1
|
Figueira MI, Carvalho TMA, Macário-Monteiro J, Cardoso HJ, Correia S, Vaz CV, Duarte AP, Socorro S. The Pros and Cons of Estrogens in Prostate Cancer: An Update with a Focus on Phytoestrogens. Biomedicines 2024; 12:1636. [PMID: 39200101 PMCID: PMC11351860 DOI: 10.3390/biomedicines12081636] [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: 06/26/2024] [Revised: 07/14/2024] [Accepted: 07/20/2024] [Indexed: 09/01/2024] Open
Abstract
The role of estrogens in prostate cancer (PCa) is shrouded in mystery, with its actions going from angelic to devilish. The findings by Huggins and Hodges establishing PCa as a hormone-sensitive cancer have provided the basis for using estrogens in therapy. However, despite the clinical efficacy in suppressing tumor growth and the panoply of experimental evidence describing its anticarcinogenic effects, estrogens were abolished from PCa treatment because of the adverse secondary effects. Notwithstanding, research work over the years has continued investigating the effects of estrogens, reporting their pros and cons in prostate carcinogenesis. In contrast with the beneficial therapeutic effects, many reports have implicated estrogens in the disruption of prostate cell fate and tissue homeostasis. On the other hand, epidemiological data demonstrating the lower incidence of PCa in Eastern countries associated with a higher consumption of phytoestrogens support the beneficial role of estrogens in counteracting cancer development. Many studies have investigated the effects of phytoestrogens and the underlying mechanisms of action, which may contribute to developing safe estrogen-based anti-PCa therapies. This review compiles the existing data on the anti- and protumorigenic actions of estrogens and summarizes the anticancer effects of several phytoestrogens, highlighting their promising features in PCa treatment.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Sílvia Socorro
- CICS-UBI, Centro de Investigação em Ciências da Saúde, Universidade da Beira Interior, 6200-506 Covilhã, Portugal; (M.I.F.)
| |
Collapse
|
2
|
Sohel M. Comprehensive exploration of Biochanin A as an oncotherapeutics potential in the treatment of multivarious cancers with molecular insights. Phytother Res 2024; 38:489-506. [PMID: 37905329 DOI: 10.1002/ptr.8050] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2023] [Revised: 09/22/2023] [Accepted: 10/01/2023] [Indexed: 11/02/2023]
Abstract
Cancer is considered a leading cause of mortality. This rising cancer death rate and several existing limitations like side effects, poor efficacies, and high cost of the present chemotherapeutic agents have increased the demand for more potent and alternative cancer treatments. This review elucidated a brief overview of Biochanin A (BCA) and its potentiality on various cancers with details of anticancer mechanism. According to our review, a number of studies including in silico, in vitro, pre-clinical, and clinical trials have tested to evaluate the efficacy of BCA. This compound is effective against 15 types of cancer, including breast, cervical, colorectal, gastric, glioblastoma, liver, lung, melanoma, oral, osteosarcoma, ovarian, pancreatic, pharynx, prostate, and umbilical vein cancer. The general anticancer activities of this compound are mediated via several molecular processes, including regulation of apoptosis, cell proliferation, metastasis and angiogenesis, signaling, enzymatic pathways, and other mechanisms. Targeting both therapeutic and oncogenic proteins, as well as different pathways, makes up the molecular mechanism underlying the anticancer action. Many signaling networks and their components, such as EFGR, PI3K/Akt/mTOR, MAPK, MMP-2, MMP-9, PARP, Caspase-3/8/9, Bax, Bcl2, PDL-1, NF-κB, TNF-α, IL-6, JAK, STAT3, VEGFR, VEGF, c-MY, Cyclin B1, D1, E1 and CDKs, Snail, and E-cadherin proteins, can be regulated in cancer cells by BCA. Such kind of anticancer properties of BCA could be a result of its correct structural chemistry. The use of BCA-based therapies as nano-carriers for the delivery of chemotherapeutic medicines has the potential to be very effective. This natural compound synergises with other natural compounds and standard drugs, including sorafenib, 5-fluorouracil, temozolomide, doxorubicin, apigenin, and genistein. Moreover, proper use of this compound can reverse multidrug resistance through numerous mechanisms. BCA has better drug-likeness and pharmacokinetic properties and is nontoxic (eye, liver, kidney, skin, cardio) in human bodies. As having a wide range of cancer-fighting mechanisms, synergistic effects, and good pharmacokinetic properties, BCA can be used as a supplementary food until standard drugs are available at pharma markets.
Collapse
Affiliation(s)
- Md Sohel
- Department of Biochemistry and Molecular Biology, Primeasia University, Dhaka, Bangladesh
- Biochemistry and Molecular Biology, Mawlana Bhashani Science and Technology University, Tangail, Bangladesh
| |
Collapse
|
3
|
Anuranjana P, Beegum F, K.P D, George KT, Viswanatha G, Nayak PG, Kanwal A, Kishore A, Shenoy RR, Nandakumar K. Mechanisms Behind the Pharmacological Application of Biochanin-A: A review. F1000Res 2023; 12:107. [PMID: 38106650 PMCID: PMC10725524 DOI: 10.12688/f1000research.126059.3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 11/30/2023] [Indexed: 12/19/2023] Open
Abstract
This review was aimed at summarizing the cellular and molecular mechanisms behind the various pharmacological actions of biochanin-A. Many studies have been reported claiming its application in cancers, metabolic disorders, airway hyperresponsiveness, cardiac disorders, neurological disorders, etc. With regard to hormone-dependent cancers like breast, prostate, and other malignancies like pancreatic, colon, lung, osteosarcoma, glioma that has limited treatment options, biochanin-A revealed agreeable results in arresting cancer development. Biochanin-A has also shown therapeutic benefits when administered for neurological disorders, diabetes, hyperlipidaemia, and other chronic diseases/disorders. Isoflavones are considered phenomenal due to their high efficiency in modifying the physiological functions of the human body. Biochanin-A is one among the prominent isoflavones found in soy (glycine max), red clover (Trifolium pratense), and alfalfa sprouts, etc., with proven potency in modulating vital cellular mechanisms in various diseases. It has been popular for ages among menopausal women in controlling symptoms. In view of the multi-targeted functions of biochanin-A, it is essential to summarize it's mechanism of action in various disorders. The safety and efficacy of biochanin-A needs to be established in clinical trials involving human subjects. Biochanin-A might be able to modify various systems of the human body like the cardiovascular system, CNS, respiratory system, etc. It has shown a remarkable effect on hormonal cancers and other cancers. Many types of research on biochanin-A, particularly in breast, lung, colon, prostate, and pancreatic cancers, have shown a positive impact. Through modulating oxidative stress, SIRT-1 expression, PPAR gamma receptors, and other multiple mechanisms biochanin-A produces anti-diabetic action. The diverse molecular mechanistic pathways involved in the pharmacological ability of biochanin-A indicate that it is a very promising molecule and can play a major impact in modifying several physiological functions.
Collapse
Affiliation(s)
- P.V. Anuranjana
- Department of Pharmacology, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, Karnataka, 576104, India
| | - Fathima Beegum
- Department of Pharmacology, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, Karnataka, 576104, India
| | - Divya K.P
- Department of Pharmacology, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, Karnataka, 576104, India
| | - Krupa Thankam George
- Department of Pharmacology, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, Karnataka, 576104, India
| | | | - Pawan G. Nayak
- Department of Pharmacology, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, Karnataka, 576104, India
| | - Abhinav Kanwal
- Department of Pharmacology, All India Institute of Medical Sciences, Bathinda, Punjab, India
| | - Anoop Kishore
- Department of Pharmacology, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, Karnataka, 576104, India
| | - Rekha R. Shenoy
- Department of Pharmacology, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, Karnataka, 576104, India
| | - K. Nandakumar
- Department of Pharmacology, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, Karnataka, 576104, India
| |
Collapse
|
4
|
Luiz-Ferreira A, Pacifico T, Cruz ÁC, Laudisi F, Monteleone G, Stolfi C. TRAIL-Sensitizing Effects of Flavonoids in Cancer. Int J Mol Sci 2023; 24:16596. [PMID: 38068921 PMCID: PMC10706592 DOI: 10.3390/ijms242316596] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2023] [Revised: 11/16/2023] [Accepted: 11/20/2023] [Indexed: 12/18/2023] Open
Abstract
Tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) represents a promising anticancer agent, as it selectively induces apoptosis in transformed cells without altering the cellular machinery of healthy cells. Unfortunately, the presence of TRAIL resistance mechanisms in a variety of cancer types represents a major hurdle, thus limiting the use of TRAIL as a single agent. Accumulating studies have shown that TRAIL-mediated apoptosis can be facilitated in resistant tumors by combined treatment with antitumor agents, ranging from synthetic molecules to natural products. Among the latter, flavonoids, the most prevalent polyphenols in plants, have shown remarkable competence in improving TRAIL-driven apoptosis in resistant cell lines as well as tumor-bearing mice with minimal side effects. Here, we summarize the molecular mechanisms, such as the upregulation of death receptor (DR)4 and DR5 and downregulation of key anti-apoptotic proteins [e.g., cellular FLICE-inhibitory protein (c-FLIP), X-linked inhibitor of apoptosis protein (XIAP), survivin], underlying the TRAIL-sensitizing properties of different classes of flavonoids (e.g., flavones, flavonols, isoflavones, chalcones, prenylflavonoids). Finally, we discuss limitations, mainly related to bioavailability issues, and future perspectives regarding the clinical use of flavonoids as adjuvant agents in TRAIL-based therapies.
Collapse
Affiliation(s)
- Anderson Luiz-Ferreira
- Inflammatory Bowel Disease Research Laboratory, Department of Biological Sciences, Institute of Biotechnology, Federal University of Catalão (UFCAT), Catalão 75704020, GO, Brazil;
| | - Teresa Pacifico
- Department of Systems Medicine, University of Rome “Tor Vergata”, 00133 Rome, Italy; (T.P.); (F.L.); (G.M.)
| | - Álefe Cardoso Cruz
- Inflammatory Bowel Disease Research Laboratory, Department of Biological Sciences, Institute of Biotechnology, Federal University of Catalão (UFCAT), Catalão 75704020, GO, Brazil;
| | - Federica Laudisi
- Department of Systems Medicine, University of Rome “Tor Vergata”, 00133 Rome, Italy; (T.P.); (F.L.); (G.M.)
| | - Giovanni Monteleone
- Department of Systems Medicine, University of Rome “Tor Vergata”, 00133 Rome, Italy; (T.P.); (F.L.); (G.M.)
| | - Carmine Stolfi
- Department of Systems Medicine, University of Rome “Tor Vergata”, 00133 Rome, Italy; (T.P.); (F.L.); (G.M.)
| |
Collapse
|
5
|
Ubaid M, Salauddin, Shadani MA, Kawish SM, Albratty M, Makeen HA, Alhazmi HA, Najmi A, Zoghebi K, Halawi MA, Ali A, Alam MS, Iqbal Z, Mirza MA. Daidzein from Dietary Supplement to a Drug Candidate: An Evaluation of Potential. ACS OMEGA 2023; 8:32271-32293. [PMID: 37780202 PMCID: PMC10538961 DOI: 10.1021/acsomega.3c03741] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/28/2023] [Accepted: 07/18/2023] [Indexed: 10/03/2023]
Abstract
Daidzein (DDZ) is a well-known nutraceutical supplement belonging to the class of isoflavones. It is isolated from various sources such as alfalfa, soybean, and red clover. It demonstrates a broad array of pharmacological/beneficial properties such as cardiovascular exercise, cholesterol reduction, and anticancer, antifibrotic, and antidiabetic effects, which make it effective in treating a wide range of diseases. Its structure and operation are the same as those of human estrogens, which are important in preventing osteoporosis, cancer, and postmenopausal diseases. It is thus a promising candidate for development as a phytopharmaceutical. Addressing safety, efficacy, and physicochemical properties are the primary prerequisites. DDZ is already ingested every day in varying amounts, so there should not be a significant safety risk; however, each indication requires a different dose to be determined. Some clinical trials are already being conducted globally to confirm its safety, efficacy, and therapeutic potential. Furthermore, as a result of its therapeutic influence on health, in order to establish intellectual property, patents are utilized. In light of the vast potential of eugenol, this review presents a detailed data collection on DDZ to substantiate the claim to develop it in the therapeutic category.
Collapse
Affiliation(s)
- Mohammed Ubaid
- School
of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi 110062, India
| | - Salauddin
- School
of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi 110062, India
| | - Md Andalib Shadani
- School
of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi 110062, India
| | - S. M. Kawish
- School
of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi 110062, India
| | - Mohammed Albratty
- Department
of Pharmaceutical Chemistry, College of Pharmacy, Jazan University, Jazan 45142, Saudi Arabia
| | - Hafiz A. Makeen
- Pharmacy
Practice Research Unit, Department of Clinical Pharmacy, College of
Pharmacy, Jazan University, Jazan 45142, Saudi Arabia
| | - Hassan A. Alhazmi
- Department
of Pharmaceutical Chemistry, College of Pharmacy, Jazan University, Jazan 45142, Saudi Arabia
- Substance
Abuse and Toxicology Research Center, Jazan University, Jazan 45142, Saudi Arabia
- Medical
Research Center, Jazan University, Jazan 45142, Saudi Arabia
| | - Asim Najmi
- Department
of Pharmaceutical Chemistry, College of Pharmacy, Jazan University, Jazan 45142, Saudi Arabia
| | - Khalid Zoghebi
- Department
of Pharmaceutical Chemistry, College of Pharmacy, Jazan University, Jazan 45142, Saudi Arabia
| | - Maryam A. Halawi
- Pharmacy
Practice, College of Pharmacy, Jazan University, Jazan 45142, Saudi Arabia
- Department
of Haematology, Division of Cancer & Genetics School of Medicine, Cardiff University, Cardiff, Wales CF14 4XN, U.K.
| | - Abuzer Ali
- Department
of Pharmacognosy, College of Pharmacy, Taif
University, P.O. Box 11099, Taif, 21944, Saudi Arabia
| | - Md Shamsher Alam
- Department
of Pharmaceutical Chemistry, College of Pharmacy, Jazan University, Jazan 45142, Saudi Arabia
| | - Zeenat Iqbal
- Department
of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi 110062, India
| | - Mohd. Aamir Mirza
- Department
of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi 110062, India
| |
Collapse
|
6
|
Li P, Li M, Lou X, Zhao B, Ma Q, Bian Y, Mi X. Evaluation of Hypoglycemic Activity and Sub-Acute Toxicity of the Novel Biochanin A–Chromium(III) Complex. Molecules 2022; 27:molecules27185786. [PMID: 36144522 PMCID: PMC9504010 DOI: 10.3390/molecules27185786] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Revised: 08/09/2022] [Accepted: 08/31/2022] [Indexed: 11/16/2022] Open
Abstract
The novel biochanin A–chromium(III) complex was synthesized by chelating chromium with biochanin A (BCA). The structure of the complex was determined and the complex ([CrBCA3]) was composed of chromium(III) and three ligands, and the chromium content was 55 μg/mg. The hypoglycemic activity of the complex was studied in db/db mice and C57 mice. The sub-acute toxicity test of the complex was carried out by the maximum limit method in KM mice. The hypoglycemic activity showed that the complex could reduce the weight of db/db mice and lower the fasting blood glucose and random blood glucose levels. The complex also improved the organ index, oral glucose tolerance test (OGTT) and insulin tolerance test (ITT) results of db/db mice, and some of the indicators were similar to those of the positive control group after treatment with the complex. The histopathology study showed significant improvements in the liver, kidney, pancreas and skeletal muscle compared with the diabetes model group. The complex also showed a significant improvement in serum biochemical indices and antioxidant enzyme activities, as well as glycogen levels. The sub-acute toxicity study showed that the complex did not cause death or any dangerous symptoms during the study. In addition, the sub-acute toxicity study showed that the complex had no significant effect on the serum biochemical indices, antioxidant capacity and organs of normal mice. This study showed that [CrBCA3] had good hypoglycemic activity in vivo and had no sub-acute toxicity. This work provides an important reference for the development of functional hypoglycemic foods or drugs.
Collapse
Affiliation(s)
- Pengshou Li
- School of Food and Drug, Luoyang Normal University, Luoyang 471934, China
- Correspondence:
| | - Mengdan Li
- School of Food and Drug, Luoyang Normal University, Luoyang 471934, China
| | - Xinhua Lou
- School of Food and Drug, Luoyang Normal University, Luoyang 471934, China
| | - Baosheng Zhao
- Beijing Academy of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Qixiang Ma
- Cancer Institute, Fudan University Cancer Hospital and Cancer Metabolism Laboratory, Institutes of Biomedical Sciences, Fudan University, Shanghai 200032, China
| | - Yumiao Bian
- School of Food and Drug, Luoyang Normal University, Luoyang 471934, China
| | - Xiliang Mi
- School of Food and Drug, Luoyang Normal University, Luoyang 471934, China
| |
Collapse
|
7
|
Pharmacological Small Molecules against Prostate Cancer by Enhancing Function of Death Receptor 5. Pharmaceuticals (Basel) 2022; 15:ph15081029. [PMID: 36015177 PMCID: PMC9413322 DOI: 10.3390/ph15081029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2022] [Revised: 08/15/2022] [Accepted: 08/19/2022] [Indexed: 02/05/2023] Open
Abstract
Death receptor 5 (DR5) is a membrane protein that mediates exogenous apoptosis. Based on its function, it is considered to be a target for the treatment of cancers including prostate cancer. It is encouraging to note that a number of drugs targeting DR5 are now progressing to different stages of clinical trial studies. We collected 38 active compounds that could produce anti-prostate-cancer effects by modulating DR5, 28 of which were natural compounds and 10 of which were synthetic compounds. In addition, 6 clinically used chemotherapeutic agents have also been shown to promote DR5 expression and thus exert apoptosis-inducing effects in prostate cancer cells. These compounds promote the expression of DR5, thereby enhancing its function in inducing apoptosis. When these compounds were used in combination with the natural ligand of DR5, the number of apoptotic cells was significantly increased. These compounds are all promising for development as anti-prostate-cancer drugs, while most of these compounds are currently being evaluated for their anti-prostate-cancer effects at the cellular level and in animal studies. A great deal of more in-depth research is needed to evaluate whether they can be developed as drugs. We collected literature reports on small molecules against prostate cancer through modulation of DR5 to understand the current dynamics in this field and to evaluate the prospects of small molecules against prostate cancer through modulation of DR5.
Collapse
|
8
|
Sohel M, Sultana H, Sultana T, Mamun AA, Amin MN, Hossain MA, Ali MC, Aktar S, Sultana A, Rahim ZB, Mitra S, Dash R. Chemotherapeutics activities of dietary phytoestrogens against prostate cancer: From observational to clinical studies. Curr Pharm Des 2022; 28:1561-1580. [PMID: 35652403 DOI: 10.2174/1381612828666220601153426] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2022] [Accepted: 05/17/2022] [Indexed: 11/22/2022]
Abstract
Prostate cancer remains one of the most frequent and deadliest malignancies in males, where the rate of disease progression is closely associated with the type of dietary intake, specifically Western-style diet. Indeed intake of the Asian diet, which contains abundant phytoestrogens, is inversely correlated with a higher risk of prostate cancer, suggesting a chemoprotective effect of phytoestrogen against cancer progression. Although the role of phytoestrogens in cancer treatment was well documented, their impact on prostate cancer is not well understood. Therefore, the present review discusses the possible chemopreventive effect of phytoestrogens, emphasizing their efficacy at the different stages of carcinogenesis. Furthermore, phytoestrogens provide a cytoprotective effect in conventional chemotherapy and enhance chemosensitivity to tumor cells, which have also been discussed. This compilation provides a solid basis for future research on phytoestrogens as a promising avenue for anticancer drug development and also recommends these beneficiary compounds in the daily diet to manage and prevent prostate cancer.
Collapse
Affiliation(s)
- Md Sohel
- Department of Biochemistry and Molecular Biology, Mawlana Bhashani Science and Technology University, Santosh, Tangail-1902, Bangladesh.,Pratyasha Health Biomedical Research Center, Dhaka-1230. Bangladesh
| | - Habiba Sultana
- Department of Biotechnology and Genetic Engineering, Faculty of life science, Mawlana Bhashani Science and Technology University, Santosh, Tangail-1902, Bangladesh
| | - Tayeba Sultana
- Department of Biotechnology and Genetic Engineering, Faculty of life science, Mawlana Bhashani Science and Technology University, Santosh, Tangail-1902, Bangladesh
| | - Abdullah Al Mamun
- Department of Biochemistry and Molecular Biology, Mawlana Bhashani Science and Technology University, Santosh, Tangail-1902, Bangladesh
| | - Mohammad Nurul Amin
- Department of Pharmacy, Atish Dipankar University of Science and Technology, Dhaka-1230. Bangladesh.,Pratyasha Health Biomedical Research Center, Dhaka-1230. Bangladesh
| | - Md Arju Hossain
- Department of Biotechnology and Genetic Engineering, Faculty of life science, Mawlana Bhashani Science and Technology University, Santosh, Tangail-1902, Bangladesh
| | - Md Chayan Ali
- Department of Immunology, Genetics and Pathology, Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| | - Suraiya Aktar
- Department of Biochemistry and Molecular Biology, Rajshahi University, Rajshahi, Bangladesh
| | - Armin Sultana
- Department of Pharmacy, BGC Trust University Bangladesh, Chittagong 4381, Bangladesh
| | - Zahed Bin Rahim
- Department of Pharmacy, BGC Trust University Bangladesh, Chittagong 4381, Bangladesh
| | - Sarmistha Mitra
- Department of Anatomy, Dongguk University College of Medicine, Gyeongju 38066, Korea
| | - Raju Dash
- Department of Anatomy, Dongguk University College of Medicine, Gyeongju 38066, Korea
| |
Collapse
|
9
|
Pendulone induces apoptosis via the ROS-mediated ER-stress pathway in human non-small cell lung cancer cells. Toxicol In Vitro 2022; 81:105346. [DOI: 10.1016/j.tiv.2022.105346] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2021] [Revised: 02/09/2022] [Accepted: 03/08/2022] [Indexed: 01/04/2023]
|
10
|
Sarkar C, Chaudhary P, Jamaddar S, Janmeda P, Mondal M, Mubarak MS, Islam MT. Redox Activity of Flavonoids: Impact on Human Health, Therapeutics, and Chemical Safety. Chem Res Toxicol 2022; 35:140-162. [PMID: 35045245 DOI: 10.1021/acs.chemrestox.1c00348] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
The cost-effectiveness of presently used therapies is a problem in overall redox-based management, which is posing a significant financial burden on communities across the world. As a result, sophisticated treatment models that provide notions of predictive diagnoses followed by targeted preventive therapies adapted to individual patient profiles are gaining global acclaim as being beneficial to patients, the healthcare sector, and society as a whole. In this context, natural flavonoids were considered due to their multifaceted antioxidant, anti-inflammatory, and anticancer effects as well as their low toxicity and ease of availability. The aim of this review is to focus on the capacity of flavonoids to modulate the responsiveness of various diseases and ailments associated with redox toxicity. The review will also focus on the flavonoids' pathway-based redox activity and the advancement of redox-based therapies as well as flavonoids' antioxidant characteristics and their influence on human health, therapeutics, and chemical safety. Research findings indicated that flavonoids significantly exhibit various redox-based therapeutic responses against several diseases such as inflammatory, neurodegenerative, cardiovascular, and hepatic diseases and various types of cancer by activating the Nrf2/Keap1 transcription system, suppressing the nuclear factor κB (NF-κB)/IκB kinase inflammatory pathway, abrogating the function of the Hsp90/Hsf1 complex, inhibiting the PTEN/PI3K/Akt pathway, and preventing mitochondrial dysfunction. Some flavonoids, especially genistein, apigenin, amentoflavone, baicalein, quercetin, licochalcone A, and biochanin A, play a potential role in redox regulation. Conclusions of this review on the antioxidant aspects of flavonoids highlight the medicinal and folk values of these compounds against oxidative stress and various diseases and ailments. In short, treatment with flavonoids could be a novel therapeutic invention in clinical trials, as we hope.
Collapse
Affiliation(s)
- Chandan Sarkar
- Department of Pharmacy, Bangabandhu Sheikh Mujibur Rahman Science and Technology University, Gopalganj 8100, Bangladesh
| | - Priya Chaudhary
- Department of Bioscience and Biotechnology, Banasthali Vidyapith, Rajasthan 304022, India
| | - Sarmin Jamaddar
- Department of Pharmacy, Bangabandhu Sheikh Mujibur Rahman Science and Technology University, Gopalganj 8100, Bangladesh
| | - Pracheta Janmeda
- Department of Bioscience and Biotechnology, Banasthali Vidyapith, Rajasthan 304022, India
| | - Milon Mondal
- Department of Pharmacy, Bangabandhu Sheikh Mujibur Rahman Science and Technology University, Gopalganj 8100, Bangladesh
| | | | - Muhammad Torequl Islam
- Department of Pharmacy, Bangabandhu Sheikh Mujibur Rahman Science and Technology University, Gopalganj 8100, Bangladesh
| |
Collapse
|
11
|
Ramachandran V, V IK, Hr KK, Tiwari R, Tiwari G. Biochanin-A: A Bioactive Natural Product with Versatile Therapeutic Perspectives. Curr Drug Res Rev 2022; 14:225-238. [PMID: 35579127 DOI: 10.2174/2589977514666220509201804] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2021] [Revised: 02/28/2022] [Accepted: 03/08/2022] [Indexed: 06/15/2023]
Abstract
BACKGROUND Biochanin-A (5,7 dihydroxy 4 methoxy isoflavone) is a phytochemical phytoestrogen that is highly effective against various diseases. Biochanin-A is a nutritional and dietary isoflavonoid naturally present in red clover, chickpea, soybeans and other herbs. Biochanin- A possesses numerous biological activities. OBJECTIVE The study focused on collective data of therapeutic activities of Biochanin-A. METHODS According to the literature, biochanin-A revealed a range of activities starting from chemoprevention, by hindering cell growth, activation of tumor cell death, hampering metastasis, angiogenic action, cell cycle regulation, neuroprotection, by controlling microglial activation, balancing antioxidants, elevating the neurochemicals, suppressing BACE-1, NADPH oxidase hindrance to inflammation, by mitigating the MAPK and NF- κB, discharge of inflammatory markers, upregulating the PPAR-γ, improving the function of heme oxygenase-1, erythroid 2 nuclear factors, detoxifying the oxygen radicals and stimulating the superoxide dismutase action, and controlling its production of transcription factors. Against pathogens, biochanin-A acts by dephosphorylating tyrosine kinase proteins, obstructing gram-negative bacteria, suppressing the development of cytokines from viruses, and improving the action of a neuraminidase cleavage of caspase-3, and acts as an efflux pump inhibitor. In metabolic disorders, biochanin-A acts by encouraging transcriptional initiation and inhibition, activating estrogen receptors, and increasing the activity of differentiation, autophagy, inflammation, and blood glucose metabolism. CONCLUSION Therefore, biochanin-A could be used as a therapeutic drug for various pathological conditions and treatments in human beings.
