Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters










Database
Language
Publication year range
1.
PLoS One ; 18(10): e0291549, 2023.
Article in English | MEDLINE | ID: mdl-37801436

ABSTRACT

RNA polymerase III transcription is pivotal in regulating cellular growth and frequently deregulated in various cancers. MAF1 negatively regulates RNA polymerase III transcription. Currently, it is unclear if MAF1 is universally deregulated in human cancers. Recently, MAF1 expression has been demonstrated to be altered in colorectal and liver carcinomas and Luminal B breast cancers. In this study, we analyzed clinical breast cancer datasets to determine if MAF1 alterations correlate with clinical outcomes in HER2-positive breast cancer. Using various bioinformatics tools, we screened breast cancer datasets for alterations in MAF1 expression. We report that MAF1 is amplified in 39% of all breast cancer sub-types, and the observed amplification co-occurs with MYC. MAF1 amplification correlated with increased methylation of the MAF1 promoter and MAF1 protein expression is significantly decreased in luminal, HER2-positive, and TNBC breast cancer subtypes. MAF1 protein expression is also significantly reduced in stage 2 and 3 breast cancer compared to normal and significantly decreased in all breast cancer patients, regardless of race and age. In SKBR3 and BT474 breast cancer cell lines treated with anti-HER2 therapies, MAF1 mRNA expression is significantly increased. In HER2-positive breast cancer patients, MAF1 expression significantly increases and correlates with five years of relapse-free survival in response to trastuzumab treatment, suggesting MAF1 is a predictive biomarker in breast cancer. These data suggest a role for MAF1 alterations in HER2-positive breast cancer. More extensive studies are warranted to determine if MAF1 serves as a predictive and prognostic biomarker in breast cancer.


Subject(s)
Breast Neoplasms , Liver Neoplasms , Humans , Female , Breast Neoplasms/pathology , Receptor, ErbB-2/genetics , Receptor, ErbB-2/metabolism , RNA Polymerase III , Neoplasm Recurrence, Local , Trastuzumab/therapeutic use
2.
Cancer Med ; 12(5): 6401-6418, 2023 03.
Article in English | MEDLINE | ID: mdl-36305848

ABSTRACT

BACKGROUND: TFIIIB, an RNA polymerase III specific transcription factor has been found to be deregulated in human cancers with much of the research focused on the TBP, BRF1, and BRF2 subunits. To date, the TFIIIB specific subunit BDP1 has not been investigated in ovarian cancer but has previously been shown to be deregulated in neuroblastoma, breast cancer, and Non-Hodgkins lymphoma. RESULTS: Using in silico analysis of clinically derived platforms, we report a decreased BDP1 expression as a result of deletion in serous ovarian cancer and a correlation with higher and advanced ovarian stages. Further analysis in the context of TP53 mutations, a major contributor to ovarian tumorigenesis, suggests that high BDP1 expression is unfavorable for overall survival and high BDP1 expression occurs in stages 2, 3 and 4 serous ovarian cancer. Additionally, high BDP1 expression is disadvantageous and unfavorable for progression-free survival. Lastly, BDP1 expression significantly decreased in patients treated with first-line chemotherapy, platin and taxane, at twelve-month relapse-free survival. CONCLUSIONS: Taken together with a ROC analysis, the data suggest BDP1 could be of clinical relevance as a predictive biomarker in serous ovarian cancer. Lastly, this study further demonstrates that both the over- and under expression of BDP1 warrants further investigation and suggests BDP1 may exhibit dual function in the context of tumorigenesis.


Subject(s)
Ovarian Neoplasms , Saccharomyces cerevisiae Proteins , TATA-Binding Protein Associated Factors , Humans , Female , Transcription Factor TFIIIB/genetics , Transcription Factor TFIIIB/metabolism , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae Proteins/metabolism , Neoplasm Recurrence, Local , Biomarkers , Ovarian Neoplasms/genetics , Carcinogenesis , TATA-Binding Protein Associated Factors/genetics
3.
Int J Mol Sci ; 23(18)2022 Sep 19.
Article in English | MEDLINE | ID: mdl-36142875

ABSTRACT

High-risk human papillomaviruses (HPV) are important agents, responsible for a large percentage of the 745,000 cases of head and neck squamous cell carcinomas (HNSCC), which were identified worldwide in 2020. In addition to being virally induced, tobacco and heavy alcohol consumption are believed to cause DNA damage contributing to the high number of HNSCC cases. Gene expression and DNA methylation differ between HNSCC based on HPV status. We used publicly available gene expression and DNA methylation profiles from the Cancer Genome Atlas and compared HPV positive and HPV negative HNSCC groups. We used differential gene expression analysis, differential methylation analysis, and a combination of these two analyses to identify the differences. Differential expression analysis identified 1854 differentially expressed genes, including PCNA, TNFRSF14, TRAF1, TRAF2, BCL2, and BIRC3. SYCP2 was identified as one of the top deregulated genes in the differential methylation analysis and in the combined differential expression and methylation analyses. Additionally, pathway and ontology analyses identified the extracellular matrix and receptor interaction pathway as the most altered between HPV negative and HPV positive HNSCC groups. Combining gene expression and DNA methylation can help in elucidating the genes involved in HPV positive HNSCC tumorigenesis, such as SYCP2 and TAF7L.


Subject(s)
Alphapapillomavirus , Carcinoma, Squamous Cell , Head and Neck Neoplasms , Papillomavirus Infections , Alphapapillomavirus/genetics , Carcinoma, Squamous Cell/pathology , DNA Methylation , Gene Expression , Head and Neck Neoplasms/complications , Head and Neck Neoplasms/genetics , Humans , Papillomaviridae/genetics , Papillomaviridae/metabolism , Papillomavirus Infections/complications , Papillomavirus Infections/genetics , Papillomavirus Infections/pathology , Proliferating Cell Nuclear Antigen/metabolism , Proto-Oncogene Proteins c-bcl-2/metabolism , Squamous Cell Carcinoma of Head and Neck/complications , Squamous Cell Carcinoma of Head and Neck/genetics , TNF Receptor-Associated Factor 1/metabolism , TNF Receptor-Associated Factor 2/metabolism
4.
Cancers (Basel) ; 14(7)2022 Mar 25.
Article in English | MEDLINE | ID: mdl-35406430

ABSTRACT

TFIIIB is deregulated in a variety of cancers. However, few studies investigate the TFIIIB subunit BDP1 in cancer. BDP1 has not been studied in breast cancer patients. Herein, we analyzed clinical breast cancer datasets to determine if BDP1 alterations correlate with clinical outcomes. BDP1 copy number (n = 1602; p = 8.03 × 10-9) and mRNA expression (n = 130; p = 0.002) are specifically decreased in patients with invasive ductal carcinoma (IDC). In IDC, BDP1 copy number negatively correlates with high grade (n = 1992; p = 2.62 × 10-19) and advanced stage (n = 1992; p = 0.005). BDP1 mRNA expression also negatively correlated with high grade (n = 55; p = 6.81 × 10-4) and advanced stage (n = 593; p = 4.66 × 10-4) IDC. Decreased BDP1 expression correlated with poor clinical outcomes (n = 295 samples): a metastatic event at three years (p = 7.79 × 10-7) and cancer reoccurrence at three years (p = 4.81 × 10-7) in IDC. Decreased BDP1 mRNA correlates with patient death at three (p = 9.90 × 10-6) and five (p = 1.02 × 10-6) years. Both BDP1 copy number (n = 3785; p = 1.0 × 10-14) and mRNA expression (n = 2434; p = 5.23 × 10-6) are altered in triple-negative invasive breast cancer (TNBC). Together, these data suggest a role for BDP1 as potential biomarker in breast cancer and additional studies are warranted.

5.
BMC Cancer ; 20(1): 1093, 2020 Nov 11.
Article in English | MEDLINE | ID: mdl-33176745

ABSTRACT

BACKGROUND: Deregulation of the RNA polymerase III specific TFIIIB subunit BRF2 occurs in subtypes of human cancers. However, correlations between BRF2 alterations and clinical outcomes in breast cancer are limited. We conducted this review to analyze BRF2 alterations in genomic data sets housed in Oncomine and cBioPortal to identify potential correlations between BRF2 alterations and clinical outcomes. METHODS: The authors queried both Oncomine and cBioPortal for alterations in BRF2 in human cancers and performed meta-analyses identifying significant correlations between BRF2 and clinical outcomes in invasive breast cancer (IBC). RESULTS: A meta cancer outlier profile analysis (COPA) of 715 data sets (86,733 samples) in Oncomine identified BRF2 as overexpressed in 60% of breast cancer data sets. COPA scores in IBC data sets (3594 patients) are comparable for HER2 (24.211, median gene rank 60) and BRF2 (29.656, median gene rank 36.5). Overall survival in IBC patients with BRF2 alterations (21%) is significantly decreased (p = 9.332e-3). IBC patients with BRF2 alterations aged 46 to 50 have a significantly poor survival outcome (p = 7.093e-3). Strikingly, in metastatic breast cancer, BRF2 is altered in 33% of women aged 45-50. BRF2 deletions are predominant in this age group. CONCLUSION: This study suggests BRF2 may be an prognostic biomarker in invasive breast carcinoma.


Subject(s)
Biomarkers, Tumor/genetics , Breast Neoplasms/pathology , Gene Deletion , Transcription Factor TFIIIB/genetics , Breast Neoplasms/genetics , Female , Humans , Neoplasm Invasiveness , Prognosis , Survival Rate
6.
BMC Cancer ; 15: 905, 2015 Nov 16.
Article in English | MEDLINE | ID: mdl-26573593

ABSTRACT

BACKGROUND: BRF2 is a transcription factor required for synthesis of a small group of non-coding RNAs by RNA polymerase III. Overexpression of BRF2 can transform human mammary epithelial cells. In both breast and lung cancers, the BRF2 gene is amplified and overexpressed and may serve as an oncogenic driver. Furthermore, elevated BRF2 can be independently prognostic of unfavorable survival. Dietary soy isoflavones increase metastasis to lungs in a model of breast cancer and a recent study reported significantly increased cell proliferation in breast cancer patients who used soy supplementation. The soy isoflavone daidzein is a major food-derived phytoestrogen that is structurally similar to estrogen. The putative estrogenic effect of soy raises concern that high consumption of soy foods by breast cancer patients may increase tumor growth. METHODS: Expression of BRF2 RNA and protein was assayed in ER-positive or -negative human breast cancer cells after exposure to daidzein. We also measured mRNA stability, promoter methylation and response to the demethylating agent 5-azacytidine. In addition, expression was compared between mice fed diets enriched or deprived of isoflavones. RESULTS: We demonstrate that the soy isoflavone daidzein specifically stimulates expression of BRF2 in ER-positive breast cancer cells, as well as the related factor BRF1. Induction is accompanied by increased levels of non-coding RNAs that are regulated by BRF2 and BRF1. Daidzein treatment stabilizes BRF2 and BRF1 mRNAs and selectively decreases methylation of the BRF2 promoter. Functional significance of demethylation is supported by induction of BRF2 by the methyltransferase inhibitor 5-azacytidine. None of these effects are observed in an ER-negative breast cancer line, when tested in parallel with ER-positive breast cancer cells. In vivo relevance is suggested by the significantly elevated levels of BRF2 mRNA detected in female mice fed a high-isoflavone commercial diet. In striking contrast, BRF2 and BRF1 mRNA levels are suppressed in matched male mice fed the same isoflavone-enriched diet. CONCLUSIONS: The BRF2 gene that is implicated in cancer can be induced in human breast cancer cells by the isoflavone daidzein, through promoter demethylation and/or mRNA stabilization. Dietary isoflavones may also induce BRF2 in female mice, whereas the converse occurs in males.


Subject(s)
Breast Neoplasms/metabolism , Isoflavones/pharmacology , Neoplasm Proteins/metabolism , Phytoestrogens/pharmacology , Transcription Factor TFIIIB/metabolism , Animals , Breast Neoplasms/genetics , Cell Line, Tumor , DNA Methylation/drug effects , Disease Models, Animal , Female , Humans , Male , Promoter Regions, Genetic/drug effects , Proto-Oncogene Mas , RNA, Messenger/metabolism , RNA, Neoplasm/metabolism , TATA-Binding Protein Associated Factors/metabolism , Transcription Factor TFIIIB/genetics
SELECTION OF CITATIONS
SEARCH DETAIL