Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
1.
Cell Physiol Biochem ; 38(1): 173-84, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-26765775

RESUMEN

BACKGROUND/AIMS: ALKBH1, an AlkB homologue in the 2-oxoglutarate and Fe2+ dependent hydroxylase family, is a histone dioxygenase that removes methyl groups from histone H2A. Studies of transgenic mice lacking Alkbh1 reveal that most Alkbh1-/- embryos die during embryonic development. Embryonic stem cells (ESCs) derived from these mice have prolonged expression of pluripotency markers and delayed induction of genes involved in neural differentiation, indicating that ALKBH1 is involved in regulation of pluripotency and differentiation. The aim of this study was to further investigate the role ALKBH1 in early development. METHODS: Double-filter methods for nitrocellulose-filter binding, dot blot, enzyme-linked immunosorbent assay (ELISA), immonocytochemistry, cell culture and differentiation of mouse ESCs, Co-IP and miRNA analysis. RESULTS: We found that SOX2 and NANOG bind the ALKBH1 promoter, and we identified protein-protein interactions between ALKBH1 and these core transcription factors of the pluripotency network. Furthermore, lack of ALKBH1 affected the expression of developmentally important miRNAs, which are involved in the regulation of NANOG, SOX2 and neural differentiation. CONCLUSION: Our results suggest that ALKBH1 interacts with the core transcriptional pluripotency network of ESCs and is involved in regulation of pluripotency and differentiation.


Asunto(s)
ADN-(Sitio Apurínico o Apirimidínico) Liasa/metabolismo , Regulación del Desarrollo de la Expresión Génica/genética , Redes Reguladoras de Genes , Factores de Transcripción/metabolismo , Regiones no Traducidas 5' , Histona H2a Dioxigenasa, Homólogo 1 de AlkB , Animales , Secuencia de Bases , Sitios de Unión , Diferenciación Celular , Línea Celular , ADN-(Sitio Apurínico o Apirimidínico) Liasa/deficiencia , ADN-(Sitio Apurínico o Apirimidínico) Liasa/genética , Técnicas de Inactivación de Genes , Proteínas de Homeodominio/química , Proteínas de Homeodominio/metabolismo , Humanos , Ratones , MicroARNs/metabolismo , Células Madre Embrionarias de Ratones/citología , Células Madre Embrionarias de Ratones/metabolismo , Proteína Homeótica Nanog , Factor 3 de Transcripción de Unión a Octámeros/química , Factor 3 de Transcripción de Unión a Octámeros/metabolismo , Regiones Promotoras Genéticas , Dominios y Motivos de Interacción de Proteínas , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo , Factores de Transcripción SOXB1/química , Factores de Transcripción SOXB1/metabolismo , Factores de Transcripción/química
2.
Nucleic Acids Res ; 40(18): 9044-59, 2012 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-22810208

RESUMEN

The structure specific flap endonuclease 1 (FEN1) plays an essential role in long-patch base excision repair (BER) and in DNA replication. We have generated a fluorescently tagged FEN1 expressing mouse which allows monitoring the localization and kinetics of FEN1 in response to DNA damage in living cells and tissues. The expression of FEN1, which is tagged at its C-terminal end with enhanced yellow fluorescent protein (FEN1-YFP), is under control of the endogenous Fen1 transcriptional regulatory elements. In line with its role in processing of Okazaki fragments during DNA replication, we found that FEN1-YFP expression is mainly observed in highly proliferating tissue. Moreover, the FEN1-YFP fusion protein allowed us to investigate repair kinetics in cells challenged with local and global DNA damage. In vivo multi-photon fluorescence microscopy demonstrates rapid localization of FEN1 to local laser-induced DNA damage sites in nuclei, providing evidence of a highly mobile protein that accumulates fast at DNA lesion sites with high turnover rate. Inhibition of poly (ADP-ribose) polymerase 1 (PARP1) disrupts FEN1 accumulation at sites of DNA damage, indicating that PARP1 is required for FEN1 recruitment to DNA repair intermediates in BER.


Asunto(s)
Reparación del ADN , Endonucleasas de ADN Solapado/metabolismo , Animales , Proteínas Bacterianas/genética , Encéfalo/metabolismo , Células Cultivadas , Daño del ADN , Endonucleasas de ADN Solapado/análisis , Endonucleasas de ADN Solapado/genética , Técnicas de Sustitución del Gen , Cinética , Proteínas Luminiscentes/genética , Ratones , Poli(ADP-Ribosa) Polimerasa-1 , Inhibidores de Poli(ADP-Ribosa) Polimerasas , Antígeno Nuclear de Célula en Proliferación/análisis , Fase S
3.
Carcinogenesis ; 32(1): 80-5, 2011 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-21062970

