4.6 Article

Lung Myofibroblasts Promote Macrophage Profibrotic Activity through Lactate-induced Histone Lactylation

出版社

AMER THORACIC SOC
DOI: 10.1165/rcmb.2020-0360OC

关键词

pulmonary fibrosis; myofibroblast; macrophage; lactate; histone lactylation

资金

  1. U.S. National Institutes of Health [HL135830]
  2. Department of Defense [W81XWH-20-1-0226]

向作者/读者索取更多资源

This study demonstrates that in lung fibrosis, myofibroblasts enhance glycolysis to produce lactate, which regulates the pathogenic phenotype of alveolar macrophages. Lactate induces profibrotic gene expression in macrophages, promoting the fibrotic process.
Augmented glycolysis due to metabolic reprogramming in lung myofibroblasts is critical to their profibrotic phenotype. The primary glycolysis byproduct, lactate, is also secreted into the extracellular milieu, together with which myofibroblasts and macrophages form a spatially restricted site usually described as fibrotic niche. Therefore, we hypothesized that myofibroblast glycolysis might have a non-cell autonomous effect through lactate regulating the pathogenic phenotype of alveolar macrophages. Here, we demonstrated that there was a markedly increased lactate in the conditioned media of TGF-beta 1 (transforming growth factor-beta 1)-induced lung myofibroblasts and in the BAL fluids (BALFs) from mice with TGF-beta 1- or bleomycin-induced lung fibrosis. Importantly, the media and BALFs promoted profibrotic mediator expression in macrophages. Mechanistically, lactate induced histone lactylation in the promoters of the profibrotic genes in macrophages, consistent with the upregulation of this epigenetic modification in these cells in the fibrotic lungs. The lactate inductions of the histone lactylation and profibrotic gene expression were mediated by p300, as evidenced by their diminished concentrations in p300-knockdown macrophages. Collectively, our study establishes that in addition to protein, lipid, and nucleic acid molecules, a metabolite can also mediate intercellular regulations in the setting of lung fibrosis. Our findings shed new light on the mechanism underlying the key contribution of myofibroblast glycolysis to the pathogenesis of lung fibrosis.

作者

我是这篇论文的作者
点击您的名字以认领此论文并将其添加到您的个人资料中。

评论

主要评分

4.6
评分不足

次要评分

新颖性
-
重要性
-
科学严谨性
-
评价这篇论文

推荐

暂无数据
暂无数据