Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 3 de 3
Filtrar
1.
Nucleic Acids Res ; 50(5): 2700-2718, 2022 03 21.
Artículo en Inglés | MEDLINE | ID: mdl-35212385

RESUMEN

The autosomal recessive genome instability disorder Ataxia-telangiectasia, caused by mutations in ATM kinase, is characterized by the progressive loss of cerebellar neurons. We find that DNA damage associated with ATM loss results in dysfunctional behaviour of human microglia, immune cells of the central nervous system. Microglial dysfunction is mediated by the pro-inflammatory RELB/p52 non-canonical NF-κB transcriptional pathway and leads to excessive phagocytic clearance of neuronal material. Activation of the RELB/p52 pathway in ATM-deficient microglia is driven by persistent DNA damage and is dependent on the NIK kinase. Activation of non-canonical NF-κB signalling is also observed in cerebellar microglia of individuals with Ataxia-telangiectasia. These results provide insights into the underlying mechanisms of aberrant microglial behaviour in ATM deficiency, potentially contributing to neurodegeneration in Ataxia-telangiectasia.


Asunto(s)
Proteínas de la Ataxia Telangiectasia Mutada , Ataxia Telangiectasia , Daño del ADN , Microglía , Ataxia Telangiectasia/genética , Ataxia Telangiectasia/patología , Proteínas de la Ataxia Telangiectasia Mutada/metabolismo , Humanos , Microglía/metabolismo , FN-kappa B/genética , FN-kappa B/metabolismo
2.
J Clin Invest ; 130(3): 1377-1391, 2020 03 02.
Artículo en Inglés | MEDLINE | ID: mdl-31877112

RESUMEN

Hematopoietic stem cell (HSC) attrition is considered the key event underlying progressive BM failure (BMF) in Fanconi anemia (FA), the most frequent inherited BMF disorder in humans. However, despite major advances, how the cellular, biochemical, and molecular alterations reported in FA lead to HSC exhaustion remains poorly understood. Here, we demonstrated in human and mouse cells that loss-of-function of FANCA or FANCC, products of 2 genes affecting more than 80% of FA patients worldwide, is associated with constitutive expression of the transcription factor microphthalmia (MiTF) through the cooperative, unscheduled activation of several stress-signaling pathways, including the SMAD2/3, p38 MAPK, NF-κB, and AKT cascades. We validated the unrestrained Mitf expression downstream of p38 in Fanca-/- mice, which display hallmarks of hematopoietic stress, including loss of HSC quiescence, DNA damage accumulation in HSCs, and reduced HSC repopulation capacity. Importantly, we demonstrated that shRNA-mediated downregulation of Mitf expression or inhibition of p38 signaling rescued HSC quiescence and prevented DNA damage accumulation. Our data support the hypothesis that HSC attrition in FA is the consequence of defects in the DNA-damage response combined with chronic activation of otherwise transiently activated signaling pathways, which jointly prevent the recovery of HSC quiescence.


Asunto(s)
Trastornos de Fallo de la Médula Ósea/metabolismo , Daño del ADN , Anemia de Fanconi/metabolismo , Células Madre Hematopoyéticas/metabolismo , Sistema de Señalización de MAP Quinasas , Factor de Transcripción Asociado a Microftalmía/metabolismo , Animales , Ácido Ascórbico , Trastornos de Fallo de la Médula Ósea/genética , Trastornos de Fallo de la Médula Ósea/patología , Línea Celular , Colecalciferol , Deshidroepiandrosterona/análogos & derivados , Quinasas MAP Reguladas por Señal Extracelular/genética , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Anemia de Fanconi/genética , Anemia de Fanconi/patología , Proteínas del Grupo de Complementación de la Anemia de Fanconi/genética , Proteínas del Grupo de Complementación de la Anemia de Fanconi/metabolismo , Células Madre Hematopoyéticas/patología , Ratones , Ratones Noqueados , Factor de Transcripción Asociado a Microftalmía/genética , Ácidos Nicotínicos , Extractos Vegetales , Proteínas Smad/genética , Proteínas Smad/metabolismo
3.
Sci Rep ; 6: 36539, 2016 11 09.
Artículo en Inglés | MEDLINE | ID: mdl-27827420

RESUMEN

Proteins involved in genetic stability maintenance and safeguarding DNA replication act not only against cancer initiation but could also play a major role in sustaining cancer progression. Here, we report that the FANC pathway is highly expressed in metastatic melanoma harboring the oncogenic microphthalmia-associated transcription factor (MiTF). We show that MiTF downregulation in melanoma cells lowers the expression of several FANC genes and proteins. Moreover, we observe that, similarly to the consequence of MiTF downregulation, FANC pathway silencing alters proliferation, migration and senescence of human melanoma cells. We demonstrate that the FANC pathway acts downstream MiTF and establish the existence of an epistatic relationship between MiTF and the FANC pathway. Our findings point to a central role of the FANC pathway in cellular and chromosomal resistance to both DNA damage and targeted therapies in melanoma cells. Thus, the FANC pathway is a promising new therapeutic target in melanoma treatment.


Asunto(s)
Proliferación Celular , Supervivencia Celular , Proteína del Grupo de Complementación D2 de la Anemia de Fanconi/fisiología , Melanoma/patología , Factor de Transcripción Asociado a Microftalmía/metabolismo , Animales , Línea Celular Tumoral , Resistencia a Antineoplásicos , Proteína del Grupo de Complementación D2 de la Anemia de Fanconi/genética , Silenciador del Gen , Humanos , Indoles/uso terapéutico , Melanoma/tratamiento farmacológico , Melanoma/metabolismo , Ratones , Ratones Desnudos , Factor de Transcripción Asociado a Microftalmía/genética , Oxidación-Reducción , Especies Reactivas de Oxígeno/metabolismo , Sulfonamidas/uso terapéutico , Vemurafenib
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA