Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 6 de 6
Filtrar
Más filtros











Intervalo de año de publicación
1.
Neurobiol Dis ; 180: 106092, 2023 05.
Artículo en Inglés | MEDLINE | ID: mdl-36948261

RESUMEN

RecQ helicase family proteins play vital roles in maintaining genome stability, including DNA replication, recombination, and DNA repair. In human cells, there are five RecQ helicases: RECQL1, Bloom syndrome (BLM), Werner syndrome (WRN), RECQL4, and RECQL5. Dysfunction or absence of RecQ proteins is associated with genetic disorders, tumorigenesis, premature aging, and neurodegeneration. The biochemical and biological roles of RecQ helicases are rather well established, however, there is no systematic study comparing the behavioral changes among various RecQ-deficient mice including consequences of exposure to DNA damage. Here, we investigated the effects of ionizing irradiation (IR) on three RecQ-deficient mouse models (RecQ1, WRN and RecQ4). We find abnormal cognitive behavior in RecQ-deficient mice in the absence of IR. Interestingly, RecQ dysfunction impairs social ability and induces depressive-like behavior in mice after a single exposure to IR, suggesting that RecQ proteins play roles in mood and cognition behavior. Further, transcriptomic and metabolomic analyses revealed significant alterations in RecQ-deficient mice, especially after IR exposure. In particular, pathways related to neuronal and microglial functions, DNA damage repair, cell cycle, and reactive oxygen responses were downregulated in the RecQ4 and WRN mice. In addition, increased DNA damage responses were found in RecQ-deficient mice. Notably, two genes, Aldolase Fructose-Bisphosphate B (Aldob) and NADPH Oxidase 4 (Nox4), were differentially expressed in RecQ-deficient mice. Our findings suggest that RecQ dysfunction contributes to social and depressive-like behaviors in mice, and that aldolase activity may be associated with these changes, representing a potential therapeutic target.


Asunto(s)
Replicación del ADN , RecQ Helicasas , Animales , Humanos , Ratones , RecQ Helicasas/genética , RecQ Helicasas/metabolismo , Reparación del ADN , Daño del ADN , Inestabilidad Genómica , Aldehído-Liasas/genética , Aldehído-Liasas/metabolismo
2.
Aging Cell ; 20(4): e13329, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33734555

RESUMEN

Senescence phenotypes and mitochondrial dysfunction are implicated in aging and in premature aging diseases, including ataxia telangiectasia (A-T). Loss of mitochondrial function can drive age-related decline in the brain, but little is known about whether improving mitochondrial homeostasis alleviates senescence phenotypes. We demonstrate here that mitochondrial dysfunction and cellular senescence with a senescence-associated secretory phenotype (SASP) occur in A-T patient fibroblasts, and in ATM-deficient cells and mice. Senescence is mediated by stimulator of interferon genes (STING) and involves ectopic cytoplasmic DNA. We further show that boosting intracellular NAD+ levels with nicotinamide riboside (NR) prevents senescence and SASP by promoting mitophagy in a PINK1-dependent manner. NR treatment also prevents neurodegeneration, suppresses senescence and neuroinflammation, and improves motor function in Atm-/- mice. Our findings suggest a central role for mitochondrial dysfunction-induced senescence in A-T pathogenesis, and that enhancing mitophagy as a potential therapeutic intervention.


Asunto(s)
Ataxia Telangiectasia/dietoterapia , Ataxia Telangiectasia/metabolismo , Suplementos Dietéticos , Proteínas de la Membrana/metabolismo , Mitofagia/efectos de los fármacos , NAD/metabolismo , Niacinamida/análogos & derivados , Compuestos de Piridinio/administración & dosificación , Fenotipo Secretor Asociado a la Senescencia/genética , Transducción de Señal/efectos de los fármacos , Animales , Ataxia Telangiectasia/genética , Proteínas de la Ataxia Telangiectasia Mutada/genética , Proteínas de la Ataxia Telangiectasia Mutada/metabolismo , Estudios de Casos y Controles , Línea Celular Tumoral , Modelos Animales de Enfermedad , Femenino , Fibroblastos/efectos de los fármacos , Fibroblastos/metabolismo , Humanos , Masculino , Proteínas de la Membrana/genética , Ratones , Ratones Noqueados , Mitocondrias/metabolismo , Mitofagia/genética , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Niacinamida/administración & dosificación , Ratas , Ratas Sprague-Dawley , Transducción de Señal/genética , Transfección , Resultado del Tratamiento
3.
EMBO J ; 39(21): e103420, 2020 11 02.
Artículo en Inglés | MEDLINE | ID: mdl-32935380

RESUMEN

Short telomeres are a principal defining feature of telomere biology disorders, such as dyskeratosis congenita (DC), for which there are no effective treatments. Here, we report that primary fibroblasts from DC patients and late generation telomerase knockout mice display lower nicotinamide adenine dinucleotide (NAD) levels, and an imbalance in the NAD metabolome that includes elevated CD38 NADase and reduced poly(ADP-ribose) polymerase and SIRT1 activities, respectively, affecting many associated biological pathways. Supplementation with the NAD precursor, nicotinamide riboside, and CD38 inhibition improved NAD homeostasis, thereby alleviating telomere damage, defective mitochondrial biosynthesis and clearance, cell growth retardation, and cellular senescence of DC fibroblasts. These findings reveal a direct, underlying role of NAD dysregulation when telomeres are short and underscore its relevance to the pathophysiology and interventions of human telomere-driven diseases.


