Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 32
Filtrar
Más filtros













Intervalo de año de publicación
1.
Cell Biochem Funct ; 41(8): 1093-1105, 2023 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-38018878

RESUMEN

One of the most important inherited metabolic disorders is GM1 gangliosidosis, which is a progressive neurological disorder. The main cause of this disease is a genetic defect in the enzyme ß-galactosidase due to a mutation in the glb1 gene. Lack of this enzyme in cells (especially neurons) leads to the accumulation of ganglioside substrate in nerve tissues, followed by three clinical forms of GM1 disease (neonatal, juvenile, and adult variants). Genetically, many mutations occur in the exons of the glb1 gene, such as exons 2, 6, 15, and 16, so the most common ones reported in scientific studies include missense/nonsense mutations. Therefore, many studies have examined the genotype-phenotype relationships of this disease and subsequently using gene therapy techniques have been able to reduce the complications of the disease and alleviate the signs and symptoms of the disease. In this regard, the present article reviews the general features of GM1 gangliosidosis and its mutations, as well as gene therapy studies and animal and human models of the disease.


Asunto(s)
Gangliosidosis GM1 , Adulto , Animales , Recién Nacido , Humanos , Gangliosidosis GM1/genética , Gangliosidosis GM1/terapia , Mutación , Mutación Missense , Neuronas , Terapia Genética
2.
EBioMedicine ; 92: 104627, 2023 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-37267847

RESUMEN

BACKGROUND: GM1 gangliosidosis is a rare, fatal, neurodegenerative disease caused by mutations in the GLB1 gene and deficiency in ß-galactosidase. Delay of symptom onset and increase in lifespan in a GM1 gangliosidosis cat model after adeno-associated viral (AAV) gene therapy treatment provide the basis for AAV gene therapy trials. The availability of validated biomarkers would greatly improve assessment of therapeutic efficacy. METHODS: The liquid chromatography-tandem mass spectrometry (LC-MS/MS) was used to screen oligosaccharides as potential biomarkers for GM1 gangliosidosis. The structures of pentasaccharide biomarkers were determined with mass spectrometry, as well as chemical and enzymatic degradations. Comparison of LC-MS/MS data of endogenous and synthetic compounds confirmed the identification. The study samples were analyzed with fully validated LC-MS/MS methods. FINDINGS: We identified two pentasaccharide biomarkers, H3N2a and H3N2b, that were elevated more than 18-fold in patient plasma, cerebrospinal fluid (CSF), and urine. Only H3N2b was detectable in the cat model, and it was negatively correlated with ß-galactosidase activity. Following intravenous (IV) AAV9 gene therapy treatment, reduction of H3N2b was observed in central nervous system, urine, plasma, and CSF samples from the cat model and in urine, plasma, and CSF samples from a patient. Reduction of H3N2b accurately reflected normalization of neuropathology in the cat model and improvement of clinical outcomes in the patient. INTERPRETATIONS: These results demonstrate that H3N2b is a useful pharmacodynamic biomarker to evaluate the efficacy of gene therapy for GM1 gangliosidosis. H3N2b will facilitate the translation of gene therapy from animal models to patients. FUNDING: This work was supported by grants U01NS114156, R01HD060576, ZIAHG200409, and P30 DK020579 from the National Institutes of Health (NIH) and a grant from National Tay-Sachs and Allied Diseases Association Inc.


Asunto(s)
Gangliosidosis GM1 , Enfermedades Neurodegenerativas , Animales , Gangliosidosis GM1/genética , Gangliosidosis GM1/terapia , Gangliosidosis GM1/patología , Enfermedades Neurodegenerativas/terapia , Cromatografía Liquida , Espectrometría de Masas en Tándem , beta-Galactosidasa/genética , beta-Galactosidasa/química , beta-Galactosidasa/uso terapéutico , Biomarcadores/líquido cefalorraquídeo , Terapia Genética
3.
CRISPR J ; 6(1): 17-31, 2023 02.
Artículo en Inglés | MEDLINE | ID: mdl-36629845

RESUMEN

Ganglioside-monosialic acid (GM1) gangliosidosis, a rare autosomal recessive disorder, is frequently caused by deleterious single nucleotide variants (SNVs) in GLB1 gene. These variants result in reduced ß-galactosidase (ß-gal) activity, leading to neurodegeneration associated with premature death. Currently, no effective therapy for GM1 gangliosidosis is available. Three ongoing clinical trials aim to deliver a functional copy of the GLB1 gene to stop disease progression. In this study, we show that 41% of GLB1 pathogenic SNVs can be replaced by adenine base editors (ABEs). Our results demonstrate that ABE efficiently corrects the pathogenic allele in patient-derived fibroblasts, restoring therapeutic levels of ß-gal activity. Off-target DNA analysis did not detect off-target editing activity in treated patient's cells, except a bystander edit without consequences on ß-gal activity based on 3D structure bioinformatics predictions. Altogether, our results suggest that gene editing might be an alternative strategy to cure GM1 gangliosidosis.


