Integration analysis identifies the role of metallothionein in the progression from hepatic steatosis to steatohepatitis

BackgroundNon-alcoholic fatty liver disease (NAFLD), a metabolic disorder that develops from non-alcoholic fatty liver (NAFL) to non-alcoholic steatohepatitis (NASH), has become an epidemic of chronic liver dysfunction worldwide. However, mechanisms that govern the transition from NAFL to NASH have...

Full description

Bibliographic Details
Main Authors: Xiaoya Li, Shaoping Zhong, Yifan Sun, Xinmei Huang, Yue Li, Lihong Wang, Yueyue Wu, Min Yang, Hai-Xin Yuan, Jun Liu, Shufei Zang
Format: Article
Language:English
Published: Frontiers Media S.A. 2022-10-01
Series:Frontiers in Endocrinology
Subjects:
Online Access:https://www.frontiersin.org/articles/10.3389/fendo.2022.951093/full
_version_ 1811182063895707648
author Xiaoya Li
Shaoping Zhong
Yifan Sun
Xinmei Huang
Yue Li
Lihong Wang
Yueyue Wu
Min Yang
Hai-Xin Yuan
Jun Liu
Shufei Zang
author_facet Xiaoya Li
Shaoping Zhong
Yifan Sun
Xinmei Huang
Yue Li
Lihong Wang
Yueyue Wu
Min Yang
Hai-Xin Yuan
Jun Liu
Shufei Zang
author_sort Xiaoya Li
collection DOAJ
description BackgroundNon-alcoholic fatty liver disease (NAFLD), a metabolic disorder that develops from non-alcoholic fatty liver (NAFL) to non-alcoholic steatohepatitis (NASH), has become an epidemic of chronic liver dysfunction worldwide. However, mechanisms that govern the transition from NAFL to NASH have not been fully elucidated.MethodsGene expression profile data of NAFLD liver tissues were obtained from Gene Expression Omnibus (GEO), including three microarray datasets with 60 NAFL and 44 NASH patients. Integrative differentially expressed genes (DEGs) between NAFL and NASH patients were identified using robust rank aggregation (RRA) analysis. Hub genes were identified combined with gene ontology functional annotation and protein–protein interaction network construction and validated using a sequencing dataset. Huh-7 cells with palmitate-induced lipid overload and NAFLD-diet mouse model of different stages were used to verify our findings.ResultsRRA analysis determined 70 robust DEGs between NAFL and NASH. The most robustly upregulated genes were SPP1, AKR1B10, CHST9, and ANXA2, while the most robustly downregulated DEGs were SNORD94, SCARNA10, SNORA20, and MT1M. Cellular response to zinc ion (GO: 0071294) ranked first in GO analysis of downregulated genes, and Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment showed that mineral absorption (hsa04978) was significantly enriched. The involvement of the metallothionein pathway was further validated by the decrease of Mt1 expression during NAFL to NASH progression in NAFLD mice and the protection from lipotoxicity in liver cells by overexpressing MT1M.ConclusionsOur integrated analysis identified novel gene signatures and provided comprehensive molecular mechanisms underlying the transition from NAFL to NASH. Metallothionein might be a potential intervention target for NAFLD progression.
first_indexed 2024-04-11T09:26:57Z
format Article
id doaj.art-7217f85f3f24465896dd420554d54baf
institution Directory Open Access Journal
issn 1664-2392
language English
last_indexed 2024-04-11T09:26:57Z
publishDate 2022-10-01
publisher Frontiers Media S.A.
record_format Article
series Frontiers in Endocrinology
spelling doaj.art-7217f85f3f24465896dd420554d54baf2022-12-22T04:32:00ZengFrontiers Media S.A.Frontiers in Endocrinology1664-23922022-10-011310.3389/fendo.2022.951093951093Integration analysis identifies the role of metallothionein in the progression from hepatic steatosis to steatohepatitisXiaoya Li0Shaoping Zhong1Yifan Sun2Xinmei Huang3Yue Li4Lihong Wang5Yueyue Wu6Min Yang7Hai-Xin Yuan8Jun Liu9Shufei Zang10Department of Endocrinology of Shanghai Fifth People’s Hospital, Fudan University, Shanghai, ChinaDepartment of Neurology, Zhongshan Hospital, Fudan University, Shanghai, ChinaDepartment of Endocrinology of Shanghai Fifth People’s Hospital, Fudan University, Shanghai, ChinaDepartment of Endocrinology of Shanghai Fifth People’s Hospital, Fudan University, Shanghai, ChinaDepartment of Endocrinology of Shanghai Fifth People’s Hospital, Fudan University, Shanghai, ChinaDepartment of Endocrinology of Shanghai Fifth People’s Hospital, Fudan University, Shanghai, ChinaDepartment of Endocrinology of Shanghai Fifth People’s Hospital, Fudan University, Shanghai, ChinaDepartment of Endocrinology of Shanghai Fifth People’s Hospital, Fudan University, Shanghai, ChinaDepartment of Endocrinology of Shanghai Fifth People’s Hospital, Fudan University, Shanghai, ChinaDepartment of Endocrinology of Shanghai Fifth People’s Hospital, Fudan University, Shanghai, ChinaDepartment of Endocrinology of Shanghai Fifth People’s Hospital, Fudan University, Shanghai, ChinaBackgroundNon-alcoholic fatty liver disease (NAFLD), a metabolic disorder that develops from non-alcoholic fatty liver (NAFL) to non-alcoholic steatohepatitis (NASH), has become an epidemic of chronic liver dysfunction worldwide. However, mechanisms that govern the transition from NAFL to NASH have not been fully elucidated.MethodsGene expression profile data of NAFLD liver tissues were obtained from Gene Expression Omnibus (GEO), including three microarray datasets with 60 NAFL and 44 NASH patients. Integrative differentially expressed genes (DEGs) between NAFL and NASH patients were identified using robust rank aggregation (RRA) analysis. Hub genes were identified combined with gene ontology functional annotation and protein–protein interaction network construction and validated using a sequencing dataset. Huh-7 cells with palmitate-induced lipid overload and NAFLD-diet mouse model of different stages were used to verify our findings.ResultsRRA analysis determined 70 robust DEGs between NAFL and NASH. The most robustly upregulated genes were SPP1, AKR1B10, CHST9, and ANXA2, while the most robustly downregulated DEGs were SNORD94, SCARNA10, SNORA20, and MT1M. Cellular response to zinc ion (GO: 0071294) ranked first in GO analysis of downregulated genes, and Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment showed that mineral absorption (hsa04978) was significantly enriched. The involvement of the metallothionein pathway was further validated by the decrease of Mt1 expression during NAFL to NASH progression in NAFLD mice and the protection from lipotoxicity in liver cells by overexpressing MT1M.ConclusionsOur integrated analysis identified novel gene signatures and provided comprehensive molecular mechanisms underlying the transition from NAFL to NASH. Metallothionein might be a potential intervention target for NAFLD progression.https://www.frontiersin.org/articles/10.3389/fendo.2022.951093/fullnon-alcoholic fatty liver diseasesteatohepatitismetallothioneinintegration analysisrobust rank aggregation
spellingShingle Xiaoya Li
Shaoping Zhong
Yifan Sun
Xinmei Huang
Yue Li
Lihong Wang
Yueyue Wu
Min Yang
Hai-Xin Yuan
Jun Liu
Shufei Zang
Integration analysis identifies the role of metallothionein in the progression from hepatic steatosis to steatohepatitis
Frontiers in Endocrinology
non-alcoholic fatty liver disease
steatohepatitis
metallothionein
integration analysis
robust rank aggregation
title Integration analysis identifies the role of metallothionein in the progression from hepatic steatosis to steatohepatitis
title_full Integration analysis identifies the role of metallothionein in the progression from hepatic steatosis to steatohepatitis
title_fullStr Integration analysis identifies the role of metallothionein in the progression from hepatic steatosis to steatohepatitis
title_full_unstemmed Integration analysis identifies the role of metallothionein in the progression from hepatic steatosis to steatohepatitis
title_short Integration analysis identifies the role of metallothionein in the progression from hepatic steatosis to steatohepatitis
title_sort integration analysis identifies the role of metallothionein in the progression from hepatic steatosis to steatohepatitis
topic non-alcoholic fatty liver disease
steatohepatitis
metallothionein
integration analysis
robust rank aggregation
url https://www.frontiersin.org/articles/10.3389/fendo.2022.951093/full
work_keys_str_mv AT xiaoyali integrationanalysisidentifiestheroleofmetallothioneinintheprogressionfromhepaticsteatosistosteatohepatitis
AT shaopingzhong integrationanalysisidentifiestheroleofmetallothioneinintheprogressionfromhepaticsteatosistosteatohepatitis
AT yifansun integrationanalysisidentifiestheroleofmetallothioneinintheprogressionfromhepaticsteatosistosteatohepatitis
AT xinmeihuang integrationanalysisidentifiestheroleofmetallothioneinintheprogressionfromhepaticsteatosistosteatohepatitis
AT yueli integrationanalysisidentifiestheroleofmetallothioneinintheprogressionfromhepaticsteatosistosteatohepatitis
AT lihongwang integrationanalysisidentifiestheroleofmetallothioneinintheprogressionfromhepaticsteatosistosteatohepatitis
AT yueyuewu integrationanalysisidentifiestheroleofmetallothioneinintheprogressionfromhepaticsteatosistosteatohepatitis
AT minyang integrationanalysisidentifiestheroleofmetallothioneinintheprogressionfromhepaticsteatosistosteatohepatitis
AT haixinyuan integrationanalysisidentifiestheroleofmetallothioneinintheprogressionfromhepaticsteatosistosteatohepatitis
AT junliu integrationanalysisidentifiestheroleofmetallothioneinintheprogressionfromhepaticsteatosistosteatohepatitis
AT shufeizang integrationanalysisidentifiestheroleofmetallothioneinintheprogressionfromhepaticsteatosistosteatohepatitis