Notoginsenoside R1 alleviates lipopolysaccharide-triggered PC-12 inflammatory damage via elevating microRNA-132

Background Delayed inflammatory response is closely associated with the severity of Spinal cord injury (SCI). Herein, the function and molecular mechanism of notoginsenoside R1 (NGR1) in the in vitro model of SCI inflammation injury were explored.Methods PC-12 neuronal cells were subjected with LPS...

Full description

Bibliographic Details
Main Authors: Yuanliang Sun, Bing Liu, Xiujun Zheng, Dechun Wang
Format: Article
Language:English
Published: Taylor & Francis Group 2019-12-01
Series:Artificial Cells, Nanomedicine, and Biotechnology
Subjects:
Online Access:https://www.tandfonline.com/doi/10.1080/21691401.2019.1610414
_version_ 1811302683134394368
author Yuanliang Sun
Bing Liu
Xiujun Zheng
Dechun Wang
author_facet Yuanliang Sun
Bing Liu
Xiujun Zheng
Dechun Wang
author_sort Yuanliang Sun
collection DOAJ
description Background Delayed inflammatory response is closely associated with the severity of Spinal cord injury (SCI). Herein, the function and molecular mechanism of notoginsenoside R1 (NGR1) in the in vitro model of SCI inflammation injury were explored.Methods PC-12 neuronal cells were subjected with LPS to construct a cell-based model of SCI inflammatory injury. NGR1 was applied in this cell model. miR-132 was silenced by transfection with miR-132 inhibitor. Cell viability and apoptosis were assessed, respectively. Then, the expression changes of pro-inflammatory cytokines and JNK pathway were examined.Results In this model, LPS was neurotoxic, with inhibiting PC-12 cell viability, inducing apoptosis, and enhancing concentrations of IL-6, IL-8 and TNF-α. However, NGR1 weakened the influence of LPS on PC-12 cells via elevating cell viability, decreasing apoptosis, decreasing pro-inflammatory cytokines expression, and suppressing activation of JNK signalling pathway. miR-132 was up-regulated by NGR1 treatment. Silence of miR-132 eliminated the influence of NGR1 on LPS-stimulated PC-12 cells.Conclusion NGR1 relieved PC-12 cells from LPS-triggered inflammatory damage via elevating miR-132 and hereafter suppressing JNK pathway.
first_indexed 2024-04-13T07:32:31Z
format Article
id doaj.art-6175e1620fb74c9cb1ade19e43fd5fd4
institution Directory Open Access Journal
issn 2169-1401
2169-141X
language English
last_indexed 2024-04-13T07:32:31Z
publishDate 2019-12-01
publisher Taylor & Francis Group
record_format Article
series Artificial Cells, Nanomedicine, and Biotechnology
spelling doaj.art-6175e1620fb74c9cb1ade19e43fd5fd42022-12-22T02:56:17ZengTaylor & Francis GroupArtificial Cells, Nanomedicine, and Biotechnology2169-14012169-141X2019-12-014711808181410.1080/21691401.2019.1610414Notoginsenoside R1 alleviates lipopolysaccharide-triggered PC-12 inflammatory damage via elevating microRNA-132Yuanliang Sun0Bing Liu1Xiujun Zheng2Dechun Wang3Department of Spine Surgery, The Affiliated Hospital of Qingdao University, Qingdao, ChinaDepartment of Critical Care Medicine, The Affiliated Hospital of Qingdao University, Qingdao, ChinaDepartment of Spine Surgery, The Affiliated Hospital of Qingdao University, Qingdao, ChinaDepartment of Spine Surgery, Qingdao Municipal Hospital, Qingdao, ChinaBackground Delayed inflammatory response is closely associated with the severity of Spinal cord injury (SCI). Herein, the function and molecular mechanism of notoginsenoside R1 (NGR1) in the in vitro model of SCI inflammation injury were explored.Methods PC-12 neuronal cells were subjected with LPS to construct a cell-based model of SCI inflammatory injury. NGR1 was applied in this cell model. miR-132 was silenced by transfection with miR-132 inhibitor. Cell viability and apoptosis were assessed, respectively. Then, the expression changes of pro-inflammatory cytokines and JNK pathway were examined.Results In this model, LPS was neurotoxic, with inhibiting PC-12 cell viability, inducing apoptosis, and enhancing concentrations of IL-6, IL-8 and TNF-α. However, NGR1 weakened the influence of LPS on PC-12 cells via elevating cell viability, decreasing apoptosis, decreasing pro-inflammatory cytokines expression, and suppressing activation of JNK signalling pathway. miR-132 was up-regulated by NGR1 treatment. Silence of miR-132 eliminated the influence of NGR1 on LPS-stimulated PC-12 cells.Conclusion NGR1 relieved PC-12 cells from LPS-triggered inflammatory damage via elevating miR-132 and hereafter suppressing JNK pathway.https://www.tandfonline.com/doi/10.1080/21691401.2019.1610414Notoginsenoside R1inflammation injuryspinal cord injury (SCI)lipopolysaccharideJNK pathway
spellingShingle Yuanliang Sun
Bing Liu
Xiujun Zheng
Dechun Wang
Notoginsenoside R1 alleviates lipopolysaccharide-triggered PC-12 inflammatory damage via elevating microRNA-132
Artificial Cells, Nanomedicine, and Biotechnology
Notoginsenoside R1
inflammation injury
spinal cord injury (SCI)
lipopolysaccharide
JNK pathway
title Notoginsenoside R1 alleviates lipopolysaccharide-triggered PC-12 inflammatory damage via elevating microRNA-132
title_full Notoginsenoside R1 alleviates lipopolysaccharide-triggered PC-12 inflammatory damage via elevating microRNA-132
title_fullStr Notoginsenoside R1 alleviates lipopolysaccharide-triggered PC-12 inflammatory damage via elevating microRNA-132
title_full_unstemmed Notoginsenoside R1 alleviates lipopolysaccharide-triggered PC-12 inflammatory damage via elevating microRNA-132
title_short Notoginsenoside R1 alleviates lipopolysaccharide-triggered PC-12 inflammatory damage via elevating microRNA-132
title_sort notoginsenoside r1 alleviates lipopolysaccharide triggered pc 12 inflammatory damage via elevating microrna 132
topic Notoginsenoside R1
inflammation injury
spinal cord injury (SCI)
lipopolysaccharide
JNK pathway
url https://www.tandfonline.com/doi/10.1080/21691401.2019.1610414
work_keys_str_mv AT yuanliangsun notoginsenosider1alleviateslipopolysaccharidetriggeredpc12inflammatorydamageviaelevatingmicrorna132
AT bingliu notoginsenosider1alleviateslipopolysaccharidetriggeredpc12inflammatorydamageviaelevatingmicrorna132
AT xiujunzheng notoginsenosider1alleviateslipopolysaccharidetriggeredpc12inflammatorydamageviaelevatingmicrorna132
AT dechunwang notoginsenosider1alleviateslipopolysaccharidetriggeredpc12inflammatorydamageviaelevatingmicrorna132