Cargando…

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

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 live...

Descripción completa

Detalles Bibliográficos
Autores principales: Liu, Tingjun, Li, Ranran, Sun, Lili, Xu, Zhongjin, Wang, Shengxuan, Zhou, Jingxuan, Wu, Xuanning, Shi, Kerong
Formato: Online Artículo Texto
Lenguaje:English
Publicado: BioMed Central 2023
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10517496/
https://www.ncbi.nlm.nih.gov/pubmed/37740216
http://dx.doi.org/10.1186/s13578-023-01119-y
_version_ 1785109334905061376
author Liu, Tingjun
Li, Ranran
Sun, Lili
Xu, Zhongjin
Wang, Shengxuan
Zhou, Jingxuan
Wu, Xuanning
Shi, Kerong
author_facet Liu, Tingjun
Li, Ranran
Sun, Lili
Xu, Zhongjin
Wang, Shengxuan
Zhou, Jingxuan
Wu, Xuanning
Shi, Kerong
author_sort Liu, Tingjun
collection PubMed
description 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: [Image: see text] SUPPLEMENTARY INFORMATION: The online version contains supplementary material available at 10.1186/s13578-023-01119-y.
format Online
Article
Text
id pubmed-10517496
institution National Center for Biotechnology Information
language English
publishDate 2023
publisher BioMed Central
record_format MEDLINE/PubMed
spelling pubmed-105174962023-09-24 Menin orchestrates hepatic glucose and fatty acid uptake via deploying the cellular translocation of SIRT1 and PPARγ Liu, Tingjun Li, Ranran Sun, Lili Xu, Zhongjin Wang, Shengxuan Zhou, Jingxuan Wu, Xuanning Shi, Kerong Cell Biosci Research 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: [Image: see text] SUPPLEMENTARY INFORMATION: The online version contains supplementary material available at 10.1186/s13578-023-01119-y. BioMed Central 2023-09-22 /pmc/articles/PMC10517496/ /pubmed/37740216 http://dx.doi.org/10.1186/s13578-023-01119-y Text en © The Author(s) 2023 https://creativecommons.org/licenses/by/4.0/Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/ (https://creativecommons.org/licenses/by/4.0/) . The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/ (https://creativecommons.org/publicdomain/zero/1.0/) ) applies to the data made available in this article, unless otherwise stated in a credit line to the data.
spellingShingle Research
Liu, Tingjun
Li, Ranran
Sun, Lili
Xu, Zhongjin
Wang, Shengxuan
Zhou, Jingxuan
Wu, Xuanning
Shi, Kerong
Menin orchestrates hepatic glucose and fatty acid uptake via deploying the cellular translocation of SIRT1 and PPARγ
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 Research
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10517496/
https://www.ncbi.nlm.nih.gov/pubmed/37740216
http://dx.doi.org/10.1186/s13578-023-01119-y
work_keys_str_mv AT liutingjun meninorchestrateshepaticglucoseandfattyaciduptakeviadeployingthecellulartranslocationofsirt1andpparg
AT liranran meninorchestrateshepaticglucoseandfattyaciduptakeviadeployingthecellulartranslocationofsirt1andpparg
AT sunlili meninorchestrateshepaticglucoseandfattyaciduptakeviadeployingthecellulartranslocationofsirt1andpparg
AT xuzhongjin meninorchestrateshepaticglucoseandfattyaciduptakeviadeployingthecellulartranslocationofsirt1andpparg
AT wangshengxuan meninorchestrateshepaticglucoseandfattyaciduptakeviadeployingthecellulartranslocationofsirt1andpparg
AT zhoujingxuan meninorchestrateshepaticglucoseandfattyaciduptakeviadeployingthecellulartranslocationofsirt1andpparg
AT wuxuanning meninorchestrateshepaticglucoseandfattyaciduptakeviadeployingthecellulartranslocationofsirt1andpparg
AT shikerong meninorchestrateshepaticglucoseandfattyaciduptakeviadeployingthecellulartranslocationofsirt1andpparg