1
|
Yuan B, Kikuchi H. Harnessing Arsenic Derivatives and Natural Agents for Enhanced Glioblastoma Therapy. Cells 2024; 13:2138. [PMID: 39768226 PMCID: PMC11674460 DOI: 10.3390/cells13242138] [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: 11/27/2024] [Revised: 12/17/2024] [Accepted: 12/21/2024] [Indexed: 01/05/2025] Open
Abstract
Glioblastoma (GBM) is the most common and lethal intracranial tumor in adults. Despite advances in the understanding of the molecular events responsible for disease development and progression, survival rates and mortality statistics for GBM patients have been virtually unchanged for decades and chemotherapeutic drugs used to treat GBM are limited. Arsenic derivatives, known as highly effective anticancer agents for leukemia therapy, has been demonstrated to exhibit cytocidal effects toward GBM cells by inducing cell death, cell cycle arrest, inhibition of migration/invasion, and angiogenesis. Differentiation induction of glioma stem-like cells (GSCs) and inhibition of neurosphere formation have also been attributed to the cytotoxicity of arsenic derivatives. Intriguingly, similar cytotoxic effects against GBM cells and GSCs have also been observed in natural agents such as anthocyanidins, tetrandrine, and bufadienolides. In the current review, we highlight the available data on the molecular mechanisms underlying the multifaceted anticancer activity of arsenic compounds and natural agents against cancer cells, especially focusing on GBM cells and GCSs. We also outline possible strategies for developing anticancer therapy by combining natural agents and arsenic compounds, as well as temozolomide, an alkylating agent used to treat GBM, in terms of improvement of chemotherapy sensitivity and minimization of side effects.
Collapse
Affiliation(s)
- Bo Yuan
- Laboratory of Pharmacology, Graduate School of Pharmaceutical Sciences, Josai University, Keyakidai, Sakado 350-0295, Saitama, Japan
| | - Hidetomo Kikuchi
- Laboratory of Pharmacotherapy, Graduate School of Pharmaceutical Sciences, Josai University, Keyakidai, Sakado 350-0295, Saitama, Japan;
| |
Collapse
|
2
|
Shahcheraghi SH, Alimardani M, Lotfi M, Lotfi M, Uversky VN, Guetchueng ST, Palakurthi SS, Charbe NB, Hromić-Jahjefendić A, Aljabali AAA, Gadewar MM, Malik S, Goyal R, El-Tanani M, Mishra V, Mishra Y, Tambuwala MM. Advances in glioblastoma multiforme: Integrating therapy and pathology perspectives. Pathol Res Pract 2024; 257:155285. [PMID: 38653089 DOI: 10.1016/j.prp.2024.155285] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/15/2024] [Revised: 03/28/2024] [Accepted: 03/30/2024] [Indexed: 04/25/2024]
Abstract
Glioblastoma, a highly lethal form of brain cancer, is characterized by its aggressive growth and resistance to conventional treatments, often resulting in limited survival. The response to therapy is notably influenced by various patient-specific genetic factors, underscoring the disease's complexity. Despite the utilization of diverse treatment modalities such as surgery, radiation, and chemotherapy, many patients experience local relapse, emphasizing the critical need for improved therapeutic strategies to effectively target these formidable tumors. Recent years have witnessed a surge in interest in natural products derived from plants, particularly alkaloids, for their potential anticancer effects. Alkaloids have shown promise in cancer chemotherapy by selectively targeting crucial signaling pathways implicated in tumor progression and survival. Specifically, they modulate the NF-κB and MAPK pathways, resulting in reduced tumor growth and altered gene expression across various cancer types. Additionally, alkaloids exhibit the capacity to induce cell cycle arrest, further impeding tumor proliferation in several malignancies. This review aims to delineate recent advances in understanding the pathology of glioblastoma multiforme (GBM) and to explore the potential therapeutic implications of alkaloids in managing this deadly disease. By segregating discussions on GBM pathology from those on alkaloid-based therapies, we provide a structured overview of the current challenges in GBM treatment and the promising opportunities presented by alkaloid-based interventions. Furthermore, we briefly discuss potential future directions in GBM research and therapy beyond alkaloids, including emerging treatment modalities or areas of investigation that hold promise for improving patient outcomes. In conclusion, our efforts offer hope for enhanced outcomes and improved quality of life for GBM patients through alkaloid-based therapies. By integrating insights from pathology and therapeutic perspectives, we underscore the significance of a comprehensive approach in addressing this devastating disease.
Collapse
Affiliation(s)
- Seyed Hossein Shahcheraghi
- Department of Medical Genetics, School of Medicine, Shahid Sadoughi University of Medical Sciences, Yazd, Iran; Infectious Diseases Research Center, Shahid Sadoughi Hospital, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Maliheh Alimardani
- Medical Genetics Research Center, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran; Department of Medical Genetics and Molecular Medicine, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran; Student Research Committee, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Malihe Lotfi
- Department of Medical Genetics and Molecular Medicine, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Marzieh Lotfi
- Abortion Research Center, Reproductive Sciences Institute, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Vladimir N Uversky
- Department of Molecular Medicine and USF Health Byrd Alzheimer's Research Institute, Morsani College of Medicine, University of South Florida, Tampa, FL 33612, USA
| | - Stephanie Tamdem Guetchueng
- Institute of Medical Research and Medicinal Plants Studies, Ministry of Scientific Research and Innovation, PO Box 6163, Yaoundé, Cameroon
| | - Sushesh Shrivastsa Palakurthi
- Department of Pharmaceutical Sciences, Irma Lerma Rangel School Of Pharmacy, Texas A&M University, Kingsville, TX 78363, USA
| | - Nitin B Charbe
- Center for Pharmacometrics & Systems Pharmacology, Department of Pharmaceutics, College of Pharmacy, University of Florida, Orlando, FL, USA
| | - Altijana Hromić-Jahjefendić
- Department of Genetics and Bioengineering, Faculty of Engineering and Natural Sciences, International University of Sarajevo, Hrasnicka cesta 15, Sarajevo 71000, Bosnia and Herzegovina
| | - Alaa A A Aljabali
- Department of Pharmaceutics and Pharmaceutical Technology, Yarmouk University, Irbid, Jordan
| | - Manoj M Gadewar
- Department of Pharmacology, School of medical and allied sciences, K.R. Mangalam University, Gurgaon, Haryana 122103, India
| | - Sumira Malik
- Amity Institute of Biotechnology, Amity University Jharkhand, Ranchi, India
| | - Rohit Goyal
- School of Pharmaceutical Sciences, Shoolini University of Biotechnology & Management Sciences, Solan, India
| | - Mohamed El-Tanani
- Ras Al Khaimah Medical and Health Sciences University, Ras Al Khaimah, United Arab Emirates
| | - Vijay Mishra
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, Punjab 144411, India
| | - Yachana Mishra
- Department of Zoology, School of Bioengineering and Biosciences, Lovely Professional University, Phagwara, Punjab 144411, India
| | - Murtaza M Tambuwala
- Ras Al Khaimah Medical and Health Sciences University, Ras Al Khaimah, United Arab Emirates; Lincoln Medical School, Brayford Pool Campus, University of Lincoln, Lincoln LN6 7TS, UK.
| |
Collapse
|
3
|
Lima EN, Lamichhane S, KC P, Ferreira ES, Koul S, Koul HK. Tetrandrine for Targeting Therapy Resistance in Cancer. Curr Top Med Chem 2024; 24:1035-1049. [PMID: 38445699 PMCID: PMC11259026 DOI: 10.2174/0115680266282360240222062032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2023] [Revised: 01/25/2024] [Accepted: 02/01/2024] [Indexed: 03/07/2024]
Abstract
During the last five decades, there has been tremendous development in our understanding of cancer biology and the development of new and novel therapeutics to target cancer. However, despite these advances, cancer remains the second leading cause of death across the globe. Most cancer deaths are attributed to the development of resistance to current therapies. There is an urgent and unmet need to address cancer therapy resistance. Tetrandrine, a bis-benzyl iso-quinoline, has shown a promising role as an anti-cancer agent. Recent work from our laboratory and others suggests that tetrandrine and its derivatives could be an excellent adjuvant to the current arsenal of anti-cancer drugs. Herein, we provide an overview of resistance mechanisms to current therapeutics and review the existing literature on the anti-cancer effects of tetrandrine and its potential use for overcoming therapy resistance in cancer.
