1
|
Fan T, Han T, Gu A, Jin J, Cui Q, Guo J, Zhang X, Yu H, Shi W. Novel Approach to Screen Endocrine-Disrupting Chemicals via Endocrine-Enhanced Reduced Human Transcriptome. ENVIRONMENTAL SCIENCE & TECHNOLOGY 2025; 59:4845-4856. [PMID: 40042996 DOI: 10.1021/acs.est.4c13159] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/19/2025]
Abstract
Endocrine-disrupting chemicals (EDCs) can interfere with multiple pathways and trigger different modes of action. Thus, the traditional EDC in vitro screening processes often require a battery of bioassays to cover multiple target pathways. Here we developed an endocrine-enhanced reduced human transcriptome (ERHT) focused on hormone receptor signaling induced by the EDCs regulating specific genes. ERHT was developed based on 1200 prioritized genes covering 110 endocrine-related biological pathways across eight potential adverse outcomes. The ability of this approach to identify EDCs was derived from machine learning of 1068 dose-dependent transcriptome profiles and enhanced by quantifying chemical-induced critical pathway responses, and thus, it demonstrated excellent classification performance (AUC = 0.84 ± 0.03) in internal cross-validation. We ultimately applied this approach to known EDCs and inactive substances to validate the reliability of this approach. Through external validation on 210 chemicals, the extrapolation accuracy exceeded 80%, demonstrating the outstanding practical performance of this approach. Meanwhile, the pathway responses induced by the same chemical were consistent with the experimental results reported by multiple sequencing platforms, highlighting the robustness of this approach. The above results demonstrate that this approach can provide novel insights for EDCs' high-throughput screening and comprehensive toxic mechanisms through biological pathways.
Collapse
Affiliation(s)
- Tianle Fan
- State Key Laboratory of Pollution Control and Resource Reuse, School of the Environment, Nanjing University, Nanjing 210023, China
| | - Tianhao Han
- State Key Laboratory of Pollution Control and Resource Reuse, School of the Environment, Nanjing University, Nanjing 210023, China
| | - Aoran Gu
- State Key Laboratory of Pollution Control and Resource Reuse, School of the Environment, Nanjing University, Nanjing 210023, China
| | - Jinsha Jin
- State Key Laboratory of Pollution Control and Resource Reuse, School of the Environment, Nanjing University, Nanjing 210023, China
| | - Qian Cui
- Nanjing Yangtze River Delta Green Development Institute, Nanjing 210093, China
| | - Jing Guo
- State Key Laboratory of Pollution Control and Resource Reuse, School of the Environment, Nanjing University, Nanjing 210023, China
| | - Xiaowei Zhang
- State Key Laboratory of Pollution Control and Resource Reuse, School of the Environment, Nanjing University, Nanjing 210023, China
| | - Hongxia Yu
- State Key Laboratory of Pollution Control and Resource Reuse, School of the Environment, Nanjing University, Nanjing 210023, China
| | - Wei Shi
- State Key Laboratory of Pollution Control and Resource Reuse, School of the Environment, Nanjing University, Nanjing 210023, China
- Jiangsu Province Ecology and Environment Protection Key Laboratory of Chemical Safety and Health Risk, Nanjing 210023, China
| |
Collapse
|
2
|
Corton JC, Ledbetter V, Cohen SM, Atlas E, Yauk CL, Liu J. A transcriptomic biomarker predictive of cell proliferation for use in adverse outcome pathway-informed testing and assessment. Toxicol Sci 2024; 201:174-189. [PMID: 39137154 DOI: 10.1093/toxsci/kfae102] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/15/2024] Open
Abstract
High-throughput transcriptomics (HTTr) is increasingly being used to identify molecular targets of chemicals that can be linked to adverse outcomes. Cell proliferation (CP) is an important key event in chemical carcinogenesis. Here, we describe the construction and characterization of a gene expression biomarker that is predictive of the CP status in human and rodent tissues. The biomarker was constructed from 30 genes known to be increased in expression in prostate cancers relative to surrounding tissues and in cycling human MCF-7 cells after estrogen receptor (ER) agonist exposure. Using a large compendium of gene expression profiles to test utility, the biomarker could identify increases in CP in (i) 308 out of 367 tumor vs. normal surrounding tissue comparisons from 6 human organs, (ii) MCF-7 cells after activation of ER, (iii) after partial hepatectomy in mice and rats, and (iv) the livers of mice and rats after exposure to nongenotoxic hepatocarcinogens. The biomarker identified suppression of CP (i) under conditions of p53 activation by DNA damaging agents in human cells, (ii) in human A549 lung cells exposed to therapeutic anticancer kinase inhibitors (dasatinib, nilotnib), and (iii) in the mouse liver when comparing high levels of CP at birth to the low background levels in the adult. The responses using the biomarker were similar to those observed using conventional markers of CP including PCNA, Ki67, and BrdU labeling. The CP biomarker will be a useful tool for interpretation of HTTr data streams to identify CP status after exposure to chemicals in human cells or in rodent tissues.
Collapse
Affiliation(s)
- J Christopher Corton
- Center for Computational Toxicology and Exposure, US Environmental Protection Agency, Research Triangle Park, NC 27711, United States
| | - Victoria Ledbetter
- Center for Computational Toxicology and Exposure, US Environmental Protection Agency, Research Triangle Park, NC 27711, United States
| | - Samuel M Cohen
- Department of Pathology and Microbiology and Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE 69198-3135, United States
| | - Ella Atlas
- Environmental Health Science and Research Bureau, Healthy Environments and Consumer Safety Branch (HECSB) Health Canada, Ottawa, ON K2K 0K9, Canada
| | - Carole L Yauk
- Department of Biology, University of Ottawa, Ottawa, ON, Canada
| | - Jie Liu
- Center for Computational Toxicology and Exposure, US Environmental Protection Agency, Research Triangle Park, NC 27711, United States
| |
Collapse
|
3
|
Corton JC, Matteo G, Chorley B, Liu J, Vallanat B, Everett L, Atlas E, Meier MJ, Williams A, Yauk CL. A 50-gene biomarker identifies estrogen receptor-modulating chemicals in a microarray compendium. Chem Biol Interact 2024; 394:110952. [PMID: 38570061 DOI: 10.1016/j.cbi.2024.110952] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Revised: 03/01/2024] [Accepted: 03/09/2024] [Indexed: 04/05/2024]
Abstract
High throughput transcriptomics (HTTr) profiling has the potential to rapidly and comprehensively identify molecular targets of environmental chemicals that can be linked to adverse outcomes. We describe here the construction and characterization of a 50-gene expression biomarker designed to identify estrogen receptor (ER) active chemicals in HTTr datasets. Using microarray comparisons, the genes in the biomarker were identified as those that exhibited consistent directional changes when ER was activated (4 ER agonists; 4 ESR1 gene constitutively active mutants) and opposite directional changes when ER was suppressed (4 antagonist treatments; 4 ESR1 knockdown experiments). The biomarker was evaluated as a predictive tool using the Running Fisher algorithm by comparison to annotated gene expression microarray datasets including those evaluating the transcriptional effects of hormones and chemicals in MCF-7 cells. Depending on the reference dataset used, the biomarker had a predictive accuracy for activation of up to 96%. To demonstrate applicability for HTTr data analysis, the biomarker was used to identify ER activators in a set of 15 chemicals that are considered potential bisphenol A (BPA) alternatives examined at up to 10 concentrations in MCF-7 cells and analyzed by full-genome TempO-Seq. Using benchmark dose (BMD) modeling, the biomarker genes stratified the ER potency of BPA alternatives consistent with previous studies. These results demonstrate that the ER biomarker can be used to accurately identify ER activators in transcript profile data derived from MCF-7 cells.
Collapse
Affiliation(s)
- J Christopher Corton
- Center for Computational Toxicology and Exposure, US Environmental Protection Agency, Research Triangle Park, NC, 27711, USA.
| | - Geronimo Matteo
- Environmental Health Science and Research Bureau, Health Canada, Ottawa, ON, K1A 0K9, Canada; Department of Biology, University of Ottawa, Ottawa, ON, K1N 6N5, Canada.
| | - Brian Chorley
- Center for Computational Toxicology and Exposure, US Environmental Protection Agency, Research Triangle Park, NC, 27711, USA.
| | - Jie Liu
- Center for Computational Toxicology and Exposure, US Environmental Protection Agency, Research Triangle Park, NC, 27711, USA.
| | - Beena Vallanat
- Center for Computational Toxicology and Exposure, US Environmental Protection Agency, Research Triangle Park, NC, 27711, USA.
| | - Logan Everett
- Center for Computational Toxicology and Exposure, US Environmental Protection Agency, Research Triangle Park, NC, 27711, USA.
| | - Ella Atlas
- Environmental Health Science and Research Bureau, Health Canada, Ottawa, ON, K1A 0K9, Canada.
| | - Matthew J Meier
- Environmental Health Science and Research Bureau, Health Canada, Ottawa, ON, K1A 0K9, Canada.
| | - Andrew Williams
- Environmental Health Science and Research Bureau, Health Canada, Ottawa, ON, K1A 0K9, Canada.
| | - Carole Lyn Yauk
- Department of Biology, University of Ottawa, Ottawa, ON, K1N 6N5, Canada.
