1. Academic Validation
  2. Repositioning of the β-Blocker Carvedilol as a Novel Autophagy Inducer That Inhibits the NLRP3 Inflammasome

Repositioning of the β-Blocker Carvedilol as a Novel Autophagy Inducer That Inhibits the NLRP3 Inflammasome

  • Front Immunol. 2018 Aug 22;9:1920. doi: 10.3389/fimmu.2018.01920.
Wei-Ting Wong 1 Lan-Hui Li 2 Yerra Koteswara Rao 3 Shih-Ping Yang 4 Shu-Meng Cheng 4 Wen-Yu Lin 4 Cheng-Chung Cheng 4 Ann Chen 5 Kuo-Feng Hua 3 5 6
Affiliations

Affiliations

  • 1 National Defense Medical Center, Graduate Institute of Life Sciences, Taipei, Taiwan.
  • 2 Department of Laboratory Medicine, Linsen, Chinese Medicine and Kunming Branch, Taipei City Hospital, Taipei, Taiwan.
  • 3 Department of Biotechnology and Animal Science, National Ilan University, Yilan City, Taiwan.
  • 4 Division of Cardiology, Department of Internal Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan.
  • 5 Department of Pathology, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan.
  • 6 Department of Medical Research, China Medical University Hospital, Taichung, Taiwan.
Abstract

The NLRP3 inflammasome is a multiprotein complex that plays a key role in the innate immune system, and aberrant activation of this complex is involved in the pathogenesis of inflammatory diseases. Carvedilol (CVL) is an α-, β-blocker used to treat high blood pressure and congestive heart failure; however, some benefits beyond decreased blood pressure were observed clinically, suggesting the potential anti-inflammatory activity of CVL. In this report, the inhibitory potential of CVL toward the NLRP3 inflammasome and the possible underlying molecular mechanisms were studied. Our results showed that CVL attenuated NLRP3 inflammasome activation and Pyroptosis in mouse macrophages, without affecting activation of the AIM2, NLRC4 and non-canonical inflammasomes. Mechanistic analysis revealed that CVL prevented lysosomal and mitochondrial damage and reduced ASC oligomerization. Additionally, CVL caused autophagic induction through a Sirt1-dependent pathway, which inhibited the NLRP3 inflammasome. In the in vivo mouse model of NLRP3-associated peritonitis, oral administration of CVL reduced (1) peritoneal recruitment of neutrophils; (2) the levels of IL-1β, IL-18, active Caspase-1, ASC, IL-6, TNF-α, MCP-1, and CXCL1 in the lavage fluids; and (3) the levels of NLRP3 and HO-1 in the peritoneal cells. Our results indicated that CVL is a novel Autophagy inducer that inhibits the NLRP3 inflammasome and can be repositioned for ameliorating NLRP3-associated complications.

Keywords

Carvedilol; NLRP3 inflammasome; autophagy; mitochondria; peritonitis.

Figures