1
|
Chuang YT, Yen CY, Liu W, Chien TM, Chang FR, Tsai YH, Tang JY, Chang HW. The protection of bisphenol A-modulated miRNAs and targets by natural products. ENVIRONMENT INTERNATIONAL 2025; 196:109299. [PMID: 39884249 DOI: 10.1016/j.envint.2025.109299] [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: 09/04/2024] [Revised: 11/04/2024] [Accepted: 01/20/2025] [Indexed: 02/01/2025]
Abstract
Bisphenol A (BPA) is a ubiquitous environmental pollutant with endocrine-disrupting functions. Identifying protective drugs and exploring the mechanisms against BPA are crucial in healthcare. Natural products exhibiting antioxidant properties are considered to be able to protect against BPA toxicity. Although BPA-modulated targets and miRNAs have been individually reported, their connections to natural products were rarely organized. With the help of a protein-protein interaction database (STRING), the relationship between individual BPA-modulated targets was interconnected to provide a systemic view. In this review, BPA-downregulated and -upregulated targets are classified, and their interactive network was innovatively analyzed using the bioinformatic database (STRING). BPA-modulated miRNAs were also retrieved and ingeniously connected to BPA-modulated targets. Moreover, a novel connection between BPA-countering natural products was integrated into BPA-modulated miRNAs and targets. All these targets-associated natural products and/or miRNAs were incorporated into the STRING network, providing systemic relationships. Overall, the BPA-modulated target-miRNA-protecting natural product axis was innovatively constructed, providing a straightforward direction for exploring the integrated BPA-countering effects and mechanisms of natural products.
Collapse
Affiliation(s)
- Ya-Ting Chuang
- Department of Biomedical Science and Environmental Biology, PhD Program in Life Sciences, College of Life Science, Kaohsiung Medical University, Kaohsiung 80708, Taiwan.
| | - Ching-Yu Yen
- School of Dentistry, Taipei Medical University, Taipei 11031, Taiwan; Department of Oral and Maxillofacial Surgery, Chi-Mei Medical Center, Tainan 71004, Taiwan.
| | - Wangta Liu
- Department of Biotechnology, Kaohsiung Medical University, Kaohsiung 80708, Taiwan.
| | - Tsu-Ming Chien
- School of Post-Baccalaureate Medicine, Kaohsiung Medical University, Kaohsiung 80708, Taiwan; Department of Urology, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 80708, Taiwan; Department of Urology, Kaohsiung Gangshan Hospital, Kaohsiung Medical University, Kaohsiung 820111, Taiwan.
| | - Fang-Rong Chang
- Graduate Institute of Natural Products, Kaohsiung Medical University, Kaohsiung 80708, Taiwan.
| | - Yi-Hong Tsai
- Department of Pharmacy and Master Program, College of Pharmacy and Health Care, Tajen University, Pingtung 907101, Taiwan.
| | - Jen-Yang Tang
- School of Post-Baccalaureate Medicine, Kaohsiung Medical University, Kaohsiung 80708, Taiwan; Department of Radiation Oncology, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 80708, Taiwan.
| | - Hsueh-Wei Chang
- Department of Biomedical Science and Environmental Biology, PhD Program in Life Sciences, College of Life Science, Kaohsiung Medical University, Kaohsiung 80708, Taiwan; Center for Cancer Research, Kaohsiung Medical University, Kaohsiung 80708, Taiwan; Department of Medical Research, Kaohsiung Medical University Hospital, Kaohsiung 80708, Taiwan.
| |
Collapse
|
2
|
Ma Y, Zhu Y, Wang F, Zhao G, Huang L, Lu R, Wang D, Tian X, Ye Y. 3,3'-Diindolylmethane promotes bone formation - A assessment in MC3T3-E1 cells and zebrafish. Biochem Pharmacol 2024; 230:116618. [PMID: 39528071 DOI: 10.1016/j.bcp.2024.116618] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2024] [Revised: 10/18/2024] [Accepted: 11/07/2024] [Indexed: 11/16/2024]
Abstract
Osteoporosis is a common degenerative bone disease in middle-aged and elderly people. The current drugs used to treat osteoporosis have many side effects and low patient compliance. Phytochemotherapy may be safer and more effective. 3,3'-diindolemethane (DIM) is the digestive product of indole-3-methanol in cruciferous vegetables in the stomach, which is a kind of anti-tumor and anti-oxidation phytochemical. However, the effects of DIM on osteoblasts and the mechanism by which DIM regulates bone formation are not fully understood. The aim of this study was to investigate the effects of DIM on the bone formation of mouse preosteoblasts MC3T3-E1 and zebrafish. DIM promotes proliferation and osteogenic differentiation of MC3T3-E1 cells in vitro, and also plays a bone promoting role by increasing the interaction between BRCA1-Associated Protein 1(BAP1) and Inositol 1,4,5-Trisphosphate Receptor(IP3R), up-regulating the expression of BAP1 and IP3R and downstream storage operation calcium entry (SOCE) related protein Recombinant Stromal Interaction Molecule 1(STIM1). The effect of DIM on osteoporosis was confirmed in zebrafish osteoporosis model, and its molecular mechanism may be related to BAP1/IP3R/SOCE signaling pathway. These findings highlight the potential therapeutic value of DIM in the prevention and treatment of osteoporosis.
Collapse
Affiliation(s)
- Ying Ma
- Department of Preventive Medicine and Public Health Laboratory Science, School of Medicine, Jiangsu University, Zhenjiang, China
| | - Yin Zhu
- Department of Oncology, Jurong Hospital Affiliated to Jiangsu University, Zhenjiang, China
| | - Feng Wang
- Department of Spine Surgery, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, China
| | - Guoyang Zhao
- Orthopedics Department, Affiliated Hospital of Jiangsu University, Zhenjiang, China
| | - Lianlian Huang
- Department of Preventive Medicine and Public Health Laboratory Science, School of Medicine, Jiangsu University, Zhenjiang, China
| | - Rongzhu Lu
- Department of Preventive Medicine and Public Health Laboratory Science, School of Medicine, Jiangsu University, Zhenjiang, China; Center for Experimental Research, Affiliated Kunshan Hospital to Jiangsu University, Suzhou, China
| | - Dongxu Wang
- School of Grain Science and Technology, Jiangsu University of Science and Technology, Zhenjiang, China
| | - Xinyu Tian
- Department of Clinical Laboratory, Nanjing Drum Tower Hospital, Nanjing Drum Tower Hospital Clinical College, Jiangsu University, Nanjing, China
| | - Yang Ye
- Department of Preventive Medicine and Public Health Laboratory Science, School of Medicine, Jiangsu University, Zhenjiang, China.
| |
Collapse
|
3
|
Tian C, Deng S, Yang M, Bai B, Pan Y, Xie G, Zhao D, Wei L. Indole-3-carbinol and its main derivative 3,3'-diindolylmethane: Regulatory roles and therapeutic potential in liver diseases. Biomed Pharmacother 2024; 180:117525. [PMID: 39388997 DOI: 10.1016/j.biopha.2024.117525] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2024] [Revised: 10/01/2024] [Accepted: 10/04/2024] [Indexed: 10/12/2024] Open
Abstract
Indole-3-carbinol (I3C), a compound found in cruciferous vegetables, has shown significant efficacy in treating both cancerous and non-cancerous diseases. Its primary derivative, 3,3'-diindolylmethane (DIM), formed during digestion, also exhibits similar therapeutic benefits. In liver disorders, I3C and DIM exhibit dual roles by inhibiting and promoting hepatocellular carcinoma (HCC) and providing relief for nonmalignant liver diseases, such as acute liver injury (ALI), hepatic fibrosis, nonalcoholic fatty liver disease (NAFLD), and alcohol-related liver disease (ALD). Mechanistically, I3C and DIM modulate various pathophysiological processes, including cell proliferation, apoptosis, oxidative stress, and lipogenesis. This review aims to enhance researchers' understanding of the regulatory roles of I3C and DIM in these liver diseases and explore the potential of plant-derived substances in liver disease treatment.
Collapse
Affiliation(s)
- Chao Tian
- Hepatopancreatobiliary Center, Ministry of Education Key Laboratory of Digital Intelligence Hepatology, Beijing Tsinghua Changgung Hospital, School of Clinical Medicine, Tsinghua Medicine of Tsinghua University, Beijing 102218, China
| | - Shizhou Deng
- Hepatopancreatobiliary Center, Ministry of Education Key Laboratory of Digital Intelligence Hepatology, Beijing Tsinghua Changgung Hospital, School of Clinical Medicine, Tsinghua Medicine of Tsinghua University, Beijing 102218, China; Research and Development Department, Guangdong Longsee Biomedical Corporation, Guangzhou 510700, China
| | - Ming Yang
- Hepatopancreatobiliary Center, Ministry of Education Key Laboratory of Digital Intelligence Hepatology, Beijing Tsinghua Changgung Hospital, School of Clinical Medicine, Tsinghua Medicine of Tsinghua University, Beijing 102218, China
| | - Baochen Bai
- Department of Cardiology, Peking University People's hospital, Beijing 100044, China
| | - Yi Pan
- Hepatopancreatobiliary Center, Ministry of Education Key Laboratory of Digital Intelligence Hepatology, Beijing Tsinghua Changgung Hospital, School of Clinical Medicine, Tsinghua Medicine of Tsinghua University, Beijing 102218, China
| | - Gangqiao Xie
- Hepatopancreatobiliary Center, Ministry of Education Key Laboratory of Digital Intelligence Hepatology, Beijing Tsinghua Changgung Hospital, School of Clinical Medicine, Tsinghua Medicine of Tsinghua University, Beijing 102218, China; Department of Biomedical Engineering, School of Medicine, Tsinghua University, Beijing 100084, China
| | - Dongliang Zhao
- Hepatopancreatobiliary Center, Ministry of Education Key Laboratory of Digital Intelligence Hepatology, Beijing Tsinghua Changgung Hospital, School of Clinical Medicine, Tsinghua Medicine of Tsinghua University, Beijing 102218, China
| | - Lai Wei
- Hepatopancreatobiliary Center, Ministry of Education Key Laboratory of Digital Intelligence Hepatology, Beijing Tsinghua Changgung Hospital, School of Clinical Medicine, Tsinghua Medicine of Tsinghua University, Beijing 102218, China.
| |
Collapse
|
4
|
Franko N, Kodila A, Sollner Dolenc M. Adverse outcomes of the newly emerging bisphenol A substitutes. CHEMOSPHERE 2024; 364:143147. [PMID: 39168390 DOI: 10.1016/j.chemosphere.2024.143147] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/02/2024] [Revised: 08/07/2024] [Accepted: 08/19/2024] [Indexed: 08/23/2024]
Abstract
BPA and its analogues are facing increasingly stringent regulations restricting their use due to the increasing knowledge of their harmful effects. It is therefore expected that novel BPA analogues and alternatives will replace them in plastic products, cans and thermal paper to circumvent restrictions imposed by legislation. This raises concerns about the safety of "BPA-free" products, as they contain BPA substitutes whose safety has not been sufficiently assessed prior to their market introduction. The regulatory agencies have recognised BPAP, BPBP, BPC2, BPE, BPFL, BPG, BPP, BPPH, BPS-MAE, BPS-MPE, BP-TMC, BPZ and the alternatives BTUM, D-90, UU and PF201 as compound with insufficient data regarding their safety. We demonstrate that the mentioned compounds are present in consumer products, food and the environment, thus exhibiting toxicological risk not only to humans, but also to other species where their toxic effects have already been described. Results of in silico, in vitro and in vivo studies examining the endocrine disruption and other effects of BPA analogues show that they disrupt the endocrine system by targeting various nuclear receptors, impairing reproductive function and causing toxic effects such as hepatotoxicity, altered behaviour and impaired reproductive function. In vitro and in vivo data on BPA alternatives are literally non-existent, although these compounds are already present in commonly used thermal papers. However, in silico studies predicted that they might cause adverse effects as well. The aim of this article is to comprehensively collate the information on selected BPA substitutes to illustrate their potential toxicity and identify safety gaps.
Collapse
Affiliation(s)
- Nina Franko
- University of Ljubljana, Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Aškerčeva cesta 7, 1000 Ljubljana, Slovenia.
| | - Anja Kodila
- University of Ljubljana, Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Aškerčeva cesta 7, 1000 Ljubljana, Slovenia.
| | - Marija Sollner Dolenc
- University of Ljubljana, Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Aškerčeva cesta 7, 1000 Ljubljana, Slovenia.
| |
Collapse
|
5
|
Xiong C, Tang Y, Li F, Ye Y, Li X, Lin J, Dai S. 3,3'-Diindolylmethane inhibits the proliferation of esophageal squamous cell carcinoma cells via downregulation of STIM1. Oncol Lett 2024; 28:339. [PMID: 38855503 PMCID: PMC11157662 DOI: 10.3892/ol.2024.14473] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2024] [Accepted: 04/15/2024] [Indexed: 06/11/2024] Open
Abstract
3,3'-Diindolylmethane (DIM) is a natural phytochemical derived from cruciferous plants that has inhibitory effects on a wide range of tumor cells; however, its relevant effects on esophageal cancer cells have been poorly studied. Therefore, in the present study, a pharmacology network approach was used to predict the possible core targets of DIM acting on esophageal cancer. Subsequently, using in vitro experiments, TE-1 human esophageal cancer cells were treated with different concentrations of DIM (0, 40, 60 and 80 µM) for 24 h. Changes in cell activity were detected by Cell Counting Kit-8 assay, and changes in the expression levels of stromal interaction molecule 1 (STIM1) and apoptosis-related proteins, B-cell lymphoma-2 (Bcl-2) and Bax, were assessed by western blotting, followed by the upregulation of STIM1 by thapsigargin (Tg). Network pharmacology analysis showed that there were 39 potential core targets of DIM in esophageal cancer. The results of the in vitro experiments showed that DIM could inhibit the viability of esophageal cancer cells, downregulate the expression of STIM1 and Bcl-2 proteins and upregulate the expression of Bax protein, all in a concentration-dependent manner. The results also demonstrated that toxic carotenoids were agonist against STIM1 protein and upregulated STIM1 and Bax protein expression. After agonizing STIM1 protein expression using Tg, DIM was able to counteract the expression trend of STIM1, Bcl-2 and Bax protein in TE-1 cells. In summary, DIM induced apoptosis and inhibited the viability of esophageal cancer cells by downregulating the expression of STIM1 protein; therefore, the natural phytochemical, DIM, may be a potential substance for the early prevention and treatment of esophageal cancer cells.
Collapse
Affiliation(s)
- Chenyi Xiong
- Department of Thoracic Surgery, Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu 212000, P.R. China
| | - Yining Tang
- Department of Thoracic Surgery, Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu 212000, P.R. China
| | - Feng Li
- Department of Thoracic Surgery, Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu 212000, P.R. China
| | - Yang Ye
- Department of Preventive Medicine and Public Health Laboratory Science, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu 212013, P.R. China
| | - Xiaoran Li
- Department of Thoracic Surgery, Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu 212000, P.R. China
| | - Jinxing Lin
- Department of Thoracic Surgery, Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu 212000, P.R. China
| | - Sunxian Dai
- Cell Medicine Laboratory, School of Medicine, Soochow University, Soochow, Jiangsu 215000, P.R. China
| |
Collapse
|
6
|
Yu J, Zhang Y, Yao H, Zhang Z, Yang X, Zhu W, Xu J. ERβ activation improves nonylphenol-induced depression and neurotransmitter secretion disruption via the TPH2/5-HT pathway. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2024; 280:116521. [PMID: 38850708 DOI: 10.1016/j.ecoenv.2024.116521] [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: 02/10/2024] [Revised: 05/24/2024] [Accepted: 05/27/2024] [Indexed: 06/10/2024]
Abstract
The aim of this study is to investigate the role of estrogen receptor β (ERβ) in nonylphenol (NP) - induced depression - like behavior in rats and its impact on the regulation of the TPH2/5-HT pathway. In the in vitro experiment, rat basophilic leukaemia cells (RBL-2H3) cells were divided into the four groups: blank group, NP group (20 μM), ERβ agonist group (0.01 μM), and NP+ERβ agonist group (20 μM+0.01 μM). For the in vivo experiment, 72 adult male Sprague-Dawley rats were randomly divided into following six groups: the Control, NP (40 mg/kg) group, ERβ agonist (2 mg/kg, Diarylpropionitrile (DPN)) group, ERβ inhibitor (0.1 mg/kg, 4-(2-phenyl-5,7-bis(trifluoromethyl)pyrazolo[1,5-a]pyrimidin-3-yl) phenol (PHTPP)) group, NP+ERβ agonist (40 mg/kg NP + 2 mg/kg DPN) group, and NP+ERβ inhibitor (40 mg/kg NP + 0.1 mg/kg PHTPP) group, with 12 rats in each group. Each rat in drug group were given NP by gavage and/or received a single intraperitoneal injection of DPN 2 mg/kg or PHTPP 0.1 mg/kg. Both in vivo and in vitro, NP group showed a decrease in the expression levels of ERβ, tryptophan hydroxylase (TPH1), and tryptophan hydroxylase-2 (TPH2) genes and proteins, and reduced levels of DA, NE, and 5-hydroxytryptophan (5-HT) neurotransmitters. RBL-2H3 cells showed signs of cell shrinkage, with rounded cells, increased suspension and more loosely arranged cells. The effectiveness of the ERβ agonist stimulation exhibited an increase exceeding 60% in RBL-2H3 cells. The application of ERβ agonist resulted in an alleviation the aforementioned alterations. ERβ agonist activated the TPH2/5-HT signaling pathways. Compared to the control group, the NP content in the brain tissue of the NP group was significantly increased. The latency to eat for the rats was longer and the amount of food consumed was lower, and the rats had prolonged immobility time in the behavioral experiment of rats. The expression levels of ERβ, TPH1, TPH2, 5-HT and 5-HITT proteins were decreased in the NP group, suggesting NP-induced depression-like behaviours as well as disturbances in the secretion of serum hormones and monoamine neurotransmitters. In the NP group, the midline raphe nucleus showed an elongated nucleus with a dark purplish-blue colour, nuclear atrophy, displacement and pale cytoplasm. ERβ might ameliorate NP-induced depression-like behaviors, and secretion disorders of serum hormones and monoamine neurotransmitters via activating TPH2/5-HT signaling pathways.
Collapse
Affiliation(s)
- Jie Yu
- School of Public Health, Zunyi Medical University, Zunyi, Guizhou 563000, PR China
| | - Yujie Zhang
- School of Public Health, Zunyi Medical University, Zunyi, Guizhou 563000, PR China
| | - Hao Yao
- School of Public Health, Zunyi Medical University, Zunyi, Guizhou 563000, PR China
| | - Ziping Zhang
- Department of Clinical Laboratory, Zunyi Medical and Pharmaceutical College, Zunyi 563006, PR China
| | - Xiao Yang
- School of Public Health, Zunyi Medical University, Zunyi, Guizhou 563000, PR China
| | - Wei Zhu
- School of Public Health, Zunyi Medical University, Zunyi, Guizhou 563000, PR China
| | - Jie Xu
- School of Public Health, Zunyi Medical University, Zunyi, Guizhou 563000, PR China.
| |
Collapse
|
7
|
He TT, Li X, Ma JZ, Yang Y, Zhu S, Zeng J, Luo L, Yin YL, Cao LY. Triclocarban and triclosan promote breast cancer progression in vitro and in vivo via activating G protein-coupled estrogen receptor signaling pathways. THE SCIENCE OF THE TOTAL ENVIRONMENT 2024; 931:172782. [PMID: 38679099 DOI: 10.1016/j.scitotenv.2024.172782] [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: 01/17/2024] [Revised: 04/21/2024] [Accepted: 04/24/2024] [Indexed: 05/01/2024]
Abstract
Triclocarban (TCC) and triclosan (TCS) have been detected ubiquitously in human body and evoked increasing concerns. This study aimed to reveal the induction risks of TCC and TCS on triple negative breast cancer through non-genomic GPER-mediated signaling pathways. Molecular simulation indicated that TCC exhibited higher GPER binding affinity than TCS theoretically. Calcium mobilization assay displayed that TCC/TCS activated GPER signaling pathway with the lowest observed effective concentrations (LOEC) of 10 nM/100 nM. TCC and TCS also upregulated MMP-2/9, EGFR, MAPK3 but downregulated MAPK8 via GPER-mediated signaling pathway. Proliferation assay showed that TCC/TCS induced 4 T1 breast cancer cells proliferation with the LOEC of 100 nM/1000 nM. Wound-healing and transwell assays showed that TCC/TCS promoted 4 T1 cells migration in a concentration-dependent manner with the LOEC of 10 nM. The effects of TCC on breast cancer cells proliferation and migration were stronger than TCS and both were regulated by GPER. TCC/TCS induced migratory effects were more significantly than proliferative effect. Mechanism study showed that TCC/TCS downregulated the expression of epithelial marker (E-cadherin) but upregulated mesenchymal markers (snail and N-cadherin), which was reversed by GPER inhibitor G15. These biomarkers results indicated that TCC/TCS-induced 4 T1 cells migration was a classic epithelial to mesenchymal transition mechanism regulated by GPER signaling pathway. Orthotopic tumor model verified that TCC promoted breast cancer in-situ tumor growth and distal tissue metastasis via GPER-mediated signaling pathway at human-exposure level of 10 mg/kg/d. TCC-induced tissue metastasis of breast cancer was more significantly than in-situ tumor growth. Overall, we demonstrated for the first time that TCC/TCS could activate the GPER signaling pathways to induce breast cancer progression.
Collapse
Affiliation(s)
- Ting-Ting He
- College of Environment and Ecology, Hunan Agricultural University, Changsha 410128, China
| | - Xin Li
- College of Environment and Ecology, Hunan Agricultural University, Changsha 410128, China
| | - Jie-Zhi Ma
- Department of Obstetrics and Gynecology, Xiangya Third Hospital, Central South University, Changsha 410013, China
| | - Yuan Yang
- College of Environment and Ecology, Hunan Agricultural University, Changsha 410128, China
| | - Shiye Zhu
- College of Environment and Ecology, Hunan Agricultural University, Changsha 410128, China
| | - Jianhua Zeng
- College of Food Science and Engineering, Ocean University of China, Qingdao 266000, China
| | - Lin Luo
- College of Environment and Ecology, Hunan Agricultural University, Changsha 410128, China
| | - Yu-Long Yin
- College of Environment and Ecology, Hunan Agricultural University, Changsha 410128, China
| | - Lin-Ying Cao
- College of Environment and Ecology, Hunan Agricultural University, Changsha 410128, China.
| |
Collapse
|
8
|
EFSA Panel on Food Contact Materials, Enzymes and Processing Aids (CEP), Lambré C, Barat Baviera JM, Bolognesi C, Chesson A, Cocconcelli PS, Crebelli R, Gott DM, Grob K, Lampi E, Mengelers M, Mortensen A, Rivière G, Silano (until 21 December 2020†) V, Steffensen I, Tlustos C, Vernis L, Zorn H, Batke M, Bignami M, Corsini E, FitzGerald R, Gundert‐Remy U, Halldorsson T, Hart A, Ntzani E, Scanziani E, Schroeder H, Ulbrich B, Waalkens‐Berendsen D, Woelfle D, Al Harraq Z, Baert K, Carfì M, Castoldi AF, Croera C, Van Loveren H. Re-evaluation of the risks to public health related to the presence of bisphenol A (BPA) in foodstuffs. EFSA J 2023; 21:e06857. [PMID: 37089179 PMCID: PMC10113887 DOI: 10.2903/j.efsa.2023.6857] [Citation(s) in RCA: 72] [Impact Index Per Article: 36.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/25/2023] Open
Abstract
In 2015, EFSA established a temporary tolerable daily intake (t-TDI) for BPA of 4 μg/kg body weight (bw) per day. In 2016, the European Commission mandated EFSA to re-evaluate the risks to public health from the presence of BPA in foodstuffs and to establish a tolerable daily intake (TDI). For this re-evaluation, a pre-established protocol was used that had undergone public consultation. The CEP Panel concluded that it is Unlikely to Very Unlikely that BPA presents a genotoxic hazard through a direct mechanism. Taking into consideration the evidence from animal data and support from human observational studies, the immune system was identified as most sensitive to BPA exposure. An effect on Th17 cells in mice was identified as the critical effect; these cells are pivotal in cellular immune mechanisms and involved in the development of inflammatory conditions, including autoimmunity and lung inflammation. A reference point (RP) of 8.2 ng/kg bw per day, expressed as human equivalent dose, was identified for the critical effect. Uncertainty analysis assessed a probability of 57-73% that the lowest estimated Benchmark Dose (BMD) for other health effects was below the RP based on Th17 cells. In view of this, the CEP Panel judged that an additional uncertainty factor (UF) of 2 was needed for establishing the TDI. Applying an overall UF of 50 to the RP, a TDI of 0.2 ng BPA/kg bw per day was established. Comparison of this TDI with the dietary exposure estimates from the 2015 EFSA opinion showed that both the mean and the 95th percentile dietary exposures in all age groups exceeded the TDI by two to three orders of magnitude. Even considering the uncertainty in the exposure assessment, the exceedance being so large, the CEP Panel concluded that there is a health concern from dietary BPA exposure.
Collapse
|
9
|
Ang HL, Mohan CD, Shanmugam MK, Leong HC, Makvandi P, Rangappa KS, Bishayee A, Kumar AP, Sethi G. Mechanism of epithelial-mesenchymal transition in cancer and its regulation by natural compounds. Med Res Rev 2023. [PMID: 36929669 DOI: 10.1002/med.21948] [Citation(s) in RCA: 105] [Impact Index Per Article: 52.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2022] [Revised: 12/19/2022] [Accepted: 02/27/2023] [Indexed: 03/18/2023]
Abstract
Epithelial-mesenchymal transition (EMT) is a complex process with a primordial role in cellular transformation whereby an epithelial cell transforms and acquires a mesenchymal phenotype. This transformation plays a pivotal role in tumor progression and self-renewal, and exacerbates resistance to apoptosis and chemotherapy. EMT can be initiated and promoted by deregulated oncogenic signaling pathways, hypoxia, and cells in the tumor microenvironment, resulting in a loss-of-epithelial cell polarity, cell-cell adhesion, and enhanced invasive/migratory properties. Numerous transcriptional regulators, such as Snail, Slug, Twist, and ZEB1/ZEB2 induce EMT through the downregulation of epithelial markers and gain-of-expression of the mesenchymal markers. Additionally, signaling cascades such as Wnt/β-catenin, Notch, Sonic hedgehog, nuclear factor kappa B, receptor tyrosine kinases, PI3K/AKT/mTOR, Hippo, and transforming growth factor-β pathways regulate EMT whereas they are often deregulated in cancers leading to aberrant EMT. Furthermore, noncoding RNAs, tumor-derived exosomes, and epigenetic alterations are also involved in the modulation of EMT. Therefore, the regulation of EMT is a vital strategy to control the aggressive metastatic characteristics of tumor cells. Despite the vast amount of preclinical data on EMT in cancer progression, there is a lack of clinical translation at the therapeutic level. In this review, we have discussed thoroughly the role of the aforementioned transcription factors, noncoding RNAs (microRNAs, long noncoding RNA, circular RNA), signaling pathways, epigenetic modifications, and tumor-derived exosomes in the regulation of EMT in cancers. We have also emphasized the contribution of EMT to drug resistance and possible therapeutic interventions using plant-derived natural products, their semi-synthetic derivatives, and nano-formulations that are described as promising EMT blockers.
Collapse
Affiliation(s)
- Hui Li Ang
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | | | - Muthu K Shanmugam
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Hin Chong Leong
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Pooyan Makvandi
- Istituto Italiano di Tecnologia Centre for Materials Interface, Pontedera, Pisa, Italy
| | | | - Anupam Bishayee
- College of Osteopathic Medicine, Lake Erie College of Osteopathic Medicine, Bradenton, Florida, USA
| | - Alan Prem Kumar
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- NUS Center for Cancer Research, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Gautam Sethi
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- NUS Center for Cancer Research, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| |
Collapse
|
10
|
Ahn C, Jeung EB. Endocrine-Disrupting Chemicals and Disease Endpoints. Int J Mol Sci 2023; 24:ijms24065342. [PMID: 36982431 PMCID: PMC10049097 DOI: 10.3390/ijms24065342] [Citation(s) in RCA: 57] [Impact Index Per Article: 28.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2023] [Accepted: 02/24/2023] [Indexed: 03/18/2023] Open
Abstract
Endocrine-disrupting chemicals (EDCs) have significant impacts on biological systems, and have been shown to interfere with physiological systems, especially by disrupting the hormone balance. During the last few decades, EDCs have been shown to affect reproductive, neurological, and metabolic development and function and even stimulate tumor growth. EDC exposure during development can disrupt normal development patterns and alter susceptibility to disease. Many chemicals have endocrine-disrupting properties, including bisphenol A, organochlorines, polybrominated flame retardants, alkylphenols, and phthalates. These compounds have gradually been elucidated as risk factors for many diseases, such as reproductive, neural, and metabolic diseases and cancers. Endocrine disruption has been spread to wildlife and species that are connected to the food chains. Dietary uptake represents an important source of EDC exposure. Although EDCs represent a significant public health concern, the relationship and specific mechanism between EDCs and diseases remain unclear. This review focuses on the disease-EDC relationship and the disease endpoints associated with endocrine disruption for a better understanding of the relationship between EDCs-disease and elucidates the development of new prevention/treatment opportunities and screening methods.
Collapse
Affiliation(s)
- Changhwan Ahn
- Laboratory of Veterinary Physiology, College of Veterinary Medicine, Jeju National University, Jeju 63243, Republic of Korea
| | - Eui-Bae Jeung
- Laboratory of Veterinary Biochemistry and Molecular Biology, College of Veterinary Medicine, Chungbuk National University, Cheongju 28644, Republic of Korea
- Correspondence: ; Tel.: +82-043-261-2397; Fax: +82-43-267-3150
| |
Collapse
|
11
|
Yu M, Xu L, Lei B, Sun S, Yang Y. Tetrachlorobisphenol A and bisphenol AF induced cell migration by activating PI3K/Akt signaling pathway via G protein-coupled estrogen receptor 1 in SK-BR-3 cells. ENVIRONMENTAL TOXICOLOGY 2023; 38:126-135. [PMID: 36190352 DOI: 10.1002/tox.23669] [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: 05/06/2022] [Revised: 08/24/2022] [Accepted: 09/16/2022] [Indexed: 06/16/2023]
Abstract
Different subtypes of breast cancer express positively G protein-coupled estrogen receptor 1 (GPER1). Our previous studies found that tetrachlorobisphenol A (TCBPA) and bisphenol AF (BPAF) significantly promoted SK-BR-3 cell proliferation by activating GPER1-regulated signals. The present study further investigated the effects of TCBPA and BPAF on the migration of SK-BR-3 cells and examined the role of phosphatidylinositol 3-kinase-protein kinase B (PI3K/Akt) and its downstream signal targets in this process. We found that low-concentration BPAF and TCBPA markedly accelerated the migration of SK-BR-3 cells and elevated the mRNA levels of target genes associated with PI3K/Akt and mitogen-activated protein kinase (MAPK) signals. TCBPA- and BPAF-induced upregulation of target genes was significantly reduced by GPER1 inhibitor G15, the PI3K/Akt inhibitor wortmannin (WM), and the epidermal growth factor receptor (EGFR) inhibitor ZD1839 (ZD). G15 and WM also decreased cell migration induced by TCBPA and BPAF. The findings revealed that TCBPA and BPAF promoted SK-BR-3 cell migration ability by activating PI3K/Akt signaling pathway via GPER1-EGFR.
Collapse
Affiliation(s)
- Mengjie Yu
- Institute of Environmental Pollution and Health, School of Environmental and Chemical Engineering, Shanghai University, Shanghai, People's Republic of China
| | - Lanbing Xu
- Institute of Environmental Pollution and Health, School of Environmental and Chemical Engineering, Shanghai University, Shanghai, People's Republic of China
| | - Bingli Lei
- Institute of Environmental Pollution and Health, School of Environmental and Chemical Engineering, Shanghai University, Shanghai, People's Republic of China
| | - Su Sun
- Institute of Environmental Pollution and Health, School of Environmental and Chemical Engineering, Shanghai University, Shanghai, People's Republic of China
| | - Yingxin Yang
- Institute of Environmental Pollution and Health, School of Environmental and Chemical Engineering, Shanghai University, Shanghai, People's Republic of China
| |
Collapse
|
12
|
Yu M, Tang Q, Lei B, Yang Y, Xu L. Bisphenol AF Promoted the Growth of Uterus and Activated Estrogen Signaling Related Targets in Various Tissues of Nude Mice with SK-BR-3 Xenograft Tumor. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2022; 19:15743. [PMID: 36497816 PMCID: PMC9741110 DOI: 10.3390/ijerph192315743] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Revised: 11/23/2022] [Accepted: 11/23/2022] [Indexed: 06/17/2023]
Abstract
Environmental estrogens can promote the growth, migration, and invasion of breast cancer. However, few studies evaluate adverse health impacts of environmental estrogens on other organs of breast cancer patients. Therefore, the present study investigated the effects of environmental estrogen bisphenol AF (BPAF) on the main organs of female Balb/cA nude mice with SK-BR-3 xenograft tumor by detecting the organ development and gene expression of targets associated with G protein-coupled estrogen receptor 1 (GPER1)-mediated phosphatidylinositol 3-kinase/protein kinase B (PI3K/Akt) and mitogen-activated protein kinase (MAPK) signaling pathways in hypothalamus, ovary, uterus, liver, and kidney. The results showed that BPAF at 20 mg/kg bw/day markedly increased the uterine weight and the uterine coefficient of nude mice compared to SK-BR-3 bearing tumor control, indicating that BPAF promoted the growth of uterus due to its estrogenic activity. Additionally, BPAF significantly up-regulated the mRNA relative expression of most targets related to nuclear estrogen receptor alpha (ERα) and GPER1-mediated signaling pathways in the hypothalamus, followed by the ovary and uterus, and the least in the liver and kidney, indicating that BPAF activated different estrogen activity related targets in different tissues. In addition, BPAF markedly up-regulated the mRNA expression of GPER1 in all tested tissues, and the molecular docking showed that BPAF could dock into GPER1. Because gene change is an early event of toxicity response, these findings suggested that BPAF might aggravate the condition of breast cancer patients through exerting its estrogenic activity via the GPER1 pathway in various organs.
Collapse
|
13
|
Tarafdar A, Sirohi R, Balakumaran PA, Reshmy R, Madhavan A, Sindhu R, Binod P, Kumar Y, Kumar D, Sim SJ. The hazardous threat of Bisphenol A: Toxicity, detection and remediation. JOURNAL OF HAZARDOUS MATERIALS 2022; 423:127097. [PMID: 34488101 DOI: 10.1016/j.jhazmat.2021.127097] [Citation(s) in RCA: 122] [Impact Index Per Article: 40.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/06/2021] [Revised: 08/23/2021] [Accepted: 08/29/2021] [Indexed: 06/13/2023]
Abstract
Bisphenol A (or BPA) is a toxic endocrine disrupting chemical that is released into the environment through modern manufacturing practices. BPA can disrupt the production, function and activity of endogenous hormones causing irregularity in the hypothalamus-pituitary-gonadal glands and also the pituitary-adrenal function. BPA has immuno-suppression activity and can downregulate T cells and antioxidant genes. The genotoxicity and cytotoxicity of BPA is paramount and therefore, there is an immediate need to properly detect and remediate its influence. In this review, we discuss the toxic effects of BPA on different metabolic systems in the human body, followed by its mechanism of action. Various novel detection techniques (LC-MS, GC-MS, capillary electrophoresis, immunoassay and sensors) involving a pretreatment step (liquid-liquid microextraction and molecularly imprinted solid-phase extraction) have also been detailed. Mechanisms of various remediation strategies, including biodegradation using native enzymes, membrane separation processes, photocatalytic oxidation, use of nanosorbents and thermal degradation has been detailed. An overview of the global regulations pertaining to BPA has been presented. More investigations are required on the efficiency of integrated remediation technologies rather than standalone methods for BPA removal. The effect of processing operations on BPA in food matrices is also warranted to restrict its transport into food products.
Collapse
Affiliation(s)
- Ayon Tarafdar
- Livestock Production and Management Section, ICAR-Indian Veterinary Research Institute, Izatnagar, Bareilly 243122, Uttar Pradesh, India
| | - Ranjna Sirohi
- Department of Chemical and Biological Engineering, Korea University, 145, Anam-ro, Seongbuk-gu, Seoul 02841, South Korea
| | - Palanisamy Athiyaman Balakumaran
- Microbial Processes and Technology Division, CSIR-National Institute for Interdisciplinary Science and Technology (CSIR-NIIST), Trivandrum 695019, Kerala, India
| | - R Reshmy
- Department of Chemistry, Bishop Moore College, Mavelikkara 690110, Kerela, India
| | - Aravind Madhavan
- Rajiv Gandhi Centre for Biotechnology, Trivandrum 695014, Kerela, India
| | - Raveendran Sindhu
- Microbial Processes and Technology Division, CSIR-National Institute for Interdisciplinary Science and Technology (CSIR-NIIST), Trivandrum 695019, Kerala, India
| | - Parameswaran Binod
- Microbial Processes and Technology Division, CSIR-National Institute for Interdisciplinary Science and Technology (CSIR-NIIST), Trivandrum 695019, Kerala, India
| | - Yogesh Kumar
- Department of Food Science and Technology, National Institute of Food Technology and Entrepreneurship and Management, Sonipat 131028, Haryana, India
| | - Deepak Kumar
- Department of Food Science and Technology, National Institute of Food Technology and Entrepreneurship and Management, Sonipat 131028, Haryana, India
| | - Sang Jun Sim
- Department of Chemical and Biological Engineering, Korea University, 145, Anam-ro, Seongbuk-gu, Seoul 02841, South Korea.
| |
Collapse
|
14
|
The effect of endocrine disrupting chemicals on the vitronectin-receptor (integrin α vβ 3)-mediated cell adhesion of human umbilical vein endothelial cells. Toxicol In Vitro 2021; 79:105275. [PMID: 34801682 DOI: 10.1016/j.tiv.2021.105275] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Revised: 10/12/2021] [Accepted: 11/16/2021] [Indexed: 12/22/2022]
Abstract
Endocrine disrupting chemicals (EDCs) are associated with cancer development and progression due to their promotion of increased cell invasiveness and metastasis formation. However, the effects of EDCs on cell adhesion mediated through integrins have not been well studied to date. Their actions are implicated by binding sites for hormones on the vitronectin receptor (VTNR; or integrin αvβ3), which is involved in tumor angiogenesis and metastasis. VTNR-expressing human umbilical vein endothelial cells (HUVECs) were used to determine the effects of EDCs and endogenous hormones on cell adhesion to vitronectin-coated surfaces, and on VTNR activation. Cell adhesion was significantly increased for bisphenol A, triclocarban, and triclosan (10, 100 nM; p < 0.05), with similar trends for bisphenols AF and S (10, 100 nM; p > 0.05). No changes in cell adhesion were seen for 5α-dihydrotestosterone, 17β-estradiol, triiodothyronine, imatinib and paroxetine. These data indicate that EDC-mediated increases in HUVEC adhesion to vitronectin are not mediated through androgenic, estrogenic, or thyroid activities, nor through activation of VTNR. Although these effects of EDCs on HUVEC adhesion require further investigation of the underlying mechanism(s) of action to define their biological relevance, the low-dose effects and nonmonotonic responses revealed here define the need for further investigation of these EDCs.
Collapse
|
15
|
Bhattacharya T, Dutta S, Akter R, Rahman MH, Karthika C, Nagaswarupa HP, Murthy HCA, Fratila O, Brata R, Bungau S. Role of Phytonutrients in Nutrigenetics and Nutrigenomics Perspective in Curing Breast Cancer. Biomolecules 2021; 11:1176. [PMID: 34439842 PMCID: PMC8394348 DOI: 10.3390/biom11081176] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2021] [Revised: 07/15/2021] [Accepted: 08/05/2021] [Indexed: 02/07/2023] Open
Abstract
Breast cancer (BC) is one of the most common type of cancer and an important contributor to female mortality. Several genes and epigenetic modifications are involved in the development and progression of BC. Research in phytochemistry, nutrigenomics, and nutrigenetics has provided strong evidence that certain phytonutrients are able to modulate gene expression at transcriptional and post-transcriptional levels. Such phytonutrients may also be beneficial to prevent and treat BC. In this review, we will focus on the nutrigenomic effects of various phytochemicals including polyphenols, phytosterols, terpenoids, alkaloids, and other compounds from different sources. Overall, these phytonutrients are found to inhibit BC cell proliferation, differentiation, invasion, metastasis, angiogenesis, and induce apoptotic cell death by targeting various molecular pathways. They also alter epigenetic mechanisms and enhance the chemosensitivity and radiosensitivity of cancer cells. Such phytochemicals may be used for the effective management of BC patients in the clinical setting in the future. The present article aims to summarize the specific molecular pathways involved in the genetic effects of phytochemicals in BC.
Collapse
Affiliation(s)
- Tanima Bhattacharya
- College of Chemistry and Chemical Engineering, Hubei University, Wuhan 430062, China;
- Techno India NJR Institute of Technology, Udaipur, Rajasthan 313003, India
| | - Soumam Dutta
- Food and Nutrition Division, University of Calcutta, Calcutta 700027, India;
| | - Rokeya Akter
- Department of Pharmacy, Jagannath University, Sadarghat, Dhaka 1100, Bangladesh;
- Department of Global Medical Science, Yonsei University Wonju College of Medicine, Yonsei University, Wonju 26426, Gangwon-do, Korea
| | - Md. Habibur Rahman
- Department of Global Medical Science, Yonsei University Wonju College of Medicine, Yonsei University, Wonju 26426, Gangwon-do, Korea
- Department of Pharmacy, Southeast University, Banani, Dhaka 1213, Bangladesh
| | - Chenmala Karthika
- Department of Pharmaceutics, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Ooty 643001, India;
| | | | - Hanabe Chowdappa Ananda Murthy
- Department of Applied Chemistry, School of Applied Natural Science, Adama Science and Technology University, Adama P.O. Box 1888, Ethiopia;
| | - Ovidiu Fratila
- Department of Medical Disciplines, Faculty of Medicine and Pharmacy, University of Oradea, 410073 Oradea, Romania; (O.F.); (R.B.)
| | - Roxana Brata
- Department of Medical Disciplines, Faculty of Medicine and Pharmacy, University of Oradea, 410073 Oradea, Romania; (O.F.); (R.B.)
| | - Simona Bungau
- Department of Pharmacy, Faculty of Medicine and Pharmacy, University of Oradea, 410028 Oradea, Romania
- Doctoral School of Biological and Biomedical Sciences, University of Oradea, 410087 Oradea, Romania
| |
Collapse
|
16
|
Deng P, Tan M, Zhou W, Chen C, Xi Y, Gao P, Ma Q, Liang Y, Chen M, Tian L, Xie J, Liu M, Luo Y, Li Y, Zhang L, Wang L, Zeng Y, Pi H, Yu Z, Zhou Z. Bisphenol A promotes breast cancer cell proliferation by driving miR-381-3p-PTTG1-dependent cell cycle progression. CHEMOSPHERE 2021; 268:129221. [PMID: 33352510 DOI: 10.1016/j.chemosphere.2020.129221] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/09/2020] [Revised: 11/22/2020] [Accepted: 12/03/2020] [Indexed: 06/12/2023]
Abstract
Bisphenol A (BPA) is a high-production-volume industrial chemical that facilitates the development of breast cancer. However, the molecular mechanism associated with BPA-induced breast cancer cell proliferation and migration remains elusive. In our study, we exposed MCF-7 cells to different concentrations of BPA (0.1, 1 and 10 μM) for 24, 48, or 72 h. We found that BPA exposure significantly promoted MCF-7 cell proliferation and migration but not invasion. To elucidate the mechanisms, the differentially expressed genes between the BPA and control groups were investigated with the Gene Expression Omnibus (GEO) database through GEO2R. Kyoto Encyclopedia of Genes and Genomes (KEGG) and pathway action network analyses demonstrated the important role of the cell cycle pathway in the effects of BPA exposure on MCF-7 cells. Importantly, analysis with the cytoHubba plugin of Cytoscape software coupled with analysis of enriched genes in the cell cycle pathway identified PTTG1 and CDC20 (two hub genes) as key targets associated with BPA-induced MCF-7 cell proliferation and migration. Interestingly, BPA significantly increased the protein expression levels of PTTG1 but not CDC20. Knockdown of PTTG1 inhibited the BPA-induced increase in proliferation and maintained cell cycle progression. In addition, we confirmed that the increased expression of PTTG1 upon BPA exposure was caused by miR-381-3p inhibition. Moreover, we verified that miR-381-3p expression was low and inversely correlated with PTTG1 expression in breast cancer tissues. Together, these findings demonstrate that BPA promotes high PTTG1 expression and alters the cell cycle to enhance MCF-7 cell proliferation by inhibiting miR-381-3p expression.
Collapse
Affiliation(s)
- Ping Deng
- Department of Occupational Health (Key Laboratory of Electromagnetic Radiation Protection, Ministry of Education), Third Military Medical University, Chongqing, China
| | - Miduo Tan
- Surgery Department of Galactophore, The Affiliated Zhuzhou Hospital Xiangya Medical College CSU (Central Hospital of Zhuzhou City), Zhuzhou, China
| | - Wei Zhou
- Surgery Department of Galactophore, The Affiliated Zhuzhou Hospital Xiangya Medical College CSU (Central Hospital of Zhuzhou City), Zhuzhou, China
| | - Chunhai Chen
- Department of Occupational Health (Key Laboratory of Electromagnetic Radiation Protection, Ministry of Education), Third Military Medical University, Chongqing, China
| | - Yu Xi
- Department of Environmental Medicine, School of Public Health, and Department of Emergency Medicine, First Affiliated Hospital, School of Medicine, Zhejiang University, China
| | - Peng Gao
- Department of Occupational Health (Key Laboratory of Electromagnetic Radiation Protection, Ministry of Education), Third Military Medical University, Chongqing, China
| | - Qinlong Ma
- Department of Occupational Health (Key Laboratory of Electromagnetic Radiation Protection, Ministry of Education), Third Military Medical University, Chongqing, China
| | - Yidan Liang
- School of Medicine, Guangxi University, Nanning, Guangxi Zhuang Autonomous Region, China
| | - Mengyan Chen
- Department of Occupational Health (Key Laboratory of Electromagnetic Radiation Protection, Ministry of Education), Third Military Medical University, Chongqing, China
| | - Li Tian
- Department of Occupational Health (Key Laboratory of Electromagnetic Radiation Protection, Ministry of Education), Third Military Medical University, Chongqing, China
| | - Jia Xie
- Department of Occupational Health (Key Laboratory of Electromagnetic Radiation Protection, Ministry of Education), Third Military Medical University, Chongqing, China
| | - Mengyu Liu
- Department of Occupational Health (Key Laboratory of Electromagnetic Radiation Protection, Ministry of Education), Third Military Medical University, Chongqing, China
| | - Yan Luo
- Department of Occupational Health (Key Laboratory of Electromagnetic Radiation Protection, Ministry of Education), Third Military Medical University, Chongqing, China
| | - Yanqi Li
- Department of Occupational Health (Key Laboratory of Electromagnetic Radiation Protection, Ministry of Education), Third Military Medical University, Chongqing, China
| | - Lei Zhang
- Department of Occupational Health (Key Laboratory of Electromagnetic Radiation Protection, Ministry of Education), Third Military Medical University, Chongqing, China
| | - Liting Wang
- Biomedical Analysis Center, Third Military Medical University, Chongqing, China
| | - Youlong Zeng
- Biomedical Analysis Center, Third Military Medical University, Chongqing, China
| | - Huifeng Pi
- Department of Occupational Health (Key Laboratory of Electromagnetic Radiation Protection, Ministry of Education), Third Military Medical University, Chongqing, China.
| | - Zhengping Yu
- Department of Occupational Health (Key Laboratory of Electromagnetic Radiation Protection, Ministry of Education), Third Military Medical University, Chongqing, China.
| | - Zhou Zhou
- Department of Environmental Medicine, School of Public Health, and Department of Emergency Medicine, First Affiliated Hospital, School of Medicine, Zhejiang University, China.
| |
Collapse
|
17
|
Alfhili MA, Hussein HAM, Park Y, Lee MH, Akula SM. Triclosan induces apoptosis in Burkitt lymphoma-derived BJAB cells through caspase and JNK/MAPK pathways. Apoptosis 2021; 26:96-110. [PMID: 33387145 DOI: 10.1007/s10495-020-01650-0] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/30/2020] [Indexed: 12/23/2022]
Abstract
Burkitt's lymphoma (BL) is the fastest growing human tumor. Current treatment consists of a multiagent regimen of cytotoxic drugs with serious side effjects including tumor lysis, cardiotoxicity, hepatic impairment, neuropathy, myelosuppression, increased susceptibility to malignancy, and death. Furthermore, therapeutic interventions in areas of BL prevalence are not as feasible as in high-income countries. Therefore, there exists an urgent need to identify new therapies with a safer profile and improved accessibility. Triclosan (TCS), an antimicrobial used in personal care products and surgical scrubs, has gained considerable interest as an antitumor agent due to its interference with fatty acid synthesis. Here, we investigate the antitumor properties and associated molecular mechanisms of TCS in BL-derived BJAB cells. Dose-dependent cell death was observed following treatment with 10-100 µM TCS for 24 h, which was associated with membrane phospholipid scrambling, compromised permeability, and cell shrinkage. TCS-induced cell death was accompanied by elevated intracellular calcium, perturbed redox balance, chromatin condensation, and DNA fragmentation. TCS upregulated Bad expression and downregulated that of Bcl2. Moreover, caspase and JNK MAPK signaling were required for the full apoptotic activity of TCS. In conclusion, this report identifies TCS as an antitumor agent and provides new insights into the molecular mechanisms governing TCS-induced apoptosis in BL cells.
Collapse
Affiliation(s)
- Mohammad A Alfhili
- Chair of Medical and Molecular Genetics Research, Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, King Saud University, Riyadh, 11433, Saudi Arabia
- Division of Hematology/Oncology, Department of Internal Medicine, Brody School of Medicine, East Carolina University, Greenville, NC, 27834, USA
| | - Hosni A M Hussein
- Department of Microbiology & Immunology, Brody School of Medicine, East Carolina University, Greenville, NC. 27834, USA
- Faculty of Science, Assiut Branch, Al Azhar University, Assiut, 71524, Egypt
| | - Youngyong Park
- Division of Hematology/Oncology, Department of Internal Medicine, Brody School of Medicine, East Carolina University, Greenville, NC, 27834, USA
| | - Myon Hee Lee
- Division of Hematology/Oncology, Department of Internal Medicine, Brody School of Medicine, East Carolina University, Greenville, NC, 27834, USA.
- Lineberger Comprehensive Cancer Center, University of North Carolina-Chapel Hill, Chapel Hill, NC, 27599, USA.
| | - Shaw M Akula
- Department of Microbiology & Immunology, Brody School of Medicine, East Carolina University, Greenville, NC. 27834, USA.
| |
Collapse
|
18
|
Stillwater BJ, Bull AC, Romagnolo DF, Neumayer LA, Donovan MG, Selmin OI. Bisphenols and Risk of Breast Cancer: A Narrative Review of the Impact of Diet and Bioactive Food Components. Front Nutr 2020; 7:581388. [PMID: 33330580 PMCID: PMC7710764 DOI: 10.3389/fnut.2020.581388] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2020] [Accepted: 10/15/2020] [Indexed: 12/13/2022] Open
Abstract
Data from preclinical studies suggest a link between increased risk of breast cancer and exposure to bisphenols at doses below what the United States Food and Drug Administration (FDA) considers as safe for consumption. Bisphenols exert estrogenic effects and are found in canned and plastic wrapped foods, food packaging, and plasticware. Mechanistically, bisphenols bind to the estrogen receptor (ER) and activate the expression of genes associated with cell proliferation and breast cancer. In this paper, we present a narrative literature review addressing bisphenol A and chemical analogs including bisphenol AF, bisphenol F, and bisphenol S selected as prototype xenoestrogens; then, we discuss biological mechanisms of action of these bisphenols in breast cells and potential impact of exposure at different stages of development (i.e., perinatal, peripubertal, and adult). Finally, we summarize studies detailing interactions, both preventative and promoting, of bisphenols with food components on breast cancer risk. We conclude the review with a discussion of current controversies in interpretation of the above research and future areas for investigation, including the impact of bisphenols and food components on breast tumor risk.
Collapse
Affiliation(s)
- Barbara J Stillwater
- Department of Surgery, Breast Surgical Oncology, University of Arizona, Tucson, AZ, United States
| | - Ashleigh C Bull
- School of Medicine, University of Utah, Salt Lake City, UT, United States
| | - Donato F Romagnolo
- Department of Nutritional Sciences, University of Arizona, Tucson, AZ, United States.,University of Arizona Cancer Center, Tucson, AZ, United States
| | - Leigh A Neumayer
- Department of Surgery, University of Florida College of Medicine-Jacksonville, Jacksonville, FL, United States
| | - Micah G Donovan
- Department of Nutritional Sciences, University of Arizona, Tucson, AZ, United States.,University of Arizona Cancer Center, Tucson, AZ, United States
| | - Ornella I Selmin
- Department of Nutritional Sciences, University of Arizona, Tucson, AZ, United States.,University of Arizona Cancer Center, Tucson, AZ, United States
| |
Collapse
|
19
|
Koli P, Reena, Indurthi HK, Sharma DK. Anticancer Activity of 3,3′‐Diindolylmethane and the Molecular Mechanism Involved in Various Cancer Cell Lines. ChemistrySelect 2020. [DOI: 10.1002/slct.202003137] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Affiliation(s)
- Papita Koli
- Department of Pharmaceutical Engg. and Tech. Indian Institute of Technology-Banaras Hindu University Varanasi, Uttar Pradesh India
| | - Reena
- Overseas Healthcare Pvt. Ltd. Phillaur, Punjab India
| | - Harish K. Indurthi
- Department of Pharmaceutical Engg. and Tech. Indian Institute of Technology-Banaras Hindu University Varanasi, Uttar Pradesh India
| | - Deepak K. Sharma
- Department of Pharmaceutical Engg. and Tech. Indian Institute of Technology-Banaras Hindu University Varanasi, Uttar Pradesh India
| |
Collapse
|
20
|
Awada Z, Nasr R, Akika R, Ghantous A, Hou L, Zgheib NK. Effect of bisphenols on telomerase expression and activity in breast cancer cell lines. Mol Biol Rep 2020; 47:3541-3549. [PMID: 32333245 DOI: 10.1007/s11033-020-05444-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2019] [Accepted: 04/09/2020] [Indexed: 12/15/2022]
Abstract
Bisphenol A (BPA), a monomer of polycarbonates and resins, was shown to induce the expression of telomerase enzyme which has been associated with breast cancer development and progression. However, the effects of BPA analogues, bisphenol F (BPF) and bisphenol S (BPS) on telomere-linked pathway have not been evaluated. Herein, MCF-7 (estrogen receptor (ER)-positive) and MDA-MB-231 (ER-negative) cells were treated with BPA, BPF and BPS ± estrogen receptor inhibitor (ERI), for 24 and/or 48 h. RNA expression and enzymatic activity of telomerase were measured using reverse transcription quantitative polymerase chain reaction (RT-qPCR) and telomeric repeat amplification protocol (TRAP); respectively. Relative telomere length (RTL) was also measured using quantitative PCR. After 24 h, the three bisphenols resulted in a 2-3 folds increase in expression and activity of telomerase in MCF-7 but not in MDA-MB-231 cells, and this increase was prevented upon co-treatment with ERI. The observed increase in the expression and activity of telomerase after 24 h of treatment with bisphenols was associated with differential and modest ER-dependent lengthening in RTL at 48 h. Our results show that telomerase potentially mediates the effects of the three bisphenols in ER-positive breast carcinoma. Hence, further investigation is warranted to elucidate the telomerase-linked pathways that could underlie bisphenol-related effects.
Collapse
Affiliation(s)
- Zainab Awada
- Department of Pharmacology and Toxicology, Faculty of Medicine, American University of Beirut, Riad El-Solh, Beirut, 1107-2020, Lebanon
| | - Rihab Nasr
- Department of Anatomy, Cell Biology and Physiology, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| | - Reem Akika
- Department of Pharmacology and Toxicology, Faculty of Medicine, American University of Beirut, Riad El-Solh, Beirut, 1107-2020, Lebanon
| | - Akram Ghantous
- Epigenetics Group, International Agency for Research On Cancer, Lyon, France
| | - Lifang Hou
- Department of Preventive Medicine, Northwestern University, Chicago, USA
| | - Nathalie K Zgheib
- Department of Pharmacology and Toxicology, Faculty of Medicine, American University of Beirut, Riad El-Solh, Beirut, 1107-2020, Lebanon.
| |
Collapse
|
21
|
Mousa DS, El-Far AH, Saddiq AA, Sudha T, Mousa SA. Nanoformulated Bioactive Compounds Derived from Different Natural Products Combat Pancreatic Cancer Cell Proliferation. Int J Nanomedicine 2020; 15:2259-2268. [PMID: 32280218 PMCID: PMC7127850 DOI: 10.2147/ijn.s238256] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2019] [Accepted: 02/26/2020] [Indexed: 12/12/2022] Open
Abstract
Purpose This study was designed to determine the potential effect of nanoencapsulated bioactive compounds from different natural sources on human pancreatic cancer. Background Pancreatic cancer carries the highest fatality rate among all human cancers because of its high metastatic potential and late presentation at the time of diagnosis. Hence there is a need for improved methods to prevent and treat it. Natural products, such as 3, 3′-diindolylmethane (DIM) and ellagic acid (EA) demonstrated anticancer efficacy against various cancer types. However, DIM is insoluble. Hence, using nanotechnology to encapsulate these compounds in combination with EA might improve their physical and chemical properties and their delivery to the cancer cells. Methods Human pancreatic cancer cells, namely SUIT2-luciferase transfected, were used to examine the effects of DIM or EA and their nanoformulation in poly(lactic-co-glycolic acid) (PLGA) and poly(ethylene glycol) (PEG) [PLGA-PEG] nanoparticles (NPs) on SUIT2-luciferase cell viability/proliferation over 24 hrs. Additionally, effects on tumor weight and angiogenesis were determined using the chick chorioallantoic membrane (CAM) tumor implant model. Results Both DIM and EA PLGA-PEG NPs resulted in rapid suppression of pancreatic cancer cell viability/proliferation within 24 hrs (P < 0.01), while the non-encapsulated DIM and EA did not show any significant effect on SUIT2 cancer cell viability or cell proliferation (MTT assay). In the CAM pancreatic cancer cell (SUIT2) implant model, results showed a greater suppression of tumor weight (P < 0.01), tumor cell viability, and tumor angiogenesis (P < 0.01) for DIM NPs and EA NPs and their combinations versus DIM or EA alone. Conclusion Nanoformulation of DIM and EA resulted in a more effective suppression of pancreatic cancer cell viability, pancreatic tumor weight, implanted cancer cell viability, and tumor angiogenesis as compared with these bioactive compounds alone.
Collapse
Affiliation(s)
| | - Ali H El-Far
- Department of Biochemistry, Faculty of Veterinary Medicine, Damanhour University, Damanhour 22511, Egypt
| | - Amna A Saddiq
- Faculty of Science, Department of Biology, University of Jeddah, Jeddah 21589, Saudi Arabia
| | - Thangirala Sudha
- Pharmaceutical Research Institute, Albany College of Pharmacy and Health Sciences, Rensselaer, NY 12144, USA
| | - Shaker A Mousa
- Pharmaceutical Research Institute, Albany College of Pharmacy and Health Sciences, Rensselaer, NY 12144, USA
| |
Collapse
|
22
|
Awada Z, Nasr R, Akika R, Cahais V, Cuenin C, Zhivagui M, Herceg Z, Ghantous A, Zgheib NK. DNA methylome-wide alterations associated with estrogen receptor-dependent effects of bisphenols in breast cancer. Clin Epigenetics 2019; 11:138. [PMID: 31601247 PMCID: PMC6785895 DOI: 10.1186/s13148-019-0725-y] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2019] [Accepted: 08/12/2019] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Bisphenol A (BPA), an estrogen-like endocrine disruptor used in plastics, has been associated with development and promotion of breast cancer, so plastic manufacturers shifted towards less-studied analogs, BPF and BPS. Studying the associated DNA methylome-wide mechanisms of these derivatives is timely, particularly in comparison with BPA. METHODS We assessed proliferation, cell cycle, and migration of breast cancer cells (estrogen receptor (ER)-positive: MCF-7 and ER-negative: MDA-MB-231) treated with BPF and BPS ± estrogen receptor inhibitor (ERI) in comparison to BPA ± ERI. RNA expression and activity of DNA (de)methylation enzymes and LINE-1 methylation were quantified. DNA methylome-wide analysis was evaluated in bisphenol-exposed cells and compared to clinical breast cancer data. RESULTS The three bisphenols caused ER-dependent increased proliferation and migration of MCF-7 but not MDA-MB-231 cells, with BPS being 10 times less potent than BPA and BPF. Although they have similar chemical structures, the three bisphenols induced differential DNA methylation alterations at several genomic clusters of or single CpG sites, with the majority of these being ER-dependent. At equipotent doses, BPA had the strongest effect on the methylome, followed by BPS then BPF. No pathways were enriched for BPF while BPA- and BPS-induced methylome alterations were enriched in focal adhesion, cGMP-PKG, and cancer pathways, which were also dysregulated in methylome-wide alterations comparing ER-positive breast cancer samples to adjacent normal tissues. CONCLUSIONS The three bisphenols have important epigenetic effects in breast cell lines, with those of BPA and BPS overlapping with cancer-related pathways in clinical breast cancer models. Hence, further investigation of their safety is warranted.
Collapse
Affiliation(s)
- Z Awada
- Pharmacology and Toxicology, Faculty of Medicine, American University of Beirut, Riad El-Solh, Beirut, 1107-2020, Lebanon
| | - R Nasr
- Anatomy, Cell Biology and Physiology, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| | - R Akika
- Pharmacology and Toxicology, Faculty of Medicine, American University of Beirut, Riad El-Solh, Beirut, 1107-2020, Lebanon
| | - V Cahais
- Epigenetics group, International Agency for Research on Cancer, Cours Albert Thomas, 69372, Lyon, France
| | - C Cuenin
- Epigenetics group, International Agency for Research on Cancer, Cours Albert Thomas, 69372, Lyon, France
| | - M Zhivagui
- Epigenetics group, International Agency for Research on Cancer, Cours Albert Thomas, 69372, Lyon, France
| | - Z Herceg
- Epigenetics group, International Agency for Research on Cancer, Cours Albert Thomas, 69372, Lyon, France
| | - A Ghantous
- Epigenetics group, International Agency for Research on Cancer, Cours Albert Thomas, 69372, Lyon, France.
| | - N K Zgheib
- Pharmacology and Toxicology, Faculty of Medicine, American University of Beirut, Riad El-Solh, Beirut, 1107-2020, Lebanon.
| |
Collapse
|
23
|
Jang YG, Go RE, Hwang KA, Choi KC. Resveratrol inhibits DHT-induced progression of prostate cancer cell line through interfering with the AR and CXCR4 pathway. J Steroid Biochem Mol Biol 2019; 192:105406. [PMID: 31185279 DOI: 10.1016/j.jsbmb.2019.105406] [Citation(s) in RCA: 57] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/09/2019] [Revised: 06/06/2019] [Accepted: 06/07/2019] [Indexed: 12/20/2022]
Abstract
Prostate cancer (PCa) is one of the most common malignancies and the second most common cause of cancer-related deaths in men world-wide and is known to be affected by the action of dihydrotestosterone (DHT) via androgen receptor (AR). Resveratrol (Res) as a phytochemical in grapes and red wine has diverse biological effects such as anti-inflammation, anti-oxidation and anti-cancer. CXCR4 as a chemokine receptor has been found to be upregulated in cancer metastasis and has been used as a prognostic marker in various types of cancer, including leukemia, breast cancer, and prostate cancer. In this study, we focused on the role of DHT in the induction of prostate cancer progression by affecting the AR and CXCR4 pathway. Also, we investigated the inhibition effect of resveratrol on DHT-induced prostate cancer metastasis. In cell viability assay, DHT increased the cell viability of LNCaP prostate cancer cells, on the other hand, Res and its combination with bicalutamide (BCT) as an AR-antagonist or AMD3100 as a CXCR4 inhibitor significantly reduced the cell viability promoted by DHT. Trans-well migration assay and wound healing assay represented the similar results with cell viability assay. According to the results of TUNEL assay, the apoptotic activity was induced by treatment of Res. As results of western blot analysis, the expression of AR, CXCR4, p-PI3K, and p-AKT and the downstream genes related with cell cycle progression and epithelial-mesenchymal transition (EMT) were decreased and the expression of the apoptosis-related genes was increased by treatment of Res and its combination with BCT or AMD3100. This study would suggest that Res and its combination with AR and CXCR4 antagonists can be used in order to suppress the metastatic behaviors of prostate cancer.
Collapse
Affiliation(s)
- Yin-Gi Jang
- Laboratory of Biochemistry and Immunology, College of Veterinary Medicine, Chungbuk National University, Cheongju, Chungbuk, Republic of Korea
| | - Ryu-Eun Go
- Laboratory of Biochemistry and Immunology, College of Veterinary Medicine, Chungbuk National University, Cheongju, Chungbuk, Republic of Korea
| | - Kyung-A Hwang
- Laboratory of Biochemistry and Immunology, College of Veterinary Medicine, Chungbuk National University, Cheongju, Chungbuk, Republic of Korea
| | - Kyung-Chul Choi
- Laboratory of Biochemistry and Immunology, College of Veterinary Medicine, Chungbuk National University, Cheongju, Chungbuk, Republic of Korea.
| |
Collapse
|
24
|
Ma Y, Liu H, Wu J, Yuan L, Wang Y, Du X, Wang R, Marwa PW, Petlulu P, Chen X, Zhang H. The adverse health effects of bisphenol A and related toxicity mechanisms. ENVIRONMENTAL RESEARCH 2019; 176:108575. [PMID: 31299621 DOI: 10.1016/j.envres.2019.108575] [Citation(s) in RCA: 426] [Impact Index Per Article: 71.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/12/2019] [Revised: 06/09/2019] [Accepted: 07/02/2019] [Indexed: 05/20/2023]
Abstract
Bisphenol A (BPA) is an industrial component commonly used in synthesis of polycarbonate plastics, epoxy resin and other polymer materials. Due to its mass productions and widespread applications, the presence of BPA is ubiquitous in the environment. BPA can enter the body via different ways such as digestive tract, respiratory tract and dermal tract. As an endocrine disruptor, BPA has estrogen-like and anti-androgen effects causing damages to different tissues and organs, including reproductive system, immune system and neuroendocrine system, etc. Recently, it has been shown that BPA could induce carcinogenesis and mutagenesis in animal models. Here, the underlying mechanisms of BPA-induced multi-organ toxicity were well summarized, involving the receptor pathways, disruption of neuroendocrine system, inhibition of enzymes, modulation of immune and inflammatory responses, as well as genotoxic and epigenetic mechanisms. The aim of this review is to compile the available current research data regarding BPA and provide an overview of the current status of BPA exposure and relevant health effects covering reproductive, developmental, metabolic, immuno, respiratory, hepatic and renal toxicity and carcinogenesis of BPA. This review provides comprehensive data of BPA toxicity on human health and related mechanisms. We also identify any missing data which should be addressed by further studies.
Collapse
Affiliation(s)
- Ya Ma
- College of Public Health, Zhengzhou University, Zhengzhou, Henan, China
| | - Haohao Liu
- College of Public Health, Zhengzhou University, Zhengzhou, Henan, China
| | - Jinxia Wu
- College of Public Health, Zhengzhou University, Zhengzhou, Henan, China
| | - Le Yuan
- College of Public Health, Zhengzhou University, Zhengzhou, Henan, China
| | - Yueqin Wang
- College of Public Health, Zhengzhou University, Zhengzhou, Henan, China
| | - Xingde Du
- College of Public Health, Zhengzhou University, Zhengzhou, Henan, China
| | - Rui Wang
- College of Public Health, Zhengzhou University, Zhengzhou, Henan, China
| | | | | | - Xinghai Chen
- Department of Chemistry and Biochemistry, St Mary's University, San Antonio, TX, USA
| | - Huizhen Zhang
- College of Public Health, Zhengzhou University, Zhengzhou, Henan, China.
| |
Collapse
|
25
|
Vimentin activation in early apoptotic cancer cells errands survival pathways during DNA damage inducer CPT treatment in colon carcinoma model. Cell Death Dis 2019; 10:467. [PMID: 31197132 PMCID: PMC6565729 DOI: 10.1038/s41419-019-1690-2] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2018] [Revised: 04/24/2019] [Accepted: 05/23/2019] [Indexed: 12/12/2022]
Abstract
Epithelial to mesenchymal transitions (EMT) is a preparatory process for cancer cells to attain motility and further metastasis to distant sites. Majority of DNA damaging drugs have shown to develop EMT as one of the major mechanisms to attain drug resistance. Here we sought to understand the resistance/survival instincts of cancer cells during initial phase of drug treatment. We provide a tangible evidence of stimulation of EMT factors in Apc knockout colorectal carcinoma model. Our results implied that CPT-treated Apc knockout cohorts depicted increased pro-invasive and pro-survival factors (Vimentin/pser38Vimentin & NFκB). Moreover, by cell sorting experiment, we have observed the expression of Vimentin in early apoptotic cells (AnnexinV positive) from 36 to 48 h of CPT treatment. We also observed the expression of chimeric Sec-AnnexinV-mvenus protein in migrated cells on transwell membrane recapitulating signatures of early apoptosis. Notably, induction of Vimentin-mediated signaling (by CPT) delayed apoptosis progression in cells conferring survival responses by modulating the promoter activity of NFκB. Furthermore, our results unveiled a novel link between Vimentin and ATM signaling, orchestrated via binding interaction between Vimentin and ATM kinase. Finally, we observed a significant alteration of crypt-villus morphology upon combination of DIM (EMT inhibitor) with CPT nullified the background EMT signals thus improving the efficacy of the DNA damaging agent. Thus, our findings revealed a resistance strategy of cancer cells within a very initial period of drug treatment by activating EMT program, which hinders the cancer cells to achieve later phases of apoptosis thus increasing the chances of early migration.
Collapse
|
26
|
Miret NV, Pontillo CA, Zárate LV, Kleiman de Pisarev D, Cocca C, Randi AS. Impact of endocrine disruptor hexachlorobenzene on the mammary gland and breast cancer: The story thus far. ENVIRONMENTAL RESEARCH 2019; 173:330-341. [PMID: 30951959 DOI: 10.1016/j.envres.2019.03.054] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/14/2018] [Revised: 02/19/2019] [Accepted: 03/21/2019] [Indexed: 06/09/2023]
Abstract
Breast cancer incidence is increasing globally and exposure to endocrine disruptors has gained importance as a potential risk factor. Hexachlorobenzene (HCB) was once used as a fungicide and, despite being banned, considerable amounts are still released into the environment. HCB acts as an endocrine disruptor in thyroid, uterus and mammary gland and was classified as possibly carcinogenic to human. This review provides a thorough analysis of results obtained in the last 15 years of research and evaluates data from assays in mammary gland and breast cancer in diverse animal models. We discuss the effects of environmentally relevant HCB concentrations on the normal mammary gland and different stages of carcinogenesis, and attempt to elucidate its mechanisms of action at molecular level. HCB weakly binds to the aryl hydrocarbon receptor (AhR), activating both membrane (c-Src) and nuclear pathways. Through c-Src stimulation, AhR signaling interacts with other membrane receptors including estrogen receptor-α, insulin-like growth factor-1 receptor, epidermal growth factor receptor and transforming growth factor beta 1 receptors. In this way, several pathways involved in mammary morphogenesis and breast cancer development are modified, inducing tumor progression. HCB thus stimulates epithelial cell proliferation, preneoplastic lesions and alterations in mammary gland development as well as neoplastic cell migration and invasion, metastasis and angiogenesis in breast cancer. In conclusion, our findings support the hypothesis that the presence and bioaccumulation of HCB in high-fat tissues and during highly sensitive time windows such as pregnancy, childhood and adolescence make exposure a risk factor for breast tumor development.
Collapse
Affiliation(s)
- Noelia V Miret
- Universidad de Buenos Aires, Facultad de Medicina, Departamento de Bioquímica Humana, Laboratorio de Efectos Biológicos de Contaminantes Ambientales, Paraguay 2155, 5to piso, CP1121, Buenos Aires, Argentina.
| | - Carolina A Pontillo
- Universidad de Buenos Aires, Facultad de Medicina, Departamento de Bioquímica Humana, Laboratorio de Efectos Biológicos de Contaminantes Ambientales, Paraguay 2155, 5to piso, CP1121, Buenos Aires, Argentina.
| | - Lorena V Zárate
- Universidad de Buenos Aires, Facultad de Medicina, Departamento de Bioquímica Humana, Laboratorio de Efectos Biológicos de Contaminantes Ambientales, Paraguay 2155, 5to piso, CP1121, Buenos Aires, Argentina.
| | - Diana Kleiman de Pisarev
- Universidad de Buenos Aires, Facultad de Medicina, Departamento de Bioquímica Humana, Laboratorio de Efectos Biológicos de Contaminantes Ambientales, Paraguay 2155, 5to piso, CP1121, Buenos Aires, Argentina.
| | - Claudia Cocca
- Universidad de Buenos Aires, Facultad de Farmacia y Bioquímica, Laboratorio de Radioisótopos, Junín 954, subsuelo, CP1113, Buenos Aires, Argentina.
| | - Andrea S Randi
- Universidad de Buenos Aires, Facultad de Medicina, Departamento de Bioquímica Humana, Laboratorio de Efectos Biológicos de Contaminantes Ambientales, Paraguay 2155, 5to piso, CP1121, Buenos Aires, Argentina.
| |
Collapse
|
27
|
Triclosan: An Update on Biochemical and Molecular Mechanisms. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2019; 2019:1607304. [PMID: 31191794 PMCID: PMC6525925 DOI: 10.1155/2019/1607304] [Citation(s) in RCA: 75] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 11/30/2018] [Revised: 02/28/2019] [Accepted: 04/01/2019] [Indexed: 12/23/2022]
Abstract
Triclosan (TCS) is a synthetic, chlorinated phenolic antimicrobial agent commonly used in commercial and healthcare products. Items made with TCS include soaps, deodorants, shampoos, cosmetics, textiles, plastics, surgical sutures, and prosthetics. A wealth of information obtained from in vitro and in vivo studies has demonstrated the therapeutic effects of TCS, particularly against inflammatory skin conditions. Nevertheless, extensive investigations on the molecular aspects of TCS action have identified numerous adversaries associated with the disinfectant including oxidative injury and influence of physiological lifespan and longevity. This review presents a summary of the biochemical alterations pertaining to TCS exposure, with special emphasis on the diverse molecular pathways responsive to TCS that have been elucidated during the present decade.
Collapse
|
28
|
Liu J, Wang S, Tian S, He Y, Lou H, Yang Z, Kong Y, Cao X. Nobiletin inhibits breast cancer via p38 mitogen-activated protein kinase, nuclear transcription factor-κB, and nuclear factor erythroid 2-related factor 2 pathways in MCF-7 cells. Food Nutr Res 2018; 62:1323. [PMID: 30574046 PMCID: PMC6294833 DOI: 10.29219/fnr.v62.1323] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2018] [Revised: 09/11/2018] [Accepted: 09/14/2018] [Indexed: 01/01/2023] Open
Abstract
INTRODUCTION Breast cancer is one of the most commonly diagnosed cancers in women, with a high mortality rate. OBJECTIVE In the present study, we evaluated the anticancer effect of nobiletin, a flavone glycoside, on the breast cancer cell line MCF-7. RESULT Cell viability and proliferation decreased and cell morphology changed from diamond to round after being treated with nobiletin. Nobiletin induced apoptosis of breast cancer MCF-7 cells via regulating the protein expression of Bax, Bcl-2, cleaved caspase-3, and p53. The expression of Bcl-2 decreased, while the expression of Bax and p53 increased in MCF-7 cells treated with nobiletin. Meanwhile, nobiletin inhibited cell migration by downregulating the protein expression of matrix metalloproteinase-2 (MMP-2) and matrix metalloproteinase-9 (MMP-9). Moreover, phosphorylation of p38 was increased, and the translocation of p65 and nuclear factor erythroid 2-related factor 2 (Nrf2) to the nucleus was decreased, which suggested that the anticancer effects of nobiletin might at least partially rely on mediating the p38 mitogen-activated protein kinase, nuclear transcription factor-κB, and Nrf2 pathways in MCF-7 breast cancer cells. CONCLUSION AND RECOMMENDATION Our data showed that nobiletin was a potential antitumor drug, and it provided some experimental basis for the clinical application of tumor therapy.
Collapse
Affiliation(s)
- Jianli Liu
- School of Life Science, Liaoning University, Shenyang, China
| | - Shuai Wang
- School of Life Science, Liaoning University, Shenyang, China
| | - Siqi Tian
- School of Life Science, Liaoning University, Shenyang, China
| | - Yin He
- School of Life Science, Liaoning University, Shenyang, China
| | - Hong Lou
- School of Life Science, Liaoning University, Shenyang, China
| | - Zhijun Yang
- School of Life Science, Liaoning University, Shenyang, China
| | - Yuchi Kong
- School of Life Science, Liaoning University, Shenyang, China
| | - Xiangyu Cao
- School of Life Science, Liaoning University, Shenyang, China
| |
Collapse
|
29
|
Pokharel A, Kolla S, Matouskova K, Vandenberg LN. Asymmetric development of the male mouse mammary gland and its response to a prenatal or postnatal estrogen challenge. Reprod Toxicol 2018; 82:63-71. [PMID: 30315872 DOI: 10.1016/j.reprotox.2018.10.003] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2018] [Revised: 09/07/2018] [Accepted: 10/09/2018] [Indexed: 12/12/2022]
Abstract
The CD-1 mouse mammary gland is sexually dimorphic, with males lacking nipples. Recent studies have revealed that the underlying epithelium in the male mammary gland is sensitive to estrogenic environmental chemicals. In ongoing investigations, we observed asymmetric morphology in the left and right male mouse mammary glands. Here, we quantified these asymmetries in the embryonic, prepubertal, pubertal and adult male mammary gland. We found that the right gland was typically larger with more branching points compared to the left gland. We next evaluated the response of the left and right glands to 17α-ethinyl estradiol (EE2) after perinatal or peripubertal exposures. We found that the right gland was more responsive to EE2 than the left at both periods of exposure. These results reveal novel aspects of male mammary gland biology and suggest that future studies should control for laterality in the evaluation of hazards associated with exposures to estrogenic chemicals.
Collapse
Affiliation(s)
- Aastha Pokharel
- Department of Environmental Health Sciences, School of Public Health and Health Sciences, University of Massachusetts, Amherst, United States
| | - SriDurgaDevi Kolla
- Department of Environmental Health Sciences, School of Public Health and Health Sciences, University of Massachusetts, Amherst, United States
| | - Klara Matouskova
- Department of Environmental Health Sciences, School of Public Health and Health Sciences, University of Massachusetts, Amherst, United States
| | - Laura N Vandenberg
- Department of Environmental Health Sciences, School of Public Health and Health Sciences, University of Massachusetts, Amherst, United States.
| |
Collapse
|
30
|
Younas M, Hano C, Giglioli-Guivarc'h N, Abbasi BH. Mechanistic evaluation of phytochemicals in breast cancer remedy: current understanding and future perspectives. RSC Adv 2018; 8:29714-29744. [PMID: 35547279 PMCID: PMC9085387 DOI: 10.1039/c8ra04879g] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2018] [Accepted: 08/15/2018] [Indexed: 12/30/2022] Open
Abstract
Breast cancer is one of the most commonly diagnosed cancers around the globe and accounts for a large proportion of fatalities in women. Despite the advancement in therapeutic and diagnostic procedures, breast cancer still represents a major challenge. Current anti-breast cancer approaches include surgical removal, radiotherapy, hormonal therapy and the use of various chemotherapeutic drugs. However, drug resistance, associated serious adverse effects, metastasis and recurrence complications still need to be resolved which demand safe and alternative strategies. In this scenario, phytochemicals have recently gained huge attention due to their safety profile and cost-effectiveness. These phytochemicals modulate various genes, gene products and signalling pathways, thereby inhibiting breast cancer cell proliferation, invasion, angiogenesis and metastasis and inducing apoptosis. Moreover, they also target breast cancer stem cells and overcome drug resistance problems in breast carcinomas. Phytochemicals as adjuvants with chemotherapeutic drugs have greatly enhanced their therapeutic efficacy. This review focuses on the recently recognized molecular mechanisms underlying breast cancer chemoprevention with the use of phytochemicals such as curcumin, resveratrol, silibinin, genistein, epigallocatechin gallate, secoisolariciresinol, thymoquinone, kaempferol, quercetin, parthenolide, sulforaphane, ginsenosides, naringenin, isoliquiritigenin, luteolin, benzyl isothiocyanate, α-mangostin, 3,3'-diindolylmethane, pterostilbene, vinca alkaloids and apigenin.
Collapse
Affiliation(s)
- Muhammad Younas
- Department of Biotechnology, Quaid-i-Azam University Islamabad-45320 Pakistan +92-51-90644121 +92-51-90644121 +33-767-97-0619
| | - Christophe Hano
- Laboratoire de Biologie des Ligneux et des Grandes Cultures (LBLGC), Plant Lignans Team, UPRES EA 1207, Université d'Orléans F 28000 Chartres France
| | | | - Bilal Haider Abbasi
- Department of Biotechnology, Quaid-i-Azam University Islamabad-45320 Pakistan +92-51-90644121 +92-51-90644121 +33-767-97-0619
- Laboratoire de Biologie des Ligneux et des Grandes Cultures (LBLGC), Plant Lignans Team, UPRES EA 1207, Université d'Orléans F 28000 Chartres France
- EA2106 Biomolecules et Biotechnologies Vegetales, Universite Francois-Rabelais de Tours Tours France
| |
Collapse
|
31
|
Lee CM, Park SH, Nam MJ. Anticarcinogenic effect of indole-3-carbinol (I3C) on human hepatocellular carcinoma SNU449 cells. Hum Exp Toxicol 2018; 38:136-147. [DOI: 10.1177/0960327118785235] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Many cruciferous vegetables, including cabbage, contain indole-3-carbinol (I3C), which is a known anticarcinogen. However, the anticarcinogenic effects of I3C on liver cancer have not been investigated. Therefore, this study was conducted to evaluate the anticarcinogenic effects of I3C in human hepatocellular carcinoma (HCC) SNU449 cells. The results of MTT and WST-1 assays indicated that treatment of SNU449 cells with I3C decreased viability in dose- and time-dependent manners, while colony formation assays indicated that I3C also inhibited proliferation of SNU449 cells. Moreover, fluorescence-activated cell sorter analysis showed that I3C induced apoptosis in SNU449 cells in dose- and time-dependent manners. Furthermore, terminal deoxynucleotidyl transferase deoxyuridine triphosphate nick-end labeling revealed that I3C induced DNA fragmentation in SNU449 cells in a time-dependent manner, while Western blotting showed that apoptotic proteins such as p53, cleaved PARP, caspase-3, and caspase-7 were activated in SNU449 cells following treatment with I3C. Finally, reactive oxygen species-related protein peroxiredoxin-1 and thioredoxin-1 expression decreased in I3C-treated SNU449 cells. The aim of our study is to investigate the unknown mechanisms responsible for the apoptotic effects of I3C on human HCC SNU449 cells, and the results suggest that I3C may be useful for the prevention and treatment of liver cancer.
Collapse
Affiliation(s)
- CM Lee
- Department of Biological Science, Gachon University, Seongnam, Republic of Korea
| | - S-H Park
- Department of Biological and Chemical Engineering, Hongik University, Seoul, Republic of Korea
| | - MJ Nam
- Department of Biological Science, Gachon University, Seongnam, Republic of Korea
| |
Collapse
|
32
|
Wu M, Zhao G, Zhuang X, Zhang T, Zhang C, Zhang W, Zhang Z. Triclosan treatment decreased the antitumor effect of sorafenib on hepatocellular carcinoma cells. Onco Targets Ther 2018; 11:2945-2954. [PMID: 29849464 PMCID: PMC5965385 DOI: 10.2147/ott.s165436] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Background Triclosan is a widely applied antimicrobial agent which affects the endocrine system and homeostasis; it may also promote the cirrhosis and hepatocellular carcinoma (HCC) growth in a mice model. The exact roles of triclosan in regulating human hepatocellular carcinoma development and treatment remain unknown. Methods MHCC97-H, a highly aggressive HCC cell line, was treated with indicated concentration of triclosan or sorafenib. The expression of drug-resistance genes was examined by qPCR. The clearance or metabolism of sorafenib was determined by liquid chromatograph-mass spectrometer/mass spectrometer (LC-MS/MS). MTT assay was used to examine the MHCC97-H cell proliferation. Nude mice were used to exam the anti-tumor effect of sorafenib on subcutaneous and intrahepatic growth of MHCC97-H cells. Results In the present study, triclosan could induce the expression of drug-resistance genes in MHCC97-H cells (a highly aggressive HCC cell line), accelerate the clearance of sorafenib, and attenuate the anti-proliferation effect of this molecular targeted agent in MHCC97-H cells. Triclosan decreased the antitumor effect of sorafenib on subcutaneous and intrahepatic growth of MHCC97-H in nude mice. Conclusion By discovering the fact that triclosan treatment enhances sorafenib resistance in HCC cells, this work suggests exposure of triclosan is detrimental to HCC patients during chemotherapy.
Collapse
Affiliation(s)
- Man Wu
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing, China.,Department of Pharmacy, The 309th Hospital of PLA, Beijing, China
| | - Guanren Zhao
- Department of Pharmacy, The 309th Hospital of PLA, Beijing, China
| | - Xiaomei Zhuang
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing, China
| | - Tianhong Zhang
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing, China
| | - Ce Zhang
- Department of Pharmacy, The 309th Hospital of PLA, Beijing, China
| | - Wenpeng Zhang
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing, China
| | - Zhenqing Zhang
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing, China
| |
Collapse
|
33
|
Kim BG, Kim JW, Kim SM, Go RE, Hwang KA, Choi KC. 3,3'-Diindolylmethane Suppressed Cyprodinil-Induced Epithelial-Mesenchymal Transition and Metastatic-Related Behaviors of Human Endometrial Ishikawa Cells via an Estrogen Receptor-Dependent Pathway. Int J Mol Sci 2018; 19:ijms19010189. [PMID: 29316692 PMCID: PMC5796138 DOI: 10.3390/ijms19010189] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2017] [Revised: 01/02/2018] [Accepted: 01/05/2018] [Indexed: 01/10/2023] Open
Abstract
Cyprodinil (CYP) is a pyrimidine amine fungicide that has been extensively used in agricultural areas. 3,3′-Diindolylmethane (DIM) is a derivative of the dietary phytoestrogen, indole-3-carbinol (I3C), which is derived from cruciferous vegetables and considered to be a cancer-preventive phytonutrient agent. In this study, the effects of CYP and DIM were examined on the cell viability, invasion, and metastasis of human endometrial cancer cells, Ishikawa, via epithelial mesenchymal transition (EMT). CYP increased the level of cell viability of Ishikawa cells compared to DMSO as a control, as did E2. Ishikawa cells lost cell-to-cell contact and obtained a spindle-shaped or fibroblast-like morphology in response to the application of E2 or CYP by the cell morphology assay. In the cell migration and invasion assay, CYP enhanced the ability of migration and invasion of Ishikawa cells, as did E2. E2 and CYP increased the expressions of N-cadherin and Snail proteins, while decreasing the expression of E-cadherin protein as EMT-related markers. In addition, E2 and CYP increased the protein expressions of cathepsin D and MMP-9, metastasis-related markers. Conversely, CYP-induced EMT, cell migration, and invasion were reversed by fulvestrant (ICI 182,780) as an estrogen receptor (ER) antagonist, indicating that CYP exerts estrogenic activity by mediating these processes via an ER-dependent pathway. Similar to ICI 182,780, DIM significantly suppressed E2 and CYP-induced proliferation, EMT, migration, and invasion of Ishikawa cancer cells. Overall, the present study revealed that DIM has an antiestrogenic chemopreventive effect to withdraw the cancer-enhancing effect of E2 and CYP, while CYP has the capacity to enhance the metastatic potential of estrogen-responsive endometrial cancer.
Collapse
Affiliation(s)
- Bo-Gyoung Kim
- Laboratory of Biochemistry and Immunology, College of Veterinary Medicine, Chungbuk National University, Cheongju 28644, Chungbuk, Korea.
| | - Jin-Wook Kim
- Laboratory of Biochemistry and Immunology, College of Veterinary Medicine, Chungbuk National University, Cheongju 28644, Chungbuk, Korea.
| | - Soo-Min Kim
- Laboratory of Biochemistry and Immunology, College of Veterinary Medicine, Chungbuk National University, Cheongju 28644, Chungbuk, Korea.
| | - Ryeo-Eun Go
- Laboratory of Biochemistry and Immunology, College of Veterinary Medicine, Chungbuk National University, Cheongju 28644, Chungbuk, Korea.
| | - Kyung-A Hwang
- Laboratory of Biochemistry and Immunology, College of Veterinary Medicine, Chungbuk National University, Cheongju 28644, Chungbuk, Korea.
| | - Kyung-Chul Choi
- Laboratory of Biochemistry and Immunology, College of Veterinary Medicine, Chungbuk National University, Cheongju 28644, Chungbuk, Korea.
| |
Collapse
|