1
|
Venkatesan J, Murugan D, Lakshminarayanan K, Smith AR, Vasanthakumari Thirumalaiswamy H, Kandhasamy H, Zender B, Zheng G, Rangasamy L. Powering up targeted protein degradation through active and passive tumour-targeting strategies: Current and future scopes. Pharmacol Ther 2024; 263:108725. [PMID: 39322067 DOI: 10.1016/j.pharmthera.2024.108725] [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: 06/01/2024] [Revised: 08/31/2024] [Accepted: 09/16/2024] [Indexed: 09/27/2024]
Abstract
Targeted protein degradation (TPD) has emerged as a prominent and vital strategy for therapeutic intervention of cancers and other diseases. One such approach involves the exploration of proteolysis targeting chimeras (PROTACs) for the selective elimination of disease-causing proteins through the innate ubiquitin-proteasome pathway. Due to the unprecedented achievements of various PROTAC molecules in clinical trials, researchers have moved towards other physiological protein degradation approaches for the targeted degradation of abnormal proteins, including lysosome-targeting chimeras (LYTACs), autophagy-targeting chimeras (AUTACs), autophagosome-tethering compounds (ATTECs), molecular glue degraders, and other derivatives for their precise mode of action. Despite numerous advantages, these molecules face challenges in solubility, permeability, bioavailability, and potential off-target or on-target off-tissue effects. Thus, an urgent need arises to direct the action of these degrader molecules specifically against cancer cells, leaving the proteins of non-cancerous cells intact. Recent advancements in TPD have led to innovative delivery methods that ensure the degraders are delivered in a cell- or tissue-specific manner to achieve cell/tissue-selective degradation of target proteins. Such receptor-specific active delivery or nano-based passive delivery of the PROTACs could be achieved by conjugating them with targeting ligands (antibodies, aptamers, peptides, or small molecule ligands) or nano-based carriers. These techniques help to achieve precise delivery of PROTAC payloads to the target sites. Notably, the successful entry of a Degrader Antibody Conjugate (DAC), ORM-5029, into a phase 1 clinical trial underscores the therapeutic potential of these conjugates, including LYTAC-antibody conjugates (LACs) and aptamer-based targeted protein degraders. Further, using bispecific antibody-based degraders (AbTACs) and delivering the PROTAC pre-fused with E3 ligases provides a solution for cell type-specific protein degradation. Here, we highlighted the current advancements and challenges associated with developing new tumour-specific protein degrader approaches and summarized their potential as single agents or combination therapeutics for cancer.
Collapse
Affiliation(s)
- Janarthanan Venkatesan
- Department of Chemistry, School of Advanced Sciences (SAS), Vellore Institute of Technology (VIT), Vellore 632014, India; Drug Discovery Unit (DDU), Centre for Biomaterials, Cellular and Molecular Theranostics (CBCMT), Vellore Institute of Technology (VIT), Vellore 632014, India
| | - Dhanashree Murugan
- Drug Discovery Unit (DDU), Centre for Biomaterials, Cellular and Molecular Theranostics (CBCMT), Vellore Institute of Technology (VIT), Vellore 632014, India; School of Biosciences and Technology (SBST), Vellore Institute of Technology (VIT), Vellore 632014, India
| | - Kalaiarasu Lakshminarayanan
- Department of Chemistry, School of Advanced Sciences (SAS), Vellore Institute of Technology (VIT), Vellore 632014, India; Drug Discovery Unit (DDU), Centre for Biomaterials, Cellular and Molecular Theranostics (CBCMT), Vellore Institute of Technology (VIT), Vellore 632014, India
| | - Alexis R Smith
- Department of Medicinal Chemistry, College of Pharmacy, University of Florida, Gainesville, FL 32610, USA; University of Florida Health Cancer Center, University of Florida, Gainesville, FL 32610, USA
| | - Harashkumar Vasanthakumari Thirumalaiswamy
- Department of Chemistry, School of Advanced Sciences (SAS), Vellore Institute of Technology (VIT), Vellore 632014, India; Drug Discovery Unit (DDU), Centre for Biomaterials, Cellular and Molecular Theranostics (CBCMT), Vellore Institute of Technology (VIT), Vellore 632014, India
| | - Hariprasath Kandhasamy
- Department of Chemistry, School of Advanced Sciences (SAS), Vellore Institute of Technology (VIT), Vellore 632014, India
| | - Boutheina Zender
- Department of Biomedical Engineering, Bahçeşehir University, Istanbul 34353, Turkey
| | - Guangrong Zheng
- Department of Medicinal Chemistry, College of Pharmacy, University of Florida, Gainesville, FL 32610, USA; University of Florida Health Cancer Center, University of Florida, Gainesville, FL 32610, USA.
| | - Loganathan Rangasamy
- Drug Discovery Unit (DDU), Centre for Biomaterials, Cellular and Molecular Theranostics (CBCMT), Vellore Institute of Technology (VIT), Vellore 632014, India.
| |
Collapse
|
2
|
Verhoog NJD, Spies LML. The anti-aromatase and anti-estrogenic activity of plant products in the treatment of estrogen receptor-positive breast cancer. J Steroid Biochem Mol Biol 2024; 243:106581. [PMID: 38997071 DOI: 10.1016/j.jsbmb.2024.106581] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/09/2023] [Revised: 06/14/2024] [Accepted: 07/09/2024] [Indexed: 07/14/2024]
Abstract
Despite being the focal point of decades of research, female breast cancer (BC) continues to be one of the most lethal cancers in the world. Given that 80 % of all diagnosed BC cases are estrogen receptor-positive (ER+) with carcinogenesis driven by estrogen-ERα signalling, current standard of care (SOC) hormone therapies are geared towards modulating the function and expression levels of estrogen and its receptors, ERα and ERβ. Currently, aromatase inhibitors (AIs), selective ER modulators (SERMs) and selective ER degraders (SERDs) are clinically prescribed for the management and treatment of ER+ BC, with the anti-aromatase activity of AIs abrogating estrogen biosynthesis, while the anti-estrogenic SERMs and SERDs antagonise and degrade the ER, respectively. The use of SOC hormone therapies is, however, significantly hampered by the onset of severe side-effects and the development of resistance. Given that numerous studies have reported on the beneficial effects of plant compounds and/or extracts and the multiple pathways through which they target ER+ breast carcinogenesis, recent research has focused on the use of dietary chemopreventive agents for BC management. When combined with SOC treatments, several of these plant components and/or extracts have demonstrated improved efficacy and/or synergistic impact. Moreover, despite a lack of in vivo investigations, plant products are generally reported to have a lower side-effect profile than SOC therapies and are therefore thought to be a safer therapeutic choice. Thus, the current review summarizes the findings from the last five years regarding the anti-aromatase and anti-estrogenic activity of plant products, as well as their synergistic anti-ER+ BC effects in combination with SOC therapies.
Collapse
Affiliation(s)
| | - Lee-Maine Lorin Spies
- Department of Biochemistry, Stellenbosch University, Van de Byl Street, Stellenbosch, 7601, South Africa
| |
Collapse
|
3
|
Maldonado J, Oliva A, Guzmán L, Molinari A, Acevedo W. Synthesis, Anticancer Activity, and Docking Studies of Novel Hydroquinone-Chalcone-Pyrazoline Hybrid Derivatives. Int J Mol Sci 2024; 25:7281. [PMID: 39000394 PMCID: PMC11242894 DOI: 10.3390/ijms25137281] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2024] [Revised: 06/20/2024] [Accepted: 06/30/2024] [Indexed: 07/16/2024] Open
Abstract
A novel series of antitumor hybrids was synthesized using 1,4-benzohydroquinone and chalcone, furane, or pyrazoline scaffolds. This were achieved through isosteric substitution of the aryl group of the chalcone β-carbon with the furanyl moiety and structural modification of the α,β-unsaturated carbonyl system. The potential antitumor activity of these hybrids was evaluated in vivo on MCF-7 breast adenocarcinoma and HT-29 colorectal carcinoma cells, demonstrating cytotoxic activity with IC50 values ranging from 28.8 to 124.6 µM. The incorporation of furan and pyrazoline groups significantly enhanced antiproliferative properties compared to their analogues and precursors (VII-X), which were inactive against both neoplastic cell lines. Compounds 4, 5, and 6 exhibited enhanced cytotoxicity against both cell lines, whereas compound 8 showed higher cytotoxic activity against HT-29 cells. Molecular docking studies revealed superior free-energy values (ΔGbin) for carcinogenic pathway-involved kinase proteins, with our in silico data suggesting that these derivatives could be promising chemotherapeutic agents targeting kinase pathways. Among all the synthesized PIBHQ compounds, derivatives 7 and 8 exhibited the best drug-likeness properties, with values of 0.53 and 0.83, respectively. ADME results collectively suggest that most of these compounds hold promise as potential candidates for preclinical assays.
Collapse
Affiliation(s)
- Javier Maldonado
- Instituto de Química, Facultad de Ciencias, Pontificia Universidad Católica de Valparaíso, Valparaíso 23732223, Chile
| | - Alfonso Oliva
- Instituto de Química, Facultad de Ciencias, Pontificia Universidad Católica de Valparaíso, Valparaíso 23732223, Chile
| | - Leda Guzmán
- Instituto de Química, Facultad de Ciencias, Pontificia Universidad Católica de Valparaíso, Valparaíso 23732223, Chile
| | - Aurora Molinari
- Instituto de Química, Facultad de Ciencias, Pontificia Universidad Católica de Valparaíso, Valparaíso 23732223, Chile
| | - Waldo Acevedo
- Instituto de Química, Facultad de Ciencias, Pontificia Universidad Católica de Valparaíso, Valparaíso 23732223, Chile
| |
Collapse
|
4
|
Xin L, Wang C, Cheng Y, Wang H, Guo X, Deng X, Deng X, Xie B, Hu H, Min C, Dong C, Zhou HB. Discovery of Novel ERα and Aromatase Dual-Targeting PROTAC Degraders to Overcome Endocrine-Resistant Breast Cancer. J Med Chem 2024; 67:8913-8931. [PMID: 38809993 DOI: 10.1021/acs.jmedchem.4c00196] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/31/2024]
Abstract
Estrogen receptor α (ERα) plays a pivotal role in the proliferation, differentiation, and migration of breast cancer (BC) cells, and aromatase (ARO) is a crucial enzyme in estrogen synthesis. Hence, it is necessary to inhibit estrogen production or the activity of ERα for the treatment of estrogen receptor-positive (ER+) BC. Herein, we present a new category of dual-targeting PROTAC degraders designed to specifically target ERα and ARO. Among them, compound 18c bifunctionally degrades and inhibits ERα/ARO, thus effectively suppressing the proliferation of MCF-7 cells while showing negligible cytotoxicity to normal cells. In vivo, 18c promotes the degradation of ERα and ARO and inhibits the growth of MCF-7 xenograft tumors. Finally, compound 18c demonstrates promising antiproliferative and ERα degradation activity against the ERαMUT cells. These findings suggest that 18c, being the inaugural dual-targeting degrader for ERα and ARO, warrants further advancement for the management of BC and the surmounting of endocrine resistance.
Collapse
Affiliation(s)
- Lilan Xin
- Department of Hematology, Zhongnan Hospital of Wuhan University, School of Pharmaceutical Sciences, Wuhan University, Wuhan 430071, China
| | - Chao Wang
- Department of Hematology, Zhongnan Hospital of Wuhan University, School of Pharmaceutical Sciences, Wuhan University, Wuhan 430071, China
| | - Yan Cheng
- Department of Hematology, Zhongnan Hospital of Wuhan University, School of Pharmaceutical Sciences, Wuhan University, Wuhan 430071, China
| | - Hongli Wang
- Department of Hematology, Zhongnan Hospital of Wuhan University, School of Pharmaceutical Sciences, Wuhan University, Wuhan 430071, China
| | - Xinyi Guo
- Department of Hematology, Zhongnan Hospital of Wuhan University, School of Pharmaceutical Sciences, Wuhan University, Wuhan 430071, China
| | - Xiaofei Deng
- Department of Hematology, Zhongnan Hospital of Wuhan University, School of Pharmaceutical Sciences, Wuhan University, Wuhan 430071, China
| | - Xiangping Deng
- Department of Hematology, Zhongnan Hospital of Wuhan University, School of Pharmaceutical Sciences, Wuhan University, Wuhan 430071, China
| | - Baohua Xie
- Department of Hematology, Zhongnan Hospital of Wuhan University, School of Pharmaceutical Sciences, Wuhan University, Wuhan 430071, China
| | - Hankun Hu
- Department of Pharmacy, Zhongnan Hospital of Wuhan University, School of Pharmaceutical Sciences, Wuhan University, Wuhan 430071, China
| | - Chang Min
- Department of Hematology, Zhongnan Hospital of Wuhan University, School of Pharmaceutical Sciences, Wuhan University, Wuhan 430071, China
| | - Chune Dong
- Department of Hematology, Zhongnan Hospital of Wuhan University, School of Pharmaceutical Sciences, Wuhan University, Wuhan 430071, China
| | - Hai-Bing Zhou
- Department of Hematology, Zhongnan Hospital of Wuhan University, School of Pharmaceutical Sciences, Wuhan University, Wuhan 430071, China
- Frontier Science Center for Immunology and Metabolism, State Key Laboratory of Virology, Provincial Key Laboratory of Developmentally Originated Disease, Key Laboratory of Combinatorial Biosynthesis and Drug Discovery (MOE) and Hubei Province Engineering and Technology Research Center for Fluorinated Pharmaceuticals, Wuhan University, Wuhan 430071, China
| |
Collapse
|
5
|
Huber D, Hatzipanagiotou M, Schüler-Toprak S, Ortmann O, Treeck O. Effects of Endocrine Interventions Targeting ERα or PR on Breast Cancer Risk in the General Population and Carriers of BRCA1/2 Pathogenic Variants. Int J Mol Sci 2024; 25:5894. [PMID: 38892081 PMCID: PMC11172552 DOI: 10.3390/ijms25115894] [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: 04/02/2024] [Revised: 05/17/2024] [Accepted: 05/25/2024] [Indexed: 06/21/2024] Open
Abstract
There is evidence suggesting that endocrine interventions such as hormone replacement therapy and hormonal contraception can increase breast cancer (BC) risk. Sexual steroid hormones like estrogens have long been known for their adverse effects on BC development and progression via binding to estrogen receptor (ER) α. Thus, in recent years, endocrine interventions that include estrogens have been discussed more and more critically, and their impact on different BC subgroups has increasingly gained interest. Carriers of pathogenic variants in BRCA1/2 genes are known to have a high risk of developing BC and ovarian cancer. However, there remain open questions to what extent endocrine interventions targeting ERα or the progesterone receptor further increase cancer risk in this subgroup. This review article aims to provide an overview and update on the effects of endocrine interventions on breast cancer risk in the general population in comparison to BRCA1/2 mutation carriers. Finally, future directions of research are addressed, to further improve the understanding of the effects of endocrine interventions on high-risk pathogenic variant carriers.
Collapse
Affiliation(s)
- Deborah Huber
- Department of Gynecology and Obstetrics, University Medical Center Regensburg, 93053 Regensburg, Germany; (D.H.); (M.H.); (S.S.-T.); (O.O.)
