Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
Mais filtros

Bases de dados
Tipo de documento
País de afiliação
Intervalo de ano de publicação
1.
Proc Natl Acad Sci U S A ; 105(7): 2616-21, 2008 Feb 19.
Artigo em Inglês | MEDLINE | ID: mdl-18268347

RESUMO

Enzyme replacement therapy has been used successfully in many lysosomal storage diseases. However, correction of brain storage has been limited by the inability of infused enzyme to cross the blood-brain barrier. The newborn mouse is an exception because recombinant enzyme is delivered to neonatal brain after mannose 6-phosphate receptor-mediated transcytosis. Access to this route is very limited after 2 weeks of age. Recently, several studies showed that multiple infusions of high doses of enzyme partially cleared storage in adult brain. These results raised the question of whether correction of brain storage by repeated high doses of enzyme depends on mannose 6-phosphate receptor-mediated uptake or whether enzyme gains access to brain storage by another route when brain capillaries are exposed to prolonged, high levels of circulating enzyme. To address this question, we used an enzyme whose carbohydrate-dependent receptor-mediated uptake was inactivated by chemical modification. Treatment of human beta-glucuronidase (GUS) with sodium metaperiodate followed by sodium borohydride reduction (PerT-GUS) eliminated uptake by mannose 6-phosphate and mannose receptors in cultured cells and dramatically slowed its plasma clearance from a t(1/2) of <10 min to 18 h. Surprisingly, PerT-GUS infused weekly for 12 weeks was more effective in clearing central nervous system storage than native GUS at the same dose. In fact, PerT-GUS resulted in almost complete reversal of storage in neocortical and hippocampal neurons. This enhanced correction of neuronal storage by long-circulating enzyme, which targets no known receptor, suggests a delivery system across the blood-brain barrier that might be exploited therapeutically.


Assuntos
Barreira Hematoencefálica/efeitos dos fármacos , Glucuronidase/química , Glucuronidase/uso terapêutico , Mucopolissacaridose VII/tratamento farmacológico , Neurônios/efeitos dos fármacos , Neurônios/enzimologia , beta-Glucosidase/química , beta-Glucosidase/uso terapêutico , Animais , Boroidretos/uso terapêutico , Células Cultivadas , Estabilidade Enzimática , Glucuronidase/genética , Glucuronidase/isolamento & purificação , Humanos , Camundongos , Mucopolissacaridose VII/enzimologia , Mucopolissacaridose VII/patologia , Ácido Periódico/uso terapêutico , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/isolamento & purificação , Proteínas Recombinantes/uso terapêutico , Temperatura , beta-Glucosidase/genética , beta-Glucosidase/isolamento & purificação
2.
Pediatr Blood Cancer ; 50(5): 1070-2, 2008 May.
Artigo em Inglês | MEDLINE | ID: mdl-18074390

RESUMO

Familial hemophagocytic lymphohistiocytosis (FHLH) is an autosomal recessive disorder of cytotoxic cell function that results in abnormal proliferation of benign lymphocytes and histiocytes in response to infectious stimuli. FHLH generally occurs in very young children, and typically presents with fever, cytopenias, coagulopathy, lymphadenopathy, and hepatosplenomegaly. Central nervous system involvement occurs frequently and may precede the development of systemic symptoms by months to years. We report a case of an 18-year-old male with a 2-year history of symptoms attributed to a demyelinating disorder, who succumbed to rapidly progressive hemophagocyte lymphohistiocytosis. Post-mortem, two distinct perforin mutations were identified. We discuss the central nervous system and genetic findings in this unusual presentation of familial hemophagocytic lymphohistiocytosis.