Collapse
Affiliation(s)
- Vadivelan Ramachandran
- Department of Pharmacology, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Ooty, Nilgiris, Tamil Nadu, India
| | - Inba Kumar V
- Department of Pharmacology, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Ooty, Nilgiris, Tamil Nadu, India
| | - Kiran Kumar Hr
- Department of Pharmacology, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Ooty, Nilgiris, Tamil Nadu, India
| | - Ruchi Tiwari
- PSIT-Pranveer Singh Institute of Technology (Pharmacy), Kalpi Road, Bhauti, Kanpur 208020, India
| | - Gaurav Tiwari
- PSIT-Pranveer Singh Institute of Technology (Pharmacy), Kalpi Road, Bhauti, Kanpur 208020, India
| |
Collapse
|
12
|
Wang J, Li Y, Li A, Liu RH, Gao X, Li D, Kou X, Xue Z. Nutritional constituent and health benefits of chickpea (Cicer arietinum L.): A review. Food Res Int 2021; 150:110790. [PMID: 34865805 DOI: 10.1016/j.foodres.2021.110790] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2021] [Revised: 10/08/2021] [Accepted: 10/24/2021] [Indexed: 11/19/2022]
Abstract
Chickpea (Cicer arietinum L.), an annual plant of the Fabaceae family, is mainly grown in temperate and semiarid regions. Its biological activity and beneficial contribution to human health have been scientifically confirmed as an essential source of nutritional components. The objective of this review was to summarize and update latest available scientific data and information, on bioactive components in chickpea, bio-activities, and molecular mechanisms, which has mainly focused on the detection of relevant biochemical indicators, the regulation of signaling pathways, essential genes and proteins. The studies have shown that chickpea have significant multifunctional activities, which are closely related to the functionally active small molecule peptides and phytochemicals of chickpea. Significantly, numerous studies have only addressed the functional activity and mechanisms of single active components of chickpea, however, overlooking the synergy and antagonism between chickpea components, changes of functional active components in different processing methods, as well as the active form of the substances after human digestion and metabolism. Additionally, due to limitations in research methods and techniques, the structure of most functional active substances have not been determined, which makes it difficult to conduct interaction mechanism studies. Consequently, the significant bio-activity of the functional components of chickpea, synergistic and antagonistic effects and activity differences between bioactive components should be further studied.
Collapse
Affiliation(s)
- Junyu Wang
- Department of Food Science, School of Chemical Engineering and Technology, Tianjin University, 300350 Tianjin, China.
| | - Yonghui Li
- Cardiovascular Department, Tianjin Fourth Center Hospital, Tianjin 300140, China.
| | - Ang Li
- Department of Food Science, School of Chemical Engineering and Technology, Tianjin University, 300350 Tianjin, China.
| | - Rui Hai Liu
- Department of Food Science, Stocking Hall, Cornell University, Ithaca, NY 14853, USA.
| | - Xin Gao
- Department of Food Science, School of Chemical Engineering and Technology, Tianjin University, 300350 Tianjin, China.
| | - Dan Li
- Department of Food Science, School of Chemical Engineering and Technology, Tianjin University, 300350 Tianjin, China.
| | - Xiaohong Kou
- Department of Food Science, School of Chemical Engineering and Technology, Tianjin University, 300350 Tianjin, China.
| | - Zhaohui Xue
- Department of Food Science, School of Chemical Engineering and Technology, Tianjin University, 300350 Tianjin, China.
| |
Collapse
|
13
|
Köksal Karayildirim Ç, Nalbantsoy A, Karabay Yavaşoğlu NÜ. Prunetin inhibits nitric oxide activity and induces apoptosis in urinary bladder cancer cells via CASP3 and TNF-α genes. Mol Biol Rep 2021; 48:7251-7259. [PMID: 34599704 DOI: 10.1007/s11033-021-06719-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2021] [Accepted: 09/27/2021] [Indexed: 11/29/2022]
Abstract
BACKGROUND Urinary bladder cancer (UBC) is considered one of the most prevalent malignant tumors worldwide. Complementary and integrative approaches for the treatment of bladder cancer, such as the intake of isoflavonoid phytoestrogens, are of increasing interest due to the risk of mortality and long-term morbidity associated with surgical procedures. The biological effects of prunetin, one of the less-studied phytoestrogens, have not yet been examined in this respect. Therefore, this study aimed to explore the efficacy of prunetin on UBC cells (RT-4). METHODS AND RESULTS: The cytotoxicity and nitric oxide synthase activities of prunetin were determined in cell cultures. The expression of apoptosis-related genes was determined with RT-PCR. Cell cycle assays were performed using a flow cytometer and cellular apoptotic rate was measured. The results suggested that prunetin has cytotoxic effects at 21.11 µg/mL on RT-4 cells. Flow cytometry analysis showed that prunetin induced apoptosis and arrested th cell cycle in the G0/G1 phase. Prunetin exposure was associated with increases in CASP3 and TNF-α gene expression in RT-4 cells at doses of 21.11 and 42.22 µg/mL, respectively. Strong nitric oxide inhibition was observed at IC50 of 5.18 µg/mL under macrophage mediated inflammatory circumstances. CONCLUSIONS Prunetin possesses anti-cancer properties and may be a candidate compound for the prevention of UBC. This is the first study that evaluated prunetin for its in vitro antitumor activities, clarified its possible apoptotic molecular mechanism and provided novel insights into its anti-inflammatory nature and effects on the expression of related key genes.
Collapse
Affiliation(s)
| | - Ayşe Nalbantsoy
- Department of Bioengineering, Faculty of Engineering, Ege University, 35100, Izmir, Turkey
| | | |
Collapse
|
14
|
Razeghian E, Suksatan W, Sulaiman Rahman H, Bokov DO, Abdelbasset WK, Hassanzadeh A, Marofi F, Yazdanifar M, Jarahian M. Harnessing TRAIL-Induced Apoptosis Pathway for Cancer Immunotherapy and Associated Challenges. Front Immunol 2021; 12:699746. [PMID: 34489946 PMCID: PMC8417882 DOI: 10.3389/fimmu.2021.699746] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2021] [Accepted: 08/05/2021] [Indexed: 01/04/2023] Open
Abstract
The immune cytokine tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) has attracted rapidly evolving attention as a cancer treatment modality because of its competence to selectively eliminate tumor cells without instigating toxicity in vivo. TRAIL has revealed encouraging promise in preclinical reports in animal models as a cancer treatment option; however, the foremost constraint of the TRAIL therapy is the advancement of TRAIL resistance through a myriad of mechanisms in tumor cells. Investigations have documented that improvement of the expression of anti-apoptotic proteins and survival or proliferation involved signaling pathways concurrently suppressing the expression of pro-apoptotic proteins along with down-regulation of expression of TRAILR1 and TRAILR2, also known as death receptor 4 and 5 (DR4/5) are reliable for tumor cells resistance to TRAIL. Therefore, it seems that the development of a therapeutic approach for overcoming TRAIL resistance is of paramount importance. Studies currently have shown that combined treatment with anti-tumor agents, ranging from synthetic agents to natural products, and TRAIL could result in induction of apoptosis in TRAIL-resistant cells. Also, human mesenchymal stem/stromal cells (MSCs) engineered to generate and deliver TRAIL can provide both targeted and continued delivery of this apoptosis-inducing cytokine. Similarly, nanoparticle (NPs)-based TRAIL delivery offers novel platforms to defeat barricades to TRAIL therapeutic delivery. In the current review, we will focus on underlying mechanisms contributed to inducing resistance to TRAIL in tumor cells, and also discuss recent findings concerning the therapeutic efficacy of combined treatment of TRAIL with other antitumor compounds, and also TRAIL-delivery using human MSCs and NPs to overcome tumor cells resistance to TRAIL.
Collapse
Affiliation(s)
- Ehsan Razeghian
- Human Genetics Division, Medical Biotechnology Department, National Institute of Genetics Engineering and Biotechnology (NIGEB), Tehran, Iran
| | - Wanich Suksatan
- Faculty of Nursing, HRH Princess Chulabhorn College of Medical Science, Chulabhorn Royal Academy, Bangkok, Thailand
| | - Heshu Sulaiman Rahman
- Department of Physiology, College of Medicine, University of Suleimanyah, Suleimanyah, Iraq
- Department of Medical Laboratory Sciences, Komar University of Science and Technology, Sulaimaniyah, Iraq
| | - Dmitry O. Bokov
- Institute of Pharmacy, Sechenov First Moscow State Medical University, Moscow, Russia
- Laboratory of Food Chemistry, Federal Research Center of Nutrition, Biotechnology and Food Safety, Moscow, Russia
| | - Walid Kamal Abdelbasset
- Department of Health and Rehabilitation Sciences, College of Applied Medical Sciences, Prince Sattam bin Abdulaziz University, Al Kharj, Saudi Arabia
- Department of Physical Therapy, Kasr Al-Aini Hospital, Cairo University, Giza, Egypt
| | - Ali Hassanzadeh
- Department of Applied Cell Sciences, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Faroogh Marofi
- Immunology Research Center (IRC), Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mahboubeh Yazdanifar
- Stem Cell Transplantation and Regenerative Medicine, Department of Pediatrics, Stanford University School of Medicine, Palo Alto, CA, United States
| | - Mostafa Jarahian
- Toxicology and Chemotherapy Unit (G401), German Cancer Research Center, Heidelberg, Germany
| |
Collapse
|
15
|
Aboushanab SA, Khedr SM, Gette IF, Danilova IG, Kolberg NA, Ravishankar GA, Ambati RR, Kovaleva EG. Isoflavones derived from plant raw materials: bioavailability, anti-cancer, anti-aging potentials, and microbiome modulation. Crit Rev Food Sci Nutr 2021; 63:261-287. [PMID: 34251921 DOI: 10.1080/10408398.2021.1946006] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Isoflavones are secondary metabolites that represent the most abundant category of plant polyphenols. Dietary soy, kudzu, and red clover contain primarily genistein, daidzein, glycitein, puerarin, formononetin, and biochanin A. The structural similarity of these compounds to β-estradiol has demonstrated protection against age-related and hormone-dependent diseases in both genders. Demonstrative shreds of evidence confirmed the fundamental health benefits of the consumption of these isoflavones. These relevant activities are complex and largely driven by the source, active ingredients, dose, and administration period of the bioactive compounds. However, the preclinical and clinical studies of these compounds are greatly variable, controversial, and still with no consensus due to the non-standardized research protocols. In addition, absorption, distribution, metabolism, and excretion studies, and the safety profile of isoflavones have been far limited. This highlights a major gap in understanding the potentially critical role of these isoflavones as prospective replacement therapy. Our general review exclusively focuses attention on the crucial role of isoflavones derived from these plant materials and critically highlights their bioavailability, possible anticancer, antiaging potentials, and microbiome modulation. Despite their fundamental health benefits, plant isoflavones reveal prospective therapeutic effects that worth further standardized analysis.
Collapse
Affiliation(s)
- Saied A Aboushanab
- Institute of Chemical Engineering, Ural Federal University named after the first President of Russia B. N. Yeltsin, Yekaterinburg, Russia
| | - Shaimaa M Khedr
- Pharmaceutical and Fermentation Industries Development Center (PFIDC), City of Scientific Research and Technological Applications, SRTA-City, Alexandria, Egypt
| | - Irina F Gette
- Institute of Chemical Engineering, Ural Federal University named after the first President of Russia B. N. Yeltsin, Yekaterinburg, Russia.,Institute of Immunology and Physiology, Ural Branch of the Russian Academy of Sciences, Yekaterinburg, Russia
| | - Irina G Danilova
- Institute of Chemical Engineering, Ural Federal University named after the first President of Russia B. N. Yeltsin, Yekaterinburg, Russia.,Institute of Immunology and Physiology, Ural Branch of the Russian Academy of Sciences, Yekaterinburg, Russia
| | - Natalia A Kolberg
- Integrated Laboratory Complex, Ural State University of Economics, Yekaterinburg, Russia
| | - Gokare A Ravishankar
- C. D. Sagar Centre for Life Sciences, Dayananda Sagar College of Engineering, Dayananda Sagar Institutions, Bangalore, Karnataka, India
| | - Ranga Rao Ambati
- Department of Biotechnology, Vignan's Foundation of Science, Technology and Research, Guntur, Andhra Pradesh, India
| | - Elena G Kovaleva
- Institute of Chemical Engineering, Ural Federal University named after the first President of Russia B. N. Yeltsin, Yekaterinburg, Russia
| |
Collapse
|
16
|
Azizi A, Mumin NH, Shafqat N. Phytochemicals With Anti 5-alpha-reductase Activity: A Prospective For Prostate Cancer Treatment. F1000Res 2021; 10:221. [PMID: 34316358 PMCID: PMC8276191 DOI: 10.12688/f1000research.51066.3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 07/01/2021] [Indexed: 11/20/2022] Open
Abstract
Prostate cancer (CaP) is one of the leading causes of death in men worldwide. Much attention has been given on its prevention and treatment strategies, including targeting the regulation of 5-alpha-Reductase (5αR) enzyme activity, aimed to limit the progression of CaP by inhibiting the conversion of potent androgen dihydrotestosterone from testosterone that is thought to play a role in pathogenesis of CaP, by using the 5-alpha-Reductase inhibitors (5αRis) such as finasteride and dutasteride. However, 5αRis are reported to exhibit numerous adverse side effects, for instance erectile dysfunction, ejaculatory dysfunction and loss of libido. This has led to a surge of interest on plant-derived alternatives that might offer favourable side effects and less toxic profiles. Phytochemicals from plants are shown to exhibit numerous medicinal properties in various studies targeting many major illnesses including CaP. Therefore, in this review, we aim to discuss the use of phytochemicals namely phytosterols, polyphenols and fatty acids, found in various plants with proven anti-CaP properties, as an alternative herbal CaP medicines as well as to outline their inhibitory activities on 5αRs isozymes based on their structural similarities with current 5αRis as part of CaP treatment approaches.
