Cargando…

Silencing of SAA1 inhibits palmitate- or high-fat diet induced insulin resistance through suppression of the NF-κB pathway

BACKGROUND: Obesity is one of the leading causes of insulin resistance. Accumulating reports have highlighted that serum amyloid A-1 (SAA1) is a potential candidate that is capable of attenuating insulin resistance. Hence, we conducted the current study with aims of investigating our proposed hypoth...

Descripción completa

Detalles Bibliográficos
Autores principales: Wang, Yong, Cao, Feng, Wang, Yang, Yu, Gang, Jia, Ben-Li
Formato: Online Artículo Texto
Lenguaje:English
Publicado: BioMed Central 2019
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC6503374/
https://www.ncbi.nlm.nih.gov/pubmed/31060494
http://dx.doi.org/10.1186/s10020-019-0075-4
_version_ 1783416397455425536
author Wang, Yong
Cao, Feng
Wang, Yang
Yu, Gang
Jia, Ben-Li
author_facet Wang, Yong
Cao, Feng
Wang, Yang
Yu, Gang
Jia, Ben-Li
author_sort Wang, Yong
collection PubMed
description BACKGROUND: Obesity is one of the leading causes of insulin resistance. Accumulating reports have highlighted that serum amyloid A-1 (SAA1) is a potential candidate that is capable of attenuating insulin resistance. Hence, we conducted the current study with aims of investigating our proposed hypothesis that silencing SAA1 could inhibit the progression of obesity-induced insulin resistance through the NF-κB pathway. METHODS: Gene expression microarray analysis was initially performed to screen differentially expressed genes (DEGs) associated with obesity. Palmitate (PA)-induced insulin resistance Huh7 cell models and high-fat diet (HFD)-induced mouse models were established to elucidate the effect of SAA1/Saa1 on insulin resistance. The NF-κB pathway-related expression was subsequently determined through the application of reverse transcription quantitative polymerase chain reaction (RT-qPCR) and Western blot analysis. RESULTS: Saa1 was identified as an obesity-related gene based on the microarray data of GSE39549. Saa1 was determined to be highly expressed in HFD-induced insulin resistance mouse models. PA-induced Huh7 cells, treated with silenced SAA1 or NF-κB pathway inhibition using BAY 11–7082, displayed a marked decrease in both Saa1 and SOCS3 as well as an elevation in 2DG, IRS1 and the extent of IRS1 phosphorylation. HFD mice treated with silenced Saa1 or inhibited NF-κB pathway exhibited improved fasting blood glucose (FBG) levels as well as fasting plasma insulin (FPI) levels, glucose tolerance and systemic insulin sensitivity. Saa1/SAA1 was determined to show a stimulatory effect on the transport of the NF-κBp65 protein from the cytoplasm to the nucleus both in vivo and in vitro, suggesting that Saa1/SAA1 could activate the NF-κB pathway. CONCLUSION: Taken together, our key findings highlight a novel mechanism by which silencing of SAA1 hinders PA or HFD-induced insulin resistance through inhibition of the NF-κB pathway. ELECTRONIC SUPPLEMENTARY MATERIAL: The online version of this article (10.1186/s10020-019-0075-4) contains supplementary material, which is available to authorized users.
format Online
Article
Text
id pubmed-6503374
institution National Center for Biotechnology Information
language English
publishDate 2019
publisher BioMed Central
record_format MEDLINE/PubMed
spelling pubmed-65033742019-05-10 Silencing of SAA1 inhibits palmitate- or high-fat diet induced insulin resistance through suppression of the NF-κB pathway Wang, Yong Cao, Feng Wang, Yang Yu, Gang Jia, Ben-Li Mol Med Research Article BACKGROUND: Obesity is one of the leading causes of insulin resistance. Accumulating reports have highlighted that serum amyloid A-1 (SAA1) is a potential candidate that is capable of attenuating insulin resistance. Hence, we conducted the current study with aims of investigating our proposed hypothesis that silencing SAA1 could inhibit the progression of obesity-induced insulin resistance through the NF-κB pathway. METHODS: Gene expression microarray analysis was initially performed to screen differentially expressed genes (DEGs) associated with obesity. Palmitate (PA)-induced insulin resistance Huh7 cell models and high-fat diet (HFD)-induced mouse models were established to elucidate the effect of SAA1/Saa1 on insulin resistance. The NF-κB pathway-related expression was subsequently determined through the application of reverse transcription quantitative polymerase chain reaction (RT-qPCR) and Western blot analysis. RESULTS: Saa1 was identified as an obesity-related gene based on the microarray data of GSE39549. Saa1 was determined to be highly expressed in HFD-induced insulin resistance mouse models. PA-induced Huh7 cells, treated with silenced SAA1 or NF-κB pathway inhibition using BAY 11–7082, displayed a marked decrease in both Saa1 and SOCS3 as well as an elevation in 2DG, IRS1 and the extent of IRS1 phosphorylation. HFD mice treated with silenced Saa1 or inhibited NF-κB pathway exhibited improved fasting blood glucose (FBG) levels as well as fasting plasma insulin (FPI) levels, glucose tolerance and systemic insulin sensitivity. Saa1/SAA1 was determined to show a stimulatory effect on the transport of the NF-κBp65 protein from the cytoplasm to the nucleus both in vivo and in vitro, suggesting that Saa1/SAA1 could activate the NF-κB pathway. CONCLUSION: Taken together, our key findings highlight a novel mechanism by which silencing of SAA1 hinders PA or HFD-induced insulin resistance through inhibition of the NF-κB pathway. ELECTRONIC SUPPLEMENTARY MATERIAL: The online version of this article (10.1186/s10020-019-0075-4) contains supplementary material, which is available to authorized users. BioMed Central 2019-05-06 /pmc/articles/PMC6503374/ /pubmed/31060494 http://dx.doi.org/10.1186/s10020-019-0075-4 Text en © The Author(s) 2019 Open AccessThis article is distributed under the terms of the Creative Commons Attribution 4.0 International License (http://creativecommons.org/licenses/by/4.0/), which permits unrestricted use, distribution, and reproduction in any medium, provided you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated.
spellingShingle Research Article
Wang, Yong
Cao, Feng
Wang, Yang
Yu, Gang
Jia, Ben-Li
Silencing of SAA1 inhibits palmitate- or high-fat diet induced insulin resistance through suppression of the NF-κB pathway
title Silencing of SAA1 inhibits palmitate- or high-fat diet induced insulin resistance through suppression of the NF-κB pathway
title_full Silencing of SAA1 inhibits palmitate- or high-fat diet induced insulin resistance through suppression of the NF-κB pathway
title_fullStr Silencing of SAA1 inhibits palmitate- or high-fat diet induced insulin resistance through suppression of the NF-κB pathway
title_full_unstemmed Silencing of SAA1 inhibits palmitate- or high-fat diet induced insulin resistance through suppression of the NF-κB pathway
title_short Silencing of SAA1 inhibits palmitate- or high-fat diet induced insulin resistance through suppression of the NF-κB pathway
title_sort silencing of saa1 inhibits palmitate- or high-fat diet induced insulin resistance through suppression of the nf-κb pathway
topic Research Article
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC6503374/
https://www.ncbi.nlm.nih.gov/pubmed/31060494
http://dx.doi.org/10.1186/s10020-019-0075-4
work_keys_str_mv AT wangyong silencingofsaa1inhibitspalmitateorhighfatdietinducedinsulinresistancethroughsuppressionofthenfkbpathway
AT caofeng silencingofsaa1inhibitspalmitateorhighfatdietinducedinsulinresistancethroughsuppressionofthenfkbpathway
AT wangyang silencingofsaa1inhibitspalmitateorhighfatdietinducedinsulinresistancethroughsuppressionofthenfkbpathway
AT yugang silencingofsaa1inhibitspalmitateorhighfatdietinducedinsulinresistancethroughsuppressionofthenfkbpathway
AT jiabenli silencingofsaa1inhibitspalmitateorhighfatdietinducedinsulinresistancethroughsuppressionofthenfkbpathway