Antidiabetic properties of dietary flavonoids: a cellular mechanism review

Ramachandran Vinayagam, Baojun Xu, Ramachandran Vinayagam, Baojun Xu

Abstract

Background: Natural food products have been used for combating human diseases for thousands of years. Naturally occurring flavonoids including flavones, flavonols, flavanones, flavonols, isoflavones and anthocyanidins have been proposed as effective supplements for management and prevention of diabetes and its long-term complications based on in vitro and animal models.

Aim: To summarize the roles of dietary flavonoids in diabetes management and their molecular mechanisms.

Findings: Tremendous studies have found that flavonoids originated from foods could improve glucose metabolism, lipid profile, regulating the hormones and enzymes in human body, further protecting human being from diseases like obesity, diabetes and their complications.

Conclusion: In the current review, we summarize recent progress in understanding the biological action, mechanism and therapeutic potential of the dietary flavonoids and its subsequent clinical outcomes in the field of drug discovery in management of diabetes mellitus.

Keywords: Diabetes mellitus; Dietary flavonoids; Glycemic control; Hyperglycemia; Insulin secretion.

Figures

Fig. 1
Fig. 1
Schematic of the proposed role of flavonoids on management of blood glucose in diabetes. AKT; v-akt murine thymoma viral oncogene homolog, IRS; Insulin receptor substrate, HB; hemoglobin, HbA1c; Glycated hemoglobin, HMG-CoA :3-hydroxy-3-methylglutaryl-coenzyme A, IL-1β; Interleukin-1 beta, PI3K; Phosphatidylinositol-3-kinase, SREBP-1c; Sterol regulatory element-binding protein, TG; Triglycerides, VLDL; Very low density lipoprotein, (↑ Increase, ↓ Decrease)

References

    1. Devendra D, Liu E, Eisenbarth GS. Type 1diabetes: Recent developments. BMJ. 2004;328:750–54. doi: 10.1136/bmj.328.7442.750.
    1. Fowler MJ. Microvascular and macrovascular complications of diabetes. Clin Diabetes. 2008;26:2.
    1. International Diabetes Federation, Sixth edition. 2014.
    1. Kumar R, Laloo D, Hemalatha S. Diabetes mellitus: An overview on its pharmacological aspects and reported medicinal plants having antidiabetic activity. Asian Pac J Trop Biomed. 2012;2:411–20.
    1. Surveswaran S, Zhong Cai Y, Corke H, Sun M. Systematic evaluation of natural phenolic antioxidants from 133 Indian medicinal plants. Food Chem. 2007;102:938–53. doi: 10.1016/j.foodchem.2006.06.033.
    1. Burgering BM, Coffer PJ. Protein kinase B (c-Akt) in phosphatidylinositol-3-OH kinase signal transduction. Nature. 1995;376:599–602. doi: 10.1038/376599a0.
    1. Weickert MO, Piffer AFH. Signalling mechanisms linking hepatic glucose and lipid metabolism. Diabetologia. 2006;49:1732–41. doi: 10.1007/s00125-006-0295-3.
    1. Kile BT, Schulman BA, Alexander WS, Nicola NA, Martin HM, Hilton DJ. The SOCS box: A tale of destruction and degradation. Trends Biochem Sci. 2002;27:235–41. doi: 10.1016/S0968-0004(02)02085-6.
    1. Schinner S, Scherbaum WA, Bornstein SR, Barthel A. Molecular mechanisms of insulin resistance. Diabet Med. 2005;22:674–82. doi: 10.1111/j.1464-5491.2005.01566.x.
    1. Kaszubska W, Falls HD, Schaefer VG, Haasch D, Frost L, Hessler P, et al. Protein tyrosine phosphatase 1B negatively regulates leptin signalling in a hypothalamic cell line. Mol Cell Endocrinol. 2002;195:109–18. doi: 10.1016/S0303-7207(02)00178-8.
    1. Clement S, Krause U, Desmedt F, Tanti JF, Behrends J, Pesesse X, et al. The lipid phosphatase SHIP2 controls insulin sensitivity. Nature. 2001;409:92–7. doi: 10.1038/35051094.
    1. Butler M, McKay RA, Popoff IJ, Gaarde WA, Witchell D, Murray SF, et al. Specific inhibition of PTEN expression reverses hyperglycemia in diabetic mice. Diabetes. 2002;51:1028–34. doi: 10.2337/diabetes.51.4.1028.
    1. Tyagi S, Gupta P, Saini AS, Kaushal C, Sharma S. The peroxisome proliferator activated receptor: A family of nuclear receptors role in various diseases. J Adv Pharm Technol Res. 2011;2:236–40. doi: 10.4103/2231-4040.90879.
    1. Furuhashi M, Hotamisligil GS. Fatty acid-binding proteins: Role in metabolic diseases and potential as drug targets. Nat Rev Drug Discov. 2008;7:489–503. doi: 10.1038/nrd2589.
    1. Richard AJ, Amini-Vaughan Z, Ribnicky DM, Stephens JM. Naringenin inhibits adipogenesis and reduces insulin sensitivity and adiponectin expression in adipocytes. Evid Based Complement Alternat Med. 2013;2013:549750. doi: 10.1155/2013/549750.
    1. Makki K, Froguel P, Wolowczuk I. Adipose tissue in obesity-related inflammation and insulin resistance: Cells, cytokines, and chemokines. ISRN Inflamm. 2013;2013:139239. doi: 10.1155/2013/139239.
    1. Moon HS, Dalamaga M, Kim SY, Polyzos SA, Hamnvik OP, Magkos F, et al. Leptin’s role in lipodystrophic and nonlipodystrophic insulin-resistant and diabetic individuals. Endocr Rev. 2013;34:377–412. doi: 10.1210/er.2012-1053.
    1. Jung UJ, Choi MS. Obesity and its metabolic complications: The role of adipokines and the relationship between obesity, inflammation, insulin resistance, dyslipidemia and nonalcoholic fatty liver disease. Int J Mol Sci. 2014;15:6184–223. doi: 10.3390/ijms15046184.
    1. Arts IC, Hollman PC. Polyphenols and disease risk in epidemiologic studies. Am J Clin Nutr. 2005;81:317S–25S.
    1. Duncan BB, Schmidt MI, Pankow JS, Ballantyne CM, Couper D, Vigo A, et al. Low-grade systemic inflammation and the development of type 2 diabetes: The atherosclerosis risk in communities study. Diabetes. 2003;52:1799–805. doi: 10.2337/diabetes.52.7.1799.
    1. Pickup JC. Inflammation and activated innate immunity in the pathogenesis of type 2 diabetes. Diabetes Care. 2004;27:813–23. doi: 10.2337/diacare.27.3.813.
    1. Rice-Evans CA, Miller NJ, Paganga G. Structure-antioxidant activity relationships of flavonoids and phenolic acids. Free Radic Biol Med. 1996;20:933–56. doi: 10.1016/0891-5849(95)02227-9.
    1. Kandaswami C, Middleton E., Jr Free radical scavenging and antioxidant activity of plant flavonoids. Adv Exp Med Biol. 1994;366:351–76. doi: 10.1007/978-1-4615-1833-4_25.
    1. Pandey KB, Rizvi SI. Plant polyphenols as dietary antioxidants in human health and disease. Oxid Med Cell Longev. 2009;2:270–8. doi: 10.4161/oxim.2.5.9498.
    1. Bahadoran Z, Mirmiran P, Azizi F. Dietary polyphenols as potential nutraceuticals in management of diabetes: A review. J Diabetes Metab Disord. 2013;12:43. doi: 10.1186/2251-6581-12-43.
    1. Hanhineva K, Törrönen R, Bondia-Pons I, Pekkinen J, Kolehmainen M, Mykkänen H, et al. Impact of dietary polyphenols on carbohydrate metabolism. Int J Mol Sci. 2010;11:1365–402. doi: 10.3390/ijms11041365.
    1. Hajiaghaalipour F, Khalilpourfarshbafi M, Arya A. Modulation of glucose transporter protein by dietary flavonoids in type 2 diabetes mellitus. Int J Biol Sci. 2015;11:508–24. doi: 10.7150/ijbs.11241.
