Por favor, use este identificador para citar o enlazar a este item: http://hdl.handle.net/10261/222074
COMPARTIR / EXPORTAR:
logo share SHARE logo core CORE BASE
Visualizar otros formatos: MARC | Dublin Core | RDF | ORE | MODS | METS | DIDL | DATACITE

Invitar a revisión por pares abierta
Título

ADAR1-mediated RNA editing is a novel oncogenic process in thyroid cancer and regulates miR-200 activity

AutorRamírez-Moya, Julia CSIC ORCID; Baker, Allison R.; Slack, Frank J.; Santisteban, Pilar CSIC ORCID
Fecha de publicación2020
EditorSpringer Nature
CitaciónOncogene 39: 3738–3753 (2020)
ResumenAdenosine deaminases acting on RNA (ADARs) convert adenosine to inosine in double-stranded RNA. A-to-I editing of RNA is a widespread posttranscriptional process that has recently emerged as an important mechanism in cancer biology. A-to-I editing levels are high in several human cancers, including thyroid cancer, but ADAR1 editase-dependent mechanisms governing thyroid cancer progression are unexplored. To address the importance of RNA A-to-I editing in thyroid cancer, we examined the role of ADAR1. Loss-of-function analysis showed that ADAR1 suppression profoundly repressed proliferation, invasion, and migration in thyroid tumor cell models. These observations were validated in an in vivo xenograft model, which showed that ADAR1-silenced cells had a diminished ability to form tumors. RNA editing of miRNAs has the potential to markedly alter target recognition. According to TCGA data, the tumor suppressor miR-200b is overedited in thyroid tumors, and its levels of editing correlate with a worse progression-free survival and disease stage. We confirmed miR-200b overediting in thyroid tumors and we showed that edited miR-200b has weakened activity against its target gene ZEB1 in thyroid cancer cells, likely explaining the reduced aggressiveness of ADAR1-silenced cells. We also found that RAS, but not BRAF, modulates ADAR1 levels, an effect mediated predominantly by PI3K and in part by MAPK. Lastly, pharmacological inhibition of ADAR1 activity with the editing inhibitor 8-azaadenosine reduced cancer cell aggressiveness. Overall, our data implicate ADAR1-mediated A-to-I editing as an important pathway in thyroid cancer progression, and highlight RNA editing as a potential therapeutic target in thyroid cancer.
Versión del editorhttps://doi.org/10.1038/s41388-020-1248-x
URIhttp://hdl.handle.net/10261/222074
DOI10.1038/s41388-020-1248-x
ISSN0950-9232
Aparece en las colecciones: (IIBM) Artículos




Ficheros en este ítem:
Fichero Descripción Tamaño Formato
adaracti.pdf1,92 MBAdobe PDFVista previa
Visualizar/Abrir
Mostrar el registro completo

CORE Recommender

PubMed Central
Citations

34
checked on 13-mar-2024

SCOPUSTM   
Citations

53
checked on 20-mar-2024

WEB OF SCIENCETM
Citations

52
checked on 25-feb-2024

Page view(s)

112
checked on 28-mar-2024

Download(s)

123
checked on 28-mar-2024

Google ScholarTM

Check

Altmetric

Altmetric


Artículos relacionados:


Este item está licenciado bajo una Licencia Creative Commons Creative Commons