Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 5 de 5
Filtrar
1.
Mol Ther ; 24(2): 276-286, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26639405

RESUMEN

Cerebrospinal fluid administration of recombinant adeno-associated viral (rAAV) vectors has been demonstrated to be effective in delivering therapeutic genes to the central nervous system (CNS) in different disease animal models. However, a quantitative and qualitative analysis of transduction patterns of the most promising rAAV serotypes for brain targeting in large animal models is missing. Here, we characterize distribution, transduction efficiency, and cellular targeting of rAAV serotypes 1, 2, 5, 7, 9, rh.10, rh.39, and rh.43 delivered into the cisterna magna of wild-type pigs. rAAV9 showed the highest transduction efficiency and the widest distribution capability among the vectors tested. Moreover, rAAV9 robustly transduced both glia and neurons, including the motor neurons of the spinal cord. Relevant cell transduction specificity of the glia was observed after rAAV1 and rAAV7 delivery. rAAV7 also displayed a specific tropism to Purkinje cells. Evaluation of biochemical and hematological markers suggested that all rAAV serotypes tested were well tolerated. This study provides a comprehensive CNS transduction map in a useful preclinical large animal model enabling the selection of potentially clinically transferable rAAV serotypes based on disease specificity. Therefore, our data are instrumental for the clinical evaluation of these rAAV vectors in human neurodegenerative diseases.


Asunto(s)
Sistema Nervioso Central/metabolismo , Dependovirus/genética , Vectores Genéticos/administración & dosificación , Vectores Genéticos/líquido cefalorraquídeo , Proteínas Fluorescentes Verdes/metabolismo , Animales , Dependovirus/inmunología , Técnicas de Transferencia de Gen , Proteínas Fluorescentes Verdes/genética , Humanos , Especificidad de Órganos , Serogrupo , Porcinos , Transducción Genética , Transgenes
2.
Mol Metab ; 86: 101965, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-38871178

RESUMEN

OBJECTIVE: Interleukin (IL)-22 is a potential therapeutic protein for the treatment of metabolic diseases such as obesity, type 2 diabetes, and metabolic dysfunction-associated steatotic liver disease due to its involvement in multiple cellular pathways and observed hepatoprotective effects. The short serum half-life of IL-22 has previously limited its use in clinical applications; however, the development of mRNA-lipid nanoparticle (LNP) technology offers a novel therapeutic approach that uses a host-generated IL-22 fusion protein. In the present study, the effects of administration of an mRNA-LNP encoding IL-22 on metabolic disease parameters was investigated in various mouse models. METHODS: C57BL/6NCrl mice were used to confirm mouse serum albumin (MSA)-IL-22 protein expression prior to assessments in C57BL/6NTac and CETP/ApoB transgenic mouse models of metabolic disease. Mice were fed either regular chow or a modified amylin liver nonalcoholic steatohepatitis-inducing diet prior to receiving either LNP-encapsulated MSA-IL-22 or MSA mRNA via intravenous or intramuscular injection. Metabolic markers were monitored for the duration of the experiments, and postmortem histology assessment and analysis of metabolic gene expression pathways were performed. RESULTS: MSA-IL-22 was detectable for ≥8 days following administration. Improvements in body weight, lipid metabolism, glucose metabolism, and lipogenic and fibrotic marker gene expression in the liver were observed in the MSA-IL-22-treated mice, and these effects were shown to be durable. CONCLUSIONS: These results support the application of mRNA-encoded IL-22 as a promising treatment strategy for metabolic syndrome and associated comorbidities in human populations.


Asunto(s)
Interleucina-22 , Interleucinas , Enfermedades Metabólicas , Ratones Endogámicos C57BL , ARN Mensajero , Animales , Ratones , Interleucinas/metabolismo , Interleucinas/genética , ARN Mensajero/metabolismo , ARN Mensajero/genética , Masculino , Enfermedades Metabólicas/metabolismo , Enfermedades Metabólicas/genética , Nanopartículas , Semivida , Ratones Transgénicos , Hígado/metabolismo , Modelos Animales de Enfermedad , Enfermedad del Hígado Graso no Alcohólico/metabolismo , Enfermedad del Hígado Graso no Alcohólico/genética , Lípidos/sangre , Liposomas
3.
Nat Commun ; 12(1): 3090, 2021 05 25.
Artículo en Inglés | MEDLINE | ID: mdl-34035281

RESUMEN

Glycogen Storage Disease 1a (GSD1a) is a rare, inherited metabolic disorder caused by deficiency of glucose 6-phosphatase (G6Pase-α). G6Pase-α is critical for maintaining interprandial euglycemia. GSD1a patients exhibit life-threatening hypoglycemia and long-term liver complications including hepatocellular adenomas (HCAs) and carcinomas (HCCs). There is no treatment for GSD1a and the current standard-of-care for managing hypoglycemia (Glycosade®/modified cornstarch) fails to prevent HCA/HCC risk. Therapeutic modalities such as enzyme replacement therapy and gene therapy are not ideal options for patients due to challenges in drug-delivery, efficacy, and safety. To develop a new treatment for GSD1a capable of addressing both the life-threatening hypoglycemia and HCA/HCC risk, we encapsulated engineered mRNAs encoding human G6Pase-α in lipid nanoparticles. We demonstrate the efficacy and safety of our approach in a preclinical murine model that phenotypically resembles the human condition, thus presenting a potential therapy that could have a significant therapeutic impact on the treatment of GSD1a.


