Long non-coding RNA MIDEAS-AS1 inhibits growth and metastasis of triple-negative breast cancer via transcriptionally activating NCALD

Abstract Background Triple-negative breast cancer (TNBC) is a subtype of breast cancer with higher aggressiveness and poorer outcomes. Recently, long non-coding RNAs (lncRNAs) have become the crucial gene regulators in the progression of human cancers. However, the function and underlying mechanisms...

Full description

Bibliographic Details
Main Authors: Dan Luo, Yiran Liang, Yajie Wang, Fangzhou Ye, Yuhan Jin, Yaming Li, Dianwen Han, Zekun Wang, Bing Chen, Wenjing Zhao, Lijuan Wang, Xi Chen, Liyu Jiang, Qifeng Yang
Format: Article
Language:English
Published: BMC 2023-09-01
Series:Breast Cancer Research
Subjects:
Online Access:https://doi.org/10.1186/s13058-023-01709-1
_version_ 1797555529804939264
author Dan Luo
Yiran Liang
Yajie Wang
Fangzhou Ye
Yuhan Jin
Yaming Li
Dianwen Han
Zekun Wang
Bing Chen
Wenjing Zhao
Lijuan Wang
Xi Chen
Liyu Jiang
Qifeng Yang
author_facet Dan Luo
Yiran Liang
Yajie Wang
Fangzhou Ye
Yuhan Jin
Yaming Li
Dianwen Han
Zekun Wang
Bing Chen
Wenjing Zhao
Lijuan Wang
Xi Chen
Liyu Jiang
Qifeng Yang
author_sort Dan Luo
collection DOAJ
description Abstract Background Triple-negative breast cancer (TNBC) is a subtype of breast cancer with higher aggressiveness and poorer outcomes. Recently, long non-coding RNAs (lncRNAs) have become the crucial gene regulators in the progression of human cancers. However, the function and underlying mechanisms of lncRNAs in TNBC remains unclear. Methods Based on public databases and bioinformatics analyses, the low expression of lncRNA MIDEAS-AS1 in breast cancer tissues was detected and further validated in a cohort of TNBC tissues. The effects of MIDEAS-AS1 on proliferation, migration, invasion were determined by in vitro and in vivo experiments. RNA pull-down assay and RNA immunoprecipitation (RIP) assay were carried out to reveal the interaction between MIDEAS-AS1 and MATR3. Luciferase reporter assay, Chromatin immunoprecipitation (ChIP) and qRT-PCR were used to evaluate the regulatory effect of MIDEAS-AS1/MATR3 complex on NCALD. Results LncRNA MIDEAS-AS1 was significantly downregulated in TNBC, which was correlated with poor overall survival (OS) and progression-free survival (PFS) in TNBC patients. MIDEAS-AS1 overexpression remarkably inhibited tumor growth and metastasis in vitro and in vivo. Mechanistically, MIDEAS-AS1 mainly located in the nucleus and interacted with the nuclear protein MATR3. Meanwhile, NCALD was selected as the downstream target, which was transcriptionally regulated by MIDEAS-AS1/MATR3 complex and further inactivated NF-κB signaling pathway. Furthermore, rescue experiment showed that the suppression of cell malignant phenotype caused by MIDEAS-AS1 overexpression could be reversed by inhibition of NCALD. Conclusions Collectively, our results demonstrate that MIDEAS-AS1 serves as a tumor-suppressor in TNBC through modulating MATR3/NCALD axis, and MIDEAS-AS1 may function as a prognostic biomarker for TNBC.
first_indexed 2024-03-10T16:48:48Z
format Article
id doaj.art-7327a182b2c044528901ba7944b4436f
institution Directory Open Access Journal
issn 1465-542X
language English
last_indexed 2024-03-10T16:48:48Z
publishDate 2023-09-01
publisher BMC
record_format Article
series Breast Cancer Research
spelling doaj.art-7327a182b2c044528901ba7944b4436f2023-11-20T11:22:02ZengBMCBreast Cancer Research1465-542X2023-09-0125112010.1186/s13058-023-01709-1Long non-coding RNA MIDEAS-AS1 inhibits growth and metastasis of triple-negative breast cancer via transcriptionally activating NCALDDan Luo0Yiran Liang1Yajie Wang2Fangzhou Ye3Yuhan Jin4Yaming Li5Dianwen Han6Zekun Wang7Bing Chen8Wenjing Zhao9Lijuan Wang10Xi Chen11Liyu Jiang12Qifeng Yang13Department of Breast Surgery, General Surgery, Qilu Hospital, Cheeloo College of Medicine, Shandong UniversityDepartment of Breast Surgery, General Surgery, Qilu Hospital, Cheeloo College of Medicine, Shandong UniversityDepartment of Breast Surgery, General Surgery, Qilu Hospital, Cheeloo College of Medicine, Shandong UniversityDepartment of Breast Surgery, General Surgery, Qilu Hospital, Cheeloo College of Medicine, Shandong UniversityDepartment of Breast Surgery, General Surgery, Qilu Hospital, Cheeloo College of Medicine, Shandong UniversityDepartment of Breast Surgery, General Surgery, Qilu Hospital, Cheeloo College of Medicine, Shandong UniversityDepartment of Breast Surgery, General Surgery, Qilu Hospital, Cheeloo College of Medicine, Shandong UniversityDepartment of Breast Surgery, General Surgery, Qilu Hospital, Cheeloo College of Medicine, Shandong UniversityResearch Institute of Breast Cancer, Shandong UniversityResearch Institute of Breast Cancer, Shandong UniversityResearch Institute of Breast Cancer, Shandong UniversityDepartment of Breast Surgery, General Surgery, Qilu Hospital, Cheeloo College of Medicine, Shandong UniversityDepartment of Breast Surgery, General Surgery, Qilu Hospital, Cheeloo College of Medicine, Shandong UniversityDepartment of Breast Surgery, General Surgery, Qilu Hospital, Cheeloo College of Medicine, Shandong UniversityAbstract Background Triple-negative breast cancer (TNBC) is a subtype of breast cancer with higher aggressiveness and poorer outcomes. Recently, long non-coding RNAs (lncRNAs) have become the crucial gene regulators in the progression of human cancers. However, the function and underlying mechanisms of lncRNAs in TNBC remains unclear. Methods Based on public databases and bioinformatics analyses, the low expression of lncRNA MIDEAS-AS1 in breast cancer tissues was detected and further validated in a cohort of TNBC tissues. The effects of MIDEAS-AS1 on proliferation, migration, invasion were determined by in vitro and in vivo experiments. RNA pull-down assay and RNA immunoprecipitation (RIP) assay were carried out to reveal the interaction between MIDEAS-AS1 and MATR3. Luciferase reporter assay, Chromatin immunoprecipitation (ChIP) and qRT-PCR were used to evaluate the regulatory effect of MIDEAS-AS1/MATR3 complex on NCALD. Results LncRNA MIDEAS-AS1 was significantly downregulated in TNBC, which was correlated with poor overall survival (OS) and progression-free survival (PFS) in TNBC patients. MIDEAS-AS1 overexpression remarkably inhibited tumor growth and metastasis in vitro and in vivo. Mechanistically, MIDEAS-AS1 mainly located in the nucleus and interacted with the nuclear protein MATR3. Meanwhile, NCALD was selected as the downstream target, which was transcriptionally regulated by MIDEAS-AS1/MATR3 complex and further inactivated NF-κB signaling pathway. Furthermore, rescue experiment showed that the suppression of cell malignant phenotype caused by MIDEAS-AS1 overexpression could be reversed by inhibition of NCALD. Conclusions Collectively, our results demonstrate that MIDEAS-AS1 serves as a tumor-suppressor in TNBC through modulating MATR3/NCALD axis, and MIDEAS-AS1 may function as a prognostic biomarker for TNBC.https://doi.org/10.1186/s13058-023-01709-1Triple-negative breast cancerMIDEAS-AS1MATR3NCALDPrognosis
spellingShingle Dan Luo
Yiran Liang
Yajie Wang
Fangzhou Ye
Yuhan Jin
Yaming Li
Dianwen Han
Zekun Wang
Bing Chen
Wenjing Zhao
Lijuan Wang
Xi Chen
Liyu Jiang
Qifeng Yang
Long non-coding RNA MIDEAS-AS1 inhibits growth and metastasis of triple-negative breast cancer via transcriptionally activating NCALD
Breast Cancer Research
Triple-negative breast cancer
MIDEAS-AS1
MATR3
NCALD
Prognosis
title Long non-coding RNA MIDEAS-AS1 inhibits growth and metastasis of triple-negative breast cancer via transcriptionally activating NCALD
title_full Long non-coding RNA MIDEAS-AS1 inhibits growth and metastasis of triple-negative breast cancer via transcriptionally activating NCALD
title_fullStr Long non-coding RNA MIDEAS-AS1 inhibits growth and metastasis of triple-negative breast cancer via transcriptionally activating NCALD
title_full_unstemmed Long non-coding RNA MIDEAS-AS1 inhibits growth and metastasis of triple-negative breast cancer via transcriptionally activating NCALD
title_short Long non-coding RNA MIDEAS-AS1 inhibits growth and metastasis of triple-negative breast cancer via transcriptionally activating NCALD
title_sort long non coding rna mideas as1 inhibits growth and metastasis of triple negative breast cancer via transcriptionally activating ncald
topic Triple-negative breast cancer
MIDEAS-AS1
MATR3
NCALD
Prognosis
url https://doi.org/10.1186/s13058-023-01709-1
work_keys_str_mv AT danluo longnoncodingrnamideasas1inhibitsgrowthandmetastasisoftriplenegativebreastcancerviatranscriptionallyactivatingncald
AT yiranliang longnoncodingrnamideasas1inhibitsgrowthandmetastasisoftriplenegativebreastcancerviatranscriptionallyactivatingncald
AT yajiewang longnoncodingrnamideasas1inhibitsgrowthandmetastasisoftriplenegativebreastcancerviatranscriptionallyactivatingncald
AT fangzhouye longnoncodingrnamideasas1inhibitsgrowthandmetastasisoftriplenegativebreastcancerviatranscriptionallyactivatingncald
AT yuhanjin longnoncodingrnamideasas1inhibitsgrowthandmetastasisoftriplenegativebreastcancerviatranscriptionallyactivatingncald
AT yamingli longnoncodingrnamideasas1inhibitsgrowthandmetastasisoftriplenegativebreastcancerviatranscriptionallyactivatingncald
AT dianwenhan longnoncodingrnamideasas1inhibitsgrowthandmetastasisoftriplenegativebreastcancerviatranscriptionallyactivatingncald
AT zekunwang longnoncodingrnamideasas1inhibitsgrowthandmetastasisoftriplenegativebreastcancerviatranscriptionallyactivatingncald
AT bingchen longnoncodingrnamideasas1inhibitsgrowthandmetastasisoftriplenegativebreastcancerviatranscriptionallyactivatingncald
AT wenjingzhao longnoncodingrnamideasas1inhibitsgrowthandmetastasisoftriplenegativebreastcancerviatranscriptionallyactivatingncald
AT lijuanwang longnoncodingrnamideasas1inhibitsgrowthandmetastasisoftriplenegativebreastcancerviatranscriptionallyactivatingncald
AT xichen longnoncodingrnamideasas1inhibitsgrowthandmetastasisoftriplenegativebreastcancerviatranscriptionallyactivatingncald
AT liyujiang longnoncodingrnamideasas1inhibitsgrowthandmetastasisoftriplenegativebreastcancerviatranscriptionallyactivatingncald
AT qifengyang longnoncodingrnamideasas1inhibitsgrowthandmetastasisoftriplenegativebreastcancerviatranscriptionallyactivatingncald