Cargando…

MALAT1 sponges miR-26a and miR-26b to regulate endothelial cell angiogenesis via PFKFB3-driven glycolysis in early-onset preeclampsia

6-phosphofructo-2-kinase (PFKFB3) is a crucial regulator of glycolysis that has been implicated in angiogenesis and the development of diverse diseases. However, the functional role and regulatory mechanism of PFKFB3 in early-onset preeclampsia (EOPE) remain to be elucidated. According to previous s...

Descripción completa

Detalles Bibliográficos
Autores principales: Li, Qi, Liu, Xiaoxia, Liu, Weifang, Zhang, Yang, Wu, Mengying, Chen, Zhirui, Zhao, Yin, Zou, Li
Formato: Online Artículo Texto
Lenguaje:English
Publicado: American Society of Gene & Cell Therapy 2021
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC7868745/
https://www.ncbi.nlm.nih.gov/pubmed/33614238
http://dx.doi.org/10.1016/j.omtn.2021.01.005
_version_ 1783648513847984128
author Li, Qi
Liu, Xiaoxia
Liu, Weifang
Zhang, Yang
Wu, Mengying
Chen, Zhirui
Zhao, Yin
Zou, Li
author_facet Li, Qi
Liu, Xiaoxia
Liu, Weifang
Zhang, Yang
Wu, Mengying
Chen, Zhirui
Zhao, Yin
Zou, Li
author_sort Li, Qi
collection PubMed
description 6-phosphofructo-2-kinase (PFKFB3) is a crucial regulator of glycolysis that has been implicated in angiogenesis and the development of diverse diseases. However, the functional role and regulatory mechanism of PFKFB3 in early-onset preeclampsia (EOPE) remain to be elucidated. According to previous studies, noncoding RNAs play crucial roles in EOPE pathogenesis. The goal of this study was to investigate the functional roles and co-regulatory mechanisms of the metastasis-associated lung adenocarcinoma transcript-1 (MALAT1)/microRNA (miR)-26/PFKFB3 axis in EOPE. In our study, decreased MALAT1 and PFKFB3 expression in EOPE tissues correlates with endothelial cell (EC) dysfunction. The results of in vitro assays revealed that PFKFB3 regulates the proliferation, migration, and tube formation of ECs by modulating glycolysis. Furthermore, MALAT1 regulates PFKFB3 expression by sponging miR-26a/26b. Finally, MALAT1 knockout reduces EC angiogenesis by inhibiting PFKFB3-mediated glycolysis flux, which is ameliorated by PFKFB3 overexpression. In conclusion, decreased MALAT1 expression in EOPE tissues reduces the glycolysis of ECs in a PFKFB3-dependent manner by sponging miR-26a/26b and inhibits EC proliferation, migration, and tube formation, which may contribute to abnormal angiogenesis in EOPE. Thus, strategies targeting PFKFB3-driven glycolysis may be a promising approach for the treatment of EOPE.
format Online
Article
Text
id pubmed-7868745
institution National Center for Biotechnology Information
language English
publishDate 2021
publisher American Society of Gene & Cell Therapy
record_format MEDLINE/PubMed
spelling pubmed-78687452021-02-19 MALAT1 sponges miR-26a and miR-26b to regulate endothelial cell angiogenesis via PFKFB3-driven glycolysis in early-onset preeclampsia Li, Qi Liu, Xiaoxia Liu, Weifang Zhang, Yang Wu, Mengying Chen, Zhirui Zhao, Yin Zou, Li Mol Ther Nucleic Acids Original Article 6-phosphofructo-2-kinase (PFKFB3) is a crucial regulator of glycolysis that has been implicated in angiogenesis and the development of diverse diseases. However, the functional role and regulatory mechanism of PFKFB3 in early-onset preeclampsia (EOPE) remain to be elucidated. According to previous studies, noncoding RNAs play crucial roles in EOPE pathogenesis. The goal of this study was to investigate the functional roles and co-regulatory mechanisms of the metastasis-associated lung adenocarcinoma transcript-1 (MALAT1)/microRNA (miR)-26/PFKFB3 axis in EOPE. In our study, decreased MALAT1 and PFKFB3 expression in EOPE tissues correlates with endothelial cell (EC) dysfunction. The results of in vitro assays revealed that PFKFB3 regulates the proliferation, migration, and tube formation of ECs by modulating glycolysis. Furthermore, MALAT1 regulates PFKFB3 expression by sponging miR-26a/26b. Finally, MALAT1 knockout reduces EC angiogenesis by inhibiting PFKFB3-mediated glycolysis flux, which is ameliorated by PFKFB3 overexpression. In conclusion, decreased MALAT1 expression in EOPE tissues reduces the glycolysis of ECs in a PFKFB3-dependent manner by sponging miR-26a/26b and inhibits EC proliferation, migration, and tube formation, which may contribute to abnormal angiogenesis in EOPE. Thus, strategies targeting PFKFB3-driven glycolysis may be a promising approach for the treatment of EOPE. American Society of Gene & Cell Therapy 2021-01-16 /pmc/articles/PMC7868745/ /pubmed/33614238 http://dx.doi.org/10.1016/j.omtn.2021.01.005 Text en © 2021 The Author(s) http://creativecommons.org/licenses/by-nc-nd/4.0/ This is an open access article under the CC BY-NC-ND license (http://creativecommons.org/licenses/by-nc-nd/4.0/).
spellingShingle Original Article
Li, Qi
Liu, Xiaoxia
Liu, Weifang
Zhang, Yang
Wu, Mengying
Chen, Zhirui
Zhao, Yin
Zou, Li
MALAT1 sponges miR-26a and miR-26b to regulate endothelial cell angiogenesis via PFKFB3-driven glycolysis in early-onset preeclampsia
title MALAT1 sponges miR-26a and miR-26b to regulate endothelial cell angiogenesis via PFKFB3-driven glycolysis in early-onset preeclampsia
title_full MALAT1 sponges miR-26a and miR-26b to regulate endothelial cell angiogenesis via PFKFB3-driven glycolysis in early-onset preeclampsia
title_fullStr MALAT1 sponges miR-26a and miR-26b to regulate endothelial cell angiogenesis via PFKFB3-driven glycolysis in early-onset preeclampsia
title_full_unstemmed MALAT1 sponges miR-26a and miR-26b to regulate endothelial cell angiogenesis via PFKFB3-driven glycolysis in early-onset preeclampsia
title_short MALAT1 sponges miR-26a and miR-26b to regulate endothelial cell angiogenesis via PFKFB3-driven glycolysis in early-onset preeclampsia
title_sort malat1 sponges mir-26a and mir-26b to regulate endothelial cell angiogenesis via pfkfb3-driven glycolysis in early-onset preeclampsia
topic Original Article
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC7868745/
https://www.ncbi.nlm.nih.gov/pubmed/33614238
http://dx.doi.org/10.1016/j.omtn.2021.01.005
work_keys_str_mv AT liqi malat1spongesmir26aandmir26btoregulateendothelialcellangiogenesisviapfkfb3drivenglycolysisinearlyonsetpreeclampsia
AT liuxiaoxia malat1spongesmir26aandmir26btoregulateendothelialcellangiogenesisviapfkfb3drivenglycolysisinearlyonsetpreeclampsia
AT liuweifang malat1spongesmir26aandmir26btoregulateendothelialcellangiogenesisviapfkfb3drivenglycolysisinearlyonsetpreeclampsia
AT zhangyang malat1spongesmir26aandmir26btoregulateendothelialcellangiogenesisviapfkfb3drivenglycolysisinearlyonsetpreeclampsia
AT wumengying malat1spongesmir26aandmir26btoregulateendothelialcellangiogenesisviapfkfb3drivenglycolysisinearlyonsetpreeclampsia
AT chenzhirui malat1spongesmir26aandmir26btoregulateendothelialcellangiogenesisviapfkfb3drivenglycolysisinearlyonsetpreeclampsia
AT zhaoyin malat1spongesmir26aandmir26btoregulateendothelialcellangiogenesisviapfkfb3drivenglycolysisinearlyonsetpreeclampsia
AT zouli malat1spongesmir26aandmir26btoregulateendothelialcellangiogenesisviapfkfb3drivenglycolysisinearlyonsetpreeclampsia