Abstract

We reported that NAD(+)-dependent SIRT1, RELB, and SIRT6 nuclear proteins in monocytes regulate a switch from the glycolysis-dependent acute inflammatory response to fatty acid oxidation-dependent sepsis adaptation. We also found that disrupting SIRT1 activity during adaptation restores immunometabolic homeostasis and rescues septic mice from death. Here, we show that nuclear SIRT1 guides RELB to differentially induce SIRT3 expression and also increases mitochondrial biogenesis, which alters bioenergetics during sepsis adaptation. We constructed this concept using TLR4-stimulated THP1 human promonocytes, a model that mimics the initiation and adaptation stages of sepsis. Following increased expression, mitochondrial SIRT3 deacetylase activates the rate-limiting tricarboxylic acid cycle (TCA) isocitrate dehydrogenase 2 and superoxide dismutase 2, concomitant with increases in citrate synthase activity. Mitochondrial oxygen consumption rate increases early and decreases during adaptation, parallel with modifications to membrane depolarization, ATP generation, and production of mitochondrial superoxide and whole cell hydrogen peroxide. Evidence of SIRT1-RELB induction of mitochondrial biogenesis included increases in mitochondrial mass, mitochondrial-to-nuclear DNA ratios, and both nuclear and mitochondrial encoded proteins. We confirmed the SIRT-RELB-SIRT3 adaptation link to mitochondrial bioenergetics in both TLR4-stimulated normal and sepsis-adapted human blood monocytes and mouse splenocytes. We also found that SIRT1 inhibition ex vivo reversed the sepsis-induced changes in bioenergetics.

Highlights

  • Nuclear SIRT1 and SIRT6 switch monocyte energy sources from glycolysis to fatty acid oxidation during sepsis adaptation

  • We reported that NAD؉-dependent SIRT1, RELB, and SIRT6 nuclear proteins in monocytes regulate a switch from the glycolysis-dependent acute inflammatory response to fatty acid oxidation-dependent sepsis adaptation

  • We found that sequential activation of SIRT1 and RELB altered mitochondrial bioenergetics during sepsis adaptation by differentially inducing SIRT3 expression, increasing mitochondrial biogenesis, and modifying mitochondrial respiration

Read more

Summary

Background

Nuclear SIRT1 and SIRT6 switch monocyte energy sources from glycolysis to fatty acid oxidation during sepsis adaptation. We reported that NAD؉-dependent SIRT1, RELB, and SIRT6 nuclear proteins in monocytes regulate a switch from the glycolysis-dependent acute inflammatory response to fatty acid oxidation-dependent sepsis adaptation. We show that nuclear SIRT1 guides RELB to differentially induce SIRT3 expression and increases mitochondrial biogenesis, which alters bioenergetics during sepsis adaptation. We constructed this concept using TLR4-stimulated THP1 human promonocytes, a model that mimics the initiation and adaptation stages of sepsis. To adapt to acute inflammation, nuclear SIRT1 guides RELB to generate silent heterochromatin at pro-immune genes like TNF-␣ and IL-1␤ [21] and to activate the euchromatin of genes responsible for fatty acid uptake and oxidation. We observed that SIRT1 inhibition in adapted cells reversed sepsisinduced changes in mitochondrial bioenergetics

EXPERIMENTAL PROCEDURES
RESULTS
DISCUSSION
Full Text
Published version (Free)

Talk to us

Join us for a 30 min session where you can share your feedback and ask us any queries you have

Schedule a call