Cited time in webofscience Cited time in scopus

Liver-Specific Deletion of Mouse CTCF Leads to Hepatic Steatosis via Augmented PPARγ Signaling

Title
Liver-Specific Deletion of Mouse CTCF Leads to Hepatic Steatosis via Augmented PPARγ Signaling
Author(s)
Choi, YeeunSong, Min-JiJung, Woong-JaeJeong, HaengduengPark, SeokjaeYang, BobaeLee, Eun-ChongJoo, Jung-SikChoi, DaheeKoo, Sseung-HoiKim, Eun-KyoungNam, Ki TaekKim, Hyoug-Pyo
Issued Date
2021-01
Citation
Cellular and Molecular Gastroenterology and Hepatology, v.12, no.5, pp.1761 - 1787
Type
Article
Author Keywords
CD36CTCFLiver SteatosisPPARγ
Keywords
IMPROVES INSULIN SENSITIVITYACTIVATED RECEPTOR-GAMMAFATTY-ACIDFUNCTIONAL-ANALYSISLIPID-METABOLISMEXPRESSIONCD36CONTRIBUTESREVEALSBINDING
ISSN
2352-345X
Abstract
Background & Aims: The liver is the major organ for metabolizing lipids, and malfunction of the liver leads to various diseases. Nonalcoholic fatty liver disease is rapidly becoming a major health concern worldwide and is characterized by abnormal retention of excess lipids in the liver. CCCTC-binding factor (CTCF) is a highly conserved zinc finger protein that regulates higher-order chromatin organization and is involved in various gene regulation processes. Here, we sought to determine the physiological role of CTCF in hepatic lipid metabolism. Methods: We generated liver-specific, CTCF-ablated and/or CD36 whole-body knockout mice. Overexpression or knockdown of peroxisome proliferator-activated receptor (PPAR)γ in the liver was achieved using adenovirus. Mice were examined for development of hepatic steatosis and inflammation. RNA sequencing was performed to identify genes affected by CTCF depletion. Genome-wide occupancy of H3K27 acetylation, PPARγ, and CTCF were analyzed by chromatin immunoprecipitation sequencing. Genome-wide chromatin interactions were analyzed by in situ Hi-C. Results: Liver-specific, CTCF-deficient mice developed hepatic steatosis and inflammation when fed a standard chow diet. Global analysis of the transcriptome and enhancer landscape revealed that CTCF-depleted liver showed enhanced accumulation of PPARγ in the nucleus, which leads to increased expression of its downstream target genes, including fat storage-related gene CD36, which is involved in the lipid metabolic process. Hepatic steatosis developed in liver-specific, CTCF-deficient mice was ameliorated by repression of PPARγ via pharmacologic blockade or adenovirus-mediated knockdown, but hardly rescued by additional knockout of CD36. Conclusions: Our data indicate that liver-specific deletion of CTCF leads to hepatosteatosis through augmented PPARγ DNA-binding activity, which up-regulates its downstream target genes associated with the lipid metabolic process. © 2021 The Authors
URI
http://hdl.handle.net/20.500.11750/15748
DOI
10.1016/j.jcmgh.2021.07.016
Publisher
Elsevier Inc.
Related Researcher
  • 김은경 Kim, Eun-Kyoung
  • Research Interests Neural functions in metabolic diseases; 뇌신경세포와 비만; 당뇨 등의 대사 질환 관련 연구
Files in This Item:
2_s2.0_85117379719.pdf

2_s2.0_85117379719.pdf

기타 데이터 / 10.14 MB / Adobe PDF download
Appears in Collections:
Department of Brain Sciences Lab of Neuro-Metabolism & Neurometabolomic Research Center 1. Journal Articles

qrcode

  • twitter
  • facebook
  • mendeley

Items in Repository are protected by copyright, with all rights reserved, unless otherwise indicated.

BROWSE