Flavones: Six Selected Flavones and Their Related Signaling Pathways That Induce Apoptosis in Cancer
Abstract
:1. Introduction
1.1. Cancer and Apoptosis
1.2. An Overview of Flavonoids and Their Structure–Activity Relationship in Anticancer Effects
1.3. An Overview of Flavones and Their Biological and Pharmacological Effects
1.4. Flavones as Excellent Anticancer Agents
2. Six Selected Flavones and Their Anticancer Effects
2.1. Apigenin
2.2. Acacetin
2.3. Baicalein
2.4. Luteolin
2.5. Tangeretin
2.6. Wogonin
3. Apoptosis Pathways
3.1. Intrinsic Pathway
3.2. Extrinsic Pathway
4. Signaling Pathways Related to Cancer Cell Apoptosis
4.1. PI3K/AKT Pathway
4.2. Wnt Pathway
4.3. JAK/STAT Pathway
4.4. MAPK Pathway
4.5. p53
5. Regulation of Signaling Pathways in Which Six Selected Flavones Induce Apoptosis in Cancer Cells
5.1. Apigenin
5.2. Acacetin
5.3. Baicalein
5.4. Luteolin
5.5. Tangeretin
5.6. Wogonin
6. Anticancer Effects of Six Selected Flavones in an In Vivo Model
7. Discussion and Conclusions
7.1. The Prospects of Compounds Such as Flavones and Flavonoids for Anticancer Effects
7.2. Noteworthy ER-Mediated Apoptosis and the Need to Study Related Flavones
7.3. Conclusions
Author Contributions
Funding
Institutional Review Board Statement
Informed Consent Statement
Data Availability Statement
Conflicts of Interest
References
- Wu, S.; Zhu, W.; Thompson, P.; Hannun, Y.A. Evaluating intrinsic and non-intrinsic cancer risk factors. Nat. Commun. 2018, 9, 3490. [Google Scholar] [CrossRef]
- Hanahan, D.; Weinberg, R.A. Hallmarks of cancer: The next generation. Cell 2011, 144, 646–674. [Google Scholar] [CrossRef]
- Wong, R.S. Apoptosis in cancer: From pathogenesis to treatment. J. Exp. Clin. Cancer Res. 2011, 30, 87. [Google Scholar] [CrossRef]
- Kroemer, G.; Galluzzi, L.; Vandenabeele, P.; Abrams, J.; Alnemri, E.S.; Baehrecke, E.H.; Blagosklonny, M.V.; El-Deiry, W.S.; Golstein, P.; Green, D.R.; et al. Classification of cell death: Recommendations of the Nomenclature Committee on Cell Death 2009. Cell Death Differ. 2009, 16, 3–11. [Google Scholar] [CrossRef]
- Kerr, J.F.; Wyllie, A.H.; Currie, A.R. Apoptosis: A basic biological phenomenon with wide-ranging implications in tissue kinetics. Br. J. Cancer 1972, 26, 239–257. [Google Scholar] [CrossRef]
- Kerr, J.F.; Winterford, C.M.; Harmon, B.V. Apoptosis. Its significance in cancer and cancer therapy. Cancer 1994, 73, 2013–2026. [Google Scholar] [CrossRef]
- Hacker, G. The morphology of apoptosis. Cell Tissue Res. 2000, 301, 5–17. [Google Scholar] [CrossRef]
- Pistritto, G.; Trisciuoglio, D.; Ceci, C.; Garufi, A.; D’Orazi, G. Apoptosis as anticancer mechanism: Function and dysfunction of its modulators and targeted therapeutic strategies. Aging 2016, 8, 603–619. [Google Scholar] [CrossRef]
- Haanen, C.; Vermes, I. Apoptosis and inflammation. Mediat. Inflamm. 1995, 4, 5–15. [Google Scholar] [CrossRef]
- Pfeffer, C.M.; Singh, A.T.K. Apoptosis: A Target for Anticancer Therapy. Int. J. Mol. Sci. 2018, 19, 448. [Google Scholar] [CrossRef] [Green Version]
- Kroemer, G.; Galluzzi, L.; Brenner, C. Mitochondrial membrane permeabilization in cell death. Physiol. Rev. 2007, 87, 99–163. [Google Scholar] [CrossRef]
- Li, P.; Nijhawan, D.; Budihardjo, I.; Srinivasula, S.M.; Ahmad, M.; Alnemri, E.S.; Wang, X. Cytochrome c and dATP-dependent formation of Apaf-1/caspase-9 complex initiates an apoptotic protease cascade. Cell 1997, 91, 479–489. [Google Scholar] [CrossRef]
- Levine, B.; Sinha, S.; Kroemer, G. Bcl-2 family members: Dual regulators of apoptosis and autophagy. Autophagy 2008, 4, 600–606. [Google Scholar] [CrossRef]
- Zhang, Z.; Wang, C.Z.; Du, G.J.; Qi, L.W.; Calway, T.; He, T.C.; Du, W.; Yuan, C.S. Genistein induces G2/M cell cycle arrest and apoptosis via ATM/p53-dependent pathway in human colon cancer cells. Int. J. Oncol. 2013, 43, 289–296. [Google Scholar] [CrossRef]
- Waring, P.; Mullbacher, A. Cell death induced by the Fas/Fas ligand pathway and its role in pathology. Immunol. Cell Biol. 1999, 77, 312–317. [Google Scholar] [CrossRef]
- Jaiswal, P.K.; Goel, A.; Mittal, R.D. Survivin: A molecular biomarker in cancer. Indian J. Med. Res. 2015, 141, 389–397. [Google Scholar]
- Li, J.; Yuan, J. Caspases in apoptosis and beyond. Oncogene 2008, 27, 6194–6206. [Google Scholar] [CrossRef]
- Solt, L.A.; May, M.J. The IkappaB kinase complex: Master regulator of NF-kappaB signaling. Immunol. Res. 2008, 42, 3–18. [Google Scholar] [CrossRef]
- Nicholson, K.M.; Anderson, N.G. The protein kinase B/Akt signalling pathway in human malignancy. Cell Signal. 2002, 14, 381–395. [Google Scholar] [CrossRef]
- Nitulescu, G.M.; Van De Venter, M.; Nitulescu, G.; Ungurianu, A.; Juzenas, P.; Peng, Q.; Olaru, O.T.; Gradinaru, D.; Tsatsakis, A.; Tsoukalas, D.; et al. The Akt pathway in oncology therapy and beyond (Review). Int. J. Oncol. 2018, 53, 2319–2331. [Google Scholar] [CrossRef]
- Abotaleb, M.; Samuel, S.M.; Varghese, E.; Varghese, S.; Kubatka, P.; Liskova, A.; Busselberg, D. Flavonoids in Cancer and Apoptosis. Cancers 2018, 11, 28. [Google Scholar] [CrossRef] [Green Version]
- Raffa, D.; Maggio, B.; Raimondi, M.V.; Plescia, F.; Daidone, G. Recent discoveries of anticancer flavonoids. Eur. J. Med. Chem. 2017, 142, 213–228. [Google Scholar] [CrossRef]
- Kopustinskiene, D.M.; Jakstas, V.; Savickas, A.; Bernatoniene, J. Flavonoids as Anticancer Agents. Nutrients 2020, 12, 457. [Google Scholar] [CrossRef]
- Kumar, S.; Pandey, A.K. Chemistry and biological activities of flavonoids: An overview. Sci. World J. 2013, 2013, 162750. [Google Scholar] [CrossRef]
- Panche, A.N.; Diwan, A.D.; Chandra, S.R. Flavonoids: An overview. J. Nutr. Sci. 2016, 5, e47. [Google Scholar] [CrossRef]
- Ramos, S. Effects of dietary flavonoids on apoptotic pathways related to cancer chemoprevention. J. Nutr. Biochem. 2007, 18, 427–442. [Google Scholar] [CrossRef]
- Liskova, A.; Samec, M.; Koklesova, L.; Brockmueller, A.; Zhai, K.; Abdellatif, B.; Siddiqui, M.; Biringer, K.; Kudela, E.; Pec, M.; et al. Flavonoids as an effective sensitizer for anti-cancer therapy: Insights into multi-faceted mechanisms and applicability towards individualized patient profiles. EPMA J. 2021, 12, 155–176. [Google Scholar] [CrossRef]
- Kawaii, S.; Tomono, Y.; Katase, E.; Ogawa, K.; Yano, M. Antiproliferative activity of flavonoids on several cancer cell lines. Biosci. Biotechnol. Biochem. 1999, 63, 896–899. [Google Scholar] [CrossRef]
- Sanchez, I.; Calderon, J.; Ruiz, B.; Tellez, J.; Calzada, L.; Taboada, J. In vitro cytotoxicity of flavonoids against MK2 and C6 tumour cells. Phytother. Res. 2001, 15, 290–293. [Google Scholar] [CrossRef]
- Jayaprakasam, B.; Vareed, S.K.; Olson, L.K.; Nair, M.G. Insulin secretion by bioactive anthocyanins and anthocyanidins present in fruits. J. Agric. Food Chem. 2005, 53, 28–31. [Google Scholar] [CrossRef]
- Benavente-Garcia, O.; Castillo, J. Update on uses and properties of citrus flavonoids: New findings in anticancer, cardiovascular, and anti-inflammatory activity. J. Agric. Food Chem. 2008, 56, 6185–6205. [Google Scholar] [CrossRef] [PubMed]
- Manthey, J.A.; Guthrie, N. Antiproliferative activities of citrus flavonoids against six human cancer cell lines. J. Agric. Food Chem. 2002, 50, 5837–5843. [Google Scholar] [CrossRef] [PubMed]
- Agullo, G.; Gamet-Payrastre, L.; Manenti, S.; Viala, C.; Remesy, C.; Chap, H.; Payrastre, B. Relationship between flavonoid structure and inhibition of phosphatidylinositol 3-kinase: A comparison with tyrosine kinase and protein kinase C inhibition. Biochem. Pharmacol. 1997, 53, 1649–1657. [Google Scholar] [CrossRef]
- Casagrande, F.; Darbon, J.M. Effects of structurally related flavonoids on cell cycle progression of human melanoma cells: Regulation of cyclin-dependent kinases CDK2 and CDK1. Biochem. Pharmacol. 2001, 61, 1205–1215. [Google Scholar] [CrossRef]
- Albini, A.; Tosetti, F.; Benelli, R.; Noonan, D.M. Tumor inflammatory angiogenesis and its chemoprevention. Cancer Res. 2005, 65, 10637–10641. [Google Scholar] [CrossRef]
- Kim, J.D.; Liu, L.; Guo, W.; Meydani, M. Chemical structure of flavonols in relation to modulation of angiogenesis and immune-endothelial cell adhesion. J. Nutr. Biochem. 2006, 17, 165–176. [Google Scholar] [CrossRef]
- Gürler, S.B.; Kiraz, Y.; Baran, Y. Chapter 21—Flavonoids in cancer therapy: Current and future trends. In Biodiversity and Biomedicine; Ozturk, M., Egamberdieva, D., Pešić, M., Eds.; Academic Press: Cambridge, MA, USA, 2020; pp. 403–440. [Google Scholar]
- Hostetler, G.L.; Ralston, R.A.; Schwartz, S.J. Flavones: Food Sources, Bioavailability, Metabolism, and Bioactivity. Adv. Nutr. 2017, 8, 423–435. [Google Scholar] [CrossRef]
- Martens, S.; Mithofer, A. Flavones and flavone synthases. Phytochemistry 2005, 66, 2399–2407. [Google Scholar] [CrossRef]
- Ollila, F.; Halling, K.; Vuorela, P.; Vuorela, H.; Slotte, J.P. Characterization of flavonoid-biomembrane interactions. Arch. Biochem. Biophys. 2002, 399, 103–108. [Google Scholar] [CrossRef]
- Verma, A.K.; Pratap, R. The biological potential of flavones. Nat. Prod. Rep. 2010, 27, 1571–1593. [Google Scholar] [CrossRef]
- Bors, W.; Heller, W.; Michel, C.; Saran, M. Flavonoids as antioxidants: Determination of radical-scavenging efficiencies. Methods Enzymol. 1990, 186, 343–355. [Google Scholar] [PubMed]
- Cao, G.; Sofic, E.; Prior, R.L. Antioxidant and prooxidant behavior of flavonoids: Structure-activity relationships. Free Radic. Biol. Med. 1997, 22, 749–760. [Google Scholar] [CrossRef]
- Su, Y.L.; Huang, Y.; Chen, Z.Y. Isolation and elucidation of antioxidant constituents from acetone extract in root of Scutellaria rehderiana. Zhongguo Zhong Yao Za Zhi 2004, 29, 863–864. [Google Scholar] [PubMed]
- Lin, S.; Zhang, G.; Liao, Y.; Pan, J.; Gong, D. Die.etary Flavonoids as Xanthine Oxidase Inhibitors: Structure-Affinity and Structure-Activity Relationships. J. Agric. Food Chem. 2015, 63, 7784–7794. [Google Scholar] [CrossRef] [PubMed]
- Zeka, K.; Ruparelia, K.; Arroo, R.R.J.; Budriesi, R.; Micucci, M. Flavonoids and Their Metabolites: Prevention in Cardiovascular Diseases and Diabetes. Diseases 2017, 5, 19. [Google Scholar] [CrossRef]
- Basu, A.; Das, A.S.; Majumder, M.; Mukhopadhyay, R. Antiatherogenic Roles of Dietary Flavonoids Chrysin, Quercetin, and Luteolin. J. Cardiovasc. Pharmacol. 2016, 68, 89–96. [Google Scholar] [CrossRef]
- Olszanecki, R.; Gebska, A.; Kozlovski, V.I.; Gryglewski, R.J. Flavonoids and nitric oxide synthase. J. Physiol. Pharmacol. 2002, 53 Pt 1, 571–584. [Google Scholar]
- Liang, Y.C.; Tsai, S.H.; Tsai, D.C.; Lin-Shiau, S.Y.; Lin, J.K. Suppression of inducible cyclooxygenase and nitric oxide synthase through activation of peroxisome proliferator-activated receptor-gamma by flavonoids in mouse macrophages. FEBS Lett. 2001, 496, 12–18. [Google Scholar] [CrossRef]
- Chi, Y.S.; Cheon, B.S.; Kim, H.P. Effect of wogonin, a plant flavone from Scutellaria radix, on the suppression of cyclooxygenase-2 and the induction of inducible nitric oxide synthase in lipopolysaccharide-treated RAW 264.7 cells. Biochem. Pharmacol. 2001, 61, 1195–1203. [Google Scholar]
- Zwaagstra, M.E.; Timmerman, H.; van de Stolpe, A.C.; de Kanter, F.J.; Tamura, M.; Wada, Y.; Zhang, M.Q. Synthesis and structure-activity relationships of carboxyflavones as structurally rigid CysLT1 (LTD4) receptor antagonists. J. Med. Chem. 1998, 41, 1428–1438. [Google Scholar] [CrossRef]
- Chapple, C.R.; Parkhouse, H.; Gardener, C.; Milroy, E.J. Double-blind, placebo-controlled, cross-over study of flavoxate in the treatment of idiopathic detrusor instability. Br. J. Urol. 1990, 66, 491–494. [Google Scholar] [CrossRef] [PubMed]
- Chen, S.; Deng, P.S.; Swiderek, K.; Li, M.; Chan, S.I. Interaction of flavones and their bromoacetyl derivatives with NAD(P)H:quinone acceptor oxidoreductase. Mol. Pharmacol. 1995, 47, 419–424. [Google Scholar] [PubMed]
- Medina, J.H.; Viola, H.; Wolfman, C.; Marder, M.; Wasowski, C.; Calvo, D.; Paladini, A.C. Overview—Flavonoids: A new family of benzodiazepine receptor ligands. Neurochem. Res. 1997, 22, 419–425. [Google Scholar] [CrossRef] [PubMed]
- Kavvadias, D.; Monschein, V.; Sand, P.; Riederer, P.; Schreier, P. Constituents of sage (Salvia officinalis) with in vitro affinity to human brain benzodiazepine receptor. Planta Med. 2003, 69, 113–117. [Google Scholar] [CrossRef] [PubMed]
