Cargando…

The COL-4A1 polypeptide destroy endothelial cells through the TGF-β/PI3K/AKT pathway

Preeclampsia (PE) is commonly considered as a placental disorder in pregnancy. Until now, the etiology and pathological mechanism of PE have remained ambiguous. Although PE can lead to a variety of maternal and infant complications, there are still no effective treatments. This study aimed to explor...

Descripción completa

Detalles Bibliográficos
Autores principales: Li, Ting, Ling, Zhonghui, Xie, Kaipeng, Wang, Yixiao, Miao, Zhijing, Ji, Xiaohong, Li, Jingyun, Hou, Wenwen, Tang, Qiuqin, Yuan, Xiaojie, Li, Nan, Li, Chanjuan, Ding, Hongjuan
Formato: Online Artículo Texto
Lenguaje:English
Publicado: Nature Publishing Group UK 2021
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC8333066/
https://www.ncbi.nlm.nih.gov/pubmed/34344927
http://dx.doi.org/10.1038/s41598-021-94801-5
_version_ 1783732963710599168
author Li, Ting
Ling, Zhonghui
Xie, Kaipeng
Wang, Yixiao
Miao, Zhijing
Ji, Xiaohong
Li, Jingyun
Hou, Wenwen
Tang, Qiuqin
Yuan, Xiaojie
Li, Nan
Li, Chanjuan
Ding, Hongjuan
author_facet Li, Ting
Ling, Zhonghui
Xie, Kaipeng
Wang, Yixiao
Miao, Zhijing
Ji, Xiaohong
Li, Jingyun
Hou, Wenwen
Tang, Qiuqin
Yuan, Xiaojie
Li, Nan
Li, Chanjuan
Ding, Hongjuan
author_sort Li, Ting
collection PubMed
description Preeclampsia (PE) is commonly considered as a placental disorder in pregnancy. Until now, the etiology and pathological mechanism of PE have remained ambiguous. Although PE can lead to a variety of maternal and infant complications, there are still no effective treatments. This study aimed to explore the correlation between the novel polypeptide COL-4A1 and PE, and to identify the underlying mechanism by which this polypeptide may function and to explore new therapeutic targets for PE. A rat model of PE was established and used to verify the function of the polypeptide COL-4A1 in vivo. Additionally, human umbilical vascular endothelial cells (HUVECs) were cultured with or without COL-4A1 and TNF-α (20 ng/ml). Cell Counting Kit-8 (CCK-8), wound-healing, Transwell and tube formation assays were used to evaluate cell proliferation, migration and angiopoiesis. RNA sequencing and mass spectrometry were conducted to explore the underlying downstream mechanism of COL-4A1. In vivo, COL-4A1 increased blood pressure and elevated the risk of fetal growth restriction (FGR) which was induced by lipopolysaccharide (LPS) in the rat model. In vitro, COL-4A1 significantly inhibited the proliferation and migration of HUVECs. After culture with COL-4A1, compared to control group the adhesive ability and level of reactive oxygen species (ROS) were enhanced and tube formation ability was decreased. Furthermore, Western blotting (WB) and pull-down assays were conducted to explore the underlying mechanism by which COL-4A1 functions, and the TGF-β/PI3K/AKT pathway was identified as the potential pathway involved in its effects. In summary, these results revealed that the polypeptide COL-4A1 caused PE-like symptoms in cells and a rat model. Through the TGF-β/PI3K/AKT pathway, COL-4A1 interferes with the pathogenesis of PE. Thus COL-4A1 is expected to become a potential target of PE, providing a basis for exploring the treatment of PE.
format Online
Article
Text
id pubmed-8333066
institution National Center for Biotechnology Information
language English
publishDate 2021
publisher Nature Publishing Group UK
record_format MEDLINE/PubMed
spelling pubmed-83330662021-08-04 The COL-4A1 polypeptide destroy endothelial cells through the TGF-β/PI3K/AKT pathway Li, Ting Ling, Zhonghui Xie, Kaipeng Wang, Yixiao Miao, Zhijing Ji, Xiaohong Li, Jingyun Hou, Wenwen Tang, Qiuqin Yuan, Xiaojie Li, Nan Li, Chanjuan Ding, Hongjuan Sci Rep Article Preeclampsia (PE) is commonly considered as a placental disorder in pregnancy. Until now, the etiology and pathological mechanism of PE have remained ambiguous. Although PE can lead to a variety of maternal and infant complications, there are still no effective treatments. This study aimed to explore the correlation between the novel polypeptide COL-4A1 and PE, and to identify the underlying mechanism by which this polypeptide may function and to explore new therapeutic targets for PE. A rat model of PE was established and used to verify the function of the polypeptide COL-4A1 in vivo. Additionally, human umbilical vascular endothelial cells (HUVECs) were cultured with or without COL-4A1 and TNF-α (20 ng/ml). Cell Counting Kit-8 (CCK-8), wound-healing, Transwell and tube formation assays were used to evaluate cell proliferation, migration and angiopoiesis. RNA sequencing and mass spectrometry were conducted to explore the underlying downstream mechanism of COL-4A1. In vivo, COL-4A1 increased blood pressure and elevated the risk of fetal growth restriction (FGR) which was induced by lipopolysaccharide (LPS) in the rat model. In vitro, COL-4A1 significantly inhibited the proliferation and migration of HUVECs. After culture with COL-4A1, compared to control group the adhesive ability and level of reactive oxygen species (ROS) were enhanced and tube formation ability was decreased. Furthermore, Western blotting (WB) and pull-down assays were conducted to explore the underlying mechanism by which COL-4A1 functions, and the TGF-β/PI3K/AKT pathway was identified as the potential pathway involved in its effects. In summary, these results revealed that the polypeptide COL-4A1 caused PE-like symptoms in cells and a rat model. Through the TGF-β/PI3K/AKT pathway, COL-4A1 interferes with the pathogenesis of PE. Thus COL-4A1 is expected to become a potential target of PE, providing a basis for exploring the treatment of PE. Nature Publishing Group UK 2021-08-03 /pmc/articles/PMC8333066/ /pubmed/34344927 http://dx.doi.org/10.1038/s41598-021-94801-5 Text en © The Author(s) 2021 https://creativecommons.org/licenses/by/4.0/Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/ (https://creativecommons.org/licenses/by/4.0/) .
spellingShingle Article
Li, Ting
Ling, Zhonghui
Xie, Kaipeng
Wang, Yixiao
Miao, Zhijing
Ji, Xiaohong
Li, Jingyun
Hou, Wenwen
Tang, Qiuqin
Yuan, Xiaojie
Li, Nan
Li, Chanjuan
Ding, Hongjuan
The COL-4A1 polypeptide destroy endothelial cells through the TGF-β/PI3K/AKT pathway
title The COL-4A1 polypeptide destroy endothelial cells through the TGF-β/PI3K/AKT pathway
title_full The COL-4A1 polypeptide destroy endothelial cells through the TGF-β/PI3K/AKT pathway
title_fullStr The COL-4A1 polypeptide destroy endothelial cells through the TGF-β/PI3K/AKT pathway
title_full_unstemmed The COL-4A1 polypeptide destroy endothelial cells through the TGF-β/PI3K/AKT pathway
title_short The COL-4A1 polypeptide destroy endothelial cells through the TGF-β/PI3K/AKT pathway
title_sort col-4a1 polypeptide destroy endothelial cells through the tgf-β/pi3k/akt pathway
topic Article
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC8333066/
https://www.ncbi.nlm.nih.gov/pubmed/34344927
http://dx.doi.org/10.1038/s41598-021-94801-5
work_keys_str_mv AT liting thecol4a1polypeptidedestroyendothelialcellsthroughthetgfbpi3kaktpathway
AT lingzhonghui thecol4a1polypeptidedestroyendothelialcellsthroughthetgfbpi3kaktpathway
AT xiekaipeng thecol4a1polypeptidedestroyendothelialcellsthroughthetgfbpi3kaktpathway
AT wangyixiao thecol4a1polypeptidedestroyendothelialcellsthroughthetgfbpi3kaktpathway
AT miaozhijing thecol4a1polypeptidedestroyendothelialcellsthroughthetgfbpi3kaktpathway
AT jixiaohong thecol4a1polypeptidedestroyendothelialcellsthroughthetgfbpi3kaktpathway
AT lijingyun thecol4a1polypeptidedestroyendothelialcellsthroughthetgfbpi3kaktpathway
AT houwenwen thecol4a1polypeptidedestroyendothelialcellsthroughthetgfbpi3kaktpathway
AT tangqiuqin thecol4a1polypeptidedestroyendothelialcellsthroughthetgfbpi3kaktpathway
AT yuanxiaojie thecol4a1polypeptidedestroyendothelialcellsthroughthetgfbpi3kaktpathway
AT linan thecol4a1polypeptidedestroyendothelialcellsthroughthetgfbpi3kaktpathway
AT lichanjuan thecol4a1polypeptidedestroyendothelialcellsthroughthetgfbpi3kaktpathway
AT dinghongjuan thecol4a1polypeptidedestroyendothelialcellsthroughthetgfbpi3kaktpathway
AT liting col4a1polypeptidedestroyendothelialcellsthroughthetgfbpi3kaktpathway
AT lingzhonghui col4a1polypeptidedestroyendothelialcellsthroughthetgfbpi3kaktpathway
AT xiekaipeng col4a1polypeptidedestroyendothelialcellsthroughthetgfbpi3kaktpathway
AT wangyixiao col4a1polypeptidedestroyendothelialcellsthroughthetgfbpi3kaktpathway
AT miaozhijing col4a1polypeptidedestroyendothelialcellsthroughthetgfbpi3kaktpathway
AT jixiaohong col4a1polypeptidedestroyendothelialcellsthroughthetgfbpi3kaktpathway
AT lijingyun col4a1polypeptidedestroyendothelialcellsthroughthetgfbpi3kaktpathway
AT houwenwen col4a1polypeptidedestroyendothelialcellsthroughthetgfbpi3kaktpathway
AT tangqiuqin col4a1polypeptidedestroyendothelialcellsthroughthetgfbpi3kaktpathway
AT yuanxiaojie col4a1polypeptidedestroyendothelialcellsthroughthetgfbpi3kaktpathway
AT linan col4a1polypeptidedestroyendothelialcellsthroughthetgfbpi3kaktpathway
AT lichanjuan col4a1polypeptidedestroyendothelialcellsthroughthetgfbpi3kaktpathway
AT dinghongjuan col4a1polypeptidedestroyendothelialcellsthroughthetgfbpi3kaktpathway