The p53 transcription factor modulates microglia behavior through microRNA-dependent regulation of c-Maf

W Su, S Hopkins, NK Nesser, B Sopher… - The Journal of …, 2014 - journals.aai.org
W Su, S Hopkins, NK Nesser, B Sopher, A Silvestroni, S Ammanuel, S Jayadev, T Möller
The Journal of Immunology, 2014journals.aai.org
Neuroinflammation occurs in acute and chronic CNS injury, including stroke, traumatic brain
injury, and neurodegenerative diseases. Microglia are specialized resident myeloid cells
that mediate CNS innate immune responses. Disease-relevant stimuli, such as reactive
oxygen species (ROS), can influence microglia activation. Previously, we observed that p53,
a ROS-responsive transcription factor, modulates microglia behaviors in vitro and in vivo,
promoting proinflammatory functions and suppressing downregulation of the inflammatory …
Abstract
Neuroinflammation occurs in acute and chronic CNS injury, including stroke, traumatic brain injury, and neurodegenerative diseases. Microglia are specialized resident myeloid cells that mediate CNS innate immune responses. Disease-relevant stimuli, such as reactive oxygen species (ROS), can influence microglia activation. Previously, we observed that p53, a ROS-responsive transcription factor, modulates microglia behaviors in vitro and in vivo, promoting proinflammatory functions and suppressing downregulation of the inflammatory response and tissue repair. In this article we describe a novel mechanism by which p53 modulates the functional differentiation of microglia both in vitro and in vivo. Adult microglia from p53-deficient mice have increased expression of the anti-inflammatory transcription factor c-Maf. To determine how p53 negatively regulates c-Maf, we examined the impact of p53 on known c-Maf regulators. MiR-155 is a microRNA that targets c-Maf. We observed that cytokine-induced expression of miR-155 was suppressed in p53-deficient microglia. Furthermore, Twist2, a transcriptional activator of c-Maf, is increased in p53-deficient microglia. We identified recognition sites in the 3′ untranslated region of Twist2 mRNA that are predicted to interact with two p53-dependent microRNAs: miR-34a and miR-145. In this article, we demonstrate that miR-34a and-145 are regulated by p53 and negatively regulate Twist2 and c-Maf expression in microglia and the RAW macrophage cell line. Taken together, these findings support the hypothesis that p53 activation induced by local ROS or accumulated DNA damage influences microglia functions and that one specific molecular target of p53 in microglia is c-Maf.
journals.aai.org