Menin orchestrates hepatic glucose and fatty acid uptake via deploying the cellular translocation of SIRT1 and PPARγ

Abstract Background Menin is a scaffold protein encoded by the Men1 gene, which interacts with various transcriptional proteins to activate or repress cellular processes and is a key mediator in multiple organs. Both liver-specific and hepatocyte-specific Menin deficiency promotes high-fat diet-indu...

Full description

Bibliographic Details
Main Authors: Tingjun Liu, Ranran Li, Lili Sun, Zhongjin Xu, Shengxuan Wang, Jingxuan Zhou, Xuanning Wu, Kerong Shi
Format: Article
Language:English
Published: BMC 2023-09-01
Series:Cell & Bioscience
Subjects:
Online Access:https://doi.org/10.1186/s13578-023-01119-y
_version_ 1797556186814349312
author Tingjun Liu
Ranran Li
Lili Sun
Zhongjin Xu
Shengxuan Wang
Jingxuan Zhou
Xuanning Wu
Kerong Shi
author_facet Tingjun Liu
Ranran Li
Lili Sun
Zhongjin Xu
Shengxuan Wang
Jingxuan Zhou
Xuanning Wu
Kerong Shi
author_sort Tingjun Liu
collection DOAJ
description Abstract Background Menin is a scaffold protein encoded by the Men1 gene, which interacts with various transcriptional proteins to activate or repress cellular processes and is a key mediator in multiple organs. Both liver-specific and hepatocyte-specific Menin deficiency promotes high-fat diet-induced liver steatosis in mice, as well as insulin resistance and type 2 diabetic phenotype. The potential link between Menin and hepatic metabolism homeostasis may provide new insights into the mechanism of fatty liver disease. Results Disturbance of hepatic Menin expression impacts metabolic pathways associated with non-alcoholic fatty liver disease (NAFLD), including the FoxO signaling pathway, which is similar to that observed in both oleic acid-induced fatty hepatocytes model and biopsied fatty liver tissues, but with elevated hepatic Menin expression and inhibited FABP1. Higher levels of Menin facilitate glucose uptake while restraining fatty acid uptake. Menin targets the expression of FABP3/4/5 and also CD36 or GK, PCK by binding to their promoter regions, while recruiting and deploying the cellular localization of PPARγ and SIRT1 in the nucleus and cytoplasm. Accordingly, Menin binds to PPARγ and/or FoxO1 in hepatocytes, and orchestrates hepatic glucose and fatty acid uptake by recruiting SIRT1. Conclusion Menin plays an orchestration role as a transcriptional activator and/or repressor to target downstream gene expression levels involved in hepatic energy uptake by interacting with the cellular energy sensor SIRT1, PPARγ, and/or FoxO1 and deploying their translocations between the cytoplasm and nucleus, thereby maintaining metabolic homeostasis. These findings provide more evidence suggesting Menin could be targeted for the treatment of hepatic steatosis, NAFLD or metabolic dysfunction-associated fatty liver disease (MAFLD), and even other hepatic diseases. Graphical Abstract
first_indexed 2024-03-10T16:58:20Z
format Article
id doaj.art-c625636974bf433f984f89ce5944e3cb
institution Directory Open Access Journal
issn 2045-3701
language English
last_indexed 2024-03-10T16:58:20Z
publishDate 2023-09-01
publisher BMC
record_format Article
series Cell & Bioscience
spelling doaj.art-c625636974bf433f984f89ce5944e3cb2023-11-20T11:03:59ZengBMCCell & Bioscience2045-37012023-09-0113112010.1186/s13578-023-01119-yMenin orchestrates hepatic glucose and fatty acid uptake via deploying the cellular translocation of SIRT1 and PPARγTingjun Liu0Ranran Li1Lili Sun2Zhongjin Xu3Shengxuan Wang4Jingxuan Zhou5Xuanning Wu6Kerong Shi7Laboratory of Animal Stem Cell and Reprogramming, College of Animal Science and Technology, Shandong Agricultural UniversityLaboratory of Animal Stem Cell and Reprogramming, College of Animal Science and Technology, Shandong Agricultural UniversityLaboratory of Animal Stem Cell and Reprogramming, College of Animal Science and Technology, Shandong Agricultural UniversityLaboratory of Animal Stem Cell and Reprogramming, College of Animal Science and Technology, Shandong Agricultural UniversityLaboratory of Animal Stem Cell and Reprogramming, College of Animal Science and Technology, Shandong Agricultural UniversityLaboratory of Animal Stem Cell and Reprogramming, College of Animal Science and Technology, Shandong Agricultural UniversityLaboratory of Animal Stem Cell and Reprogramming, College of Animal Science and Technology, Shandong Agricultural UniversityLaboratory of Animal Stem Cell and Reprogramming, College of Animal Science and Technology, Shandong Agricultural UniversityAbstract Background Menin is a scaffold protein encoded by the Men1 gene, which interacts with various transcriptional proteins to activate or repress cellular processes and is a key mediator in multiple organs. Both liver-specific and hepatocyte-specific Menin deficiency promotes high-fat diet-induced liver steatosis in mice, as well as insulin resistance and type 2 diabetic phenotype. The potential link between Menin and hepatic metabolism homeostasis may provide new insights into the mechanism of fatty liver disease. Results Disturbance of hepatic Menin expression impacts metabolic pathways associated with non-alcoholic fatty liver disease (NAFLD), including the FoxO signaling pathway, which is similar to that observed in both oleic acid-induced fatty hepatocytes model and biopsied fatty liver tissues, but with elevated hepatic Menin expression and inhibited FABP1. Higher levels of Menin facilitate glucose uptake while restraining fatty acid uptake. Menin targets the expression of FABP3/4/5 and also CD36 or GK, PCK by binding to their promoter regions, while recruiting and deploying the cellular localization of PPARγ and SIRT1 in the nucleus and cytoplasm. Accordingly, Menin binds to PPARγ and/or FoxO1 in hepatocytes, and orchestrates hepatic glucose and fatty acid uptake by recruiting SIRT1. Conclusion Menin plays an orchestration role as a transcriptional activator and/or repressor to target downstream gene expression levels involved in hepatic energy uptake by interacting with the cellular energy sensor SIRT1, PPARγ, and/or FoxO1 and deploying their translocations between the cytoplasm and nucleus, thereby maintaining metabolic homeostasis. These findings provide more evidence suggesting Menin could be targeted for the treatment of hepatic steatosis, NAFLD or metabolic dysfunction-associated fatty liver disease (MAFLD), and even other hepatic diseases. Graphical Abstracthttps://doi.org/10.1186/s13578-023-01119-yMeninHepatocytesPPARγSIRT1NAFLDMetabolism homeostatsis
spellingShingle Tingjun Liu
Ranran Li
Lili Sun
Zhongjin Xu
Shengxuan Wang
Jingxuan Zhou
Xuanning Wu
Kerong Shi
Menin orchestrates hepatic glucose and fatty acid uptake via deploying the cellular translocation of SIRT1 and PPARγ
Cell & Bioscience
Menin
Hepatocytes
PPARγ
SIRT1
NAFLD
Metabolism homeostatsis
title Menin orchestrates hepatic glucose and fatty acid uptake via deploying the cellular translocation of SIRT1 and PPARγ
title_full Menin orchestrates hepatic glucose and fatty acid uptake via deploying the cellular translocation of SIRT1 and PPARγ
title_fullStr Menin orchestrates hepatic glucose and fatty acid uptake via deploying the cellular translocation of SIRT1 and PPARγ
title_full_unstemmed Menin orchestrates hepatic glucose and fatty acid uptake via deploying the cellular translocation of SIRT1 and PPARγ
title_short Menin orchestrates hepatic glucose and fatty acid uptake via deploying the cellular translocation of SIRT1 and PPARγ
title_sort menin orchestrates hepatic glucose and fatty acid uptake via deploying the cellular translocation of sirt1 and pparγ
topic Menin
Hepatocytes
PPARγ
SIRT1
NAFLD
Metabolism homeostatsis
url https://doi.org/10.1186/s13578-023-01119-y
work_keys_str_mv AT tingjunliu meninorchestrateshepaticglucoseandfattyaciduptakeviadeployingthecellulartranslocationofsirt1andpparg
AT ranranli meninorchestrateshepaticglucoseandfattyaciduptakeviadeployingthecellulartranslocationofsirt1andpparg
AT lilisun meninorchestrateshepaticglucoseandfattyaciduptakeviadeployingthecellulartranslocationofsirt1andpparg
AT zhongjinxu meninorchestrateshepaticglucoseandfattyaciduptakeviadeployingthecellulartranslocationofsirt1andpparg
AT shengxuanwang meninorchestrateshepaticglucoseandfattyaciduptakeviadeployingthecellulartranslocationofsirt1andpparg
AT jingxuanzhou meninorchestrateshepaticglucoseandfattyaciduptakeviadeployingthecellulartranslocationofsirt1andpparg
AT xuanningwu meninorchestrateshepaticglucoseandfattyaciduptakeviadeployingthecellulartranslocationofsirt1andpparg
AT kerongshi meninorchestrateshepaticglucoseandfattyaciduptakeviadeployingthecellulartranslocationofsirt1andpparg