Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 46
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Neuropathol Appl Neurobiol ; 47(2): 251-267, 2021 02.
Artigo em Inglês | MEDLINE | ID: mdl-32841420

RESUMO

AIM: To understand the progression of CLN1 disease and develop effective therapies we need to characterize early sites of pathology. Therefore, we performed a comprehensive evaluation of the nature and timing of early CLN1 disease pathology in the spinal cord, which appears especially vulnerable, and how this may affect behaviour. METHODS: We measured the spinal volume and neuronal number, and quantified glial activation, lymphocyte infiltration and oligodendrocyte maturation, as well as cytokine profile analysis during the early stages of pathology in Ppt1-deficient (Ppt1-/- ) mouse spinal cords. We then performed quantitative gait analysis and open-field behaviour tests to investigate the behavioural correlates during this period. RESULTS: We detected significant microglial activation in Ppt1-/- spinal cords at 1 month. This was followed by astrocytosis, selective interneuron loss, altered spinal volumes and oligodendrocyte maturation at 2 months, before significant storage material accumulation and lymphocyte infiltration at 3 months. The same time course was apparent for inflammatory cytokine expression that was altered as early as one month. There was a transient early period at 2 months when Ppt1-/- mice had a significantly altered gait that resembles the presentation in children with CLN1 disease. This occurred before an anticipated decline in overall locomotor performance across all ages. CONCLUSION: These data reveal disease onset 2 months (25% of life-span) earlier than expected, while spinal maturation is still ongoing. Our multi-disciplinary data provide new insights into the spatio-temporal staging of CLN1 pathogenesis during ongoing postnatal maturation, and highlight the need to deliver therapies during the presymptomatic period.


Assuntos
Interneurônios/patologia , Lipofuscinoses Ceroides Neuronais/patologia , Medula Espinal/patologia , Tioléster Hidrolases/deficiência , Animais , Animais Recém-Nascidos , Humanos , Camundongos , Camundongos Knockout
2.
Nat Commun ; 11(1): 4798, 2020 09 23.
Artigo em Inglês | MEDLINE | ID: mdl-32968066

RESUMO

Myeloid cells are known mediators of hypertension, but their role in initiating renin-induced hypertension has not been studied. Vitamin D deficiency causes pro-inflammatory macrophage infiltration in metabolic tissues and is linked to renin-mediated hypertension. We tested the hypothesis that impaired vitamin D signaling in macrophages causes hypertension using conditional knockout of the myeloid vitamin D receptor in mice (KODMAC). These mice develop renin-dependent hypertension due to macrophage infiltration of the vasculature and direct activation of renal juxtaglomerular (JG) cell renin production. Induction of endoplasmic reticulum stress in knockout macrophages increases miR-106b-5p secretion, which stimulates JG cell renin production via repression of transcription factors E2f1 and Pde3b. Moreover, in wild-type recipient mice of KODMAC/miR106b-/- bone marrow, knockout of miR-106b-5p prevents the hypertension and JG cell renin production induced by KODMAC macrophages, suggesting myeloid-specific, miR-106b-5p-dependent effects. These findings confirm macrophage miR-106b-5p secretion from impaired vitamin D receptor signaling causes inflammation-induced hypertension.


Assuntos
Hipertensão Renal/metabolismo , Hipertensão/metabolismo , Macrófagos/metabolismo , MicroRNAs/metabolismo , Nefrite/metabolismo , Renina/metabolismo , Animais , Medula Óssea , Transplante de Medula Óssea , Modelos Animais de Doenças , Fator de Transcrição E2F1/metabolismo , Estresse do Retículo Endoplasmático , Feminino , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Células Mieloides , Receptores de Calcitriol , Vitamina D
3.
Gene Ther ; 21(1): 28-36, 2014 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-24131981

RESUMO

Canine models have many advantages for evaluating therapy of human central nervous system (CNS) diseases. In contrast to nonhuman primate models, naturally occurring canine CNS diseases are common. In contrast to murine models, the dog's lifespan is long, its brain is large and the diseases affecting it commonly have the same molecular, pathological and clinical phenotype as the human diseases. We compared the ability of four intracerebrally injected adeno-associated virus vector (AAV) serotypes to transduce the dog brain with green fluorescent protein as the first step in using these vectors to evaluate both delivery and efficacy in naturally occurring canine homologs of human diseases. Quantitative measures of transduction, maximum diameter and area, identified both AAV2/9 and AAV2/rh10 as significantly more efficient than either AAV2/1 or AAV2/5 at transducing cerebral cortex, caudate nucleus, thalamus and internal capsule. Fluorescence co-labeling with cell-type-specific antibodies demonstrated that AAV2/9 and AAV2/rh10 were capable of primarily transducing neurons, although glial transduction was also identified and found to be more efficient with the AAV2/9 vector. These data are a prerequisite to evaluating the efficacy of recombinant AAV vectors carrying disease-modifying transgenes to treat naturally occurring canine models in preclinical studies of human CNS disease therapy.


