Cargando…

Mogroside V Alleviates Lipopolysaccharide-Induced Neuroinflammation via Inhibition of TLR4-MyD88 and Activation of AKT/AMPK-Nrf2 Signaling Pathway

As innate immune effector cells in the central nervous system (CNS), microglia not only are essential for the normal development of nervous system but also act on different neurological diseases, including Alzheimer's disease (AD), Huntington's disease (HD), and other neuroinflammatory dis...

Descripción completa

Detalles Bibliográficos
Autores principales: Liu, Yuanyuan, Zhang, Boxi, Liu, Jiahe, Qiao, Chunyu, Xue, Nianyu, Lv, Hongming, Li, Shize
Formato: Online Artículo Texto
Lenguaje:English
Publicado: Hindawi 2021
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC8105091/
https://www.ncbi.nlm.nih.gov/pubmed/34012471
http://dx.doi.org/10.1155/2021/5521519
_version_ 1783689540659052544
author Liu, Yuanyuan
Zhang, Boxi
Liu, Jiahe
Qiao, Chunyu
Xue, Nianyu
Lv, Hongming
Li, Shize
author_facet Liu, Yuanyuan
Zhang, Boxi
Liu, Jiahe
Qiao, Chunyu
Xue, Nianyu
Lv, Hongming
Li, Shize
author_sort Liu, Yuanyuan
collection PubMed
description As innate immune effector cells in the central nervous system (CNS), microglia not only are essential for the normal development of nervous system but also act on different neurological diseases, including Alzheimer's disease (AD), Huntington's disease (HD), and other neuroinflammatory diseases. Mogroside V (Mog), a natural plant active ingredient and isolated form of Momordica grosvenori, has been shown to possess anti-inflammatory action, but few studies were carried out to investigate the effects of Mog on neuroinflammation. This study aimed to investigate the role of Mog in lipopolysaccharide- (LPS-) induced neuroinflammation and neuronal damage, revealing the underlying mechanisms. Our data indicated that Mog significantly inhibited the LPS-induced production of proinflammatory factors, such as tumor necrosis factor-α (TNF-α), interleukin-1β (IL-1β), IL-18, IL-6, cyclooxygenase-2 (COX-2), inducible nitric oxide synthase (iNOS), and high mobility group box 1 (HMGB1) in BV-2 cells. We found that Mog also suppressed toll-like receptor 4 (TLR4), myeloid differentiation factor 88 (MyD88), the phosphorylation of mitogen-activated protein kinases (MAPKs), adenosine 5′-monophosphate- (AMP-) activated protein kinase (AMPK), nuclear factor kappa-B (NF-κB), and protein kinase B (AKT). Moreover, Mog also enhanced the expression of γ-glutamyl cysteine synthetase catalytic subunit (GCLC), modifier subunit (GCLM), heme oxygenase-1 (HO-1), and quinine oxidoreductase 1 (NQO1) proteins, mostly depending on the nuclear translation of nuclear factor erythroid-2 related factor 2 (Nrf2). In contrast, pretreatment with inhibitors of AKT can suppress the phosphorylation of AMPK, Nrf2, and its downstream proteins expression. In summary, Mog might play a protective role against LPS-induced neurotoxicity by inhibiting the TLR4-MyD88 and activation of AMPK/AKT-Nrf2 signaling pathway.
format Online
Article
Text
id pubmed-8105091
institution National Center for Biotechnology Information
language English
publishDate 2021
publisher Hindawi
record_format MEDLINE/PubMed
spelling pubmed-81050912021-05-18 Mogroside V Alleviates Lipopolysaccharide-Induced Neuroinflammation via Inhibition of TLR4-MyD88 and Activation of AKT/AMPK-Nrf2 Signaling Pathway Liu, Yuanyuan Zhang, Boxi Liu, Jiahe Qiao, Chunyu Xue, Nianyu Lv, Hongming Li, Shize Evid Based Complement Alternat Med Research Article As innate immune effector cells in the central nervous system (CNS), microglia not only are essential for the normal development of nervous system but also act on different neurological diseases, including Alzheimer's disease (AD), Huntington's disease (HD), and other neuroinflammatory diseases. Mogroside V (Mog), a natural plant active ingredient and isolated form of Momordica grosvenori, has been shown to possess anti-inflammatory action, but few studies were carried out to investigate the effects of Mog on neuroinflammation. This study aimed to investigate the role of Mog in lipopolysaccharide- (LPS-) induced neuroinflammation and neuronal damage, revealing the underlying mechanisms. Our data indicated that Mog significantly inhibited the LPS-induced production of proinflammatory factors, such as tumor necrosis factor-α (TNF-α), interleukin-1β (IL-1β), IL-18, IL-6, cyclooxygenase-2 (COX-2), inducible nitric oxide synthase (iNOS), and high mobility group box 1 (HMGB1) in BV-2 cells. We found that Mog also suppressed toll-like receptor 4 (TLR4), myeloid differentiation factor 88 (MyD88), the phosphorylation of mitogen-activated protein kinases (MAPKs), adenosine 5′-monophosphate- (AMP-) activated protein kinase (AMPK), nuclear factor kappa-B (NF-κB), and protein kinase B (AKT). Moreover, Mog also enhanced the expression of γ-glutamyl cysteine synthetase catalytic subunit (GCLC), modifier subunit (GCLM), heme oxygenase-1 (HO-1), and quinine oxidoreductase 1 (NQO1) proteins, mostly depending on the nuclear translation of nuclear factor erythroid-2 related factor 2 (Nrf2). In contrast, pretreatment with inhibitors of AKT can suppress the phosphorylation of AMPK, Nrf2, and its downstream proteins expression. In summary, Mog might play a protective role against LPS-induced neurotoxicity by inhibiting the TLR4-MyD88 and activation of AMPK/AKT-Nrf2 signaling pathway. Hindawi 2021-04-30 /pmc/articles/PMC8105091/ /pubmed/34012471 http://dx.doi.org/10.1155/2021/5521519 Text en Copyright © 2021 Yuanyuan Liu et al. https://creativecommons.org/licenses/by/4.0/This is an open access article distributed under the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited.
spellingShingle Research Article
Liu, Yuanyuan
Zhang, Boxi
Liu, Jiahe
Qiao, Chunyu
Xue, Nianyu
Lv, Hongming
Li, Shize
Mogroside V Alleviates Lipopolysaccharide-Induced Neuroinflammation via Inhibition of TLR4-MyD88 and Activation of AKT/AMPK-Nrf2 Signaling Pathway
title Mogroside V Alleviates Lipopolysaccharide-Induced Neuroinflammation via Inhibition of TLR4-MyD88 and Activation of AKT/AMPK-Nrf2 Signaling Pathway
title_full Mogroside V Alleviates Lipopolysaccharide-Induced Neuroinflammation via Inhibition of TLR4-MyD88 and Activation of AKT/AMPK-Nrf2 Signaling Pathway
title_fullStr Mogroside V Alleviates Lipopolysaccharide-Induced Neuroinflammation via Inhibition of TLR4-MyD88 and Activation of AKT/AMPK-Nrf2 Signaling Pathway
title_full_unstemmed Mogroside V Alleviates Lipopolysaccharide-Induced Neuroinflammation via Inhibition of TLR4-MyD88 and Activation of AKT/AMPK-Nrf2 Signaling Pathway
title_short Mogroside V Alleviates Lipopolysaccharide-Induced Neuroinflammation via Inhibition of TLR4-MyD88 and Activation of AKT/AMPK-Nrf2 Signaling Pathway
title_sort mogroside v alleviates lipopolysaccharide-induced neuroinflammation via inhibition of tlr4-myd88 and activation of akt/ampk-nrf2 signaling pathway
topic Research Article
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC8105091/
https://www.ncbi.nlm.nih.gov/pubmed/34012471
http://dx.doi.org/10.1155/2021/5521519
work_keys_str_mv AT liuyuanyuan mogrosidevalleviateslipopolysaccharideinducedneuroinflammationviainhibitionoftlr4myd88andactivationofaktampknrf2signalingpathway
AT zhangboxi mogrosidevalleviateslipopolysaccharideinducedneuroinflammationviainhibitionoftlr4myd88andactivationofaktampknrf2signalingpathway
AT liujiahe mogrosidevalleviateslipopolysaccharideinducedneuroinflammationviainhibitionoftlr4myd88andactivationofaktampknrf2signalingpathway
AT qiaochunyu mogrosidevalleviateslipopolysaccharideinducedneuroinflammationviainhibitionoftlr4myd88andactivationofaktampknrf2signalingpathway
AT xuenianyu mogrosidevalleviateslipopolysaccharideinducedneuroinflammationviainhibitionoftlr4myd88andactivationofaktampknrf2signalingpathway
AT lvhongming mogrosidevalleviateslipopolysaccharideinducedneuroinflammationviainhibitionoftlr4myd88andactivationofaktampknrf2signalingpathway
AT lishize mogrosidevalleviateslipopolysaccharideinducedneuroinflammationviainhibitionoftlr4myd88andactivationofaktampknrf2signalingpathway