DOI QR코드

DOI QR Code

Quercetin attenuates fasting and postprandial hyperglycemia in animal models of diabetes mellitus

  • Kim, Ji-Hye (Department of Smart Foods and Drugs, School of Food and Life Science, Inje University) ;
  • Kang, Min-Jung (Department of Smart Foods and Drugs, School of Food and Life Science, Inje University) ;
  • Choi, Ha-Neul (Department of Smart Foods and Drugs, School of Food and Life Science, Inje University) ;
  • Jeong, Soo-Mi (Department of Smart Foods and Drugs, School of Food and Life Science, Inje University) ;
  • Lee, Young-Min (Department of Nutrition, Pusan Paik Hospital) ;
  • Kim, Jung-In (Department of Smart Foods and Drugs, School of Food and Life Science, Inje University)
  • Received : 2011.01.14
  • Accepted : 2011.02.14
  • Published : 2011.04.28

Abstract

The objective of this study was to investigate the hypoglycemic effects of quercetin (QE) in animal models of diabetes mellitus (DM). A starch solution (1 g/kg) with and without QE (100 mg/kg) or acarbose (40 mg/kg) was orally administered to streptozotocin (STZ)-induced diabetic rats after an overnight fast. Postprandial plasma glucose levels were measured and incremental areas under the response curve were calculated. To study the effects of chronic feeding of QE, five-week-old db/db mice were fed an AIN-93G diet, a diet containing QE at 0.08%, or a diet containing acarbose at 0.03% for 7 weeks after 1 week of adaptation. Plasma glucose and insulin, blood glycated hemoglobin, and maltase activity of the small intestine were measured. Oral administration of QE (100 mg/kg) or acarbose (40 mg/kg) to STZ-treated rats significantly decreased incremental plasma glucose levels 30-180 min after a single oral dose of starch and the area under the postprandial glucose response, compared with the control group. QE (0.08% of diet) or acarbose (0.03% of diet) offered to db/db mice significantly reduced both plasma glucose and blood glycated hemoglobin compared to controls without significant influence on plasma insulin. Small intestine maltase activities were significantly reduced by consumption of QE or acarbose. Thus, QE could be effective in controlling fasting and postprandial blood glucose levels in animal models of DM.

Keywords

References

  1. Cheng D. Prevalence, predisposition and prevention of type II diabetes. Nutr Metab (Lond) 2005;2:29. https://doi.org/10.1186/1743-7075-2-29
  2. American Diabetes Association (ADA). Summary of revisions for the 2008 clinical practice recommendations. Diabetes Care 2008;31:S3-4. https://doi.org/10.2337/dc08-S003
  3. Lorenzati B, Zucco C, Miglietta S, Lamberti F, Bruno G. Oral hypoglycemic drugs: pathophysiological basis of their mechanism of action. Pharmaceuticals 2010;3:3005-20. https://doi.org/10.3390/ph3093005
  4. Adewole SO, Caxton-Martins EA, Ojewole JAO. Protective effect of quercetin on the morphology of pancreatic ${\beta}-cells$ of streptozotocintreated diabetic rats. Afr J Tradit Complement Altern Med 2006;4:64-74.
  5. 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. https://doi.org/10.1016/j.phrs.2004.06.002
  6. Shetty AK, Rashmi R, Rajan MGR, Sambaiah K, Salimath PV. Antidiabetic influence of quercetin in streptozotocin-induced diabetic rats. Nutr Res 2004;24:373-81. https://doi.org/10.1016/j.nutres.2003.11.010
  7. Ramachandra R, Shetty AK, Salimath PV. Quercetin alleviates activities of intestinal and renal disaccharidases in streptozotocininduced diabetic rats. Mol Nutr Food Res 2005;49:355-60. https://doi.org/10.1002/mnfr.200400084
  8. Anjaneyulu M, Chopra K. Quercetin, an anti-oxidant bioflavonoid, attenuates diabetic nephropathy in rats. Clin Exp Pharmacol Physiol 2004;31:244-8. https://doi.org/10.1111/j.1440-1681.2004.03982.x
  9. Ishikawa A, Yamashita H, Hiemori M, Inagaki E, Kimoto M, Okamoto M, Tsuji H, Memon AN, Mohammadi A, Natori Y. Characterization of inhibitors of postprandial hyperglycemia from the leaves of Nerium indicum. J Nutr Sci Vitaminol (Tokyo) 2007;53:166-73. https://doi.org/10.3177/jnsv.53.166
  10. Jo SH, Ka EH, Lee HS, Apostolidis E, Jang HD, Kwon YI. Comparison of antioxidant potential and rat intestinal ${\alpha]-glucosidases$inihibitory activities of quercetin, rutin, and isoquercetin. Int J Appl Res Nat Prod 2009;2:52-60.
  11. Standl E, Baumgartl HJ, Fuchtenbusch M, Stemplinger J. Effect of acarbose on additional insulin therapy in type 2 diabetic patients with late failure of sulphonylurea therapy. Diabetes Obes Metab 1999;1:215-20.
  12. Abrahamson MJ. Optimal glycemic control in type 2 diabetes mellitus: Fasting and postprandial glucose in context. Arch Intern Med 2004;164:486-91. https://doi.org/10.1001/archinte.164.5.486
  13. Kim YM, Jeong YK, Wang MH, Lee WY, Rhee HI. Inhibitory effect of pine extract on ${\alpha]-glucosidase$ activity and postprandial hyperglycemia. Nutrition 2005;21:756-61. https://doi.org/10.1016/j.nut.2004.10.014
  14. Tsujita T, Takaku T. Mechanism of the inhibitory action of chestnut astringent skin extract on carbohydrate absorption. J Nutr Sci Vitaminol (Tokyo) 2008;54:416-21. https://doi.org/10.3177/jnsv.54.416
  15. Akbarzadeh A, Norouzian D, Mehrabi MR, Jamshidi Sh, Farhangi A, Allah Verdi A, Mofidian SMA, Lame Rad B. Induction of diabetes by streptozotocin in rats. Indian J Clin Biochem 2007;22:60-4. https://doi.org/10.1007/BF02913315
  16. Lee SK, Hwang JY, Song JH, Jo JR, Kim MJ, Kim ME, Kim JI. Inhibitory activity of Euonymus alatus against alphaglucosidase in vitro and in vivo. Nutr Res Pract 2007;1:184-8. https://doi.org/10.4162/nrp.2007.1.3.184
  17. Raabo E, Terkildsen TC. On the enzymatic determination of blood glucose. Scand J Clin Lab Invest 1960;12:402-7. https://doi.org/10.3109/00365516009065404
  18. Reeves PG, Nielsen FH, Fahey GC Jr. AIN-93 purified diets for laboratory rodents: final report of the American Institute of Nutrition ad hoc writing committee on the reformulation of the AIN-76A rodent diet. J Nutr 1993;123:1939-51.
  19. Schifreen RS, Hickingbotham JM, Bowers GN Jr. Accuracy, precision, and stability in measurement of hemoglobin $A_{1c}$ by "high-performance" cation-exchange chromatography. Clin Chem 1980;26:466-72.
  20. Morgan CR, Lazarow A. Immunoassay of insulin: two antibody system. Plasma insulin levels in normal, subdiabetic, and diabetic rats. Diabetes 1963;12:115-26.
  21. Dahlqvist A. Assay of intestinal disaccharidases. Scand J Clin Lab Invest 1984;44:169-72. https://doi.org/10.3109/00365518409161400
  22. Lowry OH, Rosenbrough NJ, Farr AL, Randall RJ. Protein measurement with the Folin phenol reagent. J Biol Chem 1951;193:265-75.
  23. Madar Z. The effect of acarbose and miglitol (BAY-M-1099) on postprandial glucose levels following ingestion of various sources of starch by nondiabetic and streptozotocin-induced diabetic rats. J Nutr 1989;119:2023-29.
  24. Kang MJ, Kim JH, Choi HN, Kim MJ, Han JH, Lee JH, Kim JI. Hypoglycemic effects of Welsh onion in animal model of diabetes mellitus. Nutr Res Pract 2010;4:486-91. https://doi.org/10.4162/nrp.2010.4.6.486
  25. Ceriello A. Postprandial hyperglycemia and diabetes complications: is it time to treat? Diabetes 2005;54:1-7. https://doi.org/10.2337/diabetes.54.1.1
  26. Ishida H, Takizawa M, Ozawa S, Nakamichi Y, Yamaguchi S, Katsuta H, Tanaka T, Maruyama M, Katahira H, Yoshimoto K, Itagaki E, Nagamatsu S. Pioglitazone improves insulin secretory capacity and prevents the loss of ${\beta]-cell$ mass in obese diabetic db/db mice: possible protection of ${\beta]$ cells from oxidative stress. Metabolism 2004;53:488-94. https://doi.org/10.1016/j.metabol.2003.11.021
  27. Srinivasan K, Ramarao P. Animal models in type 2 diabetes research: an overview. Indian J Med Res 2007;125:451-72.
  28. Hummel KP, Coleman DL, Lane PW. The influence of genetic background on expression of mutations at the diabetes locus in the mouse. I. C57BL/KsJ and C57BL/6J strains. Biochem Genet 1972;7:1-13. https://doi.org/10.1007/BF00487005
  29. Dimitriadis GD, Tessari P, Go VLW, Gerich JE. ${\alpha]-Glucosidase$inhibition improves postprandial hyperglycemia and decreases insulin requirements in insulin-dependent diabetes mellitus. Metabolism 1985;34:261-5. https://doi.org/10.1016/0026-0495(85)90010-1
  30. Carrascosa JM, Molero JC, Fermin Y, Martinez C, Andres A, Satrustegui J. Effects of chronic treatment with acarbose on glucose and lipid metabolism in obese diabetic Wistar rats. Diabetes Obes Metab 2001;3:240-8. https://doi.org/10.1046/j.1463-1326.2001.00102.x
  31. Kannappan S, Anuradha CV. Insulin sensitizing actions of fenugreek seed polyphenols, quercetin & metformin in a rat model. Indian J Med Res 2009;129:401-8.
  32. Juretic D, Bernik S, Cop L, Hadzija M, Petlevski R, Lukac-Bajalo J. Short-term effect of acarbose on specific intestinal disaccharidase activities and hyperglycaemia in CBA diabetic mice. J Anim Physiol Anim Nutr (Berl) 2003;87:263-8. https://doi.org/10.1046/j.1439-0396.2003.00435.x
  33. Lee SM, Bustamante S, Flores C, Bezerra J, Goda T, Koldovský O. Chronic effects of an ${\alpha]-glucosidase$ inhibitor (Bay o 1248) on intestinal disaccharidase activity in normal and diabetic mice. J Pharmacol Exp Ther 1987;240:132-7.
  34. Liu L, Yu YL, Yang JS, Li Y, Liu YW, Liang Y, Liu XD, Xie L, Wang GJ. Berberine suppresses intestinal disaccharidases with beneficial metabolic effects in diabetic states, evidences from in vivo and in vitro study. Naunyn Schmiedebergs Arch Pharmacol 2010;381:371-81. https://doi.org/10.1007/s00210-010-0502-0
  35. Bressler R, Johnson D. New pharmacological approaches to therapy of NIDDM. Diabetes Care 1992;15:792-805. https://doi.org/10.2337/diacare.15.6.792
  36. Theoharides TC, Bielory L. Mast cells and mast cell mediators as targets of dietary supplements. Ann Allergy Asthma Immunol 2004;93:S24-34. https://doi.org/10.1016/S1081-1206(10)61484-6

Cited by

  1. Effects of the hydroalcoholic extract of Phyllanthus niruri and its isolated compounds on cyclophosphamide-induced hemorrhagic cystitis in mouse vol.384, pp.3, 2011, https://doi.org/10.1007/s00210-011-0668-0
  2. A. Berger extracts in streptozotocin-induced diabetic rats vol.5, pp.4, 2011, https://doi.org/10.4162/nrp.2011.5.4.301
  3. Polyphenolic Extract Inhibits Hyperglycemia, Hyperlipidemia, and Glycation-Oxidative Stress while Improving Insulin Resistance vol.59, pp.18, 2011, https://doi.org/10.1021/jf2022379
  4. Effect of quercetin on postprandial glucose excursion after mono- and disaccharides challenge in normal and diabetic rats vol.02, pp.01, 2012, https://doi.org/10.4236/jdm.2012.21013
  5. Geriatric Obesity: Evaluating the Evidence for the Use of Flavonoids to Promote Weight Loss vol.31, pp.3, 2012, https://doi.org/10.1080/21551197.2012.698222
  6. Quercetin ameliorates hyperglycemia and dyslipidemia and improves antioxidant status in type 2 diabetic db/db mice vol.6, pp.3, 2012, https://doi.org/10.4162/nrp.2012.6.3.201
  7. Acarbose, 17-α-estradiol, and nordihydroguaiaretic acid extend mouse lifespan preferentially in males vol.13, pp.2, 2013, https://doi.org/10.1111/acel.12170
  8. Quercetin induces insulin secretion by direct activation of L-type calcium channels in pancreatic beta cells vol.169, pp.5, 2013, https://doi.org/10.1111/bph.12194
  9. -Cell Mass and Function in Fructose-Induced Hyperinsulinemia through Modulating Pancreatic Akt/FoxO1 Activation vol.2013, pp.1741-4288, 2013, https://doi.org/10.1155/2013/303902
  10. Herbal Therapies for Type 2 Diabetes Mellitus: Chemistry, Biology, and Potential Application of Selected Plants and Compounds vol.2013, pp.1741-4288, 2013, https://doi.org/10.1155/2013/378657
  11. in Mice Fed a High-Fat Diet vol.2013, pp.1741-4288, 2013, https://doi.org/10.1155/2013/921012
  12. Hypoglycemic and Hypolipidemic Effects of Orostachys japonicus with Medicinal Herbs in Streptozotocin-Induced Diabetic Rats vol.42, pp.4, 2013, https://doi.org/10.3746/jkfn.2013.42.4.587
  13. Quercetin differently regulates insulin-mediated glucose transporter 4 translocation under basal and inflammatory conditions in adipocytes vol.58, pp.5, 2013, https://doi.org/10.1002/mnfr.201300510
  14. Improved Diabetic Nephropathy via Attenuating Renal Epithelial Mesenchymal Transition vol.61, pp.31, 2013, https://doi.org/10.1021/jf4020735
  15. Potential Impact of Quercetin and Idebenone against Immuno- inflammatory and Oxidative Renal Damage Induced in Rats by Titanium Dioxide Nanoparticles Toxicity vol.62, pp.11, 2013, https://doi.org/10.5650/jos.62.961
  16. Lotus leaf alleviates hyperglycemia and dyslipidemia in animal model of diabetes mellitus vol.7, pp.3, 2013, https://doi.org/10.4162/nrp.2013.7.3.166
  17. Quercetin protects against high glucose-induced damage in bone marrow-derived endothelial progenitor cells vol.34, pp.4, 2014, https://doi.org/10.3892/ijmm.2014.1852
  18. Fructooligosaccharide augments benefits of quercetin-3-O-β-glucoside on insulin sensitivity and plasma total cholesterol with promotion of flavonoid absorption in sucrose-fed rats vol.53, pp.2, 2014, https://doi.org/10.1007/s00394-013-0546-2
  19. Quercetin reduces obesity-associated ATM infiltration and inflammation in mice: a mechanism including AMPKα1/SIRT1 vol.55, pp.3, 2014, https://doi.org/10.1194/jlr.M038786
  20. Quercetin can reduce insulin resistance without decreasing adipose tissue and skeletal muscle fat accumulation vol.9, pp.1, 2014, https://doi.org/10.1007/s12263-013-0361-7
  21. Fatty acid synthase methylation levels in adipose tissue: effects of an obesogenic diet and phenol compounds vol.9, pp.4, 2014, https://doi.org/10.1007/s12263-014-0411-9
  22. Antidiabetic properties of dietary flavonoids: a cellular mechanism review vol.12, pp.1, 2015, https://doi.org/10.1186/s12986-015-0057-7
  23. Quercetin protects RAW264.7 macrophages from glucosamine-induced apoptosis and lipid accumulation via the endoplasmic reticulum stress pathway vol.12, pp.5, 2015, https://doi.org/10.3892/mmr.2015.4340
  24. Protection of pancreatic β-cell function by dietary polyphenols vol.14, pp.6, 2015, https://doi.org/10.1007/s11101-015-9429-x
  25. Quercetin ameliorates insulin sensitivity and liver steatosis partly by increasing adiponectin expression in ob/ob mice vol.24, pp.1, 2015, https://doi.org/10.1007/s10068-015-0036-9
  26. Antidepressant‐like effects of quercetin in diabetic rats are independent of hypothalamic–pituitary–adrenal axis vol.28, pp.01, 2016, https://doi.org/10.1017/neu.2015.45
  27. Investigation of the Acute Effects of Dry Extract of Glycine Max on Postprandial Glycemia in Rats vol.59, pp.0, 2016, https://doi.org/10.1590/1678-4324-2016150085
  28. Synergistic effect of quercetin and 6-gingerol treatment in streptozotocin induced type 2 diabetic rats and poloxamer P-407 induced hyperlipidemia vol.6, pp.15, 2016, https://doi.org/10.1039/C5RA16493A
  29. Involvement of miR-539-5p in the inhibition of de novo lipogenesis induced by resveratrol in white adipose tissue vol.7, pp.3, 2016, https://doi.org/10.1039/C5FO01090J
  30. Carbohydrate and Phytochemical Digestibility in Pasta vol.8, pp.1, 2016, https://doi.org/10.1007/s12393-015-9117-z
  31. Cardiovascular disease risk reduction in diabetes through conventional and natural approaches vol.6, pp.4, 2017, https://doi.org/10.1097/XCE.0000000000000134
  32. Unfolding Novel Mechanisms of Polyphenol Flavonoids for Better Glycaemic Control: Targeting Pancreatic Islet Amyloid Polypeptide (IAPP) vol.9, pp.7, 2017, https://doi.org/10.3390/nu9070788
  33. Incorporation of whole blue maize flour increases antioxidant capacity and reduces in vitro starch digestibility of gluten-free pasta pp.00389056, 2017, https://doi.org/10.1002/star.201700126
  34. In Vivo Effects of Quercetin in Association with Moderate Exercise Training in Improving Streptozotocin-Induced Aortic Tissue Injuries vol.20, pp.12, 2015, https://doi.org/10.3390/molecules201219802
  35. Molecular Mechanisms of the Anti-Obesity and Anti-Diabetic Properties of Flavonoids vol.17, pp.4, 2016, https://doi.org/10.3390/ijms17040569
  36. Benzbromarone, Quercetin, and Folic Acid Inhibit Amylin Aggregation vol.17, pp.6, 2016, https://doi.org/10.3390/ijms17060964
  37. Coadjuvants in the Diabetic Complications: Nutraceuticals and Drugs with Pleiotropic Effects vol.17, pp.8, 2016, https://doi.org/10.3390/ijms17081273
  38. Adenosine Monophosphate (AMP)-Activated Protein Kinase: A New Target for Nutraceutical Compounds vol.18, pp.2, 2017, https://doi.org/10.3390/ijms18020288
  39. L.) extract in streptozotocin-induced diabetic rats vol.196, pp.1755-1315, 2018, https://doi.org/10.1088/1755-1315/196/1/012038
  40. Health promoting properties of blueberries: a review pp.1549-7852, 2020, https://doi.org/10.1080/10408398.2018.1518895
  41. Oral formulation of DPP-4 inhibitor plus Quercetin improves metabolic homeostasis in type 1 diabetic rats vol.8, pp.1, 2018, https://doi.org/10.1038/s41598-018-33727-x
  42. Effects of Quercetin on Lipid and Protein Damage in the Liver of Streptozotocin-Induced Experimental Diabetic Rats pp.1557-7600, 2018, https://doi.org/10.1089/jmf.2018.0030
  43. residue extracts alleviates impaired glucose regulation in mice pp.01458884, 2018, https://doi.org/10.1111/jfbc.12651
  44. Influence of 4-week intraduodenal supplementation of quercetin on performance, glucose metabolism, and mRNA abundance of genes related to glucose metabolism and antioxidative status in dairy cows vol.96, pp.11, 2013, https://doi.org/10.3168/jds.2013-6852
  45. improves postprandial hyperglycemia in diabetic mice through α-glucosidase inhibition pp.0951418X, 2019, https://doi.org/10.1002/ptr.6260
  46. Renoprotective, anti-oxidative and anti-apoptotic effects of oral low-dose quercetin in the C57BL/6J model of diabetic nephropathy vol.13, pp.None, 2011, https://doi.org/10.1186/1476-511x-13-184
  47. Concomitant Intake of Quercetin with a Grain-Based Diet Acutely Lowers Postprandial Plasma Glucose and Lipid Concentrations in Pigs vol.2014, pp.None, 2014, https://doi.org/10.1155/2014/748742
  48. Role of red grape polyphenols as antidiabetic agents vol.3, pp.3, 2011, https://doi.org/10.1016/j.imr.2014.06.001
  49. Plant-Derived Compounds Targeting Pancreatic Beta Cells for the Treatment of Diabetes vol.2015, pp.None, 2011, https://doi.org/10.1155/2015/629863
  50. The protective effects of oral low-dose quercetin on diabetic nephropathy in hypercholesterolemic mice vol.6, pp.None, 2015, https://doi.org/10.3389/fphys.2015.00247
  51. Modulation of Glucose Transporter Protein by Dietary Flavonoids in Type 2 Diabetes Mellitus vol.11, pp.5, 2011, https://doi.org/10.7150/ijbs.11241
  52. The molecular basis of the antidiabetic action of quercetin in cultured skeletal muscle cells and hepatocytes vol.11, pp.41, 2011, https://doi.org/10.4103/0973-1296.149708
  53. Quercetin potentiates transdifferentiation of bone marrow mesenchymal stem cells into the beta cells in vitro vol.40, pp.5, 2011, https://doi.org/10.1007/s40618-016-0592-8
  54. Anti-diabetic activity of quercetin extracted from Phyllanthus emblica L. fruit: In silico and in vivo approaches vol.8, pp.2, 2011, https://doi.org/10.1016/j.jpha.2017.10.005
  55. Quercetin and Idebenone Ameliorate Oxidative Stress, Inflammation, DNA damage, and Apoptosis Induced by Titanium Dioxide Nanoparticles in Rat Liver vol.16, pp.4, 2011, https://doi.org/10.1177/1559325818812188
  56. Modulating impacts of quercetin/sitagliptin combination on streptozotocin-induced diabetes mellitus in rats vol.365, pp.None, 2011, https://doi.org/10.1016/j.taap.2018.12.011
  57. Vascular impact of quercetin administration in association with moderate exercise training in experimental type 1 diabetes vol.27, pp.3, 2019, https://doi.org/10.2478/rrlm-2019-0028
  58. Glucose-responsive oral insulin delivery for postprandial glycemic regulation vol.12, pp.7, 2011, https://doi.org/10.1007/s12274-018-2264-9
  59. Characterization of Maltase and Sucrase Inhibitory Constituents from Rhodiola crenulata vol.8, pp.11, 2019, https://doi.org/10.3390/foods8110540
  60. Bioactive Compounds of the PVPP Brewery Waste Stream and their Pharmacological Effects vol.17, pp.1, 2011, https://doi.org/10.2174/1570193x16666190723112623
  61. Antioxidant (gallic acid and quercetin) profile of Sumatran wild mangoes (Mangifera spp.): a potential source for antidegenerative medicine vol.9, pp.None, 2011, https://doi.org/10.12688/f1000research.22380.2
  62. Phytochemical screening and antioxidant profiling of Sumatran wild mangoes ( Mangifera spp.): a potential source for medicine antidegenerative effects vol.9, pp.None, 2011, https://doi.org/10.12688/f1000research.22380.3
  63. Fundamental Concepts and Novel Aspects of Polycystic Ovarian Syndrome: Expert Consensus Resolutions vol.11, pp.None, 2011, https://doi.org/10.3389/fendo.2020.00516
  64. Therapeutic Potential of Quercetin: New Insights and Perspectives for Human Health vol.5, pp.20, 2011, https://doi.org/10.1021/acsomega.0c01818
  65. Quercetin liposomes ameliorate streptozotocin-induced diabetic nephropathy in diabetic rats vol.10, pp.None, 2011, https://doi.org/10.1038/s41598-020-59411-7
  66. Mechanism of antidiabetic effects of Plicosepalus Acaciae flower in streptozotocin-induced type 2 diabetic rats, as complementary and alternative therapy vol.20, pp.1, 2011, https://doi.org/10.1186/s12906-020-03087-z
  67. Effects of Fermented Houttuynia cordata Thunb. on Diabetic Rats Induced by a High-Fat Diet with Streptozotocin and on Insulin Resistance in 3T3-L1 Adipocytes vol.2021, pp.None, 2011, https://doi.org/10.1155/2021/6936025
  68. Hypoglycemic Effects of Plant Flavonoids: A Review vol.2021, pp.None, 2011, https://doi.org/10.1155/2021/2057333
  69. A Review on Antidiabetic Activity of Centaurea spp.: A New Approach for Developing Herbal Remedies vol.2021, pp.None, 2021, https://doi.org/10.1155/2021/5587938
  70. Antihyperglycemic and Antilipidemic Properties of a Tea Infusion of the Leaves from Annona cherimola Miller on Streptozocin-Induced Type 2 Diabetic Mice vol.26, pp.9, 2011, https://doi.org/10.3390/molecules26092408
  71. Botanical Interventions to Improve Glucose Control and Options for Diabetes Therapy vol.3, pp.12, 2011, https://doi.org/10.1007/s42399-021-01034-8