[HTML][HTML] Coordinate regulation of neutrophil homeostasis by liver X receptors in mice

C Hong, Y Kidani, A Noelia, T Phung… - The Journal of …, 2012 - Am Soc Clin Investig
C Hong, Y Kidani, A Noelia, T Phung, A Ito, X Rong, K Ericson, H Mikkola, SW Beaven
The Journal of clinical investigation, 2012Am Soc Clin Investig
The most abundant immune cell type is the neutrophil, a key first responder after pathogen
invasion. Neutrophil numbers in the periphery are tightly regulated to prevent opportunistic
infections and aberrant inflammation. In healthy individuals, more than 1× 109 neutrophils
per kilogram body weight are released from the bone marrow every 24 hours. To maintain
homeostatic levels, an equivalent number of senescent cells must be cleared from
circulation. Recent studies indicate that clearance of senescent neutrophils by resident …
The most abundant immune cell type is the neutrophil, a key first responder after pathogen invasion. Neutrophil numbers in the periphery are tightly regulated to prevent opportunistic infections and aberrant inflammation. In healthy individuals, more than 1 × 109 neutrophils per kilogram body weight are released from the bone marrow every 24 hours. To maintain homeostatic levels, an equivalent number of senescent cells must be cleared from circulation. Recent studies indicate that clearance of senescent neutrophils by resident tissue macrophages and DCs helps to set homeostatic levels of neutrophils via effects on the IL-23/IL-17/G-CSF cytokine axis, which stimulates neutrophil production in the bone marrow. However, the molecular events in phagocytes underlying this feedback loop have remained indeterminate. Liver X receptors (LXRs) are members of the nuclear receptor superfamily that regulate both lipid metabolic and inflammatory gene expression. Here, we demonstrate that LXRs contribute to the control of neutrophil homeostasis. Using gain- and loss-of-function models, we found that LXR signaling regulated the efficient clearance of senescent neutrophils by peripheral tissue APCs in a Mer-dependent manner. Furthermore, activation of LXR by engulfed neutrophils directly repressed the IL-23/IL-17/G-CSF granulopoietic cytokine cascade. These results provide mechanistic insight into the molecular events orchestrating neutrophil homeostasis and advance our understanding of LXRs as integrators of phagocyte function, lipid metabolism, and cytokine gene expression.
The Journal of Clinical Investigation