Asunto(s)
Gangliosidosis GM1 , Humanos , Gangliosidosis GM1/terapia , Gangliosidosis GM1/tratamiento farmacológico , beta-Galactosidasa/genética , beta-Galactosidasa/química , beta-Galactosidasa/metabolismo , Edición Génica , Sistemas CRISPR-Cas/genética , Alelos
4.
Curr Gene Ther ; 22(4): 352-365, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35249485

RESUMEN

BACKGROUND: GM1 gangliosidosis (GM1) is an autosomal recessive disorder characterized by the deficiency of beta-galactosidase (ß-gal), a ubiquitous lysosomal enzyme that catalyzes the hydrolysis of GM1 ganglioside. OBJECTIVE: The study aims to explore the application of the AAV9-coGLB1 for effective treatment in a GM1 gangliosidosis mutant mouse model. METHODS: We designed a novel adeno-associated virus 9 (AAV9) vector expressing ß-gal (AAV9- coGLB1) to treat GM1 gangliosidosis. The vector, injected via the caudal vein at 4 weeks of age, drove the widespread and sustained expression of ß-gal for up to 32 weeks in the Glb1G455R/G455R mutant mice (GM1 mice). RESULTS: The increased levels of ß-gal reduced the pathological damage occurring in GM1 mice. Histological analyses showed that myelin deficits and neuron-specific pathology were reduced in the cerebral cortex region of AAV9-coGLB1-treated mice. Immunohistochemical staining showed that the accumulation of GM1 ganglioside was also reduced after gene therapy. The reduction of the storage in these regions was accompanied by a decrease in activated microglia. In addition, AAV9 treatment reversed the blockade of autophagic flux in GM1 mice. CONCLUSION: These results show that AAV9-coGLB1 reduces the pathological signs of GM1 gangliosidosis in a mouse model.


Asunto(s)
Gangliosidosis GM1 , Animales , Sistema Nervioso Central , Dependovirus/genética , Modelos Animales de Enfermedad , Gangliósido G(M1) , Gangliosidosis GM1/genética , Gangliosidosis GM1/metabolismo , Gangliosidosis GM1/terapia , Inflamación/genética , Inflamación/terapia , Lisosomas/genética , Lisosomas/patología , Ratones
5.
Brain ; 145(2): 655-669, 2022 04 18.
Artículo en Inglés | MEDLINE | ID: mdl-34410345

RESUMEN

GM1 gangliosidosis is a fatal neurodegenerative disease caused by a deficiency of lysosomal ß-galactosidase. In its most severe form, GM1 gangliosidosis causes death by 4 years of age, and no effective treatments exist. Previous work has shown that injection of the brain parenchyma with an adeno-associated viral (AAV) vector provides pronounced therapeutic benefit in a feline GM1 model. To develop a less invasive treatment for the brain and increase systemic biodistribution, intravenous injection of AAV9 was evaluated. AAV9 expressing feline ß-galactosidase was intravenously administered at 1.5×1013 vector genomes/kg body weight to six GM1 cats at ∼1 month of age. The animals were divided into two cohorts: (i) a long-term group, which was followed to humane end point; and (ii) a short-term group, which was analysed 16 weeks post-treatment. Clinical assessments included neurological exams, CSF and urine biomarkers, and 7 T MRI and magentic resonance spectroscopy (MRS). Post-mortem analysis included ß-galactosidase and virus distribution, histological analysis and ganglioside content. Untreated GM1 animals survived 8.0 ± 0.6 months while intravenous treatment increased survival to an average of 3.5 years (n = 2) with substantial improvements in quality of life and neurological function. Neurological abnormalities, which in untreated animals progress to the inability to stand and debilitating neurological disease by 8 months of age, were mild in all treated animals. CSF biomarkers were normalized, indicating decreased CNS cell damage in the treated animals. Urinary glycosaminoglycans decreased to normal levels in the long-term cohort. MRI and MRS showed partial preservation of the brain in treated animals, which was supported by post-mortem histological evaluation. ß-Galactosidase activity was increased throughout the CNS, reaching carrier levels in much of the cerebrum and normal levels in the cerebellum, spinal cord and CSF. Ganglioside accumulation was significantly reduced by treatment. Peripheral tissues such as heart, skeletal muscle, and sciatic nerve also had normal ß-galactosidase activity in treated GM1 cats. GM1 histopathology was largely corrected with treatment. There was no evidence of tumorigenesis or toxicity. Restoration of ß-galactosidase activity in the CNS and peripheral organs by intravenous gene therapy led to profound increases in lifespan and quality of life in GM1 cats. These data support the promise of intravenous gene therapy as a safe, effective treatment for GM1 gangliosidosis.


