Evidence of Flavonoids on Disease Prevention
Abstract
:1. Introduction
2. The Effects of Flavonoids on Obesity
2.1. Anti-Adipogenesis Effects
2.2. Promotion of Energy Expenditure
2.3. Alleviation of Insulin Resistance
3. Anti-Carcinogenic Roles of Flavonoids
Flavonoids | Dosage | Duration | Models | Effects | Molecular Mechanisms | Ref. |
---|---|---|---|---|---|---|
Quercetin | 15 mg/day | - | Population-based, case-control study | ↓Lung cancer | ↓ CYP1A1 | [72] |
Quercetin | 25 mg/day | - | A case-control study in Finland | ↓Lung cancer | - | [73] |
Flavonols | 15 mg/day | - | A case-control study in Italian | ↓Breast cancer | - | [74] |
Naringenin | 10–160 μM | 24–72 h | Gastric cancer SGC7901 cell line | ↑Apoptosis | ↑Apoptotic proteins ↓AKT | [75] |
Hesperetin | 100–300 μM | 24–48 h | Esophageal cancer cells | ↑Apoptosis | ↑ROS production ↑Intracellular caspase-9, caspase-3, Apaf-1 | [76] |
Quercetin | 20–100 μM | 24–72 h | BT-474 breast cancer cells | ↑Caspase-dependent extrinsic apoptosis | ↑Caspase-8 and caspase-3 ↓STAT3 signaling | [77] |
Fisetin | 40–120 μM | 24–96 h | Prostate cancer cells | ↑Autophagy | ↓mTOR signaling pathway | [78] |
Quercetin | 30–90 μM | 48 h | Human breast cancer cell line | ↑Autophagy ↓mTOR activity | ↓Proteasome | [79] |
Apigenin | 12.5–50 μM | 24 h | HCT116 human colon cancer cells | ↑Apoptosis | ↓Autophagy | [80] |
Apigenin | 20–80 μM | 24–48 h | Human breast cancer cells | ↑Apoptosis | ↓Autophagy | [81] |
Delphinidin | 120–180 μM | 48 h | Human prostate cancer cells | ↓Cell growth | ↑Autophagy | [82] |
Anthocyanin | 30–150 μM | 72 h | Human oral cancer cells | Anti-metastatic properties | ↓Autophagy | [83] |
Naringenin | 100 mg/kg | 72 h | Breast cancer resection model mice | ↓Metastases outgrowth | ↓Treg-induced immunosuppression | [84] |
Methlut | 1–100 μM | 2–24 h | Mast cell | Inflammatory conditions | ↓Intracellular calcium ↓NF-kB activation | [85] |
3.1. Apoptosis Induction
3.2. Regulation of Autophagy
3.3. Targeting NF-κB
4. Preventative Roles of Flavonoids in Neurodegenerative Diseases
Flavonoids | Dosage | Duration | Models | Effects | Molecular Mechanisms | Ref. |
---|---|---|---|---|---|---|
EGCG and tea polyphenols | 2 and 10 mg/kg | 14 d | PD mice model | ↓Dopaminergic neurodegeneration | Antioxidative and iron-chelating properties | [125] |
Quercetin | 50 mg/kg | 14 d | PD mouse model | Antiparkinsonian properties | ↑AchE and antioxidant activities | [126] |
Anthocyanins | 20 mg/kg | 84 d | Transgenic R6/1 HD male mice | ↑Spatial cognition learning ability | ↓Oxidative status | [127] |
Quercetin | 25 mg/kg | 90 d | Aged triple transgenic AD mice model | ↓Alzheimer’s disease pathology ↑Cognitive and emotional function | - | [128] |
Fisetin | 0.05% | 180 d | AD transgenic Mice model | Maintains cognitive function | Modulation of p25 and inflammatory pathways | [129] |
Genistein | 40 μM | 48 h | Aβ-induced hippocampal cell | ↓Aβ-induced neuronal apoptosis | Antioxidative properties Estrogen receptor-mediated pathway | [130] |
Morin | 1–10 μM | 6 h | Human neuroblastoma cells | ↓Neuronal apoptosis and tau phosphorylation | ↓GSK3β | [131] |
7,8-dihydroxyflavone | 5 mg/kg | 75 d | ASL transgenic mice model | ↑Motor performance ↑Lower motor neuronal survival | - | [132] |
EGCG | 10 mg/kg | 91 d | ASL transgenic mouse model | Neuroprotective effects | ↓NF-κB and cleaved caspase-3 | [133] |
Fisetin | 9 mg/kg | 80 d | ALS transgenic mice model | Antioxidant and Neuroprotective Effects | ↑ERK | [134] |
Chrysin | 50 mg/kg | 14 d | HD rat model | ↓Mitochondrial dysfunction and striatal apoptosis | ↑Bcl-2 gene ↓Bax-Bad genes | [135] |
7,8-dihydroxyflavone | 5 mg/kg | 119 d | Male R6/1 transgenic mice | ↓Cognitive and motor deficits | ↑PLCγ1 pathway | [136] |
4.1. Parkinson’s Disease
4.2. Alzheimer’s Disease
4.3. Amyotrophic Lateral Sclerosis
4.4. Targeting NF-κB
5. Discussion and Conclusions
Author Contributions
Funding
Acknowledgments
Conflicts of Interest
References
- Mozaffarian, D. Dietary and policy priorities to reduce the global crises of obesity and diabetes. Nat. Food 2020, 1, 38–50. [Google Scholar] [CrossRef] [Green Version]
- Dariush, M.; Jason, H.Y.W. Flavonoids, Dairy Foods, and Cardiovascular and Metabolic Health: A Review of Emerging Biologic Pathways. Circ. Res. 2018, 122, 369–384. [Google Scholar]
- Cassidy, A.; O’Reilly, É.; Kay, C.; Sampson, L.; Franz, M.; Forman, J.P.; Curhan, G.; Rimm, E. Habitual intake of flavonoid subclasses and incident hypertension in adults. Am. J. Clin. Nutr. 2011, 93, 338–347. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Ding, M.; Pan, A.; Manson, J.E.; Willett, W.C.; Malik, V.; Rosner, B.; Giovannucci, E.; Hu, F.B.; Sun, Q. Consumption of soy foods and isoflavones and risk of type 2 diabetes: A pooled analysis of three US cohorts. Eur. J. Clin. Nutr. 2016, 70, 1381–1387. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Maleki, S.J.; Crespo, J.F.; Cabanillas, B. Anti-inflammatory effects of flavonoids. Food Chem. 2019, 299, 125124. [Google Scholar] [CrossRef]
- Wang, T.Y.; Li, Q.; Bi, K.S. Bioactive flavonoids in medicinal plants: Structure, activity and biological fate. Asian J. Pharm. Sci. 2018, 13, 12–23. [Google Scholar] [CrossRef]
- Atrahimovich, D.; Avni, D.; Khatib, S. Flavonoids-Macromolecules Interactions in Human Diseases with Focus on Alzheimer, Atherosclerosis and Cancer. Antioxidants 2021, 10, 423. [Google Scholar] [CrossRef]
- Ahn-Jarvis, J.H.; Parihar, A.; Doseff, A.I. Dietary Flavonoids for Immunoregulation and Cancer: Food Design for Targeting Disease. Antioxidants 2019, 8, 202. [Google Scholar] [CrossRef] [Green Version]
- Xi, X.; Wang, J.; Qin, Y.; You, Y.; Huang, W.; Zhan, J. The Biphasic Effect of Flavonoids on Oxidative Stress and Cell Proliferation in Breast Cancer Cells. Antioxidants 2022, 11, 622. [Google Scholar] [CrossRef]
- Ono, M.; Fujimori, K. Antiadipogenic effect of dietary apigenin through activation of AMPK in 3T3-L1 cells. J. Agric. Food Chem. 2011, 59, 13346–13352. [Google Scholar] [CrossRef]
- Devi, K.P.; Shanmuganathan, B.; Manayi, A.; Nabavi, S.F.; Nabavi, S.M. Molecular and Therapeutic Targets of Genistein in Alzheimer’s Disease. Mol. Neurobiol. 2017, 54, 7028–7041. [Google Scholar] [CrossRef]
- Banerjee, S.; Li, Y.; Wang, Z.; Sarkar, F.H. Multi-targeted therapy of cancer by genistein. Cancer Lett. 2008, 269, 226–242. [Google Scholar] [CrossRef] [Green Version]
- Adlercreutz, H.; Markkanen, H.; Watanabe, S. Plasma concentrations of phyto-oestrogens in Japanese men. Lancet 1993, 342, 1209–1210. [Google Scholar] [CrossRef]
- Lampe, J.W.; Nishino, Y.; Ray, R.M.; Wu, C.; Li, W.; Lin, M.G.; Gao, D.L.; Hu, Y.; Shannon, J.; Stalsberg, H.; et al. Plasma isoflavones and fibrocystic breast conditions and breast cancer among women in Shanghai, China. Cancer Epidemiol. Biomark. Prev. 2007, 16, 2579–2586. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Chen, G.L.; Fan, M.X.; Wu, J.L.; Li, N.; Guo, M.Q. Antioxidant and anti-inflammatory properties of flavonoids from lotus plumule. Food Chem. 2019, 277, 706–712. [Google Scholar] [CrossRef] [PubMed]
- XLiu, X.Y.; Lv, X.; Wang, P.; Ai, C.Z.; Zhou, Q.H.; Finel, M.; Fan, B.; Cao, Y.F.; Tang, H.; Ge, G.B. Inhibition of UGT1A1 by natural and synthetic flavonoids. Int. J. Biol. Macromol. 2019, 126, 653–661. [Google Scholar]
- del Mar Rivas-Chacón, L.; Yanes-Díaz, J.; de Lucas, B.; Riestra-Ayora, J.I.; Madrid-García, R.; Sanz-Fernández, R.; Sánchez-Rodríguez, C. Preventive Effect of Cocoa Flavonoids via Suppression of Oxidative Stress-Induced Apoptosis in Auditory Senescent Cells. Antioxidants 2022, 11, 1450. [Google Scholar] [CrossRef] [PubMed]
- Rodríguez-García, C.; Sánchez-Quesada, C.; Gaforio, J.J. Dietary Flavonoids as Cancer Chemopreventive Agents: An Updated Review of Human Studies. Antioxidants 2019, 8, 137. [Google Scholar] [CrossRef] [Green Version]
- Kawser Hossain, M.; Abdal Dayem, A.; Han, J.; Yin, Y.; Kim, K.; Kumar Saha, S.; Yang, G.M.; Choi, H.Y.; Cho, S.G. Molecular Mechanisms of the Anti-Obesity and Anti-Diabetic Properties of Flavonoids. Int. J. Mol. Sci. 2016, 17, 569. [Google Scholar] [CrossRef] [Green Version]
- Tsao, C.W.; Aday, A.W.; Almarzooq, Z.I.; Alonso, A.; Beaton, A.Z.; Bittencourt, M.S.; Boehme, A.K.; Buxton, A.E.; Carson, A.P.; Commodore-Mensah, Y.; et al. Heart Disearse and Stroke Statistics-2022 Update: A Report from the American Heart Association. Circulation 2022, 145, e153–e639. [Google Scholar] [CrossRef]
- Khalilpourfarshbafi, M.; Gholami, K.; Murugan, D.D.; Abdul Sattar, M.Z.; Abdullah, N.A. Differential effects of dietary flavonoids on adipogenesis. Eur. J. Nutr. 2019, 58, 5–25. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Kerr, A.G.; Sinha, I.; Dadvar, S.; Arner, P.; Dahlman, I. Epigenetic regulation of diabetogenic adipose morphology. Mol. Metab. 2019, 25, 159–167. [Google Scholar] [CrossRef] [PubMed]
- Ren, N.; Kim, E.; Li, B.; Pan, H.; Tong, T.; Yang, C.S.; Tu, Y. Flavonoids Alleviating Insulin Resistance through Inhibition of Inflammatory Signaling. J. Agric. Food Chem. 2019, 67, 5361–5373. [Google Scholar] [CrossRef] [PubMed]
- Xie, B.; Waters, M.J.; Schirra, H.J. Investigating potential mechanisms of obesity by metabolomics. J. Biomed. Biotechnol. 2012, 2012, 805683. [Google Scholar] [CrossRef] [Green Version]
- Kim, H.K.; Kim, J.N.; Han, S.N.; Nam, J.H.; Na, H.N.; Ha, T.J. Black soybean anthocyanins inhibit adipocyte differentiation in 3T3-L1 cells. Nutr. Res. 2012, 32, 770–777. [Google Scholar] [CrossRef] [PubMed]
- Kim, S.C.; Kim, Y.H.; Son, S.W.; Moon, E.Y.; Pyo, S.; Um, S.H. Fisetin induces Sirt1 expression while inhibiting early adipogenesis in 3T3-L1 cells. Biochem. Biophys. Res. Commun. 2015, 467, 638–644. [Google Scholar] [CrossRef]
- Kim, M.H.; Park, J.S.; Seo, M.S.; Jung, J.W.; Lee, Y.S.; Kang, K.S. Genistein and daidzein repress adipogenic differentiation of human adipose tissue-derived mesenchymal stem cells via Wnt/β-catenin signalling or lipolysis. Cell Proliferat 2010, 43, 594–605. [Google Scholar] [CrossRef]
- Zhang, X.; Zhang, Q.X.; Wang, X.; Zhang, L.; Qu, W.; Bao, B.; Liu, C.A.; Liu, J. Dietary luteolin activates browning and thermogenesis in mice through an AMPK/PGC1α pathway-mediated mechanism. Int. J. Obes. 2016, 40, 1841–1849. [Google Scholar] [CrossRef]
- Kamio, N.; Suzuki, T.; Watanabe, Y.; Suhara, Y.; Osakabe, N. A single oral dose of flavan-3-ols enhances energy expenditure by sympathetic nerve stimulation in mice. Free. Radic. Biol. Med. 2016, 91, 256–263. [Google Scholar] [CrossRef]
- Lee, M.S.; Shin, Y.; Jung, S.; Kim, Y. Effects of epigallocatechin-3-gallate on thermogenesis and mitochondrial biogenesis in brown adipose tissues of diet-induced obese mice. Food Nutr. Res. 2017, 61, 1325307. [Google Scholar] [CrossRef] [Green Version]
- Friedrich, M.; Petzke, K.J.; Raederstorff, D.; Wolfram, S.; Klaus, S. Acute effects of epigallocatechin gallate from green tea on oxidation and tissue incorporation of dietary lipids in mice fed a high-fat diet. Int. J. Obes. 2012, 36, 735–743. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Arunkumar, E.; Karthik, D.; Anuradha, C.V. Genistein sensitizes hepatic insulin signaling and modulates lipid regulatory genes through p70 ribosomal S6 kinase-1 inhibition in high-fat-high-fructose diet-fed mice. Pharm. Biol. 2013, 51, 815–824. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Liu, I.M.; Tzeng, T.F.; Liou, S.S.; Lan, T.W. Myricetin, a naturally occurring flavonol, ameliorates insulin resistance induced by a high-fructose diet in rats. Life Sci. 2007, 81, 1479–1488. [Google Scholar] [CrossRef] [PubMed]
- Tzeng, T.F.; Liou, S.S.; Liu, I.M. Myricetin Ameliorates Defective Post-Receptor Insulin Signaling via β-Endorphin Signaling in the Skeletal Muscles of Fructose-Fed Rats. Evid. Based Complement. Altern. Med. Ecam 2011, 2011, 150752. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Jung, U.J.; Lee, M.K.; Park, Y.B.; Kang, M.A.; Choi, M.S. Effect of citrus flavonoids on lipid metabolism and glucose-regulating enzyme mRNA levels in type-2 diabetic mice. Int. J. Biochem. Cell Biol. 2006, 38, 1134–1145. [Google Scholar] [CrossRef] [PubMed]
- Zhang, H.J.; Ji, B.P.; Chen, G.; Zhou, F.; Luo, Y.C.; Yu, H.Q.; Gao, F.Y.; Zhang, Z.P.; Li, H.Y. A combination of grape seed-derived procyanidins and gypenosides alleviates insulin resistance in mice and HepG2 cells. J. Food Sci. 2009, 74, H1–H7. [Google Scholar] [CrossRef] [PubMed]
- Patel, Y.M.; Lane, M.D. Mitotic clonal expansion during preadipocyte differentiation: Calpain-mediated turnover of p27. J. Biol. Chem. 2000, 275, 17653–17660. [Google Scholar] [CrossRef] [Green Version]
- Rayalam, S.; Della-Fera, M.A.; Baile, C.A. Phytochemicals and regulation of the adipocyte life cycle. J. Biol. Chem. 2008, 19, 717–726. [Google Scholar] [CrossRef]
- Leonardini, A.; Laviola, L.; Perrini, S.; Natalicchio, A.; Giorgino, F. Cross-Talk between PPARgamma and Insulin Signaling and Modulation of Insulin Sensitivity. PPAR Res. 2009, 2009, 818945. [Google Scholar] [CrossRef] [Green Version]
- Guarente, L. Sirtuins as potential targets for metabolic syndrome. Nature 2006, 444, 868–874. [Google Scholar] [CrossRef]
- Picard, F.; Kurtev, M.; Chung, N.; Topark-Ngarm, A.; Senawong, T.; Machado de Oliveira, R.; Leid, M.; McBurney, M.W.; Guarente, L. Sirt1 promotes fat mobilization in white adipocytes by repressing PPAR-gamma. Nature 2004, 429, 771–776. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Lee, J.; Lee, J.; Jung, E.; Hwang, W.; Kim, Y.S.; Park, D. Isorhamnetin-induced anti-adipogenesis is mediated by stabilization of beta-catenin protein. Life Sci. 2010, 86, 416–423. [Google Scholar] [CrossRef] [PubMed]
- Ross, S.E.; Hemati, N.; Longo, K.A.; Bennett, C.N.; Lucas, P.C.; Erickson, R.L.; MacDougald, O.A. Inhibition of adipogenesis by Wnt signaling. Science 2000, 289, 950–953. [Google Scholar] [CrossRef] [PubMed]
- Nakao, Y.; Yoshihara, H.; Fujimori, K. Suppression of Very Early Stage of Adipogenesis by Baicalein, a Plant-Derived Flavonoid through Reduced Akt-C/EBPα-GLUT4 Signaling-Mediated Glucose Uptake in 3T3-L1 Adipocytes. PLoS ONE 2016, 11, e0163640. [Google Scholar] [CrossRef] [Green Version]
- Song, D.; Cheng, L.; Zhang, X.; Wu, Z.; Zheng, X. The modulatory effect and the mechanism of flavonoids on obesity. J. Food Biochem. 2019, 43, e12954. [Google Scholar] [CrossRef]
- Kim, M.A.; Kang, K.; Lee, H.J.; Kim, M.; Kim, C.Y.; Nho, C.W. Apigenin isolated from Daphne genkwa Siebold et Zucc. inhibits 3T3-L1 preadipocyte differentiation through a modulation of mitotic clonal expansion. Life Sci. 2014, 101, 64–72. [Google Scholar] [CrossRef]
- Tsutsumi, R.; Yoshida, T.; Nii, Y.; Okahisa, N.; Iwata, S.; Tsukayama, M.; Hashimoto, R.; Taniguchi, Y.; Sakaue, H.; Hosaka, T.; et al. Sudachitin, a polymethoxylated flavone, improves glucose and lipid metabolism by increasing mitochondrial biogenesis in skeletal muscle. Nutr. Metab. 2014, 11, 32. [Google Scholar] [CrossRef] [Green Version]
- Hoek-van den Hil, E.F.; van Schothorst, E.M.; van der Stelt, I.; Swarts, H.J.; Venema, D.; Sailer, M.; Vervoort, J.J.; Hollman, P.C.; Rietjens, I.M.; Keijer, J. Quercetin decreases high-fat diet induced body weight gain and accumulation of hepatic and circulating lipids in mice. Genes Nutr. 2014, 9, 418. [Google Scholar] [CrossRef] [Green Version]
- Abdullahi, A.; Jeschke, M.G. White Adipose Tissue Browning: A Double-edged Sword. Trends Endocrinol. Metab. 2016, 27, 542–552. [Google Scholar] [CrossRef] [Green Version]
- Guo, S. Insulin signaling, resistance, and the metabolic syndrome: Insights from mouse models into disease mechanisms. J. Endocrinol. 2014, 220, T1–T23. [Google Scholar] [CrossRef] [Green Version]
- Mulvihill, E.E.; Allister, E.M.; Sutherland, B.G.; Telford, D.E.; Sawyez, C.G.; Edwards, J.Y.; Markle, J.M.; Hegele, R.A.; Huff, M.W. Naringenin prevents dyslipidemia, apolipoprotein B overproduction, and hyperinsulinemia in LDL receptor-null mice with diet-induced insulin resistance. Diabetes 2009, 58, 2198–2210. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Haas, B.; Mayer, P.; Jennissen, K.; Scholz, D.; Diaz, M.B.; Bloch, W.; Herzig, S.; Fässler, R.; Pfeifer, A. Protein kinase G controls brown fat cell differentiation and mitochondrial biogenesis. Sci. Signal. 2009, 2, ra78. [Google Scholar] [CrossRef]
- Heinonen, S.; Buzkova, J.; Muniandy, M.; Kaksonen, R.; Ollikainen, M.; Ismail, K.; Hakkarainen, A.; Lundbom, J.; Lundbom, N.; Vuolteenaho, K.; et al. Impaired Mitochondrial Biogenesis in Adipose Tissue in Acquired Obesity. Diabetes 2015, 64, 3135–3145. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Bogacka, I.; Ukropcova, B.; McNeil, M.; Gimble, J.M.; Smith, S.R. Structural and functional consequences of mitochondrial biogenesis in human adipocytes in vitro. J. Clin. Endocrinol. Metab. 2005, 90, 6650–6656. [Google Scholar] [CrossRef] [Green Version]
- YSheng, Y.; Liu, J.; Zheng, S.; Liang, F.; Luo, Y.; Huang, K.; Xu, W.; He, X. Mulberry leaves ameliorate obesity through enhancing brown adipose tissue activity and modulating gut microbiota. Food Funct. 2019, 10, 4771–4781. [Google Scholar] [CrossRef]
- Kahn, S.E.; Hull, R.L.; Utzschneider, K.M. Mechanisms linking obesity to insulin resistance and type 2 diabetes. Nature 2006, 444, 840–846. [Google Scholar] [CrossRef]
- Tong, Y.; Xu, S.; Huang, L.; Chen, C. Obesity and insulin resistance: Pathophysiology and treatment. Drug Discov. Today 2022, 27, 822–830. [Google Scholar] [CrossRef]
- Huang, X.; Liu, G.; Guo, J.; Su, Z. The PI3K/AKT pathway in obesity and type 2 diabetes. Int. J. Biol. Sci. 2018, 14, 1483–1496. [Google Scholar] [CrossRef] [Green Version]
- Maury, E.; Ehala-Aleksejev, K.; Guiot, Y.; Detry, R.; Vandenhooft, A.; Brichard, S.M. Adipokines oversecreted by omental adipose tissue in human obesity. Am. J. Physiol. Metab. 2007, 293, E656–E665. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Liang-Yi, W.; Chi-Chang, J.; Hwang, L.S.; Hsu, Y.P.; Ho, P.H.; Ho, L.T. Green tea supplementation ameliorates insulin resistance and increases glucose transporter IV content in a fructose-fed rat model. Eur. J. Nutr. 2004, 43, 116–124. [Google Scholar]
- 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 A Cancer J. Clin. 2021, 71, 209–249. [Google Scholar] [CrossRef] [PubMed]
- Bever, A.M.; Cassidy, A.; Rimm, E.B.; Stampfer, M.J.; Cote, D.J. A prospective study of dietary flavonoid intake and risk of glioma in US men and women. Am. J. Clin. Nutr. 2021, 114, 1314–1327. [Google Scholar] [CrossRef]
- Li, Y.; Zhang, T.; Chen, G.Y. Flavonoids and Colorectal Cancer Prevention. Antioxidants 2018, 7, 187. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Sudhakaran, M.; Sardesai, S.; Doseff, A.I. Flavonoids: New Frontier for Immuno-Regulation and Breast Cancer Control. Antioxidants 2019, 8, 103. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Cutler, G.J.; Nettleton, J.A.; Ross, J.A.; Harnack, L.J.; Jacobs Jr, D.R.; Scrafford, C.G.; Barraj, L.M.; Mink, P.J.; Robien, K. Dietary flavonoid intake and risk of cancer in postmenopausal women: The Iowa Women’s Health Study. Int. J. Cancer 2008, 123, 664–671. [Google Scholar] [CrossRef] [Green Version]
- Boggs, D.A.; Palmer, J.R.; Stampfer, M.J.; Spiegelman, D.; Adams-Campbell, L.L.; Rosenberg, L. Tea and coffee intake in relation to risk of breast cancer in the Black Women’s Health Study. Cancer Causes Control. 2010, 21, 1941–1948. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Christensen, K.Y.; Naidu, A.; Parent, M.É.; Pintos, J.; Abrahamowicz, M.; Siemiatycki, J.; Koushik, A. The risk of lung cancer related to dietary intake of flavonoids. Nutr. Cancer 2012, 64, 964–974. [Google Scholar] [CrossRef] [Green Version]
- Reale, G.; Russo, G.I.; Di Mauro, M.; Regis, F.; Campisi, D.; Giudice, A.L.; Marranzano, M.; Ragusa, R.; Castelli, T.; Cimino, S.; et al. Association between dietary flavonoids intake and prostate cancer risk: A case-control study in Sicily. Complement. Ther. Med. 2018, 39, 14–18. [Google Scholar] [CrossRef]
- Woo, H.D.; Lee, J.; Choi, I.J.; Kim, C.G.; Lee, J.Y.; Kwon, O.; Kim, J. Dietary flavonoids and gastric cancer risk in a Korean population. Nutrients 2014, 6, 4961–4973. [Google Scholar] [CrossRef] [Green Version]
- Nimptsch, K.; Zhang, X.; Cassidy, A.; Song, M.; O’Reilly, É.J.; Lin, J.H.; Pischon, T.; Rimm, E.B.; Willett, W.C.; Fuchs, C.S.; et al. Habitual intake of flavonoid subclasses and risk of colorectal cancer in 2 large prospective cohorts. Am. J. Clin. Nutr. 2016, 103, 184–191. [Google Scholar] [CrossRef] [Green Version]
- Zamora-Ros, R.; Guinó, E.; Henar Alonso, M.; Vidal, C.; Barenys, M.; Soriano, A.; Moreno, V. Dietary flavonoids, lignans and colorectal cancer prognosis. Sci. Rep. 2015, 5, 14148. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Le Marchand, L.; Murphy, S.P.; Hankin, J.H.; Wilkens, L.R.; Kolonel, L.N. Intake of flavonoids and lung cance. J. Natl. Cancer Inst. 2000, 92, 154–160. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Knekt, P.; Kumpulainen, J.; Järvinen, R.; Rissanen, H.; Heliövaara, M.; Reunanen, A.; Hakulinen, T.; Aromaa, A. Flavonoid intake and risk of chronic diseases. Am. J. Clin. Nutr. 2002, 76, 560–568. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Bosetti, C.; Spertini, L.; Parpinel, M.; Gnagnarella, P.; Lagiou, P.; Negri, E.; Franceschi, S.; Montella, M.; Peterson, J.; Dwyer, J.; et al. Flavonoids and breast cancer risk in Italy. Cancer Epidemiol. Biomarkers Prev. 2005, 14, 805–808. [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. J. Int. Soc. Oncodevelopmental Biol. Med. 2016, 37, 11365–11374. [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. J. Int. Soc. Oncodevelopmental Biol. Med. 2016, 37, 3451–3459. [Google Scholar] [CrossRef]
- Seo, H.S.; Ku, J.M.; Choi, H.S.; Choi, Y.K.; Woo, J.K.; Kim, M.; Kim, I.; Na, C.H.; Hur, H.; Jang, B.H.; et al. Quercetin induces caspase-dependent extrinsic apoptosis through inhibition of signal transducer and activator of transcription 3 signaling in HER2-overexpressing BT-474 breast cancer cells. Oncol. Rep. 2016, 36, 31–42. [Google Scholar] [CrossRef] [Green Version]
- Suh, Y.; Afaq, F.; Khan, N.; Johnson, J.J.; Khusro, F.H.; Mukhtar, H. Fisetin induces autophagic cell death through suppression of mTOR signaling pathway in prostate cancer cells. Carcinogenesis 2010, 31, 1424–1433. [Google Scholar] [CrossRef] [Green Version]
- Klappan, A.K.; Hones, S.; Mylonas, I.; Brüning, A. Proteasome inhibition by quercetin triggers macroautophagy and blocks mTOR activity. Histochem. Cell Biol. 2012, 137, 25–36. [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] [Green Version]
- Cao, X.; Liu, B.; Cao, W.; Zhang, W.; Zhang, F.; Zhao, H.; Meng, R.; Zhang, L.; Niu, R.; Hao, X.; et al. Autophagy inhibition enhances apigenin-induced apoptosis in human breast cancer cells. Chin. J. Cancer Res. 2013, 25, 212–222. [Google Scholar]
- Hafeez, B.B.; Siddiqui, I.A.; Asim, M.; Malik, A.; Afaq, F.; Adhami, V.M.; Saleem, M.; Din, M.; Mukhtar, H. A dietary anthocyanidin delphinidin induces apoptosis of human prostate cancer PC3 cells in vitro and in vivo: Involvement of nuclear factor-kappaB signaling. Cancer Res. 2008, 68, 8564–8572. [Google Scholar] [CrossRef] [Green Version]
- Fan, M.J.; Wang, I.C.; Hsiao, Y.T.; Lin, H.Y.; Tang, N.Y.; Hung, T.C.; Quan, C.; Lien, J.C.; Chung, J.G. Anthocyanins from black rice (Oryza sativa L.) demonstrate antimetastatic properties by reducing MMPs and NF-κB expressions in human oral cancer CAL 27 cells. Nutr. Cancer 2015, 67, 327–338. [Google Scholar] [CrossRef] [PubMed]
- Qin, L.; Jin, L.; Lu, L.; Lu, X.; Zhang, C.; Zhang, F.; Liang, W. Naringenin reduces lung metastasis in a breast cancer resection model. Protein Cell 2011, 2, 507–516. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Weng, Z.; Patel, A.B.; Panagiotidou, S.; Theoharides, T.C. The novel flavone tetramethoxyluteolin is a potent inhibitor of human mast cells. J. Allergy Clin. Immunol. 2015, 135, 1044–1052.e5. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Adams, J.M.; Cory, S. The Bcl-2 apoptotic switch in cancer development and therapy. Oncogene 2007, 26, 1324–1337. [Google Scholar] [CrossRef] [Green Version]
- Indran, I.R.; Tufo, G.; Pervaiz, S.; Brenner, C. Recent advances in apoptosis, mitochondria and drug resistance in cancer cells. Biochim. Biophys. Acta Bioenerg. 2011, 1807, 735–745. [Google Scholar] [CrossRef] [Green Version]
- Fan, T.J.; Han, L.H.; Cong, R.S.; Liang, J. Caspase family proteases and apoptosis. Acta Biochim. Biophys. Sin. 2005, 37, 719–727. [Google Scholar] [CrossRef] [Green Version]
- Igney, F.H.; Krammer, P.H. Death and anti-death: Tumour resistance to apoptosis. Nat. Rev. Cancer 2002, 2, 277–288. [Google Scholar] [CrossRef]
- Rathore, R.; McCallum, J.E.; Varghese, E.; Florea, A.M.; Büsselberg, D. Overcoming chemotherapy drug resistance by targeting inhibitors of apoptosis proteins (IAPs). Apoptosis 2017, 22, 898–919. [Google Scholar] [CrossRef] [Green Version]
- Cory, S.; Adams, J.M. The Bcl2 family: Regulators of the cellular life-or-death switch. Nat. Rev. Cancer 2002, 2, 647–656. [Google Scholar] [CrossRef]
- Li-Weber, M. Targeting apoptosis pathways in cancer by Chinese medicine. Cancer Lett. 2013, 332, 304–312. [Google Scholar] [CrossRef]
- Abotaleb, M.; Samuel, S.M.; Varghese, E.; Varghese, S.; Kubatka, P.; Liskova, A.; Büsselberg, D. Flavonoids in Cancer and Apoptosis. Cancers 2018, 11, 28. [Google Scholar] [CrossRef] [PubMed] [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] [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]
- Liu, L.; Liu, T.; Tao, W.; Liao, N.; Yan, Q.; Li, L.; Tan, J.; Shen, W.; Cheng, H.; Sun, D. Flavonoids from Scutellaria barbata D. Don exert antitumor activity in colorectal cancer through inhibited autophagy and promoted apoptosis via ATF4/sestrin2 pathway. Phytomedicine Int. J. Phytother. Phytopharm. 2022, 99, 154007. [Google Scholar] [CrossRef] [PubMed]
- Jia, X.B.; Zhang, Q.; Xu, L.; Yao, W.J.; Wei, L. Lotus leaf flavonoids induce apoptosis of human lung cancer A549 cells through the ROS/p38 MAPK pathway. Biol. Res. 2021, 54, 7. [Google Scholar] [CrossRef] [PubMed]
- Yan, W.; Yang, J.; Tang, H.; Xue, L.; Chen, K.; Wang, L.; Zhao, M.; Tang, M.; Peng, A.; Long, C.; et al. Flavonoids from the stems of Millettia pachyloba Drake mediate cytotoxic activity through apoptosis and autophagy in cancer cells. J. Adv. Res. 2019, 20, 117–127. [Google Scholar] [CrossRef] [PubMed]
- Bhosale, P.B.; Abusaliya, A.; Kim, H.H.; Ha, S.E.; Park, M.Y.; Jeong, S.H.; Vetrivel, P.; Heo, J.D.; Kim, J.A.; Won, C.K.; et al. Apigetrin Promotes TNFα-Induced Apoptosis, Necroptosis, G2/M Phase Cell Cycle Arrest, and ROS Generation through Inhibition of NF-κB Pathway in Hep3B Liver Cancer Cells. Cells 2022, 11, 2734. [Google Scholar] [CrossRef]
- Mizushima, N.; Levine, B. Autophagy in Human Diseases. N. Engl. J. Med. 2020, 383, 1564–1576. [Google Scholar] [CrossRef]
- Vargas, J.N.; Hamasaki, M.; Kawabata, T.; Youle, R.J.; Yoshimori, T. The mechanisms and roles of selective autophagy in mammals. Nat. Rev. Mol. Cell Biol. 2022, 1–19. [Google Scholar] [CrossRef] [PubMed]
- Levy, J.M.; Towers, C.G.; Thorburn, A. Targeting autophagy in cancer. Nat. Rev. Cancer 2017, 17, 528–542. [Google Scholar] [CrossRef]
- Sun, K.; Deng, W.; Zhang, S.; Cai, N.; Jiao, S.; Song, J.; Wei, L. Paradoxical roles of autophagy in different stages of tumorigenesis: Protector for normal or cancer cells. Cell Biosci. 2013, 3, 35. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Russell, R.C.; Guan, K.L. The multifaceted role of autophagy in cancer. EMBO J. 2022, 41, e110031. [Google Scholar] [CrossRef] [PubMed]
- Kondo, Y.; Kanzawa, T.; Sawaya, R.; Kondo, S. The role of autophagy in cancer development and response to therapy. Nat. Rev. Cancer 2005, 5, 726–734. [Google Scholar] [CrossRef]
- Levine, B.; Kroemer, G. Autophagy in the pathogenesis of disease. Cell 2008, 132, 27–42. [Google Scholar] [CrossRef] [Green Version]
- Tasdemir, E.; Maiuri, M.C.; Orhon, I.; Kepp, O.; Morselli, E.; Criollo, A.; Kroemer, G. p53 represses autophagy in a cell cycle-dependent fashion. Cell Cycle 2008, 7, 3006–3011. [Google Scholar] [CrossRef] [Green Version]
- Gossner, G.; Choi, M.; Tan, L.; Fogoros, S.; Griffith, K.A.; Kuenker, M.; Liu, J.R. Genistein-induced apoptosis and autophagocytosis in ovarian cancer cells. Gynecol. Oncol. 2007, 105, 23–30. [Google Scholar] [CrossRef]
- Guo, Z.; Hu, X.; Xing, Z.; Xing, R.; Lv, R.; Cheng, X.; Su, J.; Zhou, Z.; Xu, Z.; Nilsson, S.; et al. Baicalein inhibits prostate cancer cell growth and metastasis via the caveolin-1/AKT/mTOR pathway. Mol. Cell. Biochem. 2015, 406, 111–119. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Longo, L.; Platini, F.; Scardino, A.; Alabiso, O.; Vasapollo, G.; Tessitore, L. Autophagy inhibition enhances anthocyanin-induced apoptosis in hepatocellular carcinoma. Mol. Cancer Ther. 2008, 7, 2476–2485. [Google Scholar] [CrossRef] [Green Version]
- Yang, P.M.; Tseng, H.H.; Peng, C.W.; Chen, W.S.; Chiu, S.J. Dietary flavonoid fisetin targets caspase-3-deficient human breast cancer MCF-7 cells by induction of caspase-7-associated apoptosis and inhibition of autophagy. Int. J. Oncol. 2012, 40, 469–478. [Google Scholar] [PubMed] [Green Version]
- Karin, M.; Greten, F.R. NF-kappaB: Linking inflammation and immunity to cancer development and progression. Nat. Rev. Immunol. 2005, 5, 749–759. [Google Scholar] [CrossRef] [PubMed]
- Karin, M. Nuclear factor-kappaB in cancer development and progression. Nature 2006, 441, 431–436. [Google Scholar] [CrossRef] [PubMed]
- 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] [PubMed] [Green Version]
- Liyanage, U.K.; Moore, T.T.; Joo, H.G.; Tanaka, Y.; Herrmann, V.; Doherty, G.; Drebin, J.A.; Strasberg, S.M.; Eberlein, T.J.; Goedegebuure, P.S.; et al. Prevalence of regulatory T cells is increased in peripheral blood and tumor microenvironment of patients with pancreas or breast adenocarcinoma. J. Immunol. 2002, 169, 2756–2761. [Google Scholar] [CrossRef] [Green Version]
- Togashi, Y.; Shitara, K.; Nishikawa, H. Regulatory T cells in cancer immunosuppression—Implications for anticancer therapy. Nat. Rev. Clin. Oncol. 2019, 16, 356–371. [Google Scholar] [CrossRef]
- Hansson, O. Biomarkers for neurodegenerative diseases. Nat. Med. 2021, 27, 954–963. [Google Scholar] [CrossRef]
- Cully, M. Neurodegenerative diseases: Inflammasome protein seeds plaques in Alzheimer disease. Nat. Rev. Drug Discov. 2018, 17, 96. [Google Scholar] [CrossRef]
- Heemels, M.-T. Neurodegenerative diseases. Nature 2016, 539, 179. [Google Scholar] [CrossRef] [Green Version]
- Li, D.; Liu, C. Conformational strains of pathogenic amyloid proteins in neurodegenerative diseases. Nat. Rev. Neurosci. 2022, 23, 523–534. [Google Scholar] [CrossRef]
- Ravichandran, K.A.; Heneka, M.T. Inflammasome activation in neurodegenerative diseases. Essays Biochem. 2021, 65, 885–904. [Google Scholar] [PubMed]
- Nichols, E.; Szoeke, C.E.; Vollset, S.E.; Abbasi, N.; Abd-Allah, F.; Abdela, J.; Aichour, M.T.; Akinyemi, R.O.; Alahdab, F.; Asgedom, S.W.; et al. Global, regional, and national burden of Alzheimer’s disease and other dementias, 1990–2016: A systematic analysis for the Global Burden of Disease Study 2016. Lancet Neurol. 2019, 18, 88–106. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Nichols, E.; Steinmetz, J.D.; Vollset, S.E.; Fukutaki, K.; Chalek, J.; Abd-Allah, F.; Abdoli, A.; Abualhasan, A.; Abu-Gharbieh, E.; Akram, T.T.; et al. Estimation of the global prevalence of dementia in 2019 and forecasted prevalence in 2050: An analysis for the Global Burden of Disease Study 2019. Lancet Public Health 2022, 7, e105–e125. [Google Scholar] [CrossRef] [PubMed]
- Maher, P. The Potential of Flavonoids for the Treatment of Neurodegenerative Diseases. Int. J. Mol. Sci. 2019, 20, 3056. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Monteiro, A.F.; Viana, J.D.; Nayarisseri, A.; Zondegoumba, E.N.; Mendonça Junior, F.J.; Scotti, M.T.; Scotti, L. Computational Studies Applied to Flavonoids against Alzheimer’s and Parkinson’s Diseases. Oxidative Med. Cell. Longev. 2018, 2018, 7912765. [Google Scholar] [CrossRef] [PubMed]
- Levites, Y.; Weinreb, O.; Maor, G.; Youdim, M.B.; Mandel, S. Green tea polyphenol (-)-epigallocatechin-3-gallate prevents N-methyl-4-phenyl-1,2,3,6-tetrahydropyridine-induced dopaminergic neurodegeneration. J. Neurochem. 2001, 78, 1073–1082. [Google Scholar] [CrossRef] [PubMed]
- Kreilaus, F.; Spiro, A.S.; Hannan, A.J.; Garner, B.; Jenner, A.M. Therapeutic Effects of Anthocyanins and Environmental Enrichment in R6/1 Huntington’s Disease Mice. J. Huntingt. Dis. 2016, 5, 285–296. [Google Scholar] [CrossRef]
- Sabogal-Guáqueta, A.M.; Munoz-Manco, J.I.; Ramírez-Pineda, J.R.; Lamprea-Rodriguez, M.; Osorio, E.; Cardona-Gómez, G.P. The flavonoid quercetin ameliorates Alzheimer’s disease pathology and protects cognitive and emotional function in aged triple transgenic Alzheimer’s disease model mice. Neuropharmacology 2015, 93, 134–145. [Google Scholar] [CrossRef] [Green Version]
- Currais, A.; Prior, M.; Dargusch, R.; Armando, A.; Ehren, J.; Schubert, D.; Quehenberger, O.; Maher, P. Modulation of p25 and inflammatory pathways by fisetin maintains cognitive function in Alzheimer’s disease transgenic mice. Aging Cell 2014, 13, 379–390. [Google Scholar] [CrossRef]
- Zeng, H.; Chen, Q.; Zhao, B. Genistein ameliorates beta-amyloid peptide (25-35)-induced hippocampal neuronal apoptosis. Free Radic. Biol. Med. 2004, 36, 180–188. [Google Scholar] [CrossRef]
- Gong, E.J.; Park, H.R.; Kim, M.E.; Piao, S.; Lee, E.; Jo, D.G.; Chung, H.Y.; Ha, N.C.; Mattson, M.P.; Lee, J. Morin attenuates tau hyperphosphorylation by inhibiting GSK3β. Neurobiol. Dis. 2011, 44, 223–230. [Google Scholar] [CrossRef] [Green Version]
- Korkmaz, O.T.; Aytan, N.; Carreras, I.; Choi, J.K.; Kowall, N.W.; Jenkins, B.G.; Dedeoglu, A. 7,8-Dihydroxyflavone improves motor performance and enhances lower motor neuronal survival in a mouse model of amyotrophic lateral sclerosis. Neurosci. Lett. 2014, 566, 286–291. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Xu, Z.; Chen, S.; Li, X.; Luo, G.; Li, L.; Le, W. Neuroprotective effects of (-)-epigallocatechin-3-gallate in a transgenic mouse model of amyotrophic lateral sclerosis. Neurochem. Res. 2006, 31, 1263–1269. [Google Scholar] [CrossRef] [PubMed]
- Wang, T.H.; Wang, S.Y.; Wang, X.D.; Jiang, H.Q.; Yang, Y.Q.; Wang, Y.; Cheng, J.L.; Zhang, C.T.; Liang, W.W.; Feng, H.L. Fisetin Exerts Antioxidant and Neuroprotective Effects in Multiple Mutant hSOD1 Models of Amyotrophic Lateral Sclerosis by Activating ERK. Neuroscience 2018, 379, 152–166. [Google Scholar] [CrossRef] [PubMed]
- Thangarajan, S.; Ramachandran, S.; Krishnamurthy, P. Chrysin exerts neuroprotective effects against 3-Nitropropionic acid induced behavioral despair-Mitochondrial dysfunction and striatal apoptosis via upregulating Bcl-2 gene and downregulating Bax-Bad genes in male wistar rats. Biomed. Pharmacother. 2016, 84, 514–525. [Google Scholar] [CrossRef] [PubMed]
- García-Díaz Barriga, G.; Giralt, A.; Anglada-Huguet, M.; Gaja-Capdevila, N.; Orlandi, J.G.; Soriano, J.; Canals, J.M.; Alberch, J. 7,8-dihydroxyflavone ameliorates cognitive and motor deficits in a Huntington’s disease mouse model through specific activation of the PLCγ1 pathway. Hum. Mol. Genet. 2017, 26, 3144–3160. [Google Scholar] [CrossRef] [PubMed]
- Sveinbjornsdottir, S. The clinical symptoms of Parkinson’s disease. J. Neurochem. 2016, 139 (Suppl. S1), 318–324. [Google Scholar] [CrossRef] [Green Version]
- Kalia, L.V.; Lang, A.E. Parkinson’s disease. Lancet 2015, 386, 896–912. [Google Scholar] [CrossRef]
- Schulz, J.B.; Falkenburger, B.H. Neuronal pathology in Parkinson’s disease. Cell Tissue Res. 2005, 320, 211. [Google Scholar] [CrossRef] [Green Version]
- Gao, X.; Cassidy, A.; Schwarzschild, M.A.; Rimm, E.B.; Ascherio, A. Habitual intake of dietary flavonoids and risk of Parkinson disease. Neurology 2012, 78, 1138–1145. [Google Scholar] [CrossRef] [Green Version]
- Rojas, P.; Montes, P.; Rojas, C.; Serrano-García, N.; Rojas-Castañeda, J.C. Effect of a phytopharmaceutical medicine, Ginko biloba extract 761, in an animal model of Parkinson’s disease: Therapeutic perspectives. Nutrition 2012, 28, 1081–1088. [Google Scholar] [CrossRef] [PubMed]
- Mercer, L.D.; Kelly, B.L.; Horne, M.K.; Beart, P.M. Dietary polyphenols protect dopamine neurons from oxidative insults and apoptosis: Investigations in primary rat mesencephalic cultures. Biochem. Pharmacol. 2005, 69, 339–345. [Google Scholar] [CrossRef] [PubMed]
- Brookmeyer, R.; Johnson, E.; Ziegler-Graham, K.; Arrighi, H.M. Forecasting the global burden of Alzheimer’s disease. Alzheimer’s Dement. 2007, 3, 186–191. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Brown, G.C.; St George-Hyslop, P.H. Deciphering microglial diversity in Alzheimer’s disease. Science 2017, 356, 1123–1124. [Google Scholar] [CrossRef] [PubMed]
- Ransohoff, R.M. Specks of insight into AlzheimerRansohoff RM.s disease. Nature 2017, 552, 342–343. [Google Scholar] [CrossRef] [Green Version]
- Haque, A.M.; Hashimoto, M.; Katakura, M.; Tanabe, Y.; Hara, Y.; Shido, O. Long-term administration of green tea catechins improves spatial cognition learning ability in rats. J. Nutr. 2006, 136, 1043–1047. [Google Scholar] [CrossRef] [Green Version]
- Qin, X.Y.; Cheng, Y.; Yu, L.C. Potential protection of green tea polyphenols against intracellular amyloid beta-induced toxicity on primary cultured prefrontal cortical neurons of rats. Neurosci. Lett. 2012, 513, 170–173. [Google Scholar] [CrossRef]
- Bastianetto, S.; Ramassamy, C.; Doré, S.; Christen, Y.; Poirier, J.; Quirion, R. The Ginkgo biloba extract (EGb 761) protects hippocampal neurons against cell death induced by beta-amyloid. Eur. J. Neurosci. 2000, 12, 1882–1890. [Google Scholar] [CrossRef]
- Wang, S.W.; Wang, Y.J.; Su, Y.J.; Zhou, W.W.; Yang, S.G.; Zhang, R.; Zhao, M.; Li, Y.N.; Zhang, Z.P.; Zhan, D.W.; et al. Rutin inhibits β-amyloid aggregation and cytotoxicity, attenuates oxidative stress, and decreases the production of nitric oxide and proinflammatory cytokines. Neurotoxicology 2012, 33, 482–490. [Google Scholar] [CrossRef]
- Riancho, J.; Gil-Bea, F.J.; Santurtun, A.; de Munaín, A.L. Amyotrophic lateral sclerosis: A complex syndrome that needs an integrated research approach. Neural Regen. Res. 2019, 14, 193–196. [Google Scholar] [CrossRef]
- Rosen, D.R.; Siddique, T.; Patterson, D.; Figlewicz, D.A.; Sapp, P.; Hentati, A.; Donaldson, D.; Goto, J.; O’Regan, J.P.; Deng, H.X.; et al. Mutations in Cu/Zn superoxide dismutase gene are associated with familial amyotrophic lateral sclerosis. Nature 1993, 362, 59–62. [Google Scholar] [CrossRef] [PubMed]
- Walker, F.O. Huntington’s disease. Lancet 2007, 369, 218–228. [Google Scholar] [CrossRef] [PubMed]
- Brody, H. Huntington’s disease. Nature 2018, 557, S35. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Menze, E.T.; Esmat, A.; Tadros, M.G.; Khalifa, A.E.; Abdel-Naim, A.B. Genistein improves sensorimotor gating: Mechanisms related to its neuroprotective effects on the striatum. Neuropharmacology 2016, 105, 35–46. [Google Scholar] [CrossRef]
- Gopinath, K.; Sudhandiran, G. Naringin modulates oxidative stress and inflammation in 3-nitropropionic acid-induced neurodegeneration through the activation of nuclear factor-erythroid 2-related factor-2 signalling pathway. Neuroscience 2012, 227, 134–143. [Google Scholar] [CrossRef]
- Menze, E.T.; Tadros, M.G.; Abdel-Tawab, A.M.; Khalifa, A.E. Potential neuroprotective effects of hesperidin on 3-nitropropionic acid-induced neurotoxicity in rats. Neurotoxicology 2012, 33, 1265–1275. [Google Scholar] [CrossRef]
- Sandhir, R.; Mehrotra, A. Quercetin supplementation is effective in improving mitochondrial dysfunctions induced by 3-nitropropionic acid: Implications in Huntington’s disease. Acta (BBA) Mol. Basis Dis. 2013, 1832, 421–430. [Google Scholar] [CrossRef] [Green Version]
- Prusty, D.; Park, B.H.; Davis, K.E.; Farmer, S.R. Activation of MEK/ERK signaling promotes adipogenesis by enhancing peroxisome proliferator-activated receptor gamma (PPARgamma) and C/EBPalpha gene expression during the differentiation of 3T3-L1 preadipocytes. J. Biol. Chem. 2002, 277, 46226–46232. [Google Scholar] [CrossRef] [Green Version]
- Rosen, E.D.; MacDougald, O.A. Adipocyte differentiation from the inside out. Nature reviews. Mol. Cell Biol. 2006, 7, 885–896. [Google Scholar]
- Zhao, L.; Gregoire, F.; Sul, H.S. Transient induction of ENC-1, a Kelch-related actin-binding protein, is required for adipocyte differentiation. J. Biol. Chem. 2000, 275, 16845–16850. [Google Scholar] [CrossRef] [Green Version]
Flavonoids | Dosage | Duration | Models | Effects | Mechanisms | Ref. |
---|---|---|---|---|---|---|
Anthocyanins | 50 μg/mL | 1–3 d | 3T3-L1 cells | Anti-adipogenesis | ↓PPAR-γ expression | [25] |
Fisetin | 25 μM | 0–2 d | 3T3-L1 cells | Anti-adipogenesis | ↑SIRT1 expression ↓PPAR-γ | [26] |
Genistein | 20 μM | 12–42 d | AD-MSCs | Anti-adipogenesis | ↑Wnt/β-catenin signaling | [27] |
Daidzein | 20 μM | 12–24 d | AD-MSCs | Anti-adipogenesis | ↑Wnt/β-catenin signaling | [27] |
Apigenin | 50 μM | 2 d | 3T3-L1 cells | Anti- adipogenesis | ↓Adipogenic gene ↓Cell cycle | [10] |
Luteolin | 0.01% | 84 d | High-fat-fed mice | ↑Browning and thermogenesis | ↑AMPK/PGC1α pathway | [28] |
Flavan-3-ols | 10 mg/kg BW | 20 h | AR blocker-treated mice | ↑Energy expenditure | ↑Sympathetic nerve | [29] |
Epigallocatechin-3-gallate | 0.2% | 32 d | High-fat-fed mice | ↑Thermogenesis and mitochondrial biogenesis | ↑Mitochondrial DNA replication and AMPK activation | [30] |
Epigallocatechin gallate | 1.0% | 4–7 d | High-fat-fed mice | ↑Energy excretion | ↓Food digestibility ↑Fat oxidation | [31] |
Genistein | 1 mg/kg BW | 45 d | High-fat–high-fructose-fed mice | ↑Insulin sensitivity | ↑IRS phosphorylation ↑PI3K/Akt pathway | [32] |
Myricetin | 1 mg/kg BW | 14 d | High-fructose-fed mice | ↓Insulin resistance | ↑IR and IRS1 phosphorylation ↑PI3K/Akt signaling | [33] |
Myricetin | 1 mg/kg BW | 14 d | High-fructose-fed mice | ↓Insulin resistance | Binding µ-opioid receptor ↑IRS-1, PI3K, and Akt | [34] |
Hesperidin | 0.2 g/kg BW | 35 d | Type-2 diabetic mice | ↑Glycogen synthesis | ↑GK | [35] |
Naringin | 0.2 g/kg BW | 35 d | Type-2 diabetic mice | ↓Gluconeogenesis | ↓PEPCK and G6P | [35] |
procyanidins | 80 mg/kg BW | 35 d | High-fat-fed mice | ↓Insulin resistance | ↑GK and hepatic glycogen concentration | [36] |
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content. |
© 2023 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
Li, M.; Qian, M.; Jiang, Q.; Tan, B.; Yin, Y.; Han, X. Evidence of Flavonoids on Disease Prevention. Antioxidants 2023, 12, 527. https://doi.org/10.3390/antiox12020527
Li M, Qian M, Jiang Q, Tan B, Yin Y, Han X. Evidence of Flavonoids on Disease Prevention. Antioxidants. 2023; 12(2):527. https://doi.org/10.3390/antiox12020527
Chicago/Turabian StyleLi, Meng, Mengqi Qian, Qian Jiang, Bie Tan, Yulong Yin, and Xinyan Han. 2023. "Evidence of Flavonoids on Disease Prevention" Antioxidants 12, no. 2: 527. https://doi.org/10.3390/antiox12020527
APA StyleLi, M., Qian, M., Jiang, Q., Tan, B., Yin, Y., & Han, X. (2023). Evidence of Flavonoids on Disease Prevention. Antioxidants, 12(2), 527. https://doi.org/10.3390/antiox12020527