Asunto(s)
Disqueratosis Congénita/genética , Disqueratosis Congénita/metabolismo , Fibroblastos/metabolismo , NAD/metabolismo , Telomerasa/genética , Telómero/metabolismo , ADP-Ribosil Ciclasa 1/genética , Animales , Encéfalo/patología , Línea Celular , Senescencia Celular , Disqueratosis Congénita/patología , Femenino , Homeostasis , Humanos , Glicoproteínas de Membrana/genética , Ratones , Ratones Noqueados , Mitocondrias/metabolismo , Niacinamida/análogos & derivados , Niacinamida/metabolismo , Fenotipo , Poli(ADP-Ribosa) Polimerasa-1/metabolismo , Compuestos de Piridinio/metabolismo , Telomerasa/metabolismo
4.
Mech Ageing Dev ; 186: 111207, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-31923475

RESUMEN

Aging is associated with multiple human pathologies. In the past few years mitochondrial homeostasis has been well correlated with age-related disorders and longevity. Mitochondrial homeostasis involves generation, biogenesis and removal of dysfunctional mitochondria via mitophagy. Mitophagy is regulated by various mitochondrial and extra-mitochondrial factors including morphology, oxidative stress and DNA damage. For decades, DNA damage and inefficient DNA repair have been considered as major determinants for age-related disorders. Although defects in DNA damage recognition and repair and mitophagy are well documented to be major factors in age-associated diseases, interactivity between these is poorly understood. Mitophagy efficiency decreases with age leading to accumulation of dysfunctional mitochondria enhancing the severity of age-related disorders including neurodegenerative diseases, inflammatory diseases, cancer, diabetes and many more. Therefore, mitophagy is being targeted for intervention in age-associated disorders. NAD+ supplementation has emerged as one intervention to target both defective DNA repair and mitophagy. In this review, we discuss the molecular signaling pathways involved in regulation of DNA damage and repair and of mitophagy, and we highlight the opportunities for clinical interventions targeting these processes to improve the quality of life during aging.


Asunto(s)
Envejecimiento/fisiología , Daño del ADN , Mitofagia/fisiología , Envejecimiento/genética , Reparación del ADN , Humanos , Transducción de Señal
5.
Nat Neurosci ; 22(3): 401-412, 2019 03.
Artículo en Inglés | MEDLINE | ID: mdl-30742114

RESUMEN

Accumulation of damaged mitochondria is a hallmark of aging and age-related neurodegeneration, including Alzheimer's disease (AD). The molecular mechanisms of impaired mitochondrial homeostasis in AD are being investigated. Here we provide evidence that mitophagy is impaired in the hippocampus of AD patients, in induced pluripotent stem cell-derived human AD neurons, and in animal AD models. In both amyloid-ß (Aß) and tau Caenorhabditis elegans models of AD, mitophagy stimulation (through NAD+ supplementation, urolithin A, and actinonin) reverses memory impairment through PINK-1 (PTEN-induced kinase-1)-, PDR-1 (Parkinson's disease-related-1; parkin)-, or DCT-1 (DAF-16/FOXO-controlled germline-tumor affecting-1)-dependent pathways. Mitophagy diminishes insoluble Aß1-42 and Aß1-40 and prevents cognitive impairment in an APP/PS1 mouse model through microglial phagocytosis of extracellular Aß plaques and suppression of neuroinflammation. Mitophagy enhancement abolishes AD-related tau hyperphosphorylation in human neuronal cells and reverses memory impairment in transgenic tau nematodes and mice. Our findings suggest that impaired removal of defective mitochondria is a pivotal event in AD pathogenesis and that mitophagy represents a potential therapeutic intervention.


Asunto(s)
Enfermedad de Alzheimer/metabolismo , Péptidos beta-Amiloides/metabolismo , Hipocampo/metabolismo , Hipocampo/patología , Mitofagia , Neuronas/metabolismo , Neuronas/patología , Enfermedad de Alzheimer/patología , Enfermedad de Alzheimer/psicología , Animales , Animales Modificados Genéticamente , Caenorhabditis elegans , Modelos Animales de Enfermedad , Femenino , Células Madre Pluripotentes Inducidas , Masculino , Memoria , Ratones , Células-Madre Neurales
6.
Mol Brain ; 10(1): 29, 2017 07 03.
Artículo en Inglés | MEDLINE | ID: mdl-28673309

RESUMEN

Studies have implied that the circadian oscillation of mitogen-activated protein kinase (MAPK) signal pathways is crucial for hippocampus-dependent memory. NF1 mouse models (Nf1 heterozygous null mutants; Nf1 +/-) displayed enhanced MAPK activity in the hippocampus and resulted in memory deficits. We assumed a link between MAPK pathways and hippocampal rhythmic oscillations, which have never been explored in Nf1 +/- mice. We demonstrated that the level of extracellular signal-regulated kinases 1 and 2 (ERK1/2) phosphorylation in Nf1 +/- mice were significantly higher at nighttime than at daytime. Moreover, the in vivo recording revealed that for the Nf1 +/- group, the power spectral density of theta rhythm significantly decreased and the firing rates of pyramidal neurons increased. Our results indicated that the hippocampal MAPK oscillation and theta rhythmic oscillations in Nf1 +/- mice were disturbed and hinted about a possible mechanism for the brain dysfunction in Nf1 +/- mice.


Asunto(s)
Ritmo Circadiano , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Hipocampo/enzimología , Hipocampo/fisiopatología , Neurofibromina 1/genética , Ritmo Teta/fisiología , Potenciales de Acción/fisiología , Animales , Masculino , Ratones Endogámicos C57BL , Ratones Mutantes , Fosforilación
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA