Elsevier

Journal of Hepatology

Volume 58, Issue 5, May 2013, Pages 1000-1006
Journal of Hepatology

Research Article
Post-transcriptional activation of PPAR alpha by KLF6 in hepatic steatosis

https://doi.org/10.1016/j.jhep.2013.01.020Get rights and content

Background & Aims

Dysregulated glucose homeostasis and lipid accumulation characterize non-alcoholic fatty liver disease (NAFLD), but underlying mechanisms are obscure. We report here that Krüppel-like factor 6 (KLF6), a ubiquitous transcription factor that promotes adipocyte differentiation, also provokes the metabolic abnormalities of NAFLD by post-transcriptionally activating PPARα-signaling.

Methods

Mice with either hepatocyte-specific depletion of KLF6 (‘ΔHepKlf6’) or global KLF6 heterozygosity (Klf6+/−) were fed a high fat diet (HFD) or chow for 8 or 16 weeks. Glucose and insulin tolerance tests were performed to assess insulin sensitivity. Overexpression and knockdown of KLF6 in cultured cells enabled the elucidation of underlying mechanisms. In liver samples from a cohort of 28 NAFLD patients, the expression of KLF6-related target genes was quantified.

Results

Mice with global- or hepatocyte-depletion of KLF6 have reduced body fat content and improved glucose and insulin tolerance, and are protected from HFD-induced steatosis. In hepatocytes, KLF6 deficiency reduces PPARα-regulated genes (Trb3, Pepck) with diminished PPARα protein but no change in Pparα mRNA, which is explained by the discovery that KLF6 represses miRNA 10b, which leads to induction of PPARα. In NAFLD patients with advanced disease and inflammation, the expression of miRNA 10b is significantly downregulated, while PEPCK mRNA is upregulated; KLF6 mRNA expression also correlates with TRB3 as well as PEPCK gene expression.

Conclusions

KLF6 increases PPARα activity, whereas KLF6 loss leads to PPARα repression and attenuation of lipid and glucose abnormalities associated with a high fat diet. The findings establish KLF6 as a novel regulator of hepatic glucose and lipid metabolism in fatty liver.

Introduction

Non-alcoholic fatty liver disease (NAFLD) prevalence in the US and Europe is rapidly rising. NAFLD is characterized by hepatic steatosis, which can progress to non-alcoholic steatohepatitis (NASH). Subsequent progression from NASH to cirrhosis may precipitate liver failure and hepatocellular carcinoma (HCC) [1]. The mechanisms underlying the pathogenesis of NAFLD and its progression to NASH are poorly understood.

Krüppel-like factor 6 (KLF6) is a ubiquitously expressed, multifunctional transcription factor and tumor suppressor gene with emerging roles in glucose and lipid homeostasis [2], [3], [4]. KLF6 promotes adipogenesis in preadipocytes and fibroblasts, and interferes with peroxisome proliferator activated receptor γ (PPARγ) signaling [5], [6]. A functional polymorphism in the KLF6 gene that increases the generation of antagonistic KLF6 alternative splice forms protects patients from advanced NAFLD [7]. KLF6 directly transactivates hepatic glucokinase, and modulates hepatic insulin sensitivity in NAFLD [8].

Insulin resistance (IR) is a sentinel feature of the metabolic syndrome and is associated with to NASH progression. IR is mediated in part by the actions of peroxisome proliferator activated receptor alpha (PPARα), a xenobiotic and lipid sensor that regulates hepatic steatosis, lipoprotein synthesis, and hepatic gluconeogenesis. PPARα is activated by excess fatty acids (FA), thereby inducing expression of downstream genes that metabolize FA [9], including fatty acid transport proteins [10].

Effects of PPARα activation on IR are multifaceted and incompletely understood. While fibrates, which act as PPARα ligands, improve IR, the PPARα co-activator peroxisome proliferator-activated receptor gamma co-activator 1-α (PGC1α) has a paradoxical effect on IR, since PGC1α knockout mice are protected from high fat diet-induced steatosis and IR [11]. A similar observation was made for farnesoid X receptor (FXR), which also induces PPARα signaling, but FXR agonists increase hepatic gluconeogenesis via PPARα-mediated phosphoenolpyruvate carboxykinase (PEPCK) upregulation and therefore worsen hepatic IR [12]. In contrast, micro-RNA 10b (miRNA 10b) opposes the actions of PPARα signaling by repressing PPARα protein expression [13], [14]. Downregulation of the PPARα target tribbles homologue 3 (TRB3) improves IR via increased AKT phosphorylation and PPARγ activation [15], [16].

Here, we describe a functional link between KLF6 expression and hepatic PPARα signaling, in part via miRNA 10b repression. We have uncovered this link by characterizing two mouse models, one with global KLF6 heterozygosity, the second with hepatocyte KLF6 deficiency, as well as by quantifying KLF6 mRNA in a cohort of 28 NAFLD patients. Together these data reveal a regulatory role of KLF6 in insulin signaling and lipid metabolism.

Section snippets

Mouse models

Mice with a floxed KLF6 targeting vector (C57BL/6;129Sv, Genentech, San Francisco, CA [17]) were crossed with mice expressing Cre under control of the albumin promoter (B6.Cg-Tg(Alb-cre)21Mgn/J; Jackson Labs, Bar Harbor, Maine). After further backcrossing, male offspring, expressing Cre with two floxed KLF6 alleles, was used as the experimental group, while mice with two floxed alleles and no Cre expression were used as controls, as previously described [8], [18]. Male Klf6+/− mice (C57BL6)

Loss of hepatocyte KLF6 protects mice from diet-induced steatohepatitis and improves insulin response

To assess the potential impact of KLF6 on hepatic lipid and glucose homeostasis in vivo, we generated mice with hepatocyte-specific knockdown of KLF6, using a Cre-lox strategy, in which mice expressing Cre driven by the albumin promoter were crossed with animals containing two floxed KLF6 alleles, and further backcrossed into the C57BL/6 background [8], [17]. These albumin-Cre X KLF6LoxP/LoxP mice (termed ‘ΔHepKlf6 mice’) had a normal phenotype when fed standard chow, with body and liver

Discussion

Our findings substantially clarify clinical findings implicating KLF6 in the pathogenesis of NAFLD [7], [8], by demonstrating in cultured cells, mouse models, and human samples, that KLF6 mediates glucose tolerance and insulin responses in fatty liver disease. This phenotype is accompanied by KLF6-mediated induction of PPARα signaling and its downstream target Trb3. Furthermore, in ΔHepKlf6 mice, HFD-induced hepatic steatohepatitis is attenuated compared to controls, which is associated with

Financial support

Supported by grants from IFORES program of the University of Essen and the European Association for the Study of the Liver (EASL) Sheila Sherlock Short-Term Fellowship (L.P.B.); Swiss National Fund (D.V.); The European Union Seventh Framework Programme (FP7/2007-2013) under grant agreement Health-F2-2009-241762, for the project FLIP (H.L.R., G.L.P.); National Institutes of Health (NIH): DK37340, DK56621 and AA017067 (S.L.F.); Funding Program for World-Leading Innovative R&D on Science and

Conflict of interest

The authors who have taken part in this study declared that they do not have anything to disclose regarding funding or conflict of interest with respect to this manuscript.

The underlying research reported in the study was funded by the NIH Institutes of Health.

References (36)

  • G. Narla et al.

    KLF6, a candidate tumor suppressor gene mutated in prostate cancer

    Science

    (2001)
  • T. Suzuki et al.

    Isolation and initial characterization of GBF, a novel DNA-binding zinc finger protein that binds to the GC-rich binding sites of the HIV-1 promoter

    J Biochem

    (1998)
  • L. Miele et al.

    The Kruppel-like factor 6 genotype is associated with fibrosis in nonalcoholic fatty liver disease

    Gastroenterology

    (2008)
  • L.P. Bechmann et al.

    Glucokinase links Kruppel-like factor 6 to the regulation of hepatic insulin sensitivity in nonalcoholic fatty liver disease

    Hepatology

    (2012)
  • P.D. Berk

    Regulatable fatty acid transport mechanisms are central to the pathophysiology of obesity, fatty liver, and metabolic syndrome

    Hepatology

    (2008)
  • D.R. Cha et al.

    Peroxisome proliferator-activated receptor-alpha deficiency protects aged mice from insulin resistance induced by high-fat diet

    Am J Nephrol

    (2007)
  • K.R. Stayrook et al.

    Regulation of carbohydrate metabolism by the farnesoid X receptor

    Endocrinology

    (2005)
  • L. Zheng et al.

    Effect of miRNA-10b in regulating cellular steatosis level by targeting PPAR-alpha expression, a novel mechanism for the pathogenesis of NAFLD

    J Gastroenterol Hepatol

    (2010)
  • Cited by (0)

    View full text