Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Sci Rep ; 9(1): 594, 2019 01 24.
Artículo en Inglés | MEDLINE | ID: mdl-30679733

RESUMEN

Sporadic Alzheimer's disease (AD) is the most common cause of dementia. However, representative experimental models of AD have remained difficult to produce because of the disease's uncertain origin. The Polycomb group protein BMI1 regulates chromatin compaction and gene silencing. BMI1 expression is abundant in adult brain neurons but down-regulated in AD brains. We show here that mice lacking one allele of Bmi1 (Bmi1+/-) develop normally but present with age cognitive deficits and neurodegeneration sharing similarities with AD. Bmi1+/- mice also transgenic for the amyloid beta precursor protein died prematurely and present aggravated disease. Loss of heterochromatin and DNA damage response (DDR) at repetitive DNA sequences were predominant in Bmi1+/- mouse neurons and inhibition of the DDR mitigated the amyloid and Tau phenotype. Heterochromatin anomalies and DDR at repetitive DNA sequences were also found in AD brains. Aging Bmi1+/- mice may thus represent an interesting model to identify and study novel pathogenic mechanisms related to AD.


Asunto(s)
Enfermedad de Alzheimer/patología , Inestabilidad Genómica , Heterocromatina/metabolismo , Complejo Represivo Polycomb 1/genética , Proteínas Proto-Oncogénicas/genética , Enfermedad de Alzheimer/metabolismo , Enfermedad de Alzheimer/mortalidad , Péptidos beta-Amiloides/metabolismo , Animales , Encéfalo/metabolismo , Encéfalo/patología , Modelos Animales de Enfermedad , Femenino , Humanos , Estimación de Kaplan-Meier , Potenciación a Largo Plazo , Masculino , Aprendizaje por Laberinto , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Neuronas/citología , Neuronas/metabolismo , Complejo Represivo Polycomb 1/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Memoria Espacial , Proteínas tau/metabolismo
2.
Development ; 143(9): 1571-84, 2016 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-26965367

RESUMEN

Retinal development occurs through the sequential but overlapping generation of six types of neuronal cells and one glial cell type. Of these, rod and cone photoreceptors represent the functional unit of light detection and phototransduction and are frequently affected in retinal degenerative diseases. During mouse development, the Polycomb group protein Bmi1 is expressed in immature retinal progenitors and differentiated retinal neurons, including cones. We show here that Bmi1 is required to prevent post natal degeneration of cone photoreceptors and bipolar neurons and that inactivation of Chk2 or p53 could improve but not overcome cone degeneration in Bmi1(-/-) mice. The retinal phenotype of Bmi1(-/-) mice was also characterized by loss of heterochromatin, activation of tandem repeats, oxidative stress and Rip3-associated necroptosis. In the human retina, BMI1 was preferentially expressed in cones at heterochromatic foci. BMI1 inactivation in human embryonic stem cells was compatible with retinal induction but impaired cone terminal differentiation. Despite this developmental arrest, BMI1-deficient cones recapitulated several anomalies observed in Bmi1(-/-) photoreceptors, such as loss of heterochromatin, activation of tandem repeats and induction of p53, revealing partly conserved biological functions between mouse and man.


Asunto(s)
Células Madre Embrionarias/citología , Necrosis/genética , Complejo Represivo Polycomb 1/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Células Fotorreceptoras Retinianas Conos/citología , Células Fotorreceptoras Retinianas Conos/fisiología , Animales , Línea Celular , Quinasa de Punto de Control 2/genética , Heterocromatina/metabolismo , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Estrés Oxidativo , Complejo Represivo Polycomb 1/genética , Proteínas Proto-Oncogénicas/genética , Proteína Serina-Treonina Quinasas de Interacción con Receptores , Retina/embriología , Células Fotorreceptoras Retinianas Bastones/citología , Proteína p53 Supresora de Tumor/genética
3.
PLoS One ; 7(2): e31870, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22384090

RESUMEN

Aging increases the risk to develop several neurodegenerative diseases, although the underlying mechanisms are poorly understood. Inactivation of the Polycomb group gene Bmi1 in mice results in growth retardation, cerebellar degeneration, and development of a premature aging-like phenotype. This progeroid phenotype is characterized by formation of lens cataracts, apoptosis of cortical neurons, and increase of reactive oxygen species (ROS) concentrations, owing to p53-mediated repression of antioxidant response (AOR) genes. Herein we report that Bmi1 expression progressively declines in the neurons of aging mouse and human brains. In old brains, p53 accumulates at the promoter of AOR genes, correlating with a repressed chromatin state, down-regulation of AOR genes, and increased oxidative damages to lipids and DNA. Comparative gene expression analysis further revealed that aging brains display an up-regulation of the senescence-associated genes IL-6, p19(Arf) and p16(Ink4a), along with the pro-apoptotic gene Noxa, as seen in Bmi1-null mice. Increasing Bmi1 expression in cortical neurons conferred robust protection against DNA damage-induced cell death or mitochondrial poisoning, and resulted in suppression of ROS through activation of AOR genes. These observations unveil that Bmi1 genetic deficiency recapitulates aspects of physiological brain aging and that Bmi1 over-expression is a potential therapeutic modality against neurodegeneration.


Asunto(s)
Envejecimiento , Encéfalo/fisiología , Enfermedades Neurodegenerativas/metabolismo , Proteínas Nucleares/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Proteínas Represoras/metabolismo , Animales , Antioxidantes/metabolismo , Apoptosis , Encéfalo/metabolismo , Cromatina/metabolismo , ADN/metabolismo , Regulación hacia Abajo , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Genes p53 , Humanos , Peroxidación de Lípido , Lípidos/química , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Enfermedades Neurodegenerativas/prevención & control , Neuronas/metabolismo , Estrés Oxidativo , Complejo Represivo Polycomb 1 , Regiones Promotoras Genéticas , Especies Reactivas de Oxígeno , Proteína p53 Supresora de Tumor/metabolismo
4.
Antioxid Redox Signal ; 15(6): 1729-37, 2011 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-20849375

RESUMEN

Recent advances in delineating the biological functions of p53 had shed the light on its key role in the multifacets of cellular homeostasis. After its activation, via DNA damage, oxidative stress, or aberrant expression of oncogenes, p53 transduces its classical effect through several mechanisms comprising activation of the DNA repair machinery, cell cycle arrest, and initiation of apoptosis or senescence. In the mammalian brain, p53 plays critical functions in normal development, tumor suppression, neurodegenerative diseases, and aging. Herein, we focus on the constitutive pro-oxidant activity of p53 in neurons and discuss the potential implication of this finding in the context of neurodegenerative diseases and normal brain aging.


Asunto(s)
Envejecimiento/metabolismo , Encéfalo/metabolismo , Enfermedades Neurodegenerativas/metabolismo , Proteína p53 Supresora de Tumor/metabolismo , Animales , Apoptosis , Humanos , Ratones , Neuronas/metabolismo , Oxidación-Reducción
5.
J Neurosci ; 30(30): 10096-111, 2010 Jul 28.
Artículo en Inglés | MEDLINE | ID: mdl-20668194

RESUMEN

Glioblastoma multiforme (GBM) is an aggressive brain tumor that is resistant to all known therapies. Within these tumors, a CD133-positive cancer-initiating neural stem cell (NSC) population was shown to be resistant to gamma radiation through preferential activation of the DNA double-strand break (DSB) response machinery, including the ataxia-telangiectasia-mutated (ATM) kinase. The polycomb group protein BMI1 is enriched in CD133-positive GBM cells and required for their self-renewal in an INK4A/ARF-independent manner through transcriptional repression of alternate tumor suppressor pathways. We report here that BMI1 copurifies with DNA DSB response and nonhomologous end joining (NHEJ) repair proteins in GBM cells. BMI1 was enriched at the chromatin after irradiation and colocalized and copurified with ATM and the histone gammaH2AX. BMI1 also preferentially copurified with NHEJ proteins DNA-PK, PARP-1, hnRNP U, and histone H1 in CD133-positive GBM cells. BMI1 deficiency in GBM cells severely impaired DNA DSB response, resulting in increased sensitivity to radiation. In turn, BMI1 overexpression in normal NSCs enhanced ATM recruitment to the chromatin, the rate of gammaH2AX foci resolution, and resistance to radiation. BMI1 thus displays a previously uncharacterized function in controlling DNA DSB response and repair. Pharmacological inhibition of BMI1 combined with radiation therapy may provide an effective mean to target GBM stem cells.


