scholarly journals Bisphenol A Exposure Changes the Transcriptomic and Proteomic Dynamics of Human Retinoblastoma Y79 Cells

Genes ◽  
2021 ◽  
Vol 12 (2) ◽  
pp. 264
Author(s):  
Chul-Hong Kim ◽  
Mi Jin Kim ◽  
Jinhong Park ◽  
Jinho Kim ◽  
Ji-Young Kim ◽  
...  

Bisphenol A (BPA) is a xenoestrogen chemical commonly used to manufacture polycarbonate plastics and epoxy resin and might affect various human organs. However, the cellular effects of BPA on the eyes have not been widely investigated. This study aimed to investigate the cellular cytotoxicity by BPA exposure on human retinoblastoma cells. BPA did not show cytotoxic effects, such as apoptosis, alterations to cell viability and cell cycle regulation. Comparative analysis of the transcriptome and proteome profiles were investigated after long-term exposure of Y79 cells to low doses of BPA. Transcriptome analysis using RNA-seq revealed that mRNA expression of the post-transcriptional regulation-associated gene sets was significantly upregulated in the BPA-treated group. Cell cycle regulation-associated gene sets were significantly downregulated by exposure to BPA. Interestingly, RNA-seq analysis at the transcript level indicated that alternative splicing events, particularly retained introns, were noticeably altered by low-dose BPA treatment. Additionally, proteome profiling using MALDI-TOF-MS identified a total of nine differentially expressed proteins. These results suggest that alternative splicing events and altered gene/protein expression patterns are critical phenomena affected by long-term low-dose BPA exposure. This represents a novel marker for the detection of various diseases associated with environmental pollutants such as BPA.

2020 ◽  
Author(s):  
Mai Baker ◽  
Mayra Petasny ◽  
Mercedes Bentata ◽  
Gillian Kay ◽  
Eden Engal ◽  
...  

ABSTRACTChanges in the cellular environment result in chromatin structure alteration, which in turn regulates gene expression. To learn about the effect of the cellular environment on the transcriptome, we studied the H3K9 de-methylase KDM3A. Using RNA-seq, we found that KDM3A regulates the transcription and alternative splicing of genes associated with cell cycle and DNA damage. We showed that KDM3A undergoes phosphorylation by PKA at serine 265 following DNA damage, and that the phosphorylation is important for a proper cell cycle regulation. We demonstrated that SAT1 alternative splicing, regulated by KDM3A, plays a role in cell cycle regulation. Furthermore we found that KDM3A’s demethylase activity is not needed for SAT1 alternative splicing regulation. In addition, we identified KDM3A’s protein partner ARID1A, the SWI/SNF subunit, and SRSF3 as regulators of SAT1 alternative splicing and showed that KDM3A is essential for SRSF3 binding to SAT1 pre-mRNA. These results suggest that KDM3A serves as a sensor of the environment and an adaptor for splicing factor binding. Our work reveals chromatin sensing of the environment in the regulation of alternative splicing.


2009 ◽  
Vol 331 (2) ◽  
pp. 468 ◽  
Author(s):  
Xin Wang ◽  
Debbache Julien ◽  
Arnheiter Heinz

2020 ◽  
Vol 22 (Supplement_3) ◽  
pp. iii302-iii303
Author(s):  
Hannah Chatwin ◽  
Rakeb Lemma ◽  
John DeSisto ◽  
Aaron Knox ◽  
Shelby Mestnik ◽  
...  

Abstract Diffuse intrinsic pontine glioma (DIPG) is a fatal pediatric brain tumor with no curative treatments. Approximately 80% of DIPGs contain an H3K27M mutation. The implications of the mutation and how they may be targeted are not fully understood. We established an H3K27M effect-isolating model by transducing H3K27-wildtype lines (HSJD-GBM-001, normal human astrocytes) with lentiviral-packaged H3K27M. We characterized H3K27M-related changes through western blot, phenotypic assays, and RNA-seq. Drug screening of H3K27-wildtype and matched H3K27M-transduced lines was used to identify targets more effective with H3K27M present. Patient-derived pediatric glioblastoma and DIPG lines (BT-245, SU-DIPG-IV, HSJD-DIPG-007, SU-DIPG-XIII*, SF7761) were used for validation. We observed increased H3K27ac and decreased H3K27me3, as well as increased proliferative and migratory abilities, with the addition of H3K27M to H3K27-wildtype lines. RNA-seq showed downregulation of cell cycle regulation and upregulation of epithelial-mesenchymal transition. GSK1070916, an Aurora kinase B/C inhibitor, was isolated from a synthetic lethality screen with H3K27M. GSK1070916 showed strong efficacy in native H3K27M lines (IC50s=60nM-1250nM), superior to the Aurora kinase A inhibitor alisertib, to which all cell lines showed substantial resistance. Combination of both drugs was not synergistic. GSK1070916 treatment caused increased H3K27me3 and decreased H3S10ph and H3S28ph. GSK1070916 induced apoptosis and S-phase stall. The H3K27M mutation induces epigenetic, phenotypic, and cell cycle regulation changes resulting in relaxation of transcriptional controls and more aggressive growth. Aurora kinase B/C inhibition is a novel therapeutic modality for DIPG that appears capable of reversing some H3K27M-related epigenetic changes, inducing apoptosis, and repressing uncontrolled cellular division.


2020 ◽  
Vol 12 (1) ◽  
Author(s):  
Hideyuki Takeshima ◽  
Yukie Yoda ◽  
Mika Wakabayashi ◽  
Naoko Hattori ◽  
Satoshi Yamashita ◽  
...  

Abstract Background Epigenetic reprogramming using DNA demethylating drugs is a promising approach for cancer therapy, but its efficacy is highly dependent on the dosing regimen. Low-dose treatment for a prolonged period shows a remarkable therapeutic efficacy, despite its small demethylating effect. Here, we aimed to explore the mechanisms of how such low-dose treatment shows this remarkable efficacy by focusing on epigenetic reprograming at the single-cell level. Methods Expression profiles in HCT116 cells treated with decitabine (DAC) were analyzed by single-cell RNA-sequencing (scRNA-seq). Functional consequences and DNA demethylation at the single-cell level were analyzed using cloned HCT116 cells after DAC treatment. Results scRNA-seq revealed that DAC-treated cells had highly diverse expression profiles at the single-cell level, and tumor-suppressor genes, endogenous retroviruses, and interferon-stimulated genes were upregulated in random fractions of cells. DNA methylation analysis of cloned HCT116 cells revealed that, while only partial reduction of DNA methylation levels was observed in bulk cells, complete demethylation of specific cancer-related genes, such as cell cycle regulation, WNT pathway, p53 pathway, and TGF-β pathway, was observed, depending upon clones. Functionally, a clone with complete demethylation of CDKN2A (p16) had a larger fraction of cells with tetraploid than parental cells, indicating induction of cellular senescence due to normalization of cell cycle regulation. Conclusions Epigenetic reprogramming of specific cancer-related pathways at the single-cell level is likely to underlie the remarkable efficacy of low-dose DNA demethylating therapy.


Blood ◽  
2010 ◽  
Vol 116 (21) ◽  
pp. 234-234 ◽  
Author(s):  
Liang Li ◽  
Min Li ◽  
Can-Lan Sun ◽  
Liton Francisco ◽  
Melanie Sabado ◽  
...  

Abstract Abstract 234 Therapy-related myelodysplasia or acute myeloid leukemia (t-MDS/AML) is a lethal complication of cancer treatment. Study of t-MDS/AML offers a unique opportunity to understand leukemogenesis since known genotoxic exposures can be temporally and causally related to genetic changes associated with development of leukemia. Although development of t-MDS/AML is associated with known genotoxic exposures, its pathogenesis is not well understood, and methods to predict risk of development of t-MDS/AML in individual cancer survivors are not available. To better understand the pathogenetic mechanisms underlying development of t-MDS/AML we performed microarray analysis of gene expression in patients who developed t-MDS/AML after autologous hematopoietic cell transplantation (aHCT) for Hodgkin lymphoma (HL) or non-Hodgkin lymphoma (NHL) and controls that did not develop t-MDS/AML after aHCT. Peripheral blood stem cell (PBSC) samples obtained pre-aHCT from patients who subsequently developed t-MDS post-aHCT (cases) and controls matched for primary diagnosis, age, race/ethnicity, and time since aHCT were studied. In a training set of 18 t-MDS/AML cases and 37 controls, CD34+ cells were selected from PBSC samples using flow cytometry, and gene expression evaluated using Affymetrix HG U133 plus 2.0 Arrays. Differences in gene expression in CD34+ cells from cases and controls were analyzed using conditional logistic model. Significant differences in gene expression were seen in PBSC obtained pre-aHCT from patients who later developed t-MDS/AML compared to controls.(Blood, 2009; 114: 677) PBSC obtained pre-aHCT from patients who subsequently t-MDS/AML after aHCT showed significant downregulation of gene sets related to mitochondria and oxidative phosphorylation, ribosomes, aminoacyl-tRNA biosynthesis, amino acid metabolism, cell cycle regulation, and hematopoietic differentiation. G-protein coupled receptors, hematopoietic regulation, and cell adhesion related genes were upregulated in PBSC from cases. There was reduced expression of genes with binding motifs for the transcription factor NRF2, which regulates oxidative stress and drug detoxification. We then sought to identify a smaller PBSC gene signature that would identify NHL and HL patients at the pre-aHCT timepoint at high risk for developing t- MDS/AML after aHCT (Figure 1). A cross-validated 38-gene classifier was derived from the training set using prediction analysis of microarray (PAM). This gene classifier was applied to an independent test set of PBSC obtained pre-HCT from 16 patients who developed t-MDS/AML after aHCT for NHL or HL, and 20 matched controls that did not develop t-MDS/AML. Application of the 38-gene signature to the test set correctly classified 19 of the 20 subjects (95%) who did not subsequently develop t-MDS/AML, and 14 of the 16 subjects (87.5%) who did develop t-MDS/AML, with significant correlation between predicted and true disease status (P<0.001). These results indicate that the gene expression profile of hematopoietic cells pre-aHCT can identify patients at high risk for t-MDS/AML post-aHCT. GSEA analysis revealed extensive overlap of up and down-regulated gene sets in t-MDS/AML cases in the training and test sets (Figure 2). Gene expression changes related to mitochondria, metabolism, cell cycle regulation and hematopoietic progenitors that were observed in the training set were validated in the test set. These results indicate that genetic programs associated with t-MDS/AML are perturbed long before disease onset, and that PBSC gene signatures can accurately identify patients at high risk of developing this complication. Disclosures: No relevant conflicts of interest to declare.


Sign in / Sign up

Export Citation Format

Share Document