Cargando…

miR-320a/SP1 negative reciprocal interaction contributes to cell growth and invasion in colorectal cancer

BACKGROUND: Transcription factors (TFs) may be engaged in reciprocal regulatory circuits with certain miRNAs to maintain cellular homeostasis. Disequilibrium of the reciprocities by certain tumor-related stimuli may give rise to deregulation of downstream cellular signaling pathways, thus promoting...

Descripción completa

Detalles Bibliográficos
Autores principales: Zhang, Wenjing, Yang, Haitao, Wang, Zhongqiu, Wu, Yanting, Wang, Jingzhai, Duan, Guihua, Guo, Qiang, Zhang, Yu
Formato: Online Artículo Texto
Lenguaje:English
Publicado: BioMed Central 2021
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC7972346/
https://www.ncbi.nlm.nih.gov/pubmed/33731131
http://dx.doi.org/10.1186/s12935-021-01874-3
_version_ 1783666695989100544
author Zhang, Wenjing
Yang, Haitao
Wang, Zhongqiu
Wu, Yanting
Wang, Jingzhai
Duan, Guihua
Guo, Qiang
Zhang, Yu
author_facet Zhang, Wenjing
Yang, Haitao
Wang, Zhongqiu
Wu, Yanting
Wang, Jingzhai
Duan, Guihua
Guo, Qiang
Zhang, Yu
author_sort Zhang, Wenjing
collection PubMed
description BACKGROUND: Transcription factors (TFs) may be engaged in reciprocal regulatory circuits with certain miRNAs to maintain cellular homeostasis. Disequilibrium of the reciprocities by certain tumor-related stimuli may give rise to deregulation of downstream cellular signaling pathways, thus promoting malignant tumor phenotypes. Specificity Protein 1 (SP1) is the most representative member of the tumor-related transcription factors. Previous studies disclosed that SP1 can transcriptionally regulate miRNAs and coding genes to facilitate tumor progression. In our study, we used bioinformatic analysis to predict several SP1-binding sites within the miR-320a promoter and found that SP1 is a predicted target gene of miR-320a. Therefore, we hypothesize a reciprocal regulatory link between SP1 and miR-320a that participates in colorectal cancer (CRC) development METHODS: We performed bioinformatic analysis, quantitative polymerase chain reaction (qPCR), immunoblotting, dual-luciferase reporter assays, and a series of in vitro and in vivo functional assays to describe a novel SP1/miR-320a reciprocal interaction in CRC RESULTS: First, we found that miR-320a was significantly downregulated in CRC tissues and cell lines. Consistent with findings in other cancers, miR-320a exhibited inhibitory effects on cell growth and invasion of CRC in vitro and in vivo. Moreover, we identified SP1 as a target gene of miR-320a, and ectopic SP1 expression partly abolished miR-320a-induced inhibitory effects. Conversely, we confirmed that SP1 interacts with the miR-320a promoter, leading to depression of miR-320a. This illustrates a double-negative feedback loop between miR-320a and SP1. Additionally, based on the fact that SP1 promotes MACC1 transcription, we determined via immunoblotting that the oncogenic MACC1/MET signaling pathway was inactivated in the context of miR-320a-induced SP1 downregulation CONCLUSION: Taken together, our study is the first to describe a miR-320a/SP1 negative reciprocal interaction, which contributes to cell growth and invasion in CRC through modulation of the MACC1/MET signaling pathway.
format Online
Article
Text
id pubmed-7972346
institution National Center for Biotechnology Information
language English
publishDate 2021
publisher BioMed Central
record_format MEDLINE/PubMed
spelling pubmed-79723462021-03-19 miR-320a/SP1 negative reciprocal interaction contributes to cell growth and invasion in colorectal cancer Zhang, Wenjing Yang, Haitao Wang, Zhongqiu Wu, Yanting Wang, Jingzhai Duan, Guihua Guo, Qiang Zhang, Yu Cancer Cell Int Primary Research BACKGROUND: Transcription factors (TFs) may be engaged in reciprocal regulatory circuits with certain miRNAs to maintain cellular homeostasis. Disequilibrium of the reciprocities by certain tumor-related stimuli may give rise to deregulation of downstream cellular signaling pathways, thus promoting malignant tumor phenotypes. Specificity Protein 1 (SP1) is the most representative member of the tumor-related transcription factors. Previous studies disclosed that SP1 can transcriptionally regulate miRNAs and coding genes to facilitate tumor progression. In our study, we used bioinformatic analysis to predict several SP1-binding sites within the miR-320a promoter and found that SP1 is a predicted target gene of miR-320a. Therefore, we hypothesize a reciprocal regulatory link between SP1 and miR-320a that participates in colorectal cancer (CRC) development METHODS: We performed bioinformatic analysis, quantitative polymerase chain reaction (qPCR), immunoblotting, dual-luciferase reporter assays, and a series of in vitro and in vivo functional assays to describe a novel SP1/miR-320a reciprocal interaction in CRC RESULTS: First, we found that miR-320a was significantly downregulated in CRC tissues and cell lines. Consistent with findings in other cancers, miR-320a exhibited inhibitory effects on cell growth and invasion of CRC in vitro and in vivo. Moreover, we identified SP1 as a target gene of miR-320a, and ectopic SP1 expression partly abolished miR-320a-induced inhibitory effects. Conversely, we confirmed that SP1 interacts with the miR-320a promoter, leading to depression of miR-320a. This illustrates a double-negative feedback loop between miR-320a and SP1. Additionally, based on the fact that SP1 promotes MACC1 transcription, we determined via immunoblotting that the oncogenic MACC1/MET signaling pathway was inactivated in the context of miR-320a-induced SP1 downregulation CONCLUSION: Taken together, our study is the first to describe a miR-320a/SP1 negative reciprocal interaction, which contributes to cell growth and invasion in CRC through modulation of the MACC1/MET signaling pathway. BioMed Central 2021-03-17 /pmc/articles/PMC7972346/ /pubmed/33731131 http://dx.doi.org/10.1186/s12935-021-01874-3 Text en © The Author(s) 2021 Open AccessThis 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/. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated in a credit line to the data.
spellingShingle Primary Research
Zhang, Wenjing
Yang, Haitao
Wang, Zhongqiu
Wu, Yanting
Wang, Jingzhai
Duan, Guihua
Guo, Qiang
Zhang, Yu
miR-320a/SP1 negative reciprocal interaction contributes to cell growth and invasion in colorectal cancer
title miR-320a/SP1 negative reciprocal interaction contributes to cell growth and invasion in colorectal cancer
title_full miR-320a/SP1 negative reciprocal interaction contributes to cell growth and invasion in colorectal cancer
title_fullStr miR-320a/SP1 negative reciprocal interaction contributes to cell growth and invasion in colorectal cancer
title_full_unstemmed miR-320a/SP1 negative reciprocal interaction contributes to cell growth and invasion in colorectal cancer
title_short miR-320a/SP1 negative reciprocal interaction contributes to cell growth and invasion in colorectal cancer
title_sort mir-320a/sp1 negative reciprocal interaction contributes to cell growth and invasion in colorectal cancer
topic Primary Research
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC7972346/
https://www.ncbi.nlm.nih.gov/pubmed/33731131
http://dx.doi.org/10.1186/s12935-021-01874-3
work_keys_str_mv AT zhangwenjing mir320asp1negativereciprocalinteractioncontributestocellgrowthandinvasionincolorectalcancer
AT yanghaitao mir320asp1negativereciprocalinteractioncontributestocellgrowthandinvasionincolorectalcancer
AT wangzhongqiu mir320asp1negativereciprocalinteractioncontributestocellgrowthandinvasionincolorectalcancer
AT wuyanting mir320asp1negativereciprocalinteractioncontributestocellgrowthandinvasionincolorectalcancer
AT wangjingzhai mir320asp1negativereciprocalinteractioncontributestocellgrowthandinvasionincolorectalcancer
AT duanguihua mir320asp1negativereciprocalinteractioncontributestocellgrowthandinvasionincolorectalcancer
AT guoqiang mir320asp1negativereciprocalinteractioncontributestocellgrowthandinvasionincolorectalcancer
AT zhangyu mir320asp1negativereciprocalinteractioncontributestocellgrowthandinvasionincolorectalcancer