Cargando…

Downregulation of NLRP2 inhibits HUVEC viability by inhibiting the MAPK signaling pathway

Nucleotide-binding oligomerization domain (NOD)-like receptor proteins (NLRPs) are a subfamily of NOD-like receptors (NLRs) that mainly participate in innate immunity. Among the 14 NLRPs, studies on NLRP2 are few and mostly focus on its functions in reproduction and embryonic development. To the bes...

Descripción completa

Detalles Bibliográficos
Autores principales: Zhang, Xiaolu, Lu, Xinlei, Yu, Limei, Gu, Yufeng, Qu, Fuzheng
Formato: Online Artículo Texto
Lenguaje:English
Publicado: D.A. Spandidos 2019
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC6297776/
https://www.ncbi.nlm.nih.gov/pubmed/30431084
http://dx.doi.org/10.3892/mmr.2018.9625
_version_ 1783381203278102528
author Zhang, Xiaolu
Lu, Xinlei
Yu, Limei
Gu, Yufeng
Qu, Fuzheng
author_facet Zhang, Xiaolu
Lu, Xinlei
Yu, Limei
Gu, Yufeng
Qu, Fuzheng
author_sort Zhang, Xiaolu
collection PubMed
description Nucleotide-binding oligomerization domain (NOD)-like receptor proteins (NLRPs) are a subfamily of NOD-like receptors (NLRs) that mainly participate in innate immunity. Among the 14 NLRPs, studies on NLRP2 are few and mostly focus on its functions in reproduction and embryonic development. To the best of the authors' knowledge, there has been no research on the function of NLRP2 in human umbilical vein endothelial cells (HUVECs). The present study knockdown the expression of NLRP2 by transfecting a short interfering (si)RNA (siNLRP2) into HUVECs and investigating its effects on HUVECs. It was identified using a Cell Counting kit-8 assay that knockdown of NLRP2 can inhibit cell proliferation in HUVECs. The results of wound healing and Transwell assays indicated that migration and invasion were also suppressed by siNLRP2 transfection in HUVECs. Flow cytometry demonstrated that siNLRP2 induced cell cycle arrest and apoptosis in HUVECs. Western blot analysis revealed that the expression levels of cell cycle and apoptosis-associated proteins were markedly changed. In addition, knockdown of NLRP2 inhibited the mitogen-activated protein kinase (MAPK) signaling pathway by elevating extracellular signal-regulated kinase phosphorylation levels and reducing proto-oncogene serine/threonine-protein kinase expression. Taken together, it was concluded that NLRP2 served an important role in maintaining cell viability, proliferation and motility in HUVECs, mainly by promoting the MAPK signaling pathway.
format Online
Article
Text
id pubmed-6297776
institution National Center for Biotechnology Information
language English
publishDate 2019
publisher D.A. Spandidos
record_format MEDLINE/PubMed
spelling pubmed-62977762018-12-26 Downregulation of NLRP2 inhibits HUVEC viability by inhibiting the MAPK signaling pathway Zhang, Xiaolu Lu, Xinlei Yu, Limei Gu, Yufeng Qu, Fuzheng Mol Med Rep Articles Nucleotide-binding oligomerization domain (NOD)-like receptor proteins (NLRPs) are a subfamily of NOD-like receptors (NLRs) that mainly participate in innate immunity. Among the 14 NLRPs, studies on NLRP2 are few and mostly focus on its functions in reproduction and embryonic development. To the best of the authors' knowledge, there has been no research on the function of NLRP2 in human umbilical vein endothelial cells (HUVECs). The present study knockdown the expression of NLRP2 by transfecting a short interfering (si)RNA (siNLRP2) into HUVECs and investigating its effects on HUVECs. It was identified using a Cell Counting kit-8 assay that knockdown of NLRP2 can inhibit cell proliferation in HUVECs. The results of wound healing and Transwell assays indicated that migration and invasion were also suppressed by siNLRP2 transfection in HUVECs. Flow cytometry demonstrated that siNLRP2 induced cell cycle arrest and apoptosis in HUVECs. Western blot analysis revealed that the expression levels of cell cycle and apoptosis-associated proteins were markedly changed. In addition, knockdown of NLRP2 inhibited the mitogen-activated protein kinase (MAPK) signaling pathway by elevating extracellular signal-regulated kinase phosphorylation levels and reducing proto-oncogene serine/threonine-protein kinase expression. Taken together, it was concluded that NLRP2 served an important role in maintaining cell viability, proliferation and motility in HUVECs, mainly by promoting the MAPK signaling pathway. D.A. Spandidos 2019-01 2018-11-05 /pmc/articles/PMC6297776/ /pubmed/30431084 http://dx.doi.org/10.3892/mmr.2018.9625 Text en Copyright: © Zhang et al. This is an open access article distributed under the terms of the Creative Commons Attribution-NonCommercial-NoDerivs License (https://creativecommons.org/licenses/by-nc-nd/4.0/) , which permits use and distribution in any medium, provided the original work is properly cited, the use is non-commercial and no modifications or adaptations are made.
spellingShingle Articles
Zhang, Xiaolu
Lu, Xinlei
Yu, Limei
Gu, Yufeng
Qu, Fuzheng
Downregulation of NLRP2 inhibits HUVEC viability by inhibiting the MAPK signaling pathway
title Downregulation of NLRP2 inhibits HUVEC viability by inhibiting the MAPK signaling pathway
title_full Downregulation of NLRP2 inhibits HUVEC viability by inhibiting the MAPK signaling pathway
title_fullStr Downregulation of NLRP2 inhibits HUVEC viability by inhibiting the MAPK signaling pathway
title_full_unstemmed Downregulation of NLRP2 inhibits HUVEC viability by inhibiting the MAPK signaling pathway
title_short Downregulation of NLRP2 inhibits HUVEC viability by inhibiting the MAPK signaling pathway
title_sort downregulation of nlrp2 inhibits huvec viability by inhibiting the mapk signaling pathway
topic Articles
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC6297776/
https://www.ncbi.nlm.nih.gov/pubmed/30431084
http://dx.doi.org/10.3892/mmr.2018.9625
work_keys_str_mv AT zhangxiaolu downregulationofnlrp2inhibitshuvecviabilitybyinhibitingthemapksignalingpathway
AT luxinlei downregulationofnlrp2inhibitshuvecviabilitybyinhibitingthemapksignalingpathway
AT yulimei downregulationofnlrp2inhibitshuvecviabilitybyinhibitingthemapksignalingpathway
AT guyufeng downregulationofnlrp2inhibitshuvecviabilitybyinhibitingthemapksignalingpathway
AT qufuzheng downregulationofnlrp2inhibitshuvecviabilitybyinhibitingthemapksignalingpathway