4.6 Article

Ketone Bodies Rescue Mitochondrial Dysfunction Via Epigenetic Remodeling

Journal

JACC-BASIC TO TRANSLATIONAL SCIENCE
Volume 8, Issue 9, Pages 1123-1137

Publisher

ELSEVIER SCIENCE INC
DOI: 10.1016/j.jacbts.2023.03.014

Keywords

BHB; heart failure; histone methylation; ischemia; mitochondria; myocardial infarction; beta-hydroxybutyrate

Ask authors/readers for more resources

This study conducted comprehensive in vivo and in vitro experiments to investigate the molecular processes underlying ischemic cardiac disease. The findings revealed a specific pattern of chromatin remodeling induced by ischemic injury, which led to the transcriptional repression of a key regulator of mitochondrial function. Additionally, the study discovered a novel pathway linking metabolism to gene expression, demonstrating the role of ketone bodies in modulating chromatin remodeling and mitochondrial dysfunction.
Ischemic cardiac disease is a major cause of mortality worldwide. However, the exact molecular processes underlying this disorder are not fully known. This study includes a comprehensive and coordinated set of in vivo and in vitro experiments using human cardiac specimens from patients with postischemic heart failure (HF) and healthy control subjects, a murine model of HF, and cellular systems. These approaches identified for the first time a specific pattern of maladaptive chromatin remodeling, namely a double methylation of histone 3 at lysine 27 and a single methylation at lysine 36 (H3_K27me2K36me1) consistently induced by ischemic injury in all these settings: human HF; murine HF; and in vitro models. Mechanistically, this work demonstrates that this histone modification mediates the ischemia-induced transcriptional repression of PPARG coactivator 1a (PGC1a), master regulator of mitochondrial function and biogenesis. Intriguingly, both the augmented H3_K27me2K36me1 and the mitochondrial dysfunction ensued by PGC1a down-regulation were significantly attenuated by the treatment with b-hydroxybutyrate, the most abundant ketone body in humans, revealing a novel pathway coupling metabolism to gene expression. Taken together, these findings establish maladaptive chromatin remodeling as a key mechanism in postischemic heart injury, functionally modulated by ketone bodies. (J Am Coll Cardiol Basic Trans Science 2023;8:1123-1137) (c) 2023 The Authors. Published by Elsevier on behalf of the American College of Cardiology Foundation. This is an open access article under the CC BY-NC-ND license (http://creativecommons.org/licenses/by-nc-nd/4.0/).

Authors

I am an author on this paper
Click your name to claim this paper and add it to your profile.

Reviews

Primary Rating

4.6
Not enough ratings

Secondary Ratings

Novelty
-
Significance
-
Scientific rigor
-
Rate this paper

Recommended

No Data Available
No Data Available