1
|
Xiang Z, Ma B, Pei X, Wang W, Gong W. Mechanism of action of genistein on breast cancer and differential effects of different age stages. PHARMACEUTICAL BIOLOGY 2025; 63:141-155. [PMID: 39996512 PMCID: PMC11864014 DOI: 10.1080/13880209.2025.2469607] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/08/2024] [Revised: 02/12/2025] [Accepted: 02/14/2025] [Indexed: 02/26/2025]
Abstract
CONTEXT Genistein, a soy-derived isoflavone, exhibits structural similarities with 17β-estradiol and demonstrates antioxidant, anti-inflammatory, and estrogenic properties. Despite its low bioavailability limiting its clinical application, it shows potential for breast cancer prevention and treatment. OBJECTIVE This review aims to summarize the pharmacological effects and molecular mechanisms of genistein in breast cancer, focusing on its therapeutic potential, strategies to overcome bioavailability limitations, and its role in personalized medicine. Differential impacts among population subgroups are also discussed. METHODS A systematic review was conducted using PubMed, ScienceDirect, and Google Scholar databases. Studies were selected based on their focus on genistein's mechanisms of action, strategies to enhance its bioavailability, and interactions with other therapies. RESULTS Genistein exerted anticancer effects by modulating estrogen receptor β (ERβ), inhibiting angiogenesis, arresting the cell cycle, and inducing apoptosis. Its antioxidant properties help mitigate tumor-associated oxidative stress. Bioavailability enhancement strategies, such as nanoparticle and lipid-based formulations, show promise. Age-dependent effects were evident, with distinct responses observed in prepubertal, menopausal, and postmenopausal populations, underscoring its potential for personalized therapies. Furthermore, genistein influences epigenetic modifications, including DNA methylation and miRNA expression, bolstering its anticancer efficacy. CONCLUSION Genistein is a promising candidate for breast cancer therapy, particularly for personalized treatment. Strategies to enhance bioavailability and further clinical research are essential to optimize its therapeutic potential and evaluate its efficacy in combination therapies.
Collapse
Affiliation(s)
- Zhebin Xiang
- Zhejiang Chinese Medical University, Hangzhou, China
| | - Bo Ma
- Zhejiang Hospital, Hangzhou, China
| | - Xiujun Pei
- Shandong Provincial Hospital, Shandong, China
| | - Wenjie Wang
- Zhejiang Chinese Medical University, Hangzhou, China
| | - Weilun Gong
- Zhejiang Chinese Medical University, Hangzhou, China
| |
Collapse
|
2
|
Neale DA, Morris JC, Verrills NM, Ammit AJ. Understanding the regulatory landscape of protein phosphatase 2A (PP2A): Pharmacological modulators and potential therapeutics. Pharmacol Ther 2025; 269:108834. [PMID: 40023321 DOI: 10.1016/j.pharmthera.2025.108834] [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: 10/21/2024] [Revised: 01/20/2025] [Accepted: 02/20/2025] [Indexed: 03/04/2025]
Abstract
Protein phosphatase 2A (PP2A) is a ubiquitously expressed serine/threonine phosphatase with a diverse and integral role in cellular signalling pathways. Consequently, its dysfunction is frequently observed in disease states such as cancer, inflammation and Alzheimer's disease. A growing understanding of both PP2A and its endogenous regulatory proteins has presented numerous targets for therapeutic intervention. This provides important context for the dynamic control and dysregulation of PP2A function in disease states. Understanding the intricate regulation of PP2A signalling in disease has resulted in the development of novel pharmacological agents aimed at restoring cellular homeostasis. Herein we review the structure and function of PP2A together with pharmacological modulators, both endogenous (proteins) and exogenous (small molecules and peptides), with relevance to targeting PP2A as a future pharmacotherapeutic strategy.
Collapse
Affiliation(s)
- David A Neale
- School of Chemistry, UNSW Sydney, NSW 2052, Australia
| | | | - Nicole M Verrills
- School of Biomedical Sciences and Pharmacy, Faculty of Health and Medicine, University of Newcastle, NSW 2308, Australia; Precision Medicine Program, Hunter Medical Research Institute, New Lambton, NSW 2305, Australia
| | - Alaina J Ammit
- Woolcock Emphysema Centre, Woolcock Institute of Medical Research, Macquarie University, NSW, Australia; School of Life Sciences, Faculty of Science, University of Technology Sydney, NSW, Australia.
| |
Collapse
|
3
|
Kaufman-Szymczyk A, Jalmuzna J, Lubecka-Gajewska K. Soy-derived isoflavones as chemo-preventive agents targeting multiple signalling pathways for cancer prevention and therapy. Br J Pharmacol 2025; 182:2259-2286. [PMID: 38528688 DOI: 10.1111/bph.16353] [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: 08/07/2023] [Revised: 01/19/2024] [Accepted: 02/08/2024] [Indexed: 03/27/2024] Open
Abstract
The chemopreventive and chemotherapeutic properties of soy and soy-derived compounds, especially isoflavones, have been extensively studied in recent years. However, in contrast to their anticancer effects, such as cell growth inhibition, cell cycle arrest and apoptosis induction, isoflavones have also been found to promote the growth of cancer cells. Therefore, the aim of this comprehensive review article is to present the current state of knowledge regarding the molecular mechanisms by which soy-derived isoflavones target multiple cellular signalling pathways in cancer cells. Our findings indicate that soy-derived isoflavones act as, among other things, potent modulators of HOX transcript antisense RNA (HOTAIR)/SWI/SNF-related matrix-associated actin-dependent regulator of chromatin subfamily B member 1 (SMARCB1), vascular endothelial growth factor (VEGF)/C-X-C motif chemokine ligand 12 (CXCL12)/C-X-C motif chemokine receptor type 4 (CXCR4), 17-β-oestradiol (E2)/oestrogen receptor-α (ERα)/neuroglobin (NGB) and sonic hedgehog signalling pathways, epigenetic modulatory agents (i.a. miR-155, miR-34a and miR-10a-5p) and cancer stem cells and epithelial-to-mesenchymal transition inhibitors. The paper also discusses the latest epidemiological studies and clinical trials and provides an insight into recent extensive research on the chemo-preventive and therapeutic potential of soy-derived isoflavones. LINKED ARTICLES: This article is part of a themed issue Natural Products and Cancer: From Drug Discovery to Prevention and Therapy. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v182.10/issuetoc.
Collapse
Affiliation(s)
- Agnieszka Kaufman-Szymczyk
- Department of Biomedical Chemistry, Faculty of Health Sciences, Medical University of Łódź, Łódź, Poland
| | - Justyna Jalmuzna
- Department of Biomedical Chemistry, Faculty of Health Sciences, Medical University of Łódź, Łódź, Poland
| | - Katarzyna Lubecka-Gajewska
- Department of Biomedical Chemistry, Faculty of Health Sciences, Medical University of Łódź, Łódź, Poland
| |
Collapse
|
4
|
Naponelli V, Piscazzi A, Mangieri D. Cellular and Molecular Mechanisms Modulated by Genistein in Cancer. Int J Mol Sci 2025; 26:1114. [PMID: 39940882 PMCID: PMC11818640 DOI: 10.3390/ijms26031114] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2024] [Revised: 01/21/2025] [Accepted: 01/23/2025] [Indexed: 02/16/2025] Open
Abstract
Genistein (4',5,7-trihydroxyisoflavone) is a phytoestrogen belonging to a subclass of natural flavonoids that exhibits a wide range of pharmacological functions, including antioxidant and anti-inflammatory properties. These characteristics make genistein a valuable phytochemical compound for the prevention and/or treatment of cancer. Genistein effectively inhibits tumor growth and dissemination by modulating key cellular mechanisms. This includes the suppression of angiogenesis, the inhibition of epithelial-mesenchymal transition, and the regulation of cancer stem cell proliferation. These effects are mediated through pivotal signaling pathways such as JAK/STAT, PI3K/Akt/mTOR, MAPK/ERK, NF-κB, and Wnt/β-catenin. Moreover, genistein interferes with the function of specific cyclin/CDK complexes and modulates the activation of Bcl-2/Bax and caspases, playing a critical role in halting tumor cell division and promoting apoptosis. The aim of this review is to discuss in detail the key cellular and molecular mechanisms underlying the pleiotropic anticancer effects of this flavonoid.
Collapse
Affiliation(s)
- Valeria Naponelli
- Department of Medicine and Surgery, University of Parma, Plesso Biotecnologico Integrato, Via Volturno 39, 43126 Parma, Italy
| | - Annamaria Piscazzi
- Department of Clinical and Experimental Medicine, University of Foggia, Via Pinto 1, 71122 Foggia, Italy
| | - Domenica Mangieri
- Department of Clinical and Experimental Medicine, University of Foggia, Via Pinto 1, 71122 Foggia, Italy
| |
Collapse
|
5
|
Chaichian S, Nikfar B, Bidgoli SA, Moazzami B. The Role of Genistein and its Derivatives in Ovarian Cancer: New Perspectives for Molecular Mechanisms and Clinical Applications. Curr Med Chem 2025; 32:907-922. [PMID: 37921172 DOI: 10.2174/0109298673251713231019091910] [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: 03/10/2023] [Revised: 08/09/2023] [Accepted: 09/15/2023] [Indexed: 11/04/2023]
Abstract
Genistein (GEN) is a member of the polyphenol family, known chiefly for its effects on metabolic diseases and gynecological disorders. GEN has anti-cancer properties by inhibiting tumor proliferation, tumor metastasis, invasion, migration, and inducing apoptosis. Ovarian cancer (OC) is ranked 7th among the most common gynecological cancers. Despite its low incidence compared to other cancers, it is the first cause of death among gynecologic malignancies. Surgery and chemotherapy are the main options for treating this fatal cancer. Therefore, further investigations into GEN may aid in the discovery of novel therapeutics for preventing and/or treating OC. In this review, we aim to investigate the role of GEN in ovarian cancer. We investigate the anti-tumor effects of GEN on OC cell lines, including inducing apoptosis, suppressing tumor growth, and inhibiting metastasis. Also, we review the studies investigating GEN's roles as an adjuvant in therapeutic regimens with other chemotherapeutic agents (e.g., cisplatin, quercetin, and gemcitabine).
Collapse
Affiliation(s)
- Shahla Chaichian
- Endometriosis Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Banafsheh Nikfar
- Pars Advanced and Minimally Invasive Medical Manners Research Center, Pars Hospital, Iran University of Medical Sciences, Tehran, Iran
| | - Sepideh Arbabi Bidgoli
- Pars Advanced and Minimally Invasive Medical Manners Research Center, Pars Hospital, Iran University of Medical Sciences, Tehran, Iran
- Department of Toxicology and Pharmacology, Faculty of Pharmacy and Pharmaceutical Sciences, Islamic Azad University, Tehran Medical Sciences University, Tehran, Iran
| | - Bahram Moazzami
- Pars Advanced and Minimally Invasive Medical Manners Research Center, Pars Hospital, Iran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
6
|
Yapar EA, Ozdemir MN, Cavalu S, Dagıstan ÖA, Ozsoy Y, Kartal M. Phytoactive Molecules and Nanodelivery Approaches for Breast Cancer Treatment: Current and Future Perspectives. Curr Pharm Biotechnol 2025; 26:795-812. [PMID: 38859783 DOI: 10.2174/0113892010299183240529094844] [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: 01/26/2024] [Revised: 04/21/2024] [Accepted: 04/29/2024] [Indexed: 06/12/2024]
Abstract
One of the most common malignancies in women, breast cancer accounts for nearly 25% of all cancer cases. Breast cancer is a diverse cancer form that exhibits variability in both morphology and molecular characteristics and is linked to numerous risk factors. Although various approaches and research are ongoing in the treatment and prevention of breast cancer, medication resistance in the current breast cancer treatment contributes to the disease's relapse and recurrence. Phytoactive molecules are the subject of growing research in both breast cancer prevention and treatment, but currently used conventional medicines and techniques limit their application. In recent years, significant advancements have been made in the field of nanotechnology, which has proven to be essential in the fight against drug resistance. The transport of synthetic and natural anticancer molecules via nanocarriers has recently been added to breast cancer therapy, greatly alleviating the constraints of the current approach. In light of these developments, interest in nano-delivery studies of phytoactive molecules has also increased. In this review, research of phytoactive molecules for breast cancers along with their clinical studies and nanoformulations, was presented from current and future perspectives.
Collapse
Affiliation(s)
- Evren Algın Yapar
- Department of Pharmaceutical Technology, Faculty of Pharmacy, Sivas Cumhuriyet University, Sivas, Türkiye
| | - Merve Nur Ozdemir
- Department of Pharmaceutical Technology, Faculty of Pharmacy, Sivas Cumhuriyet University, Sivas, Türkiye
| | - Simona Cavalu
- Department of Preclinical Sciences, Faculty of Medicine and Pharmacy, University of Oradea, Bihor, România
| | - Özlem Akbal Dagıstan
- Department of Pharmaceutical Technology, Faculty of Pharmacy, Istanbul University, Istanbul, Türkiye
| | - Yıldız Ozsoy
- Department of Pharmaceutical Technology, Faculty of Pharmacy, Istanbul University, Istanbul, Türkiye
| | - Murat Kartal
- Faculty of Pharmacy, Bezmialem Vakıf University, Department of Pharmacognosy, Istanbul, Türkiye
| |
Collapse
|
7
|
Aly SH, Abulsoud AI, Moustafa YM, Abdel Mageed SS, Abdelmaksoud NM, El-Dakroury WA, Mohammed OA, Abdel-Reheim MA, Zaki MB, Rizk NI, Elshafei A, Elimam H, Ashraf A, Doghish AS. Harnessing natural compounds to modulate miRNAs in breast cancer therapy. Funct Integr Genomics 2024; 24:211. [PMID: 39528871 DOI: 10.1007/s10142-024-01489-7] [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: 10/14/2024] [Revised: 10/19/2024] [Accepted: 10/24/2024] [Indexed: 11/16/2024]
Abstract
Breast cancer's complexity and heterogeneity continue to present significant challenges in its treatment and management. Emerging research has underscored the pivotal role of microRNAs (miRNAs) in breast cancer pathogenesis, acting as crucial regulators of gene expression. This review delivers an in-depth analysis of miRNAs, highlighting their dual functions as both oncogenes and tumor suppressors, and detailing their impact on key biological processes, including cell proliferation, apoptosis, and metastasis. The mechanisms underlying miRNA action, particularly their interactions with target mRNAs and the factors influencing these dynamics, are thoroughly explored. Additionally, the review discusses the therapeutic prospects of miRNAs, with a focus on innovative delivery systems like nanoparticles that improve the stability and effectiveness of miRNA-based therapies. It also addresses the anticancer effects of natural compounds, such as genistein, hesperidin, quercetin, curcumin, resveratrol, epigallocatechin-3-gallate (EGCG), and glyceollins, which modulate miRNA expression and contribute to tumor growth inhibition. These advances seek to address the limitations of conventional therapies, paving the way for targeted interventions in breast cancer. By integrating current insights on miRNA biology, therapeutic strategies, and the potential of natural products to regulate miRNA expression, this review aims to shed light on miRNA- and natural product-based approaches as promising avenues for enhancing breast cancer treatment outcomes.
Collapse
Affiliation(s)
- Shaza H Aly
- Department of Pharmacognosy, Faculty of Pharmacy, Badr University in Cairo (BUC), Badr City, Cairo, 11829, Egypt
| | - Ahmed I Abulsoud
- Biochemistry and Molecular Biology Department, Faculty of Pharmacy (Boys), Al-Azhar University, Nasr City, Cairo, 11231, Egypt
- Biochemistry Department, Faculty of Pharmacy, Heliopolis University, Cairo, 11785, Egypt
| | - Yasser M Moustafa
- Pharmacology and Toxicology Department, Faculty of Pharmacy, Badr University in Cairo (BUC), Badr City, Cairo, 11829, Egypt
| | - Sherif S Abdel Mageed
- Pharmacology and Toxicology Department, Faculty of Pharmacy, Badr University in Cairo (BUC), Badr City, Cairo, 11829, Egypt
| | - Nourhan M Abdelmaksoud
- Biochemistry Department, Faculty of Pharmacy, Heliopolis University, Cairo, 11785, Egypt
| | - Walaa A El-Dakroury
- Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmacy, Badr University in Cairo (BUC), Badr City, Cairo, 11829, Egypt
| | - Osama A Mohammed
- Department of Pharmacology, College of Medicine, University of Bisha, Bisha, 61922, Saudi Arabia
| | | | - Mohamed Bakr Zaki
- Department of Biochemistry, Faculty of Pharmacy, University of Sadat City, Sadat, Menoufia, 32897, Egypt
| | - Nehal I Rizk
- Department of Biochemistry, Faculty of Pharmacy and Drug Technology, Egyptian Chinese University, Cairo, 11786, Egypt
| | - Ahmed Elshafei
- Biochemistry and Molecular Biology Department, Faculty of Pharmacy (Boys), Al-Azhar University, Nasr City, Cairo, 11231, Egypt
| | - Hanan Elimam
- Department of Biochemistry, Faculty of Pharmacy, University of Sadat City, Sadat, Menoufia, 32897, Egypt
| | - Alaa Ashraf
- Department of Clinical Pharmacy and Pharmacy Practice, Faculty of Pharmacy, Badr University in Cairo (BUC), Badr City, Cairo, 11829, Egypt
| | - Ahmed S Doghish
- Department of Biochemistry, Faculty of Pharmacy, Badr University in Cairo (BUC), Badr City, Cairo, 11829, Egypt.
- Faculty of Pharmacy (Boys), Al-Azhar University, Nasr City, Cairo, 11231, Egypt.
| |
Collapse
|
8
|
Mao K, Wang X, Hou Y, He X, Geng S, Sadiq FA, Lian Y, Sang Y. Integrated network pharmacology and transcriptomic approach reveal the role of equol in reducing colorectal cancer via regulating multiple cell cycle genes in HCT116 cells. Int J Biol Macromol 2024; 282:136832. [PMID: 39461627 DOI: 10.1016/j.ijbiomac.2024.136832] [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: 07/24/2024] [Revised: 10/15/2024] [Accepted: 10/21/2024] [Indexed: 10/29/2024]
Abstract
Equol is an isoflavone-derived metabolite known to exhibit strong estrogenic and antioxidant activities. The aim of this paper is twofold: first, to confirm the anticancer potential of equol against colorectal cancer, and second, to reveal the underlying mechanisms. After treatment with 40 μg/mL equol, cell proliferation, cell migration, and colony formation of HCT116 colon cancer cells were inhibited. Network pharmacology and transcriptomics analysis revealed the downregulation of genes related to DNA replication (CCND1, E2F1, CDC6, CDC45, MCM4), leading to G1/S cell cycle arrest and the induction of cell apoptosis, which was confirmed by flow cytometry. Genes associated with the G2-to-M transition (CDK1, CCNA2, CCNB1) were also downregulated. In addition, equol downregulated genes (FOXM1 and ASPM) that control cell migration and invasion. Our data indicate that equol can inhibit colorectal cancer by targeting multiple pathways, suggesting its potential as a key component in the adjuvant treatment of colorectal cancer.
Collapse
Affiliation(s)
- Kemin Mao
- Department of Food Science and Technology, Hebei Agricultural University, Baoding, Hebei, China
| | - Xianghong Wang
- Department of Food Science and Technology, Hebei Agricultural University, Baoding, Hebei, China
| | - Yakun Hou
- Department of Food Science and Technology, Hebei Agricultural University, Baoding, Hebei, China
| | - Xiaowei He
- Department of Food Science and Technology, Hebei Agricultural University, Baoding, Hebei, China
| | - Shuo Geng
- Department of Food Science and Technology, Hebei Agricultural University, Baoding, Hebei, China
| | - Faizan Ahmed Sadiq
- Advanced Therapies Group, School of Dentistry, College of Biomedical and Life Sciences, Cardiff University, Cardiff CF14 4XY, United Kingdom
| | - Yunhe Lian
- Chenguang Biotech Group Co., Ltd., Handan, Hebei, China
| | - Yaxin Sang
- Department of Food Science and Technology, Hebei Agricultural University, Baoding, Hebei, China.
| |
Collapse
|
9
|
Choudhury SD, Kumar P, Choudhury D. Bioactive nutraceuticals as G4 stabilizers: potential cancer prevention and therapy-a critical review. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2024; 397:3585-3616. [PMID: 38019298 DOI: 10.1007/s00210-023-02857-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/14/2023] [Accepted: 11/13/2023] [Indexed: 11/30/2023]
Abstract
G-quadruplexes (G4) are non-canonical, four-stranded, nucleic acid secondary structures formed in the guanine-rich sequences, where guanine nucleotides associate with each other via Hoogsteen hydrogen bonding. These structures are widely found near the functional regions of the mammalian genome, such as telomeres, oncogenic promoters, and replication origins, and play crucial regulatory roles in replication and transcription. Destabilization of G4 by various carcinogenic agents allows oncogene overexpression and extension of telomeric ends resulting in dysregulation of cellular growth-promoting oncogenesis. Therefore, targeting and stabilizing these G4 structures with potential ligands could aid cancer prevention and therapy. The field of G-quadruplex targeting is relatively nascent, although many articles have demonstrated the effect of G4 stabilization on oncogenic expressions; however, no previous study has provided a comprehensive analysis about the potency of a wide variety of nutraceuticals and some of their derivatives in targeting G4 and the lattice of oncogenic cell signaling cascade affected by them. In this review, we have discussed bioactive G4-stabilizing nutraceuticals, their sources, mode of action, and their influence on cellular signaling, and we believe our insight would bring new light to the current status of the field and motivate researchers to explore this relatively poorly studied arena.
Collapse
Affiliation(s)
- Satabdi Datta Choudhury
- Department of Chemistry and Biochemistry, Thapar Institute of Engineering and Technology, Patiala, Punjab, 147004, India
| | - Prateek Kumar
- School of Basic Sciences, Indian Institute of Technology (IIT), Mandi, Himachal Pradesh, 175005, India
| | - Diptiman Choudhury
- Department of Chemistry and Biochemistry, Thapar Institute of Engineering and Technology, Patiala, Punjab, 147004, India.
- Centre for Excellence in Emerging Materials, Thapar Institute of Engineering and Technology, Patiala, Punjab, 147004, India.
| |
Collapse
|
10
|
Konstantinou EK, Gioxari A, Dimitriou M, Panoutsopoulos GI, Panagiotopoulos AA. Molecular Pathways of Genistein Activity in Breast Cancer Cells. Int J Mol Sci 2024; 25:5556. [PMID: 38791595 PMCID: PMC11122029 DOI: 10.3390/ijms25105556] [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: 04/18/2024] [Revised: 05/12/2024] [Accepted: 05/16/2024] [Indexed: 05/26/2024] Open
Abstract
The most common malignancy in women is breast cancer. During the development of cancer, oncogenic transcription factors facilitate the overproduction of inflammatory cytokines and cell adhesion molecules. Antiapoptotic proteins are markedly upregulated in cancer cells, which promotes tumor development, metastasis, and cell survival. Promising findings have been found in studies on the cell cycle-mediated apoptosis pathway for medication development and treatment. Dietary phytoconstituents have been studied in great detail for their potential to prevent cancer by triggering the body's defense mechanisms. The underlying mechanisms of action may be clarified by considering the role of polyphenols in important cancer signaling pathways. Phenolic acids, flavonoids, tannins, coumarins, lignans, lignins, naphthoquinones, anthraquinones, xanthones, and stilbenes are examples of natural chemicals that are being studied for potential anticancer drugs. These substances are also vital for signaling pathways. This review focuses on innovations in the study of polyphenol genistein's effects on breast cancer cells and presents integrated chemical biology methods to harness mechanisms of action for important therapeutic advances.
Collapse
Affiliation(s)
| | | | | | | | - Athanasios A. Panagiotopoulos
- Department of Nutritional Science and Dietetics, School of Health Sciences, University of the Peloponnese, Antikalamos, 24100 Kalamata, Greece; (E.K.K.); (A.G.); (M.D.); (G.I.P.)
| |
Collapse
|
11
|
Ubhenin AE, Adefolalu AA, Oriakhi K, Adamude FA, Dingwoke EJ, Ikebuiro JO, Chiwendu BC, Muhammad ML, Omage K. Caesalpinia pulcherrima lowered serum carcinoembryonic antigen and antigen 125 in 7,12-Dimethylbenz[ a]anthracene-induced Mammary Carcinogenesis in Female Albino Rats. Heliyon 2024; 10:e23401. [PMID: 38187255 PMCID: PMC10770447 DOI: 10.1016/j.heliyon.2023.e23401] [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: 02/07/2023] [Revised: 08/07/2023] [Accepted: 12/03/2023] [Indexed: 01/09/2024] Open
Abstract
Aim This study is aimed at evaluating the anticancer effect of the aqueous extract of Caesalpinia pulcherrima (L.) Sw in 7,12-Dimethlbenz[a]anthracene (DMBA) - induced mammary cancer. Methods Tumors were induced via a single intraperitoneal injection of DMBA (dissolved in olive oil) at a dose of 80 mg/kg body weight to the test rats and allowed to develop for about four months. They were treated with cyclophosphamide and an aqueous extract of Caesalpinia pulcherrima at doses of 10 and 250 mg/kg body weight, respectively, for 28 days. Serum levels of cancer antigen 125 (CA125), carcinoembryonic antigen (CEA) activity, cyclooxygenase-2 (COX-2), and cytochrome p450 oxidase (cytp450) activity, as well as other diagnostic enzymes, were estimated. Results The result revealed that DMBA is associated with a significant (p < 0.05) increase in the serum levels of CA125, CEA, COX-2, cytp450, lactate dehydrogenase (LDH), alkaline phosphatase (ALP), aspartate aminotransferase (AST), and alanine aminotransferase (ALT) of the rats, thus suggesting tumor-promoting and hepatotoxic effects of DMBA. There was also a significant (p < 0.05) reduction of serum levels of these cancer and liver biomarker enzymes in the groups treated with cyclophosphamide and Caesalpinia pulcherrima compared to the untreated group, thus suggesting anticancer activity of Caesalpinia pulcherrima. The anticancer effect of Caesalpinia pulcherrima was further confirmed by the disappearance of infiltrative fibrous cells and the absence of inflammatory cells from the photomicrographs of the rats treated with Caesalpinia pulcherrima. Conclusion Our findings show that Caesalpinia pulcherrima possesses anticancer activity, and could protect against mammary cancer.
