17β-estradiol promotes TLR4-triggered proinflammatory mediator production through direct estrogen receptor α signaling in macrophages in vivo

B Calippe, V Douin-Echinard, L Delpy… - The Journal of …, 2010 - journals.aai.org
B Calippe, V Douin-Echinard, L Delpy, M Laffargue, K Lélu, A Krust, B Pipy, F Bayard…
The Journal of Immunology, 2010journals.aai.org
Abstract 17β-estradiol (E2) has been shown to promote the expression of inflammatory
mediators by LPS-activated tissue resident macrophages through estrogen receptor α (ERα)
signaling. However, it remained to be determined whether E2 similarly influences
macrophages effector functions under inflammatory conditions in vivo, and whether this
action of E2 resulted from a direct effect on macrophages. We show in this study that chronic
E2 administration to ovariectomized mice significantly increased both cytokine (IL-1β, IL-6 …
Abstract
17β-estradiol (E2) has been shown to promote the expression of inflammatory mediators by LPS-activated tissue resident macrophages through estrogen receptor α (ERα) signaling. However, it remained to be determined whether E2 similarly influences macrophages effector functions under inflammatory conditions in vivo, and whether this action of E2 resulted from a direct effect on macrophages. We show in this study that chronic E2 administration to ovariectomized mice significantly increased both cytokine (IL-1β, IL-6, and TNF-α) and inducible NO synthase mRNA abundance in thioglycolate (TGC)-elicited macrophages. The proinflammatory action of E2 was also evidenced at the level of released IL-1β and IL-6 by ex vivo LPS-activated macrophages. E2 concomitantly inhibited PI3K activity as well as Akt phosphorylation in TGC-elicited macrophages, suggesting that E2 promoted TLR-dependent macrophage activation by alleviating this suppressive signaling pathway. Indeed, this effect was abolished in the presence of the inhibitor wortmannin, demonstrating a key functional link between inhibition of PI3K activity and the E2 action on macrophage functions. Endogenous estrogens levels circulating in ovary-intact mice were sufficient to promote the above described actions. Finally, thanks to a CreLox strategy, targeted disruption of ERα gene in macrophages totally abolished the effect of E2 on the expression of inflammatory mediators by both resident and TGC-elicited peritoneal macrophages. In conclusion, we demonstrate that estrogens, through the activation of ERα in macrophages in vivo, enhance their ability to produce inflammatory mediators and cytokines upon subsequent TLR activation.
journals.aai.org