- Hieble, J.P.; Kolpak, D.C.; McCafferty, G.P.; Ruffolo, R.R., Jr.; Testa, R.; Leonardi, A. Effects of alpha1-adrenoceptor antagonists on agonist and tilt-induced changes in blood pressure: Relationships to uroselectivity. Eur. J. Pharmacol. 1999, 373, 51–62. [Google Scholar] [CrossRef]
- Testa, R.; Guarneri, L.; Taddei, C.; Poggesi, E.; Angelico, P.; Sartani, A.; Leonardi, A.; Gofrit, O.N.; Meretyk, S.; Caine, M. Functional antagonistic activity of Rec 15/2739, a novel alpha-1 antagonist selective for the lower urinary tract, on noradrenaline-induced contraction of human prostate and mesenteric artery. J. Pharmacol. Exp. Ther. 1996, 277, 1237–1246. [Google Scholar]
- Shin, J.S.; Kim, K.S.; Kim, M.B.; Jeong, J.H.; Kim, B.K. Synthesis and hypoglycemic effect of chrysin derivatives. Bioorg. Med. Chem. Lett. 1999, 9, 869–874. [Google Scholar] [CrossRef]
- Hari Babu, T.; Rama Subba Rao, V.; Tiwari, A.K.; Suresh Babu, K.; Srinivas, P.V.; Ali, A.Z.; Madhusudana Rao, J. Synthesis and biological evaluation of novel 8-aminomethylated oroxylin A analogues as alpha-glucosidase inhibitors. Bioorg. Med. Chem. Lett. 2008, 18, 1659–1662. [Google Scholar] [CrossRef]
- Kim, J.S.; Kwon, C.S.; Son, K.H. Inhibition of alpha-glucosidase and amylase by luteolin, a flavonoid. Biosci. Biotechnol. Biochem. 2000, 64, 2458–2461. [Google Scholar] [CrossRef]
- Kato, A.; Nasu, N.; Takebayashi, K.; Adachi, I.; Minami, Y.; Sanae, F.; Asano, N.; Watson, A.A.; Nash, R.J. Structure-activity relationships of flavonoids as potential inhibitors of glycogen phosphorylase. J. Agric. Food Chem. 2008, 56, 4469–4473. [Google Scholar] [CrossRef]
- Okuda, J.; Miwa, I.; Inagaki, K.; Horie, T.; Nakayama, M. Inhibition of aldose reductases from rat and bovine lenses by flavonoids. Biochem. Pharmacol. 1982, 31, 3807–3822. [Google Scholar] [CrossRef]
- Porcu, M.; Chiarugi, A. The emerging therapeutic potential of sirtuin-interacting drugs: From cell death to lifespan extension. Trends Pharmacol. Sci. 2005, 26, 94–103. [Google Scholar] [CrossRef] [PubMed]
- Marfak, A.; Trouillas, P.; Allais, D.P.; Calliste, C.A.; Cook-Moreau, J.; Duroux, J.L. Reactivity of flavonoids with 1-hydroxyethyl radical: A gamma-radiolysis study. Biochim. Biophys. Acta 2004, 1670, 28–39. [Google Scholar] [CrossRef] [PubMed]
- Li, Z.X.; Tian, F.J.; Wu, X.Y.; Zhang, Y.P.; Tian, L.; Shi, S. Anti-arrhythmic action of total flavones of Choerospondias axillaris fructus. Zhongguo Yao Li Xue Bao 1984, 5, 251–254. [Google Scholar] [PubMed]
- Li, G.R.; Wang, H.B.; Qin, G.W.; Jin, M.W.; Tang, Q.; Sun, H.Y.; Du, X.L.; Deng, X.L.; Zhang, X.H.; Chen, J.B.; et al. Acacetin, a natural flavone, selectively inhibits human atrial repolarization potassium currents and prevents atrial fibrillation in dogs. Circulation 2008, 117, 2449–2457. [Google Scholar] [CrossRef]
- Yang, Y.; Wang, S.; Bao, Y.R.; Li, T.J.; Yang, G.L.; Chang, X.; Meng, X.S. Anti-ulcer effect and potential mechanism of licoflavone by regulating inflammation mediators and amino acid metabolism. J. Ethnopharmacol. 2017, 199, 175–182. [Google Scholar] [CrossRef]
- Ninfali, P.; Antonelli, A.; Magnani, M.; Scarpa, E.S. Antiviral Properties of Flavonoids and Delivery Strategies. Nutrients 2020, 12, 2534. [Google Scholar] [CrossRef]
- Cushnie, T.P.; Lamb, A.J. Antimicrobial activity of flavonoids. Int. J. Antimicrob. Agents 2005, 26, 343–356. [Google Scholar] [CrossRef]
- Aboody, M.S.A.; Mickymaray, S. Anti-Fungal Efficacy and Mechanisms of Flavonoids. Antibiotics 2020, 9, 45. [Google Scholar] [CrossRef]
- Borges-Argaez, R.; Balnbury, L.; Flowers, A.; Gimenez-Turba, A.; Ruiz, G.; Waterman, P.G.; Pena-Rodriguez, L.M. Cytotoxic and antiprotozoal activity of flavonoids from Lonchocarpus spp. Phytomedicine 2007, 14, 530–533. [Google Scholar] [CrossRef]
- Nunes, A.R.; Vieira, I.G.P.; Queiroz, D.B.; Leal, A.; Maia Morais, S.; Muniz, D.F.; Calixto-Junior, J.T.; Coutinho, H.D.M. Use of Flavonoids and Cinnamates, the Main Photoprotectors with Natural Origin. Adv. Pharmacol. Sci. 2018, 2018, 5341487. [Google Scholar] [CrossRef] [PubMed]
- Arct, J.; Pytkowska, K. Flavonoids as components of biologically active cosmeceuticals. Clin. Dermatol. 2008, 26, 347–357. [Google Scholar] [CrossRef] [PubMed]
- Ferrell, J.E., Jr.; Chang Sing, P.D.; Loew, G.; King, R.; Mansour, J.M.; Mansour, T.E. Structure/activity studies of flavonoids as inhibitors of cyclic AMP phosphodiesterase and relationship to quantum chemical indices. Mol. Pharmacol. 1979, 16, 556–568. [Google Scholar] [PubMed]
- Cao, Z.; Zhang, H.; Cai, X.; Fang, W.; Chai, D.; Wen, Y.; Chen, H.; Chu, F.; Zhang, Y. Luteolin Promotes Cell Apoptosis by Inducing Autophagy in Hepatocellular Carcinoma. Cell. Physiol. Biochem. 2017, 43, 1803–1812. [Google Scholar] [CrossRef]
- Lee, D.H.; Kim, C.; Zhang, L.; Lee, Y.J. Role of p53, PUMA, and Bax in wogonin-induced apoptosis in human cancer cells. Biochem. Pharmacol. 2008, 75, 2020–2033. [Google Scholar] [CrossRef]
- Cathcart, M.C.; Useckaite, Z.; Drakeford, C.; Semik, V.; Lysaght, J.; Gately, K.; O’Byrne, K.J.; Pidgeon, G.P. Anti-cancer effects of baicalein in non-small cell lung cancer in-vitro and in-vivo. BMC Cancer 2016, 16, 707. [Google Scholar] [CrossRef]
- Pan, M.H.; Lai, C.S.; Hsu, P.C.; Wang, Y.J. Acacetin induces apoptosis in human gastric carcinoma cells accompanied by activation of caspase cascades and production of rea.active oxygen species. J. Agric. Food Chem. 2005, 53, 620–630. [Google Scholar] [CrossRef]
- Sung, H.; Ferlay, J.; Siegel, R.L.; Laversanne, M.; Soerjomataram, I.; Jemal, A.; Bray, F. Global Cancer Statistics 2020: GLOBOCAN Estimates of Incidence and Mortality Worldwide for 36 Cancers in 185 Countries. CA Cancer J. Clin. 2021, 71, 209–249. [Google Scholar] [CrossRef]
- Zhou, Y.; Zheng, J.; Li, Y.; Xu, D.P.; Li, S.; Chen, Y.M.; Li, H.B. Natural Polyphenols for Prevention and Treatment of Cancer. Nutrients 2016, 8, 515. [Google Scholar] [CrossRef]
- Yan, X.; Qi, M.; Li, P.; Zhan, Y.; Shao, H. Apigenin in cancer therapy: Anti-cancer effects and mechanisms of action. Cell Biosci. 2017, 7, 50. [Google Scholar] [CrossRef]
- Madunic, J.; Madunic, I.V.; Gajski, G.; Popic, J.; Garaj-Vrhovac, V. Apigenin: A dietary flavonoid with diverse anticancer properties. Cancer Lett. 2018, 413, 11–22. [Google Scholar] [CrossRef] [PubMed]