RESUMEN

Oxidative DNA modifications such as 7,8-dihydro-8-oxoguanine (8-oxoG) are generated endogenously in apparently all living cells. The defect of the repair of 8-oxoG in Csb(m/m)Ogg1⁻(/)⁻ mice results in elevated basal levels of these lesions and increased frequencies of spontaneous mutations, which initiate tumorigenesis in the liver if cell proliferation is stimulated. Here, we describe that the phytoalexin resveratrol, applied either for 7 days per gavage (100 mg/kg body wt) or for 3-9 months in the diet (0.04% ad libitum), reduces the endogenous oxidative DNA base damage in the livers of the Csb(m/m)Ogg1⁻(/)⁻ mice by 20-30% (P < 0.01). A small but consistent effect is also observed in the wild-type animals. The spontaneous mutation frequencies determined in the lacI gene of BigBlue® Csb(m/m)Ogg1⁻(/)⁻ mice are concomitantly reduced by resveratrol to similar extents. Mechanistically, the protection is caused by an induction of the antioxidant defense system since (i) hepatocytes isolated from all resveratrol-treated animals were less susceptible to the generation of single-strand breaks and to cell killing by H2O2, (ii) messenger RNA levels of superoxide dismutases 1 and 2 (SOD1 and SOD2) heme oxygenase-1 and glutathione peroxidase were significantly upregulated after the short-term treatment and (iii) mutations primarily ascribed to the oxidative base modification 8-oxoG (G:C to T:A transversions) were more strongly suppressed than G:C to A:T transitions ascribed to spontaneous deamination. The results thus demonstrate that spontaneous somatic mutation rates resulting from endogenous oxidative DNA damage can be reduced by application of an exogenous agent.


Asunto(s)
Antimutagênicos/administración & dosificación , Hígado/efectos de los fármacos , Mutagénesis/efectos de los fármacos , Estrés Oxidativo/efectos de los fármacos , Estilbenos/administración & dosificación , Animales , Daño del ADN/efectos de los fármacos , Dieta , Expresión Génica/efectos de los fármacos , Ratones , Ratones Transgénicos , Estrés Oxidativo/fisiología , Resveratrol , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
4.
Cell Rep ; 35(7): 109154, 2021 05 18.
Artículo en Inglés | MEDLINE | ID: mdl-34010660

RESUMEN

Human telomerase reverse transcriptase (hTERT) remains suppressed in most normal somatic cells. Resulting erosion of telomeres leads eventually to replicative senescence. Reactivation of hTERT maintains telomeres and triggers progression of >90% of cancers. However, any direct causal link between telomeres and telomerase regulation remains unclear. Here, we show that the telomere-repeat-binding-factor 2 (TRF2) binds hTERT promoter G-quadruplexes and recruits the polycomb-repressor EZH2/PRC2 complex. This is causal for H3K27 trimethylation at the hTERT promoter and represses hTERT in cancer as well as normal cells. Two highly recurrent hTERT promoter mutations found in many cancers, including ∼83% glioblastoma multiforme, that are known to destabilize hTERT promoter G-quadruplexes, showed loss of TRF2 binding in patient-derived primary glioblastoma multiforme cells. Ligand-induced G-quadruplex stabilization restored TRF2 binding, H3K27-trimethylation, and hTERT re-suppression. These results uncover a mechanism of hTERT regulation through a telomeric factor, implicating telomere-telomerase molecular links important in neoplastic transformation, aging, and regenerative therapy.


Asunto(s)
G-Cuádruplex , Telomerasa/metabolismo , Humanos , Telómero/metabolismo
5.
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
6.
Mol Cell Biol ; 30(6): 1357-67, 2010 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-20065039

RESUMEN

Mitochondrial dysfunction underlying changes in neurodegenerative diseases is often associated with apoptosis and a progressive loss of neurons, and damage to the mitochondrial genome is proposed to be involved in such pathologies. In the present study we designed a mouse model that allows us to specifically induce mitochondrial DNA toxicity in the forebrain neurons of adult mice. This is achieved by CaMKIIalpha-regulated inducible expression of a mutated version of the mitochondrial UNG DNA repair enzyme (mutUNG1). This enzyme is capable of removing thymine from the mitochondrial genome. We demonstrate that a continual generation of apyrimidinic sites causes apoptosis and neuronal death. These defects are associated with behavioral alterations characterized by increased locomotor activity, impaired cognitive abilities, and lack of anxietylike responses. In summary, whereas mitochondrial base substitution and deletions previously have been shown to correlate with premature and natural aging, respectively, we show that a high level of apyrimidinic sites lead to mitochondrial DNA cytotoxicity, which causes apoptosis, followed by neurodegeneration.


Asunto(s)
Apoptosis/efectos de los fármacos , Conducta Animal/efectos de los fármacos , ADN Mitocondrial/toxicidad , Degeneración Nerviosa/patología , Neuronas/patología , Prosencéfalo/efectos de los fármacos , Prosencéfalo/patología , Animales , Ansiedad/patología , Atrofia , Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/metabolismo , Línea Celular , Cognición/efectos de los fármacos , Espinas Dendríticas/efectos de los fármacos , Espinas Dendríticas/patología , Espinas Dendríticas/ultraestructura , Humanos , Locomoción/efectos de los fármacos , Ratones , Ratones Transgénicos , Mitocondrias/efectos de los fármacos , Mitocondrias/metabolismo , Modelos Animales , Proteínas Mutantes/metabolismo , Neuronas/efectos de los fármacos , Células Piramidales/efectos de los fármacos , Células Piramidales/patología , Células Piramidales/ultraestructura , Sinapsis/efectos de los fármacos , Sinapsis/ultraestructura , Uracil-ADN Glicosidasa/metabolismo
7.
Cancer Res ; 68(12): 4571-9, 2008 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-18559501

RESUMEN

Flap endonuclease 1 (FEN1) processes Okazaki fragments in lagging strand DNA synthesis, and FEN1 is involved in several DNA repair pathways. The interaction of FEN1 with the proliferating cell nuclear antigen (PCNA) processivity factor is central to the function of FEN1 in both DNA replication and repair. Here we present two gene-targeted mice with mutations in FEN1. The first mutant mouse carries a single amino acid point mutation in the active site of the nuclease domain of FEN1 (Fen1(E160D/E160D)), and the second mutant mouse contains two amino acid substitutions in the highly conserved PCNA interaction domain of FEN1 (Fen1(DeltaPCNA/DeltaPCNA)). Fen1(E160D/E160D) mice develop a considerably elevated incidence of B-cell lymphomas beginning at 6 months of age, particularly in females. By 16 months of age, more than 90% of the Fen1(E160D/E160D) females have tumors, primarily lymphomas. By contrast, Fen1(DeltaPCNA/DeltaPCNA) mouse embryos show extensive apoptosis in the forebrain and vertebrae area and die around stage E9.5 to E11.5.


Asunto(s)
Apoptosis , Embrión de Mamíferos/metabolismo , Embrión de Mamíferos/patología , Endonucleasas de ADN Solapado/fisiología , Linfoma/etiología , Linfoma/patología , Antígeno Nuclear de Célula en Proliferación/fisiología , Animales , Animales Recién Nacidos , Ciclo Celular/fisiología , Proliferación Celular , Células Cultivadas , Medio de Cultivo Libre de Suero , Reparación del ADN , Replicación del ADN , Embrión de Mamíferos/citología , Femenino , Genes de Inmunoglobulinas/genética , Genes Letales , Resistencia a la Insulina , Masculino , Ratones , Ratones Mutantes , Mutación/genética , Hibridación de Ácido Nucleico , Obesidad/etiología , Estructura Terciaria de Proteína , Recombinación Genética
8.
J Exp Biol ; 209(Pt 13): 2432-41, 2006 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-16788026

RESUMEN

Genes encoding the myogenic regulating factors MyoD and myogenin and the structural muscle proteins myosin light chain 2 (MyLC2) and myosin heavy chain (MyHC) were isolated from juvenile Atlantic halibut (Hippoglossus hippoglossus L.). The impact of temperature on their temporal and spatial expression during somitogenesis were examined by incubating halibut embryos at 4, 6 and 8 degrees C, and regularly sampling for whole-mount in situ hybridisation and reverse transcription (RT)-PCR. There were no significant effects of temperature on the onset of somitogenesis or number of somites at hatching. The rate of somite formation increased with increasing temperature, and the expression of MyoD, myogenin and MyHC followed the cranial-to-caudal somite formation. Hence, no significant effect of temperature on the spatial and temporal expression of the genes studied was found in relation to somite stage. MyoD, which has subsequently been shown to encode the MyoD2 isoform, displayed a novel bilaterally asymmetric expression pattern only in white muscle precursor cells during early halibut somitogenesis. The expression of myogenin resembled that previously described for other fish species, and preceded the MyHC expression by approximately five somites. Two MyLC2 cDNA sequences were for the first time described for a flatfish, probably representing embryonic (MyLC2a) and larval/juvenile (MyLC2b) isoforms. Factors regulating muscle determination, differentiation and development have so far mostly been studied in vertebrates with external bilateral symmetry. The findings of the present study suggest that more such investigations of flatfish species could provide valuable information on how muscle-regulating mechanisms work in species with different anatomical, physiological and ecological traits.


Asunto(s)
Lenguado/embriología , Lenguado/metabolismo , Proteína MioD/metabolismo , Miogenina/metabolismo , Miosinas/metabolismo , Somitos/fisiología , Secuencia de Aminoácidos , Animales , Secuencia de Bases , Clonación Molecular , ADN Complementario/análisis , Embrión no Mamífero/citología , Embrión no Mamífero/metabolismo , Proteínas de Peces/genética , Proteínas de Peces/metabolismo , Lenguado/genética , Regulación del Desarrollo de la Expresión Génica , Secuencias Hélice-Asa-Hélice , Datos de Secuencia Molecular , Proteína MioD/química , Proteína MioD/genética , Miogenina/química , Miogenina/genética , Cadenas Pesadas de Miosina/genética , Cadenas Pesadas de Miosina/metabolismo , Cadenas Ligeras de Miosina/genética , Cadenas Ligeras de Miosina/metabolismo , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Alineación de Secuencia , Somitos/citología , Somitos/metabolismo , Temperatura
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA