KLF10 integrates circadian timing and sugar signaling to coordinate hepatic metabolism

Curation statements for this article:
  • Curated by eLife

    eLife logo

    Evaluation Summary:

    This paper will be of interest in the fields of circadian biology and metabolic physiology. It provides a molecular mechanism for protection against development of fatty liver in response to high sugar consumption. Quality data support the key claims of the paper in each of the main research areas (circadian biology and metabolism) but additional efforts are needed to integrate the two parts. The current study does not thoroughly connect the in vitro and in vivo findings and misses the opportunity to fully characterize the role of KLF10 in circadian regulation in response to excessive sugar consumption.

    (This preprint has been reviewed by eLife. We include the public reviews from the reviewers here; the authors also receive private feedback with suggested changes to the manuscript. Reviewer #3 agreed to share their name with the authors.)

This article has been Reviewed by the following groups

Read the full article See related articles

Abstract

The mammalian circadian timing system and metabolism are highly interconnected, and disruption of this coupling is associated with negative health outcomes. Krüppel-like factors (KLFs) are transcription factors that govern metabolic homeostasis in various organs. Many KLFs show a circadian expression in the liver. Here, we show that the loss of the clock-controlled KLF10 in hepatocytes results in extensive reprogramming of the mouse liver circadian transcriptome, which in turn alters the temporal coordination of pathways associated with energy metabolism. We also show that glucose and fructose induce Klf10, which helps mitigate glucose intolerance and hepatic steatosis in mice challenged with a sugar beverage. Functional genomics further reveal that KLF10 target genes are primarily involved in central carbon metabolism. Together, these findings show that in the liver KLF10 integrates circadian timing and sugar metabolism-related signaling, and serves as a transcriptional brake that protects against the deleterious effects of increased sugar consumption.

Article activity feed

  1. Reviewer #3 (Public Review):

    In "KLF10 integrates circadian timing and sugar signaling to coordinate hepatic metabolism", Anthony Ruberto and colleagues characterize the role of the transcription factor KLF10 in circadian transcription and the transcriptional and physiological responses to hexose sugars in mouse hepatocytes. They confirm earlier reports that Klf10 is expressed rhythmically in mouse liver, with peak expression at ZT9. They show that Klf10 expression is induced by glucose and fructose and that hepatocyte-specific deletion of Klf10 exacerbates hyperglycemic and hepatosteatotic responses to 8 weeks of elevated sugar consumption. They use RNA sequencing and ChIP sequencing to define the complement of Klf10 target genes in hepatocytes and how they are regulated by glucose and fructose. Together their data support a model in which KLF10 limits the transcriptional induction of rate-limiting enzymes involved in gluconeogenesis and lipogenesis in response to elevated sugar consumption, thus mitigating the pathophysiological impact of high sugar diets. The experiments are mostly well designed, presented, and interpreted but several points require additional investigation and/or clarification. While the current manuscript suggests an integration of circadian timing and sugar signaling by KLF10, additional experiments to establish how some of the molecular and physiological effects are modulated by time of day are needed to better support that claim.

    Strengths:

    This study uses a combination of genetic, biochemical, and physiological approaches to investigate the hepatocyte-specific function of the transcription factor KLF10. Deletion of KLF10 specifically in hepatocytes distinguishes this study from other related work. Further, the characterization of global daily gene expression patterns in mouse liver is well designed and analyzed and establishes that hepatocyte-specific deletion of Klf10 remodels daily rhythms of gene expression in the liver. The combination of that analysis with ChIP sequencing provides powerful evidence to establish the hepatocyte-specific KLF10-dependent transcriptome and highlights its targeting of rate-limiting enzymes in lipogenic pathways. Together, the molecular and physiological analyses in this study provide compelling evidence that KLF10 plays a protective role in the context of excessive sugar consumption by limiting lipogenic gene expression pathways and thereby suppressing hepatic steatosis.

    Weaknesses:

    In its present form, this study does not thoroughly connect the in vitro and in vivo findings and misses the opportunity to fully characterize the role of KLF10 in circadian regulation of lipogenesis in response to excessive sugar consumption in vivo. It is unclear whether the concentrations of glucose and fructose used to stimulate primary hepatocytes are similar to those experienced in response to the dietary stimulus in vivo and there is no examination of the impact of sucrose on Klf10 expression or downstream gene expression. This omission complicates the interpretation of the response to the combined sugar stimulus in vivo, especially in light of a recent report that KLF10 deletion protects against hepatosteatosis caused by consumption of a high sucrose diet. It also does not examine how time of day influences KLF10-dependent gene regulation in response to sugar consumption. Without these analyses, it falls short of connecting the circadian and sugar-response pathways through KLF10.

  2. Reviewer #2 (Public Review):

    This study builds on a previously published paper from this group showing that KLF10 is under circadian control, and it in turn affects the oscillation of a set of metabolic genes in the liver. While the previous study utilized a systemic Klf10 KO mouse model, here, Ruberto et al. generated a conditional hepatocyte-specific Klf10 KO mouse model (Klf10Δhep).

    The authors find that the absence of hepatocyte KLF10 alters the circadian oscillation of a number of metabolic genes. In response to sugar consumption, Klf10Dhep mice demonstrate exacerbated adverse effects as well as significantly increased hepatic expression of many glycolysis, gluconeogenesis, and lipogenesis related genes. They conclude that Klf10 normally acts as a "transcriptional brake" to protect animals against the effects of high sugar consumption and show via ChIP-seq that KLF10 is present at a wide range of metabolic genes, particularly at those involved in acetyl-CoA metabolism. The findings are interesting, particularly in the context of the burgeoning burden of metabolic disease and its relation to high sugar consumption, and are supported by the experimental findings.

  3. Reviewer #1 (Public Review):

    Ruberto et al. utilize hepatocyte-specific Klf10 knock-out mice to demonstrate expression changes of rhythmic transcripts, highlighting dysregulated glucose and lipid metabolism as an enriched gene set. They demonstrate that KLF10 is necessary for proper glycemic control in mice and that KLF10 coordinates suppression of metabolic gene expression in the liver in response to high sugar diet. The authors corroborate their findings by analyzing gene expression changes of primary hepatocytes stimulated with fructose and high glucose. Finally, the authors identify KLF10 target genes using ChIP-seq and validate Acss2 and Acacb as target genes suppressed in mice following a high sugar diet. Novel aspects of this work include the metabolic characterization of a hepatocyte-specific Klf10 knock-out mouse, identification of KLF10 target genes in hepatocytes using ChIP-seq, and description of circadian transcript expression with Klf10 loss.

  4. Evaluation Summary:

    This paper will be of interest in the fields of circadian biology and metabolic physiology. It provides a molecular mechanism for protection against development of fatty liver in response to high sugar consumption. Quality data support the key claims of the paper in each of the main research areas (circadian biology and metabolism) but additional efforts are needed to integrate the two parts. The current study does not thoroughly connect the in vitro and in vivo findings and misses the opportunity to fully characterize the role of KLF10 in circadian regulation in response to excessive sugar consumption.

    (This preprint has been reviewed by eLife. We include the public reviews from the reviewers here; the authors also receive private feedback with suggested changes to the manuscript. Reviewer #3 agreed to share their name with the authors.)