[HTML][HTML] Inhibition of LXR controls the polarization of human inflammatory macrophages through upregulation of MAFB

AG de la Aleja, C Herrero, M Torres-Torresano… - Cellular and Molecular …, 2023 - Springer
AG de la Aleja, C Herrero, M Torres-Torresano, MT Schiaffino, A Del Castillo, B Alonso…
Cellular and Molecular Life Sciences, 2023Springer
Monocyte-derived macrophages contribute to pathogenesis in inflammatory diseases and
their effector functions greatly depend on the prevailing extracellular milieu. Whereas M-CSF
primes macrophages for acquisition of an anti-inflammatory profile, GM-CSF drives the
generation of T cell-stimulatory and pro-inflammatory macrophages. Liver X Receptors
(LXRα and LXRβ) are nuclear receptors that control cholesterol metabolism and regulate
differentiation of tissue-resident macrophages. Macrophages from rheumatoid arthritis and …
Abstract
Monocyte-derived macrophages contribute to pathogenesis in inflammatory diseases and their effector functions greatly depend on the prevailing extracellular milieu. Whereas M-CSF primes macrophages for acquisition of an anti-inflammatory profile, GM-CSF drives the generation of T cell-stimulatory and pro-inflammatory macrophages. Liver X Receptors (LXRα and LXRβ) are nuclear receptors that control cholesterol metabolism and regulate differentiation of tissue-resident macrophages. Macrophages from rheumatoid arthritis and other inflammatory pathologies exhibit an enriched LXR pathway, and recent reports have shown that LXR activation raises pro-inflammatory effects and impairs the acquisition of the anti-Inflammatory profile of M-CSF-dependent monocyte-derived macrophages (M-MØ). We now report that LXR inhibition prompts the acquisition of an anti-inflammatory gene and functional profile of macrophages generated within a pathological environment (synovial fluid from Rheumatoid Arthritis patients) as well as during the GM-CSF-dependent differentiation of human monocyte-derived macrophages (GM-MØ). Mechanistically, inhibition of LXR results in macrophages with higher expression of the v-Maf Avian Musculoaponeurotic Fibrosarcoma Oncogene Homolog B (MAFB) transcription factor, which governs the macrophage anti-inflammatory profile, as well as over-expression of MAFB-regulated genes. Indeed, gene silencing experiments on human macrophages evidenced that MAFB is required for the LXR inhibitor to enhance the anti-inflammatory nature of human macrophages. As a whole, our results demonstrate that LXR inhibition prompts the acquisition of an anti-inflammatory transcriptional and functional profile of human macrophages in a MAFB-dependent manner, and propose the use of LXR antagonists as potential therapeutic alternatives in macrophage re-programming strategies during inflammatory responses.
Springer