| |
Collapse
|
4
|
Lu L, Shen L, Cui S, Huang Y, Gao Y, Zhu X, Lu S, Zhang C, Zhuang S. Angiogenic Activity and Mechanism for Bisphenols on Endothelial Cell and Mouse: Evidence of a Structural-Selective Effect. ENVIRONMENTAL SCIENCE & TECHNOLOGY 2023; 57:11803-11813. [PMID: 37505069 DOI: 10.1021/acs.est.3c03883] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/29/2023]
Abstract
Increased epidemiological evidence indicates the association of bisphenol exposure with human vascular disorders, while the underlying mechanism has not been clarified. Here, we sought to unveil the potential angiogenic effect and the underlying mechanism of bisphenols with different structural features using endothelial cells treated with an environmentally relevant concentration of bisphenols (range: 1 nM to 10 μM) and a C57BL/6 mouse model fed with doses of 0.002, 0.02, 2, and 20 mg/kg BW/day for 5 weeks. Bisphenol A (BPA) and bisphenol S (BPS) at a 1 nM level significantly increased tube formation by 45.1 and 30.2% and induced the microvessel sprouting, while tube length and microvessel sprouting were significantly inhibited by 37.2 and 55.7% after exposure to tetrabromobisphenol S (TBBPS) at 1 μM, respectively. Mechanistically, TBBPA and TBBPS significantly inhibited the interaction between phosphatidylinositol 3-kinase (PI3K) and thyroid receptor (TR), while BPA and BPS favored the interaction between PI3K and estrogen receptor (ER), resulting in abnormal PI3K signaling with consequent distinct angiogenic activity. BPA- and BPS-induced pro-angiogenic effects and TBBPS showed anti-angiogenic effects due to their distinct disruption on the TR/ER-PI3K pathway. Our work provided new evidence and mechanistic insight on the angiogenic activity of bisphenols and expanded the scope of endocrine disruptors with interference in vascular homeostasis.
Collapse
Affiliation(s)
- Liping Lu
- Key Laboratory of Environment Remediation and Ecological Health, Ministry of Education, College of Environmental and Resource Sciences, Zhejiang University, Hangzhou, Zhejiang 310058, China
- School of Public Health, Hangzhou Normal University, Hangzhou, Zhejiang 311121, China
| | - Lilai Shen
- Key Laboratory of Environment Remediation and Ecological Health, Ministry of Education, College of Environmental and Resource Sciences, Zhejiang University, Hangzhou, Zhejiang 310058, China
| | - Shixuan Cui
- Key Laboratory of Environment Remediation and Ecological Health, Ministry of Education, College of Environmental and Resource Sciences, Zhejiang University, Hangzhou, Zhejiang 310058, China
| | - Yizhou Huang
- Department of Gynecology, Women's Hospital, Zhejiang University School of Medicine, Hangzhou 310006, Zhejiang, China
| | - Yuchen Gao
- Key Laboratory of Environment Remediation and Ecological Health, Ministry of Education, College of Environmental and Resource Sciences, Zhejiang University, Hangzhou, Zhejiang 310058, China
| | - Xiaoming Zhu
- Department of Gynecology, Women's Hospital, Zhejiang University School of Medicine, Hangzhou 310006, Zhejiang, China
| | - Shaoyong Lu
- Department of Pathophysiology, School of Medicine, Shanghai Jiao Tong University, Shanghai 200025, China
| | - Chunlong Zhang
- Department of Environmental Sciences, University of Houston-Clear Lake, 2700 Bay Area Blvd., Houston, Texas 77058, United States
| | - Shulin Zhuang
- Key Laboratory of Environment Remediation and Ecological Health, Ministry of Education, College of Environmental and Resource Sciences, Zhejiang University, Hangzhou, Zhejiang 310058, China
- Department of Gynecology, Women's Hospital, Zhejiang University School of Medicine, Hangzhou 310006, Zhejiang, China
| |
Collapse
|
5
|
Stanic B, Kokai D, Markovic Filipovic J, Samardzija Nenadov D, Pogrmic-Majkic K, Andric N. Global gene expression analysis reveals novel transcription factors associated with long-term low-level exposure of EA.hy926 human endothelial cells to bisphenol A. Chem Biol Interact 2023:110571. [PMID: 37244401 DOI: 10.1016/j.cbi.2023.110571] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2023] [Revised: 05/11/2023] [Accepted: 05/25/2023] [Indexed: 05/29/2023]
Abstract
Bisphenol A (BPA) is an endocrine disruptor that binds to estrogen receptors (ER); however, studies have shown that the ER pathway was not always the primary molecular mechanism of BPA's action in cells and that gene transcription could be altered by different exposure times and doses. Here, we sought to understand the correlation between the BPA-responsive genes that have associated biological functions and the transcription factors (TFs) involved in their regulation by repeatedly exposing human endothelial cells EA.hy926 to three nanomolar concentrations of BPA (10-9 M, 10-8 M, and 10-7 M) for 14 weeks, after which changes in global gene expression were determined by RNA sequencing. Cytoscape plug-in iRegulon was used to infer TFs involved in the control of BPA-deregulated genes. The results show a minimal overlap in deregulated genes between three concentrations of BPA, with 10-9 M BPA having the highest number of deregulated genes. TF analysis suggests that all three concentrations of BPA were active in the absence of an ER-mediated pathway. A unique set of TFs (NES≥4) has been identified for each BPA concentration, including the NFκB family and CEBPB for 10-9 M BPA, MEF family, AHR/ARNT, and ZBTB33 for 10-8 M BPA, and IRF1-7 and OVOL1/OVOL2 for 10-7 M BPA, whereas STAT1/STAT2 were common TFs for 10-9 M and 10-7 M BPA. Overall, our data suggest that long-term low-level exposure of EA.hy926 cells to BPA leads to concentration-specific changes in gene expression that are not controlled by the ER-mediated signaling but rather by other mechanisms.
Collapse
Affiliation(s)
- Bojana Stanic
- University of Novi Sad, Faculty of Sciences, Department of Biology and Ecology, Serbia.
| | - Dunja Kokai
- University of Novi Sad, Faculty of Sciences, Department of Biology and Ecology, Serbia
| | | | | | | | - Nebojsa Andric
- University of Novi Sad, Faculty of Sciences, Department of Biology and Ecology, Serbia
| |
Collapse
|
6
|
Xiong J, Tian Y, Ma G, Ling A, Shan S, Cheng G. Comparative RNA-seq analysis and ceRNA network of genistein-treated GT1-7 neurons. Mol Cell Toxicol 2022. [DOI: 10.1007/s13273-022-00279-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
7
|
Abstract
Regulatory agencies around the world depend on standardized testing approaches to evaluate environmental chemicals for endocrine disrupting properties. The US Environmental Protection Agency (EPA) has developed a two-tiered testing approach within its Endocrine Disruptor Screening Program (EDSP). The eleven Tier 1 and three Tier 2 EDSP assays can be used to identify chemicals that act as agonists or antagonists of estrogen receptor, androgen receptor, or thyroid hormone receptor, or chemicals that interfere with steroidogenesis. Additional assays have been developed in the context of Tox21, and others have been validated by the OECD. In spite of the availability of validated toxicity tests, problems have been identified with the approaches and methods used to identify endocrine disrupting chemicals (EDCs). This chapter will provide an overview of several of these issues including: (1) The way an EDC is defined by an agency impacts whether a specific test can be used to determine if a chemical is an EDC. This is especially important when considering which assays examine outcomes that are considered "adverse effects." (2) Some assumptions about the validated studies used to identify EDCs may not be true (e.g., their reproducibility has been questioned). (3) Many of the validated assays are less sensitive than other methods that have not yet been validated. Ultimately, these and other problems contribute to the current landscape, where testing approaches have failed to protect the public from known EDCs. The chapter concludes with a review of approaches that have been taken to improve current guideline studies.
Collapse
Affiliation(s)
- Laura N Vandenberg
- Department of Environmental Health Sciences, School of Public Health & Health Sciences, University of Massachusetts Amherst, Amherst, MA, United States.
| |
Collapse
|
8
|
Tran V, Kim R, Maertens M, Hartung T, Maertens A. Similarities and Differences in Gene Expression Networks Between the Breast Cancer Cell Line Michigan Cancer Foundation-7 and Invasive Human Breast Cancer Tissues. Front Artif Intell 2021; 4:674370. [PMID: 34056582 PMCID: PMC8155268 DOI: 10.3389/frai.2021.674370] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Accepted: 04/23/2021] [Indexed: 12/31/2022] Open
Abstract
Failure to adequately characterize cell lines, and understand the differences between in vitro and in vivo biology, can have serious consequences on the translatability of in vitro scientific studies to human clinical trials. This project focuses on the Michigan Cancer Foundation-7 (MCF-7) cells, a human breast adenocarcinoma cell line that is commonly used for in vitro cancer research, with over 42,000 publications in PubMed. In this study, we explore the key similarities and differences in gene expression networks of MCF-7 cell lines compared to human breast cancer tissues. We used two MCF-7 data sets, one data set collected by ARCHS4 including 1032 samples and one data set from Gene Expression Omnibus GSE50705 with 88 estradiol-treated MCF-7 samples. The human breast invasive ductal carcinoma (BRCA) data set came from The Cancer Genome Atlas, including 1212 breast tissue samples. Weighted Gene Correlation Network Analysis (WGCNA) and functional annotations of the data showed that MCF-7 cells and human breast tissues have only minimal similarity in biological processes, although some fundamental functions, such as cell cycle, are conserved. Scaled connectivity—a network topology metric—also showed drastic differences in the behavior of genes between MCF-7 and BRCA data sets. Finally, we used canSAR to compute ligand-based druggability scores of genes in the data sets, and our results suggested that using MCF-7 to study breast cancer may lead to missing important gene targets. Our comparison of the networks of MCF-7 and human breast cancer highlights the nuances of using MCF-7 to study human breast cancer and can contribute to better experimental design and result interpretation of study involving this cell line.