Asunto(s)
Gangliosidosis GM1 , Enfermedades Neurodegenerativas , Animales , Biomarcadores , Gatos , Dependovirus/genética , Gangliósido G(M1)/uso terapéutico , Gangliósidos , Gangliosidosis GM1/genética , Gangliosidosis GM1/patología , Gangliosidosis GM1/terapia , Terapia Genética/métodos , Humanos , Calidad de Vida , Distribución Tisular , beta-Galactosidasa/genética , beta-Galactosidasa/metabolismo
6.
Int J Mol Sci ; 21(23)2020 Nov 30.
Artículo en Inglés | MEDLINE | ID: mdl-33266180

RESUMEN

Morquio B disease (MBD) is an autosomal recessive GLB1-gene-related lysosomal storage disease, presenting with a peculiar type of dysostosis multiplex which is also observed in GALNS-related Morquio A disease. MBD may present as pure skeletal phenotype (pure MBD) or in combination with the neuronopathic manifestations seen in type 2 (juvenile) or type 3 (late onset) GM1 gangliosidosis (MBD plus). The main skeletal features are progressive growth impairment, kyphoscoliosis, coxa/genua valga, joint laxity, platyspondyly and odontoid hypoplasia. The main neuronopathic features are dystonia, ataxia, and intellectual/developmental/speech delay. Spinal cord compression occurs as a complication of spinal dysostosis. Chronic pain is reported, along with mobility issues and challenges with daily living and self-care activities, as the most common health concern. The most commonly reported orthopedic surgeries are hip and knee replacements. Keratan sulphate-derived oligosaccharides are characteristic biomarkers. Residual ß-galactosidase activities measured against synthetic substrates do not correlate with the phenotype. W273 L and T500A are the most frequently observed GLB1 variants in MBD, W273L being invariably associated with pure MBD. Cytokines play a role in joint destruction and pain, providing a promising treatment target. In the future, patients may benefit from small molecule therapies, and gene and enzyme replacement therapies, which are currently being developed for GM1 gangliosidosis.


Asunto(s)
Mucopolisacaridosis IV/diagnóstico , Mucopolisacaridosis IV/terapia , Biomarcadores , Citocinas/metabolismo , Diagnóstico Diferencial , Susceptibilidad a Enfermedades , Gangliosidosis GM1/diagnóstico , Gangliosidosis GM1/genética , Gangliosidosis GM1/terapia , Humanos , Mucopolisacaridosis IV/etiología , Mutación , Fenotipo , beta-Galactosidasa/genética
7.
Hum Gene Ther ; 31(21-22): 1169-1177, 2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-33045869

RESUMEN

GM1 gangliosidosis is a rare neurodegenerative lysosomal storage disease caused by loss-of-function mutations in the gene encoding beta-galactosidase (ß-gal). There are no approved treatments for GM1 gangliosidosis. Previous studies in animal models have demonstrated that adeno-associated viral (AAV) vector-mediated gene transfer to the brain can restore ß-gal expression and prevent the onset of neurological signs. We developed an optimized AAV vector expressing human ß-gal and evaluated the efficacy of a single intracerebroventricular injection of this vector into the cerebrospinal fluid (CSF) of a murine disease model. The AAV vector administration into the CSF increased ß-gal activity in the brain, reduced neuronal lysosomal storage lesions, prevented the onset of neurological signs and gait abnormalities, and increased survival. These findings demonstrate the potential therapeutic activity of this vector and support its subsequent development for the treatment of GM1 gangliosidosis.


Asunto(s)
Encéfalo/metabolismo , Líquido Cefalorraquídeo/metabolismo , Dependovirus/genética , Gangliosidosis GM1/terapia , Terapia Genética/métodos , Vectores Genéticos/administración & dosificación , beta-Galactosidasa/fisiología , Animales , Encéfalo/patología , Líquido Cefalorraquídeo/citología , Modelos Animales de Enfermedad , Gangliosidosis GM1/genética , Gangliosidosis GM1/metabolismo , Gangliosidosis GM1/patología , Vectores Genéticos/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , beta-Galactosidasa/administración & dosificación , beta-Galactosidasa/genética
8.
Neuropathology ; 40(4): 379-388, 2020 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-32219895

RESUMEN

GM1 gangliosidosis is a storage disorder with autosomal recessive inheritance caused by deficiency of ß-galactosidase (GLB1), which is a lysosomal hydrolase, due to mutations in GLB1. We describe here an autopsy case of GM1 gangliosidosis in a female patient who survived for 38 years with a long period of artificial respiratory support (ARS). She was born after a normal pregnancy and delivery. Although development was normal until one year old, she was unable to walk at two years old and started having seizures by nine years old. At 21 years old, she became unable to communicate and was bed-ridden. At 36 years old, she suffered from pneumonia and required ARS. She died of pneumonia at 40 years old. Neuropathological examination revealed severe atrophy, predominantly found in the frontal lobes. Microscopically, severe gliosis and neuronal loss were observed in the cerebral cortex, putamen, cerebellum, the latter including Purkinje cell and granule cell layers. The hippocampus was relatively preserved. Severe neuronal swelling was observed in the limbic regions and stored a material in these neurons negative for periodic acid-Schiff (PAS). A PAS-positive granular storage material in neurons and macrophages was mainly observed in the brainstem and limbic regions. Exome analysis showed a known c.152T>C (p.I51T) variant that has been described in type III patients and a novel c.1348-2A>G variant in GLB1. Detailed analysis of reverse transcription-polymerase chain reaction products of GLB1 mRNA revealed that these variants were present in a compound heterozygous state. In our case, clinical features and neuropathological findings were most consistent with type II, although the entire course was longer than any previously reported cases. This may be explained by the residual enzyme activity in this patient whose severity lay between types II and III. Our finding of relative preservation of the limbic regions suggests that neuronal loss in GM1 gangliosidosis has regional selectivity.