Assuntos
Encéfalo/metabolismo , Dependovirus/genética , Vetores Genéticos , Transdução Genética , Animais , Encéfalo/virologia , Núcleo Caudado/metabolismo , Núcleo Caudado/virologia , Córtex Cerebral/metabolismo , Córtex Cerebral/virologia , Dependovirus/classificação , Dependovirus/fisiologia , Modelos Animais de Doenças , Cães , Proteínas de Fluorescência Verde/genética , Humanos , Cápsula Interna/metabolismo , Cápsula Interna/virologia , Sorotipagem , Tálamo/metabolismo , Tálamo/virologia , Transgenes
4.
Gene Ther ; 20(9): 913-21, 2013 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-23535899

RESUMO

Mucopolysaccharidosis type IIIB (MPS IIIB) or Sanfilippo Syndrome type B is a lysosomal storage disease resulting from the deficiency of N-acetyl glucosaminidase (NAGLU) activity. We previously showed that intracranial adeno-associated virus (AAV)-based gene therapy results in partial improvements of several aspects of the disease. In an attempt to further correct the disease, MPS IIIB mice were treated at 2-4 days of age with intracranial AAV2/5-NAGLU (IC-AAV), intravenous lentiviral-NAGLU (IV-LENTI) or the combination of both (BOTH). The BOTH group had the most complete biochemical and histological improvements of any treatment group. Compared with untreated MPS IIIB animals, all treatments resulted in significant improvements in motor function (rotarod) and hearing (auditory-evoked brainstem response). In addition, each treatment group had a significantly increased median life span compared with the untreated group (322 days). The combination arm had the greatest increase (612 days), followed by IC-AAV (463 days) and IV-LENTI (358 days). Finally, the BOTH group had nearly normal circadian rhythm measures with improvement in time to activity onset. In summary, targeting both the systemic and central nervous system disease of MPS IIIB early in life appears to be the most efficacious approach for this inherited metabolic disorder.


Assuntos
Acetilglucosaminidase/genética , Encéfalo/metabolismo , Encéfalo/patologia , Dependovirus/genética , Terapia Genética , Lentivirus/genética , Mucopolissacaridose III/fisiopatologia , Mucopolissacaridose III/terapia , Acetilglucosaminidase/metabolismo , Animais , Animais Recém-Nascidos , Ritmo Circadiano , Vetores Genéticos , Humanos , Fígado/enzimologia , Fígado/patologia , Pulmão/enzimologia , Pulmão/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Atividade Motora , Mucopolissacaridose III/metabolismo , Mucopolissacaridose III/patologia , Miocárdio/enzimologia , Miocárdio/patologia , Resultado do Tratamento
5.
Neuroscience ; 169(3): 1364-75, 2010 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-20621643

RESUMO

The vulnerability of oligodendrocytes to ischemic injury may contribute to functional loss in diseases of central white matter. Immunocytochemical methods to identify oligodendrocyte injury in experimental models rely on epitope availability, and fail to discriminate structural changes in oligodendrocyte morphology. We previously described the use of a lentiviral vector (LV) carrying enhanced green fluorescent protein (eGFP) under the myelin basic protein (MBP) promoter for selective visualization of oligodendrocyte cell bodies and processes. In this study, we used LV-MBP-eGFP to label oligodendrocytes in rat cerebral white matter prior to transient focal cerebral ischemia, and examined oligodendrocyte injury 24 h, 48 h and 1 week post-reperfusion by quantifying cell survival and assaying the integrity of myelin processes. There was progressive loss of GFP+ oligodendrocytes in ischemic white matter at 24 and 48 h. Surviving GFP+ cells had non-pyknotic nuclear morphology and were terminal deoxynucleotidyl transferase dUTP nick end labeling (TUNEL)-negative, but there was marked fragmentation of myelin processes as early as 24 h after stroke. One week after stroke, we observed a restoration of GFP+ oligodendrocytes in ischemic white matter, reflected both by cell counts and by structural integrity of myelin processes. Proliferating cells were not the main source of GFP+ oligodendrocytes, as revealed by bromodeoxyuridine (BrdU) incorporation. These observations identify novel transient structural changes in oligodendrocyte cell bodies and myelinating processes, which may have consequences for white matter function after stroke.


