Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 6 de 6
Filtrar
1.
Neurodegener Dis ; 19(1): 22-34, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31167196

RESUMEN

BACKGROUND: Huntington's disease (HD) is a devastating neurodegenerative disorder caused by CAG triplet expansions in the huntingtin gene. Oxidative stress is linked to HD pathology, although it is not clear whether this is an effect or a mediator of disease. The transgenic (TgHD) minipig expresses the N-terminal part of human-mutated huntingtin and represents a unique model to investigate therapeutic strategies towards HD. A more detailed characterization of this model is needed to fully utilize its potential. METHODS: In this study, we focused on the molecular and cellular features of fibroblasts isolated from TgHD minipigs and the wild-type (WT) siblings at different ages, pre-symptomatic at the age of 24-36 months and with the onset of behavioural symptoms at the age of 48 months. We measured oxidative stress, the expression of oxidative stress-related genes, proliferation capacity along with the expression of cyclin B1 and D1 proteins, cellular permeability, and the integrity of the nuclear DNA (nDNA) and mitochondrial DNA in these cells. RESULTS: TgHD fibroblasts isolated from 48-month-old animals showed increased oxidative stress, which correlated with the overexpression of SOD2 encoding mitochondrial superoxide dismutase 2, and the NEIL3 gene encoding DNA glycosylase involved in replication-associated repair of oxidized DNA. TgHD cells displayed an abnormal proliferation capacity and permeability. We further demonstrated increased nDNA damage in pre-symptomatic TgHD fibroblasts (isolated from animals aged 24-36 months). CONCLUSIONS: Our results unravel phenotypic alterations in primary fibroblasts isolated from the TgHD minipig model at the age of 48 months. Importantly, nDNA damage appears to precede these phenotypic alterations. Our results highlight the impact of fibroblasts from TgHD minipigs in studying the molecular mechanisms of HD pathophysiology that gradually occur with age.


Asunto(s)
Envejecimiento/metabolismo , Fibroblastos/metabolismo , Proteína Huntingtina/metabolismo , Animales , Animales Modificados Genéticamente , División Celular , Daño del ADN , ADN Mitocondrial/genética , Regulación de la Expresión Génica , Humanos , Proteína Huntingtina/genética , Peroxidación de Lípido , N-Glicosil Hidrolasas/biosíntesis , N-Glicosil Hidrolasas/genética , Estrés Oxidativo , Fenotipo , Cultivo Primario de Células , Especies Reactivas de Oxígeno/metabolismo , Superóxido Dismutasa/biosíntesis , Superóxido Dismutasa/genética , Porcinos , Porcinos Enanos
2.
Dis Model Mech ; 12(7)2019 07 26.
Artículo en Inglés | MEDLINE | ID: mdl-31278192

RESUMEN

Skeletal muscle wasting and atrophy is one of the more severe clinical impairments resulting from the progression of Huntington's disease (HD). Mitochondrial dysfunction may play a significant role in the etiology of HD, but the specific condition of mitochondria in muscle has not been widely studied during the development of HD. To determine the role of mitochondria in skeletal muscle during the early stages of HD, we analyzed quadriceps femoris muscle from 24-, 36-, 48- and 66-month-old transgenic minipigs that expressed the N-terminal portion of mutated human huntingtin protein (TgHD) and age-matched wild-type (WT) siblings. We found altered ultrastructure of TgHD muscle tissue and mitochondria. There was also significant reduction of activity of citrate synthase and respiratory chain complexes (RCCs) I, II and IV, decreased quantity of oligomycin-sensitivity conferring protein (OSCP) and the E2 subunit of pyruvate dehydrogenase (PDHE2), and differential expression of optic atrophy 1 protein (OPA1) and dynamin-related protein 1 (DRP1) in the skeletal muscle of TgHD minipigs. Statistical analysis identified several parameters that were dependent only on HD status and could therefore be used as potential biomarkers of disease progression. In particular, the reduction of biomarker RCCII subunit SDH30 quantity suggests that similar pathogenic mechanisms underlie disease progression in TgHD minipigs and HD patients. The perturbed biochemical phenotype was detectable in TgHD minipigs prior to the development of ultrastructural changes and locomotor impairment, which become evident at the age of 48 months. Mitochondrial disturbances may contribute to energetic depression in skeletal muscle in HD, which is in concordance with the mobility problems observed in this model.This article has an associated First Person interview with the first author of the paper.