Collapse
Affiliation(s)
- Abraham Ehinomhen Ubhenin
- Department of Biochemistry, Faculty of Medical Sciences, Federal University Lafia, Nasarawa State, Nigeria
| | - Adedotun A. Adefolalu
- Department of Biochemistry, Faculty of Medical Sciences, Federal University Lafia, Nasarawa State, Nigeria
| | - Kelly Oriakhi
- Department of Medical Biochemistry, Faculty of Medical Sciences, University of Benin, Benin City, Nigeria
| | - Fatima Adis Adamude
- Department of Biochemistry, Faculty of Medical Sciences, Federal University Lafia, Nasarawa State, Nigeria
| | - Emeka John Dingwoke
- Department of Tropical Diseases, UNESCO-International Center for Biotechnology, University of Nigeria, Nsukka, Enugu State, Nigeria
| | - Joshua Onyeka Ikebuiro
- Department of Biochemistry, Faculty of Medical Sciences, Federal University Lafia, Nasarawa State, Nigeria
| | - Benjamin Comfort Chiwendu
- Department of Biochemistry, Faculty of Medical Sciences, Federal University Lafia, Nasarawa State, Nigeria
| | - Mabruqah Liman Muhammad
- Department of Biochemistry, Faculty of Medical Sciences, Federal University Lafia, Nasarawa State, Nigeria
| | - Kingsley Omage
- Division of Nephrology and Hypertension, Department of Medicine, Oregon Health and Science University, Portland, United States
| |
Collapse
|
12
|
Sharma P, Gupta K, Khandai SK, Malik S, Thareja S. Phytometabolites as modulators of breast cancer: a comprehensive review of mechanistic insights. Med Oncol 2024; 41:45. [PMID: 38172452 DOI: 10.1007/s12032-023-02269-2] [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: 10/12/2023] [Accepted: 11/22/2023] [Indexed: 01/05/2024]
Abstract
Breast cancer (BC) is a highly debilitating malignancy affecting females globally and imposing a substantial burden on healthcare systems in both developed and developing nations. Despite the application of conventional therapeutic modalities such as chemotherapy, radiation therapy, and hormonal intervention, BC frequently exhibits resistance, necessitating the urgent development of novel, cost-effective, and accessible treatment strategies. In this context, there is a growing scientific interest in exploring the pharmacological potential of chemical compounds derived from botanical sources, which often exhibit notable biological activity. Extensive in vitro and in vivo investigations have revealed the capacity of these compounds, referred to as phytochemicals, to attenuate the metastatic cascade and reduce the risk of cancer dissemination. These phytochemicals exert their effects through modulation of key molecular and metabolic processes, including regulation of the cell cycle, induction of apoptotic cell death, inhibition of angiogenesis, and suppression of metastatic progression. To shed light on the latest advancements in this field, a comprehensive review of the scientific literature has been conducted, focusing on secondary metabolite agents that have recently been investigated and have demonstrated promising anticancer properties. This review aims to delineate their underlying mechanisms of action and elucidate the associated signaling pathways, thereby contributing to a deeper understanding of their therapeutic potential in the context of BC management.
Collapse
Affiliation(s)
- Priyanka Sharma
- Department of Pharmaceutical Sciences and Natural Products, School of Pharmaceutical Sciences, Central University of Punjab, Bathinda, Punjab, 151401, India
| | - Khushi Gupta
- Department of Pharmaceutical Sciences and Natural Products, School of Pharmaceutical Sciences, Central University of Punjab, Bathinda, Punjab, 151401, India
| | - Sumit Kumar Khandai
- Department of Pharmaceutical Sciences and Natural Products, School of Pharmaceutical Sciences, Central University of Punjab, Bathinda, Punjab, 151401, India
| | - Sonia Malik
- Laboratory of Woody Plants and Crops Biology, University of Orleans, Orleans, France
| | - Suresh Thareja
- Department of Pharmaceutical Sciences and Natural Products, School of Pharmaceutical Sciences, Central University of Punjab, Bathinda, Punjab, 151401, India.
| |
Collapse
|
13
|
García A, Torres-Ruiz S, Vila L, Villarroel-Vicente C, Bernabeu Á, Eroles P, Cabedo N, Cortes D. Synthesis of 2-aminopropyl benzopyran derivatives as potential agents against triple-negative breast cancer. RSC Med Chem 2023; 14:2327-2341. [PMID: 38020071 PMCID: PMC10650959 DOI: 10.1039/d3md00385j] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2023] [Accepted: 09/05/2023] [Indexed: 12/01/2023] Open
Abstract
Synthesis of three series of 2-aminopropyl derivatives containing a benzopyran nucleus was performed to evaluate their performance against triple-negative breast cancer cell lines (MDA-MB-231 and MDA-MB-436) and normal breast epithelial cells (MCF10A). For the three series, the cytotoxic activity was as follows: N-methylated derivatives (tertiary amines) 5b, 6b, and 7b > secondary amine benzopyrans 5, 6, and 7 > quaternary amine salts 5c, 6c, and 7c > free phenolic derivatives 5a, 6a, and 7a. The structure-activity relationship showed the importance of the presence of an amine group and a p-fluorobenzyloxy substituent in the chromanol ring (IC50 values from 1.5 μM to 58.4 μM). In addition, 5a, 5b, 6a, and 7b displayed slight selectivity towards tumor cells. Compounds 5, 5a, 5b, 6, 6a, 6c, 7, and 7b showed apoptotic/necrotic effects due to, at least in part, an increase in reactive oxygen species generation, whereas 5b, 5c, 6b, 7a, and 7c caused cell cycle arrest in the G1 phase. Further cell-based mechanistic studies revealed that 5a, 6a, and 7b, which were the most promising compounds, downregulated the expression of Bcl-2, while 5b downregulated the expression of cyclins CCND1 and CCND2. Therefore, 2-aminopropyl benzopyran derivatives emerge as new hits and potential leads for developing useful agents against breast cancer.
Collapse
Affiliation(s)
- Ainhoa García
- Department of Pharmacology, University of Valencia 46100 Valencia Spain
- Institute of Health Research-INCLIVA, University Clinic Hospital of Valencia 46010 Valencia Spain
| | - Sandra Torres-Ruiz
- Institute of Health Research-INCLIVA, University Clinic Hospital of Valencia 46010 Valencia Spain
| | - Laura Vila
- Institute of Health Research-INCLIVA, University Clinic Hospital of Valencia 46010 Valencia Spain
| | - Carlos Villarroel-Vicente
- Department of Pharmacology, University of Valencia 46100 Valencia Spain
- Institute of Health Research-INCLIVA, University Clinic Hospital of Valencia 46010 Valencia Spain
| | - Álvaro Bernabeu
- Department of Pharmacology, University of Valencia 46100 Valencia Spain
| | - Pilar Eroles
- Institute of Health Research-INCLIVA, University Clinic Hospital of Valencia 46010 Valencia Spain
- Department of Physiology, University of Valencia 46010 Valencia Spain
- Center for Biomedical Network Research on Cancer (CIBERONC) 28019 Madrid Spain
| | - Nuria Cabedo
- Department of Pharmacology, University of Valencia 46100 Valencia Spain
- Institute of Health Research-INCLIVA, University Clinic Hospital of Valencia 46010 Valencia Spain
| | - Diego Cortes
- Department of Pharmacology, University of Valencia 46100 Valencia Spain
| |
Collapse
|
14
|
Alatawi FS, Faridi U. Anticancer and anti-metastasis activity of 1,25 dihydroxycholecalciferols and genistein in MCF-7 and MDA-MB-231 breast cancer cell lines. Heliyon 2023; 9:e21975. [PMID: 38034665 PMCID: PMC10682641 DOI: 10.1016/j.heliyon.2023.e21975] [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: 11/28/2022] [Revised: 11/01/2023] [Accepted: 11/01/2023] [Indexed: 12/02/2023] Open
Abstract
A powerful steroid hormone precursor, 1,25 dihydroxycholecalciferols (1,25(OH)2D3), and dietary phytoestrogen (genistein) are essential compounds that act by binding to nuclear receptors and altering gene expression. They have many biological benefits, some of which have anticancer properties. We studied the impact of 1,25(OH)2D3 and genistein on the proliferation, progression, and metastasis of MCF-7 and MDA-MB-231 cells when they were used alone or in combination and investigated whether there was a synergistic effect between genistein and 1,25(OH)2D3. To achieve these goals, a variety of assays, including flow cytometry, cell invasion assays, cell adhesion assays, Western blotting, and RT‒PCR, were used. Our findings showed that genistein, 1,25(OH)2D3, and the two combined all effectively declined the growth of MCF-7 and MDA-MB-231 cells by arresting the cells in the G0/G1 phase and inducing an apoptotic pathway. Stimulation of apoptosis was achieved by upregulating the expression of BAX and CASP3 genes and downregulating the expression levels of BCL-2 gene. Furthermore, both compounds suppress metastasis by reducing cell adhesion and cell migration/invasion by elevating the expression level of E-cadherin and reducing the expression level of P-cadherin and N-cadherin. Additionally, both genistein and 1,25(OH)2D3 increased the expression level of ERK1 and reduced the expression levels of JNK, p38, Ras, and MEK proteins, which reduced metastasis, enhanced the response to cancer treatment, and improved overall survival. Thus, genistein and 1,25(OH)2D3 can both be considered key candidates in the search for new breast cancer treatments.
Collapse
Affiliation(s)
- Fatema Suliman Alatawi
- Faculty of Sciences, Biochemistry Department, Science College, University of Tabuk, Tabuk Saudi Arabia
| | - Uzma Faridi
- Faculty of Sciences, Biochemistry Department, Science College, University of Tabuk, Tabuk Saudi Arabia
| |
Collapse
|
15
|
Bezerra PHA, Amaral C, Almeida CF, Correia-da-Silva G, Torqueti MR, Teixeira N. In Vitro Effects of Combining Genistein with Aromatase Inhibitors: Concerns Regarding Its Consumption during Breast Cancer Treatment. Molecules 2023; 28:4893. [PMID: 37446555 DOI: 10.3390/molecules28134893] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2023] [Revised: 06/09/2023] [Accepted: 06/15/2023] [Indexed: 07/15/2023] Open
Abstract
INTRODUCTION The third-generation of aromatase inhibitors (AIs)-Exemestane (Exe), Letrozole (Let), and Anastrozole (Ana)-is the main therapeutic approach applied for estrogen receptor-positive (ER+) breast cancer (BC), the most common neoplasm in women worldwide. Despite their success, the development of resistance limits their efficacy. Genistein (G), a phytoestrogen present in soybean, has promising anticancer properties in ER+ BC cells, even when combined with anticancer drugs. Thus, the potential beneficial effects of combining G with AIs were investigated in sensitive (MCF7-aro) and resistant (LTEDaro) BC cells. METHODS The effects on cell proliferation and expression of aromatase, ERα/ERβ, and AR receptors were evaluated. RESULTS Unlike the combination of G with Ana or Let, which negatively affects the Ais' therapeutic efficacy, G enhanced the anticancer properties of the steroidal AI Exe, increasing the antiproliferative effect and apoptosis relative to Exe. The hormone targets studied were not affected by this combination when compared with Exe. CONCLUSIONS This is the first in vitro study that highlights the potential benefit of G as an adjuvant therapy with Exe, emphasizing, however, that soy derivatives widely used in the diet or applied as auxiliary medicines may increase the risk of adverse interactions with nonsteroidal AIs used in therapy.
Collapse
Affiliation(s)
- Patrícia H A Bezerra
- Laboratory of Clinical Cytology, Department of Clinical Analyses, Toxicology and Food Science, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Ribeirão Preto 14040-903, SP, Brazil
| | - Cristina Amaral
- UCIBIO.REQUIMTE, Laboratory of Biochemistry, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, Rua Jorge Viterbo Ferreira, nº 228, 4050-313 Porto, Portugal
- Associate Laboratory i4HB-Institute for Health and Bioeconomy, Faculty of Pharmacy, University of Porto, Rua Jorge Viterbo Ferreira, nº 228, 4050-313 Porto, Portugal
| | - Cristina F Almeida
- UCIBIO.REQUIMTE, Laboratory of Biochemistry, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, Rua Jorge Viterbo Ferreira, nº 228, 4050-313 Porto, Portugal
- Associate Laboratory i4HB-Institute for Health and Bioeconomy, Faculty of Pharmacy, University of Porto, Rua Jorge Viterbo Ferreira, nº 228, 4050-313 Porto, Portugal
| | - Georgina Correia-da-Silva
- UCIBIO.REQUIMTE, Laboratory of Biochemistry, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, Rua Jorge Viterbo Ferreira, nº 228, 4050-313 Porto, Portugal
- Associate Laboratory i4HB-Institute for Health and Bioeconomy, Faculty of Pharmacy, University of Porto, Rua Jorge Viterbo Ferreira, nº 228, 4050-313 Porto, Portugal
| | - Maria Regina Torqueti
- Laboratory of Clinical Cytology, Department of Clinical Analyses, Toxicology and Food Science, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Ribeirão Preto 14040-903, SP, Brazil
| | - Natércia Teixeira
- UCIBIO.REQUIMTE, Laboratory of Biochemistry, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, Rua Jorge Viterbo Ferreira, nº 228, 4050-313 Porto, Portugal
- Associate Laboratory i4HB-Institute for Health and Bioeconomy, Faculty of Pharmacy, University of Porto, Rua Jorge Viterbo Ferreira, nº 228, 4050-313 Porto, Portugal
| |
Collapse
|
16
|
Chen B, Hu H, Chen X. From Basic Science to Clinical Practice: The Role of Cancerous Inhibitor of Protein Phosphatase 2A (CIP2A)/p90 in Cancer. Front Genet 2023; 14:1110656. [PMID: 36911405 PMCID: PMC9998691 DOI: 10.3389/fgene.2023.1110656] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Accepted: 02/03/2023] [Indexed: 03/14/2023] Open
Abstract
Cancerous inhibitor of protein phosphatase 2A (CIP2A), initially reported as a tumor-associated antigen (known as p90), is highly expressed in most solid and hematological tumors. The interaction of CIP2A/p90, protein phosphatase 2A (PP2A), and c-Myc can hinder the function of PP2A toward c-Myc S62 induction, thus stabilizing c-Myc protein, which represents a potential role of CIP2A/p90 in tumorigeneses such as cell proliferation, invasion, and migration, as well as cancer drug resistance. The signaling pathways and regulation networks of CIP2A/p90 are complex and not yet fully understood. Many previous studies have also demonstrated that CIP2A/p90 can be used as a potential therapeutic cancer target. In addition, the autoantibody against CIP2A/p90 in sera may be used as a promising biomarker in the diagnosis of certain types of cancer. In this Review, we focus on recent advances relating to CIP2A/p90 and their implications for future research.
Collapse
Affiliation(s)
- Beibei Chen
- Department of Medical Oncology, Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer Hospital, Zhengzhou, Henan, China.,Zhengzhou Key Laboratory for Precision Therapy of Gastrointestinal Cancer, Zhengzhou, Henan, China
| | - Huihui Hu
- Department of Medical Oncology, Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer Hospital, Zhengzhou, Henan, China.,Zhengzhou Key Laboratory for Precision Therapy of Gastrointestinal Cancer, Zhengzhou, Henan, China
| | - Xiaobing Chen
- Department of Medical Oncology, Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer Hospital, Zhengzhou, Henan, China.,Zhengzhou Key Laboratory for Precision Therapy of Gastrointestinal Cancer, Zhengzhou, Henan, China
| |
Collapse
|
17
|
Effects of phytoestrogens on reproductive organ health. Arch Pharm Res 2022; 45:849-864. [DOI: 10.1007/s12272-022-01417-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2022] [Accepted: 11/14/2022] [Indexed: 11/29/2022]
|
18
|
Targeting Breast Cancer Stem Cells Using Naturally Occurring Phytoestrogens. Int J Mol Sci 2022; 23:ijms23126813. [PMID: 35743256 PMCID: PMC9224163 DOI: 10.3390/ijms23126813] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2022] [Revised: 05/31/2022] [Accepted: 06/09/2022] [Indexed: 12/12/2022] Open
Abstract
Breast cancer therapies have made significant strides in improving survival for patients over the past decades. However, recurrence and drug resistance continue to challenge long-term recurrence-free and overall survival rates. Mounting evidence supports the cancer stem cell model in which the existence of a small population of breast cancer stem cells (BCSCs) within the tumor enables these cells to evade conventional therapies and repopulate the tumor, giving rise to more aggressive, recurrent tumors. Thus, successful breast cancer therapy would need to target these BCSCs, as well the tumor bulk cells. Since the Women’s Health Initiative study reported an increased risk of breast cancer with the use of conventional hormone replacement therapy in postmenopausal women, many have turned their attention to phytoestrogens as a natural alternative. Phytoestrogens are plant compounds that share structural similarities with human estrogens and can bind to the estrogen receptors to alter the endocrine responses. Recent studies have found that phytoestrogens can also target BCSCs and have the potential to complement conventional therapy eradicating BCSCs. This review summarized the latest findings of different phytoestrogens and their effect on BCSCs, along with their mechanisms of action, including selective estrogen receptor binding and inhibition of molecular pathways used by BCSCs. The latest results of phytoestrogens in clinical trials are also discussed to further evaluate the use of phytoestrogen in the treatment and prevention of breast cancer.
Collapse
|
19
|
Rasheed S, Rehman K, Shahid M, Suhail S, Akash MSH. Therapeutic potentials of genistein: New insights and perspectives. J Food Biochem 2022; 46:e14228. [PMID: 35579327 DOI: 10.1111/jfbc.14228] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2022] [Revised: 04/19/2022] [Accepted: 04/21/2022] [Indexed: 12/11/2022]
Abstract
Genistein, a polyphenolic isoflavone compound found abundantly in soy or soy-based products, is widely consumed in the Asian population. Genistein has poor bioavailability, to overcome this problem many advanced nano-drug delivery carrier systems are designed to enhance its water solubility and stability. However, further research is required to develop more efficient bioavailability improvement strategies. Genistein is a phytoestrogen which has been associated with reducing the risk of cancer, cardiovascular disorders, and diabetes mellitus. This plant-based bioactive compound possesses numerous biological activities such as anti-oxidant, anti-inflammatory, anti-obesity, anti-cancer, cardioprotective, and anti-diabetic activities to treat various disease states. Genistein has been used as an active therapeutic agent in many medications. Moreover, several clinical trials are in the ongoing stage to develop more efficient treatment therapies, especially for cancer treatment. This article highlights the protective and therapeutic benefits of genistein in the treatment of different ailments, and more specifically elaborates on the anti-cancer potential of genistein regarding various types of cancers. PRACTICAL APPLICATIONS: Genistein possesses versatile biological activities, including anti-diabetic, anti-inflammatory, anti-oxidant, anti-obesity, and anti-angiogenic. The most studied activity is anti-cancer. Currently, a number of pre-clinical and clinical trials are being carried out on anti-neoplastic and cytotoxic activities of genistein to develop novel therapeutic agents with excellent anti-cancer potential for the treatment of various kinds of cancer. Moreover, many bioavailability enhancement strategies have been developed to improve the bioavailability of genistein. Genistein shows significant hypoglycemic effects alone or in combination with other anti-diabetic agents. Genistein in combination with other chemotherapeutic agents is used for the treatment of prostate, bone, colorectal, glioma, breast, and bladder cancer.
Collapse
Affiliation(s)
- Sumbal Rasheed
- Department of Pharmaceutical Chemistry, Government College University, Faisalabad, Pakistan
| | - Kanwal Rehman
- Department of Pharmacy, The Women University, Multan, Pakistan
| | - Momina Shahid
- Department of Pharmaceutical Chemistry, Government College University, Faisalabad, Pakistan
| | - Shaleem Suhail
- Department of Pharmaceutical Chemistry, Government College University, Faisalabad, Pakistan
| | | |
Collapse
|
20
|
Luque M, Cristóbal I, Sanz-Álvarez M, Santos A, Zazo S, Eroles P, Arpí O, Rovira A, Albanell J, Madoz-Gúrpide J, García-Foncillas J, Rojo F. CIP2A as a Key Regulator for AKT Phosphorylation Has Partial Impact Determining Clinical Outcome in Breast Cancer. J Clin Med 2022; 11:jcm11061610. [PMID: 35329936 PMCID: PMC8955826 DOI: 10.3390/jcm11061610] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2022] [Revised: 03/03/2022] [Accepted: 03/12/2022] [Indexed: 12/07/2022] Open
Abstract
Together with its reported ability to modulate AKT phosphorylation (p-AKT) status in several tumor types, the oncoprotein CIP2A has been described to induce breast cancer progression and drug resistance. However, the clinical and therapeutic relevance of the CIP2A/AKT interplay in breast cancer remains to be fully clarified. Here, we found high p-AKT levels in 80 out of 220 cases (36.4%), which were associated with negative estrogen receptor expression (p = 0.049) and CIP2A overexpression (p < 0.001). Interestingly, p-AKT determined substantially shorter overall (p = 0.002) and progression-free survival (p = 0.003), and multivariate analyses showed its CIP2A-independent prognostic value. Moreover, its clinical relevance was further confirmed in the triple negative and HER2-positive subgroups after stratifying our series by molecular subtype. Functionally, we confirmed in vitro the role of CIP2A as a regulator of p-AKT levels in breast cancer cell lines, and the importance of the CIP2A/AKT axis was also validated in vivo. Finally, p-AKT also showed a higher predictive value of response to doxorubicin than CIP2A in ex vivo analyses. In conclusion, our findings suggest that CIP2A overexpression is a key contributing event to AKT phosphorylation and highlights the CIP2A/AKT axis as a promising therapeutic target in breast cancer. However, our observations highlight the existence of alternative mechanisms that regulate AKT signaling in a subgroup of breast tumors without altered CIP2A expression that determines its independent value as a marker of poor outcome in this disease.
Collapse
Affiliation(s)
- Melani Luque
- Pathology Department, IIS-Fundación Jiménez Diaz-UAM, 28040 Madrid, Spain; (M.L.); (M.S.-Á.); (S.Z.); (J.M.-G.)
| | - Ion Cristóbal
- Cancer Unit for Research on Novel Therapeutic Targets, Oncohealth Institute, ISS-FJD-UAM, 28040 Madrid, Spain;
- Translational Oncology Division, Oncohealth Institute, IIS-Fundación Jiménez Diaz-UAM, 28040 Madrid, Spain;
- Correspondence: (I.C.); (F.R.); Tel.: +34-915-504-800 (I.C. & F.R.)
| | - Marta Sanz-Álvarez
- Pathology Department, IIS-Fundación Jiménez Diaz-UAM, 28040 Madrid, Spain; (M.L.); (M.S.-Á.); (S.Z.); (J.M.-G.)
| | - Andrea Santos
- Cancer Unit for Research on Novel Therapeutic Targets, Oncohealth Institute, ISS-FJD-UAM, 28040 Madrid, Spain;
- Translational Oncology Division, Oncohealth Institute, IIS-Fundación Jiménez Diaz-UAM, 28040 Madrid, Spain;
| | - Sandra Zazo
- Pathology Department, IIS-Fundación Jiménez Diaz-UAM, 28040 Madrid, Spain; (M.L.); (M.S.-Á.); (S.Z.); (J.M.-G.)
| | - Pilar Eroles
- Institute of Health Research INCLIVA, 46010 Valencia, Spain;
| | - Oriol Arpí
- Medical Oncology Department, Hospital del Mar, 08003 Barcelona, Spain; (O.A.); (A.R.); (J.A.)
| | - Ana Rovira
- Medical Oncology Department, Hospital del Mar, 08003 Barcelona, Spain; (O.A.); (A.R.); (J.A.)
| | - Joan Albanell
- Medical Oncology Department, Hospital del Mar, 08003 Barcelona, Spain; (O.A.); (A.R.); (J.A.)
| | - Juan Madoz-Gúrpide
- Pathology Department, IIS-Fundación Jiménez Diaz-UAM, 28040 Madrid, Spain; (M.L.); (M.S.-Á.); (S.Z.); (J.M.-G.)
| | - Jesús García-Foncillas
- Translational Oncology Division, Oncohealth Institute, IIS-Fundación Jiménez Diaz-UAM, 28040 Madrid, Spain;
| | - Federico Rojo
- Pathology Department, IIS-Fundación Jiménez Diaz-UAM, 28040 Madrid, Spain; (M.L.); (M.S.-Á.); (S.Z.); (J.M.-G.)
- Correspondence: (I.C.); (F.R.); Tel.: +34-915-504-800 (I.C. & F.R.)
| |
Collapse
|
21
|
Genistein, a Potential Phytochemical against Breast Cancer Treatment-Insight into the Molecular Mechanisms. Processes (Basel) 2022. [DOI: 10.3390/pr10020415] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
Breast cancer (BC) is one of the most common malignancies in women. Although widespread successful synthetic drugs are available, natural compounds can also be considered as significant anticancer agents for treating BC. Some natural compounds have similar effects as synthetic drugs with fewer side effects on normal cells. Therefore, we aimed to unravel and analyze several molecular mechanisms of genistein (GNT) against BC. GNT is a type of dietary phytoestrogen included in the flavonoid group with a similar structure to estrogen that might provide a strong alternative and complementary medicine to existing chemotherapeutic drugs. Previous research reported that GNT could target the estrogen receptor (ER) human epidermal growth factor receptor-2 (HER2) and several signaling molecules against multiple BC cell lines and sensitize cancer cell lines to this compound when used at an optimal inhibitory concentration. More specifically, GNT mediates the anticancer mechanism through apoptosis induction, arresting the cell cycle, inhibiting angiogenesis and metastasis, mammosphere formation, and targeting and suppressing tumor growth factors. Furthermore, it acts via upregulating tumor suppressor genes and downregulating oncogenes in vitro and animal model studies. In addition, this phytochemical synergistically reverses the resistance mechanism of standard chemotherapeutic drugs, increasing their efficacy against BC. Overall, in this review, we discuss several molecular interactions of GNT with numerous cellular targets in the BC model and show its anticancer activities alone and synergistically. We conclude that GNT can have favorable therapeutic advantages when standard drugs are not available in the pharma markets.
Collapse
|
22
|
Ma X, Yu X, Min J, Chen X, Liu R, Cui X, Cheng J, Xie M, Diel P, Hu X. Genistein interferes with antitumor effects of cisplatin in an ovariectomized breast cancer xenograft tumor model. Toxicol Lett 2022; 355:106-115. [PMID: 34838996 DOI: 10.1016/j.toxlet.2021.11.013] [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: 03/10/2021] [Revised: 11/01/2021] [Accepted: 11/24/2021] [Indexed: 01/18/2023]
Abstract
Genistein (GEN) has been demonstrated to interfere with antitumor effects of cisplatin (CIS) in vitro. To analyze whether these findings are also relevant in vivo, we examined the effects of combined GEN and CIS treatment in an ovariectomized nude mouse breast cancer xenograft model. Tumor growth and markers for antitumor activity were determined after three weeks of treatment. Furthermore, the concentrations of GEN metabolites were measured in serum, liver, and xenograft tumor tissues using liquid chromatography-tandem mass spectrometry (LC-MS/MS). Three weeks' oral exposure to GEN at a dose of 5 mg kg-1·d-1 resulted in an average concentration of total GEN metabolite equivalent as high as 0.2729 nmol g-1 wet weight in xenograft tumor tissues. At this dosage, GEN significantly antagonized the antitumor effects of CIS. Mechanistically, GEN blocked both the inhibition of cell proliferation and induction of apoptosis triggered by CIS. Moreover, GEN concentrations in xenograft tumor tissues were found to be significantly higher than in serum and liver. In conclusion, our findings suggested that oral GEN exposure at a level comparable to dietary exposure in humans could interfere with CIS chemotherapy.
Collapse
Affiliation(s)
- Xing Ma
- Department of Biochemistry and Molecular Biology, School of Basic Medical Science, Nanchang University, Nanchang, 330006, China
| | - Xiaowei Yu
- Department of Biochemistry and Molecular Biology, School of Basic Medical Science, Nanchang University, Nanchang, 330006, China; State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, 510060, China
| | - Jialing Min
- State Key Laboratory of Food Science and Technology, Nanchang University, Nanchang, 330047, China; Jiangxi Biotech Vocational College, Nanchang, 330200, China
| | - Xin Chen
- Department of Biochemistry and Molecular Biology, School of Basic Medical Science, Nanchang University, Nanchang, 330006, China; State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, 510060, China
| | - Ren Liu
- Department of Biochemistry and Molecular Biology, School of Basic Medical Science, Nanchang University, Nanchang, 330006, China; State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, 510060, China
| | - Xueqing Cui
- Department of Biochemistry and Molecular Biology, School of Basic Medical Science, Nanchang University, Nanchang, 330006, China
| | - Jing Cheng
- Department of Biochemistry and Molecular Biology, School of Basic Medical Science, Nanchang University, Nanchang, 330006, China
| | - Mingyong Xie
- State Key Laboratory of Food Science and Technology, Nanchang University, Nanchang, 330047, China
| | - Patrick Diel
- Department of Molecular and Cellular Sports Medicine, German Sport University Cologne, Cologne, 50933, Germany
| | - Xiaojuan Hu
- Department of Biochemistry and Molecular Biology, School of Basic Medical Science, Nanchang University, Nanchang, 330006, China; Jiangxi Province Key Laboratory of Laboratory Animal, Nanchang, 330006, China.
| |
Collapse
|
23
|
Bhat SS, Prasad SK, Shivamallu C, Prasad KS, Syed A, Reddy P, Cull CA, Amachawadi RG. Genistein: A Potent Anti-Breast Cancer Agent. Curr Issues Mol Biol 2021; 43:1502-1517. [PMID: 34698063 PMCID: PMC8929066 DOI: 10.3390/cimb43030106] [Citation(s) in RCA: 63] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2021] [Revised: 09/28/2021] [Accepted: 10/01/2021] [Indexed: 12/15/2022] Open
Abstract
Genistein is an isoflavonoid present in high quantities in soybeans. Possessing a wide range of bioactives, it is being studied extensively for its tumoricidal effects. Investigations into mechanisms of the anti-cancer activity have revealed many pathways including induction of cell proliferation, suppression of tyrosine kinases, regulation of Hedgehog-Gli1 signaling, modulation of epigenetic activities, seizing of cell cycle and Akt and MEK signaling pathways, among others via which the cancer cell proliferation can be controlled. Notwithstanding, the observed activities have been time- and dose-dependent. In addition, genistein has also shown varying results in women depending on the physiological parameters, such as the early or post-menopausal states.
Collapse
Affiliation(s)
- Smitha S. Bhat
- Department of Biotechnology and Bioinformatics, Faculty of Life Sciences, JSS Academy of Higher Education and Research, Mysuru 570015, Karnataka, India; (S.S.B.); (S.K.P.); (C.S.)
| | - Shashanka K. Prasad
- Department of Biotechnology and Bioinformatics, Faculty of Life Sciences, JSS Academy of Higher Education and Research, Mysuru 570015, Karnataka, India; (S.S.B.); (S.K.P.); (C.S.)
| | - Chandan Shivamallu
- Department of Biotechnology and Bioinformatics, Faculty of Life Sciences, JSS Academy of Higher Education and Research, Mysuru 570015, Karnataka, India; (S.S.B.); (S.K.P.); (C.S.)
| | - Kollur Shiva Prasad
- Department of Sciences, Amrita School of Arts and Sciences, Amrita Vishwa Vidyapeetham, Mysuru Campus, Mysuru 570026, Karnataka, India;
| | - Asad Syed
- Department of Botany and Microbiology, College of Science, King Saud University, P.O. Box 2455, Riyadh 11451, Saudi Arabia;
| | - Pruthvish Reddy
- Department of Biotechnology, Acharya Institute of Technology, Bengaluru 560107, Karnataka, India;
| | | | - Raghavendra G. Amachawadi
- Department of Clinical Sciences, College of Veterinary Medicine, Kansas State University, Manhattan, KS 66506, USA
| |
Collapse
|
24
|
Sawanny R, Pramanik S, Agarwal U. Role of Phytochemicals in the Treatment of Breast Cancer: Natural Swords Battling Cancer Cells. CURRENT CANCER THERAPY REVIEWS 2021. [DOI: 10.2174/1573394716666210106123255] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Breast cancer is the most common type of malignancy among ladies (around 30% of
newly diagnosed patients every year). To date, various modern treatment modalities for breast cancer,
such as radiotherapy, surgical method, hormonal therapy, and chemotherapeutic drug utilisation,
are available. However, adverse drug reactions, therapeutic resistance, metastasis, or cancer reoccurrence
chances remain the primary causes of mortality for breast cancer patients. To overcome
all the potential drawbacks, we need to investigate novel techniques and strategies that are not considered
previously to treat breast cancer effectively with safety and efficacy. For centuries, we
utilise phytochemicals to treat various diseases because of their safety, low-cost, and least or no
side effects. Recently, naturally produced phytochemicals gain immense attention as potential
breast cancer therapeutics because of their ideal characteristics; for instance, they operate via modulating
molecular pathways associated with cancer growth and progression. The primary mechanism
involves inhibition of cell proliferation, angiogenesis, migration, invasion, increasing anti-oxidant
status, initiation of the arrest of the cell cycle, and apoptosis. Remedial viability gets effectively enhanced
when phytochemicals work as adjuvants with chemotherapeutic drugs. This comprehensive
review revolves around the latest chemopreventive, chemotherapeutic, and chemoprotective treatments
with their molecular mechanisms to treat breast cancer by utilising phytochemicals such as
vinca alkaloids, resveratrol, curcumin, paclitaxel, silibinin, quercetin, genistein, and epigallocatechin
gallate. The authors wish to extend the field of phytochemical study for its scientific validity
and its druggability.
Collapse
Affiliation(s)
- Rajni Sawanny
- Noida Institute of Engineering and Technology (Pharmacy Institute), Knowledge Park-II, Institutional Area, Greater Noida, Uttar Pradesh-201306, India
| | - Sheersha Pramanik
- Department of Biotechnology, Bhupat and Jyoti Mehta School of Biosciences, Indian Institute of Technology Madras, Chennai, Tamil Nadu-600036, India
| | - Unnati Agarwal
- School of Bioengineering and Biosciences, Lovely Professional University, Jalandhar, Delhi, Grand Trunk Road, Phagwara, Punjab-144001, India
| |
Collapse
|
25
|
Sharifi-Rad J, Quispe C, Imran M, Rauf A, Nadeem M, Gondal TA, Ahmad B, Atif M, Mubarak MS, Sytar O, Zhilina OM, Garsiya ER, Smeriglio A, Trombetta D, Pons DG, Martorell M, Cardoso SM, Razis AFA, Sunusi U, Kamal RM, Rotariu LS, Butnariu M, Docea AO, Calina D. Genistein: An Integrative Overview of Its Mode of Action, Pharmacological Properties, and Health Benefits. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2021; 2021:3268136. [PMID: 34336089 PMCID: PMC8315847 DOI: 10.1155/2021/3268136] [Citation(s) in RCA: 147] [Impact Index Per Article: 36.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/05/2021] [Revised: 06/11/2021] [Accepted: 06/28/2021] [Indexed: 12/15/2022]
Abstract
Genistein is an isoflavone first isolated from the brooming plant Dyer's Genista tinctoria L. and is widely distributed in the Fabaceae family. As an isoflavone, mammalian genistein exerts estrogen-like functions. Several biological effects of genistein have been reported in preclinical studies, such as the antioxidant, anti-inflammatory, antibacterial, and antiviral activities, the effects of angiogenesis and estrogen, and the pharmacological activities on diabetes and lipid metabolism. The purpose of this review is to provide up-to-date evidence of preclinical pharmacological activities with mechanisms of action, bioavailability, and clinical evidence of genistein. The literature was researched using the most important keyword "genistein" from the PubMed, Science, and Google Scholar databases, and the taxonomy was validated using The Plant List. Data were also collected from specialized books and other online resources. The main positive effects of genistein refer to the protection against cardiovascular diseases and to the decrease of the incidence of some types of cancer, especially breast cancer. Although the mechanism of protection against cancer involves several aspects of genistein metabolism, the researchers attribute this effect to the similarity between the structure of soy genistein and that of estrogen. This structural similarity allows genistein to displace estrogen from cellular receptors, thus blocking their hormonal activity. The pharmacological activities resulting from the experimental studies of this review support the traditional uses of genistein, but in the future, further investigations are needed on the efficacy, safety, and use of nanotechnologies to increase bioavailability and therapeutic efficacy.
Collapse
Affiliation(s)
- Javad Sharifi-Rad
- Phytochemistry Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Cristina Quispe
- Facultad de Ciencias de la Salud, Universidad Arturo Prat, Avda. Arturo Prat 2120, Iquique 1110939, Chile
| | - Muhammad Imran
- University Institute of Diet and Nutritional Sciences, Faculty of Allied Health Sciences, The University of Lahore, Lahore, Pakistan
| | - Abdur Rauf
- Department of Chemistry, University of Swabi, Anbar-, 23561 Khyber Pakhtunkhwa, Pakistan
| | - Muhammad Nadeem
- Department of Environmental Sciences, COMSATS Institute of Information Technology, Vehari-, Pakistan
| | | | - Bashir Ahmad
- Center of Biotechnology and Microbiology, University of Peshawar, Peshawar-, 25120 KPK, Pakistan
| | - Muhammad Atif
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Jouf University, Sakaka 72341, Saudi Arabia
| | | | - Oksana Sytar
- Department of Plant Biology Department, Institute of Biology, Taras Shevchenko National University of Kyiv, Volodymyrska Str., 64, Kyiv 01033, Ukraine
- Department of Plant Physiology, Slovak University of Agriculture, A. Hlinku 2, 94976 Nitra, Slovakia
| | - Oxana Mihailovna Zhilina
- Department of Organic Chemistry, Pyatigorsk Medical-Pharmaceutical Institute (PMPI), Branch of Volgograd State Medical University, Ministry of Health of Russia, Pyatigorsk 357532, Russia
| | - Ekaterina Robertovna Garsiya
- Department of Pharmacognosy, Botany and Technology of Phytopreparations, Pyatigorsk Medical-Pharmaceutical Institute (PMPI), Branch of Volgograd State Medical University, Ministry of Health of Russia, Pyatigorsk 357532, Russia
| | - Antonella Smeriglio
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Italy
| | - Domenico Trombetta
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Italy
| | - Daniel Gabriel Pons
- Grupo Multidisciplinar de Oncología Traslacional (GMOT), Institut Universitari d'Investigació en Ciències de la Salut (IUNICS), Universitat de les Illes Balears (UIB), Instituto de Investigación Sanitaria Illes Balears (IdISBa), Palma 07122, Spain
| | - Miquel Martorell
- Department of Nutrition and Dietetics, Faculty of Pharmacy, University of Concepción, Concepción 4070386, Chile
- Unidad de Desarrollo Tecnológico, Universidad de Concepción UDT, Concepción 4070386, Chile
| | - Susana M. Cardoso
- LAQV-REQUIMTE, Department of Chemistry, University of Aveiro, 3810-193 Aveiro, Portugal
| | - Ahmad Faizal Abdull Razis
- Department of Food Science, Faculty of Food Science and Technology, Universiti Putra Malaysia, 43400, UPM Serdang, Selangor, Malaysia
- Natural Medicines and Products Research Laboratory, Institute of Bioscience, Universiti Putra Malaysia, 43400 UPM Serdang, Selangor, Malaysia
| | - Usman Sunusi
- Natural Medicines and Products Research Laboratory, Institute of Bioscience, Universiti Putra Malaysia, 43400 UPM Serdang, Selangor, Malaysia
- Department of Biochemistry, Bayero University Kano, PMB 3011 Kano, Nigeria
| | - Ramla Muhammad Kamal
- Natural Medicines and Products Research Laboratory, Institute of Bioscience, Universiti Putra Malaysia, 43400 UPM Serdang, Selangor, Malaysia
- Department of Pharmacology, Federal University Dutse, PMB 7156 Dutse Jigawa State, Nigeria
| | - Lia Sanda Rotariu
- Banat's University of Agricultural Sciences and Veterinary Medicine “King Michael I of Romania” from Timisoara, Romania
| | - Monica Butnariu
- Banat's University of Agricultural Sciences and Veterinary Medicine “King Michael I of Romania” from Timisoara, Romania
| | - Anca Oana Docea
- Department of Toxicology, University of Medicine and Pharmacy of Craiova, 200349 Craiova, Romania
| | - Daniela Calina
- Department of Clinical Pharmacy, University of Medicine and Pharmacy of Craiova, 200349 Craiova, Romania
| |
Collapse
|
26
|
Saafan H, Alahdab A, Michelet R, Gohlke L, Ziemann J, Holdenrieder S, McLaughlin KM, Wass MN, Cinatl J, Michaelis M, Kloft C, Ritter CA. Constitutive Cell Proliferation Regulating Inhibitor of Protein Phosphatase 2A (CIP2A) Mediates Drug Resistance to Erlotinib in an EGFR Activating Mutated NSCLC Cell Line. Cells 2021; 10:716. [PMID: 33804833 PMCID: PMC8103245 DOI: 10.3390/cells10040716] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2020] [Revised: 03/15/2021] [Accepted: 03/17/2021] [Indexed: 11/21/2022] Open
Abstract
Exploring mechanisms of drug resistance to targeted small molecule drugs is critical for an extended clinical benefit in the treatment of non-small cell lung cancer (NSCLC) patients carrying activating epidermal growth factor receptor (EGFR) mutations. Here, we identified constitutive cell proliferation regulating inhibitor of protein phosphatase 2A (CIP2A) in the HCC4006rErlo0.5 NSCLC cell line adapted to erlotinib as a model of acquired drug resistance. Constitutive CIP2A resulted in a constitutive activation of Akt signaling. The proteasome inhibitor bortezomib was able to reduce CIP2A levels, which resulted in an activation of protein phosphatase 2A and deactivation of Akt. Combination experiments with erlotinib and bortezomib revealed a lack of interaction between the two drugs. However, the effect size of bortezomib was higher in HCC4006rErlo0.5, compared to the erlotinib-sensitive HCC4006 cells, as indicated by an increase in Emax (0.911 (95%CI 0.867-0.954) vs. 0.585 (95%CI 0.568-0.622), respectively) and decrease in EC50 (52.4 µM (95%CI 46.1-58.8 µM) vs. 73.0 µM (95%CI 60.4-111 µM), respectively) in the concentration-effect model, an earlier onset of cell death induction, and a reduced colony surviving fraction (0.38 ± 0.18 vs. 0.95 ± 0.25, respectively, n = 3, p < 0.05). Therefore, modulation of CIP2A with bortezomib could be an interesting approach to overcome drug resistance to erlotinib treatment in NSCLC.
Collapse
Affiliation(s)
- Hisham Saafan
- Institute of Pharmacy, Clinical Pharmacy, University of Greifswald, Friedrich-Ludwig-Jahn-Str. 17, 17489 Greifswald, Germany; (H.S.); (A.A.); (L.G.)
| | - Ahmad Alahdab
- Institute of Pharmacy, Clinical Pharmacy, University of Greifswald, Friedrich-Ludwig-Jahn-Str. 17, 17489 Greifswald, Germany; (H.S.); (A.A.); (L.G.)
| | - Robin Michelet
- Department of Clinical Pharmacy and Biochemistry, Institute of Pharmacy, Freie Universitaet Berlin, 14195 Berlin, Germany; (R.M.); (C.K.)
| | - Linus Gohlke
- Institute of Pharmacy, Clinical Pharmacy, University of Greifswald, Friedrich-Ludwig-Jahn-Str. 17, 17489 Greifswald, Germany; (H.S.); (A.A.); (L.G.)
| | - Janine Ziemann
- Central Unit for Infection Prevention and Control, University Medicine Greifswald, 17475 Greifswald, Germany;
| | - Stefan Holdenrieder
- Institute of Laboratory Medicine, German Heart Center, Munich Technical University, 80636 Munich, Germany;
| | - Katie-May McLaughlin
- Industrial Biotechnology Centre, School of Biosciences, University of Kent, Canterbury CT2 7NJ, UK; (K.-M.M.); (M.N.W.); (M.M.)
| | - Mark N. Wass
- Industrial Biotechnology Centre, School of Biosciences, University of Kent, Canterbury CT2 7NJ, UK; (K.-M.M.); (M.N.W.); (M.M.)
| | - Jindrich Cinatl
- Institute of Medical Virology, Goethe-University, 60596 Frankfurt am Main, Germany;
| | - Martin Michaelis
- Industrial Biotechnology Centre, School of Biosciences, University of Kent, Canterbury CT2 7NJ, UK; (K.-M.M.); (M.N.W.); (M.M.)
| | - Charlotte Kloft
- Department of Clinical Pharmacy and Biochemistry, Institute of Pharmacy, Freie Universitaet Berlin, 14195 Berlin, Germany; (R.M.); (C.K.)
| | - Christoph A Ritter
- Institute of Pharmacy, Clinical Pharmacy, University of Greifswald, Friedrich-Ludwig-Jahn-Str. 17, 17489 Greifswald, Germany; (H.S.); (A.A.); (L.G.)
| |
Collapse
|
27
|
Kumai A, Tsugami Y, Wakasa H, Suzuki N, Suzuki T, Nishimura T, Kobayashi K. Adverse Effects of Coumestrol and Genistein on Mammary Morphogenesis and Future Milk Production Ability of Mammary Epithelial Cells. ACTA ACUST UNITED AC 2020; 4:e1900187. [PMID: 32293160 DOI: 10.1002/adbi.201900187] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2019] [Revised: 01/23/2020] [Indexed: 12/15/2022]
Abstract
Isoflavones are a class of flavonoids present in legumes and are called phytoestrogens because of their estrogen-like activity. Endogenous estrogen is well known to regulate mammary gland morphogenesis during pregnancy. Each isoflavone also has different physiological activities. However, it is difficult to investigate the direct effect of each isoflavone in mammary morphogenesis in vivo because isoflavones are metabolized into different isoflavones by enteric bacteria. In this study, investigated are the direct influences of coumestrol, daidzein, and genistein on mammary structure development and future milk production ability of mammary epithelial cells (MECs) using in vitro culture models. Mouse MECs are cultured in Matrigel with basic fibroblast growth factor and epidermal growth factor to induce ductal branching and alveolar formation, respectively. Coumestrol and genistein inhibit ductal branching and alveolar formation by affecting the proliferation and migration of MECs with the induction of apoptosis. Daidzein hardly influences mammary structure development. Furthermore, pretreatment with coumestrol adversely affects the induction of milk production ability of MECs. These results suggest that each isoflavone differentially influences mammary morphogenesis and future milk production by affecting MEC behaviors. These results also suggest that the culture models are effective to study mammary epithelial morphogenesis in vitro.
Collapse
Affiliation(s)
- Aogu Kumai
- Laboratory of Cell and Tissue Biology, Research Faculty of Agriculture, Hokkaido University, North 9, West 9, 060-8589, Sapporo, Japan
| | - Yusaku Tsugami
- Laboratory of Cell and Tissue Biology, Research Faculty of Agriculture, Hokkaido University, North 9, West 9, 060-8589, Sapporo, Japan
| | - Haruka Wakasa
- Laboratory of Cell and Tissue Biology, Research Faculty of Agriculture, Hokkaido University, North 9, West 9, 060-8589, Sapporo, Japan
| | - Norihiro Suzuki
- Laboratory of Cell and Tissue Biology, Research Faculty of Agriculture, Hokkaido University, North 9, West 9, 060-8589, Sapporo, Japan
| | - Takahiro Suzuki
- Laboratory of Cell and Tissue Biology, Research Faculty of Agriculture, Hokkaido University, North 9, West 9, 060-8589, Sapporo, Japan
| | - Takanori Nishimura
- Laboratory of Cell and Tissue Biology, Research Faculty of Agriculture, Hokkaido University, North 9, West 9, 060-8589, Sapporo, Japan
| | - Ken Kobayashi
- Laboratory of Cell and Tissue Biology, Research Faculty of Agriculture, Hokkaido University, North 9, West 9, 060-8589, Sapporo, Japan
| |
Collapse
|
28
|
Tuli HS, Tuorkey MJ, Thakral F, Sak K, Kumar M, Sharma AK, Sharma U, Jain A, Aggarwal V, Bishayee A. Molecular Mechanisms of Action of Genistein in Cancer: Recent Advances. Front Pharmacol 2019; 10:1336. [PMID: 31866857 PMCID: PMC6910185 DOI: 10.3389/fphar.2019.01336] [Citation(s) in RCA: 214] [Impact Index Per Article: 35.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2019] [Accepted: 10/18/2019] [Indexed: 01/13/2023] Open
Abstract
Background: Genistein is one among the several other known isoflavones that is found in different soybeans and soy products. The chemical name of genistein is 4',5,7-trihydroxyisoflavone. Genistein has drawn attention of scientific community because of its potential beneficial effects on human grave diseases, such as cancer. Mechanistic insight of genistein reveals its potential for apoptotic induction, cell cycle arrest, as well as antiangiogenic, antimetastatic, and anti-inflammatory effects. Objective: The purpose of this review is to unravel and analyze various molecular mechanisms of genistein in diverse cancer models. Data sources: English language literature was searched using various databases, such as PubMed, ScienceDirect, EBOSCOhost, Scopus, Web of Science, and Cochrane Library. Key words used in various combinations included genistein, cancer, anticancer, molecular mechanisms prevention, treatment, in vivo, in vitro, and clinical studies. Study selection: Study selection was carried out strictly in accordance with the statement of Preferred Reporting Items for Systematic Reviews and Meta-analyses. Data extraction: Four authors independently carried out the extraction of articles. Data synthesis: One hundred one papers were found suitable for use in this review. Conclusion: This review covers various molecular interactions of genistein with various cellular targets in cancer models. It will help the scientific community understand genistein and cancer biology and will provoke them to design novel therapeutic strategies.
Collapse
Affiliation(s)
- Hardeep Singh Tuli
- Department of Biotechnology, Maharishi Markandeshwar (Deemed to be University), Mullana-Ambala, India
| | - Muobarak Jaber Tuorkey
- Division of Physiology, Zoology Department, Faculty of Science, Damanhour University, Damanhour, Egypt
| | - Falak Thakral
- Department of Biotechnology, Maharishi Markandeshwar (Deemed to be University), Mullana-Ambala, India
| | | | - Manoj Kumar
- Department of Chemistry, Maharishi Markandeshwar University, Sadopur, India
| | - Anil Kumar Sharma
- Department of Biotechnology, Maharishi Markandeshwar (Deemed to be University), Mullana-Ambala, India
| | - Uttam Sharma
- Department of Animal Sciences, Central University of Punjab, Bathinda, India
| | - Aklank Jain
- Department of Animal Sciences, Central University of Punjab, Bathinda, India
| | - Vaishali Aggarwal
- Department of Histopathology, Post Graduate Institute of Medical Education and Research, Chandigarh, India
| | - Anupam Bishayee
- Lake Erie College of Osteopathic Medicine, Bradenton, FL, United States
| |
Collapse
|
29
|
Shentu YP, Hu WT, Liang JW, Liuyang ZY, Wei H, Qun W, Wang XC, Wang JZ, Westermarck J, Liu R. Genistein Decreases APP/tau Phosphorylation and Ameliorates Aβ Overproduction Through Inhibiting CIP2A. Curr Alzheimer Res 2019; 16:732-740. [DOI: 10.2174/1567205016666190830113420] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2018] [Revised: 06/12/2019] [Accepted: 08/15/2019] [Indexed: 12/17/2022]
Abstract
Background:
Upregulation of Cancerous Inhibitor of PP2A (CIP2A) plays an important role
in disease-related phosphorylation of tau/APP and tau pathology/Aβ overproduction through inhibiting
PP2A in AD brain. Genistein has been shown to potently reduce CIP2A in experimental cancer treatment
research. Whether Genistein can ameliorate AD pathology through targeting CIP2A needs further investigation.
Methods:
The inhibitory effects of Genistein on tau/APP phosphorylation and Aβ overproduction in AD
cell models have been explored. HEK293-T cells were co-transfected with CIP2A and APP plasmids, or
CIP2A and tau plasmids, with Genistein incubation at 0, 30, 60 or 120 µM for 48 h, cell viability and
PP2A activities were measured. HEK293-T cells with CIP2A/APP overexpression treated with Genistein
at 30 µM for 48 h were collected and lyzed for Western blotting detection of CIP2A, PP2Ac, APP-T668,
total APP, PS1, BACE1, sAPPα and sAPPβ. Aβ40 and Aβ42 levels in cell supernatant, soluble fraction
(RIPA) and insoluble fraction (formic acid soluble) of cell lysates were measured by ELISA. HEK293-T
cells with CIP2A/tau overexpression treated with Genistein at 30 µM for 48 h were collected for Western
blotting detection of CIP2A, PP2Ac, tau-S396, tau-S404 and total tau.
Conclusion:
CIP2A is a target of Genistein in AD therapy. Genistein reduces APP/tau hyperphosphorylation
and Aβ production through inhibiting the effect of CIP2A on PP2A.
Results:
Genistein effectively reduced CIP2A expression, and restored PP2A activities both in
CIP2A/APP, CIP2A/tau co-expressed cells. Genistein reduced APP phosphorylation at T668 site and
inhibited Aβ production. Meantime, Genistein ameliorated tau hyperphosphorylation through repressing
the inhibitory effect of CIP2A on PP2A.
Collapse
Affiliation(s)
- Yang-Ping Shentu
- Department of Pathophysiology, Key Laboratory of Ministry of Education for Neurological Disorders, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Wen-Ting Hu
- Department of Pathophysiology, Key Laboratory of Ministry of Education for Neurological Disorders, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jia-Wei Liang
- Department of Pathophysiology, Key Laboratory of Ministry of Education for Neurological Disorders, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zhen-Yu Liuyang
- Department of Pathophysiology, Key Laboratory of Ministry of Education for Neurological Disorders, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Hui Wei
- Department of Pathophysiology, Key Laboratory of Ministry of Education for Neurological Disorders, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Wang Qun
- Department of Pathophysiology, Key Laboratory of Ministry of Education for Neurological Disorders, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiao-Chuan Wang
- Department of Pathophysiology, Key Laboratory of Ministry of Education for Neurological Disorders, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jian-Zhi Wang
- Department of Pathophysiology, Key Laboratory of Ministry of Education for Neurological Disorders, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jukka Westermarck
- Turku Centre for Biotechnology, University of Turku and Åbo Akademi University, Turku, Finland
| | - Rong Liu
- Department of Pathophysiology, Key Laboratory of Ministry of Education for Neurological Disorders, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
30
|
Kim SH, Lee WH, Seong D, An JH, Je HU, Nam HY, Kim SY, Kim SW, Han MW. The role of CIP2A as a therapeutic target of rapamycin in radioresistant head and neck cancer with TP53 mutation. Head Neck 2019; 41:3362-3371. [DOI: 10.1002/hed.25868] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2019] [Revised: 06/13/2019] [Accepted: 06/18/2019] [Indexed: 02/06/2023] Open
Affiliation(s)
- Song Hee Kim
- Department of OtolaryngologyUlsan University Hospital, University of Ulsan College of Medicine Ulsan Republic of Korea
| | - Won Hyeok Lee
- Department of OtolaryngologyUlsan University Hospital, University of Ulsan College of Medicine Ulsan Republic of Korea
| | - Daseul Seong
- Department of OtolaryngologyUlsan University Hospital, University of Ulsan College of Medicine Ulsan Republic of Korea
| | - Jae Hee An
- Department of OtolaryngologyUlsan University Hospital, University of Ulsan College of Medicine Ulsan Republic of Korea
| | - Hyoung Uk Je
- Department of Radiation OncologyUlsan University Hospital, University of Ulsan College of Medicine Ulsan Republic of Korea
| | - Hae Yun Nam
- Department of Biochemistry and Molecular BiologyAsan Medical Center, University of Ulsan College of Medicine Seoul Republic of Korea
| | - Sang Yoon Kim
- Department of OtolaryngologyAsan Medical Center, University of Ulsan College of Medicine Seoul Republic of Korea
| | - Seong Who Kim
- Department of Biochemistry and Molecular BiologyAsan Medical Center, University of Ulsan College of Medicine Seoul Republic of Korea
| | - Myung Woul Han
- Department of OtolaryngologyUlsan University Hospital, University of Ulsan College of Medicine Ulsan Republic of Korea
- Department of Otolaryngology‐Head and Neck SurgeryLondon Health Sciences Center, Schulich School of Medicine & Dentistry, Western University London Ontario Canada
| |
Collapse
|
31
|
Remmerie M, Janssens V. PP2A: A Promising Biomarker and Therapeutic Target in Endometrial Cancer. Front Oncol 2019; 9:462. [PMID: 31214504 PMCID: PMC6558005 DOI: 10.3389/fonc.2019.00462] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2018] [Accepted: 05/14/2019] [Indexed: 12/14/2022] Open
Abstract
Over the last decade, the use of targeted therapies has immensely increased in the treatment of cancer. However, treatment for endometrial carcinomas (ECs) has lagged behind, although potential molecular markers have been identified. This is particularly problematic for the type II ECs, since these aggressive tumors are usually not responsive toward the current standard therapies. Therefore, type II ECs are responsible for most EC-related deaths, indicating the need for new treatment options. Interestingly, molecular analyses of type II ECs have uncovered frequent genetic alterations (up to 40%) in PPP2R1A, encoding the Aα subunit of the tumor suppressive heterotrimeric protein phosphatase type 2A (PP2A). PPP2R1A mutations were also reported in type I ECs and other common gynecologic cancers, albeit at much lower frequencies (0-7%). Nevertheless, PP2A inactivation in the latter cancer types is common via other mechanisms, in particular by increased expression of Cancerous Inhibitor of PP2A (CIP2A) and PP2A Methylesterase-1 (PME-1) proteins. In this review, we discuss the therapeutic potential of direct and indirect PP2A targeting compounds, possibly in combination with other anti-cancer drugs, in EC. Furthermore, we investigate the potential of the PP2A status as a predictive and/or prognostic marker for type I and II ECs.
Collapse
Affiliation(s)
| | - Veerle Janssens
- Laboratory of Protein Phosphorylation and Proteomics, Department of Cellular and Molecular Medicine, KU Leuven, Leuven, Belgium
| |
Collapse
|
32
|
Possible role of phytoestrogens in breast cancer via GPER-1/GPR30 signaling. Clin Sci (Lond) 2018; 132:2583-2598. [PMID: 30545896 DOI: 10.1042/cs20180885] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2018] [Revised: 11/12/2018] [Accepted: 11/15/2018] [Indexed: 02/07/2023]
Abstract
Estrogens generated within endocrine organs and the reproductive system act as ligands for at least three types of estrogen receptors. Estrogen receptors α (ERα) and β (ERβ) belong to the so-called classical family of estrogen receptors, whereas the G protein-coupled receptor GPR30, also known as GPER-1, has been described as a novel estrogen receptor sited in the cell membrane of target cells. Furthermore, these receptors are under stimulation of a family of exogenous estrogens, known as phytoestrogens, which are a diverse group of non-steroidal plant compounds derived from plant food consumed by humans and animals. Because phytoestrogens are omnipresent in our daily diet, they are becoming increasingly important in both human health and disease. Recent evidence indicates that in addition to classical estrogen receptors, phytoestrogens also activate GPER-1 a relevant observation since GPER-1 is involved in several physiopathological disorders and especially in estrogen-dependent diseases such as breast cancer.The first estrogen receptors discovered were the classical ERα and ERβ, but from an evolutionary point of view G protein-coupled receptors trace their origins in history to over a billion years ago suggesting that estrogen receptors like GPER-1 may have been the targets of choice for ancient phytoestrogens and/or estrogens.This review provides a comprehensive and systematic literature search on phytoestrogens and its relationship with classical estrogen receptors and GPER-1 including its role in breast cancer, an issue still under discussion.
Collapse
|
33
|
Younas M, Hano C, Giglioli-Guivarc'h N, Abbasi BH. Mechanistic evaluation of phytochemicals in breast cancer remedy: current understanding and future perspectives. RSC Adv 2018; 8:29714-29744. [PMID: 35547279 PMCID: PMC9085387 DOI: 10.1039/c8ra04879g] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2018] [Accepted: 08/15/2018] [Indexed: 12/30/2022] Open
Abstract
Breast cancer is one of the most commonly diagnosed cancers around the globe and accounts for a large proportion of fatalities in women. Despite the advancement in therapeutic and diagnostic procedures, breast cancer still represents a major challenge. Current anti-breast cancer approaches include surgical removal, radiotherapy, hormonal therapy and the use of various chemotherapeutic drugs. However, drug resistance, associated serious adverse effects, metastasis and recurrence complications still need to be resolved which demand safe and alternative strategies. In this scenario, phytochemicals have recently gained huge attention due to their safety profile and cost-effectiveness. These phytochemicals modulate various genes, gene products and signalling pathways, thereby inhibiting breast cancer cell proliferation, invasion, angiogenesis and metastasis and inducing apoptosis. Moreover, they also target breast cancer stem cells and overcome drug resistance problems in breast carcinomas. Phytochemicals as adjuvants with chemotherapeutic drugs have greatly enhanced their therapeutic efficacy. This review focuses on the recently recognized molecular mechanisms underlying breast cancer chemoprevention with the use of phytochemicals such as curcumin, resveratrol, silibinin, genistein, epigallocatechin gallate, secoisolariciresinol, thymoquinone, kaempferol, quercetin, parthenolide, sulforaphane, ginsenosides, naringenin, isoliquiritigenin, luteolin, benzyl isothiocyanate, α-mangostin, 3,3'-diindolylmethane, pterostilbene, vinca alkaloids and apigenin.
Collapse
Affiliation(s)
- Muhammad Younas
- Department of Biotechnology, Quaid-i-Azam University Islamabad-45320 Pakistan +92-51-90644121 +92-51-90644121 +33-767-97-0619
| | - Christophe Hano
- Laboratoire de Biologie des Ligneux et des Grandes Cultures (LBLGC), Plant Lignans Team, UPRES EA 1207, Université d'Orléans F 28000 Chartres France
| | | | - Bilal Haider Abbasi
- Department of Biotechnology, Quaid-i-Azam University Islamabad-45320 Pakistan +92-51-90644121 +92-51-90644121 +33-767-97-0619
- Laboratoire de Biologie des Ligneux et des Grandes Cultures (LBLGC), Plant Lignans Team, UPRES EA 1207, Université d'Orléans F 28000 Chartres France
- EA2106 Biomolecules et Biotechnologies Vegetales, Universite Francois-Rabelais de Tours Tours France
| |
Collapse
|
34
|
Hayashi T, Hikichi M, Yukitake J, Wakatsuki T, Nishio E, Utsumi T, Harada N. Forskolin increases the effect of everolimus on aromatase inhibitor-resistant breast cancer cells. Oncotarget 2018; 9:23451-23461. [PMID: 29805747 PMCID: PMC5955115 DOI: 10.18632/oncotarget.25217] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2017] [Accepted: 04/06/2018] [Indexed: 01/31/2023] Open
Abstract
Aromatase inhibitor (AI) resistance is a major obstacle in the treatment of estrogen receptor-positive breast cancer. Everolimus (EVE) ameliorates AI-resistant breast cancer and is therefore used in cancer treatment. However, some patients show resistance to EVE. Here, we used 30 clones of long-term estrogen-deprived (LTED) MCF-7 cells as a model of AI-resistant breast cancer. We examined changes in protein phosphatase type 2A (PP2A) and cancerous inhibitor of PP2A (CIP2A), a negative regulator of PP2A, in LTED cells treated with EVE. In LTED cells with high sensitivity to EVE, CIP2A expression decreased at low EVE concentrations; however, in LTED cells poorly sensitive to EVE, CIP2A and PP2A did not change upon exposure to EVE. Therefore, we hypothesized that there is a relation between expression of CIP2A and sensitivity to EVE. Knockdown of CIP2A increased the sensitivity to EVE in three clones poorly sensitive to EVE. Additionally, we found that treatment with FSK, which activates PP2A, increased the sensitivity of the cells to EVE. Our data point to CIP2A and PP2A as novel therapeutic targets for AI-resistant breast cancer.
Collapse
Affiliation(s)
- Takanori Hayashi
- Department of Biochemistry, School of Medicine, Fujita Health University, Aichi, Japan
| | - Masahiro Hikichi
- Department of Breast Surgery, School of Medicine, Fujita Health University, Aichi, Japan
| | - Jun Yukitake
- Department of Clinical Immunology, School of Health Sciences, Fujita Health University, Aichi, Japan
| | - Toru Wakatsuki
- Department of Health Science, School of Medicine, Fujita Health University, Aichi, Japan
| | - Eiji Nishio
- Department of Obstetrics and Gynecology, School of Medicine, Fujita Health University, Aichi, Japan
| | - Toshiaki Utsumi
- Department of Breast Surgery, School of Medicine, Fujita Health University, Aichi, Japan
| | - Nobuhiro Harada
- Department of Biochemistry, School of Medicine, Fujita Health University, Aichi, Japan
| |
Collapse
|
35
|
Protein phosphatase 2ACα gene knock-out results in cortical atrophy through activating hippo cascade in neuronal progenitor cells. Int J Biochem Cell Biol 2018; 95:53-62. [DOI: 10.1016/j.biocel.2017.12.015] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2017] [Revised: 12/02/2017] [Accepted: 12/19/2017] [Indexed: 12/12/2022]
|
36
|
Bi YL, Min M, Shen W, Liu Y. Genistein induced anticancer effects on pancreatic cancer cell lines involves mitochondrial apoptosis, G 0/G 1cell cycle arrest and regulation of STAT3 signalling pathway. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2018; 39:10-16. [PMID: 29433670 DOI: 10.1016/j.phymed.2017.12.001] [Citation(s) in RCA: 67] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/02/2017] [Revised: 09/04/2017] [Accepted: 12/03/2017] [Indexed: 06/08/2023]
Abstract
BACKGROUND Genistein is a natural flavonoid that has been reported to exhibit anticancer effects against different types of cancers which include, but are not limited to, breast and oral squamous cell carcinoma. PURPOSE The present study was designed to evaluate the anticancer effects of the natural flavonoid genistein against pancreatic cancer cell lines and to explore the underlying mechanism. METHODS Antiproliferative activity was investigated by MTT assay. Apoptosis was detected by DAPI and annexin V/PI staining. DNA damage was assessed by comet assay. Reactive oxygen species (ROS) and reduction of mitochondrial membrane potential (MMP) were determined by flow cytometry. Cell migration was examined by wound healing assay. Protien expressions were determined by western blotting. RESULTS Antiproliferative assay revealed that genistein reduced the cell viability of pancreatic cancer cells in a dose dependent manner with an IC50 of 20 and 25 µM against Mia-PaCa2 and PANC-1 cancer cell lines respectively. However, its antiproliferative effects were less pronounced against non-cancerous pancreatic ductal epithelial cell line (H6C7) as evident from the IC50 of 120 µM. Genistein induced significant morphological changes in pancreatic cancer cells and triggered cell cycle arrest in G0/G1 phase. DAPI staining and flow cytometric analysis revealed that genistein induced apoptosis in a dose dependent manner through generation of substantial amounts of ROS and reduction of MMP. However, treatment of the pancreatic cancer with genistein and ascorbic acid could abrogate the effects of genistein on cell viability. Protien expression analysis revealed that genistein upregulated cytosolic cytochrome c, Bax, cleaved Caspase-3 and cleaved caspase-9 expressions with concomitant downregulation of Bcl-2 expression. Moreover, genistein inhibited the phosphorylation of signal transducer and activator of transcription STAT3 proteins and downregulated the expression of survivin, cyclin D1 and ALDH1A1 in Mia-PaCa2 cells in a dose dependent manner. Interestingly, genistein could inhibit the cell migration potential of the Mia-PaCa2 cells which was further associated with the downregulation of metalloproteinases (MPP-2 and MPP-9). CONCLUSION Taken together, we propose that genistein exerts anticancer activity in pancreatic cancer cells through induction of ROS mediated mitochondrial apoptosis, cell cycle arrest and regulation of STAT3 and may therefore prove beneficial in the management of pancreatic cancers cancer.
Collapse
Affiliation(s)
- Yi-Liang Bi
- Department of Gastroenterology, Academy of Military Medical Science, 307th Hospital of PLA, Beijing 100071, China
| | - Min Min
- Department of Gastroenterology, Academy of Military Medical Science, 307th Hospital of PLA, Beijing 100071, China
| | - Wei Shen
- Department of Gastroenterology, Academy of Military Medical Science, 307th Hospital of PLA, Beijing 100071, China
| | - Yan Liu
- Department of Gastroenterology, Academy of Military Medical Science, 307th Hospital of PLA, Beijing 100071, China.
| |
Collapse
|
37
|
The role of CIP2A in cancer: A review and update. Biomed Pharmacother 2017; 96:626-633. [DOI: 10.1016/j.biopha.2017.08.146] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2017] [Revised: 08/01/2017] [Accepted: 08/13/2017] [Indexed: 12/11/2022] Open
|
38
|
Ardito F, Pellegrino MR, Perrone D, Troiano G, Cocco A, Lo Muzio L. In vitro study on anti-cancer properties of genistein in tongue cancer. Onco Targets Ther 2017; 10:5405-5415. [PMID: 29180873 PMCID: PMC5692202 DOI: 10.2147/ott.s133632] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Purpose Tongue cancer is an extremely aggressive disease and is characterized by a poor prognosis. It is a complex disease to treat and current therapies have produced mediocre results with many side effects. Some facts suggest that natural essences can support traditional cancer therapy by carrying out a synergistic function with chemotherapy. Therefore, we evaluated the antitumor effects of genistein on tongue carcinoma cells. Methods Genistein 20, 50 and 100 µM were used for 24, 48 and 72 hours on 3 tongue carcinoma cell lines. xCELLigence system was used to evaluate the effects on cell adhesion, proliferation and to calculate IC50 values. Both MTT assay and Trypan blue assay were used to evaluate alterations in cell viability, scratch assay for cell migration and Western blot analysis for expression of some proteins. Results Cell adhesion was inhibited especially between 20 and 50 µM of genistein treatment. Proliferation was reduced by 50% for treatments with 20 µM at 24 hours, with 20 or 50 µM at 48 and 50 µM at 72 hours (P<0.0001). Viability tests confirmed a proportional reduction in concentration of genistein and duration of treatments. Even cell migration was reduced significantly (P<0.001). Genistein down-regulates vitronectin, OCT4 and survivin. Conclusion This in vitro study clarifies the anti-tumor effect of genistein on tongue carcinoma. In vivo studies are needed to confirm these data and develop a suitable delivery system that is capable of acting directly on tumor.
Collapse
Affiliation(s)
- Fatima Ardito
- Department of Clinical and Experimental Medicine, Foggia University, Foggia, Italy
| | - Mario R Pellegrino
- Department of Clinical and Experimental Medicine, Foggia University, Foggia, Italy
| | - Donatella Perrone
- Department of Clinical and Experimental Medicine, Foggia University, Foggia, Italy
| | - Giuseppe Troiano
- Department of Clinical and Experimental Medicine, Foggia University, Foggia, Italy
| | - Armando Cocco
- Department of Clinical and Experimental Medicine, Foggia University, Foggia, Italy
| | - Lorenzo Lo Muzio
- Department of Clinical and Experimental Medicine, Foggia University, Foggia, Italy
| |
Collapse
|
39
|
Ye D, Li Z, Wei C. Genistein inhibits the S-phase kinase-associated protein 2 expression in breast cancer cells. Exp Ther Med 2017; 15:1069-1075. [PMID: 29434697 PMCID: PMC5772955 DOI: 10.3892/etm.2017.5489] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2017] [Accepted: 09/14/2017] [Indexed: 12/17/2022] Open
Abstract
Breast cancer is one of the most lethal cancers affecting women worldwide and was estimated to account for ~30% of all new cancer diagnoses in women. Although available evidence has proved the tumor suppressor role of genistein in cancer, the underling mechanisms have remained to be fully elucidated. S-phase kinase-associated protein 2 (Skp2) has been revealed to critically enhance the pathogenesis of multiple human cancers. The present study determined whether genistein exerts its anti-tumor function by suppressing Skp2 in breast cancer cells. Genistein significantly inhibited the proliferation, invasion and migration of breast cancer cells. Furthermore, genistein treatment also induced marked apoptosis and a typical cell cycle arrest in G2/M phase. Mechanistically, genistein treatment was identified to cause a significant downregulation of Skp2. Two crucial tumor suppressors, p21 and p27, were upregulated in genistein-treated breast cancer cells. The present results revealed that genistein exerted its tumor suppressor effect at least partially via inhibition of Skp2 and promotion of its downstream targets p21 and p27. Therefore, inactivation of Skp2 by genistein may be a promising approach for breast cancer treatment.
Collapse
Affiliation(s)
- Dengfeng Ye
- Department of Oncological Surgery, The Second Hospital of Shaoxing, Shaoxing, Zhejiang 312000, P.R. China
| | - Zhian Li
- Department of Oncological Surgery, The Second Hospital of Shaoxing, Shaoxing, Zhejiang 312000, P.R. China
| | - Chunshou Wei
- Department of Oncological Surgery, The Second Hospital of Shaoxing, Shaoxing, Zhejiang 312000, P.R. China
| |
Collapse
|
40
|
Hayashi T, Hikichi M, Yukitake J, Harada N, Utsumi T. Estradiol suppresses phosphorylation of ERα serine 167 through upregulation of PP2A in breast cancer cells. Oncol Lett 2017; 14:8060-8065. [PMID: 29344249 DOI: 10.3892/ol.2017.7216] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2017] [Accepted: 09/07/2017] [Indexed: 12/23/2022] Open
Abstract
Aromatase inhibitors (AIs) are effective endocrine therapeutics for postmenopausal women with estrogen receptor (ER)α-positive breast cancer. However, the efficacy of the treatment is often limited by the onset of AI resistance, owing to the phosphorylation of ERα serine 167 (Ser167). Previous studies have indicated that hyperactivation of the phosphoinositide-3 kinase/RAC serine/threonine-protein kinase signaling pathway occurs in AI-resistant breast cancer models, which coincides with elevated levels of ERα phosphorylation at Ser167. The tumor suppressor serine/threonine-protein phosphatase 2A (PP2A) regulates the phosphatidylinositol 3-kinase/RAC serine/threonine-protein kinase signaling pathway. A previous study indicated that PP2A inhibition decreased ERα Ser167 phosphorylation and estradiol (E2)-independent cell growth. The present study investigated the potential relevance of PP2A in E2 deprivation-resistant MCF-7 cells. E2 depletion reduced the susceptibility of MCF-7 cells to inhibitors of mechanistic target of rapamycin (mTOR) and significantly increased ERα Ser167 phosphorylation and decreased expression of PP2A. Conversely, long-term E2-deprived (LTED) MCF-7 cells, a model of AI-resistant breast cancer, exhibited decreased ERα Ser167 phosphorylation and further upregulation of PP2A in E2-containing medium. The PP2A activator forskolin (FSK) significantly inhibited LTED cell proliferation by increasing the effect of everolimus (Eve), an mTOR inhibitor. In summary, the present study provides further evidence that PP2A represents a therapeutic target for AI-resistant breast cancer.
Collapse
Affiliation(s)
- Takanori Hayashi
- Department of Biochemistry, School of Medicine, Fujita Health University, Toyoake, Aichi 470-1192, Japan
| | - Masahiro Hikichi
- Department of Breast Surgery, School of Medicine, Fujita Health University, Toyoake, Aichi 470-1192, Japan
| | - Jun Yukitake
- Department of Clinical Immunology, School of Health Sciences, Fujita Health University, Toyoake, Aichi 470-1192, Japan
| | - Nobuhiro Harada
- Department of Biochemistry, School of Medicine, Fujita Health University, Toyoake, Aichi 470-1192, Japan
| | - Toshiaki Utsumi
- Department of Breast Surgery, School of Medicine, Fujita Health University, Toyoake, Aichi 470-1192, Japan
| |
Collapse
|
41
|
Dietary Natural Products for Prevention and Treatment of Breast Cancer. Nutrients 2017; 9:nu9070728. [PMID: 28698459 PMCID: PMC5537842 DOI: 10.3390/nu9070728] [Citation(s) in RCA: 173] [Impact Index Per Article: 21.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2017] [Revised: 06/30/2017] [Accepted: 06/30/2017] [Indexed: 02/07/2023] Open
Abstract
Breast cancer is the most common cancer among females worldwide. Several epidemiological studies suggested the inverse correlation between the intake of vegetables and fruits and the incidence of breast cancer. Substantial experimental studies indicated that many dietary natural products could affect the development and progression of breast cancer, such as soy, pomegranate, mangosteen, citrus fruits, apple, grape, mango, cruciferous vegetables, ginger, garlic, black cumin, edible macro-fungi, and cereals. Their anti-breast cancer effects involve various mechanisms of action, such as downregulating ER-α expression and activity, inhibiting proliferation, migration, metastasis and angiogenesis of breast tumor cells, inducing apoptosis and cell cycle arrest, and sensitizing breast tumor cells to radiotherapy and chemotherapy. This review summarizes the potential role of dietary natural products and their major bioactive components in prevention and treatment of breast cancer, and special attention was paid to the mechanisms of action.
Collapse
|