- Salmani, J.M.M.; Zhang, X.P.; Jacob, J.A.; Chen, B.A. Apigenin’s anticancer properties and molecular mechanisms of action: Recent advances and future prospectives. Chin. J. Nat. Med. 2017, 15, 321–329. [Google Scholar] [CrossRef]
- Kim, H.R.; Park, C.G.; Jung, J.Y. Acacetin (5,7-dihydroxy-4′-methoxyflavone) exhibits in vitro and in vivo anticancer activity through the suppression of NF-kappaB/Akt signaling in prostate cancer cells. Int. J. Mol. Med. 2014, 33, 317–324. [Google Scholar] [CrossRef] [PubMed]
- Singh, S.; Gupta, P.; Meena, A.; Luqman, S. Acacetin, a flavone with diverse therapeutic potential in cancer, inflammation, infections and other metabolic disorders. Food Chem. Toxicol. 2020, 145, 111708. [Google Scholar] [CrossRef]
- Prasad, N.; Sharma, J.R.; Yadav, U.C.S. Induction of growth cessation by acacetin via beta-catenin pathway and apoptosis by apoptosis inducing factor activation in colorectal carcinoma cells. Mol. Biol. Rep. 2020, 47, 987–1001. [Google Scholar] [CrossRef]
- Shim, H.Y.; Park, J.H.; Paik, H.D.; Nah, S.Y.; Kim, D.S.; Han, Y.S. Acacetin-induced apoptosis of human breast cancer MCF-7 cells involves caspase cascade, mitochondria-mediated death signaling and SAPK/JNK1/2-c-Jun activation. Mol. Cells 2007, 24, 95–104. [Google Scholar]
- Dou, J.; Wang, Z.; Ma, L.; Peng, B.; Mao, K.; Li, C.; Su, M.; Zhou, C.; Peng, G. Baicalein and baicalin inhibit colon cancer using two distinct fashions of apoptosis and senescence. Oncotarget 2018, 9, 20089–20102. [Google Scholar] [CrossRef]
- Liu, H.; Dong, Y.; Gao, Y.; Du, Z.; Wang, Y.; Cheng, P.; Chen, A.; Huang, H. The Fascinating Effects of Baicalein on Cancer: A Review. Int. J. Mol. Sci. 2016, 17, 1681. [Google Scholar] [CrossRef]
- Lee, M.N.; Lee, Y.; Wu, D.; Pae, M. Luteolin inhibits NLRP3 inflammasome activation via blocking ASC oligomerization. J. Nutr. Biochem. 2021, 92, 108614. [Google Scholar] [CrossRef]
- Imran, M.; Rauf, A.; Abu-Izneid, T.; Nadeem, M.; Shariati, M.A.; Khan, I.A.; Imran, A.; Orhan, I.E.; Rizwan, M.; Atif, M.; et al. Luteolin, a flavonoid, as an anticancer agent: A review. Biomed. Pharmacother. 2019, 112, 108612. [Google Scholar] [CrossRef]
- Lin, Y.; Shi, R.; Wang, X.; Shen, H.M. Luteolin, a flav.vonoid with potential for cancer prevention and therapy. Curr. Cancer Drug Targets 2008, 8, 634–646. [Google Scholar] [CrossRef]
- Raina, R.; Pramodh, S.; Rais, N.; Haque, S.; Shafarin, J.; Bajbouj, K.; Hamad, M.; Hussain, A. Luteolin inhibits proliferation, triggers apoptosis and modulates Akt/mTOR and MAP kinase pathways in HeLa cells. Oncol. Lett. 2021, 21, 192. [Google Scholar] [CrossRef] [PubMed]
- Ko, Y.C.; Choi, H.S.; Liu, R.; Kim, J.H.; Kim, S.L.; Yun, B.S.; Lee, D.S. Inhibitory Effects of Tangeretin, A Citrus Peel-Derived Flavonoid, on Breast Cancer Stem Cell Formation through Suppression of Stat3 Signaling. Molecules 2020, 25, 2599. [Google Scholar] [CrossRef] [PubMed]
- Dong, Y.; Cao, A.; Shi, J.; Yin, P.; Wang, L.; Ji, G.; Xie, J.; Wu, D. Tangeretin, a citrus polymethoxyflavonoid, induces apoptosis of human gastric cancer AGS cells through extrinsic and intrinsic signaling pathways. Oncol. Rep. 2014, 31, 1788–1794. [Google Scholar] [CrossRef] [PubMed]
- Arafa, E.A.; Shurrab, N.T.; Buabeid, M.A. Therapeutic Implications of a Polymethoxylated Flavone, Tangeretin, in the Management of Cancer via Modulation of Different Molecular Pathways. Adv. Pharmacol. Pharm. Sci. 2021, 2021, 4709818. [Google Scholar] [CrossRef]
- Raza, W.; Luqman, S.; Meena, A. Prospects of tangeretin as a modulator of cancer targets/pathways. Pharmacol. Res. 2020, 161, 105202. [Google Scholar] [CrossRef]
- Ruibin, J.; Bo, J.; Danying, W.; Chihong, Z.; Jianguo, F.; Linhui, G. Therapy Effects of Wogonin on Ovarian Cancer Cells. Biomed. Res. Int. 2017, 2017, 9381513. [Google Scholar] [CrossRef]
- Sharifi-Rad, J.; Herrera-Bravo, J.; Salazar, L.A.; Shaheen, S.; Abdulmajid Ayatollahi, S.; Kobarfard, F.; Imran, M.; Imran, A.; Custodio, L.; Dolores Lopez, M.; et al. The Therapeutic Potential of Wogonin Observed in Preclinical Studies. Evid. Based Complement. Alternat. Med. 2021, 2021, 9935451. [Google Scholar] [CrossRef]
- Huynh, D.L.; Sharma, N.; Kumar Singh, A.; Singh Sodhi, S.; Zhang, J.J.; Mongre, R.K.; Ghosh, M.; Kim, N.; Ho Park, Y.; Kee Jeong, D. Anti-tumor activity of wogonin, an extract from Scutellaria baicalensis, through regulating different signaling pathways. Chin. J. Nat. Med. 2017, 15, 15–40. [Google Scholar] [CrossRef]
- Tsai, C.F.; Yeh, W.L.; Huang, S.M.; Tan, T.W.; Lu, D.Y. Wogonin induces reactive oxygen species production and cell apoptosis in human glioma cancer cells. Int. J. Mol. Sci. 2012, 13, 9877–9892. [Google Scholar] [CrossRef]
- Reed, J.C. Bcl-2 family proteins: Regulators of apoptosis and chemoresistance in hematologic malignancies. Semin. Hematol. 1997, 34 (Suppl. 5), 9–19. [Google Scholar]
- Fulda, S.; Debatin, K.M. Extrinsic versus intrinsic apoptosis pathways in anticancer chemotherapy. Oncogene 2006, 25, 4798–4811. [Google Scholar] [CrossRef] [PubMed]
- Schneider, P.; Tschopp, J. Apoptosis induced by death receptors. Pharm. Acta Helv. 2000, 74, 281–286. [Google Scholar] [CrossRef]
- Annovazzi, L.; Mellai, M.; Caldera, V.; Valente, G.; Tessitore, L.; Schiffer, D. mTOR, S6 and AKT expression in relation to proliferation and apoptosis/autophagy in glioma. Anticancer Res. 2009, 29, 3087–3094. [Google Scholar]
- Ersahin, T.; Tuncbag, N.; Cetin-Atalay, R. The PI3K/AKT/mTOR interactive pathway. Mol. Biosyst. 2015, 11, 1946–1954. [Google Scholar] [CrossRef]
- Yang, J.; Nie, J.; Ma, X.; Wei, Y.; Peng, Y.; Wei, X. Targeting PI3K in cancer: Mechanisms and advances in clinical trials. Mol. Cancer 2019, 18, 26. [Google Scholar] [CrossRef]
- Fresno Vara, J.A.; Casado, E.; de Castro, J.; Cejas, P.; Belda-Iniesta, C.; Gonzalez-Baron, M. PI3K/Akt signalling pathway and cancer. Cancer Treat. Rev. 2004, 30, 193–204. [Google Scholar] [CrossRef]
- Testa, J.R.; Bellacosa, A. AKT plays a central role in tumorigenesis. Proc. Natl. Acad. Sci. USA 2001, 98, 10983–10985. [Google Scholar] [CrossRef]
- Duan, S.; Huang, W.; Liu, X.; Liu, X.; Chen, N.; Xu, Q.; Hu, Y.; Song, W.; Zhou, J. IMPDH2 promotes colorectal cancer progression through activation of the PI3K/AKT/mTOR and PI3K/AKT/FOXO1 signaling pathways. J. Exp. Clin. Cancer Res. 2018, 37, 304. [Google Scholar] [CrossRef]
- Li, H.; Gao, Q.; Guo, L.; Lu, S.H. The PTEN/PI3K/Akt pathway regulates stem-like cells in primary esophageal carcinoma cells. Cancer Biol. Ther. 2011, 11, 950–958. [Google Scholar] [CrossRef]
- Li, B.; Cheung, P.Y.; Wang, X.; Tsao, S.W.; Ling, M.T.; Wong, Y.C.; Cheung, A.L. Id-1 activation of PI3K/Akt/NFkappaB signaling pathway and its significance in promoting survival of esophageal cancer cells. Carcinogenesis 2007, 28, 2313–2320. [Google Scholar] [CrossRef] [PubMed]
- Seo, B.R.; Min, K.J.; Cho, I.J.; Kim, S.C.; Kwon, T.K. Correction: Curcumin Significantly Enhances Dual PI3K/Akt and mTOR Inhibitor NVP-BEZ235-Induced Apoptosis in Human Renal Carcinoma Caki Cells through Down-Regulation of p53-Dependent Bcl-2 Expression and Inhibition of Mcl-1 Protein Stability. PLoS ONE 2016, 11, e0151886. [Google Scholar]
- Pai, S.G.; Carneiro, B.A.; Mota, J.M.; Costa, R.; Leite, C.A.; Barroso-Sousa, R.; Kaplan, J.B.; Chae, Y.K.; Giles, F.J. Wnt/beta-catenin pathway: Modulating anticancer immune response. J. Hematol. Oncol. 2017, 10, 101. [Google Scholar] [CrossRef] [PubMed]
- Su, N.; Wang, P.; Li, Y. Role of Wnt/beta-catenin pathway in inducing autophagy and apoptosis in multiple myeloma cells. Oncol. Lett. 2016, 12, 4623–4629. [Google Scholar] [CrossRef]
- Valenta, T.; Hausmann, G.; Basler, K. The many faces and functions of beta-catenin. EMBO J. 2012, 31, 2714–2736. [Google Scholar] [CrossRef]
- Chen, S.; Guttridge, D.C.; You, Z.; Zhang, Z.; Fribley, A.; Mayo, M.W.; Kitajewski, J.; Wang, C.Y. Wnt-1 signaling inhibits apoptosis by activating beta-catenin/T cell factor-mediated transcription. J. Cell Biol. 2001, 152, 87–96. [Google Scholar] [CrossRef]
- de Almeida, G.C.; Oliveira, L.F.S.; Predes, D.; Fokoue, H.H.; Kuster, R.M.; Oliveira, F.L.; Mendes, F.A.; Abreu, J.G. Piperine suppresses the Wnt/beta-catenin pathway and has anti-cancer effects on colorectal cancer cells. Sci. Rep. 2020, 10, 11681. [Google Scholar] [CrossRef]
- Trejo-Solis, C.; Escamilla-Ramirez, A.; Jimenez-Farfan, D.; Castillo-Rodriguez, R.A.; Flores-Najera, A.; Cruz-Salgado, A. Crosstalk of the Wnt/beta-Catenin Signaling Pathway in the Induction of Apoptosis on Cancer Cells. Pharmaceuticals 2021, 14, 871. [Google Scholar] [CrossRef]
- Bowman, T.; Garcia, R.; Turkson, J.; Jove, R. STATs in oncogenesis. Oncogene 2000, 19, 2474–2488. [Google Scholar] [CrossRef]
- Morris, R.; Kershaw, N.J.; Babon, J.J. The molecular details of cytokine signaling via the JAK/STAT pathway. Protein Sci. 2018, 27, 1984–2009. [Google Scholar] [CrossRef]
- Juan, W.C.; Ong, S.T. The role of protein phosphorylation in therapy resistance and disease progression in chronic myelogenous leukemia. Prog. Mol. Biol. Transl. Sci. 2012, 106, 107–142. [Google Scholar] [PubMed]
- Seif, F.; Khoshmirsafa, M.; Aazami, H.; Mohsenzadegan, M.; Sedighi, G.; Bahar, M. The role of JAK-STAT signaling pathway and its regulators in the fate of T helper cells. Cell Commun. Signal. 2017, 15, 23. [Google Scholar] [CrossRef] [PubMed]
- Bose, S.; Banerjee, S.; Mondal, A.; Chakraborty, U.; Pumarol, J.; Croley, C.R.; Bishayee, A. Targeting the JAK/STAT Signaling Pathway Using Phytocompounds for Cancer Prevention and Therapy. Cells 2020, 9, 1451. [Google Scholar] [CrossRef] [PubMed]
- Guo, Y.J.; Pan, W.W.; Liu, S.B.; Shen, Z.F.; Xu, Y.; Hu, L.L. ERK/MAPK signalling pathway and tumorigenesis. Exp. Ther. Med. 2020, 19, 1997–2007. [Google Scholar] [CrossRef] [PubMed]
- Lee, S.; Rauch, J.; Kolch, W. Targeting MAPK Signaling in Cancer: Mechanisms of Drug Resistance and Sensitivity. Int. J. Mol. Sci. 2020, 21, 1102. [Google Scholar] [CrossRef]
- Wada, T.; Penninger, J.M. Mitogen-activated protein kinases in apoptosis regulation. Oncogene 2004, 23, 2838–2849. [Google Scholar] [CrossRef]
- Hilger, R.A.; Scheulen, M.E.; Strumberg, D. The Ras-Raf-MEK-ERK pathway in the treatment of cancer. Onkologie 2002, 25, 511–518. [Google Scholar] [CrossRef]
- English, J.M.; Cobb, M.H. Pharmacological inhibitors of MAPK pathways. Trends Pharmacol. Sci. 2002, 23, 40–45. [Google Scholar] [CrossRef]
- Yue, J.; Lopez, J.M. Understanding MAPK Signaling Pathways in Apoptosis. Int. J. Mol. Sci. 2020, 21, 2346. [Google Scholar] [CrossRef]
- Timofeev, O. Editorial: Mutant p53 in Cancer Progression and Personalized Therapeutic Treatments. Front. Oncol. 2021, 11, 740578. [Google Scholar] [CrossRef]
- Aubrey, B.J.; Kelly, G.L.; Janic, A.; Herold, M.J.; Strasser, A. How does p53 induce apoptosis and how does this relate to p53-mediated tumour suppression? Cell Death Differ. 2018, 25, 104–113. [Google Scholar] [CrossRef] [PubMed]
- Nakano, K.; Vousden, K.H. PUMA, a novel proapop.ptotic gene, is induced by p53. Mol. Cell 2001, 7, 683–694. [Google Scholar] [CrossRef]
- Oda, E.; Ohki, R.; Murasawa, H.; Nemoto, J.; Shibue, T.; Yamashita, T.; Tokino, T.; Taniguchi, T.; Tanaka, N. Noxa, a BH3-only member of the Bcl-2 family and candidate mediator of p53-induced apoptosis. Science 2000, 288, 1053–1058. [Google Scholar] [CrossRef] [PubMed]
- O’Connor, L.; Strasser, A.; O’Reilly, L.A.; Hausmann, G.; Adams, J.M.; Cory, S.; Huang, D.C. Bim: A novel member of the Bcl-2 family that promotes apoptosis. EMBO J. 1998, 17, 384–395. [Google Scholar] [CrossRef]
- Seo, H.S.; Jo, J.K.; Ku, J.M.; Choi, H.S.; Choi, Y.K.; Woo, J.K.; Kim, H.I.; Kang, S.Y.; Lee, K.M.; Nam, K.W.; et al. Induction of caspase-dependent extrinsic apoptosis by apigenin through inhibition of signal transducer and activator of transcription 3 (STAT3) signalling in HER2-overexpressing BT-474 breast cancer cells. Biosci. Rep. 2015, 35, e00276. [Google Scholar] [CrossRef]
- Seo, H.S.; Choi, H.S.; Kim, S.R.; Choi, Y.K.; Woo, S.M.; Shin, I.; Woo, J.K.; Park, S.Y.; Shin, Y.C.; Ko, S.G. Apigenin induces apoptosis via extrinsic pathway, inducing p53 and inhibiting STAT3 and NFkappaB signaling in HER2-overexpressing breast cancer cells. Mol. Cell. Biochem. 2012, 366, 319–334. [Google Scholar] [CrossRef]
- Shukla, S.; Fu, P.; Gupta, S. Apigenin induces apoptosis by targeting inhibitor of apoptosis proteins and Ku70-Bax interaction in prostate cancer. Apoptosis 2014, 19, 883–894. [Google Scholar] [CrossRef]
- Yang, J.; Pi, C.; Wang, G. Inhibition of PI3K/Akt/mTOR pathway by apigenin induces apoptosis and autophagy in hepatocellular carcinoma cells. Biomed. Pharmacother. 2018, 103, 699–707. [Google Scholar] [CrossRef]
- Lee, Y.; Sung, B.; Kang, Y.J.; Kim, D.H.; Jang, J.Y.; Hwang, S.Y.; Kim, M.; Lim, H.S.; Yoon, J.H.; Chung, H.Y.; et al. Apigenin-induced apoptosis is enhanced by inhibition of autophagy formation in HCT116 human colon cancer cells. Int. J. Oncol. 2014, 44, 1599–1606. [Google Scholar] [CrossRef]
- Koosha, S.; Alshawsh, M.A.; Looi, C.Y.; Seyedan, A.; Mohamed, Z. An Association Map on the Effect of Flavonoids on the Signaling Pathways in Colorectal Cancer. Int. J. Med. Sci. 2016, 13, 374–385. [Google Scholar] [CrossRef]
- Chidambara Murthy, K.N.; Kim, J.; Vikram, A.; Patil, B.S. Differential inhibition of human colon cancer cells by structurally similar flavonoids of citrus. Food Chem. 2012, 132, 27–34. [Google Scholar] [CrossRef]
- Maeda, Y.; Takahashi, H.; Nakai, N.; Yanagita, T.; Ando, N.; Okubo, T.; Saito, K.; Shiga, K.; Hirokawa, T.; Hara, M.; et al. Apigenin induces apoptosis by suppressing Bcl-xl and Mcl-1 simultaneously via signal transducer and activator of transcription 3 signaling in colon cancer. Int. J. Oncol. 2018, 52, 1661–1673. [Google Scholar] [CrossRef] [PubMed]
- Woo, J.S.; Choo, G.S.; Yoo, E.S.; Kim, S.H.; Lee, J.H.; Han, S.H.; Kim, H.J.; Jung, S.H.; Park, Y.S.; Kim, B.S.; et al. Apigenin induces apoptosis by regulating Akt and MAPK pathways in human melanoma cell A375SM. Mol. Med. Rep. 2020, 22, 4877–4889. [Google Scholar] [CrossRef] [PubMed]
- Lu, H.F.; Chie, Y.J.; Yang, M.S.; Lee, C.S.; Fu, J.J.; Yang, J.S.; Tan, T.W.; Wu, S.H.; Ma, Y.S.; Ip, S.W.; et al. Apigenin induces caspase-dependent apoptosis in human lung cancer A549 cells through Bax- and Bcl-2-triggered mitochondrial pathway. Int. J. Oncol. 2010, 36, 1477–1484. [Google Scholar] [PubMed]
- Chen, M.; Wang, X.; Zha, D.; Cai, F.; Zhang, W.; He, Y.; Huang, Q.; Zhuang, H.; Hua, Z.C. Apigenin potentiates TRAIL therapy of non-small cell lung cancer via upregulating DR4/DR5 expression in a p53-dependent manner. Sci. Rep. 2016, 6, 35468. [Google Scholar] [CrossRef] [PubMed]
- Lee, J.H.; Li, Y.C.; Ip, S.W.; Hsu, S.C.; Chang, N.W.; Tang, N.Y.; Yu, C.S.; Chou, S.T.; Lin, S.S.; Lino, C.C.; et al. The role of Ca2+ in baicalein-induced apoptosis in human breast MDA-MB-231 cancer cells through mitochondria- and caspase-3-dependent pathway. Anticancer Res. 2008, 28, 1701–1711. [Google Scholar]
- Yan, W.; Ma, X.; Zhao, X.; Zhang, S. Baicalein induces apoptosis and autophagy of breast cancer cells via inhibiting PI3K/AKT pathway in vivo and vitro. Drug Des. Dev. Ther. 2018, 12, 3961–3972. [Google Scholar] [CrossRef]
- Wang, Z.; Jiang, C.; Chen, W.; Zhang, G.; Luo, D.; Cao, Y.; Wu, J.; Ding, Y.; Liu, B. Baicalein induces apoptosis and autophagy via endoplasmic reticulum stress in hepatocellular carcinoma cells. Biomed. Res. Int. 2014, 2014, 732516. [Google Scholar] [CrossRef]
- Mu, J.; Liu, T.; Jiang, L.; Wu, X.; Cao, Y.; Li, M.; Dong, Q.; Liu, Y.; Xu, H. The Traditional Chinese Medicine Baicalein Potently Inhibits Gastric Cancer Cells. J. Cancer 2016, 7, 453–461. [Google Scholar] [CrossRef] [Green Version]
- Gao, C.; Zhou, Y.; Li, H.; Cong, X.; Jiang, Z.; Wang, X.; Cao, R.; Tian, W. Antitumor effects of baicalin on ovarian cancer cells through induction of cell apoptosis and inhibition of cell migration in vitro. Mol. Med. Rep. 2017, 16, 8729–8734. [Google Scholar] [CrossRef]
- Lin, C.H.; Chang, C.Y.; Lee, K.R.; Lin, H.J.; Chen, T.H.; Wan, L. Flavones inhibit breast cancer proliferation through the Akt/FOXO3a signaling pathway. BMC Cancer 2015, 15, 958. [Google Scholar] [CrossRef] [PubMed]
- Huang, L.; Jin, K.; Lan, H. Luteolin inhibits cell cycle progression and induces apoptosis of breast cancer cells through downregulation of human telomerase reverse transcriptase. Oncol. Lett. 2019, 17, 3842–3850. [Google Scholar] [CrossRef]
- Shi, R.; Huang, Q.; Zhu, X.; Ong, Y.B.; Zhao, B.; Lu, J.; Ong, C.N.; Shen, H.M. Luteolin sensitizes the anticancer effect of cisplatin via c-Jun NH2-terminal kinase-mediated p53 phosphorylation and stabilization. Mol. Cancer Ther. 2007, 6, 1338–1347. [Google Scholar] [CrossRef] [PubMed]
- Jiang, Z.Q.; Li, M.H.; Qin, Y.M.; Jiang, H.Y.; Zhang, X.; Wu, M.H. Luteolin Inhibits Tumorigenesis and Induces Apoptosis of Non-Small Cell Lung Cancer Cells via Regulation of MicroRNA-34a-5p. Int. J. Mol. Sci. 2018, 19, 447. [Google Scholar] [CrossRef] [PubMed]
- Lu, X.; Li, Y.; Li, X.; Aisa, H.A. Luteolin induces apoptosis in vitro through suppressing the MAPK and PI3K signaling pathways in gastric cancer. Oncol. Lett. 2017, 14, 1993–2000. [Google Scholar] [CrossRef] [PubMed]
- Lee, H.J.; Wang, C.J.; Kuo, H.C.; Chou, F.P.; Jean, L.F.; Tseng, T.H. Induction apoptosis of luteolin in human hepatoma HepG2 cells involving mitochondria translocation of Bax/Bak and activation of JNK. Toxicol. Appl. Pharmacol. 2005, 203, 124–131. [Google Scholar] [CrossRef]
- Fan, S.; Xu, H.; Liu, H.; Hu, Z.; Xiao, J.; Liu, H. Inhibition of cancer cell growth by Tangeretin flavone in drug-resistant MDA-MB-231 human breast carcinoma cells is facilitated via targeting cell apoptosis, cell cycle phase distribution, cell invasion and activation of numerous Caspases. J. BUON 2019, 24, 1532–1537. [Google Scholar]
- Huang, K.F.; Zhang, G.D.; Huang, Y.Q.; Diao, Y. Wogonin induces apoptosis and down-regulates survivin in human breast cancer MCF-7 cells by modulating PI3K-AKT pathway. Int. Immunopharmacol. 2012, 12, 334–341. [Google Scholar] [CrossRef]
- Kim, S.J.; Kim, H.J.; Kim, H.R.; Lee, S.H.; Cho, S.D.; Choi, C.S.; Nam, J.S.; Jung, J.Y. Antitumor actions of baicalein and wogonin in HT-29 human colorectal cancer cells. Mol. Med. Rep. 2012, 6, 1443–1449. [Google Scholar] [CrossRef]
- He, L.; Lu, N.; Dai, Q.; Zhao, Y.; Zhao, L.; Wang, H.; Li, Z.; You, Q.; Guo, Q. Wogonin induced G1 cell cycle arrest by regulating Wnt/beta-catenin signaling pathway and inactivating CDK8 in human colorectal cancer carcinoma cells. Toxicology 2013, 312, 36–47. [Google Scholar] [CrossRef]
- Tan, H.; Li, X.; Yang, W.H.; Kang, Y. A flavone, Wogonin from Scutellaria baicalensis inhibits the proliferation of human colorectal cancer cells by inducing of autophagy, apoptosis and G2/M cell cycle arrest via modulating the PI3K/AKT and STAT3 signalling pathways. J. BUON 2019, 24, 1143–1149. [Google Scholar] [PubMed]
- Banik, K.; Khatoon, E.; Harsha, C.; Rana, V.; Parama, D.; Thakur, K.K.; Bishayee, A.; Kunnumakkara, A.B. Wogonin and its analogs for the prevention and treatment of cancer: A systematic review. Phytother. Res. 2022, 36, 1854–1883. [Google Scholar] [CrossRef] [PubMed]
- Shukla, S.; Bhaskaran, N.; Babcook, M.A.; Fu, P.; Maclennan, G.T.; Gupta, S. Apigenin inhibits prostate cancer progression in TRAMP mice via targeting PI3K/Akt/FoxO pathway. Carcinogenesis 2014, 35, 452–460. [Google Scholar] [CrossRef]
- Shukla, S.; Kanwal, R.; Shankar, E.; Datt, M.; Chance, M.R.; Fu, P.; MacLennan, G.T.; Gupta, S. Apigenin blocks IKKalpha activation and suppresses prostate cancer progression. Oncotarget 2015, 6, 31216–31232. [Google Scholar] [CrossRef]
- Amawi, H.; Ashby, C.R., Jr.; Tiwari, A.K. Cancer chemoprevention through dietary flavonoids: What’s limiting? Chin. J. Cancer 2017, 36, 50. [Google Scholar] [CrossRef]
- Costea, T.; Hudita, A.; Ciolac, O.A.; Galateanu, B.; Ginghina, O.; Costache, M.; Ganea, C.; Mocanu, M.M. Chemoprevention of Colorectal Cancer by Dietary Compounds. Int. J. Mol. Sci. 2018, 19, 3787. [Google Scholar] [CrossRef]
- Yun, S.; Lee, Y.J.; Choi, J.; Kim, N.D.; Han, D.C.; Kwon, B.M. Acacetin Inhibits the Growth of STAT3-Activated DU145 Prostate Cancer Cells by Directly Binding to Signal Transducer and Activator of Transcription 3 (STAT3). Molecules 2021, 26, 6204. [Google Scholar] [CrossRef] [PubMed]
- Wang, Y.; Han, E.; Xing, Q.; Yan, J.; Arrington, A.; Wang, C.; Tully, D.; Kowolik, C.M.; Lu, D.M.; Frankel, P.H.; et al. Baicalein upregulates DDIT4 expression which mediates mTOR inhibition and growth inhibition in cancer cells. Cancer Lett. 2015, 358, 170–179. [Google Scholar] [CrossRef]
- Peng, Y.; Guo, C.; Yang, Y.; Li, F.; Zhang, Y.; Jiang, B.; Li, Q. Baicalein induces apoptosis of human cervical cancer HeLa cells in vitro. Mol. Med. Rep. 2015, 11, 2129–2134. [Google Scholar] [CrossRef]
- Zang, M.D.; Hu, L.; Fan, Z.Y.; Wang, H.X.; Zhu, Z.L.; Cao, S.; Wu, X.Y.; Li, J.F.; Su, L.P.; Li, C.; et al. Luteolin suppresses gastric cancer progression by reversing epithelial-mesenchymal transition via suppression of the Notch signaling pathway. J. Transl. Med. 2017, 15, 52. [Google Scholar] [CrossRef] [Green Version]
- Hong, Z.; Cao, X.; Li, N.; Zhang, Y.; Lan, L.; Zhou, Y.; Pan, X.; Shen, L.; Yin, Z.; Luo, L. Luteolin is effective in the non-small cell lung cancer model with L858R/T790M EGF receptor mutation and erlotinib resistance. Br. J. Pharmacol. 2014, 171, 2842–2853. [Google Scholar] [CrossRef]
- Wang, Y.; Chen, Y.; Zhang, H.; Chen, J.; Cao, J.; Chen, Q.; Li, X.; Sun, C. Polymethoxyflavones from citrus inhibited gastric cancer cell proliferation through inducing apoptosis by upregulating RARbeta, both in vitro and in vivo. Food Chem. Toxicol. 2020, 146, 111811. [Google Scholar] [CrossRef] [PubMed]
- Zhao, Y.; Zhang, L.; Wu, Y.; Dai, Q.; Zhou, Y.; Li, Z.; Yang, L.; Guo, Q.; Lu, N. Selective anti-tumor activity of wogonin targeting the Warburg effect through stablizing p53. Pharmacol. Res. 2018, 135, 49–59. [Google Scholar] [CrossRef]
- Batra, P.; Sharma, A.K. Anti-cancer potential of flavonoids: Recent trends and future perspectives. 3 Biotech 2013, 3, 439–459. [Google Scholar] [CrossRef]
- Kay, C.D. The future of flavonoid research. Br. J. Nutr. 2010, 104 (Suppl. 3), S91–S95. [Google Scholar] [CrossRef] [PubMed]
- Zhai, S.; Senderowicz, A.M.; Sausville, E.A.; Figg, W.D. Flavopiridol, a novel cyclin-dependent kinase inhibitor, in clinical development. Ann. Pharmacother. 2002, 36, 905–911. [Google Scholar] [CrossRef] [PubMed]
- Martucciello, S.; Masullo, M.; Cerulli, A.; Piacente, S. Natural Products Targeting ER Stress, and the Functional Link to Mitochondria. Int. J. Mol. Sci. 2020, 21, 1905. [Google Scholar] [CrossRef]
- Amodio, G.; Moltedo, O.; Fasano, D.; Zerillo, L.; Oliveti, M.; Di Pietro, P.; Faraonio, R.; Barone, P.; Pellecchia, M.T.; De Rosa, A.; et al. PERK-Mediated Unfolded Protein Response Activation and Oxidative Stress in PARK20 Fibroblasts. Front. Neurosci. 2019, 13, 673. [Google Scholar]
- Puthalakath, H.; O’Reilly, L.A.; Gunn, P.; Lee, L.; Kelly, P.N.; Huntington, N.D.; Hughes, P.D.; Michalak, E.M.; McKimm-Breschkin, J.; Motoyama, N.; et al. ER stress triggers apoptosis by activating BH3-only protein Bim. Cell 2007, 129, 1337–1349. [Google Scholar] [CrossRef]
- Reimertz, C.; Kogel, D.; Rami, A.; Chittenden, T.; Prehn, J.H. Gene expression during ER stress-induced apoptosis in neurons: Induction of the BH3-only protein Bbc3/PUMA and activation of the mitochondrial apoptosis pathway. J. Cell Biol. 2003, 162, 587–597. [Google Scholar] [CrossRef]
- Malhi, H.; Kaufman, R.J. Endoplasmic reticulum stress in liver disease. J. Hepatol. 2011, 54, 795–809. [Google Scholar] [CrossRef] [PubMed] [Green Version]
Biological and Pharmacological Activities of Flavones | Reference |
---|---|
| [40,41] |
| [42,43,44] |
| [45] |
| [46] |
| [47,48,49,50] |
| [51] |
| [52] |
| [56,57] |
| [74] |
| [54,55] |
| [53] |
| [65,66] |
| [67] |
| [68] |
| [69] |
| [70] |
| [58,59,60,61,62,63] |
| [71] |
| [41,64] |
| [72] |
| [73] |
| [23,41] |
Flavone | Molecular Formula | Molecular Weight | Source | Anticancer Effects | Reference |
---|---|---|---|---|---|
Apigenin | C15H10O5 | 270.24 g/mol | Parsley, grapes, apples, chamomile tea, and red wine | Breast, prostate, liver, skin, colorectal, and lung cancers | [83] |
Acacetin | C16H12O5 | 284.26 g/mol | Safflower, propolis, and Asteraceae plants | Breast, stomach, gastric, and prostate cancers | [78,84,85,86,87] |
Baicalein | C15H10O5 | 270.24 g/mol | Scutellaria baicalensis | Breast, liver, gastric, stomach, and ovarian cancers | [88,89] |
Luteolin | C15H10O6 | 286.24 g/mol | Broccoli, carrots, celery, cabbage, and parsley | Breast, lung, stomach, liver, and cervical cancers | [91,92,93] |
Tangeretin | C20H20O7 | 372.37 g/mol | Citrus family | Breast, gastric, prostate, and bladder cancers | [95,96,97] |
Woginin | C16H12O5 | 284.26 g/mol | Scutellaria baicalensis Georgi (Lamiaceae) | Breast, colorectal, lung, and ovarian cancers and glioma | [98,100,101] |
Apigenin | ||||
---|---|---|---|---|
Cancer | Cell Line | Treatment Concentration | Apoptotic Regulation of Cell Signaling Pathway | Reference |
Breast Cancer | BT-474 | 20, 40, 60, 80, 100 μM |
| [136] |
MCF-7 | 10, 20, 40 μM |
| [137] | |
Prostate Cancer | PC-3, DU145 | 5, 10, 20, 40 μM |
| [138] |
Liver Cancer | Hep G2 | 10, 20, 40 μM |
| [139] |
Colorectal (Colon) Cancer | HCT-116 | 6.5, 12.5, 25, 50 μM |
| [140] |
SW480 | 12.5, 25, 50, 100, 200 μM |
| [141,142] | |
HT29, COLO320, DLD-1, HCT-116 | 5, 15, 50 μM |
| [143] | |
Human Melanoma | A375SM | 50, 100 μM |
| [144] |
Lung Cancer | A549 | 40, 80, 120, 160 μM |
| [145] |
NSCLC | 5, 10, 20, 40, 80, 160 μM |
| [146] | |
Acacetin | ||||
Cancer | Cell Line | Treatment Concentration | Apoptotic Regulation of Cell Signaling Pathway | Reference |
Breast Cancer | MCF-7 | 25, 50, 100, 150, 200 μM |
| [87] |
Gastric Cancer | AGS | 30, 60, 100 μM |
| [78] |
Colorectal (Colon) Cancer | SW480, HCT-116 | 25 μM |
| [86] |
Prostate Cancer | DU145 | 12.5, 25 μM |
| [84] |
Baicalein | ||||
Cancer | Cell Line | Treatment Concentration | Apoptotic Regulation of Cell Signaling Pathway | Reference |
Breast Cancer | MDA-MB-231 | 25, 50, 75, 100 μM |
| [89,147] |
MCF-7, MDA-MB-231 | 10, 20, 40 μM |
| [147,148] | |
Liver Cancer | HCC | 25, 50, 100, 200 μM |
| [149] |
Gastric Cancer | SGC-7901 | 15, 30, 60 μM |
| [150] |
Colorectal (Colon) Cancer | HCT-116, SW480 | 10, 20, 50 μM |
| [88] |
Ovarian Cancer | A2780 | 20, 40, 80, 160 μM |
| [151] |
Luteolin | ||||
Cancer | Cell Line | Treatment Concentration | Apoptotic Regulation of Cell Signaling Pathway | Reference |
Breast Cancer | Hs578T, MCF-7, MDA-MB-231 | 12.5, 25, 50, 100 μM |
| [152] |
MDA-MB-231 | 10, 30 μM |
| [153] | |
Lung Cancer | SCLC | 20 or 40 μM |
| [154] |
NSCLC | 10, 20, 40 μM |
| [155] | |
Gastric Cancer | BGC-823 | 20, 40, 60 μM |
| [156] |
Liver Cancer | SMMC-7721 | 25, 50, 100 μM |
| [75] |
Hep-G2 | 40, 80 μM |
| [157] | |
Cervical Cancer | HeLa | 5, 10, 20 μM |
| [93] |
Tangeretin | ||||
Cancer | Cell Line | Treatment Concentration | Apoptotic Regulation of cell signaling pathway | Reference |
Breast Cancer | MDA-MB-231 | 4.5, 9, 18 μM |
| [158] |
Gastric Cancer | AGS | 10, 30, 60 μM |
| [95] |
Prostate Cancer | PC-3, LNCaP | 25, 50, 75, 100 μM |
| [97] |
PC-3 | 25, 50, 100 μM |
| ||
DU145 | 25, 50, 100 μM |
| ||
Bladder Cancer | BFTC-905 | 20, 40, 60 μM |
| [97] |
Wogonin | ||||
Cancer | Cell Line | Treatment Concentration | Apoptotic Regulation of Cell Signaling Pathway | Reference |
Breast Cancer | MCF-7 | 30, 60, 90 μM |
| [159] |
Colorectal (Colon) Cancer | HT-29 | 25, 50, 100 μM |
| [160] |
HCT116 | 10, 20, 40 μM |
| [161] | |
SW480 | 4, 8, 16 μM |
| [162] | |
Lung Cancer | A549 | 5, 10, 20 μM or 25, 30, 50 μM |
| [163] |
Ovarian Cancer | A2780 | 10, 20, 30 μM |
| [98] |
Glioblastoma | U251, U87 | 4, 8, 16, 24 μM |
| [101] |
Flavones | Type of Cancer | Mouse Model and Dosages | In Vivo Function | Reference |
---|---|---|---|---|
Apigenin | Colorectal cancer | AOM-injected SD rat | Suppressed colorectal cancer by reducing ACF and increasing apoptosis | [141] |
Prostate cancer | TRAMP mice (20 and 50 μg/mouse/day, gavage) | Inhibited the formation of prostate carcinoma by regulating the PI3K/AKT/FOXO pathway | [164,165] | |
Athymic nude mouse (20 and 50 μg/mouse/day, oral) | Tumor volume was reduced; XIAP, survivin, and HDAC1 were downregulated; and BAX was increased | [138] | ||
Acacetin | Prostate cancer | BALB/C nude mouse (50 mg/kg, intraperitoneal injection 5 days per week for 30 days) | Inhibited tumor growth through STAT3 regulation | [168] |
Baicalein | Lung cancer | BALB/C nude mouse (1 and 3 mg/kg, intratumoral injection twice weekly) | Inhibition of tumor growth, genes that induced apoptosis, ITGB3 and TNFRSF25 upregulated | [77] |
Breast cancer | SCID-Beige mice (20 mg/kg, intraperitoneal injection for 5 days) | mTOR inhibition prevented tumor growth | [169] | |
Luteolin | Gastric cancer | BALB/C nude mouse (10 mg/kg, intraperitoneal injection 6 times) | Inhibition of Notch1 and β-catenin | [171] |
Lung cancer | BALB/C nude mouse (10 and 30 mg/kg, intraperitoneal injection daily for 15 days) | Tumor suppression and tumor weight were reduced by inhibition of PI3K/AKT/mTOR | [172] | |
Tangeretin | Breast cancer | Nude mouse (2.5 mg/kg, intraperitoneal injection once a week 4 times) | Inhibition of tumor growth | [94] |
Gastric cancer | BALB/C nude mouse (5, 25, 125 mg kg−1·bw−1·day−1) | Inhibition of tumor growth | [173] | |
Wogonin | Ovarian cancer | Athymic BALB/C nude mouse (20, 40, 80 mg/kg, intraperitoneal injection every 3 days) | Tumor volume and weight were reduced | [174] |
Colorectal cancer | Athymic BALB/C nude mouse (20, 40, 80 mg/kg, intraperitoneal injection every 3 days) | Tumor volume and weight were reduced | [174] |
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
Jeong, S.H.; Kim, H.H.; Ha, S.E.; Park, M.Y.; Bhosale, P.B.; Abusaliya, A.; Park, K.I.; Heo, J.D.; Kim, H.W.; Kim, G.S. Flavones: Six Selected Flavones and Their Related Signaling Pathways That Induce Apoptosis in Cancer. Int. J. Mol. Sci. 2022, 23, 10965. https://doi.org/10.3390/ijms231810965
Jeong SH, Kim HH, Ha SE, Park MY, Bhosale PB, Abusaliya A, Park KI, Heo JD, Kim HW, Kim GS. Flavones: Six Selected Flavones and Their Related Signaling Pathways That Induce Apoptosis in Cancer. International Journal of Molecular Sciences. 2022; 23(18):10965. https://doi.org/10.3390/ijms231810965
Chicago/Turabian StyleJeong, Se Hyo, Hun Hwan Kim, Sang Eun Ha, Min Young Park, Pritam Bhagwan Bhosale, Abuyaseer Abusaliya, Kwang Il Park, Jeong Doo Heo, Hyun Wook Kim, and Gon Sup Kim. 2022. "Flavones: Six Selected Flavones and Their Related Signaling Pathways That Induce Apoptosis in Cancer" International Journal of Molecular Sciences 23, no. 18: 10965. https://doi.org/10.3390/ijms231810965
APA StyleJeong, S. H., Kim, H. H., Ha, S. E., Park, M. Y., Bhosale, P. B., Abusaliya, A., Park, K. I., Heo, J. D., Kim, H. W., & Kim, G. S. (2022). Flavones: Six Selected Flavones and Their Related Signaling Pathways That Induce Apoptosis in Cancer. International Journal of Molecular Sciences, 23(18), 10965. https://doi.org/10.3390/ijms231810965