1
|
Tian J, Jin L, Liu H, Hua Z. Stilbenes: a promising small molecule modulator for epigenetic regulation in human diseases. Front Pharmacol 2023; 14:1326682. [PMID: 38155902 PMCID: PMC10754530 DOI: 10.3389/fphar.2023.1326682] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2023] [Accepted: 11/24/2023] [Indexed: 12/30/2023] Open
Abstract
Stilbenes are characterized by a vinyl group connecting two benzene rings to form the basic parent nucleus. Hydrogen atoms on different positions of the benzene rings can be substituted with hydroQxyl groups. These unique structural features confer anti-inflammatory, antibacterial, antiviral, antioxidant, anticancer, cardiovascular protective, and neuroprotective pharmacological effects upon these compounds. Numerous small molecule compounds have demonstrated these pharmacological activities in recent years, including Resveratrol, and Pterostilbene, etc. Tamoxifen and Raloxifene are FDA-approved commonly prescribed synthetic stilbene derivatives. The emphasis is on the potential of these small molecules and their structural derivatives as epigenetic regulators in various diseases. Stilbenes have been shown to modulate epigenetic marks, such as DNA methylation and histone modification, which can alter gene expression patterns and contribute to disease development. This review will discuss the mechanisms by which stilbenes regulate epigenetic marks in various diseases, as well as clinical trials, with a focus on the potential of small molecule and their derivatives such as Resveratrol, Pterostilbene, and Tamoxifen.
Collapse
Affiliation(s)
- Jing Tian
- The State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, Jiangsu, China
| | - Li Jin
- The State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, Jiangsu, China
| | - Hongquan Liu
- Jiangsu Province Hospital on Integration of Chinese and Western Medicine, Nanjing, China
| | - Zichun Hua
- The State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, Jiangsu, China
- Changzhou High-Tech Research Institute of Nanjing University and Jiangsu TargetPharma Laboratories Inc., Changzhou, China
- Nanjing Generecom Biotechnology Co., Ltd., Nanjing, China
| |
Collapse
|
2
|
Hilakivi-Clarke L, de Oliveira Andrade F. Social Isolation and Breast Cancer. Endocrinology 2023; 164:bqad126. [PMID: 37586098 DOI: 10.1210/endocr/bqad126] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Revised: 08/10/2023] [Accepted: 08/11/2023] [Indexed: 08/18/2023]
Abstract
Although the role of life stressors in breast cancer remains unclear, social isolation is consistently associated with increased breast cancer risk and mortality. Social isolation can be defined as loneliness or an absence of perceived social connections. In female mice and rats, social isolation is mimicked by housing animals 1 per cage. Social isolation causes many biological changes, of which an increase in inflammatory markers and disruptions in mitochondrial and cellular metabolism are commonly reported. It is not clear how the 2 traditional stress-induced pathways, namely, the hypothalamic-pituitary-adrenocortical axis (HPA), resulting in a release of glucocorticoids from the adrenal cortex, and autonomic nervous system (ANS), resulting in a release of catecholamines from the adrenal medulla and postganglionic neurons, could explain the increased breast cancer risk in socially isolated individuals. For instance, glucocorticoid receptor activation in estrogen receptor positive breast cancer cells inhibits their proliferation, and activation of β-adrenergic receptor in immature immune cells promotes their differentiation toward antitumorigenic T cells. However, activation of HPA and ANS pathways may cause a disruption in the brain-gut-microbiome axis, resulting in gut dysbiosis. Gut dysbiosis, in turn, leads to an alteration in the production of bacterial metabolites, such as short chain fatty acids, causing a systemic low-grade inflammation and inducing dysfunction in mitochondrial and cellular metabolism. A possible causal link between social isolation-induced increased breast cancer risk and mortality and gut dysbiosis should be investigated, as it offers new tools to prevent breast cancer.
Collapse
Affiliation(s)
- Leena Hilakivi-Clarke
- Department of Food Science and Nutrition, The Hormel Institute, University of Minnesota, Austin, MN 55912, USA
| | - Fabia de Oliveira Andrade
- Department of Food Science and Nutrition, The Hormel Institute, University of Minnesota, Austin, MN 55912, USA
| |
Collapse
|
3
|
Andrade FDO, Jin L, Clarke R, Wood I, Dutton M, Anjorin C, Rubin G, Gao A, Sengupta S, FitzGerald K, Hilakivi-Clarke L. Social Isolation Activates Dormant Mammary Tumors, and Modifies Inflammatory and Mitochondrial Metabolic Pathways in the Rat Mammary Gland. Cells 2023; 12:961. [PMID: 36980301 PMCID: PMC10047513 DOI: 10.3390/cells12060961] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2023] [Revised: 02/27/2023] [Accepted: 03/16/2023] [Indexed: 03/30/2023] Open
Abstract
Although multifactorial in origin, one of the most impactful consequences of social isolation is an increase in breast cancer mortality. How this happens is unknown, but many studies have shown that social isolation increases circulating inflammatory cytokines and impairs mitochondrial metabolism. Using a preclinical Sprague Dawley rat model of estrogen receptor-positive breast cancer, we investigated whether social isolation impairs the response to tamoxifen therapy and increases the risk of tumors emerging from dormancy, and thus their recurrence. We also studied which signaling pathways in the mammary glands may be affected by social isolation in tamoxifen treated rats, and whether an anti-inflammatory herbal mixture blocks the effects of social isolation. Social isolation increased the risk of dormant mammary tumor recurrence after tamoxifen therapy. The elevated recurrence risk was associated with changes in multiple signaling pathways including an upregulation of IL6/JAK/STAT3 signaling in the mammary glands and tumors and suppression of the mitochondrial oxidative phosphorylation (OXPHOS) pathway. In addition, social isolation increased the expression of receptor for advanced glycation end-products (RAGE), consistent with impaired insulin sensitivity and weight gain linked to social isolation. In socially isolated animals, the herbal product inhibited IL6/JAK/STAT3 signaling, upregulated OXPHOS signaling, suppressed the expression of RAGE ligands S100a8 and S100a9, and prevented the increase in recurrence of dormant mammary tumors. Increased breast cancer mortality among socially isolated survivors may be most effectively prevented by focusing on the period following the completion of hormone therapy using interventions that simultaneously target several different pathways including inflammatory and mitochondrial metabolism pathways.
Collapse
Affiliation(s)
- Fabia de Oliveira Andrade
- Department of Oncology, Georgetown University, Washington, DC 20057, USA; (F.d.O.A.)
- Hormel Institute, University of Minnesota, 801 16th Ave NE, Austin, MN 55912, USA
| | - Lu Jin
- Department of Oncology, Georgetown University, Washington, DC 20057, USA; (F.d.O.A.)
- Hormel Institute, University of Minnesota, 801 16th Ave NE, Austin, MN 55912, USA
| | - Robert Clarke
- Department of Oncology, Georgetown University, Washington, DC 20057, USA; (F.d.O.A.)
- Hormel Institute, University of Minnesota, 801 16th Ave NE, Austin, MN 55912, USA
| | - Imani Wood
- Department of Oncology, Georgetown University, Washington, DC 20057, USA; (F.d.O.A.)
| | - MaryAnn Dutton
- Department of Psychiatry, Georgetown University, Washington, DC 20057, USA
| | - Chezaray Anjorin
- Department of Oncology, Georgetown University, Washington, DC 20057, USA; (F.d.O.A.)
| | - Grace Rubin
- Department of Oncology, Georgetown University, Washington, DC 20057, USA; (F.d.O.A.)
| | - Audrey Gao
- Hormel Institute, University of Minnesota, 801 16th Ave NE, Austin, MN 55912, USA
| | - Surojeet Sengupta
- Department of Oncology, Georgetown University, Washington, DC 20057, USA; (F.d.O.A.)
- Hormel Institute, University of Minnesota, 801 16th Ave NE, Austin, MN 55912, USA
| | - Kevin FitzGerald
- Department of Oncology, Georgetown University, Washington, DC 20057, USA; (F.d.O.A.)
- Department of Medical Humanities, Creighton University, Omaha, NE 68178, USA
| | - Leena Hilakivi-Clarke
- Department of Oncology, Georgetown University, Washington, DC 20057, USA; (F.d.O.A.)
- Hormel Institute, University of Minnesota, 801 16th Ave NE, Austin, MN 55912, USA
| |
Collapse
|
4
|
Wang Y, Huang Z, Sun M, Huang W, Xia L. ETS transcription factors: Multifaceted players from cancer progression to tumor immunity. Biochim Biophys Acta Rev Cancer 2023; 1878:188872. [PMID: 36841365 DOI: 10.1016/j.bbcan.2023.188872] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Revised: 01/18/2023] [Accepted: 01/28/2023] [Indexed: 02/26/2023]
Abstract
The E26 transformation specific (ETS) family comprises 28 transcription factors, the majority of which are involved in tumor initiation and development. Serving as a group of functionally heterogeneous gene regulators, ETS factors possess a structurally conserved DNA-binding domain. As one of the most prominent families of transcription factors that control diverse cellular functions, ETS activation is modulated by multiple intracellular signaling pathways and post-translational modifications. Disturbances in ETS activity often lead to abnormal changes in oncogenicity, including cancer cell survival, growth, proliferation, metastasis, genetic instability, cell metabolism, and tumor immunity. This review systematically addresses the basics and advances in studying ETS factors, from their tumor relevance to clinical translational utility, with a particular focus on elucidating the role of ETS family in tumor immunity, aiming to decipher the vital role and clinical potential of regulation of ETS factors in the cancer field.
Collapse
Affiliation(s)
- Yufei Wang
- Department of Gastroenterology, Institute of Liver and Gastrointestinal Diseases, Hubei Key Laboratory of Hepato-Pancreato-Biliary Diseases, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, Hubei Province, China
| | - Zhao Huang
- Hubei Key Laboratory of Hepato-Pancreato-Biliary Diseases, Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Clinical Medicine Research Center for Hepatic Surgery of Hubei Province, Key Laboratory of Organ Transplantation, Ministry of Education and Ministry of Public Health, Wuhan, Hubei 430030, China
| | - Mengyu Sun
- Department of Gastroenterology, Institute of Liver and Gastrointestinal Diseases, Hubei Key Laboratory of Hepato-Pancreato-Biliary Diseases, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, Hubei Province, China
| | - Wenjie Huang
- Hubei Key Laboratory of Hepato-Pancreato-Biliary Diseases, Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Clinical Medicine Research Center for Hepatic Surgery of Hubei Province, Key Laboratory of Organ Transplantation, Ministry of Education and Ministry of Public Health, Wuhan, Hubei 430030, China.
| | - Limin Xia
- Department of Gastroenterology, Institute of Liver and Gastrointestinal Diseases, Hubei Key Laboratory of Hepato-Pancreato-Biliary Diseases, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, Hubei Province, China.
| |
Collapse
|
5
|
Shan J, Geng R, Zhang Y, Wei J, Liu J, Bai J. Identification of cuproptosis-related subtypes, establishment of a prognostic model and tumor immune landscape in endometrial carcinoma. Comput Biol Med 2022; 149:105988. [PMID: 36007289 DOI: 10.1016/j.compbiomed.2022.105988] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2022] [Revised: 07/05/2022] [Accepted: 08/14/2022] [Indexed: 11/25/2022]
Abstract
Cuproptosis, the mechanism of copper-dependent cell death, is distinct from all other known forms of regulated cell death and dependents on mitochondrial respiration. Cuproptosis promises to be a novel treatment, especially for tumors resistant to conventional therapies. We investigated the changes in cuproptosis-related genes (CRGs) in endometrial cancer (EC) cohorts from the merged Gene Expression Omnibus and the Cancer Genome Atlas databases, which could be divided into three distinct CRGclusters. Patients in CRGcluster C would have higher survival probability (P = 0.007), and higher levels of tumor microenvironment (TME) cell infiltration than other CRGclusters. CRG score was calculated via the results of univariate, multivariate cox analysis and least absolute shrinkage and selection operator regression analysis. Patients were divided into two risk subgroups according to the median risk score. Low-risk patients exhibited a more favorable prognosis, higher immunogenicity, and greater immunotherapy efficacy. Besides, CRG scores were strongly correlated to copy number variation, immunophenoscore, tumor mutation load, cancer stem cell index, microsatellite instability, and chemosensitivity. The c-index of our model is 0.702, which is higher than other four published model. The results proved that our model can distinguish EC patients with high-risk and low-risk and accurately predict the prognosis of EC patients. It will provide new ideas for clinical prognosis and precise treatments.
Collapse
Affiliation(s)
- Jingsong Shan
- Division of Natural and Applied Sciences, Duke Kunshan University, Kunshan, Jiangsu, 215316, China.
| | - Rui Geng
- Department of Biostatistics, School of Public Health, Nanjing Medical University, Nanjing, Jiangsu, 211166, China.
| | - Yue Zhang
- Department of Nutrition, Nanjing Maternity and Child Health Care Hospital, Women's Hospital of Nanjing Medical University, Nanjing, Jiangsu, 210004, China.
| | - Junting Wei
- The Second Clinical School of Nanjing Medical University, Nanjing, Jiangsu, 211166, China.
| | - Jinhui Liu
- Department of Gynecology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, 210029, China.
| | - Jianling Bai
- Department of Biostatistics, School of Public Health, Nanjing Medical University, Nanjing, Jiangsu, 211166, China.
| |
Collapse
|
6
|
Fiorin LG, Matheus HR, Ervolino E, Canciani E, Pellegrini G, Dellavia C, Maiorana C, de Almeida JM. Tamoxifen improves homeostasis in the peri-implant bone remodeling of osseointegrated titanium implants. J Periodontal Res 2022; 57:880-890. [PMID: 35856857 DOI: 10.1111/jre.13026] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Revised: 04/19/2022] [Accepted: 05/29/2022] [Indexed: 11/29/2022]
Abstract
BACKGROUND The purpose of this preclinical study was to evaluate the influence of tamoxifen (TAM) on the peri-implant bone remodeling of osseointegrated titanium implants in ovariectomized female rats. MATERIALS AND METHODS Seventy-two female rats underwent bilateral ovariectomy 20 weeks before implants placement. One titanium implant was inserted in each tibia of the animals. Six weeks following the implant surgery, animals were randomly divided into two experimental groups (n = 36), which received either saline solution (SS) or tamoxifen citrate (TAM) via gavage until euthanasia. Euthanasia was performed at 30, 60, and 90 days after the first gavage. Assessments of bone to implant contact (BIC), bone ingrowth percentage (BIN), morphological description of cellular and tissue reactions, immunohistochemistry for the detection of bone morphogenetic protein 2/4 (BMP2/4), runt-related transcription factor 2 (RUNX-2), osteocalcin (OCN) and tartrate-resistant acid phosphatase (TRAP), and bone chemical composition through scanning electron microscopy with energy-dispersive x-ray spectroscopy were performed. RESULTS Tamoxifen group presented higher BIC, higher BIN, higher RUNX-2 and OCN, lower TRAP-positive cells/mm2 , and no differences regarding BMP-2/4 positive cells/mm2 than SS group in all periods. TAM group also showed higher Ca/P rate than SS group. CONCLUSION Tamoxifen enhanced the remodeling of the bone surrounding titanium implants in ovariectomized rats.
Collapse
Affiliation(s)
- Luiz Guilherme Fiorin
- Department of Diagnosis and Surgery, Division of Periodontics, São Paulo State University "Júlio de Mesquita Filho" (UNESP), Araçatuba, Brazil
- Nucleus of Study and Research in Periodontics and Implantology (NEPPI), School of Dentistry, Sao Paulo State University (UNESP), Aracatuba, Brazil
| | - Henrique Rinaldi Matheus
- Department of Diagnosis and Surgery, Division of Periodontics, São Paulo State University "Júlio de Mesquita Filho" (UNESP), Araçatuba, Brazil
- Nucleus of Study and Research in Periodontics and Implantology (NEPPI), School of Dentistry, Sao Paulo State University (UNESP), Aracatuba, Brazil
| | - Edilson Ervolino
- Department of Basics Sciences Clinic, Araçatuba School of Dentistry Sao Paulo, Sao Paulo State University (UNESP), Araçatuba, Brazil
| | - Elena Canciani
- Thin Section Laboratory, Department of Biomedical, Surgical and Dental Sciences, Università degli Studi di Milano Statale (UNIMI), Milan, Italy
| | - Gaia Pellegrini
- Thin Section Laboratory, Department of Biomedical, Surgical and Dental Sciences, Università degli Studi di Milano Statale (UNIMI), Milan, Italy
| | - Claudia Dellavia
- Thin Section Laboratory, Department of Biomedical, Surgical and Dental Sciences, Università degli Studi di Milano Statale (UNIMI), Milan, Italy
| | - Carlo Maiorana
- Implant Center for Edentulism and Jawbone Atrophies, Maxillofacial Surgery and Odontostomatology Unit, Fondazione IRCCS Ca 'Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Juliano Milanezi de Almeida
- Department of Diagnosis and Surgery, Division of Periodontics, São Paulo State University "Júlio de Mesquita Filho" (UNESP), Araçatuba, Brazil
- Nucleus of Study and Research in Periodontics and Implantology (NEPPI), School of Dentistry, Sao Paulo State University (UNESP), Aracatuba, Brazil
| |
Collapse
|
7
|
Rezaie N, Bayati M, Hamidi M, Tahaei MS, Khorasani S, Lovell NH, Breen J, Rabiee HR, Alinejad-Rokny H. Somatic point mutations are enriched in non-coding RNAs with possible regulatory function in breast cancer. Commun Biol 2022; 5:556. [PMID: 35672401 PMCID: PMC9174258 DOI: 10.1038/s42003-022-03528-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2021] [Accepted: 05/24/2022] [Indexed: 11/09/2022] Open
Abstract
Non-coding RNAs (ncRNAs) form a large portion of the mammalian genome. However, their biological functions are poorly characterized in cancers. In this study, using a newly developed tool, SomaGene, we analyze de novo somatic point mutations from the International Cancer Genome Consortium (ICGC) whole-genome sequencing data of 1,855 breast cancer samples. We identify 1030 candidates of ncRNAs that are significantly and explicitly mutated in breast cancer samples. By integrating data from the ENCODE regulatory features and FANTOM5 expression atlas, we show that the candidate ncRNAs significantly enrich active chromatin histone marks (1.9 times), CTCF binding sites (2.45 times), DNase accessibility (1.76 times), HMM predicted enhancers (2.26 times) and eQTL polymorphisms (1.77 times). Importantly, we show that the 1030 ncRNAs contain a much higher level (3.64 times) of breast cancer-associated genome-wide association (GWAS) single nucleotide polymorphisms (SNPs) than genome-wide expectation. Such enrichment has not been seen with GWAS SNPs from other cancers. Using breast cell line related Hi-C data, we then show that 82% of our candidate ncRNAs (1.9 times) significantly interact with the promoter of protein-coding genes, including previously known cancer-associated genes, suggesting the critical role of candidate ncRNA genes in the activation of essential regulators of development and differentiation in breast cancer. We provide an extensive web-based resource (https://www.ihealthe.unsw.edu.au/research) to communicate our results with the research community. Our list of breast cancer-specific ncRNA genes has the potential to provide a better understanding of the underlying genetic causes of breast cancer. Lastly, the tool developed in this study can be used to analyze somatic mutations in all cancers. The SomaGene tool is developed to identify non-coding RNAs (ncRNAs) mutated in breast cancer but can be used for other cancers. Candidate ncRNAs are shown to be enriched for regulatory features and to contain specific trait loci polymorphisms.
Collapse
Affiliation(s)
- Narges Rezaie
- Center for Complex Biological Systems, University of California Irvine, Irvine, CA, 92697, USA
| | - Masroor Bayati
- Bioinformatics and Computational Biology Lab, Department of Computer Engineering, Sharif University of Technology, Tehran, 11365, Iran
| | - Mehrab Hamidi
- Bioinformatics and Computational Biology Lab, Department of Computer Engineering, Sharif University of Technology, Tehran, 11365, Iran
| | - Maedeh Sadat Tahaei
- Bioinformatics and Computational Biology Lab, Department of Computer Engineering, Sharif University of Technology, Tehran, 11365, Iran
| | - Sadegh Khorasani
- Bioinformatics and Computational Biology Lab, Department of Computer Engineering, Sharif University of Technology, Tehran, 11365, Iran
| | - Nigel H Lovell
- Tyree Institute of Health Engineering and The Graduate School of Biomedical Engineering, UNSW Sydney, Sydney, NSW, 2052, Australia
| | - James Breen
- South Australian Health & Medical Research Institute, Adelaide, SA, 5000, Australia.,Robinson Research Institute, University of Adelaide, Adelaide, SA, 5006, Australia.,Bioinformatics Hub, University of Adelaide, Adelaide, SA, 5006, Australia
| | - Hamid R Rabiee
- Bioinformatics and Computational Biology Lab, Department of Computer Engineering, Sharif University of Technology, Tehran, 11365, Iran.
| | - Hamid Alinejad-Rokny
- BioMedical Machine Learning Lab (BML), The Graduate School of Biomedical Engineering, UNSW Sydney, Sydney, NSW, 2052, Australia. .,UNSW Data Science Hub, The University of New South Wales (UNSW Sydney), Sydney, NSW, 2052, Australia. .,Health Data Analytics Program, AI-enabled Processes (AIP) Research Centre, Macquarie University, Sydney, NSW, 2109, Australia.
| |
Collapse
|
8
|
Integrated Genomic and Bioinformatics Approaches to Identify Molecular Links between Endocrine Disruptors and Adverse Outcomes. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2022; 19:ijerph19010574. [PMID: 35010832 PMCID: PMC8744944 DOI: 10.3390/ijerph19010574] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/09/2021] [Revised: 12/13/2021] [Accepted: 12/21/2021] [Indexed: 12/04/2022]
Abstract
Exposure to Endocrine Disrupting Chemicals (EDC) has been linked with several adverse outcomes. In this review, we examine EDCs that are pervasive in the environment and are of concern in the context of human, animal, and environmental health. We explore the consequences of EDC exposure on aquatic life, terrestrial animals, and humans. We focus on the exploitation of genomics technologies and in particular whole transcriptome sequencing. Genome-wide analyses using RNAseq provides snap shots of cellular, tissue and whole organism transcriptomes under normal physiological and EDC perturbed conditions. A global view of gene expression provides highly valuable information as it uncovers gene families or more specifically, pathways that are affected by EDC exposures, but also reveals those that are unaffected. Hypotheses about genes with unknown functions can also be formed by comparison of their expression levels with genes of known function. Risk assessment strategies leveraging genomic technologies and the development of toxicology databases are explored. Finally, we review how the Adverse Outcome Pathway (AOP) has exploited this high throughput data to provide a framework for toxicology studies.
Collapse
|
9
|
Jiang W, Xu Y, Chen X, Pan S, Zhu X. E26 transformation-specific variant 4 as a tumor promotor in human cancers through specific molecular mechanisms. Mol Ther Oncolytics 2021; 22:518-527. [PMID: 34553037 PMCID: PMC8433062 DOI: 10.1016/j.omto.2021.07.012] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Abstract
E26 transformation-specific (ETS) variant 4 (ETV4) is an important transcription factor that belongs to the ETS transcription factor family and is essential for much cellular physiology. Recent evidence has revealed that ETV4 is aberrantly expressed in many types of tumors, and its overexpression is related to poor prognosis of cancer patients. Additionally, increasing studies have identified that ETV4 promotes cancer growth, invasion, metastasis, and drug resistance. Mechanistically, the level of ETV4 is regulated by some post-translation modulations in a broad spectrum of cancers. However, little progress has been made to comprehensively summarize the critical roles of ETV4 in different human cancers. Hence, this review mainly focuses on the physiological functions of ETV4 in various human tumors. In addition, the molecular mechanisms of ETV4-mediated cancer progression were elucidated, including how ETV4 modulates its downstream signaling pathways and how ETV4 is regulated by some factors. On this basis, the present review may provide a valuable therapeutics strategy for future cancer treatment by targeting ETV4-related pathways.
Collapse
Affiliation(s)
- Wenxiao Jiang
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou 325027, China
| | - Yichi Xu
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou 325027, China
| | - Xin Chen
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou 325027, China
| | - Shuya Pan
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou 325027, China
| | - Xueqiong Zhu
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou 325027, China
| |
Collapse
|
10
|
Clarke R, Jones BC, Sevigny CM, Hilakivi-Clarke LA, Sengupta S. Experimental models of endocrine responsive breast cancer: strengths, limitations, and use. CANCER DRUG RESISTANCE (ALHAMBRA, CALIF.) 2021; 4:762-783. [PMID: 34532657 PMCID: PMC8442978 DOI: 10.20517/cdr.2021.33] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Breast cancers characterized by expression of estrogen receptor-alpha (ER; ESR1) represent approximately 70% of all new cases and comprise the largest molecular subtype of this disease. Despite this high prevalence, the number of adequate experimental models of ER+ breast cancer is relatively limited. Nonetheless, these models have proved very useful in advancing understanding of how cells respond to and resist endocrine therapies, and how the ER acts as a transcription factor to regulate cell fate signaling. We discuss the primary experimental models of ER+ breast cancer including 2D and 3D cultures of established cell lines, cell line- and patient-derived xenografts, and chemically induced rodent models, with a consideration of their respective general strengths and limitations. What can and cannot be learned easily from these models is also discussed, and some observations on how these models may be used more effectively are provided. Overall, despite their limitations, the panel of models currently available has enabled major advances in the field, and these models remain central to the ability to study mechanisms of therapy action and resistance and for hypothesis testing that would otherwise be intractable or unethical in human subjects.
Collapse
Affiliation(s)
- Robert Clarke
- The Hormel Institute and Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, Austin, MN 55912, USA
| | - Brandon C Jones
- Department of Oncology, Georgetown University Medical Center, Washington, DC 20057, USA
| | - Catherine M Sevigny
- Department of Oncology, Georgetown University Medical Center, Washington, DC 20057, USA.,The Jackson Laboratory, 600 Main Street, Bar Harbor, ME 04609, USA
| | - Leena A Hilakivi-Clarke
- The Hormel Institute and Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, Austin, MN 55912, USA
| | - Surojeet Sengupta
- The Hormel Institute and Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, Austin, MN 55912, USA
| |
Collapse
|
11
|
Xiang F, Zhu Z, Zhang M, Wang J, Chen Z, Li X, Zhang T, Gu Q, Wu R, Kang X. 3,3'-Diindolylmethane Enhances Paclitaxel Sensitivity by Suppressing DNMT1-Mediated KLF4 Methylation in Breast Cancer. Front Oncol 2021; 11:627856. [PMID: 34150611 PMCID: PMC8209418 DOI: 10.3389/fonc.2021.627856] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2021] [Accepted: 05/12/2021] [Indexed: 12/11/2022] Open
Abstract
Paclitaxel (PTX) is a first-line chemotherapeutic drug for the treatment of breast cancer, but drug resistance seriously limits its clinical use. The aim of the present work was to explore the effect of 3,3’-diindolylmethane (DIM) on PTX sensitivity and its possible mechanism in breast cancer. The expression of Krüppel-like factor 4 (KLF4) and DNA-methyltransferase 1 (DNMT1) in breast cancer tissues were assessed by immunohistochemistry and Western blotting. The methylation of KLF4 was evaluated by the MassARRAY platform. The lentivirus carrying KLF4 and DNMT1 gene or shRNA targeting DNMT1 were used to overexpress KLF4 or knockdown DNMT1 in MCF-7 and T47D breast cancer cells and the role of KLF4 and DNMT1 in regulation of PTX sensitivity was investigated. The effect of PTX on inhibiting the proliferation of MCF-7 and T47D cells was measured by CCK-8 assay. Flow cytometry was used to examine cell apoptosis. The expression of mRNA and protein was evaluated by qRT-PCR and Western blotting analysis, respectively. Our data showed that the expression of DNMT1 was increased, and the methylation level of CpG sites (−148 bp) in the KLF4 promoter was increased while the KLF4 expression was significantly decreased in breast cancer tissues. Overexpression of KLF4 increased the sensitivity of MCF-7 and T47D cells to PTX. DNMT1 increased the methylation of the KLF4 promoter and decrease the expression of KLF4. Knockdown of DNMT1 increased the sensitivity of MCF-7 and T47D cells to PTX. DIM enhanced the PTX sensitivity of MCF-7 and T47D cells, decreased the expression of DNMT1 and the methylation level of KLF4 promoter, thus increasing the level of KLF4. Furthermore, overexpression of DNMT1 attenuated the effect of DIM on the regulation of PTX sensitivity. Collectively, our data indicated that DNMT1-mediated hypermethylation of KLF4 promoter leads to downregulation of KLF4 in breast cancer. The level of KLF4 is correlated with the sensitivity of MCF-7 and T47D cells to PTX. DIM could enhance the antitumor efficacy of PTX on MCF-7 and T47D cells by regulating DNMT1 and KLF4.
Collapse
Affiliation(s)
- Fenfen Xiang
- Laboratory Medicine, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Zhaowei Zhu
- Laboratory Medicine, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Mengzhe Zhang
- Laboratory Medicine, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Jie Wang
- General Surgery, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Zixi Chen
- Laboratory Medicine, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Xiaoxiao Li
- Laboratory Medicine, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Tao Zhang
- Laboratory Medicine, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Qing Gu
- Laboratory Medicine, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Rong Wu
- Laboratory Medicine, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Xiangdong Kang
- Laboratory Medicine, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| |
Collapse
|
12
|
Abstract
Despite the decline in death rate from breast cancer and recent advances in targeted therapies and combinations for the treatment of metastatic disease, metastatic breast cancer remains the second leading cause of cancer-associated death in U.S. women. The invasion-metastasis cascade involves a number of steps and multitudes of proteins and signaling molecules. The pathways include invasion, intravasation, circulation, extravasation, infiltration into a distant site to form a metastatic niche, and micrometastasis formation in a new environment. Each of these processes is regulated by changes in gene expression. Noncoding RNAs including microRNAs (miRNAs) are involved in breast cancer tumorigenesis, progression, and metastasis by post-transcriptional regulation of target gene expression. miRNAs can stimulate oncogenesis (oncomiRs), inhibit tumor growth (tumor suppressors or miRsupps), and regulate gene targets in metastasis (metastamiRs). The goal of this review is to summarize some of the key miRNAs that regulate genes and pathways involved in metastatic breast cancer with an emphasis on estrogen receptor α (ERα+) breast cancer. We reviewed the identity, regulation, human breast tumor expression, and reported prognostic significance of miRNAs that have been documented to directly target key genes in pathways, including epithelial-to-mesenchymal transition (EMT) contributing to the metastatic cascade. We critically evaluated the evidence for metastamiRs and their targets and miRNA regulation of metastasis suppressor genes in breast cancer progression and metastasis. It is clear that our understanding of miRNA regulation of targets in metastasis is incomplete.
Collapse
Affiliation(s)
- Belinda J Petri
- Department of Biochemistry and Molecular Genetics, University of Louisville School of Medicine, Louisville, KY, 40292, USA
| | - Carolyn M Klinge
- Department of Biochemistry and Molecular Genetics, University of Louisville School of Medicine, Louisville, KY, 40292, USA.
| |
Collapse
|
13
|
Zhang X, de Oliveira Andrade F, Zhang H, Cruz I, Clarke R, Gaur P, Verma V, Hilakivi-Clarke L. Maternal obesity increases offspring's mammary cancer recurrence and impairs tumor immune response. Endocr Relat Cancer 2020; 27:469-482. [PMID: 32580156 PMCID: PMC7424355 DOI: 10.1530/erc-20-0065] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/26/2020] [Accepted: 06/22/2020] [Indexed: 12/14/2022]
Abstract
Over 50% of women at a childbearing age in the United States are overweight or obese, and this can adversely affect their offspring. We studied if maternal obesity-inducing high fat diet (HFD) not only increases offspring's mammary cancer risk but also impairs response to antiestrogen tamoxifen. Female rat offspring of HFD and control diet-fed dams, in which estrogen receptor-positive (ER+) mammary tumors were induced with the carcinogen 7,12-dimethylbenz[a]anthracene (DMBA), exhibited similar initial responses to antiestrogen tamoxifen. However, after tamoxifen therapy was completed, almost all (91%) tumors recurred in HFD offspring, compared with only 29% in control offspring. The increase in local mammary tumor recurrence in HFD offspring was linked to an increase in the markers of immunosuppression (Il17f, Tgfβ1, VEGFR2) in the tumor microenvironment (TME). Protein and mRNA levels of the major histocompatibility complex II (MHC-II), but not MHC-I, were reduced in the recurring DMBA tumors of HFD offspring. Further, infiltration of CD8+ effector T cells and granzyme B+ (GZMB+) cells were lower in their recurring tumors. To determine if maternal HFD can pre-program similar changes in the TME of allografted E0771 mammary tumors in offspring of syngeneic mice, flow cytometry analysis was performed. E0771 mammary tumor growth was significantly accelerated in the HFD offspring, and a reduction in the numbers of GZMB and non-significant reduction of interferon γ (IFNγ) secreting CD8+ T cells in the TME was seen. Thus, consumption of a HFD during pregnancy increases susceptibility of the female rat and mouse offspring to tumor immune suppression and mammary tumor growth and recurrence.
Collapse
Affiliation(s)
| | | | | | | | - Robert Clarke
- Department of Oncology, Georgetown University, Washington, District of Columbia, USA
| | - Pankaj Gaur
- Department of Oncology, Georgetown University, Washington, District of Columbia, USA
| | - Vivek Verma
- Department of Oncology, Georgetown University, Washington, District of Columbia, USA
| | - Leena Hilakivi-Clarke
- Department of Oncology, Georgetown University, Washington, District of Columbia, USA
- Correspondence should be addressed to L Hilakivi-Clarke:
| |
Collapse
|
14
|
Andrade FDO, Nguyen NM, Warri A, Hilakivi-Clarke L. Reversal of increased mammary tumorigenesis by valproic acid and hydralazine in offspring of dams fed high fat diet during pregnancy. Sci Rep 2019; 9:20271. [PMID: 31889127 PMCID: PMC6937280 DOI: 10.1038/s41598-019-56854-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2019] [Accepted: 12/17/2019] [Indexed: 12/14/2022] Open
Abstract
Maternal or paternal high fat (HF) diet can modify the epigenome in germ cells and fetal somatic cells leading to an increased susceptibility among female offspring of multiple generations to develop breast cancer. We determined if combined treatment with broad spectrum DNA methyltransferase (DNMT) inhibitor hydralazine and histone deacetylase (HDAC) inhibitor valproic acid (VPA) will reverse this increased risk. C57BL/6 mouse dams were fed either a corn oil-based HF or control diet during pregnancy. Starting at age 7 weeks, female offspring were administered 3 doses of 7,12-dimethylbenz[a]anthracene (DMBA) to initiate mammary cancer. After last dose, offspring started receiving VPA/hydralazine administered via drinking water: no adverse health effects were detected. VPA/hydralazine reduced mammary tumor multiplicity and lengthened tumor latency in HF offspring when compared with non-treated HF offspring. The drug combination inhibited DNMT3a protein levels and increased expression of the tumor suppressor gene Cdkn2a/p16 in mammary tumors of HF offspring. In control mice not exposed to HF diet in utero, VPA/hydralazine increased mammary tumor incidence and burden, and elevated expression of the unfolded protein response and autophagy genes, including HIF-1α, NFkB, PERK, and SQSTM1/p62. Expression of these genes was already upregulated in HF offspring prior to VPA/hydralazine treatment. These findings suggest that breast cancer prevention strategies with HDAC/DNMT inhibitors need to be individually tailored.
Collapse
Affiliation(s)
| | - N M Nguyen
- Department of Oncology, Georgetown University, Washington, DC, USA
| | - A Warri
- Department of Oncology, Georgetown University, Washington, DC, USA.,Institute of Biomedicine, University of Turku Medical Faculty, FI-20014, Turku, Finland
| | | |
Collapse
|
15
|
Ramirez MU, Hernandez SR, Soto-Pantoja DR, Cook KL. Endoplasmic Reticulum Stress Pathway, the Unfolded Protein Response, Modulates Immune Function in the Tumor Microenvironment to Impact Tumor Progression and Therapeutic Response. Int J Mol Sci 2019; 21:ijms21010169. [PMID: 31881743 PMCID: PMC6981480 DOI: 10.3390/ijms21010169] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2019] [Revised: 12/04/2019] [Accepted: 12/09/2019] [Indexed: 01/18/2023] Open
Abstract
Despite advances in cancer therapy, several persistent issues remain. These include cancer recurrence, effective targeting of aggressive or therapy-resistant cancers, and selective treatments for transformed cells. This review evaluates the current findings and highlights the potential of targeting the unfolded protein response to treat cancer. The unfolded protein response, an evolutionarily conserved pathway in all eukaryotes, is initiated in response to misfolded proteins accumulating within the lumen of the endoplasmic reticulum. This pathway is initially cytoprotective, allowing cells to survive stressful events; however, prolonged activation of the unfolded protein response also activates apoptotic responses. This balance is key in successful mammalian immune response and inducing cell death in malignant cells. We discuss how the unfolded protein response affects cancer progression, survival, and immune response to cancer cells. The literature shows that targeting the unfolded protein response as a monotherapy or in combination with chemotherapy or immunotherapies increases the efficacy of these drugs; however, systemic unfolded protein response targeting may yield deleterious effects on immune cell function and should be taken into consideration. The material in this review shows the promise of both approaches, each of which merits further research.
Collapse
Affiliation(s)
- Manuel U. Ramirez
- Department of Physiology and Pharmacology, Wake Forest University Health Sciences, Winston-Salem, NC 27157, USA
| | | | - David R. Soto-Pantoja
- Department of Surgery, Wake Forest School of Medicine, Winston-Salem, NC 27157, USA;
- Department of Cancer Biology, Wake Forest University Health Sciences, Winston Salem, NC 27157, USA
| | - Katherine L. Cook
- Department of Surgery, Wake Forest School of Medicine, Winston-Salem, NC 27157, USA;
- Department of Cancer Biology, Wake Forest University Health Sciences, Winston Salem, NC 27157, USA
- Correspondence: ; Tel.: +01-336-716-2234
| |
Collapse
|
16
|
Linares A, Assou S, Lapierre M, Thouennon E, Duraffourd C, Fromaget C, Boulahtouf A, Tian G, Ji J, Sahin O, Badia E, Boulle N, Cavaillès V. Increased expression of the HDAC9 gene is associated with antiestrogen resistance of breast cancers. Mol Oncol 2019; 13:1534-1547. [PMID: 31099456 PMCID: PMC6599838 DOI: 10.1002/1878-0261.12505] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2018] [Revised: 04/26/2019] [Accepted: 05/15/2019] [Indexed: 12/19/2022] Open
Abstract
Estrogens play a pivotal role in breast cancer etiology, and endocrine therapy remains the main first line treatment for estrogen receptor‐alpha (ERα)‐positive breast cancer. ER are transcription factors whose activity is finely regulated by various regulatory complexes, including histone deacetylases (HDACs). Here, we investigated the role of HDAC9 in ERα signaling and response to antiestrogens in breast cancer cells. Various Michigan Cancer Foundation‐7 (MCF7) breast cancer cell lines that overexpress class IIa HDAC9 or that are resistant to the partial antiestrogen 4‐hydroxy‐tamoxifen (OHTam) were used to study phenotypic changes in response to ER ligands by using transcriptomic and gene set enrichment analyses. Kaplan–Meier survival analyses were performed using public transcriptomic datasets from human breast cancer biopsies. In MCF7 breast cancer cells, HDAC9 decreased ERα mRNA and protein expression and inhibited its transcriptional activity. Conversely, HDAC9 mRNA was strongly overexpressed in OHTam‐resistant MCF7 cells and in ERα‐negative breast tumor cell lines. Moreover, HDAC9‐overexpressing cells were less sensitive to OHTam antiproliferative effects compared with parental MCF7 cells. Several genes (including MUC1, SMC3 and S100P) were similarly deregulated in OHTam‐resistant and in HDAC9‐overexpressing MCF7 cells. Finally, HDAC9 expression was positively associated with genes upregulated in endocrine therapy‐resistant breast cancers and high HDAC9 levels were associated with worse prognosis in patients treated with OHTam. These results demonstrate the complex interactions of class IIa HDAC9 with ERα signaling in breast cancer cells and its effect on the response to hormone therapy.
Collapse
Affiliation(s)
- Aurélien Linares
- IRCM, Institut de Recherche en Cancérologie de Montpellier, France.,INSERM, U1194, Montpellier, France.,Université Montpellier, France.,ICM, Montpellier, France
| | - Said Assou
- Université Montpellier, France.,IRMB, Institute for Regenerative Medicine & Biotherapy, Montpellier, France.,INSERM, U1183, Montpellier, France
| | - Marion Lapierre
- IRCM, Institut de Recherche en Cancérologie de Montpellier, France.,INSERM, U1194, Montpellier, France.,Université Montpellier, France.,ICM, Montpellier, France
| | - Erwan Thouennon
- IRCM, Institut de Recherche en Cancérologie de Montpellier, France.,INSERM, U1194, Montpellier, France.,Université Montpellier, France.,ICM, Montpellier, France
| | - Céline Duraffourd
- Laboratoire de Biopathologie des Tumeurs, CHU Arnaud de Villeneuve, Montpellier, France
| | - Carole Fromaget
- Laboratoire de Biopathologie des Tumeurs, CHU Arnaud de Villeneuve, Montpellier, France
| | - Abdelhay Boulahtouf
- IRCM, Institut de Recherche en Cancérologie de Montpellier, France.,INSERM, U1194, Montpellier, France.,Université Montpellier, France.,ICM, Montpellier, France
| | - Gao Tian
- Key Laboratory of Carcinogenesis and Translational Research Ministry of Education, Department of Gastrointestinal Surgery, Peking University Cancer Hospital & Institute, Beijing, China
| | - Jiafu Ji
- Key Laboratory of Carcinogenesis and Translational Research Ministry of Education, Department of Gastrointestinal Surgery, Peking University Cancer Hospital & Institute, Beijing, China
| | - Ozgur Sahin
- Department of Drug Discovery and Biomedical Sciences, University of South Carolina, Columbia, SC, USA
| | - Eric Badia
- IRCM, Institut de Recherche en Cancérologie de Montpellier, France.,INSERM, U1194, Montpellier, France.,Université Montpellier, France.,ICM, Montpellier, France
| | - Nathalie Boulle
- IRCM, Institut de Recherche en Cancérologie de Montpellier, France.,INSERM, U1194, Montpellier, France.,Université Montpellier, France.,ICM, Montpellier, France.,Laboratoire de Biopathologie des Tumeurs, CHU Arnaud de Villeneuve, Montpellier, France
| | - Vincent Cavaillès
- IRCM, Institut de Recherche en Cancérologie de Montpellier, France.,INSERM, U1194, Montpellier, France.,Université Montpellier, France.,ICM, Montpellier, France
| |
Collapse
|
17
|
Clarke R, Tyson JJ, Tan M, Baumann WT, Jin L, Xuan J, Wang Y. Systems biology: perspectives on multiscale modeling in research on endocrine-related cancers. Endocr Relat Cancer 2019; 26:R345-R368. [PMID: 30965282 PMCID: PMC7045974 DOI: 10.1530/erc-18-0309] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/25/2019] [Accepted: 04/08/2019] [Indexed: 12/12/2022]
Abstract
Drawing on concepts from experimental biology, computer science, informatics, mathematics and statistics, systems biologists integrate data across diverse platforms and scales of time and space to create computational and mathematical models of the integrative, holistic functions of living systems. Endocrine-related cancers are well suited to study from a systems perspective because of the signaling complexities arising from the roles of growth factors, hormones and their receptors as critical regulators of cancer cell biology and from the interactions among cancer cells, normal cells and signaling molecules in the tumor microenvironment. Moreover, growth factors, hormones and their receptors are often effective targets for therapeutic intervention, such as estrogen biosynthesis, estrogen receptors or HER2 in breast cancer and androgen receptors in prostate cancer. Given the complexity underlying the molecular control networks in these cancers, a simple, intuitive understanding of how endocrine-related cancers respond to therapeutic protocols has proved incomplete and unsatisfactory. Systems biology offers an alternative paradigm for understanding these cancers and their treatment. To correctly interpret the results of systems-based studies requires some knowledge of how in silico models are built, and how they are used to describe a system and to predict the effects of perturbations on system function. In this review, we provide a general perspective on the field of cancer systems biology, and we explore some of the advantages, limitations and pitfalls associated with using predictive multiscale modeling to study endocrine-related cancers.
Collapse
Affiliation(s)
- Robert Clarke
- Department of Oncology, Georgetown University Medical Center, Washington, District of Columbia, USA
| | - John J Tyson
- Department of Biological Sciences, Virginia Polytechnic Institute and State University, Blacksburg, Virginia, USA
| | - Ming Tan
- Department of Biostatistics, Bioinformatics & Biomathematics, Georgetown University Medical Center, Washington, District of Columbia, USA
| | - William T Baumann
- Department of Electrical and Computer Engineering, Virginia Polytechnic Institute and State University, Blacksburg, Virginia, USA
| | - Lu Jin
- Department of Oncology, Georgetown University Medical Center, Washington, District of Columbia, USA
| | - Jianhua Xuan
- Department of Electrical and Computer Engineering, Virginia Polytechnic Institute and State University, Arlington, Virginia, USA
| | - Yue Wang
- Department of Electrical and Computer Engineering, Virginia Polytechnic Institute and State University, Arlington, Virginia, USA
| |
Collapse
|
18
|
He X, Zhang S, Chen J, Li D. Increased LGALS3 expression independently predicts shorter overall survival in patients with the proneural subtype of glioblastoma. Cancer Med 2019; 8:2031-2040. [PMID: 30848102 PMCID: PMC6536958 DOI: 10.1002/cam4.2075] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2018] [Revised: 02/05/2019] [Accepted: 02/15/2019] [Indexed: 11/24/2022] Open
Abstract
In the current study, we tried to study the expression of LGALS3 and LGALS3BP, their potential as prognostic markers and the possible genetic/epigenetic mechanisms underlying their dysregulation in different subtypes of glioblastoma (GBM). An in silico retrospective study was performed using large online databases. Results showed that LGALS3 and LGALS3BP were upregulated at both RNA and protein levels in GBM tissue and were generally associated with shorter overall survival (OS) in GBM patients. However, in subgroup analysis, we only found the association in proneural subtype. The copy number alterations did not necessarily lead to LGALS3/LGALS3BP dysregulation. In the proneural subtype of GBM patients, hypermethylation of the two CpG sites (cg19099850 and cg17403875) was associated with significantly lower expression of LGALS3. In univariate and multivariate analysis, LGALS3 expression independently predicted shorter OS in the proneural subtype of GBM (HR: 1.487, 95% CI: 1.229‐1.798, P < 0.001), after adjustment of age, gender, IDH1 mutations, temozolomide chemotherapy, radiotherapy and LGALS3BP expression. In comparison, LGALS3BP lost the prognostic value in multivariate analysis. Based on these findings, we infer that LGALS3 expression serves as an independent biomarker of shorter OS in the proneural subtype of GBM, the expression of which might be regulated in an epigenetic manner.
Collapse
Affiliation(s)
- Xia He
- Department of Integrated Traditional Chinese and Western Medicine, Sichuan Bayi Rehabilitation Center/Sichuan Provincial Rehabilitation Hospital, Chengdu, China
| | - Sunfu Zhang
- Department of Neurosurgery, The First People's Hospital of Yibin, Yibin, China
| | - Junchen Chen
- Department of Neurosurgery, Sichuan Bayi Rehabilitation Center/Sichuan Provincial Rehabilitation Hospital, Chengdu, China
| | - Dekang Li
- Department of Neurosurgery, The First People's Hospital of Yibin, Yibin, China
| |
Collapse
|
19
|
De Oliveira Andrade F, Yu W, Zhang X, Carney E, Hu R, Clarke R, FitzGerald K, Hilakivi-Clarke L. Effects of Jaeumkanghwa-tang on tamoxifen responsiveness in preclinical ER+ breast cancer model. Endocr Relat Cancer 2019; 26:339-353. [PMID: 30640711 PMCID: PMC6365679 DOI: 10.1530/erc-18-0393] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/17/2018] [Accepted: 01/14/2019] [Indexed: 12/14/2022]
Abstract
Resistance to endocrine therapy remains a clinical challenge in the treatment of estrogen receptor-positive (ER+) breast cancer. We investigated if adding a traditional Asian herbal mixture consisting of 12 herbs, called Jaeumkanghwa-tang (JEKHT), to tamoxifen (TAM) therapy might prevent resistance and recurrence in the ER+ breast cancer model of 7,12-dimethylbenz[a]anthracene (DMBA)-exposed Sprague-Dawley rats. Rats were divided into four groups treated as follows: 15 mg/kg TAM administered via diet as TAM citrate (TAM only); 500 mg/kg JEKHT administered via drinking water (JEKHT only group); TAM + JEKHT and no treatment control group. The study was replicated using two different batches of JEKHT. In both studies, a significantly higher proportion of ER+ mammary tumors responded to TAM if animals also were treated with JEKHT (experiment 1: 47% vs 65%, P = 0.015; experiment 2: 43% vs 77%, P < 0.001). The risk of local recurrence also was reduced (31% vs 12%, P = 0.002). JEKHT alone was mostly ineffective. In addition, JEKHT prevented the development of premalignant endometrial lesions in TAM-treated rats (20% in TAM only vs 0% in TAM + JEKHT). Co-treatment of antiestrogen-resistant LCC9 human breast cancer cells with 1.6 mg/mL JEKHT reversed their TAM resistance in dose-response studies in vitro. Several traditional herbal medicine preparations can exhibit anti-inflammatory properties and may increase anti-tumor immune activities in the tumor microenvironment. In the tumors of rats treated with both JEKHT and TAM, expression of Il-6 (P = 0.03), Foxp3/T regulatory cell (Treg) marker (P = 0.033) and Tgfβ1 that activates Tregs (P < 0.001) were significantly downregulated compared with TAM only group. These findings indicate that JEKHT may prevent TAM-induced evasion of tumor immune responses.
Collapse
MESH Headings
- 9,10-Dimethyl-1,2-benzanthracene/toxicity
- Animals
- Antineoplastic Agents, Hormonal/administration & dosage
- Breast Neoplasms/chemically induced
- Breast Neoplasms/drug therapy
- Breast Neoplasms/pathology
- Cell Line, Tumor
- Cell Proliferation/drug effects
- Cytokines/blood
- Drug Resistance, Neoplasm/drug effects
- Endometrium/drug effects
- Endometrium/pathology
- Estrogen Antagonists/administration & dosage
- Female
- Forkhead Transcription Factors/genetics
- Humans
- Mammary Neoplasms, Experimental
- Medicine, East Asian Traditional
- Neoplasm Recurrence, Local/prevention & control
- Plant Extracts/administration & dosage
- Plant Extracts/pharmacology
- Rats
- Rats, Sprague-Dawley
- Receptors, Estrogen/metabolism
- Tamoxifen/administration & dosage
- Transforming Growth Factor beta1/genetics
- Tumor Microenvironment/drug effects
- Tumor Microenvironment/immunology
Collapse
Affiliation(s)
| | - Wei Yu
- Department of Oncology, Georgetown University, Washington, District of Columbia, USA
| | - Xiyuan Zhang
- Department of Oncology, Georgetown University, Washington, District of Columbia, USA
| | - Elissa Carney
- Department of Oncology, Georgetown University, Washington, District of Columbia, USA
| | - Rong Hu
- Department of Oncology, Georgetown University, Washington, District of Columbia, USA
| | - Robert Clarke
- Department of Oncology, Georgetown University, Washington, District of Columbia, USA
| | - Kevin FitzGerald
- Department of Oncology, Georgetown University, Washington, District of Columbia, USA
| | - Leena Hilakivi-Clarke
- Department of Oncology, Georgetown University, Washington, District of Columbia, USA
- Correspondence should be addressed to L Hilakivi-Clarke:
| |
Collapse
|
20
|
Nguyen NM, de Oliveira Andrade F, Jin L, Zhang X, Macon M, Cruz MI, Benitez C, Wehrenberg B, Yin C, Wang X, Xuan J, de Assis S, Hilakivi-Clarke L. Maternal intake of high n-6 polyunsaturated fatty acid diet during pregnancy causes transgenerational increase in mammary cancer risk in mice. Breast Cancer Res 2017; 19:77. [PMID: 28673325 PMCID: PMC5494892 DOI: 10.1186/s13058-017-0866-x] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2016] [Accepted: 06/07/2017] [Indexed: 12/29/2022] Open
Abstract
BACKGROUND Maternal and paternal high-fat (HF) diet intake before and/or during pregnancy increases mammary cancer risk in several preclinical models. We studied if maternal consumption of a HF diet that began at a time when the fetal primordial germ cells travel to the genital ridge and start differentiating into germ cells would result in a transgenerational inheritance of increased mammary cancer risk. METHODS Pregnant C57BL/6NTac mouse dams were fed either a control AIN93G or isocaloric HF diet composed of corn oil high in n-6 polyunsaturated fatty acids between gestational days 10 and 20. Offspring in subsequent F1-F3 generations were fed only the control diet. RESULTS Mammary tumor incidence induced by 7,12-dimethylbenz[a]anthracene was significantly higher in F1 (p < 0.016) and F3 generation offspring of HF diet-fed dams (p < 0.040) than in the control offspring. Further, tumor latency was significantly shorter (p < 0.028) and burden higher (p < 0.027) in F1 generation HF offspring, and similar trends were seen in F3 generation HF offspring. RNA sequencing was done on normal mammary glands to identify signaling differences that may predispose to increased breast cancer risk by maternal HF intake. Analysis revealed 1587 and 4423 differentially expressed genes between HF and control offspring in F1 and F3 generations, respectively, of which 48 genes were similarly altered in both generations. Quantitative real-time polymerase chain reaction analysis validated 13 chosen up- and downregulated genes in F3 HF offspring, but only downregulated genes in F1 HF offspring. Ingenuity Pathway Analysis identified upregulation of Notch signaling as a key alteration in HF offspring. Further, knowledge-fused differential dependency network analysis identified ten node genes that in the HF offspring were uniquely connected to genes linked to increased cancer risk (ANKEF1, IGFBP6, SEMA5B), increased resistance to cancer treatments (SLC26A3), poor prognosis (ID4, JAM3, TBX2), and impaired anticancer immunity (EGR3, ZBP1). CONCLUSIONS We conclude that maternal HF diet intake during pregnancy induces a transgenerational increase in offspring mammary cancer risk in mice. The mechanisms of inheritance in the F3 generation may be different from the F1 generation because significantly more changes were seen in the transcriptome.
Collapse
Affiliation(s)
- Nguyen M Nguyen
- Department of Oncology, Georgetown University, Research Building, Room E407, 3970 Reservoir Road, NW, Washington, DC, 20057, USA
| | - Fabia de Oliveira Andrade
- Department of Oncology, Georgetown University, Research Building, Room E407, 3970 Reservoir Road, NW, Washington, DC, 20057, USA
| | - Lu Jin
- Department of Oncology, Georgetown University, Research Building, Room E407, 3970 Reservoir Road, NW, Washington, DC, 20057, USA
| | - Xiyuan Zhang
- Department of Oncology, Georgetown University, Research Building, Room E407, 3970 Reservoir Road, NW, Washington, DC, 20057, USA
| | - Madisa Macon
- Department of Oncology, Georgetown University, Research Building, Room E407, 3970 Reservoir Road, NW, Washington, DC, 20057, USA
| | - M Idalia Cruz
- Department of Oncology, Georgetown University, Research Building, Room E407, 3970 Reservoir Road, NW, Washington, DC, 20057, USA
| | - Carlos Benitez
- Department of Oncology, Georgetown University, Research Building, Room E407, 3970 Reservoir Road, NW, Washington, DC, 20057, USA
| | - Bryan Wehrenberg
- UTHealth Graduate School of Biomedical Sciences, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Chao Yin
- Department of Oncology, Georgetown University, Research Building, Room E407, 3970 Reservoir Road, NW, Washington, DC, 20057, USA
| | - Xiao Wang
- Department of Electrical and Computer Engineering, Virginia Tech, Arlington, VA, USA
| | - Jianhua Xuan
- Department of Electrical and Computer Engineering, Virginia Tech, Arlington, VA, USA
| | - Sonia de Assis
- Department of Oncology, Georgetown University, Research Building, Room E407, 3970 Reservoir Road, NW, Washington, DC, 20057, USA
| | - Leena Hilakivi-Clarke
- Department of Oncology, Georgetown University, Research Building, Room E407, 3970 Reservoir Road, NW, Washington, DC, 20057, USA.
| |
Collapse
|
21
|
Association of Smoking, Alcohol Use, and Betel Quid Chewing with Epigenetic Aberrations in Cancers. Int J Mol Sci 2017; 18:ijms18061210. [PMID: 28587272 PMCID: PMC5486033 DOI: 10.3390/ijms18061210] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2017] [Revised: 05/26/2017] [Accepted: 06/02/2017] [Indexed: 12/16/2022] Open
Abstract
Numerous environmental factors such as diet, alcohol use, stress, and environmental chemicals are known to elicit epigenetic changes, leading to increased rates of cancers and other diseases. The incidence of head and neck cancer, one of the most common cancers in Taiwanese males, is increasing: oral cancer and nasopharyngeal carcinoma are ranked fourth and tenth respectively, among the top ten cancers in this group, and a major cause of cancer-related deaths in Taiwanese males. Previous studies have identified smoking, alcohol use, and betel quid chewing as the three major causes of head and neck cancers; these three social habits are commonly observed in Taiwanese males, resulting in an increasing morbidity rate of head and neck cancers in this population. In this literature review, we discuss the association between specific components of betel quid, alcohol, and tobacco, and the occurrence of head and neck cancers, lung cancer, gastrointestinal cancers, and urethral cancer. We focus on regulatory mechanisms at the epigenetic level and their oncogenic effects. The review further discusses the application of FDA-approved epigenetic drugs as therapeutic strategies against cancer.
Collapse
|
22
|
Shajahan-Haq AN, Boca SM, Jin L, Bhuvaneshwar K, Gusev Y, Cheema AK, Demas DD, Raghavan KS, Michalek R, Madhavan S, Clarke R. EGR1 regulates cellular metabolism and survival in endocrine resistant breast cancer. Oncotarget 2017; 8:96865-96884. [PMID: 29228577 PMCID: PMC5722529 DOI: 10.18632/oncotarget.18292] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2016] [Accepted: 05/17/2017] [Indexed: 12/12/2022] Open
Abstract
About 70% of all breast cancers are estrogen receptor alpha positive (ER+; ESR1). Many are treated with antiestrogens. Unfortunately, de novo and acquired resistance to antiestrogens is common but the underlying mechanisms remain unclear. Since growth of cancer cells is dependent on adequate energy and metabolites, the metabolomic profile of endocrine resistant breast cancers likely contains features that are deterministic of cell fate. Thus, we integrated data from metabolomic and transcriptomic analyses of ER+ MCF7-derived breast cancer cells that are antiestrogen sensitive (LCC1) or resistant (LCC9) that resulted in a gene-metabolite network associated with EGR1 (early growth response 1). In human ER+ breast tumors treated with endocrine therapy, higher EGR1 expression was associated with a more favorable prognosis. Mechanistic studies showed that knockdown of EGR1 inhibited cell growth in both cells and EGR1 overexpression did not affect antiestrogen sensitivity. Comparing metabolite profiles in LCC9 cells following perturbation of EGR1 showed interruption of lipid metabolism. Tolfenamic acid, an anti-inflammatory drug, decreased EGR1 protein levels and synergized with antiestrogens in inhibiting cell proliferation in LCC9 cells. Collectively, these findings indicate that EGR1 is an important regulator of breast cancer cell metabolism and is a promising target to prevent or reverse endocrine resistance.
Collapse
Affiliation(s)
- Ayesha N Shajahan-Haq
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC, USA
| | - Simina M Boca
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC, USA.,Innovation Center for Biomedical Informatics (ICBI), Georgetown University Medical Center, Washington, DC, USA.,Department of Biostatistics, Bioinformatics and Biomathematics, Georgetown University, Washington, DC, USA
| | - Lu Jin
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC, USA
| | - Krithika Bhuvaneshwar
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC, USA.,Innovation Center for Biomedical Informatics (ICBI), Georgetown University Medical Center, Washington, DC, USA
| | - Yuriy Gusev
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC, USA.,Innovation Center for Biomedical Informatics (ICBI), Georgetown University Medical Center, Washington, DC, USA
| | - Amrita K Cheema
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC, USA
| | - Diane D Demas
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC, USA
| | - Kristopher S Raghavan
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC, USA
| | | | - Subha Madhavan
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC, USA.,Innovation Center for Biomedical Informatics (ICBI), Georgetown University Medical Center, Washington, DC, USA
| | - Robert Clarke
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC, USA
| |
Collapse
|
23
|
Zhang X, Cook KL, Warri A, Cruz IM, Rosim M, Riskin J, Helferich W, Doerge D, Clarke R, Hilakivi-Clarke L. Lifetime Genistein Intake Increases the Response of Mammary Tumors to Tamoxifen in Rats. Clin Cancer Res 2017; 23:814-824. [PMID: 28148690 PMCID: PMC5654585 DOI: 10.1158/1078-0432.ccr-16-1735] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2016] [Revised: 10/07/2016] [Accepted: 10/11/2016] [Indexed: 12/21/2022]
Abstract
PURPOSE Whether it is safe for estrogen receptor-positive (ER+) patients with breast cancer to consume soy isoflavone genistein remains controversial. We compared the effects of genistein intake mimicking either Asian (lifetime) or Caucasian (adulthood) intake patterns to that of starting its intake during tamoxifen therapy using a preclinical model. EXPERIMENTAL DESIGN Female Sprague-Dawley rats were fed an AIN93G diet supplemented with 0 (control diet) or 500 ppm genistein from postnatal day 15 onward (lifetime genistein). Mammary tumors were induced with 7,12-dimethylbenz(a)anthracene (DMBA), after which a group of control diet-fed rats were switched to genistein diet (adult genistein). When the first tumor in a rat reached 1.4 cm in diameter, tamoxifen was added to the diet and a subset of previously only control diet-fed rats also started genistein intake (post-diagnosis genistein). RESULTS Lifetime genistein intake reduced de novo resistance to tamoxifen, compared with post-diagnosis genistein groups. Risk of recurrence was lower both in the lifetime and in the adult genistein groups than in the post-diagnosis genistein group. We observed downregulation of unfolded protein response (UPR) and autophagy-related genes (GRP78, IRE1α, ATF4, and Beclin-1) and genes linked to immunosuppression (TGFβ and Foxp3) and upregulation of cytotoxic T-cell marker CD8a in the tumors of the lifetime genistein group, compared with controls, post-diagnosis, and/or adult genistein groups. CONCLUSIONS Genistein intake mimicking Asian consumption patterns improved response of mammary tumors to tamoxifen therapy, and this effect was linked to reduced activity of UPR and prosurvival autophagy signaling and increased antitumor immunity. Clin Cancer Res; 23(3); 814-24. ©2017 AACR.
Collapse
Affiliation(s)
- Xiyuan Zhang
- Department of Oncology, Georgetown University, Washington, District of Columbia
| | - Katherine L Cook
- Department of Surgical Sciences, Wake Forest University, Winston-Salem, North Carolina
| | - Anni Warri
- Department of Oncology, Georgetown University, Washington, District of Columbia
- Institute of Biomedicine, University of Turku Medical Faculty, Turku, Finland
| | - Idalia M Cruz
- Department of Oncology, Georgetown University, Washington, District of Columbia
| | - Mariana Rosim
- Department of Food and Experimental Nutrition, Faculty of Pharmaceutical Sciences, University of São Paulo, São Paulo, Brazil
| | - Jeffrey Riskin
- Department of Oncology, Georgetown University, Washington, District of Columbia
| | - William Helferich
- Department of Food Science and Human Nutrition, University of Illinois at Urbana-Champaign, Urbana, Illinois
| | - Daniel Doerge
- Division of Biochemical Toxicology, National Center for Toxicological Research, U.S. Food and Drug Administration, Jefferson, Arkansas
| | - Robert Clarke
- Department of Oncology, Georgetown University, Washington, District of Columbia
| | | |
Collapse
|