scholarly journals Genome-Wide Small RNA Sequencing and Gene Expression Analysis Reveals a microRNA Profile of Cancer Susceptibility in ATM-Deficient Human Mammary Epithelial Cells

PLoS ONE ◽  
2013 ◽  
Vol 8 (5) ◽  
pp. e64779 ◽  
Author(s):  
Jill E. Hesse ◽  
Liwen Liu ◽  
Cynthia L. Innes ◽  
Yuxia Cui ◽  
Stela S. Palii ◽  
...  
2021 ◽  
Vol 11 (1) ◽  
Author(s):  
Donguk Kim ◽  
Na Yeon Park ◽  
Keunsoo Kang ◽  
Stuart K. Calderwood ◽  
Dong-Hyung Cho ◽  
...  

AbstractArsenic is reportedly a biphasic inorganic compound for its toxicity and anticancer effects in humans. Recent studies have shown that certain arsenic compounds including arsenic hexoxide (AS4O6; hereafter, AS6) induce programmed cell death and cell cycle arrest in human cancer cells and murine cancer models. However, the mechanisms by which AS6 suppresses cancer cells are incompletely understood. In this study, we report the mechanisms of AS6 through transcriptome analyses. In particular, the cytotoxicity and global gene expression regulation by AS6 were compared in human normal and cancer breast epithelial cells. Using RNA-sequencing and bioinformatics analyses, differentially expressed genes in significantly affected biological pathways in these cell types were validated by real-time quantitative polymerase chain reaction and immunoblotting assays. Our data show markedly differential effects of AS6 on cytotoxicity and gene expression in human mammary epithelial normal cells (HUMEC) and Michigan Cancer Foundation 7 (MCF7), a human mammary epithelial cancer cell line. AS6 selectively arrests cell growth and induces cell death in MCF7 cells without affecting the growth of HUMEC in a dose-dependent manner. AS6 alters the transcription of a large number of genes in MCF7 cells, but much fewer genes in HUMEC. Importantly, we found that the cell proliferation, cell cycle, and DNA repair pathways are significantly suppressed whereas cellular stress response and apoptotic pathways increase in AS6-treated MCF7 cells. Together, we provide the first evidence of differential effects of AS6 on normal and cancerous breast epithelial cells, suggesting that AS6 at moderate concentrations induces cell cycle arrest and apoptosis through modulating genome-wide gene expression, leading to compromised DNA repair and increased genome instability selectively in human breast cancer cells.


2016 ◽  
Vol 83 (2) ◽  
pp. 202-208 ◽  
Author(s):  
Qiuming Chen ◽  
Yanjun Wu ◽  
Mingyuan Zhang ◽  
Wenwen Xu ◽  
Xiaoping Guo ◽  
...  

Owing to the difficulty in obtaining mammary gland tissue from lactating animals, it is difficult to test the expression levels of genes in mammary gland. The aim of the current study was to identify if milk fat globule (MFG) in buffalo milk was an alternative to mammary gland (MG) and milk somatic cell (MSC) for gene expression analysis. Six buffalos in late lactation were selected to collect MFG and MSC, and then MG was obtained by surgery. MFG was stained with acridine orange to successfully visualise RNA and several cytoplasmic crescents in MFG. The total RNA in MFG was successfully isolated and the integrity was assessed by agarose gel electrophoresis. We analysed the cellular components in MFG, MG and MSC through testing the expression of cell-specific genes by qRT-PCR. The results showed that adipocyte-specific gene (AdipoQ) and leucocyte-specific genes (CD43, CSF1 and IL1α) in MFG were not detected, whereas epithelial cell marker genes (Keratin 8 and Keratin 18) in MFG were higher than in MSC and lower than in MG, fibroblast marker gene (vimentin) in MFG was significantly lower than in MG and MSC, milk protein genes (LALBA, BLG and CSN2) and milk fat synthesis-related genes (ACC, BTN1A1, FABP3 and FAS) in MFG were higher than in MG and MSC. In conclusion, the total RNA in MFG mainly derives from mammary epithelial cells and can be used to study the functional gene expression of mammary epithelial cells.


2006 ◽  
Vol 20 (5) ◽  
Author(s):  
Monique Michelle McCallister ◽  
Manish Kumar Tripathi ◽  
Smita Misra ◽  
Gautam Chaudhuri

2021 ◽  
Author(s):  
Donguk Kim ◽  
Na Yeon Park ◽  
Keunsoo Kang ◽  
Stuart K. Calderwood ◽  
Dong-Hyung Cho ◽  
...  

ABSTRACTArsenic is reportedly a biphasic inorganic compound for its toxicity and anticancer effects in humans [1, 2]. Recent studies have shown that certain arsenic compounds including arsenic hexoxide (AS4O6; hereafter, AS6) induce programmed cell death and cell cycle arrest in human cancer cells and murine cancer models [3, 4]. However, the mechanisms by which AS6 suppresses cancer cells are incompletely understood. In this study, we report the mechanisms of AS6 through transcriptome analyses. In particular, the cytotoxicity and global gene expression regulation by AS6 were compared in human normal and cancer breast epithelial cells. Using RNA-sequencing and bioinformatics analyses, differentially expressed genes in significantly affected biological pathways in these cell types were validated by real-time quantitative polymerase chain reaction and immunoblotting assays. Our data show markedly differential effects of AS6 on cytotoxicity and gene expression in human mammary epithelial normal cells (HUMEC) and Michigan Cancer Foundation 7 (MCF7), a human mammary epithelial cancer cell line. AS6 selectively arrests cell growth and induces cell death in MCF7 cells without affecting the growth of HUMEC in a dose-dependent manner. AS6 alters the transcription of a large number of genes in MCF7 cells, but much fewer genes in HUMEC. Importantly, we found that the cell proliferation, cell cycle, and DNA repair pathways are significantly suppressed whereas cellular stress response and apoptotic pathways increase in AS6-treated MCF7 cells. Together, we provide the first evidence of differential effects of AS6 on normal and cancerous breast epithelial cells, suggesting that AS6 at moderate concentrations induces cell cycle arrest and apoptosis through modulating genome-wide gene expression, leading to compromised DNA repair and increased genome instability selectively in human breast cancer cells.


Author(s):  
Stephen J. DeMartini ◽  
Nicole B. DeHart ◽  
Jennifer R. Schroeder

Aim: To identify the oxidative stress impacts of chloro-s-triazine herbicides on human mammary epithelial cell lines. Study Design: MCF-7 mammary epithelial carcinoma and MCF-10A mammary epithelial cells were treated with levels of three triazine herbicides in concentrations flanking the US FDA safe levels. Place and Duration of Study: Department of Biology, Millikin University, in January 2015 through December 2015 and January 2019 through May 2020. Methodology: We examined the oxidative effects of two triazine herbicides, atrazine and simazine, on estrogen-dependent MCF-7 mammary epithelial carcinoma cells using three different bioluminescent assay techniques. We then utilized real time PCR to analyze gene expression through RT-PCR analysis, in both MCF-7 cells and a non-cancerous cell line, MCF-10A, for both of these triazine herbicides plus the related cyanazine. Results: At all concentrations of atrazine and simazine, no statistical differences were found in the levels of oxidized glutathione or total oxidized and reduced nicotinamide adenine dinucleotides phosphates. In stark contrast, levels of hydrogen peroxide were found to be statistically different from the control at all concentrations of atrazine and simazine tested. Using an Analysis of Variance (ANOVA) we determined that within the enzymatic portion of the hydrogen peroxide pathway there were statistically significant differences in the expression of Peroxiredoxin 1 (PRDX1), Sulfiredoxin (SRXN1), and Thioredoxin (TXN). Conclusion: Exposure to triazines alters the hydrogen peroxide pathway, which in turn can greatly affect the stability of the cell milieu.


Sign in / Sign up

Export Citation Format

Share Document