Asunto(s)
Modelos Animales de Enfermedad , Terapia Genética/métodos , Glucosa-6-Fosfatasa/genética , Enfermedad del Almacenamiento de Glucógeno/terapia , ARN Mensajero/genética , Animales , Línea Celular Tumoral , Citocinas/sangre , Citocinas/metabolismo , Glucosa-6-Fosfatasa/metabolismo , Glucógeno/metabolismo , Enfermedad del Almacenamiento de Glucógeno/genética , Enfermedad del Almacenamiento de Glucógeno/patología , Células HeLa , Humanos , Hígado/metabolismo , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Nanopartículas/administración & dosificación , Nanopartículas/química , ARN Mensajero/administración & dosificación , ARN Mensajero/química , Resultado del Tratamiento , Triglicéridos/metabolismo
4.
PLoS One ; 13(4): e0195186, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29672630

RESUMEN

Recombinant human arylsulfatase A (rhASA) is in clinical development for the treatment of patients with metachromatic leukodystrophy (MLD). Manufacturing process changes were introduced to improve robustness and efficiency, resulting in higher levels of mannose-6-phosphate and sialic acid in post-change (process B) compared with pre-change (process A) rhASA. A nonclinical comparability program was conducted to compare process A and process B rhASA. All doses were administered intrathecally. Pharmacodynamic comparability was evaluated in immunotolerant MLD mice, using immunohistochemical staining of lysosomal-associated membrane protein-1 (LAMP-1). Pharmacokinetic comparability was assessed in juvenile cynomolgus monkeys dosed once with 6.0 mg (equivalent to 100 mg/kg of brain weight) process A or process B rhASA. Biodistribution was compared by quantitative whole-body autoradiography in rats. Potential toxicity of process B rhASA was evaluated by repeated rhASA administration at doses of 18.6 mg in juvenile cynomolgus monkeys. The specific activities for process A and process B rhASA were 89 U/mg and 106 U/mg, respectively, which were both well within the target range for the assay. Pharmacodynamic assessments showed no statistically significant differences in LAMP-1 immunohistochemical staining in the spinal cord and in most of the brain areas assessed between process A and B rhASA-dosed mice. LAMP-1 staining was reduced with both process A and B rhASA compared with vehicle, supporting its activity. Concentration-time curves in cerebrospinal fluid and serum of cynomolgus monkeys were similar with process A and B rhASA. Process A and B rhASA were similar in terms of their pharmacokinetic parameters and biodistribution data. No process B rhASA-related toxicity was detected. In conclusion, manufacturing process changes did not affect the pharmacodynamic, pharmacokinetic or safety profiles of process B rhASA relative to process A rhASA.


Asunto(s)
Cerebrósido Sulfatasa/metabolismo , Proteínas Recombinantes , Animales , Cerebrósido Sulfatasa/biosíntesis , Cerebrósido Sulfatasa/aislamiento & purificación , Cerebrósido Sulfatasa/farmacología , Evaluación Preclínica de Medicamentos , Activación Enzimática , Femenino , Humanos , Inmunohistoquímica , Proteína 1 de la Membrana Asociada a los Lisosomas/metabolismo , Masculino , Ratones , Ratas , Distribución Tisular
5.
Antioxid Redox Signal ; 23(6): 550-64, 2015 Aug 20.
Artículo en Inglés | MEDLINE | ID: mdl-26094487

RESUMEN

AIMS: Loss-of-function mutations in GBA1, which cause the autosomal recessive lysosomal storage disease, Gaucher disease (GD), are also a key genetic risk factor for the α-synucleinopathies, including Parkinson's disease (PD) and dementia with Lewy bodies. GBA1 encodes for the lysosomal hydrolase glucocerebrosidase and reductions in this enzyme result in the accumulation of the glycolipid substrates glucosylceramide and glucosylsphingosine. Deficits in autophagy and lysosomal degradation pathways likely contribute to the pathological accumulation of α-synuclein in PD. In this report we used conduritol-ß-epoxide (CBE), a potent selective irreversible competitive inhibitor of glucocerebrosidase, to model reduced glucocerebrosidase activity in vivo, and tested whether sustained glucocerebrosidase inhibition in mice could induce neuropathological abnormalities including α-synucleinopathy, and neurodegeneration. RESULTS: Our data demonstrate that daily systemic CBE treatment over 28 days caused accumulation of insoluble α-synuclein aggregates in the substantia nigra, and altered levels of proteins involved in the autophagy lysosomal system. These neuropathological changes were paralleled by widespread neuroinflammation, upregulation of complement C1q, abnormalities in synaptic, axonal transport and cytoskeletal proteins, and neurodegeneration. INNOVATION: A reduction in brain GCase activity has been linked to sporadic PD and normal aging, and may contribute to the susceptibility of vulnerable neurons to degeneration. This report demonstrates that systemic reduction of GCase activity using chemical inhibition, leads to neuropathological changes in the brain reminiscent of α-synucleinopathy. CONCLUSIONS: These data reveal a link between reduced glucocerebrosidase and the development of α-synucleinopathy and pathophysiological abnormalities in mice, and support the development of GCase therapeutics to reduce α-synucleinopathy in PD and related disorders.


Asunto(s)
Complemento C1q/metabolismo , Glucosilceramidasa/antagonistas & inhibidores , Inositol/análogos & derivados , Microglía/fisiología , Agregación Patológica de Proteínas/enzimología , alfa-Sinucleína/metabolismo , Animales , Autofagia , Transporte Axonal , Corteza Cerebral/metabolismo , Corteza Cerebral/patología , Activación de Complemento , Glucosilceramidasa/metabolismo , Inositol/farmacología , Masculino , Ratones , Enfermedad de Parkinson Secundaria/inducido químicamente , Enfermedad de Parkinson Secundaria/enzimología , Agregación Patológica de Proteínas/inducido químicamente , Proteínas/metabolismo , Transmisión Sináptica
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA