A Comprehensive Analysis and Anti-Cancer Activities of Quercetin in ROS-Mediated Cancer and Cancer Stem Cells
Abstract
:1. Introduction
2. Overview of Quercetin
3. ROS Regulation within Cellular System
4. Enzymatic Mechanism of ROS Production in Cancer and Cancer Stem Cell
5. Significant Role of ROS Moieties in Cancer Cell Signaling Pathways
6. Quercetin Upregulate the ROS Levels in Cancerous Cells
6.1. Balanced Oxidative Stress in Cancerous Cells
6.2. Unbalanced Reactive Oxygen Species in Cancerous Cells by Quercetin
6.3. Quercetin-Mediated Apoptosis via Regulating Cancer Cell Signaling Pathways
7. ROS Are Responsible for Quercetin-Mediated Cell Death in Cancer
8. Molecular Mechanism for Quercetin-Induced Cancer and Cancer Stem Cell Death
8.1. ROS-Mediated Regulation of the MAPK/ERK1/2 Pathways
Cancer Type | Research/Experiment Type | Research Models/Cell Lines | Mechanism of Action | Outcomes | Reference |
---|---|---|---|---|---|
Leukemia | In vitro | U937 cell | Cell cycle arrest at G2/M, decrease in cyclin D, cyclin E and E2F, increase in the level of the cyclin B | Apoptosis and growth inhibition in The human leukemia cells | [49] |
Leukemia | In vitro and In vivo | HL-60 AML cells | Induced caspase-8, caspase-9, and caspase-3 activation, PARP cleavage, mitochondrial membrane depolarization, induced intratumoral oxidative stress | Anticancer effects in acute myeloid leukemia (AML) cells | [145] |
Leukemia | In vitro | Acute leukemia cell line, HL-60 | Induces apoptosis in a caspase-3-dependent pathway by inhibiting Cox-2 expression and regulates the expression of downstream apoptotic components, including Bcl-2 and Bax | Inhibited cell proliferation and induced apoptosis in a time- and dose-dependent manner | [146] |
Liver cancer | In vivo | Hyperplastic nodules in rat liver | Prevented DEN-mediated development of hepatocarcinoma and oxidative damage in rat liver | Potent therapeutic formulation against DEN-induced hepatocarcinoma | [147] |
Liver cancer | In vivo | HepG2 cells | Induced apoptosis, alter cell cycle in hepg2 cells, decreased the gene expression of cyclin D1 | Significantly inhibit the growth and proliferation of liver cancer cell. | [148] |
Colorectal cancer | In vivo | ApcMin mice, and HCT116 tumors | Decreased tumor proliferation and development, increased apoptosis and p53 expression | Chemical modification of quercetin generates safe and efficacious agents for colorectal Cancer | [149] |
Colon cancer | In vitro | HT-29 colon cancer cells | Induced caspase-3 cleavage, increased PARP cleavage, decreased the expression of Sp1, Sp3, Sp4 mrna, and survivin, decreased microrna-27a, and induced ZBTB10 | Cytotoxic effects in colon cancer cells, Resulting in apoptosis. | [150] |
Colon cancer | In vitro | CX-1, SW480, HT-29, HCT116 | Downregulation of transcriptional activity of β-catenin/Tcf signal pathway, and cyclin D1 and the survivin gene | Inhibited proliferation in colon cancer cells | [151] |
Colon cancer | In vivo | Male F344 rats | Decreased β-catenin accumulation in BCA-C; decreased number of ACF | Suppressed tumor growth and at high dose reduced colorectal carcinogenesis | [152] |
Lung cancer | In vitro | H460, A549 | Induction of DR5 and suppression of survivin expression | TRAIL-induced cytotoxicity in lung cancer cells | [153] |
Lung cancer | In vitro | H460 | Increased the expression of TRAILR, caspase-10, DFF45, TNFR 1, FAS, and decreased the expression of NF-κb, ikkα | Useful in the prevention and therapy of NSCLC | [154] |
Lung cancer | In vitro | Human A549 lung cancer cells | Downregulation of the expression of cdk1 and cyclin B, increased PPAR-γ expression | Inhibiton of human A549 lung cancer cell growth | [155] |
Ovarian cancer | In vitro and in vivo | A2780S ovarian cancer cells | Activated caspase-3 and caspase-9. MCL-1 downregulation, Bcl-2 downregulation, Bax upregulation, inhibited angiogenesis in vivo | Novel nano-formulation of quercetin with a potential clinical application in ovarian cancer therapy | [156] |
Ovarian cancer | In vitro | SKOV3 | Reduction in cyclin D1 level | Inhibited cell growth in ovarian carcinoma | [157] |
Breast cancer | In vitro | MCF-7, HCC1937, SK-Br3, 4T1, MDA-MB-231 | Decreased Bcl-2 expression, increasedBax expression, inhibition of PI3K-Akt pathway | Decreases proliferationand increases apoptosis in MCF-7 human breast cancer cells | [158] |
Breast cancer | In vitro | MDA-MB-231 | Induced the expression of E-cadherin and downregulated vimentin levels, modulation of β-catenin target genes such as cyclin D1 and c-Myc | Inhibited TNBC metastasis and also improve the therapeutic efficacy of existing chemotherapeutic drug | [159] |
Breast cancer | In vitro | MCF-7 | Suppressed the epithelial–mesenchymal transition process, upregulated E-cadherin expression, downregulated vimentin and MMP-2 expression, decreased Notch1 expression and induced PI3K and Akt phosphorylation | Potential therapeutic for the treatment of triple negative and hormone-sensitive breast cancer | [160] |
Breast cancer | In vivo | MCF-7/DO X | Overcoming the drug efflux by ABC transporters and promoting PCD with the arrest of cell cycle, counteracted P-gp and BCRPPumps | Reverses multidrug resistance and restores chemosensitivity to human breast cancer cells | [161] |
Gastric cancer | In vitro | GCSCs | Activation of caspase-3 and -9, downregulation of Bcl-2, upregulation of Bax and cytochrome c (Cyt-c) | Potential agent for the treatment of gastric cancer. | [162] |
Pancreatic cancer | In vivo | PANC-1, PATU-8988 | Decreased the secretion of MMP and MMP7, blocked the STAT3 signaling pathway | New therapeutic strategy for the treatment of pancreatic cancer cells That targets emt, invasion, and metastasis. | [163] |
Prostate cancer | In vitro and in vivo | PC-3, HUVECs | Reduced angiogenesis, increased TSP-1 protein and mrna expression | Good foundation for applying quercetin to clinical for human prostate cancer in the near future | [164] |
Sort of Cancer | Research/Experiment Type | Specific Cell Line/s | Core Molecular Mechanism | Doses | Final Outcomes | Reference |
---|---|---|---|---|---|---|
Colorectal cancer | In vitro | HT29 cells | Induced G2/M arrest | 75 µM | Enhanced the efficacy of low concentration of doxorubicin chemotherapy in inhibiting cell proliferation, enhance cytotoxicity and apoptosis | [165] |
Breast cancer | In vitro | MDA-MB-231 | Lowered the expression levels of proteins such as aldehyde dehydrogenase 1A1, C-X-C chemokine receptor type 4, mucin 1 and epithelial cell adhesion molecules responsible for tumorigenesis | 50 μM | Suppressed breast cancer stem cell proliferation, self-renewal, and invasiveness | [166] |
Prostate cancer | In vitro | PC-3 and LNCaP cells | Activated capase-3/7 and inhibit the expression of Bcl-2, surviving and XIAP in CSCs. Furthermore, inhibits epithelial-mesenchymal transition by inhibiting the expression of vimentin, slug, snail and nuclear β-catenin, and the activity of LEF-1/TCF responsive reporter | 20 μM | Quercetin synergized with epigallocatechin gallate inhibited the self-renewal properties of prostate CSCs, inducing apoptosis, and blocking CSC’s migration and invasion | [167] |
Breast cancer | In vitro | MCF-7 and MCF-7/dox cell lines | Downregulation of P-gp expression and eliminate BCSCs mediated by YB-1 nuclear translocation | 0.7 μm | Enhanced the antitumor activity of doxorubicin, paclitaxel and vincristine by reversing multidrug resistance | [168] |
Prostate cancer | In vitro | PC3, LNCaP and ARPE-19 cells | Down-regulated the expression of PI3K/PTEN, MAPK and NF-κB signaling pathways | 40 μM | Quercetin inhibited PC3 and CD44+/CD133+ stem cell proliferation in a time- and dose-dependent manner. | [169] |
Pancreatic cancer | In vitro | Human pancreatic CSCs (CD133þ/CD44þ/CD24þ/ESAþ) | Inhibited the expression of Bcl-2 and XIAP and activate caspase-3, attenuate transcriptional activities of Gli and TCF/LEF | 20μM | Epigallocatechin-3-gallate with quercetin had synergistic inhibitory effects on self-renewal capacity of pancreatic CSCs | [170] |
Pancreatic cancer | In vitro | PANC-1 | Affected IL-1b, TNF-α, vimentin, N-cadherin, and ACTA-2 expressions | 10μM | Quercetin could prevent Epithelial Mesenchymal Transition by reducing expression of N-cadherin | [171] |
Breast cancer | In vitro | MCF-7 and MDA-MB-231 cells | Suppressed EGFR signaling and inhibited PI3K/Akt/mTOR/GSK-3β | 50μM(MCF-7), 100μM (MDA-MB-231) | Gold nanoparticles-conjugated quercetin reduce cell proliferation through induction of apoptosis of breast cancer cell | [172] |
Pancreatic cancer | In vitro | IA Paca-2, BxPC3, AsPC-1, HPAC and PANC1 | Silencing RAGE expression by suppressing the PI3K/AKT/mTOR axis | Quercetin increased gemcitabine drug chemosensitivity in pancreatic cancer cells | [173] | |
Colon cancer | In vitro | HT-29, SW-620, and Caco-2 cells | Redistributed the TRAIL receptors and other components of the DISC complex into lipid rafts, which facilitates the formation of the DISC and the downstream signaling pathway, contributing to Bax conformational changes, release of cytochrome c, and apoptosis. | 30 μmol/L | Quercetin enhanced tumor necrosis factor-related apoptosis inducing ligand (TRAIL)-mediated apoptosis | [174] |
Breast cancer | In vivo | NOD/SCID mice | Inhibited the overexpression of Hsp27 through the regulation of epithelial mesenchymal transition and nuclear factor-kB | (50, 25 or 12.5 μM) | Effectively suppressed the overexpression of Hsp27 and inhibit the breast cancer stem cells | [175] |
8.2. ROS-Mediated Regulation of the p53 Pathway
8.3. ROS-Mediated Regulation of the JAK/STAT and TRAIL Pathways
8.4. ROS-Mediated Regulation of the AMPKα1/ASK1/p38 Pathways
8.5. ROS-Mediated Regulation of the RAGE/PI3K/AKT/mTOR Axis
8.6. ROS-Mediated Regulation of the HMGB1 and NF-κB Pathways
8.7. ROS-Mediated Regulation of the Nrf2-Induced Phase II Enzyme and Signaling Pathways
9. Quercetin Induced Cell Death through Cell Cycle Arrest → G1, G2/M, etc. Phase
10. Concluding Remarks and Future Directions
Author Contributions
Funding
Data Availability Statement
Conflicts of Interest
References
- Liu, J.; Guo, W.; Li, J.; Li, X.; Geng, J.; Chen, Q.; Gao, J. Tumor-targeting novel manganese complex induces ROS-mediated apoptotic and autophagic cancer cell death. Int. J. Mol. Med. 2015, 35, 607–616. [Google Scholar] [CrossRef] [Green Version]
- Paul, P.; Biswas, P.; Dey, D.; Saikat, A.S.M.; Islam, M.A.; Sohel, M.; Hossain, R.; Mamun, A.A.; Rahman, M.A.; Hasan, M.N.; et al. Exhaustive Plant Profile of “Dimocarpus longan Lour” with Significant Phytomedicinal Properties: A Literature Based-Review. Processes 2021, 9, 1803. [Google Scholar] [CrossRef]
- Liou, G.-Y.; Storz, P.J. Reactive oxygen species in cancer. Free. Radic. Res. 2010, 44, 479–496. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Sena, L.A.; Chandel, N.S. Physiological roles of mitochondrial reactive oxygen species. Mol. Cell 2012, 48, 158–167. [Google Scholar] [CrossRef] [Green Version]
- Covarrubias, L.; Hernández-García, D.; Schnabel, D.; Salas-Vidal, E.; Castro-Obregón, S. Function of reactive oxygen species during animal development: Passive or active? Dev. Biol. 2008, 320, 1–11. [Google Scholar] [CrossRef] [Green Version]
- Martin, K.; Barrett, J.C. Reactive oxygen species as double-edged swords in cellular processes: Low-dose cell signaling versus high-dose toxicity. Hum. Exp. Toxicol. 2002, 21, 71–75. [Google Scholar] [CrossRef] [PubMed]
- Nadege, B.; Patrick, L.; Rodrigue, R. Mitochondria: From bioenergetics to the metabolic regulation of carcinogenesis. Front. Biosci. 2009, 14, 4015–4034. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Gibellini, L.; Pinti, M.; Nasi, M.; De Biasi, S.; Roat, E.; Bertoncelli, L.; Cossarizza, A. Interfering with ROS Metabolism in Cancer Cells: The Potential Role of Quercetin. Cancers 2010, 2, 1288–1311. [Google Scholar] [CrossRef] [Green Version]
- Sullivan, L.B.; Chandel, N.S. Mitochondrial reactive oxygen species and cancer. Cancer Metab. 2014, 2, 17. [Google Scholar] [CrossRef] [Green Version]
- Redza-Dutordoir, M.; Averill-Bates, D.A. Activation of apoptosis signalling pathways by reactive oxygen species. Biochim. Biophys. Acta (BBA)-Mol. Cell Res. 2016, 1863, 2977–2992. [Google Scholar]
- Trachootham, D.; Alexandre, J.; Huang, P. Targeting cancer cells by ROS-mediated mechanisms: A radical therapeutic approach? Nat. Rev. Drug Discov. 2009, 8, 579–591. [Google Scholar] [CrossRef]
- NavaneethaKrishnan, S.; Rosales, J.L.; Lee, K.-Y.J.O. Targeting Cdk5 for killing of breast cancer cells via perturbation of redox homeostasis. Oncoscience 2018, 5, 152. [Google Scholar] [CrossRef]
- Sattler, M.; Verma, S.; Shrikhande, G.; Byrne, C.H.; Pride, Y.B.; Winkler, T.; Greenfield, E.A.; Salgia, R.; Griffin, J.D. The BCR/ABL tyrosine kinase induces production of reactive oxygen species in hematopoietic cells. J. Biol. Chem. 2000, 275, 24273–24278. [Google Scholar] [CrossRef] [Green Version]
- Rahman, M.A.; Rahman, M.D.H.; Hossain, M.S.; Biswas, P.; Islam, R.; Uddin, M.J.; Rahman, M.H.; Rhim, H. Molecular Insights into the Multifunctional Role of Natural Compounds: Autophagy Modulation and Cancer Prevention. Biomedicines 2020, 8, 517. [Google Scholar] [CrossRef]
- Rahman, M.A.; Rahman, M.H.; Biswas, P.; Hossain, M.S.; Islam, R.; Hannan, M.A.; Uddin, M.J.; Rhim, H. Potential Therapeutic Role of Phytochemicals to Mitigate Mitochondrial Dysfunctions in Alzheimer’s Disease. Antioxidants 2020, 10, 23. [Google Scholar] [CrossRef] [PubMed]
- Hashemzaei, M.; Delarami Far, A.; Yari, A.; Heravi, R.E.; Tabrizian, K.; Taghdisi, S.M.; Sadegh, S.E.; Tsarouhas, K.; Kouretas, D.; Tzanakakis, G.; et al. Anticancer and apoptosis-inducing effects of quercetin in vitro and in vivo. Oncol. Rep. 2017, 38, 819–828. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Arefin, A.; Ismail Ema, T.; Islam, T.; Hossen, S.; Islam, T.; Al Azad, S.; Uddin Badal, N.; Islam, A.; Biswas, P.; Alam, N.U.; et al. Target specificity of selective bioactive compounds in blocking α-dystroglycan receptor to suppress Lassa virus infection: An in silico approach. J. Biomed. Res. 2021, 35, 459–473. [Google Scholar] [CrossRef]
- Rahman, M.S.; Zilani, M.N.H.; Islam, M.A.; Hasan, M.M.; Islam, M.M.; Yasmin, F.; Biswas, P.; Hirashima, A.; Rahman, M.A.; Hasan, M.N.; et al. In Vivo Neuropharmacological Potential of Gomphandra tetrandra (Wall.) Sleumer and In-Silico Study against β-Amyloid Precursor Protein. Processes 2021, 9, 1449. [Google Scholar] [CrossRef]
- Khan, R.A.; Hossain, R.; Siyadatpanah, A.; Al-Khafaji, K.; Khalipha, A.B.R.; Dey, D.; Asha, U.H.; Biswas, P.; Saikat, A.S.M.; Chenari, H.A.; et al. Diterpenes/Diterpenoids and Their Derivatives as Potential Bioactive Leads against Dengue Virus: A Computational and Network Pharmacology Study. Molecules 2021, 26, 6821. [Google Scholar] [CrossRef] [PubMed]
- Schnekenburger, M.; Dicato, M.; Diederich, M. Plant-derived epigenetic modulators for cancer treatment and prevention. Biotechnol. Adv. 2014, 32, 1123–1132. [Google Scholar] [CrossRef] [PubMed]
- Butler, M.S.; Robertson, A.A.; Cooper, M.A. Natural product and natural product derived drugs in clinical trials. Nat. Prod. Rep. 2014, 31, 1612–1661. [Google Scholar] [CrossRef]
- Hashemzaei, M.; Barani, A.K.; Iranshahi, M.; Rezaee, R.; Tsarouhas, K.; Tsatsakis, A.M.; Wilks, M.F.; Tabrizian, K. Effects of resveratrol on carbon monoxide-induced cardiotoxicity in rats. Environ. Toxicol. Pharmacol. 2016, 46, 110–115. [Google Scholar] [CrossRef]
- Hashemzaei, M.; Entezari Heravi, R.; Rezaee, R.; Roohbakhsh, A.; Karimi, G. Regulation of autophagy by some natural products as a potential therapeutic strategy for cardiovascular disorders. Eur. J. Pharmacol. 2017, 802, 44–51. [Google Scholar] [CrossRef]
- Sohel, M.; Biswas, P.; Al Amin, M.; Hossain, M.A.; Sultana, H.; Dey, D.; Aktar, S.; Setu, A.; Khan, M.S.; Paul, P.; et al. Genistein, a Potential Phytochemical against Breast Cancer Treatment-Insight into the Molecular Mechanisms. Processes 2022, 10, 415. [Google Scholar] [CrossRef]
- Dey, D.; Hasan, M.M.; Biswas, P.; Papadakos, S.P.; Rayan, R.A.; Tasnim, S.; Bilal, M.; Islam, M.J.; Arshe, F.A.; Arshad, E.M.; et al. Investigating the Anticancer Potential of Salvicine as a Modulator of Topoisomerase II and ROS Signaling Cascade. Front. Oncol. 2022, 12, 899009. [Google Scholar] [CrossRef] [PubMed]
- Dey, D.; Quispe, C.; Hossain, R.; Jain, D.; Ahmed Khan, R.; Janmeda, P.; Islam, M.T.; Ansar Rasul Suleria, H.; Martorell, M.; Daştan, S.D.; et al. Ethnomedicinal Use, Phytochemistry, and Pharmacology of Xylocarpus granatum J. Koenig. Evid.-Based Complementary Altern. Med. 2021, 2021, 8922196. [Google Scholar] [CrossRef]
- Fridlender, M.; Kapulnik, Y.; Koltai, H. Plant derived substances with anti-cancer activity: From folklore to practice. Front. Plant Sci. 2015, 6, 799. [Google Scholar] [CrossRef] [PubMed]
- Islam, M.A.; Zilani, M.N.H.; Biswas, P.; Khan, D.A.; Rahman, M.H.; Nahid, R.; Nahar, N.; Samad, A.; Ahammad, F.; Hasan, M.N. Evaluation of in vitro and in silico anti-inflammatory potential of some selected medicinal plants of Bangladesh against cyclooxygenase-II enzyme. J. Ethnopharmacol. 2022, 285, 114900. [Google Scholar] [CrossRef] [PubMed]
- Biswas, P.; Dey, D.; Rahman, A.; Islam, M.A.; Susmi, T.F.; Kaium, M.A.; Hasan, M.N.; Rahman, M.D.H.; Mahmud, S.; Saleh, M.A.; et al. Analysis of SYK Gene as a Prognostic Biomarker and Suggested Potential Bioactive Phytochemicals as an Alternative Therapeutic Option for Colorectal Cancer: An In-Silico Pharmaco-Informatics Investigation. J. Pers. Med. 2021, 11, 888. [Google Scholar] [CrossRef]
- Hasan, M.M.; Zilani, M.N.H.; Akter, S.; Nasrin, P.; Shajib, G.M.A.; Islam, M.A.; Biswas, P.; Mahmud, S.; Saleh, M.A.; Hasan, M.N.; et al. UHPLC-Q/Orbitrap/MS based chemical fingerprinting and hepatoprotective potential of medicinal plant Morinda angustifolia Roxb. South Afr. J. Bot. 2022, 148, 561–572. [Google Scholar] [CrossRef]
- Karikas, G.A. Anticancer and chemopreventing natural products: Some biochemical and therapeutic aspects. J. Buon 2010, 15, 627–638. [Google Scholar] [PubMed]
- Biswas, P.; Hasan, M.M.; Dey, D.; Dos Santos Costa, A.C.; Polash, S.A.; Bibi, S.; Ferdous, N.; Kaium, M.A.; Rahman, M.D.H.; Jeet, F.K.; et al. Candidate antiviral drugs for COVID-19 and their environmental implications: A comprehensive analysis. Environ. Sci. Pollut. Res. Int. 2021, 28, 59570–59593. [Google Scholar] [CrossRef] [PubMed]
- Dey, D.; Ema, T.I.; Biswas, P.; Aktar, S.; Islam, S.; Rinik, U.R.; Firoz, M.; Ahmed, S.Z.; al Azad, S.; Rahman, A.; et al. Antiviral effects of bacteriocin against animal-to-human transmittable mutated SARS-COV-2: A systematic review. Front. Agr. Sci. Eng. 2021, 8, 603–622. [Google Scholar] [CrossRef]
- Hasan, A.; Biswas, P.; Bondhon, T.A.; Jannat, K.; Paul, T.K.; Paul, A.K.; Jahan, R.; Nissapatorn, V.; Mahboob, T.; Wilairatana, P.; et al. Can Artemisia herba-alba Be Useful for Managing COVID-19 and Comorbidities? Molecules 2022, 27, 492. [Google Scholar] [CrossRef]
- Bibi, S.; Hasan, M.M.; Biswas, P.; Shkodina, A.; Shah, M.A.; Shah, G.M.; Khan, A.; Al-Harrasi, A. Chapter 7—Phytonutrients in the management of lipids metabolism. In The Role of Phytonutrients in Metabolic Disorders; Khan, H., Akkol, E.K., Daglia, M., Eds.; Academic Press: Cambridge, MA, USA, 2022; pp. 195–236. [Google Scholar]
- Lin, P.; Tian, X.H.; Yi, Y.S.; Jiang, W.S.; Zhou, Y.J.; Cheng, W.J. Luteolin-induced protection of H2O2-induced apoptosis in PC12 cells and the associated pathway. Mol. Med. Rep. 2015, 12, 7699–7704. [Google Scholar] [CrossRef] [Green Version]
- Leung, H.W.; Kuo, C.L.; Yang, W.H.; Lin, C.H.; Lee, H.Z. Antioxidant enzymes activity involvement in luteolin-induced human lung squamous carcinoma CH27 cell apoptosis. Eur. J. Pharmacol. 2006, 534, 12–18. [Google Scholar] [CrossRef]
- Wu, T.H.; Yen, F.L.; Lin, L.T.; Tsai, T.R.; Lin, C.C.; Cham, T.M. Preparation, physicochemical characterization, and antioxidant effects of quercetin nanoparticles. Int. J. Pharm. 2008, 346, 160–168. [Google Scholar] [CrossRef]
- Zilani, M.N.H.; Islam, M.A.; Biswas, P.; Anisuzzman, M.; Hossain, H.; Shilpi, J.A.; Hasan, M.N.; Hossain, M.G. Metabolite profiling, anti-inflammatory, analgesic potentials of edible herb Colocasia gigantea and molecular docking study against COX-II enzyme. J. Ethnopharmacol. 2021, 281, 114577. [Google Scholar] [CrossRef]
- Dey, D.; Hossain, R.; Biswas, P.; Paul, P.; Islam, M.A.; Ema, T.I.; Gain, B.K.; Hasan, M.M.; Bibi, S.; Islam, M.T.; et al. Amentoflavone derivatives significantly act towards the main protease (3CL(PRO)/M(PRO)) of SARS-CoV-2: In silico admet profiling, molecular docking, molecular dynamics simulation, network pharmacology. Mol. Divers. 2022, 1–15. [Google Scholar] [CrossRef]
- Dey, D.; Biswas, P.; Paul, P.; Mahmud, S.; Ema, T.I.; Khan, A.A.; Ahmed, S.Z.; Hasan, M.M.; Saikat, A.S.M.; Fatema, B.; et al. Natural flavonoids effectively block the CD81 receptor of hepatocytes and inhibit HCV infection: A computational drug development approach. Mol. Divers. 2022, 1–14. [Google Scholar] [CrossRef]
- Paul, P.K.; Al Azad, S.; Rahman, M.H.; Farjana, M.; Uddin, M.R.; Dey, D.; Mahmud, S.; Ema, T.I.; Biswas, P.; Anjum, M.; et al. Catabolic profiling of selective enzymes in the saccharification of non-food lignocellulose parts of biomass into functional edible sugars and bioenergy: An in silico bioprospecting. J. Adv. Vet. Anim. Res. 2022, 9, 19–32. [Google Scholar] [CrossRef] [PubMed]
- Ferdausi, N.; Islam, S.; Rimti, F.H.; Quayum, S.T.; Arshad, E.M.; Ibnat, A.; Islam, T.; Arefin, A.; Ema, T.I.; Biswas, P.; et al. Point-specific interactions of isovitexin with the neighboring amino acid residues of the hACE2 receptor as a targeted therapeutic agent in suppressing the SARS-CoV-2 influx mechanism. J. Adv. Vet. Anim. Res. 2022, 9, 230–240. [Google Scholar] [CrossRef] [PubMed]
- Hossain, R.; Dey, D.; Biswas, P.; Paul, P.; Ahmed, S.Z.; Khan, A.A.; Ema, T.I.; Islam, M.T. Chlorophytum borivilianum (Musli) and Cimicifuga racemosa (Black Cohosh). In Herbs, Shrubs, and Trees of Potential Medicinal Benefits; CRC Press: Boca Raton, FL, USA, 2022; pp. 45–82. [Google Scholar]
- Hertog, M.G.; Hollman, P.C.; Katan, M.B.; Kromhout, D. Intake of potentially anticarcinogenic flavonoids and their determinants in adults in The Netherlands. Nutr. Cancer 1993, 20, 21–29. [Google Scholar] [CrossRef] [PubMed]
- Al Saber, M.; Biswas, P.; Dey, D.; Kaium, M.A.; Islam, M.A.; Tripty, M.I.A.; Rahman, M.H.; Rahaman, T.I.; Biswas, M.Y.; Paul, P.; et al. A Comprehensive Review of Recent Advancements in Cancer Immunotherapy and Generation of CAR T Cell by CRISPR-Cas9. Processes 2022, 10, 16. [Google Scholar] [CrossRef]
- Baral, S.K.; Biswas, P.; Kaium, M.A.; Islam, M.A.; Dey, D.; Saber, M.A.; Rahaman, T.I.; M, A.; Emran, T.B.; Hasan, M.N.; et al. A Comprehensive Discussion in Vaginal Cancer Based on Mechanisms, Treatments, Risk Factors and Prevention. Front. Oncol. 2022, 12, 883805. [Google Scholar] [CrossRef]
- Jeong, J.H.; An, J.Y.; Kwon, Y.T.; Rhee, J.G.; Lee, Y.J. Effects of low dose quercetin: Cancer cell-specific inhibition of cell cycle progression. J. Cell Biochem. 2009, 106, 73–82. [Google Scholar] [CrossRef] [Green Version]
- Lee, T.-J.; Kim, O.H.; Kim, Y.H.; Lim, J.H.; Kim, S.; Park, J.-W.; Kwon, T.K. Quercetin arrests G2/M phase and induces caspase-dependent cell death in U937 cells. Cancer Lett. 2006, 240, 234–242. [Google Scholar] [CrossRef]
- Ong, C.S.; Tran, E.; Nguyen, T.T.; Ong, C.K.; Lee, S.K.; Lee, J.J.; Ng, C.P.; Leong, C.; Huynh, H.J. Quercetin-induced growth inhibition and cell death in nasopharyngeal carcinoma cells are associated with increase in Bad and hypophosphorylated retinoblastoma expressions. Oncol. Rep. 2004, 11, 727–733. [Google Scholar] [CrossRef]
- Kachadourian, R.; Day, B.J. Flavonoid-induced glutathione depletion: Potential implications for cancer treatment. Free Radic. Biol. Med. 2006, 41, 65–76. [Google Scholar] [CrossRef] [Green Version]
- Ramos, A.M.; Aller, P. Quercetin decreases intracellular GSH content and potentiates the apoptotic action of the antileukemic drug arsenic trioxide in human leukemia cell lines. Biochem. Pharmacol. 2008, 75, 1912–1923. [Google Scholar] [CrossRef] [Green Version]
- Lugli, E.; Ferraresi, R.; Roat, E.; Troiano, L.; Pinti, M.; Nasi, M.; Nemes, E.; Bertoncelli, L.; Gibellini, L.; Salomoni, P.; et al. Quercetin inhibits lymphocyte activation and proliferation without inducing apoptosis in peripheral mononuclear cells. Leuk. Res. 2009, 33, 140–150. [Google Scholar] [CrossRef] [PubMed]
- Apeh, V.O.; Asogwa, E.; Chukwuma, F.I.; Okonkwo, O.F.; Nwora, F.; Uke, R. Chemical analysis and in silico anticancer and anti-inflammatory potentials of bioactive compounds from Moringaoleifera seed oil. Adv. Tradit. Med. 2022, 22, 59–74. [Google Scholar] [CrossRef]
- Yoshizumi, M.; Tsuchiya, K.; Kirima, K.; Kyaw, M.; Suzaki, Y.; Tamaki, T.J.M.P. Quercetin inhibits Shc-and phosphatidylinositol 3-kinase-mediated c-Jun N-terminal kinase activation by angiotensin II in cultured rat aortic smooth muscle cells. Mol. Pharmacol. 2001, 60, 656–665. [Google Scholar] [PubMed]
- Vulcain, E.; Goupy, P.; Caris-Veyrat, C.; Dangles, O.J. Inhibition of the metmyoglobin-induced peroxidation of linoleic acid by dietary antioxidants: Action in the aqueous vs. lipid phase. Free. Radic. Res. 2005, 39, 547–563. [Google Scholar] [CrossRef] [PubMed]
- Chien, S.Y.; Wu, Y.C.; Chung, J.G.; Yang, J.S.; Lu, H.F.; Tsou, M.F.; Wood, W.G.; Kuo, S.J.; Chen, D.R. Quercetin-induced apoptosis acts through mitochondrial- and caspase-3-dependent pathways in human breast cancer MDA-MB-231 cells. Hum Exp. Toxicol. 2009, 28, 493–503. [Google Scholar] [CrossRef] [PubMed]
- Yang, J.H.; Hsia, T.C.; Kuo, H.M.; Chao, P.D.; Chou, C.C.; Wei, Y.H.; Chung, J.G. Inhibition of lung cancer cell growth by quercetin glucuronides via G2/M arrest and induction of apoptosis. Drug Metab. Dispos. 2006, 34, 296–304. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Barbieri, D.; Abbracchio, M.P.; Salvioli, S.; Monti, D.; Cossarizza, A.; Ceruti, S.; Brambilla, R.; Cattabeni, F.; Jacobson, K.A.; Franceschi, C. Apoptosis by 2-chloro-2′-deoxy-adenosine and 2-chloro-adenosine in human peripheral blood mononuclear cells. Neurochem. Int. 1998, 32, 493–504. [Google Scholar] [CrossRef]
- Monti, D.; Salvioli, S.; Capri, M.; Malorni, W.; Straface, E.; Cossarizza, A.; Botti, B.; Piacentini, M.; Baggio, G.; Barbi, C.; et al. Decreased susceptibility to oxidative stress-induced apoptosis of peripheral blood mononuclear cells from healthy elderly and centenarians. Mech. Ageing Dev. 2000, 121, 239–250. [Google Scholar] [CrossRef]
- Ye, B.; Yang, J.L.; Chen, L.J.; Wu, X.X.; Yang, H.S.; Zhao, J.M.; Yuan, Z.P.; Li, J.; Wen, Y.J.; Mao, Y.Q.; et al. Induction of apoptosis by phenylisocyanate derivative of quercetin: Involvement of heat shock protein. Anticancer. Drugs 2007, 18, 1165–1171. [Google Scholar] [CrossRef]
- Al Azad, S.; Ahmed, S.; Biswas, P.; Mia, M.A.R.; Farjana, M.; Arshe, F.A.; Mily, S.J.; Ankhi, A.B.; Shaikat, M.M.; Sultana, S. Quantitative analysis of the factors influencing IDA and TSH downregulation in correlation to the fluctuation of activated vitamin D3 in women. J. Adv. Biotechnol. Exp. Ther. 2022, 5, 320. [Google Scholar] [CrossRef]
- NavaneethaKrishnan, S.; Rosales, J.L.; Lee, K.Y. ROS-Mediated Cancer Cell Killing through Dietary Phytochemicals. Oxid. Med. Cell. Longev. 2019, 2019, 9051542. [Google Scholar] [CrossRef] [PubMed]
- Kamaraj, S.; Vinodhkumar, R.; Anandakumar, P.; Jagan, S.; Ramakrishnan, G.; Devaki, T. The effects of quercetin on antioxidant status and tumor markers in the lung and serum of mice treated with benzo(a)pyrene. Biol. Pharm. Bull. 2007, 30, 2268–2273. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Seufi, A.M.; Ibrahim, S.S.; Elmaghraby, T.K.; Hafez, E.E. Preventive effect of the flavonoid, quercetin, on hepatic cancer in rats via oxidant/antioxidant activity: Molecular and histological evidences. J. Exp. Clin. Cancer Res. 2009, 28, 80. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Volate, S.R.; Davenport, D.M.; Muga, S.J.; Wargovich, M.J. Modulation of aberrant crypt foci and apoptosis by dietary herbal supplements (quercetin, curcumin, silymarin, ginseng and rutin). Carcinogenesis 2005, 26, 1450–1456. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Tang, S.M.; Deng, X.T.; Zhou, J.; Li, Q.P.; Ge, X.X.; Miao, L. Pharmacological basis and new insights of quercetin action in respect to its anti-cancer effects. Biomed Pharm. 2020, 121, 109604. [Google Scholar] [CrossRef]
- Harwood, M.; Danielewska-Nikiel, B.; Borzelleca, J.F.; Flamm, G.W.; Williams, G.M.; Lines, T.C. A critical review of the data related to the safety of quercetin and lack of evidence of in vivo toxicity, including lack of genotoxic/carcinogenic properties. Food Chem. Toxicol. 2007, 45, 2179–2205. [Google Scholar] [CrossRef]
- Nguyen, T.; Tran, E.; Nguyen, T.; Do, P.; Huynh, T.; Huynh, H.J.C. The role of activated MEK-ERK pathway in quercetin-induced growth inhibition and apoptosis in A549 lung cancer cells. Carcinogenesis 2004, 25, 647–659. [Google Scholar] [CrossRef]
- Vidya Priyadarsini, R.; Senthil Murugan, R.; Maitreyi, S.; Ramalingam, K.; Karunagaran, D.; Nagini, S. The flavonoid quercetin induces cell cycle arrest and mitochondria-mediated apoptosis in human cervical cancer (HeLa) cells through p53 induction and NF-κB inhibition. Eur. J. Pharmacol. 2010, 649, 84–91. [Google Scholar] [CrossRef]
- Chan, S.T.; Yang, N.C.; Huang, C.S.; Liao, J.W.; Yeh, S.L. Quercetin enhances the antitumor activity of trichostatin A through upregulation of p53 protein expression in vitro and in vivo. PLoS ONE 2013, 8, e54255. [Google Scholar] [CrossRef] [Green Version]
- Liao, Y.-R.; Lin, J.-Y.J. Quercetin, but not its metabolite quercetin-3-glucuronide, exerts prophylactic immunostimulatory activity and therapeutic antiinflammatory effects on lipopolysaccharide-treated mouse peritoneal macrophages ex vivo. J. Agric. Food Chem. 2014, 62, 2872–2880. [Google Scholar] [CrossRef]
- Rauf, A.; Imran, M.; Khan, I.A.; Ur-Rehman, M.; Gilani, S.A.; Mehmood, Z.; Mubarak, M.S. Anticancer potential of quercetin: A comprehensive review. Phytother. Res. 2018, 32, 2109–2130. [Google Scholar] [CrossRef] [PubMed]
- Williams, C.A.; Grayer, R.J. Anthocyanins and other flavonoids. Nat. Prod. Rep. 2004, 21, 539–573. [Google Scholar] [CrossRef] [PubMed]
- Khan, F.; Niaz, K.; Maqbool, F.; Ismail Hassan, F.; Abdollahi, M.; Nagulapalli Venkata, K.C.; Nabavi, S.M.; Bishayee, A. Molecular Targets Underlying the Anticancer Effects of Quercetin: An Update. Nutrients 2016, 8, 529. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Murota, K.; Shimizu, S.; Miyamoto, S.; Izumi, T.; Obata, A.; Kikuchi, M.; Terao, J. Unique uptake and transport of isoflavone aglycones by human intestinal caco-2 cells: Comparison of isoflavonoids and flavonoids. J. Nutr. 2002, 132, 1956–1961. [Google Scholar] [CrossRef] [Green Version]
- Iacopetta, D.; Grande, F.; Caruso, A.; Mordocco, R.A.; Plutino, M.R.; Scrivano, L.; Ceramella, J.; Muià, N.; Saturnino, C.; Puoci, F.; et al. New insights for the use of quercetin analogs in cancer treatment. Future Med. Chem. 2017, 9, 2011–2028. [Google Scholar] [CrossRef]
- Liu, Y.; Tang, Z.G.; Lin, Y.; Qu, X.G.; Lv, W.; Wang, G.B.; Li, C.L. Effects of quercetin on proliferation and migration of human glioblastoma U251 cells. Biomed Pharm. 2017, 92, 33–38. [Google Scholar] [CrossRef]
- Murakami, A.; Ashida, H.; Terao, J. Multitargeted cancer prevention by quercetin. Cancer Lett. 2008, 269, 315–325. [Google Scholar] [CrossRef]
- Shih, H.; Pickwell, G.V.; Quattrochi, L.C. Differential effects of flavonoid compounds on tumor promoter-induced activation of the human CYP1A2 enhancer. Arch Biochem. Biophys. 2000, 373, 287–294. [Google Scholar] [CrossRef]
- Brito, A.F.; Ribeiro, M.; Abrantes, A.M.; Pires, A.S.; Teixo, R.J.; Tralhão, J.G.; Botelho, M.F. Quercetin in Cancer Treatment, Alone or in Combination with Conventional Therapeutics? Curr. Med. Chem. 2015, 22, 3025–3039. [Google Scholar] [CrossRef] [Green Version]
- Phaniendra, A.; Jestadi, D.B.; Periyasamy, L.J. Free radicals: Properties, sources, targets, and their implication in various diseases. Indian J. Clin. Biochem. 2015, 30, 11–26. [Google Scholar] [CrossRef] [Green Version]
- Sabharwal, S.S.; Schumacker, P.T. Mitochondrial ROS in cancer: Initiators, amplifiers or an Achilles’ heel? Nat. Rev. Cancer 2014, 14, 709–721. [Google Scholar] [CrossRef] [PubMed]
- Brown, D.I.; Griendling, K.K. Regulation of signal transduction by reactive oxygen species in the cardiovascular system. Circ. Res. 2015, 116, 531–549. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Mirończuk-Chodakowska, I.; Witkowska, A.M.; Zujko, M.E. Endogenous non-enzymatic antioxidants in the human body. Adv. Med. Sci. 2018, 63, 68–78. [Google Scholar] [CrossRef] [PubMed]
- D’Autréaux, B.; Toledano, M.B. ROS as signalling molecules: Mechanisms that generate specificity in ROS homeostasis. Nat. Rev. Mol. Cell Biol. 2007, 8, 813–824. [Google Scholar] [CrossRef] [PubMed]
- Outten, F.W. Iron-sulfur clusters as oxygen-responsive molecular switches. Nat Chem. Biol. 2007, 3, 206–207. [Google Scholar] [CrossRef]
- Diebold, L.; Chandel, N.S. Mitochondrial ROS regulation of proliferating cells. Free Radic. Biol. Med. 2016, 100, 86–93. [Google Scholar] [CrossRef]
- Murphy, M.P. How mitochondria produce reactive oxygen species. Biochem. J. 2009, 417, 1–13. [Google Scholar] [CrossRef] [Green Version]
- Echtay, K.S.; Murphy, M.P.; Smith, R.A.; Talbot, D.A.; Brand, M.D. Superoxide activates mitochondrial uncoupling protein 2 from the matrix side. Studies using targeted antioxidants. J. Biol. Chem. 2002, 277, 47129–47135. [Google Scholar] [CrossRef] [Green Version]
- Martínez-Reyes, I.; Diebold, L.P.; Kong, H.; Schieber, M.; Huang, H.; Hensley, C.T.; Mehta, M.M.; Wang, T.; Santos, J.H.; Woychik, R.; et al. TCA Cycle and Mitochondrial Membrane Potential Are Necessary for Diverse Biological Functions. Mol. Cell 2016, 61, 199–209. [Google Scholar] [CrossRef] [Green Version]
- Shi, X.; Zhang, Y.; Zheng, J.; Pan, J. Reactive oxygen species in cancer stem cells. Antioxid. Redox Signal. 2012, 16, 1215–1228. [Google Scholar] [CrossRef] [Green Version]
- Pervaiz, S.; Taneja, R.; Ghaffari, S. Oxidative stress regulation of stem and progenitor cells. Antioxid. Redox Signal. 2009, 11, 2777–2789. [Google Scholar] [CrossRef] [PubMed]
- Diehn, M.; Cho, R.W.; Lobo, N.A.; Kalisky, T.; Dorie, M.J.; Kulp, A.N.; Qian, D.; Lam, J.S.; Ailles, L.E.; Wong, M.; et al. Association of reactive oxygen species levels and radioresistance in cancer stem cells. Nature 2009, 458, 780–783. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Chen, W.; Druhan, L.J.; Chen, C.-A.; Hemann, C.; Chen, Y.-R.; Berka, V.; Tsai, A.-L.; Zweier, J.L. Peroxynitrite induces destruction of the tetrahydrobiopterin and heme in endothelial nitric oxide synthase: Transition from reversible to irreversible enzyme inhibition. Biochemistry 2010, 49, 3129–3137. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Ghanbari Movahed, Z.; Rastegari-Pouyani, M.; Mohammadi, M.H.; Mansouri, K. Cancer cells change their glucose metabolism to overcome increased ROS: One step from cancer cell to cancer stem cell? Biomed. Pharmacother. 2019, 112, 108690. [Google Scholar] [CrossRef] [PubMed]
- Masoud, G.N.; Li, W. HIF-1α pathway: Role, regulation and intervention for cancer therapy. Acta Pharm. Sinica. B 2015, 5, 378–389. [Google Scholar] [CrossRef] [Green Version]
- Jung, S.N.; Yang, W.K.; Kim, J.; Kim, H.S.; Kim, E.J.; Yun, H.; Park, H.; Kim, S.S.; Choe, W.; Kang, I.; et al. Reactive oxygen species stabilize hypoxia-inducible factor-1 alpha protein and stimulate transcriptional activity via AMP-activated protein kinase in DU145 human prostate cancer cells. Carcinogenesis 2008, 29, 713–721. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Jiang, P.; Du, W.; Wu, M. Regulation of the pentose phosphate pathway in cancer. Protein Cell 2014, 5, 592–602. [Google Scholar] [CrossRef] [Green Version]
- Reya, T.; Morrison, S.J.; Clarke, M.F.; Weissman, I.L. Stem cells, cancer, and cancer stem cells. Nature 2001, 414, 105–111. [Google Scholar] [CrossRef] [Green Version]
- Ghasemi, F.; Sarabi, P.Z.; Athari, S.S.; Esmaeilzadeh, A. Therapeutics strategies against cancer stem cell in breast cancer. Int. J. Biochem. Cell Biol. 2019, 109, 76–81. [Google Scholar] [CrossRef]
- Dragu, D.L.; Necula, L.G.; Bleotu, C.; Diaconu, C.C.; Chivu-Economescu, M. Therapies targeting cancer stem cells: Current trends and future challenges. World J. Stem Cells 2015, 7, 1185–1201. [Google Scholar] [CrossRef]
- Patra, K.C.; Hay, N. The pentose phosphate pathway and cancer. Trends Biochem. Sci. 2014, 39, 347–354. [Google Scholar] [CrossRef] [PubMed]
- Schieber, M.; Chandel, N.S. ROS function in redox signaling and oxidative stress. Curr. Biol. 2014, 24, R453–R462. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Kumari, S.; Badana, A.K.; Malla, R. Reactive oxygen species: A key constituent in cancer survival. Biomark. Insights 2018, 13, 1177271918755391. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Liu, B.; Chen, Y.; Clair, D.K.S. ROS and p53: A versatile partnership. Free Radic. Biol. Med. 2008, 44, 1529–1535. [Google Scholar] [CrossRef] [Green Version]
- Moloney, J.N.; Cotter, T.G. ROS signalling in the biology of cancer. In Seminars in Cell Developmental Biology; Academic Press: Cambridge, MA, USA, 2018; pp. 50–64. [Google Scholar]
- Zhang, J.; Wang, X.; Vikash, V.; Ye, Q.; Wu, D.; Liu, Y.; Dong, W. ROS and ROS-mediated cellular signaling. Oxidative Med. Cell. Longev. 2016, 2016, 4350965. [Google Scholar] [CrossRef] [Green Version]
- Lu, Z.; Xu, S. ERK1/2 MAP kinases in cell survival and apoptosis. IUBMB Life 2006, 58, 621–631. [Google Scholar] [CrossRef]
- Dhanasekaran, D.N.; Reddy, E.P. JNK signaling in apoptosis. Oncogene 2008, 27, 6245–6251. [Google Scholar] [CrossRef] [Green Version]
- Mincheva-Tasheva, S.; Soler, R.M. NF-κB signaling pathways: Role in nervous system physiology and pathology. Neurosci. 2013, 19, 175–194. [Google Scholar] [CrossRef] [Green Version]
- Manning, B.D.; Cantley, L.C. AKT/PKB signaling: Navigating downstream. Cell 2007, 129, 1261–1274. [Google Scholar] [CrossRef] [Green Version]
- NavaneethaKrishnan, S.; Rosales, J.L.; Lee, K.-Y. Loss of Cdk5 in breast cancer cells promotes ROS-mediated cell death through dysregulation of the mitochondrial permeability transition pore. Oncogene 2018, 37, 1788–1804. [Google Scholar] [CrossRef] [Green Version]
- Youle, R.J.; Strasser, A. The BCL-2 protein family: Opposing activities that mediate cell death. Nat. Rev. Mol. Cell Biol. 2008, 9, 47–59. [Google Scholar] [CrossRef] [PubMed]
- Lee, D.H.; Rhee, J.G.; Lee, Y.J. Reactive oxygen species up-regulate p53 and Puma; a possible mechanism for apoptosis during combined treatment with TRAIL and wogonin. Br. J. Pharmacol. 2009, 157, 1189–1202. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Yoshida, K.; Miki, Y. The cell death machinery governed by the p53 tumor suppressor in response to DNA damage. Cancer Sci. 2010, 101, 831–835. [Google Scholar] [CrossRef] [PubMed]
- Hempel, N.; Trebak, M. Crosstalk between calcium and reactive oxygen species signaling in cancer. Cell 2017, 63, 70–96. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Lee, Y.-K.; Hwang, J.-T.; Kwon, D.Y.; Surh, Y.-J.; Park, O.J. Induction of apoptosis by quercetin is mediated through AMPKα1/ASK1/p38 pathway. Cancer Lett. 2010, 292, 228–236. [Google Scholar] [CrossRef]
- Hattori, K.; Naguro, I.; Runchel, C.; Ichijo, H. The roles of ASK family proteins in stress responses and diseases. Cell Commun. Signal. 2009, 7, 9. [Google Scholar] [CrossRef] [Green Version]
- Matsuzawa, A.; Ichijo, H.J.B. Redox control of cell fate by MAP kinase: Physiological roles of ASK1-MAP kinase pathway in stress signaling. Biochim. Biophys. Acta (BBA)-Gen. Subj. 2008, 1780, 1325–1336. [Google Scholar] [CrossRef]
- Lee, Y.-K.; Park, S.Y.; Kim, Y.-M.; Lee, W.S.; Park, O.J. AMP kinase/cyclooxygenase-2 pathway regulates proliferation and apoptosis of cancer cells treated with quercetin. Exp. Mol. Med. 2009, 41, 201–207. [Google Scholar] [CrossRef]
- Pelicano, H.; Carney, D.; Huang, P. ROS stress in cancer cells and therapeutic implications. Drug Resist. Updates 2004, 7, 97–110. [Google Scholar] [CrossRef]
- Schumacker, P.T. Reactive oxygen species in cancer cells: Live by the sword, die by the sword. Cancer Cell 2006, 10, 175–176. [Google Scholar] [CrossRef] [Green Version]
- Ward, A.B.; Mir, H.; Kapur, N.; Gales, D.N.; Carriere, P.P.; Singh, S. Quercetin inhibits prostate cancer by attenuating cell survival and inhibiting anti-apoptotic pathways. World J. Surg. Oncol. 2018, 16, 108. [Google Scholar] [CrossRef] [PubMed]
- Cossarizza, A.; Gibellini, L.; Pinti, M.; Nasi, M.; Montagna, J.; De Biasi, S.; Roat, E.; Bertoncelli, L.; Cooper, E. Quercetin and cancer chemoprevention. Evid. Based Complementary Altern. Med. 2011, 2011, 591356. [Google Scholar]
- Xu, D.; Hu, M.-J.; Wang, Y.-Q.; Cui, Y.-L. Antioxidant activities of quercetin and its complexes for medicinal application. Molecules 2019, 24, 1123. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Zhang, X.; Huang, J.; Yu, C.; Xiang, L.; Li, L.; Shi, D.; Lin, F. Quercetin enhanced paclitaxel therapeutic effects towards PC-3 prostate cancer through ER stress induction and ROS production. OncoTargets Ther. 2020, 13, 513. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Zhang, H.; Zhang, M.; Yu, L.; Zhao, Y.; He, N.; Yang, X. Antitumor activities of quercetin and quercetin-5′, 8-disulfonate in human colon and breast cancer cell lines. Food Chem. Toxicol. 2012, 50, 1589–1599. [Google Scholar] [CrossRef] [PubMed]
- Ferraresi, R.; Troiano, L.; Roat, E.; Lugli, E.; Nemes, E.; Nasi, M.; Pinti, M.; Fernandez, M.I.G.; Cooper, E.L.; Cossarizza, A.J.F. Essential requirement of reduced glutathione (GSH) for the anti-oxidant effect of the flavonoid quercetin. Free. Radic. Res. 2005, 39, 1249–1258. [Google Scholar] [CrossRef]
- Metodiewa, D.; Jaiswal, A.K.; Cenas, N.; Dickancaité, E.; Segura-Aguilar, J. Quercetin may act as a cytotoxic prooxidant after its metabolic activation to semiquinone and quinoidal product. Free. Radic. Biol. Med. 1999, 26, 107–116. [Google Scholar]
- Boots, A.W.; Kubben, N.; Haenen, G.R.; Bast, A. Oxidized quercetin reacts with thiols rather than with ascorbate: Implication for quercetin supplementation. Biochem. Biophys. Res. Commun. 2003, 308, 560–565. [Google Scholar] [CrossRef]
- Sahu, S.C.; Gray, G.C. Pro-oxidant activity of flavonoids: Effects on glutathione and glutathione S-transferase in isolated rat liver nuclei. Cancer Lett. 1996, 104, 193–196. [Google Scholar] [CrossRef]
- Lugli, E.; Troiano, L.; Ferraresi, R.; Roat, E.; Prada, N.; Nasi, M.; Pinti, M.; Cooper, E.L.; Cossarizza, A. Characterization of cells with different mitochondrial membrane potential during apoptosis. Cytom. Part A 2005, 68, 28–35. [Google Scholar] [CrossRef]
- Troiano, L.; Ferraresi, R.; Lugli, E.; Nemes, E.; Roat, E.; Nasi, M.; Pinti, M.; Cossarizza, A. Multiparametric analysis of cells with different mitochondrial membrane potential during apoptosis by polychromatic flow cytometry. Nat. Protoc. 2007, 2, 2719. [Google Scholar] [CrossRef] [PubMed]
- Lo, J.; Wang, H.-F.; Tam, M.; Lee, T.C. Glutathione S-transferase π in an arsenic-resistant Chinese hamster ovary cell line. Biochem. J. 1992, 288, 977–982. [Google Scholar] [CrossRef] [PubMed]
- Boots, A.W.; Haenen, G.R.; Hartog, G.J.D.; Bast, A. Oxidative damage shifts from lipid peroxidation to thiol arylation by catechol-containing antioxidants. Biochim. Biophys. Acta (BBA) Mol. Cell Biol. Lipids 2002, 1583, 279–284. [Google Scholar] [CrossRef]
- Chen, X. Protective effects of quercetin on liver injury induced by ethanol. Pharmacogn. Mag. 2010, 6, 135. [Google Scholar] [CrossRef] [Green Version]
- Kashyap, D.; Mittal, S.; Sak, K.; Singhal, P.; Tuli, H.S. Molecular mechanisms of action of quercetin in cancer: Recent advances. Tumor Biol. 2016, 37, 12927–12939. [Google Scholar] [CrossRef]
- Kumar, S.; Pandey, A.K.J. Chemistry and biological activities of flavonoids: An overview. Sci. World J. 2013, 2013, 162750. [Google Scholar] [CrossRef] [Green Version]
- Lautraite, S.; Musonda, A.; Doehmer, J.; Edwards, G.; Chipman, J.J.M. Flavonoids inhibit genetic toxicity produced by carcinogens in cells expressing CYP1A2 and CYP1A1. Mutagenesis 2002, 17, 45–53. [Google Scholar] [CrossRef] [Green Version]
- Tan, W.-f.; Lin, L.-p.; Li, M.-h.; Zhang, Y.-X.; Tong, Y.-g.; Xiao, D.; Ding, J. Quercetin, a dietary-derived flavonoid, possesses antiangiogenic potential. Eur. J. Pharmacol. 2003, 459, 255–262. [Google Scholar] [CrossRef]
- Vijayababu, M.; Arunkumar, A.; Kanagaraj, P.; Venkataraman, P.; Krishnamoorthy, G.; Arunakaran, J. Quercetin downregulates matrix metalloproteinases 2 and 9 proteins expression in prostate cancer cells (PC-3). Mol. Cell. Biochem. 2006, 287, 109–116. [Google Scholar] [CrossRef]
- Cho, S.-Y.; Park, S.-J.; Kwon, M.-J.; Jeong, T.-S.; Bok, S.-H.; Choi, W.-Y.; Jeong, W.-I.; Ryu, S.-Y.; Do, S.-H.; Lee, C.-S.J.M.; et al. Quercetin suppresses proinflammatory cytokines production through MAP kinases and NF-κB pathway in lipopolysaccharide-stimulated macrophage. Mol. Cell. Biochem. 2003, 243, 153–160. [Google Scholar] [CrossRef]
- Raman, M.; Chen, W.; Cobb, M.J.O. Differential regulation and properties of MAPKs. Oncogene 2007, 26, 3100–3112. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Lee, W.J.; Hsiao, M.; Chang, J.L.; Yang, S.F.; Tseng, T.H.; Cheng, C.W.; Chow, J.M.; Lin, K.H.; Lin, Y.W.; Liu, C.C.; et al. Quercetin induces mitochondrial-derived apoptosis via reactive oxygen species-mediated ERK activation in HL-60 leukemia cells and xenograft. Arch. Toxicol. 2015, 89, 1103–1117. [Google Scholar] [CrossRef]
- Niu, G.; Yin, S.; Xie, S.; Li, Y.; Nie, D.; Ma, L.; Wang, X.; Wu, Y. Quercetin induces apoptosis by activating caspase-3 and regulating Bcl-2 and cyclooxygenase-2 pathways in human HL-60 cells. Acta Biochim. Biophys. Sin. 2011, 43, 30–37. [Google Scholar] [CrossRef]
- Mandal, A.K.; Das, S.; Mitra, M.; Chakrabarti, R.N.; Chatterjee, M.; Das, N. Vesicular flavonoid in combating diethylnitrosamine induced hepatocarcinoma in rat model. J. Exp. Ther. Oncol. 2008, 7, 123–133. [Google Scholar] [PubMed]
- Zhou, J.; Fang, L.; Liao, J.; Li, L.; Yao, W.; Xiong, Z.; Zhou, X. Investigation of the anti-cancer effect of quercetin on HepG2 cells in vivo. PLoS ONE 2017, 12, e0172838. [Google Scholar] [CrossRef] [Green Version]
- Howells, L.M.; Britton, R.G.; Mazzoletti, M.; Greaves, P.; Broggini, M.; Brown, K.; Steward, W.P.; Gescher, A.J.; Sale, S. Preclinical colorectal cancer chemopreventive efficacy and p53-modulating activity of 3′,4′,5′-trimethoxyflavonol, a quercetin analogue. Cancer Prev. Res. 2010, 3, 929–939. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Del Follo-Martinez, A.; Banerjee, N.; Li, X.; Safe, S.; Mertens-Talcott, S. Resveratrol and quercetin in combination have anticancer activity in colon cancer cells and repress oncogenic microRNA-27a. Nutr. Cancer 2013, 65, 494–504. [Google Scholar] [CrossRef]
- Shan, B.E.; Wang, M.X.; Li, R.Q. Quercetin inhibit human SW480 colon cancer growth in association with inhibition of cyclin D1 and survivin expression through Wnt/beta-catenin signaling pathway. Cancer Investig. 2009, 27, 604–612. [Google Scholar] [CrossRef]
- Dihal, A.A.; de Boer, V.C.; van der Woude, H.; Tilburgs, C.; Bruijntjes, J.P.; Alink, G.M.; Rietjens, I.M.; Woutersen, R.A.; Stierum, R.H. Quercetin, but not its glycosidated conjugate rutin, inhibits azoxymethane-induced colorectal carcinogenesis in F344 rats. J. Nutr. 2006, 136, 2862–2867. [Google Scholar] [CrossRef] [Green Version]
- Chen, W.; Wang, X.; Zhuang, J.; Zhang, L.; Lin, Y. Induction of death receptor 5 and suppression of survivin contribute to sensitization of TRAIL-induced cytotoxicity by quercetin in non-small cell lung cancer cells. Carcinogenesis 2007, 28, 2114–2121. [Google Scholar] [CrossRef] [Green Version]
- Youn, H.; Jeong, J.C.; Jeong, Y.S.; Kim, E.J.; Um, S.J. Quercetin potentiates apoptosis by inhibiting nuclear factor-kappaB signaling in H460 lung cancer cells. Biol. Pharm. Bull. 2013, 36, 944–951. [Google Scholar] [CrossRef] [Green Version]
- Yeh, S.L.; Yeh, C.L.; Chan, S.T.; Chuang, C.H. Plasma rich in quercetin metabolites induces G2/M arrest by upregulating PPAR-γ expression in human A549 lung cancer cells. Planta Med. 2011, 77, 992–998. [Google Scholar] [CrossRef] [PubMed]
- Gao, X.; Wang, B.; Wei, X.; Men, K.; Zheng, F.; Zhou, Y.; Zheng, Y.; Gou, M.; Huang, M.; Guo, G.; et al. Anticancer effect and mechanism of polymer micelle-encapsulated quercetin on ovarian cancer. Nanoscale 2012, 4, 7021–7030. [Google Scholar] [CrossRef] [PubMed]
- Catanzaro, D.; Ragazzi, E.; Vianello, C.; Caparrotta, L.; Montopoli, M. Effect of Quercetin on Cell Cycle and Cyclin Expression in Ovarian Carcinoma and Osteosarcoma Cell Lines. Nat. Prod. Commun. 2015, 10, 1365–1368. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Duo, J.; Ying, G.G.; Wang, G.W.; Zhang, L. Quercetin inhibits human breast cancer cell proliferation and induces apoptosis via Bcl-2 and Bax regulation. Mol. Med. Rep. 2012, 5, 1453–1456. [Google Scholar] [CrossRef] [PubMed]
- Srinivasan, A.; Thangavel, C.; Liu, Y.; Shoyele, S.; Den, R.B.; Selvakumar, P.; Lakshmikuttyamma, A. Quercetin regulates β-catenin signaling and reduces the migration of triple negative breast cancer. Mol. Carcinog. 2016, 55, 743–756. [Google Scholar] [CrossRef]
- Cao, L.; Yang, Y.; Ye, Z.; Lin, B.; Zeng, J.; Li, C.; Liang, T.; Zhou, K.; Li, J. Quercetin-3-methyl ether suppresses human breast cancer stem cell formation by inhibiting the Notch1 and PI3K/Akt signaling pathways. Int. J. Mol. Med. 2018, 42, 1625–1636. [Google Scholar] [CrossRef] [Green Version]
- Iriti, M.; Kubina, R.; Cochis, A.; Sorrentino, R.; Varoni, E.M.; Kabała-Dzik, A.; Azzimonti, B.; Dziedzic, A.; Rimondini, L.; Wojtyczka, R.D. Rutin, a Quercetin Glycoside, Restores Chemosensitivity in Human Breast Cancer Cells. Phytother. Res. 2017, 31, 1529–1538. [Google Scholar] [CrossRef]
- Shen, X.; Si, Y.; Wang, Z.; Wang, J.; Guo, Y.; Zhang, X. Quercetin inhibits the growth of human gastric cancer stem cells by inducing mitochondrial-dependent apoptosis through the inhibition of PI3K/Akt signaling. Int. J. Mol. Med. 2016, 38, 619–626. [Google Scholar] [CrossRef] [Green Version]
- Yu, D.; Ye, T.; Xiang, Y.; Shi, Z.; Zhang, J.; Lou, B.; Zhang, F.; Chen, B.; Zhou, M. Quercetin inhibits epithelial-mesenchymal transition, decreases invasiveness and metastasis, and reverses IL-6 induced epithelial-mesenchymal transition, expression of MMP by inhibiting STAT3 signaling in pancreatic cancer cells. OncoTargets Ther. 2017, 10, 4719–4729. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Yang, F.; Jiang, X.; Song, L.; Wang, H.; Mei, Z.; Xu, Z.; Xing, N. Quercetin inhibits angiogenesis through thrombospondin-1 upregulation to antagonize human prostate cancer PC-3 cell growth in vitro and in vivo. Oncol. Rep. 2016, 35, 1602–1610. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Atashpour, S.; Fouladdel, S.; Movahhed, T.K.; Barzegar, E.; Ghahremani, M.H.; Ostad, S.N.; Azizi, E. Quercetin induces cell cycle arrest and apoptosis in CD133(+) cancer stem cells of human colorectal HT29 cancer cell line and enhances anticancer effects of doxorubicin. Iran. J. Basic Med. Sci. 2015, 18, 635–643. [Google Scholar] [PubMed]
- Wang, R.; Yang, L.; Li, S.; Ye, D.; Yang, L.; Liu, Q.; Zhao, Z.; Cai, Q.; Tan, J.; Li, X. Quercetin Inhibits Breast Cancer Stem Cells via Downregulation of Aldehyde Dehydrogenase 1A1 (ALDH1A1), Chemokine Receptor Type 4 (CXCR4), Mucin 1 (MUC1), and Epithelial Cell Adhesion Molecule (EpCAM). Med. Sci. Monit. 2018, 24, 412–420. [Google Scholar] [CrossRef] [PubMed]
- Tang, S.N.; Singh, C.; Nall, D.; Meeker, D.; Shankar, S.; Srivastava, R.K. The dietary bioflavonoid quercetin synergizes with epigallocathechin gallate (EGCG) to inhibit prostate cancer stem cell characteristics, invasion, migration and epithelial-mesenchymal transition. J. Mol. Signal. 2010, 5, 14. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Crans, D.C.; Yang, L.; Haase, A.; Yang, X. Health benefits of vanadium and its potential as an anticancer agent. Met. Ions Life Sci. 2018, 18, 251–279. [Google Scholar]
- Erdogan, S.; Turkekul, K.; Dibirdik, I.; Doganlar, O.; Doganlar, Z.B.; Bilir, A.; Oktem, G. Midkine downregulation increases the efficacy of quercetin on prostate cancer stem cell survival and migration through PI3K/AKT and MAPK/ERK pathway. Biomed. Pharmacother. 2018, 107, 793–805. [Google Scholar] [CrossRef]
- Tang, S.N.; Fu, J.; Nall, D.; Rodova, M.; Shankar, S.; Srivastava, R.K. Inhibition of sonic hedgehog pathway and pluripotency maintaining factors regulate human pancreatic cancer stem cell characteristics. Int. J. Cancer 2012, 131, 30–40. [Google Scholar] [CrossRef] [Green Version]
- Hoca, M.; Becer, E.; Kabadayı, H.; Yücecan, S.; Vatansever, H.S. The Effect of Resveratrol and Quercetin on Epithelial-Mesenchymal Transition in Pancreatic Cancer Stem Cell. Nutr. Cancer 2020, 72, 1231–1242. [Google Scholar] [CrossRef]
- Balakrishnan, S.; Mukherjee, S.; Das, S.; Bhat, F.A.; Raja Singh, P.; Patra, C.R.; Arunakaran, J. Gold nanoparticles-conjugated quercetin induces apoptosis via inhibition of EGFR/PI3K/Akt-mediated pathway in breast cancer cell lines (MCF-7 and MDA-MB-231). Cell Biochem. Funct. 2017, 35, 217–231. [Google Scholar] [CrossRef]
- Lan, C.Y.; Chen, S.Y.; Kuo, C.W.; Lu, C.C.; Yen, G.C. Quercetin facilitates cell death and chemosensitivity through RAGE/PI3K/AKT/mTOR axis in human pancreatic cancer cells. J. Food Drug Anal. 2019, 27, 887–896. [Google Scholar] [CrossRef] [PubMed]
- Psahoulia, F.H.; Drosopoulos, K.G.; Doubravska, L.; Andera, L.; Pintzas, A. Quercetin enhances TRAIL-mediated apoptosis in colon cancer cells by inducing the accumulation of death receptors in lipid rafts. Mol. Cancer Ther. 2007, 6, 2591–2599. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Wei, L.; Liu, T.T.; Wang, H.H.; Hong, H.M.; Yu, A.L.; Feng, H.P.; Chang, W.W. Hsp27 participates in the maintenance of breast cancer stem cells through regulation of epithelial-mesenchymal transition and nuclear factor-κB. Breast Cancer Res. BCR 2011, 13, R101. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Qin, J.J.; Li, X.; Hunt, C.; Wang, W.; Wang, H.; Zhang, R. Natural products targeting the p53-MDM2 pathway and mutant p53: Recent advances and implications in cancer medicine. Genes Dis 2018, 5, 204–219. [Google Scholar] [CrossRef]
- Kuo, P.C.; Liu, H.F.; Chao, J.I. Survivin and p53 modulate quercetin-induced cell growth inhibition and apoptosis in human lung carcinoma cells. J. Biol. Chem. 2004, 279, 55875–55885. [Google Scholar] [CrossRef]
- Tanigawa, S.; Fujii, M.; Hou, D.X. Stabilization of p53 is involved in quercetin-induced cell cycle arrest and apoptosis in HepG2 cells. Biosci. Biotechnol. Biochem. 2008, 72, 797–804. [Google Scholar] [CrossRef] [PubMed]
- Lee, Y.-J.; Lee, Y.-J.; Park, I.-S.; Song, J.-H.; Oh, M.-H.; Nam, H.-S.; Cho, M.-K.; Woo, K.-M.; Lee, S.-H. Quercetin exerts preferential cytotoxic effects on malignant mesothelioma cells by inducing p53 expression, caspase-3 activation, and apoptosis. Mol. Cell. Toxicol. 2015, 11, 295–305. [Google Scholar] [CrossRef]
- Kusaczuk, M.; Krętowski, R.; Naumowicz, M.; Stypułkowska, A.; Cechowska-Pasko, M. A Preliminary Study of the Effect of Quercetin on Cytotoxicity, Apoptosis, and Stress Responses in Glioblastoma Cell Lines. Int. J. Mol. Sci. 2022, 23, 1345. [Google Scholar] [CrossRef]
- Bao, L.; Zhang, H.; Chan, L.S. The involvement of the JAK-STAT signaling pathway in chronic inflammatory skin disease atopic dermatitis. JAK-STAT 2013, 2, e24137. [Google Scholar] [CrossRef] [Green Version]
- Horr, B.; Borck, H.; Thurmond, R.; Grösch, S.; Diel, F. STAT1 phosphorylation and cleavage is regulated by the histamine (H4) receptor in human atopic and non-atopic lymphocytes. Int. Immunopharmacol. 2006, 6, 1577–1585. [Google Scholar] [CrossRef]
- Muthian, G.; Bright, J.J. Quercetin, a flavonoid phytoestrogen, ameliorates experimental allergic encephalomyelitis by blocking IL-12 signaling through JAK-STAT pathway in T lymphocyte. J. Clin. Immunol. 2004, 24, 542–552. [Google Scholar] [CrossRef] [PubMed]
- Senggunprai, L.; Kukongviriyapan, V.; Prawan, A.; Kukongviriyapan, U. Quercetin and EGCG exhibit chemopreventive effects in cholangiocarcinoma cells via suppression of JAK/STAT signaling pathway. Phyther. Res. 2014, 28, 841–848. [Google Scholar] [CrossRef] [PubMed]
- Kim, G.T.; Lee, S.H.; Kim, J.I.; Kim, Y.M. Quercetin regulates the sestrin 2-AMPK-p38 MAPK signaling pathway and induces apoptosis by increasing the generation of intracellular ROS in a p53-independent manner. Int. J. Mol. Med. 2014, 33, 863–869. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Dutta, S.; Sadhukhan, P.; Saha, S.; Sil, P.C. Regulation of oxidative stress by different naturally occurring polyphenolic compounds: An emerging anticancer therapeutic approach. React. Oxyg. Species 2017, 3, 81–95. [Google Scholar] [CrossRef]
- Arzuman, L.; Beale, P.; Chan, C.; Yu, J.Q.; Huq, F. Synergism from combinations of tris(benzimidazole) monochloroplatinum(II) chloride with capsaicin, quercetin, curcumin and cisplatin in human ovarian cancer cell lines. Anticancer. Res. 2014, 34, 5453–5464. [Google Scholar] [PubMed]
- Matsuzawa, A.; Nishitoh, H.; Tobiume, K.; Takeda, K.; Ichijo, H. Physiological roles of ASK1-mediated signal transduction in oxidative stress- and endoplasmic reticulum stress-induced apoptosis: Advanced findings from ASK1 knockout mice. Antioxid. Redox Signal. 2002, 4, 415–425. [Google Scholar] [CrossRef] [PubMed]
- Watanabe, T.; Sekine, S.; Naguro, I.; Sekine, Y.; Ichijo, H. Apoptosis Signal-regulating Kinase 1 (ASK1)-p38 Pathway-dependent Cytoplasmic Translocation of the Orphan Nuclear Receptor NR4A2 Is Required for Oxidative Stress-induced Necrosis. J. Biol. Chem. 2015, 290, 10791–10803. [Google Scholar] [CrossRef] [Green Version]
- Maurya, A.K.; Vinayak, M. Quercetin Attenuates Cell Survival, Inflammation, and Angiogenesis via Modulation of AKT Signaling in Murine T-Cell Lymphoma. Nutr. Cancer 2017, 69, 470–480. [Google Scholar] [CrossRef]
- Pratheeshkumar, P.; Budhraja, A.; Son, Y.O.; Wang, X.; Zhang, Z.; Ding, S.; Wang, L.; Hitron, A.; Lee, J.C.; Xu, M.; et al. Quercetin inhibits angiogenesis mediated human prostate tumor growth by targeting VEGFR- 2 regulated AKT/mTOR/P70S6K signaling pathways. PLoS ONE 2012, 7, e47516. [Google Scholar] [CrossRef] [Green Version]
- Xiao, Y.; Zhou, L.; Zhang, T.; Qin, C.; Wei, P.; Luo, L.; Luo, L.; Huang, G.; Chen, A.; Liu, G. Anti-fibrosis activity of quercetin attenuates rabbit tracheal stenosis via the TGF-β/AKT/mTOR signaling pathway. Life Sci. 2020, 250, 117552. [Google Scholar] [CrossRef]
- Granato, M.; Rizzello, C.; Gilardini Montani, M.S.; Cuomo, L.; Vitillo, M.; Santarelli, R.; Gonnella, R.; D’Orazi, G.; Faggioni, A.; Cirone, M. Quercetin induces apoptosis and autophagy in primary effusion lymphoma cells by inhibiting PI3K/AKT/mTOR and STAT3 signaling pathways. J. Nutr. Biochem. 2017, 41, 124–136. [Google Scholar] [CrossRef]
- Rivera Rivera, A.; Castillo-Pichardo, L.; Gerena, Y.; Dharmawardhane, S. Anti-Breast Cancer Potential of Quercetin via the Akt/AMPK/Mammalian Target of Rapamycin (mTOR) Signaling Cascade. PLoS ONE 2016, 11, e0157251. [Google Scholar] [CrossRef] [Green Version]
- Li, X.; Zhou, N.; Wang, J.; Liu, Z.; Wang, X.; Zhang, Q.; Liu, Q.; Gao, L.; Wang, R. Quercetin suppresses breast cancer stem cells (CD44(+)/CD24(-)) by inhibiting the PI3K/Akt/mTOR-signaling pathway. Life Sci. 2018, 196, 56–62. [Google Scholar] [CrossRef] [PubMed]
- Hamidullah; Kumar, R.; Saini, K.S.; Kumar, A.; Kumar, S.; Ramakrishna, E.; Maurya, R.; Konwar, R.; Chattopadhyay, N. Quercetin-6-C-β-D-glucopyranoside, natural analog of quercetin exhibits anti-prostate cancer activity by inhibiting Akt-mTOR pathway via aryl hydrocarbon receptor. Biochimie 2015, 119, 68–79. [Google Scholar] [CrossRef]
- He, K.; Yu, X.; Wang, X.; Tang, L.; Cao, Y.; Xia, J.; Cheng, J. Baicalein and Ly294002 induces liver cancer cells apoptosis via regulating phosphatidyl inositol 3-kinase/Akt signaling pathway. J. Cancer Res. Ther. 2018, 14, 519. [Google Scholar]
- Zhao, S.; Jiang, Y.; Zhao, J.; Li, H.; Yin, X.; Wang, Y.; Xie, Y.; Chen, X.; Lu, J.; Dong, Z.; et al. Quercetin-3-methyl ether inhibits esophageal carcinogenesis by targeting the AKT/mTOR/p70S6K and MAPK pathways. Mol. Carcinog. 2018, 57, 1540–1552. [Google Scholar] [CrossRef] [PubMed]
- Roy, S.; Banerjee, S.; Chakraborty, T. Vanadium quercetin complex attenuates mammary cancer by regulating the P53, Akt/mTOR pathway and downregulates cellular proliferation correlated with increased apoptotic events. Biometals 2018, 31, 647–671. [Google Scholar] [CrossRef] [PubMed]
- Granato, M.; Rizzello, C.; Romeo, M.A.; Yadav, S.; Santarelli, R.; D’Orazi, G.; Faggioni, A.; Cirone, M. Concomitant reduction of c-Myc expression and PI3K/AKT/mTOR signaling by quercetin induces a strong cytotoxic effect against Burkitt’s lymphoma. Int. J. Biochem. Cell Biol. 2016, 79, 393–400. [Google Scholar] [CrossRef] [PubMed]
- Jia, L.; Huang, S.; Yin, X.; Zan, Y.; Guo, Y.; Han, L. Quercetin suppresses the mobility of breast cancer by suppressing glycolysis through Akt-mTOR pathway mediated autophagy induction. Life Sci. 2018, 208, 123–130. [Google Scholar] [CrossRef] [PubMed]
- Wu, H.; Pan, L.; Gao, C.; Xu, H.; Li, Y.; Zhang, L.; Ma, L.; Meng, L.; Sun, X.; Qin, H. Quercetin Inhibits the Proliferation of Glycolysis-Addicted HCC Cells by Reducing Hexokinase 2 and Akt-mTOR Pathway. Molecules 2019, 24, 1993. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Wang, K.; Liu, R.; Li, J.; Mao, J.; Lei, Y.; Wu, J.; Zeng, J.; Zhang, T.; Wu, H.; Chen, L.; et al. Quercetin induces protective autophagy in gastric cancer cells: Involvement of Akt-mTOR- and hypoxia-induced factor 1α-mediated signaling. Autophagy 2011, 7, 966–978. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Meng, F.D.; Li, Y.; Tian, X.; Ma, P.; Sui, C.G.; Fu, L.Y.; Jiang, Y.H. Synergistic effects of snail and quercetin on renal cell carcinoma Caki-2 by altering AKT/mTOR/ERK1/2 signaling pathways. Int. J. Clin. Exp. Pathol. 2015, 8, 6157–6168. [Google Scholar] [PubMed]
- Lou, M.; Zhang, L.N.; Ji, P.G.; Feng, F.Q.; Liu, J.H.; Yang, C.; Li, B.F.; Wang, L. Quercetin nanoparticles induced autophagy and apoptosis through AKT/ERK/Caspase-3 signaling pathway in human neuroglioma cells: In vitro and in vivo. Biomed. Pharm. 2016, 84, 1–9. [Google Scholar] [CrossRef]
- Rendon-Mitchell, B.; Ochani, M.; Li, J.; Han, J.; Wang, H.; Yang, H.; Susarla, S.; Czura, C.; Mitchell, R.A.; Chen, G. IFN-γ induces high mobility group box 1 protein release partly through a TNF-dependent mechanism. J. Immunol. 2003, 170, 3890–3897. [Google Scholar] [CrossRef] [Green Version]
- Andersson, U.; Wang, H.; Palmblad, K.; Aveberger, A.-C.; Bloom, O.; Erlandsson-Harris, H.; Janson, A.; Kokkola, R.; Zhang, M.; Yang, H.; et al. High mobility group 1 protein (HMG-1) stimulates proinflammatory cytokine synthesis in human monocytes. J. Exp. Med. 2000, 192, 565–570. [Google Scholar] [CrossRef] [PubMed]
- Degryse, B.; Bonaldi, T.; Scaffidi, P.; Müller, S.; Resnati, M.; Sanvito, F.; Arrigoni, G.; Bianchi, M.E. The high mobility group (HMG) boxes of the nuclear protein HMG1 induce chemotaxis and cytoskeleton reorganization in rat smooth muscle cells. J. Cell Biol. 2001, 152, 1197–1206. [Google Scholar] [CrossRef] [PubMed]
- Kim, H.; Moon, J.Y.; Ahn, K.S.; Cho, S.K. Quercetin induces mitochondrial mediated apoptosis and protective autophagy in human glioblastoma U373MG cells. Oxidative Med. Cell. Longev. 2013, 2013, 596496. [Google Scholar] [CrossRef] [Green Version]
- Han, Y.; Yu, H.; Wang, J.; Ren, Y.; Su, X.; Shi, Y.J.H. Quercetin alleviates myocyte toxic and sensitizes anti-leukemic effect of adriamycin. Hematology 2015, 20, 276–283. [Google Scholar] [CrossRef] [PubMed]
- Park, J.S.; Svetkauskaite, D.; He, Q.; Kim, J.-Y.; Strassheim, D.; Ishizaka, A.; Abraham, E. Involvement of toll-like receptors 2 and 4 in cellular activation by high mobility group box 1 protein. J. Biol. Chem. 2004, 279, 7370–7377. [Google Scholar] [CrossRef] [Green Version]
- Kokkola, R.; Andersson, Å.; Mullins, G.; Östberg, T.; Treutiger, C.J.; Arnold, B.; Nawroth, P.; Andersson, U.; Harris, R.; Harris, H. RAGE is the major receptor for the proinflammatory activity of HMGB1 in rodent macrophages. Scand. J. Immunol. 2005, 61, 1–9. [Google Scholar] [CrossRef]
- Luo, J.-L.; Kamata, H.; Karin, M. IKK/NF-κB signaling: Balancing life and death–a new approach to cancer therapy. J. Clin. Investig. 2005, 115, 2625–2632. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Kwak, M.-K.; Itoh, K.; Yamamoto, M.; Sutter, T.R.; Kensler, T.W. Role of transcription factor Nrf2 in the induction of hepatic phase 2 and antioxidative enzymes in vivo by the cancer chemoprotective agent, 3 H-1, 2-dithiole-3-thione. Mol. Med. 2001, 7, 135–145. [Google Scholar] [CrossRef] [Green Version]
- Tan, X.-L.; Spivack, S.D. Dietary chemoprevention strategies for induction of phase II xenobiotic-metabolizing enzymes in lung carcinogenesis: A review. Lung Cancer 2009, 65, 129–137. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Kansanen, E.; Jyrkkänen, H.-K.; Volger, O.L.; Leinonen, H.; Kivelä, A.M.; Häkkinen, S.-K.; Woodcock, S.R.; Schopfer, F.J.; Horrevoets, A.J.; Ylä-Herttuala, S.; et al. Nrf2-dependent and-independent responses to nitro-fatty acids in human endothelial cells. J. Biol. Chem. 2009, 284, 33233–33241. [Google Scholar] [CrossRef] [Green Version]
- Taguchi, K.; Motohashi, H.; Yamamoto, M. Molecular mechanisms of the Keap1–Nrf2 pathway in stress response and cancer evolution. Genes Cells 2011, 16, 123–140. [Google Scholar] [CrossRef] [PubMed]
- Kansanen, E.; Jyrkkänen, H.-K.; Levonen, A.-L. Activation of stress signaling pathways by electrophilic oxidized and nitrated lipids. Free Radic. Biol. Med. 2012, 52, 973–982. [Google Scholar] [CrossRef] [PubMed]
- Dinkova-Kostova, A.T.; Holtzclaw, W.D.; Cole, R.N.; Itoh, K.; Wakabayashi, N.; Katoh, Y.; Yamamoto, M.; Talalay, P. Direct evidence that sulfhydryl groups of Keap1 are the sensors regulating induction of phase 2 enzymes that protect against carcinogens and oxidants. Proc. Natl. Acad. Sci. USA 2002, 99, 11908–11913. [Google Scholar] [CrossRef] [Green Version]
- Tanigawa, S.; Fujii, M.; Hou, D.-X. Action of Nrf2 and Keap1 in ARE-mediated NQO1 expression by quercetin. Free. Radic. Biol. Med. 2007, 42, 1690–1703. [Google Scholar] [CrossRef]
- Odenthal, J.; van Heumen, B.W.; Roelofs, H.M.; te Morsche, R.H.; Marian, B.; Nagengast, F.M.; Peters, W.H. The influence of curcumin, quercetin, and eicosapentaenoic acid on the expression of phase II detoxification enzymes in the intestinal cell lines HT-29, Caco-2, HuTu 80, and LT97. Nutr. Cancer 2012, 64, 856–863. [Google Scholar] [CrossRef]
- Ramyaa, P.; Padma, V.V. Quercetin modulates OTA-induced oxidative stress and redox signalling in HepG2 cells—up regulation of Nrf2 expression and down regulation of NF-κB and COX-2. Biochim. Biophys. Acta (BBA)-Gen. Subj. 2014, 1840, 681–692. [Google Scholar] [CrossRef]
- Shi, Y.; Liang, X.-c.; Zhang, H.; Wu, Q.-l.; Qu, L.; Sun, Q. Quercetin protects rat dorsal root ganglion neurons against high glucose-induced injury in vitro through Nrf-2/HO-1 activation and NF-κB inhibition. Acta Pharmacol. Sin. 2013, 34, 1140–1148. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Lee, Y.-J.; Song, J.-H.; Oh, M.-H.; Lee, Y.-J.; Kim, Y.-B.; Im, J.-H.; Lee, S.-H. ERK1/2 activation in quercetin-treated BEAS-2B cell plays a role in Nrf2-driven HO-1 expression. Mol. Cell. Toxicol. 2011, 7, 347–355. [Google Scholar] [CrossRef]
- Chow, J.-M.; Shen, S.-C.; Huan, S.K.; Lin, H.-Y.; Chen, Y.-C. Quercetin, but not rutin and quercitrin, prevention of H2O2-induced apoptosis via anti-oxidant activity and heme oxygenase 1 gene expression in macrophages. Biochem. Pharmacol. 2005, 69, 1839–1851. [Google Scholar] [CrossRef] [PubMed]
- Yao, P.; Nussler, A.; Liu, L.; Hao, L.; Song, F.; Schirmeier, A.; Nussler, N. Quercetin protects human hepatocytes from ethanol-derived oxidative stress by inducing heme oxygenase-1 via the MAPK/Nrf2 pathways. J. Hepatol. 2007, 47, 253–261. [Google Scholar] [CrossRef] [PubMed]
- Tuli, H.S.; Sandhu, S.S.; Sharma, A.K. Pharmacological and therapeutic potential of Cordyceps with special reference to Cordycepin. 3 Biotech 2014, 4, 1–12. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Kumar, G.; Tuli, H.S.; Mittal, S.; Shandilya, J.K.; Tiwari, A.; Sandhu, S.S. Isothiocyanates: A class of bioactive metabolites with chemopreventive potential. Tumor Biol. 2015, 36, 4005–4016. [Google Scholar] [CrossRef]
- Kumar, G.; Mittal, S.; Sak, K.; Tuli, H.S. Molecular mechanisms underlying chemopreventive potential of curcumin: Current challenges and future perspectives. Life Sci. 2016, 148, 313–328. [Google Scholar] [CrossRef]
- Moon, S.-K.; Cho, G.-O.; Jung, S.-Y.; Gal, S.-W.; Kwon, T.K.; Lee, Y.-C.; Madamanchi, N.R.; Kim, C.-H. Quercetin exerts multiple inhibitory effects on vascular smooth muscle cells: Role of ERK1/2, cell-cycle regulation, and matrix metalloproteinase-9. Biochem. Biophys. Res. Commun. 2003, 301, 1069–1078. [Google Scholar] [CrossRef]
- Haghiac, M.; Walle, T. Quercetin induces necrosis and apoptosis in SCC-9 oral cancer cells. Nutr. Cancer 2005, 53, 220–231. [Google Scholar] [CrossRef]
- Mu, C.; Jia, P.; Yan, Z.; Liu, X.; Li, X.; Liu, H. Quercetin induces cell cycle G1 arrest through elevating Cdk inhibitors p2l and p27 in human hepatoma cell line (HepG2). Methods Find. Exp. Clin. Pharmacol. 2007, 29, 179–184. [Google Scholar] [CrossRef]
- Chou, C.-C.; Yang, J.-S.; Lu, H.-F.; Ip, S.-W.; Lo, C.; Wu, C.-C.; Lin, J.-P.; Tang, N.-Y.; Chung, J.-G.; Chou, M.-J. Quercetin-mediated cell cycle arrest and apoptosis involving activation of a caspase cascade through the mitochondrial pathway in human breast cancer MCF-7 cells. Arch. Pharmacal Res. 2010, 33, 1181–1191. [Google Scholar] [CrossRef]
- Casella, M.L.; Parody, J.P.; Ceballos, M.P.; Quiroga, A.D.; Ronco, M.T.; Francés, D.E.; Monti, J.A.; Pisani, G.B.; Carnovale, C.E.; Carrillo, M.C.; et al. Quercetin prevents liver carcinogenesis by inducing cell cycle arrest, decreasing cell proliferation and enhancing apoptosis. Mol. Nutr. Food Res. 2014, 58, 289–300. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Ren, M.X.; Deng, X.H.; Ai, F.; Yuan, G.Y.; Song, H.Y. Effect of quercetin on the proliferation of the human ovarian cancer cell line SKOV-3 in vitro. Exp. Ther. Med. 2015, 10, 579–583. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Lee, Y.-J.; Lee, D.M.; Lee, S.-H. Nrf2 expression and apoptosis in quercetin-treated malignant mesothelioma cells. Mol. Cells 2015, 38, 416. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Lou, G.; Liu, Y.; Wu, S.; Xue, J.; Yang, F.; Fu, H.; Zheng, M.; Chen, Z. The p53/miR-34a/SIRT1 positive feedback loop in quercetin-induced apoptosis. Cell. Physiol. Biochem. 2015, 35, 2192–2202. [Google Scholar] [CrossRef] [PubMed]
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations. |
© 2022 by the authors. Licensee MDPI, Basel, Switzerland. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license (https://creativecommons.org/licenses/by/4.0/).
Share and Cite
Biswas, P.; Dey, D.; Biswas, P.K.; Rahaman, T.I.; Saha, S.; Parvez, A.; Khan, D.A.; Lily, N.J.; Saha, K.; Sohel, M.; et al. A Comprehensive Analysis and Anti-Cancer Activities of Quercetin in ROS-Mediated Cancer and Cancer Stem Cells. Int. J. Mol. Sci. 2022, 23, 11746. https://doi.org/10.3390/ijms231911746
Biswas P, Dey D, Biswas PK, Rahaman TI, Saha S, Parvez A, Khan DA, Lily NJ, Saha K, Sohel M, et al. A Comprehensive Analysis and Anti-Cancer Activities of Quercetin in ROS-Mediated Cancer and Cancer Stem Cells. International Journal of Molecular Sciences. 2022; 23(19):11746. https://doi.org/10.3390/ijms231911746
Chicago/Turabian StyleBiswas, Partha, Dipta Dey, Polash Kumar Biswas, Tanjim Ishraq Rahaman, Shuvo Saha, Anwar Parvez, Dhrubo Ahmed Khan, Nusrat Jahan Lily, Konka Saha, Md Sohel, and et al. 2022. "A Comprehensive Analysis and Anti-Cancer Activities of Quercetin in ROS-Mediated Cancer and Cancer Stem Cells" International Journal of Molecular Sciences 23, no. 19: 11746. https://doi.org/10.3390/ijms231911746
APA StyleBiswas, P., Dey, D., Biswas, P. K., Rahaman, T. I., Saha, S., Parvez, A., Khan, D. A., Lily, N. J., Saha, K., Sohel, M., Hasan, M. M., Al Azad, S., Bibi, S., Hasan, M. N., Rahmatullah, M., Chun, J., Rahman, M. A., & Kim, B. (2022). A Comprehensive Analysis and Anti-Cancer Activities of Quercetin in ROS-Mediated Cancer and Cancer Stem Cells. International Journal of Molecular Sciences, 23(19), 11746. https://doi.org/10.3390/ijms231911746