Collapse
Affiliation(s)
- Aziemah Azizi
- PAPRSB Institute of Health Sciences, Universiti Brunei Darussalam, Jalan Tungku Link, Gadong, BE1410, Brunei
| | - Nuramalina H Mumin
- PAPRSB Institute of Health Sciences, Universiti Brunei Darussalam, Jalan Tungku Link, Gadong, BE1410, Brunei
| | - Naeem Shafqat
- PAPRSB Institute of Health Sciences, Universiti Brunei Darussalam, Jalan Tungku Link, Gadong, BE1410, Brunei
| |
Collapse
|
17
|
Xiao Y, Gong Q, Wang W, Liu F, Kong Q, Pan F, Zhang X, Yu C, Hu S, Fan F, Li S, Liu Y. The combination of Biochanin A and SB590885 potentiates the inhibition of tumour progression in hepatocellular carcinoma. Cancer Cell Int 2020; 20:371. [PMID: 32774165 PMCID: PMC7405455 DOI: 10.1186/s12935-020-01463-w] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2020] [Accepted: 07/29/2020] [Indexed: 12/15/2022] Open
Abstract
Background Hepatocellular carcinoma (HCC) is the most aggressive and frequently diagnosed malignancy of the liver. Despite aggressive therapy, life expectancy of many patients in these cases is extended by only a few months. Hepatocellular carcinoma (HCC) has a particularly poor prognosis and would greatly benefit from more effective therapies. Methods The CCK-8 assay and colony formation assays were used to test the cell proliferation and viability. The effects of combination Biochanin A and SB590885 on apoptosis and cell cycle arrest of HCC cells were analysed by flow cytometry. The expression of ERK MAPK and PI3K/AKT/mTOR signalling as well as apoptosis and cell cycle-related proteins in HCC cells were tested by western blotting. The HCC cell xenograft model was established to test the tumor proliferation. Serum and plasma were tested for liver and kidney safety markers (ALP, ALT, AST, total bilirubin, creatinine, urea nitrogen) by using SpectraMax i3X. Results The combination of natural product Biochanin A with the BRAF inhibitor SB590885 synergistically suppressed proliferation, and promoted cell cycle arrest and apoptosis in vitro. Furthermore, we demonstrated that the combination of Biochanin A and SB590885 led to increased impairment of proliferation and HCC tumour inhibition through disrupting of the ERK MAPK and the PI3K/AKT pathways in vitro. The volumes tumors and the weights of tumours were significantly reduced by the combination treatment compared to the control or single treatments in vivo. In addition, we found that there was no significant hepatorenal toxicity with the drug combination, as indicated by the hepatorenal toxicity test. Conclusion The results identify an effective combination therapy for the most aggressive form of HCC and provide the possibility of therapeutic improvement for patients with advanced HCC.
Collapse
Affiliation(s)
- Yi Xiao
- Guizhou Provincial College-based Key Lab for Tumor Prevention and Treatment with Distinctive Medicines, Zunyi Medical University, No.6 West Xuefu Road, Xinpu District, Zunyi, 563000 China.,Department of Biochemistry and Molecular Biology, Zunyi Medical University, No.6 West Xuefu Road, Xinpu District, Zunyi, 563000 China.,Research Center for Medicine & Biology, Zunyi Medical University, Zunyi, 563000 China
| | - Qiang Gong
- Guizhou Provincial College-based Key Lab for Tumor Prevention and Treatment with Distinctive Medicines, Zunyi Medical University, No.6 West Xuefu Road, Xinpu District, Zunyi, 563000 China.,Research Center for Medicine & Biology, Zunyi Medical University, Zunyi, 563000 China
| | - Wenhong Wang
- Guizhou Provincial College-based Key Lab for Tumor Prevention and Treatment with Distinctive Medicines, Zunyi Medical University, No.6 West Xuefu Road, Xinpu District, Zunyi, 563000 China.,Research Center for Medicine & Biology, Zunyi Medical University, Zunyi, 563000 China
| | - Fang Liu
- Guizhou Provincial College-based Key Lab for Tumor Prevention and Treatment with Distinctive Medicines, Zunyi Medical University, No.6 West Xuefu Road, Xinpu District, Zunyi, 563000 China
| | - Qinghong Kong
- Guizhou Provincial College-based Key Lab for Tumor Prevention and Treatment with Distinctive Medicines, Zunyi Medical University, No.6 West Xuefu Road, Xinpu District, Zunyi, 563000 China.,Research Center for Medicine & Biology, Zunyi Medical University, Zunyi, 563000 China
| | - Feng Pan
- Guizhou Provincial College-based Key Lab for Tumor Prevention and Treatment with Distinctive Medicines, Zunyi Medical University, No.6 West Xuefu Road, Xinpu District, Zunyi, 563000 China.,Research Center for Medicine & Biology, Zunyi Medical University, Zunyi, 563000 China
| | - Xiaoke Zhang
- Guizhou Provincial College-based Key Lab for Tumor Prevention and Treatment with Distinctive Medicines, Zunyi Medical University, No.6 West Xuefu Road, Xinpu District, Zunyi, 563000 China.,Research Center for Medicine & Biology, Zunyi Medical University, Zunyi, 563000 China
| | - Changyan Yu
- Guizhou Provincial College-based Key Lab for Tumor Prevention and Treatment with Distinctive Medicines, Zunyi Medical University, No.6 West Xuefu Road, Xinpu District, Zunyi, 563000 China.,Research Center for Medicine & Biology, Zunyi Medical University, Zunyi, 563000 China
| | - Shanshan Hu
- Guizhou Provincial College-based Key Lab for Tumor Prevention and Treatment with Distinctive Medicines, Zunyi Medical University, No.6 West Xuefu Road, Xinpu District, Zunyi, 563000 China.,Good Clinical Practice Center, Affiliated Hospital of Zunyi Medical University, Zunyi, 563000 China
| | - Fang Fan
- Guizhou Provincial College-based Key Lab for Tumor Prevention and Treatment with Distinctive Medicines, Zunyi Medical University, No.6 West Xuefu Road, Xinpu District, Zunyi, 563000 China.,Department of Biochemistry and Molecular Biology, Zunyi Medical University, No.6 West Xuefu Road, Xinpu District, Zunyi, 563000 China
| | - Sanhua Li
- Guizhou Provincial College-based Key Lab for Tumor Prevention and Treatment with Distinctive Medicines, Zunyi Medical University, No.6 West Xuefu Road, Xinpu District, Zunyi, 563000 China.,Research Center for Medicine & Biology, Zunyi Medical University, Zunyi, 563000 China
| | - Yun Liu
- Guizhou Provincial College-based Key Lab for Tumor Prevention and Treatment with Distinctive Medicines, Zunyi Medical University, No.6 West Xuefu Road, Xinpu District, Zunyi, 563000 China.,Department of Biochemistry and Molecular Biology, Zunyi Medical University, No.6 West Xuefu Road, Xinpu District, Zunyi, 563000 China.,Research Center for Medicine & Biology, Zunyi Medical University, Zunyi, 563000 China
| |
Collapse
|
18
|
Sarfraz A, Javeed M, Shah MA, Hussain G, Shafiq N, Sarfraz I, Riaz A, Sadiqa A, Zara R, Zafar S, Kanwal L, Sarker SD, Rasul A. Biochanin A: A novel bioactive multifunctional compound from nature. THE SCIENCE OF THE TOTAL ENVIRONMENT 2020; 722:137907. [PMID: 32208265 DOI: 10.1016/j.scitotenv.2020.137907] [Citation(s) in RCA: 79] [Impact Index Per Article: 19.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/02/2020] [Revised: 02/25/2020] [Accepted: 03/11/2020] [Indexed: 06/10/2023]
Abstract
Natural products (NPs) will continue to serve humans as matchless source of novel drug leads and an inspiration for the synthesis of non-natural drugs. As our scientific understanding of 'nature' is rapidly expanding, it would be worthwhile to illuminate the pharmacological distinctions of NPs to the scientific community and the public. Flavonoids have long fascinated scientists with their remarkable structural diversity as well as biological functions. Consequently, this review aims to shed light on the sources and pharmacological significance of a dietary isoflavone, biochanin A, which has been recently emerged as a multitargeted and multifunctional guardian of human health. Biochanin A possesses anti-inflammatory, anticancer, neuroprotective, antioxidant, anti-microbial, and hepatoprotective properties. It combats cancer development by inducing apoptosis, inhibition of metastasis and arresting cell cycle via targeting several deregulated signaling pathways of cancer. It fights inflammation by blocking the expression and activity of pro-inflammatory cytokines via modulation of NF-κB and MAPKs. Biochanin A acts as a neuroprotective agent by inhibiting microglial activation and apoptosis of neurons. As biochanin A has potential to modulate several biological networks, thus, it can be anticipated that this therapeutically potent compound might serve as a novel lead for drug development in the near future.
Collapse
Affiliation(s)
- Ayesha Sarfraz
- Department of Zoology, Faculty of Life Sciences, Government College University, Faisalabad 38000, Pakistan
| | - Maria Javeed
- Department of Zoology, Faculty of Life Sciences, Government College University, Faisalabad 38000, Pakistan
| | - Muhammad Ajmal Shah
- Department of Pharmacognosy, Faculty of Pharmaceutical Sciences, Government College University, Faisalabad 38000, Pakistan.
| | - Ghulam Hussain
- Department of Physiology, Faculty of Life Sciences, Government College University, Faisalabad 38000, Pakistan
| | - Nusrat Shafiq
- Department of Chemistry, Government College Woman University Faisalabad (GCWUF), 38000 Faisalabad, Pakistan
| | - Iqra Sarfraz
- Department of Zoology, Faculty of Life Sciences, Government College University, Faisalabad 38000, Pakistan
| | - Ammara Riaz
- Department of Zoology, Faculty of Life Sciences, Government College University, Faisalabad 38000, Pakistan
| | - Ayesha Sadiqa
- Department of Zoology, Faculty of Life Sciences, Government College University, Faisalabad 38000, Pakistan
| | - Rabia Zara
- Department of Zoology, Faculty of Life Sciences, Government College University, Faisalabad 38000, Pakistan
| | - Saba Zafar
- Department of Zoology, Faculty of Life Sciences, Government College University, Faisalabad 38000, Pakistan
| | - Lubna Kanwal
- Institute of Pure and Applied Zoology, University of Okara, Okara, Pakistan
| | - Satyajit D Sarker
- Centre for Natural Products Discovery (CNPD), School of Pharmacy & Biomolecular Sciences, Liverpool John Moores University, Byrom Street, Liverpool L3 3AF, England, UK
| | - Azhar Rasul
- Department of Zoology, Faculty of Life Sciences, Government College University, Faisalabad 38000, Pakistan.
| |
Collapse
|
19
|
Arora I, Sharma M, Tollefsbol TO. Combinatorial Epigenetics Impact of Polyphenols and Phytochemicals in Cancer Prevention and Therapy. Int J Mol Sci 2019; 20:ijms20184567. [PMID: 31540128 PMCID: PMC6769666 DOI: 10.3390/ijms20184567] [Citation(s) in RCA: 103] [Impact Index Per Article: 20.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2019] [Revised: 09/08/2019] [Accepted: 09/11/2019] [Indexed: 12/24/2022] Open
Abstract
Polyphenols are potent micronutrients that can be found in large quantities in various food sources and spices. These compounds, also known as phenolics due to their phenolic structure, play a vital nutrient-based role in the prevention of various diseases such as diabetes, cardiovascular diseases, neurodegenerative diseases, liver disease, and cancers. However, the function of polyphenols in disease prevention and therapy depends on their dietary consumption and biological properties. According to American Cancer Society statistics, there will be an expected rise of 23.6 million new cancer cases by 2030. Due to the severity of the increased risk, it is important to evaluate various preventive measures associated with cancer. Relatively recently, numerous studies have indicated that various dietary polyphenols and phytochemicals possess properties of modifying epigenetic mechanisms that modulate gene expression resulting in regulation of cancer. These polyphenols and phytochemicals, when administrated in a dose-dependent and combinatorial-based manner, can have an enhanced effect on epigenetic changes, which play a crucial role in cancer prevention and therapy. Hence, this review will focus on the mechanisms of combined polyphenols and phytochemicals that can impact various epigenetic modifications such as DNA methylation and histone modifications as well as regulation of non-coding miRNAs expression for treatment and prevention of various types of cancer.
Collapse
Affiliation(s)
- Itika Arora
- Department of Biology, University of Alabama at Birmingham, 1300 University Boulevard, Birmingham, AL 35294, USA.
| | - Manvi Sharma
- Department of Biology, University of Alabama at Birmingham, 1300 University Boulevard, Birmingham, AL 35294, USA.
| | - Trygve O Tollefsbol
- Department of Biology, University of Alabama at Birmingham, 1300 University Boulevard, Birmingham, AL 35294, USA.
- Comprehensive Center for Healthy Aging, University of Alabama Birmingham, 1530 3rd Avenue South, Birmingham, AL 35294, USA.
- Comprehensive Cancer Center, University of Alabama Birmingham, 1802 6th Avenue South, Birmingham, AL 35294, USA.
- Nutrition Obesity Research Center, University of Alabama Birmingham, 1675 University Boulevard, Birmingham, AL 35294, USA.
- Comprehensive Diabetes Center, University of Alabama Birmingham, 1825 University Boulevard, Birmingham, AL 35294, USA.
| |
Collapse
|
20
|
Ye Q, Liu K, Shen Q, Li Q, Hao J, Han F, Jiang RW. Reversal of Multidrug Resistance in Cancer by Multi-Functional Flavonoids. Front Oncol 2019; 9:487. [PMID: 31245292 PMCID: PMC6581719 DOI: 10.3389/fonc.2019.00487] [Citation(s) in RCA: 92] [Impact Index Per Article: 18.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2019] [Accepted: 05/23/2019] [Indexed: 12/22/2022] Open
Abstract
Multidrug resistance (MDR) resulting from different defensive mechanisms in cancer is one of the major obstacles of clinical treatment. To circumvent MDR many reversal agents have been developed, but most of them fail in clinical trials due to severely adverse effects. Recently, certain natural products have been reported to overcome MDR, including flavonoids which are abundant in plants, foods, and herbs. The structure of flavonoids can be abbreviated as C6-C3-C6 (C for carbon), and further categorized into flavonoids, iso-flavonoids and neo-flavonoids, according to their structural backbones. Flavonoids possess multiple bioactivities, and a growing body of research has indicated that both flavonoids and iso-flavonoids can either kill or re-sensitize conventional chemotherapeutics to resistant cancer cells. Here, we summarize the research and discuss the underlying mechanisms, concluding that these flavonoids do not function as specific regulators of target proteins, but rather as multi-functional agents that negatively regulate the key factors contributing to MDR.
Collapse
Affiliation(s)
| | - Kai Liu
- Hainan General Hospital, Haikou, China
| | - Qun Shen
- Hainan General Hospital, Haikou, China
| | | | - Jinghui Hao
- Jiaozuo Second People's Hospital, Jiaozuo, China
| | | | - Ren-Wang Jiang
- Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research, Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Development of Chinese Ministry of Education, Jinan University, Guangzhou, China
| |
Collapse
|
21
|
Network Pharmacology Integrated Molecular Docking Reveals the Antiosteosarcoma Mechanism of Biochanin A. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2019; 2019:1410495. [PMID: 30723510 PMCID: PMC6339762 DOI: 10.1155/2019/1410495] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/14/2018] [Revised: 10/29/2018] [Accepted: 11/22/2018] [Indexed: 02/06/2023]
Abstract
Background As the malignant tumor with the highest incidence in teenagers, osteosarcoma has become a major problem in oncology research. In addition to surgical management, the pharmacotherapeutic strategy for osteosarcoma treatment is an attractive way to explore. It has been demonstrated that biochanin A has an antitumor capacity on multiple kinds of solid tumor, including osteosarcoma. But the precise mechanism of biochanin A against osteosarcoma is still needed to be discovered. Objective To identify the potential therapeutic targets of biochanin A in treating osteosarcoma. Methods In present study, an integrated approach including network pharmacology and molecular docking technique was conducted, which mainly comprises target prediction, network construction, gene ontology, and pathway enrichment. CCK8 test was employed to evaluate the cell viability of MG63 cells. Western-blot was used to verify the target proteins of biochanin A. Results Ninety-six and 114 proteins were obtained as the targets of biochanin A and osteosarcoma, respectively. TP53, IGF1, JUN, BGLAP, ATM, MAPK1, ATF3, H2AFX, BAX, CDKN2A, and EGF were identified as the potential targets of biochanin A against osteosarcoma. Based on the western-blot detection, the expression of BGLAP, BAX, and ATF3 in MG63 cell line changed under the treatment of biochanin A. Conclusion Biochanin A can effectively suppress the proliferation of osteosarcoma and regulate the expression of BGLAP, BAX, and ATF3, which may act as the potential therapeutic targets of osteosarcoma.
Collapse
|
22
|
Botlagunta M, Mathi P, Musunuru N, Adurthi U. Comparative in vitro and in silico characterization of anticancer compounds piceatannol, biochanin-A and resveratrol on breast cancer cells. Pharmacogn Mag 2019. [DOI: 10.4103/pm.pm_146_19] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
|
23
|
|
24
|
Renoprotective effect of the isoflavonoid biochanin A against cisplatin induced acute kidney injury in mice: Effect on inflammatory burden and p53 apoptosis. Int Immunopharmacol 2018; 61:8-19. [DOI: 10.1016/j.intimp.2018.05.010] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2017] [Revised: 05/10/2018] [Accepted: 05/16/2018] [Indexed: 12/26/2022]
|
25
|
Xu X, Li X, Liang X. Application of ultra-performance liquid chromatography coupled with quadrupole time-of-flight mass spectrometry in identification of three isoflavone glycosides and their corresponding metabolites. RAPID COMMUNICATIONS IN MASS SPECTROMETRY : RCM 2018; 32:262-268. [PMID: 29222826 DOI: 10.1002/rcm.8038] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/18/2017] [Revised: 11/14/2017] [Accepted: 11/15/2017] [Indexed: 06/07/2023]
Abstract
RATIONALE Metabolites of isoflavones have attracted much attention in recent years due to their potential bioactivities. However, the complex constituents of the metabolic system and the low level of metabolites make them difficult to analyze. A mass spectrometry (MS) method was applied in our identification of metabolites and study of their fragmentation pathways due to the advantages of rapidity, sensitivity, and low level of sample consumption. METHODS Three isoflavone glycosides and their metabolites were identified using ultra-performance liquid chromatography coupled with quadrupole time-of-flight tandem mass spectrometry (UPLC/QTOF-MS). These metabolites were obtained by anaerobically incubating three isoflavone glycosides with human intestinal flora. The characteristic fragments of isoflavone glycosides and their metabolites were used for the identification work. RESULTS Two metabolites from ononin, three metabolites from irilone-4'-O-β-D-glucoside, and five metabolites from sissotrin were identified respectively by the retention time (RT), accurate mass, and mass spectral fragmentation patterns. The losses of the glucosyl group, CO from the [M+H]+ ion were observed for all the three isoflavone glycosides. The characteristic retro-Diels-Alder (RDA) fragmentation patterns were used to differentiate the compounds. The metabolic pathways of the three isoflavone glycosides were proposed according to the identified chemical structures of the metabolites. CONCLUSIONS A selective, sensitive and rapid method was established for detecting and identifying three isoflavone glycosides and their metabolites using UPLC/QTOF-MS. The established method can be used for further rapid structural identification studies of metabolites and natural products. Furthermore, the proposed metabolic pathways will be helpful for understanding the in vivo metabolic process of isoflavone.
Collapse
Affiliation(s)
- Xiafen Xu
- Key Laboratory for Green Pharmaceutical Technologies and Related Equipment of Ministry of Education, College of Pharmaceutical Sciences, Zhejiang University of Technology, Hangzhou, 310014, P.R. China
| | - Xinhui Li
- Key Laboratory for Green Pharmaceutical Technologies and Related Equipment of Ministry of Education, College of Pharmaceutical Sciences, Zhejiang University of Technology, Hangzhou, 310014, P.R. China
| | - Xianrui Liang
- Key Laboratory for Green Pharmaceutical Technologies and Related Equipment of Ministry of Education, College of Pharmaceutical Sciences, Zhejiang University of Technology, Hangzhou, 310014, P.R. China
| |
Collapse
|
26
|
Cho IA, You SJ, Kang KR, Kim SG, Oh JS, You JS, Lee GJ, Seo YS, Kim DK, Kim CS, Lee SY, Kim JS. Biochanin-A induces apoptosis and suppresses migration in FaDu human pharynx squamous carcinoma cells. Oncol Rep 2017; 38:2985-2992. [DOI: 10.3892/or.2017.5953] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2017] [Accepted: 08/31/2017] [Indexed: 11/05/2022] Open
|
27
|
Novel Structurally Related Flavones Augment Cell Death Induced by rhsTRAIL. Int J Mol Sci 2017; 18:ijms18061211. [PMID: 28587286 PMCID: PMC5486034 DOI: 10.3390/ijms18061211] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2017] [Revised: 05/26/2017] [Accepted: 06/01/2017] [Indexed: 12/15/2022] Open
Abstract
TRAIL (tumor necrosis factor-related apoptosis-inducing ligand) was identified as a powerful activator of apoptosis in tumor cells and one of the most promising candidates for cancer therapy with no toxicity against normal tissues. However, many tumor cells are resistant to TRAIL-induced apoptosis. The aim of this work was to analyze the improvement of the anticancer effect of rhsTRAIL (recombinant human soluble TRAIL) by nine flavones: 5-Hydroxyflavone, 6-Hydroxyflavone, 7-Hydroxyflavone and their new synthetic derivatives 5-acetoxyflavone, 5-butyryloxyflavone, 6-acetoxyflavone, 6-butyryloxyflavone, 7-acetoxyflavone and 7-butyryloxyflavone. We examined the cytotoxic and apoptotic effects of rhsTRAIL enhanced by novel structurally-related flavones on SW480 and SW620 colon cancer cells using the 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide test, the lactate dehydrogenase assay and annexin V-FITC fluorescence staining. We observed a slight difference in the activities of the flavones that was dependent on their chemical structure. Our study indicates that all nine flavones significantly augment cell death by rhsTRAIL (cytotoxicity range 36.8 ± 1.7%–91.4 ± 1.7%; apoptosis increase of 33.0 ± 0.7%–78.5 ± 0.9%). Our study demonstrates the potential use of tested flavones in TRAIL-based anticancer therapy and prevention.
Collapse
|
28
|
Dash TK, Konkimalla VB. Formulation and Optimization of Doxorubicin and Biochanin A Combinational Liposomes for Reversal of Chemoresistance. AAPS PharmSciTech 2017; 18:1116-1124. [PMID: 27600324 DOI: 10.1208/s12249-016-0614-z] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2016] [Accepted: 08/09/2016] [Indexed: 01/09/2023] Open
Abstract
Circumvention of drug resistance still remains a challenge in the development of anticancer therapeutics. Combinational nano-formulations provide many avenues for effective cancer therapy and reversal of drug resistance. In the current study, combination of biochanin A (BioA) and doxorubicin (DOX) in liposomes were prepared and studied for its potential to reverse DOX resistance in COLO205 cells. After development and validation of DOX resistant cells of COLO205 (ColoR), dosing ratio of DOX and BioA for reversal of DOX resistance was determined by co-treatment in ColoR cells. As limited solubility and analytical data available for BioA, therefore solubility was studied for BioA and analytical method was developed for the combination. Combinational liposomes were prepared and optimized for both lipid content and surface charge by evaluating size, polydispersity index, zeta potential, and encapsulation efficiency. The optimized formulation had a size about 125 nm; zeta potential of -19.5 mV and 70% encapsulation efficiency (EE) for BioA. Thus, prepared combinational liposomes of DOX and BioA were evaluated for its cellular uptake and efficacy to reverse DOX resistance. From the study, increased DOX uptake and promising effect for reversal of DOX resistance was observed.
Collapse
|
29
|
Wu X, Ge W, Shao T, Wu W, Hou J, Cui L, Wang J, Zhang Z. Enhancing the oral bioavailability of biochanin A by encapsulation in mixed micelles containing Pluronic F127 and Plasdone S630. Int J Nanomedicine 2017; 12:1475-1483. [PMID: 28260893 PMCID: PMC5328660 DOI: 10.2147/ijn.s125041] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Biochanin A (BCA), a natural dietary isoflavone, has been reported to show anticancer activities. However, its low biological availability and poor aqueous solubility limit its usefulness as a chemotherapeutic agent. We developed BCA-loaded micelles with Pluronic F127 and Plasdone S630 (BCA-FS). The optimized, spherical-shaped BCA-FS was obtained at a ratio of 1:1 (F127:S630). The particle size was 25.17±1.2 nm, and the zeta potential was −10.9±0.24 mV. BCA solubility in water increased to 5.0 mg/mL after encapsulation, and the drug-loading efficiency was 5.88%±0.76%. In vitro release experiments showed a delayed release of BCA from the mixed micelles. Furthermore, the BCA absorption permeability across a Caco-2 cell monolayer from the apical side to the basolateral side increased by 54% in BCA-FS. A pharmacokinetics evaluation showed a 2.16-fold increase in the relative oral bioavailability of BCA-FS compared with raw BCA, indicating that the mixed micelles may promote absorption in the gastrointestinal tract. A gastrointestinal safety assay was used to assess the reliability and safety of BCA-FS. On the basis of these findings, we conclude that this simple nanomicelle system could be leveraged to deliver BCA and other hydrophobic drugs.
Collapse
Affiliation(s)
- Xiaoyan Wu
- Department of Pharmacy, The Affiliated Nanjing Drum Tower Hospital of Nanjing University Medical School
| | - Weihong Ge
- Department of Pharmacy, The Affiliated Nanjing Drum Tower Hospital of Nanjing University Medical School
| | - Tengfei Shao
- Department of Pharmacy, The Affiliated Nanjing Drum Tower Hospital of Nanjing University Medical School
| | - Weijun Wu
- Department of Pharmacy, The Affiliated Nanjing Drum Tower Hospital of Nanjing University Medical School
| | - Jian Hou
- Key Laboratory of New Drug Delivery System of Chinese Materia Medica, Jiangsu Provincial Academy of Chinese Medicine, Nanjing, Jiangsu, People's Republic of China
| | - Li Cui
- Key Laboratory of New Drug Delivery System of Chinese Materia Medica, Jiangsu Provincial Academy of Chinese Medicine, Nanjing, Jiangsu, People's Republic of China
| | - Jing Wang
- Key Laboratory of New Drug Delivery System of Chinese Materia Medica, Jiangsu Provincial Academy of Chinese Medicine, Nanjing, Jiangsu, People's Republic of China
| | - Zhenghai Zhang
- Key Laboratory of New Drug Delivery System of Chinese Materia Medica, Jiangsu Provincial Academy of Chinese Medicine, Nanjing, Jiangsu, People's Republic of China
| |
Collapse
|
30
|
Tumor Necrosis Factor-Related Apoptosis-Inducing Ligand-Induced Apoptosis in Prostate Cancer Cells after Treatment with Xanthohumol-A Natural Compound Present in Humulus lupulus L. Int J Mol Sci 2016; 17:ijms17060837. [PMID: 27338375 PMCID: PMC4926371 DOI: 10.3390/ijms17060837] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2016] [Revised: 05/19/2016] [Accepted: 05/23/2016] [Indexed: 01/31/2023] Open
Abstract
TRAIL (tumor necrosis factor-related apoptosis-inducing ligand) is an endogenous ligand, which plays role in immune surveillance and anti-tumor immunity. It has ability to selectively kill tumor cells showing no toxicity to normal cells. We tested the apoptotic and cytotoxic activities of xanthohumol, a prenylated chalcone found in Humulus lupulus on androgen-sensitive human prostate adenocarcinoma cells (LNCaP) in combination with TRAIL. Cytotoxicity was measured by 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide tetrazolium reduction assay (MTT) and lactate dehydrogenase assay (LDH). The expression of death receptors (DR4/TRAIL-R1 and DR5/TRAIL-R2) and apoptosis were detected using flow cytometry. We examined mitochondrial membrane potential (ΔΨm) by DePsipher reagent using fluorescence microscopy. The intracellular expression of proteins was evaluated by Western blotting. Our study showed that xanthohumol enhanced cytotoxic and apoptotic effects of TRAIL. The tested compounds activated caspases-3, -8, -9, Bid, and increased the expression of Bax. They also decreased expression of Bcl-xL and decreased mitochondrial membrane potential, while the expression of death receptors was not changed. The findings suggest that xanthohumol is a compound of potential use in chemoprevention of prostate cancer due to its sensitization of cancer cells to TRAIL-mediated apoptosis.
Collapse
|
31
|
Rahman HP, Hofland J, Foster PA. In touch with your feminine side: how oestrogen metabolism impacts prostate cancer. Endocr Relat Cancer 2016; 23:R249-66. [PMID: 27194038 DOI: 10.1530/erc-16-0118] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/12/2016] [Accepted: 05/18/2016] [Indexed: 12/18/2022]
Abstract
Prostate cancer is the primary cancer in males, with increasing global incidence rates making this malignancy a significant healthcare burden. Androgens not only promote normal prostate maturity but also influence the development and progression of prostate cancer. Intriguingly, evidence now suggests endogenous and exogenous oestrogens, in the form of phytoestrogens, may be equally as relevant as androgens in prostate cancer growth. The prostate gland has the molecular mechanisms, catalysed by steroid sulphatase (STS), to unconjugate and utilise circulating oestrogens. Furthermore, prostate tissue also expresses enzymes essential for local oestrogen metabolism, including aromatase (CYP19A1) and 3β- and 17β-hydroxysteroid dehydrogenases. Increased expression of these enzymes in malignant prostate tissue compared with normal prostate indicates that oestrogen synthesis is favoured in malignancy and thus may influence tumour progression. In contrast to previous reviews, here we comprehensively explore the epidemiological and scientific evidence on how oestrogens impact prostate cancer, particularly focusing on pre-receptor oestrogen metabolism and subsequent molecular action. We analyse how molecular mechanisms and metabolic pathways involved in androgen and oestrogen synthesis intertwine to alter prostate tissue. Furthermore, we speculate on whether oestrogen receptor status in the prostate affects progression of this malignancy.
Collapse
Affiliation(s)
- Habibur P Rahman
- Institute of Metabolism and Systems ResearchUniversity of Birmingham, Birmingham, UK
| | - Johannes Hofland
- Department of Internal MedicineErasmus Medical Center, Rotterdam, The Netherlands
| | - Paul A Foster
- Institute of Metabolism and Systems ResearchUniversity of Birmingham, Birmingham, UK Centre for EndocrinologyDiabetes and Metabolism, Birmingham Healthcare Partners, Birmingham, UK
| |
Collapse
|
32
|
Hajrezaie M, Salehen N, Karimian H, Zahedifard M, Shams K, Batran RA, Majid NA, Khalifa SAM, Ali HM, El-Seedi H, Abdulla MA. Biochanin a gastroprotective effects in ethanol-induced gastric mucosal ulceration in rats. PLoS One 2015; 10:e0121529. [PMID: 25811625 PMCID: PMC4374864 DOI: 10.1371/journal.pone.0121529] [Citation(s) in RCA: 59] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2014] [Accepted: 02/03/2015] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND Biochanin A notable bioactive compound which is found in so many traditional medicinal plant. In vivo study was conducted to assess the protective effect of biochanin A on the gastric wall of Spraguedawley rats` stomachs. METHODOLOGY The experimental set included different animal groups. Specifically, four groups with gastric mucosal lesions were receiving either a) Ulcer control group treated with absolute ethanol (5 ml/kg), b) 20 mg/kg of omeprazole as reference group, c) 25 of biochanin A, d) 50 mg/kg of biochanin A. Histopathological sectioning followed by immunohistochemistry staining were undertaken to evaluate the influence of the different treatments on gastric wall mucosal layer. The gastric secretions were collected in the form of homogenate and exposed to superoxide dismutase (SOD) and nitric oxide enzyme (NO) and the level of malondialdehyde (MDA) and protein content were measured. Ulceration and patchy haemorrhage were clearly observed by light microscopy. The morphology of the gastric wall as confirmed by immunohistochemistry and fluorescent microscopic observations, exhibited sever deformity with notable thickness, oedematous and complete loss of the mucosal coverage however the biochanin-pretreated animals, similar to the omeprazole-pretreated animals, showed less damage compared to the ulcer control group. Moreover, up-regulation of Hsp70 protein and down-regulation of Bax protein were detected in the biochanin A pre-treated groups and the gastric glandular mucosa was positively stained with Periodic Acid Schiff (PAS) staining and the Leucocytes infiltration was commonly seen. Biochanin A displayed a great increase in SOD and NO levels and decreased the release of MDA. CONCLUSIONS This gastroprotective effect of biochanin A could be attributed to the enhancement of cellular metabolic cycles perceived as an increase in the SOD, NO activity, and decrease in the level of MDA, and also decrease in level of Bax expression and increase the Hsp70 expression level.
Collapse
Affiliation(s)
- Maryam Hajrezaie
- Department of Biomedical Science, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
- Institute of Biological Sciences, Faculty of Science, University of Malaya, Kuala Lumpur, Malaysia
| | - NurAin Salehen
- Department of Biomedical Science, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
| | - Hamed Karimian
- Department of Biomedical Science, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
| | - Maryam Zahedifard
- Department of Biomedical Science, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
- Institute of Biological Sciences, Faculty of Science, University of Malaya, Kuala Lumpur, Malaysia
| | - Keivan Shams
- Department of Biomedical Science, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
| | - Rami Al Batran
- Department of Biomedical Science, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
| | - Nazia Abdul Majid
- Institute of Biological Sciences, Faculty of Science, University of Malaya, Kuala Lumpur, Malaysia
| | - Shaden A. M. Khalifa
- Department of Experimental Hematology, Karolinska University Hospital, SE-141 86, Stockholm, Sweden
| | - Hapipah Mohd Ali
- Department of Chemistry, University of Malaya, Kuala Lumpur, Malaysia
| | - Hesham El-Seedi
- Department of Experimental Hematology, Karolinska University Hospital, SE-141 86, Stockholm, Sweden
- Division of Pharmacognosy, Department of Medicinal Chemistry, Uppsala University, SE-75 123, Uppsala, Sweden
| | - Mahmood Ameen Abdulla
- Department of Biomedical Science, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
| |
Collapse
|
33
|
Wu DQ, Zhong HM, Ding QH, Ba L. Protective effects of biochanin A on articular cartilage: in vitro and in vivo studies. Altern Ther Health Med 2014; 14:444. [PMID: 25398247 PMCID: PMC4251671 DOI: 10.1186/1472-6882-14-444] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2014] [Accepted: 10/29/2014] [Indexed: 01/28/2023]
Abstract
Background Increased production of matrix metalloproteinases (MMPs) is closely related to the progression of osteoarthritis (OA). The present study was performed to investigate the potential value of biochanin A in inhibition of MMP expression in both rabbit chondrocytes and an animal model of OA. Methods MTT assay was performed to assess chondrocyte survival in monolayers. The mRNA and protein expression of MMPs (including MMP-1, MMP-3, and MMP-13) and tissue inhibitor of metalloproteinase-1 (TIMP-1) in interleukin-1 < beta > (IL-1β)-induced rabbit chondrocytes were determined by quantitative real-time PCR and enzyme-linked immunosorbent assay (ELISA), respectively. The involvement of the NF-kappaB (NF-κB) pathway activated by IL-1β was determined by western blotting. The in vivo effects of biochanin A were evaluated by intra-articular injection in an experimental OA rabbit model induced by anterior cruciate ligament transection (ACLT). Results Biochanin A downregulated the expression of MMPs and upregulated TIMP-1 at both the mRNA and protein levels in IL-1β-induced chondrocytes in a dose-dependent manner. In addition, IL-1β-induced activation of NF-κB was attenuated by biochanin A, as determined by western blotting. Moreover, biochanin A decreased cartilage degradation as determined by both morphological and histological analyses in vivo. Conclusions Taken together, these findings suggest that biochanin A may be a useful agent in the treatment and prevention of OA.
Collapse
|
34
|
Choi S, Jung WS, Cho NS, Ryu KH, Jun JY, Shin BC, Chung JH, Yeum CH. Mechanisms of phytoestrogen biochanin A-induced vasorelaxation in renovascular hypertensive rats. Kidney Res Clin Pract 2014; 33:181-6. [PMID: 26885474 PMCID: PMC4714256 DOI: 10.1016/j.krcp.2014.08.003] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2014] [Revised: 08/20/2014] [Accepted: 08/26/2014] [Indexed: 11/19/2022] Open
Abstract
Background The plant-derived estrogen biochanin A is known to cause vasodilation, but its mechanism of action in hypertension remains unclear. This study was undertaken to investigate the effects and mechanisms of biochanin A on the thoracic aorta in two-kidney, one clip (2K1C) renovascular hypertensive rats. Methods Hypertension was induced by clipping the left renal artery, and control age-matched rats were sham treated. Thoracic aortae were mounted in tissue baths to measure isometric tension. Results Biochanin A caused concentration-dependent relaxation in aortic rings from 2K1C hypertensive and sham-treated rats, which was greater in 2K1C rats than in sham rats. Biochanin A-induced relaxation was significantly attenuated by removing the endothelium in aortic rings from 2K1C rats, but not in sham rats. Nω-Nitro-l-arginine methyl ester, a nitric oxide synthase inhibitor, or indomethacin, a cyclooxygenase inhibitor, did not affect the biochanin A-induced relaxation in aortic rings from 2K1C and sham rats. By contrast, treatment with glibenclamide, a selective inhibitor of adenosine triphosphate-sensitive K+ channels, or tetraethylammonium, an inhibitor of Ca2+-activated K+ channels, significantly reduced biochanin A-induced relaxation in aortic rings from both groups. However, 4-aminopyridine, a selective inhibitor of voltage-dependent K+ channels, inhibited the relaxation induced by biochanin A in 2K1C rats, whereas no significant differences were observed in sham rats. Conclusion These results suggest that the enhanced relaxation caused by biochanin A in aortic rings from hypertensive rats is endothelium dependent. Vascular smooth muscle K+ channels may be involved in biochanin A-induced relaxation in aortae from hypertensive and normotensive rats. In addition, an endothelium-derived activation of voltage-dependent K+ channels contributes, at least in part, to the relaxant effect of biochanin A in renovascular hypertension.
Collapse
Affiliation(s)
- Seok Choi
- Department of Physiology, College of Medicine, Chosun University, Gwangju, Korea
| | - Won Suk Jung
- Department of Emergency Medicine, College of Medicine, Chosun University, Gwangju, Korea
| | - Nam Soo Cho
- Department of Emergency Medicine, College of Medicine, Chosun University, Gwangju, Korea
| | - Kwon Ho Ryu
- Department of Physiology, College of Medicine, Chosun University, Gwangju, Korea
| | - Jae Yeoul Jun
- Department of Physiology, College of Medicine, Chosun University, Gwangju, Korea
| | - Byung Chul Shin
- Department of Internal Medicine, College of Medicine, Chosun University, Gwangju, Korea
| | - Jong Hoon Chung
- Department of Internal Medicine, College of Medicine, Chosun University, Gwangju, Korea
| | - Cheol Ho Yeum
- Department of Physiology, College of Medicine, Chosun University, Gwangju, Korea
- Corresponding author. Department of Physiology, College of Medicine, Chosun University, 309, Pilmundaero, Dong-Gu, Gwangju 501-759, Korea.
| |
Collapse
|
35
|
Pei N, Jie F, Luo J, Wan R, Zhang Y, Chen X, Liang Z, Du H, Li A, Chen B, Zhang Y, Sumners C, Li J, Gu W, Li H. Gene expression profiling associated with angiotensin II type 2 receptor-induced apoptosis in human prostate cancer cells. PLoS One 2014; 9:e92253. [PMID: 24658029 PMCID: PMC3962398 DOI: 10.1371/journal.pone.0092253] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2013] [Accepted: 02/19/2014] [Indexed: 11/30/2022] Open
Abstract
Increased expression of angiotensin II type 2 receptor (AT2R) induces apoptosis in numerous tumor cell lines, with either Angiotensin II-dependent or Angiotensin II-independent regulation, but its molecular mechanism remains poorly understood. Here, we used PCR Array analysis to determine the gene and microRNA expression profiles in human prostate cancer cell lines transduced with AT2R recombinant adenovirus. Our results demonstrated that AT2R over expression leads to up-regulation of 6 apoptosis-related genes (TRAIL-R2, BAG3, BNIPI, HRK, Gadd45a, TP53BP2), 2 cytokine genes (IL6 and IL8) and 1 microRNA, and down-regulation of 1 apoptosis-related gene TNFSF10 and 2 cytokine genes (BMP6, BMP7) in transduced DU145 cells. HRK was identified as an up-regulated gene in AT2R-transduced PC-3 cells by real-time RT-PCR. Next, we utilized siRNAs to silence the up-regulated genes to further determine their roles on AT2R overexpression mediated apoptosis. The results showed downregulation of Gadd45a reduced the apoptotic effect by ∼30% in DU145 cells, downregulation of HRK reduced AT2R-mediated apoptosis by more than 50% in PC-3 cells, while downregulation of TRAIL-R2 enhanced AT2R-mediated apoptosis more than 4 times in DU145 cells. We also found that the effects on AT2R-mediated apoptosis caused by downregulation of Gadd45a, TRAIL-R2 and HRK were independent in activation of p38 MAPK, p44/42 MAPK and p53. Taken together, our results demonstrated that TRAIL-R2, Gadd45a and HRK may be novel target genes for further study of the mechanism of AT2R-mediated apoptosis in prostate cancer cells.
Collapse
Affiliation(s)
- Nana Pei
- School of Biotechnology, Southern Medical University, Guangzhou, Guangdong, China
| | - Feilong Jie
- School of Biotechnology, Southern Medical University, Guangzhou, Guangdong, China
| | - Jie Luo
- School of Biotechnology, Southern Medical University, Guangzhou, Guangdong, China
| | - Renqiang Wan
- Department of Otolaryngology-Head and Neck Surgery, Guangdong No. 2 Provincial People’s Hospital, Guangzhou, Guangdong, China
| | - Yanling Zhang
- School of Biotechnology, Southern Medical University, Guangzhou, Guangdong, China
| | - Xinglu Chen
- School of Biotechnology, Southern Medical University, Guangzhou, Guangdong, China
| | - Zhibing Liang
- School of Biotechnology, Southern Medical University, Guangzhou, Guangdong, China
| | - Hongyan Du
- School of Biotechnology, Southern Medical University, Guangzhou, Guangdong, China
| | - Andrew Li
- Department of Neuroscience, University of Florida, Gainesville, Florida, United States of America
| | - Baihong Chen
- School of Biotechnology, Southern Medical University, Guangzhou, Guangdong, China
| | - Yi Zhang
- Department of Pharmacology, University of Florida, Gainesville, Florida, United States of America
| | - Colin Sumners
- Department of Physiology and Functional Genomics, University of Florida, Gainesville, Florida, United States of America
| | - Jinlong Li
- School of Biotechnology, Southern Medical University, Guangzhou, Guangdong, China
- * E-mail: (JL); (WG); (HL)
| | - Weiwang Gu
- Institute of Comparative Medicine and Center of Laboratory Animals, Southern Medical University, Guangzhou, Guangdong, China
- * E-mail: (JL); (WG); (HL)
| | - Hongwei Li
- School of Biotechnology, Southern Medical University, Guangzhou, Guangdong, China
- * E-mail: (JL); (WG); (HL)
| |
Collapse
|
36
|
Mathi P, Nikhil K, Ambatipudi N, Roy P, Bokka VR, Botlagunta M. In-Vitro and in-Silico characterization of Sophora interrupta plant extract as an anticancer activity. Bioinformation 2014; 10:144-51. [PMID: 24748754 PMCID: PMC3974241 DOI: 10.6026/97320630010144] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2014] [Accepted: 03/06/2014] [Indexed: 11/23/2022] Open
Abstract
Sophora interrupta belongs to the family of Fabaceae and the species in this genus have a diverse medicinal importance as a folk
medicine for preventing many ailments including cancer. In order to evaluate the anticancer activity of S.interrupta, we have
performed in vitro anti-oxidant, anti-inflammatory, anti-proliferative, and cell based anticancer activity in MCF-7 and PC-3 cell
lines. Secondary metabolites of S.interrupta were used to identify anticancer compounds using Open Eye software. The antioxidant
activity of the S.interrupta root ethylacetate (SEA) extract at 100 µg/ml is equal to that of ascorbic acid at 50 µg/ml. The antiinflammatory
activity of SEA is half of that of diclofenac at 50 µg/ml. Anticancer activity was detected by measuring the
mitochondrial dehydrogenase activity (MTT assay). The half maximal inhibitory concentrations (IC50) for MCF-7 and PC-3 cell lines
are 250 and 700 µg/ml respectively. This was supported by the morphological changes such as membrane blebbing, cell
detachment and rounded cell morphology when compared to the parental cells. In addition, we observed few green cells (live)
over red cells (dead) based on the uptake of acridine orange and ethidium bromide dyes. Kaempferol-3-O-b-D-glucopyranoside, a
Secondary metabolite of S.interrupta form 6 hydrogen bond interactions with Arg 202, Gln 207, Gly 227, Gly 229, Thr 231 and Ala
232 human DEAD box RNA helicase, DDX3 protein and is equivalent to crystal structure of adenosine mono phosphate to DDX3.
Overall, it suggests that the SEA extract has anticancer compounds, and it can be used to enhance death receptor mediated cancer
cell death.
Collapse
Affiliation(s)
- Pardhasaradhi Mathi
- Biomedical research Laboratory, Department of Biotechnology, K L University, Green fields, Vaddeswaram, Guntur 522 502, AndhraPradesh, India
| | - Kumar Nikhil
- Molecular Endocrinology Laboratory, Department of Biotechnology, Indian Institute of Technology Roorkee, Roorkee 247 667, Uttarakhand, India
| | | | - Partha Roy
- Molecular Endocrinology Laboratory, Department of Biotechnology, Indian Institute of Technology Roorkee, Roorkee 247 667, Uttarakhand, India
| | - Venkata Raman Bokka
- Department of Basic Sciences-Chemistry, Madanapalle Institute of Technology and Science (MITS), Madanapalle 517 325, Chittoor District, Andhra Pradesh, India
| | - Mahendran Botlagunta
- Biomedical research Laboratory, Department of Biotechnology, K L University, Green fields, Vaddeswaram, Guntur 522 502, AndhraPradesh, India
| |
Collapse
|
37
|
Ethanolic Extract of Polish Propolis: Chemical Composition and TRAIL-R2 Death Receptor Targeting Apoptotic Activity against Prostate Cancer Cells. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2013; 2013:757628. [PMID: 24324518 PMCID: PMC3845518 DOI: 10.1155/2013/757628] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/10/2013] [Accepted: 09/18/2013] [Indexed: 12/03/2022]
Abstract
Propolis possesses chemopreventive properties through direct anticancer and indirect immunomodulatory activities. Tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) plays a significant role in immunosurveillance and defense against cancer cells. TRAIL triggers apoptosis upon binding to TRAIL-R1 (DR4) and TRAIL-R2 (DR5) death receptors expressed on cancer cell surface. The activation of TRAIL apoptotic signaling is considered an attractive option for cancer prevention. However, as more tumor cells are reported to be resistant to TRAIL-mediated death, it is important to develop new strategies to overcome this resistance. The aim of this study was to investigate the chemical composition and proapoptotic mechanism of ethanolic extract of Polish propolis (EEP-P) against cancer cells. The identification and quantification of phenolic compounds in propolis extract were performed using HPLC-DAD and UPLC-Q-TOF-MS methods. TRAIL-resistant LNCaP prostate cancer cells were treated with EEP-P and TRAIL. Cytotoxicity was measured by MTT and LDH assays. Apoptosis was detected using annexin V-FITC staining by flow cytometry and fluorescence microscopy. Death receptors expression was analyzed using flow cytometry. Pinobanksin, chrysin, methoxyflavanone, p-coumaric acid, ferulic acid and caffeic acid were the main phenolics found in EEP-P. Propolis sensitized LNCaP cells through upregulation of TRAIL-R2. These results suggest that EEP-P supports TRAIL-mediated immunochemoprevention in prostate cancer cells.
Collapse
|
38
|
Lim TG, Kim JE, Jung SK, Li Y, Bode AM, Park JS, Yeom MH, Dong Z, Lee KW. MLK3 is a direct target of biochanin A, which plays a role in solar UV-induced COX-2 expression in human keratinocytes. Biochem Pharmacol 2013; 86:896-903. [PMID: 23948065 PMCID: PMC4241970 DOI: 10.1016/j.bcp.2013.08.002] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2013] [Revised: 08/01/2013] [Accepted: 08/02/2013] [Indexed: 12/22/2022]
Abstract
Solar UV (sUV) is an important environmental carcinogen. Recent studies have shown that sUV is associated with numerous human skin disorders, such as wrinkle formation and inflammation. In this study, we found that the isoflavone, biochanin A, inhibited the expression of sUV-induced COX-2, which is a well-characterized sUV-induced enzyme, in both human HaCaT keratinocytes and JB6 P+ mouse skin epidermal cells. Several studies have demonstrated the beneficial effects of biochanin A. However, its direct molecular target is unknown. We found that biochanin A inhibited sUV-induced phosphorylation of MKK4/JNK/c-Jun and MKK3/6/p38/MSK1. Mixed-lineage kinase 3 (MLK3) is an upstream kinase of MKK4 and MKK3/6. Thus, we evaluated the effect of biochanin A on MLK3. We found that sUV-induced MLK3 phosphorylation was not affected, whereas MLK3 kinase activity was significantly suppressed by biochanin A. Furthermore, direct binding of biochanin A in the MLK3 ATP-binding pocket was detected using pull-down assays. Computer modeling supported our observation that MLK3 is a novel target of biochanin A. These results suggest that biochanin A exerts chemopreventive effects by suppressing sUV-induced COX-2 expression mediated through MLK3 inhibition.
Collapse
Affiliation(s)
- Tae-Gyu Lim
- The Hormel Institute, University of Minnesota, 801 16th Avenue NE, Austin, MN, 55912, USA
- Advanced Institutes of Convergence Technology, Seoul National University, Suwon, 443-270, Republic of Korea
- WCU Biomodulation Major, Department of Agricultural Biotechnology and Center for Food and Bioconvergence, Seoul National University, Seoul, 151-921, Republic of Korea
| | - Jong-Eun Kim
- The Hormel Institute, University of Minnesota, 801 16th Avenue NE, Austin, MN, 55912, USA
- Advanced Institutes of Convergence Technology, Seoul National University, Suwon, 443-270, Republic of Korea
- WCU Biomodulation Major, Department of Agricultural Biotechnology and Center for Food and Bioconvergence, Seoul National University, Seoul, 151-921, Republic of Korea
| | - Sung Keun Jung
- The Hormel Institute, University of Minnesota, 801 16th Avenue NE, Austin, MN, 55912, USA
- Advanced Institutes of Convergence Technology, Seoul National University, Suwon, 443-270, Republic of Korea
- Division of Metabolism and Functionality Research, Korea Food Research Institute, Seongnam, Republic of Korea
| | - Yan Li
- The Hormel Institute, University of Minnesota, 801 16th Avenue NE, Austin, MN, 55912, USA
| | - Ann M. Bode
- The Hormel Institute, University of Minnesota, 801 16th Avenue NE, Austin, MN, 55912, USA
| | - Jun-Seong Park
- Skin Research Institute, Amorepacific Corporation R&D Center, Yongin-si, Gyeonggi-do, 341-1, Republic of Korea
| | - Myeong Hun Yeom
- Skin Research Institute, Amorepacific Corporation R&D Center, Yongin-si, Gyeonggi-do, 341-1, Republic of Korea
| | - Zigang Dong
- The Hormel Institute, University of Minnesota, 801 16th Avenue NE, Austin, MN, 55912, USA
| | - Ki Won Lee
- Advanced Institutes of Convergence Technology, Seoul National University, Suwon, 443-270, Republic of Korea
- WCU Biomodulation Major, Department of Agricultural Biotechnology and Center for Food and Bioconvergence, Seoul National University, Seoul, 151-921, Republic of Korea
| |
Collapse
|
39
|
Szliszka E, Mertas A, Czuba ZP, Król W. Inhibition of Inflammatory Response by Artepillin C in Activated RAW264.7 Macrophages. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE : ECAM 2013; 2013:735176. [PMID: 23781267 PMCID: PMC3679718 DOI: 10.1155/2013/735176] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/04/2013] [Accepted: 05/06/2013] [Indexed: 11/17/2022]
Abstract
Artepillin C (3,5-diprenyl-4-hydroxycinnamic acid) is the main bioactive component of Brazilian green propolis. The aim of this study was to investigate the anti-inflammatory effect of artepillin C on LPS + IFN- γ - or PMA-stimulated RAW264.7 macrophages. The cell viability was evaluated by MTT and LDH assays. The radical scavenging ability was determined using DPPH(•) and ABTS(•+). ROS and RNS generation was analyzed by chemiluminescence. NO concentration was detected by the Griess reaction. The release of various cytokines by activated RAW264.7 cells was measured in the culture supernatants using a multiplex bead array system based on xMAP technology. NF- κ B activity was confirmed by the ELISA-based TransAM NF- κ B kit. At the tested concentrations, the compound did not decrease the cell viability and did not cause the cytotoxicity. Artepillin C exerted strong antioxidant activity, significantly inhibited the production of ROS, RNS, NO, and cytokine IL-1 β , IL-3, IL-4, IL-5, IL-9, IL-12p40, IL-13, IL-17, TNF- α , G-CSF, GM-CSF, MCP-1, MIP-1 α , MIP-1 β , RANTES, and KC, and markedly blocked NF- κ B expression in stimulated RAW264.7 macrophages. Our findings provide new insights for understanding the mechanism involved in the anti-inflammatory effect of artepillin C and support the application of Brazilian green propolis in complementary and alternative medicine.
Collapse
Affiliation(s)
- Ewelina Szliszka
- Department of Microbiology and Immunology, Medical University of Silesia in Katowice, Jordana 19, 41 808 Zabrze, Poland
| | - Anna Mertas
- Department of Microbiology and Immunology, Medical University of Silesia in Katowice, Jordana 19, 41 808 Zabrze, Poland
| | - Zenon P. Czuba
- Department of Microbiology and Immunology, Medical University of Silesia in Katowice, Jordana 19, 41 808 Zabrze, Poland
| | - Wojciech Król
- Department of Microbiology and Immunology, Medical University of Silesia in Katowice, Jordana 19, 41 808 Zabrze, Poland
| |
Collapse
|
40
|
Szliszka E, Krol W. Polyphenols Isolated from Propolis Augment TRAIL-Induced Apoptosis in Cancer Cells. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE : ECAM 2013; 2013:731940. [PMID: 23573148 PMCID: PMC3615595 DOI: 10.1155/2013/731940] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/07/2013] [Accepted: 02/04/2013] [Indexed: 11/17/2022]
Abstract
Epidemiological data support the concept that phenols and polyphenols in diet are safe and nontoxic, and have long-lasting beneficial effects on human health. The potential target for complementary and alternative medicine (CAM) research has been on the discovery of natural compounds that can be used in the prevention and treatment of cancer. Propolis is one of the richest sources of plant phenolics (flavonoids and phenolic acids). The ethanolic extract of propolis (EEP) and its polyphenols possess immunomodulatory, chemopreventive, and antitumor effects. Tumor necrosis factor-related apoptosis inducing ligand (TRAIL) is a naturally occurring anticancer agent that preferentially induces apoptosis in cancer cells and is not toxic to normal cells. Endogenous TRAIL plays a significant role in immunosurveillance and defense against cancer cells. However, as more tumor cells are reported to be resistant to TRAIL-mediated death, it is important to develop new strategies to overcome this resistance. EEP and polyphenols isolated from propolis have been shown to sensitize cancer cells to TRAIL-induced apoptosis. In this paper we demonstrate for the first time the crucial role of the main phenolics isolated from propolis in enhancing TRAIL-mediated death in tumor cells for cancer chemoprevention.
Collapse
Affiliation(s)
| | - Wojciech Krol
- Department of Microbiology and Immunology, Medical University of Silesia in Katowice, Jordana 19, 41 808 Zabrze, Poland
| |
Collapse
|
41
|
Tan JW, Tham CL, Israf DA, Lee SH, Kim MK. Neuroprotective effects of biochanin A against glutamate-induced cytotoxicity in PC12 cells via apoptosis inhibition. Neurochem Res 2012; 38:512-8. [PMID: 23224778 DOI: 10.1007/s11064-012-0943-6] [Citation(s) in RCA: 55] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2012] [Revised: 11/02/2012] [Accepted: 11/27/2012] [Indexed: 02/07/2023]
Abstract
L-Glutamate plays a crucial role in neuronal cell death, which is known to be associated with various neurodegenerative diseases, such as Alzheimer's, Parkinson's, and Huntington's diseases. In this study, we investigated the protective effects of biochanin A, a phytoestrogen compound found mainly in Trifolium pratense, against L-glutamate-induced cytotoxicity in a PC12 cell line. Exposure of the cells to 10 mM L-glutamate was found to significantly increase cell viability loss and apoptosis, whereas pretreatment with various concentrations of biochanin A attenuated the cytotoxic effects of L-glutamate. Specifically, the pretreatment led to not only decreases in the release of lactate dehydrogenase, the number of apoptotic cells, and the activity of caspase-3 but also an increase in the total glutathione level in the L-glutamate-treated PC12 cells. These results indicate that biochanin A may be able to exert neuroprotective effects against L-glutamate-induced cytotoxicity. Furthermore, our findings also imply that biochanin A may act as an antiapoptotic agent in order to perform its protective function.
Collapse
Affiliation(s)
- Ji Wei Tan
- Institute of Bioscience, Universiti Putra Malaysia, 43400, Serdang, Selangor, Malaysia
| | | | | | | | | |
Collapse
|
42
|
Szliszka E, Jaworska D, Ksek M, Czuba ZP, Król W. Targeting death receptor TRAIL-R2 by chalcones for TRAIL-induced apoptosis in cancer cells. Int J Mol Sci 2012. [PMID: 23203129 PMCID: PMC3509645 DOI: 10.3390/ijms131115343] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) induces apoptosis in cancer cells without toxicity to normal cells. TRAIL binds to death receptors, TRAIL-R1 (DR4) and TRAIL-R2 (DR5) expressed on cancer cell surface and activates apoptotic pathways. Endogenous TRAIL plays an important role in immune surveillance and defense against cancer cells. However, as more tumor cells are reported to be resistant to TRAIL mediated death, it is important to search for and develop new strategies to overcome this resistance. Chalcones can sensitize cancer cells to TRAIL-induced apoptosis. We examined the cytotoxic and apoptotic effects of TRAIL in combination with four chalcones: chalcone, isobavachalcone, licochalcone A and xanthohumol on HeLa cancer cells. The cytotoxicity was measured by MTT and LDH assays. The apoptosis was detected using annexin V-FITC staining by flow cytometry and fluorescence microscopy. Death receptor expression was analyzed using flow cytometry. The decreased expression of death receptors in cancer cells may be the cause of TRAIL-resistance. Chalcones enhance TRAIL-induced apoptosis in HeLa cells through increased expression of TRAIL-R2. Our study has indicated that chalcones augment the antitumor activity of TRAIL and confirm their cancer chemopreventive properties.
Collapse
Affiliation(s)
- Ewelina Szliszka
- Chair and Department of Microbiology and Immunology, Medical University of Silesia in Katowice, Jordana 19, 41-808 Zabrze, Poland.
| | | | | | | | | |
Collapse
|
43
|
Szliszka E, Kostrzewa-Susłow E, Bronikowska J, Jaworska D, Janeczko T, Czuba ZP, Krol W. Synthetic flavanones augment the anticancer effect of tumor necrosis factor-related apoptosis-inducing ligand (TRAIL). Molecules 2012; 17:11693-711. [PMID: 23027370 PMCID: PMC6268189 DOI: 10.3390/molecules171011693] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2012] [Revised: 09/03/2012] [Accepted: 09/24/2012] [Indexed: 12/12/2022] Open
Abstract
Tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) is considered as the most promising anticancer agent in the TNF superfamily because of its selective cytotoxicity against tumor cells versus normal primary cells. However, as more tumor cells are reported to be resistant to TRAIL-mediated death, it is important to develop new therapeutic strategies to overcome this resistance. Flavonoids have been shown to sensitize cancer cells to TRAIL-induced apoptosis. The aim of this study was to examine the cytotoxic and apoptotic activities of TRAIL on HeLa cancer cells in combination with two synthetic compounds: 6-hydroxyflavanone (6-HF) and its derivative 6-propionoxy-flavanone (6-PF) and to determine the mechanism by which the flavanones overcome the TRAIL-resistance. The cytotoxicity was measured by MTT and LDH assays. The apoptosis was detected by annexin V-FITC fluorescence staining in flow cytometry and microscopy. Death receptor (TRAIL-R1/DR4 and TRAIL-R2/DR5) expression were analysed using flow cytometry. Mitochondrial membrane potential was evaluated using DePsipher staining by fluorescence microscopy. The synthetic flavanones enhanced TRAIL-induced apoptosis in HeLa cells through increased expression of TRAIL-R2 death receptor and reduction of mitochondrial membrane potential. Our study indicates that the 6-HF and 6-PF augmented the anticancer effects of TRAIL and confirm a potential use of flavanones in TRAIL-based anticancer therapy and prevention.
Collapse
Affiliation(s)
- Ewelina Szliszka
- Department of Microbiology and Immunology, Medical University of Silesia, Katowice, Jordana 19, Zabrze 41-808, Poland; (E.S.); (J.B.); (D.J.); (Z.P.C.)
| | - Edyta Kostrzewa-Susłow
- Department of Chemistry, Wrocław University of Environmental and Life Sciences, Norwida 25, Wrocław 50-375, Poland; (E.K.-S.); (T.J.)
| | - Joanna Bronikowska
- Department of Microbiology and Immunology, Medical University of Silesia, Katowice, Jordana 19, Zabrze 41-808, Poland; (E.S.); (J.B.); (D.J.); (Z.P.C.)
| | - Dagmara Jaworska
- Department of Microbiology and Immunology, Medical University of Silesia, Katowice, Jordana 19, Zabrze 41-808, Poland; (E.S.); (J.B.); (D.J.); (Z.P.C.)
| | - Tomasz Janeczko
- Department of Chemistry, Wrocław University of Environmental and Life Sciences, Norwida 25, Wrocław 50-375, Poland; (E.K.-S.); (T.J.)
| | - Zenon P. Czuba
- Department of Microbiology and Immunology, Medical University of Silesia, Katowice, Jordana 19, Zabrze 41-808, Poland; (E.S.); (J.B.); (D.J.); (Z.P.C.)
| | - Wojciech Krol
- Department of Microbiology and Immunology, Medical University of Silesia, Katowice, Jordana 19, Zabrze 41-808, Poland; (E.S.); (J.B.); (D.J.); (Z.P.C.)
- Author to whom correspondence should be addressed; ; Tel./Fax: +48-322-722-554
| |
Collapse
|
44
|
Szliszka E, Zydowicz G, Mizgala E, Krol W. Artepillin C (3,5-diprenyl-4-hydroxycinnamic acid) sensitizes LNCaP prostate cancer cells to TRAIL-induced apoptosis. Int J Oncol 2012; 41:818-28. [PMID: 22735465 PMCID: PMC3582787 DOI: 10.3892/ijo.2012.1527] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2012] [Accepted: 05/18/2012] [Indexed: 12/04/2022] Open
Abstract
Naturally occurring phenolic compounds have been shown to sensitize prostate cancer cells to tumour necrosis factor-related apoptosis-inducing ligand (TRAIL)-induced apoptosis. TRAIL is a potent stimulator of apoptosis in cancer cells and an important immune effector molecule in the surveillance and elimination of developing tumours. However, many cancer cells are resistant to TRAIL-mediated death. In this study, we aimed to determine the mechanisms by which TRAIL resistance can be overcome in prostate cancer cells by 3,5-diprenyl-4-hydroxycinnamic acid (artepillin C). Artepillin C is a bioactive component of Brazilian green propolis that possesses antitumour and chemopreventive activities. TRAIL-resistant LNCaP prostate cancer cells were treated with TRAIL and artepillin C. Cytotoxicity was measured by MTT and lactate dehydrogenase (LDH) assays. Apoptosis was detected using Annexin V-FITC staining by flow cytometry and fluorescence microscopy. Death receptor (DR) (TRAIL-R1/DR4 and TRAIL-R2/DR5) expression was analyzed using flow cytometry. Mitochondrial membrane potential (ΔΨm) was evaluated using DePsipher staining by fluorescence micro scopy. The inhibition of NF-κB (p65) activation was confirmed with the ELISA-based TransAM NF-κB kit. Caspase-8 and caspase-3 activities were determined by colorimetric protease assays. The results showed that artepillin C sensitized the TRAIL-resistant LNCaP cells by engaging the extrinsic (receptor-mediated) and intrinsic (mitochondrial) apoptotic pathways. Artepillin C increased the expression of TRAIL-R2 and decreased the activity of NF-κB. Co-treatment with TRAIL and artepillin C induced the significant activation of caspase-8 and caspase-3, as well as the disruption of ΔΨm. These findings show that prostate cancer cells can be sensitized to TRAIL-mediated immunoprevention by artepillin C and confirm the role of phenolic compounds in prostate cancer immunochemoprevention.
Collapse
Affiliation(s)
- Ewelina Szliszka
- Department of Microbiology and Immunology, Medical University of Silesia in Katowice, Zabrze, Poland
| | | | | | | |
Collapse
|
45
|
The coumarin psoralidin enhances anticancer effect of tumor necrosis factor-related apoptosis-inducing ligand (TRAIL). Molecules 2012; 17:6449-64. [PMID: 22643355 PMCID: PMC6268812 DOI: 10.3390/molecules17066449] [Citation(s) in RCA: 70] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2012] [Revised: 05/15/2012] [Accepted: 05/21/2012] [Indexed: 11/22/2022] Open
Abstract
Coumarins are a very common type of secondary plant metabolites with a broad spectrum of biological activities. Psoralidin is a naturally occurring furanocoumarin isolated from Psoralea corylifolia possessing anticancer and chemopreventive properties. Tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) triggers apoptosis in cancer cells with no toxicity toward normal tissues. Endogenous TRAIL plays an important role in immune surveillance and defence against cancer cells. Coumarins can modulate TRAIL-mediated apoptosis in cancer cells. We examined the cytotoxic and apoptotic activities of psoralidin in combination with TRAIL on HeLa cancer cells. The cytotoxicity was measured by MTT and LDH assays. The apoptosis was detected using annexin V-FITC staining and mitochondrial membrane potential was evaluated using DePsipher staining by fluorescence microscopy. Death receptor (TRAIL-R1/DR4 and TRAIL-R2/DR5) expression was analyzed using flow cytometry. Psoralidin enhanced TRAIL-induced apoptosis in HeLa cells through increased expression of TRAIL-R2 death receptor and depolarization of mitochondrial membrane potential. Our study indicated that psoralidin augmented the anticancer effects of TRAIL and confirmed a potential use of coumarins in cancer chemoprevention.
Collapse
|