Induction of IL-4Rα–dependent microRNAs identifies PI3K/Akt signaling as essential for IL-4–driven murine macrophage proliferation in vivo

D Rückerl, SJ Jenkins, NN Laqtom… - Blood, The Journal …, 2012 - ashpublications.org
D Rückerl, SJ Jenkins, NN Laqtom, IJ Gallagher, TE Sutherland, S Duncan, AH Buck…
Blood, The Journal of the American Society of Hematology, 2012ashpublications.org
Macrophage (MΦ) activation must be tightly controlled to preclude overzealous responses
that cause self-damage. MicroRNAs promote classical MΦ activation by blocking
antiinflammatory signals and transcription factors but also can prevent excessive TLR
signaling. In contrast, the microRNA profile associated with alternatively activated MΦ and
their role in regulating wound healing or antihelminthic responses has not been described.
By using an in vivo model of alternative activation in which adult Brugia malayi nematodes …
Abstract
Macrophage (MΦ) activation must be tightly controlled to preclude overzealous responses that cause self-damage. MicroRNAs promote classical MΦ activation by blocking antiinflammatory signals and transcription factors but also can prevent excessive TLR signaling. In contrast, the microRNA profile associated with alternatively activated MΦ and their role in regulating wound healing or antihelminthic responses has not been described. By using an in vivo model of alternative activation in which adult Brugia malayi nematodes are implanted surgically in the peritoneal cavity of mice, we identified differential expression of miR-125b-5p, miR-146a-5p, miR-199b-5p, and miR-378-3p in helminth-induced MΦ. In vitro experiments demonstrated that miR-378-3p was specifically induced by IL-4 and revealed the IL-4–receptor/PI3K/Akt-signaling pathway as a target. Chemical inhibition of this pathway showed that intact Akt signaling is an important enhancement factor for alternative activation in vitro and in vivo and is essential for IL-4–driven MΦ proliferation in vivo. Thus, identification of miR-378-3p as an IL-4Rα–induced microRNA led to the discovery that Akt regulates the newly discovered mechanism of IL-4–driven macrophage proliferation. Together, the data suggest that negative regulation of Akt signaling via microRNAs might play a central role in limiting MΦ expansion and alternative activation during type 2 inflammatory settings.
ashpublications.org