RETRACTED ARTICLE: Targeting a novel LncRNA SNHG15/miR-451/c-Myc signaling cascade is effective to hamper the pathogenesis of breast cancer (BC) in vitro and in vivo

Abstract Background To our knowledge, LncRNA SNHG15 exerted its tumor-promoting effects to facilitate the development of breast cancer (BC), but there still needed more data to elucidate the potential underlying mechanisms. Methods We examined genes expression status by performing Real-Time qPCR and...

Full description

Bibliographic Details
Main Authors: Jiang Du, Hong Zhong, Binlin Ma
Format: Article
Language:English
Published: BMC 2021-03-01
Series:Cancer Cell International
Subjects:
Online Access:https://doi.org/10.1186/s12935-021-01885-0
_version_ 1797675038442258432
author Jiang Du
Hong Zhong
Binlin Ma
author_facet Jiang Du
Hong Zhong
Binlin Ma
author_sort Jiang Du
collection DOAJ
description Abstract Background To our knowledge, LncRNA SNHG15 exerted its tumor-promoting effects to facilitate the development of breast cancer (BC), but there still needed more data to elucidate the potential underlying mechanisms. Methods We examined genes expression status by performing Real-Time qPCR and Western Blot analysis, and cellular functions, including cell proliferation, viability, apoptosis, mobility, were measured by using the CCK-8 assay, colony formation assay, trypan blue staining assay, flow cytometer (FCM), transwell assay and wound scratch assay, respectively. The predicted targeting sites in LncRNA SNHG15, miR-451 and c-Myc 3′UTR were validated by dual-luciferase reporter gene system assay. Finally, we established the tumor-bearing mice models, and the expression status, including its enrichment and cellular localization were examined by immunohistochemistry (IHC) assay. Results Our data indicated LncRNA SNHG15 upregulated c-Myc to facilitate BC progression by sponging miR-451 in a competing endogenous RNA (ceRNA)-dependent manner in vitro and in vivo. Specifically, LncRNA SNHG15 and c-Myc were upregulated, while miR-451 was downregulated in BC cells and clinical tissues, compared to their normal counterparts. In addition, miR-451 negatively correlated with LncRNA SNHG15 and c-Myc, and LncRNA SNHG15 was positively relevant to c-Myc in BC tissues. Next, we validated that LncRNA SNHG15 sponged miR-451 to upregulate c-Myc in BC cells. Further gain- and loss-of-function experiments evidenced that LncRNA SNHG15 promoted, while miR-451 inhibited malignant phenotypes, including cell proliferation, viability, migration, invasion and epithelial-mesenchymal transition (EMT) in BC cells. Interestingly, the inhibiting effects of LncRNA SNHG15 ablation on BC progression were abrogated by both silencing miR-451 and overexpressing c-Myc. Conclusions We concluded that targeting the LncRNA SNHG15/miR-451/c-Myc signaling cascade was novel to hamper BC progression, which broadened our knowledge in this field, and provided potential biomarkers for BC diagnosis and treatment.
first_indexed 2024-03-11T22:07:54Z
format Article
id doaj.art-6380867885aa4bbdb8cf8bdd9d7a8e3c
institution Directory Open Access Journal
issn 1475-2867
language English
last_indexed 2024-03-11T22:07:54Z
publishDate 2021-03-01
publisher BMC
record_format Article
series Cancer Cell International
spelling doaj.art-6380867885aa4bbdb8cf8bdd9d7a8e3c2023-09-24T11:31:57ZengBMCCancer Cell International1475-28672021-03-0121111110.1186/s12935-021-01885-0RETRACTED ARTICLE: Targeting a novel LncRNA SNHG15/miR-451/c-Myc signaling cascade is effective to hamper the pathogenesis of breast cancer (BC) in vitro and in vivoJiang Du0Hong Zhong1Binlin Ma2Department of Breast and Thyroid Surgery, The 3rd Affiliated Teaching Hospital of Xinjiang Medical University (Affiliated Cancer Hospital)Department of Breast and Thyroid Surgery, The 3rd Affiliated Teaching Hospital of Xinjiang Medical University (Affiliated Cancer Hospital)Department of Breast and Thyroid Surgery, The 3rd Affiliated Teaching Hospital of Xinjiang Medical University (Affiliated Cancer Hospital)Abstract Background To our knowledge, LncRNA SNHG15 exerted its tumor-promoting effects to facilitate the development of breast cancer (BC), but there still needed more data to elucidate the potential underlying mechanisms. Methods We examined genes expression status by performing Real-Time qPCR and Western Blot analysis, and cellular functions, including cell proliferation, viability, apoptosis, mobility, were measured by using the CCK-8 assay, colony formation assay, trypan blue staining assay, flow cytometer (FCM), transwell assay and wound scratch assay, respectively. The predicted targeting sites in LncRNA SNHG15, miR-451 and c-Myc 3′UTR were validated by dual-luciferase reporter gene system assay. Finally, we established the tumor-bearing mice models, and the expression status, including its enrichment and cellular localization were examined by immunohistochemistry (IHC) assay. Results Our data indicated LncRNA SNHG15 upregulated c-Myc to facilitate BC progression by sponging miR-451 in a competing endogenous RNA (ceRNA)-dependent manner in vitro and in vivo. Specifically, LncRNA SNHG15 and c-Myc were upregulated, while miR-451 was downregulated in BC cells and clinical tissues, compared to their normal counterparts. In addition, miR-451 negatively correlated with LncRNA SNHG15 and c-Myc, and LncRNA SNHG15 was positively relevant to c-Myc in BC tissues. Next, we validated that LncRNA SNHG15 sponged miR-451 to upregulate c-Myc in BC cells. Further gain- and loss-of-function experiments evidenced that LncRNA SNHG15 promoted, while miR-451 inhibited malignant phenotypes, including cell proliferation, viability, migration, invasion and epithelial-mesenchymal transition (EMT) in BC cells. Interestingly, the inhibiting effects of LncRNA SNHG15 ablation on BC progression were abrogated by both silencing miR-451 and overexpressing c-Myc. Conclusions We concluded that targeting the LncRNA SNHG15/miR-451/c-Myc signaling cascade was novel to hamper BC progression, which broadened our knowledge in this field, and provided potential biomarkers for BC diagnosis and treatment.https://doi.org/10.1186/s12935-021-01885-0LncRNA SNHG15Breast cancerC-MycMiR-451Malignant phenotypes
spellingShingle Jiang Du
Hong Zhong
Binlin Ma
RETRACTED ARTICLE: Targeting a novel LncRNA SNHG15/miR-451/c-Myc signaling cascade is effective to hamper the pathogenesis of breast cancer (BC) in vitro and in vivo
Cancer Cell International
LncRNA SNHG15
Breast cancer
C-Myc
MiR-451
Malignant phenotypes
title RETRACTED ARTICLE: Targeting a novel LncRNA SNHG15/miR-451/c-Myc signaling cascade is effective to hamper the pathogenesis of breast cancer (BC) in vitro and in vivo
title_full RETRACTED ARTICLE: Targeting a novel LncRNA SNHG15/miR-451/c-Myc signaling cascade is effective to hamper the pathogenesis of breast cancer (BC) in vitro and in vivo
title_fullStr RETRACTED ARTICLE: Targeting a novel LncRNA SNHG15/miR-451/c-Myc signaling cascade is effective to hamper the pathogenesis of breast cancer (BC) in vitro and in vivo
title_full_unstemmed RETRACTED ARTICLE: Targeting a novel LncRNA SNHG15/miR-451/c-Myc signaling cascade is effective to hamper the pathogenesis of breast cancer (BC) in vitro and in vivo
title_short RETRACTED ARTICLE: Targeting a novel LncRNA SNHG15/miR-451/c-Myc signaling cascade is effective to hamper the pathogenesis of breast cancer (BC) in vitro and in vivo
title_sort retracted article targeting a novel lncrna snhg15 mir 451 c myc signaling cascade is effective to hamper the pathogenesis of breast cancer bc in vitro and in vivo
topic LncRNA SNHG15
Breast cancer
C-Myc
MiR-451
Malignant phenotypes
url https://doi.org/10.1186/s12935-021-01885-0
work_keys_str_mv AT jiangdu retractedarticletargetinganovellncrnasnhg15mir451cmycsignalingcascadeiseffectivetohamperthepathogenesisofbreastcancerbcinvitroandinvivo
AT hongzhong retractedarticletargetinganovellncrnasnhg15mir451cmycsignalingcascadeiseffectivetohamperthepathogenesisofbreastcancerbcinvitroandinvivo
AT binlinma retractedarticletargetinganovellncrnasnhg15mir451cmycsignalingcascadeiseffectivetohamperthepathogenesisofbreastcancerbcinvitroandinvivo