- Department of Obstetrics and Gynecology, Technical University of Munich, 80333 Munich, Germany
| | - Maria Hatzipanagiotou
- Department of Gynecology and Obstetrics, University Medical Center Regensburg, 93053 Regensburg, Germany; (D.H.); (M.H.); (S.S.-T.); (O.O.)
| | - Susanne Schüler-Toprak
- Department of Gynecology and Obstetrics, University Medical Center Regensburg, 93053 Regensburg, Germany; (D.H.); (M.H.); (S.S.-T.); (O.O.)
| | - Olaf Ortmann
- Department of Gynecology and Obstetrics, University Medical Center Regensburg, 93053 Regensburg, Germany; (D.H.); (M.H.); (S.S.-T.); (O.O.)
| | - Oliver Treeck
- Department of Gynecology and Obstetrics, University Medical Center Regensburg, 93053 Regensburg, Germany; (D.H.); (M.H.); (S.S.-T.); (O.O.)
| |
Collapse
|
6
|
Stevanović MZ, Bekić SS, Petri ET, Ćelić AS, Jakimov DS, Sakač MN, Kuzminac IZ. Synthesis, in vitro and in silico anticancer evaluation of novel pyridin-2-yl estra-1,3,5(10)-triene derivatives. Future Med Chem 2024; 16:1127-1145. [PMID: 38629440 PMCID: PMC11221553 DOI: 10.4155/fmc-2024-0039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2024] [Accepted: 03/26/2024] [Indexed: 06/26/2024] Open
Abstract
Aim: The aim of this study was the synthesis of steroid compounds with heterocyclic rings and good anticancer properties. Materials & methods: The synthesis, in silico and in vitro anticancer testing of novel pyridin-2-yl estra-1,3,5(10)-triene derivatives was performed. Results: All synthesized compounds have shown promising results for, antiproliferative activity, relative binding affinities for the ligand binding domains of estrogen receptors α, β and androgen receptor, aromatase binding potential, and inhibition of AKR1C3 enzyme. Conclusion: 3-Benzyloxy (17E)-pycolinilidene derivative 9 showed the best antitumor potential against MDA-MB-231 cell line, an activity that can be explained by its moderate inhibition of AKR1C3. Molecular docking simulation indicates that it binds to AKR1C3 in a very similar orientation and geometry as steroidal inhibitor EM1404.
Collapse
Affiliation(s)
- Milica Z Stevanović
- Department of Chemistry, Biochemistry & Environmental Protection, University of Novi Sad, Faculty of Sciences, Trg Dositeja Obradovića 3, 21000 Novi Sad, Serbia
| | - Sofija S Bekić
- Department of Chemistry, Biochemistry & Environmental Protection, University of Novi Sad, Faculty of Sciences, Trg Dositeja Obradovića 3, 21000 Novi Sad, Serbia
| | - Edward T Petri
- Department of Biology & Ecology, University of Novi Sad, Faculty of Sciences, Trg Dositeja Obradovića 2, 21000 Novi Sad, Serbia
| | - Andjelka S Ćelić
- Department of Biology & Ecology, University of Novi Sad, Faculty of Sciences, Trg Dositeja Obradovića 2, 21000 Novi Sad, Serbia
| | - Dimitar S Jakimov
- Oncology Institute of Vojvodina, Faculty of Medicine, University of Novi Sad, Put Dr Goldmana 4, 21204 Sremska Kamenica, Serbia
| | - Marija N Sakač
- Department of Chemistry, Biochemistry & Environmental Protection, University of Novi Sad, Faculty of Sciences, Trg Dositeja Obradovića 3, 21000 Novi Sad, Serbia
| | - Ivana Z Kuzminac
- Department of Chemistry, Biochemistry & Environmental Protection, University of Novi Sad, Faculty of Sciences, Trg Dositeja Obradovića 3, 21000 Novi Sad, Serbia
| |
Collapse
|
7
|
Xiong S, Song K, Xiang H, Luo G. Dual-target inhibitors based on ERα: Novel therapeutic approaches for endocrine resistant breast cancer. Eur J Med Chem 2024; 270:116393. [PMID: 38588626 DOI: 10.1016/j.ejmech.2024.116393] [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: 05/25/2023] [Revised: 04/04/2024] [Accepted: 04/04/2024] [Indexed: 04/10/2024]
Abstract
Estrogen receptor alpha (ERα), a nuclear transcription factor, is a well-validated therapeutic target for more than 70% of all breast cancers (BCs). Antagonizing ERα either by selective estrogen receptor modulators (SERMs) or selective estrogen receptor degraders (SERDs) forms the foundation of endocrine therapy and has achieved great success in the treatment of ERα positive (ERα+) BCs. Unfortunately, despite initial effectiveness, endocrine resistance eventually emerges in up to 30% of ERα+ BC patients and remains a significant medical challenge. Several mechanisms implicated in endocrine resistance have been extensively studied, including aberrantly activated growth factor receptors and downstream signaling pathways. Hence, the crosstalk between ERα and another oncogenic signaling has led to surge of interest to develop combination therapies and dual-target single agents. This review briefly introduces the synergisms between ERα and another anticancer target and summarizes the recent advances of ERα-based dual-targeting inhibitors from a medicinal chemistry perspective. Accordingly, their rational design strategies, structure-activity relationships (SARs) and biological activities are also dissected to provide some perspectives on future directions for ERα-based dual target drug discovery in BC therapy.
Collapse
Affiliation(s)
- Shuangshuang Xiong
- Jiangsu Key Laboratory of Drug Design and Optimization, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, 210009, China; Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing, 210009, China
| | - Ke Song
- Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing, 210009, China
| | - Hua Xiang
- Jiangsu Key Laboratory of Drug Design and Optimization, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, 210009, China; Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing, 210009, China.
| | - Guoshun Luo
- Jiangsu Key Laboratory of Drug Design and Optimization, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, 210009, China; Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing, 210009, China.
| |
Collapse
|
8
|
Meyer C, Brockmueller A, Buhrmann C, Shakibaei M. Prevention and Co-Management of Breast Cancer-Related Osteoporosis Using Resveratrol. Nutrients 2024; 16:708. [PMID: 38474838 DOI: 10.3390/nu16050708] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2024] [Revised: 02/21/2024] [Accepted: 02/26/2024] [Indexed: 03/14/2024] Open
Abstract
Breast cancer (BC) is currently one of the most common cancers in women worldwide with a rising tendency. Epigenetics, generally inherited variations in gene expression that occur independently of changes in DNA sequence, and their disruption could be one of the main causes of BC due to inflammatory processes often associated with different lifestyle habits. In particular, hormone therapies are often indicated for hormone-positive BC, which accounts for more than 50-80% of all BC subtypes. Although the cure rate in the early stage is more than 70%, serious negative side effects such as secondary osteoporosis (OP) due to induced estrogen deficiency and chemotherapy are increasingly reported. Approaches to the management of secondary OP in BC patients comprise adjunctive therapy with bisphosphonates, non-steroidal anti-inflammatory drugs (NSAIDs), and cortisone, which partially reduce bone resorption and musculoskeletal pain but which are not capable of stimulating the necessary intrinsic bone regeneration. Therefore, there is a great therapeutic need for novel multitarget treatment strategies for BC which hold back the risk of secondary OP. In this review, resveratrol, a multitargeting polyphenol that has been discussed as a phytoestrogen with anti-inflammatory and anti-tumor effects at the epigenetic level, is presented as a potential adjunct to both support BC therapy and prevent osteoporotic risks by positively promoting intrinsic regeneration. In this context, resveratrol is also known for its unique role as an epigenetic modifier in the regulation of essential signaling processes-both due to its catabolic effect on BC and its anabolic effect on bone tissue.
Collapse
Affiliation(s)
- Christine Meyer
- Chair of Vegetative Anatomy, Institute of Anatomy, Faculty of Medicine, LMU Munich, Pettenkoferstr. 11, 80336 Munich, Germany
| | - Aranka Brockmueller
- Chair of Vegetative Anatomy, Institute of Anatomy, Faculty of Medicine, LMU Munich, Pettenkoferstr. 11, 80336 Munich, Germany
| | - Constanze Buhrmann
- Institute of Anatomy and Cell Biology, Faculty of Medicine, University of Augsburg, 86159 Augsburg, Germany
| | - Mehdi Shakibaei
- Chair of Vegetative Anatomy, Institute of Anatomy, Faculty of Medicine, LMU Munich, Pettenkoferstr. 11, 80336 Munich, Germany
| |
Collapse
|
9
|
Li J, Yu J, Zou H, Zhang J, Ren L. Estrogen receptor-mediated health benefits of phytochemicals: a review. Food Funct 2023; 14:10681-10699. [PMID: 38047630 DOI: 10.1039/d3fo04702d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/05/2023]
Abstract
Estrogen receptors (ERs) are transcription factors with two subtypes: estrogen receptor alpha (ERα) and estrogen receptor beta (ERβ), which are essential for the maintenance of human health and play a regulatory role in common diseases such as breast cancer, osteoporosis, neurodegenerative disorders, liver injuries and lung cancers. A number of phytochemicals extracted from various fruits and vegetables have been demonstrated to exhibit estrogenic effects and are termed phytoestrogens. As modulators of ERs, phytoestrogens can be involved in the prevention and treatment of multiple diseases as complementary or alternative therapeutic agents and have a variety of health benefits for humans. This article reviews the health benefits of phytoestrogens in clinical and epidemiologic studies for several diseases and also provides a detailed description of the molecular mechanisms of their action. A brief comparison of the advantages and disadvantages of natural phytochemicals compared to synthetic drugs is also presented. The role of phytoestrogens in the treatment of diseases and human health requires further research to fully realize their therapeutic potential.
Collapse
Affiliation(s)
- Junfeng Li
- College of Food Science and Engineering, Jilin University, Changchun 130062, China.
| | - Jia Yu
- College of Food Science and Engineering, Jilin University, Changchun 130062, China.
| | - Haoyang Zou
- College of Food Science and Engineering, Jilin University, Changchun 130062, China.
| | - Jie Zhang
- College of Food Science and Engineering, Jilin University, Changchun 130062, China.
| | - Li Ren
- College of Food Science and Engineering, Jilin University, Changchun 130062, China.
| |
Collapse
|
10
|
Yue Z, He S, Wang J, Jiang Q, Wang H, Wu J, Li C, Wang Z, He X, Jia N. Glyceollins from soybean: Their pharmacological effects and biosynthetic pathways. Heliyon 2023; 9:e21874. [PMID: 38034638 PMCID: PMC10682181 DOI: 10.1016/j.heliyon.2023.e21874] [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: 04/15/2023] [Revised: 10/30/2023] [Accepted: 10/31/2023] [Indexed: 12/02/2023] Open
Abstract
Flavonoids are a highly abundant class of secondary metabolites present in plants. Isoflavonoids, in particular, are primarily synthesized in leguminous plants within the subfamily Papilionoideae. Numerous reports have established the favorable role of isoflavonoids in preventing a range of human diseases. Among the isoflavonoid components, glyceollins are synthesized specifically in soybean plants and have displayed promising effects in mitigating the occurrence and progression of breast and ovarian cancers as well as other diseases. Consequently, glyceollins have become a sought-after natural component for promoting women's health. In recent years, extensive research has focused on investigating the molecular mechanism underlying the preventative properties of glyceollins against various diseases. Substantial progress has also been made toward elucidating the biosynthetic pathway of glyceollins and exploring potential regulatory factors. Herein, we provide a review of the research conducted on glyceollins since their discovery five decades ago (1972-2023). We summarize their pharmacological effects, biosynthetic pathways, and advancements in chemical synthesis to enhance our understanding of the molecular mechanisms of their function and the genes involved in their biosynthetic pathway. Such knowledge may facilitate improved glyceollin synthesis and the creation of health products based on glyceollins.
Collapse
Affiliation(s)
- Zhiyong Yue
- School of Medicine, Xi'an International University, 18 Yudou Road, Yanta District, Xi'an Shaanxi, 710077, China
- Engineering Research Center of Personalized Anti-aging Health Product Development and Transformation, Universities of Shaanxi Province, 18 Yudou Road, Yanta District, Xi'an Shaanxi, 710077, China
| | - Shanhong He
- School of Medicine, Xi'an International University, 18 Yudou Road, Yanta District, Xi'an Shaanxi, 710077, China
| | - Jinpei Wang
- School of Medicine, Xi'an International University, 18 Yudou Road, Yanta District, Xi'an Shaanxi, 710077, China
- Engineering Research Center of Personalized Anti-aging Health Product Development and Transformation, Universities of Shaanxi Province, 18 Yudou Road, Yanta District, Xi'an Shaanxi, 710077, China
| | - Qi Jiang
- School of Medicine, Xi'an International University, 18 Yudou Road, Yanta District, Xi'an Shaanxi, 710077, China
- Engineering Research Center of Personalized Anti-aging Health Product Development and Transformation, Universities of Shaanxi Province, 18 Yudou Road, Yanta District, Xi'an Shaanxi, 710077, China
| | - Hanping Wang
- School of Medicine, Xi'an International University, 18 Yudou Road, Yanta District, Xi'an Shaanxi, 710077, China
- Engineering Research Center of Personalized Anti-aging Health Product Development and Transformation, Universities of Shaanxi Province, 18 Yudou Road, Yanta District, Xi'an Shaanxi, 710077, China
| | - Jia Wu
- School of Medicine, Xi'an International University, 18 Yudou Road, Yanta District, Xi'an Shaanxi, 710077, China
- Engineering Research Center of Personalized Anti-aging Health Product Development and Transformation, Universities of Shaanxi Province, 18 Yudou Road, Yanta District, Xi'an Shaanxi, 710077, China
| | - Chenxi Li
- School of Medicine, Xi'an International University, 18 Yudou Road, Yanta District, Xi'an Shaanxi, 710077, China
| | - Zixian Wang
- School of Medicine, Xi'an International University, 18 Yudou Road, Yanta District, Xi'an Shaanxi, 710077, China
| | - Xuan He
- School of Engineering, Xi'an International University, 18 Yudou Road, Yanta District, Xi'an Shaanxi, 710077, China
| | - Nannan Jia
- School of Medicine, Xi'an International University, 18 Yudou Road, Yanta District, Xi'an Shaanxi, 710077, China
| |
Collapse
|
11
|
Maldonado J, Oliva A, Molinari A, Acevedo W. 2-Acetyl-5,8-dihydro-6-(4-methyl-3-pentenyl)-1,4-naphthohydroquinone-Derived Chalcones as Potential Anticancer Agents. Molecules 2023; 28:7172. [PMID: 37894650 PMCID: PMC10609043 DOI: 10.3390/molecules28207172] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2023] [Revised: 09/29/2023] [Accepted: 10/01/2023] [Indexed: 10/29/2023] Open
Abstract
Based on previous results with benzoindazolequinone (BIZQ) and 3-methylnaphtho [2,3-d]isoxazole-4,9-quinone (NIQ) derivatives, a novel series of chalcone-1,4-naphthoquinone/benzohydroquinone (CNQ and CBHQ) compounds were synthesized from 2-acetyl-5,8-dihydro-6-(4-methyl-3-pentenyl)-1,4-naphthohydroquinone. Their structures were elucidated via spectroscopy. These hybrids were assessed in vivo for their antiproliferative activity on MCF-7 breast adenocarcinoma and HT-29 colorectal carcinoma cells, revealing cytotoxicity with IC50 values between 6.0 and 110.5 µM. CBHQ hybrids 5e and 5f displayed enhanced cytotoxicity against both cell lines, whereas CNQ hybrids 6a-c and 6e exhibited higher cytotoxic activity against MCF-7 cells. Docking studies showed strong binding energies (ΔGbin) of CNQs to kinase proteins involved in carcinogenic pathways. Furthermore, our in silico analysis of drug absorption, distribution, metabolism, and excretion (ADME) properties suggests their potential as candidates for cancer pre-clinical assays.
Collapse
Affiliation(s)
| | | | - Aurora Molinari
- Instituto de Química, Facultad de Ciencias, Pontificia Universidad Católica de Valparaíso, Valparaíso 23732223, Chile; (J.M.); (A.O.)
| | - Waldo Acevedo
- Instituto de Química, Facultad de Ciencias, Pontificia Universidad Católica de Valparaíso, Valparaíso 23732223, Chile; (J.M.); (A.O.)
| |
Collapse
|
12
|
Liu J, Zhang L, Tang M, Chen X, Yang C, Li Y, Feng J, Deng Y, Wang X, Zhang Y. Functional variant rs10175368 which affects the expression of CYP1B1 plays a protective role against breast cancer in a Chinese Han population. Eur J Cancer Prev 2023; 32:450-459. [PMID: 37038992 PMCID: PMC10373845 DOI: 10.1097/cej.0000000000000800] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Accepted: 03/10/2023] [Indexed: 04/12/2023]
Abstract
OBJECTIVE Cytochrome P450 1B1 ( CYP1B1 ) genetic variants are relevant in the pathogenesis of breast cancer. Exploring the relationships between CYP1B1 functional variants and breast cancer could improve our understanding of breast cancer molecular pathophysiology. METHODS This is a two-stage hospital-based case-control study of a Chinese Han population. Genotyping was performed to identify candidate gene variants. 3DSNP, ANNOVAR, and RegulomeDB were used to determine functional single nucleotide polymorphisms (SNPs). The relationship between candidate variants and breast cancer risk was evaluated through unconditional logistic regression analysis. The PancanQTL platform was used to perform cis and trans expression quantitative trait loci (eQTL) analysis of positive SNPs. The GSCA platform was then used to compare the gene expression levels of potential target genes between breast cancer tissue and normal tissue adjacent to the cancer. RESULTS rs10175368-T acted as a protective factor against breast cancer based on an additive model [odds ratio (OR) = 0.722, 95% confidence interval (CI) = 0.613-0.850; P < 0.001], and was identified as a protective factor in the postmenopausal population (OR = 0.601; 95% CI, 0.474-0.764; P < 0.001). eQTL analysis and analysis of differential expression in carcinoma and paracancerous tissues revealed that the expression level of CYP1B1 - AS1 was associated with rs10175368 and that CYP1B1-AS1 had significantly higher expression levels in breast cancer tissues than in paracancerous tissues. CONCLUSION We show, for the first time in a Chinese Han population, that the functional variant rs10175368 plays a protective role against breast cancer, especially in the postmenopausal population.
Collapse
Affiliation(s)
- Jiarui Liu
- Department of Hygiene Toxicology, School of Public Health, Zunyi Medical University
- Department of Preventive Medicine, Guizhou Province Preventive Medicine Experimental Teaching Demonstration Centre, Zunyi, Guizhou
| | - Lijia Zhang
- Department of Hygiene Toxicology, School of Public Health, Zunyi Medical University
- Department of Preventive Medicine, Guizhou Province Preventive Medicine Experimental Teaching Demonstration Centre, Zunyi, Guizhou
| | - Mingwen Tang
- Department of Hygiene Toxicology, School of Public Health, Zunyi Medical University
- Department of Preventive Medicine, Guizhou Province Preventive Medicine Experimental Teaching Demonstration Centre, Zunyi, Guizhou
| | - Xinyu Chen
- Department of Hygiene Toxicology, School of Public Health, Zunyi Medical University
- Department of Preventive Medicine, Guizhou Province Preventive Medicine Experimental Teaching Demonstration Centre, Zunyi, Guizhou
| | - Caiyun Yang
- Department of Hygiene Toxicology, School of Public Health, Zunyi Medical University
- Department of Preventive Medicine, Guizhou Province Preventive Medicine Experimental Teaching Demonstration Centre, Zunyi, Guizhou
| | - Yong Li
- Department of Oncology, GuiZhou Provincial People’s Hospital, Guiyang
| | - Jin Feng
- Department of Clinical Laboratory, Affiliated Hospital of Zunyi Medical University, Zunyi
| | - Yan Deng
- Department of Scientific Research and Education, The First People's Hospital of Bijie city, Bijie Guizhou, China
| | - Xin Wang
- Department of Orthopaedic Surgery, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou, China
| | - Yi Zhang
- Department of Hygiene Toxicology, School of Public Health, Zunyi Medical University
- Department of Preventive Medicine, Guizhou Province Preventive Medicine Experimental Teaching Demonstration Centre, Zunyi, Guizhou
| |
Collapse
|
13
|
Ajabnoor GMA. The Molecular and Genetic Interactions between Obesity and Breast Cancer Risk. MEDICINA (KAUNAS, LITHUANIA) 2023; 59:1338. [PMID: 37512149 PMCID: PMC10384495 DOI: 10.3390/medicina59071338] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/18/2023] [Revised: 07/14/2023] [Accepted: 07/19/2023] [Indexed: 07/30/2023]
Abstract
Breast cancer (BC) is considered the leading cause of death among females worldwide. Various risk factors contribute to BC development, such as age, genetics, reproductive factors, obesity, alcohol intake, and lifestyle. Obesity is considered to be a pandemic health problem globally, affecting millions of people worldwide. Obesity has been associated with a high risk of BC development. Determining the impact of obesity on BC development risk in women by demonstrating the molecular and genetic association in pre- and post-menopause females and risk to BC initiation is crucial in order to improve the diagnosis and prognosis of BC disease. In epidemiological studies, BC in premenopausal women was shown to be protective in a certain pattern. These altered effects between the two phases could be due to various physiological changes, such as estrogen/progesterone fluctuating levels. In addition, the relationship between BC risk and obesity is indicated by different molecular alterations as metabolic pathways and genetic mutation or epigenetic DNA changes supporting a strong connection between obesity and BC risk. However, these molecular and genetic alteration remain incompletely understood. The aim of this review is to highlight and elucidate the different molecular mechanisms and genetic changes occurring in obese women and their association with BC risk and development.
Collapse
Affiliation(s)
- Ghada M A Ajabnoor
- Department of Clinical Biochemistry, Faculty of Medicine, King Abdulaziz University, Jeddah 21589, Saudi Arabia
- Food, Nutrition and Lifestyle Unit, King Fahd Medical Research Centre, King Abdulaziz University, Jeddah 21551, Saudi Arabia
- Saudi Diabetes Research Group, Faculty of Medicine, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| |
Collapse
|
14
|
[Therapeutic strategies for the treatment of endocrine resistant hormone receptor positive advanced breast cancer]. Bull Cancer 2023; 110:69-87. [PMID: 36307325 DOI: 10.1016/j.bulcan.2022.09.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Revised: 09/10/2022] [Accepted: 09/13/2022] [Indexed: 11/06/2022]
Abstract
HR+ breast cancers are defined by the prominence of signaling pathways dependent on the estrogen receptor. Endocrine therapy is the standard treatment for these advanced diseases. Resistance to these treatments, called hormone resistance, appears invariably with biological mechanisms that have led to the development of therapeutic opportunities. An exhaustive literature review was carried out concerning the biology of the hormone resistance pathways, the therapeutic options before the era of CDK4/6 inhibitors, the rise of CDK4/6 inhibitors and the therapeutic prospects in a situation of hormone resistance. Various biological abnormalities have been identified in the mechanisms of hormone resistance such as changes in the estrogen receptor, mutations in the ESR1 gene, aberrant activation of the PI3K pathway or cell cycle deregulations. Historical strategies for circumventing this hormone resistance have been based on hormonal manipulation, on the development of new endocrine therapy such as fulvestrant (selective estrogen receptor inhibitor, SERD), on combinations of treatments such as everolimus, a mTOR inhibitor. This strategy combining endocrine therapy and targeted therapy has led to the development of combinations with CDK4/6 inhibitors which have now become a standard treatment in the hormone resistance phase. The future of this therapeutic era remains to be written with new combinations of hormone therapy and targeted therapy such as PI3K inhibitors or even with the positioning of new SERDs in clinical development.
Collapse
|
15
|
Furth PA, Wang W, Kang K, Rooney BL, Keegan G, Muralidaran V, Zou X, Flaws JA. Esr1 but Not CYP19A1 Overexpression in Mammary Epithelial Cells during Reproductive Senescence Induces Pregnancy-Like Proliferative Mammary Disease Responsive to Anti-Hormonals. THE AMERICAN JOURNAL OF PATHOLOGY 2023; 193:84-102. [PMID: 36464512 PMCID: PMC9768685 DOI: 10.1016/j.ajpath.2022.09.007] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/10/2022] [Revised: 08/22/2022] [Accepted: 09/16/2022] [Indexed: 12/04/2022]
Abstract
Molecular-level analyses of breast carcinogenesis benefit from vivo disease models. Estrogen receptor 1 (Esr1) and cytochrome P450 family 19 subfamily A member 1 (CYP19A1) overexpression targeted to mammary epithelial cells in genetically engineered mouse models induces largely similar rates of proliferative mammary disease in prereproductive senescent mice. Herein, with natural reproductive senescence, Esr1 overexpression compared with CYP19A1 overexpression resulted in significantly higher rates of preneoplasia and cancer. Before reproductive senescence, Esr1, but not CYP19A1, overexpressing mice are tamoxifen resistant. However, during reproductive senescence, Esr1 mice exhibited responsiveness. Both Esr1 and CYP19A1 are responsive to letrozole before and after reproductive senescence. Gene Set Enrichment Analyses of RNA-sequencing data sets showed that higher disease rates in Esr1 mice were accompanied by significantly higher expression of cell proliferation genes, including members of prognostic platforms for women with early-stage hormone receptor-positive disease. Tamoxifen and letrozole exposure induced down-regulation of these genes and resolved differences between the two models. Both Esr1 and CYP19A1 overexpression induced abnormal developmental patterns of pregnancy-like gene expression. This resolved with progression through reproductive senescence in CYP19A1 mice, but was more persistent in Esr1 mice, resolving only with tamoxifen and letrozole exposure. In summary, genetically engineered mouse models of Esr1 and CYP19A1 overexpression revealed a diversion of disease processes resulting from the two distinct molecular pathophysiological mammary gland-targeted intrusions into estrogen signaling during reproductive senescence.
Collapse
Affiliation(s)
- Priscilla A Furth
- Department of Oncology, Georgetown University, Washington, District of Columbia; Department of Medicine, Georgetown University, Washington, District of Columbia.
| | - Weisheng Wang
- Department of Oncology, Georgetown University, Washington, District of Columbia
| | - Keunsoo Kang
- Department of Microbiology, College of Science and Technology, Dankook University, Cheonan, Republic of Korea
| | - Brendan L Rooney
- Department of Oncology, Georgetown University, Washington, District of Columbia
| | - Grace Keegan
- Department of Oncology, Georgetown University, Washington, District of Columbia
| | - Vinona Muralidaran
- Department of Oncology, Georgetown University, Washington, District of Columbia
| | - Xiaojun Zou
- Department of Oncology, Georgetown University, Washington, District of Columbia
| | - Jodi A Flaws
- Department of Comparative Biosciences, University of Illinois Urbana-Champaign, Urbana, Illinois
| |
Collapse
|
16
|
Ardevines S, Marqués-López E, Herrera RP. Heterocycles in Breast Cancer Treatment: The Use of Pyrazole Derivatives. Curr Med Chem 2023; 30:1145-1174. [PMID: 36043746 PMCID: PMC11475274 DOI: 10.2174/0929867329666220829091830] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2021] [Revised: 04/21/2022] [Accepted: 05/05/2022] [Indexed: 11/22/2022]
Abstract
Among the aromatic heterocycle rings, pyrazole -a five-membered ring with two adjacent nitrogen atoms in its structure has been postulated as a potent candidate in the pharmacological context. This moiety is an interesting therapeutic target covering a broad spectrum of biological activities due to its presence in many natural substances. Hence, the potential of the pyrazole derivatives as antitumor agents has been explored in many investigations, showing promising results in some cases. In this sense, breast cancer, which is already the leading cause of cancer mortality in women in some countries, has been the topic selected for this review, which covers a range of different research from the earliest studies published in 2003 to the most recent ones in 2021.
Collapse
Affiliation(s)
- Sandra Ardevines
- Laboratorio de Organocatálisis Asimétrica, Departamento de Química Orgánica. Instituto de Síntesis Química y Catálisis Homogénea (ISQCH), CSIC-Universidad de Zaragoza. C/Pedro Cerbuna 12, E-50009 Zaragoza, Spain
| | - Eugenia Marqués-López
- Laboratorio de Organocatálisis Asimétrica, Departamento de Química Orgánica. Instituto de Síntesis Química y Catálisis Homogénea (ISQCH), CSIC-Universidad de Zaragoza. C/Pedro Cerbuna 12, E-50009 Zaragoza, Spain
| | - Raquel P. Herrera
- Laboratorio de Organocatálisis Asimétrica, Departamento de Química Orgánica. Instituto de Síntesis Química y Catálisis Homogénea (ISQCH), CSIC-Universidad de Zaragoza. C/Pedro Cerbuna 12, E-50009 Zaragoza, Spain
| |
Collapse
|
17
|
Zhang J, Xie M. Graph regularized non-negative matrix factorization with prior knowledge consistency constraint for drug-target interactions prediction. BMC Bioinformatics 2022; 23:564. [PMID: 36581822 PMCID: PMC9798666 DOI: 10.1186/s12859-022-05119-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2022] [Accepted: 12/20/2022] [Indexed: 12/30/2022] Open
Abstract
BACKGROUND Identifying drug-target interactions (DTIs) plays a key role in drug development. Traditional wet experiments to identify DTIs are expensive and time consuming. Effective computational methods to predict DTIs are useful to narrow the searching scope of potential drugs and speed up the process of drug discovery. There are a variety of non-negativity matrix factorization based methods to predict DTIs, but the convergence of the algorithms used in the matrix factorization are often overlooked and the results can be further improved. RESULTS In order to predict DTIs more accurately and quickly, we propose an alternating direction algorithm to solve graph regularized non-negative matrix factorization with prior knowledge consistency constraint (ADA-GRMFC). Based on known DTIs, drug chemical structures and target sequences, ADA-GRMFC at first constructs a DTI matrix, a drug similarity matrix and a target similarity matrix. Then DTI prediction is modeled as the non-negative factorization of the DTI matrix with graph dual regularization terms and a prior knowledge consistency constraint. The graph dual regularization terms are used to integrate the information from the drug similarity matrix and the target similarity matrix, and the prior knowledge consistency constraint is used to ensure the matrix decomposition result should be consistent with the prior knowledge of known DTIs. Finally, an alternating direction algorithm is used to solve the matrix factorization. Furthermore, we prove that the algorithm can converge to a stationary point. Extensive experimental results of 10-fold cross-validation show that ADA-GRMFC has better performance than other state-of-the-art methods. In the case study, ADA-GRMFC is also used to predict the targets interacting with the drug olanzapine, and all of the 10 highest-scoring targets have been accurately predicted. In predicting drug interactions with target estrogen receptors alpha, 17 of the 20 highest-scoring drugs have been validated.
Collapse
Affiliation(s)
- Junjun Zhang
- grid.411427.50000 0001 0089 3695Key Laboratory of Computing and Stochastic Mathematics (LCSM) (Ministry of Education), School of Mathematics and Statistics, Hunan Normal University, Changsha, 410081 China
| | - Minzhu Xie
- grid.411427.50000 0001 0089 3695Key Laboratory of Computing and Stochastic Mathematics (LCSM) (Ministry of Education), School of Mathematics and Statistics, Hunan Normal University, Changsha, 410081 China ,grid.411427.50000 0001 0089 3695College of Information Science and Engineering, Hunan Normal University, Changsha, 410081 China
| |
Collapse
|
18
|
Optimization Method of an Antibreast Cancer Drug Candidate Based on Machine Learning. COMPUTATIONAL AND MATHEMATICAL METHODS IN MEDICINE 2022; 2022:4133663. [PMID: 36105244 PMCID: PMC9467812 DOI: 10.1155/2022/4133663] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Revised: 08/17/2022] [Accepted: 08/22/2022] [Indexed: 12/03/2022]
Abstract
Breast cancer is a common but serious and even lethal disease. Fortunately, compared with other cancers, breast cancer treatments currently are relatively well developed. The use of specific drugs is typically essential in the majority of breast cancer treatment strategies. Given the aforementioned factors, it is important to continue researching effective antibreast cancer drug design. Machine learning-based computer-aided drug design is currently a common practice in both drug industries and academic institutes. According to the characteristics of breast cancer, we selected multiple candidate compounds; based on the corresponding molecular descriptors, biological activities, and pharmacokinetic properties, a dataset of inhibition potency and pharmacokinetic properties paired with multiple features of compounds was constructed. On this basis, the random forest method was utilized to choose greater-influenced feature embeddings; thus, 224 main operating variables were selected for further analysis; we then employed the efficient MobileNetV3 deep neural network as the backbone to establish the prediction models for the inhibition potency and pharmacokinetic properties of the compounds. After data preprocessing, the weights are obtained by training on the refined dataset. Finally, we define an optimization problem to discover compounds with the best properties. The problem is solved using the genetic algorithm with the acquired prediction model, and the solution value for the corresponding operating variables with the best clinical properties in theory is then obtained. Analysis demonstrates that our approach could be used to aid the screening process of antibreast cancer drug candidates.
Collapse
|
19
|
Wetzel EA, Marks KJ, Gleason AA, Brown-Ford S, Reid TE, Chaudhury S, Lindeman S, Sem DS, Donaldson WA. Discovery of two novel (4-hydroxyphenyl) substituted polycyclic carbocycles as potent and selective estrogen receptor beta agonists. Bioorg Med Chem Lett 2022; 73:128906. [PMID: 35870729 DOI: 10.1016/j.bmcl.2022.128906] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2022] [Revised: 07/12/2022] [Accepted: 07/17/2022] [Indexed: 11/02/2022]
Abstract
Two (4-hydroxyphenyl) substituted polycyclic carbocycles were prepared and assayed for estrogen receptor activity. 4-(4-Hydroxyphenyl)tricyclo[3.3.1.13,7]decane-1-methanol (5a/b) and 7-(4-hydroxyphenyl)spiro[3.5]nonan-2-ol ((±)-11) were found to be potent ERβ agonists (1.9 ± 0.4 nM and 6.2 ± 1.4 nM respectively) in a cell-based functional assay. Furthermore, both 5a/b and 11 were highly selective for ERβ over ERα (377 and 1,100-fold selective respectively). While neither compound inhibited CYP2D6 or CYP3A4 at concentrations up to 62.5 μM, 5a/b did have weak binding to CYP2C9 with an IC50 of 10 ± 0.5 μM. Computational assessment of 5a/b and 11 predicted the most probable site of metabolism would be ortho to the phenolic hydroxyl group.
Collapse
Affiliation(s)
- Edward A Wetzel
- Department of Chemistry, Marquette University, P. O. Box 1881, Milwaukee, WI 53201-1881, United States
| | - Kylee J Marks
- School of Pharmacy, Center for Structure-based Drug Design and Development, Concordia University Wisconsin, Mequon, WI 53097, United States
| | - Alexandra A Gleason
- School of Pharmacy, Center for Structure-based Drug Design and Development, Concordia University Wisconsin, Mequon, WI 53097, United States
| | - Sandra Brown-Ford
- School of Pharmacy, Center for Structure-based Drug Design and Development, Concordia University Wisconsin, Mequon, WI 53097, United States
| | - Terry-Elinor Reid
- School of Pharmacy, Center for Structure-based Drug Design and Development, Concordia University Wisconsin, Mequon, WI 53097, United States
| | - Subhabrata Chaudhury
- Department of Chemistry, Marquette University, P. O. Box 1881, Milwaukee, WI 53201-1881, United States
| | - Sergey Lindeman
- Department of Chemistry, Marquette University, P. O. Box 1881, Milwaukee, WI 53201-1881, United States
| | - Daniel S Sem
- School of Pharmacy, Center for Structure-based Drug Design and Development, Concordia University Wisconsin, Mequon, WI 53097, United States
| | - William A Donaldson
- Department of Chemistry, Marquette University, P. O. Box 1881, Milwaukee, WI 53201-1881, United States.
| |
Collapse
|
20
|
Vijayan S, Loganathan C, Sakayanathan P, Thayumanavan P. Synthesis and Characterization of Plumbagin S-Allyl Cysteine Ester: Determination of Anticancer Activity In Silico and In Vitro. Appl Biochem Biotechnol 2022; 194:5827-5847. [PMID: 35819687 DOI: 10.1007/s12010-022-04079-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2022] [Accepted: 07/05/2022] [Indexed: 11/29/2022]
Abstract
In recent years, derivatives of natural compounds are synthesized to increase the bioavailability, pharmacology, and pharmacokinetics properties. The naphthoquinone, plumbagin (PLU), is well known for its anticancer activity. However, the clinical use of PLU is hindered due to its toxicity. Previous reports have shown that modification of PLU at 5'-hydroxyl group has reduced its toxicity towards normal cell line. In accordance, in the present study, 5'-hydroxyl group of PLU was esterified with S-allyl cysteine (SAC) to obtain PLU-SAC ester. The drug-likeness of PLU-SAC was understood by in silico ADME analysis. PLU-SAC was characterized by UV-visible spectroscopy, mass spectroscopy, and nuclear magnetic resonance (NMR) spectroscopy. Molecular docking and dynamics simulation analysis revealed the interaction of PLU-SAC with proteins of interest in cancer therapy such as human estrogen receptor α, tumor protein p53 negative regulator mouse double minute 2, and cyclin-dependent kinase 2. MMGBSA calculation showed the favorable binding energy which in turn demonstrated the stable binding of PLU-SAC with these proteins. PLU-SAC showed apoptosis in breast cancer cell line (MCF-7) by inducing oxidative stress, disturbing mitochondrial function, arresting cells at G1 phase of cell cycle, and initiating DNA fragmentation. However, PLU-SAC did not show toxicity towards normal Vero cell line. PLU-SAC was synthesized and structurally characterized, and its anticancer activity was determined by in silico and in vitro analysis.
Collapse
Affiliation(s)
- Sudha Vijayan
- Department of Biochemistry, Periyar University, Salem, Tamil Nadu, 636011, India
| | - Chitra Loganathan
- Department of Biochemistry, Periyar University, Salem, Tamil Nadu, 636011, India.,Research and Development Center, Bioinnov Solutions LLP, Salem, Tamil Nadu, 636002, India
| | | | | |
Collapse
|
21
|
Lesser C, Mericq V, Reyes M, Garmendia ML, Shepherd JA, Michels KB, Corvalán C, Pereira A. Habitual Phytoestrogen Intake Is Associated with Breast Composition in Girls at 2 Years after Menarche Onset. Cancer Epidemiol Biomarkers Prev 2022; 31:1334-1340. [PMID: 35477112 PMCID: PMC9250624 DOI: 10.1158/1055-9965.epi-22-0016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Revised: 03/23/2022] [Accepted: 04/15/2022] [Indexed: 01/03/2023] Open
Abstract
BACKGROUND High phytoestrogen intake during adolescence is associated with a reduced risk of breast cancer. Breast density (BD) is a strong predictor of breast cancer and can be considered an early marker. We aim to assess the association between the mean habitual intake of isoflavones, lignans, and total phytoestrogens intake during puberty until 2 years after menarche onset and absolute fibroglandular volume (AFGV) and percentage of fibroglandular volume (%FGV) in Hispanic girls at the end of puberty. METHODS Longitudinal study set up in the Growth and Obesity Chilean Cohort Study (GOCS). We included 329 girls with dietary data (multiple 24-hours recalls) from puberty until 2 years after menarche onset (81% had 2-4 recalls). Two international datasets were used to estimate isoflavones, lignans, and total phytoestrogens in the diet. Breast composition was measured by dual energy X-ray absorptiometry at 2 years after menarche. Multiple linear regression models were used to assess the association between isoflavones, lignans, and total phytoestrogens intake and AFGV and %FGV. RESULTS The average total phytoestrogen intake was 1 mg/day and %FGV was 50.7% (SD = 15.2) and AFGV 218.8 cm3 (SD = 79.3). An inverse association was found between consumption of isoflavones and AFGV, as well as, with total phytoestrogens [Q4 vs. Q1 adjusted model ß = -49.2 cm3; 95% CI (-85.5 to -13.0)]. CONCLUSIONS Girls with a higher intake of total phytoestrogens and isoflavones during puberty until 2 years after menarche onset had significantly lower AFGV. IMPACT Although the intake of phytoestrogens is low in Western populations, higher consumption of them during a critical period of life like puberty could be beneficial to reduce breast cancer during adulthood.
Collapse
Affiliation(s)
- Constanza Lesser
- Institute of Nutrition and Food Technology, University of Chile, Santiago, Chile
| | - Verónica Mericq
- Institute of Maternal and Child Research (IDIMI), Faculty of Medicine, University of Chile, Santiago, Chile
| | - Marcela Reyes
- Institute of Nutrition and Food Technology, University of Chile, Santiago, Chile
| | | | - John A Shepherd
- Population Sciences in the Pacific Program (Cancer Epidemiology), University of Hawaiʻi Cancer Center, University of Hawaiʻi, Honolulu, HI, USA
| | - Karin B Michels
- Department of Epidemiology, Fielding School of Public Health, University of California, Los Angeles, USA.,Institute for Prevention and Cancer Epidemiology, Faculty of Medicine and Medical Center, University of Freiburg, Germany
| | - Camila Corvalán
- Institute of Nutrition and Food Technology, University of Chile, Santiago, Chile
| | - Ana Pereira
- Institute of Nutrition and Food Technology, University of Chile, Santiago, Chile
| |
Collapse
|
22
|
Choo SW, Zhong Y, Sendler E, Goustin AS, Cai J, Ju D, Kosir MA, Giordo R, Lipovich L. Estrogen distinctly regulates transcription and translation of lncRNAs and pseudogenes in breast cancer cells. Genomics 2022; 114:110421. [PMID: 35779786 DOI: 10.1016/j.ygeno.2022.110421] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Revised: 05/25/2022] [Accepted: 06/25/2022] [Indexed: 11/04/2022]
Abstract
Estrogen drives key transcriptional changes in breast cancer and stimulates breast cancer cells' growth with multiple mechanisms to coordinate transcription and translation. In addition to protein-coding transcripts, estrogen can regulate long non-coding RNA (lncRNA) transcripts, plus diverse non-coding RNAs including antisense, enhancer, and intergenic. LncRNA genes comprise the majority of human genes. The accidental, or regulated, translation of their short open reading frames by ribosomes remains a controversial topic. Here we report for the first time an integrated analysis of RNA abundance and ribosome occupancy level, using Ribo-seq combined with RNA-Seq, in the estrogen-responsive, estrogen receptor α positive, human breast cancer cell model MCF7, before and after hormone treatment. Translational profiling can determine, in an unbiased manner, which fraction of the genome is actually translated into proteins, as well as resolving whether transcription and translation respond concurrently, or differentially, to estrogen treatment. Our data showed specific transcripts more robustly detected in RNA-Seq than in the ribosome-profiling data, and vice versa, suggesting distinct gene-specific estrogen responses at the transcriptional and the translational level, respectively. Here, we showed that estrogen stimulation affects the expression levels of numerous lncRNAs, but not their association with ribosomes, and that most lncRNAs are not ribosome-bound. For the first time, we also demonstrated the transcriptional and translational response of expressed pseudogenes to estrogen, pointing to new perspectives for drug-target development in breast cancer in the future.
Collapse
Affiliation(s)
- Siew-Woh Choo
- Department of Biology, College of Science and Technology, Wenzhou-Kean University, 88 Daxue Road, Ouhai, Wenzhou, Zhejiang Province, China; Zhejiang Bioinformatics International Science and Technology Cooperation Center, Ouhai, Wenzhou, Zhejiang Province, China; Wenzhou Municipal Key Laboratory for Applied Biomedical and Biopharmaceutical Informatics, Ouhai, Wenzhou, Zhejiang Province, China.
| | - Yu Zhong
- Department of Biological Sciences, Xi'an Jiaotong-Liverpool University, Suzhou, China
| | - Edward Sendler
- Center for Molecular Medicine and Genetics, School of Medicine, Wayne State University, Detroit, USA
| | - Anton-Scott Goustin
- Center for Molecular Medicine and Genetics, School of Medicine, Wayne State University, Detroit, USA
| | - Juan Cai
- Department of Biochemistry, Microbiology and Immunology School of Medicine, Wayne State University, Detroit, USA
| | - Donghong Ju
- Center for Molecular Medicine and Genetics, School of Medicine, Wayne State University, Detroit, USA; Department of Surgery and Karmanos Cancer Institute, School of Medicine, Wayne State University, Detroit, USA
| | - Mary Ann Kosir
- Department of Surgery and Karmanos Cancer Institute, School of Medicine, Wayne State University, Detroit, USA
| | - Roberta Giordo
- College of Medicine, Mohammed Bin Rashid University of Medicine and Health Sciences, Dubai, United Arab Emirates.
| | - Leonard Lipovich
- College of Medicine, Mohammed Bin Rashid University of Medicine and Health Sciences, Dubai, United Arab Emirates.
| |
Collapse
|
23
|
Human Estrogen Receptor Alpha Antagonists, Part 3: 3-D Pharmacophore and 3-D QSAR Guided Brefeldin A Hit-to-Lead Optimization toward New Breast Cancer Suppressants. Molecules 2022; 27:molecules27092823. [PMID: 35566172 PMCID: PMC9101642 DOI: 10.3390/molecules27092823] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Revised: 04/22/2022] [Accepted: 04/23/2022] [Indexed: 02/01/2023] Open
Abstract
The estrogen receptor α (ERα) is an important biological target mediating 17β-estradiol driven breast cancer (BC) development. Aiming to develop innovative drugs against BC, either wild-type or mutated ligand-ERα complexes were used as source data to build structure-based 3-D pharmacophore and 3-D QSAR models, afterward used as tools for the virtual screening of National Cancer Institute datasets and hit-to-lead optimization. The procedure identified Brefeldin A (BFA) as hit, then structurally optimized toward twelve new derivatives whose anticancer activity was confirmed both in vitro and in vivo. Compounds as SERMs showed picomolar to low nanomolar potencies against ERα and were then investigated as antiproliferative agents against BC cell lines, as stimulators of p53 expression, as well as BC cell cycle arrest agents. Most active leads were finally profiled upon administration to female Wistar rats with pre-induced BC, after which 3DPQ-12, 3DPQ-3, 3DPQ-9, 3DPQ-4, 3DPQ-2, and 3DPQ-1 represent potential candidates for BC therapy.
Collapse
|
24
|
Herber CB, Yuan C, Chang A, Wang JC, Cohen I, Leitman DC. 2',3',4'-Trihydroxychalcone changes estrogen receptor α regulation of genes and breast cancer cell proliferation by a reprogramming mechanism. Mol Med 2022; 28:44. [PMID: 35468719 PMCID: PMC9036729 DOI: 10.1186/s10020-022-00470-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2021] [Accepted: 04/06/2022] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND Menopausal hormone therapy (MHT) is recommended for only five years to treat vasomotor symptoms and vulvovaginal atrophy because of safety concerns with long-term treatment. We investigated the ability of 2',3',4'-trihydroxychalcone (2',3',4'-THC) to modulate estrogen receptor (ER)-mediated responses in order to find drug candidates that could potentially prevent the adverse effects of long-term MHT treatment. METHODS Transfection assays, real time-polymerase chain reaction, and microarrays were used to evaluate the effects of 2',3',4'-THC on gene regulation. Radioligand binding studies were used to determine if 2',3',4'-THC binds to ERα. Cell proliferation was examined in MCF-7 breast cancer cells by using growth curves and flow cytometry. Western blots were used to determine if 2',3',4'-THC alters the E2 activation of the MAPK pathway and degradation of ERα. Chromatin immunoprecipitation was used to measure ERα binding to genes. RESULTS The 2',3',4'-THC/E2 combination produced a synergistic activation with ERα on reporter and endogenous genes in human U2OS osteosarcoma cells. Microarrays identified 824 genes that we termed reprogrammed genes because they were not regulated in U2OS-ERα cells unless they were treated with 2',3',4'-THC and E2 at the same time. 2',3',4'-THC blocked the proliferation of MCF-7 cells by preventing the E2-induced activation of MAPK and c-MYC transcription. The antiproliferative mechanism of 2',3',4'-THC differs from selective estrogen receptor modulators (SERMs) because 2',3',4'-THC did not bind to the E2 binding site in ERα like SERMs. CONCLUSION Our study suggests that 2',3',4'-THC may represent a new class of ERα modulators that do not act as a direct agonists or antagonists. We consider 2',3',4'-THC to be a reprogramming compound, since it alters the activity of ERα on gene regulation and cell proliferation without competing with E2 for binding to ERα. The addition of a reprogramming drug to estrogens in MHT may offer a new strategy to overcome the adverse proliferative effects of estrogen in MHT by reprogramming ERα as opposed to an antagonist mechanism that involves blocking the binding of estrogen to ERα.
Collapse
Affiliation(s)
- Candice B Herber
- Department of Nutritional Sciences and Toxicology, University of California, Berkeley, CA, 94720-3104, USA
- DENALI Therapeutics, 161 Oyster Point Blvd, South San Francisco, CA, 94080, USA
| | - Chaoshen Yuan
- Department of Nutritional Sciences and Toxicology, University of California, Berkeley, CA, 94720-3104, USA
- Iaterion, University of California, QB3, 1700 4th Street Byers Hall, Suite 214, San Francisco, CA, 94158, USA
| | - Anthony Chang
- Department of Nutritional Sciences and Toxicology, University of California, Berkeley, CA, 94720-3104, USA
- Biomedical Sciences Program, University of California San Francisco, San Francisco, CA, 94143, USA
| | - Jen-Chywan Wang
- Department of Nutritional Sciences and Toxicology, University of California, Berkeley, CA, 94720-3104, USA
| | - Isaac Cohen
- Iaterion, University of California, QB3, 1700 4th Street Byers Hall, Suite 214, San Francisco, CA, 94158, USA
| | - Dale C Leitman
- Department of Nutritional Sciences and Toxicology, University of California, Berkeley, CA, 94720-3104, USA.
- Iaterion, University of California, QB3, 1700 4th Street Byers Hall, Suite 214, San Francisco, CA, 94158, USA.
| |
Collapse
|
25
|
Tran LTT, Dang NYT, Nguyen Le NT, Nguyen HT, Ho DV, Do TT, Tran MH, Nguyen TK, Pham PTV. In Silico and in Vitro Evaluation of Alkaloids from Goniothalamus elegans Ast. for Breast Cancer Treatment. Nat Prod Commun 2022. [DOI: 10.1177/1934578x221088110] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Breast cancer is the leading cause of cancer mortality in women. In this study, liriodenine and lysicamine from Goniothalamus elegans Ast. were investigated for their anti-breast cancer activity based on their molecular interactions with three proteins related to breast cancer. Liriodenine had predicted binding affinities for BRCA1, BRCA2, and estrogen receptor alpha of −6.2, −7.9, and −8.3 kcal/mol, respectively. Lysicamine had predicted binding affinities of −5.8, −7.2, and 7.6 kcal/mol. To evaluate the biological activity of liriodenine and lysicamine, we studied their in vitro cytotoxic effects on MCF-7 cells. These alkaloids showed significant inhibitory effects with IC50 values of 33.31 and 70.03 µM. These results suggest that Goniothalamus elegans could be a promising medical plant for breast cancer treatment. Further studies are needed to understand the molecular mechanisms and improve the toxicity of liriodenine and lysicamine for clinical use.
Collapse
Affiliation(s)
- Linh Thuy Thi Tran
- Faculty of Pharmacy, Hue University of Medicine and Pharmacy, Hue University, Hue City, Thua Thien Hue Province, Vietnam
| | - Nhi Yen Thi Dang
- Faculty of Pharmacy, Hue University of Medicine and Pharmacy, Hue University, Hue City, Thua Thien Hue Province, Vietnam
| | - Nguyen Thao Nguyen Le
- Faculty of Pharmacy, Hue University of Medicine and Pharmacy, Hue University, Hue City, Thua Thien Hue Province, Vietnam
| | - Hoai Thi Nguyen
- Faculty of Pharmacy, Hue University of Medicine and Pharmacy, Hue University, Hue City, Thua Thien Hue Province, Vietnam
| | - Duc Viet Ho
- Faculty of Pharmacy, Hue University of Medicine and Pharmacy, Hue University, Hue City, Thua Thien Hue Province, Vietnam
| | - Thao Thi Do
- Institute of Biotechnology, Vietnam Academy of Science and Technology, , Hanoi, Vietnam
- Graduate University of Science and Technology, Vietnam Academy of Science and Technology, Hanoi, Vietnam
| | - Manh Hung Tran
- UDA-Institute of Applied Life Sciences (UDA-IALS), Dong A University, Da Nang city, Vietnam
- Scientific Management Department, Dong A University, Da Nang city, Vietnam
| | - Tan Khanh Nguyen
- UDA-Institute of Applied Life Sciences (UDA-IALS), Dong A University, Da Nang city, Vietnam
- Scientific Management Department, Dong A University, Da Nang city, Vietnam
| | - Phu Tran Vinh Pham
- Faculty of Medicine, Dong A University, Da Nang city, Vietnam
- UDA-Institute of Applied Life Sciences (UDA-IALS), Dong A University, Da Nang city, Vietnam
| |
Collapse
|
26
|
Liu R, Liu L, Bian Y, Zhang S, Wang Y, Chen H, Jiang X, Li G, Chen Q, Xue C, Li M, Liu L, Liu X, Ma S. The Dual Regulation Effects of ESR1/NEDD4L on SLC7A11 in Breast Cancer Under Ionizing Radiation. Front Cell Dev Biol 2022; 9:772380. [PMID: 35252218 PMCID: PMC8888677 DOI: 10.3389/fcell.2021.772380] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Accepted: 12/20/2021] [Indexed: 11/13/2022] Open
Abstract
Radiotherapy is one of the most important treatments for breast cancer. Ferroptosis is a recently recognized form of regulated cell death that is characterized by lipid peroxidation. However, whether ionizing radiation (IR) could induce ferroptosis in breast cancer and how it works remain unknown. Bioinformatics analysis were performed to screen ferroptosis-related genes differentially expressed in breast tumor tissue and normal tissue. Then, breast cancer cell lines with different estrogen receptor (ER) phenotypes were used for studies in vitro, including ER-positive (MCF-7 and ZR-75-1) and ER-negative (MDA-MB-231) cells. The dynamic changes of mRNA and protein levels were examined after x-ray of 8 Gy by qRT-PCR and Western blotting, respectively. Immunoprecipitation (IP) was used to explore the interaction between proteins. Luciferase assay was used to analyze the transcriptional regulation effect of ESR1 on SLC7A11. BODIPY C11 and trypan blue dyes were used to determine lipid peroxidation and cell death, respectively. The result showed that the ferroptosis-related gene SLC7A11 was higher in breast cancer tissues compared with normal tissues and associated with poor survival. A positive correlation exists between ESR1 and SLC7A11 expression. ESR1 promoted SLC7A11 expression at the early stage after IR. ESR1/SLC7A11 knockdown significantly enhanced IR-induced ferroptosis in ER-positive cells. At 12 h after IR, the IP data showed the interaction between E3 ubiquitin ligase NEDD4L and SLC7A11 increased, followed by the ubiquitylation and degradation of SLC7A11. Thus, SLC7A11 expression was regulated by both ESR1 and NEDD4L, in opposite ways. For the first time, we elucidated that ESR1 and NEDD4L functioned together after radiation treatment and finally induced ferroptosis in breast cancer cells, which provides novel insight into the guidance of clinical treatment of breast cancer.
Collapse
Affiliation(s)
- Rui Liu
- School of Public Health and Management, Wenzhou Medical University, Wenzhou, China.,NHC Key Laboratory of Radiobiology, School of Public Health of Jilin University, Changchun, China
| | - Lin Liu
- School of Public Health and Management, Wenzhou Medical University, Wenzhou, China
| | - Yan Bian
- School of Public Health and Management, Wenzhou Medical University, Wenzhou, China
| | - Shinan Zhang
- School of Public Health and Management, Wenzhou Medical University, Wenzhou, China
| | - Yue Wang
- School of Public Health and Management, Wenzhou Medical University, Wenzhou, China
| | - Huajian Chen
- School of Public Health and Management, Wenzhou Medical University, Wenzhou, China
| | - Xinyue Jiang
- School of Public Health and Management, Wenzhou Medical University, Wenzhou, China
| | - Guanghui Li
- School of Public Health and Management, Wenzhou Medical University, Wenzhou, China
| | - Qing Chen
- School of Public Health and Management, Wenzhou Medical University, Wenzhou, China
| | - Chang Xue
- School of Public Health and Management, Wenzhou Medical University, Wenzhou, China
| | - Mengke Li
- School of Public Health and Management, Wenzhou Medical University, Wenzhou, China
| | - Lianchang Liu
- NHC Key Laboratory of Radiobiology, School of Public Health of Jilin University, Changchun, China.,The Second Affiliated Hospital of Jilin University, Changchun, China
| | - Xiaodong Liu
- School of Public Health and Management, Wenzhou Medical University, Wenzhou, China.,Key Laboratory of Watershed Science and Health of Zhejiang Province, Wenzhou Medical University, Wenzhou, China
| | - Shumei Ma
- School of Public Health and Management, Wenzhou Medical University, Wenzhou, China.,NHC Key Laboratory of Radiobiology, School of Public Health of Jilin University, Changchun, China.,Key Laboratory of Watershed Science and Health of Zhejiang Province, Wenzhou Medical University, Wenzhou, China
| |
Collapse
|
27
|
Pharmacological Properties to Pharmacological Insight of Sesamin in Breast Cancer Treatment: A Literature-Based Review Study. Int J Breast Cancer 2022; 2022:2599689. [PMID: 35223101 PMCID: PMC8872699 DOI: 10.1155/2022/2599689] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Revised: 01/20/2022] [Accepted: 01/26/2022] [Indexed: 12/28/2022] Open
Abstract
The use of dietary phytochemical rather than conventional therapies to treat numerous cancers is now a well-known approach in medical science. Easily available and less toxic dietary phytochemicals present in plants should be introduced in the list of phytochemical-based treatment areas. Sesamin, a natural phytochemical, may be a promising chemopreventive agent aiming to manage breast cancer. In this study, we discussed the pharmacological properties of sesamin that determine its therapeutics opportunity to be used in breast cancer treatment and other diseases. Sesamin is available in medicinal plants, especially in Sesamum indicum, and is easily metabolized by the liver. To better understand the antibreast cancer consequence of sesamin, we postulate some putative pathways related to the antibreast cancer mechanism: (1) regulation of estrogen receptor (ER-α and ER-β) activities, (2) suppressing programmed death-ligand 1 (PD-L1) overexpression, (3) growth factor receptor inhibition, and (4) some tyrosine kinase pathways. Targeting these pathways, sesamin can modulate cell proliferation, cell cycle arrest, cell growth and viability, metastasis, angiogenesis, apoptosis, and oncogene inactivation in various in vitro and animal models. Although the actual tumor intrinsic signaling mechanism targeted by sesamin in cancer treatment is still unknown, this review summarized that this phytoestrogen suppressed NF-κB, STAT, MAPK, and PIK/AKT signaling pathways and activated some tumor suppressor protein in numerous breast cancer models. Cotreatment with γ-tocotrienol, conventional drugs, and several drug carriers systems increased the anticancer potentiality of sesamin. Furthermore, sesamin exhibited promising pharmacokinetics properties with less toxicity in the bodies. Overall, the shreds of evidence highlight that sesamin can be a potent candidate to design drugs against breast cancer. So, like other phytochemicals, sesamin can be consumed for better therapeutic advantages due to having the ability to target a plethora of molecular pathways until clinically trialed standard drugs are not available in pharma markets.
Collapse
|
28
|
Chimento A, De Luca A, Avena P, De Amicis F, Casaburi I, Sirianni R, Pezzi V. Estrogen Receptors-Mediated Apoptosis in Hormone-Dependent Cancers. Int J Mol Sci 2022; 23:1242. [PMID: 35163166 PMCID: PMC8835409 DOI: 10.3390/ijms23031242] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Revised: 01/14/2022] [Accepted: 01/17/2022] [Indexed: 02/04/2023] Open
Abstract
It is known that estrogen stimulates growth and inhibits apoptosis through estrogen receptor(ER)-mediated mechanisms in many cancer cell types. Interestingly, there is strong evidence that estrogens can also induce apoptosis, activating different ER isoforms in cancer cells. It has been observed that E2/ERα complex activates multiple pathways involved in both cell cycle progression and apoptotic cascade prevention, while E2/ERβ complex in many cases directs the cells to apoptosis. However, the exact mechanism of estrogen-induced tumor regression is not completely known. Nevertheless, ERs expression levels of specific splice variants and their cellular localization differentially affect outcome of estrogen-dependent tumors. The goal of this review is to provide a general overview of current knowledge on ERs-mediated apoptosis that occurs in main hormone dependent-cancers. Understanding the molecular mechanisms underlying the induction of ER-mediated cell death will be useful for the development of specific ligands capable of triggering apoptosis to counteract estrogen-dependent tumor growth.
Collapse
Affiliation(s)
- Adele Chimento
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Via Pietro Bucci, Arcavacata di Rende, 87036 Cosenza, Italy
| | - Arianna De Luca
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Via Pietro Bucci, Arcavacata di Rende, 87036 Cosenza, Italy
| | - Paola Avena
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Via Pietro Bucci, Arcavacata di Rende, 87036 Cosenza, Italy
| | - Francesca De Amicis
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Via Pietro Bucci, Arcavacata di Rende, 87036 Cosenza, Italy
| | - Ivan Casaburi
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Via Pietro Bucci, Arcavacata di Rende, 87036 Cosenza, Italy
| | - Rosa Sirianni
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Via Pietro Bucci, Arcavacata di Rende, 87036 Cosenza, Italy
| | - Vincenzo Pezzi
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Via Pietro Bucci, Arcavacata di Rende, 87036 Cosenza, Italy
| |
Collapse
|
29
|
Chen D, Wang M, Zhang H, Zhou S, Luo C. Estrogen receptor β2 (ERβ2)-mediated upregulation of hsa_circ_0000732 promotes tumor progression via sponging microRNA-1184 in triple-negative breast cancer (TNBC). Inflamm Res 2022; 71:255-266. [DOI: 10.1007/s00011-021-01536-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Revised: 12/06/2021] [Accepted: 12/14/2021] [Indexed: 12/25/2022] Open
|
30
|
Qiu L, Meng Y, Wang L, Gunewardena S, Liu S, Han J, Krieg AJ. Histone lysine demethylase 4B regulates general and unique gene expression signatures in hypoxic cancer cells. MedComm (Beijing) 2021; 2:414-429. [PMID: 34766154 PMCID: PMC8554665 DOI: 10.1002/mco2.85] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2021] [Revised: 07/28/2021] [Accepted: 07/30/2021] [Indexed: 02/05/2023] Open
Abstract
The hypoxic tumor microenvironment promotes tumor survival by inducing the expression of genes involved in angiogenesis and metastasis. As a direct target of hypoxia-inducible factor, lysine demethylase 4B (KDM4B) is overexpressed in multiple cancers, suggesting that a general KDM4B regulatory mechanism may exist in these cancer types. In this study, we sought to further investigate the general and unique roles of KDM4B in ovarian, colon, and renal cancer cells. We first identified a set of potential KDM4B targets shared by SKOV3ip.1, HCT116, and RCC4 cell lines, as well as numerous genes specifically regulated in each cell line. Through Gene Ontology, KEGG, and Oncobox pathway analyses, we found that KDM4B primarily regulated biosynthetic and cell cycle pathways in normoxia, whereas in hypoxia, it regulated pathways associated with inflammatory response and migration. TCGA data analyses reveal high expression of KDM4B in multiple cancer types and differential expression across cancer stages. Kaplan-Meier plots suggest that elevated KDM4B expression may contribute to a better or worse prognosis in a manner specific to each cancer type. Overall, our findings suggest that KDM4B plays complex roles in regulating multiple cancer processes, providing a useful resource for the future development of cancer therapies that target KDM4B expression.
Collapse
Affiliation(s)
- Lei Qiu
- Research Laboratory of Cancer Epigenetics and Genomics Department of General Surgery Frontiers Science Center for Disease-related Molecular Network Cancer Center West China Hospital Sichuan University Chengdu China.,Department of Obstetrics and Gynecology University of Kansas Medical Center Kansas City Kansas USA.,Department of Pathology and Laboratory Medicine University of Kansas Medical Center Kansas City Kansas USA
| | - Yang Meng
- Research Laboratory of Cancer Epigenetics and Genomics Department of General Surgery Frontiers Science Center for Disease-related Molecular Network Cancer Center West China Hospital Sichuan University Chengdu China
| | - Lingli Wang
- Research Laboratory of Cancer Epigenetics and Genomics Department of General Surgery Frontiers Science Center for Disease-related Molecular Network Cancer Center West China Hospital Sichuan University Chengdu China
| | - Sumedha Gunewardena
- Department of Molecular and Integrative Physiology University of Kansas Medical Center Kansas City Kansas USA
| | - Sicheng Liu
- Research Laboratory of Cancer Epigenetics and Genomics Department of General Surgery Frontiers Science Center for Disease-related Molecular Network Cancer Center West China Hospital Sichuan University Chengdu China
| | - Junhong Han
- Research Laboratory of Cancer Epigenetics and Genomics Department of General Surgery Frontiers Science Center for Disease-related Molecular Network Cancer Center West China Hospital Sichuan University Chengdu China
| | - Adam J Krieg
- Department of Obstetrics and Gynecology University of Kansas Medical Center Kansas City Kansas USA.,Department of Pathology and Laboratory Medicine University of Kansas Medical Center Kansas City Kansas USA.,Department of Obstetrics and Gynecology Oregon Health and Science University USA.,Division of Reproductive and Developmental Sciences Oregon National Primate Research Center Beaverton Oregon USA
| |
Collapse
|
31
|
Porras L, Ismail H, Mader S. Positive Regulation of Estrogen Receptor Alpha in Breast Tumorigenesis. Cells 2021; 10:cells10112966. [PMID: 34831189 PMCID: PMC8616513 DOI: 10.3390/cells10112966] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2021] [Revised: 10/23/2021] [Accepted: 10/24/2021] [Indexed: 12/31/2022] Open
Abstract
Estrogen receptor alpha (ERα, NR3A1) contributes through its expression in different tissues to a spectrum of physiological processes, including reproductive system development and physiology, bone mass maintenance, as well as cardiovascular and central nervous system functions. It is also one of the main drivers of tumorigenesis in breast and uterine cancer and can be targeted by several types of hormonal therapies. ERα is expressed in a subset of luminal cells corresponding to less than 10% of normal mammary epithelial cells and in over 70% of breast tumors (ER+ tumors), but the basis for its selective expression in normal or cancer tissues remains incompletely understood. The mapping of alternative promoters and regulatory elements has delineated the complex genomic structure of the ESR1 gene and shed light on the mechanistic basis for the tissue-specific regulation of ESR1 expression. However, much remains to be uncovered to better understand how ESR1 expression is regulated in breast cancer. This review recapitulates the current body of knowledge on the structure of the ESR1 gene and the complex mechanisms controlling its expression in breast tumors. In particular, we discuss the impact of genetic alterations, chromatin modifications, and enhanced expression of other luminal transcription regulators on ESR1 expression in tumor cells.
Collapse
|
32
|
Mihović N, Tomašević N, Matić S, Mitrović MM, Kostić DA, Sabatino M, Antonini L, Ragno R, Mladenović M. Human Estrogen Receptor α Antagonists. Part 1: 3-D QSAR-Driven Rational Design of Innovative Coumarin-Related Antiestrogens as Breast Cancer Suppressants through Structure-Based and Ligand-Based Studies. J Chem Inf Model 2021; 61:5028-5053. [PMID: 34648283 DOI: 10.1021/acs.jcim.1c00530] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
The estrogen receptor α (ERα) represents a 17β-estradiol-inducible transcriptional regulator that initiates the RNA polymerase II-dependent transcriptional machinery, pointed for breast cancer (BC) development via either genomic direct or genomic indirect (i.e., tethered) pathway. To develop innovative ligands, structure-based (SB) three-dimensional (3-D) quantitative structure-activity relationship (QSAR) studies have been undertaken from structural data taken from partial agonists, mixed agonists/antagonists (selective estrogen receptor modulators (SERMs)), and full antagonists (selective ERα downregulators (SERDs)) correlated with either wild-type or mutated ERα receptors. SB and ligand-based (LB) alignments allow us to rule out guidelines for the SB/LB alignment of untested compounds. 3-D QSAR models for ERα ligands, coupled with SB/LB alignment, were revealed to be useful tools to dissect the chemical determinants for ERα-based anticancer activity as well as to predict their potency. The herein developed protocol procedure was verified through the design and potency prediction of 12 new coumarin-based SERMs, namely, 3DQ-1a to 3DQ-1e, that upon synthesis turned to be potent ERα antagonists by means of either in vitro or in vivo assays (described in the second part of this study).
Collapse
Affiliation(s)
- Nezrina Mihović
- Kragujevac Center for Computational Biochemistry, Department of Chemistry, Faculty of Science, University of Kragujevac, Radoja Domanovića 12, P.O. Box 60, 34000 Kragujevac, Serbia
| | - Nevena Tomašević
- Kragujevac Center for Computational Biochemistry, Department of Chemistry, Faculty of Science, University of Kragujevac, Radoja Domanovića 12, P.O. Box 60, 34000 Kragujevac, Serbia
| | - Sanja Matić
- Institute for Informational Technologies, University of Kragujevac, Jovana Cvijića bb, 34000 Kragujevac, Serbia
| | - Marina M Mitrović
- Department of Biochemistry, Faculty of Medical Sciences, University of Kragujevac, Svetozara Markovića 69, 34000 Kragujevac, Serbia
| | - Danijela A Kostić
- Department of Chemistry, Faculty of Sciences and Mathematics, University of Niš, Višegradska 33, 18000 Niš, Serbia
| | - Manuela Sabatino
- Rome Center for Molecular Design, Department of Drug Chemistry and Technology, Faculty of Pharmacy and Medicine, Sapienza University of Rome, P.le A. Moro 5, 00185 Rome, Italy
| | - Lorenzo Antonini
- Rome Center for Molecular Design, Department of Drug Chemistry and Technology, Faculty of Pharmacy and Medicine, Sapienza University of Rome, P.le A. Moro 5, 00185 Rome, Italy
| | - Rino Ragno
- Rome Center for Molecular Design, Department of Drug Chemistry and Technology, Faculty of Pharmacy and Medicine, Sapienza University of Rome, P.le A. Moro 5, 00185 Rome, Italy
| | - Milan Mladenović
- Kragujevac Center for Computational Biochemistry, Department of Chemistry, Faculty of Science, University of Kragujevac, Radoja Domanovića 12, P.O. Box 60, 34000 Kragujevac, Serbia
| |
Collapse
|
33
|
Dai YH, Chen GY, Tang CH, Huang WC, Yang JC, Wu YC. Drug Screening of Potential Multiple Target Inhibitors for Estrogen Receptor-α-positive Breast Cancer. In Vivo 2021; 35:761-777. [PMID: 33622869 DOI: 10.21873/invivo.12317] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2020] [Revised: 11/21/2020] [Accepted: 11/24/2020] [Indexed: 02/07/2023]
Abstract
BACKGROUND/AIM Estrogen receptor α (ERα) antagonist is the most common treatment for ERα-positive breast cancer. However, compensatory signaling contributes to resistance to ERα antagonists. Thus, to explore the potential agents for targeting compensatory signaling, we screened multiple target inhibitors for breast cancer treatment. MATERIALS AND METHODS We attempted to build a structure-based virtual screening model that can find potential compounds and assay the anticancer ability of these drugs by overall cell survival assay. The downstream compensatory phosphorylated signaling was measured by immunoblotting. RESULTS Hamamelitannin and glucocheirolin were hits for ERα, phosphoinositide 3-kinase (PI3K), and KRAS proto-oncogene, GTPase (KRAS), which were active against estrogen and epidermal growth factor-triggered proliferation. Additionally, we select aminopterin as a hit for ERα, PI3K, KRAS, and SRC proto-oncogene, non-receptor tyrosine kinase (SRC) with inhibitory activities toward AKT serine/threonine kinase 1 (AKT) and mitogen-activated protein kinase kinase (MEK) signaling. CONCLUSION Our structure-based virtual screening model selected hamamelitannin, glucocheirolin, aminopterin, and pemetrexed as compounds that may act as potential inhibitors for improving endocrine therapies for breast cancer.
Collapse
Affiliation(s)
- Yun-Hao Dai
- School of Pharmacy, China Medical University, Taichung, Taiwan, R.O.C.,Chinese Medicine Research and Development Center, Center for Molecular Medicine, China Medical University Hospital, China Medical University, Taichung, Taiwan, R.O.C
| | - Guan-Yu Chen
- Chinese Medicine Research and Development Center, Center for Molecular Medicine, China Medical University Hospital, China Medical University, Taichung, Taiwan, R.O.C
| | - Chih-Hsin Tang
- Department of Pharmacology, School of Medicine, China Medical University, Taichung, Taiwan, R.O.C.,Chinese Medicine Research Center, Drug Development Center, China Medical University, Taichung, Taiwan, R.O.C.,Department of Biotechnology, College of Medical and Health Science, Asia University, Taichung, Taiwan, R.O.C
| | - Wei-Chien Huang
- Chinese Medicine Research and Development Center, Center for Molecular Medicine, China Medical University Hospital, China Medical University, Taichung, Taiwan, R.O.C.,Chinese Medicine Research Center, Drug Development Center, China Medical University, Taichung, Taiwan, R.O.C.,Graduate Institute of Biomedical Sciences, China Medical University, Taichung, Taiwan, R.O.C.,The Biotechnology Department, College of Medical and Health Science, Asia University, Taichung, Taiwan, R.O.C
| | - Juan-Cheng Yang
- Chinese Medicine Research and Development Center, Center for Molecular Medicine, China Medical University Hospital, China Medical University, Taichung, Taiwan, R.O.C.;
| | - Yang-Chang Wu
- Chinese Medicine Research and Development Center, Center for Molecular Medicine, China Medical University Hospital, China Medical University, Taichung, Taiwan, R.O.C.; .,The Biotechnology Department, College of Medical and Health Science, Asia University, Taichung, Taiwan, R.O.C.,Graduate Institute of Integrated Medicine, College of Chinese Medicine, China Medical University, Taichung, Taiwan, R.O.C
| |
Collapse
|
34
|
Binienda A, Ziolkowska S, Pluciennik E. The Anticancer Properties of Silibinin: Its Molecular Mechanism and Therapeutic Effect in Breast Cancer. Anticancer Agents Med Chem 2021; 20:1787-1796. [PMID: 31858905 DOI: 10.2174/1871520620666191220142741] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2019] [Revised: 10/29/2019] [Accepted: 11/12/2019] [Indexed: 02/08/2023]
Abstract
BACKGROUND Silibinin (SB), the main component of Silymarin (SM), is a natural substance obtained from the seeds of the milk thistle. SM contains up to 70% of SB as two isoforms: A and B. It has an antioxidant and anti-inflammatory effect on hepatocytes and is known to inhibit cell proliferation, induce apoptosis, and curb angiogenesis. SB has demonstrated activity against many cancers, such as skin, liver, lung, bladder, and breast carcinomas. METHODS This review presents current knowledge of the use of SM in breast cancer, this being one of the most common types of cancer in women. It describes selected molecular mechanisms of the action of SM; for example, although SB influences both Estrogen Receptors (ER), α and β, it has opposite effects on the two. Its action on ERα influences the PI3K/AKT/mTOR and RAS/ERK signaling pathways, while by up-regulating ERβ, it increases the numbers of apoptotic cells. In addition, ERα is involved in SB-induced autophagy, while ERβ is not. Interestingly, SB also inhibits metastasis by suppressing TGF-β2 expression, thus suppressing Epithelial to Mesenchymal Transition (EMT). It also influences migration and invasive potential via the Jak2/STAT3 pathway. RESULTS SB may be a promising enhancement of BC treatment: when combined with chemotherapeutic drugs such as carboplatin, cisplatin, and doxorubicin, the combination exerts a synergistic effect against cancer cells. This may be of value when treating aggressive types of mammary carcinoma. CONCLUSION Summarizing, SB inhibits proliferation, induces apoptosis, and restrains metastasis via several mechanisms. It is possible to combine SB with different anticancer drugs, an approach that represents a promising therapeutic strategy for patients suffering from BC.
Collapse
Affiliation(s)
- Agata Binienda
- Faculty of Biomedical Sciences and Postgraduate Education, Medical University of Lodz, Lodz, Poland
| | - Sylwia Ziolkowska
- Faculty of Biomedical Sciences and Postgraduate Education, Medical University of Lodz, Lodz, Poland
| | - Elzbieta Pluciennik
- Department of Molecular Carcinogenesis, Medical University of Lodz, Lodz, Poland
| |
Collapse
|
35
|
Estrogen suppresses HOXB2 expression via ERα in breast cancer cells. Gene 2021; 794:145746. [PMID: 34062258 DOI: 10.1016/j.gene.2021.145746] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2020] [Revised: 04/27/2021] [Accepted: 05/27/2021] [Indexed: 11/20/2022]
Abstract
The expression of HOXB2, a homeobox transcription factor, is altered in a variety of solid tumors. Using an in vivo screen to identify regulators of breast tumor growth in murine mammary fat pads, Boimel and co-workers recently identified HOXB2 as a tumor suppressor. However, the mechanistic underpinnings of its role in breast cancer is not understood. Given the emerging interaction of estrogen-regulated gene expression and altered HOX gene expression network in the pathophysiology of breast cancer, this study addressed the relationship between estrogen signaling and HOXB2 expression. Using a mouse model and human breast cancer cell lines, we show that estrogen suppresses HOXB2 expression. Suppression of HOXB2 by PPT, a known ERα agonist, in MCF-7 and T47D cells indicated the involvement of ERα, which was confirmed by siRNA-mediated ERα knockdown experiments. In-silico analysis of the upstream promoter region revealed the presence of three putative EREs. Chromatin immunoprecipitation experiments showed that upon estrogen binding, ERα engaged with EREs in the 5' upstream region of HOXB2 in MCF-7 and T47D cells. Future investigations should address the implications of estrogen-mediated suppression on the proposed tumor suppressor function of HOXB2.
Collapse
|
36
|
Linares-Anaya O, Avila-Sorrosa A, Díaz-Cedillo F, Gil-Ruiz LÁ, Correa-Basurto J, Salazar-Mendoza D, Orjuela AL, Alí-Torres J, Ramírez-Apan MT, Morales-Morales D. Synthesis, Characterization, and Preliminary In Vitro Cytotoxic Evaluation of a Series of 2-Substituted Benzo [ d] [1,3] Azoles. Molecules 2021; 26:molecules26092780. [PMID: 34066820 PMCID: PMC8125891 DOI: 10.3390/molecules26092780] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2021] [Revised: 04/09/2021] [Accepted: 04/13/2021] [Indexed: 11/16/2022] Open
Abstract
A series of benzo [d] [1,3] azoles 2-substituted with benzyl- and allyl-sulfanyl groups were synthesized, and their cytotoxic activities were in vitro evaluated against a panel of six human cancer cell lines. The results showed that compounds BTA-1 and BMZ-2 have the best inhibitory effects, compound BMZ-2 being comparable in some cases with the reference drug tamoxifen and exhibiting a low cytotoxic effect against healthy cells. In silico molecular coupling studies at the tamoxifen binding site of ERα and GPER receptors revealed affinity and the possible mode of interaction of both compounds BTA-1 and BMZ-2.
Collapse
Affiliation(s)
- Ozvaldo Linares-Anaya
- Instituto Politécnico Nacional, Departamento de Química Orgánica, Carpio y Plan de Ayala S/N, Escuela Nacional de Ciencias Biológicas, Colonia Santo Tomás, Ciudad de México 11340, Mexico; (O.L.-A.); (F.D.-C.); (L.Á.G.-R.)
| | - Alcives Avila-Sorrosa
- Instituto Politécnico Nacional, Departamento de Química Orgánica, Carpio y Plan de Ayala S/N, Escuela Nacional de Ciencias Biológicas, Colonia Santo Tomás, Ciudad de México 11340, Mexico; (O.L.-A.); (F.D.-C.); (L.Á.G.-R.)
- Correspondence: ; Tel.: +52-555-729-6000
| | - Francisco Díaz-Cedillo
- Instituto Politécnico Nacional, Departamento de Química Orgánica, Carpio y Plan de Ayala S/N, Escuela Nacional de Ciencias Biológicas, Colonia Santo Tomás, Ciudad de México 11340, Mexico; (O.L.-A.); (F.D.-C.); (L.Á.G.-R.)
| | - Luis Ángel Gil-Ruiz
- Instituto Politécnico Nacional, Departamento de Química Orgánica, Carpio y Plan de Ayala S/N, Escuela Nacional de Ciencias Biológicas, Colonia Santo Tomás, Ciudad de México 11340, Mexico; (O.L.-A.); (F.D.-C.); (L.Á.G.-R.)
- Laboratorio de Diseño y Desarrollo de Nuevos Fármacos e Innovación Biotecnológica, Instituto Politécnico Nacional, Escuela Superior de Medicina, Ciudad de México 11340, Mexico;
| | - José Correa-Basurto
- Laboratorio de Diseño y Desarrollo de Nuevos Fármacos e Innovación Biotecnológica, Instituto Politécnico Nacional, Escuela Superior de Medicina, Ciudad de México 11340, Mexico;
| | - Domingo Salazar-Mendoza
- Carretera a Acatlima, Huajuapan de León, Universidad Tecnológica de la Mixteca, Oaxaca 69000, Mexico;
| | - Adrian L. Orjuela
- Departamento de Química, Universidad Nacional de Colombia-Sede, Bogotá 111321, Colombia; (A.L.O.); (J.A.-T.)
| | - Jorge Alí-Torres
- Departamento de Química, Universidad Nacional de Colombia-Sede, Bogotá 111321, Colombia; (A.L.O.); (J.A.-T.)
| | - María Teresa Ramírez-Apan
- Instituto de Química, Universidad Nacional Autónoma de México, Circuito Exterior, Ciudad Universitaria, Ciudad de México 04510, Mexico; (M.T.R.-A.); (D.M.-M.)
| | - David Morales-Morales
- Instituto de Química, Universidad Nacional Autónoma de México, Circuito Exterior, Ciudad Universitaria, Ciudad de México 04510, Mexico; (M.T.R.-A.); (D.M.-M.)
| |
Collapse
|
37
|
Kauffman RP, Young C, Castracane VD. Perils of prolonged ovarian suppression and hypoestrogenism in the treatment of breast cancer: Is the risk of treatment worse than the risk of recurrence? Mol Cell Endocrinol 2021; 525:111181. [PMID: 33529690 DOI: 10.1016/j.mce.2021.111181] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/01/2020] [Revised: 01/04/2021] [Accepted: 01/21/2021] [Indexed: 01/18/2023]
Abstract
Premenopausal breast cancer is usually estrogen receptor positive, and hence, prolonged ovarian suppression by medical or surgical means to prevent recurrence has become standard of management to improve disease-free survival. Ten-year adjuvant tamoxifen therapy is associated with 3.5% fewer recurrences compared to five years. The SOFT trial demonstrated small but statistically significant incremental improvements in long-term disease-free survival by the addition of gonadotropin-releasing hormone analog treatment (triptorelin) to an aromatase inhibitor (exemestane). Profound hypoestrogenism in the premenopausal age group may not be well tolerated due to a host of bothersome side effects (primarily vasomotor symptoms, musculoskeletal complaints, genitourinary syndrome of menopause, and mood disorders). Prolonged hypoestrogenism in younger women is associated with premature development of cardiovascular disease, bone loss, cognitive decline, and all-cause mortality. This paper explores multi-system consequences of prolonged hypoestrogenism in premenopausal women derived from studies of women with and without breast cancer. Pretreatment counseling in estrogen receptor positive breast cancer should emphasize the benefit of prolonged estrogen suppression on breast cancer recurrence and established risks of lifelong hypoestrogenism on quality of life and all-cause mortality. Future genomic research may help identify the best candidates for extended ovarian suppression to avoid treating many women when only a minority benefit.
Collapse
Affiliation(s)
- Robert P Kauffman
- Department of Obstetrics and Gynecology, Texas Tech University Health Sciences Center, School of Medicine, 1400 S. Coulter Rd, Amarillo, TX, 79106, USA.
| | - Christina Young
- Department of Obstetrics and Gynecology, Texas Tech University Health Sciences Center, School of Medicine, 1400 S. Coulter Rd, Amarillo, TX, 79106, USA
| | - V Daniel Castracane
- Department of Obstetrics and Gynecology, Texas Tech University Health Sciences Center, School of Medicine, 1400 S. Coulter Rd, Amarillo, TX, 79106, USA
| |
Collapse
|
38
|
Calaf GM. Role of organophosphorous pesticides and acetylcholine in breast carcinogenesis. Semin Cancer Biol 2021; 76:206-217. [PMID: 33766648 DOI: 10.1016/j.semcancer.2021.03.016] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2021] [Accepted: 03/18/2021] [Indexed: 12/16/2022]
Abstract
Breast cancer is the leading cause of cancer-related death in women worldwide. Several studies have addressed the association between cancer in humans and agricultural pesticide exposure. Evidence indicates that exposure to organophosphorous pesticides such as parathion and malathion occurs as a result of occupational factors since they are extensively used to control insects. On the other hand, estrogens have been considered beneficial to the organism; however, epidemiological studies have pointed out an increased breast cancer risk in both humans and animals. Experimental female rat mammary gland cancer models were developed after exposure to parathion, malathion, eserine, an acetylcholinesterase inhibitor, and estrogen allowing the analysis of the signs of carcinogenicity as alteration of cell proliferation, receptor expression, genomic instability, and cell metabolism in vivo and in vitro. Thus, pesticides increased proliferative ducts followed by ductal carcinoma; and 17β-estradiol increased proliferative lobules followed by lobular carcinomas. The combination of both pesticides and either eserine or estrogen induced tumors with both types of structures followed by mammary gland tumors and metastasis to the lung and kidneys after 240 days of a 5-day treatment. Studies also showed that these pesticides and eserine decreased three to five times the acetylcholinesterase activity in the serum compared to controls whereas terminal end buds increased in number, being inhibited by atropine. Genomic instability was analyzed in such tissues (mp53, CYP1A2, c-myc, c-fos, ERα, M2R) and pesticides increased protein expression that was stimulated by estrogens but inhibited by atropine. Eserine also transformed the epithelium of the rat mammary gland in the presence of estrogen and increased the number of terminal end buds after treatment inducing mammary carcinomas. Then, enzymatic digestion of such structures gave rise to cells with increased DNA synthesis and induced anchorage independence. Thus, there were changes in the epithelium of the mammary gland influencing breast carcinogenesis. Furthermore, these substances and acetylcholine also showed the signs of carcinogenicity in vitro as cell proliferation, receptor expression (ERα, ErbB2, M2R), genomic instability (c-myc, mp53, ERα, M2R), and cell metabolism. A unique cellular model is also presented here based on the use of MCF-10 F, a non-tumorigenic cell line that represents a valuable clinically translatable experimental approach that identifies mechanistic links for pesticides and estrogen as suspect human carcinogenic agents.
Collapse
Affiliation(s)
- Gloria M Calaf
- Instituto de Alta Investigación, Universidad de Tarapacá, Arica, 1000000, Chile; Center for Radiological Research, Columbia University Medical Center, New York, NY, 10032, USA.
| |
Collapse
|
39
|
Bhat-Nakshatri P, Gao H, Sheng L, McGuire PC, Xuei X, Wan J, Liu Y, Althouse SK, Colter A, Sandusky G, Storniolo AM, Nakshatri H. A single-cell atlas of the healthy breast tissues reveals clinically relevant clusters of breast epithelial cells. CELL REPORTS MEDICINE 2021; 2:100219. [PMID: 33763657 PMCID: PMC7974552 DOI: 10.1016/j.xcrm.2021.100219] [Citation(s) in RCA: 47] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Revised: 11/10/2020] [Accepted: 02/18/2021] [Indexed: 01/21/2023]
Abstract
Single-cell RNA sequencing (scRNA-seq) is an evolving technology used to elucidate the cellular architecture of adult organs. Previous scRNA-seq on breast tissue utilized reduction mammoplasty samples, which are often histologically abnormal. We report a rapid tissue collection/processing protocol to perform scRNA-seq of breast biopsies of healthy women and identify 23 breast epithelial cell clusters. Putative cell-of-origin signatures derived from these clusters are applied to analyze transcriptomes of ~3,000 breast cancers. Gene signatures derived from mature luminal cell clusters are enriched in ~68% of breast cancers, whereas a signature from a luminal progenitor cluster is enriched in ~20% of breast cancers. Overexpression of luminal progenitor cluster-derived signatures in HER2+, but not in other subtypes, is associated with unfavorable outcome. We identify TBX3 and PDK4 as genes co-expressed with estrogen receptor (ER) in the normal breasts, and their expression analyses in >550 breast cancers enable prognostically relevant subclassification of ER+ breast cancers.
Collapse
Affiliation(s)
- Poornima Bhat-Nakshatri
- Department of Surgery, Indiana University of School of Medicine, Indianapolis, IN 46202, USA
| | - Hongyu Gao
- Center for Computational Biology and Bioinformatics, Indiana University School of Medicine, Indianapolis, IN 46202, USA.,Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Liu Sheng
- Center for Computational Biology and Bioinformatics, Indiana University School of Medicine, Indianapolis, IN 46202, USA.,Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Patrick C McGuire
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Xiaoling Xuei
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Jun Wan
- Center for Computational Biology and Bioinformatics, Indiana University School of Medicine, Indianapolis, IN 46202, USA.,Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Yunlong Liu
- Center for Computational Biology and Bioinformatics, Indiana University School of Medicine, Indianapolis, IN 46202, USA.,Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Sandra K Althouse
- Department of Biostatistics, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Austyn Colter
- Department of Pathology and Laboratory Medicine, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - George Sandusky
- Department of Pathology and Laboratory Medicine, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Anna Maria Storniolo
- Department of Medicine, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Harikrishna Nakshatri
- Department of Surgery, Indiana University of School of Medicine, Indianapolis, IN 46202, USA.,Center for Computational Biology and Bioinformatics, Indiana University School of Medicine, Indianapolis, IN 46202, USA.,Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN 46202, USA.,Roudebush VA Medical Center, Indianapolis, IN 46202, USA
| |
Collapse
|
40
|
The Secreted Protein C10orf118 Is a New Regulator of Hyaluronan Synthesis Involved in Tumour-Stroma Cross-Talk. Cancers (Basel) 2021; 13:cancers13051105. [PMID: 33807583 PMCID: PMC7961460 DOI: 10.3390/cancers13051105] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2021] [Accepted: 02/26/2021] [Indexed: 12/20/2022] Open
Abstract
Simple Summary Hyaluronan is a main glycosaminoglycan in extracellular matrix with an important role in breast cancer progression. Alterations in its synthesis and size may affect tu-mour growth and metastasis. Communication between stromal and breast cancer cells consists of the secretion of factors that provoke a series of cell signalling that influence cell fate and tis-sue microenvironment, by favouring tumour cell survival and motility. Here, we present the c10orf118 protein expressed in high amounts by breast tumour cells as a new regulator in hya-luronan synthesis. This protein is found both in Golgi and secreted in the extracellular matrix, whereas its role is still unknown. The secreted c10orf118 is found to induce hyaluronan synthase 2 in normal fibroblasts. Importantly, high expression of c10orf118 is positively correlated to pa-tient’s survival and to a low metastasis. Abstract Interaction between cancer cells and their microenvironment is central in defining the fate of cancer development. Tumour cells secrete signals (cytokines, chemokines, growth factors) that modify the surrounding area, while the niche supplies structures and activities necessary for tumour maintenance and growth. Hyaluronan (HA) is a glycosaminoglycan that constitute cancer cell niche and is known to influence tumour functions such as proliferation, migration and neoangiogenesis. The knowledge of the factors regulating HA synthesis and size is crucial in understanding the mechanisms sustaining tumour development. Here we show that a yet uncharacterized protein secreted by breast tumour cell lines, named c10orf118 (accession number NM_018017 in NCBI/BLAST, and Q7z3E2 according to the Uniprot identifier), with a predicted length of 898 amino acids, can induce the secretion of HA by stromal fibroblasts through the up-regulation of the hyaluronan synthase 2 gene (HAS2). Intracellularly, this protein is localized in the Golgi apparatus with a possible role in vesicle maturation and transport. The expression of c10orf118 was verified in breast cancer patient specimens and was found to be associated with the presence of estrogen receptor that characterizes a good patient survival. We suggest c10orf118 as a new player that influences the HA amount in breast cancer microenvironment and is associated with low aggressiveness of cancer.
Collapse
|
41
|
Emad A, Sinha S. Inference of phenotype-relevant transcriptional regulatory networks elucidates cancer type-specific regulatory mechanisms in a pan-cancer study. NPJ Syst Biol Appl 2021; 7:9. [PMID: 33558504 PMCID: PMC7870953 DOI: 10.1038/s41540-021-00169-7] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2019] [Accepted: 01/05/2021] [Indexed: 01/30/2023] Open
Abstract
Reconstruction of transcriptional regulatory networks (TRNs) is a powerful approach to unravel the gene expression programs involved in healthy and disease states of a cell. However, these networks are usually reconstructed independent of the phenotypic (or clinical) properties of the samples. Therefore, they may confound regulatory mechanisms that are specifically related to a phenotypic property with more general mechanisms underlying the full complement of the analyzed samples. In this study, we develop a method called InPheRNo to identify "phenotype-relevant" TRNs. This method is based on a probabilistic graphical model that models the simultaneous effects of multiple transcription factors (TFs) on their target genes and the statistical relationship between the target genes' expression and the phenotype. Extensive comparison of InPheRNo with related approaches using primary tumor samples of 18 cancer types from The Cancer Genome Atlas reveals that InPheRNo can accurately reconstruct cancer type-relevant TRNs and identify cancer driver TFs. In addition, survival analysis reveals that the activity level of TFs with many target genes could distinguish patients with poor prognosis from those with better prognosis.
Collapse
Affiliation(s)
- Amin Emad
- Department of Electrical and Computer Engineering, McGill University, Montreal, QC, Canada.
| | - Saurabh Sinha
- Carl R. Woese Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Urbana, IL, USA.
- Department of Computer Science, University of Illinois at Urbana-Champaign, Urbana, IL, USA.
- Cancer Center at Illinois, University of Illinois at Urbana-Champaign, Urbana, IL, USA.
| |
Collapse
|
42
|
El Mohtadi M, Whitehead K, Dempsey-Hibbert N, Belboul A, Ashworth J. Estrogen deficiency - a central paradigm in age-related impaired healing? EXCLI JOURNAL 2021; 20:99-116. [PMID: 33510594 PMCID: PMC7838826 DOI: 10.17179/excli2020-3210] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/26/2020] [Accepted: 01/04/2021] [Indexed: 11/10/2022]
Abstract
Wound healing is a dynamic biological process achieved through four sequential, overlapping phases; hemostasis, inflammation, tissue proliferation and remodeling. For effective wound healing, all four phases must occur in the appropriate order and time frame. It is well accepted that the wound healing process becomes disrupted in the elderly, increasing the propensity of non-healing wound states that can lead to substantial patient morbidity and an enormous financial burden on healthcare systems. Estrogen deprivation in the elderly has been identified as the key driver of age-related delayed wound healing in both genders, with topical and systemic estrogen replacement reversing the detrimental effects of aging on wound repair. Evidence suggests estrogen deprivation may contribute to the development of chronic wound healing states in the elderly but research in this area is somewhat limited, warranting further investigations. Moreover, although the beneficial effects of estrogen on cutaneous healing have been widely explored, the development of estrogen-based treatments to enhance wound repair in the elderly have yet to be widely exploited. This review explores the critical role of estrogen in reversing age-related impaired healing and evaluates the prospect of developing more focused novel therapeutic strategies that enhance wound repair in the elderly via activation of specific estrogen signaling pathways in regenerating tissues, whilst leaving non-target tissues largely unaffected.
Collapse
Affiliation(s)
- Mohamed El Mohtadi
- Department of Biology, Edge Hill University, Ormskirk, Lancashire, L39 4QP, UK
| | - Kathryn Whitehead
- Centre for Bioscience, Manchester Metropolitan University, Chester Street, Manchester, M1 5GD, UK
| | - Nina Dempsey-Hibbert
- Centre for Bioscience, Manchester Metropolitan University, Chester Street, Manchester, M1 5GD, UK
| | - Amina Belboul
- Centre for Bioscience, Manchester Metropolitan University, Chester Street, Manchester, M1 5GD, UK
| | - Jason Ashworth
- Centre for Bioscience, Manchester Metropolitan University, Chester Street, Manchester, M1 5GD, UK
| |
Collapse
|
43
|
Rathinaraj Benjamin S, de Lima F. Current and Prospective of Breast Cancer Biomarkers. Mol Biotechnol 2021. [DOI: 10.5772/intechopen.91151] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Biomarkers have shown great promise over the past decade the process of drug development more effective and have become an integral part of diagnosis of diseases. Biosensors were integrated with biomarker detection and point-of-care detection for signal amplification, high specificity and sensitivity, rapid response time, low cost, simplicity and multi-analytical testing. In order to detect more sensitively, these particular biomarkers have been explored with the possibility of real-time measurements in order to develop simple and compact systems which can analyze complex specimens. Various biosensors including electrochemical biosensors have recently been developed based on disease-specific biomarkers in the diagnosis of cancer disease. The main objective of the book chapter is to review research with new materials/methods in electrochemical biosensing techniques to detection of breast cancer biomarkers and evaluating latest techniques for detection of important analytes in real samples. In this book chapter, the recent development of electrochemical biosensors of breast cancer biomarkers will be reviewed. Furthermore, recent and future trend application of breast cancer biomarkers will be discussed.
Collapse
|
44
|
Gul HF, Ilhan N, Ilhan N, Ozercan IH, Kuloglu T. The combined effect of pomegranate extract and tangeretin on the DMBA-induced breast cancer model. J Nutr Biochem 2020; 89:108566. [PMID: 33326843 DOI: 10.1016/j.jnutbio.2020.108566] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2020] [Revised: 10/16/2020] [Accepted: 11/25/2020] [Indexed: 12/14/2022]
Abstract
The aim of this study was to investigate the protective effects of pomegranate extract and tangeretin alone or in combination in DMBA-induced rat breast cancer model. A total of 68 female rats were randomly divided into 8 groups. The first 4 groups were designed as controls for cancer and treatment groups, and the control groups were composed of only control (C), Pomegranate (P), Tangeretin (T), and Pomegranate+Tangeretin (P+T) groups. The other four groups were designed as cancer and treatment groups and were composed of DMBA (D) and DMBA+Pomegranate (D+P), DMBA+Tangeretin (D+T), DMBA+Pomegranate+Tangeretin (D+P+T) groups. Tumor markers and angiogenesis parameters were studied from plasma samples obtained from rats. Histopathological, immunohistochemical, and TUNEL analyses and expressions of proteins affecting apoptosis and cell cycle were determined in breast tissue samples. In the DMBA group, plasma CA15-3, CEA, VEGF, MMP-9, and NF-κB levels were significantly increased compared to the controls, but significant decreases were observed in these parameters except MMP-9 in the treatment groups. It was observed that p53 and Bax expressions significantly increased in both D+P and D+P+T groups compared to the DMBA group, and these findings were supported by Tunel and immunohistochemical findings. Cyclin D1 expressions were found to be significantly decreased only in the D+T group and supported by TUNEL and immunohistochemical findings. Immunohistochemical ER-α and Ki-67 immune reactivities were significantly decreased in all treatment groups compared to the DMBA group. Our results showed that combined application of pomegranate extract and tangeretin may be more beneficial in preventing breast cancer development.
Collapse
Affiliation(s)
- Huseyin Fatih Gul
- Department of Medical Biochemistry, Kafkas University, Faculty of Medicine, Kars, Turkey.
| | - Necip Ilhan
- Department of Medical Biochemistry, Firat University, Faculty of Medicine, Elazıg, Turkey
| | - Nevin Ilhan
- Department of Medical Biochemistry, Firat University, Faculty of Medicine, Elazıg, Turkey
| | | | - Tuncay Kuloglu
- Department of Histology and Embryology, Firat University, Faculty of Medicine, Elazıg, Turkey
| |
Collapse
|
45
|
Pather K, Augustine TN. Tamoxifen induces hypercoagulation and alterations in ERα and ERβ dependent on breast cancer sub-phenotype ex vivo. Sci Rep 2020; 10:19256. [PMID: 33159119 PMCID: PMC7648622 DOI: 10.1038/s41598-020-75779-y] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2020] [Accepted: 09/23/2020] [Indexed: 12/24/2022] Open
Abstract
Tamoxifen shows efficacy in reducing breast cancer-related mortality but clinically, is associated with increased risk for thromboembolic events. We aimed to determine whether breast tumour sub-phenotype could predict propensity for thrombosis. We present two ex vivo Models of Tamoxifen-therapy, Model 1 in which treatment recapitulates accumulation within breast tissue, by treating MCF7 and T47D cells directly prior to exposure to blood constituents; and Model 2 in which we recreate circulating Tamoxifen by treating blood constituents prior to exposure to cancer cells. Blood constituents included whole blood, platelet-rich plasma and platelet-poor plasma. Hypercoagulation was assessed as a function of thrombin activity, expression of CD62P and CD63 activation markers defined as an index of platelet activation, and platelet morphology; while oestrogen receptor expression was assessed using immunocytochemistry with quantitative analysis. We determined, in concert with clinical studies and contrary to selected laboratory investigations, that Tamoxifen induces hypercoagulation, dependent on sub-phenotypes, with the T47D cell line capacity most enhanced. We determined a weak positive correlation between oestrogen receptor expression, and CD62P and CD63; indicating an association between tumour invasion profiles and hypercoagulation, however, other yet unknown factors may play a predictive role in defining hypercoagulation.
Collapse
Affiliation(s)
- K Pather
- School of Anatomical Sciences, Faculty of Health Sciences, University of the Witwatersrand, 7 York Road, Parktown, Johannesburg, 2193, South Africa.
| | - T N Augustine
- School of Anatomical Sciences, Faculty of Health Sciences, University of the Witwatersrand, 7 York Road, Parktown, Johannesburg, 2193, South Africa.
| |
Collapse
|
46
|
Lu JJ, Zhou FM, Hu XJ, Fang JJ, Liu CX, Zhu BQ, Ding ZS. Molecular docking simulation and in vitro studies on estrogenic activities of flavonoids from leaves of Carya cathayensis Sarg. Steroids 2020; 163:108726. [PMID: 32889051 DOI: 10.1016/j.steroids.2020.108726] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/23/2020] [Revised: 08/18/2020] [Accepted: 08/25/2020] [Indexed: 12/11/2022]
Abstract
The main purpose of this study was to evaluate the estrogenic properties of total flavonoids (TFs) and five flavonoid monomers (cardamonin (Car), pinostrobin chalcone (PC), wogonin (Wo), chrysin (Chr) and Pinocembrin (PI)) from leaves of Carya cathayensis Sarg (LCC). TFs from LCC were isolated and determined using HPLC. The 3-[4,5-dimethylthiazole-2-yl]-2,5-diphenyltetrazolium bromide (MTT) assay and flow cytometry were performed to assess the effects of flavonoids on cell proliferation and cell cycle, respectively. The molecular docking technique was applied to investigate binding conformations of the monomers from LCC to the estrogen receptor ERα and ERβ. Gene and protein expression patterns were assessed using quantitative real-time PCR (qRT-PCR) and western blot, respectively. The results showed that TFs, Car, PC, Wo and Chr promoted proliferation of MCF-7 cells and cell transition from the G1 to S phase, and inhabitation of MCF-7 cell proliferation was observed after the treatment of PI. Molecular docking studies confirmed ERs as molecular targets for the monomers. TFs, Car, PC, Wo and Chr from LCC promoted gene expression of ERα, ERβ, progesterone receptor (PR) and pS2. Our collective results demonstrated that TFs and monomers from LCC may exert ER agonist activity through competitively bind to ER, inducing ER upregulation and active ER to estrogen response element (ERE)- independent gene regulation. As an abundant natural product, LCC may provide a novel medicinal source for treatment of diseases caused by estrogen deficiency.
Collapse
Affiliation(s)
- Jing-Jing Lu
- College of Life Science, Zhejiang Chinese Medical University, Hangzhou 310053, Zhejiang, China
| | - Fang-Mei Zhou
- College of Medical Technology, Zhejiang Chinese Medical University, Hangzhou 310053, Zhejiang, China
| | - Xu-Jiao Hu
- Yinzhou People's Hospital, Ningbo, Zhejiang Province, China
| | - Jing-Jing Fang
- Yinzhou People's Hospital, Ningbo, Zhejiang Province, China
| | - Cai-Xia Liu
- College of Life Science, Zhejiang Chinese Medical University, Hangzhou 310053, Zhejiang, China
| | - Bing-Qi Zhu
- College of Medical Technology, Zhejiang Chinese Medical University, Hangzhou 310053, Zhejiang, China
| | - Zhi-Shan Ding
- College of Medical Technology, Zhejiang Chinese Medical University, Hangzhou 310053, Zhejiang, China.
| |
Collapse
|
47
|
Treeck O, Schüler-Toprak S, Ortmann O. Estrogen Actions in Triple-Negative Breast Cancer. Cells 2020; 9:cells9112358. [PMID: 33114740 PMCID: PMC7692567 DOI: 10.3390/cells9112358] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2020] [Revised: 10/15/2020] [Accepted: 10/21/2020] [Indexed: 12/11/2022] Open
Abstract
Triple-negative breast cancer (TNBC) lacks estrogen receptor (ER) α, but the expression of estrogen receptors ERβ and G protein-coupled estrogen receptor 1 (GPER-1) is able to trigger estrogen-responsivity in TNBC. Estrogen signaling in TNBC can also be activated and modulated by the constitutively active estrogen-related receptors (ERRs). In this review article, we discuss the role of ERβ and GPER-1 as mediators of E2 action in TNBC as well as the function of ERRs as activators and modulators of estrogen signaling in this cancer entity. For this purpose, original research articles on estrogen actions in TNBC were considered, which are listed in the PubMed database. Additionally, we performed meta-analyses of publicly accessible integrated gene expression and survival data to elucidate the association of ERβ, GPER-1, and ERR expression levels in TNBC with survival. Finally, options for endocrine therapy strategies for TNBC were discussed.
Collapse
|
48
|
Cross-talk between the ER pathway and the lncRNA MAFG-AS1/miR-339-5p/ CDK2 axis promotes progression of ER+ breast cancer and confers tamoxifen resistance. Aging (Albany NY) 2020; 12:20658-20683. [PMID: 33098638 PMCID: PMC7655217 DOI: 10.18632/aging.103966] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2020] [Accepted: 07/07/2020] [Indexed: 12/12/2022]
Abstract
Hormone receptor-positive breast cancer accounts for around 75% of breast cancers. The estrogen receptor pathway promotes tumor progression and endocrine resistance. Recently, the cross-talk between the ER signaling pathway and cell cycle regulation has been identified. It is necessary to determine the underlying molecular mechanisms involved in the ER signaling pathway and find new target genes for prognosis and drug resistance in ER+ breast cancer. In this study, lncRNA MAFG-AS1 was shown to be up-regulated and associated with poor prognosis in ER+ breast cancer. Functionally, down-regulation of MAFG-AS1 could inhibit cell proliferation and promote apoptosis. In addition, MAFG-AS1 which contained an estrogen-responsive element could promote CDK2 expression by sponging miR-339-5p. Subsequently, MAFG-AS1 and CDK2 were found to be up-regulated in tamoxifen-resistant MCF-7 cells. Cross-talk between the ER signaling pathway and cell cycle conducted by MAFG-AS1 and CDK2 could promote tamoxifen resistance. In conclusion, our study indicated that estrogen-responsive lncRNA MAFG-AS1 up-regulated CDK2 by sponging miR-339-5p, which promoted ER+ breast cancer proliferation. Cross-talk between the ER signaling pathway and cell cycle suggested that lncRNA MAFG-AS1 is a potential biomarker and therapeutic target in ER+ breast cancer. CDK2 inhibitors may be applied to endocrine resistance therapy.
Collapse
|
49
|
Ye J, Tian T, Chen X. The efficacy of gefitinib supplementation for breast cancer: A meta-analysis of randomized controlled studies. Medicine (Baltimore) 2020; 99:e22613. [PMID: 33120749 PMCID: PMC7581042 DOI: 10.1097/md.0000000000022613] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
Abstract
INTRODUCTION The efficacy of gefitinib supplementation for breast cancer remains controversial. We conduct a systematic review and meta-analysis to explore the influence of gefitinib supplementation vs placebo on the efficacy of breast cancer. METHODS We have searched PubMed, EMbase, Web of science, EBSCO, and Cochrane library databases through February 2019 and included randomized controlled trials assessing the effect of gefitinib supplementation vs placebo on overall response for breast cancer patients. This meta-analysis was performed using the random-effect model. RESULTS Seven randomized controlled trials involving 927 patients were included in the meta-analysis. Overall, compared with control group for breast cancer, gefitinib supplementation revealed no obvious impact on complete response (risk ration [RR] = 1.19; 95% confidence interval [CI] = 0.58 to 2.44; P = .63), progressive disease (RR = 0.81; 95% CI = 0.59-1.11; P = .18), partial response (RR = 0.67; 95% CI = 0.36-1.25; P = .21), stable disease (RR = 1.02; 95% CI = 0.65-1.60; P = .92), nausea or vomiting (RR = 0.99; 95% CI = 0.73-1.33; P = .93), but was associated with increased incidence of diarrhea (RR = 2.80; 95% CI = 2.23-3.52; P < .00001), decreased incidence of hot flash (RR = 0.53; 95% CI = 0.37-0.78; P = .001), and improved incidence of adverse events (RR = 1.12; 95% CI = 1.05-1.19; P = .0006). CONCLUSIONS Gefitinib supplementation may provide no positive effect on complete response, progressive disease, partial response or stable disease for breast cancer patients, but with the increase in adverse events.
Collapse
Affiliation(s)
- Jing Ye
- Department of Surgery, Chongqing Yongchuan Health Center for Women and Children
| | - Tian Tian
- Department of General Surgery, Yongchuan Hospital of Chongqing Medical University, Chongqing, China
| | - Xiaopin Chen
- Department of General Surgery, Yongchuan Hospital of Chongqing Medical University, Chongqing, China
| |
Collapse
|
50
|
Niu Z, Li X, Feng S, Huang Q, Zhuang T, Yan C, Qian H, Ding Y, Zhu J, Xu W. The deubiquitinating enzyme USP1 modulates ERα and modulates breast cancer progression. J Cancer 2020; 11:6992-7000. [PMID: 33123289 PMCID: PMC7591989 DOI: 10.7150/jca.50477] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2020] [Accepted: 09/27/2020] [Indexed: 12/27/2022] Open
Abstract
Breast cancer is one of the most common malignancies worldwide, while the luminal types (ERα positive) accounts for two third of all breast cancer cases. Although ERα positive breast cancer could be effective controlled by endocrine therapy, most of the patients will develop endocrine resistance, which becomes a headache clinical issue for breast cancer field. Endocrine resistance could be caused by multiple pathway disorders, the dys-regulation of ERα signaling might be a critical factor, which makes it urgent and important to reveal the potential molecular mechanism of ERα signaling. In our current study, we identified a new deubiquitination enzyme USP1 through screening the whole DUB (Deubiquitinases) siRNA library. The expression of USP1 is elevated in human breast cancer compared with normal mammary tissues. Importantly, USP1 expression levels are specially correlated with poor survival in ERα positive patients. USP1 depletion inhibited breast cancer cell progression and ERα signaling activity. Immuno-precipitation assays indicate that USP1 associates with ERα and promotes its stability possibly via inhibiting ERα K48-linked poly-ubiquitination. In conclusion, our data implicate a non-genomic mechanism by USP1 via stabilizing ERα protein controls ERα target gene expression linked to breast cancer progression.
Collapse
Affiliation(s)
- Zhiguo Niu
- Jiangsu Key Laboratory of Medical Science and Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu, 212000, China.,Henan Key Laboratory of Immunology and Targeted Drugs, School of Laboratory Medicine, Xinxiang Medical University, 453000, China
| | - Xin Li
- Henan Key Laboratory of Immunology and Targeted Drugs, School of Laboratory Medicine, Xinxiang Medical University, 453000, China
| | - Suyin Feng
- Department of Neurosurgery, Affiliated Hospital of Jiangnan University, 200 Huihe Road, Wuxi, 214000, Jiangsu, China
| | - Qingsong Huang
- Henan Key Laboratory of Immunology and Targeted Drugs, School of Laboratory Medicine, Xinxiang Medical University, 453000, China
| | - Ting Zhuang
- Henan Key Laboratory of Immunology and Targeted Drugs, School of Laboratory Medicine, Xinxiang Medical University, 453000, China
| | - Cheng Yan
- School of Medicine, Xinxiang University, Xinxiang, 453003 Henan P.R. China
| | - Hui Qian
- Jiangsu Key Laboratory of Medical Science and Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu, 212000, China
| | - Yinlu Ding
- Department of general surgery, the Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China, 250033
| | - Jian Zhu
- Department of general surgery, the Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China, 250033.,Henan Key Laboratory of Immunology and Targeted Drugs, School of Laboratory Medicine, Xinxiang Medical University, 453000, China
| | - Wenrong Xu
- Jiangsu Key Laboratory of Medical Science and Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu, 212000, China
| |
Collapse
|