Collapse
Affiliation(s)
- Vy Tran
- Department of Environmental Health and Engineering, Center for Alternatives to Animal Testing, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, United States
| | - Robert Kim
- Department of Environmental Health and Engineering, Center for Alternatives to Animal Testing, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, United States
| | - Mikhail Maertens
- Department of Environmental Health and Engineering, Center for Alternatives to Animal Testing, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, United States
| | - Thomas Hartung
- Department of Environmental Health and Engineering, Center for Alternatives to Animal Testing, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, United States.,Department of Biology, Center for Alternatives to Animal Testing-Europe, University of Konstanz, Konstanz, Germany.,Department of Environmental Health and Engineering, Doerenkamp-Zbinden Professor and Chair for Evidence-Based Toxicology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, United States
| | - Alexandra Maertens
- Department of Environmental Health and Engineering, Center for Alternatives to Animal Testing, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, United States
| |
Collapse
|
9
|
Deng P, Tan M, Zhou W, Chen C, Xi Y, Gao P, Ma Q, Liang Y, Chen M, Tian L, Xie J, Liu M, Luo Y, Li Y, Zhang L, Wang L, Zeng Y, Pi H, Yu Z, Zhou Z. Bisphenol A promotes breast cancer cell proliferation by driving miR-381-3p-PTTG1-dependent cell cycle progression. CHEMOSPHERE 2021; 268:129221. [PMID: 33352510 DOI: 10.1016/j.chemosphere.2020.129221] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/09/2020] [Revised: 11/22/2020] [Accepted: 12/03/2020] [Indexed: 06/12/2023]
Abstract
Bisphenol A (BPA) is a high-production-volume industrial chemical that facilitates the development of breast cancer. However, the molecular mechanism associated with BPA-induced breast cancer cell proliferation and migration remains elusive. In our study, we exposed MCF-7 cells to different concentrations of BPA (0.1, 1 and 10 μM) for 24, 48, or 72 h. We found that BPA exposure significantly promoted MCF-7 cell proliferation and migration but not invasion. To elucidate the mechanisms, the differentially expressed genes between the BPA and control groups were investigated with the Gene Expression Omnibus (GEO) database through GEO2R. Kyoto Encyclopedia of Genes and Genomes (KEGG) and pathway action network analyses demonstrated the important role of the cell cycle pathway in the effects of BPA exposure on MCF-7 cells. Importantly, analysis with the cytoHubba plugin of Cytoscape software coupled with analysis of enriched genes in the cell cycle pathway identified PTTG1 and CDC20 (two hub genes) as key targets associated with BPA-induced MCF-7 cell proliferation and migration. Interestingly, BPA significantly increased the protein expression levels of PTTG1 but not CDC20. Knockdown of PTTG1 inhibited the BPA-induced increase in proliferation and maintained cell cycle progression. In addition, we confirmed that the increased expression of PTTG1 upon BPA exposure was caused by miR-381-3p inhibition. Moreover, we verified that miR-381-3p expression was low and inversely correlated with PTTG1 expression in breast cancer tissues. Together, these findings demonstrate that BPA promotes high PTTG1 expression and alters the cell cycle to enhance MCF-7 cell proliferation by inhibiting miR-381-3p expression.
Collapse
Affiliation(s)
- Ping Deng
- Department of Occupational Health (Key Laboratory of Electromagnetic Radiation Protection, Ministry of Education), Third Military Medical University, Chongqing, China
| | - Miduo Tan
- Surgery Department of Galactophore, The Affiliated Zhuzhou Hospital Xiangya Medical College CSU (Central Hospital of Zhuzhou City), Zhuzhou, China
| | - Wei Zhou
- Surgery Department of Galactophore, The Affiliated Zhuzhou Hospital Xiangya Medical College CSU (Central Hospital of Zhuzhou City), Zhuzhou, China
| | - Chunhai Chen
- Department of Occupational Health (Key Laboratory of Electromagnetic Radiation Protection, Ministry of Education), Third Military Medical University, Chongqing, China
| | - Yu Xi
- Department of Environmental Medicine, School of Public Health, and Department of Emergency Medicine, First Affiliated Hospital, School of Medicine, Zhejiang University, China
| | - Peng Gao
- Department of Occupational Health (Key Laboratory of Electromagnetic Radiation Protection, Ministry of Education), Third Military Medical University, Chongqing, China
| | - Qinlong Ma
- Department of Occupational Health (Key Laboratory of Electromagnetic Radiation Protection, Ministry of Education), Third Military Medical University, Chongqing, China
| | - Yidan Liang
- School of Medicine, Guangxi University, Nanning, Guangxi Zhuang Autonomous Region, China
| | - Mengyan Chen
- Department of Occupational Health (Key Laboratory of Electromagnetic Radiation Protection, Ministry of Education), Third Military Medical University, Chongqing, China
| | - Li Tian
- Department of Occupational Health (Key Laboratory of Electromagnetic Radiation Protection, Ministry of Education), Third Military Medical University, Chongqing, China
| | - Jia Xie
- Department of Occupational Health (Key Laboratory of Electromagnetic Radiation Protection, Ministry of Education), Third Military Medical University, Chongqing, China
| | - Mengyu Liu
- Department of Occupational Health (Key Laboratory of Electromagnetic Radiation Protection, Ministry of Education), Third Military Medical University, Chongqing, China
| | - Yan Luo
- Department of Occupational Health (Key Laboratory of Electromagnetic Radiation Protection, Ministry of Education), Third Military Medical University, Chongqing, China
| | - Yanqi Li
- Department of Occupational Health (Key Laboratory of Electromagnetic Radiation Protection, Ministry of Education), Third Military Medical University, Chongqing, China
| | - Lei Zhang
- Department of Occupational Health (Key Laboratory of Electromagnetic Radiation Protection, Ministry of Education), Third Military Medical University, Chongqing, China
| | - Liting Wang
- Biomedical Analysis Center, Third Military Medical University, Chongqing, China
| | - Youlong Zeng
- Biomedical Analysis Center, Third Military Medical University, Chongqing, China
| | - Huifeng Pi
- Department of Occupational Health (Key Laboratory of Electromagnetic Radiation Protection, Ministry of Education), Third Military Medical University, Chongqing, China.
| | - Zhengping Yu
- Department of Occupational Health (Key Laboratory of Electromagnetic Radiation Protection, Ministry of Education), Third Military Medical University, Chongqing, China.
| | - Zhou Zhou
- Department of Environmental Medicine, School of Public Health, and Department of Emergency Medicine, First Affiliated Hospital, School of Medicine, Zhejiang University, China.
| |
Collapse
|
10
|
Wang TN, Yang PJ, Tseng YT, Tsai YS, Kuo PL, Chiu CC, Liang SS, Hsieh TH, Hou MF, Tsai EM. Visceral obesity and cell cycle pathways serve as links in the association between bisphenol A exposure and breast cancer. Oncol Lett 2020; 20:33-42. [PMID: 32565931 PMCID: PMC7285711 DOI: 10.3892/ol.2020.11553] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2018] [Accepted: 08/16/2018] [Indexed: 11/05/2022] Open
Abstract
It has been identified that bisphenol A (BPA) exposure causes developmental toxicity in breast cells. However, the exact molecular mechanisms underlying the association between exposure to BPA and breast cancer remain unclear. The aim of the present study was to investigate the BPA-regulated signaling pathways associated with the aggressiveness and the development of breast cancer. Microarray technology and functional gene set analyses were used to evaluate BPA and breast cancer-associated biomarkers and pathways in a discovery-driven manner. Using individual dataset analyses, it was indicated that two BPA-associated gene sets, the visceral obesity pathway, involved in visceral fat deposits and the metabolic syndrome, and the cell cycle pathway, involved in cyclins and cell cycle regulation, were significantly associated with a high grade of aggressiveness and the development of estrogen receptor (ER)-positive breast cancer (between P<0.05 and 0.0001). The pooled analysis indicated that the most significant pathway was G1/S checkpoint regulation, and the cyclin and cell cycle regulation pathway for BPA-associated ER-positive cancer. Cancer cell signaling pathways were associated with healthy breast cells developing into breast cancer. The visceral obesity and the cell cycle pathways were indicated to link BPA exposure to breast cancer. The results of the present study demonstrate a significant association between breast cancer and BPA-regulated gene pathways.
Collapse
Affiliation(s)
- Tsu-Nai Wang
- Department of Public Health, College of Health Science, Kaohsiung Medical University, Kaohsiung 80708, Taiwan, R.O.C.,Research Center for Environmental Medicine, Kaohsiung Medical University, Kaohsiung 80708, Taiwan, R.O.C
| | - Pei-Jing Yang
- Department of Public Health, College of Health Science, Kaohsiung Medical University, Kaohsiung 80708, Taiwan, R.O.C
| | - Yu-Ting Tseng
- Department of Public Health, College of Health Science, Kaohsiung Medical University, Kaohsiung 80708, Taiwan, R.O.C
| | - Yi-Shan Tsai
- Department of Public Health, College of Health Science, Kaohsiung Medical University, Kaohsiung 80708, Taiwan, R.O.C
| | - Po-Lin Kuo
- Research Center for Environmental Medicine, Kaohsiung Medical University, Kaohsiung 80708, Taiwan, R.O.C.,Graduate Institute of Clinical Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 80708, Taiwan, R.O.C
| | - Chien-Chih Chiu
- Research Center for Environmental Medicine, Kaohsiung Medical University, Kaohsiung 80708, Taiwan, R.O.C.,Department of Biotechnology, College of Life Science, Kaohsiung Medical University, Kaohsiung 80708, Taiwan, R.O.C
| | - Shih-Shin Liang
- Department of Biotechnology, College of Life Science, Kaohsiung Medical University, Kaohsiung 80708, Taiwan, R.O.C.,Institute of Biomedical Science, College of Science, National Sun Yat-Sen University, Kaohsiung 80424, Taiwan, R.O.C
| | - Tsung-Hua Hsieh
- Department of Obstetrics and Gynecology, Kaohsiung Medical University Hospital, Kaohsiung 80756, Taiwan, R.O.C
| | - Ming-Feng Hou
- Graduate Institute of Clinical Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 80708, Taiwan, R.O.C.,Department of Surgery, Kaohsiung Medical University Hospital, Kaohsiung 80756, Taiwan, R.O.C.,Department of Cancer Center, Kaohsiung Medical University Hospital, Kaohsiung 80756, Taiwan, R.O.C.,Department of Surgery, Kaohsiung Municipal Siaogang Hospital, Kaohsiung 81267, Taiwan, R.O.C.,Department of Biological Science and Technology, National Chiao Tung University, Hsin-Chu 30010, Taiwan, R.O.C
| | - Eing-Mei Tsai
- Research Center for Environmental Medicine, Kaohsiung Medical University, Kaohsiung 80708, Taiwan, R.O.C.,Department of Obstetrics and Gynecology, Kaohsiung Medical University Hospital, Kaohsiung 80756, Taiwan, R.O.C.,Center for Research Resources and Development, Kaohsiung Medical University Hospital, Kaohsiung 80756, Taiwan, R.O.C.,Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 80708, Taiwan, R.O.C
| |
Collapse
|
11
|
PD-1 Imposes Qualitative Control of Cellular Transcriptomes in Response to T Cell Activation. Mol Cell 2020; 77:937-950.e6. [PMID: 31926851 DOI: 10.1016/j.molcel.2019.12.012] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2019] [Revised: 11/07/2019] [Accepted: 12/12/2019] [Indexed: 01/08/2023]
Abstract
Targeted blockade of programmed cell death 1 (PD-1), an immune-checkpoint receptor that inhibits T cell activation, provides clinical benefits in various cancers. However, how PD-1 modulates gene expression in T cells remains enigmatic. Here we investigated how PD-1 affects transcriptome changes induced by T cell receptor (TCR) activation. Intriguingly, we identified a huge variance in PD-1 sensitivity among TCR-inducible genes. When we quantified the half maximal effective concentration (EC50) as the relationship between change in gene expression and TCR signal strength, we found that genes associated with survival and proliferation were efficiently expressed upon TCR activation and resistant to PD-1-mediated inhibition. Conversely, genes encoding cytokines and effector molecules were expressed less efficiently and sensitive to PD-1-mediated inhibition. We further demonstrated that transcription factor binding motifs and CpG frequency in the promoter region affect EC50 and thus the PD-1 sensitivity of genes. Our findings explain how PD-1, dependent on the TCR signal strength, calibrates cellular transcriptomes to shape functional properties of T cell populations.
Collapse
|
12
|
Sharma S, Ahmad S, Afjal MA, Habib H, Parvez S, Raisuddin S. Dichotomy of bisphenol A-induced expression of peroxisome proliferator-activated receptors in hepatic and testicular tissues in mice. CHEMOSPHERE 2019; 236:124264. [PMID: 31319301 DOI: 10.1016/j.chemosphere.2019.06.234] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/02/2019] [Revised: 06/23/2019] [Accepted: 06/30/2019] [Indexed: 06/10/2023]
Abstract
Environmental and dietary exposure to bisphenol A (BPA) and its toxicological consequences are extensively reported. BPA has multiple cellular targets. One of the mechanisms of action of BPA involves interaction with and activation of nuclear receptors (NRs) including peroxisome proliferator activated-receptors (PPARs). PPARs regulate genes involved in adipogenesis, and metabolism of glucose, lipid and cholesterol. Study of tissue and dose specific PPAR expression may decipher the toxicity outcome of BPA exposure. We studied expression of three forms of PPARs in mouse liver and testes exposed to BPA for 14 days. mRNA and protein expression of all forms of PPAR increased linearly (monotonic) with the dose in the liver while non-monotonic but dose specific effects were observed in the testes showing a differential pattern of expression. However, histopathological study showed a dose-dependent pattern of changes in liver as well as testes demonstrating a monotonic effect. These findings imply that other PPAR-independent mechanisms may play a role in BPA-induced pathological changes. The present study warrants exploration of the role of PPARs in BPA-induced effects on male reproductive functions and offers an insight into the peculiar response of BPA at low subchronic levels which may be helpful in designing appropriate risk assessment framework.
Collapse
Affiliation(s)
- Shikha Sharma
- Molecular Toxicology Laboratory, Department of Medical Elementology & Toxicology, Jamia Hamdard (Hamdard University), New Delhi, 110062, India
| | - Shahzad Ahmad
- Molecular Toxicology Laboratory, Department of Medical Elementology & Toxicology, Jamia Hamdard (Hamdard University), New Delhi, 110062, India
| | - Mohd Amir Afjal
- Molecular Toxicology Laboratory, Department of Medical Elementology & Toxicology, Jamia Hamdard (Hamdard University), New Delhi, 110062, India
| | - Haroon Habib
- Molecular Toxicology Laboratory, Department of Medical Elementology & Toxicology, Jamia Hamdard (Hamdard University), New Delhi, 110062, India
| | - Suhel Parvez
- Molecular Toxicology Laboratory, Department of Medical Elementology & Toxicology, Jamia Hamdard (Hamdard University), New Delhi, 110062, India
| | - Sheikh Raisuddin
- Molecular Toxicology Laboratory, Department of Medical Elementology & Toxicology, Jamia Hamdard (Hamdard University), New Delhi, 110062, India.
| |
Collapse
|
13
|
Maertens A, Tran V, Kleensang A, Hartung T. Weighted Gene Correlation Network Analysis (WGCNA) Reveals Novel Transcription Factors Associated With Bisphenol A Dose-Response. Front Genet 2018; 9:508. [PMID: 30483308 PMCID: PMC6240694 DOI: 10.3389/fgene.2018.00508] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2018] [Accepted: 10/10/2018] [Indexed: 11/13/2022] Open
Abstract
Despite Bisphenol-A (BPA) being subject to extensive study, a thorough understanding of molecular mechanism remains elusive. Here we show that using weighted gene correlation network analysis (WGCNA), which takes advantage of a graph theoretical approach to understanding correlations amongst genes and grouping genes into modules that typically have co-ordinated biological functions and regulatory mechanisms, that despite some commonality in altered genes, there is minimal overlap between BPA and estrogen in terms of network topology. We confirmed previous findings that ZNF217 and TFAP2C are involved in the estrogen pathway, and are implicated in BPA as well, although for BPA they appear to be active in the absence of canonical estrogen-receptor driven gene expression. Furthermore, our study suggested that PADI4 and RACK7/ZMYNDB8 may be involved in the overlap in gene expression between estradiol and BPA. Lastly, we demonstrated that even at low doses there are unique transcription factors that appear to be driving the biology of BPA, such as SREBF1. Overall, our data is consistent with other reports that BPA leads to subtle gene changes rather than profound aberrations of a conserved estrogen signaling (or other) pathways.
Collapse
Affiliation(s)
- Alexandra Maertens
- Center for Alternatives to Animal Testing, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, United States
| | - Vy Tran
- Center for Alternatives to Animal Testing, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, United States
| | - Andre Kleensang
- Center for Alternatives to Animal Testing, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, United States
| | - Thomas Hartung
- Center for Alternatives to Animal Testing, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, United States.,Center for Alternatives to Animal Testing - Europe, University of Konstanz, Konstanz, Germany.,Doerenkamp-Zbinden Professor and Chair for Evidence-Based Toxicology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, United States
| |
Collapse
|
14
|
Perrot-Applanat M, Kolf-Clauw M, Michel C, Beausoleil C. Alteration of mammary gland development by bisphenol a and evidence of a mode of action mediated through endocrine disruption. Mol Cell Endocrinol 2018; 475:29-53. [PMID: 30048677 DOI: 10.1016/j.mce.2018.06.015] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/28/2017] [Revised: 06/26/2018] [Accepted: 06/26/2018] [Indexed: 01/08/2023]
Abstract
The development and function of the mammary gland are endocrine-dependent processes, depending on the stage of development. Foetal and/or postnatal exposure to low doses of BPA alters tissue organisation through epithelial proliferation and stroma-epithelial interactions. BPA also alters the expression of E2-dependent epithelial and stroma transcriptomes. Several signalling pathways are consistent with the observed phenotype: proliferation and apoptosis, a focal adhesion pathway indicating changes in biomechanical properties of the extracellular matrix, and immune function. Some of BPA's effects are reversed by oestrogen and/or GPER inhibitors. BPA also alters the expression of epigenetic marks (EZH2, HOTAIR), which would explain the delayed effect of foetal BPA exposure. In conclusion, experimental evidence shows that pre- or postnatal BPA exposure consistently causes endocrine modifications in the mammary tissue of different animal species, disrupting stromal-epithelial interactions and ultimately increasing its susceptibility to carcinogens. An interspecies comparison highlights why and how these effects apply to humans.
Collapse
Affiliation(s)
| | - Martine Kolf-Clauw
- CREFRE, Toulouse University, INSERM, Toulouse Veterinary School, 23 chemin des Capelles, BP 87614, F 310176, Toulouse Cedex 3, France
| | - Cécile Michel
- ANSES, Risk Assessment Department, Maisons-Alfort, France.
| | | |
Collapse
|
15
|
Abstract
Endocrine disruption has been gathering increasing attention in the past 25 years as a possible new threat for health and safety. Exposure to endocrine disruptor has been progressively linked with a growing number of increasing disease in the human population. The mechanics through which endocrine disruptors act are not yet completely clear, however a number of pathways have been identified. A key concern is the cumulative and synergic effects that endocrine disruptors could have when mixed in consumer products. We reviewed the available literature to identify known or potential endocrine disruptors, as well as endocrine active substances that could contribute to cumulative effects, in topical consumer products. The number of endocrine actives used daily in consumer products is staggering and even though most if not all are used in concentrations that are considered to be safe, we believe that the possibility of combined effects in mixtures and non-monotonic dose/response is enough to require further precautions. A combined in vitro approach based on existing, validated OECD test methods is suggested to screen consumer products and mixtures for potential interaction with estrogen and androgen hormone receptors, in order to identify products that could have cumulative effects or support their safety concerning direct endocrine disruption capabilities.
Collapse
|
16
|
Watford SM, Grashow RG, De La Rosa VY, Rudel RA, Friedman KP, Martin MT. Novel application of normalized pointwise mutual information (NPMI) to mine biomedical literature for gene sets associated with disease: use case in breast carcinogenesis. COMPUTATIONAL TOXICOLOGY (AMSTERDAM, NETHERLANDS) 2018; 7:46-57. [PMID: 32274464 PMCID: PMC7144681 DOI: 10.1016/j.comtox.2018.06.003] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Advances in technology within biomedical sciences have led to an inundation of data across many fields, raising new challenges in how best to integrate and analyze these resources. For example, rapid chemical screening programs like the US Environmental Protection Agency's ToxCast and the collaborative effort, Tox21, have produced massive amounts of information on putative chemical mechanisms where assay targets are identified as genes; however, systematically linking these hypothesized mechanisms with in vivo toxicity endpoints like disease outcomes remains problematic. Herein we present a novel use of normalized pointwise mutual information (NPMI) to mine biomedical literature for gene associations with biological concepts as represented by Medical Subject Headings (MeSH terms) in PubMed. Resources that tag genes to articles were integrated, then cross-species orthologs were identified using UniRef50 clusters. MeSH term frequency was normalized to reflect the MeSH tree structure, and then the resulting GeneID-MeSH associations were ranked using NPMI. The resulting network, called Entity MeSH Co-occurrence Network (EMCON), is a scalable resource for the identification and ranking of genes for a given topic of interest. The utility of EMCON was evaluated with the use case of breast carcinogenesis. Topics relevant to breast carcinogenesis were used to query EMCON and retrieve genes important to each topic. A breast cancer gene set was compiled through expert literature review (ELR) to assess performance of the search results. We found that the results from EMCON ranked the breast cancer genes from ELR higher than randomly selected genes with a recall of 0.98. Precision of the top five genes for selected topics was calculated as 0.87. This work demonstrates that EMCON can be used to link in vitro results to possible biological outcomes, thus aiding in generation of testable hypotheses for furthering understanding of biological function and the contribution of chemical exposures to disease.
Collapse
Affiliation(s)
- Sean M Watford
- ORAU, contractor to U.S. Environmental Protection Agency through the National Student Services Contract, Oak Ridge, TN
- Department of Environmental Sciences and Engineering, Gillings School of Global Public Health, UNC-Chapel Hill, Chapel Hill, North Carolina, United States
| | - Rachel G Grashow
- Silent Spring Institute, Newton, MA
- Department of Environmental Health, Harvard T.H. Chan School of Public Health, Boston, MA
| | - Vanessa Y De La Rosa
- Silent Spring Institute, Newton, MA
- Social Science Environmental Health Research Institute, Northeastern University, Boston, MA
| | | | | | - Matthew T Martin
- U.S. Environmental Protection Agency, National Center for Computational Toxicology, Research Triangle Park, NC, USA
- Currently at Pfizer Worldwide Research & Development, Groton, CT, USA
| |
Collapse
|
17
|
Mesnage R, Phedonos A, Arno M, Balu S, Corton JC, Antoniou MN. Editor's Highlight: Transcriptome Profiling Reveals Bisphenol A Alternatives Activate Estrogen Receptor Alpha in Human Breast Cancer Cells. Toxicol Sci 2018; 158:431-443. [PMID: 28591870 PMCID: PMC5837682 DOI: 10.1093/toxsci/kfx101] [Citation(s) in RCA: 83] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Plasticizers with estrogenic activity, such as bisphenol A (BPA), have potential adverse
health effects in humans. Due to mounting evidence of these health effects, BPA is being
phased out and replaced by other bisphenol variants in “BPA-free” products. We have
compared estrogenic activity of BPA with 6 bisphenol analogues [bisphenol S (BPS);
bisphenol F (BPF); bisphenol AP (BPAP); bisphenol AF (BPAF); bisphenol Z (BPZ); bisphenol
B (BPB)] in 3 human breast cancer cell lines. Estrogenicity was assessed
(10−11–10−4 M) by cell growth in an estrogen receptor
(ER)-mediated cell proliferation assay, and by the induction of estrogen response
element-mediated transcription in a luciferase assay. BPAF was the most potent bisphenol,
followed by BPB > BPZ ∼ BPA > BPF ∼ BPAP > BPS. The addition of ICI 182,780
antagonized the activation of ERs. Data mining of ToxCast high-throughput screening assays
confirm our results but also show divergence in the sensitivities of the assays. Gene
expression profiles were determined in MCF-7 cells by microarray analysis. The comparison
of transcriptome profile alterations resulting from BPA alternatives with an ERα gene
expression biomarker further indicates that all BPA alternatives act as ERα agonists in
MCF-7 cells. These results were confirmed by Illumina-based RNA sequencing. In conclusion,
BPA alternatives are not necessarily less estrogenic than BPA in human breast cancer
cells. BPAF, BPB, and BPZ were more estrogenic than BPA. These findings point to the
importance of better understanding the risk of adverse effects from exposure to BPA
alternatives, including hormone-dependent breast cancer.
Collapse
Affiliation(s)
- Robin Mesnage
- Gene Expression and Therapy Group, Faculty of Life Sciences & Medicine, Department of Medical and Molecular Genetics, King's College London, London SE1 9RT, United Kingdom
| | - Alexia Phedonos
- Gene Expression and Therapy Group, Faculty of Life Sciences & Medicine, Department of Medical and Molecular Genetics, King's College London, London SE1 9RT, United Kingdom
| | - Matthew Arno
- Genomics Centre, King's College London, London SE1 9NH, United Kingdom
| | - Sucharitha Balu
- Genomics Centre, King's College London, London SE1 9NH, United Kingdom
| | - J Christopher Corton
- Integrated Systems Toxicology Division National Health and Environmental Effects Research Lab, US Environmental Protection Agency, Research Triangle Park, North Carolina 27711
| | - Michael N Antoniou
- Gene Expression and Therapy Group, Faculty of Life Sciences & Medicine, Department of Medical and Molecular Genetics, King's College London, London SE1 9RT, United Kingdom
| |
Collapse
|
18
|
Grashow RG, De La Rosa VY, Watford SM, Ackerman JM, Rudel RA. BCScreen: A gene panel to test for breast carcinogenesis in chemical safety screening. COMPUTATIONAL TOXICOLOGY (AMSTERDAM, NETHERLANDS) 2018; 5:16-24. [PMID: 31218268 PMCID: PMC6583811 DOI: 10.1016/j.comtox.2017.11.003] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Targeted gene lists have been used in clinical settings to specify breast tumor type, and to predict breast cancer prognosis and response to treatment. Separately, panels have been curated to predict systemic toxicity and xenoestrogen activity as a part of chemical screening strategies. However, currently available panels do not specifically target biological processes relevant to breast development and carcinogenesis. We have developed a gene panel called the Breast Carcinogen Screen (BCScreen) as a tool to identify potential breast carcinogens and characterize mechanisms of toxicity. First, we used four seminal reviews to identify 14 key characteristics of breast carcinogenesis, such as apoptosis, immunomodulation, and genotoxicity. Then, using a hybrid data and knowledge-driven framework, we systematically combined information from whole transcriptome data from genomic databases, biomedical literature, the CTD chemical-gene interaction database, and primary literature review to generate a panel of 500 genes relevant to breast carcinogenesis. We used normalized pointwise mutual information (NPMI) to rank genes that frequently co-occurred with key characteristics in biomedical literature. We found that many genes identified for BCScreen were not included in prognostic breast cancer or systemic toxicity panels. For example, more than half of BCScreen genes were not included in the Tox21 S1500+ general toxicity gene list. Of the 230 that did overlap between the two panels, representation varied across characteristics of carcinogenesis ranging from 21% for genes associated with epigenetics to 82% for genes associated with xenobiotic metabolism. Enrichment analysis of BCScreen identified pathways and processes including response to steroid hormones, cancer, cell cycle, apoptosis, DNA damage and breast cancer. The biologically-based systematic approach to gene prioritization demonstrated here provides a flexible framework for creating disease-focused gene panels to support discovery related to etiology. With validation, BCScreen may also be useful for toxicological screening relevant to breast carcinogenesis.
Collapse
Affiliation(s)
- Rachel G. Grashow
- Silent Spring Institute, 320 Nevada Street, Newton, MA 02460, United States
| | - Vanessa Y. De La Rosa
- Silent Spring Institute, 320 Nevada Street, Newton, MA 02460, United States
- Social Science Environmental Health Research Institute, Northeastern University, Boston, MA, United States
| | - Sean M. Watford
- Department of Environmental Sciences and Engineering, Gillings School of Global Public Health, UNC-Chapel Hill, Chapel Hill, NC, United States
| | - Janet M. Ackerman
- Silent Spring Institute, 320 Nevada Street, Newton, MA 02460, United States
| | - Ruthann A. Rudel
- Silent Spring Institute, 320 Nevada Street, Newton, MA 02460, United States
| |
Collapse
|
19
|
Meta-analysis of microarray and RNA-Seq gene expression datasets for carcinogenic risk: An assessment of Bisphenol A. Mol Cell Toxicol 2017. [DOI: 10.1007/s13273-017-0026-5] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
|
20
|
Soto AM, Schaeberle C, Maier MS, Sonnenschein C, Maffini MV. Evidence of Absence: Estrogenicity Assessment of a New Food-Contact Coating and the Bisphenol Used in Its Synthesis. ENVIRONMENTAL SCIENCE & TECHNOLOGY 2017; 51:1718-1726. [PMID: 28098991 DOI: 10.1021/acs.est.6b04704] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/06/2023]
Abstract
Consumer concerns about exposure to substances found in food contact materials with estrogenic activity (EA) have created substantial demand for alternatives. We assessed the potential EA of both a new bisphenol monomer used to synthesize polymeric coatings for metal food-contact applications and the nonintentionally added substances (NIAS) that may migrate into food. We evaluated tetramethyl bisphenol F (TMBPF) using in vitro and in vivo assays. We extracted the polymeric coating using food simulants ethanol (50% v/v) and acetic acid (3% w/v) and measured migration using tandem liquid chromatography (LC)/mass spectrometry (MS) and LC time-of-flight MS for TMBPF and NIAS, respectively. We also tested migrants for EA using the E-SCREEN assay. TMBPF did not show estrogenic activity in the uterotrophic assay and did not alter puberty in male and female rats or mammary gland development in female rats. Neither TMBPF nor the migrants from the final polymeric coating increased proliferation of estrogen-sensitive MCF7 cells. TMBPF did not show estrogen-agonist or antagonist activity in the estrogen receptor-transactivation assay. TMBPF migration was below the 0.2 parts per billion detection limit. Our findings provide compelling evidence for the absence of EA by TMBPF and the polymeric coating derived from it and that human exposure to TMBPF would be negligible.
Collapse
Affiliation(s)
- Ana M Soto
- Department of Integrative Physiology and Pathobiology, Tufts University School of Medicine , Boston, Massachusetts 02111, United States
| | - Cheryl Schaeberle
- Department of Integrative Physiology and Pathobiology, Tufts University School of Medicine , Boston, Massachusetts 02111, United States
| | - Mark S Maier
- The Valspar Corporation , Packaging Division, Sewickley, Pennsylvania 15143, United States
| | - Carlos Sonnenschein
- Department of Integrative Physiology and Pathobiology, Tufts University School of Medicine , Boston, Massachusetts 02111, United States
| | | |
Collapse
|
21
|
Potratz S, Tarnow P, Jungnickel H, Baumann S, von Bergen M, Tralau T, Luch A. Combination of Metabolomics with Cellular Assays Reveals New Biomarkers and Mechanistic Insights on Xenoestrogenic Exposures in MCF-7 Cells. Chem Res Toxicol 2016; 30:883-892. [PMID: 27514991 DOI: 10.1021/acs.chemrestox.6b00106] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
The disruptive potential of xenoestrogens like bisphenol A (BPA) lies in their 17β-estradiol (E2)-like binding to estrogen receptors (ERs) followed by concomitant modulation of ER target gene expression. Unsurprisingly, most endocrine testing systems focus on the quantification of canonical transcripts or ER-sensitive reporters. However, only little information is available about the corresponding metabolomic changes in vitro. This knowledge gap becomes particularly relevant in the context of potential mixture effects, for example, as a consequence of coexposure to potentially estrogenically active pollutants (e.g., Cd2+). Such effects are often difficult to dissect with molecular tools, especially with regard to potential physiological relevance. Metabolomic biomarkers are well-suited to address this latter aspect as they provide a comprehensive readout of whole-cell physiology. Applying a targeted metabolomics approach (FIA-MS/MS), this study looked for biomarkers indicative of xenoestrogenic exposure in MCF-7 cells. Cells were treated with E2 and BPA in the presence or absence of Cd2+. Statistical analysis revealed a total of 11 amino acids and phospholipids to be related to the compound's estrogenic potency. Co-exposure to Cd2+ modulated the estrogenic profile. However, the corresponding changes were found to be moderate with cellular assays such as the E-screen failing to record any Cd2+-specific estrogenic effects. Overall, metabolomics analysis identified proline as the most prominent estrogenic biomarker. Its increase could clearly be related to estrogenic exposure and concomitant ERα-mediated induction of proliferation. Involvement of the latter was confirmed by siRNA-mediated knockdown studies as well as by receptor inhibition. Further, the underlying signaling was also found to involve the oncoprotein MYC. Taken together, this study provides insights into the underlying mechanisms of xenoestrogenic effects and exemplify the strength of the complementary use of metabolomics and cellular and molecular assays.
Collapse
Affiliation(s)
- Sarah Potratz
- Department of Chemical and Product Safety, German Federal Institute for Risk Assessment (BfR) , Max-Dohrn-Strasse 8-10, 10589 Berlin, Germany
| | - Patrick Tarnow
- Department of Chemical and Product Safety, German Federal Institute for Risk Assessment (BfR) , Max-Dohrn-Strasse 8-10, 10589 Berlin, Germany
| | - Harald Jungnickel
- Department of Chemical and Product Safety, German Federal Institute for Risk Assessment (BfR) , Max-Dohrn-Strasse 8-10, 10589 Berlin, Germany
| | - Sven Baumann
- Department of Molecular Systems Biology, UFZ-Helmholtz-Centre for Environmental Research , Permoserstrasse 15, 04318 Leipzig, Germany.,Institute of Pharmacy, Faculty of Biosciences, Pharmacology and Psychology, University of Leipzig , Brüderstrasse 34, 04103 Leipzig, Germany
| | - Martin von Bergen
- Department of Molecular Systems Biology, UFZ-Helmholtz-Centre for Environmental Research , Permoserstrasse 15, 04318 Leipzig, Germany.,Institute of Biochemistry, Faculty of Biosciences, Pharmacology and Psychology, University of Leipzig , Brüderstrasse 34, 04103 Leipzig, Germany.,Department of Biotechnology, Chemistry and Environmental Engineering, Aalborg University , Fredrik Bajers Vej 7K, 9220 Aalborg, Denmark
| | - Tewes Tralau
- Department of Chemical and Product Safety, German Federal Institute for Risk Assessment (BfR) , Max-Dohrn-Strasse 8-10, 10589 Berlin, Germany
| | - Andreas Luch
- Department of Chemical and Product Safety, German Federal Institute for Risk Assessment (BfR) , Max-Dohrn-Strasse 8-10, 10589 Berlin, Germany
| |
Collapse
|
22
|
Naciff JM, Khambatta ZS, Carr GJ, Tiesman JP, Singleton DW, Khan SA, Daston GP. Dose- and Time-Dependent Transcriptional Response of Ishikawa Cells Exposed to Genistein. Toxicol Sci 2016; 151:71-87. [PMID: 26865667 DOI: 10.1093/toxsci/kfw024] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
To further define the utility of the Ishikawa cells as a reliable in vitro model to determine the potential estrogenic activity of chemicals of interest, transcriptional changes induced by genistein (GES) in Ishikawa cells at various doses (10 pM, 1 nM, 100 nM, and 10 μM) and time points (8, 24, and 48 h) were identified using a comprehensive microarray approach. Trend analysis indicated that the expression of 5342 unique genes was modified by GES in a dose- and time-dependent manner (P ≤ 0.0001). However, the majority of gene expression changes induced in Ishikawa cells were elicited by the highest dose of GES evaluated (10 μM). The GES' estrogenic activity was identified by comparing the Ishikawa cells' response to GES versus 17 α-ethynyl estradiol (EE, at equipotent doses, ie, 10 μM vs 1 μM, respectively) and was defined by changes in the expression of 284 unique genes elicited by GES and EE in the same direction, although the magnitude of the change for some genes was different. Further, comparing the response of the Ishikawa cells exposed to high doses of GES and EE versus the response of the juvenile rat uterus exposed to EE, we identified 66 unique genes which were up- or down regulated in a similar manner in vivo as well as in vitro Genistein elicits changes in multiple molecular pathways affecting various biological processes particularly associated with cell organization and biogenesis, regulation of translation, cell proliferation, and intracellular transport; processes also affected by estrogen exposure in the uterus of the rat. These results indicate that Ishikawa cells are capable of generating a biologically relevant estrogenic response and offer an in vitro model to assess this mode of action.
Collapse
Affiliation(s)
- Jorge M Naciff
- *Mason Business Center, The Procter and Gamble Company, Mason, Ohio 45040
| | - Zubin S Khambatta
- *Mason Business Center, The Procter and Gamble Company, Mason, Ohio 45040
| | - Gregory J Carr
- *Mason Business Center, The Procter and Gamble Company, Mason, Ohio 45040
| | - Jay P Tiesman
- *Mason Business Center, The Procter and Gamble Company, Mason, Ohio 45040
| | - David W Singleton
- Department of Cell Biology, Neurobiology, and Anatomy, Vontz Center for Molecular Studies, University of Cincinnati, Cincinnati, Ohio 45267
| | - Sohaib A Khan
- Department of Cell Biology, Neurobiology, and Anatomy, Vontz Center for Molecular Studies, University of Cincinnati, Cincinnati, Ohio 45267
| | - George P Daston
- *Mason Business Center, The Procter and Gamble Company, Mason, Ohio 45040
| |
Collapse
|
23
|
Wang RL, Biales AD, Garcia-Reyero N, Perkins EJ, Villeneuve DL, Ankley GT, Bencic DC. Fish connectivity mapping: linking chemical stressors by their mechanisms of action-driven transcriptomic profiles. BMC Genomics 2016; 17:84. [PMID: 26822894 PMCID: PMC4730593 DOI: 10.1186/s12864-016-2406-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2015] [Accepted: 01/19/2016] [Indexed: 12/21/2022] Open
Abstract
Background A very large and rapidly growing collection of transcriptomic profiles in public repositories is potentially of great value to developing data-driven bioinformatics applications for toxicology/ecotoxicology. Modeled on human connectivity mapping (Cmap) in biomedical research, this study was undertaken to investigate the utility of an analogous Cmap approach in ecotoxicology. Over 3500 zebrafish (Danio rerio) and fathead minnow (Pimephales promelas) transcriptomic profiles, each associated with one of several dozen chemical treatment conditions, were compiled into three distinct collections of rank-ordered gene lists (ROGLs) by species and microarray platforms. Individual query signatures, each consisting of multiple gene probes differentially expressed in a chemical condition, were used to interrogate the reference ROGLs. Results Informative connections were established at high success rates within species when, as defined by their mechanisms of action (MOAs), both query signatures and ROGLs were associated with the same or similar chemicals. Thus, a simple query signature functioned effectively as an exposure biomarker without need for a time-consuming process of development and validation. More importantly, a large reference database of ROGLs also enabled a query signature to cross-interrogate other chemical conditions with overlapping MOAs, leading to novel groupings and subgroupings of seemingly unrelated chemicals at a finer resolution. This approach confirmed the identities of several estrogenic chemicals, as well as a polycyclic aromatic hydrocarbon and a neuro-toxin, in the largely uncharacterized water samples near several waste water treatment plants, and thus demonstrates its future potential utility in real world applications. Conclusions The power of Cmap should grow as chemical coverages of ROGLs increase, making it a framework easily scalable in the future. The feasibility of toxicity extrapolation across fish species using Cmap needs more study, however, as more gene expression profiles linked to chemical conditions common to multiple fish species are needed. Electronic supplementary material The online version of this article (doi:10.1186/s12864-016-2406-y) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Rong-Lin Wang
- Exposure Methods & Measurements Division, National Exposure Research Laboratory, US Environmental Protection Agency, 26 W Martin Luther King Dr., MS 587, Cincinnati, OH, 45268, USA.
| | - Adam D Biales
- Exposure Methods & Measurements Division, National Exposure Research Laboratory, US Environmental Protection Agency, 26 W Martin Luther King Dr., MS 587, Cincinnati, OH, 45268, USA.
| | - Natalia Garcia-Reyero
- Environmental Laboratory, US Army Engineer Research and Development Center, US Army Corps of Engineers, 3909 Halls Ferry Rd, Vicksburg, MS, 39180, USA.
| | - Edward J Perkins
- Environmental Laboratory, US Army Engineer Research and Development Center, US Army Corps of Engineers, 3909 Halls Ferry Rd, Vicksburg, MS, 39180, USA.
| | - Daniel L Villeneuve
- Mid-Continent Ecology Division, National Health and Environmental Effects Research Laboratory, US Environmental Protection Agency, 6201 Congdon Boulevard, Duluth, MN, 55804, USA.
| | - Gerald T Ankley
- Mid-Continent Ecology Division, National Health and Environmental Effects Research Laboratory, US Environmental Protection Agency, 6201 Congdon Boulevard, Duluth, MN, 55804, USA.
| | - David C Bencic
- Exposure Methods & Measurements Division, National Exposure Research Laboratory, US Environmental Protection Agency, 26 W Martin Luther King Dr., MS 587, Cincinnati, OH, 45268, USA.
| |
Collapse
|
24
|
Nodes-and-connections RNAi knockdown screening: identification of a signaling molecule network involved in fulvestrant action and breast cancer prognosis. Oncogenesis 2015; 4:e172. [PMID: 26479444 PMCID: PMC4632093 DOI: 10.1038/oncsis.2015.32] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2015] [Accepted: 09/11/2015] [Indexed: 11/08/2022] Open
Abstract
Although RNA interference (RNAi) knockdown screening of cancer cell cultures is an effective approach to predict drug targets or therapeutic/prognostic biomarkers, interactions among identified targets often remain obscure. Here, we introduce the nodes-and-connections RNAi knockdown screening that generates a map of target interactions through systematic iterations of in silico prediction of targets and their experimental validation. An initial RNAi knockdown screening of MCF-7 human breast cancer cells targeting 6560 proteins identified four signaling molecules required for their fulvestrant-induced apoptosis. Signaling molecules physically or functionally interacting with these four primary node targets were computationally predicted and experimentally validated, resulting in identification of four second-generation nodes. Three rounds of further iterations of the prediction–validation cycle generated third, fourth and fifth generation of nodes, completing a 19-node interaction map that contained three predicted nodes but without experimental validation because of technical limitations. The interaction map involved all three members of the death-associated protein kinases (DAPKs) as well as their upstream and downstream signaling molecules (calmodulins and myosin light chain kinases), suggesting that DAPKs play critical roles in the cytocidal action of fulvestrant. The in silico Kaplan–Meier analysis of previously reported human breast cancer cohorts demonstrated significant prognostic predictive power for five of the experimentally validated nodes and for three of the prediction-only nodes. Immunohistochemical studies on the expression of 10 nodal proteins in human breast cancer tissues not only supported their prognostic prediction power but also provided statistically significant evidence of their synchronized expression, implying functional interactions among these nodal proteins. Thus, the Nodes-and-Connections approach to RNAi knockdown screening yields biologically meaningful outcomes by taking advantage of the existing knowledge of the physical and functional interactions between the predicted target genes. The resulting interaction maps provide useful information on signaling pathways cooperatively involved in clinically important features of the malignant cells, such as drug resistance.
Collapse
|
25
|
Cichon MA, Nelson CM, Radisky DC. Regulation of epithelial-mesenchymal transition in breast cancer cells by cell contact and adhesion. Cancer Inform 2015; 14:1-13. [PMID: 25698877 PMCID: PMC4325704 DOI: 10.4137/cin.s18965] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2014] [Revised: 12/29/2014] [Accepted: 01/04/2015] [Indexed: 02/06/2023] Open
Abstract
Epithelial-mesenchymal transition (EMT) is a physiological program that is activated during cancer cell invasion and metastasis. We show here that EMT-related processes are linked to a broad and conserved program of transcriptional alterations that are influenced by cell contact and adhesion. Using cultured human breast cancer and mouse mammary epithelial cells, we find that reduced cell density, conditions under which cell contact is reduced, leads to reduced expression of genes associated with mammary epithelial cell differentiation and increased expression of genes associated with breast cancer. We further find that treatment of cells with matrix metalloproteinase-3 (MMP-3), an inducer of EMT, interrupts a defined subset of cell contact-regulated genes, including genes encoding a variety of RNA splicing proteins known to regulate the expression of Rac1b, an activated splice isoform of Rac1 known to be a key mediator of MMP-3-induced EMT in breast, lung, and pancreas. These results provide new insights into how MMPs act in cancer progression and how loss of cell-cell interactions is a key step in the earliest stages of cancer development.
Collapse
Affiliation(s)
- Magdalena A Cichon
- Department of Cancer Biology, Mayo Clinic Cancer Center, Jacksonville, FL USA
| | - Celeste M Nelson
- Department of Chemical and Biological Engineering, Princeton University, Princeton, NJ, USA
- Department of Molecular Biology, Princeton University, Princeton, NJ, USA
| | - Derek C Radisky
- Department of Cancer Biology, Mayo Clinic Cancer Center, Jacksonville, FL USA
| |
Collapse
|
26
|
Vaiman D. Reproductive performance: at the cross-road of genetics, technologies and environment. Reprod Fertil Dev 2015; 27:1-13. [DOI: 10.1071/rd14316] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
Sexual reproduction depends on a negotiation between the sexes at the level of the cells (gametes), tissue (trophectoderm of the blastocyst and endometrium in the uterus) and organisms (to allow sexual intercourse). This review evaluates new questions linked to sexual reproduction in the biosphere in the context of the 21st century, in light of current knowledge in genetics and epigenetics. It presents the challenge of ‘forcing reproductive efficiency’ using ineffective gametes, or despite other fertility problems, through medically assisted reproduction and presents the reproductive challenge of high production farm animals, which are in a situation of chronically negative energy balance. It also analyses the situation created by the release of endocrine disruptors into the environment and discusses the possible transgenerational consequences of environmental modifications linked to these compounds.
Collapse
|
27
|
Zoeller RT, Bergman Å, Becher G, Bjerregaard P, Bornman R, Brandt I, Iguchi T, Jobling S, Kidd KA, Kortenkamp A, Skakkebaek NE, Toppari J, Vandenberg LN. A path forward in the debate over health impacts of endocrine disrupting chemicals. Environ Health 2014; 13:118. [PMID: 25533907 PMCID: PMC4298083 DOI: 10.1186/1476-069x-13-118] [Citation(s) in RCA: 79] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2014] [Accepted: 12/08/2014] [Indexed: 05/17/2023]
Abstract
Several recent publications reflect debate on the issue of "endocrine disrupting chemicals" (EDCs), indicating that two seemingly mutually exclusive perspectives are being articulated separately and independently. Considering this, a group of scientists with expertise in basic science, medicine and risk assessment reviewed the various aspects of the debate to identify the most significant areas of dispute and to propose a path forward. We identified four areas of debate. The first is about the definitions for terms such as "endocrine disrupting chemical", "adverse effects", and "endocrine system". The second is focused on elements of hormone action including "potency", "endpoints", "timing", "dose" and "thresholds". The third addresses the information needed to establish sufficient evidence of harm. Finally, the fourth focuses on the need to develop and the characteristics of transparent, systematic methods to review the EDC literature. Herein we identify areas of general consensus and propose resolutions for these four areas that would allow the field to move beyond the current and, in our opinion, ineffective debate.
Collapse
Affiliation(s)
| | - Åke Bergman
- />Swedish Toxicology Sciences Research Center (Swetox), Forskargatan 20, SE-151 36 Sodertalje, Sweden
| | - Georg Becher
- />Norwegian Institute of Public Health, Oslo, Norway
| | | | - Riana Bornman
- />School of Health Systems and Public Health, University of Pretoria, Pretoria, South Africa
| | | | - Taisen Iguchi
- />National Institute for Basic Biology, Okazaki, Japan
| | | | - Karen A Kidd
- />University of New Brunswick, Saint John, New Brunswick, Canada
| | | | | | | | | |
Collapse
|
28
|
Shike M, Doane AS, Russo L, Cabal R, Reis-Filho JS, Gerald W, Cody H, Khanin R, Bromberg J, Norton L. The effects of soy supplementation on gene expression in breast cancer: a randomized placebo-controlled study. J Natl Cancer Inst 2014; 106:dju189. [PMID: 25190728 PMCID: PMC4817128 DOI: 10.1093/jnci/dju189] [Citation(s) in RCA: 88] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2014] [Revised: 05/22/2014] [Accepted: 05/28/2014] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND There are conflicting reports on the impact of soy on breast carcinogenesis. This study examines the effects of soy supplementation on breast cancer-related genes and pathways. METHODS Women (n = 140) with early-stage breast cancer were randomly assigned to soy protein supplementation (n = 70) or placebo (n = 70) for 7 to 30 days, from diagnosis until surgery. Adherence was determined by plasma isoflavones: genistein and daidzein. Gene expression changes were evaluated by NanoString in pre- and posttreatment tumor tissue. Genome-wide expression analysis was performed on posttreatment tissue. Proliferation (Ki67) and apoptosis (Cas3) were assessed by immunohistochemistry. RESULTS Plasma isoflavones rose in the soy group (two-sided Wilcoxon rank-sum test, P < .001) and did not change in the placebo group. In paired analysis of pre- and posttreatment samples, 21 genes (out of 202) showed altered expression (two-sided Student's t-test, P < .05). Several genes including FANCC and UGT2A1 revealed different magnitude and direction of expression changes between the two groups (two-sided Student's t-test, P < .05). A high-genistein signature consisting of 126 differentially expressed genes was identified from microarray analysis of tumors. This signature was characterized by overexpression (>2-fold) of cell cycle transcripts, including those that promote cell proliferation, such as FGFR2, E2F5, BUB1, CCNB2, MYBL2, CDK1, and CDC20 (P < .01). Soy intake did not result in statistically significant changes in Ki67 or Cas3. CONCLUSIONS Gene expression associated with soy intake and high plasma genistein defines a signature characterized by overexpression of FGFR2 and genes that drive cell cycle and proliferation pathways. These findings raise the concerns that in a subset of women soy could adversely affect gene expression in breast cancer.
Collapse
MESH Headings
- Adult
- Aged
- Apoptosis/drug effects
- Biomarkers/blood
- Breast Neoplasms/blood
- Breast Neoplasms/drug therapy
- Breast Neoplasms/metabolism
- Breast Neoplasms/pathology
- Carcinoma, Ductal, Breast/blood
- Carcinoma, Ductal, Breast/drug therapy
- Carcinoma, Ductal, Breast/metabolism
- Carcinoma, Ductal, Breast/pathology
- Caspase 3/metabolism
- Cell Proliferation/drug effects
- Dietary Supplements/adverse effects
- Female
- Gene Expression Regulation, Neoplastic/drug effects
- Genistein/blood
- Humans
- Immunohistochemistry
- Isoflavones/blood
- Ki-67 Antigen/metabolism
- Middle Aged
- Receptor, Fibroblast Growth Factor, Type 2/metabolism
- Soybean Proteins/administration & dosage
- Soybean Proteins/adverse effects
- Tissue Array Analysis
- Up-Regulation
Collapse
Affiliation(s)
- Moshe Shike
- Department of Medicine (MS, AD, LR, JB, LN) and Department of Pathology (RC, JRF, WG) and Department of Surgery (HC) and Department of Computational Biology (RK), Memorial Sloan-Kettering Cancer Center and Weill Cornell Medical College (MS, JRF, WG, HC, JB, LN).
| | - Ashley S Doane
- Department of Medicine (MS, AD, LR, JB, LN) and Department of Pathology (RC, JRF, WG) and Department of Surgery (HC) and Department of Computational Biology (RK), Memorial Sloan-Kettering Cancer Center and Weill Cornell Medical College (MS, JRF, WG, HC, JB, LN)
| | - Lianne Russo
- Department of Medicine (MS, AD, LR, JB, LN) and Department of Pathology (RC, JRF, WG) and Department of Surgery (HC) and Department of Computational Biology (RK), Memorial Sloan-Kettering Cancer Center and Weill Cornell Medical College (MS, JRF, WG, HC, JB, LN)
| | - Rafael Cabal
- Department of Medicine (MS, AD, LR, JB, LN) and Department of Pathology (RC, JRF, WG) and Department of Surgery (HC) and Department of Computational Biology (RK), Memorial Sloan-Kettering Cancer Center and Weill Cornell Medical College (MS, JRF, WG, HC, JB, LN)
| | - Jorge S Reis-Filho
- Department of Medicine (MS, AD, LR, JB, LN) and Department of Pathology (RC, JRF, WG) and Department of Surgery (HC) and Department of Computational Biology (RK), Memorial Sloan-Kettering Cancer Center and Weill Cornell Medical College (MS, JRF, WG, HC, JB, LN)
| | - William Gerald
- Department of Medicine (MS, AD, LR, JB, LN) and Department of Pathology (RC, JRF, WG) and Department of Surgery (HC) and Department of Computational Biology (RK), Memorial Sloan-Kettering Cancer Center and Weill Cornell Medical College (MS, JRF, WG, HC, JB, LN)
| | - Hiram Cody
- Department of Medicine (MS, AD, LR, JB, LN) and Department of Pathology (RC, JRF, WG) and Department of Surgery (HC) and Department of Computational Biology (RK), Memorial Sloan-Kettering Cancer Center and Weill Cornell Medical College (MS, JRF, WG, HC, JB, LN)
| | - Raya Khanin
- Department of Medicine (MS, AD, LR, JB, LN) and Department of Pathology (RC, JRF, WG) and Department of Surgery (HC) and Department of Computational Biology (RK), Memorial Sloan-Kettering Cancer Center and Weill Cornell Medical College (MS, JRF, WG, HC, JB, LN)
| | - Jacqueline Bromberg
- Department of Medicine (MS, AD, LR, JB, LN) and Department of Pathology (RC, JRF, WG) and Department of Surgery (HC) and Department of Computational Biology (RK), Memorial Sloan-Kettering Cancer Center and Weill Cornell Medical College (MS, JRF, WG, HC, JB, LN)
| | - Larry Norton
- Department of Medicine (MS, AD, LR, JB, LN) and Department of Pathology (RC, JRF, WG) and Department of Surgery (HC) and Department of Computational Biology (RK), Memorial Sloan-Kettering Cancer Center and Weill Cornell Medical College (MS, JRF, WG, HC, JB, LN)
| |
Collapse
|