1
|
Chawapun P, Khamto N, Utama K, Siriphong S, Dechsupa N, Kantapan J, Meerak J, Meepowpan P, Sangthong P. Synthesis and biological evaluation of imidazolium conjugated with dimethylcardamonin (DMC) as a novel potential agent against MDA-MB-231 triple-negative breast cancer cells. Biomed Pharmacother 2024; 178:117249. [PMID: 39111077 DOI: 10.1016/j.biopha.2024.117249] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Revised: 07/29/2024] [Accepted: 07/30/2024] [Indexed: 08/25/2024] Open
Abstract
A new imidazolium ionic liquid (IL) halide conjugated with dimethylcardamonin (DMC, 1), namely [Bbim]Br-DMC (3), was synthesised to improve the biological activity of the natural chalcone. DMC was isolated from seeds of Syzygium nervosum A. Cunn. ex DC. which was an effective anti-breast cancer agent. The compound 1 and 3 showed anticancer activity in MDA-MB-231 cells with IC50 values of 14.54 ± 0.99 μM and 7.40 ± 0.15 μM, respectively. MTT assay showed that compound 3 had cytotoxic effect at least two-fold greater than compound 1 but was low toxic to normal cells of Hs 578Bst. After 48 h, compound 3 at concentration of IC50 value inhibited the proliferation and induced morphological changes of MDA-MB-231 cells in a time-dependent manner. The cell cycle profile also showed that compound 3 exerted anti-proliferation activity with the cell cycle arrest at G0/G1 phase and compound 3 also induced apoptosis and reduced mitochondrial membrane potential in MDA-MB-231 cells in a dose-dependent manner. In gene expression assay, compound 3 up-regulated pro-apoptotic genes such as Bax and p53 and suppressed anti-apoptotic Bcl-2 whereas there was no effect on DNA repair gene such as PARP1. The Bax/Bcl-2 ratio was significantly increased after treated with compound 3. In the molecular docking study, the interactions between compound 3 and B-DNA structure in the minor groove region via hydrogen bonds was reported. In conclusion, [Bbim]Br-DMC or compound 3 is a potential candidate to induce apoptosis and inhibits proliferation via cell cycle arrest and decreases mitochondrial membrane of triple-negative breast cancer MDA-MB-231 cells.
Collapse
Affiliation(s)
- Pornthip Chawapun
- Program in Biotechnology, Multidisciplinary and Interdisciplinary School, Chiang Mai University, Chiang Mai 50200, Thailand; Department of Chemistry, Faculty of Science, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Nopawit Khamto
- Department of Chemistry, Faculty of Science, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Kraikrit Utama
- Department of Chemistry, Faculty of Science, Chiang Mai University, Chiang Mai 50200, Thailand; Office of Research Administration, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Sadanon Siriphong
- Program in Biotechnology, Multidisciplinary and Interdisciplinary School, Chiang Mai University, Chiang Mai 50200, Thailand; Department of Chemistry, Faculty of Science, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Nathupakorn Dechsupa
- Molecular Imaging and Therapy Research Unit, Department of Radiologic Technology, Faculty of Associated Medical Sciences, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Jiraporn Kantapan
- Molecular Imaging and Therapy Research Unit, Department of Radiologic Technology, Faculty of Associated Medical Sciences, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Jomkhwan Meerak
- Department of Biology, Faculty of Science, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Puttinan Meepowpan
- Department of Chemistry, Faculty of Science, Chiang Mai University, Chiang Mai 50200, Thailand; Center of Excellence in Materials Science and Technology, Faculty of Science, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Padchanee Sangthong
- Department of Chemistry, Faculty of Science, Chiang Mai University, Chiang Mai 50200, Thailand; Division of Biochemistry and Biochemical Innovation, Department of Chemistry, Faculty of Science, Chiang Mai University, Chiang Mai 50200, Thailand.
| |
Collapse
|
2
|
Wehbe N, Badran A, Baydoun S, Al-Sawalmih A, Maresca M, Baydoun E, Mesmar JE. The Antioxidant Potential and Anticancer Activity of Halodule uninervis Ethanolic Extract against Triple-Negative Breast Cancer Cells. Antioxidants (Basel) 2024; 13:726. [PMID: 38929164 PMCID: PMC11200955 DOI: 10.3390/antiox13060726] [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: 04/28/2024] [Revised: 06/07/2024] [Accepted: 06/09/2024] [Indexed: 06/28/2024] Open
Abstract
Natural remedies have been indispensable to traditional medicine practices for generations, offering therapeutic solutions for various ailments. In modern times, these natural products continue to play a pivotal role in the discovery of new drugs, especially for cancer treatment. The marine ecosystem offers a wide range of plants with potential anticancer activities due to their distinct biochemical diversity and adaptation to extreme situations. The seagrass Halodule uninervis is rich in diverse bioactive metabolites that bestow the plant with various pharmacological properties. However, its anticancer activity against invasive triple-negative breast cancer (TNBC) is still poorly investigated. In the present study, the phytochemical composition of an ethanolic extract of H. uninervis (HUE) was screened, and its antioxidant potential was evaluated. Moreover, the anticancer potential of HUE against MDA-MB-231 cells was investigated along with the possible underlying mechanisms of action. Our results showed that HUE is rich in diverse phytochemicals that are known for their antioxidant and anticancer effects. In MDA-MB-231 cells, HUE targeted the hallmarks of cancer, including cell proliferation, adhesion, migration, invasion, and angiogenesis. The HUE-mediated anti-proliferative and anti-metastatic effects were associated with the downregulation of the proto-oncogenic STAT3 signaling pathway. Taken together, H. uninervis could serve as a valuable source for developing novel drugs targeting TNBC.
Collapse
Affiliation(s)
- Nadine Wehbe
- Department of Biology, Faculty of Arts and Sciences, American University of Beirut, Riad El Solh, Beirut 1107 2020, Lebanon; (N.W.); (E.B.)
| | - Adnan Badran
- Department of Nutrition, Faculty of Pharmacy and Medical Sciences, University of Petra, Amman 11196, Jordan;
| | - Serine Baydoun
- Breast Imaging Section, Imaging Institute, Cleveland Clinic Foundation, Cleveland, OH 44195, USA;
| | - Ali Al-Sawalmih
- Marine Science Station, University of Jordan, Aqaba 11942, Jordan;
| | - Marc Maresca
- Aix-Marseille Univ, CNRS, Centrale Marseille, iSM2, 13013 Marseille, France
| | - Elias Baydoun
- Department of Biology, Faculty of Arts and Sciences, American University of Beirut, Riad El Solh, Beirut 1107 2020, Lebanon; (N.W.); (E.B.)
| | - Joelle Edward Mesmar
- Department of Biology, Faculty of Arts and Sciences, American University of Beirut, Riad El Solh, Beirut 1107 2020, Lebanon; (N.W.); (E.B.)
| |
Collapse
|
3
|
Soni D, Anjum Z, Raza K, Verma S. A Review on Picrosides Targeting NFκB and its Proteins for Treatment of Breast Cancer. Cell Biochem Biophys 2024; 82:575-591. [PMID: 38724755 DOI: 10.1007/s12013-024-01281-1] [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] [Accepted: 04/17/2024] [Indexed: 08/25/2024]
Abstract
Breast cancer is the most frequently diagnosed disease causing most deaths in women worldwide. Chemotherapy and neo-adjuvant therapy are the standard method of treatment in early stages of breast cancer. However drug resistance in breast cancer limit the use of these methods for treatment. Research focus is now shifted towards identifying natural phytochemicals with lower toxicity. This review illustrates the NF κB interaction with different signaling pathways in normal condition, breast cancer and other cancer and thus represent a potential target for treatment. No reports are available on the action of picrosides on NFκB and its associated proteins for anticancer activity. In the present review, potential interaction of picrosides with NF-κB and its associated proteins is reviewed for anticancer action. Further, an important facet of this review entails the ADMET analysis of Picroside, elucidating key ADMET properties which serves to underscore the crucial characteristics of Picroside as a potential drug for treating breast cancer. Furthermore, in silico analysis of Picrosides was executed in order to get potential binding modes between ligand (Picrosides II) and NFκB.
Collapse
Affiliation(s)
- Deepika Soni
- National Institute of Pathology, Safdarjung Hospital Campus, New Delhi, India
| | - Zubina Anjum
- Department of Bioinformatics, Central University of South Bihar, Gaya, Bihar, India
| | - Khalid Raza
- Department of Computer Science, Jamia Millia Islamia, New Delhi, India
| | - Saurabh Verma
- Indian Council of Medical Research, HRD Division, V. Ramalingaswami Bhawan, Ansari Nagar, New Delhi, India.
| |
Collapse
|
4
|
Oh HM, Cho CK, Lee NH, Son CG. Experimental evidence for anti-metastatic actions of apigenin: a mini review. Front Oncol 2024; 14:1380194. [PMID: 38515580 PMCID: PMC10954815 DOI: 10.3389/fonc.2024.1380194] [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: 02/01/2024] [Accepted: 02/20/2024] [Indexed: 03/23/2024] Open
Abstract
Cancer metastasis is responsible for the majority of cancer-related deaths. Accordingly, to reduce metastasis remains a vital challenge in clinical practice, and phytochemicals have taken an attention as anti-metastatic agents. Apigenin, a plant flavone, showed anti-cancer effects against in various animal models, moreover its potentials inhibiting tumor metastasis have been reported. Herein, we analyzed the overall features at what apigenin inhibited metastasis and its action modes. We searched for articles in MEDLINE (Pubmed), EMBASE and Cochrane Central Register of Controlled Trials (CENTRAL) through March 2023. Total 6 animal studies presented anti-metastatic effects of apigenin using 5 difference experimental models, while the mechanisms involved modulations of epithelial-mesenchymal transition (EMT), matrix metalloproteinases (MMPs), angiogenesis, and various metastasis-related signaling pathways. This review provides an overall potential of apigenin as a candidate reducing the risk of cancer metastasis.
Collapse
Affiliation(s)
- Hyeon-Muk Oh
- College of Korean Medicine, Daejeon University, Daejeon, Republic of Korea
- East-West Cancer Center, Daejeon Korean Medicine Hospital of Daejeon University, Daejeon, Republic of Korea
| | - Chong-Kwan Cho
- College of Korean Medicine, Daejeon University, Daejeon, Republic of Korea
- East-West Cancer Center, Daejeon Korean Medicine Hospital of Daejeon University, Daejeon, Republic of Korea
| | - Nam-Hun Lee
- College of Korean Medicine, Daejeon University, Daejeon, Republic of Korea
- East-West Cancer Center, Cheonan Korean Medicine Hospital of Daejeon University, Daejeon, Republic of Korea
| | - Chang-Gue Son
- College of Korean Medicine, Daejeon University, Daejeon, Republic of Korea
- East-West Cancer Center, Daejeon Korean Medicine Hospital of Daejeon University, Daejeon, Republic of Korea
| |
Collapse
|
5
|
da Silva FC, Brandão DC, Ferreira EA, Siqueira RP, Ferreira HSV, Da Silva Filho AA, Araújo TG. Tailoring Potential Natural Compounds for the Treatment of Luminal Breast Cancer. Pharmaceuticals (Basel) 2023; 16:1466. [PMID: 37895937 PMCID: PMC10610388 DOI: 10.3390/ph16101466] [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: 08/29/2023] [Revised: 09/24/2023] [Accepted: 09/29/2023] [Indexed: 10/29/2023] Open
Abstract
Breast cancer (BC) is the most diagnosed cancer worldwide, mainly affecting the epithelial cells from the mammary glands. When it expresses the estrogen receptor (ER), the tumor is called luminal BC, which is eligible for endocrine therapy with hormone signaling blockade. Hormone therapy is essential for the survival of patients, but therapeutic resistance has been shown to be worrying, significantly compromising the prognosis. In this context, the need to explore new compounds emerges, especially compounds of plant origin, since they are biologically active and particularly promising. Natural products are being continuously screened for treating cancer due to their chemical diversity, reduced toxicity, lower side effects, and low price. This review summarizes natural compounds for the treatment of luminal BC, emphasizing the activities of these compounds in ER-positive cells. Moreover, their potential as an alternative to endocrine resistance is explored, opening new opportunities for the design of optimized therapies.
Collapse
Affiliation(s)
- Fernanda Cardoso da Silva
- Laboratory of Genetics and Biotechnology, Institute of Biotechnology, Universidade Federal de Uberlândia, Patos de Minas 38700-002, MG, Brazil; (F.C.d.S.); (D.C.B.); (R.P.S.); (H.S.V.F.)
| | - Douglas Cardoso Brandão
- Laboratory of Genetics and Biotechnology, Institute of Biotechnology, Universidade Federal de Uberlândia, Patos de Minas 38700-002, MG, Brazil; (F.C.d.S.); (D.C.B.); (R.P.S.); (H.S.V.F.)
| | - Everton Allan Ferreira
- Department of Pharmaceutical Sciences, Faculty of Pharmacy, Federal University of Juiz de Fora, Juiz de Fora 36036-900, MG, Brazil; (E.A.F.); (A.A.D.S.F.)
| | - Raoni Pais Siqueira
- Laboratory of Genetics and Biotechnology, Institute of Biotechnology, Universidade Federal de Uberlândia, Patos de Minas 38700-002, MG, Brazil; (F.C.d.S.); (D.C.B.); (R.P.S.); (H.S.V.F.)
| | - Helen Soares Valença Ferreira
- Laboratory of Genetics and Biotechnology, Institute of Biotechnology, Universidade Federal de Uberlândia, Patos de Minas 38700-002, MG, Brazil; (F.C.d.S.); (D.C.B.); (R.P.S.); (H.S.V.F.)
| | - Ademar Alves Da Silva Filho
- Department of Pharmaceutical Sciences, Faculty of Pharmacy, Federal University of Juiz de Fora, Juiz de Fora 36036-900, MG, Brazil; (E.A.F.); (A.A.D.S.F.)
| | - Thaise Gonçalves Araújo
- Laboratory of Genetics and Biotechnology, Institute of Biotechnology, Universidade Federal de Uberlândia, Patos de Minas 38700-002, MG, Brazil; (F.C.d.S.); (D.C.B.); (R.P.S.); (H.S.V.F.)
- Laboratory of Nanobiotechnology Prof. Dr. Luiz Ricardo Goulart Filho, Institute of Biotechnology, Universidade Federal de Uberlândia, Uberlandia 38405-302, MG, Brazil
| |
Collapse
|
6
|
Farghadani R, Naidu R. The anticancer mechanism of action of selected polyphenols in triple-negative breast cancer (TNBC). Biomed Pharmacother 2023; 165:115170. [PMID: 37481930 DOI: 10.1016/j.biopha.2023.115170] [Citation(s) in RCA: 20] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2023] [Revised: 07/10/2023] [Accepted: 07/12/2023] [Indexed: 07/25/2023] Open
Abstract
Breast cancer is a leadingcause of cancer-related deaths in women globally, with triple-negative breast cancer (TNBC) being an aggressive subtype that lacks targeted therapies and is associated with a poor prognosis. Polyphenols, naturally occurring compounds in plants, have been investigated as a potential therapeutic strategy for TNBC. This review provides an overview of the anticancer effects of polyphenols in TNBC and their mechanisms of action. Several polyphenols, including resveratrol, quercetin, kaempferol, genistein, epigallocatechin-3-gallate, apigenin, fisetin, hesperetin and luteolin, have been shown to inhibit TNBC cell proliferation, induce cell cycle arrest, promote apoptosis, and suppress migration/invasion in preclinical models. The molecular mechanisms underlying their anticancer effects involve the modulation of several signalling pathways, such as PI3K/Akt, MAPK, STATT, and NF-κB pathways. Polyphenols also exhibit synergistic effects with chemotherapy drugs, making them promising candidates for combination therapy. The review also highlights clinical trials investigating the potential use of polyphenols, individually or in combination therapy, against breast cancer. This review deepens the under-standing of the mechanism of action of respective polyphenols and provides valuable insights into the potential use of polyphenols as a therapeutic strategy for TNBC, and lays the groundwork for future research in this area.
Collapse
Affiliation(s)
- Reyhaneh Farghadani
- Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Jalan Lagoon Selatan, Bandar Sunway, Selangor Darul Ehsan 47500, Malaysia.
| | - Rakesh Naidu
- Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Jalan Lagoon Selatan, Bandar Sunway, Selangor Darul Ehsan 47500, Malaysia.
| |
Collapse
|
7
|
Bi C, Han W, Yu J, Zhang H, Xing G, Liu Z. Insights into the pharmacological and therapeutic effects of apigenin in liver injuries and diseases. Heliyon 2023; 9:e15609. [PMID: 37144193 PMCID: PMC10151377 DOI: 10.1016/j.heliyon.2023.e15609] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2023] [Revised: 04/12/2023] [Accepted: 04/17/2023] [Indexed: 05/06/2023] Open
Abstract
Background Liver diseases are a spectrum of diseases that include hepatic steatosis, nonalcoholic fatty liver disease, hepatitis, liver fibrosis, cirrhosis, and hepatic cancer. These diseases not only severely decrease the quality of life for patients, but also cause financial burden. Although apigenin (APG) has recently become the primary treatment for liver injuries and diseases (LIADs), there has been no systematic review of its use. Purpose To review the existing literature and put forward novel strategies for future APG research on LIADs. Methods A search was conducted in PubMed, Science Direct, Research Gate, Web of Science, VIP, Wanfang, and CNKI, and 809 articles were obtained. After applying inclusion and exclusion criteria, 135 articles were included. Results APG is promising in treating LIADs via various mechanisms arising from its anti-inflammation, anti-proliferation, anti-infection, anti-oxidation, and anti-cancer properties. Conclusion This review summarizes the evidence supporting the use of APG as a treatment for LIADs and provides an insight into the intestinal microbiota, which may have important implications in its future clinical use.
Collapse
Affiliation(s)
- Chenchen Bi
- Department of Pharmacology, School of Medicine, Shaoxing University, Shaoxing, Zhejiang, P. R. China
- Department of Clinical Medicine, School of Medicine, Shaoxing University, Shaoxing, Zhejiang, P. R. China
| | - Wenwen Han
- Department of Clinical Medicine, School of Medicine, Shaoxing University, Shaoxing, Zhejiang, P. R. China
| | - Jingru Yu
- Department of Clinical Medicine, School of Medicine, Shaoxing University, Shaoxing, Zhejiang, P. R. China
| | - Huafang Zhang
- Department of Pharmacology, School of Medicine, Shaoxing University, Shaoxing, Zhejiang, P. R. China
| | - Guiying Xing
- Department of Pharmacology, School of Medicine, Shaoxing University, Shaoxing, Zhejiang, P. R. China
- Corresponding author.
| | - Zheng Liu
- Department of Pharmacology, School of Medicine, Shaoxing University, Shaoxing, Zhejiang, P. R. China
- Corresponding author.
| |
Collapse
|
8
|
Wen SY, Wei BY, Ma JQ, Wang L, Chen YY. Phytochemicals, Biological Activities, Molecular Mechanisms, and Future Prospects of Plantago asiatica L. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2023; 71:143-173. [PMID: 36545763 DOI: 10.1021/acs.jafc.2c07735] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/17/2023]
Abstract
Plantago asiatica L. has been used as a vegetable and nutritious food in Asia for thousands of years. According to recent phytochemical and pharmacological research, the active compositions of the plant contribute to various health benefits, such as antioxidant, anti-inflammatory, antibacterial, antiviral, and anticancer. This article reviews the 87 components of the plant and their structures, as well as their biological activities and molecular research progress, in detail. This review provides valuable reference material for further study, production, and application of P. asiatica, as well as its components in functional foods and therapeutic agents.
Collapse
Affiliation(s)
- Shi-Yuan Wen
- College of Basic Medical Sciences, Shanxi Medical University, Taiyuan 030000, China
| | - Bing-Yan Wei
- College of Basic Medical Sciences, Shanxi Medical University, Taiyuan 030000, China
| | - Jie-Qiong Ma
- College of Basic Medical Sciences, Shanxi Medical University, Taiyuan 030000, China
| | - Li Wang
- College of Basic Medical Sciences, Shanxi Medical University, Taiyuan 030000, China
| | - Yan-Yan Chen
- School of Medicine, Jiangsu University, Zhenjiang 212013, China
| |
Collapse
|
9
|
Ye Y, Ma Y, Kong M, Wang Z, Sun K, Li F. Effects of Dietary Phytochemicals on DNA Damage in Cancer Cells. Nutr Cancer 2023; 75:761-775. [PMID: 36562548 DOI: 10.1080/01635581.2022.2157024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
With the increasing incidence of cancer worldwide, the prevention and treatment of cancer have garnered considerable scientific attention. Traditional chemotherapeutic drugs are highly toxic and associated with substantial side effects; therefore, there is an urgent need for developing new therapeutic agents. Dietary phytochemicals are important in tumor prevention and treatment because of their low toxicity and side effects at low concentrations; however, their exact mechanisms of action remain obscure. DNA damage is mainly caused by physical or chemical factors in the environment, such as ultraviolet light, alkylating agents and reactive oxygen species that cause changes in the DNA structure of cells. Several phytochemicals have been shown inhibit the occurrence and development of tumors by inducing DNA damage. This article reviews the advances in phytochemical research; particularly regarding the mechanisms related to DNA damage and provide a theoretical basis for future chemoprophylaxis research.
Collapse
Affiliation(s)
- Yang Ye
- Department of Preventive Medicine and Public Health Laboratory Science, School of Medicine, Jiangsu University, Zhenjiang, China
| | - Ying Ma
- Department of Preventive Medicine and Public Health Laboratory Science, School of Medicine, Jiangsu University, Zhenjiang, China
| | - Mei Kong
- Department of Gastroenterology, Affiliated Hospital of Jiangsu University, Zhenjiang, China
| | - Zhihua Wang
- Department of Gastroenterology, Affiliated Hospital of Jiangsu University, Zhenjiang, China
| | - Kang Sun
- Department of Gastrointestinal Surgery, Affiliated Hospital of Jiangsu University, Zhenjiang, China
| | - Fang Li
- Department of Preventive Medicine and Public Health Laboratory Science, School of Medicine, Jiangsu University, Zhenjiang, China
| |
Collapse
|
10
|
Ngernyuang N, Wongwattanakul M, Charusirisawad W, Shao R, Limpaiboon T. Green synthesized apigenin conjugated gold nanoparticles inhibit cholangiocarcinoma cell activity and endothelial cell angiogenesis in vitro. Heliyon 2022; 8:e12028. [PMID: 36506385 PMCID: PMC9732323 DOI: 10.1016/j.heliyon.2022.e12028] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2022] [Revised: 11/05/2022] [Accepted: 11/25/2022] [Indexed: 12/02/2022] Open
Abstract
Cholangiocarcinoma (CCA) is a rare malignancy of the biliary tract with extremely poor clinical outcomes due to a lack of effective therapies to improve disease management. The emerging green synthesis of gold nanoparticles (AuNPs) has extensively provided their use in biomedical applications. In this study, we developed AuNPs via reducing gold salts with apigenin (4',5,7-trihydroxyflavone). The synthesized apigenin-conjugated AuNPs (api-AuNPs) were physicochemically characterized by various techniques before evaluation their biological and functional inhibition in a CCA cell line, KKU-M055. The mean size of api-AuNPs was 90.34 ± 22.82 nm with zeta potential of -36 ± 0.55. The half-maximal inhibitory concentration (IC50, 0.8 mg/mL) of api-AuNPs on cell proliferation of KKU-M055 was 1.9-fold less than that of an immortalized human cholangiocyte cell line, MMNK1 (IC50, 1.5 mg/mL). Moreover, api-AuNPs induced cell apoptosis via the up-regulation of Bax, Bid, and Caspase 3, and down-regulation of Bcl2, leading to elevated caspase 3/7, 8, 9 activities and reactive oxygen species (ROS) production. The api-AuNPs significantly inhibited the migration of KKU-M055 cells and suppressed the proliferation, migration, and in vitro tube formation of vascular endothelial cells. Collectively, our findings indicate the dual abilities of api-AuNPs that potentially inhibit cancer cell growth and motility as well as endothelial cell-mediated angiogenesis, which may offer a novel therapeutic avenue to treat CCA patients effectively.
Collapse
Affiliation(s)
- Nipaporn Ngernyuang
- Chulabhorn International College of Medicine, Thammasat University, Pathum Thani 12120, Thailand
| | - Molin Wongwattanakul
- Centre for Research and Development of Medical Diagnostic Laboratories, Faculty of Associated Medical Science, Khon Kaen University, Khon Kaen 40002, Thailand
| | - Wannit Charusirisawad
- Chulabhorn International College of Medicine, Thammasat University, Pathum Thani 12120, Thailand
| | - Rong Shao
- Development of Pharmacology, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Temduang Limpaiboon
- Centre for Research and Development of Medical Diagnostic Laboratories, Faculty of Associated Medical Science, Khon Kaen University, Khon Kaen 40002, Thailand
| |
Collapse
|
11
|
Talib WH, Abuawad A, Thiab S, Alshweiat A, Mahmod AI. Flavonoid-based nanomedicines to target tumor microenvironment. OPENNANO 2022. [DOI: 10.1016/j.onano.2022.100081] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
12
|
Nipun VB, Amin KA. Recent Advances in Protein Kinase CK2, a Potential Therapeutic Target in Cancer. RUSSIAN JOURNAL OF BIOORGANIC CHEMISTRY 2022; 48:919-931. [DOI: 10.1134/s1068162022050144] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Affiliation(s)
- V. B. Nipun
- Cancer Research Center, Shantou University Medical Collage, Shantou, Guangdong, 515041, PR China
- Department of Chemistry, Faculty of Science, University of Imam Abdulrahman Bin Faisal university, P.O. Box 1982, Dammam, 31441, Saudi Arabia
| | - K. A. Amin
- Department of Chemistry, Faculty of Science, University of Imam Abdulrahman Bin Faisal university, P.O. Box 1982, Dammam, 31441, Saudi Arabia
- Basic and Applied Scientific Research Center, Imam Abdulrahman Bin Faisal university, P.O. Box 1982, Dammam, 31441, Saudi Arabia
| |
Collapse
|
13
|
Park MY, Kim Y, Ha SE, Kim HH, Bhosale PB, Abusaliya A, Jeong SH, Kim GS. Function and Application of Flavonoids in the Breast Cancer. Int J Mol Sci 2022; 23:7732. [PMID: 35887080 PMCID: PMC9323071 DOI: 10.3390/ijms23147732] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Revised: 06/23/2022] [Accepted: 07/09/2022] [Indexed: 11/28/2022] Open
Abstract
Breast cancer is one of the top causes of death, particularly among women, and it affects many women. Cancer can also be caused by various factors, including acquiring genetic alteration. Doctors use radiation to detect and treat breast cancer. As a result, breast cancer becomes radiation-resistant, necessitating a new strategy for its treatment. The approach discovered by the researchers is a flavonoid, which is being researched to see if it might help treat radiation-resistant breast cancer more safely than an approved medicine already being used in the field. As a result, this study focuses on the role of flavonoids in breast cancer suppression, breast cancer gene anomalies, and the resulting apoptotic mechanism.
Collapse
Affiliation(s)
- Min Yeong Park
- Research Institute of Life Science, College of Veterinary Medicine, Gyeongsang National University, Gazwa, Jinju 52828, Korea; (M.Y.P.); (S.E.H.); (H.H.K.); (P.B.B.); (A.A.); (S.H.J.)
| | - Yoonjung Kim
- College of Nursing, Konyang University Medical Campus, 158, Gwanjeodong-ro, Seo-gu, Daejeon 35365, Korea;
| | - Sang Eun Ha
- Research Institute of Life Science, College of Veterinary Medicine, Gyeongsang National University, Gazwa, Jinju 52828, Korea; (M.Y.P.); (S.E.H.); (H.H.K.); (P.B.B.); (A.A.); (S.H.J.)
- Biological Resources Research Group, Gyeongnam Department of Environment Toxicology and Chemistry, Korea Institute of Toxicology, 17 Jegok-gil, Jinju 52834, Korea
| | - Hun Hwan Kim
- Research Institute of Life Science, College of Veterinary Medicine, Gyeongsang National University, Gazwa, Jinju 52828, Korea; (M.Y.P.); (S.E.H.); (H.H.K.); (P.B.B.); (A.A.); (S.H.J.)
| | - Pritam Bhangwan Bhosale
- Research Institute of Life Science, College of Veterinary Medicine, Gyeongsang National University, Gazwa, Jinju 52828, Korea; (M.Y.P.); (S.E.H.); (H.H.K.); (P.B.B.); (A.A.); (S.H.J.)
| | - Abuyaseer Abusaliya
- Research Institute of Life Science, College of Veterinary Medicine, Gyeongsang National University, Gazwa, Jinju 52828, Korea; (M.Y.P.); (S.E.H.); (H.H.K.); (P.B.B.); (A.A.); (S.H.J.)
| | - Se Hyo Jeong
- Research Institute of Life Science, College of Veterinary Medicine, Gyeongsang National University, Gazwa, Jinju 52828, Korea; (M.Y.P.); (S.E.H.); (H.H.K.); (P.B.B.); (A.A.); (S.H.J.)
| | - Gon Sup Kim
- Research Institute of Life Science, College of Veterinary Medicine, Gyeongsang National University, Gazwa, Jinju 52828, Korea; (M.Y.P.); (S.E.H.); (H.H.K.); (P.B.B.); (A.A.); (S.H.J.)
| |
Collapse
|
14
|
Underlying Anticancer Mechanisms and Synergistic Combinations of Phytochemicals with Cancer Chemotherapeutics: Potential Benefits and Risks. J FOOD QUALITY 2022. [DOI: 10.1155/2022/1189034] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Cancer therapies are associated with various challenges including the emergence of multidrug resistant tumors, toxicological issues, severe side effects, and economic burden. To counteract these effects, natural products as substitutes and adjuvant therapies have received considerable attention owing to their safety, efficacy, and economic aspects. Various preclinical and clinical studies revealed that natural products and their combinations with chemotherapeutics mediate their anticancer effects via modulation of various signaling pathways implicated in promoting apoptosis, inhibiting excessive cellular proliferation, and mobilizing the immune system. Several lead phytochemicals including curcumin, resveratrol, quercetin, and cannabinoids synergistically act with cancer chemotherapeutics reducing cell proliferation and inducing apoptosis and cell cycle arrest. However, clinical studies on the subject matter are limited and need further extensive studies. It has been observed that patients undergoing chemotherapy use alternative therapies to ameliorate the symptoms associated with the use of chemotherapeutic agents. Nevertheless, some of the patients inform their physicians regarding herbal medicine during chemotherapy while others do not, and even most of the patients do not know the composition of herbal medicine they consume during chemotherapy. Herbal interactions with chemotherapeutics are associated with both beneficial and harmful aspects, but the beneficial aspect overweighs the harmful ones in terms of controlling the symptoms associated with the chemotherapy. Nonetheless, a large number of herbal medicines have been demonstrated to have synergistic effect with chemotherapy and alleviate the side effects of chemotherapeutic agents. The concomitant use of the majority of herbal medicines with chemotherapy has been demonstrated to be beneficial in multiple malignant tumors like cancer of blood, lungs, kidneys, liver, skin, and gastrointestinal tract. However, herbal medicines which possess positive interaction and improve the quality of life of patients should be sorted out and integrated with the chemotherapy. There should be a quality control system for the appraisal of herbal medicine, and there should also be an appropriate system of patient-doctor communication to counsel the patients regarding the beneficial and deleterious effects of the herbal medicine in combination with chemotherapy.
Collapse
|
15
|
Sioud F, Dhouafi Z, Lahmar A, Elgueder D, Chekir-Ghedira L. A Novel Anticancer Effect of Ephedra alata Decne in Breast Cancer Cells. Nutr Cancer 2022; 74:3403-3412. [PMID: 35545961 DOI: 10.1080/01635581.2022.2072907] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Cancer is a class of diseases characterized by uncontrolled cell growth. One of the main aims of developing new therapies is to use natural resources to induce apoptosis. LC-ms/ms analysis of a methanolic extract of Ephedra alata (E.A.) allowed the identification of 20 secondary metabolites, including flavonoids, phenolic acids, and proanthocyanidins. Antiproliferative effect was assessed by crystal violet assay. Antimigration effect was tested by wound healing assay and apoptosis induction was determined by annexin binding assays, Hoechst staining, ROS production, and activation of apoptotic proteins. The results indicated that exposure of breast cancer cells to E.A. extract significantly reduced cell viability in a dose and time-dependent manner and inhibited the migration of 4T1 cells at a low dose. Moreover, treatment of cells with E.A. extract induced apoptosis, as it was detected by Annexin V/7 AAD, Hoechst staining, ROS production, and the activation of caspases.
Collapse
Affiliation(s)
- Fairouz Sioud
- Research Unit Bioactive Natural Products and Biotechnology UR17ES49, Faculty of Dental Medicine of Monastir, University of Monastir, Monastir, Tunisia.,Laboratory of Pharmacognosy, Faculty of Pharmacy, University of Monastir, Monastir, Tunisia
| | - Zaineb Dhouafi
- Research Unit Bioactive Natural Products and Biotechnology UR17ES49, Faculty of Dental Medicine of Monastir, University of Monastir, Monastir, Tunisia
| | - Aida Lahmar
- Research Unit Bioactive Natural Products and Biotechnology UR17ES49, Faculty of Dental Medicine of Monastir, University of Monastir, Monastir, Tunisia.,Laboratory of Pharmacognosy, Faculty of Pharmacy, University of Monastir, Monastir, Tunisia
| | - Dorra Elgueder
- Research Unit Bioactive Natural Products and Biotechnology UR17ES49, Faculty of Dental Medicine of Monastir, University of Monastir, Monastir, Tunisia
| | - Leila Chekir-Ghedira
- Research Unit Bioactive Natural Products and Biotechnology UR17ES49, Faculty of Dental Medicine of Monastir, University of Monastir, Monastir, Tunisia.,Laboratory of Pharmacognosy, Faculty of Pharmacy, University of Monastir, Monastir, Tunisia
| |
Collapse
|
16
|
Moral R, Escrich E. Influence of Olive Oil and Its Components on Breast Cancer: Molecular Mechanisms. MOLECULES (BASEL, SWITZERLAND) 2022; 27:molecules27020477. [PMID: 35056792 PMCID: PMC8780060 DOI: 10.3390/molecules27020477] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/24/2021] [Revised: 12/31/2021] [Accepted: 01/08/2022] [Indexed: 02/06/2023]
Abstract
Breast cancer is the most frequent malignant neoplasia and a leading cause of mortality in women worldwide. The Mediterranean diet has been proposed as a healthy dietary pattern with protective effects in several chronic diseases, including breast cancer. This diet is characterized by the consumption of abundant plant foods and olive oil as the principal source of fat, which is considered one of the main components with potential antioxidant, anti-inflammatory and anticancer effects. Extra-virgin olive oil (EVOO) has several bioactive compounds, mainly including monounsaturated fatty acids, triterpenes and polyphenols, such as phenolic alcohols (e.g., hydroxytyrosol), secoiridoids (e.g., oleuropein and oleocanthal), lignans (e.g., pinoresinol) or flavonoids (e.g., luteolin). While epidemiological evidence is still limited, experimental in vivo and in vitro data have shown a protective effect of this oil and its compounds on mammary carcinogenesis. Such effects account through complex and multiple mechanisms, including changes in epigenetics, transcriptome and protein expression that modulate several signaling pathways. Molecular targets of EVOO compounds have a role in the acquisition of cancer hallmarks. Although further research is needed to elucidate their beneficial effects on human prevention and progression of the disease, evidence points to EVOO in the context of the Mediterranean diet as a heathy choice, while EVOO components may be promising adjuvants in anticancer strategies.
Collapse
|
17
|
Oyenihi OR, Oyenihi AB, Alabi TD, Tade OG, Adeyanju AA, Oguntibeju OO. Reactive oxygen species: Key players in the anticancer effects of apigenin? J Food Biochem 2022; 46:e14060. [PMID: 34997605 DOI: 10.1111/jfbc.14060] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2021] [Revised: 12/07/2021] [Accepted: 12/10/2021] [Indexed: 01/04/2023]
Abstract
Reactive oxygen species (ROS) exhibit a double-edged sword in cancer-hence their modulation has been an attractive strategy in cancer prevention and therapy. The abundance of scientific information on the pro-oxidant effects of apigenin in cancer cells suggests the crucial role of ROS in its mechanisms of action. Although apigenin is known to enhance the cellular ROS levels to cytotoxic degrees in cancer cells in vitro, it remains to be determined if these pro-oxidant effects prevail or are relevant in experimental tumor models and clinical trials. Here, we critically examine the pro-oxidant and antioxidant effects of apigenin in cancer to provide insightful perspectives on the association between its ROS-modulating action and anticancer potential. We also discussed these effects in a cell/tissue type-specific context to highlight the factors influencing the switch between antioxidant and pro-oxidant effects. Finally, we raised some questions that need addressing for the potential translation of these studies into clinical applications. Further research into this duality in oxidant actions of apigenin, especially in vivo, may enable better exploitation of its anticancer potential. PRACTICAL APPLICATION: Apigenin is a naturally occurring compound found in chamomile flowers, parsley, celery, peppermint, and citrus fruits. Many human trials of dietary interventions with apigenin-containing herbs and flavonoid mixture on oxidative stress markers, for instance, point to their antioxidant effects and health benefits in many diseases. Preclinical studies suggest that apigenin alone or its combination with chemotherapeutics has a strong anti-neoplastic effect and can induce ROS-mediated cytotoxicity at concentrations in the micromolar (μM) range, which may not be feasible with dietary interventions. Enhancing the in vivo pharmacokinetic properties of apigenin may be indispensable for its potential cancer-specific pro-oxidant therapy and may provide relevant information for clinical studies of apigenin either as a single agent or an adjuvant to chemotherapeutics.
Collapse
Affiliation(s)
- Omolola R Oyenihi
- Phytomedicine and Phytochemistry Group, Department of Biomedical Sciences, Faculty of Health and Wellness Sciences, Cape Peninsula University of Technology, Bellville, South Africa
| | - Ayodeji B Oyenihi
- Functional Foods Research Unit, Faculty of Applied Sciences, Cape Peninsula University of Technology, Bellville, South Africa
| | - Toyin D Alabi
- Phytomedicine and Phytochemistry Group, Department of Biomedical Sciences, Faculty of Health and Wellness Sciences, Cape Peninsula University of Technology, Bellville, South Africa
| | - Oluwatosin G Tade
- School of Physiology, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Anne A Adeyanju
- Department of Biological Sciences, Faculty of Applied Sciences, KolaDaisi University, Ibadan, Oyo State, Nigeria
| | - Oluwafemi O Oguntibeju
- Phytomedicine and Phytochemistry Group, Department of Biomedical Sciences, Faculty of Health and Wellness Sciences, Cape Peninsula University of Technology, Bellville, South Africa
| |
Collapse
|
18
|
Lipovka Y, Alday E, Hernandez J, Velazquez C. Molecular Mechanisms of Biologically Active Compounds from Propolis in Breast Cancer: State of the Art and Future Directions. FOOD REVIEWS INTERNATIONAL 2021. [DOI: 10.1080/87559129.2021.2003380] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Affiliation(s)
- Yulia Lipovka
- Department of Chemistry-Biology, University of Sonora, Hermosillo, Mexico
| | - Efrain Alday
- Department of Chemistry-Biology, University of Sonora, Hermosillo, Mexico
| | - Javier Hernandez
- Unidad de Servicios de Apoyo en Resolución Analítica, Universidad Veracruzana, Xalapa, Mexico
| | - Carlos Velazquez
- Department of Chemistry-Biology, University of Sonora, Hermosillo, Mexico
| |
Collapse
|
19
|
Kim TW, Lee HG. Apigenin Induces Autophagy and Cell Death by Targeting EZH2 under Hypoxia Conditions in Gastric Cancer Cells. Int J Mol Sci 2021; 22:ijms222413455. [PMID: 34948250 PMCID: PMC8706813 DOI: 10.3390/ijms222413455] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Revised: 12/07/2021] [Accepted: 12/13/2021] [Indexed: 01/17/2023] Open
Abstract
Hypoxia is a major obstacle to gastric cancer (GC) therapy and leads to chemoresistance as GC cells are frequently exposed to the hypoxia environment. Apigenin, a flavonoid found in traditional medicine, fruits, and vegetables and an HDAC inhibitor, is a powerful anti-cancer agent against various cancer cell lines. However, detailed mechanisms involved in the treatment of GC using APG are not fully understood. In this study, we investigated the biological activity of and molecular mechanisms involved in APG-mediated treatment of GC under hypoxia. APG promoted autophagic cell death by increasing ATG5, LC3-II, and phosphorylation of AMPK and ULK1 and down-regulating p-mTOR and p62 in GC. Furthermore, our results show that APG induces autophagic cell death via the activation of the PERK signaling, indicating an endoplasmic reticulum (ER) stress response. The inhibition of ER stress suppressed APG-induced autophagy and conferred prolonged cell survival, indicating autophagic cell death. We further show that APG induces ER stress- and autophagy-related cell death through the inhibition of HIF-1α and Ezh2 under normoxia and hypoxia. Taken together, our findings indicate that APG activates autophagic cell death by inhibiting HIF-1α and Ezh2 under hypoxia conditions in GC cells.
Collapse
Affiliation(s)
- Tae Woo Kim
- Immunotherapy Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon 34141, Korea
- Department of Preventive Medicine, College of Korean Medicine, Kyung Hee University, 1 Hoegi, Seoul 130-701, Korea
- Department of Biomedicine & Health Sciences, College of Medicine, The Catholic University of Korea, Seoul 06591, Korea
- Department of Biomolecular Science, University of Science and Technology, Daejeon 34113, Korea
- Correspondence: (T.W.K.); (H.G.L.); Tel.: +82-2-961-0329 (T.W.K.); +82-42-860-4182 (H.G.L.); Fax: +82-2-961-1165 (T.W.K.); +82-42-860-4593 (H.G.L.)
| | - Hee Gu Lee
- Immunotherapy Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon 34141, Korea
- Department of Biomolecular Science, University of Science and Technology, Daejeon 34113, Korea
- Correspondence: (T.W.K.); (H.G.L.); Tel.: +82-2-961-0329 (T.W.K.); +82-42-860-4182 (H.G.L.); Fax: +82-2-961-1165 (T.W.K.); +82-42-860-4593 (H.G.L.)
| |
Collapse
|
20
|
Park MY, Ha SE, Vetrivel P, Kim HH, Bhosale PB, Abusaliya A, Kim GS. Differences of Key Proteins between Apoptosis and Necroptosis. BIOMED RESEARCH INTERNATIONAL 2021; 2021:3420168. [PMID: 34934768 PMCID: PMC8684821 DOI: 10.1155/2021/3420168] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/23/2021] [Revised: 11/08/2021] [Accepted: 11/13/2021] [Indexed: 12/31/2022]
Abstract
Many different types of programmed cell death (PCD) have been identified, including apoptosis and necroptosis. Apoptosis is a type of cell death that is controlled by various genes. It is in charge of eliminating aberrant cells such as cancer cells, replenishing normal cells, and molding the body as it develops. Necroptosis is a type of programmed cell death that combines necrosis and apoptosis. In other words, it takes on a necrotic appearance, although cells die in a controlled manner. Various investigations of these two pathways have revealed that caspase-8, receptor-interacting serine/threonine-protein kinase 1 (RIPK1), and RIPK3 are crucial proteins in charge of the switching between these two pathways, resulting in the activation or inhibition of necroptosis. In this review, we have summarized the key proteins between apoptosis and necroptosis.
Collapse
Affiliation(s)
- Min Yeong Park
- Research Institute of Life Science and College of Veterinary Medicine, Gyeongsang National University, Gazwa, Jinju 52828, Republic of Korea
| | - Sang Eun Ha
- Research Institute of Life Science and College of Veterinary Medicine, Gyeongsang National University, Gazwa, Jinju 52828, Republic of Korea
| | - Preethi Vetrivel
- Research Institute of Life Science and College of Veterinary Medicine, Gyeongsang National University, Gazwa, Jinju 52828, Republic of Korea
| | - Hun Hwan Kim
- Research Institute of Life Science and College of Veterinary Medicine, Gyeongsang National University, Gazwa, Jinju 52828, Republic of Korea
| | - Pritam Bhangwan Bhosale
- Research Institute of Life Science and College of Veterinary Medicine, Gyeongsang National University, Gazwa, Jinju 52828, Republic of Korea
| | - Abuyaseer Abusaliya
- Research Institute of Life Science and College of Veterinary Medicine, Gyeongsang National University, Gazwa, Jinju 52828, Republic of Korea
| | - Gon Sup Kim
- Research Institute of Life Science and College of Veterinary Medicine, Gyeongsang National University, Gazwa, Jinju 52828, Republic of Korea
| |
Collapse
|
21
|
Harmine Hydrochloride Mediates the Induction of G2/M Cell Cycle Arrest in Breast Cancer Cells by Regulating the MAPKs and AKT/FOXO3a Signaling Pathways. Molecules 2021; 26:molecules26216714. [PMID: 34771123 PMCID: PMC8588485 DOI: 10.3390/molecules26216714] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2021] [Revised: 10/24/2021] [Accepted: 11/03/2021] [Indexed: 11/30/2022] Open
Abstract
Breast cancer (BC) is one of the most common causes of death among women worldwide. Recently, interest in novel approaches for BC has increased by developing new drugs derived from natural products with reduced side effects. This study aimed to treat BC cells with harmine hydrochloride (HMH) to identify its anticancer effects and mechanisms. HMH treatment suppressed cell growth, migration, invasion, and colony formation in MCF-7 and MDA-MB-231 cells, regardless of the hormone signaling. It also reduced the phosphorylation of PI3K, AKT, and mTOR and increased FOXO3a expression. Additionally, HMH treatment increased p38 phosphorylation in MCF-7 cells and activated c-Jun N-terminal kinase (JNK) phosphorylation in MDA-MB-231 cells in a dose-dependent manner, where activated p38 and JNK increased FOXO3a expression. Activated FOXO3a increased the expression of p53, p21, and their downstream proteins, including p-cdc25, p-cdc2, and cyclin B1, to induce G2/M cell cycle arrest. Furthermore, HMH inhibited the PI3K/AKT/mTOR pathway by significantly reducing p-AKT expression in combination with LY294002, an AKT inhibitor. These results indicate that mitogen-activated protein kinases (MAPKs) and AKT/FOXO3a signaling pathways mediate the induction of cell cycle arrest following HMH treatment. Therefore, HMH could be a potential active compound for anticancer bioactivity in BC cells.
Collapse
|
22
|
Mohsin A, Haneef K, Ilyas A, Zarina S, Hashim Z. Oxidative Stress Induced Cell Cycle Arrest: Potential Role of PRX-2 and GSTP-1 as Therapeutic Targets in Hepatocellular Carcinoma. Protein Pept Lett 2021; 28:1323-1329. [PMID: 34749598 DOI: 10.2174/0929866528666211105105953] [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: 08/05/2021] [Revised: 09/22/2021] [Accepted: 10/12/2021] [Indexed: 11/22/2022]
Abstract
BACKGROUND The increasing incidence and mortality rate of HCC is a major concern, especially for developing countries of the world. Hence, extensive research is being carried out in order to explore new approaches for developing successful therapeutic strategies for HCC. The controversial role of oxidative stress in the prognosis and treatment of various diseases such as cancer has become the area of great interest and intrigue for many scientists throughout the world. OBJECTIVE We aim to investigate the role of induced oxidative stress on the suppression of HCC Huh-7 cancerous cells as therapeutic approach. METHODS Induction of oxidative stress via H2O2 treatment produced cell cytotoxicity in a dose dependent manner and also led to the over expression of GSTP-1 and PRX-2. The expression of GSTP-1 and PRX-2 was compared in HCC Huh-7 treated, untreated cells and normal hepatocytes using immunocytochemistry. Furthermore, the effects of oxidative stress on cell cycle arrest were also studied through flow cytometry. RESULTS Our study demonstrated the inhibition of cancer cell proliferation as a result of H2O2 induction by arresting the cell cycle at G2 phase. CONCLUSION The induction of oxidative stress could be a potential therapeutic approach for treating HCC in the future. GSTP-1 and PRX-2 can serve as substantial therapeutic targets for the treatment of HCC.
Collapse
Affiliation(s)
- Abeer Mohsin
- Dr. Zafar H. Zaidi Center for Proteomics, University of Karachi, Karachi 75270. Pakistan
| | - Kanwal Haneef
- Dr. Zafar H. Zaidi Center for Proteomics, University of Karachi, Karachi 75270. Pakistan
| | - Amber Ilyas
- Dr. Zafar H. Zaidi Center for Proteomics, University of Karachi, Karachi 75270. Pakistan
| | - Shamshad Zarina
- Dr. Zafar H. Zaidi Center for Proteomics, University of Karachi, Karachi 75270. Pakistan
| | - Zehra Hashim
- Dr. Zafar H. Zaidi Center for Proteomics, University of Karachi, Karachi 75270. Pakistan
| |
Collapse
|
23
|
Awwad A, Poucheret P, Idres YA, Tshibangu DST, Servent A, Ferrare K, Lazennec F, Bidel LPR, Cazals G, Tousch D. In Vitro Tests for a Rapid Evaluation of Antidiabetic Potential of Plant Species Containing Caffeic Acid Derivatives: A Validation by Two Well-Known Antidiabetic Plants, Ocimum gratissimum L. Leaf and Musanga cecropioides R. Br. ex Tedlie (Mu) Stem Bark. Molecules 2021; 26:5566. [PMID: 34577036 PMCID: PMC8465390 DOI: 10.3390/molecules26185566] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Revised: 09/01/2021] [Accepted: 09/06/2021] [Indexed: 01/04/2023] Open
Abstract
Plant bioactive extracts represent a major resource for identifying drugs and adjuvant therapy for type 2 diabetes. To promote early screening of plants' antidiabetic potential, we designed a four in vitro tests strategy to anticipate in vivo bioactivity. Two antidiabetic plants were studied: Ocimum gratissimum L. (Oc) leaf extract and Musanga cecropoides R. Br. ex Tedlie (Mu) stem bark extract. Chemical compositions were analyzed by LCMS and HPLC. Antidiabetic properties were measured based on (1) INS-1 cells for insulin secretion, (2) L6 myoblast cells for insulin sensitization (Glut-4 translocation), (3) L6 myoblast cells for protection against hydrogen peroxide (H2O2) oxidative stress (cell mortality), and (4) liver microsomial fraction for glucose-6-phosphastase activity (G6P). Oc extract increased insulin secretion and insulin sensitivity, whereas it decreased oxidative stress-induced cell mortality and G6P activity. Mu extract decreased insulin secretion and had no effect on insulin sensitivity or G6P activity, but it increased oxidative stress-induced cell mortality. Results were compared with NCRAE, an antidiabetic plant extract used as reference, previously characterized and reported with increased insulin secretion and insulin sensitivity, protection against oxidative stress, and decreased G6P activity. The proposed set of four in vitro tests combined with chemical analysis provided insight into the interest in rapid early screening of plant extract antidiabetic potential to anticipate pharmaco-toxicological in vivo effects.
Collapse
Affiliation(s)
- Abdulmomem Awwad
- Qualisud, University Montpellier, CIRAD, Institut Agro, Avignon Université, University de La Réunion, 34090 Montpellier, France; (A.A.); (Y.A.I.); (A.S.); (K.F.); (F.L.); (D.T.)
| | - Patrick Poucheret
- Qualisud, University Montpellier, CIRAD, Institut Agro, Avignon Université, University de La Réunion, 34090 Montpellier, France; (A.A.); (Y.A.I.); (A.S.); (K.F.); (F.L.); (D.T.)
| | - Yanis A. Idres
- Qualisud, University Montpellier, CIRAD, Institut Agro, Avignon Université, University de La Réunion, 34090 Montpellier, France; (A.A.); (Y.A.I.); (A.S.); (K.F.); (F.L.); (D.T.)
| | - Damien S. T. Tshibangu
- Laboratory of Natural Substances and Medicinal Chemistry, Faculty of Sciences, University of Kinshasa, Kinshasa 190 Kinshasa XI, Democratic Republic of the Congo;
| | - Adrien Servent
- Qualisud, University Montpellier, CIRAD, Institut Agro, Avignon Université, University de La Réunion, 34090 Montpellier, France; (A.A.); (Y.A.I.); (A.S.); (K.F.); (F.L.); (D.T.)
| | - Karine Ferrare
- Qualisud, University Montpellier, CIRAD, Institut Agro, Avignon Université, University de La Réunion, 34090 Montpellier, France; (A.A.); (Y.A.I.); (A.S.); (K.F.); (F.L.); (D.T.)
| | - Françoise Lazennec
- Qualisud, University Montpellier, CIRAD, Institut Agro, Avignon Université, University de La Réunion, 34090 Montpellier, France; (A.A.); (Y.A.I.); (A.S.); (K.F.); (F.L.); (D.T.)
| | - Luc P. R. Bidel
- INRA, UMR AGAP, CIRAD, SupAgro, 2 Place Pierre Viala, 34060 Montpellier, France;
| | - Guillaume Cazals
- Guillaume Cazals, University Montpellier, IBMM UMR5247, CNRS, ENSCM, Place Eugène Bataillon, CEDEX 5, 34095 Montpellier, France;
| | - Didier Tousch
- Qualisud, University Montpellier, CIRAD, Institut Agro, Avignon Université, University de La Réunion, 34090 Montpellier, France; (A.A.); (Y.A.I.); (A.S.); (K.F.); (F.L.); (D.T.)
| |
Collapse
|
24
|
Jiang D, Xu J, Liu S, Nasser MI, Wei W, Mao T, Liu X, Zou X, Li J, Li X. Rosmanol induces breast cancer cells apoptosis by regulating PI3K/AKT and STAT3/JAK2 signaling pathways. Oncol Lett 2021; 22:631. [PMID: 34267823 PMCID: PMC8258625 DOI: 10.3892/ol.2021.12892] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2020] [Accepted: 06/07/2021] [Indexed: 12/31/2022] Open
Abstract
Breast cancer is one of the most frequently diagnosed cancers amongst women; however, there is currently no effective treatment. Natural compounds are considered to contribute to cancer prevention and have a pivotal role in modulating apoptosis. Rosmanol is a phenolic diterpene compound with antioxidant and anti-inflammatory properties. In the present study, the effects of Rosmanol on breast cancer cell proliferation/apoptosis were investigated, and it was demonstrated that it inhibited the proliferation of MCF-7 and MDA-MB 231 cells but did not have a significant effect on normal human breast MCF-10A cells. In addition, the apoptotic process was accelerated by Rosmanol, through mitochondrial pathways and reactive oxygen species (ROS) production caused by DNA damage, which function further demonstrated by the attenuation and addition of the ROS inhibitor, N-acetyl-cysteine. It was also demonstrated that Rosmanol accelerated cell apoptosis, and arrested breast cancer cells in the S phase. Moreover, Rosmanol inhibited proliferation and promoted apoptosis of cancer cells via the inhibition of ERK and STAT3 signals, attributable to the increase in p-p38, the overexpression of protein inhibitor of activated STAT3, and the decrease in PI3K/AKT, ERK and JAK2/STAT3.
Collapse
Affiliation(s)
- Dongjun Jiang
- The Key Laboratory of Molecular Epigenetics of Ministry of Education (MOE), School of Life Sciences, Northeast Normal University, Changchun, Jilin 130024, P.R. China
| | - Jiaqi Xu
- The Key Laboratory of Molecular Epigenetics of Ministry of Education (MOE), School of Life Sciences, Northeast Normal University, Changchun, Jilin 130024, P.R. China
| | - Sitong Liu
- The Key Laboratory of Molecular Epigenetics of Ministry of Education (MOE), School of Life Sciences, Northeast Normal University, Changchun, Jilin 130024, P.R. China
| | - Moussa Ide Nasser
- The Key Laboratory of Molecular Epigenetics of Ministry of Education (MOE), School of Life Sciences, Northeast Normal University, Changchun, Jilin 130024, P.R. China
| | - Wei Wei
- The Key Laboratory of Molecular Epigenetics of Ministry of Education (MOE), School of Life Sciences, Northeast Normal University, Changchun, Jilin 130024, P.R. China
| | - Tianjiao Mao
- Department of Stomatogy, Affiliated Stomatological Hospital of Guangzhou Medical University, Guangzhou, Guangdong 510180, P.R. China
| | - Xintong Liu
- Bioprobe Application Research Unit, Chemical Biology Department, RIKEN-Max Planck Joint Research Division, RIkagaku KENkyusho/Institute of Physical and Chemical Research (RIKEN) Center for Sustainable Resource Science, Wako, Saitama 351-0198, Japan
| | - Xiaopan Zou
- The Key Laboratory of Molecular Epigenetics of Ministry of Education (MOE), School of Life Sciences, Northeast Normal University, Changchun, Jilin 130024, P.R. China
| | - Jiang Li
- Department of Stomatogy, Affiliated Stomatological Hospital of Guangzhou Medical University, Guangzhou, Guangdong 510180, P.R. China
| | - Xiaomeng Li
- The Key Laboratory of Molecular Epigenetics of Ministry of Education (MOE), School of Life Sciences, Northeast Normal University, Changchun, Jilin 130024, P.R. China
| |
Collapse
|
25
|
An In Vitro-In Vivo Evaluation of the Antiproliferative and Antiangiogenic Effect of Flavone Apigenin against SK-MEL-24 Human Melanoma Cell Line. ACTA ACUST UNITED AC 2021; 2021:5552664. [PMID: 34239802 PMCID: PMC8241515 DOI: 10.1155/2021/5552664] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2021] [Revised: 04/09/2021] [Accepted: 06/05/2021] [Indexed: 01/02/2023]
Abstract
One of the most important class of natural compounds with successful preclinical results in the management of cancer is the flavonoids. Due to the plethora of biological activities, apigenin (4',5,7 trihydroxyflavone) is a main representant of the flavone subclass. Although the antiproliferative and antiangiogenic effects of apigenin were studied on a significant number of human and murine melanoma cell lines, in order to complete the data existing in the literature, the aim of this study is to evaluate the in vitro effect of apigenin on SK-MEL-24 human melanoma cell line as well as in vivo on tumor angiogenesis using the aforementioned cell line on the chorioallantoic membrane assay. Results have shown that in the range of tested doses, the phytocompound presents significant antiproliferative, cytotoxic, and antimigratory potential at 30 μM, respectively, 60 μM. Moreover, the phytocompound in both tested concentrations limited melanoma cell growth and migration and induced a reduced angiogenic reaction limiting melanoma cell development.
Collapse
|
26
|
Klein S, Distel LVR, Neuhuber W, Kryschi C. Caffeic Acid, Quercetin and 5-Fluorocytidine-Functionalized Au-Fe 3O 4 Nanoheterodimers for X-ray-Triggered Drug Delivery in Breast Tumor Spheroids. NANOMATERIALS (BASEL, SWITZERLAND) 2021; 11:1167. [PMID: 33947086 PMCID: PMC8146450 DOI: 10.3390/nano11051167] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/10/2021] [Revised: 04/25/2021] [Accepted: 04/26/2021] [Indexed: 01/11/2023]
Abstract
Au-Fe3O4 nanoheterodimers (NHD) were functionalized with the natural and synthetic anticancer drugs caffeic acid (CA), quercetin (Q) and 5-fluorocytidine (5FC). Their X-radiation dose-enhancing potential and chemotherapeutic efficacy for bimodal cancer therapy were investigated by designing multicellular tumor spheroids (MCTS) to in vitro avascular tumor models. MCTS were grown from the breast cancer cell lines MCF-7, MDA-MB-231, and MCF-10A. The MCF-7, MDA-MB-231 and MCF-10A MCTS were incubated with NHD-CA, NHD-Q, or NHD-5FC and then exposed to fractionated X-radiation comprising either a single 10 Gy dose, 2 daily single 5 Gy doses or 5 daily single 2 Gy doses. The NHD-CA, NHD-Q, and NHD-5FC affected the growth of X-ray irradiated and non-irradiated MCTS in a different manner. The impact of the NHDs on the glycolytic metabolism due to oxygen deprivation inside MCTS was assessed by measuring lactate secretion and glucose uptake by the MCTS. The NHD-CA and NHD-Q were found to act as X-radiation dose agents in MCF-7 MCTS and MDA-MB-231 MCTS and served as radioprotector in MCF-10A MCTS. X-ray triggered release of CA and Q inhibited lactate secretion and thereupon disturbed glycolytic reprogramming, whereas 5FC exerted their cytotoxic effects on both, healthy and tumor cells, after their release into the cytosol.
Collapse
Affiliation(s)
- Stefanie Klein
- Department of Chemistry and Pharmacy, Physical Chemistry I and ICMM, Friedrich-Alexander University of Erlangen-Nuremberg, Egerlandstr. 3, D-91058 Erlangen, Germany;
| | - Luitpold V. R. Distel
- Department of Radiation Oncology, Friedrich-Alexander University of Erlangen-Nuremberg, Universitätsstr. 27, D-91054 Erlangen, Germany;
| | - Winfried Neuhuber
- Institute of Anatomy, Chair of Anatomy and Cell Biology, Friedrich Alexander University Erlangen-Nuremberg, Krankenhausstr. 9, D-91054 Erlangen, Germany;
| | - Carola Kryschi
- Department of Chemistry and Pharmacy, Physical Chemistry I and ICMM, Friedrich-Alexander University of Erlangen-Nuremberg, Egerlandstr. 3, D-91058 Erlangen, Germany;
| |
Collapse
|
27
|
Buranrat B. Growth Inhibition, Apoptosis Induction and Migratory Suppression by Careya arborea Leaf Extract in HeLa Cervical Cells. INT J PHARMACOL 2021. [DOI: 10.3923/ijp.2021.339.349] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
|
28
|
Gospodinova ZI, Zupkó I, Bózsity N, Manova VI, Georgieva MS, Todinova SJ, Taneva SG, Ocsovszki I, Krasteva ME. Cotinus coggygria Scop. induces cell cycle arrest, apoptosis, genotoxic effects, thermodynamic and epigenetic events in MCF7 breast cancer cells. ACTA ACUST UNITED AC 2021; 76:129-140. [PMID: 32975208 DOI: 10.1515/znc-2020-0087] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2020] [Accepted: 09/05/2020] [Indexed: 12/24/2022]
Abstract
Current plant-derived anticancer therapeutics aim to reach higher effectiveness, to potentiate chemosensitivity and minimize the toxic side effects compared to conventional chemotherapy. Cotinus coggygria Scop. is a herb with high pharmacological potential, widely applied in traditional phytotherapy. Our previous study revealed that leaf aqueous ethanolic extract from C. coggygria exerts in vitro anticancer activity on human breast, ovarian and cervical cancer cell lines. The objective of the present research was to investigate possible molecular mechanisms and targets of the antitumor activity of the extract in breast cancer MCF7 cells through analysis of cell cycle and apoptosis, clonogenic ability assessment, evaluation of the extract genotoxic capacity, characterization of cells thermodynamic properties, and analysis on the expression of genes involved in cellular epigenetic processes. The obtained results indicated that in MCF7 cells C. coggygria extract causes S phase cell cycle arrest and triggers apoptosis, reduces colony formation, induces DNA damage, affects cellular thermodynamic parameters, and tends to inhibit the relative expression of DNMT1, DNMT3a, MBD3, and p300. Further studies on the targeted molecules and the extract anti-breast cancer potential on animal experimental model system, need to be performed in the future.
Collapse
Affiliation(s)
- Zlatina I Gospodinova
- Laboratory of Genome Dynamics and Stability, Institute of Plant Physiology and Genetics, Bulgarian Academy of Sciences, Acad. G. Bonchev Str., Bldg. 21, 1113 Sofia, Bulgaria
| | - Istvan Zupkó
- Department of Pharmacodynamics and Biopharmacy, University of Szeged, Eotvos Str. 6, H-6720 Szeged, Hungary
| | - Noémi Bózsity
- Department of Pharmacodynamics and Biopharmacy, University of Szeged, Eotvos Str. 6, H-6720 Szeged, Hungary
| | - Vasilissa I Manova
- Laboratory of Genome Dynamics and Stability, Institute of Plant Physiology and Genetics, Bulgarian Academy of Sciences, Acad. G. Bonchev Str., Bldg. 21, 1113 Sofia, Bulgaria
| | - Mariyana S Georgieva
- Laboratory of Regulation of Gene Expression, Institute of Plant Physiology and Genetics, Bulgarian Academy of Sciences, Acad. G. Bonchev Str., Bldg. 21, 1113 Sofia, Bulgaria
| | - Svetla J Todinova
- Department of Biomacromolecules and Biomolecular Interactions, Institute of Biophysics and Biomedical Engineering, Bulgarian Academy of Sciences, Acad. G. Bonchev Str., Bldg. 21, 1113 Sofia, Bulgaria
| | - Stefka G Taneva
- Department of Biomacromolecules and Biomolecular Interactions, Institute of Biophysics and Biomedical Engineering, Bulgarian Academy of Sciences, Acad. G. Bonchev Str., Bldg. 21, 1113 Sofia, Bulgaria
| | - Imre Ocsovszki
- Department of Biochemistry, University of Szeged, Dóm tér 9, H-6720 Szeged, Hungary
| | - Maria E Krasteva
- Laboratory of Genome Dynamics and Stability, Institute of Plant Physiology and Genetics, Bulgarian Academy of Sciences, Acad. G. Bonchev Str., Bldg. 21, 1113 Sofia, Bulgaria
| |
Collapse
|
29
|
Anaya-Eugenio GD, Blanco Carcache PJ, Ninh TN, Ren Y, Soejarto DD, Kinghorn AD. A pentamethoxylated flavone from Glycosmis ovoidea promotes apoptosis through the intrinsic pathway and inhibits migration of MCF-7 breast cancer cells. Phytother Res 2021; 35:1634-1645. [PMID: 33124130 PMCID: PMC8005457 DOI: 10.1002/ptr.6930] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2020] [Revised: 10/02/2020] [Accepted: 10/09/2020] [Indexed: 12/15/2022]
Abstract
The rare flavone 5,3'-dihydroxy-3,6,7,8,4'-pentamethoxyflavone (PMF) has been isolated from several plant species, and its cytotoxic activity has been reported against many types of cancer cells. In this study, PMF was purified from Glycomis ovoidea collected in Vietnam, and its antiproliferative effects and underlying mechanism of action were investigated against MCF-7 cells. PMF inhibited growth in MCF-7 > MCF-10A > MDA-MB-231 cells after 72 hr treatment, with IC50 values of 1.5, 1.9, and 8.6 μg/ml, respectively. Further experiments conducted with this compound in MCF-7 cells, showed the loss of mitochondrial membrane potential, reactive oxygen species overproduction, upregulation of BAX, cytochrome c, caspase-3 and PARP-1 and down-regulation of BCL-2 proteins as well as an increase in caspase-3/-7 activity, suggesting induction of the apoptotic intrinsic pathway. Furthermore, PMF increased cell cycle arrest in the G1 phase, which correlated with increments in the p53 and p21 levels. Additionally, MCF-7 cell migration was inhibited, which could be related to NF-κB p65 downregulation. Finally, PMF did not show toxicity in vivo in a zebrafish (Danio rerio) model. In conclusion, PMF induces cell death in MCF-7 cells through regulation of the BCL-2 protein family and may be proposed as a lead as a potential alternative for breast cancer therapy.
Collapse
Affiliation(s)
- Gerardo D. Anaya-Eugenio
- Division of Medicinal Chemistry and Pharmacognosy, College of Pharmacy, The Ohio State University, Columbus, Ohio 43210, United States
| | - Peter J. Blanco Carcache
- Division of Medicinal Chemistry and Pharmacognosy, College of Pharmacy, The Ohio State University, Columbus, Ohio 43210, United States
| | - Tran Ngoc Ninh
- Institute of Ecology and Biological Resources, Vietnam Academy of Science and Technology, Hoang Quoc Viet, Cau Giay, Hanoi, Vietnam
| | - Yulin Ren
- Division of Medicinal Chemistry and Pharmacognosy, College of Pharmacy, The Ohio State University, Columbus, Ohio 43210, United States
| | - Djaja D. Soejarto
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Illinois at Chicago, Chicago, Illinois 60612, United States
| | - A. Douglas Kinghorn
- Division of Medicinal Chemistry and Pharmacognosy, College of Pharmacy, The Ohio State University, Columbus, Ohio 43210, United States
| |
Collapse
|
30
|
Wu L, Guo T, Deng R, Liu L, Yu Y. Apigenin Ameliorates Insulin Resistance and Lipid Accumulation by Endoplasmic Reticulum Stress and SREBP-1c/SREBP-2 Pathway in Palmitate-Induced HepG2 Cells and High-Fat Diet-Fed Mice. J Pharmacol Exp Ther 2021; 377:146-156. [PMID: 33509902 DOI: 10.1124/jpet.120.000162] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2021] [Accepted: 01/21/2021] [Indexed: 02/06/2023] Open
Abstract
Insulin resistance (IR) is the common basis of diabetes and cardiovascular diseases, and its development is closely associated with lipid metabolism disorder. Flavonoids have definite chemical defense effects, including anti-inflammatory effects, anticancer effects, and antimutation effects. However, the function and mechanism of apigenin (AP, a kind of flavonoid) in IR are still unclear. In our study, intracellular fat accumulation model cells and high-fat diet (HFD)-fed model mice were established using palmitate (PA) and HFD. Mechanistically, we first demonstrated that AP could notably downregulate sterol regulatory element-binding protein 1c (SREBP-1c), sterol regulatory element-binding protein 2 (SREBP-2), fatty acid synthase, stearyl-CoA desaturase 1, and 3-hydroxy-3-methyl-glutaryl-CoA reductase in PA-induced hyperlipidemic cells and mice. Functionally, we verified that AP could markedly reduce lipid accumulation in PA-induced hyperlipidemic cells and decrease the body weight, visceral fat weight, IR, and lipid accumulation in HFD-induced hyperlipidemic mice. Besides, we showed that PA could significantly downregulate endoplasmic reticulum stress (ERS)-related proteins and inhibit ERS. Furthermore, we proved that AP could reduce blood lipids by inhibiting ERS in PA-induced hyperlipidemic cells. Meanwhile, 4-phenyl butyric acid (also called ERS alleviator), like AP, could significantly reduce blood lipids and alleviate IR in HFD-fed model mice. Therefore, we concluded that AP could substantially improve the disorder of lipid metabolism, and its mechanism might be related to the decrease of SREBP-1c, SREBP-2, and downstream genes, the inhibition of ERS, and the reduction of blood lipids and IR. SIGNIFICANCE STATEMENT: Apigenin, a nontoxic and naturally sourced flavonoid, has antihyperlipidemic properties in mice and hepatocyte. This study highlights a new mechanism of apigenin and proposes that these hypolipidemic effects are associated with the mitigation of endoplasmic reticulum stress and insulin resistance in diet-induced obesity. This study might provide translational insight into the prevention and treatment of apigenin in hyperlipidemia-related diseases.
Collapse
Affiliation(s)
- Liling Wu
- College of Animal Science and Technology, Southwest University, Chongqing, China (L.W., L.L., Y.Y.) and Department of Pharmacy, Nanchong Central Hospital, The Second Clinical Medical College (L.W., T.G.), Department of Clinical Medicine (R.D.), North Sichuan Medical College, Nanchong, Sichuan, China
| | - Tingdong Guo
- College of Animal Science and Technology, Southwest University, Chongqing, China (L.W., L.L., Y.Y.) and Department of Pharmacy, Nanchong Central Hospital, The Second Clinical Medical College (L.W., T.G.), Department of Clinical Medicine (R.D.), North Sichuan Medical College, Nanchong, Sichuan, China
| | - Ranxi Deng
- College of Animal Science and Technology, Southwest University, Chongqing, China (L.W., L.L., Y.Y.) and Department of Pharmacy, Nanchong Central Hospital, The Second Clinical Medical College (L.W., T.G.), Department of Clinical Medicine (R.D.), North Sichuan Medical College, Nanchong, Sichuan, China
| | - Lusheng Liu
- College of Animal Science and Technology, Southwest University, Chongqing, China (L.W., L.L., Y.Y.) and Department of Pharmacy, Nanchong Central Hospital, The Second Clinical Medical College (L.W., T.G.), Department of Clinical Medicine (R.D.), North Sichuan Medical College, Nanchong, Sichuan, China
| | - Yongxiong Yu
- College of Animal Science and Technology, Southwest University, Chongqing, China (L.W., L.L., Y.Y.) and Department of Pharmacy, Nanchong Central Hospital, The Second Clinical Medical College (L.W., T.G.), Department of Clinical Medicine (R.D.), North Sichuan Medical College, Nanchong, Sichuan, China
| |
Collapse
|
31
|
Ahmed SA, Parama D, Daimari E, Girisa S, Banik K, Harsha C, Dutta U, Kunnumakkara AB. Rationalizing the therapeutic potential of apigenin against cancer. Life Sci 2020; 267:118814. [PMID: 33333052 DOI: 10.1016/j.lfs.2020.118814] [Citation(s) in RCA: 55] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Revised: 11/14/2020] [Accepted: 11/20/2020] [Indexed: 12/12/2022]
Abstract
BACKGROUND Despite the remarkable advances made in the diagnosis and treatment of cancer during the past couple of decades, it remains the second largest cause of mortality in the world, killing approximately 9.6 million people annually. The major challenges in the treatment of the advanced stage of this disease are the development of chemoresistance, severe adverse effects of the drugs, and high treatment cost. Therefore, the development of drugs that are safe, efficacious, and cost-effective remains a 'Holy Grail' in cancer research. However, the research over the past four decades shed light on the cancer-preventive and therapeutic potential of natural products and their underlying mechanism of action. Apigenin is one such compound, which is known to be safe and has significant potential in the prevention and therapy of this disease. AIM To assess the literature available on the potential of apigenin and its analogs in modulating the key molecular targets leading to the prevention and treatment of different types of cancer. METHOD A comprehensive literature search has been carried out on PubMed for obtaining information related to the sources and analogs, chemistry and biosynthesis, physicochemical properties, biological activities, bioavailability and toxicity of apigenin. KEY FINDINGS The literature search resulted in many in vitro, in vivo and a few cohort studies that evidenced the effectiveness of apigenin and its analogs in modulating important molecular targets and signaling pathways such as PI3K/AKT/mTOR, JAK/STAT, NF-κB, MAPK/ERK, Wnt/β-catenin, etc., which play a crucial role in the development and progression of cancer. In addition, apigenin was also shown to inhibit chemoresistance and radioresistance and make cancer cells sensitive to these agents. Reports have further revealed the safety of the compound and the adaptation of nanotechnological approaches for improving its bioavailability. SIGNIFICANCE Hence, the present review recapitulates the properties of apigenin and its pharmacological activities against different types of cancer, which warrant further investigation in clinical settings.
Collapse
Affiliation(s)
- Semim Akhtar Ahmed
- Cell and Molecular Biology Laboratory, Department of Zoology, Cotton University, Pan Bazar, Guwahati, Assam 781001, India
| | - Dey Parama
- Cancer Biology Laboratory and DBT-AIST International Center for Translational and Environmental Research (DAICENTER), Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Assam 781039, India
| | - Enush Daimari
- Cell and Molecular Biology Laboratory, Department of Zoology, Cotton University, Pan Bazar, Guwahati, Assam 781001, India
| | - Sosmitha Girisa
- Cancer Biology Laboratory and DBT-AIST International Center for Translational and Environmental Research (DAICENTER), Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Assam 781039, India
| | - Kishore Banik
- Cancer Biology Laboratory and DBT-AIST International Center for Translational and Environmental Research (DAICENTER), Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Assam 781039, India
| | - Choudhary Harsha
- Cancer Biology Laboratory and DBT-AIST International Center for Translational and Environmental Research (DAICENTER), Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Assam 781039, India
| | - Uma Dutta
- Cell and Molecular Biology Laboratory, Department of Zoology, Cotton University, Pan Bazar, Guwahati, Assam 781001, India.
| | - Ajaikumar B Kunnumakkara
- Cancer Biology Laboratory and DBT-AIST International Center for Translational and Environmental Research (DAICENTER), Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Assam 781039, India.
| |
Collapse
|
32
|
Anticancer natural medicines: An overview of cell signaling and other targets of anticancer phytochemicals. Eur J Pharmacol 2020; 888:173488. [DOI: 10.1016/j.ejphar.2020.173488] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2020] [Revised: 07/23/2020] [Accepted: 08/13/2020] [Indexed: 02/07/2023]
|
33
|
Yehia S, Abdel-Salam IM, Elgamal BM, El-Agamy B, Hamdy GM, Aldesouki HM. Cytotoxic and Apoptotic Effects of Luffa Cylindrica Leaves Extract against Acute Lymphoblastic Leukemic Stem Cells. Asian Pac J Cancer Prev 2020; 21:3661-3668. [PMID: 33369466 PMCID: PMC8046306 DOI: 10.31557/apjcp.2020.21.12.3661] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2020] [Accepted: 12/15/2020] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Acute lymphoblastic leukemia (ALL) is an aggressive malignancy defined by accumulation of lymphoblasts in the bone marrow. Leukemic stem cells (LSCs) are the major cause of the recurrence and metastasis of ALL. This study aimed to develop an effective anti-cancer agent targeting these LSCs. Luffa Cylindrica (L.C.) leaves extract was selected to evaluate its effect on ALL via eradicating the LSCs as it contains many active anti-cancer flavonoids. METHODS Thirty-two bone marrow samples of ALL patients were used in this study. LSCs population was identified in the selected samples. Cell viability was measured by MTT assay and flow cytometry. Cell cycle, apoptosis, proliferation marker; ki-67 and colony forming assay were further analyzed. RESULTS This study revealed the expression of CD34+/CD38+ cells in addition to CD34+/CD38- population and the extract was effective against the two LSCs populations. MTT assay showed that treated leukemic cells exhibited significant reduction in the viable cells in a dose dependent manner with IC50 of 3 µg/µl which was then confirmed by flow cytometry. Cell cycle analysis results showed significant reduction in the percentage of cells treated with L.C. extract in both the S and G0/G1 phases, with concomitant increase in the G2/M phase. Also, L.C. extract could effectively induce apoptosis, inhibit proliferation and suppress colonogenecity of leukemic cells. CONCLUSION This study validated the medicinal potential of L.C. leaves extract as a promising anti-leukemic agent targeting both LSCs and blasts in ALL patients, which may be explained by the synergy found between its potent flavonoids especially apigenin, luteolin and kaempferol.
Collapse
Affiliation(s)
- Shimaa Yehia
- Biochemistry Department, Faculty of Science, Ain Shams University, Cairo, Egypt.
| | | | - Basma M. Elgamal
- Clinical Pathology Department, National Cancer Institute, Cairo University, Cairo, Egypt.
| | - Basma El-Agamy
- Biochemistry Department, Faculty of Science, Ain Shams University, Cairo, Egypt.
| | - Germine M. Hamdy
- Biochemistry Department, Faculty of Science, Ain Shams University, Cairo, Egypt.
| | - Hala M. Aldesouki
- Biochemistry Department, Faculty of Science, Ain Shams University, Cairo, Egypt.
| |
Collapse
|
34
|
Apigenin and Hesperidin Downregulate DNA Repair Genes in MCF-7 Breast Cancer Cells and Augment Doxorubicin Toxicity. Molecules 2020; 25:molecules25194421. [PMID: 32993087 PMCID: PMC7582946 DOI: 10.3390/molecules25194421] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2020] [Revised: 09/22/2020] [Accepted: 09/23/2020] [Indexed: 01/03/2023] Open
Abstract
A number of studies have confirmed anti-tumor activity of flavonoids and their ability to enhance the effectiveness of classical anticancer drugs. The mechanism of this phenomenon is difficult to explain because of the ambivalent nature of these compounds. Many therapeutic properties of these compounds are attributed to their antioxidant activity; however, it is known that they can act as oxidants. The aim of this study was to assess the influence of apigenin and hesperidin on MCF-7 breast cancer cells with doxorubicin. The cytotoxic effect was determined using an MTT test and cell cycle analysis. To evaluate the possible interaction mechanism, reduced glutathione levels, as well as the DNA oxidative damage and the double strand breaks, were evaluated. Additionally, mRNA expression of genes related to DNA repair was assessed. It was demonstrated that flavonoids intensified the cytotoxic effect of doxorubicin despite flavonoids reduced oxidative damage caused by the drug. At the same time, the number of double strand breaks significantly increased and expression of tested genes was downregulated. In conclusion, both apigenin and hesperidin enhance the cytotoxic effects of doxorubicin on breast cancer cells, and this phenomenon occurs regardless of oxidative stress but is accompanied by disorders of DNA damage response mechanisms.
Collapse
|
35
|
García-Martínez DJ, Calzada Funes J, Martín Saborido C, Santos C. Grape Polyphenols to Arrest in Vitro Proliferation of Human Leukemia Cells: A Systematic Review and Meta-analysis. FOOD REVIEWS INTERNATIONAL 2020. [DOI: 10.1080/87559129.2020.1810700] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Affiliation(s)
| | - Javier Calzada Funes
- Instituto De Nanociencia Y Materiales De Aragón (INMA), CSIC-Universidad De Zaragoza, Zaragoza, Spain
- Department of Chemical and Environmental Engineering, Universidad De Zaragoza, Zaragoza, Spain
| | - Carlos Martín Saborido
- ERN-Transplant Child, Hospital La Paz Institute for Health Research (Idipaz), Madrid, Spain
| | - Cruz Santos
- Faculty of Experimental Sciences, Universidad Francisco De Vitoria, Madrid, Spain
| |
Collapse
|
36
|
Ren G, Hao X, Yang S, Chen J, Qiu G, Ang KP, Mohd Tamrin MI. 10H-3,6-Diazaphenothiazines triggered the mitochondrial-dependent and cell death receptor-dependent apoptosis pathways and further increased the chemosensitivity of MCF-7 breast cancer cells via inhibition of AKT1 pathways. J Biochem Mol Toxicol 2020; 34:e22544. [PMID: 32619082 DOI: 10.1002/jbt.22544] [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: 02/25/2020] [Revised: 04/21/2020] [Accepted: 05/29/2020] [Indexed: 11/11/2022]
Abstract
Breast cancer is one of the leading causes of death in cancer categories, followed by lung, colorectal, and ovarian among the female gender across the world. 10H-3,6-diazaphenothiazine (PTZ) is a thiazine derivative compound that exhibits many pharmacological activities. Herein, we proceed to investigate the pharmacological activities of PTZ toward breast cancer MCF-7 cells as a representative in vitro breast cancer cell model. The PTZ exhibited a proliferation inhibition (IC50 = 0.895 µM) toward MCF-7 cells. Further, cell cycle analysis illustrated that the S-phase checkpoint was activated to achieve proliferation inhibition. In vitro cytotoxicity test on three normal cell lines (HEK293 normal kidney cells, MCF-10A normal breast cells, and H9C2 normal heart cells) demonstrated that PTZ was more potent toward cancer cells. Increase in the levels of reactive oxygen species results in polarization of mitochondrial membrane potential (ΔΨm), together with suppression of mitochondrial thioredoxin reductase enzymatic activity suggested that PTZ induced oxidative damages toward mitochondria and contributed to improved drug efficacy toward treatment. The RT2 PCR Profiler Array (human apoptosis pathways) proved that PTZ induced cell death via mitochondria-dependent and cell death receptor-dependent pathways, through a series of modulation of caspases, and the respective morphology of apoptosis was observed. Mechanistic studies of apoptosis suggested that PTZ inhibited AKT1 pathways resulting in enhanced drug efficacy despite it preventing invasion of cancer cells. These results showed the effectiveness of PTZ in initiation of apoptosis, programmed cell death, toward highly chemoresistant MCF-7 cells, thus suggesting its potential as a chemotherapeutic drug.
Collapse
Affiliation(s)
- Guanghui Ren
- Department of General Surgery, Shenzhen Hospital of Southern Medical University, Shenzhen, Guangdong, China
| | - Xiaoyan Hao
- Department of Thyroid and Breast Surgery, Longgang Central Hospital of Shenzhen, Shenzhen, Guangdong, China
| | - Shuyi Yang
- Department of General Surgery, Shenzhen Hospital of Southern Medical University, Shenzhen, Guangdong, China
| | - Jun Chen
- Department of General Surgery, Shenzhen Hospital of Southern Medical University, Shenzhen, Guangdong, China
| | - Guobin Qiu
- Department of General Surgery, Shenzhen Hospital of Southern Medical University, Shenzhen, Guangdong, China
| | - Kok Pian Ang
- Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Serdang, Selangor, Malaysia
| | - Mohd Islahuddin Mohd Tamrin
- Department of Surgery, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Serdang, Selangor, Malaysia
| |
Collapse
|
37
|
Shendge AK, Chaudhuri D, Basu T, Mandal N. A natural flavonoid, apigenin isolated from Clerodendrum viscosum leaves, induces G2/M phase cell cycle arrest and apoptosis in MCF-7 cells through the regulation of p53 and caspase-cascade pathway. Clin Transl Oncol 2020; 23:718-730. [PMID: 32715386 DOI: 10.1007/s12094-020-02461-0] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2020] [Accepted: 07/15/2020] [Indexed: 12/12/2022]
Abstract
BACKGROUND With 9.6 million deaths in 2018, cancer remains the second leading cause of death worldwide. Breast cancer is the most deadly type of cancer among females, with 55.2% of crude incidence rate and 16.6% of crude mortality rate. PURPOSE The present study was aimed to investigate the anti-breast cancer potential of natural dietary flavonoid, apigenin isolated from Clerodendrum viscosum leaves. METHODS Apigenin was evaluated for in-depth anticancer activity in MCF-7 cells using cell viability assay, cell cycle analysis, Annexin-V-FLUOS staining, ROS induction, morphological analysis, and western blot analysis. RESULTS Apigenin showed selective cytotoxicity on MCF-7 cells with an IC50-56.72 ± 2.35 µM, while negligible cytotoxicity was observed on WI-38 cells. Further, the flow cytometer-based analysis showed that apigenin halted MCF-7 cells in the G2/M phase arrest followed by dose-dependent apoptosis. Moreover, the FACS and confocal microscopy results confirmed the elevation of intracellular ROS and nuclear fragmentation in apigenin-treated MCF-7 cells. Western blots showed up-regulation of cell cycle regulatory proteins, increased p53 expression, Bax/Bcl-2 ratio, activation of caspases, and cleavage of PARP. Finally, apigenin treatment in the presence of Pifithrin-µ showed decreased apoptotic population and it was further confirmed through western blotting study. The results revealed the vital role of p53 in apigenin-induced apoptosis in MCF-7 cells. CONCLUSIONS In the present findings, treatment of apigenin-induced intracellular ROS in MCF-7 cells followed by induction of G2/M phase cell cycle arrest and further apoptosis through the regulation of p53 and caspase-cascade signaling pathway.
Collapse
Affiliation(s)
- A K Shendge
- Division of Molecular Medicine, Bose Institute, P-1/12 CIT Scheme - VIIM, Kolkata, West Bengal, 700054, India
| | - D Chaudhuri
- Division of Molecular Medicine, Bose Institute, P-1/12 CIT Scheme - VIIM, Kolkata, West Bengal, 700054, India
| | - T Basu
- Division of Molecular Medicine, Bose Institute, P-1/12 CIT Scheme - VIIM, Kolkata, West Bengal, 700054, India
| | - N Mandal
- Division of Molecular Medicine, Bose Institute, P-1/12 CIT Scheme - VIIM, Kolkata, West Bengal, 700054, India.
| |
Collapse
|
38
|
Moradi M, Gholipour H, Sepehri H, Attari F, Delphi L, Arefian E, Moridi Farimani M. Flavonoid calycopterin triggers apoptosis in triple-negative and ER-positive human breast cancer cells through activating different patterns of gene expression. Naunyn Schmiedebergs Arch Pharmacol 2020; 393:2145-2156. [PMID: 32617603 DOI: 10.1007/s00210-020-01917-y] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2020] [Accepted: 06/02/2020] [Indexed: 12/17/2022]
Abstract
Breast cancer is the most common cause of death related to cancer in women, and several studies proved that flavonoids could induce apoptosis in this cancer through different pathways. Calycopterin is a flavonoid which was shown to induce preferential antiproliferative effects on some cancers; however, no information is available on its effect on breast cancer. Therefore, in this paper, the apoptotic effect of calycopterin and its underlying mechanism in two different breast cancer cells, MDA-MB-231 and MCF7 cell lines were investigated. MTT assay showed that calycopterin reduced the proliferation of both cancer lines with no adverse effect on normal cells. The clonogenic assay showed that calycopterin treatments decreased the colony numbers and sizes, and wound healing assay demonstrated the inhibition of migration in both cancer cells. Cell cycle and annexin/PI analyses indicated that calycopterin augmented sub-G1 population and annexin/PI-positive cells. Gene expression revealed that Bax/Bcl2 increased in the MDA-MB-231 cell line, while no change was observed in that of the MCF7 line. Expression of gene caspase-8 was augmented in both lines, although increased expression of caspase-3 was found just in MDA-MB-231 cells. Our results validated the apoptotic effect of calycopterin on both breast cancer lines with more potency on triple-negative ones.
Collapse
Affiliation(s)
- Mehrnaz Moradi
- Department of Animal Biology, School of Biology, College of Science, University of Tehran, Tehran, Iran
| | - Hajar Gholipour
- Department of Animal Biology, School of Biology, College of Science, University of Tehran, Tehran, Iran
| | - Houri Sepehri
- Department of Animal Biology, School of Biology, College of Science, University of Tehran, Tehran, Iran.
| | - Farnoosh Attari
- Department of Animal Biology, School of Biology, College of Science, University of Tehran, Tehran, Iran.
| | - Ladan Delphi
- Department of Animal Biology, School of Biology, College of Science, University of Tehran, Tehran, Iran
| | - Ehsan Arefian
- Molecular Virology Lab, Department of Microbiology, School of Biology, College of Science, University of Tehran, Tehran, Iran
| | - Mahdi Moridi Farimani
- Department of Phytochemistry, Medicinal Plants and Drugs Research Institute, Shahid Beheshti University, Tehran, Iran
| |
Collapse
|
39
|
Qi Y, Ding Z, Yao Y, Ren F, Yin M, Yang S, Chen A. Apigenin induces apoptosis and counteracts cisplatin-induced chemoresistance via Mcl-1 in ovarian cancer cells. Exp Ther Med 2020; 20:1329-1336. [PMID: 32742367 PMCID: PMC7388300 DOI: 10.3892/etm.2020.8880] [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: 03/15/2019] [Accepted: 02/25/2020] [Indexed: 12/27/2022] Open
Abstract
Ovarian cancer (OC) is one of the prominent causes of mortality in female patients diagnosed with gynecologic malignancies. While it has previously been demonstrated that apigenin inhibits cell growth in colon and breast cancer cells, the effect of apigenin in OC cells is not fully understood. Therefore, the aim of the present study was to investigate the impact of apigenin on cell death and resistance to cisplatin in OC cells. It was found that apigenin inhibited proliferation, hindered cell cycle progression and promoted SKOV3 cell apoptosis. Moreover, these effects were also observed in cisplatin-resistant SKOV3/DDP cells. Furthermore, apigenin reduced the mitochondrial transmembrane potential, and elevated the ratios of cleaved caspase-3/caspase-3 and Bax/Bcl-2 in the two cell types. Reverse transcription-quantitative PCR and western blotting results demonstrated that apigenin significantly downregulated Mcl-1 at the transcriptional and translational levels in SKOV3 and SKOV3/DDP cells, which was responsible for its cytotoxic functions and chemosensitizing effects. Collectively, the present results identified the impact of apigenin on OC cell death and resistance to cisplatin, and the potential molecular mechanisms. However, additional studies are required to further elucidate the underlying mechanisms.
Collapse
Affiliation(s)
- Yuyan Qi
- Department of Gynecology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong 266000, P.R. China
| | - Zhaoxia Ding
- Department of Gynecology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong 266000, P.R. China
| | - Yushuang Yao
- Department of Gynecology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong 266000, P.R. China
| | - Feifei Ren
- Department of Obstetrics and Gynecology, The Affiliated Hospital of Taishan Medical University, Taian, Shandong 271000, P.R. China
| | - Min Yin
- Department of Gynecology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong 266000, P.R. China
| | - Songbin Yang
- Department of Gynecology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong 266000, P.R. China
| | - Aiping Chen
- Department of Gynecology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong 266000, P.R. China
| |
Collapse
|
40
|
Malla RR, Deepak K, Merchant N, Dasari VR. Breast Tumor Microenvironment: Emerging target of therapeutic phytochemicals. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2020; 70:153227. [PMID: 32339885 DOI: 10.1016/j.phymed.2020.153227] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/02/2019] [Revised: 03/31/2020] [Accepted: 04/11/2020] [Indexed: 06/11/2023]
Abstract
Triple negative breast cancer (TNBC) is the most aggressive and challenging form of breast cancers. Tumor microenvironment (TME) of TNBC is associated with induction of metastasis, immune system suppression, escaping immune detection and drug resistance. TME is highly complex and heterogeneous, consists of tumor cells, stromal cells and immune cells. The rapid expansion of tumors induce hypoxia, which concerns the reprogramming of TME components. The reciprocal communication of tumor cells and TME cells predisposes cancer cells to metastasis by modulation of developmental pathways, Wnt, notch, hedgehog and their related mechanisms in TME. Dietary phytochemicals are non-toxic and associated with various human health benefits and remarkable spectrum of biological activities. The phytochemicals serve as vital resources for drug discovery and also as a source for breast cancer therapy. The novel properties of dietary phytochemicals propose platform for modulation of tumor signaling, overcoming drug resistance, and targeting TME. Therefore, TME could serve as promising target for the treatment of TNBC. This review presents current status and implications of experimentally evaluated therapeutic phytochemicals as potential targeting agents of TME, potential nanosystems for targeted delivery of phytochemicals and their current challenges and future implications in TNBC treatment. The dietary phytochemicals especially curcumin with significant delivery system could prevent TNBC development as it is considered safe and well tolerated in phase II clinical trials.
Collapse
Affiliation(s)
- Rama Rao Malla
- Cancer Biology Lab, Department of Biochemistry and Bioinformatics, Institute of Science, GITAM (Deemed to be University), Visakhapatnam, 530045, India.
| | - Kgk Deepak
- Cancer Biology Lab, Department of Biochemistry and Bioinformatics, Institute of Science, GITAM (Deemed to be University), Visakhapatnam, 530045, India
| | - Neha Merchant
- Department of Hematology and Medical Oncology, Winship Cancer Institute, Emory University, Atlanta, GA, 30322, USA
| | - Venkata Ramesh Dasari
- Department of Molecular and Functional Genomics, Geisinger Clinic, 100 Academy Ave, Danville, PA, 17822, USA
| |
Collapse
|
41
|
First phytochemical study and biological activity of the leaves ethanolic extract from Cissus spinosa Cambess. SCIENTIA MEDICA 2020. [DOI: 10.15448/1980-6108.2020.1.34860] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
Abstract
AIMS: The objective of this study was to identify the phytochemical profile and to evaluate the biological effects of the crude ethanolic extract (EE) and the ethanolic fraction (EF) of leaves of the species Cissus spinosa Cambess, after oxidative stress induced by cyclophosphamide (CP) in mice.METHODS: Phytochemical profile was performed detecting functional groups and, analysis of total flavonoids and phenols concentration, as well as the antiradical activity in EE and EF. The phytochemical characterization was done for the identification of flavonoids present in the leaves of the plant. In the biochemical tests, hematological parameters, glucose and total cholesterol dosages in plasma, enzymatic and non-enzymatic antioxidants and lipid damage marker were evaluated in different tissues (liver, kidney and heart), besides genotoxic and immunological analyzes. The animals received 15 days of treatment, via gavage, with EE (50 mg kg-1) or EF (50 mg kg-1) and on the 15th day, an intraperitoneal injection of CP (100 mg kg-1) or saline (0.9%). After 24 h the last treatment, the animals were anesthetized for blood withdrawal, sacrificed and removal of the organs.RESULTS: In the phytochemical analyzes, the presence of alkaloids, flavonoids and phenols was identified, the latter presented a higher concentration for EF. Eight flavonoids were identified - Rutin, Quercetin-3-β-D-glucoside, Quercitrin, Taxifolin, Quercetin, Canferol, Luteolin and Apigenin. In the biochemical analyzes, in general, EE showed a better antioxidant action against oxidative damages, hypoglycemic and antitilipemic action when comparing with EF, probably due to the synergism caused by flavonoids. It was observed the reduction and an increase of micronucleated polychromatic erythrocytes, due to the action of antioxidant compounds and alkaloids present in the plant, also considering the question of the seasonal period that directly interferes in the production of these compounds. In the immunological analysis, the extracts did not stimulate the spontaneous production of oxygen peroxide (H2O2) and nitric oxide (NO•). CONCLUSIONS: Other studies, such as the variation of the chemical composition of the plant by local seasonality, hypoglycemic and antilipemic action, should be carried out to better delineate the biological action present in this plant.
Collapse
|
42
|
Won YS, Seo KI. Lupiwighteone induces caspase-dependent and -independent apoptosis on human breast cancer cells via inhibiting PI3K/Akt/mTOR pathway. Food Chem Toxicol 2019; 135:110863. [PMID: 31604113 DOI: 10.1016/j.fct.2019.110863] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2019] [Revised: 09/04/2019] [Accepted: 09/27/2019] [Indexed: 12/25/2022]
Abstract
Breast cancer is one of the most common causes of mortality in women. Lupiwighteone has anticancer effects in prostate cancer cells and neuroblastoma cells. However, the molecular and cellular mechanisms of lupiwighteone effects on human breast cancer cells are not as well known. In the present study, we investigated the effects of lupiwighteone on the proliferation and apoptosis of two different human cancer cells; MCF-7, an estrogen receptor (ER)-positive human breast cancer cell, and MDA-MB-231, a triple negative human breast cancer cell. Lupiwighteone treatment decreased the viability of MCF-7 and MDA-MB-231 cells. Lupiwighteone treatment resulted in apoptotic cell death in breast cancer cells, which was characterized by DNA fragmentation, accumulation of apoptotic cells, and nuclear condensation. We also showed that treatment with lupiwighteone induced caspase-dependent apoptosis (up-regulation of caspase-3, -7, -8, -9, PARP, and Bax or down-regulation of Bid, Bcl-2), induction of caspase-independent apoptosis (up-regulation of AIF and Endo G on cytosol), and inhibition of the PI3K/Akt/mTOR signaling pathway (down-regulation of PI3K, p-Akt, and p-mTOR) in both MCF-7 and MDA-MB-231 cells. These results suggest that lupiwighteone induces caspase-dependent and -independent apoptosis in both breast cancer cell lines via inhibiting PI3K/Akt/mTOR pathway.
Collapse
Affiliation(s)
- Yeong-Seon Won
- Department of Biotechnology, Dong-A University, Busan, 49315, Republic of Korea
| | - Kwon-Il Seo
- Department of Biotechnology, Dong-A University, Busan, 49315, Republic of Korea.
| |
Collapse
|
43
|
Chen Y, Chen WN, Hu N, Banwell MG, Ma C, Gardiner MG, Lan P. Cytotoxicity and Anti-inflammatory Properties of Apigenin-Derived Isolaxifolin. JOURNAL OF NATURAL PRODUCTS 2019; 82:2451-2459. [PMID: 31465218 DOI: 10.1021/acs.jnatprod.9b00113] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
The rare flavonoid isolaxifolin, a potent insecticide, has been touted as a potential grain-protecting agent. In order to assess any impact of this natural product on human health and to explore its various other biological properties, we have established a semisynthesis from the simpler but structurally related and more abundant natural product apigenin. The five-step reaction sequence has provided, for the first time, sufficient material for an in-depth evaluation of the cytotoxic properties of the title natural product. The impact of isolaxifolin on certain pro-inflammatory cytokines in murine macrophage RAW 264.7 cells has also been examined. Such studies have revealed that isolaxifolin displays no toxic effects toward normal cells while displaying greater cytotoxicities against certain cancer cell lines than its synthetic precursor apigenin. Furthermore, unlike apigenin, isolaxifolin only reduced NO, TNF-α, and IL-6 secretions in LPS-induced RAW 264.7 cells in a rather modest and dose-independent manner.
Collapse
Affiliation(s)
- Yongsheng Chen
- Institute for Advanced and Applied Chemical Synthesis , Jinan University , Guangzhou , 510632 , People's Republic of China
- Department of Food Science and Engineering , Jinan University , Guangzhou , 510632 , People's Republic of China
| | - Wan-Na Chen
- College of Pharmacy , Jinan University , Guangzhou , 510632 , People's Republic of China
| | - Nan Hu
- Institute for Advanced and Applied Chemical Synthesis , Jinan University , Guangzhou , 510632 , People's Republic of China
| | - Martin G Banwell
- Institute for Advanced and Applied Chemical Synthesis , Jinan University , Guangzhou , 510632 , People's Republic of China
- Research School of Chemistry, Institute of Advanced Studies , The Australian National University , Canberra , Australian Capital Territory 2601 , Australia
| | - Chenxi Ma
- Research School of Chemistry, Institute of Advanced Studies , The Australian National University , Canberra , Australian Capital Territory 2601 , Australia
| | - Michael G Gardiner
- Research School of Chemistry, Institute of Advanced Studies , The Australian National University , Canberra , Australian Capital Territory 2601 , Australia
| | - Ping Lan
- Institute for Advanced and Applied Chemical Synthesis , Jinan University , Guangzhou , 510632 , People's Republic of China
- College of Pharmacy , Jinan University , Guangzhou , 510632 , People's Republic of China
| |
Collapse
|
44
|
Korga A, Ostrowska M, Jozefczyk A, Iwan M, Wojcik R, Zgorka G, Herbet M, Vilarrubla GG, Dudka J. Apigenin and hesperidin augment the toxic effect of doxorubicin against HepG2 cells. BMC Pharmacol Toxicol 2019; 20:22. [PMID: 31053173 PMCID: PMC6499973 DOI: 10.1186/s40360-019-0301-2] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2018] [Accepted: 04/11/2019] [Indexed: 01/22/2023] Open
Abstract
BACKGROUND Hepatocellular carcinoma (HCC) is one of the most common malignancies, with an increasing incidence. Despite the fact that systematic chemotherapy with a doxorubicin provides only marginal improvements in survival of the HCC patients, the doxorubicin is being used in transarterial therapies or combined with the target drug - sorafenib. The aim of the study was to evaluate the effect of natural flavonoids on the cytotoxicity of the doxorubicin against human hepatocellular carcinoma cell line HepG2. METHODS The effect of apigenin and its glycosides - cosmosiin, rhoifolin; baicalein and its glycosides - baicalin as well as hesperetin and its glycosides - hesperidin on glycolytic genes expression of HepG2 cell line, morphology and cells' viability at the presence of doxorubicin have been tested. In an attempt to elucidate the mechanism of observed results, the fluorogenic probe for reactive oxygen species (ROS), the DNA oxidative damage, the lipid peroxidation and the double strand breaks were evaluated. To assess impact on the glycolysis pathway, the mRNA expression for a hexokinase 2 (HK2) and a lactate dehydrogenase A (LDHA) enzymes were measured. The results were analysed statistically with the one-way analysis of variance (ANOVA) and post hoc multiple comparisons. RESULTS The apigenin and the hesperidin revealed the strongest effect on the toxicity of doxorubicin. Both flavonoids simultaneously changed the expression of the glycolytic pathway genes - HK2 and LDHA, which play a key role in the Warburg effect. Although separate treatment with doxorubicin, apigenin and hesperidin led to a significant oxidative DNA damage and double strand breaks, simultaneous administration of doxorubicin and apigenin or hesperidin abolished these damage with the simultaneous increase in the doxorubicin toxicity. CONCLUSION The obtained results indicate the existence of a very effective cytotoxic mechanism in the HepG2 cells of the combined effect of doxorubicin and apigenin (or hesperidin), not related to the oxidative stress. To explain this synergy mechanism, further research is needed, The observed intensification of the cytotoxic effect of doxorubicin by this flavonoids may be a promising direction of the research on the therapy of hepatocellular carcinoma, especially in a chemoembolization.
Collapse
Affiliation(s)
- Agnieszka Korga
- Independent Medical Biology Unit, Medical University of Lublin, 8b Jaczewski Street, 20-090 Lublin, Poland
| | - Marta Ostrowska
- Department of Toxicology, Medical University of Lublin, 8b Jaczewski Street, 20-090 Lublin, Poland
| | - Aleksandra Jozefczyk
- Department of Pharmacognosy with Medicinal Plant Laboratory, Medical University of Lublin, 1 Chodzko Street, 20-093 Lublin, Poland
| | - Magdalena Iwan
- Independent Medical Biology Unit, Medical University of Lublin, 8b Jaczewski Street, 20-090 Lublin, Poland
| | - Rafal Wojcik
- Department of Human Anatomy, Medical University of Lublin, 4 Jaczewski Street, 20-090 Lublin, Poland
| | - Grazyna Zgorka
- Department of Pharmacognosy with Medicinal Plant Laboratory, Medical University of Lublin, 1 Chodzko Street, 20-093 Lublin, Poland
| | - Mariola Herbet
- Department of Toxicology, Medical University of Lublin, 8b Jaczewski Street, 20-090 Lublin, Poland
| | - Gemma Gomez Vilarrubla
- Independent Medical Biology Unit, Medical University of Lublin, 8b Jaczewski Street, 20-090 Lublin, Poland
| | - Jaroslaw Dudka
- Department of Toxicology, Medical University of Lublin, 8b Jaczewski Street, 20-090 Lublin, Poland
| |
Collapse
|
45
|
Sudhakaran M, Sardesai S, Doseff AI. Flavonoids: New Frontier for Immuno-Regulation and Breast Cancer Control. Antioxidants (Basel) 2019; 8:E103. [PMID: 30995775 PMCID: PMC6523469 DOI: 10.3390/antiox8040103] [Citation(s) in RCA: 56] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2019] [Revised: 04/01/2019] [Accepted: 04/05/2019] [Indexed: 12/15/2022] Open
Abstract
Breast cancer (BC) remains the second most common cause of cancer-related deaths in women in the US, despite advances in detection and treatment. In addition, breast cancer survivors often struggle with long-term treatment related comorbidities. Identifying novel therapies that are effective while minimizing toxicity is critical in curtailing this disease. Flavonoids, a subclass of plant polyphenols, are emerging as promising treatment options for the prevention and treatment of breast cancer. Recent evidence suggests that in addition to anti-oxidant properties, flavonoids can directly interact with proteins, making them ideal small molecules for the modulation of enzymes, transcription factors and cell surface receptors. Of particular interest is the ability of flavonoids to modulate the tumor associated macrophage function. However, clinical applications of flavonoids in cancer trials are limited. Epidemiological and smaller clinical studies have been largely hypothesis generating. Future research should aim at addressing known challenges with a broader use of preclinical models and investigating enhanced dose-delivery systems that can overcome limited bioavailability of dietary flavonoids. In this review, we discuss the structure-functional impact of flavonoids and their action on breast tumor cells and the tumor microenvironment, with an emphasis on their clinical role in the prevention and treatment of breast cancer.
Collapse
Affiliation(s)
- Meenakshi Sudhakaran
- Department Physiology, Michigan State University, East Lansing, MI 48824, USA.
- Physiology Graduate Program, Michigan State University, East Lansing, MI 48824, USA.
| | - Sagar Sardesai
- Division of Hematology, Department of Internal Medicine, The Ohio State University, Columbus, OH 43210, USA.
| | - Andrea I Doseff
- Department Physiology, Michigan State University, East Lansing, MI 48824, USA.
- Department Pharmacology and Toxicology, Michigan State University, East Lansing, MI 48824, USA.
| |
Collapse
|
46
|
Licochalcone A Inhibits Cellular Motility by Suppressing E-cadherin and MAPK Signaling in Breast Cancer. Cells 2019; 8:cells8030218. [PMID: 30841634 PMCID: PMC6468539 DOI: 10.3390/cells8030218] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2019] [Revised: 02/16/2019] [Accepted: 03/01/2019] [Indexed: 02/06/2023] Open
Abstract
A compound isolated from Glycyrrhiza uralensis, licochalcone A (LA) exhibits anti-inflammatory and anti-tumor properties in various cell lines. LA has been found to promote autophagy and suppress specificity protein 1, inducing apoptosis in breast cancer cells. However, the regulation of breast cancer cell invasion and migration by LA is elusive. Thus, the present study investigated whether LA induces apoptosis and cellular motility in MDA-MB-231 breast cells, and investigated the underlying molecular mechanisms. MDA-MB-231 cells treated with LA and cell viability measured by cell counting kit-8 assay. Apoptotic signal proteins checked by flow cytometry, fluorescent staining, and Western blot. LA effectively suppressed cell migration, and modulated E-cadherin and vimentin expression by blocking MAPK and AKT signaling. LA inhibited cell proliferation and cell cycle, modulated mitochondrial membrane potential and DNA damage, and reduced oxidative stress in MDA-MB-231 cells. LA also activated cleaved-caspase 3 and 9, significantly decreased Bcl-2 expression, ultimately causing the release of cytochrome c from the mitochondria into the cytoplasm. Overall, our findings suggest that LA decreases cell proliferation and increases reactive oxygen species production for induced apoptosis, and regulates E-cadherin and vimentin by reducing MAPK and AKT signaling, resulting in suppressed MDA-MB-231 cell migration and invasion.
Collapse
|
47
|
Evaluation of the Antioxidant and Wound-Healing Properties of Extracts from Different Parts of Hylocereus polyrhizus. AGRONOMY-BASEL 2019. [DOI: 10.3390/agronomy9010027] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Hylocereus polyrhizus cultivation started in Taiwan around the 1980s. The pulp of the fruit is edible and contains small, black, and soft seeds. The peel of the fruits are covered with bracts. The H. polyrhizus fruit is known to be rich in nutrients and minerals. To evaluate the potential applications of the agricultural wastes of H. polyrhizus, the stem, peel, and flower of H. polyrhizus were extracted with solutions of ethanol and water mixed in different ratios. Data was collected for the H. polyrhizus extract including the yield of total phenolics, the total flavonoids, and antioxidant activity, as determined by the 2-2-diphenyl-1-picrylhydrazyl (DPPH) and 2,2′-azino-bis (3-ethylbenzothiazoline-6-sulphonic acid) (ABTS) radical scavenging assay. The protective effects of H. polyrhizus extract on DNA was investigated using an assay with the pUC19 plasmid. The cell proliferation and migration effects were evaluated in the NIH-3T3 fibroblast cell line. The greatest yield of extract from the stem of H. polyrhizus was 44.70 ± 1.77% which was obtained using 50% aqueous ethanol and the greatest yield of extract from the peel was 43.47% using distilled water. The stem extract, which was prepared with 95% aqueous ethanol, had the highest composition of phenolics and flavonoids as well as the best DPPH radical scavenging activity. The stem extract had excellent ABTS radical scavenging activity as well. The stem, peel, and flower extracts, which were prepared using 95% aqueous ethanol, showed excellent results in protecting themselves from DNA damage, similar to the effect of 0.3 mg/mL ferulic acid. None of the extracts were able to promote cell proliferation at concentrations of 250 μg/mL to 2,000 μg/mL in a 24 h period. The 1000 μg/mL stem and flower extracts in 95% aqueous ethanol promoted considerable cell migration after a 24 h period.
Collapse
|
48
|
Apigenin Protects the Brain against Ischemia/Reperfusion Injury via Caveolin-1/VEGF In Vitro and In Vivo. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2018; 2018:7017204. [PMID: 30622670 PMCID: PMC6304859 DOI: 10.1155/2018/7017204] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/31/2018] [Accepted: 10/09/2018] [Indexed: 12/17/2022]
Abstract
Apigenin is a natural flavonoid found in several dietary plant foods as vegetables and fruits. To investigate potential anti-ischemia/reperfusion injury properties of apigenin in vitro, cell proliferation assay, tube formation, cell migration, apoptosis, and autophagy were performed in human brain microvascular endothelial cells (HBMVECs) after oxygen-glucose deprivation/reoxygenation (OGD/R). The effect of apigenin was also explored in rats after middle cerebral artery occlusion/reperfusion (MCAO/R) via neurobehavioral scores, pathological examination, and measurement of markers involved in ischemia/reperfusion injury. Data in vitro indicated that apigenin could prompt cell proliferation, tube formation, and cell migration while inhibiting apoptosis and autophagy by affecting Caveolin-1/VEGF, Bcl-2, Caspase-3, Beclin-1, and mTOR expression. Results in vivo showed that apigenin significantly reduced neurobehavioral scores and volume of cerebral infarction while prompting vascular endothelial cell proliferation by upregulating VEGFR2/CD34 double-labeling endothelial progenitor cell (EPC) number and affecting Caveolin-1, VEGF, and eNOS expression in brain tissue of MCAO/R rats. All the data suggested that apigenin may be protective for the brain against ischemia/reperfusion injury by alleviating apoptosis and autophagy, promoting cell proliferation in HBMVECs of OGD/R, and attenuating brain damage and improved neurological function in rats of MCAO/R through the Caveolin-1/VEGF pathway.
Collapse
|
49
|
Cucurbitacin B induces mitochondrial-mediated apoptosis pathway in cholangiocarcinoma cells via suppressing focal adhesion kinase signaling. Naunyn Schmiedebergs Arch Pharmacol 2018; 392:271-278. [DOI: 10.1007/s00210-018-1584-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2018] [Accepted: 11/12/2018] [Indexed: 01/08/2023]
|
50
|
Chen YT, Xie JY, Sun Q, Mo WJ. Novel drug candidates for treating esophageal carcinoma: A study on differentially expressed genes, using connectivity mapping and molecular docking. Int J Oncol 2018; 54:152-166. [PMID: 30387840 PMCID: PMC6254996 DOI: 10.3892/ijo.2018.4618] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2018] [Accepted: 10/23/2018] [Indexed: 12/14/2022] Open
Abstract
Patients with esophageal carcinoma (ESCA) have a poor prognosis and high mortality rate. Although standard therapies have had effect, there is an urgent requirement to develop novel options, as increasing drug tolerance has been identified in clinical practice. In the present study, differentially expressed genes (DEGs) of ESCA were identified in The Cancer Genome Atlas and Genotype-Tissue Expression databases. Functional and protein-protein interaction (PPI) analyses were performed. The Connectivity Map (CMAP) was selected to predict drugs for the treatment of ESCA, and their target genes were acquired from the Search Tool for Interactions of Chemicals (STITCH) by uploading the Simplified Molecular-Input Line-Entry System structure. Additionally, significant target genes and ESCA-associated hub genes were extracted using another PPI analysis, and the corresponding drugs were added to construct a network. Furthermore, the binding affinity between predicted drug candidates and ESCA-associated hub genes was calculated using molecular docking. Finally, 827 DEGs (|log2 fold-change|≥2; q-value <0.05), which are principally involved in protein digestion and absorption (P<0.005), the plasminogen-activating cascade (P<0.01), as well as the ‘biological regulation’ of the Biological Process, ‘membrane’ of the Cellular Component and ‘protein binding’ of the Molecular Function categories, were obtained. Additionally, 11 hub genes were obtained from the PPI network (all degrees ≥30). Furthermore, the 15 first screen drugs were extracted from CMAP (score <−0.85) and the 9 second screen drugs with 70 target genes were extracted from STITCH. Furthermore, another PPI analysis extracted 51 genes, and apigenin, baclofen, Prestwick-685, menadione, butyl hydroxybenzoate, gliclazide and valproate were selected as drug candidates for ESCA. Those molecular docking results with a docking score of >5.52 indicated the significance of apigenin, Prestwick-685 and menadione. The results of the present study may lead to novel drug candidates for ESCA, among which Prestwick-685 and menadione were identified to be significant new drug candidates.
Collapse
Affiliation(s)
- Yu-Ting Chen
- Department of Pathology, First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi 530021, P.R. China
| | - Jia-Yi Xie
- Department of Pathology, First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi 530021, P.R. China
| | - Qi Sun
- Department of Pathology, First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi 530021, P.R. China
| | - Wei-Jia Mo
- Department of Pathology, First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi 530021, P.R. China
| |
Collapse
|