1
|
Tian CB, Qin ML, Qian YL, Qin SS, Shi ZQ, Zhao YL, Luo XD. Liver injury protection of Artemisia stechmanniana besser through PI3K/AKT pathway. JOURNAL OF ETHNOPHARMACOLOGY 2024; 334:118590. [PMID: 39029542 DOI: 10.1016/j.jep.2024.118590] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Revised: 07/02/2024] [Accepted: 07/15/2024] [Indexed: 07/21/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Artemisia stechmanniana Besser, one of the most prevalent botanical medicines in Chinese, has been traditionally used for hepatitis treatment. However, the bioactive components and pharmacological mechanism on alcohol-induced liver injury remains unclear. AIM OF THE STUDY To investigate the effect of A. stechmanniana on alcohol-induced liver damage, and further explore its mechanism. MATERIALS AND METHODS Phytochemical isolation and structural identification were used to determine the chemical constituents of A. stechmanniana. Then, the alcohol-induced liver damage animal and cell model were established to evaluate its hepato-protective potential. Network pharmacology, molecular docking and bioinformatics were integrated to explore the mechanism and then the prediction was further supported by experiments. Moreover, both compounds were subjected to ADMET prediction through relevant databases. RESULTS 28 compounds were isolated from the most bioactive fraction, ethyl acetate extract A. stechmanniana, in which five compounds (abietic acid, oplopanone, oplodiol, hydroxydavanone, linoleic acid) could attenuate mice livers damage caused by alcohol intragastration, reduce the degree of oxidative stress, and serum AST and ALT, respectively. Furthermore, abietic acid and hydroxydavanone exhibited best protective effect against alcohol-stimulated L-O2 cells injury among five bioactive compounds. Network pharmacology and bioinformatics analysis suggested that abietic acid and hydroxydavanone exhibiting drug likeliness characteristics, were the principal active compounds acting on liver injury treatment, primarily impacting to cell proliferation, oxidative stress and inflammation-related PI3K-AKT signaling pathways. Both of them displayed strong binding energies with five target proteins (HRAS, HSP90AA1, AKT1, CDK2, NF-κB p65) via molecular docking. Western blotting results further supported the predication with up-regulation of protein expressions of CDK2, and down-regulation of HRAS, HSP90AA1, AKT1, NF-κB p65 by abietic acid and hydroxydavanone. CONCLUSION Alcohol-induced liver injury protection by A. stechmanniana was verified in vivo and in vitro expanded its traditional use, and its two major bioactive compounds, abietic acid and hydroxydavanone exerted hepatoprotective effect through the regulation of PI3K-AKT signaling pathway.
Collapse
Affiliation(s)
- Cai-Bo Tian
- Yunnan Characteristic Plant Extraction Laboratory, Key Laboratory of Medicinal Chemistry for Natural Resource, Ministry of Education and Yunnan Province, School of Chemical Science and Technology, Yunnan University, Kunming, 650500, PR China
| | - Ma-Long Qin
- Yunnan Characteristic Plant Extraction Laboratory, Key Laboratory of Medicinal Chemistry for Natural Resource, Ministry of Education and Yunnan Province, School of Chemical Science and Technology, Yunnan University, Kunming, 650500, PR China
| | - Yan-Ling Qian
- Yunnan Characteristic Plant Extraction Laboratory, Key Laboratory of Medicinal Chemistry for Natural Resource, Ministry of Education and Yunnan Province, School of Chemical Science and Technology, Yunnan University, Kunming, 650500, PR China
| | - Shi-Shi Qin
- Yunnan Characteristic Plant Extraction Laboratory, Key Laboratory of Medicinal Chemistry for Natural Resource, Ministry of Education and Yunnan Province, School of Chemical Science and Technology, Yunnan University, Kunming, 650500, PR China
| | - Zhuo-Qi Shi
- Yunnan Characteristic Plant Extraction Laboratory, Key Laboratory of Medicinal Chemistry for Natural Resource, Ministry of Education and Yunnan Province, School of Chemical Science and Technology, Yunnan University, Kunming, 650500, PR China
| | - Yun-Li Zhao
- Yunnan Characteristic Plant Extraction Laboratory, Key Laboratory of Medicinal Chemistry for Natural Resource, Ministry of Education and Yunnan Province, School of Chemical Science and Technology, Yunnan University, Kunming, 650500, PR China.
| | - Xiao-Dong Luo
- Yunnan Characteristic Plant Extraction Laboratory, Key Laboratory of Medicinal Chemistry for Natural Resource, Ministry of Education and Yunnan Province, School of Chemical Science and Technology, Yunnan University, Kunming, 650500, PR China; State Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming, 650201, PR China.
| |
Collapse
|
2
|
Ray PP, Islam MA, Islam MS, Han A, Geng P, Aziz MA, Mamun AA. A comprehensive evaluation of the therapeutic potential of silibinin: a ray of hope in cancer treatment. Front Pharmacol 2024; 15:1349745. [PMID: 38487172 PMCID: PMC10937417 DOI: 10.3389/fphar.2024.1349745] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Accepted: 02/13/2024] [Indexed: 03/17/2024] Open
Abstract
Natural compounds hold promise in the search for cancer therapies due to their unique chemical structures and combinations that may effectively combat cancer while minimizing toxicity and side effects compared to conventional treatments. Silibinin, a natural lignan, has been found to possess strong anti-cancer activity against several types of human cancers based on emerging research. This study aims to provide an overview of the therapeutic potential of silibinin in the treatment and prevention of cancers. A comprehensive search was conducted using various internet databases such as PubMed, Google Scholar, and ScienceDirect to identify relevant research papers. Silibinin has been shown to exhibit anticancer activity against several types of cancers, including liver, lungs, breast, prostate, colorectal, skin, and bladder cancers. Its multifaceted mechanisms of action contribute to its therapeutic effects. Silibinin exerts antioxidant, anti-inflammatory, anti-proliferative, pro-apoptotic, anti-metastatic, and anti-angiogenic activities, making it a promising candidate for cancer therapy. One of the key mechanisms underlying the anticancer effects of silibinin is its ability to modulate multiple signaling pathways involved in cancer development and progression. It can inhibit the activation of various oncogenic pathways, including PI3K/Akt, NF-κB, Wnt/β-catenin, and MAPK pathways, thereby suppressing cancer cell proliferation, inducing cell cycle arrest, and promoting apoptosis. Silibinin possesses great potential as an effective treatment agent for cancer. The multifaceted mechanisms of action, favorable safety profile, and potential synergistic effects of silibinin with conventional therapies make it an attractive candidate for further investigation and development as a cancer treatment. However, more extensive clinical studies are necessary to fully establish the efficacy, optimal dosage, and long-term effects of silibinin in cancer treatment.
Collapse
Affiliation(s)
- Pantha Prodip Ray
- Department of Pharmacy, State University of Bangladesh, Dhaka, Bangladesh
| | | | - Mohammad Safiqul Islam
- Department of Pharmacy, Noakhali Science and Technology University, Noakhali, Bangladesh
| | - Aixia Han
- Key Laboratory of Joint Diagnosis and Treatment of Chronic Liver Disease and Liver Cancer of Lishui of The Sixth Affiliated Hospital of Wenzhou Medical University, Lishui People’s Hospital, Lishui, China
| | - Peiwu Geng
- Key Laboratory of Joint Diagnosis and Treatment of Chronic Liver Disease and Liver Cancer of Lishui of The Sixth Affiliated Hospital of Wenzhou Medical University, Lishui People’s Hospital, Lishui, China
| | - Md. Abdul Aziz
- Department of Pharmacy, State University of Bangladesh, Dhaka, Bangladesh
| | - Abdullah Al Mamun
- Key Laboratory of Joint Diagnosis and Treatment of Chronic Liver Disease and Liver Cancer of Lishui of The Sixth Affiliated Hospital of Wenzhou Medical University, Lishui People’s Hospital, Lishui, China
| |
Collapse
|
3
|
Hosseinzadeh A, Poursoleiman F, Biregani AN, Esmailzadeh A. Flavonoids target different molecules of autophagic and metastatic pathways in cancer cells. Cancer Cell Int 2023; 23:114. [PMID: 37308913 DOI: 10.1186/s12935-023-02960-4] [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: 03/12/2023] [Accepted: 05/30/2023] [Indexed: 06/14/2023] Open
Abstract
Despite the success of cancer therapy, it has encountered a major obstacle due to the complicated nature of cancer, namely resistance. The recurrence and metastasis of cancer occur when anti-cancer therapeutic agents fail to eradicate all cancer cells. Cancer therapy aims to find the best agent that targets all cancer cells, including those sensitive or resistant to treatment. Flavonoids, natural products from our diet, show anti-cancer effects in different studies. They can inhibit metastasis and the recurrence of cancers. This review discusses metastasis, autophagy, anoikis in cancer cells, and their dynamic relationship. We present evidence that flavonoids can block metastasis and induce cell death in cancer cells. Our research suggests that flavonoids can serve as potential therapeutic agents in cancer therapy.
Collapse
Affiliation(s)
- Aysooda Hosseinzadeh
- Department of Immunology, Faculty of Medicine, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Faezeh Poursoleiman
- Department of Cellular and Molecular Nutrition, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Akram Naghdipour Biregani
- Department of Nutrition, School of Health, Shahid Sadoughi University of Medical Scinences, Yazd, Iran
| | - Ahmad Esmailzadeh
- Students' Scientific Center, Tehran University of Medical Sciences, Tehran, Iran.
- Endocrinology and Metabolism Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran, Islamic Republic of Iran.
- Food Security Research Center, Department of Community Nutrition, School of Nutrition and Food Science, Isfahan University of Medical Sciences, Isfahan, Iran.
| |
Collapse
|
4
|
Estrogenic flavonoids and their molecular mechanisms of action. J Nutr Biochem 2023; 114:109250. [PMID: 36509337 DOI: 10.1016/j.jnutbio.2022.109250] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Revised: 12/02/2022] [Accepted: 12/07/2022] [Indexed: 12/13/2022]
Abstract
Flavonoids are a major group of phytoestrogens associated with physiological effects, and ecological and social impacts. Although the estrogenic activity of flavonoids was reported by researchers in the fields of medical, environmental and food studies, their molecular mechanisms of action have not been comprehensively reviewed. The estrogenic activity of the respective classes of flavonoids, anthocyanidins/anthocyanins, 2-arylbenzofurans/3-arylcoumarins/α-methyldeoxybenzoins, aurones/chalcones/dihydrochalcones, coumaronochromones, coumestans, flavans/flavan-3-ols/flavan-4-ols, flavanones/dihydroflavonols, flavones/flavonols, homoisoflavonoids, isoflavans, isoflavanones, isoflavenes, isoflavones, neoflavonoids, oligoflavonoids, pterocarpans/pterocarpenes, and rotenone/rotenoids, was summarized through a comprehensive literature search, and their structure-activity relationship, biological activities, signaling pathways, and applications were discussed. Although the respective classes of flavonoids contained at least one chemical mimicking estrogen, the mechanisms varied, such as those with estrogenic, anti-estrogenic, non-estrogenic, and biphasic activities, and additional activities through crosstalk/bypassing, which exert biological activities through cell signaling pathways. Such mechanistic variations of estrogen action are not limited to flavonoids and are observed among other broad categories of chemicals, thus this group of chemicals can be termed as the "estrogenome". This review article focuses on the connection of estrogen action mainly between the outer and the inner environments, which represent variations of chemicals and biological activities/signaling pathways, respectively, and form the basis to understand their applications. The applications of chemicals will markedly progress due to emerging technologies, such as artificial intelligence for precision medicine, which is also true of the study of the estrogenome including estrogenic flavonoids.
Collapse
|
5
|
Wang Y, Deng X, Liu Y, Wang Y, Luo X, Zhao T, Wang Z, Cheng G. Protective effect of Anneslea fragrans ethanolic extract against CCl4-induced liver injury by inhibiting inflammatory response, oxidative stress and apoptosis. Food Chem Toxicol 2023; 175:113752. [PMID: 37004906 DOI: 10.1016/j.fct.2023.113752] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2023] [Revised: 03/09/2023] [Accepted: 03/27/2023] [Indexed: 04/03/2023]
Abstract
Anneslea Fragrans Wall. (AF) is a medicinal and edible plant distributed in China. Its leaves and bark generally used for the treatments of diarrhea, fever, and liver diseases. While its ethnopharmacological application against liver diseases has not been fully studied. This study was aimed to evaluate the hepatoprotective effect of ethanolic extract from A. fragrans (AFE) on CCl4 induced liver injury in mice. The results showed that AFE could effectively reduce plasma activities of ALT and AST, increase antioxidant enzymes activities (SOD and CAT) and GSH level, and decrease MDA content in CCl4 induced mice. AFE effectively decreased the expressions of inflammatory cytokines (IL-1β, IL-6, TNF-α, COX-2 and iNOS), cell apoptosis-related proteins (Bax, caspase-3 and caspase-9) and increased Bcl-2 protein expression via inhibiting MAPK/ERK pathway. Additionally, TUNEL staining, Masson and Sirius red staining, immunohistochemical analyses revealed that AFE could inhibit the CCl4-induced hepatic fibrosis formation via reducing depositions of α-SMA, collagen I and collagen III. Conclusively, the present study demonstrated that AFE had an hepatoprotective effect by MAPK/ERK pathway to inhibit oxidative stress, inflammatory response and apoptosis in CCl4-induced liver injury mice, suggesting that AFE might be served as a hepatoprotective ingredient in the prevention and treatment of liver injury.
Collapse
Affiliation(s)
- Yudan Wang
- Faculty of Food Science and Engineering, Kunming University of Science and Technology, Kunming, 650500, China; National and Local Joint Engineering Research Center for Green Preparation Technology of Biobased Materials, Yunnan Minzu University, Kunming, 650500, China
| | - Xiaocui Deng
- Faculty of Food Science and Engineering, Kunming University of Science and Technology, Kunming, 650500, China
| | - Yaping Liu
- Faculty of Food Science and Engineering, Kunming University of Science and Technology, Kunming, 650500, China
| | - Yifen Wang
- Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, 650223, China
| | - Xiaodong Luo
- Key Laboratory of Medicinal Chemistry for Natural Resource, Ministry of Education and Yunnan Province, School of Chemical Science and Technology, Yunnan University, Kunming, 650091, China
| | - Tianrui Zhao
- Faculty of Food Science and Engineering, Kunming University of Science and Technology, Kunming, 650500, China
| | - Zhengxuan Wang
- Faculty of Food Science and Engineering, Kunming University of Science and Technology, Kunming, 650500, China.
| | - Guiguang Cheng
- Faculty of Food Science and Engineering, Kunming University of Science and Technology, Kunming, 650500, China.
| |
Collapse
|
6
|
Gogacz M, Peszke J, Natorska-Chomicka D, Ruszała M, Dos Santos Szewczyk K. Anticancer Effects of Propolis Extracts Obtained Using the Cold Separation Method on Breast Cancer Cell Lines. PLANTS (BASEL, SWITZERLAND) 2023; 12:884. [PMID: 36840233 PMCID: PMC9958691 DOI: 10.3390/plants12040884] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/26/2022] [Revised: 02/11/2023] [Accepted: 02/13/2023] [Indexed: 06/18/2023]
Abstract
Propolis and its extracts show a wide spectrum of biological activity. Due to the necessity to use high temperatures and high polarity in the eluent, the obtained extracts are depleted of active compounds. The new, cold separation method allows obtaining a qualitatively better product containing a number of chemical compounds absent in extracts obtained using high-temperature methods. The purpose of our study was to evaluate the biological activity of propolis extracts produced with the cold separation method in four female breast cancer cell lines: MDA-MB-231, MDA-MB-468, MCF-7, and T-47D. The results of the breast cancer cell viability were obtained using the MTT test. Propolis extracts at 75 and 80% showed similar cytotoxicity against cancer cells, with the polyphenol fraction 75% being slightly more negative for cells. Propolis extracts at concentrations of 50, 75, and 100 µg/mL significantly reduced cell viability. With the exception of the MDA-MB-231 line, cell viability was also decreased after incubation with a concentration of 25 µg/mL. Our results suggest that propolis extracts obtained with the cold separation method may be considered as promising compounds for the production of health-promoting supplements.
Collapse
Affiliation(s)
- Marek Gogacz
- Chair and Department of Gynecology, Medical University of Lublin, 20-090 Lublin, Poland
| | - Jerzy Peszke
- Department of Experimental Biotechnology, Decont LLC, 08-500 Ryki, Poland
| | - Dorota Natorska-Chomicka
- Chair and Department of Toxicology, Faculty of Pharmacy, Medical University of Lublin, 20-090 Lublin, Poland
| | - Monika Ruszała
- Chair and Department of Obstetrics and Perinatology, Medical University of Lublin, 20-090 Lublin, Poland
| | | |
Collapse
|
7
|
Jovankić JV, Nikodijević DD, Milutinović MG, Nikezić AG, Kojić VV, Cvetković AM, Cvetković DM. Potential of Orlistat to induce apoptotic and antiangiogenic effects as well as inhibition of fatty acid synthesis in breast cancer cells. Eur J Pharmacol 2023; 939:175456. [PMID: 36528070 DOI: 10.1016/j.ejphar.2022.175456] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2022] [Revised: 12/04/2022] [Accepted: 12/12/2022] [Indexed: 12/23/2022]
Abstract
Breast cancer as most often women's cancer is the second cause of mortality worldwide. Research interest increased in testing non-standard drugs to suppress breast cancer progression and become significant supplements in anticancer therapy. The anti-obesity drug Orlistat showed significant ability for modulation of cancer cell metabolism via antiproliferative, proapoptotic, antiangiogenic, antimetastatic, and hypolipidemic effects. The anticancer potential of Orlistat was evaluated by cytotoxicity (MTT assay), type of cell death (AO/EB double staining), determination of redox status parameters (superoxide, hydrogen peroxide, lipid peroxidation, reduced glutathione), and total lipid levels with colorimetric methods, as well on angiogenesis-related (VEGF, MMP-9, CXCR4/CXCL12) and fatty acid synthesis-related (ACLY, ACC, FASN) parameters on gene and protein levels (immunocytochemistry and qPCR). Based on obtained results Orlistat induces significant cytotoxic, proapoptotic, and anti-angiogenic effects in MDA-MB-231, MDA-MB-468 and MCF-7 breast cancer cells, without significant cytotoxic effects on normal MRC-5 cells. It decreased total lipid levels and changed redox status parameters and cancer cell metabolism via suppression of genes and proteins involved and fatty acid synthesis. Based on showed, Orlistat may be an important supplement in antiangiogenic therapy against breast cancer with no side effects on normal cells, making it a good candidate for future clinical trials.
Collapse
Affiliation(s)
- Jovana V Jovankić
- University of Kragujevac, Faculty of Science, Department of Biology and Ecology, Radoja Domanovića 12, 34000, Kragujevac, Serbia
| | - Danijela D Nikodijević
- University of Kragujevac, Faculty of Science, Department of Biology and Ecology, Radoja Domanovića 12, 34000, Kragujevac, Serbia
| | - Milena G Milutinović
- University of Kragujevac, Faculty of Science, Department of Biology and Ecology, Radoja Domanovića 12, 34000, Kragujevac, Serbia.
| | - Aleksandra G Nikezić
- University of Kragujevac, Faculty of Science, Department of Biology and Ecology, Radoja Domanovića 12, 34000, Kragujevac, Serbia
| | - Vesna V Kojić
- University of Novi Sad, Faculty of Medicine, Oncology Institute of Vojvodina, Put Dr Goldmana 4, Sremska Kamenica, 21204, Serbia
| | - Aleksandar M Cvetković
- University of Kragujevac, Faculty of Medical Sciences, Department of Surgery, Svetozara Markovića 69, 34000, Kragujevac, Serbia
| | - Danijela M Cvetković
- University of Kragujevac, Institute for Information Technologies Kragujevac, Department of Natural Sciences, Jovana Cvijića bb, 34000, Kragujevac, Serbia
| |
Collapse
|
8
|
Vachetta VS, Marder M, Troncoso MF, Elola MT. Opportunities, obstacles and current challenges of flavonoids for luminal and triple-negative breast cancer therapy. EUROPEAN JOURNAL OF MEDICINAL CHEMISTRY REPORTS 2022; 6:100077. [DOI: 10.1016/j.ejmcr.2022.100077] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/09/2025]
|
9
|
Solanki R, Jodha B, Prabina KE, Aggarwal N, Patel S. Recent advances in phytochemical based nano-drug delivery systems to combat breast cancer: A review. J Drug Deliv Sci Technol 2022. [DOI: 10.1016/j.jddst.2022.103832] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
10
|
Hussain A, Bourguet-Kondracki ML, Hussain F, Rauf A, Ibrahim M, Khalid M, Hussain H, Hussain J, Ali I, Khalil AA, Alhumaydhi FA, Khan M, Hussain R, Rengasamy KRR. The potential role of dietary plant ingredients against mammary cancer: a comprehensive review. Crit Rev Food Sci Nutr 2022; 62:2580-2605. [DOI: 10.1080/10408398.2020.1855413] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Affiliation(s)
- Amjad Hussain
- Department of Chemistry, University of Okara, Okara, Pakistan
- Laboratoire Molécules de Communication et Adaptation des Micro-organismes, UMR 7245 MNHN-CNRS, Muséum National d’Histoire Naturelle, Paris, France
- Department of Applied Chemistry, Government College University, Faisalabad, Pakistan
| | - Marie-Lise Bourguet-Kondracki
- Laboratoire Molécules de Communication et Adaptation des Micro-organismes, UMR 7245 MNHN-CNRS, Muséum National d’Histoire Naturelle, Paris, France
| | - Farhad Hussain
- Department of Applied Chemistry, Government College University, Faisalabad, Pakistan
| | - Abdur Rauf
- Department of Chemistry, University of Swabi, Swabi, Anbar, Khyber Pukhtanuk (KP), Pakistan
| | - Muhammad Ibrahim
- Department of Applied Chemistry, Government College University, Faisalabad, Pakistan
| | - Muhammad Khalid
- Department of Chemistry, Khwaja Fareed University of Engineering & Information Technology, Punjab, Pakistan
| | - Hidayat Hussain
- Department of Bioorganic Chemistry, Leibniz Institute of Plant Biochemistry, Halle (Salle), Germany
| | - Javid Hussain
- Department of Biological Sciences & Chemistry, College of Arts and Sciences, University of Nizwa, Nizwa, Sultanate of Oman
| | - Iftikhar Ali
- Department of Chemistry, Karakoram International University, Gilgit, Pakistan
| | - Anees Ahmed Khalil
- University Institute of Diet and Nutritional Sciences, Faculty of Allied Health Sciences, The University of Lahore, Lahore, Pakistan
| | - Fahad A. Alhumaydhi
- Department of Medical Laboratories, College of Applied Medical Sciences, Qassim University, Buraydah, Saudi Arabia
| | - Muhammad Khan
- Department of Chemistry, University of Okara, Okara, Pakistan
| | - Riaz Hussain
- Department of Chemistry, University of Okara, Okara, Pakistan
| | - Kannan R. R. Rengasamy
- Institute of Research and Development, Duy Tan University, Da Nang, Vietnam
- Faculty of Environment and Chemical Engineering, Duy Tan University, Da Nang, Vietnam
- Indigenous Knowledge Systems Centre, Faculty of Natural and Agricultural Sciences, North-West University, Mmabatho, South Africa
| |
Collapse
|
11
|
Bai Y, Chen J, Hu W, Wang L, Wu Y, Yu S. Silibinin Therapy Improves Cholangiocarcinoma Outcomes by Regulating ERK/Mitochondrial Pathway. Front Pharmacol 2022; 13:847905. [PMID: 35401195 PMCID: PMC8983842 DOI: 10.3389/fphar.2022.847905] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2022] [Accepted: 03/07/2022] [Indexed: 11/13/2022] Open
Abstract
Background: Silibinin is widely utilized drug in various cancer treatments, though its application in cholangiocarcinoma has not yet been explored. For the first time, we evaluated the anticancer potential and underlying molecular mechanism of silibinin in treatment of cholangiocarcinoma treatment. Methods: HuCCT-1 and CCLP-1 cells were chosen to be an in vitro study model and were exposed to various concentrations of silibinin for indicated times. Cell viability was evaluated by the cell counting kit-8 (CCK-8) assay and half maximal inhibitory (IC50) concentrations were calculated. Cell proliferation capacity was determined through the use of colony formation and 5-Ethynyl-2′- deoxyuridine (EdU) assays. Cell apoptosis and cycle arrest were assessed by Live/Dead staining assay and flow cytometry (FCM). The protein levels of extracellular regulated protein kinases (ERK)/mitochondrial apoptotic pathway were evaluated through western blotting (WB). Mitochondrial membrane potential changes were determined via 5,5′,6,6′-Tetrachloro-1,1′,3,3′-tetraethyl-imidacarbocyanine iodide (JC-1). A cholangiocarcinoma cell line xenograft model was used to assess the anti-tumor activity of silibinin in vivo. Results: Inhibition of the ERK protein by silibinin led to a significant decrease in mitochondrial membrane potential, which, in turn, caused Cytochrome C to be released from the mitochondria. The activation of downstream apoptotic pathways led to apoptosis of cholangiocarcinoma cells. In general, silibinin inhibited the growth of cholangiocarcinoma cell line xenograft tumors. Conclusions: Silibinin is able to inhibit cholangiocarcinoma through the ERK/mitochondrial apoptotic pathway, which makes silibinin a potential anti-tumor drug candidate for cholangiocarcinoma treatment.
Collapse
Affiliation(s)
- Yang Bai
- Department of Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Department of Hepatobiliary and Pancreatic Surgery, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua, China
| | - Jiaqi Chen
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Clinical Research Center for Oral Diseases of Zhejiang Province, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Hangzhou, China
| | - Weijian Hu
- Department of Hepatobiliary and Pancreatic Surgery, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua, China
| | - Lei Wang
- Department of Urology Surgery, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua, China
| | - Yulian Wu
- Department of Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- *Correspondence: Shi’an Yu, ; Yulian Wu,
| | - Shi’an Yu
- Department of Hepatobiliary and Pancreatic Surgery, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua, China
- *Correspondence: Shi’an Yu, ; Yulian Wu,
| |
Collapse
|
12
|
THE EFFECT OF QUERCETIN AND QUERCETIN-3-D-XYLOSIDE ON BREAST CANCER PROLIFERATION AND MIGRATION. JOURNAL OF BASIC AND CLINICAL HEALTH SCIENCES 2022. [DOI: 10.30621/jbachs.1056769] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
|
13
|
Liu R, Liu L, Bian Y, Zhang S, Wang Y, Chen H, Jiang X, Li G, Chen Q, Xue C, Li M, Liu L, Liu X, Ma S. The Dual Regulation Effects of ESR1/NEDD4L on SLC7A11 in Breast Cancer Under Ionizing Radiation. Front Cell Dev Biol 2022; 9:772380. [PMID: 35252218 PMCID: PMC8888677 DOI: 10.3389/fcell.2021.772380] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Accepted: 12/20/2021] [Indexed: 11/13/2022] Open
Abstract
Radiotherapy is one of the most important treatments for breast cancer. Ferroptosis is a recently recognized form of regulated cell death that is characterized by lipid peroxidation. However, whether ionizing radiation (IR) could induce ferroptosis in breast cancer and how it works remain unknown. Bioinformatics analysis were performed to screen ferroptosis-related genes differentially expressed in breast tumor tissue and normal tissue. Then, breast cancer cell lines with different estrogen receptor (ER) phenotypes were used for studies in vitro, including ER-positive (MCF-7 and ZR-75-1) and ER-negative (MDA-MB-231) cells. The dynamic changes of mRNA and protein levels were examined after x-ray of 8 Gy by qRT-PCR and Western blotting, respectively. Immunoprecipitation (IP) was used to explore the interaction between proteins. Luciferase assay was used to analyze the transcriptional regulation effect of ESR1 on SLC7A11. BODIPY C11 and trypan blue dyes were used to determine lipid peroxidation and cell death, respectively. The result showed that the ferroptosis-related gene SLC7A11 was higher in breast cancer tissues compared with normal tissues and associated with poor survival. A positive correlation exists between ESR1 and SLC7A11 expression. ESR1 promoted SLC7A11 expression at the early stage after IR. ESR1/SLC7A11 knockdown significantly enhanced IR-induced ferroptosis in ER-positive cells. At 12 h after IR, the IP data showed the interaction between E3 ubiquitin ligase NEDD4L and SLC7A11 increased, followed by the ubiquitylation and degradation of SLC7A11. Thus, SLC7A11 expression was regulated by both ESR1 and NEDD4L, in opposite ways. For the first time, we elucidated that ESR1 and NEDD4L functioned together after radiation treatment and finally induced ferroptosis in breast cancer cells, which provides novel insight into the guidance of clinical treatment of breast cancer.
Collapse
Affiliation(s)
- Rui Liu
- School of Public Health and Management, Wenzhou Medical University, Wenzhou, China.,NHC Key Laboratory of Radiobiology, School of Public Health of Jilin University, Changchun, China
| | - Lin Liu
- School of Public Health and Management, Wenzhou Medical University, Wenzhou, China
| | - Yan Bian
- School of Public Health and Management, Wenzhou Medical University, Wenzhou, China
| | - Shinan Zhang
- School of Public Health and Management, Wenzhou Medical University, Wenzhou, China
| | - Yue Wang
- School of Public Health and Management, Wenzhou Medical University, Wenzhou, China
| | - Huajian Chen
- School of Public Health and Management, Wenzhou Medical University, Wenzhou, China
| | - Xinyue Jiang
- School of Public Health and Management, Wenzhou Medical University, Wenzhou, China
| | - Guanghui Li
- School of Public Health and Management, Wenzhou Medical University, Wenzhou, China
| | - Qing Chen
- School of Public Health and Management, Wenzhou Medical University, Wenzhou, China
| | - Chang Xue
- School of Public Health and Management, Wenzhou Medical University, Wenzhou, China
| | - Mengke Li
- School of Public Health and Management, Wenzhou Medical University, Wenzhou, China
| | - Lianchang Liu
- NHC Key Laboratory of Radiobiology, School of Public Health of Jilin University, Changchun, China.,The Second Affiliated Hospital of Jilin University, Changchun, China
| | - Xiaodong Liu
- School of Public Health and Management, Wenzhou Medical University, Wenzhou, China.,Key Laboratory of Watershed Science and Health of Zhejiang Province, Wenzhou Medical University, Wenzhou, China
| | - Shumei Ma
- School of Public Health and Management, Wenzhou Medical University, Wenzhou, China.,NHC Key Laboratory of Radiobiology, School of Public Health of Jilin University, Changchun, China.,Key Laboratory of Watershed Science and Health of Zhejiang Province, Wenzhou Medical University, Wenzhou, China
| |
Collapse
|
14
|
Koltai T, Fliegel L. Role of Silymarin in Cancer Treatment: Facts, Hypotheses, and Questions. J Evid Based Integr Med 2022; 27:2515690X211068826. [PMID: 35018864 PMCID: PMC8814827 DOI: 10.1177/2515690x211068826] [Citation(s) in RCA: 44] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2021] [Revised: 10/20/2021] [Accepted: 12/06/2021] [Indexed: 12/14/2022] Open
Abstract
The flavonoid silymarin extracted from the seeds of Sylibum marianum is a mixture of 6 flavolignan isomers. The 3 more important isomers are silybin (or silibinin), silydianin, and silychristin. Silybin is functionally the most active of these compounds. This group of flavonoids has been extensively studied and they have been used as hepato-protective substances for the mushroom Amanita phalloides intoxication and mainly chronic liver diseases such as alcoholic cirrhosis and nonalcoholic fatty liver. Hepatitis C progression is not, or slightly, modified by silymarin. Recently, it has also been proposed for SARS COVID-19 infection therapy. The biochemical and molecular mechanisms of action of these substances in cancer are subjects of ongoing research. Paradoxically, many of its identified actions such as antioxidant, promoter of ribosomal synthesis, and mitochondrial membrane stabilization, may seem protumoral at first sight, however, silymarin compounds have clear anticancer effects. Some of them are: decreasing migration through multiple targeting, decreasing hypoxia inducible factor-1α expression, inducing apoptosis in some malignant cells, and inhibiting promitotic signaling among others. Interestingly, the antitumoral activity of silymarin compounds is limited to malignant cells while the nonmalignant cells seem not to be affected. Furthermore, there is a long history of silymarin use in human diseases without toxicity after prolonged administration. The ample distribution and easy accessibility to milk thistle-the source of silymarin compounds, its over the counter availability, the fact that it is a weed, some controversial issues regarding bioavailability, and being a nutraceutical rather than a drug, has somehow led medical professionals to view its anticancer effects with skepticism. This is a fundamental reason why it never achieved bedside status in cancer treatment. However, in spite of all the antitumoral effects, silymarin actually has dual effects and in some cases such as pancreatic cancer it can promote stemness. This review deals with recent investigations to elucidate the molecular actions of this flavonoid in cancer, and to consider the possibility of repurposing it. Particular attention is dedicated to silymarin's dual role in cancer and to some controversies of its real effectiveness.
Collapse
Affiliation(s)
- Tomas Koltai
- Hospital del Centro Gallego de Buenos Aires, Buenos Aires, Argentina
| | | |
Collapse
|
15
|
Brimson JM, Prasanth MI, Malar DS, Thitilertdecha P, Kabra A, Tencomnao T, Prasansuklab A. Plant Polyphenols for Aging Health: Implication from Their Autophagy Modulating Properties in Age-Associated Diseases. Pharmaceuticals (Basel) 2021; 14:ph14100982. [PMID: 34681206 PMCID: PMC8538309 DOI: 10.3390/ph14100982] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2021] [Revised: 09/13/2021] [Accepted: 09/19/2021] [Indexed: 02/06/2023] Open
Abstract
Polyphenols are a family of naturally occurring organic compounds, majorly present in fruits, vegetables, and cereals, characterised by multiple phenol units, including flavonoids, tannic acid, and ellagitannin. Some well-known polyphenols include resveratrol, quercetin, curcumin, epigallocatechin gallate, catechin, hesperetin, cyanidin, procyanidin, caffeic acid, and genistein. They can modulate different pathways inside the host, thereby inducing various health benefits. Autophagy is a conserved process that maintains cellular homeostasis by clearing the damaged cellular components and balancing cellular survival and overall health. Polyphenols could maintain autophagic equilibrium, thereby providing various health benefits in mediating neuroprotection and exhibiting anticancer and antidiabetic properties. They could limit brain damage by dismantling misfolded proteins and dysfunctional mitochondria, thereby activating autophagy and eliciting neuroprotection. An anticarcinogenic mechanism is stimulated by modulating canonical and non-canonical signalling pathways. Polyphenols could also decrease insulin resistance and inhibit loss of pancreatic islet β-cell mass and function from inducing antidiabetic activity. Polyphenols are usually included in the diet and may not cause significant side effects that could be effectively used to prevent and treat major diseases and ailments.
Collapse
Affiliation(s)
- James Michael Brimson
- Natural Products for Neuroprotection and Anti-Ageing Research Unit, Chulalongkorn University, Bangkok 10330, Thailand; (J.M.B.); (M.I.P.); (D.S.M.)
- Department of Clinical Chemistry, Faculty of Allied Health Sciences, Chulalongkorn University, Bangkok 10330, Thailand
| | - Mani Iyer Prasanth
- Natural Products for Neuroprotection and Anti-Ageing Research Unit, Chulalongkorn University, Bangkok 10330, Thailand; (J.M.B.); (M.I.P.); (D.S.M.)
- Department of Clinical Chemistry, Faculty of Allied Health Sciences, Chulalongkorn University, Bangkok 10330, Thailand
| | - Dicson Sheeja Malar
- Natural Products for Neuroprotection and Anti-Ageing Research Unit, Chulalongkorn University, Bangkok 10330, Thailand; (J.M.B.); (M.I.P.); (D.S.M.)
- Department of Clinical Chemistry, Faculty of Allied Health Sciences, Chulalongkorn University, Bangkok 10330, Thailand
| | - Premrutai Thitilertdecha
- Siriraj Research Group in Immunobiology and Therapeutic Sciences, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok 10330, Thailand;
| | - Atul Kabra
- Department of Pharmacology, University Institute of Pharma Sciences, Chandigarh University, Sahibzad Ajit Singh Nagar 140413, Punjab, India;
| | - Tewin Tencomnao
- Natural Products for Neuroprotection and Anti-Ageing Research Unit, Chulalongkorn University, Bangkok 10330, Thailand; (J.M.B.); (M.I.P.); (D.S.M.)
- Department of Clinical Chemistry, Faculty of Allied Health Sciences, Chulalongkorn University, Bangkok 10330, Thailand
- Correspondence: (T.T.); (A.P.)
| | - Anchalee Prasansuklab
- Natural Products for Neuroprotection and Anti-Ageing Research Unit, Chulalongkorn University, Bangkok 10330, Thailand; (J.M.B.); (M.I.P.); (D.S.M.)
- College of Public Health Sciences, Chulalongkorn University, Bangkok 10330, Thailand
- Correspondence: (T.T.); (A.P.)
| |
Collapse
|
16
|
Tuli HS, Mittal S, Aggarwal D, Parashar G, Parashar NC, Upadhyay SK, Barwal TS, Jain A, Kaur G, Savla R, Sak K, Kumar M, Varol M, Iqubal A, Sharma AK. Path of Silibinin from diet to medicine: A dietary polyphenolic flavonoid having potential anti-cancer therapeutic significance. Semin Cancer Biol 2021; 73:196-218. [PMID: 33130037 DOI: 10.1016/j.semcancer.2020.09.014] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2020] [Revised: 09/11/2020] [Accepted: 09/29/2020] [Indexed: 02/07/2023]
Abstract
In the last few decades, targeting cancer by the use of dietary phytochemicals has gained enormous attention. The plausible reason and believe or mind set behind this fact is attributed to either lesser or no side effects of natural compounds as compared to the modern chemotherapeutics, or due to their conventional use as dietary components by mankind for thousands of years. Silibinin is a naturally derived polyphenol (a flavonolignans), possess following biochemical features; molecular formula C25H22O10, Molar mass: 482.44 g/mol, Boiling point 793 °C, with strikingly high antioxidant and anti-tumorigenic properties. The anti-cancer properties of Silibinin are determined by a variety of cellular pathways which include induction of apoptosis, cell cycle arrest, inhibition of angiogenesis and metastasis. In addition, Silibinin controls modulation of the expression of aberrant miRNAs, inflammatory response, and synergism with existing anti-cancer drugs. Therefore, modulation of a vast array of cellular responses and homeostatic aspects makes Silibinin an attractive chemotherapeutic agent. However, like other polyphenols, the major hurdle to declare Silibinin a translational chemotherapeutic agent, is its lesser bioavailability. After summarizing the chemistry and metabolic aspects of Silibinin, this extensive review focuses on functional aspects governed by Silibinin in chemoprevention with an ultimate goal of summarizing the evidence supporting the chemopreventive potential of Silibinin and clinical trials that are currently ongoing, at a single platform.
Collapse
Affiliation(s)
- Hardeep Singh Tuli
- Department of Biotechnology, Maharishi Markandeshwar (Deemed to be University), Mullana-Ambala, 133 207, Haryana, India
| | - Sonam Mittal
- School of Biotechnology, Jawaharlal Nehru University, New Delhi, India
| | - Diwakar Aggarwal
- Department of Biotechnology, Maharishi Markandeshwar (Deemed to be University), Mullana-Ambala, 133 207, Haryana, India
| | - Gaurav Parashar
- Department of Biotechnology, Maharishi Markandeshwar (Deemed to be University), Mullana-Ambala, 133 207, Haryana, India
| | | | - Sushil Kumar Upadhyay
- Department of Biotechnology, Maharishi Markandeshwar (Deemed to be University), Mullana-Ambala, 133 207, Haryana, India
| | - Tushar Singh Barwal
- Department of Zoology, Central University of Punjab, Bathinda, 151 001, Punjab, India
| | - Aklank Jain
- Department of Zoology, Central University of Punjab, Bathinda, 151 001, Punjab, India
| | - Ginpreet Kaur
- Department of Pharmacology, Shobhaben Pratapbhai Patel School of Pharmacy and Technology Management, SVKM's, NMIMS, Mumbai, 400 056, Maharastra, India
| | - Raj Savla
- Department of Pharmacology, Shobhaben Pratapbhai Patel School of Pharmacy and Technology Management, SVKM's, NMIMS, Mumbai, 400 056, Maharastra, India
| | | | - Manoj Kumar
- Department of Chemistry, Maharishi Markandeshwar University, Sadopur, India
| | - Mehmet Varol
- Department of Molecular Biology and Genetics, Faculty of Science, Mugla Sitki Kocman University, Mugla, TR48000, Turkey
| | - Ashif Iqubal
- Department of Pharmacology, School of Pharmaceutical Education and Research (Formerly Faculty of Pharmacy), Jamia Hamdard (Deemed to be University), Delhi, India
| | - Anil Kumar Sharma
- Department of Biotechnology, Maharishi Markandeshwar (Deemed to be University), Mullana-Ambala, 133 207, Haryana, India.
| |
Collapse
|
17
|
Binienda A, Ziolkowska S, Pluciennik E. The Anticancer Properties of Silibinin: Its Molecular Mechanism and Therapeutic Effect in Breast Cancer. Anticancer Agents Med Chem 2021; 20:1787-1796. [PMID: 31858905 DOI: 10.2174/1871520620666191220142741] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2019] [Revised: 10/29/2019] [Accepted: 11/12/2019] [Indexed: 02/08/2023]
Abstract
BACKGROUND Silibinin (SB), the main component of Silymarin (SM), is a natural substance obtained from the seeds of the milk thistle. SM contains up to 70% of SB as two isoforms: A and B. It has an antioxidant and anti-inflammatory effect on hepatocytes and is known to inhibit cell proliferation, induce apoptosis, and curb angiogenesis. SB has demonstrated activity against many cancers, such as skin, liver, lung, bladder, and breast carcinomas. METHODS This review presents current knowledge of the use of SM in breast cancer, this being one of the most common types of cancer in women. It describes selected molecular mechanisms of the action of SM; for example, although SB influences both Estrogen Receptors (ER), α and β, it has opposite effects on the two. Its action on ERα influences the PI3K/AKT/mTOR and RAS/ERK signaling pathways, while by up-regulating ERβ, it increases the numbers of apoptotic cells. In addition, ERα is involved in SB-induced autophagy, while ERβ is not. Interestingly, SB also inhibits metastasis by suppressing TGF-β2 expression, thus suppressing Epithelial to Mesenchymal Transition (EMT). It also influences migration and invasive potential via the Jak2/STAT3 pathway. RESULTS SB may be a promising enhancement of BC treatment: when combined with chemotherapeutic drugs such as carboplatin, cisplatin, and doxorubicin, the combination exerts a synergistic effect against cancer cells. This may be of value when treating aggressive types of mammary carcinoma. CONCLUSION Summarizing, SB inhibits proliferation, induces apoptosis, and restrains metastasis via several mechanisms. It is possible to combine SB with different anticancer drugs, an approach that represents a promising therapeutic strategy for patients suffering from BC.
Collapse
Affiliation(s)
- Agata Binienda
- Faculty of Biomedical Sciences and Postgraduate Education, Medical University of Lodz, Lodz, Poland
| | - Sylwia Ziolkowska
- Faculty of Biomedical Sciences and Postgraduate Education, Medical University of Lodz, Lodz, Poland
| | - Elzbieta Pluciennik
- Department of Molecular Carcinogenesis, Medical University of Lodz, Lodz, Poland
| |
Collapse
|
18
|
Daphnane-type diterpenes from genus Daphne and their anti-tumor activity. CHINESE HERBAL MEDICINES 2021; 13:145-156. [PMID: 36117500 PMCID: PMC9476389 DOI: 10.1016/j.chmed.2020.09.006] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Revised: 08/15/2020] [Accepted: 09/01/2020] [Indexed: 11/22/2022] Open
Abstract
Daphnane-type diterpenenoids are the major biologically active constituents in the genus Daphne. We find that there are about 101 Daphnane-type diterpenes in this genus, most of those compounds show different degrees of inhibitory effect on various cancer cell. Some of them have been studied in depth and the potent molecular mechanisms might be associated with modulation of different cell-signaling pathways. In addition, some compounds of this type also can inhibit the synthesis of protein and DNA. Absolutely, the anti-tumor activity of Daphnane-type diterpenes is worthy of attention. Unfortunately, most of the current research on the activity of these compounds is focused on simple drug efficacy, and its in-depth mechanism research is far from enough. On the other point of view, there still exists wide growing space on the depth of these compounds.
Collapse
|
19
|
Liu W, Wang F, Li C, Otkur W, Hayashi T, Mizuno K, Hattori S, Fujisaki H, Onodera S, Ikejima T. Silibinin treatment protects human skin cells from UVB injury through upregulation of estrogen receptors. JOURNAL OF PHOTOCHEMISTRY AND PHOTOBIOLOGY B-BIOLOGY 2021; 216:112147. [PMID: 33561689 DOI: 10.1016/j.jphotobiol.2021.112147] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/21/2020] [Revised: 11/27/2020] [Accepted: 01/28/2021] [Indexed: 02/07/2023]
Abstract
Ultraviolet B (UVB) from the sunlight is a major environmental cause for human skin damages, inducing cell death, inflammation, senescence and even carcinogenesis. The natural flavonoid silibinin, clinically used as liver protectant, has protective effects against UVB-caused skin injury in vivo and in vitro. Silibinin is often classified as a phytoestrogen, because it modulates the activation of estrogen receptors (ERs). However, whether silibinin's estrogenic effect contributes to the skin protection against UVB injury remains to be elucidated. The issue was explored in this study by using the human foreskin dermal fibroblasts (HFF) and human non-malignant immortalized keratinocytes (HaCaT). In HFF, pre-treatment with silibinin rescued UVB-irradiated cells from apoptosis. Interestingly, silibinin increased the whole cellular and nuclear levels of ERα and ERβ in UVB-irradiated cells. Activation of ERs by treatment with estradiol elevated the cell survival and reduced apoptosis in UVB-treated cells. ERα agonist increased cell survival, while its antagonist decreased it. ERβ agonist also increased cell survival, but the antagonist had no effect on cell survival. Transfection of the cells with the small interfering RNAs (si-RNAs) to ERα or ERβ diminished the protective effect of silibinin on UVB-irradiated cells. In UVB-treated HaCaT cells, both ERα and ERβ were increased by silibinin treatment. Inhibition of activation and expression of ERα or ERβ by specific antagonists and si-RNAs, respectively, reduced cell survival in UVB-treated HaCaT cells regardless of silibinin treatment. Taken together, it is summarized that silibinin up-regulates both ERα and ERβ pathways in UVB-treated dermal HFF cells and epidermal HaCaT cells, leading to protection of skin from UVB-damage.
Collapse
Affiliation(s)
- Weiwei Liu
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, Liaoning, China
| | - Fang Wang
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, Liaoning, China
| | - Can Li
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, Liaoning, China
| | - Wuxiyar Otkur
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, Liaoning, China; CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, China
| | - Toshihiko Hayashi
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, Liaoning, China; Department of Chemistry and Life science, School of Advanced Engineering, Kogakuin University, 2665-1, Nakanomachi, Hachioji, Tokyo 192-0015, Japan; Nippi Research Institute of Biomatrix, Toride, Ibaraki 302-0017, Japan
| | - Kazunori Mizuno
- Nippi Research Institute of Biomatrix, Toride, Ibaraki 302-0017, Japan
| | - Shunji Hattori
- Nippi Research Institute of Biomatrix, Toride, Ibaraki 302-0017, Japan
| | - Hitomi Fujisaki
- Nippi Research Institute of Biomatrix, Toride, Ibaraki 302-0017, Japan
| | - Satoshi Onodera
- Medical Research Institute of Curing Mibyo, 1-6-28 Narusedai Mechida Tokyo, 194-0042, Japan
| | - Takashi Ikejima
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, Liaoning, China; Key Laboratory of Computational Chemistry-Based Natural Antitumor Drug Research & Development, Shenyang Pharmaceutical University, Shenyang 110016, Liaoning, China.
| |
Collapse
|
20
|
Kaipa JM, Starkuviene V, Erfle H, Eils R, Gladilin E. Transcriptome profiling reveals Silibinin dose-dependent response network in non-small lung cancer cells. PeerJ 2020; 8:e10373. [PMID: 33362957 PMCID: PMC7749657 DOI: 10.7717/peerj.10373] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2020] [Accepted: 10/26/2020] [Indexed: 12/20/2022] Open
Abstract
Silibinin (SIL), a natural flavonolignan from the milk thistle (Silybum marianum), is known to exhibit remarkable hepatoprotective, antineoplastic and EMT inhibiting effects in different cancer cells by targeting multiple molecular targets and pathways. However, the predominant majority of previous studies investigated effects of this phytocompound in a one particular cell line. Here, we carry out a systematic analysis of dose-dependent viability response to SIL in five non-small cell lung cancer (NSCLC) lines that gradually differ with respect to their intrinsic EMT stage. By correlating gene expression profiles of NSCLC cell lines with the pattern of their SIL IC50 response, a group of cell cycle, survival and stress responsive genes, including some prominent targets of STAT3 (BIRC5, FOXM1, BRCA1), was identified. The relevancy of these computationally selected genes to SIL viability response of NSCLC cells was confirmed by the transient knockdown test. In contrast to other EMT-inhibiting compounds, no correlation between the SIL IC50 and the intrinsic EMT stage of NSCLC cells was observed. Our experimental results show that SIL viability response of differently constituted NSCLC cells is linked to a subnetwork of tightly interconnected genes whose transcriptomic pattern can be used as a benchmark for assessment of individual SIL sensitivity instead of the conventional EMT signature. Insights gained in this study pave the way for optimization of customized adjuvant therapy of malignancies using Silibinin.
Collapse
Affiliation(s)
- Jagan Mohan Kaipa
- Helmholtz Center for Infection Research, Braunschweig, Germany.,BioQuant, University Heidelberg, Heidelberg, Germany.,Theoretical Bioinformatics, German Cancer Research Center, Heidelberg, Germany
| | - Vytaute Starkuviene
- BioQuant, University Heidelberg, Heidelberg, Germany.,Institute of Biosciences, Vilnius University Life Science Center, Vilnius, Lithuania
| | - Holger Erfle
- BioQuant, University Heidelberg, Heidelberg, Germany
| | - Roland Eils
- Center for Digital Health, Berlin Institute of Health and Charité Universitätsmedizin Berlin, Berlin, Germany.,Health Data Science Unit, Heidelberg University Hospital, Heidelberg, Germany
| | - Evgeny Gladilin
- BioQuant, University Heidelberg, Heidelberg, Germany.,Leibniz Institute of Plant Genetics and Crop Plant Research, Seeland, Germany.,Applied Bioinformatics, German Cancer Research Center, Heidelberg, Germany
| |
Collapse
|
21
|
Mashhadi Akbar Boojar M, Mashhadi Akbar Boojar M, Golmohammad S. Overview of Silibinin anti-tumor effects. J Herb Med 2020. [DOI: 10.1016/j.hermed.2020.100375] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
|
22
|
Liu W, Ji Y, Sun Y, Si L, Fu J, Hayashi T, Onodera S, Ikejima T. Estrogen receptors participate in silibinin-caused nuclear translocation of apoptosis-inducing factor in human breast cancer MCF-7 cells. Arch Biochem Biophys 2020; 689:108458. [DOI: 10.1016/j.abb.2020.108458] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2020] [Revised: 06/02/2020] [Accepted: 06/04/2020] [Indexed: 02/08/2023]
|
23
|
Wang J, Zhang X, Zhang L, Yan T, Wu B, Xu F, Jia Y. Silychristin A activates Nrf2-HO-1/SOD2 pathway to reduce apoptosis and improve GLP-1 production through upregulation of estrogen receptor α in GLUTag cells. Eur J Pharmacol 2020; 881:173236. [PMID: 32497626 DOI: 10.1016/j.ejphar.2020.173236] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2020] [Revised: 05/21/2020] [Accepted: 05/29/2020] [Indexed: 01/08/2023]
Abstract
Glucagon-like peptide-1 (GLP-1), a glucagon-like peptide secreted mainly from intestinal L cells, possesses the functions of promoting synthesis and secretion of insulin in pancreatic β-cells, and maintaining glucose homeostasis in an insulin-independent manner. Silychristin A, a major flavonolignan from silymarin, was reported to protect pancreatic β-cells from oxidative damage in streptozotocin (STZ)-induced diabetic rats. However, the role of silychristin A in the protection of intestinal L-cells is still unknown. Our current study demonstrated that palmitate (PA) inhibited protein expression of NF-E2-related factor 2 (Nrf2), heme oxygenase-1 (HO-1) and superoxide dismutase 2 (SOD2), and subsequently increased reactive oxygen species level to induce apoptosis and decrease GLP-1 content in intestinal L-cell line GLUTag cells. Pre-incubation of silychristin A effectively reversed PA-inactivated Nrf2-HO-1/SOD2 antioxidative pathway accompanied with decreased apoptosis level and increased GLP-1 level in GLUTag cells. As a potential target of silychristin A, estrogen receptor α was shown to be downregulated by PA stimulation, and the expression of which was improved by silychristin A in a concentration-dependent manner. Further study revealed that the treatment of estrogen receptor α antagonist MPP induced apoptosis and blocked the stimulation of GLP-1 production by silychristin A through the activation of Nrf2-HO-1/SOD2 pathway in GLUTag cells. Taken together, our study found silychristin A activated estrogen receptor α-dependent Nrf2-HO-1/SOD2 pathway to decrease apoptosis and upregulate GLP-1 production in GLUTag cells.
Collapse
Affiliation(s)
- Jinyu Wang
- School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, Shenyang, 110016, PR China
| | - Xiaoying Zhang
- School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, Shenyang, 110016, PR China
| | - Luxin Zhang
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, 110016, PR China
| | - Tingxu Yan
- Faculty of Functional Food and Wine, Shenyang Pharmaceutical University, Shenyang, 110016, PR China
| | - Bo Wu
- Faculty of Functional Food and Wine, Shenyang Pharmaceutical University, Shenyang, 110016, PR China
| | - Fanxing Xu
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, 110016, PR China.
| | - Ying Jia
- Faculty of Functional Food and Wine, Shenyang Pharmaceutical University, Shenyang, 110016, PR China.
| |
Collapse
|
24
|
Chu C, Gao X, Li X, Zhang X, Ma R, Jia Y, Li D, Wang D, Xu F. Involvement of Estrogen Receptor-α in the Activation of Nrf2-Antioxidative Signaling Pathways by Silibinin in Pancreatic β-Cells. Biomol Ther (Seoul) 2020; 28:163-171. [PMID: 31649209 PMCID: PMC7059807 DOI: 10.4062/biomolther.2019.071] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2019] [Revised: 08/18/2019] [Accepted: 09/04/2019] [Indexed: 01/10/2023] Open
Abstract
Silibinin exhibits antidiabetic potential by preserving the mass and function of pancreatic β-cells through up-regulation of estrogen receptor-α (ERα) expression. However, the underlying protective mechanism of silibinin in pancreatic β-cells is still unclear. In the current study, we sought to determine whether ERα acts as the target of silibinin for the modulation of antioxidative response in pancreatic β-cells under high glucose and high fat conditions. Our in vivo study revealed that a 4-week oral administration of silibinin (100 mg/kg/day) decreased fasting blood glucose with a concurrent increase in levels of serum insulin in high-fat diet/streptozotocin-induced type 2 diabetic rats. Moreover, expression of ERα, NF-E2-related factor 2 (Nrf2), and heme oxygenase-1 (HO-1) in pancreatic β-cells in pancreatic islets was increased by silibinin treatment. Accordingly, silibinin (10 μM) elevated viability, insulin biosynthesis, and insulin secretion of high glucose/palmitate-treated INS-1 cells accompanied by increased expression of ERα, Nrf2, and HO-1 as well as decreased reactive oxygen species production in vitro. Treatment using an ERα antagonist (MPP) in INS-1 cells or silencing ERα expression in INS-1 and NIT-1 cells with siRNA abolished the protective effects of silibinin. Our study suggests that silibinin activates the Nrf2-antioxidative pathways in pancreatic β-cells through regulation of ERα expression.
Collapse
Affiliation(s)
- Chun Chu
- Department of Pharmaceutics, School of Pharmacy, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Xiang Gao
- Department of Natural Products Chemistry, School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Xiang Li
- Qiqihaer Middle School, Qiqihaer 161099, China
| | - Xiaoying Zhang
- Department of Food Quality and Safety, Faculty of Functional Food and Wine, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Ruixin Ma
- Department of Food Quality and Safety, Faculty of Functional Food and Wine, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Ying Jia
- Department of Food Quality and Safety, Faculty of Functional Food and Wine, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Dahong Li
- Department of Natural Products Chemistry, School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Dongkai Wang
- Department of Pharmaceutics, School of Pharmacy, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Fanxing Xu
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, China
| |
Collapse
|
25
|
Si L, Fu J, Liu W, Hayashi T, Mizuno K, Hattori S, Fujisaki H, Onodera S, Ikejima T. Silibinin-induced mitochondria fission leads to mitophagy, which attenuates silibinin-induced apoptosis in MCF-7 and MDA-MB-231 cells. Arch Biochem Biophys 2020; 685:108284. [PMID: 32014401 DOI: 10.1016/j.abb.2020.108284] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2019] [Revised: 01/19/2020] [Accepted: 01/27/2020] [Indexed: 02/08/2023]
Abstract
We reported previously that higher doses (150-250 μM) of silibinin enhanced fission and inhibited fusion of mitochondria, accompanying apoptosis of double-positive breast cancer cell line MCF-7 cells and triple-negative breast cancer cell line MDA-MB-231 cells. We report here three important questions yet unclarified in the previous study; 1) Whether enhanced fission of mitochondria by the treatment of silibinin leads to mitophagy, 2) Whether mitophagy positively contributes to apoptosis and 3) Whether estrogen receptor-positive (ER+) MCF-7 cells and estrogen receptor-negative (ER-) MDA-MB-231 cells are affected in a different way by silibinin treatment, since silibinin often works through ERs signaling pathway. Mitophagy driven by Pink1/Parkin signaling, plays an important role in eliminating damaged mitochondria. Indeed, increased expression of Pink1 and the recruitment of Parkin and LC3-II to mitochondria by the treatment with silibinin account for silibinin induction of mitophagy. In this study, the effects of mitochondrial division inhibitor 1 (mdivi-1) and small interfering RNA targeting dynamin-related protein 1 (DRP1) were examined to reveal the effect of mitochondrial fission on mitophagy. As expected, mdivi-1 or siRNA targeting DRP1 reversed silibinin-induced mitochondrial fission due to down-regulation in the expression of DRP1. Inhibition of mitochondrial fission by mdivi-1 prevented induction of mitophagy as well as autophagy in both MCF-7 and MDA-MB-231 cells, indicating that silibinin-induced mitochondrial fission leads to mitophagy. Inhibition of mitochondrial fission efficiently prevented silibinin-induced apoptosis in MCF-7 and MDA-MB-231 cells in our previous work, and the second point of the present study, inhibition of mitophagy by Pink1 or Parkin knockdown increased silibinin-induced apoptosis of these cells, respectively, suggesting that the mitophagy induced by silibinin treatment serves as a cytoprotective effect, resulting in reduction of apoptosis of cancer cells in both cells. In the third point, we studied whether estrogen receptors (ERs) played a role in silibinin-induced mitophagy and apoptosis in MCF-7 and MDA-MB-231 cells. ERα and ERβ are not involved in silibinin-induced mitophagic process in MCF-7 and MDA-MB-231 cells. These findings demonstrated that silibinin induced mitochondria fission leads to mitophagy, which attenuates silibinin-induced apoptosis not through ERs-Pink1 or -Parkin pathway in MCF-7 and MDA-MB-231.
Collapse
Affiliation(s)
- Lingling Si
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, 110016, Liaoning, PR China
| | - Jianing Fu
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, 110016, Liaoning, PR China
| | - Weiwei Liu
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, 110016, Liaoning, PR China
| | - Toshihiko Hayashi
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, 110016, Liaoning, PR China; Department of Chemistry and Life Science, School of Advanced Engineering, Kogakuin University, 2665-1, Nakanomachi, Hachioji, Tokyo, 192-0015, Japan
| | - Kazunori Mizuno
- Nippi Research Institute of Biomatrix, Toride, Ibaraki, 302-0017, Japan
| | - Shunji Hattori
- Nippi Research Institute of Biomatrix, Toride, Ibaraki, 302-0017, Japan
| | - Hitomi Fujisaki
- Nippi Research Institute of Biomatrix, Toride, Ibaraki, 302-0017, Japan
| | - Satoshi Onodera
- Medical Research Institute of Curing Mibyo, 1-6-28 Narusedai Machida Tokyo, 194-0042, Japan
| | - Takashi Ikejima
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, 110016, Liaoning, PR China; Key Laboratory of Computational Chemistry-Based Natural Antitumor Drug Research & Development, Shenyang Pharmaceutical University, Shenyang, 110016, Liaoning, PR China.
| |
Collapse
|
26
|
Kong Y, Feng Z, Chen A, Qi Q, Han M, Wang S, Zhang Y, Zhang X, Yang N, Wang J, Huang B, Zhang Q, Xiang G, Li W, Zhang D, Wang J, Li X. The Natural Flavonoid Galangin Elicits Apoptosis, Pyroptosis, and Autophagy in Glioblastoma. Front Oncol 2019; 9:942. [PMID: 31612107 PMCID: PMC6776614 DOI: 10.3389/fonc.2019.00942] [Citation(s) in RCA: 94] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2019] [Accepted: 09/06/2019] [Indexed: 12/31/2022] Open
Abstract
Galangin (GG), a flavonoid, elicits a potent antitumor activity in diverse cancers. Here, we evaluated the efficacy of GG in the treatment of human glioblastoma multiforme (GBM) and investigated the molecular basis for its inhibitory effects in the disease. GG inhibited viability and proliferation of GBM cells (U251, U87MG, and A172) in a dose-dependent manner (IC50 = 221.8, 262.5, 273.9 μM, respectively; P < 0.001; EdU, ~40% decrease at 150 μM, P < 0.001), and the number of colonies formed was significantly reduced (at 50 μM, P < 0.001). However, normal human astrocytes were more resistant to its cytotoxic effects (IC50 >450 μM). Annexin-V/PI staining was increased indicating that GG induced apoptosis in GBM cells (26.67 and 30.42%, U87MG and U251, respectively) and associated proteins including BAX and cleaved PARP-1 were increased (~3×). Cells also underwent pyroptosis as determined under phase-contrast microscopy. Knockdown of gasdermin E (GSDME), a protein involved in pyroptosis, alleviated pyroptosis induced by GG through aggravating nuclear DNA damage in GBM cells. Meanwhile, fluorescent GFP-RFP-MAP1LC3B puncta associated with autophagy increased under GG treatment, and transmission electron microscopy confirmed the formation of autophagic vesicles. Inhibition of autophagy enhanced GG-induced apoptosis and pyroptosis in GBM cells. Finally, in an orthotopic xenograft model in nude mice derived from U87MG cells, treatment with GG in combination with an inhibitor of autophagy, chloroquine, suppressed tumor growth, and enhanced survival compared to GG monotherapy (P < 0.05). Our results demonstrated that GG simultaneously induces apoptosis, pytoptosis, and protective autophagy in GBM cells, indicating that combination treatment of GG with autophagy inhibitors may be an effective therapeutic strategy for GBM.
Collapse
Affiliation(s)
- Yang Kong
- Department of Neurosurgery, Qilu Hospital of Shandong University and Institute of Brain and Brain-Inspired Science, Shandong University, Jinan, China.,Shandong Key Laboratory of Brain Function Remodeling, Jinan, China
| | - Zichao Feng
- Department of Neurosurgery, Qilu Hospital of Shandong University and Institute of Brain and Brain-Inspired Science, Shandong University, Jinan, China.,Shandong Key Laboratory of Brain Function Remodeling, Jinan, China.,Department of Biomedicine, University of Bergen, Bergen, Norway
| | - Anjing Chen
- Department of Neurosurgery, Qilu Hospital of Shandong University and Institute of Brain and Brain-Inspired Science, Shandong University, Jinan, China.,Shandong Key Laboratory of Brain Function Remodeling, Jinan, China
| | - Qichao Qi
- Department of Neurosurgery, Qilu Hospital of Shandong University and Institute of Brain and Brain-Inspired Science, Shandong University, Jinan, China.,Shandong Key Laboratory of Brain Function Remodeling, Jinan, China
| | - Mingzhi Han
- Department of Neurosurgery, Qilu Hospital of Shandong University and Institute of Brain and Brain-Inspired Science, Shandong University, Jinan, China.,Shandong Key Laboratory of Brain Function Remodeling, Jinan, China.,Department of Biomedicine, University of Bergen, Bergen, Norway
| | - Shuai Wang
- Department of Neurosurgery, Qilu Hospital of Shandong University and Institute of Brain and Brain-Inspired Science, Shandong University, Jinan, China.,Shandong Key Laboratory of Brain Function Remodeling, Jinan, China
| | - Yulin Zhang
- Department of Neurosurgery, Qilu Hospital of Shandong University and Institute of Brain and Brain-Inspired Science, Shandong University, Jinan, China.,Shandong Key Laboratory of Brain Function Remodeling, Jinan, China
| | - Xin Zhang
- Department of Neurosurgery, Qilu Hospital of Shandong University and Institute of Brain and Brain-Inspired Science, Shandong University, Jinan, China
| | - Ning Yang
- Department of Neurosurgery, Qilu Hospital of Shandong University and Institute of Brain and Brain-Inspired Science, Shandong University, Jinan, China.,Shandong Key Laboratory of Brain Function Remodeling, Jinan, China
| | - Jiwei Wang
- Department of Neurosurgery, Qilu Hospital of Shandong University and Institute of Brain and Brain-Inspired Science, Shandong University, Jinan, China.,Shandong Key Laboratory of Brain Function Remodeling, Jinan, China
| | - Bin Huang
- Department of Neurosurgery, Qilu Hospital of Shandong University and Institute of Brain and Brain-Inspired Science, Shandong University, Jinan, China.,Shandong Key Laboratory of Brain Function Remodeling, Jinan, China
| | - Qing Zhang
- Department of Neurosurgery, Qilu Hospital of Shandong University and Institute of Brain and Brain-Inspired Science, Shandong University, Jinan, China.,Shandong Key Laboratory of Brain Function Remodeling, Jinan, China
| | - Guo Xiang
- Department of Neurosurgery, Qilu Hospital of Shandong University and Institute of Brain and Brain-Inspired Science, Shandong University, Jinan, China.,Shandong Key Laboratory of Brain Function Remodeling, Jinan, China
| | - Wenjie Li
- Department of Neurosurgery, Qilu Hospital of Shandong University and Institute of Brain and Brain-Inspired Science, Shandong University, Jinan, China.,Shandong Key Laboratory of Brain Function Remodeling, Jinan, China
| | - Di Zhang
- Department of Neurosurgery, Qilu Hospital of Shandong University and Institute of Brain and Brain-Inspired Science, Shandong University, Jinan, China.,Shandong Key Laboratory of Brain Function Remodeling, Jinan, China
| | - Jian Wang
- Department of Neurosurgery, Qilu Hospital of Shandong University and Institute of Brain and Brain-Inspired Science, Shandong University, Jinan, China.,Shandong Key Laboratory of Brain Function Remodeling, Jinan, China.,Department of Biomedicine, University of Bergen, Bergen, Norway
| | - Xingang Li
- Department of Neurosurgery, Qilu Hospital of Shandong University and Institute of Brain and Brain-Inspired Science, Shandong University, Jinan, China.,Shandong Key Laboratory of Brain Function Remodeling, Jinan, China
| |
Collapse
|
27
|
Si L, Liu W, Hayashi T, Ji Y, Fu J, Nie Y, Mizuno K, Hattori S, Onodera S, Ikejima T. Silibinin-induced apoptosis of breast cancer cells involves mitochondrial impairment. Arch Biochem Biophys 2019; 671:42-51. [PMID: 31085166 DOI: 10.1016/j.abb.2019.05.009] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2019] [Revised: 04/22/2019] [Accepted: 05/10/2019] [Indexed: 01/20/2023]
Abstract
Mitochondria are dynamically regulated by fission and fusion processes. Silibinin induces apoptosis of MCF-7 and MDA-MB-231 human breast cancer cells. However, whether or not mitochondria dysfunction is involved in the apoptosis induction with silibinin of both types of the cells remains unknown. We here report that silibinin decreases the mitochondrial mass in terms of MitoTracker Green staining in both breast cancer cells. Silibinin induces morphological changes of mitochondria from oval to truncated or fragmented shapes accordingly. Condensed crests are observed in mitochondria by transmission electron microscopy. Silibinin causes mitochondrial membrane potential reduced. The expression of mitochondrial fission-associated proteins including dynamin-related protein 1 (DRP1) is up-regulated, whereas expression of the mitochondrial fusion-associated proteins, optic atrophy 1 and mitofusin 1, is down-regulated. In addition, silibinin treatment down-regulates ATP content as well as the levels of mitochondrial biogenesis-regulators including mitochondrial transcription factor A, peroxisome proliferator-activated receptor gamma coactivator 1 and nuclear respiratory factor 2. Moreover, treatments with DRP1 inhibitor, mdivi-1, or with DRP1-targetted siRNA efficiently prevent silibinin-induced apoptosis in the breast cancer cells, whereas inhibition of DRP1 phosphorylation with staurosporine increases apoptosis furthermore. Taken together, we conclude that silibinin impairs mitochondrial dynamics and biogenesis, leading to apoptosis of MCF-7 and MDA-MB-123 cells.
Collapse
Affiliation(s)
- Lingling Si
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, Liaoning, PR China
| | - Weiwei Liu
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, Liaoning, PR China
| | - Toshihiko Hayashi
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, Liaoning, PR China; Department of Chemistry and Life Science, School of Advanced Engineering, Kogakuin University, 2665-1, Nakanomachi, Hachioji, Tokyo, 192-0015, Japan
| | - Yachao Ji
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, Liaoning, PR China
| | - Jianing Fu
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, Liaoning, PR China
| | - Yuheng Nie
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, Liaoning, PR China
| | - Kazunori Mizuno
- Nippi Research Institute of Biomatrix, Toride, Ibaraki 302-0017, Japan
| | - Shunji Hattori
- Nippi Research Institute of Biomatrix, Toride, Ibaraki 302-0017, Japan
| | - Satoshi Onodera
- Medical Research Institute of Curing Mibyo, 1-6-28 Narusedai Mechida Tokyo, 194-0042, Japan
| | - Takashi Ikejima
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, Liaoning, PR China; Key Laboratory of Computational Chemistry-Based Natural Antitumor Drug Research & Development, Shenyang Pharmaceutical University, Shenyang 110016, Liaoning, PR China.
| |
Collapse
|
28
|
Ju S, Liang Z, Li C, Ding C, Xu C, Song X, Zhao J. The effect and mechanism of miR-210 in down-regulating the autophagy of lung cancer cells. Pathol Res Pract 2019; 215:453-458. [PMID: 30573163 DOI: 10.1016/j.prp.2018.12.018] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/21/2018] [Revised: 11/27/2018] [Accepted: 12/11/2018] [Indexed: 02/07/2023]
Abstract
This project aims to investigate the roles of miR-210 in autophagy of lung cancer cells and the related mechanism. The expressions of miR-210 and ATG7 in 30 cancer tissues and the adjacent tissues in patients with lung cancer were compared using RT-qPCR methods, Western Blot assay was carried out to test the expression of ATG7 in protein. Moreover, the dual luciferase reporter gene assay system was used to confirm ATG7 is a target gene of miR-210. Furthermore, lung cancer cell line A549 was transfected with either miR-210 mimics or inhibitors and RT-qPCR methods was used to detect the expression of miR-210 and ATG7. Next, MTT assay was used to examine the effect of miR-210 on the growth of the lung cancer cells, and finally, the expression of autophagy related genes, ATG7, LC3-II/LC3-I and Beclin-1 were detected by Western Blot and ICC assay. We observed that miR-210 was significantly increased and ATG7 was markedly decreased in cancer tissue of patients with lung cancer compared with normal tissue. Moreover, results of dual luciferase reporter assay indicated that ATG7 is a direct target of miR-210. Next, transfection of miR-210 mimics in lung cancer cells induced significant increase in cell proliferation, and transfection of miR-210 inhibitors lead to inhibited cell proliferation. Furthermore, over-expression of miR-210 induced marked decrease in the expression of ATG7, LC3-II/LC3-I and Beclin-1, while transfection of miR-210 inhibitors induced significant increase in the expression of ATG7, LC3-II/LC3-I and beclin-1. Our results suggested that miR-210 plays a great role in autophagy of lung cancer cell by targeting ATG7.
Collapse
Affiliation(s)
- Sheng Ju
- Department of Thoracic Surgery, The First Affiliated Hospital of Soochow University, Suzhou, 215006, China
| | - Zhipan Liang
- Department of Thoracic Surgery, The First Affiliated Hospital of Soochow University, Suzhou, 215006, China
| | - Chang Li
- Department of Thoracic Surgery, The First Affiliated Hospital of Soochow University, Suzhou, 215006, China
| | - Cheng Ding
- Department of Thoracic Surgery, The First Affiliated Hospital of Soochow University, Suzhou, 215006, China
| | - Chun Xu
- Department of Thoracic Surgery, The First Affiliated Hospital of Soochow University, Suzhou, 215006, China
| | - Xinyu Song
- Department of Thoracic Surgery, The First Affiliated Hospital of Soochow University, Suzhou, 215006, China
| | - Jun Zhao
- Department of Thoracic Surgery, The First Affiliated Hospital of Soochow University, Suzhou, 215006, China.
| |
Collapse
|
29
|
Razak NA, Abu N, Ho WY, Zamberi NR, Tan SW, Alitheen NB, Long K, Yeap SK. Cytotoxicity of eupatorin in MCF-7 and MDA-MB-231 human breast cancer cells via cell cycle arrest, anti-angiogenesis and induction of apoptosis. Sci Rep 2019; 9:1514. [PMID: 30728391 PMCID: PMC6365513 DOI: 10.1038/s41598-018-37796-w] [Citation(s) in RCA: 110] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2018] [Accepted: 12/13/2018] [Indexed: 01/26/2023] Open
Abstract
Eupatorin has been reported with in vitro cytotoxic effect on several human cancer cells. However, reports on the mode of action and detail mechanism of eupatorin in vitro in breast cancer disease are limited. Hence, eupatorin's effect on the human breast carcinoma cell line MCF-7 and MDA-MB-231 was investigated. MTT assay showed that eupatorin had cytotoxic effects on MCF-7 and MDA-MB-231 cells but was non-toxic to the normal cells of MCF-10a in a time-dose dependent manner. At 24 h, the eupatorin showed mild cytotoxicity on both MCF-7 and MDA-MB-231 cells with IC50 values higher than 20 μg/mL. After 48 h, eupatorin at 5 μg/mL inhibited the proliferation of MCF-7 and MDA-MB-231 cells by 50% while the IC50 of MCF-10a was significantly (p < 0.05) high with 30 μg/mL. The concentration of eupatorin at 5 μg/mL induced apoptosis mainly through intrinsic pathway by facilitating higher fold of caspase 9 compared to caspase 8 at 48 h. The cell cycle profile also showed that eupatorin (5 μg/mL) exerted anti-proliferation activity with the cell cycle arrest of MCF-7 and MDA-MB-231 cells at sub Gθ/G1 in a time-dependent manner. In addition, wound healing assay showed an incomplete wound closure of scratched MDA-MB-231 cells, and more than 60% of the MDA-MB-231 cells were prevented to migrate and invade the membrane in the Boyden chamber after 24 h. Eupatorin also inhibited angiogenic sprouting of new blood vessels in ex vivo mouse aorta ring assay. In gene expression assay, eupatorin up-regulated pro-apoptotic genes such as Bak1, HIF1A, Bax, Bad, cytochrome c and SMAC/Diablo and blocked the Phospho-Akt pathway. In conclusion, eupatorin is a potent candidate to induce apoptosis and concurrently inhibit the invasion, migration and angiogenesis of MDA-MB-231 and MCF-7 cells through inhibition of Phospho-Akt pathway and cell cycle blockade.
Collapse
Affiliation(s)
- Nursyamirah Abd Razak
- Laborotary of Vaccines and Immunotherapeutics, Institute of Bioscience, Universiti Putra Malaysia, Serdang, 43400, Selangor, Malaysia
| | - Nadiah Abu
- UKM Molecular Biology Institute (UMBI), UKM Medical Centre, Jalan Yaa'cob Latiff, Bandar Tun Razak, Cheras, 56000, Kuala Lumpur, Malaysia
| | - Wan Yong Ho
- School of Biomedical Sciences, The University of Nottingham Malaysia Campus, Jalan Broga, Semenyih, 43500, Selangor, Malaysia
| | - Nur Rizi Zamberi
- Laborotary of Vaccines and Immunotherapeutics, Institute of Bioscience, Universiti Putra Malaysia, Serdang, 43400, Selangor, Malaysia
| | - Sheau Wei Tan
- Laborotary of Vaccines and Immunotherapeutics, Institute of Bioscience, Universiti Putra Malaysia, Serdang, 43400, Selangor, Malaysia
| | - Noorjahan Banu Alitheen
- Department of Cell and Molecular Biology, Faculty of Biotechnology and Biomolecular Sciences, Universiti Putra Malaysia, Serdang, 43400, Selangor, Malaysia
| | - Kamariah Long
- Malaysian Agricultural Research and Development Institute (MARDI), Serdang, 43400, Selangor, Malaysia
| | - Swee Keong Yeap
- Laborotary of Vaccines and Immunotherapeutics, Institute of Bioscience, Universiti Putra Malaysia, Serdang, 43400, Selangor, Malaysia.
- China-ASEAN College of Marine Sciences, Xiamen University Malaysia, Jalan Sunsuria, Bandar Sunsuria, Sepang, 43900, Selangor, Malaysia.
| |
Collapse
|
30
|
Yang J, Wen L, Jiang Y, Yang B. Natural Estrogen Receptor Modulators and Their Heterologous Biosynthesis. Trends Endocrinol Metab 2019; 30:66-76. [PMID: 30527917 DOI: 10.1016/j.tem.2018.11.002] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/28/2018] [Revised: 10/24/2018] [Accepted: 11/09/2018] [Indexed: 12/23/2022]
Abstract
Estrogen receptors (ERs) are transcription factors highly involved in physiological development and metabolism in the human body. They also play important roles in the treatment of cancer and metabolic diseases. Chemicals that interact with ERs can be used to treat diseases and maintain health. Phytoestrogens are natural chemicals that have been documented to possess significant ER modulatory activities. However, since phytoestrogens usually exist at low quantities in nature, heterologous biosynthesis techniques have quickly developed in recent years in order meet the demands for needed therapeutic amounts. In this review, the performance of phytoestrogens as ER modulators is described along with recent advances in biosynthesis techniques.
Collapse
Affiliation(s)
- Jiali Yang
- Key Laboratory of Plant Resources Conservation and Sustainable Utilization, Guangdong Provincial Key Laboratory of Applied Botany, South China Botanical Garden, Chinese Academy of Sciences, Guangzhou 510650, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Lingrong Wen
- Key Laboratory of Plant Resources Conservation and Sustainable Utilization, Guangdong Provincial Key Laboratory of Applied Botany, South China Botanical Garden, Chinese Academy of Sciences, Guangzhou 510650, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yueming Jiang
- Key Laboratory of Plant Resources Conservation and Sustainable Utilization, Guangdong Provincial Key Laboratory of Applied Botany, South China Botanical Garden, Chinese Academy of Sciences, Guangzhou 510650, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Bao Yang
- Key Laboratory of Plant Resources Conservation and Sustainable Utilization, Guangdong Provincial Key Laboratory of Applied Botany, South China Botanical Garden, Chinese Academy of Sciences, Guangzhou 510650, China; University of Chinese Academy of Sciences, Beijing 100049, China.
| |
Collapse
|
31
|
Younas M, Hano C, Giglioli-Guivarc'h N, Abbasi BH. Mechanistic evaluation of phytochemicals in breast cancer remedy: current understanding and future perspectives. RSC Adv 2018; 8:29714-29744. [PMID: 35547279 PMCID: PMC9085387 DOI: 10.1039/c8ra04879g] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2018] [Accepted: 08/15/2018] [Indexed: 12/30/2022] Open
Abstract
Breast cancer is one of the most commonly diagnosed cancers around the globe and accounts for a large proportion of fatalities in women. Despite the advancement in therapeutic and diagnostic procedures, breast cancer still represents a major challenge. Current anti-breast cancer approaches include surgical removal, radiotherapy, hormonal therapy and the use of various chemotherapeutic drugs. However, drug resistance, associated serious adverse effects, metastasis and recurrence complications still need to be resolved which demand safe and alternative strategies. In this scenario, phytochemicals have recently gained huge attention due to their safety profile and cost-effectiveness. These phytochemicals modulate various genes, gene products and signalling pathways, thereby inhibiting breast cancer cell proliferation, invasion, angiogenesis and metastasis and inducing apoptosis. Moreover, they also target breast cancer stem cells and overcome drug resistance problems in breast carcinomas. Phytochemicals as adjuvants with chemotherapeutic drugs have greatly enhanced their therapeutic efficacy. This review focuses on the recently recognized molecular mechanisms underlying breast cancer chemoprevention with the use of phytochemicals such as curcumin, resveratrol, silibinin, genistein, epigallocatechin gallate, secoisolariciresinol, thymoquinone, kaempferol, quercetin, parthenolide, sulforaphane, ginsenosides, naringenin, isoliquiritigenin, luteolin, benzyl isothiocyanate, α-mangostin, 3,3'-diindolylmethane, pterostilbene, vinca alkaloids and apigenin.
Collapse
Affiliation(s)
- Muhammad Younas
- Department of Biotechnology, Quaid-i-Azam University Islamabad-45320 Pakistan +92-51-90644121 +92-51-90644121 +33-767-97-0619
| | - Christophe Hano
- Laboratoire de Biologie des Ligneux et des Grandes Cultures (LBLGC), Plant Lignans Team, UPRES EA 1207, Université d'Orléans F 28000 Chartres France
| | | | - Bilal Haider Abbasi
- Department of Biotechnology, Quaid-i-Azam University Islamabad-45320 Pakistan +92-51-90644121 +92-51-90644121 +33-767-97-0619
- Laboratoire de Biologie des Ligneux et des Grandes Cultures (LBLGC), Plant Lignans Team, UPRES EA 1207, Université d'Orléans F 28000 Chartres France
- EA2106 Biomolecules et Biotechnologies Vegetales, Universite Francois-Rabelais de Tours Tours France
| |
Collapse
|
32
|
Draz H, Goldberg AA, Tomlinson Guns ES, Fazli L, Safe S, Sanderson JT. Autophagy inhibition improves the chemotherapeutic efficacy of cruciferous vegetable-derived diindolymethane in a murine prostate cancer xenograft model. Invest New Drugs 2018; 36:718-725. [PMID: 29607466 DOI: 10.1007/s10637-018-0595-8] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2018] [Accepted: 03/21/2018] [Indexed: 01/07/2023]
Abstract
Prostate cancer is the second leading cause of cancer-related deaths in men in North America and there is an urgent need for development of more effective therapeutic treatments against this disease. We have recently shown that diindolylmethane (DIM) and several of its halogenated derivatives (ring-DIMs) induce death and protective autophagy in human prostate cancer cells. However, the in vivo efficacy of ring-DIMs and the use of autophagy inhibitors as adjuvant therapy have not yet been studied in vivo. The objective of this study was to determine these effects on tumor growth in nude CD-1 mice bearing bioluminescent androgen-independent PC-3 human prostate cancer cells. We found that chloroquine (CQ) significantly sensitized PC-3 cells to death in the presence of sub-toxic concentrations of DIM or 4,4'-Br2DIM in vitro. Moreover, a combination of DIM (10 mg/kg) and CQ (60 mg/kg), 3× per week, significantly decreased PC-3 tumor growth in vivo after 3 and 4 weeks of treatment. Furthermore, 4,4'-Br2DIM at 10 mg/kg (3× per week) significantly inhibited tumour growth after 4 weeks of treatment. Tissues microarray analysis showed that DIM alone or combined with CQ induced apoptosis marker TUNEL; the combination also significantly inhibited the cell proliferation marker Ki67. In conclusion, we have confirmed that DIM and 4,4'-Br2DIM are effective agents against prostate cancer in vivo and shown that inhibition of autophagy with CQ enhances the anticancer efficacy of DIM. Our results suggest that including selective autophagy inhibitors as adjuvants may improve the efficacy of existing and novel drug therapies against prostate cancer.
Collapse
Affiliation(s)
- Hossam Draz
- INRS-Institut Armand-Frappier, 531 boulevard des Prairies, Laval, QC, H7V 1B7, Canada
- Department of Biochemistry, National Research Centre, Dokki, Cairo, Egypt
| | - Alexander A Goldberg
- INRS-Institut Armand-Frappier, 531 boulevard des Prairies, Laval, QC, H7V 1B7, Canada
| | | | - Ladan Fazli
- Vancouver Prostate Centre, University of British Columbia, Vancouver, BC, Canada
| | - Stephen Safe
- Veterinary Physiology and Pharmacology, Texas A&M University, College Station, TX, USA
| | - J Thomas Sanderson
- INRS-Institut Armand-Frappier, 531 boulevard des Prairies, Laval, QC, H7V 1B7, Canada.
| |
Collapse
|
33
|
Silibinin-induced autophagy mediated by PPARα-sirt1-AMPK pathway participated in the regulation of type I collagen-enhanced migration in murine 3T3-L1 preadipocytes. Mol Cell Biochem 2018; 450:1-23. [DOI: 10.1007/s11010-018-3368-y] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2017] [Accepted: 05/17/2018] [Indexed: 12/21/2022]
|
34
|
Involvement of estrogen receptors in silibinin protection of pancreatic β-cells from TNFα- or IL-1β-induced cytotoxicity. Biomed Pharmacother 2018; 102:344-353. [PMID: 29571019 DOI: 10.1016/j.biopha.2018.01.128] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2017] [Revised: 01/26/2018] [Accepted: 01/28/2018] [Indexed: 01/09/2023] Open
Abstract
Silibinin is a polyphenolic flavonoid that exhibits anticarcinogenic, anti-inflammatory and cytoprotective effects. The effect of silibinin on pancreatic islet β-cell is yet largely unknown in spite of well documented-hepatoprotective effects. Protecting the functional insulin-producing β-cells in the pancreas is a major therapeutic challenge in the patients with type 1 (T1DM) or type 2 diabetes mellitus (T2DM). This study reports the effect of silibinin on the rat pancreatic β-cell line, INS-1, damaged with pro-inflammatory cytokine, TNFα or IL-1β. Exposure to TNFα or IL-1β for 48 h caused INS-1 cells to reduce the production of insulin as well as cell viability. These actions of TNFα or IL-1β are associated with suppression of the expression of estrogen receptors (ERs). Further study revealed that silibinin protected the suppression in the expression of both ERα and ERβ that were involved in insulin synthesis and release, respectively. Furthermore, evidence is obtained that silibinin may impede the loss of critical INS-1 cells by promoting autophagy and preventing apoptosis. Direct cytoprotective effects of silibinin on INS-1 cells suggest that silibinin may be therapeutically beneficial for diabetes.
Collapse
|
35
|
Egea J, Fabregat I, Frapart YM, Ghezzi P, Görlach A, Kietzmann T, Kubaichuk K, Knaus UG, Lopez MG, Olaso-Gonzalez G, Petry A, Schulz R, Vina J, Winyard P, Abbas K, Ademowo OS, Afonso CB, Andreadou I, Antelmann H, Antunes F, Aslan M, Bachschmid MM, Barbosa RM, Belousov V, Berndt C, Bernlohr D, Bertrán E, Bindoli A, Bottari SP, Brito PM, Carrara G, Casas AI, Chatzi A, Chondrogianni N, Conrad M, Cooke MS, Costa JG, Cuadrado A, My-Chan Dang P, De Smet B, Debelec-Butuner B, Dias IHK, Dunn JD, Edson AJ, El Assar M, El-Benna J, Ferdinandy P, Fernandes AS, Fladmark KE, Förstermann U, Giniatullin R, Giricz Z, Görbe A, Griffiths H, Hampl V, Hanf A, Herget J, Hernansanz-Agustín P, Hillion M, Huang J, Ilikay S, Jansen-Dürr P, Jaquet V, Joles JA, Kalyanaraman B, Kaminskyy D, Karbaschi M, Kleanthous M, Klotz LO, Korac B, Korkmaz KS, Koziel R, Kračun D, Krause KH, Křen V, Krieg T, Laranjinha J, Lazou A, Li H, Martínez-Ruiz A, Matsui R, McBean GJ, Meredith SP, Messens J, Miguel V, Mikhed Y, Milisav I, Milković L, Miranda-Vizuete A, Mojović M, Monsalve M, Mouthuy PA, Mulvey J, Münzel T, Muzykantov V, Nguyen ITN, Oelze M, Oliveira NG, Palmeira CM, Papaevgeniou N, et alEgea J, Fabregat I, Frapart YM, Ghezzi P, Görlach A, Kietzmann T, Kubaichuk K, Knaus UG, Lopez MG, Olaso-Gonzalez G, Petry A, Schulz R, Vina J, Winyard P, Abbas K, Ademowo OS, Afonso CB, Andreadou I, Antelmann H, Antunes F, Aslan M, Bachschmid MM, Barbosa RM, Belousov V, Berndt C, Bernlohr D, Bertrán E, Bindoli A, Bottari SP, Brito PM, Carrara G, Casas AI, Chatzi A, Chondrogianni N, Conrad M, Cooke MS, Costa JG, Cuadrado A, My-Chan Dang P, De Smet B, Debelec-Butuner B, Dias IHK, Dunn JD, Edson AJ, El Assar M, El-Benna J, Ferdinandy P, Fernandes AS, Fladmark KE, Förstermann U, Giniatullin R, Giricz Z, Görbe A, Griffiths H, Hampl V, Hanf A, Herget J, Hernansanz-Agustín P, Hillion M, Huang J, Ilikay S, Jansen-Dürr P, Jaquet V, Joles JA, Kalyanaraman B, Kaminskyy D, Karbaschi M, Kleanthous M, Klotz LO, Korac B, Korkmaz KS, Koziel R, Kračun D, Krause KH, Křen V, Krieg T, Laranjinha J, Lazou A, Li H, Martínez-Ruiz A, Matsui R, McBean GJ, Meredith SP, Messens J, Miguel V, Mikhed Y, Milisav I, Milković L, Miranda-Vizuete A, Mojović M, Monsalve M, Mouthuy PA, Mulvey J, Münzel T, Muzykantov V, Nguyen ITN, Oelze M, Oliveira NG, Palmeira CM, Papaevgeniou N, Pavićević A, Pedre B, Peyrot F, Phylactides M, Pircalabioru GG, Pitt AR, Poulsen HE, Prieto I, Rigobello MP, Robledinos-Antón N, Rodríguez-Mañas L, Rolo AP, Rousset F, Ruskovska T, Saraiva N, Sasson S, Schröder K, Semen K, Seredenina T, Shakirzyanova A, Smith GL, Soldati T, Sousa BC, Spickett CM, Stancic A, Stasia MJ, Steinbrenner H, Stepanić V, Steven S, Tokatlidis K, Tuncay E, Turan B, Ursini F, Vacek J, Vajnerova O, Valentová K, Van Breusegem F, Varisli L, Veal EA, Yalçın AS, Yelisyeyeva O, Žarković N, Zatloukalová M, Zielonka J, Touyz RM, Papapetropoulos A, Grune T, Lamas S, Schmidt HHHW, Di Lisa F, Daiber A. European contribution to the study of ROS: A summary of the findings and prospects for the future from the COST action BM1203 (EU-ROS). Redox Biol 2017; 13:94-162. [PMID: 28577489 PMCID: PMC5458069 DOI: 10.1016/j.redox.2017.05.007] [Show More Authors] [Citation(s) in RCA: 202] [Impact Index Per Article: 25.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2017] [Accepted: 05/08/2017] [Indexed: 12/12/2022] Open
Abstract
The European Cooperation in Science and Technology (COST) provides an ideal framework to establish multi-disciplinary research networks. COST Action BM1203 (EU-ROS) represents a consortium of researchers from different disciplines who are dedicated to providing new insights and tools for better understanding redox biology and medicine and, in the long run, to finding new therapeutic strategies to target dysregulated redox processes in various diseases. This report highlights the major achievements of EU-ROS as well as research updates and new perspectives arising from its members. The EU-ROS consortium comprised more than 140 active members who worked together for four years on the topics briefly described below. The formation of reactive oxygen and nitrogen species (RONS) is an established hallmark of our aerobic environment and metabolism but RONS also act as messengers via redox regulation of essential cellular processes. The fact that many diseases have been found to be associated with oxidative stress established the theory of oxidative stress as a trigger of diseases that can be corrected by antioxidant therapy. However, while experimental studies support this thesis, clinical studies still generate controversial results, due to complex pathophysiology of oxidative stress in humans. For future improvement of antioxidant therapy and better understanding of redox-associated disease progression detailed knowledge on the sources and targets of RONS formation and discrimination of their detrimental or beneficial roles is required. In order to advance this important area of biology and medicine, highly synergistic approaches combining a variety of diverse and contrasting disciplines are needed.
Collapse
Affiliation(s)
- Javier Egea
- Institute Teofilo Hernando, Department of Pharmacology, School of Medicine. Univerisdad Autonoma de Madrid, Spain
| | - Isabel Fabregat
- Bellvitge Biomedical Research Institute (IDIBELL) and University of Barcelona (UB), L'Hospitalet, Barcelona, Spain
| | - Yves M Frapart
- LCBPT, UMR 8601 CNRS - Paris Descartes University, Sorbonne Paris Cité, Paris, France
| | | | - Agnes Görlach
- Experimental and Molecular Pediatric Cardiology, German Heart Center Munich at the Technical University Munich, Munich, Germany; DZHK (German Centre for Cardiovascular Research), partner site Munich Heart Alliance, Munich, Germany
| | - Thomas Kietzmann
- Faculty of Biochemistry and Molecular Medicine, and Biocenter Oulu, University of Oulu, Oulu, Finland
| | - Kateryna Kubaichuk
- Faculty of Biochemistry and Molecular Medicine, and Biocenter Oulu, University of Oulu, Oulu, Finland
| | - Ulla G Knaus
- Conway Institute, School of Medicine, University College Dublin, Dublin, Ireland
| | - Manuela G Lopez
- Institute Teofilo Hernando, Department of Pharmacology, School of Medicine. Univerisdad Autonoma de Madrid, Spain
| | | | - Andreas Petry
- Experimental and Molecular Pediatric Cardiology, German Heart Center Munich at the Technical University Munich, Munich, Germany
| | - Rainer Schulz
- Institute of Physiology, JLU Giessen, Giessen, Germany
| | - Jose Vina
- Department of Physiology, University of Valencia, Spain
| | - Paul Winyard
- University of Exeter Medical School, St Luke's Campus, Exeter EX1 2LU, UK
| | - Kahina Abbas
- LCBPT, UMR 8601 CNRS - Paris Descartes University, Sorbonne Paris Cité, Paris, France
| | - Opeyemi S Ademowo
- Life & Health Sciences and Aston Research Centre for Healthy Ageing, Aston University, Aston Triangle, Birmingham B4 7ET, UK
| | - Catarina B Afonso
- School of Life & Health Sciences, Aston University, Aston Triangle, Birmingham B47ET, UK
| | - Ioanna Andreadou
- Laboratory of Pharmacology, Faculty of Pharmacy, National and Kapodistrian University of Athens, Greece
| | - Haike Antelmann
- Institute for Biology-Microbiology, Freie Universität Berlin, Berlin, Germany
| | - Fernando Antunes
- Departamento de Química e Bioquímica and Centro de Química e Bioquímica, Faculdade de Ciências, Portugal
| | - Mutay Aslan
- Department of Medical Biochemistry, Faculty of Medicine, Akdeniz University, Antalya, Turkey
| | - Markus M Bachschmid
- Vascular Biology Section & Whitaker Cardiovascular Institute, Boston University School of Medicine, Boston, MA, USA
| | - Rui M Barbosa
- Center for Neurosciences and Cell Biology, University of Coimbra and Faculty of Pharmacy, University of Coimbra, Coimbra, Portugal
| | - Vsevolod Belousov
- Molecular technologies laboratory, Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Miklukho-Maklaya 16/10, Moscow 117997, Russia
| | - Carsten Berndt
- Department of Neurology, Medical Faculty, Heinrich-Heine University, Düsseldorf, Germany
| | - David Bernlohr
- Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota - Twin Cities, USA
| | - Esther Bertrán
- Bellvitge Biomedical Research Institute (IDIBELL) and University of Barcelona (UB), L'Hospitalet, Barcelona, Spain
| | | | - Serge P Bottari
- GETI, Institute for Advanced Biosciences, INSERM U1029, CNRS UMR 5309, Grenoble-Alpes University and Radio-analysis Laboratory, CHU de Grenoble, Grenoble, France
| | - Paula M Brito
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Lisboa, Portugal; Faculdade de Ciências da Saúde, Universidade da Beira Interior, Covilhã, Portugal
| | - Guia Carrara
- Department of Pathology, University of Cambridge, Cambridge, UK
| | - Ana I Casas
- Department of Pharmacology & Personalized Medicine, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Maastricht, The Netherlands
| | - Afroditi Chatzi
- Institute of Molecular Cell and Systems Biology, College of Medical Veterinary and Life Sciences, University of Glasgow, University Avenue, Glasgow, UK
| | - Niki Chondrogianni
- National Hellenic Research Foundation, Institute of Biology, Medicinal Chemistry and Biotechnology, 48 Vas. Constantinou Ave., 116 35 Athens, Greece
| | - Marcus Conrad
- Helmholtz Center Munich, Institute of Developmental Genetics, Neuherberg, Germany
| | - Marcus S Cooke
- Oxidative Stress Group, Dept. Environmental & Occupational Health, Florida International University, Miami, FL 33199, USA
| | - João G Costa
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Lisboa, Portugal; CBIOS, Universidade Lusófona Research Center for Biosciences & Health Technologies, Lisboa, Portugal
| | - Antonio Cuadrado
- Instituto de Investigaciones Biomédicas "Alberto Sols" UAM-CSIC, Instituto de Investigación Sanitaria La Paz (IdiPaz), Department of Biochemistry, Faculty of Medicine, Autonomous University of Madrid. Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | - Pham My-Chan Dang
- Université Paris Diderot, Sorbonne Paris Cité, INSERM-U1149, CNRS-ERL8252, Centre de Recherche sur l'Inflammation, Laboratoire d'Excellence Inflamex, Faculté de Médecine Xavier Bichat, Paris, France
| | - Barbara De Smet
- Department of Plant Systems Biology, VIB, 9052 Ghent, Belgium; Structural Biology Research Center, VIB, 1050 Brussels, Belgium; Department of Biomedical Sciences and CNR Institute of Neuroscience, University of Padova, Padova, Italy; Pharmahungary Group, Szeged, Hungary
| | - Bilge Debelec-Butuner
- Department of Pharmaceutical Biotechnology, Faculty of Pharmacy, Ege University, Bornova, Izmir 35100, Turkey
| | - Irundika H K Dias
- Life & Health Sciences and Aston Research Centre for Healthy Ageing, Aston University, Aston Triangle, Birmingham B4 7ET, UK
| | - Joe Dan Dunn
- Department of Biochemistry, Science II, University of Geneva, 30 quai Ernest-Ansermet, 1211 Geneva-4, Switzerland
| | - Amanda J Edson
- Department of Molecular Biology, University of Bergen, Bergen, Norway
| | - Mariam El Assar
- Fundación para la Investigación Biomédica del Hospital Universitario de Getafe, Getafe, Spain
| | - Jamel El-Benna
- Université Paris Diderot, Sorbonne Paris Cité, INSERM-U1149, CNRS-ERL8252, Centre de Recherche sur l'Inflammation, Laboratoire d'Excellence Inflamex, Faculté de Médecine Xavier Bichat, Paris, France
| | - Péter Ferdinandy
- Department of Pharmacology and Pharmacotherapy, Medical Faculty, Semmelweis University, Budapest, Hungary; Pharmahungary Group, Szeged, Hungary
| | - Ana S Fernandes
- CBIOS, Universidade Lusófona Research Center for Biosciences & Health Technologies, Lisboa, Portugal
| | - Kari E Fladmark
- Department of Molecular Biology, University of Bergen, Bergen, Norway
| | - Ulrich Förstermann
- Department of Pharmacology, Johannes Gutenberg University Medical Center, Mainz, Germany
| | - Rashid Giniatullin
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | - Zoltán Giricz
- Department of Pharmacology and Pharmacotherapy, Medical Faculty, Semmelweis University, Budapest, Hungary; Pharmahungary Group, Szeged, Hungary
| | - Anikó Görbe
- Department of Pharmacology and Pharmacotherapy, Medical Faculty, Semmelweis University, Budapest, Hungary; Pharmahungary Group, Szeged, Hungary
| | - Helen Griffiths
- Life & Health Sciences and Aston Research Centre for Healthy Ageing, Aston University, Aston Triangle, Birmingham B4 7ET, UK; Faculty of Health and Medical Sciences, University of Surrey, Guildford GU2 7XH, UK
| | - Vaclav Hampl
- Department of Physiology, 2nd Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Alina Hanf
- Molecular Cardiology, Center for Cardiology, Cardiology 1, University Medical Center Mainz, Mainz, Germany
| | - Jan Herget
- Department of Physiology, 2nd Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Pablo Hernansanz-Agustín
- Servicio de Immunología, Hospital Universitario de La Princesa, Instituto de Investigación Sanitaria Princesa (IIS-IP), Madrid, Spain; Departamento de Bioquímica, Facultad de Medicina, Universidad Autónoma de Madrid (UAM) and Instituto de Investigaciones Biomédicas Alberto Sols, Madrid, Spain
| | - Melanie Hillion
- Institute for Biology-Microbiology, Freie Universität Berlin, Berlin, Germany
| | - Jingjing Huang
- Department of Plant Systems Biology, VIB, 9052 Ghent, Belgium; Structural Biology Research Center, VIB, 1050 Brussels, Belgium; Department of Plant Biotechnology and Bioinformatics, Ghent University, 9052 Ghent, Belgium; Brussels Center for Redox Biology, Structural Biology Brussels, Vrije Universiteit Brussel, 1050 Brussels, Belgium
| | - Serap Ilikay
- Harran University, Arts and Science Faculty, Department of Biology, Cancer Biology Lab, Osmanbey Campus, Sanliurfa, Turkey
| | - Pidder Jansen-Dürr
- Institute for Biomedical Aging Research, University of Innsbruck, Innsbruck, Austria
| | - Vincent Jaquet
- Dept. of Pathology and Immunology, Centre Médical Universitaire, Geneva, Switzerland
| | - Jaap A Joles
- Department of Nephrology & Hypertension, University Medical Center Utrecht, The Netherlands
| | | | | | - Mahsa Karbaschi
- Oxidative Stress Group, Dept. Environmental & Occupational Health, Florida International University, Miami, FL 33199, USA
| | - Marina Kleanthous
- Molecular Genetics Thalassaemia Department, The Cyprus Institute of Neurology and Genetics, Nicosia, Cyprus
| | - Lars-Oliver Klotz
- Institute of Nutrition, Department of Nutrigenomics, Friedrich Schiller University, Jena, Germany
| | - Bato Korac
- University of Belgrade, Institute for Biological Research "Sinisa Stankovic" and Faculty of Biology, Belgrade, Serbia
| | - Kemal Sami Korkmaz
- Department of Bioengineering, Cancer Biology Laboratory, Faculty of Engineering, Ege University, Bornova, 35100 Izmir, Turkey
| | - Rafal Koziel
- Institute for Biomedical Aging Research, University of Innsbruck, Innsbruck, Austria
| | - Damir Kračun
- Experimental and Molecular Pediatric Cardiology, German Heart Center Munich at the Technical University Munich, Munich, Germany
| | - Karl-Heinz Krause
- Dept. of Pathology and Immunology, Centre Médical Universitaire, Geneva, Switzerland
| | - Vladimír Křen
- Institute of Microbiology, Laboratory of Biotransformation, Czech Academy of Sciences, Videnska 1083, CZ-142 20 Prague, Czech Republic
| | - Thomas Krieg
- Department of Medicine, University of Cambridge, UK
| | - João Laranjinha
- Center for Neurosciences and Cell Biology, University of Coimbra and Faculty of Pharmacy, University of Coimbra, Coimbra, Portugal
| | - Antigone Lazou
- School of Biology, Aristotle University of Thessaloniki, Thessaloniki 54124, Greece
| | - Huige Li
- Department of Pharmacology, Johannes Gutenberg University Medical Center, Mainz, Germany
| | - Antonio Martínez-Ruiz
- Servicio de Immunología, Hospital Universitario de La Princesa, Instituto de Investigación Sanitaria Princesa (IIS-IP), Madrid, Spain; Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain
| | - Reiko Matsui
- Vascular Biology Section & Whitaker Cardiovascular Institute, Boston University School of Medicine, Boston, MA, USA
| | - Gethin J McBean
- School of Biomolecular and Biomedical Science, Conway Institute, University College Dublin, Dublin, Ireland
| | - Stuart P Meredith
- School of Life & Health Sciences, Aston University, Aston Triangle, Birmingham B47ET, UK
| | - Joris Messens
- Structural Biology Research Center, VIB, 1050 Brussels, Belgium; Brussels Center for Redox Biology, Structural Biology Brussels, Vrije Universiteit Brussel, 1050 Brussels, Belgium
| | - Verónica Miguel
- Centro de Biología Molecular "Severo Ochoa" (CSIC-UAM), Madrid, Spain
| | - Yuliya Mikhed
- Molecular Cardiology, Center for Cardiology, Cardiology 1, University Medical Center Mainz, Mainz, Germany
| | - Irina Milisav
- University of Ljubljana, Faculty of Medicine, Institute of Pathophysiology and Faculty of Health Sciences, Ljubljana, Slovenia
| | - Lidija Milković
- Ruđer Bošković Institute, Division of Molecular Medicine, Zagreb, Croatia
| | - Antonio Miranda-Vizuete
- Instituto de Biomedicina de Sevilla, Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Sevilla, Spain
| | - Miloš Mojović
- University of Belgrade, Faculty of Physical Chemistry, Studentski trg 12-16, 11000 Belgrade, Serbia
| | - María Monsalve
- Instituto de Investigaciones Biomédicas "Alberto Sols" (CSIC-UAM), Madrid, Spain
| | - Pierre-Alexis Mouthuy
- Laboratory for Oxidative Stress, Rudjer Boskovic Institute, Bijenicka 54, 10000 Zagreb, Croatia
| | - John Mulvey
- Department of Medicine, University of Cambridge, UK
| | - Thomas Münzel
- Molecular Cardiology, Center for Cardiology, Cardiology 1, University Medical Center Mainz, Mainz, Germany
| | - Vladimir Muzykantov
- Department of Pharmacology, Center for Targeted Therapeutics & Translational Nanomedicine, ITMAT/CTSA Translational Research Center University of Pennsylvania The Perelman School of Medicine, Philadelphia, PA, USA
| | - Isabel T N Nguyen
- Department of Nephrology & Hypertension, University Medical Center Utrecht, The Netherlands
| | - Matthias Oelze
- Molecular Cardiology, Center for Cardiology, Cardiology 1, University Medical Center Mainz, Mainz, Germany
| | - Nuno G Oliveira
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Lisboa, Portugal
| | - Carlos M Palmeira
- Center for Neurosciences & Cell Biology of the University of Coimbra, Coimbra, Portugal; Department of Life Sciences of the Faculty of Sciences & Technology of the University of Coimbra, Coimbra, Portugal
| | - Nikoletta Papaevgeniou
- National Hellenic Research Foundation, Institute of Biology, Medicinal Chemistry and Biotechnology, 48 Vas. Constantinou Ave., 116 35 Athens, Greece
| | - Aleksandra Pavićević
- University of Belgrade, Faculty of Physical Chemistry, Studentski trg 12-16, 11000 Belgrade, Serbia
| | - Brandán Pedre
- Structural Biology Research Center, VIB, 1050 Brussels, Belgium; Brussels Center for Redox Biology, Structural Biology Brussels, Vrije Universiteit Brussel, 1050 Brussels, Belgium
| | - Fabienne Peyrot
- LCBPT, UMR 8601 CNRS - Paris Descartes University, Sorbonne Paris Cité, Paris, France; ESPE of Paris, Paris Sorbonne University, Paris, France
| | - Marios Phylactides
- Molecular Genetics Thalassaemia Department, The Cyprus Institute of Neurology and Genetics, Nicosia, Cyprus
| | | | - Andrew R Pitt
- School of Life & Health Sciences, Aston University, Aston Triangle, Birmingham B47ET, UK
| | - Henrik E Poulsen
- Laboratory of Clinical Pharmacology, Rigshospitalet, University Hospital Copenhagen, Denmark; Department of Clinical Pharmacology, Bispebjerg Frederiksberg Hospital, University Hospital Copenhagen, Denmark; Department Q7642, Rigshospitalet, Blegdamsvej 9, DK-2100 Copenhagen, Denmark
| | - Ignacio Prieto
- Instituto de Investigaciones Biomédicas "Alberto Sols" (CSIC-UAM), Madrid, Spain
| | - Maria Pia Rigobello
- Department of Biomedical Sciences, University of Padova, via Ugo Bassi 58/b, 35131 Padova, Italy
| | - Natalia Robledinos-Antón
- Instituto de Investigaciones Biomédicas "Alberto Sols" UAM-CSIC, Instituto de Investigación Sanitaria La Paz (IdiPaz), Department of Biochemistry, Faculty of Medicine, Autonomous University of Madrid. Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | - Leocadio Rodríguez-Mañas
- Fundación para la Investigación Biomédica del Hospital Universitario de Getafe, Getafe, Spain; Servicio de Geriatría, Hospital Universitario de Getafe, Getafe, Spain
| | - Anabela P Rolo
- Center for Neurosciences & Cell Biology of the University of Coimbra, Coimbra, Portugal; Department of Life Sciences of the Faculty of Sciences & Technology of the University of Coimbra, Coimbra, Portugal
| | - Francis Rousset
- Dept. of Pathology and Immunology, Centre Médical Universitaire, Geneva, Switzerland
| | - Tatjana Ruskovska
- Faculty of Medical Sciences, Goce Delcev University, Stip, Republic of Macedonia
| | - Nuno Saraiva
- CBIOS, Universidade Lusófona Research Center for Biosciences & Health Technologies, Lisboa, Portugal
| | - Shlomo Sasson
- Institute for Drug Research, Section of Pharmacology, Diabetes Research Unit, The Hebrew University Faculty of Medicine, Jerusalem, Israel
| | - Katrin Schröder
- Institute for Cardiovascular Physiology, Goethe-University, Frankfurt, Germany; DZHK (German Centre for Cardiovascular Research), partner site Rhine-Main, Mainz, Germany
| | - Khrystyna Semen
- Danylo Halytsky Lviv National Medical University, Lviv, Ukraine
| | - Tamara Seredenina
- Dept. of Pathology and Immunology, Centre Médical Universitaire, Geneva, Switzerland
| | - Anastasia Shakirzyanova
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | | | - Thierry Soldati
- Department of Biochemistry, Science II, University of Geneva, 30 quai Ernest-Ansermet, 1211 Geneva-4, Switzerland
| | - Bebiana C Sousa
- School of Life & Health Sciences, Aston University, Aston Triangle, Birmingham B47ET, UK
| | - Corinne M Spickett
- Life & Health Sciences and Aston Research Centre for Healthy Ageing, Aston University, Aston Triangle, Birmingham B4 7ET, UK
| | - Ana Stancic
- University of Belgrade, Institute for Biological Research "Sinisa Stankovic" and Faculty of Biology, Belgrade, Serbia
| | - Marie José Stasia
- Université Grenoble Alpes, CNRS, Grenoble INP, CHU Grenoble Alpes, TIMC-IMAG, F38000 Grenoble, France; CDiReC, Pôle Biologie, CHU de Grenoble, Grenoble, F-38043, France
| | - Holger Steinbrenner
- Institute of Nutrition, Department of Nutrigenomics, Friedrich Schiller University, Jena, Germany
| | - Višnja Stepanić
- Ruđer Bošković Institute, Division of Molecular Medicine, Zagreb, Croatia
| | - Sebastian Steven
- Molecular Cardiology, Center for Cardiology, Cardiology 1, University Medical Center Mainz, Mainz, Germany
| | - Kostas Tokatlidis
- Institute of Molecular Cell and Systems Biology, College of Medical Veterinary and Life Sciences, University of Glasgow, University Avenue, Glasgow, UK
| | - Erkan Tuncay
- Department of Biophysics, Ankara University, Faculty of Medicine, 06100 Ankara, Turkey
| | - Belma Turan
- Department of Biophysics, Ankara University, Faculty of Medicine, 06100 Ankara, Turkey
| | - Fulvio Ursini
- Department of Molecular Medicine, University of Padova, Padova, Italy
| | - Jan Vacek
- Department of Medical Chemistry and Biochemistry, Faculty of Medicine and Dentistry, Palacký University, Hnevotinska 3, Olomouc 77515, Czech Republic
| | - Olga Vajnerova
- Department of Physiology, 2nd Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Kateřina Valentová
- Institute of Microbiology, Laboratory of Biotransformation, Czech Academy of Sciences, Videnska 1083, CZ-142 20 Prague, Czech Republic
| | - Frank Van Breusegem
- Department of Plant Systems Biology, VIB, 9052 Ghent, Belgium; Department of Plant Biotechnology and Bioinformatics, Ghent University, 9052 Ghent, Belgium
| | - Lokman Varisli
- Harran University, Arts and Science Faculty, Department of Biology, Cancer Biology Lab, Osmanbey Campus, Sanliurfa, Turkey
| | - Elizabeth A Veal
- Institute for Cell and Molecular Biosciences, and Institute for Ageing, Newcastle University, Framlington Place, Newcastle upon Tyne, UK
| | - A Suha Yalçın
- Department of Biochemistry, School of Medicine, Marmara University, İstanbul, Turkey
| | | | - Neven Žarković
- Laboratory for Oxidative Stress, Rudjer Boskovic Institute, Bijenicka 54, 10000 Zagreb, Croatia
| | - Martina Zatloukalová
- Department of Medical Chemistry and Biochemistry, Faculty of Medicine and Dentistry, Palacký University, Hnevotinska 3, Olomouc 77515, Czech Republic
| | | | - Rhian M Touyz
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, UK
| | - Andreas Papapetropoulos
- Laboratoty of Pharmacology, Faculty of Pharmacy, National and Kapodistrian University of Athens, Greece
| | - Tilman Grune
- German Institute of Human Nutrition, Department of Toxicology, Arthur-Scheunert-Allee 114-116, 14558 Nuthetal, Germany
| | - Santiago Lamas
- Centro de Biología Molecular "Severo Ochoa" (CSIC-UAM), Madrid, Spain
| | - Harald H H W Schmidt
- Department of Pharmacology & Personalized Medicine, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Maastricht, The Netherlands
| | - Fabio Di Lisa
- Department of Biomedical Sciences and CNR Institute of Neuroscience, University of Padova, Padova, Italy.
| | - Andreas Daiber
- Molecular Cardiology, Center for Cardiology, Cardiology 1, University Medical Center Mainz, Mainz, Germany; DZHK (German Centre for Cardiovascular Research), partner site Rhine-Main, Mainz, Germany.
| |
Collapse
|
36
|
Park Y, Woo SH, Seo SK, Kim H, Noh WC, Lee JK, Kwon BM, Min KN, Choe TB, Park IC. Ginkgetin induces cell death in breast cancer cells via downregulation of the estrogen receptor. Oncol Lett 2017; 14:5027-5033. [PMID: 29085516 DOI: 10.3892/ol.2017.6742] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2016] [Accepted: 06/15/2017] [Indexed: 01/08/2023] Open
Abstract
Ginkgetin is a natural biflavonoid isolated from the leaves of Ginkgo biloba, and is characterized by its anti-inflammatory and anti-viral activities. Although numerous studies state that it has also antitumor activity, the anti-proliferative effect of ginkgetin and the underlying mechanism in breast cancer cells have not yet been investigated. In the present study, ginkgetin inhibited the cell viability of MCF-7 and T-47D cells dose-dependently, and suppressed the expression of the estrogen receptor (ER) at the mRNA and protein levels. Among the targets of the ER, 6-phosphofructo-2-kinase/fructose-2,6-bisphosphatase 3 (PFKFB3), cyclin D1 and survivin were also downregulated by ginkgetin treatment. The anti-proliferative effects of ginkgetin were sufficient to suppress the growth by estradiol stimulation. However, ginkgetin did not significantly affect the viability of MDA-MB-231 cells, which are ER-negative cells. Furthermore, the knockdown of the ER and an inhibitor of PFKFB3 significantly sensitized MCF-7 and T-47D cells to ginkgetin. These findings suggest that ginkgetin induces cell death in ER-positive breast cancer cells via the inhibition of ER expression and that it is a promising agent for breast cancer treatment.
Collapse
Affiliation(s)
- Yoonhwa Park
- Division of Radiation Cancer Research, Korea Institute of Radiological and Medical Sciences, Nowon, Seoul, Gyeonggi 01812, Republic of Korea.,School of Life Science and Biotechnology, Korea University, Seongbuk, Seoul, Gyeonggi 02841, Republic of Korea
| | - Sang Hyeok Woo
- Division of Radiation Cancer Research, Korea Institute of Radiological and Medical Sciences, Nowon, Seoul, Gyeonggi 01812, Republic of Korea.,KIRAMS Radiation Biobank, Korea Institute of Radiological and Medical Sciences, Nowon, Seoul, Gyeonggi 01812, Republic of Korea
| | - Sung-Keum Seo
- Division of Radiation Cancer Research, Korea Institute of Radiological and Medical Sciences, Nowon, Seoul, Gyeonggi 01812, Republic of Korea
| | - Hyunggee Kim
- School of Life Science and Biotechnology, Korea University, Seongbuk, Seoul, Gyeonggi 02841, Republic of Korea
| | - Woo Chul Noh
- Department of Surgery, Korea Cancer Center Hospital, Korea Institute of Radiological and Medical Sciences, Nowon, Seoul, Gyeonggi 01812, Republic of Korea
| | - Jin Kyung Lee
- KIRAMS Radiation Biobank, Korea Institute of Radiological and Medical Sciences, Nowon, Seoul, Gyeonggi 01812, Republic of Korea
| | - Byoung-Mog Kwon
- Laboratory of Chemical Biology and Genomics, Korea Research Institute of Bioscience and Biotechnology, Yuseong, Daejeon, Chungcheong 34141, Republic of Korea
| | - Kyung Nam Min
- Department of Microbiological Engineering, Kon-Kuk University, Gwangjin, Seoul, Gyeonggi 05029, Republic of Korea
| | - Tae-Boo Choe
- Department of Microbiological Engineering, Kon-Kuk University, Gwangjin, Seoul, Gyeonggi 05029, Republic of Korea
| | - In-Chul Park
- Division of Radiation Cancer Research, Korea Institute of Radiological and Medical Sciences, Nowon, Seoul, Gyeonggi 01812, Republic of Korea
| |
Collapse
|
37
|
Crosstalk of ROS/RNS and autophagy in silibinin-induced apoptosis of MCF-7 human breast cancer cells in vitro. Acta Pharmacol Sin 2017; 38:277-289. [PMID: 27867187 DOI: 10.1038/aps.2016.117] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/12/2016] [Accepted: 09/02/2016] [Indexed: 02/06/2023]
Abstract
Reactive oxygen species (ROS) and reactive nitrogen species (RNS) play important roles in regulating cell survival and death. Silibinin is a natural polyphenolic flavonoid isolated from milk thistle with anti-tumor activities, but it was found to induce cytoprotective ROS/RNS in human breast cancer MCF-7 cells. Furthermore, treatment with silibinin down-regulates ERα expression in MCF-7 cells, and inducing both autophagy and apoptosis. In this study we explored the relationship between ER-associated pathways and RNS/ROS in MCF-7 cells. We also investigated the molecular mechanisms underlying the reciprocal regulation between ROS/RNS levels and autophagy in the death signaling pathways in silibinin-treated MCF-7 cells. Silibinin (100-300 μmol/L) dose-dependently increased ROS/RNS generation in MCF-7 cells (with high expression of ERα and low expression of ERβ) and MDA-MB-231 cells (with low expression of ERα and high expression of ERβ). Scavenging ROS/RNS significantly enhanced silibinin-induced death of MCF-7 cells, but not MDA-MB231 cells. Pharmacological activation or blockade of ERα in MCF-7 cells significantly enhanced or decreased, respectively, silibinin-induced ROS/RNS generation, whereas activation or block of ERβ had no effect. In silibinin-treated MCF-7 cells, exposure to the ROS/RNS donators decreased the autophagic levels, whereas inhibition of autophagy with 3-MA significantly increased ROS/RNS levels. We further showed that increases in ROS/RNS generation, ERα activation or autophagy down-regulation had protective roles in silibinin-treated MCF-7 cells. Under a condition of ERα activation, scavenging ROS/RNS or stimulating autophagy enhanced the cytotoxicity of silibinin. These results demonstrate the existence of two conflicting pathways in silibinin-induced death of MCF-7 cells: one involves the down-regulation of ERα and thereby augmenting the pro-apoptotic autophagy downstream, leading to cell death; the other involves the up-regulation of pro-survival ROS/RNS; and that the generation of ROS/RNS and autophagy form a negative feedback loop whose balance is regulated by ERα.
Collapse
|
38
|
Jiang C, Jin S, Jiang Z, Wang J. Inhibitory effects of silibinin on proliferation and lung metastasis of human high metastasis cell line of salivary gland adenoid cystic carcinoma via autophagy induction. Onco Targets Ther 2016; 9:6609-6618. [PMID: 27822066 PMCID: PMC5087781 DOI: 10.2147/ott.s107101] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
OBJECTIVE To investigate the possible mechanisms and effects of silibinin (SIL) on the proliferation and lung metastasis of human lung high metastasis cell line of salivary gland adenoid cystic carcinoma (ACC-M). METHODS A methyl thiazolyl tetrazolium assay was performed to detect the inhibitory effects of SIL on the proliferation of ACC-M cells in vitro. Fluorescence microscopy and transmission electron microscopy were used to observe the autophagic process. Western blot was performed to detect the expression of microtube-related protein 1 light-chain 3 (LC3). An experimental adenoid cystic carcinoma (ACC) lung metastasis model was established in nude mice to detect the impacts of SIL on lung weight and lung cancer nodules. Immunohistochemistry was used to detect the expressions of LC3 in human ACC samples and normal salivary gland tissue samples. RESULTS SIL inhibited the proliferation of ACC-M cells in a dose- and time-dependent manner, and inductively increased the autophagic bodies in ACC-M cells. Furthermore, SIL could increase the expression of LC3 in ACC-M cells and promote the conversion of LC3-I into LC3-II in a dose- and time-dependent manner. In the ACC lung metastasis model, the lung weight and left and right lung nodules in the SIL-treated group were significantly less than those in the control group (P<0.05). The expressions of LC3-I and LC3-II as well as the positive expression rate of LC3 (80%) significantly increased, but the positive expression of LC3 in human ACC (42.22%) reduced significantly. CONCLUSION SIL could inhibit the proliferation and lung metastasis of ACC-M cells by possibly inducing tumor cells autophagy.
Collapse
Affiliation(s)
- Canhua Jiang
- Department of Oral and Maxillofacial Surgery, Xiangya Hospital
| | - Shufang Jin
- Department of Oral and Maxillofacial Surgery, Xiangya Hospital
| | - Zhisheng Jiang
- Department of Oral and Maxillofacial Surgery, Xiangya Hospital
| | - Jie Wang
- Department of Immunology, Xiangya School of Medicine, Central South University, Changsha, Hunan, People's Republic of China
| |
Collapse
|
39
|
The Anti-Cancer Effect of Polyphenols against Breast Cancer and Cancer Stem Cells: Molecular Mechanisms. Nutrients 2016; 8:nu8090581. [PMID: 27657126 PMCID: PMC5037565 DOI: 10.3390/nu8090581] [Citation(s) in RCA: 102] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2016] [Revised: 08/25/2016] [Accepted: 09/09/2016] [Indexed: 02/07/2023] Open
Abstract
The high incidence of breast cancer in developed and developing countries, and its correlation to cancer-related deaths, has prompted concerned scientists to discover novel alternatives to deal with this challenge. In this review, we will provide a brief overview of polyphenol structures and classifications, as well as on the carcinogenic process. The biology of breast cancer cells will also be discussed. The molecular mechanisms involved in the anti-cancer activities of numerous polyphenols, against a wide range of breast cancer cells, in vitro and in vivo, will be explained in detail. The interplay between autophagy and apoptosis in the anti-cancer activity of polyphenols will also be highlighted. In addition, the potential of polyphenols to target cancer stem cells (CSCs) via various mechanisms will be explained. Recently, the use of natural products as chemotherapeutics and chemopreventive drugs to overcome the side effects and resistance that arise from using chemical-based agents has garnered the attention of the scientific community. Polyphenol research is considered a promising field in the treatment and prevention of breast cancer.
Collapse
|
40
|
Cho YJ, Woo JH, Lee JS, Jang DS, Lee KT, Choi JH. Eclalbasaponin II induces autophagic and apoptotic cell death in human ovarian cancer cells. J Pharmacol Sci 2016; 132:6-14. [DOI: 10.1016/j.jphs.2016.02.006] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2015] [Revised: 12/29/2015] [Accepted: 02/18/2016] [Indexed: 02/09/2023] Open
|
41
|
|
42
|
Zhu XX, Ding YH, Wu Y, Qian LY, Zou H, He Q. Silibinin: a potential old drug for cancer therapy. Expert Rev Clin Pharmacol 2016; 9:1323-1330. [PMID: 27362364 DOI: 10.1080/17512433.2016.1208563] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
INTRODUCTION Silibinin is mixture of flavonolignans extracted from milk thistle and often has been used in the treatment of acute and chronic liver disorders caused by toxins, drug, alcohol and hepatitis and gall bladder disorders for its antioxidant and hepatoprotective properties. Areas covered: However, increasing evidence suggest that silibinin is not solely limited in the treatment of these diseases. Further research suggests that silymarin may function diversely and may serve as a novel therapy for cancer therapy, such as lung cancer, prostatic cancer, colon cancer, breast cancer, bladder cancer and hepatocellular carcinoma by regulating cancer cells growth, proliferation, apoptosis, angiogenesis and many other mechanism. Expert commentary: In this review, in order to provide potential new treatment for these cancer, we summarize the recent anti-cancer findings of silibinin in these cancer and clarify the mechanisms of this effect.
Collapse
Affiliation(s)
- Xing-Xing Zhu
- a Department of Nephrology , Zhejiang Provincial People's Hospital , Hangzhou , China
| | - Ya-Hui Ding
- b Department of Cardiology , Zhejiang Provincial People's Hospital , Hangzhou , China
| | - Yi Wu
- c Department of Hematology , Zhejiang Provincial People's Hospital , Hangzhou , China
| | - Lin-Yan Qian
- b Department of Cardiology , Zhejiang Provincial People's Hospital , Hangzhou , China
| | - Hai Zou
- b Department of Cardiology , Zhejiang Provincial People's Hospital , Hangzhou , China
| | - Qiang He
- a Department of Nephrology , Zhejiang Provincial People's Hospital , Hangzhou , China
| |
Collapse
|
43
|
Li F, Zheng X, Liu Y, Li P, Liu X, Ye F, Zhao T, Wu Q, Jin X, Li Q. Different Roles of CHOP and JNK in Mediating Radiation-Induced Autophagy and Apoptosis in Breast Cancer Cells. Radiat Res 2016; 185:539-48. [PMID: 27135967 DOI: 10.1667/rr14344.1] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
Unfolded protein response (UPR) is comprised of complex and conserved stress pathways that function as a short-term adaptive mechanism to reduce levels of unfolded or misfolded proteins and maintain homeostasis in the endoplasmic reticulum (ER). UPR can be triggered by prolonged or persistent ER stress under many physiological or pathological conditions, including radiation exposure. Radiation-induced ER stress elicits autophagy and apoptosis in cancer cells, where C/EBP homologous protein (CHOP) and c-Jun NH2-terminal kinase (JNK) may play crucial roles. However, the specific mechanisms that regulate autophagy and apoptosis through CHOP and JNK after radiation exposure and how the balance of these activities determines the cellular radiosensitivity remain largely unclear. In this study, we found that exposure to X-ray radiation induced ER stress, UPR and high expression of CHOP and JNK. Furthermore, autophagy and apoptosis occurred in sequential order when breast cancer MDA-MB-231 and MCF-7 cells were exposed to X-ray radiation. CHOP gene knockdown with RNA interference inhibited autophagy and enhanced radiosensitivity in MDA-MB-231 cells, while impacting apoptosis and subsequently increasing radioresistance in MCF-7 cells. However, treatment with JNK inhibitor decreased autophagy while promoting apoptosis, thereby leading to radiosensitivity in both cell lines. Our results indicate that CHOP mediates radiation-induced autophagy and apoptosis in a cellular environment. Importantly, the functional consistency of regulating apoptosis and autophagy in these two irradiated breast cancer cell lines suggests that JNK may be more useful as a potential target for maximizing the efficacy of radiation therapy for breast cancers.
Collapse
Affiliation(s)
- Feifei Li
- a Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou 730000, China;,b Key Laboratory of Heavy Ion Radiation Biology and Medicine of Chinese Academy of Sciences, Lanzhou 730000, China;,c Key Laboratory of Basic Research on Heavy Ion Radiation Application in Medicine, Gansu Province, Lanzhou 730000, China; and.,d University of Chinese Academy of Sciences, Beijing 100049, China
| | - Xiaogang Zheng
- a Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou 730000, China;,b Key Laboratory of Heavy Ion Radiation Biology and Medicine of Chinese Academy of Sciences, Lanzhou 730000, China;,c Key Laboratory of Basic Research on Heavy Ion Radiation Application in Medicine, Gansu Province, Lanzhou 730000, China; and.,d University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yan Liu
- a Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou 730000, China;,b Key Laboratory of Heavy Ion Radiation Biology and Medicine of Chinese Academy of Sciences, Lanzhou 730000, China;,c Key Laboratory of Basic Research on Heavy Ion Radiation Application in Medicine, Gansu Province, Lanzhou 730000, China; and.,d University of Chinese Academy of Sciences, Beijing 100049, China
| | - Ping Li
- a Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou 730000, China;,b Key Laboratory of Heavy Ion Radiation Biology and Medicine of Chinese Academy of Sciences, Lanzhou 730000, China;,c Key Laboratory of Basic Research on Heavy Ion Radiation Application in Medicine, Gansu Province, Lanzhou 730000, China; and
| | - Xiongxiong Liu
- a Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou 730000, China;,b Key Laboratory of Heavy Ion Radiation Biology and Medicine of Chinese Academy of Sciences, Lanzhou 730000, China;,c Key Laboratory of Basic Research on Heavy Ion Radiation Application in Medicine, Gansu Province, Lanzhou 730000, China; and
| | - Fei Ye
- a Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou 730000, China;,b Key Laboratory of Heavy Ion Radiation Biology and Medicine of Chinese Academy of Sciences, Lanzhou 730000, China;,c Key Laboratory of Basic Research on Heavy Ion Radiation Application in Medicine, Gansu Province, Lanzhou 730000, China; and.,d University of Chinese Academy of Sciences, Beijing 100049, China
| | - Ting Zhao
- a Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou 730000, China;,b Key Laboratory of Heavy Ion Radiation Biology and Medicine of Chinese Academy of Sciences, Lanzhou 730000, China;,c Key Laboratory of Basic Research on Heavy Ion Radiation Application in Medicine, Gansu Province, Lanzhou 730000, China; and
| | - Qingfeng Wu
- a Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou 730000, China
| | - Xiaodong Jin
- a Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou 730000, China;,b Key Laboratory of Heavy Ion Radiation Biology and Medicine of Chinese Academy of Sciences, Lanzhou 730000, China;,c Key Laboratory of Basic Research on Heavy Ion Radiation Application in Medicine, Gansu Province, Lanzhou 730000, China; and
| | - Qiang Li
- a Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou 730000, China;,b Key Laboratory of Heavy Ion Radiation Biology and Medicine of Chinese Academy of Sciences, Lanzhou 730000, China;,c Key Laboratory of Basic Research on Heavy Ion Radiation Application in Medicine, Gansu Province, Lanzhou 730000, China; and
| |
Collapse
|
44
|
Zheng N, Liu L, Liu W, Zhang P, Huang H, Zang L, Hayashi T, Tashiro SI, Onodera S, Xia M, Ikejima T. ERβ up-regulation was involved in silibinin-induced growth inhibition of human breast cancer MCF-7 cells. Arch Biochem Biophys 2016; 591:141-9. [DOI: 10.1016/j.abb.2016.01.002] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2015] [Revised: 12/30/2015] [Accepted: 01/03/2016] [Indexed: 12/11/2022]
|
45
|
Vriend J, Reiter RJ. Breast cancer cells: Modulation by melatonin and the ubiquitin-proteasome system--a review. Mol Cell Endocrinol 2015; 417:1-9. [PMID: 26363225 DOI: 10.1016/j.mce.2015.09.001] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/11/2015] [Accepted: 09/01/2015] [Indexed: 02/07/2023]
Abstract
Melatonin inhibits human breast cancer cells stimulated with estrogen. This antiproliferative action depends on the presence of the estrogen receptor alpha (ERα) in the human MCF-7 cell line and is strictly dose-dependent. Since researchers concerned with melatonin and breast cancer have not considered the relevance of the ubiquitin-proteasome system to this research in this review we do so. The fact that the first breast cancer susceptibility gene to be identified, Brca1, functions as a ubiquitin ligase indicates that the ubiquitin-proteasome system has a role in regulating susceptibility to breast cancer. While mutations of this gene increase the incidence of breast cancer, the wild type gene suppresses estrogen-dependent transcriptional events relying on the estrogen receptor ERα. Three other ubiquitin ligases, SCF(Skp2), E6AP and APC, interact directly with ERα at the ERE and AP-1 promoters of ERα target genes. Melatonin, like proteasome inhibitors, decreases estrogen-induced gene transcription. Indeed, it has been reported that melatonin specifically inhibits estrogen-induced transcription mediated by ERα at the ERE and AP1 gene promoters. Herein, we present a model in which the inhibitory action of melatonin on MCF-7 cells is mediated, directly or indirectly, by the ubiquitin-proteasome system. In this model ERα, apoptotic proteins, and cell cycle proteins, all influenced by melatonin, are substrates of key ubiquitin ligases including SCF(Skp2), E6AP, and SCF(B-TrCP). Since dysfunction of the ubiquitin-proteasome system is a risk factor for breast cancer, this model provides a context in which to test the clinical potential, and limitations, of melatonin and proteasome inhibitors.
Collapse
Affiliation(s)
- Jerry Vriend
- Department of Human Anatomy and Cell Science, University of Manitoba, Winnipeg, MB, Canada.
| | - Russel J Reiter
- Department of Cellular and Structural Biology, UT Health Science Center San Antonio, TX, USA
| |
Collapse
|
46
|
Resveratrol, a potential radiation sensitizer for glioma stem cells both in vitro and in vivo. J Pharmacol Sci 2015; 129:216-25. [DOI: 10.1016/j.jphs.2015.11.001] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2015] [Revised: 10/27/2015] [Accepted: 10/29/2015] [Indexed: 11/19/2022] Open
|
47
|
Ma Z, Liu W, Zeng J, Zhou J, Guo P, Xie H, Yang Z, Zheng L, Xu S, Wang X, Chang LS, He D, Li L. Silibinin induces apoptosis through inhibition of the mTOR-GLI1-BCL2 pathway in renal cell carcinoma. Oncol Rep 2015; 34:2461-8. [PMID: 26323996 DOI: 10.3892/or.2015.4224] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2015] [Accepted: 07/22/2015] [Indexed: 11/05/2022] Open
Abstract
The downstream transcriptional factor of the hedgehog (Hh) pathway, GLI family zinc finger 1 (GLI1), plays a crucial role in regulating tumor progression. In the present study, we demonstrated that silibinin, a natural flavonoid antioxidant isolated from extracts of the milk thistle herb, exerts its anticancer capabilities by restraining GLI1 function in renal cell carcinoma (RCC) cells in vitro and in vivo. In the present study, we confirmed that silibinin induced growth inhibition of RCC through caspase-dependent apoptosis and downregulation of GLI1 and BCL2, which could be partially reversed by GLI1 overexpression. Moreover, we determined that the decreased GLI1 expression by silibinin was mediated by the mammalian target of rapamycin (mTOR) pathway. The in vivo mouse xenograft study also showed that silibinin significantly reduced RCC tumor growth and specifically targeted the mTOR-GLI1-BCL2 signaling pathway. In conclusion, our findings demonstrated for the first time that silibinin induces apoptosis of RCC cells through inhibition of the mTOR-GLI1‑BCL2 pathway. These findings also indicate that GLI1 is a novel regulator for the potential therapeutic application of silibinin against RCC.
Collapse
Affiliation(s)
- Zhenkun Ma
- Department of Urology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| | - Wei Liu
- Department of Urology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| | - Jin Zeng
- Department of Urology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| | - Jiancheng Zhou
- Department of Urology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| | - Peng Guo
- Department of Urology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| | - Hongjun Xie
- Department of Urology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| | - Zhao Yang
- Department of Urology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| | - Long Zheng
- Department of Urology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| | - Shan Xu
- Department of Urology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| | - Xinyang Wang
- Department of Urology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| | - Luke S Chang
- Department of Urology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| | - Dalin He
- Department of Urology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| | - Lei Li
- Department of Urology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| |
Collapse
|