Collapse
Affiliation(s)
- Ellen Nogueira Lima
- Department of Interdisciplinary Oncology, LSUHSC-New Orleans
- Southeast Louisiana Veterans Health Care System, New Orleans – LA
- LSU-LCMC Cancer Center, School of Medicine, Louisiana State University Health Sciences Center, New Orleans, LA, USA
| | - Santosh Lamichhane
- Department of Interdisciplinary Oncology, LSUHSC-New Orleans
- Southeast Louisiana Veterans Health Care System, New Orleans – LA
- LSU-LCMC Cancer Center, School of Medicine, Louisiana State University Health Sciences Center, New Orleans, LA, USA
| | - Pramod KC
- Department of Interdisciplinary Oncology, LSUHSC-New Orleans
- LSU-LCMC Cancer Center, School of Medicine, Louisiana State University Health Sciences Center, New Orleans, LA, USA
| | - Elisa Silva Ferreira
- Brazilian Nanotechnology National Laboratory (LNNano) Brazilian Center for Research in Energy and Materials (CNPEM) Campinas, SP, Brazil
| | - Sweaty Koul
- Department of Interdisciplinary Oncology, LSUHSC-New Orleans
- Department of Urology, LSUHSC-New Orleans
- LSU-LCMC Cancer Center, School of Medicine, Louisiana State University Health Sciences Center, New Orleans, LA, USA
| | - Hari K Koul
- Department of Interdisciplinary Oncology, LSUHSC-New Orleans
- Department of Biochemistry & Molecular Biology, LSUHSC-New Orleans
- Department of Urology, LSUHSC-New Orleans
- Southeast Louisiana Veterans Health Care System, New Orleans – LA
- LSU-LCMC Cancer Center, School of Medicine, Louisiana State University Health Sciences Center, New Orleans, LA, USA
| |
Collapse
|
4
|
Ali R, Alhaj Sulaiman A, Memon B, Pradhan S, Algethami M, Aouida M, McKay G, Madhusudan S, Abdelalim EM, Ramotar D. Altered Regulation of the Glucose Transporter GLUT3 in PRDX1 Null Cells Caused Hypersensitivity to Arsenite. Cells 2023; 12:2682. [PMID: 38067110 PMCID: PMC10705171 DOI: 10.3390/cells12232682] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2023] [Revised: 11/13/2023] [Accepted: 11/18/2023] [Indexed: 12/18/2023] Open
Abstract
Targeting tumour metabolism through glucose transporters is an attractive approach. However, the role these transporters play through interaction with other signalling proteins is not yet defined. The glucose transporter SLC2A3 (GLUT3) is a member of the solute carrier transporter proteins. GLUT3 has a high affinity for D-glucose and regulates glucose uptake in the neurons, as well as other tissues. Herein, we show that GLUT3 is involved in the uptake of arsenite, and its level is regulated by peroxiredoxin 1 (PRDX1). In the absence of PRDX1, GLUT3 mRNA and protein expression levels are low, but they are increased upon arsenite treatment, correlating with an increased uptake of glucose. The downregulation of GLUT3 by siRNA or deletion of the gene by CRISPR cas-9 confers resistance to arsenite. Additionally, the overexpression of GLUT3 sensitises the cells to arsenite. We further show that GLUT3 interacts with PRDX1, and it forms nuclear foci, which are redistributed upon arsenite exposure, as revealed by immunofluorescence analysis. We propose that GLUT3 plays a role in mediating the uptake of arsenite into cells, and its homeostatic and redox states are tightly regulated by PRDX1. As such, GLUT3 and PRDX1 are likely to be novel targets for arsenite-based cancer therapy.
Collapse
Affiliation(s)
- Reem Ali
- College of Health and Life Sciences, Hamad Bin Khalifa University (HBKU), Qatar Foundation (QF), Education City, Doha 34110, Qatar; (R.A.); (A.A.S.); (B.M.); (M.A.); (E.M.A.)
| | - Abdallah Alhaj Sulaiman
- College of Health and Life Sciences, Hamad Bin Khalifa University (HBKU), Qatar Foundation (QF), Education City, Doha 34110, Qatar; (R.A.); (A.A.S.); (B.M.); (M.A.); (E.M.A.)
| | - Bushra Memon
- College of Health and Life Sciences, Hamad Bin Khalifa University (HBKU), Qatar Foundation (QF), Education City, Doha 34110, Qatar; (R.A.); (A.A.S.); (B.M.); (M.A.); (E.M.A.)
- Diabetes Research Center, Qatar Biomedical Research Institute (QBRI), Hamad Bin Khalifa University (HBKU), Qatar Foundation (QF), Doha 34110, Qatar
| | - Singdhendubala Pradhan
- Division of Sustainable Development, College of Science and Engineering, Hamad Bin Khalifa University, Doha 34110, Qatar; (S.P.); (G.M.)
| | - Mashael Algethami
- Nottingham Biodiscovery Institute, School of Medicine, University of Nottingham, University Park, Nottingham NG7 3RD, UK; (M.A.); (S.M.)
| | - Mustapha Aouida
- College of Health and Life Sciences, Hamad Bin Khalifa University (HBKU), Qatar Foundation (QF), Education City, Doha 34110, Qatar; (R.A.); (A.A.S.); (B.M.); (M.A.); (E.M.A.)
| | - Gordon McKay
- Division of Sustainable Development, College of Science and Engineering, Hamad Bin Khalifa University, Doha 34110, Qatar; (S.P.); (G.M.)
| | - Srinivasan Madhusudan
- Nottingham Biodiscovery Institute, School of Medicine, University of Nottingham, University Park, Nottingham NG7 3RD, UK; (M.A.); (S.M.)
- Department of Oncology, Nottingham University Hospitals, Nottingham NG5 1PB, UK
| | - Essam M. Abdelalim
- College of Health and Life Sciences, Hamad Bin Khalifa University (HBKU), Qatar Foundation (QF), Education City, Doha 34110, Qatar; (R.A.); (A.A.S.); (B.M.); (M.A.); (E.M.A.)
- Diabetes Research Center, Qatar Biomedical Research Institute (QBRI), Hamad Bin Khalifa University (HBKU), Qatar Foundation (QF), Doha 34110, Qatar
| | - Dindial Ramotar
- College of Health and Life Sciences, Hamad Bin Khalifa University (HBKU), Qatar Foundation (QF), Education City, Doha 34110, Qatar; (R.A.); (A.A.S.); (B.M.); (M.A.); (E.M.A.)
| |
Collapse
|
5
|
Wang H, He Y, Zheng Q, Yang Q, Wang J, Zhu J, Zhan X. Toxicity of photoaged polyvinyl chloride microplastics to wheat seedling roots. JOURNAL OF HAZARDOUS MATERIALS 2023; 463:132816. [PMID: 39491995 DOI: 10.1016/j.jhazmat.2023.132816] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/18/2023] [Revised: 10/17/2023] [Accepted: 10/18/2023] [Indexed: 11/05/2024]
Abstract
Photoaging-prone and additive-rich polyvinyl chloride microplastics (PVC-MPs) are abundant in the terrestrial environment, However, current knowledge about the effects of PVC-MPs on terrestrial plants is lacking. Herein, we investigated the physicochemical toxicity mechanisms of photoaged PVC-MP components, i.e. leachate (L), leached PVC-particles (P), and unleached PVC-MPs (UAMP), to wheat seedling roots. 108-h photoaged components were more detrimental to root growth than unaged ones, with root length decline by 3.56%- 7.45%, indicating enhanced ecotoxicity. Notably, 108-h aged UAMP displayed more pronounced inhibition to root architecture, nutrient content and root activity, and more significant stimulation on antioxidant systems compared to 108-h aged L and P. The abovementioned phenomena suggested the presence of a synergistic effect between physical damage from P and chemical harm from L. Surface adsorption experiments demonstrated that the adsorption of photoaging induced smaller particles caused physical damage to root system. Exposure treatment suggested that there was appreciable environmental risk posed by photoaged PVC-MP-derived additives, e.g., Irgafos 168-ox and Irganox 1076. Based on principal component analysis (PCA), additives from leachate played a greater role in UAMP ecotoxicity. Therefore, PVC-MP-derived additives require more consideration and put forward an important new aspect for the impact assessment of PVC-MPs in the environment.
Collapse
Affiliation(s)
- Huiqian Wang
- College of Resources and Environmental Sciences, Nanjing Agricultural University, Nanjing, Jiangsu 210095, People's Republic of China; Stockbridge School of Agriculture, University of Massachusetts, Amherst, MA 01003, United States
| | - Yuan He
- College of Resources and Environmental Sciences, Nanjing Agricultural University, Nanjing, Jiangsu 210095, People's Republic of China
| | - Qiuping Zheng
- College of Resources and Environmental Sciences, Nanjing Agricultural University, Nanjing, Jiangsu 210095, People's Republic of China
| | - Qian Yang
- College of Resources and Environmental Sciences, Nanjing Agricultural University, Nanjing, Jiangsu 210095, People's Republic of China
| | - Jiawei Wang
- College of Resources and Environmental Sciences, Nanjing Agricultural University, Nanjing, Jiangsu 210095, People's Republic of China
| | - Jiahui Zhu
- College of Resources and Environmental Sciences, Nanjing Agricultural University, Nanjing, Jiangsu 210095, People's Republic of China
| | - Xinhua Zhan
- College of Resources and Environmental Sciences, Nanjing Agricultural University, Nanjing, Jiangsu 210095, People's Republic of China.
| |
Collapse
|
6
|
Kan LLY, Chan BCL, Leung PC, Wong CK. Natural-Product-Derived Adjunctive Treatments to Conventional Therapy and Their Immunoregulatory Activities in Triple-Negative Breast Cancer. Molecules 2023; 28:5804. [PMID: 37570775 PMCID: PMC10421415 DOI: 10.3390/molecules28155804] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2023] [Revised: 07/27/2023] [Accepted: 07/31/2023] [Indexed: 08/13/2023] Open
Abstract
Triple-negative breast cancer (TNBC) is an invasive and persistent subtype of breast cancer that is likely to be resistant to conventional treatments. The rise in immunotherapy has created new modalities to treat cancer, but due to high costs and unreliable efficacy, adjunctive and complementary treatments have sparked interest in enhancing the efficacy of currently available treatments. Natural products, which are bioactive compounds derived from natural sources, have historically been used to treat or ameliorate inflammatory diseases and symptoms. As TNBC patients have shown little to no response to immunotherapy, the potential of natural products as candidates for adjuvant immunotherapy is being explored, as well as their immunomodulatory effects on cancer. Due to the complexity of TNBC and the ever-changing tumor microenvironment, there are challenges in determining the feasibility of using natural products to enhance the efficacy or counteract the toxicity of conventional treatments. In view of technological advances in molecular docking, pharmaceutical networking, and new drug delivery systems, natural products show promise as potential candidates in adjunctive therapy. In this article, we summarize the mechanisms of action of selected natural-product-based bioactive compounds and analyze their roles and applications in combination treatments and immune regulation.
Collapse
Affiliation(s)
- Lea Ling-Yu Kan
- Institute of Chinese Medicine, The Chinese University of Hong Kong, Hong Kong, China; (L.L.-Y.K.); (B.C.-L.C.); (P.-C.L.)
- State Key Laboratory of Research on Bioactivities and Clinical Applications of Medicinal Plants, The Chinese University of Hong Kong, Hong Kong, China
| | - Ben Chung-Lap Chan
- Institute of Chinese Medicine, The Chinese University of Hong Kong, Hong Kong, China; (L.L.-Y.K.); (B.C.-L.C.); (P.-C.L.)
- State Key Laboratory of Research on Bioactivities and Clinical Applications of Medicinal Plants, The Chinese University of Hong Kong, Hong Kong, China
| | - Ping-Chung Leung
- Institute of Chinese Medicine, The Chinese University of Hong Kong, Hong Kong, China; (L.L.-Y.K.); (B.C.-L.C.); (P.-C.L.)
- State Key Laboratory of Research on Bioactivities and Clinical Applications of Medicinal Plants, The Chinese University of Hong Kong, Hong Kong, China
| | - Chun-Kwok Wong
- Institute of Chinese Medicine, The Chinese University of Hong Kong, Hong Kong, China; (L.L.-Y.K.); (B.C.-L.C.); (P.-C.L.)
- State Key Laboratory of Research on Bioactivities and Clinical Applications of Medicinal Plants, The Chinese University of Hong Kong, Hong Kong, China
- Department of Chemical Pathology, The Chinese University of Hong Kong, Hong Kong, China
- Li Dak Sum Yip Yio Chin R & D Centre for Chinese Medicine, The Chinese University of Hong Kong, Hong Kong, China
| |
Collapse
|
7
|
Yuan B, Kikuchi H, Li J, Kawabata A, Yao K, Takagi N, Okazaki M. Cytotoxic Effects of Darinaparsin, a Novel Organic Arsenical, against Human Leukemia Cells. Int J Mol Sci 2023; 24:2282. [PMID: 36768603 PMCID: PMC9916914 DOI: 10.3390/ijms24032282] [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: 12/13/2022] [Revised: 01/10/2023] [Accepted: 01/16/2023] [Indexed: 01/26/2023] Open
Abstract
To explore the molecular mechanisms of action underlying the antileukemia activities of darinaparsin, an organic arsenical approved for the treatment of peripheral T-cell lymphoma in Japan, cytotoxicity of darinaparsin was evaluated in leukemia cell lines NB4, U-937, MOLT-4 and HL-60. Darinaparsin was a more potent cytotoxic than sodium arsenite, and induced apoptosis/necrosis in NB4 and HL-60 cells. In NB4 cells exhibiting the highest susceptibility to darinaparsin, apoptosis induction was accompanied by the activation of caspase-8/-9/-3, a substantial decrease in Bid expression, and was suppressed by Boc-D-FMK, a pancaspase inhibitor, suggesting that darinaparsin triggered a convergence of the extrinsic and intrinsic pathways of apoptosis via Bid truncation. A dramatic increase in the expression level of γH2AX, a DNA damage marker, occurred in parallel with G2/M arrest. Activation of p53 and the inhibition of cdc25C/cyclin B1/cdc2 were concomitantly observed in treated cells. Downregulation of c-Myc, along with inactivation of E2F1 associated with the activation of Rb, was observed, suggesting the critical roles of p53 and c-Myc in darinaparsin-mediated G2/M arrest. Trolox, an antioxidative reagent, suppressed the apoptosis induction but failed to correct G2/M arrest, suggesting that oxidative stress primarily contributed to apoptosis induction. Suppression of Notch1 signaling was also confirmed. Our findings provide novel insights into molecular mechanisms underlying the cytotoxicity of darinaparsin and strong rationale for its new clinical application for patients with different types of cancer.
Collapse
Affiliation(s)
- Bo Yuan
- Laboratory of Pharmacology, Graduate School of Pharmaceutical Sciences, Josai University, Sakado 350-0295, Japan
| | - Hidetomo Kikuchi
- Laboratory of Pharmacotherapy, Graduate School of Pharmaceutical Sciences, Josai University, Sakado 350-0295, Japan
| | - Jingmei Li
- Laboratory of Pharmacology, Graduate School of Pharmaceutical Sciences, Josai University, Sakado 350-0295, Japan
| | - Atsushi Kawabata
- Laboratory of Pharmacology, Graduate School of Pharmaceutical Sciences, Josai University, Sakado 350-0295, Japan
| | - Kozo Yao
- Product Development Division, Solasia Pharma K.K., Tokyo 105-0011, Japan
| | - Norio Takagi
- Department of Applied Biochemistry, School of Pharmacy, Tokyo University of Pharmacy & Life Sciences, Hachioji 192-0392, Japan
| | - Mari Okazaki
- Laboratory of Pharmacology, Graduate School of Pharmaceutical Sciences, Josai University, Sakado 350-0295, Japan
| |
Collapse
|
8
|
Breast Cancer and Arsenic Anticancer Effects: Systematic Review of the Experimental Data from In Vitro Studies. BIOMED RESEARCH INTERNATIONAL 2022; 2022:8030931. [PMID: 36619302 PMCID: PMC9815927 DOI: 10.1155/2022/8030931] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Revised: 11/17/2022] [Accepted: 12/02/2022] [Indexed: 12/31/2022]
Abstract
Arsenic is a known environmental carcinogenic agent. However, under certain circumstances, it may exert anticancer effects. In this systematic review, we aim to provide information on recent developments in studies on arsenic antitumor effects in breast cancer. Research included in the review refers to experimental data from in vitro studies. The data was collected using search terms "breast cancer," "arsenic," and "anticancer" (25.05.2021). Only studies in English and published in the last 10 years were included. The search identified 123 studies from the EBSCOhost, PubMed, and Scopus databases. In the selection process, thirty full-texts were evaluated as eligible for the review. The literature of the last decade provides a lot of information on mechanisms behind anticancer effects of arsenic on breast cancer. Similar to arsenic-induced carcinogenesis, these mechanisms include the activation of the redox system and the increased production of free radicals. Targets of arsenic action are systems of cell membranes, mitochondria, pathways of intracellular transmission, and the genetic apparatus of the cell. Beneficial effects of arsenic use are possible due to significant metabolic differences between cancer and healthy cells. Further efforts are needed in order to establish modes and doses of treatment with arsenic that would provide anticancer activity with minimal toxicity.
Collapse
|
9
|
Yuan B, Li J, Miyashita SI, Kikuchi H, Xuan M, Matsuzaki H, Iwata N, Kamiuchi S, Sunaga K, Sakamoto T, Hibino Y, Okazaki M. Enhanced Cytotoxic Effects of Arenite in Combination with Active Bufadienolide Compounds against Human Glioblastoma Cell Line U-87. MOLECULES (BASEL, SWITZERLAND) 2022; 27:molecules27196577. [PMID: 36235115 PMCID: PMC9571627 DOI: 10.3390/molecules27196577] [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: 09/08/2022] [Revised: 09/25/2022] [Accepted: 09/29/2022] [Indexed: 11/16/2022]
Abstract
The cytotoxicity of a trivalent arsenic derivative (arsenite, AsIII) combined with arenobufagin or gamabufotalin was evaluated in human U-87 glioblastoma cells. Synergistic cytotoxicity with upregulated intracellular arsenic levels was observed, when treated with AsIII combined with arenobufagin instead of gamabufotalin. Apoptosis and the activation of caspase-9/-8/-3 were induced by AsIII and further strengthened by arenobufagin. The magnitude of increase in the activities of caspase-9/-3 was much greater than that of caspase-8, suggesting that the intrinsic pathway played a much more important role in the apoptosis. An increase in the number of necrotic cells, enhanced LDH leakage, and intensified G2/M phase arrest were observed. A remarkable increase in the expression level of γH2AX, a DNA damage marker, was induced by AsIII+arenobufagin. Concomitantly, the activation of autophagy was observed, suggesting that autophagic cell death associated with DNA damage was partially attributed to the cytotoxicity of AsIII+arenobufagin. Suppression of Notch signaling was confirmed in the combined regimen-treated cells, suggesting that inactivation of Jagged1/Notch signaling would probably contribute to the synergistic cytotoxic effect of AsIII+arenobufagin. Given that both AsIII and arenobufagin are capable of penetrating into the blood-brain barrier, our findings may provide fundamental insight into the clinical application of the combined regimen for glioblastoma.
Collapse
Affiliation(s)
- Bo Yuan
- Laboratory of Pharmacology, Graduate School of Pharmaceutical Sciences, Josai University, Keyakidai, Sakado 350-0295, Saitama, Japan
- Correspondence: ; Tel./Fax: +81-49-271-8026
| | - Jingmei Li
- Laboratory of Immunobiochemistry, Graduate School of Pharmaceutical Sciences, Josai University, Keyakidai, Sakado 350-0295, Saitama, Japan
| | - Shin-Ich Miyashita
- National Institute of Advanced Industrial Science and Technology (AIST), AIST Tsukuba Central 3, 1-1-1 Umezono, Tsukuba 305-8563, Ibaraki, Japan
| | - Hidetomo Kikuchi
- Laboratory of Pharmacotherapy, Graduate School of Pharmaceutical Sciences, Josai University, Keyakidai, Sakado 350-0295, Saitama, Japan
| | - Meiyan Xuan
- Laboratory of Organic and Medicinal Chemistry; Graduate School of Pharmaceutical Sciences, Josai University, Keyakidai, Sakado 350-0295, Saitama, Japan
| | - Hirokazu Matsuzaki
- Laboratory of Pharmacology, Graduate School of Pharmaceutical Sciences, Josai University, Keyakidai, Sakado 350-0295, Saitama, Japan
| | - Naohiro Iwata
- Laboratory of Immunobiochemistry, Graduate School of Pharmaceutical Sciences, Josai University, Keyakidai, Sakado 350-0295, Saitama, Japan
| | - Shinya Kamiuchi
- Laboratory of Immunobiochemistry, Graduate School of Pharmaceutical Sciences, Josai University, Keyakidai, Sakado 350-0295, Saitama, Japan
| | - Katsuyoshi Sunaga
- Laboratory of Pharmacotherapy, Graduate School of Pharmaceutical Sciences, Josai University, Keyakidai, Sakado 350-0295, Saitama, Japan
| | - Takeshi Sakamoto
- Laboratory of Organic and Medicinal Chemistry; Graduate School of Pharmaceutical Sciences, Josai University, Keyakidai, Sakado 350-0295, Saitama, Japan
| | - Yasuhide Hibino
- Laboratory of Immunobiochemistry, Graduate School of Pharmaceutical Sciences, Josai University, Keyakidai, Sakado 350-0295, Saitama, Japan
| | - Mari Okazaki
- Laboratory of Pharmacology, Graduate School of Pharmaceutical Sciences, Josai University, Keyakidai, Sakado 350-0295, Saitama, Japan
| |
Collapse
|
10
|
Reactive oxygen species-dependent upregulation of death receptor, tumor necrosis factor receptor 1, is responsible for theophylline-mediated cytotoxicity in MDA-MB-231 breast cancer cells. Anticancer Drugs 2022; 33:731-740. [PMID: 35946512 DOI: 10.1097/cad.0000000000001322] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Theophylline, a methylxanthine drug, has been used as a therapy for respiratory diseases. Recently, it has also been shown to have a potential in treating different cancers. Also, it has shown promising results in clinical trials for AML in combination therapy. Subsequently, studies have shown theophylline to kill breast cancer cells but not normal breast cells. Therefore, in this study, we have explored the molecular mechanism underlying the cytotoxic effect of theophylline on breast cancer cells. Theophylline-treated cancer cells were analyzed for the transcript and protein expression of candidate apoptotic genes such as TNFR1, caspase-8, -9, -3 using qPCR and immunoblotting, respectively. Cell viability and apoptosis was measured in the presence or absence of TNFR1 inhibitor, R7050, using AO/EtBr staining and MTT assay, respectively. Similarly, oxidative stress was studied by analyzing ROS in the presence or absence of ROS inhibitor, NAC, using DCFDA assay. Theophylline caused reduced cell viability in cancer but not normal cells. Theophylline-treated breast cancer cells showed increased expression of death receptor, TNFR1, along with elevated levels of active caspase-8, -9 and -3. Inhibition of TNFR1 reduced caspase-dependent apoptosis even in the presence of theophylline. Theophylline further caused increased ROS generation, inhibition of which resulted in reduced TNFR1-mediated apoptosis. Theophylline also increased cathepsin activity, which was reduced on exposure of cells to TNFR1 inhibitor, R7050. We conclude that ROS-mediated activation of TNFR1 is responsible for caspase-3 and cathepsin-dependent cell death in breast cancer cells on exposure to theophylline.
Collapse
|
11
|
Mo L, Zhang F, Chen F, Xia L, Huang Y, Mo Y, Zhang L, Huang D, He S, Deng J, Hao E, Du Z. Progress on structural modification of Tetrandrine with wide range of pharmacological activities. Front Pharmacol 2022; 13:978600. [PMID: 36052124 PMCID: PMC9424556 DOI: 10.3389/fphar.2022.978600] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2022] [Accepted: 07/18/2022] [Indexed: 11/23/2022] Open
Abstract
Tetrandrine (Tet), derived from the traditional Chinese herb Fangji, is a class of natural alkaloids with the structure of bisbenzylisoquinoline, which has a wide range of physiological activities and significant pharmacfological effects. However, studies and clinical applications have revealed a series of drawbacks such as its poor water solubility, low bioavailability, and the fact that it can be toxic to humans. The results of many researchers have confirmed that chemical structural modifications and nanocarrier delivery can address the limited application of Tet and improve its efficacy. In this paper, we summarize the anti-tumor efficacy and mechanism of action, anti-inflammatory efficacy and mechanism of action, and clinical applications of Tet, and describe the progress of Tet based on chemical structure modification and nanocarrier delivery, aiming to explore more diverse structures to improve the pharmacological activity of Tet and provide ideas to meet clinical needs.
Collapse
Affiliation(s)
- Liuying Mo
- Guangxi Scientific Experimental Center of Traditional Chinese Medicine, Guangxi University of Chinese Medicine, Nanning, China
- Guangxi Collaborative Innovation Center of Study on Functional Ingredients of Agricultural Residues, Nanning, China
- Guangxi Key Laboratory of Efficacy Study on Chinese Materia Medica, Nanning, China
| | - Fan Zhang
- Guangxi Scientific Experimental Center of Traditional Chinese Medicine, Guangxi University of Chinese Medicine, Nanning, China
- Guangxi Collaborative Innovation Center of Study on Functional Ingredients of Agricultural Residues, Nanning, China
- Guangxi Key Laboratory of Efficacy Study on Chinese Materia Medica, Nanning, China
- Guangxi International Zhuang Medicine Hospital Affiliated to Guangxi University of Chinese Medicine, Nanning, China
| | - Feng Chen
- Guangxi Scientific Experimental Center of Traditional Chinese Medicine, Guangxi University of Chinese Medicine, Nanning, China
- Guangxi Collaborative Innovation Center of Study on Functional Ingredients of Agricultural Residues, Nanning, China
- Guangxi Key Laboratory of Efficacy Study on Chinese Materia Medica, Nanning, China
| | - Lei Xia
- Guangxi Scientific Experimental Center of Traditional Chinese Medicine, Guangxi University of Chinese Medicine, Nanning, China
- Guangxi Collaborative Innovation Center of Study on Functional Ingredients of Agricultural Residues, Nanning, China
- Guangxi Key Laboratory of Efficacy Study on Chinese Materia Medica, Nanning, China
| | - Yi Huang
- Office of the President, Guangxi University of Chinese Medicine, Nanning, China
| | - Yuemi Mo
- Guangxi Scientific Experimental Center of Traditional Chinese Medicine, Guangxi University of Chinese Medicine, Nanning, China
- Guangxi Collaborative Innovation Center of Study on Functional Ingredients of Agricultural Residues, Nanning, China
- Guangxi Key Laboratory of Efficacy Study on Chinese Materia Medica, Nanning, China
| | - Lingqiu Zhang
- Guangxi Scientific Experimental Center of Traditional Chinese Medicine, Guangxi University of Chinese Medicine, Nanning, China
- Guangxi Collaborative Innovation Center of Study on Functional Ingredients of Agricultural Residues, Nanning, China
- Guangxi Key Laboratory of Efficacy Study on Chinese Materia Medica, Nanning, China
| | - Daquan Huang
- Guangxi Dahai Sunshine Pharmaceutical, Nanning, China
| | - Shunli He
- Guangxi Heli Pharmaceutical, Nanning, China
| | - Jiagang Deng
- Guangxi Scientific Experimental Center of Traditional Chinese Medicine, Guangxi University of Chinese Medicine, Nanning, China
- Guangxi Collaborative Innovation Center of Study on Functional Ingredients of Agricultural Residues, Nanning, China
- Guangxi Key Laboratory of Efficacy Study on Chinese Materia Medica, Nanning, China
- *Correspondence: Jiagang Deng, ; Erwei Hao, ; Zhengcai Du,
| | - Erwei Hao
- Guangxi Scientific Experimental Center of Traditional Chinese Medicine, Guangxi University of Chinese Medicine, Nanning, China
- Guangxi Collaborative Innovation Center of Study on Functional Ingredients of Agricultural Residues, Nanning, China
- Guangxi Key Laboratory of Efficacy Study on Chinese Materia Medica, Nanning, China
- *Correspondence: Jiagang Deng, ; Erwei Hao, ; Zhengcai Du,
| | - Zhengcai Du
- Guangxi Scientific Experimental Center of Traditional Chinese Medicine, Guangxi University of Chinese Medicine, Nanning, China
- Guangxi Collaborative Innovation Center of Study on Functional Ingredients of Agricultural Residues, Nanning, China
- Guangxi Key Laboratory of Efficacy Study on Chinese Materia Medica, Nanning, China
- *Correspondence: Jiagang Deng, ; Erwei Hao, ; Zhengcai Du,
| |
Collapse
|
12
|
Bhagya N, Chandrashekar KR. Autophagy and cancer: Can tetrandrine be a potent anticancer drug in the near future? Biomed Pharmacother 2022; 148:112727. [PMID: 35219119 DOI: 10.1016/j.biopha.2022.112727] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2022] [Revised: 02/14/2022] [Accepted: 02/15/2022] [Indexed: 11/02/2022] Open
Abstract
Autophagy is an essential catabolic process in mammalian cells to maintain cellular integrity and viability by degrading the old and damaged cell organelles and other contents with the help of lysosomes. Deregulation in autophagy can be one of the major contributors leading to the continuous cell proliferation and development of tumors. Tetrandrine, a bisbenzylisoquinoline alkaloid known to have potent bioactivities such as anticancer, antimicrobial, anti-inflammatory, antidiabetic, antioxidant, immunosuppressive, cardiovascular, and calcium channel blocking effects. The present review evaluated the effectiveness of tetrandrine in targeting key proteins in the autophagy pathway to induce anticancer effect based on the available literature. An attempt is also made to understand the influence of tetrandrine in regulating autophagy by mTOR dependant and mTOR-independent pathways. In addition, the review also highlights the limitations involved and future perspectives in developing tetrandrine as a chemotherapeutic drug to treat cancer.
Collapse
Affiliation(s)
- N Bhagya
- Yenepoya Research Center, Yenepoya (Deemed to be University), Deralakatte, Mangalore, Karnataka 575018, India
| | - K R Chandrashekar
- Yenepoya (Deemed to be University), Deralakatte, Mangalore, Karnataka 575018, India.
| |
Collapse
|
13
|
Islam MR, Islam F, Nafady MH, Akter M, Mitra S, Das R, Urmee H, Shohag S, Akter A, Chidambaram K, Alhumaydhi FA, Emran TB, Cavalu S. Natural Small Molecules in Breast Cancer Treatment: Understandings from a Therapeutic Viewpoint. Molecules 2022; 27:2165. [PMID: 35408561 PMCID: PMC9000328 DOI: 10.3390/molecules27072165] [Citation(s) in RCA: 78] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2022] [Revised: 03/21/2022] [Accepted: 03/23/2022] [Indexed: 12/12/2022] Open
Abstract
Breast cancer (BrCa) is the most common malignancy in women and the second most significant cause of death from cancer. BrCa is one of the most challenging malignancies to treat, and it accounts for a large percentage of cancer-related deaths. The number of cases requiring more effective BrCa therapy has increased dramatically. Scientists are looking for more productive agents, such as organic combinations, for BrCa prevention and treatment because most chemotherapeutic agents are linked to cancer metastasis, the resistance of the drugs, and side effects. Natural compounds produced by living organisms promote apoptosis and inhibit metastasis, slowing the spread of cancer. As a result, these compounds may delay the spread of BrCa, enhancing survival rates and reducing the number of deaths caused by BrCa. Several natural compounds inhibit BrCa production while lowering cancer cell proliferation and triggering cell death. Natural compounds, in addition to therapeutic approaches, are efficient and potential agents for treating BrCa. This review highlights the natural compounds demonstrated in various studies to have anticancer properties in BrCa cells. Future research into biological anti-BrCa agents may pave the way for a new era in BrCa treatment, with natural anti-BrCa drugs playing a key role in improving BrCa patient survival rates.
Collapse
Affiliation(s)
- Md. Rezaul Islam
- Department of Pharmacy, Faculty of Allied Health Sciences, Daffodil International University, Dhaka 1207, Bangladesh; (M.R.I.); (F.I.); (M.A.); (A.A.)
| | - Fahadul Islam
- Department of Pharmacy, Faculty of Allied Health Sciences, Daffodil International University, Dhaka 1207, Bangladesh; (M.R.I.); (F.I.); (M.A.); (A.A.)
| | - Mohamed H. Nafady
- Faculty of Applied Health Science Technology, Misr University for Science and Technology, Giza 12568, Egypt;
| | - Muniya Akter
- Department of Pharmacy, Faculty of Allied Health Sciences, Daffodil International University, Dhaka 1207, Bangladesh; (M.R.I.); (F.I.); (M.A.); (A.A.)
| | - Saikat Mitra
- Department of Pharmacy, Faculty of Pharmacy, University of Dhaka, Dhaka 1000, Bangladesh; (S.M.); (R.D.)
| | - Rajib Das
- Department of Pharmacy, Faculty of Pharmacy, University of Dhaka, Dhaka 1000, Bangladesh; (S.M.); (R.D.)
| | - Humaira Urmee
- Department of Pharmaceutical Science, North South University, Dhaka 1229, Bangladesh;
| | - Sheikh Shohag
- Department of Biochemistry and Molecular Biology, Faculty of Life Science, Bangabandhu Sheikh Mujibur Rahman Science and Technology University, Gopalganj 8100, Bangladesh;
| | - Aklima Akter
- Department of Pharmacy, Faculty of Allied Health Sciences, Daffodil International University, Dhaka 1207, Bangladesh; (M.R.I.); (F.I.); (M.A.); (A.A.)
| | - Kumarappan Chidambaram
- Department of Pharmacology and Toxicology, College of Pharmacy, King Khalid University, Abha 62529, Saudi Arabia;
| | - Fahad A. Alhumaydhi
- Department of Medical Laboratories, College of Applied Medical Sciences, Qassim University, Buraydah 52571, Saudi Arabia;
| | - Talha Bin Emran
- Department of Pharmacy, BGC Trust University Bangladesh, Chittagong 4381, Bangladesh
| | - Simona Cavalu
- Faculty of Medicine and Pharmacy, University of Oradea, 410087 Oradea, Romania
| |
Collapse
|
14
|
Zhang Y, Yuan B, Bian B, Zhao H, Kiyomi A, Hayashi H, Iwatani Y, Sugiura M, Takagi N. Cytotoxic Effects of Hellebrigenin and Arenobufagin Against Human Breast Cancer Cells. Front Oncol 2021; 11:711220. [PMID: 34513690 PMCID: PMC8427765 DOI: 10.3389/fonc.2021.711220] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Accepted: 08/05/2021] [Indexed: 12/12/2022] Open
Abstract
Development of new therapeutic strategies for breast cancer is urgently needed due to the sustained emergence of drug resistance, tumor recurrence and metastasis. To gain a novel insight into therapeutic approaches to fight against breast cancer, the cytocidal effects of hellebrigenin (Helle) and arenobufagin (Areno) were investigated in human estrogen receptor (ER)-positive breast cancer cell line MCF-7 and triple-negative breast cancer cell line MDA-MB-231. Helle exhibited more potent cytotoxicity than Areno in both cancer cells, and MCF-7 cells were more susceptible to both drugs in comparison with MDA-MB-231 cells. Apoptotic-like morphological characteristics, along with the downregulation of the expression level of Bcl-2 and Bcl-xL and the upregulation of the expression level of Bad, were observed in Helle-treated MCF-7 cells. Helle also caused the activation of caspase-8, caspase-9, along with the cleavage of poly(ADP-ribose) polymerase in MCF-7 cells. Helle-mediated necrosis-like phenotype, as evidenced by the increased propidium iodide (PI)-positive cells was further observed. G2/M cell cycle arrest was also induced by Helle in the cells. Upregulation of the expression level of p21 and downregulation of the expression level of cyclin D1, cyclin E1, cdc25C and survivin were observed in MCF-7 cells treated with Helle and occurred in parallel with G2/M arrest. Autophagy was triggered in MCF-7 cells and the addition of wortmannin or 3-MA, two well-known autophagy inhibitors, slightly but significantly rescued the cells. Furthermore, similar alterations of some key molecules associated with the aforementioned biological phenomena were observed in MDA-MB-231 cells. Intriguingly, the numbers of PI-positive cells in Helle-treated MCF-7 cells were significantly reduced by wortmannin and 3-MA, respectively. In addition, Helle-triggered G2/M arrest was significantly corrected by wortmannin, suggesting autophagy induction contributed to Helle-induced cytotoxicity of breast cancer cells by modulating necrosis and cell cycle arrest. Collectively, our results suggested potential usefulness of both Helle and Areno in developing therapeutic strategies to treat patients with different types of breast cancer, especially ER-positive breast cancer.
Collapse
Affiliation(s)
- Yu Zhang
- Department of Applied Biochemistry, Tokyo University of Pharmacy & Life Sciences, Hachioji, Japan.,Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, China
| | - Bo Yuan
- Laboratory of Pharmacology, School of Pharmacy, Faculty of Pharmaceutical Sciences, Josai University, Sakado, Japan
| | - Baolin Bian
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, China
| | - Haiyu Zhao
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, China
| | - Anna Kiyomi
- Department of Drug Safety and Risk Management, Tokyo University of Pharmacy & Life Sciences, Hachioji, Japan
| | - Hideki Hayashi
- Department of Applied Biochemistry, Tokyo University of Pharmacy & Life Sciences, Hachioji, Japan
| | - Yui Iwatani
- Department of Applied Biochemistry, Tokyo University of Pharmacy & Life Sciences, Hachioji, Japan
| | - Munetoshi Sugiura
- Department of Drug Safety and Risk Management, Tokyo University of Pharmacy & Life Sciences, Hachioji, Japan
| | - Norio Takagi
- Department of Applied Biochemistry, Tokyo University of Pharmacy & Life Sciences, Hachioji, Japan
| |
Collapse
|
15
|
The Anticancer Effects of Flavonoids through miRNAs Modulations in Triple-Negative Breast Cancer. Nutrients 2021; 13:nu13041212. [PMID: 33916931 PMCID: PMC8067583 DOI: 10.3390/nu13041212] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2021] [Revised: 03/31/2021] [Accepted: 04/02/2021] [Indexed: 12/31/2022] Open
Abstract
Triple- negative breast cancer (TNBC) incidence rate has regularly risen over the last decades and is expected to increase in the future. Finding novel treatment options with minimum or no toxicity is of great importance in treating or preventing TNBC. Flavonoids are new attractive molecules that might fulfill this promising therapeutic option. Flavonoids have shown many biological activities, including antioxidant, anti-inflammatory, and anticancer effects. In addition to their anticancer effects by arresting the cell cycle, inducing apoptosis, and suppressing cancer cell proliferation, flavonoids can modulate non-coding microRNAs (miRNAs) function. Several preclinical and epidemiological studies indicate the possible therapeutic potential of these compounds. Flavonoids display a unique ability to change miRNAs' levels via different mechanisms, either by suppressing oncogenic miRNAs or activating oncosuppressor miRNAs or affecting transcriptional, epigenetic miRNA processing in TNBC. Flavonoids are not only involved in the regulation of miRNA-mediated cancer initiation, growth, proliferation, differentiation, invasion, metastasis, and epithelial-to-mesenchymal transition (EMT), but also control miRNAs-mediated biological processes that significantly impact TNBC, such as cell cycle, immune system, mitochondrial dysregulation, modulating signaling pathways, inflammation, and angiogenesis. In this review, we highlighted the role of miRNAs in TNBC cancer progression and the effect of flavonoids on miRNA regulation, emphasizing their anticipated role in the prevention and treatment of TNBC.
Collapse
|
16
|
Yuan B, Xu K, Shimada R, Li J, Hayashi H, Okazaki M, Takagi N. Cytotoxic Effects of Arsenite in Combination With Gamabufotalin Against Human Glioblastoma Cell Lines. Front Oncol 2021; 11:628914. [PMID: 33796463 PMCID: PMC8009626 DOI: 10.3389/fonc.2021.628914] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2020] [Accepted: 01/29/2021] [Indexed: 12/13/2022] Open
Abstract
Glioblastoma is a fatal primary malignant brain tumor, and the 5-year survival rate of treated glioblastoma patients still remains <5%. Considering the sustained development of metastasis, tumor recurrence, and drug resistance, there is an urgent need for the novel therapeutic approaches to combat glioblastoma. Trivalent arsenic derivative (arsenite, AsIII) with remarkable clinical efficacy in leukemia has been shown to exert cytocidal effect against glioblastoma cells. Gamabufotalin, an active bufadienolide compound, also shows selective cytocidal effect against glioblastoma cells, and has been suggested to serve as a promising adjuvant therapeutic agent to potentiate therapeutic effect of conventional anticancer drugs. In order to gain novel insight into therapeutic approaches against glioblastoma, the cytotoxicity of AsIII and gamabufotalin was explored in the human glioblastoma cell lines U-87 and U-251. In comparison with U-251 cells, U-87 cells were highly susceptible to the two drugs, alone or in combination. More importantly, clinically achieved concentrations of AsIII combined with gamabufotalin exhibited synergistic cytotoxicity against U-87 cells, whereas showed much less cytotoxicity to human normal peripheral blood mononuclear cells. G2/M cell cycle arrest was induced by each single drug, and further augmented by their combination in U-87 cells. Downregulation of the expression levels of cdc25C, Cyclin B1, cdc2, and survivin was observed in U-87 cells treated with the combined regimen and occurred in parallel with G2/M arrest. Concomitantly, lactate dehydrogenase leakage was also observed. Intriguingly, SB203580, a specific inhibitor of p38 MAPK, intensified the cytotoxicity of the combined regimen in U-87 cells, whereas wortmannin, a potent autophagy inhibitor, significantly rescued the cells. Collectively, G2/M arrest, necrosis and autophagy appeared to cooperatively contribute to the synergistic cytotoxicity of AsIII and gamabufotalin. Given that p38 MAPK serves an essential role in promoting glioblastoma cell survival, developing a possible strategy composed of AsIII, gamabufotalin, and a p38 MAPK inhibitor may provide novel insight into approaches designed to combat glioblastoma.
Collapse
Affiliation(s)
- Bo Yuan
- Laboratory of Pharmacology, Faculty of Pharmaceutical Sciences, Josai University, Sakado, Japan.,Department of Applied Biochemistry, Tokyo University of Pharmacy & Life Sciences, Hachioji, Japan
| | - Kang Xu
- Department of Applied Biochemistry, Tokyo University of Pharmacy & Life Sciences, Hachioji, Japan
| | - Ryota Shimada
- Department of Applied Biochemistry, Tokyo University of Pharmacy & Life Sciences, Hachioji, Japan
| | - JingZhe Li
- Beijing Key Laboratory of Research of Chinese Medicine on Prevention and Treatment for Major Diseases, Experimental Research Center, China Academy of Chinese Medical Sciences, Beijing, China
| | - Hideki Hayashi
- Department of Applied Biochemistry, Tokyo University of Pharmacy & Life Sciences, Hachioji, Japan
| | - Mari Okazaki
- Laboratory of Pharmacology, Faculty of Pharmaceutical Sciences, Josai University, Sakado, Japan
| | - Norio Takagi
- Department of Applied Biochemistry, Tokyo University of Pharmacy & Life Sciences, Hachioji, Japan
| |
Collapse
|
17
|
Elsewedy HS, Aldhubiab BE, Mahdy MA, Elnahas HM. Brucine PEGylated nanoemulsion: In vitro and in vivo evaluation. Colloids Surf A Physicochem Eng Asp 2021. [DOI: 10.1016/j.colsurfa.2020.125618] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
|
18
|
Elsewedy HS, Dhubiab BEA, Mahdy MA, Elnahas HM. Development, optimization, and evaluation of PEGylated brucine-loaded PLGA nanoparticles. Drug Deliv 2020; 27:1134-1146. [PMID: 32729331 PMCID: PMC7470130 DOI: 10.1080/10717544.2020.1797237] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2020] [Revised: 07/13/2020] [Accepted: 07/13/2020] [Indexed: 12/19/2022] Open
Abstract
The application of nanotechnology to drug delivery systems for cancer therapy has progressively received great attention. The most heavily investigated approach is the development of nanoparticles (NPs) utilizing biodegradable and biocompatible polymers such as poly (lactic-co-glycolic acid) (PLGA). These NPs could be further improved by surface modification utilizing a hydrophilic biodegradable polymer such as polyethylene glycol (PEG) to achieve passive targeting. Modified NPs can deliver drugs such as brucine (BRU), which has shown its potential in cancer therapy. The objective of the current investigation was to develop and evaluate the passive targeting of long-circulating PLGA NPs loaded with BRU. NPs were characterized in terms of drug-excipient compatibility studies, including FTIR and DSC; physicochemical evaluations including particle size, zeta potential, morphological evaluation, entrapment efficiency and percentage yield; total serum protein adsorbed onto NP surfaces; and in vitro release of the loaded drug. Factorial design was employed to attain optimal PLGA-loaded NPs. Finally, the in vivo anti-tumor activity of BRU-loaded PLGA NPs was evaluated in tumor-bearing mice. The NPs obtained had smooth surfaces with particle sizes ranged from 94 ± 3.05 to 253 ± 8.7 nm with slightly positive surface charge ranged from 1.09 ± 0.15 to 3.71 ± 0.44 mV. Entrapment of BRU ranged between 37.5 ± 1.8% and 77 ± 1.3% with yields not less than 70.8%. Total protein adsorbed was less than 25.5 µg total protein/1 mg NP. In vitro drug release was less than 99.1% at 168 h. Finally, significant reductions in tumor growth rate and mortality rate were observed for PEG PLGA NP formulations compared to both BRU solution and naked NPs.
Collapse
Affiliation(s)
- Heba S. Elsewedy
- Department of Pharmaceutics and Industrial
Pharmacy, Faculty of Pharmacy, Zagazig University, Zagazig,
Egypt
- Department of Pharmaceutical Sciences, College
of Clinical Pharmacy, King Faisal University, Saudi
Arabia
| | - Bandar E. Al Dhubiab
- Department of Pharmaceutical Sciences, College
of Clinical Pharmacy, King Faisal University, Saudi
Arabia
| | - Mahmoud A. Mahdy
- Department of Pharmaceutics and Industrial
Pharmacy, Faculty of Pharmacy, Zagazig University, Zagazig,
Egypt
| | - Hanan M. Elnahas
- Department of Pharmaceutics and Industrial
Pharmacy, Faculty of Pharmacy, Zagazig University, Zagazig,
Egypt
| |
Collapse
|
19
|
Anti-Tumor Effects of Sodium Meta-Arsenite in Glioblastoma Cells with Higher Akt Activities. Int J Mol Sci 2020; 21:ijms21238982. [PMID: 33256086 PMCID: PMC7729740 DOI: 10.3390/ijms21238982] [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: 10/28/2020] [Revised: 11/23/2020] [Accepted: 11/24/2020] [Indexed: 11/18/2022] Open
Abstract
Glioblastoma is a type of aggressive brain tumor that grows very fast and evades surrounding normal brain, lead to treatment failure. Glioblastomas are associated with Akt activation due to somatic alterations in PI3 kinase/Akt pathway and/or PTEN tumor suppressor. Sodium meta-arsenite, KML001 is an orally bioavailable, water-soluble, and trivalent arsenical and it shows antitumoral effects in several solid tumor cells via inhibiting oncogenic signaling, including Akt and MAPK. Here, we evaluated the effect of sodium meta-arsenite, KML001, on the growth of human glioblastoma cell lines with different PTEN expression status and Akt activation, including PTEN-deficient cells (U87-MG and U251) and PTEN-positive cells (LN229). The growth-inhibitory effect of KML001 was stronger in U87-MG and U251 cells, which exhibited higher Akt activity than LN229 cells. KML001 deactivated Akt and decreased its protein levels via proteasomal degradation in U87-MG cells. KML001 upregulated mutant PTEN levels via inhibition of its proteasomal degradation. KML001 inhibited cell growth more effectively in active Akt-overexpressing LN229 cells than in mock-expressing LN229 cells. Consistent with these results, KML001 sensitized PTEN-deficient cells more strongly to growth inhibition than it did PTEN-positive cells in prostate and breast cancer cell lines. Finally, we illustrated in vivo anti-tumor effects of KML001 using an intracranial xenograft mouse model. These results suggest that KML001 could be an effective chemotherapeutic drug for the treatment of glioblastoma cancer patients with higher Akt activity and PTEN loss.
Collapse
|
20
|
Luan F, He X, Zeng N. Tetrandrine: a review of its anticancer potentials, clinical settings, pharmacokinetics and drug delivery systems. J Pharm Pharmacol 2020; 72:1491-1512. [PMID: 32696989 DOI: 10.1111/jphp.13339] [Citation(s) in RCA: 60] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2020] [Accepted: 06/21/2020] [Indexed: 12/18/2022]
Abstract
OBJECTIVES Tetrandrine, a natural bisbenzylisoquinoline alkaloid, possesses promising anticancer activities on diverse tumours. This review provides systematically organized information on cancers of tetrandrine in vivo and in vitro, discuss the related molecular mechanisms and put forward some new insights for the future investigations. KEY FINDINGS Anticancer activities of tetrandrine have been reported comprehensively, including lung cancer, colon cancer, bladder cancer, prostate cancer, ovarian cancer, gastric cancer, breast cancer, pancreatic cancer, cervical cancer and liver cancer. The potential molecular mechanisms corresponding to the anticancer activities of tetrandrine might be related to induce cancer cell apoptosis, autophagy and cell cycle arrest, inhibit cell proliferation, migration and invasion, ameliorate metastasis and suppress tumour cell growth. Pharmaceutical applications of tetrandrine combined with nanoparticle delivery system including liposomes, microspheres and nanoparticles with better therapeutic efficiency have been designed and applied encapsulate tetrandrine to enhance its stability and efficacy in cancer treatment. SUMMARY Tetrandrine was proven to have definite antitumour activities. However, the safety, bioavailability and pharmacokinetic parameter studies on tetrandrine are very limited in animal models, especially in clinical settings. Our present review on anticancer potentials of tetrandrine would be necessary and highly beneficial for providing guidelines and directions for further research of tetrandrine.
Collapse
Affiliation(s)
- Fei Luan
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Xirui He
- Department of Bioengineering, Zhuhai Campus of Zunyi Medical University, Zhuhai, China
| | - Nan Zeng
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| |
Collapse
|
21
|
Yu B, Yuan B, Li J, Kiyomi A, Kikuchi H, Hayashi H, Hu X, Okazaki M, Sugiura M, Hirano T, Fan Y, Pei X, Takagi N. JNK and Autophagy Independently Contributed to Cytotoxicity of Arsenite combined With Tetrandrine via Modulating Cell Cycle Progression in Human Breast Cancer Cells. Front Pharmacol 2020; 11:1087. [PMID: 32765280 PMCID: PMC7379898 DOI: 10.3389/fphar.2020.01087] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2020] [Accepted: 07/03/2020] [Indexed: 12/12/2022] Open
Abstract
Novel therapeutic strategies for breast cancer are urgently needed due to the sustained development of drug resistance and tumor recurrence. Trivalent arsenic derivative (arsenite, AsIII) has been reported to induce cytotoxicity in breast cancer cells. We recently demonstrated that AsIII plus tetrandrine (Tetra), a Chinese plant-derived alkaloid, exerted potent antitumor activity against human breast cancer cells, however, the underlying mechanisms for their action have not been well defined. In order to provide fundamental insights for understanding the action of AsIII plus Tetra, the effects of the combined regimen on two breast cancer cell lines T47D and MDA-MB-231 were evaluated. Compared to T47D cells, MDA-MB-231 cells were much more susceptible to the synergistic cytotoxic effects of AsIII and Tetra. Besides the induction of apoptotic/necrotic cell death, S-phase arrest and autophagic cell death were also observed in MDA-MB-231 cells. Exposure of MDA-MB-231 cells to AsIII and Tetra caused the activation of MAPKs. Cytotoxicity of the combined regimen in MDA-MB-231 cell was significantly abrogated by SP600125, a potent c-Jun N-terminal kinase (JNK) inhibitor. However, similar abrogation was not caused by p38 and ERK inhibitors. The addition of either autophagy inhibitors (3-methyladenine or wortmannin) or SP600125 corrected the combined regimen-triggered S-phase arrest, whereas had little effect on the apoptosis/necrosis induction in the cells. Surprisingly, SP600125NC, a negative control for SP600125, significantly strengthened S-phase arrest and the cytotoxicity induced by the combined regimen. The addition of SP600125 did not alter autophagy induction. In conclusion, the cytotoxicity of AsIII combined with Tetra was attributed to the induction of S-phase arrest, apoptotic/necrotic and autophagic cell death. The enhanced cytotoxicity of the two drugs by SP600125NC might be explained by its capability to strengthen S-phase arrest. Our results suggested that JNK and autophagy independently contributed to the cytotoxicity via modulating cell cycle progression. The study further provides fundamental insights for the development of AsIII in combination with Tetra for patients with different types of breast cancer.
Collapse
Affiliation(s)
- Bowen Yu
- Department of Applied Biochemistry, School of Pharmacy, Tokyo University of Pharmacy & Life Sciences, Tokyo, Japan.,Galactophore Department, Beijing University of Chinese Medicine Third Affiliated Hospital, Beijing, China
| | - Bo Yuan
- Department of Applied Biochemistry, School of Pharmacy, Tokyo University of Pharmacy & Life Sciences, Tokyo, Japan.,Laboratory of Pharmacology, School of Pharmacy, Josai University, Saitama, Japan
| | - JingZhe Li
- Beijing Key Laboratory of Research of Chinese Medicine on Prevention and Treatment for Major Diseases, Experimental Research Center, China Academy of Chinese Medical Sciences, Beijing, China
| | - Anna Kiyomi
- Drug Safety and Risk Management, School of Pharmacy, Tokyo University of Pharmacy & Life Sciences, Tokyo, Japan
| | - Hidetomo Kikuchi
- Laboratory of Pharmacotherapy, Department of Clinical Dietetics and Human Nutrition, Faculty of Pharmacy and Pharmaceutical Sciences, Josai University, Saitama, Japan
| | - Hideki Hayashi
- Department of Applied Biochemistry, School of Pharmacy, Tokyo University of Pharmacy & Life Sciences, Tokyo, Japan
| | - Xiaomei Hu
- Hematology Department, XiYuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Mari Okazaki
- Laboratory of Pharmacology, School of Pharmacy, Josai University, Saitama, Japan
| | - Munetoshi Sugiura
- Drug Safety and Risk Management, School of Pharmacy, Tokyo University of Pharmacy & Life Sciences, Tokyo, Japan
| | - Toshihiko Hirano
- Clinical Pharmacology, School of Pharmacy, Tokyo University of Pharmacy & Life Sciences, Tokyo, Japan
| | - Yingyi Fan
- Galactophore Department, Beijing University of Chinese Medicine Third Affiliated Hospital, Beijing, China
| | - Xiaohua Pei
- Galactophore Department, Beijing University of Chinese Medicine Third Affiliated Hospital, Beijing, China
| | - Norio Takagi
- Department of Applied Biochemistry, School of Pharmacy, Tokyo University of Pharmacy & Life Sciences, Tokyo, Japan
| |
Collapse
|
22
|
Noel B, Singh SK, Lillard JW, Singh R. Role of natural compounds in preventing and treating breast cancer. Front Biosci (Schol Ed) 2020; 12:137-160. [PMID: 32114452 DOI: 10.2741/s544] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Breast cancer (BrCa) is the most commonly diagnosed cancer and the second leading cause of cancer-related death in women. Alarming increases in the cases quests for more effective treatment of BrCa. As most chemotherapeutic drugs are associated with drug resistance, cancer relapse, and side effects, scientists are turning to agents with more efficacy, such as natural compounds for treatment and prevention of BrCa. Selected natural compounds, substances derived from living organisms, promote apoptosis and inhibit metastasis, preventing cancer growth. As a result, these compounds have the potential to suppress BrCa progression, thus increasing patient survival rates and decreasing the number of BrCa-related deaths. In this review, we summarize natural compounds that have displayed, anti-cancer effects on BrCa cells in various studies. These natural compounds inhibit the development of BrCa, suppress the growth of cancer cells, and promote cell death. We conclude that natural compounds are efficient, effective and promising agents for treating BrCa other than therapeutic methods.
Collapse
Affiliation(s)
- Brianna Noel
- Morehouse School of Medicine, 720 Westview Drive SW, Atlanta GA 30310
| | - Santosh Kumar Singh
- Department of Microbiology, Biochemistry and Immunology,Morehouse School of Medicine, 720 Westview drive, SW, Atlanta- 30310 USA
| | - James W Lillard
- Morehouse School of Medicine, 720 Westview Drive SW, Atlanta GA 30310
| | - Rajesh Singh
- Morehouse School of Medicine, 720 Westview Drive SW, Atlanta,
| |
Collapse
|
23
|
Yu B, Yuan B, Kiyomi A, Kikuchi H, Hayashi H, Hu X, Okazaki M, Sugiura M, Hirano T, Pei X, Takagi N. Differentiation induction of human breast cancer cells by arsenite in combination with tetrandrine. Am J Transl Res 2019; 11:7310-7323. [PMID: 31934280 PMCID: PMC6943472] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2019] [Accepted: 10/25/2019] [Indexed: 06/10/2023]
Abstract
BACKGROUND To provide novel insight into the development of new therapeutic strategies to combat breast cancer, differentiation-inducing activity of clinically achievable concentrations of arsenite (AsIII) and tetrandrine (Tetra) was investigated in breast cancer cell lines MDA-MB-231 and MCF-7. METHODS Differentiation induction of cancer cells was analyzed by flow cytometer. Alterations of genes related to differentiation, and proliferation of human normal peripheral blood mononuclear cells (PBMCs) were analyzed using western blotting and cell viability assay, respectively. RESULTS Exposure to Tetra alone or in combination with AsIII induced differentiation of both cells characterized by upregulation of ICAM-1, downregulation of Her2/neu. In comparison with MCF-7, the combination of lower concentrations of AsIII and Tetra induced differentiation of MDA-MB-231, indicating that MDA-MB-231 cells were highly susceptible to differentiation. The differentiation occurred in parallel with activation of Erk signaling pathway, and was abolished by PD98059, a potent Erk inhibitor. Consistent with in vitro experimental results, the upregulation of ICAM-1 and the activation of Erk signaling pathway were also observed in MDA-MB-231 breast tumors in xenograft mouse obtained from our previous study. No obvious proliferation inhibition of PBMCs was observed following the exposure to AsIII combined with Tetra at the concentrations capable of inducing differentiation of MDA-MB-231 cells. CONCLUSION The Erk signaling pathway may be crucially involved in the differentiation induction of breast cancer cells in vitro and in vivo. Collectively, our results suggest that the combination can probably serve as promising candidates for the development of novel therapeutic approaches for different types of breast cancer.
Collapse
Affiliation(s)
- Bowen Yu
- Department of Applied Biochemistry, School of Pharmacy, Tokyo University of Pharmacy & Life Sciences1432-1 Horinouchi, Hachioji, Tokyo 192-0392, Japan
- Beijing University of Chinese Medicine Third Affiliated HospitalBeijing 100029, P. R. China
| | - Bo Yuan
- Department of Applied Biochemistry, School of Pharmacy, Tokyo University of Pharmacy & Life Sciences1432-1 Horinouchi, Hachioji, Tokyo 192-0392, Japan
- Laboratory of Pharmacology, School of Pharmacy, Faculty of Pharmacy and Pharmaceutical Sciences, Josai University1-1 Keyakidai, Sakado, Saitama 350-0295, Japan
| | - Anna Kiyomi
- Department of Drug Safety and Risk Management, School of Pharmacy, Tokyo University of Pharmacy & Life Sciences1432-1 Horinouchi, Hachioji, Tokyo 192-0392, Japan
| | - Hidetomo Kikuchi
- Laboratory of Pharmacotherapy, Department of Clinical Dietetics and Human Nutrition, Faculty of Pharmacy and Pharmaceutical Sciences, Josai University1-1 Keyakidai, Sakado, Saitama 350-0295, Japan
| | - Hideki Hayashi
- Department of Applied Biochemistry, School of Pharmacy, Tokyo University of Pharmacy & Life Sciences1432-1 Horinouchi, Hachioji, Tokyo 192-0392, Japan
| | - Xiaomei Hu
- Xiyuan Hospital, China Academy of Chinese Medical SciencesBeijing 100091, P. R. China
| | - Mari Okazaki
- Laboratory of Pharmacology, School of Pharmacy, Faculty of Pharmacy and Pharmaceutical Sciences, Josai University1-1 Keyakidai, Sakado, Saitama 350-0295, Japan
| | - Munetoshi Sugiura
- Department of Drug Safety and Risk Management, School of Pharmacy, Tokyo University of Pharmacy & Life Sciences1432-1 Horinouchi, Hachioji, Tokyo 192-0392, Japan
| | - Toshihiko Hirano
- Department of Clinical Pharmacology, School of Pharmacy, Tokyo University of Pharmacy & Life Sciences1432-1 Horinouchi, Hachioji, Tokyo 192-0392, Japan
| | - Xiaohua Pei
- Beijing University of Chinese Medicine Third Affiliated HospitalBeijing 100029, P. R. China
| | - Norio Takagi
- Department of Applied Biochemistry, School of Pharmacy, Tokyo University of Pharmacy & Life Sciences1432-1 Horinouchi, Hachioji, Tokyo 192-0392, Japan
| |
Collapse
|
24
|
Yuan B, Shimada R, Xu K, Han L, Si N, Zhao H, Bian B, Hayashi H, Okazaki M, Takagi N. Multiple cytotoxic effects of gamabufotalin against human glioblastoma cell line U-87. Chem Biol Interact 2019; 314:108849. [DOI: 10.1016/j.cbi.2019.108849] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2019] [Revised: 08/06/2019] [Accepted: 10/09/2019] [Indexed: 12/13/2022]
|
25
|
Sun J, Zhang Y, Zhen Y, Cui J, Hu G, Lin Y. Antitumor activity of tetrandrine citrate in human glioma U87 cells in vitro and in vivo. Oncol Rep 2019; 42:2345-2354. [PMID: 31638254 PMCID: PMC6826321 DOI: 10.3892/or.2019.7372] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2019] [Accepted: 09/11/2019] [Indexed: 12/18/2022] Open
Abstract
Since the current methods of treatment for malignant glioma, radiotherapy and chemotherapy, are unsatisfactory, the development of novel therapeutic compounds is required. In the present study, the inhibitory effect of tetrandrine citrate (TetC) on the proliferation of human glioma U87 cells, as well as its mechanism of action, were investigated. An MTT assay was used to assess cell viability in vitro, and the production of intracellular reactive oxygen species (ROS) was determined by assessing the fluorescence intensity of 2,7-dichlorofluorescein (DCF). Flow cytometry was used to determine the level of apoptosis and cell cycle status, and the protein expression levels of apoptosis-associated proteins were determined using western blotting. Additionally, the antitumor activity of TetC was assessed in vivo using a nude mouse xenograft model. The results revealed that in vitro, the proliferative rate of U87, U251 and human umbilical vein endothelial cells (HUVECs) was significantly reduced in a dose-dependent manner following treatment with TetC, although TetC had the greatest inhibitory effect on U87 cells. The vacuolization and apoptosis of U87 cells was induced using 10 and 20 µmol/l TetC, respectively. The overall proliferative inhibition was associated with an increase in the levels of ROS and apoptosis. In TetC-treated cells, the expression levels of apoptosis-related proteins, including cleaved (CL) caspase-3, Fas, phosphorylated (p)-p38 and p-JNK, were increased, whereas those of caspase-3 and Bcl-2 were decreased. In vivo, TetC was highly effective at inhibiting the growth of human glioma U87 ×enografts in BALB/c nude mice, with a percentage growth inhibition of ≥68.7%. These findings indicated that the potent antitumor activity of TetC may be mediated through an increase in ROS levels, the downregulation of Bcl-2, and the upregulation of CL caspase-3, Fas, p-p38 and p-JNK expression levels.
Collapse
Affiliation(s)
- Jingyu Sun
- Key Laboratory of Geriatrics, Beijing Hospital, National Center of Gerontology, Beijing 100730, P.R. China
| | - Yang Zhang
- Peking University Fifth School of Clinical Medicine, Beijing 100730, P.R. China
| | - Yongzhan Zhen
- Tangshan Key Laboratory for Preclinical and Basic Research on Chronic Diseases, School of Basic Medical Sciences, North China University of Science and Technology, Tangshan 063000, P.R. China
| | - Ju Cui
- Key Laboratory of Geriatrics, Beijing Hospital, National Center of Gerontology, Beijing 100730, P.R. China
| | - Gang Hu
- Key Laboratory of Geriatrics, Beijing Hospital, National Center of Gerontology, Beijing 100730, P.R. China
| | - Yajun Lin
- Key Laboratory of Geriatrics, Beijing Hospital, National Center of Gerontology, Beijing 100730, P.R. China
| |
Collapse
|
26
|
Kikuchi H, Yuan B, Hu X, Okazaki M. Chemopreventive and anticancer activity of flavonoids and its possibility for clinical use by combining with conventional chemotherapeutic agents. Am J Cancer Res 2019; 9:1517-1535. [PMID: 31497340 PMCID: PMC6726994] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2019] [Accepted: 06/11/2019] [Indexed: 06/10/2023] Open
Abstract
Cancer is a diverse class of diseases characterized by uncontrolled cell growth with the potential to invade and spread to other parts of the body, and continues to be one of the leading causes of death worldwide. Conventional cancer treatment modalities include antitumor drugs, surgical resection, locally targeted therapies such as radiation therapy. Along with improved understanding of the molecular pathogenesis of various cancers, generation and the use of smart targeted anti-cancer drugs have been challenged. The need for novel therapeutic strategies remains paramount given the sustained development of drug resistance, tumor recurrence, and metastasis. Development of new strategies aimed at improving chemotherapy sensitivity and minimizing the adverse side effects is thus essential for obtaining satisfied therapeutic outcomes for patients and enhancing their quality of life. Emerging evidence has reported that many cancer patients use either herbs employed in complementary therapies or dietary agents that influence cellular signaling worldwide. Numerous components of edible plants, collectively termed phytochemicals that have beneficial effects for health, are being reported increasingly in the scientific literature. Of those, flavonoids have attracted much attention by virtue of its wide variety of biological functions including antioxidant, anti-inflammatory, and anticancer activity. In this review, we highlight the molecular mechanisms underlying its multiple pharmacological effects, especially focusing on cancer chemoprevention. We further discuss possible strategies to develop anticancer therapy by combining flavonoids nutraceuticals and conventional chemotherapeutic agents. We also highlight numerous pharmacokinetic challenges such as bioavailability, drug-drug interactions, which are still fundamental questions concerning its future clinical application.
Collapse
Affiliation(s)
- Hidetomo Kikuchi
- Laboratory of Pharmacotherapy, Department of Clinical Dietetics and Human Nutrition, School of Pharmacy, Faculty of Pharmacy and Pharmaceutical Sciences, Josai University1-1 Keyakidai, Sakado, Saitama 350-0295, Japan
| | - Bo Yuan
- Laboratory of Pharmacology, School of Pharmacy, Faculty of Pharmacy and Pharmaceutical Sciences, Josai University1-1 Keyakidai, Sakado, Saitama 350-0295, Japan
| | - Xiaomei Hu
- Xiyuan Hospital, China Academy of Chinese Medical SciencesBeijing 100091, People’s Republic of China
| | - Mari Okazaki
- Laboratory of Pharmacology, School of Pharmacy, Faculty of Pharmacy and Pharmaceutical Sciences, Josai University1-1 Keyakidai, Sakado, Saitama 350-0295, Japan
| |
Collapse
|