Asunto(s)
Encéfalo/patología , Gangliosidosis GM1/patología , Adulto , Autopsia , Femenino , Gangliosidosis GM1/genética , Gangliosidosis GM1/terapia , Humanos , Respiración Artificial , Adulto Joven , beta-Galactosidasa/genética
9.
J Biol Chem ; 295(39): 13532-13555, 2020 09 25.
Artículo en Inglés | MEDLINE | ID: mdl-31481471

RESUMEN

Autosomal recessive mutations in the galactosidase ß1 (GLB1) gene cause lysosomal ß-gal deficiency, resulting in accumulation of galactose-containing substrates and onset of the progressive and fatal neurodegenerative lysosomal storage disease, GM1 gangliosidosis. Here, an enzyme replacement therapy (ERT) approach in fibroblasts from GM1 gangliosidosis patients with recombinant human ß-gal (rhß-gal) produced in Chinese hamster ovary cells enabled direct and precise rhß-gal delivery to acidified lysosomes. A single, low dose (3 nm) of rhß-gal was sufficient for normalizing ß-gal activity and mediating substrate clearance for several weeks. We found that rhß-gal uptake by the fibroblasts is dose-dependent and saturable and can be competitively inhibited by mannose 6-phosphate, suggesting cation-independent, mannose 6-phosphate receptor-mediated endocytosis from the cell surface. A single intracerebroventricularly (ICV) administered dose of rhß-gal (100 µg) resulted in broad bilateral biodistribution of rhß-gal to critical regions of pathology in a mouse model of GM1 gangliosidosis. Weekly ICV dosing of rhß-gal for 8 weeks substantially reduced brain levels of ganglioside and oligosaccharide substrates and reversed well-established secondary neuropathology. Of note, unlike with the ERT approach, chronic lentivirus-mediated GLB1 overexpression in the GM1 gangliosidosis patient fibroblasts caused accumulation of a prelysosomal pool of ß-gal, resulting in activation of the unfolded protein response and endoplasmic reticulum stress. This outcome was unsurprising in light of our in vitro biophysical findings for rhß-gal, which include pH-dependent and concentration-dependent stability and dynamic self-association. Collectively, our results highlight that ICV-ERT is an effective therapeutic intervention for managing GM1 gangliosidosis potentially more safely than with gene therapy approaches.


Asunto(s)
Terapia de Reemplazo Enzimático , Gangliosidosis GM1/terapia , beta-Galactosidasa/metabolismo , Animales , Gangliosidosis GM1/metabolismo , Gangliosidosis GM1/patología , Ratones
10.
Mol Genet Metab ; 121(2): 170-179, 2017 06.
Artículo en Inglés | MEDLINE | ID: mdl-28476546

RESUMEN

BACKGROUND: Infantile gangliosidoses include GM1 gangliosidosis and GM2 gangliosidosis (Tay-Sachs disease, Sandhoff disease). To date, natural history studies in infantile GM2 (iGM2) have been retrospective and conducted through surveys. Compared to iGM2, there is even less natural history information available on infantile GM1 disease (iGM1). There are no approved treatments for infantile gangliosidoses. Substrate reduction therapy using miglustat has been tried, but is limited by gastrointestinal side effects. Development of effective treatments will require identification of meaningful outcomes in the setting of rapidly progressive and fatal diseases. OBJECTIVES: This study aimed to establish a timeline of clinical changes occurring in infantile gangliosidoses, prospectively, to: 1) characterize the natural history of these diseases; 2) improve planning of clinical care; and 3) identify meaningful future treatment outcome measures. METHODS: Patients were evaluated prospectively through ongoing clinical care. RESULTS: Twenty-three patients were evaluated: 8 infantile GM1, 9 infantile Tay-Sachs disease, 6 infantile Sandhoff disease. Common patterns of clinical change included: hypotonia before 6months of age; severe motor skill impairment within first year of life; seizures; dysphagia and feeding-tube placement before 18months of age. Neurodevelopmental testing scores reached the floor of the testing scale by 20 to 28months of age. Vertebral beaking, kyphosis, and scoliosis were unique to patients with infantile GM1. Chest physiotherapy was associated with increased survival in iGM1 (p=0.0056). Miglustat combined with a low-carbohydrate ketogenic diet (the Syner-G regimen) in patients who received a feeding-tube was associated with increased survival in infantile GM1 (p=0.025). CONCLUSIONS: This is the first prospective study of the natural history of infantile gangliosidoses and the very first natural history of infantile GM1. The homogeneity of the infantile gangliosidoses phenotype as demonstrated by the clinical events timeline in this study provides promising secondary outcome measure candidates. This study indicates that overall survival is a meaningful primary outcome measure for future clinical trials due to reliable timing and early occurrence of this event. Combination therapy approaches, instead of monotherapy approaches, will likely be the best way to optimize clinical outcomes. Combination therapy approaches include palliative therapies (e.g., chest physiotherapy) along with treatments that address the underlying disease pathology (e.g. miglustat or future gene therapies).


