Cargando…

Micropeptide MIAC inhibits the tumor progression by interacting with AQP2 and inhibiting EREG/EGFR signaling in renal cell carcinoma

BACKGROUND: Although, micropeptides encoded by non-coding RNA have been shown to have an important role in a variety of tumors processes, there have been no reports on micropeptide in renal cell carcinoma (RCC). Based on the micropeptide MIAC (micropeptide inhibiting actin cytoskeleton) discovered a...

Descripción completa

Detalles Bibliográficos
Autores principales: Li, Mengwei, Liu, Guangxiang, Jin, Xinrong, Guo, Hongqian, Setrerrahmane, Sarra, Xu, Xindi, Li, Tiantian, Lin, Yunfei, Xu, Hanmei
Formato: Online Artículo Texto
Lenguaje:English
Publicado: BioMed Central 2022
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC9484220/
https://www.ncbi.nlm.nih.gov/pubmed/36117171
http://dx.doi.org/10.1186/s12943-022-01654-1
_version_ 1784791834522812416
author Li, Mengwei
Liu, Guangxiang
Jin, Xinrong
Guo, Hongqian
Setrerrahmane, Sarra
Xu, Xindi
Li, Tiantian
Lin, Yunfei
Xu, Hanmei
author_facet Li, Mengwei
Liu, Guangxiang
Jin, Xinrong
Guo, Hongqian
Setrerrahmane, Sarra
Xu, Xindi
Li, Tiantian
Lin, Yunfei
Xu, Hanmei
author_sort Li, Mengwei
collection PubMed
description BACKGROUND: Although, micropeptides encoded by non-coding RNA have been shown to have an important role in a variety of tumors processes, there have been no reports on micropeptide in renal cell carcinoma (RCC). Based on the micropeptide MIAC (micropeptide inhibiting actin cytoskeleton) discovered and named in the previous work, this study screened its tumor spectrum, and explored its mechanism of action and potential diagnosis and treatment value in the occurrence and development of renal carcinoma. METHODS: The clinical significance of MIAC in RCC was explored by bioinformatics analysis through high-throughput RNA-seq data from 530 patients with kidney renal clear cell carcinoma (KIRC) in the TCGA database, and the detection of clinical samples of 70 cases of kidney cancer. In vitro and in vivo experiments to determine the role of MIAC in renal carcinoma cell growth and metastasis; High-throughput transcriptomics, western blotting, immunoprecipitation, molecular docking, affinity experiments, and Streptavidin pulldown experiments identify MIAC direct binding protein and key regulatory pathways. RESULTS: The analysis of 600 renal carcinoma samples from different sources revealed that the expression level of MIAC is significantly decreased, and corelated with the prognosis and clinical stage of tumors in patients with renal carcinoma. Overexpression of MIAC in renal carcinoma cells can significantly inhibit the proliferation and migration ability, promote apoptosis of renal carcinoma cells, and affect the distribution of cells at various stages. After knocking down MIAC, the trend is reversed. In vivo experiments have found that MIAC overexpression inhibit the growth and metastasis of RCC, while the synthetized MIAC peptides can significantly inhibit the occurrence and development of RCC in vitro and in vivo. Further mechanistic studies have demonstrated that MIAC directly bind to AQP2 protein, inhibit EREG/EGFR expression and activate downstream pathways PI3K/AKT and MAPK to achieve anti-tumor effects. CONCLUSIONS: This study revealed for the first time the tumor suppressor potential of the lncRNA-encoded micropeptide MIAC in RCC, which inhibits the activation of the EREG/EGFR signaling pathway by direct binding to AQP2 protein, thereby inhibiting renal carcinoma progression and metastasis. This result emphasizes that the micropeptide MIAC can provide a new strategy for the diagnosis and treatment of RCC. SUPPLEMENTARY INFORMATION: The online version contains supplementary material available at 10.1186/s12943-022-01654-1.
format Online
Article
Text
id pubmed-9484220
institution National Center for Biotechnology Information
language English
publishDate 2022
publisher BioMed Central
record_format MEDLINE/PubMed
spelling pubmed-94842202022-09-20 Micropeptide MIAC inhibits the tumor progression by interacting with AQP2 and inhibiting EREG/EGFR signaling in renal cell carcinoma Li, Mengwei Liu, Guangxiang Jin, Xinrong Guo, Hongqian Setrerrahmane, Sarra Xu, Xindi Li, Tiantian Lin, Yunfei Xu, Hanmei Mol Cancer Research BACKGROUND: Although, micropeptides encoded by non-coding RNA have been shown to have an important role in a variety of tumors processes, there have been no reports on micropeptide in renal cell carcinoma (RCC). Based on the micropeptide MIAC (micropeptide inhibiting actin cytoskeleton) discovered and named in the previous work, this study screened its tumor spectrum, and explored its mechanism of action and potential diagnosis and treatment value in the occurrence and development of renal carcinoma. METHODS: The clinical significance of MIAC in RCC was explored by bioinformatics analysis through high-throughput RNA-seq data from 530 patients with kidney renal clear cell carcinoma (KIRC) in the TCGA database, and the detection of clinical samples of 70 cases of kidney cancer. In vitro and in vivo experiments to determine the role of MIAC in renal carcinoma cell growth and metastasis; High-throughput transcriptomics, western blotting, immunoprecipitation, molecular docking, affinity experiments, and Streptavidin pulldown experiments identify MIAC direct binding protein and key regulatory pathways. RESULTS: The analysis of 600 renal carcinoma samples from different sources revealed that the expression level of MIAC is significantly decreased, and corelated with the prognosis and clinical stage of tumors in patients with renal carcinoma. Overexpression of MIAC in renal carcinoma cells can significantly inhibit the proliferation and migration ability, promote apoptosis of renal carcinoma cells, and affect the distribution of cells at various stages. After knocking down MIAC, the trend is reversed. In vivo experiments have found that MIAC overexpression inhibit the growth and metastasis of RCC, while the synthetized MIAC peptides can significantly inhibit the occurrence and development of RCC in vitro and in vivo. Further mechanistic studies have demonstrated that MIAC directly bind to AQP2 protein, inhibit EREG/EGFR expression and activate downstream pathways PI3K/AKT and MAPK to achieve anti-tumor effects. CONCLUSIONS: This study revealed for the first time the tumor suppressor potential of the lncRNA-encoded micropeptide MIAC in RCC, which inhibits the activation of the EREG/EGFR signaling pathway by direct binding to AQP2 protein, thereby inhibiting renal carcinoma progression and metastasis. This result emphasizes that the micropeptide MIAC can provide a new strategy for the diagnosis and treatment of RCC. SUPPLEMENTARY INFORMATION: The online version contains supplementary material available at 10.1186/s12943-022-01654-1. BioMed Central 2022-09-19 /pmc/articles/PMC9484220/ /pubmed/36117171 http://dx.doi.org/10.1186/s12943-022-01654-1 Text en © The Author(s) 2022 https://creativecommons.org/licenses/by/4.0/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/ (https://creativecommons.org/licenses/by/4.0/) . The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/ (https://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 Research
Li, Mengwei
Liu, Guangxiang
Jin, Xinrong
Guo, Hongqian
Setrerrahmane, Sarra
Xu, Xindi
Li, Tiantian
Lin, Yunfei
Xu, Hanmei
Micropeptide MIAC inhibits the tumor progression by interacting with AQP2 and inhibiting EREG/EGFR signaling in renal cell carcinoma
title Micropeptide MIAC inhibits the tumor progression by interacting with AQP2 and inhibiting EREG/EGFR signaling in renal cell carcinoma
title_full Micropeptide MIAC inhibits the tumor progression by interacting with AQP2 and inhibiting EREG/EGFR signaling in renal cell carcinoma
title_fullStr Micropeptide MIAC inhibits the tumor progression by interacting with AQP2 and inhibiting EREG/EGFR signaling in renal cell carcinoma
title_full_unstemmed Micropeptide MIAC inhibits the tumor progression by interacting with AQP2 and inhibiting EREG/EGFR signaling in renal cell carcinoma
title_short Micropeptide MIAC inhibits the tumor progression by interacting with AQP2 and inhibiting EREG/EGFR signaling in renal cell carcinoma
title_sort micropeptide miac inhibits the tumor progression by interacting with aqp2 and inhibiting ereg/egfr signaling in renal cell carcinoma
topic Research
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC9484220/
https://www.ncbi.nlm.nih.gov/pubmed/36117171
http://dx.doi.org/10.1186/s12943-022-01654-1
work_keys_str_mv AT limengwei micropeptidemiacinhibitsthetumorprogressionbyinteractingwithaqp2andinhibitingeregegfrsignalinginrenalcellcarcinoma
AT liuguangxiang micropeptidemiacinhibitsthetumorprogressionbyinteractingwithaqp2andinhibitingeregegfrsignalinginrenalcellcarcinoma
AT jinxinrong micropeptidemiacinhibitsthetumorprogressionbyinteractingwithaqp2andinhibitingeregegfrsignalinginrenalcellcarcinoma
AT guohongqian micropeptidemiacinhibitsthetumorprogressionbyinteractingwithaqp2andinhibitingeregegfrsignalinginrenalcellcarcinoma
AT setrerrahmanesarra micropeptidemiacinhibitsthetumorprogressionbyinteractingwithaqp2andinhibitingeregegfrsignalinginrenalcellcarcinoma
AT xuxindi micropeptidemiacinhibitsthetumorprogressionbyinteractingwithaqp2andinhibitingeregegfrsignalinginrenalcellcarcinoma
AT litiantian micropeptidemiacinhibitsthetumorprogressionbyinteractingwithaqp2andinhibitingeregegfrsignalinginrenalcellcarcinoma
AT linyunfei micropeptidemiacinhibitsthetumorprogressionbyinteractingwithaqp2andinhibitingeregegfrsignalinginrenalcellcarcinoma
AT xuhanmei micropeptidemiacinhibitsthetumorprogressionbyinteractingwithaqp2andinhibitingeregegfrsignalinginrenalcellcarcinoma