Asunto(s)
Modelos Animales de Enfermedad , Metabolismo Energético , Enfermedad de Huntington/metabolismo , Mitocondrias Musculares/metabolismo , Músculo Esquelético/metabolismo , Animales , Animales Modificados Genéticamente , Peso Corporal , ADN/metabolismo , Progresión de la Enfermedad , Transporte de Electrón , Humanos , Proteína Huntingtina/genética , Enfermedad de Huntington/patología , Mitocondrias Musculares/ultraestructura , Proteínas Mitocondriales/metabolismo , Músculo Esquelético/ultraestructura , Mutación , Fosforilación Oxidativa , Porcinos , Porcinos Enanos
3.
Sci Rep ; 8(1): 9817, 2018 06 29.
Artículo en Inglés | MEDLINE | ID: mdl-29959348

RESUMEN

Huntington's disease (HD) is a progressive neurodegenerative disorder primarily affecting the basal ganglia and is caused by expanded CAG repeats in the huntingtin gene. Except for CAG sizing, mitochondrial and nuclear DNA (mtDNA and nDNA) parameters have not yet proven to be representative biomarkers for disease and future therapy. Here, we identified a general suppression of genes associated with aerobic metabolism in peripheral blood mononuclear cells (PBMCs) from HD patients compared to controls. In HD, the complex II subunit SDHB was lowered although not sufficiently to affect complex II activity. Nevertheless, we found decreased level of factors associated with mitochondrial biogenesis and an associated dampening of the mitochondrial DNA damage frequency in HD, implying an early defect in mitochondrial activity. In contrast to mtDNA, nDNA from HD patients was four-fold more modified than controls and demonstrated that nDNA integrity is severely reduced in HD. Interestingly, the level of nDNA damage correlated inversely with the total functional capacity (TFC) score; an established functional score of HD. Our data show that PBMCs are a promising source to monitor HD progression and highlights nDNA damage and diverging mitochondrial and nuclear genome responses representing early cellular impairments in HD.


Asunto(s)
Daño del ADN , ADN Mitocondrial/análisis , Inestabilidad Genómica , Enfermedad de Huntington/patología , Leucocitos Mononucleares/patología , Mitocondrias/patología , Adulto , Anciano , Estudios de Casos y Controles , ADN Mitocondrial/genética , Femenino , Humanos , Enfermedad de Huntington/genética , Leucocitos Mononucleares/metabolismo , Masculino , Persona de Mediana Edad , Mitocondrias/metabolismo , Adulto Joven
4.
Dis Model Mech ; 11(10)2018 10 24.
Artículo en Inglés | MEDLINE | ID: mdl-30254085

RESUMEN

Huntington's disease (HD) is a monogenic, progressive, neurodegenerative disorder with currently no available treatment. The Libechov transgenic minipig model for HD (TgHD) displays neuroanatomical similarities to humans and exhibits slow disease progression, and is therefore more powerful than available mouse models for the development of therapy. The phenotypic characterization of this model is still ongoing, and it is essential to validate biomarkers to monitor disease progression and intervention. In this study, the behavioral phenotype (cognitive, motor and behavior) of the TgHD model was assessed, along with biomarkers for mitochondrial capacity, oxidative stress, DNA integrity and DNA repair at different ages (24, 36 and 48 months), and compared with age-matched controls. The TgHD minipigs showed progressive accumulation of the mutant huntingtin (mHTT) fragment in brain tissue and exhibited locomotor functional decline at 48 months. Interestingly, this neuropathology progressed without any significant age-dependent changes in any of the other biomarkers assessed. Rather, we observed genotype-specific effects on mitochondrial DNA (mtDNA) damage, mtDNA copy number, 8-oxoguanine DNA glycosylase activity and global level of the epigenetic marker 5-methylcytosine that we believe is indicative of a metabolic alteration that manifests in progressive neuropathology. Peripheral blood mononuclear cells (PBMCs) were relatively spared in the TgHD minipig, probably due to the lack of detectable mHTT. Our data demonstrate that neuropathology in the TgHD model has an age of onset of 48 months, and that oxidative damage and electron transport chain impairment represent later states of the disease that are not optimal for assessing interventions.This article has an associated First Person interview with the first author of the paper.


Asunto(s)
Conducta Animal , Enfermedad de Huntington/patología , 8-Hidroxi-2'-Desoxicoguanosina , Animales , Animales Modificados Genéticamente , Daño del ADN , Reparación del ADN , Desoxiguanosina/análogos & derivados , Desoxiguanosina/metabolismo , Modelos Animales de Enfermedad , Metabolismo Energético , Genoma , Humanos , Proteína Huntingtina/metabolismo , Enfermedad de Huntington/metabolismo , Mitocondrias/metabolismo , Degeneración Nerviosa/patología , Especificidad de Órganos , Porcinos , Porcinos Enanos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA