1
|
Yu H, Wu B, He J, Yi J, Wu W, Wang H, Yang Q, Sun D, Zheng H. Exploring the epigenetic impacts of atrazine in zebrafish: Unveiling mechanisms of neurotoxicity, reproductive toxicity, and implications for human health. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2025; 371:125941. [PMID: 40023241 DOI: 10.1016/j.envpol.2025.125941] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/21/2024] [Revised: 02/07/2025] [Accepted: 02/26/2025] [Indexed: 03/04/2025]
Abstract
Atrazine (ATZ), a widely utilized herbicide, is notable for its long environmental half-life and high solubility, raising significant concerns regarding its ecological and health impacts. While debates continue over its role as an endocrine disruptor, increasing attention has been directed toward its potential epigenetic effects. Utilizing the zebrafish model, a vertebrate with considerable genetic similarity to humans, provides valuable insights into how ATZ exposure may translate into human health risks. This review systematically examines the differential DNA methylation induced by ATZ's non-competitive inhibition of DNA methyltransferases, miRNA dysregulation resulting from mutations in miRNA processing enzymes, and the complex epigenetic interactions affecting histone modifications. Additionally, potential epigenetic biomarkers for ATZ exposure are proposed, which could advance targeted treatment strategies and improve health risk assessments. This synthesis of current understanding identifies knowledge gaps and guides future research towards a more comprehensive understanding of ATZ's epigenetic mechanisms.
Collapse
Affiliation(s)
- Haiyang Yu
- National & Local Joint Engineering Research Center for Ecological Treatment Technology of Urban Water Pollution, College of Life and Environmental Science, Wenzhou University, Wenzhou, 325035, China
| | - Baihui Wu
- Institute of Life Science & Biomedical Collaborative Innovation Center of Zhejiang Province, Wenzhou University, Wenzhou, 325035, China
| | - Jiaxuan He
- Institute of Life Science & Biomedical Collaborative Innovation Center of Zhejiang Province, Wenzhou University, Wenzhou, 325035, China
| | - Jia Yi
- Institute of Life Science & Biomedical Collaborative Innovation Center of Zhejiang Province, Wenzhou University, Wenzhou, 325035, China
| | - Wei Wu
- Bioengineering College of Chongqing University, Chongqing, 400030, China
| | - Hanbing Wang
- Department of Biotechnology, The University of Hong Kong, Hong Kong SAR, 999077, China
| | - Qinsi Yang
- Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, 325000, China
| | - Da Sun
- National & Local Joint Engineering Research Center for Ecological Treatment Technology of Urban Water Pollution, College of Life and Environmental Science, Wenzhou University, Wenzhou, 325035, China; Institute of Life Science & Biomedical Collaborative Innovation Center of Zhejiang Province, Wenzhou University, Wenzhou, 325035, China.
| | - Hongliang Zheng
- Department of Pharmacy, The Quzhou Affiliated Hospital of Wenzhou Medical University, Quzhou People's Hospital, Quzhou, 324000, China.
| |
Collapse
|
2
|
Al Hashami ZS, van der Vegt B, Mourits MJ, Kluiver J, van den Berg A. miRNA-dependent resistance mechanisms to anti-hormonal therapies in estrogen receptor-positive breast cancer patients. MOLECULAR THERAPY. ONCOLOGY 2025; 33:200941. [PMID: 40190354 PMCID: PMC11969448 DOI: 10.1016/j.omton.2025.200941] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 04/09/2025]
Abstract
The estrogen receptor (ERα) is expressed in 70%-80% of breast cancers and is a target of endocrine therapy. However, resistance to endocrine therapy poses a significant clinical challenge. MicroRNAs (miRNAs) have emerged as critical players in oncogenesis and as modulators of therapy response. This review provides an overview of miRNAs that modulate anti-hormonal drug responses. We identified 56 miRNAs associated with resistance to endocrine therapy. These miRNAs had a total of 40 proven target genes that were grouped based on their function under currently known resistance mechanisms, including ER modulation, signaling pathway activation, cell-cycle modulation, and other mechanisms. For a limited number of miRNA-target gene interactions, the relevance of the identified target gene(s) was confirmed by copy or rescue of the miRNA-induced phenotype. Overall, this review highlights critical roles of miRNAs as crucial mediators of resistance to anti-hormonal therapy. The identified miRNA-target gene interactions can serve as a foundation for future functional studies exploring the potential of selected miRNAs in overcoming drug resistance, which might improve outcomes for breast cancer patients.
Collapse
Affiliation(s)
- Zainab Salam Al Hashami
- Department of Pathology and Medical Biology, University of Groningen, Groningen, the Netherlands
- University Medical Centre Groningen, Groningen, the Netherlands
- Department of Biochemistry, College of Medicine and Health Sciences, Sultan Qaboos University, Muscat, Oman
| | - Bert van der Vegt
- Department of Pathology and Medical Biology, University of Groningen, Groningen, the Netherlands
- University Medical Centre Groningen, Groningen, the Netherlands
| | - Marian J.E. Mourits
- Department of Gynaecological Oncology, University of Groningen, University Medical Centre Groningen, Groningen, the Netherlands
| | - Joost Kluiver
- Department of Pathology and Medical Biology, University of Groningen, Groningen, the Netherlands
- University Medical Centre Groningen, Groningen, the Netherlands
| | - Anke van den Berg
- Department of Pathology and Medical Biology, University of Groningen, Groningen, the Netherlands
- University Medical Centre Groningen, Groningen, the Netherlands
| |
Collapse
|
3
|
Vidafar P, Spitschan M. Light on Shedding: A Review of Sex and Menstrual Cycle Differences in the Physiological Effects of Light in Humans. J Biol Rhythms 2023; 38:15-33. [PMID: 36367137 PMCID: PMC9902977 DOI: 10.1177/07487304221126785] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
The human circadian system responds to light as low as 30 photopic lux. Furthermore, recent evidence shows that there are huge individual differences in light sensitivity, which may help to explain why some people are more susceptible to sleep and circadian disruption than others. The biological mechanisms underlying the differences in light sensitivity remain largely unknown. A key variable of interest in understanding these individual differences in light sensitivity is biological sex. It is possible that in humans, males and females differ in their sensitivity to light, but the evidence is inconclusive. This is in part due to the historic exclusion of women in biomedical research. Hormonal fluctuations across the menstrual cycle in women has often been cited as a confound by researchers. Attitudes, however, are changing with funding and publication agencies advocating for more inclusive research frameworks and mandating that women and minorities participate in scientific research studies. In this article, we distill the existing knowledge regarding the relationship between light and the menstrual cycle. There is some evidence of a relationship between light and the menstrual cycle, but the nature of this relationship seems dependent on the timing of the light source (sunlight, moonlight, and electric light at night). Light sensitivity may be influenced by biological sex and menstrual phase but there might not be any effect at all. To better understand the relationship between light, the circadian system, and the menstrual cycle, future research needs to be designed thoughtfully, conducted rigorously, and reported transparently.
Collapse
Affiliation(s)
- Parisa Vidafar
- Turner Institute for Brain and Mental Health, School of Psychological Sciences, Monash University, Melbourne, VIC, Australia
- Translational Sensory and Circadian Neuroscience, Max Planck Institute for Biological Cybernetics, Tübingen, Germany
| | - Manuel Spitschan
- Translational Sensory and Circadian Neuroscience, Max Planck Institute for Biological Cybernetics, Tübingen, Germany
- TUM Department of Sport and Health Sciences, Technical University of Munich, Munich, Germany
- TUM Institute for Advanced Study, Technical University of Munich, Garching, Germany
| |
Collapse
|
4
|
Darwati, Nurlelasari, Mayanti T, Ambardhani N, Kurnia D. Morelloflavone as Potential Anticancer Agent Against MCF-7 Breast
Cancer Cell Lines: In vitro and In silico Studies. LETT DRUG DES DISCOV 2022. [DOI: 10.2174/1570180818666210706110538] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Background:
Breast cancer is most commonly reported to contribute to people's death. Nowadays,
cancer treatment is focused on investigating anticancer drugs from natural compounds. Various
methods, including in vitro, in vivo, and in silico methods, are used to assess the potential of anticancer
compounds. The efficacy of bioactive compounds from medicinal plant origin lies in their affordability
and minimized side effects. The Garcinia genus contains bioactive compounds, such as xanthones, benzophenones,
triterpenes, biflavonoids, and benzoquinones.
Purpose:
The study aimed at investigating an active compound that can inhibit cancer cell growth and
proteins that contribute to cancer cell growth, such as Caspase-9, TNF-α, ER-α, and HER-2.
Methods:
This study is divided into three steps. The first step is the isolation of the active compound from
G. cymosa. The second step is an assessment of cytotoxic activity against MCF-7 cell by using MTT assay,
and the last one is an investigation of the molecular mechanism of an active compound against
Caspase-9, TNF-α, ER-α, and HER-2 by using in silico studies utilizing various programs, such as PyRx
0.8, PYMOL, and Discovery Studio.
Results:
Morelloflavone from G. cymosa stem barks has exhibited anticancer activity (55.84 μg/mL)
eight times lower than doxorubicin (6.99 μg/mL), but it can block the activity of Caspase-9, TNF-α, ER-
α, and HER-2. The binding affinity of morelloflavone is the strongest of all ligands.
Conclusion:
The natural flavonoid, morelloflavone, may be a new lead candidate for anticancer agent
inhibiting action mechanism of Caspase-9, TNF-α, ER-α, and HER-2, respectively.
Collapse
Affiliation(s)
- Darwati
- Department of Chemistry, Faculty of Mathematics and Natural Sciences, Universitas Padjdjaran, Jatinangor 45363,
Jawa Barat, Indonesia
| | - Nurlelasari
- Department of Chemistry, Faculty of Mathematics and Natural Sciences, Universitas Padjdjaran, Jatinangor 45363,
Jawa Barat, Indonesia
| | - Tri Mayanti
- Department of Chemistry, Faculty of Mathematics and Natural Sciences, Universitas Padjdjaran, Jatinangor 45363,
Jawa Barat, Indonesia
| | - Nurul Ambardhani
- Department of Chemistry, Faculty of Mathematics and Natural Sciences, Universitas Padjdjaran, Jatinangor 45363,
Jawa Barat, Indonesia
| | - Dikdik Kurnia
- Department of Chemistry, Faculty of Mathematics and Natural Sciences, Universitas Padjdjaran, Jatinangor 45363,
Jawa Barat, Indonesia
| |
Collapse
|
5
|
In silico molecular docking and dynamic simulation of eugenol compounds against breast cancer. J Mol Model 2021; 28:17. [PMID: 34962586 DOI: 10.1007/s00894-021-05010-w] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2021] [Accepted: 12/14/2021] [Indexed: 10/19/2022]
Abstract
Breast cancer is one of the most severe problems, and it is the primary cause of cancer-related death in females worldwide. The adverse effects and therapeutic resistance development are among the most potent clinical issues for potent medications for breast cancer treatment. The eugenol molecules have a significant affinity for breast cancer receptors. The aim of the study has been on the eugenol compounds, which has potent actions on Erα, PR, EGFR, CDK2, mTOR, ERBB2, c-Src, HSP90, and chemokines receptors inhibition. Initially, the drug-likeness property was examined to evaluate the anti-breast cancer activity by applying Lipinski's rule of five on 120 eugenol molecules. Further, structure-based virtual screening was performed via molecular docking, as protein-like interactions play a vital role in drug development. The 3D structure of the receptors has been acquired from the protein data bank and is docked with 87 3D PubChem and ZINC structures of eugenol compounds, and five FDA-approved anti-cancer drugs using AutoDock Vina. Then, the compounds were subjected to three replica molecular dynamic simulations run of 100 ns per system. The results were evaluated using root mean square deviation (RMSD), root mean square fluctuation (RMSF), and protein-ligand interactions to indicate protein-ligand complex stability. The results confirm that Eugenol cinnamaldehyde has the best docking score for breast cancer, followed by Aspirin eugenol ester and 4-Allyl-2-methoxyphenyl cinnamate. From the results obtained from in silico studies, we propose that the selected eugenols can be further investigated and evaluated for further lead optimization and drug development.
Collapse
|
6
|
Lee HS, Lee IH, Kang K, Park SI, Jung M, Yang SG, Kwon TW, Lee DY. Network Pharmacology-Based Dissection of the Comprehensive Molecular Mechanisms of the Herbal Prescription FDY003 Against Estrogen Receptor-Positive Breast Cancer. Nat Prod Commun 2021. [DOI: 10.1177/1934578x211044377] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Estrogen receptor-positive breast cancer (ERPBC) is the commonest subtype of breast cancer, with a high prevalence, incidence, and mortality. Herbal drugs are increasingly being used to treat ERPBC, although their mechanisms of action are not fully understood. Therefore, in this study, we aimed to analyze the therapeutic properties of FDY003, a herbal anti-ERPBC prescription, using a network pharmacology approach. FDY003 decreased the viability of human ERPBC cells and sensitized them to tamoxifen, an endocrine drug that is widely used in the treatment of ERPBC. The network pharmacology analysis revealed 18 pharmacologically active components in FDY003 that may interact with and regulate 66 therapeutic targets. The enriched gene ontology terms for the FDY003 targets were associated with the modulation of cell survival and death, cell proliferation and growth arrest, and estrogen-associated cellular processes. Analysis of the pathway enrichment of the targets showed that FDY003 may target a variety of ERPBC-associated pathways, including the PIK3-Akt, focal adhesion, MAPK, and estrogen pathways. Overall, these data provide a comprehensive mechanistic insight into the anti-ERPBC activity of FDY003.
Collapse
Affiliation(s)
- Ho-Sung Lee
- The Fore, Seoul, Republic of Korea
- Forest Hospital, Seoul, Republic of Korea
| | | | | | | | - Minho Jung
- Forest Hospital, Seoul, Republic of Korea
| | | | | | - Dae-Yeon Lee
- The Fore, Seoul, Republic of Korea
- Forest Hospital, Seoul, Republic of Korea
| |
Collapse
|
7
|
Yeh SHH, Chang WC, Hsu SM, Lin MH, Chung MC, Ke CS, Lee YC, Hwang CJ, Yang DJ. Chelation-Tamoxifen Conjugates for Imaging of Estrogen Receptors. Cancer Biother Radiopharm 2021; 37:30-40. [PMID: 34491835 DOI: 10.1089/cbr.2021.0169] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Background: The differential diagnosis of estrogen receptor-positive (ER+) pathway-activated systems by using a labeled antiestrogen helps to select the patients for optimal response to endocrine therapy and to discontinue the treatment when resistance occurs. The authors' purpose was to synthesize chelator-tamoxifen conjugates for imaging ER (+) diseases. Materials and Methods: A hydroxypropyl linker was incorporated between either cyclam or cyclam diacetic acid and tamoxifen analog to produce SC-05-L-1 (Z-1-(1,4,8,11-tetraazacyclotetradecan-1-yl)-3-((5-(4-(2-(diethylamino)ethoxy) phenyl) -4,5-diphenylpent-4-en-1-yl)oxy)propan-2-ol) and SC-05-N-1 (Z-2,2'-(4-(3-((5-(4-(2-(diethylamino)ethoxy)phenyl)-4,5-diphenylpent-4-en-1-yl)oxy)-2-hydroxy-propyl) -1,4,8,11-tetraazacyclotetradecane-1,8-diyl)diacetic acid), respectively. In vitro cell uptake and cell/media ratios of 99mTc-SC-05-L-1 and 99mTc- SC-05-N-1 in ER (+) ovarian cancer cells (TOV-112D and OVCAR3) were performed. To ascertain the specificity of cell uptake, the cell uptake was blocked with estrone. In vivo 99mTc-SC-05-L-1 or 99mTc-SC-05-N-1 single-photon emission computed tomography/computed tomography was conducted in tumor-bearing rodents and compared to 18F-fluoro-2-deoxy-d-glucose (18F-FDG) positron emission tomography/magnetic resonance imaging (a reference technology). Results: The radiochemical purities of 99mTc-SC-05-L-1 and 99mTc-SC-05-N-1 were greater than 99% (n = 10). 99mTc-SC-05-L-1 had higher cell/media ratios than 99mTc-SC-05-N-1 in OVCAR-3 ER (+) cells. The cell uptake of 99mTc-SC-05-L-1 was blocked 80% by estrone indicating an ER-mediated process occurred. 99mTc-SC-05-N-1 was further selected for in vivo imaging studies due to higher maximum tolerated dose and superior water solubility than 99mTc-SC-05-L-1. 99mTc-SC-05-N-1 showed higher tumor uptake and tumor/muscle count density ratios than 18F-FDG in tumor-bearing rodents. Conclusion: 99mTc-SC-05-N-1 showed better differential diagnosis of ovarian tumors than 18F-FDG, indicating great promising in chelator-tamoxifen conjugate for ER pathway-directed systems imaging.
Collapse
Affiliation(s)
- Skye Hsin-Hsien Yeh
- Brain Research Center, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | | | - Shu-Meng Hsu
- Institute of Neuroscience, National Yang Ming Chaio Tung University, Taipei, Taiwan
| | - Ming Hsien Lin
- Department of Nuclear Medicine, Camillian Saint Mary's Hospital Luodong, Yilan, Taiwan
| | | | | | | | - Chorng-Jer Hwang
- Management Center, Camillian Saint Mary's Hospital Luodong, Yilan, Taiwan
| | | |
Collapse
|
8
|
Darwati D, Safitri AN, Ambardhani N, Mayanti T, Nurlelasari N, Kurnia D. Effectiveness and Anticancer Activity of a Novel Phenolic Compound from Garcinia porrecta Against the MCF-7 Breast Cancer Cell Line in vitro and in silico. DRUG DESIGN DEVELOPMENT AND THERAPY 2021; 15:3523-3533. [PMID: 34408404 PMCID: PMC8366943 DOI: 10.2147/dddt.s321824] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/09/2021] [Accepted: 07/15/2021] [Indexed: 12/09/2022]
Abstract
Background Cancer is a leading cause of death worldwide, with breast cancer being the most common invasive cancer type in women. Several therapeutic strategies have been explored to reduce the mortality rates of breast cancer. Chemotherapy is the most commonly used systemic treatment, but associated with numerous side-effects. Development of anticancer agents with high efficacy and minimal negative effects is therefore an important focus of research. Natural materials provide an excellent source of bioactive compounds. For instance, Garcinia porrecta from the Clusiaceae family has multiple pharmacological activities, including antioxidant, anti-inflammatory, antibacterial, antiviral, anti-HIV, antidepressant, and anticancer properties. Purpose The main objective of this study was to investigate the potential anticancer effects of compounds extracted from the bark of G. porrecta. Materials and Methods Our experiments were divided into three steps: (1) chromatographic isolation of compounds using various separation techniques, such as extraction, separation and purification, (2) characterization via infrared (IR), nuclear magnetic resonance (NMR) and mass spectroscopy, and (3) evaluation of anticancer activity in vitro (MTT assay) and in silico (via analysis of molecular docking against caspase-9, tumor necrosis factor alpha (TNF-α), estrogen receptor alpha (ER-α), and human epidermal growth factor receptor 2 (HER-2)). Results Depsidone (1) and benzophenone (2) from the ethyl acetate extract of bark of G. porrecta were identified as bioactive components. Examination of the activities of these compounds against MCF-7 cells revealed an IC50 value of 119.3 µg/mL for benzophenone, whereas IC50 for depsidone could not be estimated. Benzophenone activity was lower than that of the positive control doxorubicin (6.9 µg/mL). Depsidone showed the highest binding affinity for HER-2 (−9.2 kcal.mol-1) and benzophenone for ER-α (−8.0 kcal.mol-1). Conclusion Benzophenone displays potency as an anticancer agent through blocking ER-α.
Collapse
Affiliation(s)
- Darwati Darwati
- Department of Chemistry, Faculty of Mathematics and Natural Sciences, Universitas Padjadjaran, Jatinangor, 45363, West Java, Indonesia
| | - Ayu Nadila Safitri
- Department of Chemistry, Faculty of Mathematics and Natural Sciences, Universitas Padjadjaran, Jatinangor, 45363, West Java, Indonesia
| | - Nurul Ambardhani
- Department of Chemistry, Faculty of Mathematics and Natural Sciences, Universitas Padjadjaran, Jatinangor, 45363, West Java, Indonesia
| | - Tri Mayanti
- Department of Chemistry, Faculty of Mathematics and Natural Sciences, Universitas Padjadjaran, Jatinangor, 45363, West Java, Indonesia
| | - Nurlelasari Nurlelasari
- Department of Chemistry, Faculty of Mathematics and Natural Sciences, Universitas Padjadjaran, Jatinangor, 45363, West Java, Indonesia
| | - Dikdik Kurnia
- Department of Chemistry, Faculty of Mathematics and Natural Sciences, Universitas Padjadjaran, Jatinangor, 45363, West Java, Indonesia
| |
Collapse
|
9
|
Wang M, Allen GI. Integrative Generalized Convex Clustering Optimization and Feature Selection for Mixed Multi-View Data. JOURNAL OF MACHINE LEARNING RESEARCH : JMLR 2021; 22:55. [PMID: 34744522 PMCID: PMC8570363] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Figures] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
In mixed multi-view data, multiple sets of diverse features are measured on the same set of samples. By integrating all available data sources, we seek to discover common group structure among the samples that may be hidden in individualistic cluster analyses of a single data view. While several techniques for such integrative clustering have been explored, we propose and develop a convex formalization that enjoys strong empirical performance and inherits the mathematical properties of increasingly popular convex clustering methods. Specifically, our Integrative Generalized Convex Clustering Optimization (iGecco) method employs different convex distances, losses, or divergences for each of the different data views with a joint convex fusion penalty that leads to common groups. Additionally, integrating mixed multi-view data is often challenging when each data source is high-dimensional. To perform feature selection in such scenarios, we develop an adaptive shifted group-lasso penalty that selects features by shrinking them towards their loss-specific centers. Our so-called iGecco+ approach selects features from each data view that are best for determining the groups, often leading to improved integrative clustering. To solve our problem, we develop a new type of generalized multi-block ADMM algorithm using sub-problem approximations that more efficiently fits our model for big data sets. Through a series of numerical experiments and real data examples on text mining and genomics, we show that iGecco+ achieves superior empirical performance for high-dimensional mixed multi-view data.
Collapse
Affiliation(s)
- Minjie Wang
- Department of Statistics, Rice University, Houston, TX 77005, USA
| | - Genevera I Allen
- Departments of Electrical and Computer Engineering, Statistics, and Computer Science, Rice University, Houston, TX 77005, USA; Jan and Dan Duncan Neurological Research Institute, Baylor College of Medicine, Houston, TX 77030, USA
| |
Collapse
|
10
|
Bafna D, Ban F, Rennie PS, Singh K, Cherkasov A. Computer-Aided Ligand Discovery for Estrogen Receptor Alpha. Int J Mol Sci 2020; 21:E4193. [PMID: 32545494 PMCID: PMC7352601 DOI: 10.3390/ijms21124193] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Revised: 05/30/2020] [Accepted: 06/09/2020] [Indexed: 02/08/2023] Open
Abstract
Breast cancer (BCa) is one of the most predominantly diagnosed cancers in women. Notably, 70% of BCa diagnoses are Estrogen Receptor α positive (ERα+) making it a critical therapeutic target. With that, the two subtypes of ER, ERα and ERβ, have contrasting effects on BCa cells. While ERα promotes cancerous activities, ERβ isoform exhibits inhibitory effects on the same. ER-directed small molecule drug discovery for BCa has provided the FDA approved drugs tamoxifen, toremifene, raloxifene and fulvestrant that all bind to the estrogen binding site of the receptor. These ER-directed inhibitors are non-selective in nature and may eventually induce resistance in BCa cells as well as increase the risk of endometrial cancer development. Thus, there is an urgent need to develop novel drugs with alternative ERα targeting mechanisms that can overcome the limitations of conventional anti-ERα therapies. Several functional sites on ERα, such as Activation Function-2 (AF2), DNA binding domain (DBD), and F-domain, have been recently considered as potential targets in the context of drug research and discovery. In this review, we summarize methods of computer-aided drug design (CADD) that have been employed to analyze and explore potential targetable sites on ERα, discuss recent advancement of ERα inhibitor development, and highlight the potential opportunities and challenges of future ERα-directed drug discovery.
Collapse
Affiliation(s)
| | | | | | | | - Artem Cherkasov
- Vancouver Prostate Centre, University of British Columbia, 2660 Oak Street, Vancouver, BC V6H 3Z6, Canada; (D.B.); (F.B.); (P.S.R.); (K.S.)
| |
Collapse
|
11
|
Davila-Avila N, Muñiz-Ruvalcaba FP, Hernandez-Zimbron LF, Gonzalez-Salinas R, Corredor-Ortega C, Perez-Vazquez J, Soberon S, Quiroz-Mercado H. Expression of Fibulin-2 and Fibulin-5 on subretinal fluid in human primary rhegmatogenous retinal detachment. Exp Eye Res 2020; 194:107992. [PMID: 32151522 DOI: 10.1016/j.exer.2020.107992] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2019] [Revised: 02/25/2020] [Accepted: 03/02/2020] [Indexed: 10/24/2022]
Affiliation(s)
- Ned Davila-Avila
- Asociación Para Evitar La Ceguera en México I.A.P, Vicente García Torres 46, Barrio San Lucas, 04030, Coyoacán, Ciudad de México, Mexico
| | - Frida P Muñiz-Ruvalcaba
- Research Department, Asociación Para Evitar La Ceguera en México I.A.P, Vicente García Torres 46, Barrio San Lucas, 04030, Coyoacán, Ciudad de México, Mexico
| | - Luis Fernando Hernandez-Zimbron
- Research Department, Asociación Para Evitar La Ceguera en México I.A.P, Vicente García Torres 46, Barrio San Lucas, 04030, Coyoacán, Ciudad de México, Mexico; Department of Biochemistry, Facultad de Medicina, Universidad Nacional Autónoma de México, 04510, Ciudad Universitaria, Ciudad de México, Mexico.
| | - Roberto Gonzalez-Salinas
- Research Department, Asociación Para Evitar La Ceguera en México I.A.P, Vicente García Torres 46, Barrio San Lucas, 04030, Coyoacán, Ciudad de México, Mexico
| | - Claudia Corredor-Ortega
- Asociación Para Evitar La Ceguera en México I.A.P, Vicente García Torres 46, Barrio San Lucas, 04030, Coyoacán, Ciudad de México, Mexico; Anterior Segment Department, Asociación para Evitar la Ceguera en México, México City, Mexico
| | - Jose Perez-Vazquez
- Asociación Para Evitar La Ceguera en México I.A.P, Vicente García Torres 46, Barrio San Lucas, 04030, Coyoacán, Ciudad de México, Mexico
| | - Santiago Soberon
- Asociación Para Evitar La Ceguera en México I.A.P, Vicente García Torres 46, Barrio San Lucas, 04030, Coyoacán, Ciudad de México, Mexico
| | - Hugo Quiroz-Mercado
- Research Department, Asociación Para Evitar La Ceguera en México I.A.P, Vicente García Torres 46, Barrio San Lucas, 04030, Coyoacán, Ciudad de México, Mexico.
| |
Collapse
|
12
|
Maqbool SN, Lim SC, Park KC, Hanif R, Richardson DR, Jansson PJ, Kovacevic Z. Overcoming tamoxifen resistance in oestrogen receptor-positive breast cancer using the novel thiosemicarbazone anti-cancer agent, DpC. Br J Pharmacol 2020; 177:2365-2380. [PMID: 31975484 DOI: 10.1111/bph.14985] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2019] [Revised: 12/02/2019] [Accepted: 12/22/2019] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND AND PURPOSE Breast cancer is the leading cause of death in women worldwide, with resistance to current therapeutic strategies, including tamoxifen, causing major clinical challenges and leading to more aggressive and metastatic disease. To address this, novel strategies that can inhibit the mechanisms responsible for tamoxifen resistance need to be assessed. EXPERIMENTAL APPROACH We examined the effect of the novel, clinically-trialled, thiosemicarbazone anti-cancer agent, DpC, and its potential as a combination therapy with the clinically used estrogen receptor (ER) antagonist, tamoxifen, using both tamoxifen-resistant and -sensitive, human breast cancer cells (MDA-MB-453, MDA-MB-231 and MCF-7) in 2D and 3D cell-culture. Synergy was assessed using the Chou-Talalay method. The molecular and anti-proliferative effects of these agents and their combination was examined via Western blot, immunofluorescence and colony formation assays. KEY RESULTS Combinations of tamoxifen with DpC were highly synergistic, leading to potent inhibition of cell proliferation, colony formation, and ER-α transcriptional activity. The combination also more efficiently reduced major molecular drivers of proliferation of tamoxifen-resistant cells, including c-Myc, cyclin D1, and p-AKT, while up-regulating the cell cycle inhibitor, p27, and inhibiting oncogenic phosphorylation of ER-α at Ser167. Assessing these effects using 3D cell culture further confirmed the greater effects of DpC combined with tamoxifen in reducing ER-α expression, and that of the proliferation marker, Ki-67, in both tamoxifen-sensitive and -resistant MCF-7 spheroids. CONCLUSIONS AND IMPLICATIONS These studies demonstrate that the synergistic combination of DpC with tamoxifen could be a promising new therapeutic strategy to overcome tamoxifen resistance in ER-positive breast cancer.
Collapse
Affiliation(s)
- Sundus N Maqbool
- Molecular Pathology and Pharmacology Program, Department of Pathology and Bosch Institute, University of Sydney, Sydney, NSW, Australia.,Atta-ur Rahman School of Applied Biosciences, National University of Sciences and Technology, Islamabad, Pakistan
| | - Syer C Lim
- Molecular Pathology and Pharmacology Program, Department of Pathology and Bosch Institute, University of Sydney, Sydney, NSW, Australia
| | - Kyung Chan Park
- Molecular Pathology and Pharmacology Program, Department of Pathology and Bosch Institute, University of Sydney, Sydney, NSW, Australia
| | - Rumeza Hanif
- Atta-ur Rahman School of Applied Biosciences, National University of Sciences and Technology, Islamabad, Pakistan
| | - Des R Richardson
- Molecular Pathology and Pharmacology Program, Department of Pathology and Bosch Institute, University of Sydney, Sydney, NSW, Australia
| | - Patric J Jansson
- Molecular Pathology and Pharmacology Program, Department of Pathology and Bosch Institute, University of Sydney, Sydney, NSW, Australia
| | - Zaklina Kovacevic
- Molecular Pathology and Pharmacology Program, Department of Pathology and Bosch Institute, University of Sydney, Sydney, NSW, Australia
| |
Collapse
|
13
|
Korkmaz G, Manber Z, Lopes R, Prekovic S, Schuurman K, Kim Y, Teunissen H, Flach K, Wit ED, Galli GG, Zwart W, Elkon R, Agami R. A CRISPR-Cas9 screen identifies essential CTCF anchor sites for estrogen receptor-driven breast cancer cell proliferation. Nucleic Acids Res 2019; 47:9557-9572. [PMID: 31372638 PMCID: PMC6765117 DOI: 10.1093/nar/gkz675] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2019] [Revised: 07/11/2019] [Accepted: 07/24/2019] [Indexed: 01/07/2023] Open
Abstract
Estrogen receptor α (ERα) is an enhancer activating transcription factor, a key driver of breast cancer and a main target for cancer therapy. ERα-mediated gene regulation requires proper chromatin-conformation to facilitate interactions between ERα-bound enhancers and their target promoters. A major determinant of chromatin structure is the CCCTC-binding factor (CTCF), that dimerizes and together with cohesin stabilizes chromatin loops and forms the boundaries of topologically associated domains. However, whether CTCF-binding elements (CBEs) are essential for ERα-driven cell proliferation is unknown. To address this question in a global manner, we implemented a CRISPR-based functional genetic screen targeting CBEs located in the vicinity of ERα-bound enhancers. We identified four functional CBEs and demonstrated the role of one of them in inducing chromatin conformation changes in favor of activation of PREX1, a key ERα target gene in breast cancer. Indeed, high PREX1 expression is a bona-fide marker of ERα-dependency in cell lines, and is associated with good outcome after anti-hormonal treatment. Altogether, our data show that distinct CTCF-mediated chromatin structures are required for ERα- driven breast cancer cell proliferation.
Collapse
Affiliation(s)
- Gozde Korkmaz
- Division of Oncogenomics, Oncode Institute, The Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX Amsterdam, The Netherlands
| | - Zohar Manber
- Department of Human Genetics, Sackler School of Medicine, Tel Aviv University, Tel Aviv 69978, Israel
| | - Rui Lopes
- Division of Oncogenomics, Oncode Institute, The Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX Amsterdam, The Netherlands
| | - Stefan Prekovic
- Division of Oncogenomics, Oncode Institute, The Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX Amsterdam, The Netherlands
| | - Karianne Schuurman
- Division of Oncogenomics, Oncode Institute, The Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX Amsterdam, The Netherlands
| | - Yongsoo Kim
- Division of Oncogenomics, Oncode Institute, The Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX Amsterdam, The Netherlands
| | - Hans Teunissen
- Division of Gene Regulation, The Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX Amsterdam, The Netherlands
| | - Koen Flach
- Division of Gene Regulation, The Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX Amsterdam, The Netherlands
| | - Elzo de Wit
- Division of Gene Regulation, The Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX Amsterdam, The Netherlands
| | - Giorgio G Galli
- Disease area Oncology, Novartis Institute for Biomedical Research, CH-4002 Basel, Switzerland
| | - Wilbert Zwart
- Division of Oncogenomics, Oncode Institute, The Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX Amsterdam, The Netherlands.,Laboratory of Chemical Biology and Institute for Complex Molecular Systems, Department of Biomedical Engineering, Eindhoven University of Technology, PO Box 513, 5600MB, Eindhoven, The Netherlands
| | - Ran Elkon
- Department of Human Genetics, Sackler School of Medicine, Tel Aviv University, Tel Aviv 69978, Israel
| | - Reuven Agami
- Division of Oncogenomics, Oncode Institute, The Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX Amsterdam, The Netherlands.,Erasmus MC, Rotterdam University, Doctor Molewaterplein 40, 3015 GD Rotterdam, The Netherlands
| |
Collapse
|
14
|
Chen Y, Yang L, Liu J, Chen Z. Estrogen conjugated fluorescent silica nanoparticles as optical probes for breast cancer cells imaging. BIOMICROFLUIDICS 2019; 13:044113. [PMID: 31531151 PMCID: PMC6735662 DOI: 10.1063/1.5117769] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/01/2019] [Accepted: 08/15/2019] [Indexed: 06/10/2023]
Abstract
Fluorescent nanoparticles are promising tools for living cancer cell imaging and cancer targeting. In this study, estrogen conjugated dye-doped fluorescent nanoparticles (estrogen conjugated FNPs) were synthesized and characterized. The functionalized nanoparticles with low toxicity have shown high selectivity and sensitivity toward target cells. Based on the specific recognition between the estrogen and the estrogen receptor, estrogen conjugated FNPs have been employed as optical probes for specific targeting of estrogen receptor-positive cancer cells with fluorescence microscopy imaging technology. The results demonstrate that the estrogen conjugated FNPs can effectively recognize breast cancer cells with good sensitivity and exceptional photostability, which would offer a novel approach for the diagnosis of breast cancer cells, as well as a new method in detecting estrogen receptors.
Collapse
Affiliation(s)
| | | | - Jing Liu
- Key Laboratory of Combinatorial Biosynthesis and
Drug Discovery (Wuhan University), Ministry of Education, and Wuhan University School of
Pharmaceutical Sciences, Wuhan 430072, China
| | - Zilin Chen
- Key Laboratory of Combinatorial Biosynthesis and
Drug Discovery (Wuhan University), Ministry of Education, and Wuhan University School of
Pharmaceutical Sciences, Wuhan 430072, China
| |
Collapse
|
15
|
Oncogenic Signaling in Tumorigenesis and Applications of siRNA Nanotherapeutics in Breast Cancer. Cancers (Basel) 2019; 11:cancers11050632. [PMID: 31064156 PMCID: PMC6562835 DOI: 10.3390/cancers11050632] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2019] [Revised: 04/06/2019] [Accepted: 04/08/2019] [Indexed: 12/16/2022] Open
Abstract
Overexpression of oncogenes and cross-talks of the oncoproteins-regulated signaling cascades with other intracellular pathways in breast cancer could lead to massive abnormal signaling with the consequence of tumorigenesis. The ability to identify the genes having vital roles in cancer development would give a promising therapeutics strategy in combating the disease. Genetic manipulations through siRNAs targeting the complementary sequence of the oncogenic mRNA in breast cancer is one of the promising approaches that can be harnessed to develop more efficient treatments for breast cancer. In this review, we highlighted the effects of major signaling pathways stimulated by oncogene products on breast tumorigenesis and discussed the potential therapeutic strategies for targeted delivery of siRNAs with nanoparticles in suppressing the stimulated signaling pathways.
Collapse
|
16
|
Curcumae Radix Extract Decreases Mammary Tumor-Derived Lung Metastasis via Suppression of C-C Chemokine Receptor Type 7 Expression. Nutrients 2019; 11:nu11020410. [PMID: 30781353 PMCID: PMC6412318 DOI: 10.3390/nu11020410] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2019] [Revised: 02/11/2019] [Accepted: 02/11/2019] [Indexed: 12/29/2022] Open
Abstract
Curcumae radix is the dry root of Curcuma longa L. (turmeric) that can be used either as a spice or traditional medicine. The aim of this study was to investigate the survival benefits and the anti-metastatic activity of curcumae radix extract (CRE) in MCF7 cells and in MMTV-PyMT transgenic mice—a mouse model of breast cancer metastasis. In vitro wound scratch assay revealed that CRE treatment inhibited cell motility and cell migration in a dose-dependent manner. To investigate the effect of CRE in breast cancer metastasis, MMTV-PyMT transgenic female virgin mice were used and randomly divided into two groups. For survival curve analysis, CRE was administered in a dose of 50 mg/kg to 8–20-week-old mice. Interestingly, CRE treatment significantly increased the median and prolonged survival of MMTV-PyMT mice. Furthermore, CRE treatment decreased tumor burden and inhibited cell proliferation in primary breast tumor, and also suppressed mammary tumor-derived lung metastasis. The size of the lung metastases substantially decreased in the CRE-treated group compared with the ones in the control group. Curcumae radix extract showed anti-metastatic activity through regulating the expression of metastasis markers including C-C Chemokine Receptor Type 7, Matrix Metalloproteinase 9 and the proto-oncogenes c-fos and c-jun. We demonstrated that these metastatic regulators were decreased when CCR7 expression was suppressed in MCF7 cells transfected with CCR7 siRNA. The results of this study show that curcumae radix exerts antitumor and anti-metastatic activities, and we suggest that curcumae radix might be a potential supplement for the treatment and prevention of breast cancer metastasis.
Collapse
|
17
|
Lee SR, Park YJ, Han YB, Lee JC, Lee S, Park HJ, Lee HJ, Kim KH. Isoamericanoic Acid B from Acer tegmentosum as a Potential Phytoestrogen. Nutrients 2018; 10:nu10121915. [PMID: 30518114 PMCID: PMC6315828 DOI: 10.3390/nu10121915] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2018] [Revised: 11/20/2018] [Accepted: 11/28/2018] [Indexed: 12/25/2022] Open
Abstract
Phytoestrogens derived from plants have attracted the attention of the general public and the medical community due to their potentially beneficial role in relieving menopausal symptoms. The deciduous tree Acer tegmentosum Maxim (Aceraceae) has long been utilized in Korean folk medicine to alleviate many physiological disorders, including abscesses, surgical bleeding, and liver diseases. In order to explore structurally and/or biologically new constituents from Korean medicinal plants, a comprehensive phytochemical study was carried out on the bark of A. tegmentosum. One new phenolic compound with a 1,4-benzodioxane scaffold, isoamericanoic acid B (1), as well as with nine known phenolic compounds (2–10), were successfully isolated from the aqueous extracts of the bark of A. tegmentosum. A detailed analysis using 1D and 2D NMR spectroscopy, electronic circular dichroism (ECD) spectral data, and LC/MS afforded the unambiguous structural determination of all isolated compounds, including the new compound 1. In addition, compounds 2, 4, 5, and 9 were isolated and identified from the bark of A. tegmentosum for the first time. All isolated compounds were tested for their estrogenic activities using an MCF-7 BUS cell proliferation assay, which revealed that compounds 1, 2, and 10 showed moderate estrogenic activity. To study the mechanism of this estrogenic effect, a docking simulation of compound 1, which showed the best estrogenic activity, was conducted with estrogen receptor (ER) -α and ER-β, which revealed that it interacts with the key residues of ER-α and ER-β. In addition, compound 1 had slightly higher affinity for ER-β than ER-α in the calculated Gibbs free energy for 1:ER-α and 1:ER-β. Thus, the present experimental evidence demonstrated that active compound 1 from A. tegmentosum could be a promising phytoestrogen for the development of natural estrogen supplements.
Collapse
Affiliation(s)
- Seoung Rak Lee
- School of Pharmacy, Sungkyunkwan University, Suwon 16419, Korea.
| | - Yong Joo Park
- School of Pharmacy, Sungkyunkwan University, Suwon 16419, Korea.
| | - Yu Bin Han
- School of Pharmacy, Sungkyunkwan University, Suwon 16419, Korea.
| | - Joo Chan Lee
- School of Pharmacy, Sungkyunkwan University, Suwon 16419, Korea.
| | - Seulah Lee
- School of Pharmacy, Sungkyunkwan University, Suwon 16419, Korea.
| | - Hyun-Ju Park
- School of Pharmacy, Sungkyunkwan University, Suwon 16419, Korea.
| | - Hae-Jeung Lee
- Department of Food and Nutrition, Gachon University, Seongnam 13120, Korea.
| | - Ki Hyun Kim
- School of Pharmacy, Sungkyunkwan University, Suwon 16419, Korea.
| |
Collapse
|
18
|
Jakubaszek M, Goud B, Ferrari S, Gasser G. Mechanisms of action of Ru(ii) polypyridyl complexes in living cells upon light irradiation. Chem Commun (Camb) 2018; 54:13040-13059. [PMID: 30398487 DOI: 10.1039/c8cc05928d] [Citation(s) in RCA: 74] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The unique photophysical properties of Ru(ii) polypyridyl complexes make them very attractive candidates as photosensitisers in Photodynamic Therapy (PDT). However, to date, there are not many studies exploring in detail the mechanism(s) of action of such compounds in living systems upon light irradiation. This feature article provides an overview of the most in-depth biological studies on such compounds.
Collapse
Affiliation(s)
- Marta Jakubaszek
- Chimie ParisTech, PSL University, Laboratory for Inorganic Chemical Biology, Paris, France.
| | | | | | | |
Collapse
|
19
|
CUEDC1 is a primary target of ERα essential for the growth of breast cancer cells. Cancer Lett 2018; 436:87-95. [DOI: 10.1016/j.canlet.2018.08.018] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2018] [Revised: 08/14/2018] [Accepted: 08/16/2018] [Indexed: 01/31/2023]
|
20
|
Efficient Synthesis of Glutamate Peptide-Estradiol Conjugate for Imaging Estrogen Receptor-Positive Diseases. BIOMED RESEARCH INTERNATIONAL 2018; 2018:5208964. [PMID: 30356372 PMCID: PMC6176321 DOI: 10.1155/2018/5208964] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/14/2018] [Revised: 06/19/2018] [Accepted: 08/16/2018] [Indexed: 11/17/2022]
Abstract
Molecular imaging of estrogen receptor-positive (ER+) pathway-activated system serves the basis of ER+ disease management such as cancers and endometriosis. ER+ patients have better response to endocrine therapy and survive twice as long as negative ER patients. However, tumor resistance resulting from clinical used aromatase inhibitors and antiestrogens is unpredictable. Radiolabeled ER+ ligand could quantify ER+ tissue uptake which helps to stage and restage of the cancer as well as endometriosis. The differential diagnosis of ER+ lesions by using a labeled ligand helps to select the patients for optimal response to endocrine therapy and to discontinue the treatment when resistance occurs. In addition, radiolabeled ER+ ligand serves as basis for image-guided response follow-up. Glutamate receptors are cell surface receptors which are overexpressed in inflammation and infection. Using glutamate peptide as a drug carrier helps to target intracellular genes via glutamate receptor-mediated process. Reports have shown that polyglutamate is a drug carrier that could alter drug solubility and enhance estrogen receptor-ligand binding pocket. However, polyglutamate was a blend of mixed polymer with a wide range of molecular weight. Thus, the structural confirmation and purity of the conjugates were not optimized. To overcome this problem, the efficient synthesis of glutamate peptide-estradiol (GAP-EDL) conjugate was achieved with high purity. EDL was conjugated site-specific at the first glutamate of GAP. The average cell uptake of 68Ga-GAP-EDL was 5-fold higher than the previous reported synthesis. The efficient synthesis of GAP-EDL has greatly enhanced sensitivity and specificity in cell uptake studies. In vivo PET imaging studies indicated that 68Ga-GAP-EDL could image ER (+) tumors in MCF-7 tumor-bearing mice. Therefore, GAP-EDL makes it possible to image ER-enriched endometriosis and cancer.
Collapse
|
21
|
Huq AM, Wai LK, Rullah K, Mohd Aluwi MFF, Stanslas J, Jamal JA. Oestrogenic activity of mimosine on MCF-7 breast cancer cell line through the ERα-mediated pathway. Chem Biol Drug Des 2018; 93:222-231. [PMID: 30251480 DOI: 10.1111/cbdd.13404] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2018] [Revised: 08/13/2018] [Accepted: 09/10/2018] [Indexed: 11/29/2022]
Abstract
Hormone replacement therapy has been a conventional treatment for postmenopausal symptoms in women. However, it has potential risks of breast and endometrial cancers. The aim of this study was to evaluate the oestrogenicity of a plant-based compound, mimosine, in MCF-7 cells by in silico model. Cell viability and proliferation, ERα-SRC1 coactivator activity and expression of specific ERα-dependent marker TFF1 and PGR genes were evaluated. Binding modes of 17β-oestradiol and mimosine at the ERα ligand binding domain were compared using docking and molecular dynamics simulation experiments followed by binding interaction free energy calculation with molecular mechanics/Poisson-Boltzmann surface area. Mimosine showed increased cellular viability (64,450 cells/ml) at 0.1 μM with significant cell proliferation (120.5%) compared to 17β-oestradiol (135.2%). ER antagonist tamoxifen significantly reduced proliferative activity mediated by mimosine (49.9%). Mimosine at 1 μM showed the highest ERα binding activity through increased SRC1 recruitment at 186.9%. It expressed TFF1 (11.1-fold at 0.1 μM) and PGR (13.9-fold at 0.01 μM) genes. ERα-mimosine binding energy was -49.9 kJ/mol, and it interacted with Thr347, Gly521 and His524 of ERα-LBD. The results suggested that mimosine has oestrogenic activity.
Collapse
Affiliation(s)
- Akm Moyeenul Huq
- Drug and Herbal Research Centre, Faculty of Pharmacy, Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia
| | - Lam Kok Wai
- Drug and Herbal Research Centre, Faculty of Pharmacy, Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia
| | - Kamal Rullah
- Drug and Herbal Research Centre, Faculty of Pharmacy, Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia
| | | | - Johnson Stanslas
- Pharmacotherapeutics Unit, Department of Medicine, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Selangor, Malaysia
| | - Jamia Azdina Jamal
- Drug and Herbal Research Centre, Faculty of Pharmacy, Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia
| |
Collapse
|
22
|
Wang J, Zhang X, Ling J, Wang Y, Xu X, Liu Y, Jin C, Ju J, Yuan Y, He F, Zhao C, Wang J, Tian C. KRAB-containing zinc finger protein ZNF496 inhibits breast cancer cell proliferation by selectively repressing ERα activity. BIOCHIMICA ET BIOPHYSICA ACTA. GENE REGULATORY MECHANISMS 2018; 1861:S1874-9399(18)30048-8. [PMID: 30012466 DOI: 10.1016/j.bbagrm.2018.07.003] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/06/2018] [Revised: 06/25/2018] [Accepted: 07/10/2018] [Indexed: 12/20/2022]
Abstract
KRAB-containing zinc finger proteins (KZNF) constitute the largest family of transcriptional regulators in humans and play critical roles in normal development and tumorigenesis. However, the function and mechanism of most KZNFs remain unclear. Here, we report that ZNF496, a KZNF family member, interacts with the DNA binding domain (DBD) of estrogen receptor alpha (ERα) via its C2H2 domain. This interaction decreases ERα binding to chromatin DNA and results in the repression of ERα transactivation, the selective suppression of ERα target genes, and ultimately in a reduction of ERα-positive cell growth in the presence of E2. An analysis of clinical data revealed that the downregulation of ZNF496 expression is observed only in ERα-positive and not in ERα-negative breast cancer tissues when compared with that in matched adjacent tissues. Lastly, we also observed that the downregulation of ZNF496 is associated with poor recurrence-free survival among patients with breast cancer. Collectively, our findings demonstrate that ZNF496 is a novel ERα-binding protein that acts as a target gene-specific ERα corepressor and inhibits the growth of ERα-positive breast cancer cells.
Collapse
Affiliation(s)
- Jinlong Wang
- School of Bioscience and Technology, Weifang Medical University, Weifang, Shandong Province 261053, China; State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing 102206, China; Department of Pathology, The 422th Hospital of PLA, Zhanjiang, Guangdong Province 524000, China
| | - Xiuyuan Zhang
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing 102206, China
| | - Jiming Ling
- School of Bioscience and Technology, Weifang Medical University, Weifang, Shandong Province 261053, China; State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing 102206, China
| | - Yun Wang
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing 102206, China; College of Animal Science, Shandong Agricultural University, Taian, Shandong Province 271018, China
| | - Xiaolin Xu
- School of Bioscience and Technology, Weifang Medical University, Weifang, Shandong Province 261053, China; State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing 102206, China
| | - Yuchen Liu
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing 102206, China
| | - Chaozhi Jin
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing 102206, China
| | - Jiyu Ju
- School of Bioscience and Technology, Weifang Medical University, Weifang, Shandong Province 261053, China
| | - Yanzhi Yuan
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing 102206, China
| | - Fuchu He
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing 102206, China
| | - Chunling Zhao
- School of Bioscience and Technology, Weifang Medical University, Weifang, Shandong Province 261053, China.
| | - Jian Wang
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing 102206, China.
| | - Chunyan Tian
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing 102206, China.
| |
Collapse
|
23
|
Ying J, Wang Q, Xu T, Lyu J. Establishment of a nine-gene prognostic model for predicting overall survival of patients with endometrial carcinoma. Cancer Med 2018; 7:2601-2611. [PMID: 29665298 PMCID: PMC6010780 DOI: 10.1002/cam4.1498] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2018] [Revised: 03/04/2018] [Accepted: 03/21/2018] [Indexed: 12/11/2022] Open
Abstract
Endometrial carcinoma (EC) is the most common malignant tumor of the female genital tract in developed countries. The prognosis of early stage EC is favorable, but a subset faces high risk of cancer progression or recurrence. EC has a poor prognosis upon progression to advanced or metastatic stages. Therefore, our goal is to build a robust prognostic model for predicting overall survival (OS) in EC patients. In this study, 1571 genes were identified as being associated with OS based on genomewide expression profiles using a training dataset. Kyoto Encyclopedia of Genes and Genomes enrichment analysis revealed that these genes were involved in various cancer-related signaling pathways. Nine signature genes were further selected using stepwise selection, and their potential role in the development of EC was demonstrated by performing differential expression analysis between EC and normal uterine tissues. A prognostic model that aggregated these nine signature genes was ultimately established and effectively divided EC patients into two risk groups. OS for patients in the high-risk group was significantly poorer compared with that of the low-risk group. This nine-gene model was subsequently validated and evaluated using the TCGA dataset and shown to have a high discriminating power to distinguish EC patients with an elevated risk of mortality based on the FIGO staging system and other prognostic factors. This study provides a novel prognostic model for the identification of EC patients with elevated risk of mortality and will help to improve our understanding of the underlying mechanisms involved in prognostic EC factors.
Collapse
Affiliation(s)
- Jianchao Ying
- Key Laboratory of Laboratory MedicineMinistry of EducationZhejiang Provincial Key Laboratory of Medical GeneticsSchool of Laboratory Medicine and Life ScienceWenzhou Medical UniversityWenzhouChina
| | - Qian Wang
- Department of Clinical LaboratoryWenzhou People's HospitalThe Third Clinical Institute Affiliated to Wenzhou Medical UniversityWenzhouChina
| | - Teng Xu
- Key Laboratory of Laboratory MedicineMinistry of EducationZhejiang Provincial Key Laboratory of Medical GeneticsSchool of Laboratory Medicine and Life ScienceWenzhou Medical UniversityWenzhouChina
| | - Jianxin Lyu
- Key Laboratory of Laboratory MedicineMinistry of EducationZhejiang Provincial Key Laboratory of Medical GeneticsSchool of Laboratory Medicine and Life ScienceWenzhou Medical UniversityWenzhouChina
| |
Collapse
|
24
|
Estrogenic effects of phytoestrogens derived from Flemingia strobilifera in MCF-7 cells and immature rats. Arch Pharm Res 2018; 41:519-529. [PMID: 29797242 DOI: 10.1007/s12272-018-1027-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2017] [Accepted: 03/22/2018] [Indexed: 01/19/2023]
Abstract
Phytoestrogen (PE) has received considerable attention due to the physiological significance of its estrogenicity. Flemingia strobilifera (FS) has been used as a folk medicine in Asia for the treatment of inflammation, cancer, and infection; however, the estrogenic effects and chemical components of FS have not yet been reported. We aimed to uncover the estrogenic properties and PEs derived from FS using phytochemical and pharmacological evaluation. PEs from FS extract (FSE) were analyzed by NMR, HPLC, and MS. To evaluate estrogenic activity, FSE and its compounds were evaluated by in vitro and in vivo assays, including human estrogen receptor alpha (hERα) binding, estrogen response element (ERE)-luciferase reporter assays, and uterotrophic assays. FSE and its compounds 1-5 showed binding affinities for hERα and activated ERE transcription in MCF-7 cells. Additionally, FSE and compounds 1-5 induced MCF-7 cell proliferation and trefoil factor 1 (pS2) expression. In immature female rats, significant increases in uterine weight and pS2 gene were observed in FSE-treated groups. We identified estrogenic activities of FSE and its bioactive compounds, suggesting their possible roles as PEs via ERs. PEs derived from FSE are promising candidates for ER-targeted therapy for post-menopausal symptoms.
Collapse
|
25
|
A trisubstituted pyrazole derivative reduces DMBA-induced mammary tumor growth in rats by inhibiting estrogen receptor-α expression. Mol Cell Biochem 2018; 449:137-144. [DOI: 10.1007/s11010-018-3350-8] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2017] [Accepted: 04/05/2018] [Indexed: 01/06/2023]
|
26
|
Molecular mechanisms affecting estrogen receptor levels in breast cancer. JOURNAL OF SURGERY AND MEDICINE 2018. [DOI: 10.28982/josam.412314] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
|
27
|
Murata M, Kang JH. Bisphenol A (BPA) and cell signaling pathways. Biotechnol Adv 2018; 36:311-327. [DOI: 10.1016/j.biotechadv.2017.12.002] [Citation(s) in RCA: 227] [Impact Index Per Article: 32.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2017] [Revised: 12/01/2017] [Accepted: 12/07/2017] [Indexed: 01/09/2023]
|
28
|
Phytoestrogenic Activity of Blackcurrant Anthocyanins Is Partially Mediated through Estrogen Receptor Beta. Molecules 2017; 23:molecules23010074. [PMID: 29286333 PMCID: PMC6017224 DOI: 10.3390/molecules23010074] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2017] [Revised: 12/22/2017] [Accepted: 12/27/2017] [Indexed: 01/09/2023] Open
Abstract
Phytoestrogens are plant compounds with estrogenic effects found in many foods. We have previously reported phytoestrogen activity of blackcurrant anthocyanins (cyanidin-3-glucoside, cyanidin-3-rutinoside, delphinidin-3-glucoside, and delphinidin-3-rutinoside) via the estrogen receptor (ER)α. In this study, we investigated the participation of ERβ in the phytoestrogen activity of these anthocyanins. Blackcurrant anthocyanin induced ERβ-mediated transcriptional activity, and the IC50 of ERβ was lower than that of ERα, indicating that blackcurrant anthocyanins have a higher binding affinity to ERβ. In silico docking analysis of cyanidin and delphinidin, the core portions of the compound that fits within the ligand-binding pocket of ERβ, showed that similarly to 17β-estradiol, hydrogen bonds formed with the ERβ residues Glu305, Arg346, and His475. No fitting placement of glucoside or rutinoside sugar chains within the ligand-binding pocket of ERβ-estradiol complex was detected. However, as the conformation of helices 3 and 12 in ERβ varies depending on the ligand, we suggest that the surrounding structure, including these helices, adopts a conformation capable of accommodating glucoside or rutinoside. Comparison of ERα and ERβ docking structures revealed that the selectivity for ERβ is higher than that for ERα, similar to genistein. These results show that blackcurrant anthocyanins exert phytoestrogen activity via ERβ.
Collapse
|
29
|
Velloso FJ, Bianco AFR, Farias JO, Torres NEC, Ferruzo PYM, Anschau V, Jesus-Ferreira HC, Chang THT, Sogayar MC, Zerbini LF, Correa RG. The crossroads of breast cancer progression: insights into the modulation of major signaling pathways. Onco Targets Ther 2017; 10:5491-5524. [PMID: 29200866 PMCID: PMC5701508 DOI: 10.2147/ott.s142154] [Citation(s) in RCA: 53] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Cancer is the disease with highest public health impact in developed countries. Particularly, breast cancer has the highest incidence in women worldwide and the fifth highest mortality in the globe, imposing a significant social and economic burden to society. The disease has a complex heterogeneous etiology, being associated with several risk factors that range from lifestyle to age and family history. Breast cancer is usually classified according to the site of tumor occurrence and gene expression profiling. Although mutations in a few key genes, such as BRCA1 and BRCA2, are associated with high breast cancer risk, the large majority of breast cancer cases are related to mutated genes of low penetrance, which are frequently altered in the whole population. Therefore, understanding the molecular basis of breast cancer, including the several deregulated genes and related pathways linked to this pathology, is essential to ensure advances in early tumor detection and prevention. In this review, we outline key cellular pathways whose deregulation has been associated with breast cancer, leading to alterations in cell proliferation, apoptosis, and the delicate hormonal balance of breast tissue cells. Therefore, here we describe some potential breast cancer-related nodes and signaling concepts linked to the disease, which can be positively translated into novel therapeutic approaches and predictive biomarkers.
Collapse
Affiliation(s)
| | | | | | | | | | - Valesca Anschau
- Department of Genetics and Evolutionary Biology, Institute of Biosciences, University of São Paulo, São Paulo, Brazil
| | | | - Ted Hung-Tse Chang
- Cancer Genomics Group, International Center for Genetic Engineering and Biotechnology (ICGEB), Cape Town, South Africa
| | | | - Luiz F Zerbini
- Cancer Genomics Group, International Center for Genetic Engineering and Biotechnology (ICGEB), Cape Town, South Africa
| | - Ricardo G Correa
- Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, USA
| |
Collapse
|
30
|
Liu CY, Wu CY, Petrossian K, Huang TT, Tseng LM, Chen S. Treatment for the endocrine resistant breast cancer: Current options and future perspectives. J Steroid Biochem Mol Biol 2017; 172:166-175. [PMID: 28684381 DOI: 10.1016/j.jsbmb.2017.07.001] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/07/2017] [Revised: 05/31/2017] [Accepted: 07/01/2017] [Indexed: 02/07/2023]
Abstract
Endocrine resistance remains a challenge and an unmet need for managing hormone receptor-positive breast cancer. The mechanisms of endocrine resistance are multifaceted and are likely to evolve over time following various single or combination therapies. The purpose of this review article is to provide general understanding of molecular basis of endocrine resistance of breast cancer and to offer comprehensive review on current treatment options and potential new treatment strategies for endocrine resistant breast cancers. Last but not the least, we discuss current challenges and future directions for management of endocrine resistant breast cancers.
Collapse
Affiliation(s)
- Chun-Yu Liu
- Division of Medical Oncology, Department of Oncology, Taiwan; School of Medicine, National Yang-Ming University, Taipei, Taiwan
| | - Chia-Yun Wu
- Division of Medical Oncology, Department of Oncology, Taiwan; School of Medicine, National Yang-Ming University, Taipei, Taiwan
| | - Karineh Petrossian
- Department of Cancer Biology, Beckman Research Institute of the City of Hope, United States
| | - Tzu-Ting Huang
- Division of Medical Oncology, Department of Oncology, Taiwan
| | - Ling-Ming Tseng
- Department of Surgery, Taipei Veterans General Hospital, Taipei, Taiwan; School of Medicine, National Yang-Ming University, Taipei, Taiwan
| | - Shiuan Chen
- Department of Cancer Biology, Beckman Research Institute of the City of Hope, United States.
| |
Collapse
|
31
|
Kuruca SE, Karadenizli S, Akgun-Dar K, Kapucu A, Kaptan Z, Uzum G. The effects of 17β-estradiol on blood brain barrier integrity in the absence of the estrogen receptor alpha; an in-vitro model. Acta Histochem 2017; 119:638-647. [PMID: 28803749 DOI: 10.1016/j.acthis.2017.07.005] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2017] [Revised: 07/25/2017] [Accepted: 07/25/2017] [Indexed: 12/17/2022]
Abstract
The blood-brain barrier (BBB), which saves the brain from toxic substances, is formed by endothelial cells. It is mainly composed of tight junction (TJ) proteins existing between endothelial cells. Estrogen is an important regulatory hormone of BBB permeability. It protects the BBB before menopause, but may increase BBB permeability with aging. In addition, nitric oxide modulates BBB permeability. Alcohol impairs the integrity of the BBB with oxidants and inflammatory mediators such as iNOS. We investigated the effects of estrogen on BBB integrity in an in vitro BBB model created with ERα-free HUVEC (human umbilical vein endothelial-like cells) to mimics the menopausal period. In vitro BBB model is created with HUVEC/C6 (rat glioma cells) co-culture. The effect of 17β-estradiol on ethanol-induced BBB disruption and change/or increase of iNOS activity, which modulate BBB integrity, were evaluated. Inducibility and functionality of BBB were investigated using transendothelial electrical resistance (TEER) and the expression of proteins TJ proteins (occludin and claudin-1) and iNOS activity by immunostaining. Our results revealed that 17β-estradiol treatment before and after ethanol decrease expression of occludin and claudin-1 and value of TEER which are BBB disrupt indicators. In addition, ethanol and 17β-estradiol separately and pre- and post-ethanol 17β-estradiol treatment increased iNOS expression. Thus our study suggests caution in the use of 17β-estradiol after menopause because 17β-estradiol at this time may both increase the inflammatory process as well as damage the BBB. We think that beneficial effects of 17β-estradiol may be through ERα but it needs further studies.
Collapse
|
32
|
Frycz BA, Murawa D, Borejsza-Wysocki M, Wichtowski M, Spychała A, Marciniak R, Murawa P, Drews M, Jagodziński PP. mRNA expression of steroidogenic enzymes, steroid hormone receptors and their coregulators in gastric cancer. Oncol Lett 2017; 13:3369-3378. [PMID: 28521442 PMCID: PMC5431337 DOI: 10.3892/ol.2017.5881] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2016] [Accepted: 12/12/2016] [Indexed: 02/07/2023] Open
Abstract
Epidemiological and experimental findings suggest that the development of gastric cancer (GC) is regulated by steroid hormones. In postmenopausal women and older men, the majority of steroid hormones are produced locally in peripheral tissue through the enzymatic conversion of steroid precursors. Therefore, using reverse transcription-quantitative polymerase chain reaction analysis, the mRNA expression of genes encoding steroidogenic enzymes, including steroid sulfatase (STS), hydroxy-delta-5-steroid dehydrogenase 3 beta- and steroid delta-isomerase 1 (HSD3B1), 17β-hydroxysteroid dehydrogenase type 7 and aromatase (CYP19A1), was investigated in primary tumoral and adjacent healthy gastric mucosa from 60 patients with GC. Furthermore, the mRNA levels for estrogen receptor α, estrogen receptor β (ESR2) and androgen receptor (AR), along with their coregulators, including proline, glutamate and leucine rich protein 1, CREB binding protein, nuclear receptor coactivator 1 (NCOA1), nuclear receptor corepressor 1 (NCOR1) and nuclear receptor subfamily 2 group F member 1 (NR2F1), were investigated. Additionally, the association between the mRNA expression of these genes and the clinicopathological features of patients with GC was examined. Significantly decreased levels of STS, HSD3B1, ESR2, AR, NCOA1 and NCOR1 mRNA, in addition to significantly increased levels of CYP19A1 mRNA were demonstrated in tumoral tissue samples compared with adjacent healthy gastric tissue samples. Deregulated expression of these genes in the analyzed tissue samples was associated with certain clinicopathological features of GC, such as age and localization of the tumor. The results of the current study suggest that all of the genes analyzed are expressed in tumoral and adjacent healthy gastric mucosa. In addition, the results indicate that abnormal expression of STS, ESR2, AR, NCOA1 and NCOR1 may serve a role in the development and progression of GC, and may be associated with specific clinicopathological features in patients with GC.
Collapse
Affiliation(s)
- Bartosz Adam Frycz
- Department of Biochemistry and Molecular Biology, Poznań University of Medical Sciences, 60-781 Poznań, Poland
| | - Dawid Murawa
- First Department of Surgical Oncology and General Surgery, Greater Poland Cancer Centre, 61-866 Poznań, Poland.,Research and Development Centre, Regional Specialist Hospital of Wrocław, 51-124 Wrocław, Poland
| | - Maciej Borejsza-Wysocki
- Department of General and Endocrine Surgery and Gastroenterological Oncology, Heliodor Święcicki Clinical Hospital, Poznań University of Medical Sciences, 60-355 Poznań, Poland
| | - Mateusz Wichtowski
- First Department of Surgical Oncology and General Surgery, Greater Poland Cancer Centre, 61-866 Poznań, Poland
| | - Arkadiusz Spychała
- First Department of Surgical Oncology and General Surgery, Greater Poland Cancer Centre, 61-866 Poznań, Poland
| | - Ryszard Marciniak
- Department of General and Endocrine Surgery and Gastroenterological Oncology, Heliodor Święcicki Clinical Hospital, Poznań University of Medical Sciences, 60-355 Poznań, Poland
| | - Paweł Murawa
- First Department of Surgical Oncology and General Surgery, Greater Poland Cancer Centre, 61-866 Poznań, Poland
| | - Michał Drews
- Department of General and Endocrine Surgery and Gastroenterological Oncology, Heliodor Święcicki Clinical Hospital, Poznań University of Medical Sciences, 60-355 Poznań, Poland
| | - Paweł Piotr Jagodziński
- Department of Biochemistry and Molecular Biology, Poznań University of Medical Sciences, 60-781 Poznań, Poland
| |
Collapse
|
33
|
Wu LM, Amidi A. Cognitive impairment following hormone therapy: current opinion of research in breast and prostate cancer patients. Curr Opin Support Palliat Care 2017; 11:38-45. [PMID: 27926544 PMCID: PMC5297865 DOI: 10.1097/spc.0000000000000251] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
PURPOSE OF REVIEW Hormone therapy is a common cancer treatment that may be associated with numerous side and late effects, and in recent years, has been linked to changes in cognition. Here, we present the most important recent findings from empirical studies and reviews that have focused on the effects of hormone therapy on cognitive functioning in breast and prostate cancer populations, underline some general shortcomings, and propose directions for future research. RECENT FINDINGS Recent research indicates that cognitive impairment may occur in breast and prostate cancer patients following onset of hormone therapy. However, because of methodological shortcomings and heterogeneity of current research, conclusions regarding the effects of hormone therapy on cognitive functions remain tentative. SUMMARY The review highlights the general findings while also describing the many methodological shortcomings that need to be addressed in future research. It is clear that larger scale neuropsychological studies that also evaluate the impact of impairments on daily life functioning will improve our understanding of the effects of hormone therapy on cognition and inform the development of appropriate interventions.
Collapse
Affiliation(s)
- Lisa M. Wu
- Department of Medical Social Sciences, Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - Ali Amidi
- Unit for Psycho-oncology and Health Psychology, Department of Oncology, Aarhus University Hospital & Department of Psychology, Aarhus University, Aarhus, Denmark
| |
Collapse
|
34
|
Synthesis and anticancer activity of new azo compounds containing extended π-conjugated systems. CHEMICAL PAPERS 2017. [DOI: 10.1007/s11696-017-0140-9] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
35
|
Physical interaction of estrogen receptor with MnSOD: implication in mitochondrial O 2.- upregulation and mTORC2 potentiation in estrogen-responsive breast cancer cells. Oncogene 2016; 36:1829-1839. [PMID: 27721400 DOI: 10.1038/onc.2016.346] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2015] [Revised: 08/04/2016] [Accepted: 08/10/2016] [Indexed: 12/28/2022]
Abstract
Augmented reactive oxygen species levels consequential to functional alteration of key mitochondrial attributes contribute to carcinogenesis, either directly via oxidative DNA damage infliction or indirectly via activation of oncogenic signaling cascades. We previously reported activation of a key oncogenic signaling cascade via mammalian target of rapamycin (mTOR) signaling complex-2 (mTORC2) owing to estrogen receptor (ER-α)-dependent augmentation of O2.- within the mitochondria of 17-β-estradiol (E2)-stimulated breast cancer cells. Manganese superoxide dismutase (MnSOD) is the principal mitochondrial attribute governing mitochondrial O2.- homeostasis, raising the possibility that its functional alteration could be instrumental in augmenting mitochondrial O2.- levels in breast cancer cells. Here we show ER-dependent transient inhibition of MnSOD catalytic function in breast cancer cells. Catalytic function of MnSOD is tightly regulated at the post-translational level. Post-translational modifications such as phosphorylation, nitration and acetylation represent key regulatory means governing the catalytic function of MnSOD. Acetylation at lysine-68 (K68) inhibits MnSOD catalytic activity and thus represents an important post-translational regulatory mechanism in human cells. Using reciprocal immunoprecipitation and proximity ligation assay, we demonstrate the occurrence of direct physical interaction between ER-α and MnSOD in human breast cancer cells, which in turn was associated with potentiated acetylation of MnSOD at K68. In addition, we also observed diminished interaction of MnSOD with sirtuin-3, the key mitochondrial deacetylase that deacetylates MnSOD at critical K68 and thereby activates it for scavenging O2.-. Consequently, compromised deacetylation of MnSOD at K68 leading to its inhibition and a resultant buildup of O2.- within the mitochondria culminated in the activation of mTORC2. In agreement with this, human breast cancer tissue specimen exhibited a positive correlation between acetyl-MnSODK68 levels and phospho-Ser2481 mTOR levels. In addition to exposing the crosstalk of ER-α with MnSOD post-translational regulatory mechanisms, these data also unravel a regulatory role of ER/MnSOD interaction as an important control switch for redox regulation of ER-α-responsive oncogenic signaling cascades. Furthermore, our study provides a mechanistic link for ER-α-dependent O2.- potentiation and resultant mTORC2 activation in breast cancer cells.
Collapse
|
36
|
Morgan MM, Johnson BP, Livingston MK, Schuler LA, Alarid ET, Sung KE, Beebe DJ. Personalized in vitro cancer models to predict therapeutic response: Challenges and a framework for improvement. Pharmacol Ther 2016; 165:79-92. [PMID: 27218886 PMCID: PMC5439438 DOI: 10.1016/j.pharmthera.2016.05.007] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Personalized cancer therapy focuses on characterizing the relevant phenotypes of the patient, as well as the patient's tumor, to predict the most effective cancer therapy. Historically, these methods have not proven predictive in regards to predicting therapeutic response. Emerging culture platforms are designed to better recapitulate the in vivo environment, thus, there is renewed interest in integrating patient samples into in vitro cancer models to assess therapeutic response. Successful examples of translating in vitro response to clinical relevance are limited due to issues with patient sample acquisition, variability and culture. We will review traditional and emerging in vitro models for personalized medicine, focusing on the technologies, microenvironmental components, and readouts utilized. We will then offer our perspective on how to apply a framework derived from toxicology and ecology towards designing improved personalized in vitro models of cancer. The framework serves as a tool for identifying optimal readouts and culture conditions, thus maximizing the information gained from each patient sample.
Collapse
Affiliation(s)
- Molly M Morgan
- Biomedical Engineering, University of Wisconsin-Madison, Madison, WI, United States
| | - Brian P Johnson
- Biomedical Engineering, University of Wisconsin-Madison, Madison, WI, United States
| | - Megan K Livingston
- Biomedical Engineering, University of Wisconsin-Madison, Madison, WI, United States
| | - Linda A Schuler
- Department of Comparative Biosciences, University of Wisconsin-Madison, Madison, WI, United States
| | - Elaine T Alarid
- Department of Oncology, University of Wisconsin-Madison, Madison, WI, United States
| | - Kyung E Sung
- Biomedical Engineering, University of Wisconsin-Madison, Madison, WI, United States.
| | - David J Beebe
- Biomedical Engineering, University of Wisconsin-Madison, Madison, WI, United States; Department of Oncology, University of Wisconsin-Madison, Madison, WI, United States.
| |
Collapse
|
37
|
Flach KD, Zwart W. The first decade of estrogen receptor cistromics in breast cancer. J Endocrinol 2016; 229:R43-56. [PMID: 26906743 DOI: 10.1530/joe-16-0003] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/11/2016] [Accepted: 02/23/2016] [Indexed: 02/03/2023]
Abstract
The advent of genome-wide transcription factor profiling has revolutionized the field of breast cancer research. Estrogen receptor α (ERα), the major drug target in hormone receptor-positive breast cancer, has been known as a key transcriptional regulator in tumor progression for over 30 years. Even though this function of ERα is heavily exploited and widely accepted as an Achilles heel for hormonal breast cancer, only since the last decade we have been able to understand how this transcription factor is functioning on a genome-wide scale. Initial ChIP-on-chip (chromatin immunoprecipitation coupled with tiling array) analyses have taught us that ERα is an enhancer-associated factor binding to many thousands of sites throughout the human genome and revealed the identity of a number of directly interacting transcription factors that are essential for ERα action. More recently, with the development of massive parallel sequencing technologies and refinements thereof in sample processing, a genome-wide interrogation of ERα has become feasible and affordable with unprecedented data quality and richness. These studies have revealed numerous additional biological insights into ERα behavior in cell lines and especially in clinical specimens. Therefore, what have we actually learned during this first decade of cistromics in breast cancer and where may future developments in the field take us?
Collapse
Affiliation(s)
- Koen D Flach
- Division of Molecular PathologyThe Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Wilbert Zwart
- Division of Molecular PathologyThe Netherlands Cancer Institute, Amsterdam, The Netherlands
| |
Collapse
|
38
|
Chaudhary S, Madhukrishna B, Adhya AK, Keshari S, Mishra SK. Overexpression of caspase 7 is ERα dependent to affect proliferation and cell growth in breast cancer cells by targeting p21(Cip). Oncogenesis 2016; 5:e219. [PMID: 27089142 PMCID: PMC4848833 DOI: 10.1038/oncsis.2016.12] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2015] [Revised: 12/01/2015] [Accepted: 12/17/2015] [Indexed: 12/20/2022] Open
Abstract
Caspase 7 (CASP7) expression has important function during cell cycle progression and cell growth in certain cancer cells and is also involved in the development and differentiation of dental tissues. However, the function of CASP7 in breast cancer cells is unclear. The aim of this study was to analyze the expression of CASP7 in breast carcinoma patients and determine the role of CASP7 in regulating tumorigenicity in breast cancer cells. In this study, we show that the CASP7 expression is high in breast carcinoma tissues compared with normal counterpart. The ectopic expression of CASP7 is significantly associated with ERα expression status and persistently elevated in different stages of the breast tumor grades. High level of CASP7 expression showed better prognosis in breast cancer patients with systemic endocrine therapy as observed from Kaplan–Meier analysis. S3 and S4, estrogen responsive element (ERE) in the CASP7 promoter, is important for estrogen-ERα-mediated CASP7 overexpression. Increased recruitment of p300, acetylated H3 and pol II in the ERE region of CASP7 promoter is observed after hormone stimulation. Ectopic expression of CASP7 in breast cancer cells results in cell growth and proliferation inhibition via p21Cip reduction, whereas small interfering RNA (siRNA) mediated reduction of CASP7 rescued p21Cip levels. We also show that pro- and active forms of CASP7 is located in the nucleus apart from cytoplasmic region of breast cancer cells. The proliferation and growth of breast cancer cells is significantly reduced by broad-spectrum peptide inhibitors and siRNA of CASP7. Taken together, our findings show that CASP7 is aberrantly expressed in breast cancer and contributes to cell growth and proliferation by downregulating p21Cip protein, suggesting that targeting CASP7-positive breast cancer could be one of the potential therapeutic strategies.
Collapse
Affiliation(s)
- S Chaudhary
- Cancer Biology Laboratory, Gene Function and Regulation group, Institute of Life Sciences, Nalco Square, Chandrasekharpur, Bhubaneswar, Odisha, India
| | - B Madhukrishna
- Cancer Biology Laboratory, Gene Function and Regulation group, Institute of Life Sciences, Nalco Square, Chandrasekharpur, Bhubaneswar, Odisha, India
| | - A K Adhya
- Department of Pathology, Kalinga Institute of Medical Sciences, KIIT Rd, Chandaka Industrial Estate, Patia, Bhubaneshwar, Odisha, India
| | - S Keshari
- Cancer Biology Laboratory, Gene Function and Regulation group, Institute of Life Sciences, Nalco Square, Chandrasekharpur, Bhubaneswar, Odisha, India
| | - S K Mishra
- Cancer Biology Laboratory, Gene Function and Regulation group, Institute of Life Sciences, Nalco Square, Chandrasekharpur, Bhubaneswar, Odisha, India
| |
Collapse
|
39
|
A triterpenoid from wild bitter gourd inhibits breast cancer cells. Sci Rep 2016; 6:22419. [PMID: 26926586 PMCID: PMC4772478 DOI: 10.1038/srep22419] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2015] [Accepted: 02/15/2016] [Indexed: 01/06/2023] Open
Abstract
The antitumor activity of 3β,7β,25-trihydroxycucurbita-5,23(E)-dien-19-al (TCD), a triterpenoid isolated from wild bitter gourd, in breast cancer cells was investigated. TCD suppressed the proliferation of MCF-7 and MDA-MB-231 breast cancer cells with IC50 values at 72 h of 19 and 23 μM, respectively, via a PPARγ−independent manner. TCD induced cell apoptosis accompanied with pleiotrophic biological modulations including down-regulation of Akt-NF-κB signaling, up-regulation of p38 mitogen-activated protein kinase and p53, increased reactive oxygen species generation, inhibition of histone deacetylases protein expression, and cytoprotective autophagy. Together, these findings provided the translational value of TCD and wild bitter gourd as an antitumor agent for patients with breast cancer.
Collapse
|
40
|
Guo L, Zhang YU, Yilamu D, Liu S, Guo C. ERβ overexpression results in endocrine therapy resistance and poor prognosis in postmenopausal ERα-positive breast cancer patients. Oncol Lett 2016; 11:1531-1536. [PMID: 26893775 DOI: 10.3892/ol.2016.4095] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2014] [Accepted: 10/28/2015] [Indexed: 12/21/2022] Open
Abstract
The aim of the present study was to investigate the role of estrogen receptor (ER) β in the prognosis of ERα-positive breast cancer in postmenopausal women, and its effect on the efficacy of endocrine therapy. Tissue specimens from 195 patients with postmenopausal breast cancer were analyzed. ERβ expression levels were detected using immunohistochemical staining. Kaplan-Meier analysis was performed to assess patient survival, and the difference in survival was analyzed using the log-rank test. Cox regression was utilized to evaluate prognostic factors. The results revealed that the disease-free survival rate decreased dramatically as ERβ expression levels increased in all postmenopausal ERα-positive breast cancer patients, and ERβ expression was identified to be an indicator of poor prognosis in cases of this disease. Similarly, in postmenopausal ERα-positive breast cancer patients undergoing endocrine therapy, high ERβ expression levels reduced the disease-free survival rate and were correlated with poor patient prognosis. However, in such patients who were not treated with endocrine therapy, disease-free survival rate and prognosis were not significantly affected by ERβ expression. In conclusion, ERβ overexpression led to endocrine therapy resistance and poor prognosis in postmenopausal ERα-positive breast cancer patients, suggesting that ERβ may affect breast cancer prognosis via an increase in endocrine therapy resistance.
Collapse
Affiliation(s)
- Liying Guo
- Department of Breast Cancer, Digestive and Vascular Center, First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang 830054, P.R. China
| | - Y U Zhang
- Department of General Surgery, Yantai Affiliated Hospital of Binzhou Medical University, Yantai, Shandong 264100, P.R. China
| | - Dilimina Yilamu
- Department of Breast Cancer, Digestive and Vascular Center, First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang 830054, P.R. China
| | - Sha Liu
- Department of Breast Cancer, Digestive and Vascular Center, First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang 830054, P.R. China
| | - Chenming Guo
- Department of Breast Cancer, Digestive and Vascular Center, First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang 830054, P.R. China
| |
Collapse
|
41
|
Jeong SY, Ahn HN, Bae GU, Chang M, Liu X, Rhee HK, Lee J, Chin YW, Oh SR, Song YS. Isoguaiacins, Arylnaphthalene Types Identified as Novel Potent Estrogenic Signaling Molecules from Larrea nitida. B KOREAN CHEM SOC 2015. [DOI: 10.1002/bkcs.10437] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Affiliation(s)
- Si-Yeon Jeong
- College of Pharmacy; Sookmyung Women's University; Seoul 140-742 Republic of Korea
| | - Hye-na Ahn
- College of Pharmacy; Sookmyung Women's University; Seoul 140-742 Republic of Korea
| | - Gyu-Un Bae
- College of Pharmacy; Sookmyung Women's University; Seoul 140-742 Republic of Korea
| | - Minsun Chang
- Department of Medical and Pharmaceutical Science, College of Science; Sookmyung Women's University; Seoul 140-742 Republic of Korea
| | - Xiyuan Liu
- Graduate School of Life Systems Sciences; Sookmyung Women's University; Seoul 140-742 Republic of Korea
| | - Hee-Kyung Rhee
- Department of Medical and Pharmaceutical Science, College of Science; Sookmyung Women's University; Seoul 140-742 Republic of Korea
| | - Joongku Lee
- International Biological Material Research Center; KRIBB; Daejeon 305-806 Republic of Korea
| | - Young-Won Chin
- College of Pharmacy; Dongguk University-Seoul; Goyang 410-820 Republic of Korea
| | - Sei-Ryang Oh
- Natural Medicine Research Center; KRIBB; ChungBuk 363-883 Republic of Korea
| | - Yun Seon Song
- College of Pharmacy; Sookmyung Women's University; Seoul 140-742 Republic of Korea
| |
Collapse
|
42
|
Wu VS, Kanaya N, Lo C, Mortimer J, Chen S. From bench to bedside: What do we know about hormone receptor-positive and human epidermal growth factor receptor 2-positive breast cancer? J Steroid Biochem Mol Biol 2015; 153:45-53. [PMID: 25998416 PMCID: PMC4568143 DOI: 10.1016/j.jsbmb.2015.05.005] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/17/2015] [Revised: 05/08/2015] [Accepted: 05/12/2015] [Indexed: 12/11/2022]
Abstract
Breast cancer is a heterogeneous disease. Thanks to extensive efforts from research scientists and clinicians, treatment for breast cancer has advanced into the era of targeted medicine. With the use of several well-established biomarkers, such as hormone receptors (HRs) (i.e., estrogen receptor [ER] and progesterone receptor [PgR]) and human epidermal growth factor receptor-2 (HER2), breast cancer patients can be categorized into multiple subgroups with specific targeted treatment strategies. Although therapeutic strategies for HR-positive (HR+) HER2-negative (HER2-) breast cancer and HR-negative (HR-) HER2-positive (HER2+) breast cancer are well-defined, HR+ HER2+ breast cancer is still an overlooked subgroup without tailored therapeutic options. In this review, we have summarized the molecular characteristics, etiology, preclinical tools and therapeutic options for HR+ HER2+ breast cancer. We hope to raise the attention of both the research and the medical community on HR+ HER2+ breast cancer, and to advance patient care for this subtype of disease.
Collapse
Affiliation(s)
- Victoria Shang Wu
- Department of Cancer Biology, Beckman Research Institute of the City of Hope, Duarte, CA, United States
| | - Noriko Kanaya
- Department of Cancer Biology, Beckman Research Institute of the City of Hope, Duarte, CA, United States
| | - Chiao Lo
- Department of Surgery, National Taiwan University Hospital, Taipei, Taiwan
| | - Joanne Mortimer
- Department of Medical Oncology and Experimental Therapeutics, City of Hope Medical Center Duarte, CA, United States
| | - Shiuan Chen
- Department of Cancer Biology, Beckman Research Institute of the City of Hope, Duarte, CA, United States.
| |
Collapse
|
43
|
Saraiva AL, Payan-Carreira R, Gärtner F, Fortuna da Cunha MR, Rêma A, Faria F, Lourenço LM, Pires MDA. An immunohistochemical study on the expression of sex steroid receptors, Ki-67 and cytokeratins 7 and 20 in feline endometrial adenocarcinomas. BMC Vet Res 2015; 11:204. [PMID: 26268561 PMCID: PMC4535787 DOI: 10.1186/s12917-015-0530-6] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2015] [Accepted: 08/04/2015] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Endometrial adenocarcinomas are a rare type of tumour in cats. Though different morphologies have been reported, the most frequent histological type of feline endometrial adenocarcinoma (FEA) is the papillary serous. Characterization of molecular markers expression in FEA may contribute to clarify the pathogenesis of these tumours and to assess the differences between normal endometrium and FEA regarding the expression pattern of several proteins. Therefore, this study aimed to evaluate the immunohistochemical profile of a wide panel of antibodies (specific for ER-α, PR, Ki-67, CK7 and CK20) in twenty-four cases of FEA. Comparisons were made between FEA and feline normal cyclic endometrium in follicular (n = 13) and luteal (n = 10) stages. Except for Ki-67, all other molecular markers were assessed independently for the intensity of immunolabeling and for the percentage of cells expressing the protein. RESULTS This study showed that in FEA a loss of expression occurs for ER-α (P ≤ 0.0001) and less markedly also for PR. The lost in sex steroid receptors concerns a decrease in both the proportion of labelled cells and the intensity of immunolabelling (P = 0.002 and P = 0.024, respectively). Proliferative activity, estimated via Ki-67 immunoreaction, significantly increased in FEA as compared to normal endometrium (P ≤ 0.0001). Feline endometrial adenocarcinomas maintained the CK7+/CK20+ status of normal endometrium. However, FEA showed decreased CK7 intensity of labelling compared to normal endometria (P ≤ 0.0001) and loss of CK20 expression, both in intensity (P ≤ 0.0001) and in percentage of positive cells (P = 0.01), compared to normal tissues. CONCLUSIONS Data gathered in this study suggest that proliferation in FEA accompanies ER-α down-regulation, possibly following activation of pathways mediated by local growth factors. Moreover, FEA retains combined expression of CK7 and CK20, as evidenced in normal endometrial epithelia, although a decrease in CK7 expression was observed.
Collapse
Affiliation(s)
- Ana Laura Saraiva
- CECAV, Centro de Ciência Animal e Veterinária, Universidade de Trás-os-Montes e Alto Douro, Quinta de Prados, 5000-801, Vila Real, Portugal. .,Escola Universitária Vasco da Gama (EUVG), Avenida José R. Sousa Fernandes, Campus Universitário, Bloco B, Lordemão, 3020-210, Coimbra, Portugal.
| | - Rita Payan-Carreira
- CECAV, Centro de Ciência Animal e Veterinária, Universidade de Trás-os-Montes e Alto Douro, Quinta de Prados, 5000-801, Vila Real, Portugal.
| | - Fátima Gärtner
- Abel Salazar Institute of Biomedical Sciences (ICBAS), University of Porto, Rua de Jorge Viterbo Ferreira n.° 228, 4050-313, Porto, Portugal. .,Institute of Molecular Pathology and Immunology of the University of Porto (IPATIMUP), Rua Júlio Amaral de Carvalho, 45, 4200-135, Porto, Portugal.
| | - Marta R Fortuna da Cunha
- CECAV, Centro de Ciência Animal e Veterinária, Universidade de Trás-os-Montes e Alto Douro, Quinta de Prados, 5000-801, Vila Real, Portugal.
| | - Alexandra Rêma
- Abel Salazar Institute of Biomedical Sciences (ICBAS), University of Porto, Rua de Jorge Viterbo Ferreira n.° 228, 4050-313, Porto, Portugal.
| | - Fátima Faria
- Abel Salazar Institute of Biomedical Sciences (ICBAS), University of Porto, Rua de Jorge Viterbo Ferreira n.° 228, 4050-313, Porto, Portugal.
| | - Lígia M Lourenço
- CECAV, Centro de Ciência Animal e Veterinária, Universidade de Trás-os-Montes e Alto Douro, Quinta de Prados, 5000-801, Vila Real, Portugal.
| | - Maria Dos Anjos Pires
- CECAV, Centro de Ciência Animal e Veterinária, Universidade de Trás-os-Montes e Alto Douro, Quinta de Prados, 5000-801, Vila Real, Portugal.
| |
Collapse
|
44
|
Suganya J, Radha M, Naorem DL, Nishandhini M. In Silico docking studies of selected flavonoids--natural healing agents against breast cancer. Asian Pac J Cancer Prev 2015; 15:8155-9. [PMID: 25338999 DOI: 10.7314/apjcp.2014.15.19.8155] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Breast cancer is the serious health concern in India causing the highest mortality rate in females, which occurs due to uncontrolled cell division and can be metastasize to other parts of the human body. Interactions with estrogen receptor (ER) alpha are mainly responsible for the malignant tumors with regulation of the transcription of various genes as a transcription factor. Most of the drugs currently used for the breast cancer treatment produce various side effects and hence we focused on natural compounds which do not exhibit any toxic effect against normal human cells. MATERIALS AND METHODS Structure of human ER was retrieved from the Protein Data Bank and the structures of flavonoid compounds have been collected from PubChem database. Molecular docking and drug likeness studies were performed for those natural compounds to evaluate and analyze the anti-breast cancer activity. RESULTS Finally two compounds satisfying the Lipinski's rule of five were reported. The two compounds also exhibited highest binding affinity with human ER greater than 10.5 Kcal/mol. CONCLUSIONS The results of this study can be implemented in the drug designing pipeline.
Collapse
Affiliation(s)
- Jeyabaskar Suganya
- Department of Bioinformatics, School of Life Sciences, Vels University, Pallavaram, Chennai, Tamil Nadu, India E-mail : ,
| | | | | | | |
Collapse
|
45
|
Lee CG, Lee HW, Kim BO, Rhee DK, Pyo S. Allicin inhibits invasion and migration of breast cancer cells through the suppression of VCAM-1: Regulation of association between p65 and ER-α. J Funct Foods 2015. [DOI: 10.1016/j.jff.2015.03.017] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
|
46
|
Blackburn HL, Ellsworth DL, Shriver CD, Ellsworth RE. Role of cytochrome P450 genes in breast cancer etiology and treatment: effects on estrogen biosynthesis, metabolism, and response to endocrine therapy. Cancer Causes Control 2015; 26:319-32. [PMID: 25554091 DOI: 10.1007/s10552-014-0519-7] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2014] [Accepted: 12/19/2014] [Indexed: 10/24/2022]
Abstract
PURPOSE The cytochrome P450 (CYP) genes are oxygenases involved in estrogen biosynthesis and metabolism, generation of DNA damaging procarcinogens, and response to anti-estrogen therapies. Since lifetime estrogen exposure is an established risk factor for breast cancer, determining the role of CYP genes in breast cancer etiology may provide critical information for understanding tumorigenesis and response to treatment. METHODS This review summarizes literature available in PubMed published between 1993 and 2013 that focuses on studies evaluating the effects of DNA variants in CYP genes on estrogen synthesis, metabolism, and generation of procarcinogens in addition to response to anti-estrogen therapies. RESULTS Evaluation of DNA variants in estrogen metabolism genes was largely inconclusive. Meta-analyses of data from CYP19A1 support an association between the number of (TTTA) n repeats in intron 4 and breast cancer risk, but the biological mechanism for this relationship is unknown. Associations between single nucleotide polymorphism in CYP1B1 and DNA damage caused by procarcinogenic estrogen metabolites were ambiguous. Variants in CYP2D6 are associated with altered metabolism tamoxifen; however, current data do not support widespread clinical testing. The effect of variants in CYP19A1 in response to aromatase inhibitors is also questionable. CONCLUSION Evaluation of DNA variants in CYP genes involved with estrogen metabolism or treatment response has been inconclusive, reflecting small samples sizes, tumor heterogeneity, and differences between populations. Better-powered studies that account for genetic backgrounds and tumor phenotypes are thus necessary.
Collapse
|
47
|
Brennan M, Lim B. The Actual Role of Receptors as Cancer Markers, Biochemical and Clinical Aspects: Receptors in Breast Cancer. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2015; 867:327-37. [DOI: 10.1007/978-94-017-7215-0_20] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
|
48
|
Rossdeutscher L, Li J, Luco AL, Fadhil I, Ochietti B, Camirand A, Huang DC, Reinhardt TA, Muller W, Kremer R. Chemoprevention activity of 25-hydroxyvitamin D in the MMTV-PyMT mouse model of breast cancer. Cancer Prev Res (Phila) 2014; 8:120-8. [PMID: 25468832 DOI: 10.1158/1940-6207.capr-14-0110] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
Development of oncologic conditions is often accompanied by inadequate vitamin D status. The chemoprevention ability of this molecule is of high interest for breast cancer, the most common malignancy in women worldwide. Because current effective vitamin D analogues, including the naturally occurring active metabolite 1,25-dihydroxycholecalciferol (1,25(OH)2D), frequently cause hypercalcemia at pharmacologic doses, the development of safer molecules for clinical chemopreventive use is essential. This study examines whether exogenously supplied prohormone 25-hydroxycholecalciferol (25(OH)D) can delay tumor progression in vivo without hypercalcemic effects. A low vitamin D diet (25 IU/kg) in the non-immunodeficient MMTV-PyMT mouse model of metastatic breast cancer revealed a significant acceleration of mammary neoplasia compared with normal diet (1,000 IU/kg). Systemic perfusion of MMTV-PyMT mice with 25(OH)D or 1,25(OH)2D delayed tumor appearance and significantly decreased lung metastasis, and both metabolites reduced Ki-67, cyclin D1, and ErbB2 levels in tumors. Perfusion with 25(OH)D caused a 50% raise in tumor 1,25(OH)2D levels, indicating good tumor penetration and effective activation. Importantly, in contrast with 1,25(OH)2D, perfusion with 25(OH)D did not cause hypercalcemia. In vitro treatment of cultured MMTV-PyMT mammary tumor cells with 25(OH)D inhibited proliferation, confirming local activation of the prohormone in this system. This study provides an in vivo demonstration in a non-immunodeficient model of spontaneous breast cancer that exogenous 25(OH)D delays neoplasia, tumor growth, and metastasis, and that its chemoprevention efficacy is not accompanied by hypercalcemia.
Collapse
Affiliation(s)
- Lionel Rossdeutscher
- Department of Medicine, McGill University Health Centre, Montréal, Quebec, Canada
| | - Jiarong Li
- Department of Medicine, McGill University Health Centre, Montréal, Quebec, Canada
| | - Aimée-Lee Luco
- Department of Medicine, McGill University Health Centre, Montréal, Quebec, Canada
| | - Ibtihal Fadhil
- Department of Medicine, McGill University Health Centre, Montréal, Quebec, Canada
| | - Benoit Ochietti
- Department of Medicine, McGill University Health Centre, Montréal, Quebec, Canada
| | - Anne Camirand
- Department of Medicine, McGill University Health Centre, Montréal, Quebec, Canada
| | - Dao Chao Huang
- Department of Medicine, McGill University Health Centre, Montréal, Quebec, Canada
| | - Timothy A Reinhardt
- United States Dept of Agriculture (ARS), National Animal Disease Center, Ames, Iowa
| | - William Muller
- Goodman Cancer Research Centre, McGill University, Montréal, Quebec, Canada
| | - Richard Kremer
- Department of Medicine, McGill University Health Centre, Montréal, Quebec, Canada.
| |
Collapse
|
49
|
van der Born D, Sewing C, Herscheid JKDM, Windhorst AD, Orru RVA, Vugts DJ. A Universal Procedure for the [18F]Trifluoromethylation of Aryl Iodides and Aryl Boronic Acids with Highly Improved Specific Activity. Angew Chem Int Ed Engl 2014. [DOI: 10.1002/ange.201406221] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
|
50
|
van der Born D, Sewing C, Herscheid JKDM, Windhorst AD, Orru RVA, Vugts DJ. A Universal Procedure for the [18F]Trifluoromethylation of Aryl Iodides and Aryl Boronic Acids with Highly Improved Specific Activity. Angew Chem Int Ed Engl 2014; 53:11046-50. [DOI: 10.1002/anie.201406221] [Citation(s) in RCA: 74] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2014] [Indexed: 11/09/2022]
|