor alpha, interferon gamma, and interleukin 2) were measured, and immunohistochemistry was performed. RESULTS: For lung graft function, the arterial oxygenation was considerably higher in the cotransfected group receiving transforming growth factor beta(1) plus interleukin 10 compared with that in all other groups (P < or =.03). The wet/dry ratio, reflecting lung edema, was reduced in the cotransfected group compared with that in control animals (nontreated, P <.02; chloramphenicol acetyltransferase, P <.03; chloramphenicol acetyltransferase plus beta-galactosidase, P <.01). Myeloperoxidase, which measures neutrophil sequestration, was also reduced with cotransfection compared with that seen in control animals (P < or =.03). All proinflammatory cytokines were decreased in the cotransfected group compared with those in all other groups (interleukin 1beta, P <.04; tumor necrosis factor alpha, P <.002; interferon gamma, P <.0001; interleukin 2, P <.03). These results indicate that cotransfection provides a synergistic benefit in graft function versus either cytokine alone, neutrophil sequestration, or inflammatory cytokine expression. Immunohistochemistry showed positive staining of transforming growth factor beta(1) plus interleukin 10 in type I and II pneumocytes and localized edema fluid. CONCLUSIONS: Recipient intramuscular naked plasmid cotransfection of transforming growth factor beta(1) and interleukin 10 provides a synergistic effect in ameliorating lung reperfusion injury after prolonged ischemia.


Assuntos
Expressão Gênica , Técnicas de Transferência de Genes , Interleucina-10/genética , Transplante de Pulmão , Pulmão/metabolismo , Traumatismo por Reperfusão/prevenção & controle , Fator de Crescimento Transformador beta/genética , Análise de Variância , Animais , Citocinas/metabolismo , Modelos Animais de Doenças , Ensaio de Imunoadsorção Enzimática , Imuno-Histoquímica , Interleucina-10/farmacologia , Masculino , Peroxidase/metabolismo , Plasmídeos , Ratos , Ratos Endogâmicos F344 , Traumatismo por Reperfusão/etiologia , Transfecção , Fator de Crescimento Transformador beta/farmacologia
10.
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
11.
Mol Ther ; 5(6): 745-54, 2002 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-12027559

RESUMO

Gene therapy efforts have focused primarily on the use of either the liver or skeletal muscle as depot organs for the production of a variety of therapeutic proteins that act systemically. Here we examined the lung to determine whether it could function as yet another portal for the secretion of proteins into the circulation. Fabry disease is caused by a deficiency of the lysosomal hydrolase alpha-galactosidase A, resulting in the abnormal deposition of the glycosphingolipid globotriaosylceramide (GL-3) in vascular lysosomes. Pulmonary instillation of a recombinant adenoviral vector (Ad2/CMVHI-alpha(gal)) encoding human alpha-galactosidase A into Fabry mice resulted in high-level transduction and expression of the enzyme in the lung. Importantly, enzymatic activity was also detected in the plasma, liver, spleen, heart, and kidneys of the Fabry mice. The detection of enzymatic activity outside of the lung, along with the finding that viral DNA was limited to the lung, indicates that the enzyme crossed the air/blood barrier, entered the systemic circulation, and was internalized by the distal visceral organs. The levels of alpha-galactosidase A attained in these tissues were sufficient to reduce GL-3 to basal levels in the lung, liver, and spleen and to approximately 50% of untreated levels in the heart. Together, these results suggest that the lung may be a viable alternate depot organ for the production and systemic secretion of alpha-galactosidase A for Fabry disease.


Assuntos
Adenoviridae/genética , Doença de Fabry/terapia , Terapia Genética , Pulmão/metabolismo , Transdução Genética , alfa-Galactosidase/genética , Adenoviridae/metabolismo , Animais , Vetores Genéticos , Humanos , Pulmão/citologia , Lisossomos/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Camundongos SCID , alfa-Galactosidase/sangue , alfa-Galactosidase/uso terapêutico
12.
Hum Gene Ther ; 13(8): 935-45, 2002 May 20.
Artigo em Inglês | MEDLINE | ID: mdl-12031126

RESUMO

Systemic administration of recombinant adenoviral vectors for gene therapy of chronic diseases such as Fabry disease can be limited by dose-dependent toxicity. Because administration of a high dose of Ad2/CMVHI-alpha gal encoding human alpha-galactosidase A results in expression of supraphysiological levels of the enzyme, we sought to determine whether lower doses would suffice to correct the enzyme deficiency and lysosomal storage abnormality observed in Fabry mice. Reducing the dose of Ad2/CMVHI-alpha gal by 10-fold (from 10(11) to 10(10) particles/mouse) resulted in a greater than 200-fold loss in transgene expression. In Fabry mice, the reduced expression of alpha-galactosidase A, using the lower dose of Ad2/CMVHI-alpha gal, was associated with less than optimal clearance of the accumulated glycosphingolipid (GL-3) from the affected lysosomes. It was determined that this lack of linearity in dose response was not due to an inability to deliver the recombinant viral vectors to the liver but rather to sequestration, at least in part, of the viral vectors by the Kupffer cells. This lack of correlation between dose and expression levels could be obviated by supplementing the low dose of Ad2/CMVHI-alpha gal with an unrelated adenoviral vector or by depleting the Kupffer cells before administration of Ad2/CMVHI-alpha gal. Prior removal of the Kupffer cells, using clodronate liposomes, facilitated the use of a 100-fold lower dose of Ad2/CMVHI-alpha gal (10(9) particles/mouse) to effect the nearly complete clearance of GL-3 from the affected organs of Fabry mice. These results suggest that practical strategies that minimize the interaction between the recombinant adenoviral vectors and the reticuloendothelial system (RES) may improve the therapeutic window of this vector system. In this regard, we showed that pretreatment of mice with gamma globulins also resulted in significantly enhanced adenovirus-mediated transduction and expression of alpha-galactosidase A in the liver.


Assuntos
Adenoviridae/genética , Doença de Fabry/terapia , Terapia Genética , Vetores Genéticos , Animais , Ácido Clodrônico/farmacologia , Relação Dose-Resposta a Droga , Feminino , Humanos , Células de Kupffer/metabolismo , Fígado/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Transdução Genética , alfa-Galactosidase/genética , alfa-Galactosidase/metabolismo , gama-Globulinas/farmacologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA