scholarly journals Comprehensive landscape of epigenetic-dysregulated lncRNAs reveals a profound role of enhancers in carcinogenesis in breast cancer subtypes

Author(s):  
Hongying Zhao ◽  
Xiaoqin Liu ◽  
Lei Yu ◽  
Shihua Lin ◽  
Caiyu Zhang ◽  
...  
2017 ◽  
Vol 28 ◽  
pp. vi33
Author(s):  
L. Bastianelli ◽  
M. Pistelli ◽  
G.M. Giuseppetti ◽  
M. De Lisa ◽  
M. Macchini ◽  
...  

2018 ◽  
Vol 38 (2) ◽  
pp. 54 ◽  
Author(s):  
Jyh-Cherng Yu ◽  
Guo-Shiou Liao ◽  
Huan-Ming Hsu ◽  
Chi-Hong Chu ◽  
Zhi-Jie Hong ◽  
...  

Cancers ◽  
2020 ◽  
Vol 12 (5) ◽  
pp. 1239 ◽  
Author(s):  
Rajeev Vikram ◽  
Wen Cheng Chou ◽  
Shih-Chieh Hung ◽  
Chen-Yang Shen

Cells with high CD44 but low CD24 expression (CD44high/CD24−/low) and high aldehyde dehydrogenase activity (ALDHbr) are widely considered to be drivers of metastasis, therapy resistance and tumor recurrence in breast cancer. However, the role of the CD44high/CD24−/low and ALDHbr phenotypes in identifying tumorigenic cells in breast cancer remains controversial due to the discrepancy in their distribution and tumorigenic potential in intrinsic breast cancer subtypes. In this study, we analyzed the cells expressing these markers in six different breast cancer cell lines representing major breast cancer subtypes (T47D, MCF-7, BT-474, AU-565, Hs578T and MDA-MB-231). CD44high/CD24−/low, ALDHbr and CD44−/low/CD24−/low cell populations were isolated by flow cytometry and analyzed for hallmark stem cell characteristics of differentiation, migration, invasiveness and metastasis using in vitro and in vivo techniques. Our results demonstrate that the CD44−/low/CD24−/low cell population, which is enriched in luminal cell lines (T47D, MCF-7 and BT-474), possesses metastatic and tumorigenic properties. We also show that, contrary to previous claims, the expression of the ALDH1 isoform ALDH1A1 does not affect the tumorigenic potential of cell lines with high ALDH activity (BT-474 and AU-565). Further transcriptomic and clinical studies are needed to determine the potential of these markers as early diagnostic tools and treatment targets.


2018 ◽  
Vol 123 (10) ◽  
pp. 753-764 ◽  
Author(s):  
Marco Macchini ◽  
Martina Ponziani ◽  
Andrea Prochowski Iamurri ◽  
Mirco Pistelli ◽  
Mariagrazia De Lisa ◽  
...  

Author(s):  
Romain Teinturier ◽  
Razan Abou Ziki ◽  
Loay Kassem ◽  
Yakun Luo ◽  
Lucie Malbeteau ◽  
...  

Abstract Purpose Menin, encoded by the MEN1 gene, was recently reported to be involved in breast cancers, though the underlying mechanisms remain elusive. In the current study, we sought to further determine its role in mammary cells. Methods Menin expression in mammary lesions from mammary-specific Men1 mutant mice was detected using immunofluorescence staining. RT-qPCR and western blot were performed to determine the role of menin in ERα expression in human breast cancer cell lines. ChIP-qPCR and reporter gene assays were carried out to dissect the action of menin on the proximal ESR1 promoter. Menin expression in female patients with breast cancer was analyzed and its correlation with breast cancer subtypes was investigated. Results Immunofluorescence staining revealed that early mammary neoplasia in Men1 mutant mice displayed weak ERα expression. Furthermore, MEN1 silencing led to both reduced ESR1 mRNA and ERα protein expression in MCF7 and T47D cells. To further dissect the regulation of ESR1 transcription by menin, we examined whether and in which way menin could regulate the proximal ESR1 promoter, which has not been fully explored. Using ChIP analysis and reporter gene assays covering − 2500 bp to + 2000 bp of the TSS position, we showed that the activity of the proximal ESR1 promoter was markedly reduced upon menin downregulation independently of H3K4me3 status. Importantly, by analyzing the expression of menin in 354 human breast cancers, we found that a lower expression was associated with ER-negative breast cancer (P = 0.041). Moreover, among the 294 ER-positive breast cancer samples, reduced menin expression was not only associated with larger tumors (P = 0.01) and higher SBR grades (P = 0.005) but also with the luminal B-like breast cancer subtype (P = 0.006). Consistent with our clinical data, we demonstrated that GATA3 and FOXA1, co-factors in ESR1 regulation, interact physically with menin in MCF7 cells, and MEN1 knockdown led to altered protein expression of GATA3, the latter being a known marker of the luminal A subtype, in MCF7 cells. Conclusion Taken together, our data provide clues to the important role of menin in ERα regulation and the formation of breast cancer subtypes.


2021 ◽  
Vol 12 (1) ◽  
Author(s):  
Hao Huang ◽  
Jianyang Hu ◽  
Alishba Maryam ◽  
Qinghua Huang ◽  
Yuchen Zhang ◽  
...  

AbstractBreast cancer is a heterogeneous disease, affecting over 3.5 million women worldwide, yet the functional role of cis-regulatory elements including super-enhancers in different breast cancer subtypes remains poorly characterized. Triple-negative breast cancer (TNBC) is an aggressive subtype of breast cancer with a poor prognosis. Here we apply integrated epigenomic and transcriptomic profiling to uncover super-enhancer heterogeneity between breast cancer subtypes, and provide clinically relevant biological insights towards TNBC. Using CRISPR/Cas9-mediated gene editing, we identify genes that are specifically regulated by TNBC-specific super-enhancers, including FOXC1 and MET, thereby unveiling a mechanism for specific overexpression of the key oncogenes in TNBC. We also identify ANLN as a TNBC-specific gene regulated by super-enhancer. Our studies reveal a TNBC-specific epigenomic landscape, contributing to the dysregulated oncogene expression in breast tumorigenesis.


Sign in / Sign up

Export Citation Format

Share Document