Assuntos
Ataque Isquêmico Transitório/patologia , Oligodendroglia/patologia , Animais , Proliferação de Células , Proteínas de Fluorescência Verde/biossíntese , Proteínas de Fluorescência Verde/genética , HIV/genética , Humanos , Masculino , Proteína Básica da Mielina/genética , Bainha de Mielina/patologia , Células-Tronco Neurais/patologia , Ratos , Ratos Sprague-Dawley , Fatores de Tempo
6.
J Inherit Metab Dis ; 30(2): 227-38, 2007 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-17308887

RESUMO

Mucopolysaccharidosis type VII (MPS VII) is a lysosomal storage disease caused by beta-glucuronidase (GUSB) deficiency. This disease exhibits a broad spectrum of clinical signs including skeletal dysplasia, retinal degeneration, cognitive deficits and hearing impairment. Sustained, high-level expression of GUSB significantly improves the clinical course of the disease in the murine model of MPS VII. Low levels of enzyme expression (1-5% of normal) can significantly reduce the biochemical and histopathological manifestations of MPS VII. However, it has not been clear from previous studies whether persistent, low levels of circulating GUSB lead to significant improvements in the clinical presentation of this disease. We generated a rAAV2 vector that mediates persistent, low-level GUSB expression in the liver. Liver and serum levels of GUSB were maintained at approximately 5% and approximately 2.5% of normal, respectively, while other tissue ranged from background levels to 0.9%. This level of activity significantly reduced the secondary elevations of alpha-galactosidase and the levels of glycosaminoglycans in multiple tissues. Interestingly, this level of GUSB was also sufficient to reduce lysosomal storage in neurons in the brain. Although there were small but statistically significant improvements in retinal function, auditory function, skeletal dysplasia, and reproduction in rAAV-treated MPS VII mice, the clinical deficits were still profound and there was no improvement in lifespan. These data suggest that circulating levels of GUSB greater than 2.5% will be required to achieve substantial clinical improvements in MPS VII.


Assuntos
Técnicas de Transferência de Genes , Glucuronidase/genética , Mucopolissacaridose VII/fisiopatologia , Animais , Doenças do Desenvolvimento Ósseo/etiologia , Doenças do Desenvolvimento Ósseo/patologia , Dependovirus/genética , Modelos Animais de Doenças , Vetores Genéticos , Glucuronidase/sangue , Glucuronidase/metabolismo , Glicosaminoglicanos/metabolismo , Audição , Fígado/enzimologia , Longevidade , Lisossomos/ultraestrutura , Camundongos , Mucopolissacaridose VII/complicações , Mucopolissacaridose VII/metabolismo , Mucopolissacaridose VII/patologia , Reprodução , Retina/fisiopatologia , Distribuição Tecidual , alfa-Galactosidase/metabolismo
7.
Bone ; 30(2): 352-9, 2002 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-11856642

RESUMO

Mucopolysaccharidosis type VII (MPS VII) is a heritable lysosomal storage disease caused by a deficiency in beta-glucuronidase (GUSB) activity, leading to progressive accumulation of undegraded glycosaminoglycans in many tissues. Clinical features include growth and mental retardation, hearing and visual defects, shortened lifespan, and skeletal deformities. A murine model of MPS VII has been described that shares many of the manifestations of the human disease, including the skeletal dysplasia. In this study we describe abnormalities in the cellular morphology and function of osteoclasts and a localized defect in bone formation rate in the MPS VII mouse. Ultrastructural analysis revealed that MPS VII osteoclasts fail to form ruffled border membranes and many appeared to be detached from the bone surface. Following bone marrow transplantation, osteoclasts derived from wild-type donors showed normal morphology and were closely associated with the bone surface in MPS VII recipients. In vitro bone resorption assays demonstrated that MPS VII osteoclasts formed significantly smaller and fewer pits than those formed by osteoclasts derived from normal mice of the same strain. Although osteoclast morphology and function appeared to be abnormal in the MPS VII mouse, interleukin-1 (IL-1)-induced osteoclastogenesis in vivo was not affected. In addition to the osteoclast defects, MPS VII mice demonstrated a slower rate of bone matrix deposition in the epiphysis by in vivo calcein labeling experiments. These data suggest that abnormal morphology and function of MPS VII osteoclasts, combined with deficient matrix deposition, may contribute to the skeletal defects observed in this lysosomal storage disease.


Assuntos
Remodelação Óssea/fisiologia , Reabsorção Óssea/patologia , Mucopolissacaridose VII/patologia , Osteoclastos/patologia , Animais , Modelos Animais de Doenças , Fêmur/patologia , Fluoresceínas , Corantes Fluorescentes , Glucuronidase/biossíntese , Camundongos , Camundongos Mutantes , Osteoclastos/enzimologia
8.
Gene Ther ; 8(17): 1291-8, 2001 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-11571565

RESUMO

For many inborn errors of metabolism, early treatment is critical to prevent long-term developmental sequelae. We have previously shown that systemic treatment of neonatal mucopolysaccharidosis type VII (MPS VII) mice with recombinant adeno-associated virus (AAV) vectors results in relatively long-term expression of beta-glucuronidase (GUSB) in multiple tissues, and a reduction in lysosomal storage. Here, we demonstrate that therapeutic levels of enzyme persist for at least 1 year following a single intravenous injection of virus in neonatal MPS VII mice. The level and distribution of GUSB expression achieved is sufficient to prevent the development of many aspects of clinical disease over the life of the animal. Following treatment, bone lengths, weights and retinal function were maintained at nearly normal levels throughout the life of the animal. In addition, significant improvements in survival and auditory function were seen in AAV-treated MPS VII mice when compared with untreated mutant siblings. These data suggest that AAV-mediated gene transfer in the neonatal period can lead to prevention of many of the clinical symptoms associated with MPS VII in the murine model of this disease.


Assuntos
Terapia Genética/métodos , Glucuronidase/genética , Mucopolissacaridose VII/terapia , Transdução Genética , Animais , Animais Recém-Nascidos , Surdez/fisiopatologia , Eletrorretinografia , Potenciais Evocados Auditivos do Tronco Encefálico , Expressão Gênica , Camundongos , Camundongos Mutantes , Modelos Animais , Mucopolissacaridose VII/enzimologia , Mucopolissacaridose VII/fisiopatologia , Retina/fisiopatologia , Taxa de Sobrevida
9.
Gene Ther ; 8(17): 1343-6, 2001 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-11571571

RESUMO

Gene therapy using recombinant adeno-associated virus vectors (rAAV) is generally considered safe. During the course of a study designed to determine the long-term efficacy of rAAV-mediated gene therapy initiated in newborn mice with the lysosomal storage disease, mucopolysaccharidosis type VII (MPSVII), a significant incidence of hepatocellular carcinomas and angiosarcomas was discovered. A hepatocellular carcinoma was first detected in a 35-week-old mouse and by 72 weeks of age, three out of five rAAV-treated MPSVII mice had similar lesions. These types of tumors had not been seen previously in long-term studies of MPSVII mice using recombinant enzyme or bone marrow transplantation. In an attempt to ascertain whether mouse strain or GUSB expression confers susceptibility to tumor formation, we histopathologically examined untreated normal mice of the same strain, untreated MPSVII mice, and normal mice overexpressing human GUSB for the presence of tumors and increased hepatocyte replication. The results of these studies do not indicate that MPSVII mice or mice overexpressing human GUSB are susceptible to tumor formation; however, the number of animals examined is too small to draw definitive conclusions. Results from quantitative PCR performed on the tumor samples suggest that the tumors are probably not caused by an insertional mutagenesis event followed by the clonal expansion of a transformed cell. In a separate study, a relatively large group of mice injected with varying doses and types of rAAV vectors had no evidence of hepatic or vascular tumors. Although the mechanism of tumor formation is currently unknown, the tumorigenic potential of rAAV vectors must be rigorously determined in long-term in vivo studies.


Assuntos
Carcinoma Hepatocelular/etiologia , Dependovirus/genética , Vetores Genéticos/administração & dosagem , Hemangiossarcoma/etiologia , Neoplasias Hepáticas/etiologia , Doenças por Armazenamento dos Lisossomos/terapia , Animais , Animais Recém-Nascidos , Carcinoma Hepatocelular/patologia , Hemangiossarcoma/patologia , Neoplasias Hepáticas/patologia , Camundongos , Camundongos Mutantes , Modelos Animais
10.
J Biol Chem ; 276(46): 43160-5, 2001 Nov 16.
Artigo em Inglês | MEDLINE | ID: mdl-11562370

RESUMO

Enzyme replacement therapy (ERT) has been shown to be effective at reducing the accumulation of undegraded substrates in lysosomal storage diseases. Most ERT studies have been performed with recombinant proteins that are mixtures of phosphorylated and non-phosphorylated enzyme. Because different cell types use different receptors to take up phosphorylated or non-phosphorylated enzyme, it is difficult to determine which form of enzyme contributed to the clinical response. Here we compare the uptake, distribution, and efficacy of highly phosphorylated and non-phosphorylated beta-glucuronidase (GUSB) in the MPS VII mouse. Highly phosphorylated murine GUSB was efficiently taken up by a wide range of tissues. In contrast, non-phosphorylated murine GUSB was taken up primarily by tissues of the reticuloendothelial (RE) system. Although the tissue distribution was different, the half-lives of both enzymes in any particular tissue were similar. Both preparations of enzyme were capable of preventing the accumulation of lysosomal storage in cell types they targeted. An important difference in clinical efficacy emerged in that phosphorylated GUSB was more efficient than non-phosphorylated enzyme at preventing the hearing loss associated with this disease. These data suggest that both forms of enzyme contribute to the clinical responses of ERT in MPS VII mice but that enzyme preparations containing phosphorylated GUSB are more broadly effective than non-phosphorylated enzyme.


Assuntos
Glucuronidase/metabolismo , Glucuronidase/farmacocinética , Mucopolissacaridose VII/enzimologia , Animais , Animais Recém-Nascidos , Tronco Encefálico/metabolismo , Modelos Animais de Doenças , Potenciais Evocados Auditivos do Tronco Encefálico , Fibroblastos/metabolismo , Glucuronidase/química , Humanos , Insetos , Cinética , Lisossomos/metabolismo , Camundongos , Mutação , Fenótipo , Fosforilação , Ligação Proteica , Proteínas Recombinantes/metabolismo , Fatores de Tempo , Distribuição Tecidual
11.
Hum Gene Ther ; 12(5): 563-73, 2001 Mar 20.
Artigo em Inglês | MEDLINE | ID: mdl-11268288

RESUMO

Although AAV vectors show promise for hepatic gene therapy, the optimal transcriptional regulatory elements have not yet been identified. In this study, we show that an AAV vector with the CMV enhancer/chicken beta-actin promoter results in 9.5-fold higher expression after portal vein injection than an AAV vector with the EF1 alpha promoter, and 137-fold higher expression than an AAV vector with the CMV promoter/enhancer. Although induction of the acute-phase response with the administration of lipopolysaccharide (LPS) activated the CMV promoter/enhancer from the context of an adenoviral vector in a previous study, LPS resulted in only a modest induction of this promoter from an AAV vector in vivo. An AAV vector with the CMV-beta-actin promoter upstream of the coagulation protein human factor X (hFX) was injected intravenously into neonatal mice. This resulted in expression of hFX at 548 ng/ml (6.8% of normal) for up to 1.2 years, and 0.6 copies of AAV vector per diploid genome in the liver at the time of sacrifice. Neonatal intramuscular injection resulted in expression of hFX at 248 ng/ml (3.1% of normal), which derived from both liver and muscle. We conclude that neonatal gene therapy with an AAV vector with the CMV-beta-actin promoter might correct hemophilia due to hFX deficiency.


Assuntos
Actinas/genética , Citomegalovirus/genética , Dependovirus/genética , Fator X/genética , Hemofilia A/genética , Hemofilia A/terapia , Fígado/metabolismo , Fator 1 de Elongação de Peptídeos/genética , Regiões Promotoras Genéticas , Animais , Sítios de Ligação , Southern Blotting , Galinhas , DNA/metabolismo , DNA Complementar/metabolismo , Elementos Facilitadores Genéticos , Ensaio de Imunoadsorção Enzimática , Feminino , Terapia Genética/métodos , Vetores Genéticos , Humanos , Interleucina-6/metabolismo , Lipopolissacarídeos/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , RNA/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Fatores de Tempo , Transcrição Gênica
12.
Pediatr Res ; 49(3): 342-8, 2001 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-11228259

RESUMO

We describe the clinical and pathologic findings in a murine model of mucopolysaccharidosis VII (Sly disease) that arose spontaneously in the C3H/HeOuJ mouse strain. Affected gus(mps2J)/gus(mps2J) mice are deficient in beta-glucuronidase because of insertion of an intracisternal A particle element into intron 8 of the gus structural gene. This is the first model of a human lysosomal storage disease caused by an intracisternal A particle element insertion. Mice with the gus(mps2J)/gus(mps2J) genotype have < 1% of normal beta-glucuronidase activity and secondary elevations of other lysosomal enzymes. The phenotype includes shortened life-span, dysmorphic features, and skeletal dysplasia. Lysosomal storage of glycosaminoglycans is widespread and affects the brain, skeleton, eye, ear, heart valves, aorta, and the fixed tissue macrophage system. Thus the phenotypic and pathologic alterations in gus(mps2J)/gus(mps2J) mice are similar to those in patients with mucopolysaccharidosis VII. The finding of antibodies to beta-glucuronidase in some older gus(mps2J)/gus(mps2J) mice suggests the mice produce sufficient enzyme to elicit an immune response. The gus(mps2J)/gus(mps2J) model provides another well-defined genetic system for the study of the pathophysiology of mucopolysaccharidosis and for evaluation of experimental therapies for lysosomal storage diseases. The disease in gus(mps2J)/gus(mps2J) mice is less severe than that seen in the previously characterized B6.C-H2(bm1)/ByBir-gus(mps)/gus(mps) mouse model. Furthermore, unlike gus(mps)/gus(mps) mice, gus(mps2J)/gus(mps2J) mice are fertile and breed to produce litters, all of which are mucopolysaccharidosis VII pups. This feature makes them extremely useful for testing intrauterine therapies.