Assuntos
Linfo-Histiocitose Hemofagocítica/complicações , Doenças do Sistema Nervoso/etiologia , Perforina/deficiência , Adolescente , Transplante de Medula Óssea , Humanos , Linfo-Histiocitose Hemofagocítica/metabolismo , Linfo-Histiocitose Hemofagocítica/patologia , Masculino , Doenças do Sistema Nervoso/terapia
3.
Mol Ther ; 15(1): 44-52, 2007 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-17164774

RESUMO

Globoid-cell leukodystrophy (GLD) is a rapidly progressing inherited neurodegenerative disorder caused by a deficiency in galactosylceramidase activity. Previous studies in the murine model of GLD (Twitcher mouse) have shown that both bone marrow transplantation (BMT) and central nervous system (CNS)-directed gene therapy can be moderately effective at ameliorating certain aspects of GLD. As BMT and CNS-directed gene therapy target fundamentally different tissues, we tested the hypothesis that combining these disparate therapies would be more efficacious than either therapy alone. Mice receiving myeloreductive conditioning at birth followed by syngeneic BMT had approximately 25-35% donor chimerism. Untreated Twitcher mice, Twitcher mice treated with BMT alone, AAV2/5 alone, or a combination of BMT and AAV2/5 had mean lifespans of 39, 44, 49, and 104 days, respectively. Twitcher mice treated with a combination of BMT and AAV2/5 also had significantly improved performance in several behavioral tests and greater reduction in demyelination, astrocytosis, and macrophage infiltration compared to untreated Twitcher mice or mice that received either therapy alone. These data suggest that CNS-directed gene therapy synergizes with BMT. The combination of these disparate therapeutic approaches may form the basis for more effective treatment of this inherited neurodegenerative disorder.


Assuntos
Transplante de Medula Óssea , Sistema Nervoso Central/metabolismo , Dependovirus/genética , Modelos Animais de Doenças , Terapia Genética , Leucodistrofia de Células Globoides/genética , Leucodistrofia de Células Globoides/terapia , Animais , Comportamento Animal , Biomarcadores , Peso Corporal , Quimerismo , Leucodistrofia de Células Globoides/patologia , Leucodistrofia de Células Globoides/cirurgia , Longevidade , Camundongos , Camundongos Endogâmicos C57BL , Especificidade de Órgãos , Doadores de Tecidos , Condicionamento Pré-Transplante
4.
J Neurosci ; 23(8): 3302-7, 2003 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-12716937

RESUMO

The mucopolysaccharidoses (MPSs) are lysosomal storage diseases resulting from impaired catabolism of sulfated glycosaminoglycans. MPS VII mice lack lysosomal beta-glucuronidase (GUSB) activity, leading to the accumulation of partially degraded chondroitin, dermatan, and heparan sulfates in most tissues. Consequently, these mice develop most of the symptoms exhibited by human MPS VII patients, including progressive visual and cognitive deficits. To investigate the effects of reducing lysosomal storage in nervous tissues, we injected recombinant adeno-associated virus encoding GUSB directly into the vitreous humor of young adult mice. Interestingly, GUSB activity was subsequently detected in the brains of the recipients. At 8-12 weeks after treatment, increased GUSB activity and reduced lysosomal distension were found in regions of the thalamus and tectum that received inputs from the injected eye. Lysosomal storage was also reduced in adjacent nonvisual regions, including the hippocampus, as well as in the visual cortex. The findings suggest that both diffusion and trans-synaptic transfer contribute to the dissemination of enzyme activity within the CNS. Intravitreal injection may thus provide a means of delivering certain therapeutic gene products to specific areas within the CNS.


Assuntos
Sistema Nervoso Central/enzimologia , Terapia Genética/métodos , Glucuronidase/genética , Lisossomos/enzimologia , Mucopolissacaridose VII/enzimologia , Animais , Sistema Nervoso Central/patologia , Sistema Nervoso Central/virologia , DNA Viral/análise , Dependovirus/genética , Modelos Animais de Doenças , Vias de Administração de Medicamentos , Ativação Enzimática/genética , Olho/enzimologia , Olho/virologia , Vetores Genéticos/administração & dosagem , Vetores Genéticos/genética , Glucuronidase/biossíntese , Glucuronidase/deficiência , Hipocampo/enzimologia , Hipocampo/patologia , Hipocampo/virologia , Humanos , Lisossomos/patologia , Camundongos , Camundongos Mutantes Neurológicos , Mucopolissacaridose VII/genética , Mucopolissacaridose VII/patologia , Reação em Cadeia da Polimerase , Colículos Superiores/enzimologia , Colículos Superiores/patologia , Colículos Superiores/virologia , Tálamo/enzimologia , Tálamo/patologia , Tálamo/virologia , Resultado do Tratamento , Córtex Visual/enzimologia , Córtex Visual/patologia , Córtex Visual/virologia , Vias Visuais/enzimologia , Vias Visuais/patologia , Vias Visuais/virologia
6.
Pediatr Dev Pathol ; 11(3): 185-92, 2008.
Artigo em Inglês | MEDLINE | ID: mdl-17990914

RESUMO

Infantile neuronal ceroid lipofuscinosis (INCL), also known as Santavuori-Haltia disease, is an inherited neurodegenerative disorder caused by a mutation in the gene encoding the lysosomal enzyme palmitoyl-protein-thioesterase-1 (PPT1). Fatty acid-modified proteins are not degraded and accumulate as granular osmiophilic deposits in cells in the central nervous system; patients have blindness, seizures, progressive psychomotor deterioration, and die in early childhood. Although the disease manifests clinically primarily with neurological symptoms, visceral storage also accumulates. A murine model of INCL due to PPT1 deficiency exhibits clinical findings and pathology similar to those seen in patients with INCL. Homozygous PPT1-deficient mice have a shortened life span and neurological abnormalities including seizures, blindness, and mental and motor deficits. Widespread granular osmiophilic deposits (GRODs) accumulate in lysosomes in neurons and glia in the brain, retinal cells, kidney glomerular cells, aortic smooth muscle cells, and, in lesser amounts, in the fixed-tissue macrophage system. Accumulation of GRODs in aortic smooth muscle cells is accompanied by abnormalities in cardiac function and aortic root dilatation. This PPT1-deficient murine model is a well-defined genetic system that can be used to test potential therapies for lysosomal storage disease and to study the pathophysiology of INCL.


Assuntos
Encéfalo/ultraestrutura , Coração/fisiopatologia , Corpos de Inclusão/ultraestrutura , Lipofuscinoses Ceroides Neuronais/patologia , Vísceras/patologia , Animais , Modelos Animais de Doenças , Ecocardiografia Doppler , Camundongos , Camundongos Knockout , Microscopia Eletrônica de Transmissão , Miocárdio/patologia , Retina/ultraestrutura , Tioléster Hidrolases/deficiência
7.
Mol Genet Metab ; 91(1): 61-8, 2007 May.
Artigo em Inglês | MEDLINE | ID: mdl-17321776

RESUMO

Treatment of brain disease with recombinant proteins is difficult due to the blood-brain barrier. As an alternative to direct injections into the brain, we studied whether application of high concentrations of therapeutic enzymes via intrathecal (IT) injections could successfully drive uptake across the ependyma to treat brain disease. We studied IT enzyme replacement therapy with recombinant human iduronidase (rhIDU) in canine mucopolysaccharidosis I (MPS I, Hurler syndrome), a lysosomal storage disorder with brain and meningeal involvement. Monthly or quarterly IT treatment regimens with rhIDU achieved supranormal iduronidase enzyme levels in the brain, spinal cord, and spinal meninges. All regimens normalized total brain glycosaminoglycan (GAG) storage and reduced spinal meningeal GAG storage by 58-70%. The improvement in GAG storage levels persisted three months after the final IT dose. The successful use of enzyme therapy via the CSF represents a potentially useful approach for lysosomal storage disorders.


Assuntos
Encefalopatias/tratamento farmacológico , Iduronidase/administração & dosagem , Mucopolissacaridose I/tratamento farmacológico , Animais , Encéfalo/efeitos dos fármacos , Encéfalo/metabolismo , Encéfalo/patologia , Encefalopatias/patologia , Modelos Animais de Doenças , Cães , Relação Dose-Resposta a Droga , Glicosaminoglicanos/metabolismo , Humanos , Iduronidase/líquido cefalorraquidiano , Iduronidase/uso terapêutico , Injeções Espinhais , Meninges/efeitos dos fármacos , Meninges/metabolismo , Meninges/patologia , Mucopolissacaridose I/patologia , Proteínas Recombinantes/administração & dosagem , Proteínas Recombinantes/líquido cefalorraquidiano , Proteínas Recombinantes/uso terapêutico , Medula Espinal/efeitos dos fármacos , Medula Espinal/metabolismo , Medula Espinal/patologia , Distribuição Tecidual
8.
Proc Natl Acad Sci U S A ; 103(41): 15172-7, 2006 Oct 10.
Artigo em Inglês | MEDLINE | ID: mdl-17015822

RESUMO

Enzyme replacement therapy (ERT) is available for several lysosomal storage diseases. Except for Gaucher disease, for which an enzyme with exposed mannosyl residues targets mannose receptors (MR) on macrophages, ERT targets primarily the mannose 6-phosphate receptor (MPR). Most recombinant lysosomal enzymes contain oligosaccharides with both terminal mannosyl and mannose 6-phosphate residues. Effective MPR-mediated delivery may be compromised by rapid clearance of infused enzyme by the MR on fixed tissue macrophages, especially Kupffer cells. To evaluate the impact of this obstacle to ERT, we introduced the MR-null mutation onto the mucopolysaccharidosis type VII (MPS VII) background and produced doubly deficient MR-/- MPS VII mice. The availability of both MR+/+ and MR-/- mice allowed us to study the effects of eliminating the MR on MR- and MPR-mediated plasma clearance and tissue distribution of infused phosphorylated (P) and nonphosphorylated (NP) forms of human beta-glucuronidase (GUS). In MR+/+ MPS VII mice, the MR clearance system predominated at doses up to 6.4 mg/kg P-GUS. Genetically eliminating the MR slowed plasma clearance of both P- and NP-GUS and enhanced the effectiveness of P-GUS in clearing storage in kidney, bone, and retina. Saturating the MR clearance system by high doses of enzyme also improved targeting to MPR-containing tissues such as muscle, kidney, heart, and hepatocytes. Although ablating the MR clearance system genetically is not practical clinically, blocking the MR-mediated clearance system with high doses of enzyme is feasible. This approach delivers a larger fraction of enzyme to MPR-expressing tissues, thus enhancing the effectiveness of MPR-targeted ERT.


Assuntos
Glucuronidase/uso terapêutico , Lectinas Tipo C/deficiência , Lectinas Tipo C/fisiologia , Lectinas de Ligação a Manose/deficiência , Lectinas de Ligação a Manose/fisiologia , Manosefosfatos/metabolismo , Mucopolissacaridose VII/tratamento farmacológico , Mucopolissacaridose VII/genética , Receptor IGF Tipo 2/fisiologia , Receptores de Superfície Celular/deficiência , Receptores de Superfície Celular/fisiologia , Animais , Glucuronidase/deficiência , Glucuronidase/genética , Glucuronidase/metabolismo , Lectinas Tipo C/genética , Receptor de Manose , Lectinas de Ligação a Manose/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Mucopolissacaridose VII/enzimologia , Fosforilação , Receptores de Superfície Celular/genética
9.
Clin Immunol ; 119(2): 166-79, 2006 May.
Artigo em Inglês | MEDLINE | ID: mdl-16487752

RESUMO

Treatment of nonmalignant childhood disorders by bone marrow transplantation (BMT) is limited by toxicity from preparatory regimens and immune consequences associated with engraftment of allogeneic donor cells. Using costimulatory blockade (anti-CD40L mAb and CTLA-4Ig) combined with high-dose BMT in nonablated neonates, we obtained engraftment and established tolerance using both partially MHC mismatched (H2g7 into H2b) and fully mismatched BM (H2s into H2b). Recipients were mucopolysaccharidosis type VII (MPS VII) mice with lysosomal storage disease in order to assess therapeutic outcome. Recipients treated with donor lymphocyte infusion (DLI) amplified microchimerism to full donor. Recipients without DLI maintained long-term engraftment, tolerance, and had extended life spans. DLI increased donor cell mediated replacement of beta-glucuronidase (GUSB) activity in all tissues and maintained clearance of lysosomes better than in non-DLI-treated mice. DLI amplification of partially mismatched BM and fully mismatched BM caused late onset chronic GvHD in 56% and 100% of recipients, respectively.


Assuntos
Transplante de Medula Óssea , Ativação Linfocitária , Transfusão de Linfócitos , Doenças por Armazenamento dos Lisossomos/terapia , Agonistas Mieloablativos , Animais , Quimera , Doença Enxerto-Hospedeiro/metabolismo , Doença Enxerto-Hospedeiro/patologia , Cardiopatias/terapia , Imuno-Histoquímica , Rim/patologia , Teste de Cultura Mista de Linfócitos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos NOD , Transplante Homólogo
10.
Pediatr Dev Pathol ; 8(4): 453-62, 2005.
Artigo em Inglês | MEDLINE | ID: mdl-16222480

RESUMO

Lysosomal storage diseases (LSDs), due to deficiency of a lysosomal enzyme, are inherited, progressive disorders that are often fatal during childhood. The mucopolysaccharidoses (MPS) are LSDs caused by deficiency of a lysosomal enzyme needed for the stepwise degradation of glycosaminoglycans. A murine model of MPS VII shares many clinical, biochemical, and pathologic features with human MPS and has proved valuable for the study of the pathophysiology of MPS and for evaluation of therapies for LSDs. Early therapy of MPS VII mice, initiated in the first weeks of life, is much more effective in decreasing clinical and morphologic evidence of disease than treatment begun in mature animals. Whether such early therapy decreases existing storage or prevents its accumulation is incompletely investigated. We performed an analysis of storage in very young MPS VII mice to define the extent of disease at and before the time of initiation of early treatments. MPS VII pups from 12 days postcoitus (dpc) to 31 days postnatal (dpn) were studied. Storage accumulated in fixed tissue macrophages in the liver and cartilage as soon as 12 dpc and was present in central nervous system glia, leptomeninges, and perivascular cells by 15 dpc. Osteoblast and primitive neocortical cell storage was apparent at 18 to 19 dpc. At 2 dpn, lysosomal distention appeared in circulating leukocytes. Abundant lysosomal storage was present in many sites by 14 dpn. Secondary accumulation of beta-hexosaminidase paralleled increasing glycosaminoglycan storage. These results confirm the presence of widespread storage even in utero and in the very young MPS VII mouse and highlight the importance of early treatment to prevent storage accumulation.


Assuntos
Animais Recém-Nascidos/metabolismo , Feto/metabolismo , Mucopolissacaridose VII/patologia , Animais , Osso e Ossos/anormalidades , Osso e Ossos/diagnóstico por imagem , Cartilagem/metabolismo , Cartilagem/patologia , Sistema Nervoso Central/metabolismo , Sistema Nervoso Central/patologia , Modelos Animais de Doenças , Feminino , Fígado/metabolismo , Fígado/patologia , Lisossomos/metabolismo , Lisossomos/ultraestrutura , Macrófagos/metabolismo , Macrófagos/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Mutantes , Mucopolissacaridose VII/metabolismo , Osteoblastos/metabolismo , Osteoblastos/patologia , Gravidez , Radiografia , Vísceras/metabolismo , beta-N-Acetil-Hexosaminidases/metabolismo
11.
Proc Natl Acad Sci U S A ; 102(41): 14777-82, 2005 Oct 11.
Artigo em Inglês | MEDLINE | ID: mdl-16162667

RESUMO

Enzyme replacement therapy (ERT) effectively reverses storage in several lysosomal storage diseases. However, improvement in brain is limited by the blood-brain barrier except in the newborn period. In this study, we asked whether this barrier could be overcome by higher doses of enzyme than are used in conventional trials. We measured the distribution of recombinant human beta-glucuronidase (hGUS) and reduction in storage by weekly doses of 0.3-40 mg/kg administered i.v. over 1-13 weeks to mucopolysaccharidosis type VII mice immunotolerant to recombinant hGUS. Mice given up to 5 mg/kg enzyme weekly over 3 weeks had moderate reduction in meningeal storage but no change in neo-cortical neurons. Mice given 20-40 mg/kg three times over 1 week showed no reduction in storage in any area of the CNS except the meninges. In contrast, mice receiving 4 mg/kg per week for 13 weeks showed clearance not only in meninges but also in parietal neocortical and hippocampal neurons and glia. Mice given 20 mg/kg once weekly for 4 weeks also had decreased neuronal, glial, and meningeal storage and averaged 2.5% of wild-type hGUS activity in brain. These results indicate that therapeutic enzyme can be delivered across the blood-brain barrier in the adult mucopolysaccharidosis type VII mouse if administered at higher doses than are used in conventional ERT trials and if the larger dose of enzyme is administered over a sufficient period. These results may have important implications for ERT for lysosomal storage diseases with CNS involvement.


Assuntos
Barreira Hematoencefálica/metabolismo , Glucuronidase/farmacocinética , Glucuronidase/uso terapêutico , Mucopolissacaridose VII/tratamento farmacológico , Mucopolissacaridose VII/enzimologia , Animais , Relação Dose-Resposta a Droga , Glucuronidase/metabolismo , Técnicas Histológicas , Humanos , Meninges/metabolismo , Meninges/patologia , Camundongos , Camundongos Transgênicos , Neuroglia/metabolismo , Neuroglia/patologia , Neurônios/metabolismo , Neurônios/patologia
12.
Mol Ther ; 12(3): 422-30, 2005 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-15996520

RESUMO

Globoid-cell leukodystrophy (GLD) is an autosomal recessive lysosomal storage disorder caused by mutations in the galactosylceramidase (GALC) gene. Infantile GLD has a lethal course with severe cerebral demyelination that progresses to death by 2 years of age. In the current study twitcher mice, an authentic murine model of infantile GLD, were given intracranial injections of either recombinant adeno-associated virus serotype 2 encoding the murine Galc cDNA (AAV2-GALC) or the same genome pseudotyped with AAV5 capsid proteins (AAV2/5-GALC) on day 3 of age. The group injected intracranially with AAV2/5-GALC had approximately 25-fold greater than normal Galc levels in the brain, while AAV2-GALC-injected animals had 28% normal levels. The average life expectancy of twitcher mice ( approximately 38 days) was significantly (P < 0.0001) increased to 48 and 52 days for the AAV2-GALC- and AAV2/5-GALC-treated groups, respectively. The AAV2/5-GALC group performed significantly better in a battery of behavioral tests compared to untreated, AAV2-GFP-treated, or AAV2-treated twitcher animals. This longitudinal study demonstrated that AAV2/5-GALC-mediated gene therapy resulted in higher levels of Galc expression and slowed the neurologic deterioration more completely than AAV2-GALC in the murine model of globoid-cell leukodystrophy. However, the clinical improvements, as assessed by behavioral tests and life span, were only modest.


Assuntos
Doenças do Sistema Nervoso Central/enzimologia , Dependovirus/genética , Galactosilceramidase/genética , Terapia Genética/instrumentação , Vetores Genéticos/genética , Leucodistrofia de Células Globoides/terapia , Animais , Comportamento Animal , Peso Corporal , Encéfalo/metabolismo , Encéfalo/patologia , Doenças do Sistema Nervoso Central/genética , DNA Complementar/metabolismo , Modelos Animais de Doenças , Técnicas de Transferência de Genes , Terapia Genética/métodos , Genótipo , Proteínas de Fluorescência Verde/metabolismo , Homozigoto , Camundongos , Mutação , Neurônios/metabolismo , Fenótipo , Reação em Cadeia da Polimerase , Fatores de Tempo
13.
Mol Ther ; 12(2): 345-52, 2005 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-16043103

RESUMO

We used recombinant forms of human beta-glucuronidase (GUS) purified from secretions from stably transfected CHO cells to compare the native enzyme to a GUS-Tat C-terminal fusion protein containing the 11-amino-acid HIV Tat protein transduction domain for: (1) susceptibility to endocytosis by cultured cells, (2) rate of clearance following intravenous infusion, and (3) tissue distribution and effectiveness in clearing lysosomal storage following infusion in the MPS VII mouse. We found: (1) Native GUS was more efficiently taken up by cultured human fibroblasts and its endocytosis was exclusively mediated by the M6P receptor. The GUS-Tat fusion protein showed only 30-50% as much M6P-receptor-mediated uptake, but also was taken up by adsorptive endocytosis through binding of the positively charged Tat peptide to cell surface proteoglycans. (2) GUS-Tat was less rapidly cleared from the circulation in the rat (t(1/2) = 13 min vs 7 min). (3) Delivery to most tissues of the MPS VII mouse was similar, but GUS-Tat was more efficiently delivered to kidney. Histology showed that GUS-Tat more efficiently reduced storage in renal tubules, retina, and bone. These studies demonstrate that Tat modification can extend the range of tissues corrected by infused enzyme.


Assuntos
Produtos do Gene tat/metabolismo , Glucuronidase/metabolismo , Mucopolissacaridose VII/metabolismo , Animais , Células CHO , Células Cultivadas , Cricetinae , Cricetulus , Endocitose , Fibroblastos/metabolismo , Terapia Genética , Glucuronidase/química , Glucuronidase/genética , Masculino , Manosefosfatos/metabolismo , Camundongos , Camundongos Transgênicos , Mucopolissacaridose VII/genética , Mucopolissacaridose VII/terapia , Ratos , Ratos Sprague-Dawley , Proteínas Recombinantes/metabolismo
14.
Proc Natl Acad Sci U S A ; 100(5): 2669-73, 2003 Mar 04.
Artigo em Inglês | MEDLINE | ID: mdl-12591953

RESUMO

beta-Glucuronidase (GUSB) is a lysosomal enzyme important in the normal step-wise degradation of glycosaminoglycans. Deficiency of GUSB causes the lysosomal storage disease mucopolysaccharidosis VII (MPS VII, Sly disease). Affected patients have widespread progressive accumulation of beta-glucuronide-containing glycosaminoglycans in lysosomes. Enzyme replacement, bone marrow transplantation, and gene therapy can correct lysosomal storage in the MPS VII mouse model. Gene therapy in MPS VII patients and animals may result in massive overexpression of GUSB in individual tissues, and the toxicity of such overexpression is incompletely investigated. To gain insight into the effect of massive overexpression of GUSB, we established 19 transgenic mouse lines, two of which expressed very high levels of human GUSB in many tissues. The founder overexpressing mice had from >100- to several thousand-fold increases in tissue and serum GUSB. The enzyme expression in most tissues decreased in subsequent generations in one line, and expression in liver and marrow fell in subsequent generations of the other. Both lines had morphologically similar widespread lysosomal storage of GUSB and secondary elevations of other lysosomal enzymes, a finding characteristic of lysosomal storage disease. One line developed tumors, and one did not. These transgenic models show that massive overexpression of a lysosomal enzyme can be associated with dramatic morphological alterations, which, at least in one of the two lines, had little clinical consequence. For the other transgenic line, the high frequency of tumor development in F(2) FVB progeny suggests that the vector used to generate the transgenic lines has an integration site-dependent potential to be oncogenic, at least in this strain background.


Assuntos
Glucuronidase/biossíntese , Glucuronidase/genética , Lisossomos/enzimologia , Mucopolissacaridose VII/terapia , Transgenes , Animais , Linhagem Celular , DNA Complementar/metabolismo , Feminino , Terapia Genética/métodos , Glucuronidase/fisiologia , Imuno-Histoquímica , Lisossomos/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Microscopia Eletrônica , Mucopolissacaridose VII/genética , Oncogenes/genética , Fatores Sexuais
15.
Neurobiol Dis ; 16(2): 360-9, 2004 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-15193292

RESUMO

Infantile neuronal ceroid lipofuscinosis (INCL) is the earliest onset form of a class of inherited neurodegenerative disease called Batten disease. INCL is caused by a deficiency in the lysosomal enzyme palmitoyl protein thioesterase-1 (PPT1). Autofluorescent storage material accumulates in virtually all tissues in INCL patients, including the brain, and leads to widespread neuronal loss and cortical atrophy. To determine the efficacy of viral-mediated gene therapy, we injected a recombinant adeno-associated virus 2 vector encoding human PPT1 (rAAV-PPT1) intracranially (I.C.) into a murine model of INCL. INCL mice given four I.C. injections of rAAV-PPT1 as newborns exhibited PPT1 activity near the injection sites and decreased secondary elevations of another lysosomal enzyme. In addition, storage material was decreased in cortical, hippocampal, and cerebellar neurons, and brain weights and cortical thicknesses were increased. These data demonstrate that an adeno-associated virus 2 (AAV2)-mediated gene therapy approach may provide some therapeutic benefit for INCL.


Assuntos
Adenoviridae/genética , Terapia Genética , Lipofuscinoses Ceroides Neuronais/terapia , Tioléster Hidrolases/genética , Animais , Encéfalo/patologia , Modelos Animais de Doenças , Fluorescência , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Mutantes , Lipofuscinoses Ceroides Neuronais/patologia
16.
Blood Cells Mol Dis ; 32(1): 199-213, 2004.
Artigo em Inglês | MEDLINE | ID: mdl-14757436

RESUMO

Lysosomal storage diseases (LSD) respond to bone marrow (BM) transplantation when donor-derived cells deliver needed enzyme. Hypothetically, the ubiquitous resident macrophages (MPhi) are the primary delivery vehicle of therapeutic protein. In mucopolysaccharidosis type VII (MPS VII) mice with LSD, transplanted mature MPhi reduce undegraded glycosaminoglycans (GAG) in the lysosome but are incapable of self-renewal, leading to return of storage after 1 month. We show here that a population of early BM-derived myeloid progenitors devoid of long-term hematopoietic stem cells (LT-HSC) engrafted MPS VII BM, released monocytes into peripheral blood (PBL), and engrafted tissues at known sites of resident MPhi. These primitive Mac-1- cells were sorted from normal whole BM and were defined by ER-MP12hi20-58med/hi labeling. Lysosomal storage was reduced in liver, spleen, thymus, heart, kidney, and bone. Cells persisted for 3 months, suggesting self-renewal capacity or a long half-life. Cells sorted from BM by ER-MP12-20hi marker expression (which are maturer myeloid cells that express Mac-1) engrafted tissues instead of BM and quantitatively repopulated less than cells derived from the ER-MP12hi20-58med/hi population. Also, reduction of lysosomal storage was variable and generally less when compared to that following transplantation of immature ER-MP12hi20-58med/hi cells. We conclude that primitive myeloid progenitors are more therapeutic for LSD than mature myeloid cells due to their greater longevity and increased capacity to seed tissues. The ability of cells derived from these primitive precursors to seed deep within tissues make them excellent candidates for both cellular therapy and gene transfer techniques to cure a wide range of metabolic diseases.


Assuntos
Transplante de Células-Tronco Hematopoéticas/métodos , Doenças por Armazenamento dos Lisossomos/terapia , Macrófagos/citologia , Células Progenitoras Mieloides/citologia , Células Progenitoras Mieloides/transplante , Animais , Células da Medula Óssea , Movimento Celular , Separação Celular , Sobrevivência de Enxerto , Antígeno de Macrófago 1/análise , Camundongos , Monócitos/citologia , Mucopolissacaridose VII/terapia , Especificidade de Órgãos
17.
Hum Mol Genet ; 12(24): 3349-58, 2003 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-14583446

RESUMO

Mucopolysaccharidosis IVA is an autosomal recessive disorder caused by a deficiency of N-acetylgalactosamine-6-sulfate sulfatase (GALNS), a lysosomal enzyme required for the stepwise degradation of keratan sulfate (KS) and chondroitin-6-sulfate (C6S). To generate a model for studies of the pathophysiology and of potential therapies, we disrupted exon 2 of Galns, the homologous murine gene. Homozygous Galns-/- mice have no detectable GALNS enzyme activity and show increased urinary glycosaminoglycan (GAGs) levels. These mice accumulate GAGs in multiple tissues including liver, kidney, spleen, heart, brain and bone marrow. At 2 months old, lysosomal storage is present primarily within reticuloendothelial cells such as Kupffer cells and cells of the sinusoidal lining of the spleen. Additionally, by 12 months old, vacuolar change is observed in the visceral epithelial cells of glomeruli and cells at the base of heart valves but it is not present in parenchymal cells such as hepatocytes and renal tubular epithelial cells. In the brain, hippocampal and neocortical neurons and meningeal cells had lysosomal storage. KS and C6S were more abundant in the cytoplasm of corneal epithelial cells of Galns-/- mice compared with wild-type mice by immunohistochemistry. Radiographs revealed no change in the skeletal bones of mice up to 12 months old. Thus, targeted disruption of the murine Galns gene has produced a murine model, which shows visceral storage of GAGs but lacks the skeletal features. The complete absence of GALNS in mutant mice makes them useful for studies of pharmacokinetics and tissue targeting of recombinant GALNS designed for enzyme replacement.


Assuntos
Condroitina Sulfatases/genética , Modelos Animais de Doenças , Mucopolissacaridose IV/genética , Animais , Quimera , Condroitina Sulfatases/deficiência , Marcação de Genes , Camundongos , Camundongos Knockout , Mucopolissacaridose IV/patologia , Fenótipo , Recombinação Genética , Células-Tronco
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA