Flavonoids as Anticancer Agents
Abstract
:1. Introduction
2. Chemical Properties of Flavonoids
3. Bioavailability of Flavonoids
4. Anticancer Effects of Flavonoids
4.1. Flavonoids in Oxidative Stress
4.2. Flavonoids in Apoptosis
4.3. Immunomodulatory and Anti-Inflammatory Effects of Flavonoids
4.4. Effects of Flavonoids on Mitochondrial Functions
4.5. Effects of Flavonoids on Gut Microbiota
5. Conclusions and Future Perspectives
Author Contributions
Funding
Acknowledgments
Conflicts of Interest
References
- Nabavi, S.M.; Samec, D.; Tomczyk, M.; Milella, L.; Russo, D.; Habtemariam, S.; Suntar, I.; Rastrelli, L.; Daglia, M.; Xiao, J.; et al. Flavonoid biosynthetic pathways in plants: Versatile targets for metabolic engineering. Biotechnol. Adv. 2018. [Google Scholar] [CrossRef] [PubMed]
- Scarano, A.; Chieppa, M.; Santino, A. Looking at Flavonoid Biodiversity in Horticultural Crops: A Colored Mine with Nutritional Benefits. Plants 2018, 7, 98. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Liu, J.; Wang, X.; Yong, H.; Kan, J.; Jin, C. Recent advances in flavonoid-grafted polysaccharides: Synthesis, structural characterization, bioactivities and potential applications. Int. J. Biol. Macromol. 2018, 116, 1011–1025. [Google Scholar] [CrossRef] [PubMed]
- Kofink, M.; Papagiannopoulos, M.; Galensa, R. (-)-Catechin in cocoa and chocolate: Occurrence and analysis of an atypical flavan-3-ol enantiomer. Molecules 2007, 12, 1274–1288. [Google Scholar] [CrossRef] [PubMed]
- Braicu, C.; Ladomery, M.R.; Chedea, V.S.; Irimie, A.; Berindan-Neagoe, I. The relationship between the structure and biological actions of green tea catechins. Food Chem. 2013, 141, 3282–3289. [Google Scholar] [CrossRef] [PubMed]
- Arts, I.C.; van De Putte, B.; Hollman, P.C. Catechin contents of foods commonly consumed in The Netherlands. 2. Tea, wine, fruit juices, and chocolate milk. J. Agric. Food Chem. 2000, 48, 1752–1757. [Google Scholar] [CrossRef]
- Maatta-Riihinen, K.R.; Kamal-Eldin, A.; Torronen, A.R. Identification and quantification of phenolic compounds in berries of Fragaria and Rubus species (family Rosaceae). J. Agric. Food Chem. 2004, 52, 6178–6187. [Google Scholar] [CrossRef]
- Wu, X.; Gu, L.; Prior, R.L.; McKay, S. Characterization of anthocyanins and proanthocyanidins in some cultivars of Ribes, Aronia, and Sambucus and their antioxidant capacity. J. Agric. Food Chem. 2004, 52, 7846–7856. [Google Scholar] [CrossRef]
- De Pascual-Teresa, S.; Santos-Buelga, C.; Rivas-Gonzalo, J.C. Quantitative analysis of flavan-3-ols in Spanish foodstuffs and beverages. J. Agric. Food Chem. 2000, 48, 5331–5337. [Google Scholar] [CrossRef]
- Arts, I.C.; van de Putte, B.; Hollman, P.C. Catechin contents of foods commonly consumed in The Netherlands. 1. Fruits, vegetables, staple foods, and processed foods. J. Agric. Food Chem. 2000, 48, 1746–1751. [Google Scholar] [CrossRef]
- Vrhovsek, U.; Rigo, A.; Tonon, D.; Mattivi, F. Quantitation of polyphenols in different apple varieties. J. Agric. Food Chem. 2004, 52, 6532–6538. [Google Scholar] [CrossRef] [PubMed]
- Landberg, R.; Naidoo, N.; van Dam, R.M. Diet and endothelial function: From individual components to dietary patterns. Curr. Opin. Lipidol. 2012, 23, 147–155. [Google Scholar] [CrossRef] [PubMed]
- Mejri, F.; Selmi, S.; Martins, A.; Benkhoud, H.; Baati, T.; Chaabane, H.; Njim, L.; Serralheiro, M.L.M.; Rauter, A.P.; Hosni, K. Broad bean (Vicia faba L.) pods: A rich source of bioactive ingredients with antimicrobial, antioxidant, enzyme inhibitory, anti-diabetic and health-promoting properties. Food Funct. 2018, 9, 2051–2059. [Google Scholar] [CrossRef] [PubMed]
- Romani, A.; Mulinacci, N.; Pinelli, P.; Vincieri, F.F.; Cimato, A. Polyphenolic content in five tuscany cultivars of Olea europaea L. J. Agric. Food Chem. 1999, 47, 964–967. [Google Scholar] [CrossRef]
- Slimestad, R.; Fossen, T.; Vagen, I.M. Onions: A source of unique dietary flavonoids. J. Agric. Food Chem. 2007, 55, 10067–10080. [Google Scholar] [CrossRef]
- Pandjaitan, N.; Howard, L.R.; Morelock, T.; Gil, M.I. Antioxidant capacity and phenolic content of spinach as affected by genetics and maturation. J. Agric. Food Chem. 2005, 53, 8618–8623. [Google Scholar] [CrossRef]
- Fattorusso, E.; Iorizzi, M.; Lanzotti, V.; Taglialatela-Scafati, O. Chemical composition of shallot (Allium ascalonicum Hort.). J. Agric. Food Chem. 2002, 50, 5686–5690. [Google Scholar] [CrossRef]
- Rodriguez-Garcia, C.; Sanchez-Quesada, C. Dietary Flavonoids as Cancer Chemopreventive Agents: An Updated Review of Human Studies. Antioxidants 2019, 8, 137. [Google Scholar] [CrossRef] [Green Version]
- Yahfoufi, N.; Alsadi, N.; Jambi, M.; Matar, C. The Immunomodulatory and Anti-Inflammatory Role of Polyphenols. Nutrients 2018, 10, 1618. [Google Scholar] [CrossRef] [Green Version]
- 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]
- Chirumbolo, S.; Bjorklund, G.; Lysiuk, R.; Vella, A.; Lenchyk, L.; Upyr, T. Targeting Cancer with Phytochemicals via Their Fine Tuning of the Cell Survival Signaling Pathways. Int. J. Mol. Sci. 2018, 19, 3568. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Panche, A.N.; Diwan, A.D.; Chandra, S.R. Flavonoids: An overview. J. Nutr. Sci. 2016, 5, e47. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Durazzo, A.; Lucarini, M.; Souto, E.B.; Cicala, C.; Caiazzo, E.; Izzo, A.A.; Novellino, E.; Santini, A. Polyphenols: A concise overview on the chemistry, occurrence, and human health. Phytother. Res. 2019, 33, 2221–2243. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Rosen, T. Green tea catechins: Biologic properties, proposed mechanisms of action, and clinical implications. J. Drugs Derm. 2012, 11, e55–e60. [Google Scholar]
- Babu, P.V.; Liu, D. Green tea catechins and cardiovascular health: An update. Curr. Med. Chem. 2008, 15, 1840–1850. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Ravanel, P. Uncoupling activity of a series of flavones and flavonols on isolated plant mitochondria. Phytochemistry 1986, 25, 1015–1020. [Google Scholar] [CrossRef]
- van Dijk, C.; Driessen, A.J.; Recourt, K. The uncoupling efficiency and affinity of flavonoids for vesicles. Biochem. Pharmacol. 2000, 60, 1593–1600. [Google Scholar] [CrossRef] [Green Version]
- Dorta, D.J.; Pigoso, A.A.; Mingatto, F.E.; Rodrigues, T.; Prado, I.M.; Helena, A.F.; Uyemura, S.A.; Santos, A.C.; Curti, C. The interaction of flavonoids with mitochondria: Effects on energetic processes. Chem. Biol. Interact. 2005, 152, 67–78. [Google Scholar] [CrossRef]
- Scholz, S.; Williamson, G. Interactions affecting the bioavailability of dietary polyphenols in vivo. Int. J. Vitam Nutr. Res. 2007, 77, 224–235. [Google Scholar] [CrossRef]
- Jakobek, L. Interactions of polyphenols with carbohydrates, lipids and proteins. Food Chem. 2015, 175, 556–567. [Google Scholar] [CrossRef]
- Gonzales, G.B.; Smagghe, G.; Grootaert, C.; Zotti, M.; Raes, K.; Van Camp, J. Flavonoid interactions during digestion, absorption, distribution and metabolism: A sequential structure-activity/property relationship-based approach in the study of bioavailability and bioactivity. Drug Metab. Rev. 2015, 47, 175–190. [Google Scholar] [CrossRef] [PubMed]
- Swieca, M.; Gawlik-Dziki, U.; Dziki, D.; Baraniak, B.; Czyz, J. The influence of protein-flavonoid interactions on protein digestibility in vitro and the antioxidant quality of breads enriched with onion skin. Food Chem. 2013, 141, 451–458. [Google Scholar] [CrossRef] [PubMed]
- Hollman, P.C.; Katan, M.B. Absorption, metabolism and health effects of dietary flavonoids in man. Biomed. Pharmacother. 1997, 51, 305–310. [Google Scholar] [CrossRef]
- Cassidy, A.; Minihane, A.M. The role of metabolism (and the microbiome) in defining the clinical efficacy of dietary flavonoids. Am. J. Clin. Nutr. 2017, 105, 10–22. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Lotito, S.B.; Frei, B. Consumption of flavonoid-rich foods and increased plasma antioxidant capacity in humans: Cause, consequence, or epiphenomenon? Free Radic. Biol. Med. 2006, 41, 1727–1746. [Google Scholar] [CrossRef] [PubMed]
- Perez-Vizcaino, F.; Fraga, C.G. Research trends in flavonoids and health. Arch. Biochem. Biophys. 2018, 646, 107–112. [Google Scholar] [CrossRef]
- Gorlach, S.; Fichna, J.; Lewandowska, U. Polyphenols as mitochondria-targeted anticancer drugs. Cancer Lett. 2015, 366, 141–149. [Google Scholar] [CrossRef]
- Neagu, M.; Constantin, C.; Popescu, I.D.; Zipeto, D.; Tzanakakis, G.; Nikitovic, D.; Fenga, C.; Stratakis, C.A.; Spandidos, D.A.; Tsatsakis, A.M. Inflammation and Metabolism in Cancer Cell-Mitochondria Key Player. Front. Oncol. 2019, 9, 348. [Google Scholar] [CrossRef] [Green Version]
- Kroemer, G.; Pouyssegur, J. Tumor cell metabolism: cancer's Achilles' heel. Cancer Cell 2008, 13, 472–482. [Google Scholar] [CrossRef]
- Blackadar, C.B. Historical review of the causes of cancer. World J. Clin. Oncol. 2016, 7, 54–86. [Google Scholar] [CrossRef]
- Bock, F.J.; Tait, S.W.G. Mitochondria as multifaceted regulators of cell death. Nat. Rev. Mol. Cell Biol. 2019, 21, 85–100. [Google Scholar] [CrossRef] [PubMed]
- Lebelo, M.T.; Joubert, A.M.; Visagie, M.H. Warburg effect and its role in tumourigenesis. Arch. Pharmacal Res. 2019, 42, 833–847. [Google Scholar] [CrossRef]
- Chiche, J.; Brahimi-Horn, M.C.; Pouyssegur, J. Tumour hypoxia induces a metabolic shift causing acidosis: A common feature in cancer. J. Cell. Mol. Med. 2010, 14, 771–794. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Zaidi, N.; Lupien, L.; Kuemmerle, N.B.; Kinlaw, W.B.; Swinnen, J.V.; Smans, K. Lipogenesis and lipolysis: The pathways exploited by the cancer cells to acquire fatty acids. Prog. Lipid Res. 2013, 52, 585–589. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Weinberg, F.; Ramnath, N.; Nagrath, D. Reactive Oxygen Species in the Tumor Microenvironment: An Overview. Cancers 2019, 11, 1191. [Google Scholar] [CrossRef] [Green Version]
- Lu, S.; Wang, Y. Nonmetabolic functions of metabolic enzymes in cancer development. Cancer Commun. 2018, 38, 63. [Google Scholar] [CrossRef] [Green Version]
- Lee, N.; Kim, D. Cancer Metabolism: Fueling More than Just Growth. Mol. Cells 2016, 39, 847–854. [Google Scholar] [CrossRef]
- Vegliante, R.; Di Leo, L.; Ciccarone, F.; Ciriolo, M.R. Hints on ATGL implications in cancer: Beyond bioenergetic clues. Cell Death Dis. 2018, 9, 316. [Google Scholar] [CrossRef]
- Seyfried, T.N.; Flores, R.E.; Poff, A.M.; D’Agostino, D.P. Cancer as a metabolic disease: Implications for novel therapeutics. Carcinogenesis 2014, 35, 515–527. [Google Scholar] [CrossRef]
- Zhong, H.; Xiao, M.; Zarkovic, K.; Zhu, M.; Sa, R.; Lu, J.; Tao, Y.; Chen, Q.; Xia, L.; Cheng, S.; et al. Mitochondrial control of apoptosis through modulation of cardiolipin oxidation in hepatocellular carcinoma: A novel link between oxidative stress and cancer. Free Radic. Biol. Med. 2017, 102, 67–76. [Google Scholar] [CrossRef]
- Kiebish, M.A.; Han, X.; Cheng, H.; Chuang, J.H.; Seyfried, T.N. Cardiolipin and electron transport chain abnormalities in mouse brain tumor mitochondria: Lipidomic evidence supporting the Warburg theory of cancer. J. Lipid Res. 2008, 49, 2545–2556. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Murphy, M.P. How mitochondria produce reactive oxygen species. Biochem. J. 2009, 417, 1–13. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Hadi, S.M.; Asad, S.F.; Singh, S.; Ahmad, A. Putative mechanism for anticancer and apoptosis-inducing properties of plant-derived polyphenolic compounds. IUBMB Life 2000, 50, 167–171. [Google Scholar] [PubMed]
- Link, A.; Balaguer, F.; Goel, A. Cancer chemoprevention by dietary polyphenols: Promising role for epigenetics. Biochem. Pharmacol. 2010, 80, 1771–1792. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Youn, H.S.; Lee, J.Y.; Saitoh, S.I.; Miyake, K.; Kang, K.W.; Choi, Y.J.; Hwang, D.H. Suppression of MyD88- and TRIF-dependent signaling pathways of Toll-like receptor by (-)-epigallocatechin-3-gallate, a polyphenol component of green tea. Biochem. Pharmacol. 2006, 72, 850–859. [Google Scholar] [CrossRef]
- Fraga, C.G.; Galleano, M.; Verstraeten, S.V.; Oteiza, P.I. Basic biochemical mechanisms behind the health benefits of polyphenols. Mol. Asp. Med. 2010, 31, 435–445. [Google Scholar] [CrossRef]
- Oliveira-Marques, V.; Marinho, H.S.; Cyrne, L.; Antunes, F. Modulation of NF-kappaB-dependent gene expression by H2O2: A major role for a simple chemical process in a complex biological response. Antioxid Redox Signal 2009, 11, 2043–2053. [Google Scholar] [CrossRef]
- Valko, M.; Leibfritz, D.; Moncol, J.; Cronin, M.T.; Mazur, M.; Telser, J. Free radicals and antioxidants in normal physiological functions and human disease. Int. J. Biochem. Cell Biol. 2007, 39, 44–84. [Google Scholar] [CrossRef]
- Kaushik, S.; Shyam, H.; Agarwal, S.; Sharma, R.; Nag, T.C.; Dwivedi, A.K.; Balapure, A.K. Genistein potentiates Centchroman induced antineoplasticity in breast cancer via PI3K/Akt deactivation and ROS dependent induction of apoptosis. Life Sci. 2019, 117073. [Google Scholar] [CrossRef]
- Jin, S.; Zhang, Q.Y.; Kang, X.M.; Wang, J.X.; Zhao, W.H. Daidzein induces MCF-7 breast cancer cell apoptosis via the mitochondrial pathway. Ann. Oncol. 2010, 21, 263–268. [Google Scholar] [CrossRef]
- Pandey, P.; Sayyed, U.; Tiwari, R.K.; Siddiqui, M.H.; Pathak, N.; Bajpai, P. Hesperidin Induces ROS-Mediated Apoptosis along with Cell Cycle Arrest at G2/M Phase in Human Gall Bladder Carcinoma. Nutr. Cancer 2019, 71, 676–687. [Google Scholar] [CrossRef] [PubMed]
- Wu, D.; Zhang, J.; Wang, J.; Li, J.; Liao, F.; Dong, W. Hesperetin induces apoptosis of esophageal cancer cells via mitochondrial pathway mediated by the increased intracellular reactive oxygen species. Tumour Biol. 2016, 37, 3451–3459. [Google Scholar] [CrossRef] [PubMed]
- Zhang, J.; Song, J.; Wu, D.; Wang, J.; Dong, W. Hesperetin induces the apoptosis of hepatocellular carcinoma cells via mitochondrial pathway mediated by the increased intracellular reactive oxygen species, ATP and calcium. Med. Oncol. 2015, 32, 101. [Google Scholar] [CrossRef] [PubMed]
- Palit, S.; Kar, S.; Sharma, G.; Das, P.K. Hesperetin Induces Apoptosis in Breast Carcinoma by Triggering Accumulation of ROS and Activation of ASK1/JNK Pathway. J. Cell. Physiol. 2015, 230, 1729–1739. [Google Scholar] [CrossRef] [PubMed]
- Park, S.; Lim, W.; Bazer, F.W.; Song, G. Naringenin suppresses growth of human placental choriocarcinoma via reactive oxygen species-mediated P38 and JNK MAPK pathways. Phytomedicine 2018, 50, 238–246. [Google Scholar] [CrossRef] [PubMed]
- Ahamad, M.S.; Siddiqui, S.; Jafri, A.; Ahmad, S.; Afzal, M.; Arshad, M. Induction of apoptosis and antiproliferative activity of naringenin in human epidermoid carcinoma cell through ROS generation and cell cycle arrest. PLoS ONE 2014, 9, e110003. [Google Scholar] [CrossRef] [Green Version]
- Lim, W.; Park, S.; Bazer, F.W.; Song, G. Naringenin-Induced Apoptotic Cell Death in Prostate Cancer Cells Is Mediated via the PI3K/AKT and MAPK Signaling Pathways. J. Cell. Biochem. 2017, 118, 1118–1131. [Google Scholar] [CrossRef]
- Zaidun, N.H.; Thent, Z.C.; Latiff, A.A. Combating oxidative stress disorders with citrus flavonoid: Naringenin. Life Sci. 2018, 208, 111–122. [Google Scholar] [CrossRef]
- Taparia, S.S.; Khanna, A. Procyanidin-rich extract of natural cocoa powder causes ROS-mediated caspase-3 dependent apoptosis and reduction of pro-MMP-2 in epithelial ovarian carcinoma cell lines. Biomed. Pharmacother. 2016, 83, 130–140. [Google Scholar] [CrossRef]
- Martin, M.A.; Serrano, A.B.; Ramos, S.; Pulido, M.I.; Bravo, L.; Goya, L. Cocoa flavonoids up-regulate antioxidant enzyme activity via the ERK1/2 pathway to protect against oxidative stress-induced apoptosis in HepG2 cells. J. Nutr. Biochem. 2010, 21, 196–205. [Google Scholar] [CrossRef]
- Rodriguez-Ramiro, I.; Martin, M.A.; Ramos, S.; Bravo, L.; Goya, L. Comparative effects of dietary flavanols on antioxidant defences and their response to oxidant-induced stress on Caco2 cells. Eur. J. Nutr. 2011, 50, 313–322. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Martin, M.A.; Goya, L.; Ramos, S. Preventive Effects of Cocoa and Cocoa Antioxidants in Colon Cancer. Diseases 2016, 4. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Martin, M.A.; Goya, L.; Ramos, S. Potential for preventive effects of cocoa and cocoa polyphenols in cancer. Food Chem. Toxicol. 2013, 56, 336–351. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Rather, R.A.; Bhagat, M. Quercetin as an innovative therapeutic tool for cancer chemoprevention: Molecular mechanisms and implications in human health. Cancer Med. 2019. [Google Scholar] [CrossRef] [PubMed]
- 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. Pharmacother. 2019, 121, 109604. [Google Scholar] [CrossRef] [PubMed]
- Jeon, J.S.; Kwon, S.; Ban, K.; Kwon Hong, Y.; Ahn, C.; Sung, J.S.; Choi, I. Regulation of the Intracellular ROS Level Is Critical for the Antiproliferative Effect of Quercetin in the Hepatocellular Carcinoma Cell Line HepG2. Nutr. Cancer 2019, 71, 861–869. [Google Scholar] [CrossRef]
- Shang, H.S.; Lu, H.F.; Lee, C.H.; Chiang, H.S.; Chu, Y.L.; Chen, A.; Lin, Y.F.; Chung, J.G. Quercetin induced cell apoptosis and altered gene expression in AGS human gastric cancer cells. Environ. Toxicol. 2018, 33, 1168–1181. [Google Scholar] [CrossRef] [PubMed]
- Wu, Q.; Kroon, P.A.; Shao, H.; Needs, P.W.; Yang, X. Differential Effects of Quercetin and Two of Its Derivatives, Isorhamnetin and Isorhamnetin-3-glucuronide, in Inhibiting the Proliferation of Human Breast-Cancer MCF-7 Cells. J. Agric. Food Chem. 2018, 66, 7181–7189. [Google Scholar] [CrossRef]
- Wu, P.; Meng, X.; Zheng, H.; Zeng, Q.; Chen, T.; Wang, W.; Zhang, X.; Su, J. Kaempferol Attenuates ROS-Induced Hemolysis and the Molecular Mechanism of Its Induction of Apoptosis on Bladder Cancer. Molecules 2018, 23, 2592. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Choi, J.B.; Kim, J.H.; Lee, H.; Pak, J.N.; Shim, B.S.; Kim, S.H. Reactive Oxygen Species and p53 Mediated Activation of p38 and Caspases is Critically Involved in Kaempferol Induced Apoptosis in Colorectal Cancer Cells. J. Agric. Food Chem. 2018, 66, 9960–9967. [Google Scholar] [CrossRef] [PubMed]
- Seydi, E.; Salimi, A.; Rasekh, H.R.; Mohsenifar, Z.; Pourahmad, J. Selective Cytotoxicity of Luteolin and Kaempferol on Cancerous Hepatocytes Obtained from Rat Model of Hepatocellular Carcinoma: Involvement of ROS-Mediated Mitochondrial Targeting. Nutr. Cancer 2018, 70, 594–604. [Google Scholar] [CrossRef] [PubMed]
- Tavsan, Z.; Kayali, H.A. Flavonoids showed anticancer effects on the ovarian cancer cells: Involvement of reactive oxygen species, apoptosis, cell cycle and invasion. Biomed. Pharmacother. 2019, 116, 109004. [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]
- Souza, R.P.; Bonfim-Mendonca, P.S.; Gimenes, F.; Ratti, B.A.; Kaplum, V.; Bruschi, M.L.; Nakamura, C.V.; Silva, S.O.; Maria-Engler, S.S.; Consolaro, M.E. Oxidative Stress Triggered by Apigenin Induces Apoptosis in a Comprehensive Panel of Human Cervical Cancer-Derived Cell Lines. Oxidative Med. Cell. Longev. 2017, 2017, 1512745. [Google Scholar] [CrossRef]
- Park, W.; Park, S.; Lim, W.; Song, G. Chrysin disrupts intracellular homeostasis through mitochondria-mediated cell death in human choriocarcinoma cells. Biochem. Biophys. Res. Commun. 2018, 503, 3155–3161. [Google Scholar] [CrossRef] [PubMed]
- Xu, Y.; Tong, Y.; Ying, J.; Lei, Z.; Wan, L.; Zhu, X.; Ye, F.; Mao, P.; Wu, X.; Pan, R.; et al. Chrysin induces cell growth arrest, apoptosis, and ER stress and inhibits the activation of STAT3 through the generation of ROS in bladder cancer cells. Oncol. Lett. 2018, 15, 9117–9125. [Google Scholar] [CrossRef]
- Lim, W.; Ryu, S.; Bazer, F.W.; Kim, S.M.; Song, G. Chrysin attenuates progression of ovarian cancer cells by regulating signaling cascades and mitochondrial dysfunction. J. Cell. Physiol. 2018, 233, 3129–3140. [Google Scholar] [CrossRef]
- Alipour, B.; Rashidkhani, B.; Edalati, S. Dietary flavonoid intake, total antioxidant capacity and lipid oxidative damage: A cross-sectional study of Iranian women. Nutrition 2016, 32, 566–572. [Google Scholar] [CrossRef]
- Sorrenti, V.; Vanella, L.; Acquaviva, R.; Cardile, V.; Giofre, S.; Di Giacomo, C. Cyanidin induces apoptosis and differentiation in prostate cancer cells. Int. J. Oncol. 2015, 47, 1303–1310. [Google Scholar] [CrossRef] [Green Version]
- Cvorovic, J.; Tramer, F.; Granzotto, M.; Candussio, L.; Decorti, G.; Passamonti, S. Oxidative stress-based cytotoxicity of delphinidin and cyanidin in colon cancer cells. Arch. Biochem. Biophys. 2010, 501, 151–157. [Google Scholar] [CrossRef]
- McArthur, K.; Kile, B.T. Apoptotic Caspases: Multiple or Mistaken Identities? Trends Cell Biol. 2018, 28, 475–493. [Google Scholar] [CrossRef] [PubMed]
- Jan, R.; Chaudhry, G.E. Understanding Apoptosis and Apoptotic Pathways Targeted Cancer Therapeutics. Adv. Pharm. Bull. 2019, 9, 205–218. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Choi, E.J.; Jung, J.Y.; Kim, G.H. Genistein inhibits the proliferation and differentiation of MCF-7 and 3T3-L1 cells via the regulation of ERalpha expression and induction of apoptosis. Exp. Ther. Med. 2014, 8, 454–458. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Solomon, L.A.; Ali, S.; Banerjee, S.; Munkarah, A.R.; Morris, R.T.; Sarkar, F.H. Sensitization of ovarian cancer cells to cisplatin by genistein: The role of NF-kappaB. J. Ovarian Res. 2008, 1, 9. [Google Scholar] [CrossRef] [Green Version]
- Shafiee, G.; Saidijam, M.; Tavilani, H.; Ghasemkhani, N.; Khodadadi, I. Genistein Induces Apoptosis and Inhibits Proliferation of HT29 Colon Cancer Cells. Int. J. Mol. Cell. Med. 2016, 5, 178–191. [Google Scholar] [PubMed]
- Qin, J.; Teng, J.; Zhu, Z.; Chen, J.; Huang, W.J. Genistein induces activation of the mitochondrial apoptosis pathway by inhibiting phosphorylation of Akt in colorectal cancer cells. Pharm. Biol. 2016, 54, 74–79. [Google Scholar] [CrossRef] [Green Version]
- Danciu, C.; Avram, S.; Pavel, I.Z.; Ghiulai, R.; Dehelean, C.A.; Ersilia, A.; Minda, D.; Petrescu, C.; Moaca, E.A.; Soica, C. Main Isoflavones Found in Dietary Sources as Natural Anti-inflammatory Agents. Curr. Drug Targets 2018, 19, 841–853. [Google Scholar] [CrossRef]
- Liu, X.; Suzuki, N.; Santosh Laxmi, Y.R.; Okamoto, Y.; Shibutani, S. Anti-breast cancer potential of daidzein in rodents. Life Sci. 2012, 91, 415–419. [Google Scholar] [CrossRef]
- Park, H.J.; Jeon, Y.K.; You, D.H.; Nam, M.J. Daidzein causes cytochrome c-mediated apoptosis via the Bcl-2 family in human hepatic cancer cells. Food Chem. Toxicol. 2013, 60, 542–549. [Google Scholar] [CrossRef]
- Zhang, J.; Wu, D.; Vikash; Song, J.; Wang, J.; Yi, J.; Dong, W. Hesperetin Induces the Apoptosis of Gastric Cancer Cells via Activating Mitochondrial Pathway by Increasing Reactive Oxygen Species. Dig. Dis. Sci. 2015, 60, 2985–2995. [Google Scholar] [CrossRef]
- Sivagami, G.; Vinothkumar, R.; Bernini, R.; Preethy, C.P.; Riyasdeen, A.; Akbarsha, M.A.; Menon, V.P.; Nalini, N. Role of hesperetin (a natural flavonoid) and its analogue on apoptosis in HT-29 human colon adenocarcinoma cell line--a comparative study. Food Chem. Toxicol. 2012, 50, 660–671. [Google Scholar] [CrossRef] [PubMed]
- Elango, R.; Athinarayanan, J.; Subbarayan, V.P.; Lei, D.K.Y.; Alshatwi, A.A. Hesperetin induces an apoptosis-triggered extrinsic pathway and a p53- independent pathway in human lung cancer H522 cells. J. Asian Nat. Prod. Res. 2018, 20, 559–569. [Google Scholar] [CrossRef] [PubMed]
- Sambantham, S.; Radha, M.; Paramasivam, A.; Anandan, B.; Malathi, R.; Chandra, S.R.; Jayaraman, G. Molecular mechanism underlying hesperetin-induced apoptosis by in silico analysis and in prostate cancer PC-3 cells. Asian Pac. J. Cancer Prev. 2013, 14, 4347–4352. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Bao, L.; Liu, F.; Guo, H.B.; Li, Y.; Tan, B.B.; Zhang, W.X.; Peng, Y.H. Naringenin inhibits proliferation, migration, and invasion as well as induces apoptosis of gastric cancer SGC7901 cell line by downregulation of AKT pathway. Tumour Biol. 2016, 37, 11365–11374. [Google Scholar] [CrossRef]
- Zhang, H.; Zhong, X.; Zhang, X.; Shang, D.; Zhou, Y.I.; Zhang, C. Enhanced anticancer effect of ABT-737 in combination with naringenin on gastric cancer cells. Exp. Ther. Med. 2016, 11, 669–673. [Google Scholar] [CrossRef]
- Shirakami, Y.; Sakai, H.; Kochi, T.; Seishima, M.; Shimizu, M. Catechins and Its Role in Chronic Diseases. Adv. Exp. Med. Biol. 2016, 929, 67–90. [Google Scholar]
- Moradzadeh, M.; Hosseini, A.; Erfanian, S.; Rezaei, H. Epigallocatechin-3-gallate promotes apoptosis in human breast cancer T47D cells through down-regulation of PI3K/AKT and Telomerase. Pharmacol. Rep. 2017, 69, 924–928. [Google Scholar] [CrossRef]
- Chen, L.; Zhang, H.Y. Cancer preventive mechanisms of the green tea polyphenol (-)-epigallocatechin-3-gallate. Molecules 2007, 12, 946–957. [Google Scholar] [CrossRef] [Green Version]
- Liu, J.; Xing, J.; Fei, Y. Green tea (Camellia sinensis) and cancer prevention: A systematic review of randomized trials and epidemiological studies. Chin. Med. 2008, 3, 12. [Google Scholar] [CrossRef] [Green Version]
- Sutherland, B.A.; Rahman, R.M.; Appleton, I. Mechanisms of action of green tea catechins, with a focus on ischemia-induced neurodegeneration. J. Nutr. Biochem. 2006, 17, 291–306. [Google Scholar] [CrossRef]
- Granado-Serrano, A.B.; Martin, M.A.; Izquierdo-Pulido, M.; Goya, L.; Bravo, L.; Ramos, S. Molecular mechanisms of (-)-epicatechin and chlorogenic acid on the regulation of the apoptotic and survival/proliferation pathways in a human hepatoma cell line. J. Agric. Food Chem. 2007, 55, 2020–2027. [Google Scholar] [CrossRef] [PubMed]
- Granado-Serrano, A.B.; Martin, M.A.; Haegeman, G.; Goya, L.; Bravo, L.; Ramos, S. Epicatechin induces NF-kappaB, activator protein-1 (AP-1) and nuclear transcription factor erythroid 2p45-related factor-2 (Nrf2) via phosphatidylinositol-3-kinase/protein kinase B (PI3K/AKT) and extracellular regulated kinase (ERK) signalling in HepG2 cells. Br. J. Nutr. 2010, 103, 168–179. [Google Scholar] [PubMed] [Green Version]
- Ranganathan, S.; Halagowder, D.; Sivasithambaram, N.D. Quercetin Suppresses Twist to Induce Apoptosis in MCF-7 Breast Cancer Cells. PLoS ONE 2015, 10, e0141370. [Google Scholar] [CrossRef] [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] [PubMed]
- 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.; et al. 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] [PubMed]
- Wang, P.; Heber, D.; Henning, S.M. Quercetin increased the antiproliferative activity of green tea polyphenol (-)-epigallocatechin gallate in prostate cancer cells. Nutr. Cancer 2012, 64, 580–587. [Google Scholar] [CrossRef] [PubMed]
- 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] [Green Version]
- Granado-Serrano, A.B.; Martin, M.A.; Bravo, L.; Goya, L.; Ramos, S. Quercetin induces apoptosis via caspase activation, regulation of Bcl-2, and inhibition of PI-3-kinase/Akt and ERK pathways in a human hepatoma cell line (HepG2). J. Nutr. 2006, 136, 2715–2721. [Google Scholar] [CrossRef] [Green Version]
- Sun, S.; Gong, F.; Liu, P.; Miao, Q. Metformin combined with quercetin synergistically repressed prostate cancer cells via inhibition of VEGF/PI3K/Akt signaling pathway. Gene 2018, 664, 50–57. [Google Scholar] [CrossRef]
- Imran, M.; Rauf, A.; Shah, Z.A.; Saeed, F.; Imran, A.; Arshad, M.U.; Ahmad, B.; Bawazeer, S.; Atif, M.; Peters, D.G.; et al. Chemo-preventive and therapeutic effect of the dietary flavonoid kaempferol: A comprehensive review. Phytother. Res. 2019, 33, 263–275. [Google Scholar] [CrossRef]
- Luo, H.; Rankin, G.O.; Li, Z.; Depriest, L.; Chen, Y.C. Kaempferol induces apoptosis in ovarian cancer cells through activating p53 in the intrinsic pathway. Food Chem. 2011, 128, 513–519. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Kashafi, E.; Moradzadeh, M.; Mohamadkhani, A.; Erfanian, S. Kaempferol increases apoptosis in human cervical cancer HeLa cells via PI3K/AKT and telomerase pathways. Biomed. Pharmacother. 2017, 89, 573–577. [Google Scholar] [CrossRef] [PubMed]
- Dean, M.; Murphy, B.T.; Burdette, J.E. Phytosteroids beyond estrogens: Regulators of reproductive and endocrine function in natural products. Mol. Cell. Endocrinol. 2017, 442, 98–105. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Pandey, M.; Kaur, P.; Shukla, S.; Abbas, A.; Fu, P.; Gupta, S. Plant flavone apigenin inhibits HDAC and remodels chromatin to induce growth arrest and apoptosis in human prostate cancer cells: In vitro and in vivo study. Mol. Carcinog. 2012, 51, 952–962. [Google Scholar] [CrossRef] [Green Version]
- 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]
- Meng, S.; Zhu, Y.; Li, J.F.; Wang, X.; Liang, Z.; Li, S.Q.; Xu, X.; Chen, H.; Liu, B.; Zheng, X.Y.; et al. Apigenin inhibits renal cell carcinoma cell proliferation. Oncotarget 2017, 8, 19834–19842. [Google Scholar] [CrossRef] [Green Version]
- Shi, M.D.; Shiao, C.K.; Lee, Y.C.; Shih, Y.W. Apigenin, a dietary flavonoid, inhibits proliferation of human bladder cancer T-24 cells via blocking cell cycle progression and inducing apoptosis. Cancer Cell Int. 2015, 15, 33. [Google Scholar] [CrossRef] [Green Version]
- Zhu, Y.; Mao, Y.; Chen, H.; Lin, Y.; Hu, Z.; Wu, J.; Xu, X.; Xu, X.; Qin, J.; Xie, L. Apigenin promotes apoptosis, inhibits invasion and induces cell cycle arrest of T24 human bladder cancer cells. Cancer Cell Int. 2013, 13, 54. [Google Scholar] [CrossRef] [Green Version]
- Liu, R.; Ji, P.; Liu, B.; Qiao, H.; Wang, X.; Zhou, L.; Deng, T.; Ba, Y. Apigenin enhances the cisplatin cytotoxic effect through p53-modulated apoptosis. Oncol. Lett. 2017, 13, 1024–1030. [Google Scholar] [CrossRef] [Green Version]
- Khoo, B.Y.; Chua, S.L.; Balaram, P. Apoptotic effects of chrysin in human cancer cell lines. Int. J. Mol. Sci. 2010, 11, 2188–2199. [Google Scholar] [CrossRef] [Green Version]
- Xue, C.; Chen, Y.; Hu, D.N.; Iacob, C.; Lu, C.; Huang, Z. Chrysin induces cell apoptosis in human uveal melanoma cells via intrinsic apoptosis. Oncol. Lett. 2016, 12, 4813–4820. [Google Scholar] [CrossRef] [PubMed]
- Chen, Y.; Wang, S.; Geng, B.; Yi, Z. Pelargonidin induces antitumor effects in human osteosarcoma cells via autophagy induction, loss of mitochondrial membrane potential, G2/M cell cycle arrest and downregulation of PI3K/AKT signalling pathway. J. BUON 2018, 23, 735–740. [Google Scholar] [PubMed]
- Lopez de Las Hazas, M.C.; Mosele, J.I.; Macia, A.; Ludwig, I.A.; Motilva, M.J. Exploring the Colonic Metabolism of Grape and Strawberry Anthocyanins and Their in Vitro Apoptotic Effects in HT-29 Colon Cancer Cells. J. Agric. Food Chem. 2017, 65, 6477–6487. [Google Scholar] [CrossRef] [PubMed]
- Hosseini, M.M.; Karimi, A.; Behroozaghdam, M.; Javidi, M.A.; Ghiasvand, S.; Bereimipour, A.; Aryan, H.; Nassiri, F.; Jangholi, E. Cytotoxic and Apoptogenic Effects of Cyanidin-3-Glucoside on the Glioblastoma Cell Line. World Neurosurg. 2017, 108, 94–100. [Google Scholar] [CrossRef] [PubMed]
- Lim, W.; Song, G. Inhibitory effects of delphinidin on the proliferation of ovarian cancer cells via PI3K/AKT and ERK 1/2 MAPK signal transduction. Oncol. Lett. 2017, 14, 810–818. [Google Scholar] [CrossRef] [Green Version]
- Pal, H.C.; Sharma, S.; Strickland, L.R.; Agarwal, J.; Athar, M.; Elmets, C.A.; Afaq, F. Delphinidin reduces cell proliferation and induces apoptosis of non-small-cell lung cancer cells by targeting EGFR/VEGFR2 signaling pathways. PLoS ONE 2013, 8, e77270. [Google Scholar] [CrossRef] [Green Version]
- Gupta, S.C.; Kunnumakkara, A.B.; Aggarwal, S.; Aggarwal, B.B. Inflammation, a Double-Edge Sword for Cancer and Other Age-Related Diseases. Front. Immunol. 2018, 9, 2160. [Google Scholar] [CrossRef]
- Perez-Cano, F.J.; Castell, M. Flavonoids, Inflammation and Immune System. Nutrients 2016, 8, 659. [Google Scholar] [CrossRef]
- Ding, S.; Jiang, H.; Fang, J. Regulation of Immune Function by Polyphenols. J. Immunol. Res. 2018, 2018, 1264074. [Google Scholar] [CrossRef] [Green Version]
- Hosseinzade, A.; Sadeghi, O.; Naghdipour Biregani, A.; Soukhtehzari, S.; Brandt, G.S.; Esmaillzadeh, A. Immunomodulatory Effects of Flavonoids: Possible Induction of T CD4+ Regulatory Cells Through Suppression of mTOR Pathway Signaling Activity. Front. Immunol. 2019, 10, 51. [Google Scholar] [CrossRef] [Green Version]
- Mooradian, M.J.; Sullivan, R.J. Immunomodulatory effects of current cancer treatment and the consequences for follow-up immunotherapeutics. Future Oncol. 2017, 13, 1649–1663. [Google Scholar] [CrossRef] [PubMed]
- Xu, L.; Zhang, Y.; Tian, K.; Chen, X.; Zhang, R.; Mu, X.; Wu, Y.; Wang, D.; Wang, S.; Liu, F.; et al. Apigenin suppresses PD-L1 expression in melanoma and host dendritic cells to elicit synergistic therapeutic effects. J. Exp. Clin. Cancer Res. 2018, 37, 261. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Li, W.; Kim, T.I.; Kim, J.H.; Chung, H.S. Immune Checkpoint PD-1/PD-L1 CTLA-4/CD80 are Blocked by Rhus verniciflua Stokes and its Active Compounds. Molecules 2019, 24, 4062. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Bao, F.; Bai, H.Y.; Wu, Z.R.; Yang, Z.G. Phenolic compounds from cultivated Glycyrrhiza uralensis and their PD-1/PD-L1 inhibitory activities. Nat. Prod. Res. 2019, 1–8. [Google Scholar] [CrossRef] [PubMed]
- Sundaram, M.K.; Unni, S.; Somvanshi, P.; Bhardwaj, T.; Mandal, R.K.; Hussain, A.; Haque, S. Genistein Modulates Signaling Pathways and Targets Several Epigenetic Markers in HeLa Cells. Genes 2019, 10, 955. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Pons, D.G.; Vilanova-Llompart, J.; Gaya-Bover, A.; Alorda-Clara, M.; Oliver, J.; Roca, P.; Sastre-Serra, J. The phytoestrogen genistein affects inflammatory-related genes expression depending on the ERalpha/ERbeta ratio in breast cancer cells. Int. J. Food Sci. Nutr. 2019, 70, 941–949. [Google Scholar] [CrossRef]
- Ning, Y.; Feng, W.; Cao, X.; Ren, K.; Quan, M.; Chen, A.; Xu, C.; Qiu, Y.; Cao, J.; Li, X.; et al. Genistein inhibits stemness of SKOV3 cells induced by macrophages co-cultured with ovarian cancer stem-like cells through IL-8/STAT3 axis. J. Exp. Clin. Cancer Res. 2019, 38, 19. [Google Scholar] [CrossRef] [Green Version]
- Sakamoto, Y.; Kanatsu, J.; Toh, M.; Naka, A.; Kondo, K.; Iida, K. The Dietary Isoflavone Daidzein Reduces Expression of Pro-Inflammatory Genes through PPARalpha/gamma and JNK Pathways in Adipocyte and Macrophage Co-Cultures. PLoS ONE 2016, 11, e0149676. [Google Scholar] [CrossRef]
- Ren, H.; Hao, J.; Liu, T.; Zhang, D.; Lv, H.; Song, E.; Zhu, C. Hesperetin Suppresses Inflammatory Responses in Lipopolysaccharide-Induced RAW 264.7 Cells via the Inhibition of NF-kappaB and Activation of Nrf2/HO-1 Pathways. Inflammation 2016, 39, 964–973. [Google Scholar]
- Nalini, N.; Aranganathan, S.; Kabalimurthy, J. Chemopreventive efficacy of hesperetin (citrus flavonone) against 1,2-dimethylhydrazine-induced rat colon carcinogenesis. Toxicol. Mech. Methods 2012, 22, 397–408. [Google Scholar] [CrossRef]
- Ahmed, O.M.; Ahmed, A.A.; Fahim, H.I.; Zaky, M.Y. Quercetin and naringenin abate diethylnitrosamine/acetylaminofluorene-induced hepatocarcinogenesis in Wistar rats: The roles of oxidative stress, inflammation and cell apoptosis. Drug Chem. Toxicol. 2019. [Google Scholar] [CrossRef] [PubMed]
- Zhao, Z.; Jin, G.; Ge, Y.; Guo, Z. Naringenin inhibits migration of breast cancer cells via inflammatory and apoptosis cell signaling pathways. Inflammopharmacology 2019, 27, 1021–1036. [Google Scholar] [CrossRef] [PubMed]
- Chen, Y.Y.; Chang, Y.M.; Wang, K.Y.; Chen, P.N.; Hseu, Y.C.; Chen, K.M.; Yeh, K.T.; Chen, C.J.; Hsu, L.S. Naringenin inhibited migration and invasion of glioblastoma cells through multiple mechanisms. Environ. Toxicol. 2019, 34, 233–239. [Google Scholar] [CrossRef] [PubMed]
- Rodriguez-Ramiro, I.; Ramos, S.; Lopez-Oliva, E.; Agis-Torres, A.; Bravo, L.; Goya, L.; Martin, M.A. Cocoa polyphenols prevent inflammation in the colon of azoxymethane-treated rats and in TNF-alpha-stimulated Caco-2 cells. Br. J. Nutr. 2013, 110, 206–215. [Google Scholar] [CrossRef] [Green Version]
- 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. Pharmacother. 2017, 92, 33–38. [Google Scholar] [CrossRef]
- 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]
- Han, M.; Song, Y.; Zhang, X. Quercetin Suppresses the Migration and Invasion in Human Colon Cancer Caco-2 Cells Through Regulating Toll-like Receptor 4/Nuclear Factor-kappa B Pathway. Pharmacogn. Mag. 2016, 12, S237–S244. [Google Scholar]
- Bruning, A. Inhibition of mTOR signaling by quercetin in cancer treatment and prevention. Anti Cancer Agents Med. Chem. 2013, 13, 1025–1031. [Google Scholar] [CrossRef]
- Lee, S.; Kim, Y.J.; Kwon, S.; Lee, Y.; Choi, S.Y.; Park, J.; Kwon, H.J. Inhibitory effects of flavonoids on TNF-alpha-induced IL-8 gene expression in HEK 293 cells. BMB Rep. 2009, 42, 265–270. [Google Scholar] [CrossRef] [Green Version]
- Ren, J.; Lu, Y.; Qian, Y.; Chen, B.; Wu, T.; Ji, G. Recent progress regarding kaempferol for the treatment of various diseases. Exp. Ther. Med. 2019, 18, 2759–2776. [Google Scholar] [CrossRef] [Green Version]
- Devi, K.P.; Malar, D.S.; Nabavi, S.F.; Sureda, A.; Xiao, J.; Nabavi, S.M.; Daglia, M. Kaempferol and inflammation: From chemistry to medicine. Pharmacol. Res. 2015, 99, 1–10. [Google Scholar] [CrossRef] [PubMed]
- Kadioglu, O.; Nass, J.; Saeed, M.E.; Schuler, B.; Efferth, T. Kaempferol Is an Anti-Inflammatory Compound with Activity towards NF-kappaB Pathway Proteins. Anticancer Res. 2015, 35, 2645–2650. [Google Scholar] [PubMed]
- Ghitu, A.; Schwiebs, A.; Radeke, H.H.; Avram, S.; Zupko, I.; Bor, A.; Pavel, I.Z.; Dehelean, C.A.; Oprean, C.; Bojin, F.; et al. A Comprehensive Assessment of Apigenin as an Antiproliferative, Proapoptotic, Antiangiogenic and Immunomodulatory Phytocompound. Nutrients 2019, 11, 858. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Ai, X.Y.; Qin, Y.; Liu, H.J.; Cui, Z.H.; Li, M.; Yang, J.H.; Zhong, W.L.; Liu, Y.R.; Chen, S.; Sun, T.; et al. Apigenin inhibits colonic inflammation and tumorigenesis by suppressing STAT3-NF-kappaB signaling. Oncotarget 2017, 8, 100216–100226. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Ginwala, R.; Bhavsar, R.; Chigbu, D.I.; Jain, P.; Khan, Z.K. Potential Role of Flavonoids in Treating Chronic Inflammatory Diseases with a Special Focus on the Anti-Inflammatory Activity of Apigenin. Antioxidants 2019, 8, 35. [Google Scholar] [CrossRef] [Green Version]
- Rehman, M.U.; Tahir, M.; Khan, A.Q.; Khan, R.; Lateef, A.; Oday, O.H.; Qamar, W.; Ali, F.; Sultana, S. Chrysin suppresses renal carcinogenesis via amelioration of hyperproliferation, oxidative stress and inflammation: Plausible role of NF-kappaB. Toxicol. Lett. 2013, 216, 146–158. [Google Scholar] [CrossRef]
- Liu, C.; Zhu, L.; Fukuda, K.; Ouyang, S.; Chen, X.; Wang, C.; Zhang, C.J.; Martin, B.; Gu, C.; Qin, L.; et al. The flavonoid cyanidin blocks binding of the cytokine interleukin-17A to the IL-17RA subunit to alleviate inflammation in vivo. Sci. Signal. 2017, 10, eaaf8823. [Google Scholar] [CrossRef] [Green Version]
- Lee, B.S.; Lee, C.; Yang, S.; Park, E.K.; Ku, S.K.; Bae, J.S. Suppressive effects of pelargonidin on lipopolysaccharide-induced inflammatory responses. Chem. Biol. Interact. 2019, 302, 67–73. [Google Scholar] [CrossRef]
- Venancio, V.P.; Cipriano, P.A.; Kim, H.; Antunes, L.M.; Talcott, S.T.; Mertens-Talcott, S.U. Cocoplum (Chrysobalanus icaco L.) anthocyanins exert anti-inflammatory activity in human colon cancer and non-malignant colon cells. Food Funct. 2017, 8, 307–314. [Google Scholar] [CrossRef]
- Im, N.K.; Jang, W.J.; Jeong, C.H.; Jeong, G.S. Delphinidin suppresses PMA-induced MMP-9 expression by blocking the NF-kappaB activation through MAPK signaling pathways in MCF-7 human breast carcinoma cells. J. Med. Food 2014, 17, 855–861. [Google Scholar] [CrossRef]
- Porporato, P.E.; Filigheddu, N.; Pedro, J.M.B.; Kroemer, G.; Galluzzi, L. Mitochondrial metabolism and cancer. Cell Res. 2018, 28, 265–280. [Google Scholar] [CrossRef]
- Kwak, T.W.; Park, S.B.; Kim, H.J.; Jeong, Y.I.; Kang, D.H. Anticancer activities of epigallocatechin-3-gallate against cholangiocarcinoma cells. Oncotargets Ther. 2017, 10, 137–144. [Google Scholar] [CrossRef] [PubMed]
- Klingenberg, M. The ADP and ATP transport in mitochondria and its carrier. Biochim. Biophys. Acta 2008, 1778, 1978–2021. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Halestrap, A.P. Mitochondrial permeability: Dual role for the ADP/ATP translocator? Nature 2004, 430, 984. [Google Scholar] [CrossRef]
- Ortega, R.; Garcia, N. The flavonoid quercetin induces changes in mitochondrial permeability by inhibiting adenine nucleotide translocase. J. Bioenerg. Biomembr. 2009, 41, 41–47. [Google Scholar] [CrossRef] [PubMed]
- Oishi, M.; Iizumi, Y.; Taniguchi, T.; Goi, W.; Miki, T.; Sakai, T. Apigenin sensitizes prostate cancer cells to Apo2L/TRAIL by targeting adenine nucleotide translocase-2. PLoS ONE 2013, 8, e55922. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Moini, H.; Arroyo, A.; Vaya, J.; Packer, L. Bioflavonoid effects on the mitochondrial respiratory electron transport chain and cytochrome c redox state. Redox Rep. 1999, 4, 35–41. [Google Scholar] [CrossRef]
- Salvi, M.; Brunati, A.M.; Clari, G.; Toninello, A. Interaction of genistein with the mitochondrial electron transport chain results in opening of the membrane transition pore. Biochim. Biophys. Acta 2002, 1556, 187–196. [Google Scholar] [CrossRef]
- Valenti, D.; de Bari, L.; Manente, G.A.; Rossi, L.; Mutti, L.; Moro, L.; Vacca, R.A. Negative modulation of mitochondrial oxidative phosphorylation by epigallocatechin-3 gallate leads to growth arrest and apoptosis in human malignant pleural mesothelioma cells. Biochim. Biophys. Acta 2013, 1832, 2085–2096. [Google Scholar] [CrossRef]
- Oliveira, M.R.; Nabavi, S.F.; Daglia, M.; Rastrelli, L.; Nabavi, S.M. Epigallocatechin gallate and mitochondria-A story of life and death. Pharmacol. Res. 2016, 104, 70–85. [Google Scholar] [CrossRef]
- Bendokas, V.; Skemiene, K.; Trumbeckaite, S.; Stanys, V.; Passamonti, S.; Borutaite, V.; Liobikas, J. Anthocyanins: From plant pigments to health benefits at mitochondrial level. Crit. Rev. Food Sci. Nutr. 2019, 1–14. [Google Scholar] [CrossRef] [PubMed]
- Liobikas, J.; Skemiene, K.; Trumbeckaite, S.; Borutaite, V. Anthocyanins in cardioprotection: A path through mitochondria. Pharmacol. Res. 2016, 113, 808–815. [Google Scholar] [CrossRef] [PubMed]
- Buss, G.D.; Constantin, J.; de Lima, L.C.; Teodoro, G.R.; Comar, J.F.; Ishii-Iwamoto, E.L.; Bracht, A. The action of quercetin on the mitochondrial NADH to NAD(+) ratio in the isolated perfused rat liver. Planta Med. 2005, 71, 1118–1122. [Google Scholar] [CrossRef] [PubMed]
- Wang, J.; Fang, F.; Huang, Z.; Wang, Y.; Wong, C. Kaempferol is an estrogen-related receptor alpha and gamma inverse agonist. FEBS Lett. 2009, 583, 643–647. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Constantin, R.P.; do Nascimento, G.S.; Constantin, R.P.; Salgueiro, C.L.; Bracht, A.; Ishii-Iwamoto, E.L.; Yamamoto, N.S.; Constantin, J. Citrus flavanones affect hepatic fatty acid oxidation in rats by acting as prooxidant agents. Biomed. Res. Int. 2013, 2013, 342973. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Chen, L.B. Mitochondrial membrane potential in living cells. Annu. Rev. Cell Biol. 1988, 4, 155–181. [Google Scholar] [CrossRef] [PubMed]
- Fantin, V.R.; St-Pierre, J.; Leder, P. Attenuation of LDH-A expression uncovers a link between glycolysis, mitochondrial physiology, and tumor maintenance. Cancer Cell 2006, 9, 425–434. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Bernatoniene, J.; Kopustinskiene, D.M.; Jakstas, V.; Majiene, D.; Baniene, R.; Kursvietiene, L.; Masteikova, R.; Savickas, A.; Toleikis, A.; Trumbeckaite, S. The effect of Leonurus cardiaca herb extract and some of its flavonoids on mitochondrial oxidative phosphorylation in the heart. Planta Med. 2014, 80, 525–532. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Bernatoniene, J.; Trumbeckaite, S.; Majiene, D.; Baniene, R.; Baliutyte, G.; Savickas, A.; Toleikis, A. The effect of crataegus fruit extract and some of its flavonoids on mitochondrial oxidative phosphorylation in the heart. Phytother. Res. 2009, 23, 1701–1707. [Google Scholar] [CrossRef] [Green Version]
- Trumbeckaite, S.; Bernatoniene, J.; Majiene, D.; Jakstas, V.; Savickas, A.; Toleikis, A. The effect of flavonoids on rat heart mitochondrial function. Biomed. Pharmacother. 2006, 60, 245–248. [Google Scholar] [CrossRef]
- Kopustinskiene, D.M.; Savickas, A.; Vetchy, D.; Masteikova, R.; Kasauskas, A.; Bernatoniene, J. Direct effects of (-)-epicatechin and procyanidin B2 on the respiration of rat heart mitochondria. Biomed. Res. Int. 2015, 2015, 232836. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Oteiza, P.I.; Fraga, C.G.; Mills, D.A.; Taft, D.H. Flavonoids and the gastrointestinal tract: Local and systemic effects. Mol. Asp. Med. 2018, 61, 41–49. [Google Scholar] [CrossRef] [PubMed]
- Wells, J.M.; Brummer, R.J.; Derrien, M.; MacDonald, T.T.; Troost, F.; Cani, P.D.; Theodorou, V.; Dekker, J.; Meheust, A.; de Vos, W.M.; et al. Homeostasis of the gut barrier and potential biomarkers. Am. J. Physiology. Gastrointest. Liver Physiol. 2017, 312, G171–G193. [Google Scholar] [CrossRef] [PubMed]
- Rowland, I.; Gibson, G.; Heinken, A.; Scott, K.; Swann, J.; Thiele, I.; Tuohy, K. Gut microbiota functions: Metabolism of nutrients and other food components. Eur. J. Nutr. 2018, 57, 1–24. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Qu, G.; Chen, J.; Guo, X. The beneficial and deleterious role of dietary polyphenols on chronic degenerative diseases by regulating gene expression. Biosci. Trends 2019, 12, 526–536. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Espin, J.C.; Gonzalez-Sarrias, A.; Tomas-Barberan, F.A. The gut microbiota: A key factor in the therapeutic effects of (poly)phenols. Biochem. Pharmacol. 2017, 139, 82–93. [Google Scholar] [CrossRef]
- Bettaieb, A.; Vazquez Prieto, M.A.; Rodriguez Lanzi, C.; Miatello, R.M.; Haj, F.G.; Fraga, C.G.; Oteiza, P.I. (-)-Epicatechin mitigates high-fructose-associated insulin resistance by modulating redox signaling and endoplasmic reticulum stress. Free Radic. Biol. Med. 2014, 72, 247–256. [Google Scholar] [CrossRef] [Green Version]
- Gutierrez-Salmean, G.; Ortiz-Vilchis, P.; Vacaseydel, C.M.; Garduno-Siciliano, L.; Chamorro-Cevallos, G.; Meaney, E.; Villafana, S.; Villarreal, F.; Ceballos, G.; Ramirez-Sanchez, I. Effects of (-)-epicatechin on a diet-induced rat model of cardiometabolic risk factors. Eur. J. Pharmacol. 2014, 728, 24–30. [Google Scholar] [CrossRef]
- Vazquez Prieto, M.A.; Bettaieb, A.; Rodriguez Lanzi, C.; Soto, V.C.; Perdicaro, D.J.; Galmarini, C.R.; Haj, F.G.; Miatello, R.M.; Oteiza, P.I. Catechin and quercetin attenuate adipose inflammation in fructose-fed rats and 3T3-L1 adipocytes. Mol. Nutr. Food Res. 2015, 59, 622–633. [Google Scholar] [CrossRef]
- Cremonini, E.; Mastaloudis, A.; Hester, S.N.; Verstraeten, S.V.; Anderson, M.; Wood, S.M.; Waterhouse, A.L.; Fraga, C.G.; Oteiza, P.I. Anthocyanins inhibit tumor necrosis alpha-induced loss of Caco-2 cell barrier integrity. Food Funct. 2017, 8, 2915–2923. [Google Scholar] [CrossRef] [Green Version]
- Olejnik, A.; Kowalska, K.; Kidon, M.; Czapski, J.; Rychlik, J.; Olkowicz, M.; Dembczynski, R. Purple carrot anthocyanins suppress lipopolysaccharide-induced inflammation in the co-culture of intestinal Caco-2 and macrophage RAW264.7 cells. Food Funct. 2016, 7, 557–564. [Google Scholar] [CrossRef] [PubMed]
- He, X.; Sun, L.M. Dietary intake of flavonoid subclasses and risk of colorectal cancer: Evidence from population studies. Oncotarget 2016, 7, 26617–26627. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Grosso, G.; Godos, J.; Lamuela-Raventos, R.; Ray, S.; Micek, A.; Pajak, A.; Sciacca, S.; D'Orazio, N.; Del Rio, D.; Galvano, F. A comprehensive meta-analysis on dietary flavonoid and lignan intake and cancer risk: Level of evidence and limitations. Mol. Nutr. Food Res. 2017, 61. [Google Scholar] [CrossRef] [PubMed]
- Rossi, M.; Bosetti, C.; Negri, E.; Lagiou, P.; La Vecchia, C. Flavonoids, proanthocyanidins, and cancer risk: A network of case-control studies from Italy. Nutr. Cancer 2010, 62, 871–877. [Google Scholar] [CrossRef]
- Li, Y.; Zhang, T.; Chen, G.Y. Flavonoids and Colorectal Cancer Prevention. Antioxidants 2018, 7, 187. [Google Scholar] [CrossRef] [Green Version]
- Masumoto, S.; Terao, A.; Yamamoto, Y.; Mukai, T.; Miura, T.; Shoji, T. Non-absorbable apple procyanidins prevent obesity associated with gut microbial and metabolomic changes. Sci. Rep. 2016, 6, 31208. [Google Scholar] [CrossRef]
- Sugiyama, H.; Akazome, Y.; Shoji, T.; Yamaguchi, A.; Yasue, M.; Kanda, T.; Ohtake, Y. Oligomeric procyanidins in apple polyphenol are main active components for inhibition of pancreatic lipase and triglyceride absorption. J. Agric. Food Chem. 2007, 55, 4604–4609. [Google Scholar] [CrossRef]
- Fei, Q.; Gao, Y.; Zhang, X.; Sun, Y.; Hu, B.; Zhou, L.; Jabbar, S.; Zeng, X. Effects of Oolong tea polyphenols, EGCG, and EGCG3″Me on pancreatic alpha-amylase activity in vitro. J. Agric. Food Chem. 2014, 62, 9507–9514. [Google Scholar] [CrossRef]
- Cardona, F.; Andres-Lacueva, C.; Tulipani, S.; Tinahones, F.J.; Queipo-Ortuno, M.I. Benefits of polyphenols on gut microbiota and implications in human health. J. Nutr. Biochem. 2013, 24, 1415–1422. [Google Scholar] [CrossRef] [Green Version]
- David, L.A.; Maurice, C.F.; Carmody, R.N.; Gootenberg, D.B.; Button, J.E.; Wolfe, B.E.; Ling, A.V.; Devlin, A.S.; Varma, Y.; Fischbach, M.A.; et al. Diet rapidly and reproducibly alters the human gut microbiome. Nature 2014, 505, 559–563. [Google Scholar] [CrossRef] [Green Version]
- De Leoz, M.L.; Kalanetra, K.M.; Bokulich, N.A.; Strum, J.S.; Underwood, M.A.; German, J.B.; Mills, D.A.; Lebrilla, C.B. Human milk glycomics and gut microbial genomics in infant feces show a correlation between human milk oligosaccharides and gut microbiota: A proof-of-concept study. J. Proteome Res. 2015, 14, 491–502. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Marchesi, J.R.; Adams, D.H.; Fava, F.; Hermes, G.D.; Hirschfield, G.M.; Hold, G.; Quraishi, M.N.; Kinross, J.; Smidt, H.; Tuohy, K.M.; et al. The gut microbiota and host health: A new clinical frontier. Gut 2016, 65, 330–339. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Walle, T. Absorption and metabolism of flavonoids. Free Radic. Biol. Med. 2004, 36, 829–837. [Google Scholar] [CrossRef] [PubMed]
- Kumar, S.; Pandey, A.K. Chemistry and biological activities of flavonoids: An overview. Sci. World J. 2013, 2013, 162750. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Williamson, G.; Clifford, M.N. Role of the small intestine, colon and microbiota in determining the metabolic fate of polyphenols. Biochem. Pharmacol. 2017, 139, 24–39. [Google Scholar] [CrossRef] [PubMed]
- Duda-Chodak, A. The inhibitory effect of polyphenols on human gut microbiota. J. Physiol. Pharmacol. 2012, 63, 497–503. [Google Scholar]
- Nohynek, L.J.; Alakomi, H.L.; Kahkonen, M.P.; Heinonen, M.; Helander, I.M.; Oksman-Caldentey, K.M.; Puupponen-Pimia, R.H. Berry phenolics: Antimicrobial properties and mechanisms of action against severe human pathogens. Nutr. Cancer 2006, 54, 18–32. [Google Scholar] [CrossRef]
- Espley, R.V.; Butts, C.A.; Laing, W.A.; Martell, S.; Smith, H.; McGhie, T.K.; Zhang, J.; Paturi, G.; Hedderley, D.; Bovy, A.; et al. Dietary flavonoids from modified apple reduce inflammation markers and modulate gut microbiota in mice. J. Nutr. 2014, 144, 146–154. [Google Scholar] [CrossRef] [Green Version]
- Gwiazdowska, D.; Jus, K.; Jasnowska-Malecka, J.; Kluczynska, K. The impact of polyphenols on Bifidobacterium growth. Acta Biochim. Pol. 2015, 62, 895–901. [Google Scholar] [CrossRef]
- Ridlon, J.M.; Kang, D.J.; Hylemon, P.B.; Bajaj, J.S. Bile acids and the gut microbiome. Curr. Opin. Gastroenterol. 2014, 30, 332–338. [Google Scholar] [CrossRef] [Green Version]
- Maynard, C.L.; Elson, C.O.; Hatton, R.D.; Weaver, C.T. Reciprocal interactions of the intestinal microbiota and immune system. Nature 2012, 489, 231–241. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Shortt, C.; Hasselwander, O.; Meynier, A.; Nauta, A.; Fernandez, E.N.; Putz, P.; Rowland, I.; Swann, J.; Turk, J.; Vermeiren, J.; et al. Systematic review of the effects of the intestinal microbiota on selected nutrients and non-nutrients. Eur. J. Nutr. 2018, 57, 25–49. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Kuo, S.M. Dietary flavonoid and cancer prevention: Evidence and potential mechanism. Crit. Rev. Oncog. 1997, 8, 47–69. [Google Scholar] [CrossRef] [PubMed]
- Galati, G.; O'Brien, P.J. Potential toxicity of flavonoids and other dietary phenolics: Significance for their chemopreventive and anticancer properties. Free Radic. Biol. Med. 2004, 37, 287–303. [Google Scholar] [CrossRef] [PubMed]
- Skibola, C.F.; Smith, M.T. Potential health impacts of excessive flavonoid intake. Free Radic. Biol. Med. 2000, 29, 375–383. [Google Scholar] [CrossRef]
© 2020 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 (http://creativecommons.org/licenses/by/4.0/).
Share and Cite
Kopustinskiene, D.M.; Jakstas, V.; Savickas, A.; Bernatoniene, J. Flavonoids as Anticancer Agents. Nutrients 2020, 12, 457. https://doi.org/10.3390/nu12020457
Kopustinskiene DM, Jakstas V, Savickas A, Bernatoniene J. Flavonoids as Anticancer Agents. Nutrients. 2020; 12(2):457. https://doi.org/10.3390/nu12020457
Chicago/Turabian StyleKopustinskiene, Dalia M., Valdas Jakstas, Arunas Savickas, and Jurga Bernatoniene. 2020. "Flavonoids as Anticancer Agents" Nutrients 12, no. 2: 457. https://doi.org/10.3390/nu12020457
APA StyleKopustinskiene, D. M., Jakstas, V., Savickas, A., & Bernatoniene, J. (2020). Flavonoids as Anticancer Agents. Nutrients, 12(2), 457. https://doi.org/10.3390/nu12020457