Asunto(s)
Daño del ADN/fisiología , Células Madre Embrionarias/efectos de la radiación , Células Madre Neoplásicas/efectos de la radiación , Proteínas Nucleares/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Tolerancia a Radiación , Proteínas Represoras/metabolismo , Antígeno AC133 , Antígenos CD/metabolismo , Neoplasias Encefálicas/patología , Línea Celular Tumoral , Transformación Celular Neoplásica/patología , Transformación Celular Neoplásica/efectos de la radiación , Quinasa de Punto de Control 2 , Cromatografía Liquida/métodos , Ensayo Cometa/métodos , Roturas del ADN de Doble Cadena/efectos de la radiación , Proteínas de Unión al ADN/metabolismo , Células Madre Embrionarias/fisiología , Feto , Citometría de Flujo/métodos , Glioblastoma/patología , Glicoproteínas/metabolismo , Proteínas Fluorescentes Verdes/genética , Humanos , Inmunoprecipitación/métodos , Células Madre Neoplásicas/fisiología , Proteínas Nucleares/genética , Péptidos/metabolismo , Fosforilación/genética , Complejo Represivo Polycomb 1 , Proteínas del Grupo Polycomb , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Proto-Oncogénicas/genética , Especies Reactivas de Oxígeno/metabolismo , Proteínas Represoras/genética , Espectrometría de Masas en Tándem/métodos , Transfección/métodos
6.
Stem Cells ; 28(8): 1412-23, 2010 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-20549707

RESUMEN

The developing mammalian retina is generated by the proliferation and differentiation of multipotent retinal progenitor cells (RPCs) giving rise to neuronal and glial lineages. Whether an immature progenitor/stem cell subpopulation is present in the developing mammalian retina remains undefined. Deficiency in the polycomb group gene Bmi1 results in reduced proliferation and postnatal depletion of neural and hematopoietic stem cells. Here, we show that Bmi1 is required for the self-renewal of most immature RPCs and for postnatal retinal development. In the embryo, Bmi1 is highly enriched in a rare stage-specific embryonic antigen-1-positive RPC subpopulation expressing the stem cell markers Sox2, Lhx2, and Musashi. Gain-of-function experiments revealed that Bmi1 overexpression could convert RPCs having limited proliferation capacity into RPCs showing extensive proliferation and multiple differentiation capacities over time. At all developmental stages analyzed using the neurosphere assay, Bmi1 deficiency resulted in reduced proliferation and self-renewal of most immature RPCs. Reduced RPCs proliferation was also observed in the peripheral retina of Bmi1(-/-) fetus and newborn mice. The biological impact of these developmental anomalies was revealed by the reduced retinal diameter of Bmi1-deficient pups. P19(Arf) and p16(Ink4a) were upregulated in vivo and in vitro and coinactivation of p53, which lies downstream of p19(Arf), partially restored Bmi1-deficient RPCs self-renewal phenotype. Bmi1 thus distinguishes immature RPCs from the main RPC population and is required for normal retinal development.


Asunto(s)
Proteínas Nucleares/fisiología , Proteínas Proto-Oncogénicas/fisiología , Proteínas Represoras/fisiología , Retina/citología , Retina/metabolismo , Células Madre/citología , Células Madre/metabolismo , Animales , Diferenciación Celular/genética , Diferenciación Celular/fisiología , Citometría de Flujo , Ratones , Proteínas Nucleares/genética , Complejo Represivo Polycomb 1 , Proteínas Proto-Oncogénicas/genética , Proteínas Represoras/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
7.
J Neurosci ; 29(28): 8884-96, 2009 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-19605626

RESUMEN

Glioblastoma multiforme (GBM) is one of the most common and aggressive types of brain tumors. In GBM, a subpopulation of CD133-positive cancer initiating cells displays stem cell characteristics. The Polycomb group (PcG) and oncogene BMI1 is part of the Polycomb repressive complex 1 (PRC1) that regulates gene expression by modifying chromatin organization. Here we show that BMI1 is expressed in human GBM tumors and highly enriched in CD133-positive cells. Stable BMI1 knockdown using short hairpin RNA-expressing lentiviruses resulted in inhibition of clonogenic potential in vitro and of brain tumor formation in vivo. Cell biology studies support the notion that BMI1 prevents CD133-positive cell apoptosis and/or differentiation into neurons and astrocytes, depending on the cellular context. Gene expression analyses suggest that BMI1 represses alternate tumor suppressor pathways that attempt to compensate for INK4A/ARF/P53 deletion and PI(3)K/AKT hyperactivity. Inhibition of EZH2, the main component of the PRC2, also impaired GBM tumor growth. Our results reveal that PcG proteins are involved in GBM tumor growth and required to sustain cancer initiating stem cell renewal.


Asunto(s)
Neoplasias Encefálicas/patología , Glioblastoma/patología , Células Madre Neoplásicas/fisiología , Proteínas Nucleares/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Proteínas Represoras/metabolismo , Antígeno AC133 , Análisis de Varianza , Animales , Antígenos CD/metabolismo , Apoptosis/genética , Diferenciación Celular/fisiología , Línea Celular Tumoral , Transdiferenciación Celular , Transformación Celular Neoplásica/metabolismo , Transformación Celular Neoplásica/patología , Células Cultivadas , Inmunoprecipitación de Cromatina/métodos , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Modelos Animales de Enfermedad , Proteína Potenciadora del Homólogo Zeste 2 , Citometría de Flujo , Regulación Neoplásica de la Expresión Génica/genética , Silenciador del Gen/fisiología , Glicoproteínas/metabolismo , Humanos , Ratones , Ratones Endogámicos NOD/fisiología , Ratones Endogámicos NOD/cirugía , Ratones SCID/cirugía , Células Madre Neoplásicas/patología , Proteínas Nucleares/genética , Análisis de Secuencia por Matrices de Oligonucleótidos/métodos , Péptidos/metabolismo , Complejo Represivo Polycomb 1 , Complejo Represivo Polycomb 2 , Proteínas Proto-Oncogénicas/genética , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo , Proteínas Represoras/genética , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Ensayo de Tumor de Célula Madre/métodos
8.
J Neurosci ; 29(2): 529-42, 2009 Jan 14.
Artículo en Inglés | MEDLINE | ID: mdl-19144853

RESUMEN

Aging may be determined by a genetic program and/or by the accumulation rate of molecular damages. Reactive oxygen species (ROS) generated by the mitochondrial metabolism have been postulated to be the central source of molecular damages and imbalance between levels of intracellular ROS and antioxidant defenses is a characteristic of the aging brain. How aging modifies free radicals concentrations and increases the risk to develop most neurodegenerative diseases is poorly understood, however. Here we show that the Polycomb group and oncogene Bmi1 is required in neurons to suppress apoptosis and the induction of a premature aging-like program characterized by reduced antioxidant defenses. Before weaning, Bmi1(-/-) mice display a progeroid-like ocular and brain phenotype, while Bmi1(+/-) mice, although apparently normal, have reduced lifespan. Bmi1 deficiency in neurons results in increased p19(Arf)/p53 levels, abnormally high ROS concentrations, and hypersensitivity to neurotoxic agents. Most Bmi1 functions on neurons' oxidative metabolism are genetically linked to repression of p53 pro-oxidant activity, which also operates in physiological conditions. In Bmi1(-/-) neurons, p53 and corepressors accumulate at antioxidant gene promoters, correlating with a repressed chromatin state and antioxidant gene downregulation. These findings provide a molecular mechanism explaining how Bmi1 regulates free radical concentrations and reveal the biological impact of Bmi1 deficiency on neuronal survival and aging.


Asunto(s)
Peroxidación de Lípido/genética , Neuronas/metabolismo , Proteínas Nucleares/metabolismo , Peroxirredoxinas/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Proteínas Represoras/metabolismo , Proteína p53 Supresora de Tumor/metabolismo , Factores de Edad , Análisis de Varianza , Animales , Apoptosis/genética , Proliferación Celular , Supervivencia Celular/genética , Células Cultivadas , Corteza Cerebral/citología , Embrión de Mamíferos , Proteínas del Ojo/metabolismo , Regulación de la Expresión Génica/genética , Proteína Ácida Fibrilar de la Glía/metabolismo , Histonas/metabolismo , Proteínas de Homeodominio/metabolismo , Peróxido de Hidrógeno/farmacología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas Asociadas a Microtúbulos/metabolismo , Neuronas/efectos de los fármacos , Neurotoxinas/farmacología , Proteínas Nucleares/deficiencia , Factor de Transcripción PAX6 , Factores de Transcripción Paired Box/metabolismo , Peroxirredoxinas/genética , Fosfopiruvato Hidratasa/metabolismo , Complejo Represivo Polycomb 1 , Proteínas Proto-Oncogénicas/deficiencia , Proteína p53 Supresora de Tumor/genética , beta-Galactosidasa/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...