Assuntos
Genes de Partícula A Intracisternal/genética , Glucuronidase/genética , Mucopolissacaridose VII , Animais , Modelos Animais de Doenças , Humanos , Camundongos , Mucopolissacaridose VII/genética , Mucopolissacaridose VII/patologia , Mucopolissacaridose VII/fisiopatologia , Mutagênese Insercional
13.
Mol Ther ; 3(3): 351-8, 2001 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-11273777

RESUMO

Mucopolysaccharidosis type VII (MPS VII) is a lysosomal storage disease caused by the lack of beta-glucuronidase (GUSB) activity. GUSB deficiency leads to the progressive accumulation of undegraded glycosaminoglycans (GAGs) in cells of most tissues, including the brain, and is associated with mental retardation. Reduction of lysosomal storage in the central nervous system and prevention of cognitive dysfunction may require intracranial delivery of a therapeutic agent during the newborn period that provides a continuous source of GUSB. Therefore, we injected recombinant adeno-associated virus encoding human GUSB into both the anterior cortex and the hippocampus of newborn MPS VII mice. Total GUSB activity in the brain approached normal levels by 18 weeks. Although GUSB activity was concentrated near the injection sites, lysosomal distension was reduced in most areas of the brain. In addition to histopathologic evidence of GAG reduction, the previously undescribed accumulation of GM2 and GM3 gangliosides in the brain was also prevented. Furthermore, GUSB expression and reduced lysosomal distension correlated with improvements in cognitive function as measured in the Morris Water Maze test. These findings indicate that localized overexpression of GUSB has positive effects on the pathology and cognitive function and does not have overt toxicity.


Assuntos
Dependovirus/genética , Terapia Genética , Vetores Genéticos , Glucuronidase/genética , Mucopolissacaridose VII/terapia , Animais , Animais Recém-Nascidos , Córtex Cerebral , Expressão Gênica , Técnicas de Transferência de Genes , Glucuronidase/deficiência , Glucuronidase/uso terapêutico , Hipocampo , Injeções , Camundongos , Modelos Animais , Mucopolissacaridose VII/fisiopatologia , Proteínas Recombinantes/administração & dosagem , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo
14.
Pediatr Dev Pathol ; 4(5): 421-33, 2001.
Artigo em Inglês | MEDLINE | ID: mdl-11779044

RESUMO

The mucopolysaccharidoses are a group of lysosomal storage diseases caused by deficiency of an enzyme required for the normal degradation of glycosaminoglycans. Patients with mucopolysaccharidosis typically have widespread lysosomal storage, skeletal and central nervous system disease, and hepatosplenomegaly. Some patients with mucopolysaccharidosis may benefit from enzyme replacement therapy or bone marrow transplantation. Animal models of mucopolysaccharidosis have proven valuable for the evaluation of the effectiveness of potential treatments for patients with lysosomal storage disease. A murine model of MPS VII (Sly syndrome) has proven particularly useful because of its well-defined genetics and its well-characterized clinical, pathologic, and biochemical alterations, which resemble those seen in patients with mucopolysaccharidosis. Correction of these alterations forms the basis for evaluation of the effectiveness of novel treatments. A wide range of therapies have been tested using this model, including enzyme replacement therapy, bone marrow, stem cell, and neural progenitor cell transplantation, and a variety of viral-mediated gene therapies. The inferences drawn from these therapeutic studies using the murine MPS VII model are likely generalizable to other lysosomal storage diseases.


Assuntos
Modelos Animais de Doenças , Mucopolissacaridose VII , Animais , Transplante de Medula Óssea , Feminino , Terapia Genética , Glucuronidase/deficiência , Glucuronidase/genética , Glucuronidase/uso terapêutico , Masculino , Camundongos , Mucopolissacaridose VII/enzimologia , Mucopolissacaridose VII/genética , Mucopolissacaridose VII/patologia , Mucopolissacaridose VII/terapia , Fenótipo
15.
Mol Ther ; 2(6): 552-61, 2000 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-11124056

RESUMO

Gene therapy has been at least partially effective in several mouse disease models, but treatment of large mammals has been more difficult to achieve. One major limitation is that only low levels of expression of the corrective gene are often maintained in vivo. In a mouse model of the lysosomal storage disease mucopolysaccharidosis (MPS) type VII (Sly disease) with a null mutation in beta-glucuronidase, gene transfer experiments have shown that only 1-2% of normal beta-glucuronidase can correct the storage in some major organs. In contrast, MPS VII dogs, cats, and humans that have residual beta-glucuronidase activity levels in this range are affected. Thus, higher levels of transferred gene expression may be needed to achieve a therapeutic effect in large animals and humans. We tested this by examining liver pathology in MPS VII dogs after intraperitoneal transplantation of neo-organs containing retrovirus vector-corrected autologous fibroblasts that expressed low levels of beta-glucuronidase. The enzyme secreted from the neo-organs was taken up by the liver and significantly reduced the substrate content compared with untreated dogs. This suggests that small amounts of normal enzyme, when delivered to target tissues, may be therapeutically effective in human MPS VII patients.


Assuntos
Técnicas de Transferência de Genes , Glucuronidase/genética , Fígado/metabolismo , Mucopolissacaridose VII/terapia , Animais , Modelos Animais de Doenças , Cães , Glucuronidase/fisiologia , Fígado/enzimologia , Fígado/ultraestrutura , Camundongos , Camundongos Knockout , Microscopia Eletrônica , Mucopolissacaridose VII/enzimologia
16.
Hum Gene Ther ; 11(15): 2117-27, 2000 Oct 10.
Artigo em Inglês | MEDLINE | ID: mdl-11044913

RESUMO

Most lysosomal enzyme deficiencies are catastrophic illnesses with no generally available treatments. We have used the beta-glucuronidase-deficient mouse model of mucopolysaccharidosis type VII (MPS VII) to develop an alternative approach to therapy. A "universal" cell line engineered to secrete the missing enzyme is implanted in all recipients requiring the same enzyme replacement. The cells, although nonautologous, are rendered immunologically tolerant by encapsulation in microcapsules that provide protection from immune mediators. Using this strategy, we injected beta-glucuronidase-secreting fibroblasts enclosed in alginate microcapsules into mutant MPS VII mice. After 24 hr, beta-glucuronidase activity was detected in the plasma, reaching 66% of physiological levels by 2 weeks postimplantation. Significant beta-glucuronidase activity was detected in liver and spleen for the duration of the 8-week experiment. Concomitantly, the intralysosomal accumulation of undegraded glycosaminoglycans was dramatically reduced in liver and spleen tissue sections and urinary glycosaminoglycan content was reduced to normal levels. Elevated secondary lysosomal enzymes beta-hexosaminidase and alpha-galactosidase were also reduced. However, implanted mutant MPS VII mice developed antibodies against the murine beta-glucuronidase, demonstrating a potential obstacle in patients with a null mutation who react against the replaced enzyme as a foreign antigen. The antibody response was transiently circumvented with a single treatment of purified anti-CD4 antibody coadministered with the microcapsules. This resulted in increased levels and duration of beta-glucuronidase delivery. Similarly, treated heterozygous mice maintained elevated levels of beta-glucuronidase and did not develop antibodies. This novel cell-based therapy demonstrates a potentially cost-effective and nonviral treatment applicable to all lysosomal storage diseases.


Assuntos
Mucopolissacaridose VII/genética , Mucopolissacaridose VII/terapia , Alelos , Animais , Antígenos CD4/imunologia , Linhagem Celular , DNA Complementar/metabolismo , Composição de Medicamentos , Fibroblastos/metabolismo , Terapia Genética/métodos , Glucuronidase/biossíntese , Glucuronidase/sangue , Glucuronidase/deficiência , Glicosaminoglicanos/urina , Heterozigoto , Humanos , Rim/metabolismo , Fígado/metabolismo , Camundongos , Camundongos Mutantes , Baço/metabolismo , Fatores de Tempo , alfa-Galactosidase/metabolismo , beta-N-Acetil-Hexosaminidases/metabolismo
17.
Exp Eye Res ; 71(5): 469-81, 2000 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-11040082

RESUMO

Mucopolysaccharidoses are heritable lysosomal storage diseases caused by deficiencies in acid hydrolases involved in the sequential degradation of complex glycosaminoglycans (GAGs). In many mucopolysaccharidoses, GAGs accumulate in the retinal pigment epithelial (RPE) cells of the eye resulting in pronounced lysosomal distension. It is not clear how the progressive accumulation of GAGs affects retinal function. Bone marrow transplantation (BMT) is a relatively effective therapy for many lysosomal storage diseases and can result in a dramatic reduction in lysosomal distention in the RPE. Although effective at reducing lysosomal storage, it is not clear how effective syngeneic BMT is at treating retinal dysfunction. Here we show that there is a progressive decrease in the amplitudes of both the dark-adapted (rod-cone) and light-adapted (cone-dominated) flash electroretinograms (ERG) between 8 and 20 weeks of age in a murine model of mucopolysaccharidosis type VII (MPS VII). By 20 weeks, the average dark-adapted b-wave amplitude was 118 microV in MPS VII mice as compared to 469 microV in normal mice of the same strain. MPS VII mice receiving syngeneic BMT at 4 weeks of age have reduced lysosomal storage in retinal pigment epithelial cells and normal ERG amplitudes at 20 weeks of age. Retinal function is impaired in untreated 8 week old MPS VII mice. Following BMT at 8 weeks, rod-cone- and cone-dominated responses recovered to within the normal range by 20 weeks of age. These data demonstrate the temporal pattern of retinal dysfunction in the MPS VII mouse and indicate that BMT can reduce the lysosomal storage and improve retinal function.


Assuntos
Transplante de Medula Óssea , Mucopolissacaridose VII/fisiopatologia , Doenças Retinianas/fisiopatologia , Animais , Progressão da Doença , Eletrorretinografia/métodos , Glucuronidase/análise , Camundongos , Modelos Biológicos , Mucopolissacaridose VII/terapia , Neutrófilos/química , Epitélio Pigmentado Ocular/fisiopatologia , Valores de Referência , Doenças Retinianas/terapia , Irradiação Corporal Total
18.
Mol Ther ; 2(3): 233-44, 2000 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-10985954

RESUMO

Mucopolysaccharidosis VII (MPS VII) is caused by beta-glucuronidase (beta-gluc) deficiency and results in lysosomal storage due to the inability to degrade glycosaminoglycans. Transfer of a beta-gluc gene into the liver reduces hepatic pathology as well as storage in other organs via uptake of secreted protein. A Moloney murine leukemia-based retroviral vector expressing the human beta-gluc cDNA was injected intravascularly into MPS VII mice during hepatocyte replication, which was induced with im injection of an adenoviral vector that transiently expressed hepatocyte growth factor (Ad.CMV. HGF). This procedure resulted in transduction of approximately 1% of hepatocytes, 1% of normal liver enzyme activity, and a reduction in lysosomal storage in the liver at 3.5 months. Surprisingly, controls that received retroviral vector without HGF had transduction of nonparenchymal cells in the liver, significant levels of enzyme and RNA in the liver at 2 but not 3.5 months, and reduced lysosomal storage at 3.5 months. Transduction was also achieved in the replicating cells of the spleen, where lysosomal storage was reduced. An approach using a retroviral vector without a growth factor might temporarily reduce lysosomal storage in the liver and spleen in humans. Addition of HGF might be used to augment and prolong gene transfer.


Assuntos
Vetores Genéticos , Glucuronidase/genética , Fígado/enzimologia , Lisossomos/metabolismo , Mucopolissacaridose VII/enzimologia , Retroviridae/genética , Baço/enzimologia , Animais , Sequência de Bases , Divisão Celular/efeitos dos fármacos , Primers do DNA , Fator de Crescimento de Hepatócito/farmacologia , Humanos , Fígado/efeitos dos fármacos , Camundongos , Mucopolissacaridose VII/terapia , Baço/citologia
19.
Pediatr Res ; 47(6): 757-62, 2000 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-10832733

RESUMO

Lysosomal storage diseases are a class of inborn errors of metabolism that lead to widespread disease in multiple tissues. The murine model of mucopolysaccharidosis type VII (MPS VII) closely parallels the human syndrome and has been extensively used to investigate the natural history and therapeutic strategies for lysosomal storage diseases in general. Here we demonstrate a previously undescribed immune defect in the MPS VII mouse. Although the normal populations of cells are present in lymph nodes of these mice, MPS VII mice show a blunted T cell proliferative response and decreased antibody production after immunization with antigens. One mechanism of this defect is ineffective processing of protein antigens, as responses to peptide antigens are normal. This phenotype is presumably caused by the lysosomal disorder, as the defect can be corrected in vivo by direct enzyme replacement therapy. These findings have implications for the use of this animal model, and may have clinical significance for other, more-common lysosomal storage diseases.


Assuntos
Mucopolissacaridose VII/imunologia , Animais , Divisão Celular , Modelos Animais de Doenças , Ensaio de Imunoadsorção Enzimática , Glucuronidase/uso terapêutico , Humanos , Subpopulações de Linfócitos , Camundongos , Proteínas Recombinantes/uso terapêutico
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...