1. Academic Validation
  2. Trichostatin A modulates the macrophage phenotype by enhancing autophagy to reduce inflammation during polymicrobial sepsis

Trichostatin A modulates the macrophage phenotype by enhancing autophagy to reduce inflammation during polymicrobial sepsis

  • Int Immunopharmacol. 2019 Dec;77:105973. doi: 10.1016/j.intimp.2019.105973.
Shu-Nan Cui 1 Zhao-Yuan Chen 1 Xiao-Bo Yang 1 Lin Chen 2 Yi-Yi Yang 2 Shang-Wen Pan 1 Ya-Xin Wang 1 Ji-Qian Xu 1 Ting Zhou 1 Hai-Rong Xiao 2 Lu Qin 2 Shi-Ying Yuan 3 You Shang 4
Affiliations

Affiliations

  • 1 Department of Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Institute of Anesthesiology and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
  • 2 Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Institute of Anesthesiology and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
  • 3 Department of Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Institute of Anesthesiology and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China. Electronic address: [email protected].
  • 4 Department of Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Institute of Anesthesiology and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China. Electronic address: [email protected].
Abstract

Sepsis is a syndrome of life-threatening organ dysfunction caused by dysregulated host responses to Infection. Macrophage polarization is a key process involved in the pathogenesis of sepsis. Recent evidence has demonstrated that Autophagy participates in the regulation of macrophage polarization in different phases of inflammation. Here, we investigated whether trichostatin A (TSA), a histone deacetylase (HDAC) inhibitor, promotes the macrophage M2 phenotype by enhancing Autophagy to counteract excessive inflammation in a cecal ligation and puncture (CLP) mouse model. TSA stimulation increased the proportions of M2 marker (CD206, CD124 and CD23)-labeled RAW264.7 macrophages. Furthermore, with increasing TSA doses, Autophagy was enhanced gradually. Interestingly, the Autophagy activator rapamycin (Rap), also known as an mTOR Inhibitor, unexpectedly decreased the proportions of M2 marker-labeled macrophages. However, TSA treatment reversed the Rap-induced decreases in CD206-labeled macrophages. Next, we stimulated different groups of RAW264.7 cells with the Autophagy inhibitors MHY1485 or 3-methyladenine (3-MA). Inhibition of Autophagy at any stage in the process suppressed TSA-induced macrophage M2 polarization, but the effect was not associated with mTOR activity. In vivo, TSA administration promoted peritoneal macrophage M2 polarization, increased LC3 II expression, attenuated sepsis-induced organ (lung, liver and kidney) injury, and altered systemic inflammatory cytokine secretion. However, 3-MA abolished the protective effects of TSA in CLP mice and decreased the number of M2 peritoneal macrophages. Therefore, TSA promotes the macrophage M2 phenotype by enhancing Autophagy to reduce systemic inflammation and ultimately improves the survival of mice with polymicrobial sepsis.

Keywords

Autophagy; Inflammation; Macrophage polarization; Sepsis; Trichostatin A.

Figures
Products