Cargando…

N(6)-methyladenosine promotes induction of ADAR1-mediated A-to-I RNA editing to suppress aberrant antiviral innate immune responses

Among over 150 distinct RNA modifications, N(6)-methyladenosine (m(6)A) and adenosine-to-inosine (A-to-I) RNA editing represent 2 of the most studied modifications on mammalian mRNAs. Although both modifications occur on adenosine residues, knowledge on potential functional crosstalk between these 2...

Descripción completa

Detalles Bibliográficos
Autores principales: Terajima, Hideki, Lu, Mijia, Zhang, Linda, Cui, Qi, Shi, Yanhong, Li, Jianrong, He, Chuan
Formato: Online Artículo Texto
Lenguaje:English
Publicado: Public Library of Science 2021
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC8320976/
https://www.ncbi.nlm.nih.gov/pubmed/34324489
http://dx.doi.org/10.1371/journal.pbio.3001292
Descripción
Sumario:Among over 150 distinct RNA modifications, N(6)-methyladenosine (m(6)A) and adenosine-to-inosine (A-to-I) RNA editing represent 2 of the most studied modifications on mammalian mRNAs. Although both modifications occur on adenosine residues, knowledge on potential functional crosstalk between these 2 modifications is still limited. Here, we show that the m(6)A modification promotes expression levels of the ADAR1, which encodes an A-to-I RNA editing enzyme, in response to interferon (IFN) stimulation. We reveal that YTH N(6)-methyladenosine RNA binding protein 1 (YTHDF1) mediates up-regulation of ADAR1; YTHDF1 is a reader protein that can preferentially bind m(6)A-modified transcripts and promote translation. Knockdown of YTHDF1 reduces the overall levels of IFN-induced A-to-I RNA editing, which consequently activates dsRNA-sensing pathway and increases expression of various IFN-stimulated genes. Physiologically, YTHDF1 deficiency inhibits virus replication in cells through regulating IFN responses. The A-to-I RNA editing activity of ADAR1 plays important roles in the YTHDF1-dependent IFN responses. Therefore, we uncover that m(6)A and YTHDF1 affect innate immune responses through modulating the ADAR1-mediated A-to-I RNA editing.