1
|
Chen M, Zhang S, Huang X, Zhang D, Zhu D, Ouyang C, Li Y. The protective effects and mechanism of myricetin in liver diseases (Review). Mol Med Rep 2025; 31:87. [PMID: 39917997 PMCID: PMC11811602 DOI: 10.3892/mmr.2025.13452] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2024] [Accepted: 01/22/2025] [Indexed: 02/13/2025] Open
Abstract
Liver diseases have become one of the significant threats to global health. However, there is a lack of effective targeted therapeutic drugs in this field and the existing drugs used for liver disease treatment usually have side‑effects. Traditional Chinese medicine (TCM) has the distinctive advantages of multi‑target and low side‑effects. As a flavonoid with various pharmacological activities such as anti‑tumour, anti‑oxidant, anti‑inflammatory and anti‑bacterial, the TCM myricetin has been widely used in liver disease research. The present work focuses on the role and molecular mechanism of myricetin in liver diseases such as acute liver injury, fatty liver, liver fibrosis and hepatocellular carcinoma. It is a promising reference for further research and application of myricetin in the treatment of liver diseases.
Collapse
Affiliation(s)
- Mi Chen
- Hubei Key Laboratory of Diabetes and Angiopathy, School of Pharmacy, Hubei University of Science and Technology, Xianning, Hubei 437100, P.R. China
| | - Shengnan Zhang
- Hubei Key Laboratory of Diabetes and Angiopathy, School of Pharmacy, Hubei University of Science and Technology, Xianning, Hubei 437100, P.R. China
- School of Resources and Environmental Science and Engineering, Hubei University of Science and Technology, Xianning, Hubei 437100, P.R. China
| | - Xingqiong Huang
- Hubei Key Laboratory of Diabetes and Angiopathy, School of Pharmacy, Hubei University of Science and Technology, Xianning, Hubei 437100, P.R. China
| | - Dandan Zhang
- Hubei Key Laboratory of Diabetes and Angiopathy, School of Pharmacy, Hubei University of Science and Technology, Xianning, Hubei 437100, P.R. China
| | - Dan Zhu
- Hubei Key Laboratory of Diabetes and Angiopathy, School of Pharmacy, Hubei University of Science and Technology, Xianning, Hubei 437100, P.R. China
| | - Changhan Ouyang
- Hubei Key Laboratory of Diabetes and Angiopathy, School of Pharmacy, Hubei University of Science and Technology, Xianning, Hubei 437100, P.R. China
| | - Yankun Li
- Hubei Key Laboratory of Diabetes and Angiopathy, School of Pharmacy, Hubei University of Science and Technology, Xianning, Hubei 437100, P.R. China
| |
Collapse
|
2
|
Kustiawan PM, Siregar KAAK, Jauhar MM, Ramadhan D, Mardliyati E, Syaifie PH. Network pharmacology and bioinformatic integrative analysis reveals candidate gene targets and potential therapeutic of East Kalimantan propolis against hepatocellular carcinoma. Heliyon 2024; 10:e39142. [PMID: 39524833 PMCID: PMC11544044 DOI: 10.1016/j.heliyon.2024.e39142] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2024] [Revised: 10/01/2024] [Accepted: 10/08/2024] [Indexed: 11/16/2024] Open
Abstract
Introduction Hepatocellular Carcinoma (HCC) is commonly treated with surgery, liver transplantation, and chemotherapy, but recurrence and metastasis remain challenges. Natural complementary therapies like propolis, known for its hepatoprotective properties, are gaining interest due to limited efficacy and toxicity of conventional chemotherapy. This study aims to identify core targets for HCC, assess the therapeutic potential of East Kalimantan propolis (EKP) from stingless bees, and analyze the molecular interactions. Methods EKP compounds were analyzed using target prediction tools related to HCC, alongside clinical data from the Gene Expression Omnibus (GEO) database, to identify overlapping genes with clinical relevance. The selected genes were then subjected to protein-protein interaction (PPI), GO and KEGG enrichment, immunohistochemical comparison and survival analysis to identify potential core targets and related pathways for HCC therapy. Furthermore, molecular docking and dynamics were conducted to verify the molecular interactions and stability of EKP compounds with targets. Results 108 genes have been selected as HCC potential targets, which mostly associated with MicroRNAs in cancer, chemical carcinogenesis, and viral carcinogenesis pathways. These targets were obtained by overlapping genes from GEO clinical databases and target predictors. PPI network analysis revealed 4 main targets of propolis in HCC. Furthermore, differential expression genes, survival analysis, and Immunohistochemical analysis from databases suggested that AKR1C3 and MAPK1 promote HCC progression and shorten survival rate of HCC patients. Molecular docking and dynamic studies confirmed strong binding affinity and stability of Baicalein, Chrysin, Quercetin, and Myricetin with receptor targets within simulation time. Conclusions This study provides insight into the mechanism of action of EKP on HCC and identifies AKR1C3 and MAPK1 as candidate target treatments for future drug development.
Collapse
Affiliation(s)
- Paula Mariana Kustiawan
- Faculty of Pharmacy, Universitas Muhammadiyah Kalimantan Timur, Samarinda, East Kalimantan, 75124, Indonesia
| | - Khalish Arsy Al Khairy Siregar
- Faculty of Pharmacy, Universitas Muhammadiyah Kalimantan Timur, Samarinda, East Kalimantan, 75124, Indonesia
- Center of Excellence Life Sciences, Nano Center Indonesia, South Tangerang, 15314, Indonesia
| | - Muhammad Miftah Jauhar
- Center of Excellence Life Sciences, Nano Center Indonesia, South Tangerang, 15314, Indonesia
| | - Donny Ramadhan
- Research Center for Pharmaceutical Ingredients and Traditional Medicine, National Research and Innovation Agency (BRIN), Bogor, Indonesia
| | - Etik Mardliyati
- Research Center for Vaccine and Drug, National Research and Innovation Agency (BRIN), Bogor, 16911, Indonesia
| | - Putri Hawa Syaifie
- Center of Excellence Life Sciences, Nano Center Indonesia, South Tangerang, 15314, Indonesia
| |
Collapse
|
3
|
Li S, Hao L, Li N, Hu X, Yan H, Dai E, Shi X. Targeting the Hippo/YAP1 signaling pathway in hepatocellular carcinoma: From mechanisms to therapeutic drugs (Review). Int J Oncol 2024; 65:88. [PMID: 39092548 DOI: 10.3892/ijo.2024.5676] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Accepted: 07/15/2024] [Indexed: 08/04/2024] Open
Abstract
The Hippo signaling pathway plays a pivotal role in regulating cell growth and organ size. Its regulatory effects on hepatocellular carcinoma (HCC) encompass diverse aspects, including cell proliferation, invasion and metastasis, tumor drug resistance, metabolic reprogramming, immunomodulatory effects and autophagy. Yes‑associated protein 1 (YAP1), a potent transcriptional coactivator and a major downstream target tightly controlled by the Hippo pathway, is influenced by various molecules and pathways. The expression of YAP1 in different cell types within the liver tumor microenvironment exerts varying effects on tumor outcomes, warranting careful consideration. Therefore, research on YAP1‑targeted therapies merits attention. This review discusses the composition and regulation mechanism of the Hippo/YAP1 signaling pathway and its relationship with HCC, offering insights for future research and cancer prevention strategies.
Collapse
Affiliation(s)
- Shenghao Li
- Clinical Medical College, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan 610075, P.R. China
| | - Liyuan Hao
- Clinical Medical College, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan 610075, P.R. China
| | - Na Li
- Clinical Medical College, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan 610075, P.R. China
| | - Xiaoyu Hu
- Department of Infectious Diseases, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan 610072, P.R. China
| | - Huimin Yan
- Clinical Research Center, Shijiazhuang Fifth Hospital, Shijiazhuang, Hebei 050024, P.R. China
| | - Erhei Dai
- Clinical Research Center, Shijiazhuang Fifth Hospital, Shijiazhuang, Hebei 050024, P.R. China
| | - Xinli Shi
- Center of Experimental Management, Shanxi University of Chinese Medicine, Jinzhong, Shanxi 030619, P.R. China
| |
Collapse
|
4
|
Yang J, Chen J, Li Q, Xu RA, Chen X. Effects of three flavonoids on the metabolism of lenvatinib. Front Pharmacol 2024; 15:1438259. [PMID: 39228528 PMCID: PMC11368737 DOI: 10.3389/fphar.2024.1438259] [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: 05/25/2024] [Accepted: 07/26/2024] [Indexed: 09/05/2024] Open
Abstract
Lenvatinib is a first-line therapy for the treatment of hepatocellular carcinoma (HCC), an active multi-target tyrosine kinase inhibitor (TKI). The interaction between Traditional Chinese Medicine (TCM) and chemicals has increasingly become a research hotspot. The objective of this study was to pinpoint the effects of three flavonoids on the metabolism of lenvatinib. Enzyme reaction system was established and optimized in vitro, and in vivo experiments were conducted in Sprague-Dawley (SD) rats, where the analytes were detected by ultra performance liquid chromatography tandem mass spectrometry (UPLC-MS/MS). We found that among three flavonoids, luteolin and myricetin had strong inhibitory effects on lenvatinib metabolism, with half-maximal inhibitory concentration (IC50) values of 11.36 ± 0.46 µM and 11.21 ± 0.81 µM in rat liver microsomes (RLM), respectively, and 6.89 ± 0.43 µM and 12.32 ± 1.21 µM in human liver microsomes (HLM), respectively. In Sprague-Dawley rats, the combined administration of lenvatinib and luteolin obviously expanded the exposure to lenvatinib; however, co-administered with myricetin did not have any changes, which may be due to the poor bioavailability of myricetin in vivo. Furthermore, the inhibitory type of luteolin on lenvatinib showed an un-competitive in RLM and a mixed in HLM. Collectively, flavonoids with liver protection, especially luteolin, may inhibit lenvatinib metabolism in vitro and in vivo.
Collapse
Affiliation(s)
- Jinzhao Yang
- Wenzhou People’s Hospital, The Third Clinical Institute Affiliated to Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Jie Chen
- The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Qingqing Li
- The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Ren-ai Xu
- The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Xiaohai Chen
- The Third Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| |
Collapse
|
5
|
Li J, Luo T, Zhao Y, Wang D, Jin Y, Wu Z, Yang G, Qi X. Cardioprotective potentials of myricetin on doxorubicin-induced cardiotoxicity based on biochemical and transcriptomic analysis. Biomed Pharmacother 2024; 175:116748. [PMID: 38776683 DOI: 10.1016/j.biopha.2024.116748] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Revised: 05/10/2024] [Accepted: 05/10/2024] [Indexed: 05/25/2024] Open
Abstract
Doxorubicin (DOX) is a commonly used anthracycline in cancer chemotherapy. The clinical application of DOX is constrained by its cardiotoxicity. Myricetin (MYR) is a natural flavonoid widely present in many plants with antioxidant and anti-inflammatory properties. However, MYR's beneficial effects and mechanisms in alleviating DOX-induced cardiotoxicity (DIC) remain unknown. C57BL/6 mice were injected with 15 mg/kg of DOX to establish the DIC, and MYR solutions were administrated by gavage to investigate its cardioprotective potentials. Histopathological analysis, physiological indicators assessment, transcriptomics analysis, and RT-qPCR were used to elucidate the potential mechanism of MYR in DIC treatment. MYR reduced cardiac injury produced by DOX, decreased levels of cTnI, AST, LDH, and BNP, and improved myocardial injury and fibrosis. MYR effectively prevented DOX-induced oxidative stress, such as lowered MDA levels and elevated SOD, CAT, and GSH activities. MYR effectively suppressed NLRP3 and ASC gene expression levels to inhibit pyroptosis while regulating Caspase1 and Bax levels to reduce cardiac cell apoptosis. According to the transcriptomic analysis, glucose and fatty acid metabolism were associated with differential gene expression. KEGG pathway analysis revealed differential gene enrichment in PPAR and AMPK pathways, among others. Following validation, MYR was found to alleviate DIC by regulating glycolipid metabolism and AMPK pathway-related genes. Our findings demonstrated that MYR could mitigate DIC by regulating the processes of oxidative stress, apoptosis, and pyroptosis. MYR is critical in improving DOX-induced myocardial energy metabolism abnormalities mediated by the AMPK signaling pathway. In conclusion, MYR holds promise as a therapeutic strategy for DIC.
Collapse
Affiliation(s)
- Jaili Li
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-Products, Ningbo University, Ningbo, Zhejiang Province 315832, China; State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-Products, Laboratory (Hangzhou) for Risk Assessment of Agricultural Products of Ministry of Agriculture, Institute of Agro-Product Safety and Nutrition, Zhejiang Academy of Agricultural Sciences, Hangzhou, Zhejiang Province 310021, China
| | - Ting Luo
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-Products, Ningbo University, Ningbo, Zhejiang Province 315832, China; State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-Products, Laboratory (Hangzhou) for Risk Assessment of Agricultural Products of Ministry of Agriculture, Institute of Agro-Product Safety and Nutrition, Zhejiang Academy of Agricultural Sciences, Hangzhou, Zhejiang Province 310021, China.
| | - Yao Zhao
- Xianghu Laboratory, Hangzhou, Zhejiang Province 311231, China
| | - Dou Wang
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-Products, Ningbo University, Ningbo, Zhejiang Province 315832, China; State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-Products, Laboratory (Hangzhou) for Risk Assessment of Agricultural Products of Ministry of Agriculture, Institute of Agro-Product Safety and Nutrition, Zhejiang Academy of Agricultural Sciences, Hangzhou, Zhejiang Province 310021, China; Key Laboratory of Traceability for Agricultural Genetically Modified Organisms, Ministry of Agriculture and Rural Affairs, China
| | - Yuanxiang Jin
- Xianghu Laboratory, Hangzhou, Zhejiang Province 311231, China; College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou, Zhejiang Province 310032, China
| | - Zufang Wu
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-Products, Ningbo University, Ningbo, Zhejiang Province 315832, China.
| | - Guiling Yang
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-Products, Ningbo University, Ningbo, Zhejiang Province 315832, China; State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-Products, Laboratory (Hangzhou) for Risk Assessment of Agricultural Products of Ministry of Agriculture, Institute of Agro-Product Safety and Nutrition, Zhejiang Academy of Agricultural Sciences, Hangzhou, Zhejiang Province 310021, China; Xianghu Laboratory, Hangzhou, Zhejiang Province 311231, China.
| | - Xingjiang Qi
- Xianghu Laboratory, Hangzhou, Zhejiang Province 311231, China.
| |
Collapse
|
6
|
Singh B, Semwal BC. A Compressive Review on Source, Toxicity and Biological Activity of Flavonoid. Curr Top Med Chem 2024; 24:2093-2116. [PMID: 39108008 DOI: 10.2174/0115680266316032240718050055] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2024] [Revised: 06/05/2024] [Accepted: 06/25/2024] [Indexed: 10/22/2024]
Abstract
Flavonoids are biologically active chemicals in various fruits, plants, vegetables, and leaves, which have promising uses in medicinal science. The health properties of these natural chemicals are widely accepted, and efforts are underway to extract the specific components referred to as flavonoids. Flavonoids demonstrate a diverse range of bio-activities, anticancer, antioxidant activity, anti-cholinesterase activity, antiinflammatory activity, antimalarial activity, antidiabetic activity, neurodegenerative disease, cardiovascular effect, hepatoprotective effects, and antiviral and antimicrobial activity. This study aims to examine the prevailing trends in flavonoid investigation studies, elucidate the activity of flavonoids, examine their various functions and uses, assess the potential of flavonoids as preventive medications for chronic diseases, and outline future research opportunities in this field. This review explores the diverse functions of flavonoids in preventing and managing various diseases.
Collapse
Affiliation(s)
- Bhoopendra Singh
- Department of Pharmacology, GLA University, NH#2 Delhi Mathura Highway, Uttar Pradesh, India
| | - Bhupesh Chander Semwal
- Department of Pharmacology, GLA University, NH#2 Delhi Mathura Highway, Uttar Pradesh, India
| |
Collapse
|
7
|
Trivedi A, Hasan A, Ahmad R, Siddiqui S, Srivastava A, Misra A, Mir SS. Flavonoid Myricetin as Potent Anticancer Agent: A Possibility towards Development of Potential Anticancer Nutraceuticals. Chin J Integr Med 2024; 30:75-84. [PMID: 37340205 DOI: 10.1007/s11655-023-3701-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/01/2023] [Indexed: 06/22/2023]
Abstract
Good nutrition plays a crucial role in maintaining a balanced lifestyle. The beneficial effects of nutrition have been found to counteract nutritional disturbances with the expanded use of nutraceuticals to treat and manage cardiovascular diseases, cancer, and other developmental defects over the last decade. Flavonoids are found abundantly in plant-derived foods such as fruits, vegetables, tea, cocoa, and wine. Fruits and vegetables contain phytochemicals like flavonoids, phenolics, alkaloids, saponins, and terpenoids. Flavonoids can act as anti-inflammatory, anti-allergic, anti-microbial (antibacterial, antifungal, and antiviral) antioxidant, anti-cancer, and anti-diarrheal agents. Flavonoids are also reported to upregulate apoptotic activity in several cancers such as hepatic, pancreatic, breast, esophageal, and colon. Myricetin is a flavonol which is naturally present in fruits and vegetables and has shown possible nutraceutical value. Myricetin has been portrayed as a potent nutraceutical that may protect against cancer. The focus of the present review is to present an updated account of studies demonstrating the anticancer potential of myricetin and the molecular mechanisms involved therein. A better understanding of the molecular mechanism(s) underlying its anticancer activity would eventually help in its development as a novel anticancer nutraceutical having minimal side effects.
Collapse
Affiliation(s)
- Anchal Trivedi
- Department of Biochemistry, Era's Lucknow Medical College & Hospital, Era University, Lucknow, 226003, India
| | - Adria Hasan
- Molecular Cell Biology Laboratory, Integral Information and Research Centre-4 (IIRC-4), Department of Bioengineering, Faculty of Engineering, Integral University, Lucknow, 226026, India
| | - Rumana Ahmad
- Department of Biochemistry, Era's Lucknow Medical College & Hospital, Era University, Lucknow, 226003, India
| | - Sahabjada Siddiqui
- Department of Biotechnology, Era's Lucknow Medical College & Hospital, Era University, Lucknow, 226003, India
| | - Aditi Srivastava
- Department of Biochemistry, Era's Lucknow Medical College & Hospital, Era University, Lucknow, 226003, India
| | - Aparna Misra
- Department of Biochemistry, Era's Lucknow Medical College & Hospital, Era University, Lucknow, 226003, India
| | - Snober S Mir
- Molecular Cell Biology Laboratory, Integral Information and Research Centre-4 (IIRC-4), Department of Bioengineering, Faculty of Engineering, Integral University, Lucknow, 226026, India.
- Department of Biosciences, Faculty of Science, Integral University, Lucknow, 226026, India.
| |
Collapse
|
8
|
Shi H, Zou Y, Zhong W, Li Z, Wang X, Yin Y, Li D, Liu Y, Li M. Complex roles of Hippo-YAP/TAZ signaling in hepatocellular carcinoma. J Cancer Res Clin Oncol 2023; 149:15311-15322. [PMID: 37608027 DOI: 10.1007/s00432-023-05272-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Accepted: 08/09/2023] [Indexed: 08/24/2023]
Abstract
BACKGROUND The Hippo signaling pathway is an evolutionarily conserved signaling module that controls organ size in different species, and the disorder of the Hippo pathway can induce liver cancer in organisms, especially hepatocellular carcinoma (HCC). The exact mechanism that causes cancer is still unknown. Recent studies have shown that it is a classical kinase cascade that phosphorylates the Mst1/2-sav1 complex and activates the phosphorylation of the Lats1/2-mob1A/B complex for inactivating Yap and Taz. These kinases and scaffolds are regarded as primary regulators of the Hippo pathway, and help in activating a variety of carcinogenic processes. Among them, Yap/Taz is seen to be the main effector molecule, which is downstream of the Hippo pathway, and its abnormal activation is related to a variety of human cancers including liver cancer. Currently, since Yap/Taz plays a variety of roles in cancer promotion and tumor regeneration, the Hippo pathway has emerged as an attractive target in recent drug development research. METHODS We collect and review relevant literature in web of Science and Pubmed. CONCLUSION This review highlights the important roles of Yap/Taz in activating Hippo pathway in liver cancer. The recent findings on the crosstalks between the Hippo and other cancer associated pathways and moleculars are also discussed. In this review, we summarized and discussed recent breakthroughs in our understanding of how key components of the Hippo-YAP/TAZ pathway influence the hepatocellular carcinoma, including their effects on tumor occurrence and development, their roles in regulating metastasis, and their function in chemotherapy resistance. Further, the molecular mechanism and roles in regulating cross talk between Hippo-YAP/TAZ pathway and other cancer-associated pathways or oncogenes/cancer suppressor genes were summarized and discussed. More, many other inducers and inhibitors of this signaling cascade and available experimental therapies against the YAP/TAZ/TEAD axis were discussed. Targeting this pathway for cancer therapy may have great significance in the treatment of hepatocellular carcinoma. Graphical summary of the complex role of Hippo-YAP/TAZ signaling in hepatocellular carcinoma.
Collapse
Affiliation(s)
- Hewen Shi
- Featured Laboratory for Biosynthesis and Target Discovery of Active Components of Traditional Chinese Medicine, School of Integrated Traditional Chinese and Western Medicine, Binzhou Medical University, Yantai, 264003, Shandong, People's Republic of China
| | - Ying Zou
- Featured Laboratory for Biosynthesis and Target Discovery of Active Components of Traditional Chinese Medicine, School of Integrated Traditional Chinese and Western Medicine, Binzhou Medical University, Yantai, 264003, Shandong, People's Republic of China
| | - Weiwei Zhong
- Featured Laboratory for Biosynthesis and Target Discovery of Active Components of Traditional Chinese Medicine, School of Integrated Traditional Chinese and Western Medicine, Binzhou Medical University, Yantai, 264003, Shandong, People's Republic of China
| | - Zhaoying Li
- Traditional Chinese Medicine Research Center, Shandong Public Health Clinical Center, Jinan, 250102, People's Republic of China
| | - Xiaoxue Wang
- Featured Laboratory for Biosynthesis and Target Discovery of Active Components of Traditional Chinese Medicine, School of Integrated Traditional Chinese and Western Medicine, Binzhou Medical University, Yantai, 264003, Shandong, People's Republic of China
| | - Yancun Yin
- School of Basic Medical Sciences, Binzhou Medical University, Yantai, 264003, People's Republic of China
| | - Defang Li
- Featured Laboratory for Biosynthesis and Target Discovery of Active Components of Traditional Chinese Medicine, School of Integrated Traditional Chinese and Western Medicine, Binzhou Medical University, Yantai, 264003, Shandong, People's Republic of China
| | - Ying Liu
- Featured Laboratory for Biosynthesis and Target Discovery of Active Components of Traditional Chinese Medicine, School of Integrated Traditional Chinese and Western Medicine, Binzhou Medical University, Yantai, 264003, Shandong, People's Republic of China.
| | - Minjing Li
- Featured Laboratory for Biosynthesis and Target Discovery of Active Components of Traditional Chinese Medicine, School of Integrated Traditional Chinese and Western Medicine, Binzhou Medical University, Yantai, 264003, Shandong, People's Republic of China.
| |
Collapse
|
9
|
Gupta M, Ahmad J, Ahamad J, Kundu S, Goel A, Mishra A. Flavonoids as promising anticancer therapeutics: Contemporary research, nanoantioxidant potential, and future scope. Phytother Res 2023; 37:5159-5192. [PMID: 37668281 DOI: 10.1002/ptr.7975] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2023] [Revised: 06/30/2023] [Accepted: 07/21/2023] [Indexed: 09/06/2023]
Abstract
Flavonoids are natural polyphenolic compounds considered safe, pleiotropic, and readily available molecules. It is widely distributed in various food products such as fruits and vegetables and beverages such as green tea, wine, and coca-based products. Many studies have reported the anticancer potential of flavonoids against different types of cancers, including solid tumors. The chemopreventive effect of flavonoids is attributed to various mechanisms, including modulation of autophagy, induction of cell cycle arrest, apoptosis, and antioxidant defense. Despite of significant anticancer activity of flavonoids, their clinical translation is limited due to their poor biopharmaceutical attributes (such as low aqueous solubility, limited permeability across the biological membranes (intestinal and blood-brain barrier), and stability issue in biological systems). A nanoparticulate system is an approach that is widely utilized to improve the biopharmaceutical performance and therapeutic efficacy of phytopharmaceuticals. The present review discusses the significant anticancer potential of promising flavonoids in different cancers and the utilization of nanoparticulate systems to improve their nanoantioxidant activity further to enhance the anticancer activity of loaded promising flavonoids. Although, various plant-derived secondary metabolites including flavonoids have been recommended for treating cancer, further vigilant research is warranted to prove their translational values.
Collapse
Affiliation(s)
- Mukta Gupta
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, Punjab, India
| | - Javed Ahmad
- Department of Pharmaceutics, College of Pharmacy, Najran University, Najran, Saudi Arabia
| | - Javed Ahamad
- Department of Pharmacognosy, Faculty of Pharmacy, Tishk International University, Erbil, Iraq
| | - Snehashis Kundu
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Guwahati, Assam, India
| | - Archit Goel
- All India Institute of Medical Sciences (AIIMS), Bathinda, Punjab, India
| | - Awanish Mishra
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Guwahati, Assam, India
| |
Collapse
|
10
|
Kumar S, Swamy N, Tuli HS, Rani S, Garg A, Mishra D, Abdulabbas HS, Sandhu SS. Myricetin: a potential plant-derived anticancer bioactive compound-an updated overview. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2023; 396:2179-2196. [PMID: 37083713 DOI: 10.1007/s00210-023-02479-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/17/2023] [Accepted: 03/28/2023] [Indexed: 04/22/2023]
Abstract
The globe is currently confronting a global fight against the deadliest cancer sickness. Chemotherapy, hormonal therapy, surgery, and radiation therapy are among cancer treatment options. Still, these treatments can induce patient side effects, including recurrence, multidrug resistance, fever, and weakness. As a result, the scientific community is always working on natural phytochemical substances. Numerous phytochemical compounds, including taxol analogues, vinca alkaloids such as vincristine and vinblastine, and podophyllotoxin analogues, are currently undergoing testing and have shown promising results against a number of the deadliest diseases, as well as considerable advantages due to their safety and low cost. According to research, secondary plant metabolites such as myricetin, a flavonoid in berries, herbs, and walnuts, have emerged as valuable bio-agents for cancer prevention. Myricetin and its derivatives have antiinflammatory, anticancer, apoptosis-inducing, and anticarcinogenic properties and can prevent cancer cell proliferation. Multiple studies have found that myricetin has anticancer characteristics in various malignancies, including colon, breast, prostate, bladder, and pancreatic cancers. Current knowledge of the anticancer effects of myricetin reveals its promise as a potentially bioactive chemical produced from plants for the prevention and treatment of cancer. This review aimed to study the numerous bioactivities, mode of action, and modification of several cellular processes that myricetin possesses to impede the spread of cancer cells. This review also addresses the challenges and future prospects of using myricetin as a anticancer drug.
Collapse
Affiliation(s)
- Suneel Kumar
- Department of Botany, Government Girls College Khargone, 451001, Khargone, Madhya Pradesh, India
| | - Nitin Swamy
- Fungal Biotechnology and Invertebrate Pathology Laboratory, Department of Biological Sciences, Rani Durgavati University, Jabalpur, 482001, Madhya Pradesh, India
| | - Hardeep Singh Tuli
- Department of Biotechnology, Maharishi Markandeshwar (Deemed to Be University), Mullana, Ambala, 133207, Haryana, India
| | - Seema Rani
- Department of Chemistry, Government M. H. College of Home Science & Science for Women, Autonomous, Jabalpur, 482002, Madhya Pradesh, India
| | - Abhijeet Garg
- Fungal Biotechnology and Invertebrate Pathology Laboratory, Department of Biological Sciences, Rani Durgavati University, Jabalpur, 482001, Madhya Pradesh, India
| | - Deepa Mishra
- Department of Biotechnology, Mata Gujri Mahila Mahavidyalaya Jabalpur, 482001, Jabalpur, Madhya Pradesh, India
| | - Hadi Sajid Abdulabbas
- Continuous Education Department, Faculty of Dentistry, University of Al-Ameed, Karbala, 56001, Iraq
| | - Sardul Singh Sandhu
- Bio-Design Innovation Centre, Rani Durgavati University, Jabalpur, 482001, Madhya Pradesh, India.
| |
Collapse
|
11
|
Al-Ghamdi MA, Alsulami RR, Bakkar A, Kumosani TA, Barrbour EK, Abulnaja KO, Huwait E, Moselhy SS. Khalas date flavonoids inhibited cell viability, induced apoptosis and expression of the pro-autophagy LC3-B gene in human hepatocellular carcinoma cells (HepG2). Nat Prod Res 2023; 37:3109-3113. [PMID: 36346382 DOI: 10.1080/14786419.2022.2140803] [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: 09/13/2022] [Revised: 10/18/2022] [Accepted: 10/20/2022] [Indexed: 11/09/2022]
Abstract
Autophagy is a protective mechanism important in human diseases as cancer. We evaluated the impact of khalas date extract (KDE) (20-60 mg/mL) on cell viability, morphological changes, DNA fragmentation and gene expression of LC3B-II associated with autophagosome on HepG2 cell line. The GC/MS identification of KDE showed its high content of flavonoids including quercetin, myricetin, kaempferol and catechol. KDE reduced cell viability of HepG2 with IC50 (31.52 mg/mL). Cells treated with KDE showed two band of DNA fragments at (30 and 40 mg) indicating that KDE induced DNA damage and apoptosis in HepG2. The analysis RT-PCR data showed a 0.2-fold increase in the expression of LC3-B in the cells treated with KDE versus control. We concluded that, KDE flavonoids such as quercetin, myricetin kaempferol exhibited anticancer properties manifested by inhibition of HepG2 cell viability and induction of apoptosis and upregulation of the pro-autophagy LC3-B gene.
Collapse
Affiliation(s)
- Maryam Abdu Al-Ghamdi
- Department of Biochemistry, Faculty of Science, King Abdulaziz University. Jeddah, Saudi Arabia
| | - Rawyah Radi Alsulami
- Department of Biochemistry, Faculty of Science, King Abdulaziz University. Jeddah, Saudi Arabia
| | - Ashraf Bakkar
- Modern Sciences and Arts University (MSA), Giza, Egypt
| | - Taha Abullah Kumosani
- Department of Biochemistry, Faculty of Science, King Abdulaziz University. Jeddah, Saudi Arabia
- Production of natural products for industrial purposes Research Group, King Abdulaziz University, Saudi Arabia
- Experimental Biochemistry Unit, King Fahd Medical Research Centre, King Abdulaziz University, Saudi Arabia
| | - Elie Kamil Barrbour
- Department of Biochemistry, Faculty of Science, King Abdulaziz University. Jeddah, Saudi Arabia
- Production of natural products for industrial purposes Research Group, King Abdulaziz University, Saudi Arabia
- Experimental Biochemistry Unit, King Fahd Medical Research Centre, King Abdulaziz University, Saudi Arabia
- Director of R and D Department, Opticon Hygiene Consulting, Oechsli, Zurich, Switzerland
| | - Khalid Omar Abulnaja
- Department of Biochemistry, Faculty of Science, King Abdulaziz University. Jeddah, Saudi Arabia
- Experimental Biochemistry Unit, King Fahd Medical Research Centre, King Abdulaziz University, Saudi Arabia
- Bioactive natural products Research Group, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Etimad Huwait
- Department of Biochemistry, Faculty of Science, King Abdulaziz University. Jeddah, Saudi Arabia
- Production of natural products for industrial purposes Research Group, King Abdulaziz University, Saudi Arabia
- Experimental Biochemistry Unit, King Fahd Medical Research Centre, King Abdulaziz University, Saudi Arabia
| | - Said Salama Moselhy
- Department of Biochemistry, Faculty of Science, Ain Shams University, Cairo, Egypt
| |
Collapse
|
12
|
Cai F, Li B, Li J, Ding Y, Xu D, Huang F. Myricetin is effective and selective in inhibiting imatinib-resistant chronic myeloid leukemia stem and differentiated cells through targeting eIF4E. Anticancer Drugs 2023; 34:620-626. [PMID: 36730418 DOI: 10.1097/cad.0000000000001421] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Although imatinib has revolutionized the treatment of chronic myeloid leukemia (CML), s develop resistance to imatinib when progress to blast phase and relapse. Myricetin, a flavonoid compound found in natural plants, has multiple biological functions. In this study, we show that myricetin demonstrated potent efficacy in imatinib-resistant CML CD34 + stem/progenitor cells with less toxicity in normal bone marrow. Myricetin is also active against imatinib-resistant CML bulk cells. The in vitro observations on the therapeutic effects of myricetin were translatable to in vivo imatinib-resistant CML xenograft mouse models. Mechanism studies showed that myricetin decreased the phosphorylation of eIF4E and Ak strain transforming, and the protein level of c-Myc and Cyclin D1. Rescue studies using eIF4E (S209D) and (S209A) confirmed that eIF4E phosphorylation inhibition was the mechanism of myricetin's action in CML. Our results suggest that myricetin may be a potential lead for drug development to overcome imatinib resistance in CML.
Collapse
Affiliation(s)
- Fangfang Cai
- Department of Rehabilitation Medicine, Hubei Provincial Hospital of Traditional Chinese Medicine, Affiliated Hospital of Hubei University of Traditional Chinese Medicine, Wuhan, Hubei, China
| | | | | | | | | | | |
Collapse
|
13
|
Shi H, Zou Y, Wang X, Wang G, Gao Y, Yi F, Xu J, Yin Y, Li D, Li M. Activating the Hippo pathway by nevadensin overcomes Yap-drived resistance to sorafenib in hepatocellular carcinoma. Discov Oncol 2023; 14:83. [PMID: 37243813 DOI: 10.1007/s12672-023-00699-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Accepted: 05/22/2023] [Indexed: 05/29/2023] Open
Abstract
BACKGROUND Hepatocellular carcinoma (HCC) is a highly malignant type of tumor that is insensitive to cytotoxic chemotherapy and often develops drug resistance. Nevadensin, a bioflavonoid, exhibits anti-cancer properties in some cancers. However, the precise underlying mechanism of nevadensin against liver cancer are poorly understood. We aim to evaluate the efficacy as well as the molecular mechanism of nevadensin in the treatment of liver cancer. METHODS Effects of nevadensin on HCC cell proliferation and apoptosis were detected using EdU labeling and flow cytometry assays. The molecular mechanism of nevadensin on HCC was determined using RNAseq. The effects of nevadensin on hippo-Yap signaling were verified using western blot and RT-PCR. RESULTS In this study, we show that nevadensin significantly inhibits growth of HCC cells via inducing cell cycle arrest and apoptosis. RNAseq analysis showed that nevadensin regulates multiple functional signaling pathways associated with cancer including Hippo signaling. Western Blot analysis revealed that nevadensin notably induces activation of the MST1/2- LATS1/2 kinase in HCC cells, further resulting in the primary effector molecule YAP phosphorylation and subsequent degradation. These results indicated that nevadensin might exert its anti-HCC activity through the Hippo-ON mechanism. Moreover, nevadensin could increase the sensitivity of HCC cells to sorafenib by down-regulating YAP and its downstream targets. CONCLUSIONS The present study indicates that nevadensin could be a potential effective approach to treating HCC, and overcoming sorafeni resistance via inducing activation of Hippo signaling.
Collapse
Affiliation(s)
- Hewen Shi
- Featured Laboratory for Biosynthesis and Target Discovery of Active Components of Traditional Chinese Medicine, School of Integrated Traditional Chinese and Western Medicine, Binzhou Medical University, Yantai, Shandong, People's Republic of China
| | - Ying Zou
- Featured Laboratory for Biosynthesis and Target Discovery of Active Components of Traditional Chinese Medicine, School of Integrated Traditional Chinese and Western Medicine, Binzhou Medical University, Yantai, Shandong, People's Republic of China
| | - Xiaoxue Wang
- Featured Laboratory for Biosynthesis and Target Discovery of Active Components of Traditional Chinese Medicine, School of Integrated Traditional Chinese and Western Medicine, Binzhou Medical University, Yantai, Shandong, People's Republic of China
| | - Guoli Wang
- Featured Laboratory for Biosynthesis and Target Discovery of Active Components of Traditional Chinese Medicine, School of Integrated Traditional Chinese and Western Medicine, Binzhou Medical University, Yantai, Shandong, People's Republic of China
| | - Yijia Gao
- School of Basic Medical Sciences, Binzhou Medical University, Yantai, Shandong, People's Republic of China
| | - Fan Yi
- School of Basic Medical Sciences, Binzhou Medical University, Yantai, Shandong, People's Republic of China
| | - Junqing Xu
- Department of Hematology, Qingdao University Medical College, Affiliated Yantai Yuhuangding Hoepital, Yantai, Shandong, People's Republic of China
| | - Yancun Yin
- School of Basic Medical Sciences, Binzhou Medical University, Yantai, Shandong, People's Republic of China.
| | - Defang Li
- Featured Laboratory for Biosynthesis and Target Discovery of Active Components of Traditional Chinese Medicine, School of Integrated Traditional Chinese and Western Medicine, Binzhou Medical University, Yantai, Shandong, People's Republic of China.
| | - Minjing Li
- Featured Laboratory for Biosynthesis and Target Discovery of Active Components of Traditional Chinese Medicine, School of Integrated Traditional Chinese and Western Medicine, Binzhou Medical University, Yantai, Shandong, People's Republic of China.
| |
Collapse
|
14
|
Identification of MAP Kinase Kinase 3 as a protein target of myricetin in non-small cell lung cancer cells. Biomed Pharmacother 2023; 161:114460. [PMID: 36870282 DOI: 10.1016/j.biopha.2023.114460] [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: 12/04/2022] [Revised: 02/21/2023] [Accepted: 02/26/2023] [Indexed: 03/06/2023] Open
Abstract
Myricetin is a typical flavonol with various pharmacological effects which shows favorable biological activities in cancer. However, the underlying mechanisms and potential targets of myricetin in NSCLC (non-small cell lung cancer) cells remain unclear. First, we demonstrated that myricetin not only inhibited the proliferation, migration and invasion, but also induced apoptosis in A549 and H1299 cells in a dose-dependent manner. Then, we confirmed myricetin may play an anti-NSCLC effect through modulating MAPK-related functions and signaling pathway by Network pharmacology. Furthermore, MKK3 (MAP Kinase Kinase 3) was identified and confirmed as a potential target of myricetin by biolayer interferometry (BLI) and molecular docking, revealing that myricetin directly bound to MKK3. Moreover, three mutations (D208, L240, and Y245) of key amino acids predicted by molecular docking obviously decreased the affinity between myricetin and MKK3. Finally, enzyme activity assay was utilized to determine the effect of myricetin on MKK3 activity in vitro, and the result showed that myricetin attenuated MKK3 activity. Subsequently, myricetin decreased the phosphorylation of p38 MAPK. Furthermore, knockdown of MKK3 reduced the susceptibility of A549 and H1299 cells to myricetin. These results suggested that myricetin inhibited the growth of NSCLC cells via targeting MKK3 and influencing the downstream p38 MAPK signaling pathway. The findings revealed that MKK3 is a potential target of myricetin in the NSCLC and myricetin is considered to be a small-molecular inhibitor of MKK3, which can improve comprehension of the molecular mechanisms of myricetin pharmacological effects in cancer and further development of MKK3 inhibitors.
Collapse
|
15
|
Duan N, Hu X, Zhou R, Li Y, Wu W, Liu N. A Review on Dietary Flavonoids as Modulators of the Tumor Microenvironment. Mol Nutr Food Res 2023; 67:e2200435. [PMID: 36698331 DOI: 10.1002/mnfr.202200435] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
The tumor microenvironment (TME) is the local environment where malignant cells strive and survive, composed of cancer cells and their surroundings, regulating essential tumor survival, and promotion functions. Dietary flavonoids are abundantly present in common vegetables and fruits and exhibit good anti-cancer activities, which significantly inhibit tumorigenesis by targeting TME constituents and their interaction with cancer cells. This review aims to synthesize information concerning the modulation of TME by dietary flavonoids, as well as to provide insights into the molecular basis of its potential anti-tumor activities, with an emphasis on its ability to control intracellular signaling cascades that regulate the TME processes, involving cell proliferation, invasion and migration, continuous angiogenesis, and immune inflammation. This study will provide a theoretical basis for the development of the leading compound targeting TME for anti-cancer therapies from these dietary flavonoids.
Collapse
Affiliation(s)
- Namin Duan
- College of Food Science and Technology, Shanghai Ocean University, Shanghai, 201306, China
| | - Xiaohui Hu
- College of Food Science and Technology, Shanghai Ocean University, Shanghai, 201306, China
| | - Rui Zhou
- College of Food Science and Technology, Shanghai Ocean University, Shanghai, 201306, China
| | - Yuru Li
- College of Food Science and Technology, Shanghai Ocean University, Shanghai, 201306, China
| | - Wenhui Wu
- College of Food Science and Technology, Shanghai Ocean University, Shanghai, 201306, China
| | - Ning Liu
- College of Food Science and Technology, Shanghai Ocean University, Shanghai, 201306, China.,National R&D Branch Center for Freshwater Aquatic Products Processing Technology, Shanghai, 201306, China.,National Experimental Teaching Demonstration Center for Food Science and Engineering, Shanghai Ocean University, Shanghai, 201306, China.,Marine Biomedical Science and Technology Innovation Platform of Lin-gang Special Area, Shanghai, 201306, China
| |
Collapse
|
16
|
Crosstalk between xanthine oxidase (XO) inhibiting and cancer chemotherapeutic properties of comestible flavonoids- a comprehensive update. J Nutr Biochem 2022; 110:109147. [PMID: 36049673 DOI: 10.1016/j.jnutbio.2022.109147] [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: 06/03/2021] [Revised: 12/17/2021] [Accepted: 08/10/2022] [Indexed: 01/13/2023]
Abstract
Gout is an inflammatory disease caused by metabolic disorder or genetic inheritance. People throughout the world are strongly dependent on ethnomedicine for the treatment of gout and some receive satisfactory curative treatment. The natural remedies as well as established drugs derived from natural sources or synthetically made exert their action by mechanisms that are closely associated with anticancer treatment mechanisms regarding inhibition of xanthine oxidase, feedback inhibition of de novo purine synthesis, depolymerization and disappearance of microtubule, inhibition of NF-ĸB activation, induction of TRAIL, promotion of apoptosis, and caspase activation and proteasome inhibition. Some anti-gout and anticancer novel compounds interact with same receptors for their action, e.g., colchicine and colchicine analogues. Dietary flavonoids, i.e., chrysin, kaempferol, quercetin, fisetin, pelargonidin, apigenin, luteolin, myricetin, isorhamnetin, phloretinetc etc. have comparable IC50 values with established anti-gout drug and effective against both cancer and gout. Moreover, a noticeable number of newer anticancer compounds have already been isolated from plants that have been using by local traditional healers and herbal practitioners to treat gout. Therefore, the anti-gout plants might have greater potentiality to become selective candidates for screening of newer anticancer leads.
Collapse
|
17
|
Cerquido AS, Vojtek M, Ribeiro-Oliveira R, Viegas O, Sousa JB, Ferreira IMPLVO, Diniz C. Unravelling Potential Health-Beneficial Properties of Corema album Phenolic Compounds: A Systematic Review. Pharmaceuticals (Basel) 2022; 15:ph15101231. [PMID: 36297345 PMCID: PMC9610266 DOI: 10.3390/ph15101231] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2022] [Revised: 09/26/2022] [Accepted: 09/27/2022] [Indexed: 11/17/2022] Open
Abstract
Corema (C.) album belongs to the family Ericaceae and can be found in the Iberian Peninsula, especially on the coastal areas facing the Atlantic coast. C. album berries have been used for centuries in traditional medicine. Recent studies have revealed that not only the berries but also the leaves have relevant antioxidant, antiproliferative, and anti-inflammatory properties, bringing this plant to the forefront of discussion. A systematic review of the literature was carried out to summarize the phenolic compounds and bioactive properties identified in C. album berries and leaves and to search for research gaps on this topic. The search was conducted in three electronic databases (PubMed, SCOPUS, and Web of Science) using PRISMA methodology. The inclusion criteria were the chemical compositions of the berries, leaves, or their extracts and their bioactive properties. The exclusion criteria were agronomic and archaeological research. The number of studies concerning phenolic compounds' composition and the bioactive properties of C. album berries and leaves is still limited (11 articles). However, the variety of polyphenolic compounds identified make it possible to infer new insights into their putative mechanism of action towards the suppression of NF-kB transcription factor activation, the modulation of inflammatory mediators/enzymes, the induction of apoptosis, the modulation of mitogen activated protein kinase, cell cycle arrest, and the reduction of oxidative stress. These factors can be of major relevance concerning the future use of C. album as nutraceuticals, food supplements, or medicines. Nevertheless, more scientific evidence concerning C. album's bioactivity is required.
Collapse
Affiliation(s)
- Ana Sofia Cerquido
- LAQV/REQUIMTE, Laboratory of Pharmacology, Department of Drug Sciences, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal
| | - Martin Vojtek
- LAQV/REQUIMTE, Laboratory of Pharmacology, Department of Drug Sciences, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal
| | - Rita Ribeiro-Oliveira
- LAQV/REQUIMTE, Laboratory of Pharmacology, Department of Drug Sciences, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal
| | - Olga Viegas
- LAQV/REQUIMTE, Laboratory of Bromatology and Hydrology, Department of Chemical Sciences, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal
- Faculty of Nutrition and Food Sciences, University of Porto, 4150-180 Porto, Portugal
| | - Joana Beatriz Sousa
- LAQV/REQUIMTE, Laboratory of Pharmacology, Department of Drug Sciences, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal
- Correspondence: (J.B.S.); (I.M.P.L.V.O.F.); (C.D.)
| | - Isabel M. P. L. V. O. Ferreira
- LAQV/REQUIMTE, Laboratory of Bromatology and Hydrology, Department of Chemical Sciences, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal
- Correspondence: (J.B.S.); (I.M.P.L.V.O.F.); (C.D.)
| | - Carmen Diniz
- LAQV/REQUIMTE, Laboratory of Pharmacology, Department of Drug Sciences, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal
- Correspondence: (J.B.S.); (I.M.P.L.V.O.F.); (C.D.)
| |
Collapse
|
18
|
Jiang C, Xie L, Wang Y, Liang J, Li H, Luo L, Li T, Liang Z, Tang L, Ning D, Ya Y, Yan F. Highly sensitive electrochemical detection of myricetin in food samples based on the enhancement effect of Al-MOFs. ANALYTICAL METHODS : ADVANCING METHODS AND APPLICATIONS 2022; 14:3521-3528. [PMID: 36018228 DOI: 10.1039/d2ay00957a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Microporous aluminum-based metal-organic frameworks (CAU-1) are used to develop a simple and sensitive electrochemical sensor for myricetin (MYR) based on a modified carbon paste electrode (CPE) for the first time. The morphologies and electrochemical properties of the as-synthesized CAU-1 are studied utilizing various analytical methods including scanning electron microscopy, transmission electron microscopy, X-ray diffraction, Fourier transform infrared spectroscopy, N2 adsorption-desorption, and electrochemical impedance spectroscopy. In terms of electrochemical oxidation of MYR, CAU-1/CPE with its large number of active micropores and rapid electron transfer demonstrates superior performance compared to the bare CPE. Under optimized conditions, the calibration curve for MYR exhibits a linear range of 1.0-10 μg L-1 and 10-1000 μg L-1 with a detection limit of 0.50 μg L-1. The developed CAU-1/CPE exhibits superior analytical characteristics, compared to previously reported electrochemical sensors for MYR detection. Furthermore, CAU-1/CPE is employed to determine MYR in Myrica bark samples, and the results are consistent with those obtained by high-performance liquid chromatography, demonstrating the excellent potential of CAU-1/CPE for the rapid analysis of MYR in complicated real samples.
Collapse
Affiliation(s)
- Cuiwen Jiang
- Institute for Agricultural Product Quality Safety and Testing Technology, Guangxi Academy of Agricultural Sciences, Nanning 530007, China.
| | - Liping Xie
- Institute for Agricultural Product Quality Safety and Testing Technology, Guangxi Academy of Agricultural Sciences, Nanning 530007, China.
| | - Yanli Wang
- Institute for Agricultural Product Quality Safety and Testing Technology, Guangxi Academy of Agricultural Sciences, Nanning 530007, China.
| | - Jing Liang
- Institute for Agricultural Product Quality Safety and Testing Technology, Guangxi Academy of Agricultural Sciences, Nanning 530007, China.
| | - Huiling Li
- Institute for Agricultural Product Quality Safety and Testing Technology, Guangxi Academy of Agricultural Sciences, Nanning 530007, China.
| | - Lihong Luo
- Institute for Agricultural Product Quality Safety and Testing Technology, Guangxi Academy of Agricultural Sciences, Nanning 530007, China.
| | - Tao Li
- Institute for Agricultural Product Quality Safety and Testing Technology, Guangxi Academy of Agricultural Sciences, Nanning 530007, China.
| | - Zhongdan Liang
- Institute for Agricultural Product Quality Safety and Testing Technology, Guangxi Academy of Agricultural Sciences, Nanning 530007, China.
| | - Li Tang
- Institute for Agricultural Product Quality Safety and Testing Technology, Guangxi Academy of Agricultural Sciences, Nanning 530007, China.
| | - Dejiao Ning
- Institute for Agricultural Product Quality Safety and Testing Technology, Guangxi Academy of Agricultural Sciences, Nanning 530007, China.
| | - Yu Ya
- Institute for Agricultural Product Quality Safety and Testing Technology, Guangxi Academy of Agricultural Sciences, Nanning 530007, China.
| | - Feiyan Yan
- Institute for Agricultural Product Quality Safety and Testing Technology, Guangxi Academy of Agricultural Sciences, Nanning 530007, China.
| |
Collapse
|
19
|
Hu H, Hu Z, Zhang Y, Wan H, Yin Z, Li L, Liang X, Zhao X, Yin L, Ye G, Zou YF, Tang H, Jia R, Chen Y, Zhou H, Song X. Myricetin inhibits pseudorabies virus infection through direct inactivation and activating host antiviral defense. Front Microbiol 2022; 13:985108. [PMID: 36187970 PMCID: PMC9520584 DOI: 10.3389/fmicb.2022.985108] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2022] [Accepted: 08/22/2022] [Indexed: 11/21/2022] Open
Abstract
Myricetin, a polyhydroxyflavone compound, is one of the main ingredients of various human foods and therefore also known as dietary flavonoids. Due to the continuous emergence of resistant strains of herpesviruses, novel control measures are required. In the present study, myricetin exhibited potent antiviral activity against pseudorabies virus (PRV), a model organism of herpesvirus. The suppression rate could reach up to 96.4% at a concentration of 500 μM in cells, and the 50% inhibitory concentration (IC50) was 42.69 μM. Moreover, the inhibitory activity was not attenuated by the increased amount of infective dose, and a significant reduction of intracellular PRV virions was observed by indirect immunofluorescence. A mode of action study indicated that myricetin could directly inactivate the virus in vitro, leading to inhibition of viral adsorption, penetration and replication in cells. In addition to direct killing effect, myricetin could also activate host antiviral defense through regulation of apoptosis-related gene expressions (Bcl-2, Bcl-xl, Bax), NF-κB and MAPK signaling pathways and cytokine gene expressions (IL-1α, IL-1β, IL-6, c-Jun, STAT1, c-Fos, and c-Myc). In PRV-infected mouse model, myricetin could enhance the survival rate by 40% at 5 days post infection, and viral loads in kidney, liver, lung, spleen, and brain were significantly decreased. The pathological changes caused by PRV infection were improved by myricetin treatment. The gene expressions of inflammatory factors (MCP-1, G-CSF, IL-1α, IL-1β, and IL-6) and apoptotic factors (Bcl-xl, Bcl-2, and Bax) were regulated by myricetin in PRV-infected mice. The present findings suggest that myricetin can effectively inhibit PRV infection and become a candidate for development of new anti-herpesvirus drugs.
Collapse
Affiliation(s)
- Huaiyue Hu
- Natural Medicine Research Center, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Zhiqiang Hu
- Shandong New Hope Liuhe Agriculture and Animal Husbandry Technology Co., Ltd., Dezhou, China
| | - Yingying Zhang
- College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, China
| | - Hongping Wan
- Natural Medicine Research Center, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Zhongqiong Yin
- Natural Medicine Research Center, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Lixia Li
- Natural Medicine Research Center, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Xiaoxia Liang
- Natural Medicine Research Center, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Xinghong Zhao
- Natural Medicine Research Center, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Lizi Yin
- Natural Medicine Research Center, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Gang Ye
- Natural Medicine Research Center, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Yuan-Feng Zou
- Natural Medicine Research Center, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Huaqiao Tang
- Natural Medicine Research Center, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Renyong Jia
- Natural Medicine Research Center, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Yaqin Chen
- Natural Medicine Research Center, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Hao Zhou
- College of Medical Technology, Chengdu University of Traditional Chinese Medicine, Chengdu, China
- Department of Microbiology, NYU Grossman School of Medicine, New York, NY, United States
| | - Xu Song
- Natural Medicine Research Center, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| |
Collapse
|
20
|
Javed Z, Khan K, Herrera-Bravo J, Naeem S, Iqbal MJ, Raza Q, Sadia H, Raza S, Bhinder M, Calina D, Sharifi-Rad J, Cho WC. Myricetin: targeting signaling networks in cancer and its implication in chemotherapy. Cancer Cell Int 2022; 22:239. [PMID: 35902860 PMCID: PMC9336020 DOI: 10.1186/s12935-022-02663-2] [Citation(s) in RCA: 51] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2022] [Accepted: 07/22/2022] [Indexed: 02/06/2023] Open
Abstract
The gaps between the complex nature of cancer and therapeutics have been narrowed down due to extensive research in molecular oncology. Despite gathering massive insight into the mysteries of tumor heterogeneity and the molecular framework of tumor cells, therapy resistance and adverse side effects of current therapeutic remain the major challenge. This has shifted the attention towards therapeutics with less toxicity and high efficacy. Myricetin a natural flavonoid has been under the spotlight for its anti-cancer, anti-oxidant, and anti-inflammatory properties. The cutting-edge molecular techniques have shed light on the interplay between myricetin and dysregulated signaling cascades in cancer progression, invasion, and metastasis. However, there are limited data available regarding the nano-delivery platforms composed of myricetin in cancer. In this review, we have provided a comprehensive detail of myricetin-mediated regulation of different cellular pathways, its implications in cancer prevention, preclinical and clinical trials, and its current available nano-formulations for the treatment of various cancers.
Collapse
Affiliation(s)
- Zeeshan Javed
- Office of Research Innovation and Commercialization, Lahore Garrison University, Lahore, Pakistan
| | - Khushbukhat Khan
- Department of Healthcare Biotechnology, Atta-ur-Rahman School of Applied Biosciences, National University of Sciences and Technology, Sector H-12, Islamabad, 44000 Pakistan
| | - Jesús Herrera-Bravo
- Departamento de Ciencias Básicas, Facultad de Ciencias, Universidad Santo Tomas, Santiago, Chile
- Center of Molecular Biology and Pharmacogenetics, Scientific and Technological Bioresource Nucleus, Universidad de La Frontera, 4811230 Temuco, Chile
| | - Sajid Naeem
- Gansu Key Laboratory of Biomonitoring and Bioremediation for Environmental Pollution, School of Life Sciences, Lanzhou University, Lanzhou, 730000 China
| | - Muhammad Javed Iqbal
- Department of Biotechnology, Faculty of Sciences, University of Sialkot, Sialkot, Pakistan
| | - Qamar Raza
- Institute of Biochemistry and Biotechnology, University of Veterinary and Animal Sciences, Lahore, Punjab Pakistan
| | - Haleema Sadia
- Department of Biotechnology, Balochistan University of Information Technology, Engineering and Management Sciences, Quetta, 87100 Pakistan
| | - Shahid Raza
- Office of Research Innovation and Commercialization, Lahore Garrison University, Lahore, Pakistan
| | - Munir Bhinder
- Department of Human Genetics & Molecular Biology, University of Health Sciences, Lahore, 54600 Pakistan
| | - Daniela Calina
- Department of Clinical Pharmacy, University of Medicine and Pharmacy of Craiova, 200349 Craiova, Romania
| | | | - William C. Cho
- Department of Clinical Oncology, Queen Elizabeth Hospital, Kowloon, Hong Kong China
| |
Collapse
|
21
|
Li Y, Zhang J, Zhou H, Du Z. Anticancer effects of natural phytochemicals in anaplastic thyroid cancer (Review). Oncol Rep 2022; 48:156. [PMID: 35856443 PMCID: PMC9471558 DOI: 10.3892/or.2022.8368] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Accepted: 06/23/2022] [Indexed: 11/13/2022] Open
Abstract
Anaplastic thyroid cancer (ATC) is an aggressive and lethal malignancy having a dismal prognosis. Phytochemicals are bioactive components obtained from plants that have been proven useful to treat numerous diseases. Phytochemicals are also an important source of novel anticancer drugs and an important area of research due to the numerous available candidates that can potentially treat cancers. This review discusses naturally occurring phytochemicals and their derivatives that show promising anticancer effects in anaplastic thyroid cancer. Anticancer effects include cell growth inhibition, induction of apoptosis, promoting cell cycle arrest, suppressing angiogenesis, modulating autophagy, and increasing the production of reactive oxygen species. Phytochemicals are not only prospective candidates in the therapy of anaplastic thyroid cancer but also exhibit potential as adjuvants to improve the anticancer effects of other drugs. Although some phytochemicals have excellent anticancer properties, drug resistance observed during the use of resveratrol and artemisinin in different anaplastic thyroid cancer cell lines is still a problem. Anaplastic thyroid cancer cells have several biological, clinical, and drug-resistance features that differ from differentiated thyroid cancer cells. Phytochemicals such as resveratrol and quercetin exhibit different biological effects in anaplastic thyroid cancer and differentiated thyroid cancer. Tumor cells depend on increased aerobic glycolysis by mitochondrial oxidative phosphorylation to provide energy for their rapid growth, invasiveness, and drug resistance. Phytochemicals can alter signaling cascades, modulate the metabolic properties of cancer cells, and influence the mitochondrial membrane potential of anaplastic thyroid cancer cells. These findings enrich our knowledge of the anticancer effects of phytochemicals and highlight alternative therapies to prevent drug resistance in anaplastic thyroid cancer.
Collapse
Affiliation(s)
- Yitian Li
- Department of Hygiene, Public Health College, Jining Medical University, Jining, Shandong 272067, P.R. China
| | - Jing Zhang
- Department of Hygiene, Public Health College, Jining Medical University, Jining, Shandong 272067, P.R. China
| | - Huihui Zhou
- Department of Hygiene, Public Health College, Jining Medical University, Jining, Shandong 272067, P.R. China
| | - Zhen Du
- Department of Hygiene, Public Health College, Jining Medical University, Jining, Shandong 272067, P.R. China
| |
Collapse
|
22
|
Yang L, Gao Y, Gong J, Wang H, Farag MA, Simal‐Gandara J, Zhao Y, Nie S, Xiao J. Myricetin ameliorated prediabetes via immunomodulation and gut microbiota interaction. FOOD FRONTIERS 2022. [DOI: 10.1002/fft2.152] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Affiliation(s)
- Li Yang
- Institute of Chinese Medical Sciences State Key Laboratory of Quality Research in Chinese Medicine University of Macau Macau China
| | - Yongchao Gao
- Department of Clinical Pharmacology Xiangya Hospital Central South University Changsha China
| | - Jupeng Gong
- Guangdong Provincial Key Laboratory of Aquatic Product Processing and Safety College of Food Science and Technology Guangdong Ocean University Zhanjiang China
| | - Hui Wang
- State Key Laboratory of Food Science and Technology China‐Canada Joint Lab of Food Science and Technology (Nanchang) Nanchang University Nanchang China
| | - Mohamed A. Farag
- Pharmacognosy Department College of Pharmacy Cairo University Cairo Egypt
| | - Jesus Simal‐Gandara
- Nutrition and Bromatology Group Department of Analytical and Food Chemistry Faculty of Sciences Universidade de Vigo Ourense Spain
| | - Yonghua Zhao
- Institute of Chinese Medical Sciences State Key Laboratory of Quality Research in Chinese Medicine University of Macau Macau China
| | - Shaoping Nie
- State Key Laboratory of Food Science and Technology China‐Canada Joint Lab of Food Science and Technology (Nanchang) Nanchang University Nanchang China
| | - Jianbo Xiao
- Nutrition and Bromatology Group Department of Analytical and Food Chemistry Faculty of Sciences Universidade de Vigo Ourense Spain
- Institute of Food Safety and Nutrition Jinan University Guangzhou China
| |
Collapse
|
23
|
Xu B, Mo X, Chen J, Yu H, Liu Y. Myricetin Inhibits α-Synuclein Amyloid Aggregation by Delaying the Liquid-to-Solid Phase Transition. Chembiochem 2022; 23:e202200216. [PMID: 35657723 DOI: 10.1002/cbic.202200216] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2022] [Revised: 06/02/2022] [Indexed: 11/12/2022]
Abstract
The aggregation of α-synuclein (α-Syn) is a critical pathological hallmark of Parkinson's disease (PD). Prevention of α-Syn aggregation has become a key strategy for treating PD. Recent studies have suggested that α-Syn undergoes liquid-liquid phase separation (LLPS) to facilitate nucleation and amyloid formation. Here, we examined the modulation of α-Syn aggregation by myricetin, a polyhydroxyflavonol compound, under the conditions of LLPS. Unexpectedly, neither the initial morphology nor the phase-separated fraction of α-Syn was altered by myricetin. However, the dynamics of α-Syn condensates decreased upon myricetin binding. Further studies showed that myricetin dose-dependently inhibits amyloid aggregation in the condensates by delaying the liquid-to-solid phase transition. In addition, myricetin could disassemble the preformed α-Syn amyloid aggregates matured from the condensates. Together, our study shows that myricetin inhibits α-Syn amyloid aggregation in the condensates by retarding the liquid-to-solid phase transition and reveals that α-Syn phase transition can be targeted to inhibit amyloid aggregation.
Collapse
Affiliation(s)
- Bingkuan Xu
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University, No. 1 Wenyuan Road, Nanjing, 210046, (P. R. China)
| | - Xiaoli Mo
- Biology Department, Clark University 950 Main Street, Worcester, Massachusetts (USA) 01610
| | - Jing Chen
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University, No. 1 Wenyuan Road, Nanjing, 210046, (P. R. China)
| | - Haijia Yu
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University, No. 1 Wenyuan Road, Nanjing, 210046, (P. R. China)
| | - Yinghui Liu
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University, No. 1 Wenyuan Road, Nanjing, 210046, (P. R. China)
| |
Collapse
|
24
|
Molecular Pathways Involved in the Anti-Cancer Activity of Flavonols: A Focus on Myricetin and Kaempferol. Int J Mol Sci 2022; 23:ijms23084411. [PMID: 35457229 PMCID: PMC9026553 DOI: 10.3390/ijms23084411] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2022] [Revised: 04/04/2022] [Accepted: 04/14/2022] [Indexed: 12/22/2022] Open
Abstract
Natural compounds have always represented valuable allies in the battle against several illnesses, particularly cancer. In this field, flavonoids are known to modulate a wide panel of mechanisms involved in tumorigenesis, thus rendering them worthy candidates for both cancer prevention and treatment. In particular, it was reported that flavonoids regulate apoptosis, as well as hamper migration and proliferation, crucial events for the progression of cancer. In this review, we collect recent evidence concerning the anti-cancer properties of the flavonols myricetin and kaempferol, discussing their mechanisms of action to give a thorough overview of their noteworthy capabilities, which are comparable to those of their most famous analogue, namely quercetin. On the whole, these flavonols possess great potential, and hence further study is highly advised to allow a proper definition of their pharmaco-toxicological profile and assess their potential use in protocols of chemoprevention and adjuvant therapies.
Collapse
|
25
|
Han SH, Lee JH, Woo JS, Jung GH, Jung SH, Han EJ, Kim B, Cho SD, Nam JS, Che JH, Jung JY. Myricetin induces apoptosis and autophagy in human gastric cancer cells through inhibition of the PI3K/Akt/mTOR pathway. Heliyon 2022; 8:e09309. [PMID: 35521506 PMCID: PMC9065623 DOI: 10.1016/j.heliyon.2022.e09309] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2021] [Revised: 01/06/2022] [Accepted: 04/19/2022] [Indexed: 12/02/2022] Open
Abstract
Myricetin, a natural flavonoid present in berries, nuts, and green tea, is well-known for its anticancer properties. Even though several previous studies have reported the anticancer effects induced by myricetin, these effects have not yet been confirmed in the adenocarcinoma gastric cell line (AGS). Moreover, the exact mechanisms of myricetin-induced apoptosis and autophagy have not been clearly identified either. Therefore, in this study, we aimed to examine the role of myricetin in inducing apoptosis and autophagy in AGS gastric cancer cells. First, the survival rate of AGS gastric cancer cells was assessed using the 3-(4, 5-dimethylthiazolyl-2)-2, 5-diphenyltetrazolium bromide (MTT) cell viability assay. Thereafter, the rate of apoptosis was analyzed using4′,6-diamidino-2-phenylindole (DAPI) staining as well as annexin V and propidium iodide (PI) staining, and the expression of the proteins associated with apoptosis, PI3K/Akt/mTOR pathway, and autophagy was examined by western blotting. We observed that myricetin reduced the survival rate of AGS gastric cancer cells by inhibiting the PI3K/Akt/mTOR pathway, thereby inducing apoptosis and autophagy. Similar results were also obtained in vivo, and tumor growth was inhibited. Therefore, in the AGS gastric cancer cells, myricetin seems to inhibit the PI3K/Akt/mTOR pathway, which in turn leads to apoptosis in vitroand in vivo, cell-protective autophagy, as well as inhibition of cancer cell proliferation. These results indicate the potential of myricetin as a natural anticancer agent.
Collapse
Affiliation(s)
- So-Hee Han
- Department of Companion and Laboratory Animal Science, Kongju National University, Yesan 32439, Republic of Korea
| | - Jae-Han Lee
- Department of Companion and Laboratory Animal Science, Kongju National University, Yesan 32439, Republic of Korea
| | - Joong-Seok Woo
- Department of Companion and Laboratory Animal Science, Kongju National University, Yesan 32439, Republic of Korea
| | - Gi-Hwan Jung
- Department of Companion and Laboratory Animal Science, Kongju National University, Yesan 32439, Republic of Korea
| | - Soo-Hyun Jung
- Department of Companion and Laboratory Animal Science, Kongju National University, Yesan 32439, Republic of Korea
| | - Eun-Ji Han
- Department of Companion and Laboratory Animal Science, Kongju National University, Yesan 32439, Republic of Korea
| | - Bumseok Kim
- College of Veterinary Medicine and Bio-safety Research Institute, Jeonbuk National University, Iksan 54596, Republic of Korea
| | - Sung Dae Cho
- Department of Oral Pathology, School of Dentistry and Dental Research Institute, Seoul National University, Seoul 03080, Republic of Korea
| | - Jeong Seok Nam
- Gwangju Institute of Science and Technology, School of Life Sciences, Gwangju 61005, Republic of Korea
| | - Jeong Hwan Che
- Biomedical Center for Animal Resource Development, Seoul National University College of Medicine, Seoul 03080, Republic of Korea
- Biomedical Research Institute, Seoul National University Hospital, Seoul 03080, Republic of Korea
| | - Ji-Youn Jung
- Department of Companion and Laboratory Animal Science, Kongju National University, Yesan 32439, Republic of Korea
- Corresponding author.
| |
Collapse
|
26
|
Wang M, Ren S, Bi Z, Zhang L, Cui M, Sun R, Bao J, Gao D, Yang B, Li X, Li M, Xiao T, Zhou H, Yang C. Myricetin reverses epithelial–endothelial transition and inhibits vasculogenic mimicry and angiogenesis of hepatocellular carcinoma by directly targeting
PAR1. Phytother Res 2022; 36:1807-1821. [DOI: 10.1002/ptr.7427] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2021] [Revised: 01/27/2022] [Accepted: 01/30/2022] [Indexed: 11/06/2022]
Affiliation(s)
- Ming Wang
- State Key Laboratory of Medicinal Biology, College of Pharmacy and Tianjin Key Laboratory of Molecular Drug Research Nankai University Tianjin People's Republic of China
| | - Shanfa Ren
- State Key Laboratory of Medicinal Biology, College of Pharmacy and Tianjin Key Laboratory of Molecular Drug Research Nankai University Tianjin People's Republic of China
- Tianjin Key Laboratory of Molecular Drug Research, Tianjin International Joint Academy of Biomedicine Tianjin People's Republic of China
| | - Zhun Bi
- State Key Laboratory of Medicinal Biology, College of Pharmacy and Tianjin Key Laboratory of Molecular Drug Research Nankai University Tianjin People's Republic of China
| | - Liang Zhang
- Department of Thoracic Surgery Tianjin First Central Hospital, Nankai University Tianjin People's Republic of China
| | - Mengqi Cui
- State Key Laboratory of Medicinal Biology, College of Pharmacy and Tianjin Key Laboratory of Molecular Drug Research Nankai University Tianjin People's Republic of China
- Tianjin Key Laboratory of Molecular Drug Research, Tianjin International Joint Academy of Biomedicine Tianjin People's Republic of China
| | - Ronghao Sun
- State Key Laboratory of Medicinal Biology, College of Pharmacy and Tianjin Key Laboratory of Molecular Drug Research Nankai University Tianjin People's Republic of China
| | - Jiali Bao
- State Key Laboratory of Medicinal Biology, College of Pharmacy and Tianjin Key Laboratory of Molecular Drug Research Nankai University Tianjin People's Republic of China
- Tianjin Key Laboratory of Molecular Drug Research, Tianjin International Joint Academy of Biomedicine Tianjin People's Republic of China
| | - Dandi Gao
- State Key Laboratory of Medicinal Biology, College of Pharmacy and Tianjin Key Laboratory of Molecular Drug Research Nankai University Tianjin People's Republic of China
- Tianjin Key Laboratory of Molecular Drug Research, Tianjin International Joint Academy of Biomedicine Tianjin People's Republic of China
| | - Bo Yang
- Department of Thoracic Surgery Tianjin First Central Hospital, Nankai University Tianjin People's Republic of China
| | - Xiaoping Li
- Department of Thoracic Surgery Tianjin First Central Hospital, Nankai University Tianjin People's Republic of China
| | - Mingjiang Li
- Department of Thoracic Surgery Tianjin First Central Hospital, Nankai University Tianjin People's Republic of China
| | - Ting Xiao
- State Key Laboratory of Medicinal Biology, College of Pharmacy and Tianjin Key Laboratory of Molecular Drug Research Nankai University Tianjin People's Republic of China
- Tianjin Key Laboratory of Molecular Drug Research, Tianjin International Joint Academy of Biomedicine Tianjin People's Republic of China
| | - Hong‐gang Zhou
- State Key Laboratory of Medicinal Biology, College of Pharmacy and Tianjin Key Laboratory of Molecular Drug Research Nankai University Tianjin People's Republic of China
- Tianjin Key Laboratory of Molecular Drug Research, Tianjin International Joint Academy of Biomedicine Tianjin People's Republic of China
| | - Cheng Yang
- State Key Laboratory of Medicinal Biology, College of Pharmacy and Tianjin Key Laboratory of Molecular Drug Research Nankai University Tianjin People's Republic of China
- Tianjin Key Laboratory of Molecular Drug Research, Tianjin International Joint Academy of Biomedicine Tianjin People's Republic of China
| |
Collapse
|
27
|
Han SH, Lee JH, Woo JS, Jung GH, Jung SH, Han EJ, Park YS, Kim BS, Kim SK, Park BK, Choi C, Jung JY. Myricetin induces apoptosis through the MAPK pathway and regulates JNK‑mediated autophagy in SK‑BR‑3 cells. Int J Mol Med 2022; 49:54. [PMID: 35234274 PMCID: PMC8904074 DOI: 10.3892/ijmm.2022.5110] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Accepted: 02/14/2022] [Indexed: 11/05/2022] Open
Abstract
Myricetin, a flavonoid found in fruits and vegetables, is known to have antioxidant and anticancer effects. However, the anticancer effects of myricetin on SK-BR-3 human breast cancer cells have not been elucidated. In the present study, the anticancer effects of myricetin were confirmed in human breast cancer SK-BR-3 cells. As the concentration of myricetin increased, the cell viability decreased. DAPI (4′,6-diamidino-2-phenylindole) and Annexin V/PI staining also revealed a significant increase in apoptotic bodies and apoptosis. Western blot analysis was performed to confirm the myricetin-induced expression of apoptosis-related proteins. The levels of cleaved PARP and Bax proteins were increased, and that of Bcl-2 was decreased. The levels of proteins in the mitogen-activated protein kinase (MAPK) pathway were examined to confirm the mechanism of myricetin-induced apoptosis, and it was found that the expression levels of phosphorylated c-Jun N-terminal kinase (p-JNK) and phosphorylated mitogen-activated protein kinases (p-p38) were increased, whereas that of phosphorylated extracellular-regulated kinase (p-ERK) was decreased. It was also demonstrated that myricetin induced autophagy by promoting autophagy-related proteins such as microtubule-associated protein 1A/1B-light chain 3 (LC 3) and beclin 1. In addition, 3-methyladenine (3-MA) was used to evaluate the association between cell viability and autophagy in cells treated with myricetin. The results showed that simultaneous treatment with 3-MA and myricetin promoted the apoptosis of breast cancer cells. Furthermore, treatment with a JNK inhibitor reduced cell viability, promoted Bax expression, and reduced the expression of p-JNK, Bcl-2, and LC 3-II/I. These results suggest that myricetin induces apoptosis via the MAPK pathway and regulates JNK-mediated autophagy in SK-BR-3 cells. In conclusion, myricetin shows potential as a natural anticancer agent in SK-BR-3 cells.
Collapse
Affiliation(s)
- So-Hee Han
- Department of Companion and Laboratory Animal Science, Kongju National University, Yesan-eup, Chungcheongnamdo 32439, Republic of Korea
| | - Jae-Han Lee
- Department of Companion and Laboratory Animal Science, Kongju National University, Yesan-eup, Chungcheongnamdo 32439, Republic of Korea
| | - Joong-Seok Woo
- Department of Companion and Laboratory Animal Science, Kongju National University, Yesan-eup, Chungcheongnamdo 32439, Republic of Korea
| | - Gi-Hwan Jung
- Department of Companion and Laboratory Animal Science, Kongju National University, Yesan-eup, Chungcheongnamdo 32439, Republic of Korea
| | - Soo-Hyun Jung
- Department of Companion and Laboratory Animal Science, Kongju National University, Yesan-eup, Chungcheongnamdo 32439, Republic of Korea
| | - Eun-Ji Han
- Department of Companion and Laboratory Animal Science, Kongju National University, Yesan-eup, Chungcheongnamdo 32439, Republic of Korea
| | - Young-Seok Park
- Department of Companion and Laboratory Animal Science, Kongju National University, Yesan-eup, Chungcheongnamdo 32439, Republic of Korea
| | - Byeong-Soo Kim
- Department of Companion and Laboratory Animal Science, Kongju National University, Yesan-eup, Chungcheongnamdo 32439, Republic of Korea
| | - Sang-Ki Kim
- Department of Companion and Laboratory Animal Science, Kongju National University, Yesan-eup, Chungcheongnamdo 32439, Republic of Korea
| | - Byung-Kwon Park
- Department of Companion and Laboratory Animal Science, Kongju National University, Yesan-eup, Chungcheongnamdo 32439, Republic of Korea
| | - Changsun Choi
- School of Food Science and Technology, Chung‑ang University, Ansung, Gyeonggi-do 17546, Republic of Korea
| | - Ji-Youn Jung
- Department of Companion and Laboratory Animal Science, Kongju National University, Yesan-eup, Chungcheongnamdo 32439, Republic of Korea
| |
Collapse
|
28
|
Chen YC, He XL, Qi L, Shi W, Yuan LW, Huang MY, Xu YL, Chen X, Gu L, Zhang LL, Lu JJ. Myricetin inhibits interferon-γ-induced PD-L1 and IDO1 expression in lung cancer cells. Biochem Pharmacol 2022; 197:114940. [PMID: 35120895 DOI: 10.1016/j.bcp.2022.114940] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2021] [Revised: 01/26/2022] [Accepted: 01/27/2022] [Indexed: 11/02/2022]
Abstract
Programmed death ligand-1 (PD-L1) and indoleamine 2, 3-dioxygenase 1 (IDO1) are immune checkpoints induced by interferon-γ (IFN-γ) in the tumor microenvironment, leading to immune escape of tumors. Myricetin (MY) is a flavonoid distributed in many edible and medicinal plants. In this study, MY was identified to inhibit IFN-γ-induced PD-L1 expression in human lung cancer cells. It also reduced the expression of IDO1 and the production of kynurenine which is the product catalyzed by IDO1, while didn't show obvious effect on the expression of major histocompatibility complex-I (MHC-I), a crucial molecule for antigen presentation. In addition, the function of T cells was evaluated using a co-culture system consist of lung cancer cells and the Jurkat-PD-1 T cell line overexpressing PD-1. MY restored the survival, proliferation, CD69 expression and interleukin-2 (IL-2) secretion of Jurkat-PD-1 T cells suppressed by IFN-γ-treated lung cancer cells. Mechanistically, IFN-γ up-regulated PD-L1 and IDO1 at the transcriptional level through the JAK-STAT-IRF1 axis, which was targeted and inhibited by MY. Together, our research revealed a new mechanism of MY mediated anti-tumor activity and highlighted the potential implications of MY in tumor immunotherapy.
Collapse
Affiliation(s)
- Yu-Chi Chen
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao, China
| | - Xin-Ling He
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao, China
| | - Lu Qi
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao, China
| | - Wei Shi
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao, China
| | - Luo-Wei Yuan
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao, China
| | - Mu-Yang Huang
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao, China
| | - Yu-Lian Xu
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao, China
| | - Xiuping Chen
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao, China
| | - Lei Gu
- Epigenetics Laboratory, Max Planck Institute for Heart and Lung Research, 61231 Bad Nauheim, Germany; Cardiopulmonary Institute (CPI), 61231 Bad Nauheim, Germany
| | - Le-Le Zhang
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao, China
| | - Jin-Jian Lu
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao, China; Department of Pharmaceutical Sciences, Faculty of Health Sciences, University of Macau, Macao, China; MoE Frontiers Science Center for Precision Oncology, University of Macau, Macao, China; Guangdong-Hong Kong-Macau Joint Lab on Chinese Medicine and Immune Disease Research, University of Macau, Macao, China.
| |
Collapse
|
29
|
Agraharam G, Girigoswami A, Girigoswami K. Myricetin: a Multifunctional Flavonol in Biomedicine. CURRENT PHARMACOLOGY REPORTS 2022; 8:48-61. [PMID: 35036292 PMCID: PMC8743163 DOI: 10.1007/s40495-021-00269-2] [Citation(s) in RCA: 45] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Accepted: 11/17/2021] [Indexed: 12/13/2022]
Abstract
PURPOSE OF REVEIW The root cause of many diseases like CVD, cancer, and aging is free radicals which exert their effect by interfering with different metabolic pathways. The sources of free radicals can be exogenous, like UV rays from sunlight, and endogenous due to different metabolic by-products.In our body, there are defense mechanisms present, such as antioxidant enzymes and antioxidant molecules to combat these free radicals, but if there is an overload of these free radicals in our body, the defense system may not be sufficient to neutralize these free radicals. In such situations, we are exposed to a chronic low dose of oxidants creating oxidative stress, which is responsible for eliciting different diseases. RECENT FINDINGS Pubmed and Google Scholar are the search engines used to sort out relevant papers on myricetin and its role in combating many diseases. Myricetin is present in many fruits and vegetables and is a known antioxidant. It can elevate the antioxidant enzyme levels; reduces the lipid peroxidation; and is known to protect against cancer. In the case of myocardial dysfunction, myricetin has been shown to suppress the inflammatory cytokines and reduced the mortality rate. Myricetin has also been found to reduce platelet aggregation and control the viral infections by interfering in the DNA replication pathways. SUMMARY In this paper, we have briefly reviewed about the different type and site of free radicals and the role of myricetin in addressing the ROS and different diseases.
Collapse
Affiliation(s)
- Gopikrishna Agraharam
- Faculty of Allied Health Sciences, Chettinad Hospital and Research Institute, Chettinad Academy of Research and Education, Kelambakkam, 603103 Tamilnadu India
| | - Agnishwar Girigoswami
- Faculty of Allied Health Sciences, Chettinad Hospital and Research Institute, Chettinad Academy of Research and Education, Kelambakkam, 603103 Tamilnadu India
| | - Koyeli Girigoswami
- Faculty of Allied Health Sciences, Chettinad Hospital and Research Institute, Chettinad Academy of Research and Education, Kelambakkam, 603103 Tamilnadu India
| |
Collapse
|
30
|
Li M, Zha G, Chen R, Chen X, Sun Q, Jiang H. Anticancer effects of myricetin derivatives in non-small cell lung cancer in vitro and in vivo. Pharmacol Res Perspect 2021; 10:e00905. [PMID: 34964301 PMCID: PMC8929361 DOI: 10.1002/prp2.905] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2021] [Accepted: 11/29/2021] [Indexed: 02/06/2023] Open
Abstract
Lung cancer is the most common cause of cancer‐related deaths. Moreover, exploring efficient tumor‐killing drugs is urgently needed. In our study, several derivative compounds of myricetin were synthesized and tested. Experiments on non‐small cell lung cancer (NSCLC) showed that S4‐2‐2 (5,7‐dimethoxy‐3‐(4‐(methyl(1‐(naphthalen‐2‐ylsulfonyl)piperidin‐4‐yl)amino)butoxy)‐2‐(3,4,5‐trimethoxyphenyl)‐4H‐chromen‐4‐one) had the strongest effect on A549 cell inhibition across all compounds. Furthermore, S4‐2‐2‐treated A549 cells were also suppressed when transplanted into immunodeficient mice. Particularly, we found that the migration and invasiveness of A549 cells became suppressed upon treatment with S4‐2‐2. Furthermore, the compound significantly induced cell apoptosis, but did not affect the cell cycle of A549 cells. Finally, we revealed that S4‐2‐2 inhibited the biological function of NSCLC cells by regulating the protein process in the endoplasmic reticulum, and then by inducing the expression of apoptosis‐related proteins. Taken together, S4‐2‐2 was shown to act as a potential molecular inhibitor of A549 cells.
Collapse
Affiliation(s)
- Mengmeng Li
- Department of Radiation Oncology, The First Affiliated Hospital of Bengbu Medical College, Bengbu, China
| | - Genlan Zha
- Department of Radiation Oncology, The First Affiliated Hospital of Bengbu Medical College, Bengbu, China
| | - Rujun Chen
- Department of Radiation Oncology, The First Affiliated Hospital of Bengbu Medical College, Bengbu, China
| | - Xin Chen
- Department of Radiation Oncology, The First Affiliated Hospital of Bengbu Medical College, Bengbu, China
| | - Qian Sun
- Department of Radiation Oncology, The First Affiliated Hospital of Bengbu Medical College, Bengbu, China
| | - Hao Jiang
- Department of Radiation Oncology, The First Affiliated Hospital of Bengbu Medical College, Bengbu, China
| |
Collapse
|
31
|
Gao L, Luo H, Wang Q, Hu G, Xiong Y. Synergistic Effect of Hydrogen Bonds and Chemical Bonds to Construct a Starch-Based Water-Absorbing/Retaining Hydrogel Composite Reinforced with Cellulose and Poly(ethylene glycol). ACS OMEGA 2021; 6:35039-35049. [PMID: 34963985 PMCID: PMC8697600 DOI: 10.1021/acsomega.1c05614] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/13/2021] [Accepted: 11/24/2021] [Indexed: 06/14/2023]
Abstract
The hydrogel prepared by graft copolymerization of starch (ST) and acrylamide (AM) is a commonly used absorbent material; however, due to their irregular network structure and a limited number of hydrophilic groups, starch-based hydrogels have poor water absorption and water retention. To overcome this, here, we provide a new preparation method for starch-based hydrogels. Using cerium ammonium nitrate (CAN) as an initiator, the starch-acrylamide-cellulose (CMC)/poly(ethylene glycol) (S-A-M/PEG) superabsorbent hydrogel was prepared by graft copolymerization. The starch-acrylamide-cellulose/poly(ethylene glycol) hydrogel network is constructed through the synergistic effect of hydrogen bonds and chemical bonds. The experimental results showed that the starch-acrylamide-cellulose/poly(ethylene glycol) superabsorbent hydrogel has a complete network structure that does not easily collapse due to its superior mechanical properties. The water swelling rate reached 80.24 times, and it reached 50.61% water retention after 16 days. This hydrogel has excellent water-absorbing and water-retaining properties, biocompatibility, and degradability, making it useful for further studies in medical, agricultural, and other fields.
Collapse
Affiliation(s)
- Longfei Gao
- Department of Polymer Materials
and Engineering, Guizhou University, Guiyang 550025, P. R. China
| | - Huiyuan Luo
- Department of Polymer Materials
and Engineering, Guizhou University, Guiyang 550025, P. R. China
| | - Qian Wang
- Department of Polymer Materials
and Engineering, Guizhou University, Guiyang 550025, P. R. China
| | - Guirong Hu
- Department of Polymer Materials
and Engineering, Guizhou University, Guiyang 550025, P. R. China
| | - Yuzhu Xiong
- Department of Polymer Materials
and Engineering, Guizhou University, Guiyang 550025, P. R. China
| |
Collapse
|
32
|
Bai Y, Liu X, Chen Q, Chen T, Jiang N, Guo Z. Myricetin ameliorates ox-LDL-induced HUVECs apoptosis and inflammation via lncRNA GAS5 upregulating the expression of miR-29a-3p. Sci Rep 2021; 11:19637. [PMID: 34608195 PMCID: PMC8490408 DOI: 10.1038/s41598-021-98916-7] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2021] [Accepted: 08/03/2021] [Indexed: 02/06/2023] Open
Abstract
Oxidized low-density lipoprotein (ox-LDL)-induced endothelial cell dysfunction is a significant event in the progression of atherosclerosis. Even Myricetin (Myr) has been exhibited strong antioxidant potency, the effect on atherosclerosis is still elusive. HUVECs were subjected to ox-LDL, before which cells were preconditioned with Myr. Cell Counting Kit-8 assay, flow cytometry, quantitative real-time polymerase chain reaction and Western blot were carried out to assess the impacts of ox-LDL and Myr on HUVECs. The expression of EndMT markers was determined by Western blot analysis and immunocytochemistry. In addition, the relationship of GAS5 and miR-29a-3p was evaluated by RNA Fluorescent in Situ Hybridization and RNA immunoprecipitation assay. Myr preconditioning prevented ox-LDL-induced apoptosis, inflammatory response, and EndMT. GAS5 was upregulated in response to ox-LDL while it was down-regulated by Myr preconditioning. GAS5 over-expression attenuates Myr protective effects against ox-LDL–mediated HUVEC injury. Besides, miR-29a-3p is a target of GAS5 and down-regulated miR-29a-3p could further reduce the effects of GAS5 in ox-LDL–mediated HUVEC. Furthermore, Myr inactivated the TLR4/NF-κB signalling pathway in ox-LDL-treated HUVEC by down-regulating GAS5 or upregulating miR-26a-5p. Myr possessed an anti-inflammatory and anti-EndMT function against ox-LDL-induced HUVEC injury by regulating the GAS5/miR-29a-3p, indicating that Myr may have an important therapeutic function for atherosclerosis.
Collapse
Affiliation(s)
- Yunpeng Bai
- Chest hospital, Tianjin university, Tianjin, 300222, China.,Tianjin chest hospital, Tianjin medical university, Tianjin, 300222, China
| | - Xiankun Liu
- Tianjin chest hospital, Tianjin medical university, Tianjin, 300222, China.,Graduate School, Tianjin Medical University, Tianjin, 300070, P. R. China
| | - Qingliang Chen
- Tianjin chest hospital, Tianjin medical university, Tianjin, 300222, China
| | - Tongyun Chen
- Tianjin chest hospital, Tianjin medical university, Tianjin, 300222, China
| | - Nan Jiang
- Tianjin chest hospital, Tianjin medical university, Tianjin, 300222, China.
| | - Zhigang Guo
- Tianjin chest hospital, Tianjin medical university, Tianjin, 300222, China.
| |
Collapse
|
33
|
Imran M, Saeed F, Hussain G, Imran A, Mehmood Z, Gondal TA, El‐Ghorab A, Ahmad I, Pezzani R, Arshad MU, Bacha U, Shariarti MA, Rauf A, Muhammad N, Shah ZA, Zengin G, Islam S. Myricetin: A comprehensive review on its biological potentials. Food Sci Nutr 2021; 9:5854-5868. [PMID: 34646551 PMCID: PMC8498061 DOI: 10.1002/fsn3.2513] [Citation(s) in RCA: 68] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Revised: 06/27/2021] [Accepted: 07/07/2021] [Indexed: 12/13/2022] Open
Abstract
Myricetin is a critical nutritive component of diet providing immunological protection and beneficial for maintaining good health. It is found in fruits, vegetables, tea, and wine. The families Myricaceae, Polygonaceae, Primulaceae, Pinaceae, and Anacardiaceae are the richest sources of myricetin. Different researchers explored the therapeutic potential of this valuable constituent such as anticancer, antidiabetic, antiobesity, cardiovascular protection, osteoporosis protection, anti-inflammatory, and hepatoprotective. In addition to these, the compound has been tested for cancer and diabetic mellitus during clinical trials. Health benefits of myricetin are related to its impact on different cell processes, such as apoptosis, glycolysis, cell cycle, energy balance, lipid level, serum protein concentrations, and osteoclastogenesis. This review explored the potential health benefits of myricetin with a specific emphasis on its mechanism of action, considering the most updated and novel findings in the field.
Collapse
Affiliation(s)
- Muhammad Imran
- Faculty of Allied Health SciencesUniversity Institute of Diet and Nutritional SciencesThe University of LahoreLahorePakistan
| | - Farhan Saeed
- Department of Food ScienceInstitute of Home and Food SciencesGovernment College UniversityFaisalabadPakistan
| | - Ghulam Hussain
- Neurochemicalbiology and Genetics Laboratory (NGL)Department of PhysiologyFaculty of Life SciencesGovernment College UniversityFaisalabadPakistan
| | - Ali Imran
- Department of Food ScienceInstitute of Home and Food SciencesGovernment College UniversityFaisalabadPakistan
| | - Zaffar Mehmood
- School of Life SciencesForman Christian College (A Chartered University)LahorePakistan
| | - Tanweer Aslam Gondal
- School of Exercise and NutritionFaculty of HealthDeakin UniversityBurwoodVictoriaAustralia
| | - Ahmed El‐Ghorab
- College of Science, Chemistry DepartmentJouf UniversitySakakaSaudi Arabia
| | - Ishtiaque Ahmad
- Department of Dairy TechnologyUniversity of Veterinary and Animal SciencesLahorePakistan
| | - Raffaele Pezzani
- Endocrinology UnitDepartment of Medicine (DIMED)University of PadovaPadovaItaly
- AIROBAssociazione Italiana per la Ricerca Oncologica di BasePadovaItaly
| | - Muhammad Umair Arshad
- Department of Food ScienceInstitute of Home and Food SciencesGovernment College UniversityFaisalabadPakistan
| | - Umar Bacha
- School of Health Sciences (SHS)University of Management and TechnologyJohar Town, LahorePakistan
| | - Mohammad Ali Shariarti
- Department of Technology of Food ProductionsK.G. RazumovskyMoscow State University of Technologies and Management (the First Cossack University)MoscowRussian Federation
| | - Abdur Rauf
- Department of ChemistryUniversity of SwabiSwabiKhyber Pakhtunkhwa (KP)Pakistan
| | - Naveed Muhammad
- Department of PharmacyAbdul Wali Khan UniversityMardanPakistan
| | - Zafar Ali Shah
- Department of ChemistryUniversity of SwabiSwabiKhyber Pakhtunkhwa (KP)Pakistan
| | - Gokhan Zengin
- Department of BiologyScience FacultySelcuk UniversityKonyaTurkey
| | - Saiful Islam
- Institute of Nutrition and Food ScienceUniversity of DhakaDhakaBangladesh
| |
Collapse
|
34
|
Zhao Z, Wang H, Tian N, Yan H, Wang J. Synthesis and biological evaluation of N 4 -hydrazone derivatives of 5,7-dihydro-6H-pyrrolo[2,3-d]pyrimidin-6-one as novel anticancer agents with antimetastatic adjunct efficacy. Arch Pharm (Weinheim) 2021; 354:e2100213. [PMID: 34368988 DOI: 10.1002/ardp.202100213] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2021] [Revised: 07/19/2021] [Accepted: 07/20/2021] [Indexed: 12/13/2022]
Abstract
To obtain new anticancer agents with antimetastatic adjunct efficacy, a series of novel N4 -hydrazone derivatives of 5,7-dihydro-6H-pyrrolo[2,3-d]pyrimidin-6-one were designed and synthesized by an eight-step reaction, with appropriate yields. All the synthesized compounds were evaluated for their antiproliferative activity against A549 and MCF-7 cells and for antiplatelet aggregation activity in vitro. The results showed that compounds 25 and 35 not only showed potent antiproliferative activity against the A549 (IC50 = 15.3 and 21.4 μM) and MCF-7 (IC50 = 15.6 and 10.9 μM) cell lines but also showed certain antiplatelet aggregation activity (inhibition rates: 47.0% and 45.8%). These results indicated that the structural modification on the N4 -hydrazone moiety of 5,7-dihydro-6H-pyrrolo[2,3-d]pyrimidin-6-one is promising to obtain novel anticancer compounds with antimetastatic adjunct efficacy. In addition, a molecular docking study was performed to investigate the possible targets, and these results indicated that compounds 25 and 35 have the potential to target EGFR, HER2, and P2Y12 .
Collapse
Affiliation(s)
- Zhichang Zhao
- Beijing Key Laboratory of Environmental and Viral Oncology, Faculty of Environment and Life, Beijing University of Technology, Beijing, China
| | - Hongjun Wang
- Beijing Tide Pharmaceutical Co., Beijing Economic Technological Development Area (BDA), Beijing, China
| | - Nana Tian
- Beijing Key Laboratory of Environmental and Viral Oncology, Faculty of Environment and Life, Beijing University of Technology, Beijing, China.,Beijing Tide Pharmaceutical Co., Beijing Economic Technological Development Area (BDA), Beijing, China
| | - Hong Yan
- Beijing Key Laboratory of Environmental and Viral Oncology, Faculty of Environment and Life, Beijing University of Technology, Beijing, China
| | - Juan Wang
- Beijing Key Laboratory of Environmental and Viral Oncology, Faculty of Environment and Life, Beijing University of Technology, Beijing, China
| |
Collapse
|
35
|
Ma J, Huang X. Research progress in role of Hippo signaling pathway in diagnosis and treatment for hepatocellular carcinoma. ZHONG NAN DA XUE XUE BAO. YI XUE BAN = JOURNAL OF CENTRAL SOUTH UNIVERSITY. MEDICAL SCIENCES 2021; 46:637-643. [PMID: 34275933 PMCID: PMC10930194 DOI: 10.11817/j.issn.1672-7347.2021.200243] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 03/13/2020] [Indexed: 11/03/2022]
Abstract
Hepatocellular carcinoma (HCC) is one of the most common malignant tumor worldwide, with high incidence and mortality. However, the exact mechanisms leading to HCC development remain unclear. The cores of the Hippo signaling pathway consist of a kinase cascade to transmit signals, which inhibits the transcriptional coactivator translocate into the nucleus and reduces the transcription of downstream proliferation-related genes. Hippo signaling pathway regulates liver development and regeneration after liver resection, and it is also related to the occurrence of HCC. The Hippo pathway regulates proliferation, apoptosis, metastasis, autophagy, metabolic reprogramming of HCC cells, affects the tumor immune microenvironment, and participates multiple-drug resistance. Further study on the role of Hippo signaling pathway in HCC is important to develop new therapeutic targets.
Collapse
Affiliation(s)
- Jiamei Ma
- Department of Gastroenterology, Affiliated Haikou Hospital of Xiangya School of Medicine, Central South University, Haikou 570208, China.
| | - Xiaoxi Huang
- Department of Gastroenterology, Affiliated Haikou Hospital of Xiangya School of Medicine, Central South University, Haikou 570208, China.
| |
Collapse
|
36
|
Miao RR, Zhan S, Hu XT, Yuan WM, Wu LJ, Cui SX, Qu XJ. Myricetin and M10, a myricetin-3-O-β-d-lactose sodium salt, modify composition of gut microbiota in mice with ulcerative colitis. Toxicol Lett 2021; 346:7-15. [PMID: 33811973 DOI: 10.1016/j.toxlet.2021.03.009] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2021] [Revised: 01/20/2021] [Accepted: 03/27/2021] [Indexed: 12/26/2022]
Abstract
Our previous studies found that M10, a myricetin-3-O-β-d-lactose sodium salt, possessed higher effects of ameliorating ulcerative colitis (UC) than Myricetin in mice. Here, we aim to investigate whether the inhibition of UC is the consequence of the effects of M10 that leads to the changed microbiota. Mice model of UC was induced by dextran sulfate sodium (DSS) treatment. M10 and Myricetin were orally administrated for 12 weeks. We performed 16S rDNA sequencing assay to analyze the composition of gut microbiota isolated from ileocecum. Both M10 and Myricetin normalized the composition of Firmicutes and Actinobacteria as healthy mice had. At genus level, the effects of M10 and Myricetin on colitis were associated to the increase of probiotics, such as Akkermansia, and the inhibition of pathogenic microorganisms, such as Ruminococcus and Parabacteroides. M10 had stronger activity than Myricetin in the improvement of biosynthesis and degradation activities, resulting to increasing metabolism of sulfur, pyruvate, steroid biosynthesis and unsaturated fatty acid biosynthesis in gut. Furthermore, M10 normalized the proportion of Firmicutes and Actinobacteria in gut microbiota. It suggests that the improvements in UC are the consequence of the effect of M10 that leads to the changed intestinal microbiota. Conclusion: M10 contributed the pharmacological effects on UC by modification of the intestinal microbiota.
Collapse
Affiliation(s)
- Rong-Rong Miao
- Department of Pharmacology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Sheng Zhan
- Department of Pharmacology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Xue-Tao Hu
- Department of Pharmacology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Wen-Min Yuan
- Drug Evaluation Center, Marine Biomedical Research Institute of Qingdao, China
| | - Li-Juan Wu
- Drug Evaluation Center, Marine Biomedical Research Institute of Qingdao, China
| | - Shu-Xiang Cui
- Toxicology and Sanitary Chemistry, School of Public Health, Capital Medical University, Beijing, China
| | - Xian-Jun Qu
- Department of Pharmacology, School of Basic Medical Sciences, Capital Medical University, Beijing, China.
| |
Collapse
|
37
|
LeBlanc L, Ramirez N, Kim J. Context-dependent roles of YAP/TAZ in stem cell fates and cancer. Cell Mol Life Sci 2021; 78:4201-4219. [PMID: 33582842 PMCID: PMC8164607 DOI: 10.1007/s00018-021-03781-2] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2020] [Revised: 12/30/2020] [Accepted: 01/28/2021] [Indexed: 02/06/2023]
Abstract
Hippo effectors YAP and TAZ control cell fate and survival through various mechanisms, including transcriptional regulation of key genes. However, much of this research has been marked by conflicting results, as well as controversy over whether YAP and TAZ are redundant. A substantial portion of the discordance stems from their contradictory roles in stem cell self-renewal vs. differentiation and cancer cell survival vs. apoptosis. In this review, we present an overview of the multiple context-dependent functions of YAP and TAZ in regulating cell fate decisions in stem cells and organoids, as well as their mechanisms of controlling programmed cell death pathways in cancer.
Collapse
Affiliation(s)
- Lucy LeBlanc
- Department of Molecular Biosciences, The University of Texas at Austin, Austin, TX, 78712, USA. .,Interdisciplinary Life Sciences Graduate Program, The University of Texas at Austin, Austin, TX, 78712, USA.
| | - Nereida Ramirez
- Department of Molecular Biosciences, The University of Texas at Austin, Austin, TX, 78712, USA.,Harvard Medical School, 25 Shattuck St, Boston, MA, 02115, USA
| | - Jonghwan Kim
- Department of Molecular Biosciences, The University of Texas at Austin, Austin, TX, 78712, USA. .,Interdisciplinary Life Sciences Graduate Program, The University of Texas at Austin, Austin, TX, 78712, USA. .,Center for Systems and Synthetic Biology, The University of Texas at Austin, Austin, TX, 78712, USA.
| |
Collapse
|
38
|
Gao X, Yanan J, Santhanam RK, Wang Y, Lu Y, Zhang M, Chen H. Garlic flavonoids alleviate H 2 O 2 induced oxidative damage in L02 cells and induced apoptosis in HepG2 cells by Bcl-2/Caspase pathway. J Food Sci 2021; 86:366-375. [PMID: 33448034 DOI: 10.1111/1750-3841.15599] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2020] [Revised: 12/07/2020] [Accepted: 12/09/2020] [Indexed: 12/18/2022]
Abstract
Liver damage is a common liver disorder, which could induce liver cancer. Oral antioxidant is one of the effective treatments to prevent and alleviate liver damage. In this study, three flavonoids namely myricetin, isoquercitrin, and isorhamnetin were isolated and identified from Laba garlic. The isolated compounds were investigated on the protective effects against H2 O2 -induced oxidative damages in hepatic L02 cells and apoptosis inducing mechanism in hepatic cancer cells HepG2 by using MTT assay, flow cytometry and western blotting analysis. Myricetin, isoquercitrin, and isorhamnetin showed proliferation inhibition on HepG2 cells with IC50 value of 44.32 ± 0.213 µM, 49.68 ± 0.192 µM, and 54.32 ± 0.176 µM, respectively. While they showed low toxicity on normal cell lines L02. They could significantly alleviate the oxidative damage towards L02 cells (P < 0.05), via inhibiting the morphological changes in mitochondria and upholding the integrity of mitochondrial structure and function. The fluorescence intensity of L02 cells pre-treated with myricetin, isoquercitrin, and isorhamnetin (100 µM) was 89.23 ± 1.26%, 89.35 ± 1.43% and 88.97 ± 0.79%, respectively. Moreover, the flavonoids could induce apoptosis in HepG2 cells via Bcl-2/Caspase pathways, where it could up-regulate the expression of Bax and down-regulate the expression of Bcl-2, Bcl-xL, pro-Caspase-3, and pro-Caspase-9 proteins in a dose dependent manner. Overall, the results suggested that the flavonoids from Laba garlic might be a promising candidate for the treatment of various liver disorders. PRACTICAL APPLICATION: Flavonoids from Laba garlic showed selective toxicity towards HepG2 cells in comparison to L02 cells via regulating Bcl-2/caspase pathway. Additionally, the isolated flavonoids expressively barred the oxidative damage induced by H2 O2 in L02 cells. These results suggested that the flavonoids from laba garlic could be a promising agent towards the development of functional foods.
Collapse
Affiliation(s)
- Xudong Gao
- Tianjin Key Laboratory for Modern Drug Delivery & High-Efficiency, School of Pharmaceutical Science and Technology, Tianjin University, Tianjin, 300072, P. R. China
| | - Jia Yanan
- Tianjin Key Laboratory for Modern Drug Delivery & High-Efficiency, School of Pharmaceutical Science and Technology, Tianjin University, Tianjin, 300072, P. R. China
| | - Ramesh Kumar Santhanam
- Faculty of Science and Marine Environment, University Malaysia Terengganu, Kuala Nerus, 21030, Malaysia
| | - Yajie Wang
- Tianjin Key Laboratory for Modern Drug Delivery & High-Efficiency, School of Pharmaceutical Science and Technology, Tianjin University, Tianjin, 300072, P. R. China
| | - Yangpeng Lu
- Tianjin Key Laboratory for Modern Drug Delivery & High-Efficiency, School of Pharmaceutical Science and Technology, Tianjin University, Tianjin, 300072, P. R. China
| | - Min Zhang
- State Key Laboratory of Food Nutrition and Safety, Tianjin Agricultural University, Tianjin, 300384, P.R. China
| | - Haixia Chen
- Tianjin Key Laboratory for Modern Drug Delivery & High-Efficiency, School of Pharmaceutical Science and Technology, Tianjin University, Tianjin, 300072, P. R. China
| |
Collapse
|
39
|
Shang XF, Dai LX, Yang CJ, Guo X, Liu YQ, Miao XL, Zhang JY. A value-added application of eugenol as acaricidal agent: The mechanism of action and the safety evaluation. J Adv Res 2020; 34:149-158. [PMID: 35024187 PMCID: PMC8655235 DOI: 10.1016/j.jare.2020.12.010] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2020] [Revised: 11/27/2020] [Accepted: 12/18/2020] [Indexed: 12/21/2022] Open
Abstract
Introduction Eugenol is a major component of essential oils of several plants, it exhibits significant antiparasitic and acaricidal activities, yet its molecular targets remain unknown. Objectives We aimed to systematically investigate the mechanism of action and the potential targets of eugenol against P. cuniculi, and evaluate the safety for laying the theoretical foundation for clinical application as an acaricide. Methods Using RNA-Seq analysis, surface plasmon resonance analysis and RNA interference assay, the mode of action of eugenol against Psoroptes cuniculi was investigated. The effect on the mitochondrial membrane potential and complex I of PC12 cells and C6/36 cells was assayed to investigate the species specificity of eugenol in insects and mammals. Finally, a safety evaluation of eugenol in vivo was performed. Results Eugenol inhibited complex I activity of the mitochondrial respiratory chain in the oxidative phosphorylation pathway by binding to NADH dehydrogenase chain 2 and resulted in the death of mites. The inhibition rates were 37.89% for 50 μg/mL and 60.26% for 100 μg/mL, respectively. Further experiments indicated that the difference in the complex I sequence between insects and mammals led to the different affinity of eugenol to specific peptide, resulting in species specificity. Eugenol exhibited significant inhibitory effects against the mitochondrial membrane potential and complex I in Aedes albopictus C6/36 cells but was not active in rat PC12 cells. Insect cells were particularly sensitive to eugenol. In contrast to the known inhibitor rotenone, eugenol had better safety and did not result in Parkinson's disease or other diseases in rats. Conclusion This is the first report on acaricidal eugenol targeting complex I of the mitochondrial respiratory chain. This work lays the foundation for the development of eugenol as an environmentally alternative acaricidal agent.
Collapse
Affiliation(s)
- Xiao-Fei Shang
- Key Laboratory of New Animal Drug Project, Gansu Province, Key Laboratory of Veterinary Pharmaceutical Development of Ministry of Agriculture, Lanzhou Institute of Husbandry and Pharmaceutical Sciences, Chinese Academy of Agricultural Sciences, Lanzhou 730050, PR China.,School of Pharmacy, Lanzhou University, Lanzhou 730000, P.R. China
| | - Li-Xia Dai
- Key Laboratory of New Animal Drug Project, Gansu Province, Key Laboratory of Veterinary Pharmaceutical Development of Ministry of Agriculture, Lanzhou Institute of Husbandry and Pharmaceutical Sciences, Chinese Academy of Agricultural Sciences, Lanzhou 730050, PR China
| | - Chen-Jie Yang
- School of Pharmacy, Lanzhou University, Lanzhou 730000, P.R. China
| | - Xiao Guo
- Tibetan Medicine Research Center, Qinghai University, Xining 810016, P.R. China
| | - Ying-Qian Liu
- School of Pharmacy, Lanzhou University, Lanzhou 730000, P.R. China
| | - Xiao-Lou Miao
- Key Laboratory of New Animal Drug Project, Gansu Province, Key Laboratory of Veterinary Pharmaceutical Development of Ministry of Agriculture, Lanzhou Institute of Husbandry and Pharmaceutical Sciences, Chinese Academy of Agricultural Sciences, Lanzhou 730050, PR China
| | - Ji-Yu Zhang
- Key Laboratory of New Animal Drug Project, Gansu Province, Key Laboratory of Veterinary Pharmaceutical Development of Ministry of Agriculture, Lanzhou Institute of Husbandry and Pharmaceutical Sciences, Chinese Academy of Agricultural Sciences, Lanzhou 730050, PR China
| |
Collapse
|
40
|
Myricetin: A review of the most recent research. Biomed Pharmacother 2020; 134:111017. [PMID: 33338751 DOI: 10.1016/j.biopha.2020.111017] [Citation(s) in RCA: 177] [Impact Index Per Article: 35.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2020] [Revised: 11/09/2020] [Accepted: 11/11/2020] [Indexed: 12/13/2022] Open
Abstract
Myricetin(MYR) is a flavonoid compound widely found in many natural plants including bayberry. So far, MYR has been proven to have multiple biological functions and it is a natural compound with promising research and development prospects. This review comprehensively retrieved and collected the latest pharmacological abstracts on MYR, and discussed the potential molecular mechanisms of its effects. The results of our review indicated that MYR has a therapeutic effect on many diseases, including tumors of different types, inflammatory diseases, atherosclerosis, thrombosis, cerebral ischemia, diabetes, Alzheimer's disease and pathogenic microbial infections. Furthermore, it regulates the expression of Hippo, MAPK, GSK-3β, PI3K/AKT/mTOR, STAT3, TLR, IκB/NF-κB, Nrf2/HO-1, ACE, eNOS / NO, AChE and BrdU/NeuN. MYR also enhances the immunomodulatory functions, suppresses cytokine storms, improves cardiac dysfunction, possesses an antiviral potential, can be used as an adjuvant treatment against cancer, cardiovascular injury and nervous system diseases, and it may be a potential drug against COVID-19 and other viral infections. Generally, this article provides a theoretical basis for the clinical application of MYR and a reference for its further use.
Collapse
|
41
|
Sajedi N, Homayoun M, Mohammadi F, Soleimani M. Myricetin Exerts its Apoptotic Effects on MCF-7 Breast Cancer Cells through Evoking the BRCA1-GADD45 Pathway. Asian Pac J Cancer Prev 2020; 21:3461-3468. [PMID: 33369440 PMCID: PMC8046300 DOI: 10.31557/apjcp.2020.21.12.3461] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2019] [Accepted: 12/06/2020] [Indexed: 02/07/2023] Open
Abstract
OBJECTIVE Myricetin is a polyphenol flavonoid with nutraceutical values which is abundantly found as the main ingredient of various foods and beverages. It has been reported that the function of myricetin is to trigger apoptosis in several types of cancers. The present study intended to investigate the apoptotic effects of myricetin on MCF-7 breast cancer cells and to assess its possible mechanisms of action. MATERIALS AND METHODS MCF-7 breast cancer cells were assigned to four groups: Control (cells in normal condition); myricetin (cells treated with the IC50 dosage of myricetin) in three different incubation times (24, 48, and 72 h). The 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay, annexin V assay, flow cytometry, real-time polymerase chain reaction (PCR), and caspase-3 assay were used to estimate the apoptosis function of myricetin in breast cancer. RESULTS The expression levels of apoptosis-related genes caspase-3, caspase-8, caspase-9, and the BAX /Bcl-2 ratio as well as the expression of p53, BRCA1, GADD45 genes were significantly increased following the treatment of MCF-7 breast cancer cells with myricetin. The annexin V assay demonstrated the significant expression of annexin which was also detected by flow cytometry. CONCLUSION Myricetin efficiently induces apoptosis in MCF-7 breast cancer cells by evoking both extrinsic and intrinsic apoptotic pathways. Myricetin may exert its apoptotic effects on MCF-7 cells by inducing the BRCA1- GADD45 pathway. .
Collapse
Affiliation(s)
- Nayereh Sajedi
- Department of Anatomical Sciences, Isfahan University of Medical Sciences, Iran.
| | - Mansour Homayoun
- Department of Anatomical Sciences, Isfahan University of Medical Sciences, Iran.
| | | | - Mitra Soleimani
- Department of Anatomical Sciences, Isfahan University of Medical Sciences, Iran.
| |
Collapse
|
42
|
Nutraceutical Properties of Polyphenols against Liver Diseases. Nutrients 2020; 12:nu12113517. [PMID: 33203174 PMCID: PMC7697723 DOI: 10.3390/nu12113517] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2020] [Revised: 11/08/2020] [Accepted: 11/12/2020] [Indexed: 02/07/2023] Open
Abstract
Current food tendencies, suboptimal dietary habits and a sedentary lifestyle are spreading metabolic disorders worldwide. Consequently, the prevalence of liver pathologies is increasing, as it is the main metabolic organ in the body. Chronic liver diseases, with non-alcoholic fatty liver disease (NAFLD) as the main cause, have an alarming prevalence of around 25% worldwide. Otherwise, the consumption of certain drugs leads to an acute liver failure (ALF), with drug-induced liver injury (DILI) as its main cause, or alcoholic liver disease (ALD). Although programs carried out by authorities are focused on improving dietary habits and lifestyle, the long-term compliance of the patient makes them difficult to follow. Thus, the supplementation with certain substances may represent a more easy-to-follow approach for patients. In this context, the consumption of polyphenol-rich food represents an attractive alternative as these compounds have been characterized to be effective in ameliorating liver pathologies. Despite of their structural diversity, certain similar characteristics allow to classify polyphenols in 5 groups: stilbenes, flavonoids, phenolic acids, lignans and curcuminoids. Herein, we have identified the most relevant compounds in each group and characterized their main sources. By this, authorities should encourage the consumption of polyphenol-rich products, as most of them are available in quotidian life, which might reduce the socioeconomical burden of liver diseases.
Collapse
|
43
|
Antineoplastic Activity of Chrysin against Human Hepatocellular Carcinoma: New Insight on GPC3/SULF2 Axis and lncRNA-AF085935 Expression. Int J Mol Sci 2020; 21:ijms21207642. [PMID: 33076548 PMCID: PMC7589298 DOI: 10.3390/ijms21207642] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2020] [Revised: 10/06/2020] [Accepted: 10/13/2020] [Indexed: 12/17/2022] Open
Abstract
The natural flavonoid chrysin possesses antiproliferative activity against various types of cancers, including hepatocellular carcinoma (HCC), which is a common malignancy. However, the exact mechanism of chrysin antiproliferative activity remains unclear. This research was executed to explore the impact of chrysin on glypican-3 (GPC3)/sulfatase-2 (SULF2) axis and lncRNA-AF085935 expression in HCC using HepG2 cells. Cisplatin (20, 50, 100 μg/mL), chrysin (15, 30, and 60 μg/mL) and the combination of 50 μg/mL cisplatin with different concentrations of chrysin were applied for 24/48 h. Cell viability was determined by MTT assay. Protein levels of GPC3 and SULF2 were measured by ELISA at 24/48 h. GPC3 immunoreactivity was detected by immunocytochemistry. Moreover, GPC3 and SULF2 mRNA expressions in addition to lncRNA-AF085935 expression were assessed by qPCR at 48 h. The GPC3 protein, immunostaining and mRNA levels, SULF2 protein and mRNA levels, as well as lncRNA-AF085935 expression, were decreased significantly with cisplatin and chrysin alone when compared with the control untreated HepG2 cells. However, the combination treatment exhibited a better chemopreventive effect in a dose- and time-dependent manner. This study demonstrated, for the first time, the antiproliferative activity of chrysin against HCC through the suppression of the GPC3/SULF2 axis along with the downregulation of lncRNA-AF085935 expression. Synergistic effect of chrysin with cisplatin could potentiate their antiproliferative action in a dose- and time-dependent manner.
Collapse
|
44
|
Gupta G, Siddiqui MA, Khan MM, Ajmal M, Ahsan R, Rahaman MA, Ahmad MA, Arshad M, Khushtar M. Current Pharmacological Trends on Myricetin. Drug Res (Stuttg) 2020; 70:448-454. [DOI: 10.1055/a-1224-3625] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
AbstractMyricetin is a member of the group of flavonoids called flavonols. Myricetin is obtained from various fruit, vegetables, tea, berries and red wine. Myricetin is characterized by the pysrogallol B-ring, and the more hydroxylated structure is known to be capable for its increased biological properties compared with other flavonols. Myricetin is produced by the Myricaceae, Anacardiaceae, Polygonaceae, Pinaceae and Primulacea families. It is soluble in organic solvent such as ethanol, DMSO (dimethyl sulfoxide), and dimethyl formamide (DMF). It is sparingly soluble in aqueous buffers. Myricetin shows its various pharmacological activities including antioxidant, anti-amyloidogenic, antibacterial, antiviral, antidiabetic, anticancer, anti-inflammatory, anti-epileptic and anti-ulcer. This review article focuses on pharmacological effects of Myricetin on different diseases such as osteoporotic disorder, anti-inflammatory disorder, alzheimer’s disease, anti-epileptic, cancer, cardiac disorder, diabetic metabolic disorder, hepatoprotective disorder and gastro protective disorder.
Collapse
Affiliation(s)
- Gudiya Gupta
- Department of Pharmacology, Faculty of Pharmacy, Integral University, Lucknow, India
| | - Mohd Aftab Siddiqui
- Department of Pharmacology, Faculty of Pharmacy, Integral University, Lucknow, India
| | - Mohd Muazzam Khan
- Department of Pharmacology, Faculty of Pharmacy, Integral University, Lucknow, India
| | - Mohd Ajmal
- Department of Pharmacology, Faculty of Pharmacy, Integral University, Lucknow, India
| | - Rabiya Ahsan
- Department of Pharmacology, Faculty of Pharmacy, Integral University, Lucknow, India
| | - Md Azizur Rahaman
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Integral University, Lucknow, India
| | - Md Afroz Ahmad
- Department of Pharmacology, Faculty of Pharmacy, Integral University, Lucknow, India
| | - Md Arshad
- Department of Zoology, Aligarh Muslim University, Aligarh, India
| | - Mohammad Khushtar
- Department of Pharmacology, Faculty of Pharmacy, Integral University, Lucknow, India
| |
Collapse
|
45
|
Tuttis K, Costa DLMGD, Serpeloni JM, Santos LCD, Varanda EA, Vilegas W, Martínez-López W, Cólus IMDS. Phytochemical Profile, and Antiproliferative and Proapoptotic Effects of Pouteria ramiflora (Mart.) Radlk. Leaf Extract, and Its Synergism with Cisplatin in HepG2 Cells. J Med Food 2020; 24:452-463. [PMID: 32757998 DOI: 10.1089/jmf.2020.0045] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Different species of the genus Pouteria have been used in folk medicine for the treatment of inflammation, fever, ulcers, diabetes, and diarrhea. We analyzed the phytochemical profile of the hydroethanolic extract from Pouteria ramiflora leaves by electrospray ionization ion trap tandem mass spectrometry and high-performance liquid chromatography-diode array detection, and examined whether it alone and in combination with cisplatin interfered with cell proliferation and death processes in HepG2 (human hepatocellular carcinoma) and FGH (human gingival fibroblasts) cells. Five compounds were identified in the extract: gallic acid, myricetin-3-O-α-l-arabinopyranoside, quercetin-3-O-β-d-galactopyranoside, myricetin-3-O-α-l-rhamnopyranoside, and myricetin-3-O-β-d-galactopyranoside. The extract was cytotoxic to both cell lines by inducing apoptotic cell death and acted in synergy with cisplatin; such effect was stronger in HepG2 cells than in FGH cells, demonstrating some selectivity to tumor cells. In HepG2 cells, the extract exerted antiproliferative effect mediated by induction of cell cycle arrest at the S and G2/M phases. Association of the extract with cisplatin enhanced the latter's antiproliferative effect, arrested the cell cycle at the S phase by CDK2 modulation, and reduced the number of anti-cyclin D1-stained HepG2 cells. Simultaneous treatment with the extract and cisplatin increased the latter's cytotoxicity, apoptotic cell death, and BAX expression in HepG2 cells. Altogether, the results reported herein indicate that P. ramiflora extract is a possible adjuvant to cancer therapy, which can circumvent the cisplatin-mediated resistance mechanisms in cancer cells.
Collapse
Affiliation(s)
- Katiuska Tuttis
- Department of General Biology, Biological Science Center, Londrina State University-UEL, Londrina, PR, Brazil
| | - Daryne Lu Maldonado Gomes da Costa
- Department of Organic Chemistry, Institute of Chemistry, São Paulo State University-UNESP, Araraquara, SP, Brazil.,Federal Institute of Mato Grosso, Bela Vista Campus-IFMT, Cuiabá, MT, Brazil
| | - Juliana Mara Serpeloni
- Department of General Biology, Biological Science Center, Londrina State University-UEL, Londrina, PR, Brazil
| | - Lourdes Campaner Dos Santos
- Department of Organic Chemistry, Institute of Chemistry, São Paulo State University-UNESP, Araraquara, SP, Brazil
| | - Eliana Aparecida Varanda
- Department of Biological Sciences, Faculty of Pharmaceutical Sciences of Araraquara, São Paulo State University-UNESP, Araraquara, SP, Brazil
| | - Wagner Vilegas
- Experimental Campus of the Paulista Coast, São Paulo State University-UNESP, São Vicente, SP, Brazil
| | | | - Ilce Mara de Syllos Cólus
- Department of General Biology, Biological Science Center, Londrina State University-UEL, Londrina, PR, Brazil
| |
Collapse
|
46
|
Braicu C, Zanoaga O, Zimta AA, Tigu AB, Kilpatrick KL, Bishayee A, Nabavi SM, Berindan-Neagoe I. Natural compounds modulate the crosstalk between apoptosis- and autophagy-regulated signaling pathways: Controlling the uncontrolled expansion of tumor cells. Semin Cancer Biol 2020; 80:218-236. [PMID: 32502598 DOI: 10.1016/j.semcancer.2020.05.015] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2020] [Revised: 05/22/2020] [Accepted: 05/24/2020] [Indexed: 02/07/2023]
Abstract
Due to the high number of annual cancer-related deaths, and the economic burden that this malignancy affects today's society, the study of compounds isolated from natural sources should be encouraged. Most cancers are the result of a combined effect of lifestyle, environmental factors, and genetic and hereditary components. Recent literature reveals an increase in the interest for the study of phytochemicals from traditional medicine, this being a valuable resource for modern medicine to identify novel bioactive agents with potential medicinal applications. Phytochemicals are components of traditional medicine that are showing promising application in modern medicine due to their antitumor activities. Recent studies regarding two major mechanisms underlying cancer development and regulation, apoptosis and autophagy, have shown that the signaling pathways of both these processes are significantly interconnected through various mechanisms of crosstalk. Phytochemicals are able to activate pro-autophagic and pro-apoptosis mechanisms. Understanding the molecular mechanism involved in apoptosis-autophagy relationship modulated by phytochemicals plays a key role in development of a new therapeutic strategy for cancer treatment. The purpose of this review is to outline the bioactive properties of the natural phytochemicals with validated antitumor activity, focusing particularly on their role in the regulation of apoptosis and autophagy crosstalk that triggers the uncontrolled expansion of tumor cells. Furthermore, we have also critically discussed the limitations and challenges of existing research strategies and the prospective research directions in this field.
Collapse
Affiliation(s)
- Cornelia Braicu
- Research Center for Functional Genomics and Translational Medicine, Iuliu Hatieganu University of Medicine and Pharmacy, 40015, Cluj-Napoca, Romania
| | - Oana Zanoaga
- Research Center for Functional Genomics and Translational Medicine, Iuliu Hatieganu University of Medicine and Pharmacy, 40015, Cluj-Napoca, Romania
| | - Alina-Andreea Zimta
- MEDFUTURE-Research Center for Advanced Medicine, Iuliu Hatieganu University of Medicine and Pharmacy, 40015, Cluj-Napoca, Romania
| | - Adrian Bogdan Tigu
- MEDFUTURE-Research Center for Advanced Medicine, Iuliu Hatieganu University of Medicine and Pharmacy, 40015, Cluj-Napoca, Romania; Babeș-Bolyai University, Faculty of Biology and Geology, 42 Republicii Street, 400015, Cluj-Napoca, Romania
| | | | - Anupam Bishayee
- Lake Erie College of Osteopathic Medicine, Bradenton, FL, 34211, USA
| | - Seyed Mohammad Nabavi
- Applied Biotechnology Research Center, Baqiyatallah University of Medical Sciences, Tehran, 1435916471, Iran
| | - Ioana Berindan-Neagoe
- Research Center for Functional Genomics and Translational Medicine, Iuliu Hatieganu University of Medicine and Pharmacy, 40015, Cluj-Napoca, Romania; Department of Functional Genomics and Experimental Pathology, The Oncology Institute "Prof. Dr. Ion Chiricuta", 400015, Cluj-Napoca, Romania.
| |
Collapse
|
47
|
Yamada N, Matsushima-Nishiwaki R, Kozawa O. Quercetin suppresses the migration of hepatocellular carcinoma cells stimulated by hepatocyte growth factor or transforming growth factor-α: Attenuation of AKT signaling pathway. Arch Biochem Biophys 2020; 682:108296. [PMID: 32032576 DOI: 10.1016/j.abb.2020.108296] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2019] [Revised: 01/27/2020] [Accepted: 02/03/2020] [Indexed: 02/06/2023]
|
48
|
Anti-tumor effects and associated molecular mechanisms of myricetin. Biomed Pharmacother 2019; 120:109506. [PMID: 31586904 DOI: 10.1016/j.biopha.2019.109506] [Citation(s) in RCA: 82] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2019] [Revised: 09/18/2019] [Accepted: 09/26/2019] [Indexed: 12/14/2022] Open
Abstract
Myricetin (3, 5, 7, 3', 4', 5'-hexahydroxyflavone) is a natural flavonol compound found in a large variety of plants, including berries, oranges, grapes, herbs, teas, and wine. In the last decade, a convergence of evidence has demonstrated that myricetin has good biological activity as an anti-tumor, anti-inflammatory, and anti-oxidation agent. In studies involving various types of cancer cells, myricetin has been shown to suppress cancer cell invasion and metastasis, to induce cell cycle arrest and apoptosis of cancer cells, and to inhibit their proliferation. These findings have raised interest in myricetin as a potential tumor inhibitor in human patients. In this review, evidence of myricetin's anti-tumor activity and its underlying molecular mechanisms published in the last decade are summarized.
Collapse
|
49
|
Anti-Tumor Potential of IMP Dehydrogenase Inhibitors: A Century-Long Story. Cancers (Basel) 2019; 11:cancers11091346. [PMID: 31514446 PMCID: PMC6770829 DOI: 10.3390/cancers11091346] [Citation(s) in RCA: 77] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2019] [Revised: 09/01/2019] [Accepted: 09/02/2019] [Indexed: 01/15/2023] Open
Abstract
The purine nucleotides ATP and GTP are essential precursors to DNA and RNA synthesis and fundamental for energy metabolism. Although de novo purine nucleotide biosynthesis is increased in highly proliferating cells, such as malignant tumors, it is not clear if this is merely a secondary manifestation of increased cell proliferation. Suggestive of a direct causative effect includes evidence that, in some cancer types, the rate-limiting enzyme in de novo GTP biosynthesis, inosine monophosphate dehydrogenase (IMPDH), is upregulated and that the IMPDH inhibitor, mycophenolic acid (MPA), possesses anti-tumor activity. However, historically, enthusiasm for employing IMPDH inhibitors in cancer treatment has been mitigated by their adverse effects at high treatment doses and variable response. Recent advances in our understanding of the mechanistic role of IMPDH in tumorigenesis and cancer progression, as well as the development of IMPDH inhibitors with selective actions on GTP synthesis, have prompted a reappraisal of targeting this enzyme for anti-cancer treatment. In this review, we summarize the history of IMPDH inhibitors, the development of new inhibitors as anti-cancer drugs, and future directions and strategies to overcome existing challenges.
Collapse
|