    1. Campanero MA, Escolar M, Perez G, Garcia-Quetglas E, Sadaba B, Azanza JR. Simultaneous determination of diosmin and diosmetin in human plasma by ion trap liquid chromatography-atmospheric pressure chemical ionization tandem mass spectrometry: application to a clinical pharmacokinetic study. J Pharm Biomed Anal. 2010;51:875–881. doi: 10.1016/j.jpba.2009.09.012.
    1. Manuel Y, Keenoy B, Vertommen J, De Leeuw I. The effect of flavonoid treatment on the glycation and antioxidant status in type I diabetic patients. Diabetes Nutr Metab. 1999;12:256–63.
    1. Pari L, Srinivasan S. Antihyperglycemic effect of diosmin on hepatic key enzymes of carbohydrate metabolism in streptozotocin-nicotinamide-induced diabetic rats. Biomed Pharmacother. 2010;64:477–81. doi: 10.1016/j.biopha.2010.02.001.
    1. Srinivasan S, Pari L. Antihyperlipidemic effect of diosmin: A citrus flavonoid on lipid metabolism in experimental diabetic rats. J Funct Foods. 2012;5:484–92. doi: 10.1016/j.jff.2012.12.004.
    1. Jain D, Bansal MK, Dalvi R, Upganlawar A, Somani R. Protective effect of diosmin against diabetic neuropathy in experimental rats. J Integr Med. 2014;12:35–41. doi: 10.1016/S2095-4964(14)60001-7.
    1. Srinivasan S, Pari L. Ameliorative effect of diosmin, a citrus flavonoid against streptozotocin-nicotinamide generated oxidative stress induced diabetic rats. Chem Biol Interact. 2012;195:43–51. doi: 10.1016/j.cbi.2011.10.003.
    1. Vertommen J, van den Enden M, Simoens L, de Leeuw I. Flavonoid treatment reduces glycation and lipid peroxidation in experimental diabetic rats. Phytother Res. 1994;8:430–32. doi: 10.1002/ptr.2650080711.
    1. Du HQ. Isolation and identification of fisetin in Cotinus coggygria. Zhong Yao Tong Bao. 1983;8:29–30.
    1. Arai Y, Watanabe S, Kimira M, Shimoi K, Mochizuki R, Kinae N. Dietary intakes of flavonols, flavones and isoflavones by Japanese women and the inverse correlation between quercetin intake and plasma LDL cholesterol concentration. J Nutr. 2000;130:2243–50.
    1. Constantin RP, Constantin J, Pagadigorria CL, Ishii-Iwamoto EL, Bracht A, Ono Mde K, et al. The actions of fisetin on glucose metabolism in the rat liver. Cell Biochem Funct. 2010;28:149–58. doi: 10.1002/cbf.1635.
    1. Prasath GS, Pillai S, Subramanian SP. Fisetin improves glucose homeostasis through the inhibition of gluconeogenic enzymes in hepatic tissues of streptozotocin induced diabetic rats. Eur J Pharmacol. 2014;740:248–54. doi: 10.1016/j.ejphar.2014.06.065.
    1. Prasath GS, Sundaram CS, Subramanian SP. Fisetin averts oxidative stress in pancreatic tissues of streptozotocin-induced diabetic rats. Endocrine. 2013;44:359–68. doi: 10.1007/s12020-012-9866-x.
    1. Kim HJ, Kim SH, Yun JM. Fisetin inhibits hyperglycemia-induced proinflammatory cytokine production by epigenetic mechanisms. Evid Based Complement Alternat Med. 2012;2012:639469.
    1. Kan E, Kiliçkan E, Ayar A, Colak R. Effects of two antioxidants; α-lipoic acid and fisetin against diabetic cataract in mice. Int Ophthalmol. 2015;35:115–20. doi: 10.1007/s10792-014-0029-3.
    1. Prasath GS, Subramanian SP. Antihyperlipidemic effect of fisetin, a bioflavonoid of strawberries, studied in streptozotocin-induced diabetic rats. J Biochem Mol Toxicol. 2014;28:442–9. doi: 10.1002/jbt.21583.
    1. Kwak S, Ku SK, Bae JS. Fisetin inhibits high-glucose-induced vascular inflammation in vitro and in vivo. Inflamm Res. 2014;63:779–87. doi: 10.1007/s00011-014-0750-4.
    1. Prasath GS, Subramanian SP. Fisetin, a tetra hydroxy flavone recuperates antioxidant status and protects hepatocellular ultrastructure from hyperglycemia mediated oxidative stress in streptozotocin induced experimental diabetes in rats. Food Chem Toxicol. 2013;59:249–55. doi: 10.1016/j.fct.2013.05.062.
    1. Prasath GS, Subramanian SP. Modulatory effects of fisetin, a bioflavonoid, on hyperglycemia by attenuating the key enzymes of carbohydrate metabolism in hepatic and renal tissues in streptozotocin-induced diabetic rats. Eur J Pharmacol. 2011;668:492–6. doi: 10.1016/j.ejphar.2011.07.021.
    1. Maher P, Dargusch R, Ehren JL, Okada S, Sharma K, Schubert D. Fisetin lowers methylglyoxal dependent protein glycation and limits the complications of diabetes. PLoS One. 2011;6:e21226. doi: 10.1371/journal.pone.0021226.
    1. Ricardo KFS, Toledo de Oliveira T, Nagem TJ, Pinto AS, Oliveira MGA, Soares JF. Effect of flavonoids morin; quercetin and nicotinic acid on lipid metabolism of rats experimentally fed with triton. Braz Arch Biol Technol. 2001;44:263–67. doi: 10.1590/S1516-89132001000300007.
    1. Sreedharan V, Venkatachalam KK, Namasivayam N. Effect of morin on tissue lipid peroxidation and antioxidant status in 1, 2-dimethylhydrazine induced experimental colon carcinogenesis. Invest New Drugs. 2009;27:21–30. doi: 10.1007/s10637-008-9136-1.
    1. Sendrayaperumal V, Iyyam Pillai S, Subramanian S. Design, synthesis and characterization of zinc-morin, a metal flavonol complex and evaluation of its antidiabetic potential in HFD-STZ induced type 2 diabetes in rats. Chem Biol Interact. 2014;219:9–17. doi: 10.1016/j.cbi.2014.05.003.
    1. Abuohashish HM, Al-Rejaie SS, Al-Hosaini KA, Parmar MY, Ahmed MM. Alleviating effects of morin against experimentally-induced diabetic osteopenia. Diabetol Metab Syndr. 2013;5:5. doi: 10.1186/1758-5996-5-5.
    1. Wang X, Zhang DM, Gu TT, Ding XQ, Fan CY, Zhu Q, et al. Morin reduces hepatic inflammation-associated lipid accumulation in high fructose-fed rats via inhibiting sphingosine kinase 1/sphingosine 1-phosphate signaling pathway. Biochem Pharmacol. 2013;86:1791–804. doi: 10.1016/j.bcp.2013.10.005.
    1. Paoli P, Cirri P, Caselli A, Ranaldi F, Bruschi G, Santi A, et al. The insulin-mimetic effect of morin: A promising molecule in diabetes treatment. Biochim Biophys Acta. 2013;30:3102–11. doi: 10.1016/j.bbagen.2013.01.017.
    1. Vanitha P, Uma C, Suganya N, Bhakkiyalakshmi E, Suriyanarayanan S, Gunasekaran P, et al. Modulatory effects of morin on hyperglycemia by attenuating the hepatic key enzymes of carbohydrate metabolism and β-cell function in streptozotocin-induced diabetic rats. Environ Toxicol Pharmacol. 2014;37:326–35. doi: 10.1016/j.etap.2013.11.017.
    1. Kapoor R, Kakkar P. Protective role of morin, a flavonoid, against high glucose induced oxidative stress mediated apoptosis in primary rat hepatocytes. PLoS One. 2012;7:e41663. doi: 10.1371/journal.pone.0041663.
    1. Miyake Y, Yamamoto K, Tsujihara N, Osawa T. Protective effects of lemon flavonoids on oxidative stress in diabetic rats. Lipids. 1998;33:689–95. doi: 10.1007/s11745-998-0258-y.
    1. Zhang WY, Lee JJ, Kim Y, Kim IS, Han JH, Lee SG, et al. Effect of eriodictyol on glucose uptake and insulin resistance in vitro. J Agric Food Chem. 2012;60:7652–8. doi: 10.1021/jf300601z.
    1. Bucolo C, Leggio GM, Drago F, Salomone S. Eriodictyol prevents early retinal and plasma abnormalities in streptozotocin-induced diabetic rats. Biochem Pharmacol. 2012;84:88–92. doi: 10.1016/j.bcp.2012.03.019.
    1. Emim JA, Oliveira AB, Lapa AJ. Pharmacological evaluation of the anti-inflammatory activity of a citrus bioflavonoid, hesperidin, and the isoflavonoids, duartin and claussequinone, in rats and mice. J Pharm Pharmacol. 1994;46:118–22. doi: 10.1111/j.2042-7158.1994.tb03753.x.
    1. Kawaguchi K, Mizuno T, Aida K, Uchino K. Hesperidin as an inhibitor of lipases from porcine pancreas and Pseudomonas. Biosci Biotechnol Biochem. 1997;61:102–4. doi: 10.1271/bbb.61.102.
    1. Visnagri A, Kandhare AD, Chakravarty S, Ghosh P, Bodhankar SL. Hesperidin, a flavanoglycone attenuates experimental diabetic neuropathy via modulation of cellular and biochemical marker to improve nerve functions. Pharm Biol. 2014;52:814–28. doi: 10.3109/13880209.2013.870584.
    1. Gumieniczek A. Effect of the new thiazolidinedione-pioglitazone on the development of oxidative stress in liver and kidney of diabetic rabbits. Life Sci. 2003;74:553–62. doi: 10.1016/j.lfs.2003.03.004.
    1. Shi X, Liao S, Mi H, Guo C, Qi D, Li F, et al. Hesperidin prevents retinal and plasma abnormalities in streptozotocin-induced diabetic rats. Molecules. 2012;17:12868–81. doi: 10.3390/molecules171112868.
    1. Akiyama S, Katsumata S, Suzuki K, Ishimi Y, Wu J, Uehara M. Dietary hesperidin exerts hypoglycemic and hypolipidemic effects in streptozotocin-induced marginal type 1 diabetic rats. J Clin Biochem Nutr. 2010;46:87–92. doi: 10.3164/jcbn.09-82.
    1. Yo A, Sharma PK, Shrivastava B, Ojha S, Upadhya HM, Arya DS, et al. Hesperidin produces cardioprotective activity via PPAR-γ pathway in ischemic heart disease model in diabetic rats. PLoS One. 2014;9:e111212. doi: 10.1371/journal.pone.0111212.
    1. Mahmoud AM, Ashour MB, Abdel-Moneim A, Ahmed OM. Hesperidin and naringin attenuate hyperglycemia-mediated oxidative stress and proinflammatory cytokine production in high fat fed/streptozotocin-induced type 2 diabetic rats. J. Diabetes Complicat. 2012;26:483–90. doi: 10.1016/j.jdiacomp.2012.06.001.
    1. Kakadiya J, Mulani H, Shah N. Protective effect of hesperidin on cardiovascular complication in experimentally induced myocardial infarction in diabetes in rats. J Basic Clin Pharm. 2010;1:85–91.
    1. Akiyama S, Katsumata S, Suzuki K, Nakaya Y, Ishimi Y, Uehara M. Hypoglycemic and hypolipidemic effects of hesperidin and cyclodextrin-clathrated hesperetin in Goto-Kakizaki rats with type 2 diabetes. Biosci Biotechnol Biochem. 2009;73:2779–82. doi: 10.1271/bbb.90576.
    1. Jung UJ, Lee MK, Jeong KS, Choi MS. The hypoglycemic effects of hesperidin and naringin are partly mediated by hepatic glucose-regulating enzymes in C57BL/KsJ-db/db mice. J Nutr. 2004;134:2499–503.
    1. Ashafaq M, Varshney L, Khan MH, Salman M, Naseem M, Wajid S, et al. Neuromodulatory effects of hesperidin in mitigating oxidative stress in streptozotocin induced diabetes. Biomed Res Int. 2014;249031:9.
    1. Wilcox LJ, Borradaile NM, Huff MW. Antiatherogenic properties of naringenin, a citrus flavonoid. Cardiovasc Drug Rev. 1999;17:160–78. doi: 10.1111/j.1527-3466.1999.tb00011.x.
    1. Van Acker FA, Schouten O, Haenen GR, van der Vijgh WJ, Bast A. Flavonoids can replace alpha-tocopherol as an antioxidant. FEBS Lett. 2000;473:145–8. doi: 10.1016/S0014-5793(00)01517-9.
    1. Sanchez-Salgado JC, Ortiz-Andrade RR, Aguirre-Crespo F, Vergara-Galicia J, Leon-Rivera I, Montes S, et al. Hypoglycemic, vasorelaxant and hepatoprotective effects of Cochlospermum vitifolium (willd.) sprengel: A potential agent for the treatment of metabolic syndrome. J Ethnopharmacol. 2007;109:400–5. doi: 10.1016/j.jep.2006.08.008.
    1. Priscilla DH, Roy D, Suresh A, Kumar V, Thirumurugan K. Naringenin inhibits α-glucosidase activity: A promising strategy for the regulation of postprandial hyperglycemia in high fat diet fed streptozotocin induced diabetic rats. Chem Biol Interact. 2014;210:77–85. doi: 10.1016/j.cbi.2013.12.014.
    1. Li JM, Che CT, Lau CBS, Leung PS, Cheng CHK. Inhibition of intestinal andrenal Na+-glucose co-transporter by naringenin. Int J Biochem Cell Biol. 2006;38:985–95. doi: 10.1016/j.biocel.2005.10.002.
    1. Fallahi F, Roghani M, Moghadami S. Citrus flavonoid naringenin improves aortic reactivity in streptozotocin-diabetic rats. Indian J Pharmacol. 2012;44:382–96. doi: 10.4103/0253-7613.96350.
    1. Pu P, Gao DM, Mohamed S, Chen J, Zhang J, Zhou XY, et al. Naringin ameliorates metabolic syndrome by activating AMP-activated protein kinase in mice fed a high-fat diet. Arch Biochem Biophys. 2012;518:61–70. doi: 10.1016/j.abb.2011.11.026.
    1. Zygmunt K, Faubert B, MacNeil J, Tsiani E. Naringenin, a citrus flavonoid, increases muscle cell glucose uptake via AMPK. Biochem Biophys Res Commun. 2010;398:178–83. doi: 10.1016/j.bbrc.2010.06.048.
    1. Choi JS, Yokozawa T, Oura H. Improvement of hyperglycemia, hyperlipemiain streptozotocin-diabetic rats by a methanolic extract of Prunus davidiana stems, its main component. Prunin Planta Med. 1991;57:208–11. doi: 10.1055/s-2006-960075.
    1. Kannappan S, Anuradha CV. Naringenin enhances insulin-stimulated tyrosine phosphorylation and improves the cellular actions of insulin in a dietary model of metabolic syndrome. Eur J Nutr. 2010;49:101–9. doi: 10.1007/s00394-009-0054-6.
    1. Mulvihill EE, Allister EM, Sutherland BG, Telford DE, Sawyez CG, Edwards JY, et al. Naringenin prevents dyslipidemia, apolipoprotein B overproduction, and hyperinsulinemia in LDL receptor-null mice with diet-induced insulin resistance. Diabetes. 2009;58:2198–10. doi: 10.2337/db09-0634.
    1. Priscilla DH, Jayakumar M, Thirumurugan K. Flavanone naringenin: An effective antihyperglycemic and antihyperlipidemic nutraceutical agent on high fat diet fed streptozotocin induced type 2 diabetic rats. J Funct Foods. 2015;14:363–73. doi: 10.1016/j.jff.2015.02.005.
    1. Yoshida H, Watanabe H, Ishida A, Watanabe W, Narumi K, Atsumi T, et al. Naringenin suppresses macrophage infiltration into adipose tissue in an early phase of high-fat diet-induced obesity. Biochem Biophys Res Commun. 2014;454:95–101. doi: 10.1016/j.bbrc.2014.10.038.
    1. Hasanein P, Fazeli F. Role of naringenin in protection against diabetic hyperalgesia and tactile allodynia in male Wistar rats. J Physiol Biochem. 2014;70:997–1006. doi: 10.1007/s13105-014-0369-5.
    1. Bhattacharya S, Oksbjerg N, Young JF, Jeppesen PB. Caffeic acid, naringenin and quercetin enhance glucose-stimulated insulin secretion and glucose sensitivity in INS-1E cells. Diabetes Obes Metab. 2014;16:602–12. doi: 10.1111/dom.12236.
    1. Yoshida H, Watanabe W, Oomagari H, Tsuruta E, Shida M, Kurokawa M. Citrus flavonoid naringenin inhibits TLR2 expression in adipocytes. J Nutr Biochem. 2013;24:1276–84. doi: 10.1016/j.jnutbio.2012.10.003.
    1. Rahigude A, Bhutada P, Kaulaskar S, Aswar M, Otari K. Participation of antioxidant and cholinergic system in protective effect of naringenin against type-2 diabetes-induced memory dysfunction in rats. Neuroscience. 2012;226:62–72. doi: 10.1016/j.neuroscience.2012.09.026.
    1. Ross JA, Kasum CM. Dietary flavonoids: Bioavailability, metabolic effects, and safety. Annu Rev Nutr. 2002;22:19–34. doi: 10.1146/annurev.nutr.22.111401.144957.
    1. Panda S, Kar A. Apigenin (4',5,7-trihydroxyflavone) regulates hyperglycaemia, thyroid dysfunction and lipid peroxidation in alloxan-induced diabetic mice. J Pharm Pharmacol. 2007;59:1543–8. doi: 10.1211/jpp.59.11.0012.
    1. Mrazek AA, Porro LJ, Bhatia V, Falzon M, Spratt H, Zhou J, et al. Apigenin inhibits pancreatic stellate cell activity in pancreatitis. J Surg Res. 2015;196:8–16. doi: 10.1016/j.jss.2015.02.032.
    1. Suh KS, Oh S, Woo JT, Kim SW, Kim JW, Kim YS, et al. Apigenin attenuates 2-deoxy-D-ribose-induced oxidative cell damage in HIT-T15 pancreatic β-cells. Biol Pharm Bull. 2012;35:121–6. doi: 10.1248/bpb.35.121.
    1. Hossain CM, Ghosh MK, Satapathy BS, Dey NS, Mukherjee B. Apigenin causes biochemical modulation, GLUT4 and Cd38 alterations to improve diabetes and to protect damages of some vital organs in experimental diabetes. Am J Pharmacol Toxicol. 2014;9:39–52. doi: 10.3844/ajptsp.2014.39.52.
    1. Zhang X, Wang G, Gurley EC, Zhou H. Flavonoid apigenin inhibits lipopolysaccharide-induced inflammatory response through multiple mechanisms in macrophages. PLoS One. 2014;9:e107072. doi: 10.1371/journal.pone.0107072.
    1. Yamagata K, Miyashita A, Matsufuji H, Chino M. Dietary flavonoid apigenin inhibits high glucose and tumor necrosis factor alpha-induced adhesion molecule expression in human endothelial cells. J Nutr Biochem. 2010;21:116–24. doi: 10.1016/j.jnutbio.2008.11.003.
    1. Cazarolli LH, Folador P, Moresco HH, Brighente IM, Pizzolatti MG, Silva FR. Mechanism of action of the stimulatory effect of apigenin-6-C-(2''-O-alpha-l-rhamnopyranosyl)-beta-L-fucopyranoside on 14C-glucose uptake. Chem Biol Interact. 2009;179:407–12. doi: 10.1016/j.cbi.2008.11.012.
    1. Kim YO, Leem K, Park J, Lee P, Ahn DK, Lee BC, et al. Cytoprotective effect of Scutellaria baicalensis in CA1 hippocampal neurons of rats after global cerebral ischemia. J Ethnopharmacol. 2001;77:183–88. doi: 10.1016/S0378-8741(01)00283-5.
    1. Lapchak PA, Maher P, Schubert D, Zivin JA. Baicalein, an antioxidant12/15-lipoxygenase inhibitor improves clinical rating scores following multiple infarct embolic strokes. Neuroscience. 2007;150:585–91. doi: 10.1016/j.neuroscience.2007.09.033.
    1. Fu Y, Luo J, Jia Z, Zhen W, Zhou K, Gilbert E, et al. Baicalein protects against type 2 diabetes via promoting islet β-cell function in obese diabetic mice. Int J Endocrinol. 2014;2014:846742.
    1. Stavniichuk R, Drel VR, Shevalye H, Maksimchyk Y, Kuchmerovska TM, Nadler JL, et al. Baicalein alleviates diabetic peripheral neuropathy through inhibition of oxidative-nitrosative stress and p38 MAPK activation. Exp Neurol. 2011;230:106–13. doi: 10.1016/j.expneurol.2011.04.002.
    1. Ahad A, Mujeeb M, Ahsan H, Siddiqui WA. Prophylactic effect of baicalein against renal dysfunction in type 2 diabetic rats. Biochimie. 2014;106:101–10. doi: 10.1016/j.biochi.2014.08.006.
    1. El-Bassossy HM, Hassan NA, Mahmoud MF, Fahmy A. Baicalein protects against hypertension associated with diabetes: Effect on vascular reactivity and stiffness. Phytomedicine. 2014;21:1742–45. doi: 10.1016/j.phymed.2014.08.012.
    1. Pu P, Wang XA, Salim M, Zhu LH, Wang L, Chen KJ, et al. Baicalein, a natural product, selectively activating AMPKα(2) and ameliorates metabolic disorder in diet-induced mice. Mol Cell Endocrinol. 2012;362:128–38. doi: 10.1016/j.mce.2012.06.002.
    1. Qi Z, Xu Y, Liang Z, Li S, Wang J, Wei Y, et al. Baicalein alters PI3K/Akt/GSK3β signaling pathway in rats with diabetes-associated cognitive deficits. Int J Clin Exp Med. 2015;8:1993–2000.
    1. Siess MH, LeBon AM, Canivenc-Laver MC, Amiot MJ, Sabatier S, Aubert SY, et al. Flavonoids of honey and propolis: Characterization and effects on hepatic drug-metabolising enzymes and benzo[a]pyrene-DNA binding in rats. J Agric Food Chem. 1996;44:2379–83. doi: 10.1021/jf9504733.
    1. Dhawan K, Kumar S, Sharma A. Beneficial effects of chrysin and benzoflavone on virility in 2-year-old male rats. J Med Food. 2002;5:43–8. doi: 10.1089/109662002753723214.
    1. Sandborn WJ, Faubion WA. Clinical pharmacology of inflammatory bowel disease therapies. Curr Gastroenterol Rep. 2000;2:440–5. doi: 10.1007/s11894-000-0005-0.
    1. Williams CA, Harborne JB, Newman M, Greenham J, Eagles J. Chrysin and other leaf exudate flavonoids in the genus Pelargonium. Phytochemistry. 1997;46:1349–53. doi: 10.1016/S0031-9422(97)00514-1.
    1. Ahad A, Ganai AA, Mujeeb M, Siddiqui WA. Chrysin, an anti-inflammatory molecule, abrogates renal dysfunction in type 2 diabetic rats. Toxicol Appl Pharmacol. 2014;279:1–7. doi: 10.1016/j.taap.2014.05.007.
    1. Li R, Zang A, Zhang L, Zhang H, Zhao L, Qi Z, et al. Chrysin ameliorates diabetes-associated cognitive deficits in Wistar rats. Neurol Sci. 2014;35:1527–32. doi: 10.1007/s10072-014-1784-7.
    1. Sirovina D, Orsolić N, Koncić MZ, Kovacević G, Benković V, Gregorović G. Quercetin vs chrysin: Effect on liver histopathology in diabetic mice. Hum Exp Toxicol. 2013;32:1058–66. doi: 10.1177/0960327112472993.
    1. El-Bassossy HM, Abo-Warda SM, Fahmy A. Chrysin and luteolin attenuate diabetes-induced impairment in endothelial-dependent relaxation: Effect on lipid profile, AGEs and NO generation. Phytother Res. 2013;27:1678–84. doi: 10.1002/ptr.4917.
    1. Neuhouser ML. Dietary flavonoids and cancer risk: Evidence from human population studies. Nutr Cancer. 2004;50:1–7. doi: 10.1207/s15327914nc5001_1.
    1. Miean KH, Mohamed S. Flavonoid (myricetin, quercetin, kaempferol, luteolin, and apigenin) content of edible tropical plants. J Agric Food Chem. 2001;49:3106–12. doi: 10.1021/jf000892m.
    1. Gates MA, Tworoger SS, Hecht JL, De Vivo I, Rosner B, Hankinson SE, et al. A prospective study of dietary flavonoid intake and incidence of epithelial ovarian cancer. Int J Cancer. 2007;121:2225–32. doi: 10.1002/ijc.22790.
    1. Sun T, Xu Z, Wu CT, Janes M, Prinyawiwatkul W, No HK. Antioxidant activities of different colored sweet bell peppers (Capsicum annuum L) J Food Sci. 2007;72:98–102. doi: 10.1111/j.1750-3841.2006.00245.x.
    1. Ding L, Jin D, Chen X. Luteolin enhances insulin sensitivity via activation of PPARγ transcriptional activity in adipocytes. J Nutr Biochem. 2010;21:941–47. doi: 10.1016/j.jnutbio.2009.07.009.
    1. Liu Y, Fu X, Lan N, Li S, Zhang J, Wang S, et al. Luteolin protects against high fat diet-induced cognitive deficits in obesity mice. Behav Brain Res. 2014;267:178–188. doi: 10.1016/j.bbr.2014.02.040.
    1. Wang GG, Lu XH, Li W, Zhao X, Zhang C. Protective effects of luteolin on diabetic nephropathy in STZ-induced diabetic rats. Evid Based Complement Alternat Med. 2011;2011:323171.
    1. Deqiu Z, Kang L, Jiali Y, Baolin L, Gaolin L. Luteolin inhibits inflammatory response and improves insulin sensitivity in the endothelium. Biochimie. 2011;93:506–12. doi: 10.1016/j.biochi.2010.11.002.
    1. Ding Y, Shi X, Shuai X, Xu Y, Liu Y, Liang X, et al. Luteolin prevents uric acid-induced pancreatic β-cell dysfunction. J Biomed Res. 2014;28:292–8. doi: 10.7555/JBR.28.20130170.
    1. Kim HJ, Lee W, Yun JM. Luteolin inhibits hyperglycemia-induced proinflammatory cytokine production and its epigenetic mechanism in human monocytes. Phytother Res. 2014;28:1383–91. doi: 10.1002/ptr.5141.
    1. Kwon EY, Jung UJ, Park T, Yun JW, Choi MS. Luteolin attenuates hepatic steatosis and insulin resistance through the interplay between the liver and adipose tissue in mice with diet-induced obesity. Diabetes. 2015;64:1658–69. doi: 10.2337/db14-0631.
    1. Yang JT, Qian LB, Zhang FJ, Wang J, Ai H, Tang LH, et al. Cardioprotective effects of luteolin on ischemia/reperfusion injury in diabetic rats are modulated by eNOS and the mitochondrial permeability transition pathway. J Cardiovasc Pharmacol. 2015;65:349–56. doi: 10.1097/FJC.0000000000000202.
    1. Xu N, Zhang L, Dong J, Zhang X, Chen YG, Bao B, et al. Low-dose diet supplement of a natural flavonoid, luteolin, ameliorates diet-induced obesity and insulin resistance in mice. Mol Nutr Food Res. 2014;58:1258–68. doi: 10.1002/mnfr.201300830.
    1. Kim MS, Hur HJ, Kwon DY, Hwang JT. Tangeretin stimulates glucose uptake via regulation of AMPK signaling pathways in C2C12 myotubes and improves glucose tolerance in high-fat diet-induced obese mice. Mol Cell Endocrinol. 2012;358:127–34. doi: 10.1016/j.mce.2012.03.013.
    1. Sundaram R, Shanthi P, Sachdanandam P. Effect of tangeretin, a polymethoxylated flavone on glucose metabolism in streptozotocin-induced diabetic rats. Phytomedicine. 2014;21:793–9. doi: 10.1016/j.phymed.2014.01.007.
    1. Miyata Y, Tanaka H, Shimada A, Sato T, Ito A, Yamanouchi T, et al. Regulation of adipocytokine secretion and adipocyte hypertrophy by polymethoxy flavonoids, nobiletin and tangeretin. Life Sci. 2011;88:613–18. doi: 10.1016/j.lfs.2011.01.024.
    1. Tai MC, Tsang SY, Chang LY, Xue H. Therapeutic potential of wogonin: A naturally occurring flavonoid. CNS Drug Rev. 2005;11:141e50.
    1. Bak EJ, Kim J, Choi YH, Kim JH, Lee DE, Woo GH, et al. Wogonin ameliorates hyperglycemia and dyslipidemia via PPARα activation in db/db mice. Clin Nutr. 2014;33:156–63. doi: 10.1016/j.clnu.2013.03.013.
    1. Ku SK, Bae JS. Baicalin, baicalein and wogonin inhibits high glucose-induced vascular inflammation in vitro and in vivo. BMB Rep. 2015;48:519–24. doi: 10.5483/BMBRep.2015.48.9.017.
    1. Zhang YM, Li MX, Tang Z, Wang CH. Wogonin suppresses osteopontin expression in adipocytes by activating PPARα. Acta Pharmacol Sin. 2015;36:987–97. doi: 10.1038/aps.2015.37.
    1. Yokozawa T, Kim HY, Cho EJ, Choi JS, Chung HY. Antioxidant effects of isorhamnetin 3,7-di-O-beta-D-glucopyranoside isolated from mustard leaf (Brassica juncea) in rats with streptozotocin-induced diabetes. J Agric Food Chem. 2002;50:5490–95. doi: 10.1021/jf0202133.
    1. Lee YS, Lee S, Lee HS, Kim BK, Ohuchi K, Shin KH. Inhibitory effects of isorhamnetin-3-O-beta-D-glucoside from Salicornia herbacea on rat lens aldose reductase and sorbitol accumulation in streptozotocin-induced diabetic rat tissues. Biol Pharm Bull. 2005;28:916–8. doi: 10.1248/bpb.28.916.
    1. Rodríguez-Rodríguez C, Torres N, Gutiérrez-Uribe JA, Noriega LG, Torre-Villalvazo I, Leal-Díaz AM, et al. The effect of isorhamnetin glycosides extracted from Opuntia ficus-indica in a mouse model of diet induced obesity. Food Funct. 2015;6:805–15. doi: 10.1039/C4FO01092B.
    1. Lee J, Jung E, Lee J, Kim S, Huh S, Kim Y, et al. Isorhamnetin represses adipogenesis in 3 T3-L1 cells. Obesity (Silver Spring) 2009;17:226–32. doi: 10.1038/oby.2008.472.
    1. An G, Gallegos J, Morris ME. The bioflavonoid kaempferol is an abcg2 substrateand inhibits abcg2-mediated quercetin efflux. Drug Metab Dispos. 2011;39:426–32. doi: 10.1124/dmd.110.035212.
    1. Hakkinen SH, Karenlampi SO, Heinonen IM, Mykkanen HM, Torronen AR. Content of the flavonolsquercetin, myricetin, and kaempferolin 25 edible berries. J Agric Food Chem. 1999;47:2274–79. doi: 10.1021/jf9811065.
    1. Nirmala P, Ramanathan M. Effect of kaempferol on lipid peroxidation and antioxidant status in 1,2-dimethyl hydrazine induced colorectal carcinoma in rats. Eur J Pharmacol. 2011;654:75–9. doi: 10.1016/j.ejphar.2010.11.034.
    1. Jorge AP, Horst H, de Sousa E, Pizzolatti MG, Silva FR. Insulinomimetic effects of kaempferitrin on glycaemia and on 14C-glucose uptake in rat soleus muscle. Chem Biol Interact. 2004;149:89–96. doi: 10.1016/j.cbi.2004.07.001.
    1. Zhang Y, Liu D. Flavonol kaempferol improves chronic hyperglycemia-impaired pancreatic beta-cell viability and insulin secretory function. Eur J Pharmacol. 2011;670:325–32. doi: 10.1016/j.ejphar.2011.08.011.
    1. Zanatta L, Rosso A, Folador P, Figueiredo MS, Pizzolatti MG, Leite LD, et al. Insulinomimetic effect of kaempferol 3-neohesperidoside on the rat soleus muscle. J Nat Prod. 2008;71:532–5. doi: 10.1021/np070358+.
    1. Al-Numair KS, Chandramohan G, Veeramani C, Alsaif MA. Ameliorative effect of kaempferol, a flavonoid, on oxidative stress in streptozotocin-induced diabetic rats. Redox Rep. 2015;20:198–209. doi: 10.1179/1351000214Y.0000000117.
    1. Abo-Salem OM. Kaempferol attenuates the development of diabetic neuropathic pain in mice: Possible anti-inflammatory and anti-oxidant mechanisms. Macedonian J Med Sci. 2014;7:424–30. doi: 10.3889/oamjms.2014.073.
    1. Zang Y, Zhang L, Igarashi K, Yu C. The anti-obesity and anti-diabetic effects of kaempferol glycosides from unripe soybean leaves in high-fat-diet mice. Food Funct. 2015;6:834–41. doi: 10.1039/C4FO00844H.
    1. Al-Numair KS, Veeramani C, Alsaif MA, Chandramohan G. Influence of kaempferol, a flavonoid compound, on membrane-bound ATPases in streptozotocin-induced diabetic rats. Pharm Biol. 2015;8:1–7.
    1. Zhang Y, Zhen W, Maechler P, Liu D. Small molecule kaempferol modulates PDX-1 protein expression and subsequently promotes pancreatic β-cell survival and function via CREB. J Nutr Biochem. 2013;24:638–46. doi: 10.1016/j.jnutbio.2012.03.008.
    1. Alkhalidy H, Moore W, Zhang Y, McMillan R, Wang A, Ali M, et al. Small molecule kaempferol promotes insulin sensitivity and preserved pancreatic β-cell mass in middle-aged obese diabetic mice. J Diabetes Res. 2015;2015:532984. doi: 10.1155/2015/532984.
    1. Kreft S, Knapp M, Kreft I. Extraction of rutin from buckwheat (Fagopyrum esculentum Moench) seeds and determination by capillary electrophoresis. J Agric Food Chem. 1999;47:4649–52. doi: 10.1021/jf990186p.
    1. Huang WY, Zhang HC, Liu WX, Li CY. Survey of antioxidant capacity and phenolic composition of blueberry, blackberry, and strawberry in Nanjing. J Zhejiang Univ Sci B. 2012;13:94–102. doi: 10.1631/jzus.B1100137.
    1. Prince PSM, Kamalakkannan N. Rutin improves glucose homeostasis in streptozotocin diabetic tissues by altering glycolytic and gluconeogenic enzymes. J Biochem Mol Toxicol. 2006;20:96–102. doi: 10.1002/jbt.20117.
    1. Fernandes AA, Novelli EL, Okoshi K, Okoshi MP, Di Muzio BP, Guimarães JF, et al. Influence of rutin treatment on biochemical alterations in experimental diabetes. Biomed Pharmacother. 2010;64:214–19. doi: 10.1016/j.biopha.2009.08.007.
    1. Prince PSM, Kamalakkannan N. Protective effect of rutin on lipids, lipoproteins, lipid metabolizing enzymes and glycoproteins in streptozotocin-induced diabetic rats. J Pharm Pharmacol. 2006;58:1373–83. doi: 10.1211/jpp.58.10.0011.
    1. Hao HH, Shao ZM, Tang DQ, Lu Q, Chen X, Yin XX, et al. Preventive effects of rutin on the development of experimental diabetic nephropathy in rats. Life Sci. 2012;91:959–67. doi: 10.1016/j.lfs.2012.09.003.
    1. Kappel VD, Cazarolli LH, Pereira DF, Postal BG, Zamoner A, Reginatto FH, et al. Involvement of GLUT-4 in the stimulatory effect of rutin on glucose uptake in rat soleus muscle. J Pharm Pharmacol. 2013;65:1179–86. doi: 10.1111/jphp.12066.
    1. Niture NT, Ansari AA, Naik SR. Anti-hyperglycemic activity of rutin in streptozotocin-induced diabetic rats: an effect mediated through cytokines, antioxidants and lipid biomarkers. Indian J Exp Biol. 2014;52:720–7.
    1. Wang YB, Ge ZM, Kang WQ, Lian ZX, Yao J, Zhou CY. Rutin alleviates diabetic cardiomyopathy in a rat model of type 2 diabetes. Exp Ther Med. 2015;9:451–55.
    1. Ola MS, Ahmed MM, Ahmad R, Abuohashish HM, Al-Rejaie SS, Alhomida AS. Neuroprotective effects of rutin in streptozotocin-induced diabetic rat retina. J Mol Neurosci. 2015;56:440–8. doi: 10.1007/s12031-015-0561-2.
    1. Kamalakkannan N, Prince PS. Antihyperglycaemic and antioxidant effect of rutin, a polyphenolic flavonoid, in streptozotocin-induced diabetic wistar rats. Basic Clin Pharmacol Toxicol. 2006;98:97–103. doi: 10.1111/j.1742-7843.2006.pto_241.x.
    1. Butchi Akondi R, Kumar P, Annapurna A, Pujari M. Protective effect of rutin and naringin on sperm quality in streptozotocin induced type 1 diabetic rats. Iran J Pharm Res. 2011;10:585–96.
    1. Hollman PCH, de Vries JHM, van Leeuwen SD, Mengelers MJB, Katan MB. Absorption of dietary quercetin glycosides and quercetin in healthy ileostomy volunteers. Am J Clin Nutr. 1995;62:1276–82.
    1. Alinezhad H, Azimi A, Zare M, Ebrahimzadeh MA, Eslami S, Nabavi SF, et al. Antioxidant and antihemolytic activities of ethanolic extract of flowers, leaves, and stems of Hyssopus officinalis L. var. angustifolius. Int J Food Prop. 2013;16:1169–78. doi: 10.1080/10942912.2011.578319.
    1. Coskun O, Kanter M, Korkmaz A, Oter S. Quercetin, a flavonoid antioxidant, prevents and protects streptozotocin-induced oxidative stress and beta-cell damage in rat pancreas. Pharmacol Res. 2005;51:117–23. doi: 10.1016/j.phrs.2004.06.002.
    1. Stewart LK, Wang Z, Ribnicky D, Soileau JL, Cefalu WT, Gettys TW. Failure of dietary quercetin to alter the temporal progression of insulin resistance among tissues of C57BL/6 J mice during the development of diet-induced obesity. Diabetologia. 2009;52:514–23. doi: 10.1007/s00125-008-1252-0.
    1. Kwon O, Eck P, Chen S, Corpe CP, Lee JH, Kruhlak M, et al. Inhibition of the intestinal glucose transporter GLUT2 by flavonoids. FASEB J. 2007;21:366–77. doi: 10.1096/fj.06-6620com.
    1. Eid HM, Nachar A, Thong F, Sweeney G, Haddad PS. The molecular basis of the antidiabetic action of quercetin in cultured skeletal muscle cells and hepatocytes. Pharmacogn Mag. 2015;11:74–81. doi: 10.4103/0973-1296.149708.
    1. Panchal SK, Poudyal H, Brown L. Quercetin ameliorates cardiovascular, hepatic, and metabolic changes in diet-induced metabolic syndrome in rats. J Nutr. 2012;142:1026–32. doi: 10.3945/jn.111.157263.
    1. Kobori M, Masumoto S, Akimoto Y, Takahashi Y. Dietary quercetin alleviates diabetic symptoms and reduces streptozotocin-induced disturbance of hepatic gene expression in mice. Mol Nutr Food Res. 2009;53:859–68. doi: 10.1002/mnfr.200800310.
    1. Lu Q, Ji XJ, Zhou YX, Yao XQ, Liu YQ, Zhang F, et al. Quercetin inhibits the mTORC1/p70S6K signaling-mediated renal tubular epithelial-mesenchymal transition and renal fibrosis in diabetic nephropathy. Pharmacol Res. 2015;99:237–47. doi: 10.1016/j.phrs.2015.06.006.
    1. Kumar B, Gupta SK, Nag TC, Srivastava S, Saxena R, Jha KA, et al. Retinal neuroprotective effects of quercetin in streptozotocin-induced diabetic rats. Exp Eye Res. 2014;125:193–202. doi: 10.1016/j.exer.2014.06.009.
    1. Alam MM, Meerza D, Naseem I. Protective effect of quercetin on hyperglycemia, oxidative stress and DNA damage in alloxan induced type 2 diabetic mice. Life Sci. 2014;109:8–14. doi: 10.1016/j.lfs.2014.06.005.
    1. Dai X, Ding Y, Zhang Z, Cai X, Li Y. Quercetin and quercitrin protect against cytokine-induced injuries in RINm5F β-cells via the mitochondrial pathway and NF-κB signaling. Int J Mol Med. 2013;31:265–71.
    1. Jeong SM, Kang MJ, Choi HN, Kim JH, Kim JI. Quercetin ameliorates hyperglycemia and dyslipidemia and improves antioxidant status in type 2 diabetic db/db mice. Nutr Res Pract. 2012;6:201–7. doi: 10.4162/nrp.2012.6.3.201.
    1. Lai PB, Zhang L, Yang LY. Quercetin ameliorates diabetic nephropathy by reducing the expressions of transforming growth factor-β1 and connective tissue growth factor in streptozotocin-induced diabetic rats. Ren Fail. 2012;34:83–7. doi: 10.3109/0886022X.2011.623564.
    1. Kim JH, Kang MJ, Choi HN, Jeong SM, Lee YM, Kim JI. Quercetin attenuates fasting and postprandial hyperglycemia in animal models of diabetes mellitus. Nutr Res Pract. 2011;5:107–11. doi: 10.4162/nrp.2011.5.2.107.
    1. Patisaul HB, Jefferson W. The pros and cons of phytoestrogens. Front Neuroendocrinol. 2010;31:400–19. doi: 10.1016/j.yfrne.2010.03.003.
    1. Guo TL, Wang Y, Xiong T, Ling X, Zheng J. Genistein modulation of streptozotocin diabetes in male B6C3F1 mice can be induced by diet. Toxicol Appl Pharmacol. 2014;280:455–66. doi: 10.1016/j.taap.2014.08.028.
    1. Babu PV, Si H, Fu Z, Zhen W, Liu D. Genistein prevents hyperglycemia-induced monocyte adhesion to human aortic endothelial cells through preservation of the cAMP signaling pathway and ameliorates vascular inflammation in obese diabetic mice. J Nutr. 2012;142:724–30. doi: 10.3945/jn.111.152322.
    1. Palanisamy N, Viswanathan P, Anuradha CV. Effect of genistein, a soy isoflavone, on whole body insulin sensitivity and renal damage induced by a high-fructose diet. Ren Fail. 2008;30:645–54. doi: 10.1080/08860220802134532.
    1. Elmarakby AA, Ibrahim AS, Faulkner J, Mozaffari MS, Liou GI, Abdelsayed R. Tyrosine kinase inhibitor, genistein, reduces renal inflammation and injury in streptozotocininduced diabetic mice. Vascular Pharma. 2011;55:149–56. doi: 10.1016/j.vph.2011.07.007.
    1. Fu Z, Gilbert ER, Pfeiffer L, Zhang Y, Fu Y, Liu D. Genistein ameliorates hyperglycemia in a mouse model of nongenetic type 2 diabetes. Appl Physiol Nutr Metab. 2012;37:480–8. doi: 10.1139/h2012-005.
    1. Valsecchi AE, Franchi S, Panerai AE, Rossi A, Sacerdote P, Colleoni M. The soy isoflavone genistein reverses oxidative and inflammatory state, neuropathic pain, neurotrophic and vasculature deficits in diabetes mouse model. Eur J Pharmacol. 2011;650:694–702. doi: 10.1016/j.ejphar.2010.10.060.
    1. El-Kordy EA, Alshahrani AM. Effect of genistein, a natural soy isoflavone, on pancreatic-cells of streptozotocin-induced diabetic rats: Histological and immunohistochemical study. J Microsc Ultrastruct. 2015;3:108–119. doi: 10.1016/j.jmau.2015.03.005.
    1. Weigt C, Hertrampf T, Flenker U, Hülsemann F, Kurnaz P, Fritzemeier KH, et al. Effects of estradiol, estrogen receptor subtype-selective agonists and genistein on glucose metabolism in leptin resistant female Zucker diabetic fatty (ZDF) rats. J Steroid Biochem Mol Biol. 2015;154:12–22. doi: 10.1016/j.jsbmb.2015.06.002.
    1. Gupta SK, Dongare S, Mathur R, Mohanty IR, Srivastava S, Mathur S, et al. Genistein ameliorates cardiac inflammation and oxidative stress in streptozotocin-induced diabetic cardiomyopathy in rats. Mol Cell Biochem. 2015;408:63–72. doi: 10.1007/s11010-015-2483-2.
    1. Kim MJ, Lim Y. Protective effect of short-term genistein supplementation on the early stage in diabetes-induced renal damage. Mediators Inflamm. 2013;510212:14.
    1. Tie L, An Y, Han J, Xiao Y, Xiaokaiti Y, Fan S, et al. Genistein accelerates refractory wound healing by suppressing superoxide and FoxO1/iNOS pathway in type 1 diabetes. J Nutr Biochem. 2013;24:88–96. doi: 10.1016/j.jnutbio.2012.02.011.
    1. Yang W, Wang S, Li L, Liang Z, Wang L. Genistein reduces hyperglycemia and islet cell loss in a high-dosage manner in rats with alloxan-induced pancreatic damage. Pancreas. 2011;40:396–402. doi: 10.1097/MPA.0b013e318204e74d.
    1. Fu Z, Zhang W, Zhen W, Lum H, Nadler J, Bassaganya-Riera J, et al. Genistein induces pancreatic beta-cell proliferation through activation of multiple signaling pathways and prevents insulin-deficient diabetes in mice. Endocrinology. 2010;151:3026–37. doi: 10.1210/en.2009-1294.
    1. Yuan WJ, Jia FY, Meng JZ. Effects of genistein on secretion of extracellular matrix components and transforming growth factor beta in high-glucose-cultured rat mesangial cells. J Artif Organs. 2009;12:242–6. doi: 10.1007/s10047-009-0479-y.
    1. Zhang M, Ikeda K, Xu JW, Yamori Y, Gao XM, Zhang BL. Genistein suppresses adipogenesis of 3 T3-L1 cells via multiple signal pathways. Phytother Res. 2009;23:713–8. doi: 10.1002/ptr.2724.
    1. Kim YS, Kim NH, Jung DH, Jang DS, Lee YM, Kim JM, et al. Genistein inhibits aldose reductase activity and high glucose-induced TGF-beta2 expression in human lens epithelial cells. Eur J Pharmacol. 2008;594:18–25. doi: 10.1016/j.ejphar.2008.07.033.
    1. Choi MS, Jung UJ, Yeo J, Kim MJ, Lee MK. Genistein and daidzein prevent diabetes onset by elevating insulin level and altering hepatic gluconeogenic and lipogenic enzyme activities in non-obese diabetic (NOD) mice. Diabetes Metab Res Rev. 2008;24:74–81. doi: 10.1002/dmrr.780.
    1. Liggins J, Bluck LJ, Runswick S, Atkinson C, Coward WA, Bingham SA. Daidzein and genistein content of fruits and nuts. J Nutr Biochem. 2000;11:326–31. doi: 10.1016/S0955-2863(00)00085-1.
    1. Kapiotis S, Jilma B, Szalay Y. Evidence against an effect of endothelin-1 on blood coagulation, fibrinolysis, and endothelial cell integrity in healthy men. Arterioscler Thromb Vasc Biol. 1997;17:2861–67. doi: 10.1161/01.ATV.17.11.2861.
    1. Park SA, Choi MS, Cho SY, Seo JS, Jung UJ, Kim MJ, et al. Genistein and daidzein modulate hepatic glucose and lipid regulating enzyme activities in C57BL/Ks J-db/db mice. Life Sci. 2006;79:1207–13. doi: 10.1016/j.lfs.2006.03.022.
    1. Cederroth CR, Vinciguerra M, Gjinovci A, Kuhne F, Klein M, Cederroth M, et al. Dietary phytoestrogens activate AMP-activated protein kinase with improvement in lipid and glucose metabolism. Diabetes. 2008;57:1176–85. doi: 10.2337/db07-0630.
    1. Cheong SH, Furuhashi K, Ito K, Nagaoka M, Yonezawa T, Miura Y, et al. Daidzein promotes glucose uptake through glucose transporter 4 translocation to plasma membrane in L6 myocytes and improves glucose homeostasis in type II diabetic model mice. J Nutr Biochem. 2014;25:136–43. doi: 10.1016/j.jnutbio.2013.09.012.
    1. Roghani M, Vaez Mahdavi MR, Jalali-Nadoushan MR, Baluchnejadmojarad T, Naderi G, Roghani-Dehkordi F, et al. Chronic administration of daidzein, a soybean isoflavone, improves endothelial dysfunction and attenuates oxidative stress in streptozotocin-induced diabetic rats. Phytother Res. 2013;27:112–7. doi: 10.1002/ptr.4699.
    1. Cho KW, Lee OH, Banz WJ, Moustaid-Moussa N, Shay NF, Kim YC. Daidzein and the daidzein metabolite, equol, enhance adipocyte differentiation and PPAR γ transcriptional activity. J Nutr Biochem. 2010;21:841–7. doi: 10.1016/j.jnutbio.2009.06.012.
    1. Akkarachiyasit S, Charoenlertkul P, Yibchok-Anun S, Adisakwattana S. Inhibitory activities of cyanidin and its glycosides and synergistic effect with acarbose against intestinal α-glucosidase and pancreatic α-amylase. Int J Mol Sci. 2010;11:3387–96. doi: 10.3390/ijms11093387.
    1. Nizamutdinova IT, Jin YC, Chung JI, Shin SC, Lee SJ, Seo HG, et al. The anti-diabetic effect of anthocyanins in streptozotocin-induced diabetic rats through glucose transporter 4 regulation and prevention of insulin resistance and pancreatic apoptosis. Mol Nutr Food Res. 2009;53:1419–29. doi: 10.1002/mnfr.200800526.
    1. Nasri S, Roghani M, Baluchnejadmojarad T, Rabani T, Balvardi M. Vascular mechanisms of cyanidin-3-glucoside response in streptozotocin-diabetic rats. Pathophysiology. 2011;18:273–8. doi: 10.1016/j.pathophys.2011.03.001.
    1. Zhu W, Jia Q, Wang Y, Zhang Y, Xia M. The anthocyanin cyanidin-3-O-betaglucoside, a flavonoid, increases hepatic glutathione synthesis and protects hepatocytes against reactive oxygen species during hyperglycemia: Involvement of a cAMP-PKA-dependent signaling pathway. Free RadicBiol Med. 2012;52:314–27. doi: 10.1016/j.freeradbiomed.2011.10.483.
    1. Kurimoto Y, Shibayama Y, Inoue S, Soga M, Takikawa M, Ito C, et al. Black soybean seed coat extract ameliorates hyperglycemia and insulin sensitivityvia the activation of AMP-activated protein kinase in diabetic mice. J Agric Food Chem. 2013;61:5558–64. doi: 10.1021/jf401190y.
    1. Jiang X, Tang X, Zhang P, Liu G, Guo H. Cyanidin-3-O-β-glucoside protects primary mouse hepatocytes against high glucose-induced apoptosis by modulating mitochondrial dysfunction and the PI3K/Akt pathway. Biochem Pharmacol. 2014;90:135–44. doi: 10.1016/j.bcp.2014.04.018.
    1. Guo H, Xia M, Zou T, Ling W, Zhong R, Zhang W. Cyanidin 3-glucoside attenuates obesity-associated insulin resistance and hepatic steatosis in high-fat diet-fed and db/db mice via the transcription factor FoxO1. J Nutr Biochem. 2012;23:349–60. doi: 10.1016/j.jnutbio.2010.12.013.
    1. Park S, Kang S, Jeong DY, Jeong SY, Park JJ, Yun HS. Cyanidin and malvidin in aqueous extracts of black carrots fermented with Aspergillus oryzae prevent the impairment of energy, lipid and glucose metabolism in estrogen-deficient rats by AMPK activation. Genes Nutr. 2015;10:455.
    1. Matsukawa T, Inaguma T, Han J, Villareal MO, Isoda H. Cyanidin-3-glucoside derived from black soybeans ameliorate type 2 diabetes through the induction of differentiation of preadipocytes into smaller and insulin-sensitive adipocytes. J Nutr Biochem. 2015;26:860–7. doi: 10.1016/j.jnutbio.2015.03.006.
    1. Sasaki R, Nishimura N, Hoshino H, Isa Y, Kadowaki M, Ichi T, et al. Cyanidin 3-glucoside ameliorates hyperglycemia and insulin sensitivity due to down-regulation of retinol binding protein 4 expression in diabetic mice. Biochem Pharmacol. 2007;74:1619–27. doi: 10.1016/j.bcp.2007.08.008.
    1. Tsao TS, Burcelin R, Charron MJ. Regulation of hexokinase II gene expression by glucose flux in skeletal muscle. J Biol Chem. 1996;271:14959–63. doi: 10.1074/jbc.271.9.4993.
    1. Sun CD, Zhang B, Zhang JK, Xu CJ, Wu YL, Li X, et al. Cyanidin-3-glucoside-rich extract from Chinese bayberry fruit protects pancreatic β-cells and ameliorates hyperglycemia in streptozotocin-induced diabetic mice. J Med Food. 2012;15:288–98. doi: 10.1089/jmf.2011.1806.
    1. Bertuglia S, Malandrino S, Colantuoni A. Effects of the natural flavonoid delphinidin on diabetic microangiopathy. Arzneimittelforschung. 1995;45:481–85.
    1. Gharib A, Faezizadeh Z, Godarzee M. Treatment of diabetes in the mouse model by delphinidin and cyanidin hydrochloride in free and liposomal forms. Planta Med. 2013;79:1599–604. doi: 10.1055/s-0033-1350908.
    1. Mazza G. Compositional and functional properties of saskatoon berry and blueberry. Int J Fruit Sci. 2005;5:101. doi: 10.1300/J492v05n03_10.
    1. Roy M, Sen S, Chakraborti AS. Action of pelargonidin on hyperglycemia and oxidative damage in diabetic rats: Implication for glycation-induced hemoglobin modification. Life Sci. 2008;82:1102–10. doi: 10.1016/j.lfs.2008.03.011.
    1. Mirshekar M, Roghani M, Khalili M, Baluchnejadmojarad T, Arab MS. Chronic oral pelargonidin alleviates streptozotocin-induced diabetic neuropathic hyperalgesia in rat: Involvement of oxidative stress. Iran Biomed J. 2010;14:33–9.
    1. Jayaprakasam B, Vareed SK, Olson LK, Nair MG. Insulin secretion by anthocyanins and anthocyanidins. J Agric Food Chem. 2005;53:2519–23. doi: 10.1021/jf049018+.
    1. Mirshekar M, Roghani M, Khalili M, Baluchnejadmojarad T. Chronic oral pelargonidin alleviates learning and memory disturbances in streptozotocin diabetic rats. Iran J Pharm Res. 2011;10:569–75.

Source: PubMed

3
Subscribe