Cargando…

Systemic Deficiency of GHR in Pigs leads to Hepatic Steatosis via Negative Regulation of AHR Signaling

Laron syndrome (LS) is an autosomal recessive genetic disease mainly caused by mutations in the human growth hormone receptor (GHR) gene. Previous studies have focused on Ghr mutant mice, but compared with LS patients, Ghr knockout (KO) mice exhibit differential lipid metabolism. To elucidate the re...

Descripción completa

Detalles Bibliográficos
Autores principales: Han, Qi, Chen, Huiling, Wang, Likai, An, Yang, Hu, Xiaoxiang, Zhao, Yaofeng, Zhang, Hao, Zhang, Ran
Formato: Online Artículo Texto
Lenguaje:English
Publicado: Ivyspring International Publisher 2021
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC8579453/
https://www.ncbi.nlm.nih.gov/pubmed/34803486
http://dx.doi.org/10.7150/ijbs.64894
_version_ 1784596432138797056
author Han, Qi
Chen, Huiling
Wang, Likai
An, Yang
Hu, Xiaoxiang
Zhao, Yaofeng
Zhang, Hao
Zhang, Ran
author_facet Han, Qi
Chen, Huiling
Wang, Likai
An, Yang
Hu, Xiaoxiang
Zhao, Yaofeng
Zhang, Hao
Zhang, Ran
author_sort Han, Qi
collection PubMed
description Laron syndrome (LS) is an autosomal recessive genetic disease mainly caused by mutations in the human growth hormone receptor (GHR) gene. Previous studies have focused on Ghr mutant mice, but compared with LS patients, Ghr knockout (KO) mice exhibit differential lipid metabolism. To elucidate the relationship between GHR mutation and lipid metabolism, the role of GHR in lipid metabolism was examined in GHR KO pigs and hepatocytes transfected with siGHR. We observed high levels of free fatty acids and hepatic steatosis in GHR KO pigs, which recapitulates the abnormal lipid metabolism in LS patients. RNAseq analysis revealed that genes related to the fatty acid oxidation pathway were significantly altered in GHR KO pigs. AHR, a transcription factor related to lipid metabolism, was significantly downregulated in GHR KO pigs and siGHR-treated human hepatocytes. We found that AHR directly regulated fatty acid oxidation by directly binding to the promoters of ACOX1 and CPT1A and activating their expression. These data indicate that loss of GHR disturbs the ERK-AHR-ACOX1/CPT1A pathway and consequently leads to hepatic steatosis. Our results established AHR as a modulator of hepatic steatosis, thereby providing a therapeutic target for lipid metabolism disorder.
format Online
Article
Text
id pubmed-8579453
institution National Center for Biotechnology Information
language English
publishDate 2021
publisher Ivyspring International Publisher
record_format MEDLINE/PubMed
spelling pubmed-85794532021-11-19 Systemic Deficiency of GHR in Pigs leads to Hepatic Steatosis via Negative Regulation of AHR Signaling Han, Qi Chen, Huiling Wang, Likai An, Yang Hu, Xiaoxiang Zhao, Yaofeng Zhang, Hao Zhang, Ran Int J Biol Sci Research Paper Laron syndrome (LS) is an autosomal recessive genetic disease mainly caused by mutations in the human growth hormone receptor (GHR) gene. Previous studies have focused on Ghr mutant mice, but compared with LS patients, Ghr knockout (KO) mice exhibit differential lipid metabolism. To elucidate the relationship between GHR mutation and lipid metabolism, the role of GHR in lipid metabolism was examined in GHR KO pigs and hepatocytes transfected with siGHR. We observed high levels of free fatty acids and hepatic steatosis in GHR KO pigs, which recapitulates the abnormal lipid metabolism in LS patients. RNAseq analysis revealed that genes related to the fatty acid oxidation pathway were significantly altered in GHR KO pigs. AHR, a transcription factor related to lipid metabolism, was significantly downregulated in GHR KO pigs and siGHR-treated human hepatocytes. We found that AHR directly regulated fatty acid oxidation by directly binding to the promoters of ACOX1 and CPT1A and activating their expression. These data indicate that loss of GHR disturbs the ERK-AHR-ACOX1/CPT1A pathway and consequently leads to hepatic steatosis. Our results established AHR as a modulator of hepatic steatosis, thereby providing a therapeutic target for lipid metabolism disorder. Ivyspring International Publisher 2021-10-03 /pmc/articles/PMC8579453/ /pubmed/34803486 http://dx.doi.org/10.7150/ijbs.64894 Text en © The author(s) https://creativecommons.org/licenses/by/4.0/This is an open access article distributed under the terms of the Creative Commons Attribution License (https://creativecommons.org/licenses/by/4.0/). See http://ivyspring.com/terms for full terms and conditions.
spellingShingle Research Paper
Han, Qi
Chen, Huiling
Wang, Likai
An, Yang
Hu, Xiaoxiang
Zhao, Yaofeng
Zhang, Hao
Zhang, Ran
Systemic Deficiency of GHR in Pigs leads to Hepatic Steatosis via Negative Regulation of AHR Signaling
title Systemic Deficiency of GHR in Pigs leads to Hepatic Steatosis via Negative Regulation of AHR Signaling
title_full Systemic Deficiency of GHR in Pigs leads to Hepatic Steatosis via Negative Regulation of AHR Signaling
title_fullStr Systemic Deficiency of GHR in Pigs leads to Hepatic Steatosis via Negative Regulation of AHR Signaling
title_full_unstemmed Systemic Deficiency of GHR in Pigs leads to Hepatic Steatosis via Negative Regulation of AHR Signaling
title_short Systemic Deficiency of GHR in Pigs leads to Hepatic Steatosis via Negative Regulation of AHR Signaling
title_sort systemic deficiency of ghr in pigs leads to hepatic steatosis via negative regulation of ahr signaling
topic Research Paper
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC8579453/
https://www.ncbi.nlm.nih.gov/pubmed/34803486
http://dx.doi.org/10.7150/ijbs.64894
work_keys_str_mv AT hanqi systemicdeficiencyofghrinpigsleadstohepaticsteatosisvianegativeregulationofahrsignaling
AT chenhuiling systemicdeficiencyofghrinpigsleadstohepaticsteatosisvianegativeregulationofahrsignaling
AT wanglikai systemicdeficiencyofghrinpigsleadstohepaticsteatosisvianegativeregulationofahrsignaling
AT anyang systemicdeficiencyofghrinpigsleadstohepaticsteatosisvianegativeregulationofahrsignaling
AT huxiaoxiang systemicdeficiencyofghrinpigsleadstohepaticsteatosisvianegativeregulationofahrsignaling
AT zhaoyaofeng systemicdeficiencyofghrinpigsleadstohepaticsteatosisvianegativeregulationofahrsignaling
AT zhanghao systemicdeficiencyofghrinpigsleadstohepaticsteatosisvianegativeregulationofahrsignaling
AT zhangran systemicdeficiencyofghrinpigsleadstohepaticsteatosisvianegativeregulationofahrsignaling