fbpx

Science

Study Title
Ketone body β-hydroxybutyrate blocks the NLRP3 inflammasome-mediated inflammatory disease
Publication
Natural Medicine
Author(s)

Yun-Hee Youm, Kim Y. Nguyen, Ryan W. Grant, Emily L. Goldberg, Monica Bodogai, Dongin Kim, Dominic D’Agostino, Noah Planavsky, Christopher Lupfer, Thirumala D. Kanneganti, Seokwon Kang, Tamas L. Horvath, Tarek M. Fahmy, Peter A. Crawford, Arya Biragyn, Emad Alnemri, and Vishwa Deep Dixit

Abstract

Ketone bodies , β-hydroxybutyrate (BHB) and acetoacetate support mammalian survival during states of energy deficit by serving as alternative source of ATP1. BHB levels are elevated during starvation, high-intensity exercise or by the low carbohydrate ketogenic diet2. Prolonged caloric restriction or fasting reduces inflammation as immune system adapts to low glucose supply and energy metabolism switches towards mitochondrial fatty acid oxidation, ketogenesis and ketolysis2-6. However, role of ketones bodies in regulation of innate immune response is unknown. We report that BHB, but neither acetoacetate nor structurally-related short chain fatty acids, butyrate and acetate, suppresses activation of the NLRP3 inflammasome in response to several structurally unrelated NLRP3 activators, without impacting NLRC4, AIM2 or non- canonical caspase-11 inflammasome activation. Mechanistically, BHB inhibits NLRP3 inflammasome by preventing K+ efflux and reducing ASC oligomerization and speck formation. The inhibitory effects of BHB on NLRP3 were not dependent on chirality or classical starvation regulated mechanisms like AMPK, reactive oxygen species (ROS), autophagy or glycolytic inhibition. BHB blocked NLRP3 inflammasome without undergoing oxidation in TCA cycle, independently of uncoupling protein-2 (UCP2), Sirt2, receptor Gpr109a and inhibition of NLRP3 did not correlate with magnitude of histone acetylation in macrophages. BHB reduced the NLRP3 inflammasome mediated IL-1β and IL-18 production in human monocytes. In vivo, BHB attenuates caspase-1 activation and IL-1β secretion in mouse models of NLRP3-mediated diseases like Muckle-Wells Syndrome (MWS), Familial Cold Autoinflammatory syndrome (FCAS) and urate crystal induce body cavity inflammation. Taken together, these findings suggest that the anti- inflammatory effects of caloric restriction or ketogenic diets may be mechanistically linked to BHB-mediated inhibition of the NLRP3 inflammasome, and point to the potential use of interventions that elevate circulating BHB against NLRP3-mediated pro-inflammatory diseases.

Date
March 21, 2015
View study

Share This

Related Topics

KetosisKetogenic DietInflammation

Dr. Perlmutter is one of the leading lights in medicine today, illuminating the path for solving chronic illness

Mark Hyman, MD