Cargando…

The Novel Peptide AEDPPE Alleviates Trophoblast Cell Dysfunction Associated With Preeclampsia by Regulating the NF-κB Signaling Pathway

Background: Preeclampsia (PE) is a serious risk to the health of pregnant women and fetuses during pregnancy, and there is no effective treatment for this condition. Although many reports have confirmed the therapeutic effects of peptides in diseases, the role of peptides in PE remains poorly unders...

Descripción completa

Detalles Bibliográficos
Autores principales: Wang, Yixiao, Cao, Yan, Ji, Xiaohong, Li, Ting, Xue, Lu, Li, Chanjuan, Jia, Ruizhe, Ding, Hongjuan
Formato: Online Artículo Texto
Lenguaje:English
Publicado: Frontiers Media S.A. 2021
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC8719592/
https://www.ncbi.nlm.nih.gov/pubmed/34977169
http://dx.doi.org/10.3389/fcvm.2021.738378
_version_ 1784624968115421184
author Wang, Yixiao
Cao, Yan
Ji, Xiaohong
Li, Ting
Xue, Lu
Li, Chanjuan
Jia, Ruizhe
Ding, Hongjuan
author_facet Wang, Yixiao
Cao, Yan
Ji, Xiaohong
Li, Ting
Xue, Lu
Li, Chanjuan
Jia, Ruizhe
Ding, Hongjuan
author_sort Wang, Yixiao
collection PubMed
description Background: Preeclampsia (PE) is a serious risk to the health of pregnant women and fetuses during pregnancy, and there is no effective treatment for this condition. Although many reports have confirmed the therapeutic effects of peptides in diseases, the role of peptides in PE remains poorly understood. Methods: A differentially expressed peptide in PE (AEDPPE) is derived from heat-shock protein beta-1 (HSPB1), amino acids 100 to 109 (DVNHFAPDEL), which we identified in a previous study. We synthesized AEDPPE and investigated its effect on HTR-8/SVneo cell function using a Cell Counting Kit-8, flow cytometric assay, and Transwell and wound-healing assays. Quantitative reverse transcription-PCR and ELISA were used to determine cytokine expression. Pull-down assay, mass spectrometry, Western blot analysis, and immunofluorescence were used to explore the potential targets and signaling pathways regulated by AEDPPE. Finally, we assessed the effect of AEDPPE in the lipopolysaccharide (LPS)-induced PE-like rat model. Results: AEDPPE significantly promoted the migration and invasion of HTR-8/SVneo cells, and it decreased the expression of interleukins 1 beta (IL-1β), interleukin 6 (IL-6), and interleukin 8 (IL-8). These functions performed by AEDPPE remained evident after injury to HTR-8/SVneo cells with tumor necrosis factor-alpha (TNF-α), and AEDPPE reversed the elevated sFlt-1/PlGF ratio induced by TNF-α. AEDPPE may exert these biological effects by binding to heat-shock protein 90β (HSP 90β) and, thus, affect the NF-κB signaling pathway. In an LPS-induced PE-like rat model, AEDPPE significantly improved PE symptoms and fetal rat outcomes. Conclusion: Our study showed that AEDPPE enhanced trophoblast migration and invasion and reduced inflammatory cytokine expression, and we hypothesized that these actions involved the NF-κB signaling pathway. The use of AEDPPE may thus develop into a novel modality in the treatment of PE.
format Online
Article
Text
id pubmed-8719592
institution National Center for Biotechnology Information
language English
publishDate 2021
publisher Frontiers Media S.A.
record_format MEDLINE/PubMed
spelling pubmed-87195922022-01-01 The Novel Peptide AEDPPE Alleviates Trophoblast Cell Dysfunction Associated With Preeclampsia by Regulating the NF-κB Signaling Pathway Wang, Yixiao Cao, Yan Ji, Xiaohong Li, Ting Xue, Lu Li, Chanjuan Jia, Ruizhe Ding, Hongjuan Front Cardiovasc Med Cardiovascular Medicine Background: Preeclampsia (PE) is a serious risk to the health of pregnant women and fetuses during pregnancy, and there is no effective treatment for this condition. Although many reports have confirmed the therapeutic effects of peptides in diseases, the role of peptides in PE remains poorly understood. Methods: A differentially expressed peptide in PE (AEDPPE) is derived from heat-shock protein beta-1 (HSPB1), amino acids 100 to 109 (DVNHFAPDEL), which we identified in a previous study. We synthesized AEDPPE and investigated its effect on HTR-8/SVneo cell function using a Cell Counting Kit-8, flow cytometric assay, and Transwell and wound-healing assays. Quantitative reverse transcription-PCR and ELISA were used to determine cytokine expression. Pull-down assay, mass spectrometry, Western blot analysis, and immunofluorescence were used to explore the potential targets and signaling pathways regulated by AEDPPE. Finally, we assessed the effect of AEDPPE in the lipopolysaccharide (LPS)-induced PE-like rat model. Results: AEDPPE significantly promoted the migration and invasion of HTR-8/SVneo cells, and it decreased the expression of interleukins 1 beta (IL-1β), interleukin 6 (IL-6), and interleukin 8 (IL-8). These functions performed by AEDPPE remained evident after injury to HTR-8/SVneo cells with tumor necrosis factor-alpha (TNF-α), and AEDPPE reversed the elevated sFlt-1/PlGF ratio induced by TNF-α. AEDPPE may exert these biological effects by binding to heat-shock protein 90β (HSP 90β) and, thus, affect the NF-κB signaling pathway. In an LPS-induced PE-like rat model, AEDPPE significantly improved PE symptoms and fetal rat outcomes. Conclusion: Our study showed that AEDPPE enhanced trophoblast migration and invasion and reduced inflammatory cytokine expression, and we hypothesized that these actions involved the NF-κB signaling pathway. The use of AEDPPE may thus develop into a novel modality in the treatment of PE. Frontiers Media S.A. 2021-12-17 /pmc/articles/PMC8719592/ /pubmed/34977169 http://dx.doi.org/10.3389/fcvm.2021.738378 Text en Copyright © 2021 Wang, Cao, Ji, Li, Xue, Li, Jia and Ding. https://creativecommons.org/licenses/by/4.0/This is an open-access article distributed under the terms of the Creative Commons Attribution License (CC BY). The use, distribution or reproduction in other forums is permitted, provided the original author(s) and the copyright owner(s) are credited and that the original publication in this journal is cited, in accordance with accepted academic practice. No use, distribution or reproduction is permitted which does not comply with these terms.
spellingShingle Cardiovascular Medicine
Wang, Yixiao
Cao, Yan
Ji, Xiaohong
Li, Ting
Xue, Lu
Li, Chanjuan
Jia, Ruizhe
Ding, Hongjuan
The Novel Peptide AEDPPE Alleviates Trophoblast Cell Dysfunction Associated With Preeclampsia by Regulating the NF-κB Signaling Pathway
title The Novel Peptide AEDPPE Alleviates Trophoblast Cell Dysfunction Associated With Preeclampsia by Regulating the NF-κB Signaling Pathway
title_full The Novel Peptide AEDPPE Alleviates Trophoblast Cell Dysfunction Associated With Preeclampsia by Regulating the NF-κB Signaling Pathway
title_fullStr The Novel Peptide AEDPPE Alleviates Trophoblast Cell Dysfunction Associated With Preeclampsia by Regulating the NF-κB Signaling Pathway
title_full_unstemmed The Novel Peptide AEDPPE Alleviates Trophoblast Cell Dysfunction Associated With Preeclampsia by Regulating the NF-κB Signaling Pathway
title_short The Novel Peptide AEDPPE Alleviates Trophoblast Cell Dysfunction Associated With Preeclampsia by Regulating the NF-κB Signaling Pathway
title_sort novel peptide aedppe alleviates trophoblast cell dysfunction associated with preeclampsia by regulating the nf-κb signaling pathway
topic Cardiovascular Medicine
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC8719592/
https://www.ncbi.nlm.nih.gov/pubmed/34977169
http://dx.doi.org/10.3389/fcvm.2021.738378
work_keys_str_mv AT wangyixiao thenovelpeptideaedppealleviatestrophoblastcelldysfunctionassociatedwithpreeclampsiabyregulatingthenfkbsignalingpathway
AT caoyan thenovelpeptideaedppealleviatestrophoblastcelldysfunctionassociatedwithpreeclampsiabyregulatingthenfkbsignalingpathway
AT jixiaohong thenovelpeptideaedppealleviatestrophoblastcelldysfunctionassociatedwithpreeclampsiabyregulatingthenfkbsignalingpathway
AT liting thenovelpeptideaedppealleviatestrophoblastcelldysfunctionassociatedwithpreeclampsiabyregulatingthenfkbsignalingpathway
AT xuelu thenovelpeptideaedppealleviatestrophoblastcelldysfunctionassociatedwithpreeclampsiabyregulatingthenfkbsignalingpathway
AT lichanjuan thenovelpeptideaedppealleviatestrophoblastcelldysfunctionassociatedwithpreeclampsiabyregulatingthenfkbsignalingpathway
AT jiaruizhe thenovelpeptideaedppealleviatestrophoblastcelldysfunctionassociatedwithpreeclampsiabyregulatingthenfkbsignalingpathway
AT dinghongjuan thenovelpeptideaedppealleviatestrophoblastcelldysfunctionassociatedwithpreeclampsiabyregulatingthenfkbsignalingpathway
AT wangyixiao novelpeptideaedppealleviatestrophoblastcelldysfunctionassociatedwithpreeclampsiabyregulatingthenfkbsignalingpathway
AT caoyan novelpeptideaedppealleviatestrophoblastcelldysfunctionassociatedwithpreeclampsiabyregulatingthenfkbsignalingpathway
AT jixiaohong novelpeptideaedppealleviatestrophoblastcelldysfunctionassociatedwithpreeclampsiabyregulatingthenfkbsignalingpathway
AT liting novelpeptideaedppealleviatestrophoblastcelldysfunctionassociatedwithpreeclampsiabyregulatingthenfkbsignalingpathway
AT xuelu novelpeptideaedppealleviatestrophoblastcelldysfunctionassociatedwithpreeclampsiabyregulatingthenfkbsignalingpathway
AT lichanjuan novelpeptideaedppealleviatestrophoblastcelldysfunctionassociatedwithpreeclampsiabyregulatingthenfkbsignalingpathway
AT jiaruizhe novelpeptideaedppealleviatestrophoblastcelldysfunctionassociatedwithpreeclampsiabyregulatingthenfkbsignalingpathway
AT dinghongjuan novelpeptideaedppealleviatestrophoblastcelldysfunctionassociatedwithpreeclampsiabyregulatingthenfkbsignalingpathway