Cargando…

Amelioration of Mitochondrial Dysfunction-Induced Insulin Resistance in Differentiated 3T3-L1 Adipocytes via Inhibition of NF-κB Pathways

A growing body of evidence suggests that activation of nuclear factor kappa B (NF-κB) signaling pathways is among the inflammatory mechanism involved in the development of insulin resistance and chronic low-grade inflammation in adipose tissues derived from obese animal and human subjects. Neverthel...

Descripción completa

Detalles Bibliográficos
Autores principales: Hafizi Abu Bakar, Mohamad, Sarmidi, Mohamad Roji, Kai, Cheng Kian, Huri, Hasniza Zaman, Yaakob, Harisun
Formato: Online Artículo Texto
Lenguaje:English
Publicado: MDPI 2014
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC4284705/
https://www.ncbi.nlm.nih.gov/pubmed/25474091
http://dx.doi.org/10.3390/ijms151222227
_version_ 1782351441545920512
author Hafizi Abu Bakar, Mohamad
Sarmidi, Mohamad Roji
Kai, Cheng Kian
Huri, Hasniza Zaman
Yaakob, Harisun
author_facet Hafizi Abu Bakar, Mohamad
Sarmidi, Mohamad Roji
Kai, Cheng Kian
Huri, Hasniza Zaman
Yaakob, Harisun
author_sort Hafizi Abu Bakar, Mohamad
collection PubMed
description A growing body of evidence suggests that activation of nuclear factor kappa B (NF-κB) signaling pathways is among the inflammatory mechanism involved in the development of insulin resistance and chronic low-grade inflammation in adipose tissues derived from obese animal and human subjects. Nevertheless, little is known about the roles of NF-κB pathways in regulating mitochondrial function of the adipose tissues. In the present study, we sought to investigate the direct effects of celastrol (potent NF-κB inhibitor) upon mitochondrial dysfunction-induced insulin resistance in 3T3-L1 adipocytes. Celastrol ameliorates mitochondrial dysfunction by altering mitochondrial fusion and fission in adipocytes. The levels of oxidative DNA damage, protein carbonylation and lipid peroxidation were down-regulated. Further, the morphology and quantification of intracellular lipid droplets revealed the decrease of intracellular lipid accumulation with reduced lipolysis. Moreover, massive production of the pro-inflammatory mediators tumor necrosis factor-α (TNF-α) and interleukin-1β (IL-1β) were markedly depleted. Insulin-stimulated glucose uptake activity was restored with the enhancement of insulin signaling pathways. This study signified that the treatments modulated towards knockdown of NF-κB transcription factor may counteract these metabolic insults exacerbated in our model of synergy between mitochondrial dysfunction and inflammation. These results demonstrate for the first time that NF-κB inhibition modulates mitochondrial dysfunction induced insulin resistance in 3T3-L1 adipocytes.
format Online
Article
Text
id pubmed-4284705
institution National Center for Biotechnology Information
language English
publishDate 2014
publisher MDPI
record_format MEDLINE/PubMed
spelling pubmed-42847052015-01-21 Amelioration of Mitochondrial Dysfunction-Induced Insulin Resistance in Differentiated 3T3-L1 Adipocytes via Inhibition of NF-κB Pathways Hafizi Abu Bakar, Mohamad Sarmidi, Mohamad Roji Kai, Cheng Kian Huri, Hasniza Zaman Yaakob, Harisun Int J Mol Sci Article A growing body of evidence suggests that activation of nuclear factor kappa B (NF-κB) signaling pathways is among the inflammatory mechanism involved in the development of insulin resistance and chronic low-grade inflammation in adipose tissues derived from obese animal and human subjects. Nevertheless, little is known about the roles of NF-κB pathways in regulating mitochondrial function of the adipose tissues. In the present study, we sought to investigate the direct effects of celastrol (potent NF-κB inhibitor) upon mitochondrial dysfunction-induced insulin resistance in 3T3-L1 adipocytes. Celastrol ameliorates mitochondrial dysfunction by altering mitochondrial fusion and fission in adipocytes. The levels of oxidative DNA damage, protein carbonylation and lipid peroxidation were down-regulated. Further, the morphology and quantification of intracellular lipid droplets revealed the decrease of intracellular lipid accumulation with reduced lipolysis. Moreover, massive production of the pro-inflammatory mediators tumor necrosis factor-α (TNF-α) and interleukin-1β (IL-1β) were markedly depleted. Insulin-stimulated glucose uptake activity was restored with the enhancement of insulin signaling pathways. This study signified that the treatments modulated towards knockdown of NF-κB transcription factor may counteract these metabolic insults exacerbated in our model of synergy between mitochondrial dysfunction and inflammation. These results demonstrate for the first time that NF-κB inhibition modulates mitochondrial dysfunction induced insulin resistance in 3T3-L1 adipocytes. MDPI 2014-12-02 /pmc/articles/PMC4284705/ /pubmed/25474091 http://dx.doi.org/10.3390/ijms151222227 Text en © 2014 by the authors; licensee MDPI, Basel, Switzerland. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution license (http://creativecommons.org/licenses/by/4.0/).
spellingShingle Article
Hafizi Abu Bakar, Mohamad
Sarmidi, Mohamad Roji
Kai, Cheng Kian
Huri, Hasniza Zaman
Yaakob, Harisun
Amelioration of Mitochondrial Dysfunction-Induced Insulin Resistance in Differentiated 3T3-L1 Adipocytes via Inhibition of NF-κB Pathways
title Amelioration of Mitochondrial Dysfunction-Induced Insulin Resistance in Differentiated 3T3-L1 Adipocytes via Inhibition of NF-κB Pathways
title_full Amelioration of Mitochondrial Dysfunction-Induced Insulin Resistance in Differentiated 3T3-L1 Adipocytes via Inhibition of NF-κB Pathways
title_fullStr Amelioration of Mitochondrial Dysfunction-Induced Insulin Resistance in Differentiated 3T3-L1 Adipocytes via Inhibition of NF-κB Pathways
title_full_unstemmed Amelioration of Mitochondrial Dysfunction-Induced Insulin Resistance in Differentiated 3T3-L1 Adipocytes via Inhibition of NF-κB Pathways
title_short Amelioration of Mitochondrial Dysfunction-Induced Insulin Resistance in Differentiated 3T3-L1 Adipocytes via Inhibition of NF-κB Pathways
title_sort amelioration of mitochondrial dysfunction-induced insulin resistance in differentiated 3t3-l1 adipocytes via inhibition of nf-κb pathways
topic Article
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC4284705/
https://www.ncbi.nlm.nih.gov/pubmed/25474091
http://dx.doi.org/10.3390/ijms151222227
work_keys_str_mv AT hafiziabubakarmohamad ameliorationofmitochondrialdysfunctioninducedinsulinresistanceindifferentiated3t3l1adipocytesviainhibitionofnfkbpathways
AT sarmidimohamadroji ameliorationofmitochondrialdysfunctioninducedinsulinresistanceindifferentiated3t3l1adipocytesviainhibitionofnfkbpathways
AT kaichengkian ameliorationofmitochondrialdysfunctioninducedinsulinresistanceindifferentiated3t3l1adipocytesviainhibitionofnfkbpathways
AT hurihasnizazaman ameliorationofmitochondrialdysfunctioninducedinsulinresistanceindifferentiated3t3l1adipocytesviainhibitionofnfkbpathways
AT yaakobharisun ameliorationofmitochondrialdysfunctioninducedinsulinresistanceindifferentiated3t3l1adipocytesviainhibitionofnfkbpathways