Silencing of c-Fos expression by microRNA-155 is critical for dendritic cell maturation and function

I Dunand-Sauthier, ML Santiago-Raber… - Blood, The Journal …, 2011 - ashpublications.org
I Dunand-Sauthier, ML Santiago-Raber, L Capponi, CE Vejnar, O Schaad, M Irla
Blood, The Journal of the American Society of Hematology, 2011ashpublications.org
MicroRNAs (miRNAs) are small, noncoding RNAs that regulate target mRNAs by binding to
their 3′ untranslated regions. There is growing evidence that microRNA-155 (miR155)
modulates gene expression in various cell types of the immune system and is a prominent
player in the regulation of innate and adaptive immune responses. To define the role of
miR155 in dendritic cells (DCs) we performed a detailed analysis of its expression and
function in human and mouse DCs. A strong increase in miR155 expression was found to be …
Abstract
MicroRNAs (miRNAs) are small, noncoding RNAs that regulate target mRNAs by binding to their 3′ untranslated regions. There is growing evidence that microRNA-155 (miR155) modulates gene expression in various cell types of the immune system and is a prominent player in the regulation of innate and adaptive immune responses. To define the role of miR155 in dendritic cells (DCs) we performed a detailed analysis of its expression and function in human and mouse DCs. A strong increase in miR155 expression was found to be a general and evolutionarily conserved feature associated with the activation of DCs by diverse maturation stimuli in all DC subtypes tested. Analysis of miR155-deficient DCs demonstrated that miR155 induction is required for efficient DC maturation and is critical for the ability of DCs to promote antigen-specific T-cell activation. Expression-profiling studies performed with miR155−/− DCs and DCs overexpressing miR155, combined with functional assays, revealed that the mRNA encoding the transcription factor c-Fos is a direct target of miR155. Finally, all of the phenotypic and functional defects exhibited by miR155−/− DCs could be reproduced by deregulated c-Fos expression. These results indicate that silencing of c-Fos expression by miR155 is a conserved process that is required for DC maturation and function.
ashpublications.org