Asunto(s)
Gangliosidosis GM2/fisiopatología , Gangliosidosis/fisiopatología , Gangliosidosis/terapia , Gangliosidosis GM1/fisiopatología , 1-Desoxinojirimicina/efectos adversos , 1-Desoxinojirimicina/análogos & derivados , 1-Desoxinojirimicina/uso terapéutico , Dieta Cetogénica , Disacaridasas/antagonistas & inhibidores , Femenino , Gangliosidosis/complicaciones , Gangliosidosis GM2/terapia , Gangliosidosis GM1/terapia , Inhibidores de Glicósido Hidrolasas/efectos adversos , Inhibidores de Glicósido Hidrolasas/uso terapéutico , Humanos , Lactante , Masculino , Estudios Prospectivos , Estudios Retrospectivos
11.
Mol Ther ; 25(4): 892-903, 2017 04 05.
Artículo en Inglés | MEDLINE | ID: mdl-28236574

RESUMEN

GM1 gangliosidosis is a fatal neurodegenerative disease that affects individuals of all ages. Favorable outcomes using adeno-associated viral (AAV) gene therapy in GM1 mice and cats have prompted consideration of human clinical trials, yet there remains a paucity of objective biomarkers to track disease status. We developed a panel of biomarkers using blood, urine, cerebrospinal fluid (CSF), electrodiagnostics, 7 T MRI, and magnetic resonance spectroscopy in GM1 cats-either untreated or AAV treated for more than 5 years-and compared them to markers in human GM1 patients where possible. Significant alterations were noted in CSF and blood of GM1 humans and cats, with partial or full normalization after gene therapy in cats. Gene therapy improved the rhythmic slowing of electroencephalograms (EEGs) in GM1 cats, a phenomenon present also in GM1 patients, but nonetheless the epileptiform activity persisted. After gene therapy, MR-based analyses revealed remarkable preservation of brain architecture and correction of brain metabolites associated with microgliosis, neuroaxonal loss, and demyelination. Therapeutic benefit of AAV gene therapy in GM1 cats, many of which maintain near-normal function >5 years post-treatment, supports the strong consideration of human clinical trials, for which the biomarkers described herein will be essential for outcome assessment.


Asunto(s)
Biomarcadores , Gangliosidosis GM1/genética , Gangliosidosis GM1/metabolismo , Terapia Genética , Animales , Biomarcadores/sangre , Biomarcadores/líquido cefalorraquídeo , Biomarcadores/orina , Gatos , Dependovirus/clasificación , Dependovirus/genética , Modelos Animales de Enfermedad , Electroencefalografía , Gangliosidosis GM1/mortalidad , Gangliosidosis GM1/terapia , Terapia Genética/métodos , Vectores Genéticos/administración & dosificación , Vectores Genéticos/genética , Humanos , Hipocalcemia/metabolismo , Imagen por Resonancia Magnética , Espectroscopía de Resonancia Magnética , Resultado del Tratamiento
12.
Pediatr Endocrinol Rev ; 13 Suppl 1: 663-73, 2016 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-27491214

RESUMEN

The gangliosidoses are lysosomal storage disorders caused by accumulation of GM1 or GM2 gangliosides. GM1 gangliosidosis has both central nervous system and systemic findings; while, GM2 gangliosidosis is restricted primarily to the central nervous system. Both disorders have autosomal recessive modes of inheritance and a continuum of clinical presentations from a severe infantile form to a milder, chronic adult form. Both are devastating diseases without cure or specific treatment however, with the use of supportive aggressive medical management, the lifespan and quality of life has been extended for both diseases. Naturally occurring and engineered animal models that mimic the human diseases have enhanced our understanding of the pathogenesis of disease progression. Some models have shown significant improvement in symptoms and lifespan with enzyme replacement, substrate reduction, and anti-inflammatory treatments alone or in combination. More recently gene therapy has shown impressive results in large and small animal models. Treatment with FDA-approved glucose analogs to reduce the amount of ganglioside substrate is used as off-label treatments for some patients. Therapies also under clinical development include small molecule chaperones and gene therapy.


Asunto(s)
Antiinflamatorios/uso terapéutico , Terapia de Reemplazo Enzimático/métodos , Gangliosidosis GM2/terapia , Gangliosidosis GM1/terapia , Terapia Genética/métodos , Progresión de la Enfermedad , Gangliosidosis GM2/fisiopatología , Gangliosidosis GM1/fisiopatología , Glucosa/análogos & derivados , Humanos , Índice de Severidad de la Enfermedad
13.
Hum Mol Genet ; 24(15): 4353-64, 2015 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-25964428

RESUMEN

GM1 gangliosidosis (GM1) is an autosomal recessive lysosomal storage disease where GLB1 gene mutations result in a reduction or absence of lysosomal acid ß-galactosidase (ßgal) activity. ßgal deficiency leads to accumulation of GM1-ganglioside in the central nervous system (CNS). GM1 is characterized by progressive neurological decline resulting in generalized paralysis, extreme emaciation and death. In this study, we assessed the therapeutic efficacy of an adeno-associated virus (AAV) 9-mßgal vector infused systemically in adult GM1 mice (ßGal(-/-)) at 1 × 10(11) or 3 × 10(11) vector genomes (vg). Biochemical analysis of AAV9-treated GM1 mice showed high ßGal activity in liver and serum. Moderate ßGal levels throughout CNS resulted in a 36-76% reduction in GM1-ganglioside content in the brain and 75-86% in the spinal cord. Histological analyses of the CNS of animals treated with 3 × 10(11) vg dose revealed increased presence of ßgal and clearance of lysosomal storage throughout cortex, hippocampus, brainstem and spinal cord. Storage reduction in these regions was accompanied by a marked decrease in astrogliosis. AAV9 treatment resulted in improved performance in multiple tests of motor function and behavior. Also the majority of GM1 mice in the 3 × 10(11) vg cohort retained ambulation and rearing despite reaching the humane endpoint due to weight loss. Importantly, the median survival of AAV9 treatment groups (316-576 days) was significantly increased over controls (250-264 days). This study shows that moderate widespread expression of ßgal in the CNS of GM1 gangliosidosis mice is sufficient to achieve significant biochemical impact with phenotypic amelioration and extension in lifespan.


Asunto(s)
Sistema Nervioso Central/metabolismo , Gangliosidosis GM1/genética , Terapia Genética , beta-Galactosidasa/genética , Animales , Astrocitos/metabolismo , Astrocitos/patología , Tronco Encefálico/metabolismo , Tronco Encefálico/patología , Sistema Nervioso Central/patología , Dependovirus/genética , Modelos Animales de Enfermedad , Gangliósidos/metabolismo , Gangliosidosis GM1/metabolismo , Gangliosidosis GM1/terapia , Vectores Genéticos , Humanos , Ratones , Médula Espinal/metabolismo , Médula Espinal/patología , beta-Galactosidasa/biosíntesis , beta-Galactosidasa/sangre
14.
Brain Dev ; 35(6): 515-23, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23290321

RESUMEN

Chaperone therapy is a newly developed molecular therapeutic approach to lysosomal diseases, a group of human genetic diseases causing severe brain damage. Based on early molecular studies during the last decade of the 20th century and early years of the 21st century, mainly on Fabry disease and GM1-gangliosidosis, we found some mutant enzyme proteins were unstable in the cell, and unable to express catalytic activities. Subsequently galactose and other active-site binding substrate analogs were found stabilized and enhance the mutant enzyme activity in culture cells. We concluded that the mutant misfolding enzyme protein and substrate analog competitive inhibitor (chemical chaperone) form a stable complex to be transported to the lysosome, to restore the catalytic activity of mutant enzyme after spontaneous dissociation under the acidic condition. This gene mutation-specific molecular interaction is a paradoxical phenomenon that an enzyme inhibitor in vitro serves as an enzyme stabilizer in situ. First we developed a commercially available compound 1-deoxygalactonojirimycin (DGJ) for Fabry disease, and confirmed the above molecular phenomenon. Currently DGJ has become a new candidate of oral medicine for Fabry disease, generalized vasculopathy involving the kidneys, heart and central nervous system in the middle age. This drug development has reached the phase 3 of human clinical study. Then we found two valienamine derivatives, N-octyl-4-epi-ß-valienamine (NOEV) and N-octyl-ß-valienamine (NOV), as promising therapeutic agents for human ß-galactosidase deficiency disorders (GM1-gangliosidosis and Morquio B disease) and ß-glucosidase deficiency disorders (phenotypic variations of Gaucher disease), respectively. Originally NOEV and NOV had been discovered as competitive inhibitors, and then their paradoxical bioactivities as chaperones were confirmed in cultured fibroblasts from patients with these disorders. Subsequently GM1-gangliosidosis model mice have been used for confirmation of clinical effectiveness, adverse effects and pharmacokinetic studies. Orally administered NOEV entered the brain through the blood-brain barrier, enhanced ß-galactosidase activity, reduced substrate storage, and improved neurological deterioration clinically. Computational analysis revealed pH-dependent enzyme-chaperone interactions. Our recent study indicated chaperone activity of a new DGJ derivative, MTD118, for ß-galactosidase complementary to NOEV. NOV also showed the chaperone effect toward several ß-glucosidase gene mutants in Gaucher disease. Furthermore a commercial expectorant drug ambroxol was found to be a chaperone for ß-glucosidase. A few Gaucher patients responded to this drug with remarkable improvement of oculomotor dysfunction and myoclonus. We hope chaperone therapy will become available for some patients with Fabry disease, GM1-gangliosidosis, Gaucher disease, and other lysosomal storage diseases particularly with central nervous system involvement.


Asunto(s)
Enfermedad de Fabry , Gangliosidosis GM1 , Enfermedad de Gaucher , Chaperonas Moleculares/uso terapéutico , Animales , Enfermedad de Fabry/genética , Enfermedad de Fabry/terapia , Gangliosidosis GM1/enzimología , Gangliosidosis GM1/genética , Gangliosidosis GM1/terapia , Enfermedad de Gaucher/genética , Enfermedad de Gaucher/terapia , Humanos , Chaperonas Moleculares/biosíntesis , Chaperonas Moleculares/genética
15.
Mol Genet Metab ; 106(1): 92-8, 2012 May.
Artículo en Inglés | MEDLINE | ID: mdl-22436580

RESUMEN

Therapeutic chaperone effect of a valienamine derivative N-octyl 4-epi-ß-valienamine (NOEV) was studied in G(M1)-gangliosidosis model mice. Phamacokinetic analysis revealed rapid intestinal absorption and renal excretion after oral administration. Intracellular accumulation was not observed after continuous treatment. NOEV was delivered to the central nervous system through the blood-brain barrier to induce high expression of the apparently deficient ß-galactosidase activity. NOEV treatment starting at the early stage of disease resulted in remarkable arrest of neurological progression within a few months. Survival time was significantly prolonged. This result suggests that NOEV chaperone therapy will be clinically effective for prevention of neuronal damage if started early in life hopefully also in human patients with G(M1)-gangliosidosis.


Asunto(s)
Gangliosidosis GM1/terapia , Hexosaminas/administración & dosificación , Chaperonas Moleculares/administración & dosificación , beta-Galactosidasa/genética , beta-Glucosidasa/genética , Animales , Barrera Hematoencefálica , Sistema Nervioso Central/enzimología , Sistema Nervioso Central/patología , Modelos Animales de Enfermedad , Gangliosidosis GM1/enzimología , Gangliosidosis GM1/patología , Regulación de la Expresión Génica/efectos de los fármacos , Hexosaminas/farmacocinética , Humanos , Mucosa Intestinal/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Chaperonas Moleculares/farmacocinética , Urinálisis
16.
PLoS One ; 5(10): e13468, 2010 Oct 18.
Artículo en Inglés | MEDLINE | ID: mdl-20976108

RESUMEN

BACKGROUND: GM1-gangliosidosis is a glycosphingolipid (GSL) lysosomal storage disease caused by a genetic deficiency of acid ß-galactosidase (ßgal), which results in the accumulation of GM1-ganglioside and its asialo-form (GA1) primarily in the CNS. Age of onset ranges from infancy to adulthood, and excessive ganglioside accumulation produces progressive neurodegeneration and psychomotor retardation in humans. Currently, there are no effective therapies for the treatment of GM1-gangliosidosis. METHODOLOGY/PRINCIPAL FINDINGS: In this study we examined the effect of thalamic infusion of AAV2/1-ßgal vector in adult GM1 mice on enzyme distribution, activity, and GSL content in the CNS, motor behavior, and survival. Six to eight week-old GM1 mice received bilateral injections of AAV vector in the thalamus, or thalamus and deep cerebellar nuclei (DCN) with pre-determined endpoints at 1 and 4 months post-injection, and the humane endpoint, or 52 weeks of age. Enzyme activity was elevated throughout the CNS of AAV-treated GM1 mice and GSL storage nearly normalized in most structures analyzed, except in the spinal cord which showed ∼50% reduction compared to age-matched untreated GM1 mice spinal cord. Survival was significantly longer in AAV-treated GM1 mice (52 wks) than in untreated mice. However the motor performance of AAV-treated GM1 mice declined over time at a rate similar to that observed in untreated GM1 mice. CONCLUSIONS/SIGNIFICANCE: Our studies show that the AAV-modified thalamus can be used as a 'built-in' central node network for widespread distribution of lysosomal enzymes in the mouse cerebrum. In addition, this study indicates that thalamic delivery of AAV vectors should be combined with additional targets to supply the cerebellum and spinal cord with therapeutic levels of enzyme necessary to achieve complete correction of the neurological phenotype in GM1 mice.


Asunto(s)
Dependovirus/genética , Gangliosidosis GM1/terapia , Vectores Genéticos , Transfección , Animales , Cromatografía en Capa Delgada , Potenciales Evocados Visuales , Gangliosidosis GM1/genética , Ratones , Reacción en Cadena de la Polimerasa , Prueba de Desempeño de Rotación con Aceleración Constante , Análisis de Supervivencia
17.
Brain Dev ; 31(10): 717-24, 2009 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-19118961

RESUMEN

We performed a cell transplantation study to treat the brain involvement in lysosomal storage diseases. We used acid beta-galactosidase knock-out mice (BKO) from C57BL/6 as recipients. To minimize immune responses, we used cells derived from transgenic mice of C57BL/6 overexpressing the normal human beta-galactosidase. Fetal brain cells (FBC), bone marrow-derived mesenchymal stem cells (MSC), and mixed FBC and MSC cells were prepared and injected into the ventricle of newborn BKO mouse brain. The mice were examined at 1, 2, 4, and 8 weeks and 6 months after injection. In each experiment, the injected cells migrated into the whole brain effectively and survived for at least 8 weeks. Decrease in ganglioside GM1 level was also observed. FBC could survive for 6 months in recipient brain. However, the number of transplanted FBC decreased. In the brains of MSC- or mixed cell-treated mice, no grafted cells could be found at 6 months. To achieve sufficient long-term effects on the brain, a method of steering the immune response away from cytotoxic responses or of inducing tolerance to the products of therapeutic genes must be developed.


Asunto(s)
Corteza Cerebral/trasplante , Gangliosidosis GM1/terapia , Trasplante de Células Madre Mesenquimatosas , Animales , Western Blotting , Trasplante de Tejido Encefálico , Células Cultivadas , Trasplante de Tejido Fetal , Gangliosidosis GM1/genética , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , beta-Galactosidasa/genética , beta-Galactosidasa/metabolismo
18.
Gene Ther ; 16(2): 303-8, 2009 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-18818671

RESUMEN

GM1-gangliosidosis is a lysosomal storage disease (LSD) caused by an autosomal recessive deficiency of lysosomal acid beta-galactosidase (betagal). This leads to accumulation of GM1-ganglioside and its asialo derivative GA1 in the central nervous system (CNS), and progressive neurodegeneration. Therapeutic AAV-mediated gene delivery to the brain for LSDs has proven very successful in several animal models. GM1-gangliosidosis is also a prime candidate for AAV-mediated gene therapy in the CNS. As global neuropathology characterizes the most severe forms of this disease, therapeutic interventions need to achieve distribution of betagal throughout the entire CNS. Therefore, careful consideration of routes of administration and target structures from where metabolically active enzyme can be produced, released and distributed throughout the CNS, is necessary. The goal of this study was to investigate the pattern and mechanism of distribution of betagal in the adult GM1-gangliosidosis mouse brain upon hippocampal injection of an AAV vector-encoding betagal. We found evidence that three different mechanisms contribute to its distribution in the brain: (1) diffusion; (2) axonal transport within neurons from the site of production; (3) CSF flow in the perivascular space of Virchow-Robin. In addition, we found evidence of axonal transport of vector-encoded mRNA.


Asunto(s)
Encéfalo/enzimología , Gangliosidosis GM1/enzimología , Terapia Genética/métodos , beta-Galactosidasa/genética , Animales , Transporte Axonal , Dependovirus/genética , Modelos Animales de Enfermedad , Gangliosidosis GM1/terapia , Vectores Genéticos/farmacocinética , Hipocampo/enzimología , Ratones , Ratones Noqueados , Neuronas/fisiología , ARN Mensajero/genética , Distribución Tisular , beta-Galactosidasa/biosíntesis , beta-Galactosidasa/deficiencia , beta-Galactosidasa/farmacocinética
19.
Mol Genet Metab ; 94(4): 391-396, 2008 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-18524657

RESUMEN

GM(1) gangliosidosis is a lysosomal storage disorder due to deficiency of the beta-galactosidase enzyme. This deficiency results in accumulation of GM(1) gangliosides and related glycoconjugates in the lysosomes leading to lysosomal swelling, cellular damage, and organ dysfunction. The disease is lethal in the infantile and juvenile forms. To date, up to 102 mutations distributed along the beta-galactosidase gene (GLB1) have been reported. This review gives an overview of the clinical and molecular findings in patients with GM(1) gangliosidosis. Furthermore, it describes therapeutic approaches which are currently under investigation in animal models of the disease.


Asunto(s)
Gangliosidosis GM1/genética , Gangliosidosis GM1/terapia , beta-Galactosidasa/genética , Adolescente , Adulto , Secuencia de Aminoácidos , Animales , Niño , Preescolar , Modelos Animales de Enfermedad , Femenino , Gangliosidosis GM1/metabolismo , Gangliosidosis GM1/patología , Humanos , Lactante , Masculino , Mutación , beta-Galactosidasa/química , beta-Galactosidasa/metabolismo
20.
Braz J Med Biol Res ; 41(4): 283-8, 2008 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-18392450

RESUMEN

GM1 gangliosidosis is an autosomal recessive disorder caused by the deficiency of lysosomal acid hydrolase beta-galactosidase (beta-Gal). It is one of the most frequent lysosomal storage disorders in Brazil, with an estimated frequency of 1:17,000. The enzyme is secreted and can be captured by deficient cells and targeted to the lysosomes. There is no effective treatment for GM1 gangliosidosis. To determine the efficiency of an expression vector for correcting the genetic defect of GM1 gangliosidosis, we tested transfer of the beta-Gal gene (Glb1) to fibroblasts in culture using liposomes. Beta-Gal cDNA was cloned into the expression vectors pSCTOP and pREP9. Transfection was performed using 4 microL lipofectamine 2000 and 1.5-2.0 microg DNA. Cells (2 x 10(5)/well) were harvested 24 h, 48 h, and 7 days after transfection. Enzyme specific activity was measured in cell lysate and supernatant by fluorometric assay. Twenty-four hours after transfection, treated cells showed a higher enzyme specific activity (pREP9-beta-Gal: 621.5 +/- 323.0, pSCTOP-beta-Gal: 714.5 +/- 349.5, pREP9-beta-Gal + pSCTOP-beta-Gal: 1859.0 +/- 182.4, and pREP9-ss-Gal + pTRACER: 979.5 +/- 254.9 nmol x h-1 x mg-1 protein) compared to untreated cells (18.0 +/- 3.1 for cell and 32.2 +/- 22.2 nmol x h-1 x mg-1 protein for supernatant). However, cells maintained in culture for 7 days showed values similar to those of untreated patients. In the present study, we were able to transfect primary patients' skin fibroblasts in culture using a non-viral vector which overexpresses the beta-Gal gene for 24 h. This is the first attempt to correct fibroblasts from patients with GM1 gangliosidosis by gene therapy using a non-viral vector.


Asunto(s)
Fibroblastos/enzimología , Gangliosidosis GM1/enzimología , Vectores Genéticos , Transfección/métodos , beta-Galactosidasa/metabolismo , ADN Complementario , Fluorometría , Gangliosidosis GM1/terapia , Humanos , Liposomas , Plásmidos/genética , beta-Galactosidasa/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA