Next Article in Journal
The Effect of IFT80 Deficiency in Osteocytes on Orthodontic Loading-Induced and Physiologic Bone Remodeling: In Vivo Study
Next Article in Special Issue
Cryopreservation of Pig Semen Using a Quercetin-Supplemented Freezing Extender
Previous Article in Journal
Feasibility of Novel Software-Based Perfusion Indicators for the Ileal J-Pouch—On the Path towards Objective and Quantifiable Intraoperative Perfusion Assessment with Indocyanine Green Near-Infrared Fluorescence
Previous Article in Special Issue
A Flavonoid on the Brain: Quercetin as a Potential Therapeutic Agent in Central Nervous System Disorders
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Therapeutic Potential of Quercetin in the Management of Type-2 Diabetes Mellitus

1
Department of Pharmacy, School of Pharmacy and Public Health, Independent University, Bangladesh (IUB), Dhaka 1229, Bangladesh
2
School of Biomedical Sciences, Ulster University, Coleraine BT52 1SA, UK
3
Department of Public Health, School of Pharmacy and Public Health, Independent University, Bangladesh (IUB), Dhaka 1229, Bangladesh
4
Natural Products Research Laboratory, Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow G4 0RE, UK
*
Author to whom correspondence should be addressed.
Life 2022, 12(8), 1146; https://doi.org/10.3390/life12081146
Submission received: 25 June 2022 / Revised: 24 July 2022 / Accepted: 27 July 2022 / Published: 28 July 2022
(This article belongs to the Special Issue Quercetin and Its Complexes for Medicinal Applications)

Abstract

:
Diabetes Mellitus (DM) is a metabolic disorder that is spreading alarmingly around the globe. Type-2 DM (T2DM) is characterized by low-grade inflammation and insulin resistance and is closely linked to obesity. T2DM is mainly controlled by lifestyle/dietary changes and oral antidiabetic drugs but requires insulin in severe cases. Many of the drugs that are currently used to treat DM are costly and present adverse side effects. Several cellular, animal, and clinical studies have provided compelling evidence that flavonoids have therapeutic potential in the management of diabetes and its complications. Quercetin is a flavonoid, present in various natural sources, which has demonstrated in vitro and in vivo antidiabetic properties. It improves oral glucose tolerance, as well as pancreatic β-cell function to secrete insulin. It inhibits the α-glucosidase and DPP-IV enzymes, which prolong the half-life of glucagon-like peptide-1 (GLP-1) and glucose-dependent insulinotropic polypeptide (GIP). Quercetin also suppresses the release of pro-inflammatory markers such as IL-1β, IL-4, IL-6, and TNF-α. Further studies are warranted to elucidate the mode(s) of action of quercetin at the molecular level. This review demonstrates the therapeutic potential of quercetin in the management of T2DM.

1. Introduction

Diabetes mellitus (DM) is a chronic disease that is one of the leading causes of illness and mortality across the globe. DM is diagnosed as a result of an elevated blood glucose level (hyperglycaemia) caused by inadequate insulin secretion, defective insulin action, or both. The improper control of insulin has also been linked to abnormalities in the metabolism of lipids and proteins. If proper treatment is not received on time, or if left untreated, DM can lead to hyperglycaemic coma, and severe damage to the eyes, kidneys, blood vessels, and nervous and cardiovascular system. It can even lead to death due to ketoacidosis and nonketotic hyperosmolar syndrome [1,2]. These metabolic disruptions result from low insulin levels or insulin resistance in skeletal muscles, adipose tissue, and other target tissues. The development, pathogenesis, and complications of DM have been strongly correlated with high levels of oxidative stress, free radicals, and other metabolic stressors [3,4]. According to reports from 2021, 465 million people suffer from DM worldwide [5]. This number is anticipated to rise to 700 million by 2045. The majority of DM sufferers are from middle and low-income countries [5].
The American Diabetes Association has categorized diabetes as Type 1, Type 2, and gestational DM [6]. Type 1 diabetes, also known as juvenile diabetes, causes a decrease in glucose sensitivity to clonal pancreatic β-cells [7]. It has no cure but can be controlled by lifestyle changes, blood sugar monitoring, and the administration of insulin. This type of diabetes occurs in approximately 80–90% of children and adolescents [8]. Type 2 diabetes mellitus (T2DM) is the most prevalent and occurs due to the insufficient production of insulin by the body, insulin resistance, and obesity [9]. It can be controlled by lifestyle/dietary changes and oral antidiabetic drugs but requires insulin in severe cases [10]. Whilst the majority of T2DM sufferers are adults (more than 90% of the patient population), it affects people of all ages. Individuals over 40 years of age and with obesity issues and a family history of the disease are at a higher risk of developing T2DM [11].
A range of antidiabetic drugs such as metformin, sulfonylureas, meglitinides, thiazolidinediones, GLP-1 mimetics, DPP-IV, and SGLT2 inhibitors are currently used to treat T2DM. However, many of these are costly and present notable adverse side effects (Table 1) [12]. Plant-based medicines have emerged as an alternative treatment for DM, particularly as these are affordable, widely accessible to rural populations, and have been associated with low side effects [13,14]. According to the World Health Organization, 75% of the world’s population uses herbal medicine for basic healthcare needs [14]. Traditional medicinal plants are often used for a wide variety of ailments, including DM [15]. Phytoconstituents from medicinal plants are well-known for their valuable therapeutic potential owing to their various biological effects including antidiabetic, anti-inflammatory, cardioprotective, antiviral, and antibacterial activities [16]. Flavonoids, including quercetin, are found in several medicinal plants including Momordica charantia, Dracocephalum moldavica, Euphorbia helioscopia, and Brassica rapa (Table 2). Medicinal plants containing quercetin have been used traditionally for the treatment of diabetes, infections, and cancer [16]. Recent studies have revealed that quercetin reduces the risk of cardiovascular diseases by lowering hyperglycaemia, high blood pressure, hyperlipidaemia, and promoting weight loss [17]. Some studies have demonstrated that this flavonoid is beneficial in chronic hypertension, dyslipidemia, obesity, and T2DM [18]. Quercetin has been proven to decrease blood glucose, liver glucose content, and enzyme levels, and lower serum cholesterol levels [18,19]. It has also been shown to prevent oxidative damage, enhancing the regeneration of pancreatic β-cell islets, and the subsequent release of insulin [19]. In comparison to current synthetic drugs, which have many adverse effects, quercetin has proven to be an exceptional template for the development of novel antidiabetic drugs. The purpose of this review is to explore the therapeutic potential of quercetin in the management of T2DM.

2. Chemistry of Quercetin

The term quercetin is derived from the Latin word “Quercetum” which means oak forest. The main dietary sources of quercetin are fruits, vegetables, and various medicinal plants (Table 2). Quercetin (3,3′,4′,5,7-pentahydroxyflavone) is a compound yellow in color, fully soluble in lipids and alcohol, insoluble in cold water, and sparingly soluble in hot water, that was isolated as a flavonoid glycoside for the first time in 1854. Its chemical structure was elucidated in 1899 [37]. Quercetin belongs to the flavonol subclass of flavonoids, with two aromatic rings (A and B) interlinked by a three-carbon linked γ-pyrone ring (C), and five hydroxyl (OH) groups that can be variously substituted (Figure 1).
The majority of quercetin derivatives are found in a glycoside form in which one or more hydroxyl group is substituted by different types of sugars [38]. Its polyphenolic structure, catechol moiety in the B ring, OH groups at positions 3 and 5 in the A ring, and 2,3-double bond conjugated with a 4-oxo function in the C ring have been identified as important features responsible for the well-known antioxidant effect of quercetin [39,40].

3. Pharmacological Actions of Quercetin in Diabetes and Associated Metabolic Disorders

Quercetin possesses various pharmacological properties and has been reported as one of the most widely used flavonoids to treat metabolic and inflammatory disorders [14]. In vitro studies on human retinal endothelial cells demonstrated that quercetin could inhibit the proliferation of high-glucose-induced cells by lowering the production of vascular endothelial growth factor (VEGF) (Figure 2) [41]. Quercetin also inhibited carbohydrate digesting-enzymes (intestinal α-glucosidase and pancreatic α-amylase), reduced starch hydrolysis, decreased the rate of glucose absorption, as well as slowed down the progression of postprandial hyperglycaemia in in vitro settings (Figure 2) [42,43].
Studies carried out on streptozotocin (STZ)-induced diabetic rats have revealed that quercetin could reduce blood glucose levels and improve glucose tolerance [44]. Quercetin decreased plasma glucose levels in Type 2 diabetic rats [45]. In hyperlipidaemic animals, quercetin lowered the levels of triglycerides (TG), total cholesterol (TC), LDL, and VLDL cholesterol, inhibited 3-hydroxy-3-methylglutaryl-CoA (HMG-CoA) reductase, and increased adiponectin and HDL cholesterol levels (Table 2) [45,46]. Previous findings also indicated that quercetin could improve the high-fat diet (HFD)-induced dyslipidaemia in Swiss albino mice [47]. Other studies have shown that quercetin inhibited the overexpression of connective tissue growth factor (CTGF) and transforming growth factor beta-1 (TGF-β1) and contributed to improving renal function in diabetic nephropathic rats (Figure 2) [48].
Quercetin has the potential to prevent diabetic liver oxidative damage by suppressing the CYP2E1 liver enzyme in diabetic mice (Figure 2) [49]. Additionally, it decreases oxidative stress in diabetic renal tissue (Table 2) [50]. The administration of quercetin decreased body weight, fat accumulation, hyperglycaemia, dyslipidemia, and hyperinsulinemia in high-fat-fed obese mice [50]. Quercetin reduced blood glucose levels in mice and rats with T2DM [45,51]. It also decreased oxidative damage in the pancreatic tissue of high-fat-fed mice [52]. When combined with resveratrol, quercetin significantly upregulated gene-associated glucose or lipid metabolism, as well as liver function, in HFD animal models [53]. Furthermore, quercetin with/without resveratrol reduced the damage to pancreatic β-cells by restoring serum C-peptide and glycosylated hemoglobin (HbA1c) levels in diabetic rats (Figure 2) [54]. Histological investigations demonstrated that quercetin, with/without resveratrol, preserved pancreatic tissue and regulated insulin levels, thereby exerting hypoglycemic activity, and enhancing the function of pancreatic β-cells in diabetic rats [55]. Quercetin reduced serum lipids levels and showed beneficial effects on dyslipidemia-associated complications including atherosclerosis, myocardial attack, and coronary diseases [50]. Quercetin significantly decreased plasma glucose levels in STZ-induced diabetic rats. In addition, it improved glucose tolerance and hepatic glucokinase activity [44]. Quercetin increased the number of pancreatic islets in both normal and diabetic mice. It also regenerated the pancreatic islets and enhanced insulin secretion in STZ/alloxan-induced diabetic mice [56].
In a randomized, double-blind, placebo-controlled clinical trial, quercetin at a dose of 100 mg/day for 12 weeks reduced body fat and body mass index (BMI) of obese subjects [57]. It also downregulated triacylglycerol levels at a dose of 150 mg/day in overweight individuals [57]. Quercetin also lowered maltose-induced postprandial hyperglycaemia but had no significant effect on glucose-induced postprandial hyperglycaemia [58]. The oral administration of multiple doses of quercetin decreased blood glucose and HbA1c levels, enhanced glycogen synthesis, decreased α-glucosidase activity, and insulin resistance. In addition, it minimized β-cell insufficiency, enhancing pancreatic insulin secretion and controlling blood glucose levels in diabetic patients by reducing oxidative stress [59].

4. Other Activities and Side Effects of Quercetin

Due to its polyphenolic structure and its catechol moiety, quercetin displays antioxidant/radical scavenging properties [60,61]. These effects help to protect against the oxidative stress-induced damage to pancreatic β-cells associated with diabetes [62]. In addition, quercetin has cardioprotective, anti-tumour, anti-arthritis, and antimicrobial properties [63,64,65,66] and it also prevents tyrosinase enzyme activity [67]. Quercetin has been reported to treat non-alcoholic fatty liver disease (NAFLD) by decreasing the level of liver enzymes, such as alanine transaminase (ALT) and aspartate transaminase (AST) (Figure 2), oxidative stress, and inflammation, and by regenerating altered metabolites and gut microbiota [44,68]. Quercetin has shown immunomodulatory activity, reducing the release of pro-inflammatory cytokines such as interleukin (IL)-1, IL-6, IL-8, IL-4, and tumour necrosis factor (TNF)-α (Figure 2) [69]. At doses higher than 945 mg/m2, quercetin can cause emesis, hypertension, nephrotoxicity, and decrease serum potassium levels [46]. Quercetin was also found to increase insulin secretion from BRIN-BD11 cells in a dose-dependent manner; however, it showed toxicity at doses above 50 μM [15].
Table 2. Pharmacological actions of quercetin-containing plants.
Table 2. Pharmacological actions of quercetin-containing plants.
Plant NamesPlant Part(s)Diabetic Model/sPharmacological Actions of PlantsDose of QuercetinDuration of TreatmentPharmacological Actions of QuercetinReferences
Acanthopanax senticosusRootAlloxan-induced diabetic rats↓ Blood glucose, total cholesterol, total bilirubin, creatinine, urea
↓ Oxidative stress
50 mg/kg30 daysInhibits α-glucosidase activity
Reduces oxidative stress
[66,70,71]
Ginkgo bilobaLeafSTZ-induced diabetic rats↑ β-cell mass and insulin secretion
↓ Amyloid-β neurotoxicity
90 mg/kg10 weeksDelays the progression of STZ-induced diabetic cataracts
Reduces AGE products activity
[72,73]
Psidium guajavaLeafNA-STZ-induced diabetic rats↓ Oxidative stress
↓ Protein glycation
↓ Inflammation
10- 50 mg/kg28 daysReduces blood glucose levels
Increases insulin secretion
Improves T2DM-mediated cardiovascular disease
[74,75]
Momordica charantiaFruitHFF obese rats↓ Blood glucose, total cholesterol
↑ Insulin secretion
50 mg/kg12 weeksReduces oxidation stress by inhibiting the release of chemokines and cytokines[76,77]
Polygonum perfoliatumLeafHFF obese rats↓ Blood glucose
↓ Inflammation
60–240 mg/kg4 weeksInhibits α-glucosidase activity[78]
Phyllanthus EmblicaFruitSTZ-induced diabetic rats↓ Triglycerides, LDL, VLDL, total cholesterol
↑ HDL cholesterol
25–75 mg/kg28 daysDecreases blood glucose
Increases insulin secretion
[79]
Cuscuta chinensisSeedAlloxan-induced diabetic mice↓ Fasting blood glucose
↑ Insulin secretion
Inhibits DPP-IV activity
20 mg/kg3 weeksReduces fasting blood glucose level
Enhances GLUT4 expression
[65,80]
Euphorbia helioscopiaLeaf, rootSTZ-induced diabetic rats↑ Insulin secretion
↓ Blood glucose
100 mg/kg7 weeksReduces blood glucose and blood glycated hemoglobin levels[81,82]
Brassica rapaRootSTZ-induced diabetic rats↓ Fasting blood glucose
↓ Inflammation
↓ Hypertension
Inhibits DPP-IV activity
15 mg/kg25 daysDecreases blood glucose levels
Improves glucose tolerance
[83,84]
Crataegus pinnatifidaLeaf, fruitSTZ-induced diabetic rats↓ Fasting blood glucose
↓ VLDL and LDL cholesterol
100 mg/kg14 daysDecreases blood glucose
Increases plasma insulin
[85,86]
Sophora japonicaBud,
flower
STZ-induced diabetic rats↑ Insulin release
Inhibits DPP-IV activity
10–15 mg/kg10 daysReduces blood glucose levels
Improves glucose tolerance
[50,87]
Coriandrum sativumHerbSTZ-induced diabetic rats↑ Insulin secretion
↓ Blood glucose
↓ Inflammation
50 mg/kg8 weeksDecreases fasting blood glucose
Suppresses TNF-α, IL-1β, and production of AGEs
[88,89,90]
Cymbopogon citratusHerbSTZ-induced diabetic rats↓ Fasting blood glucose
↓ Inflammation
↓ Hypertension
↑ Insulin secretion
20–50 mg/kg6 weeksReduces blood glucose levels
Decreases the production of reactive oxygen species (ROS)
Improves T2DM-mediated testicular damage
[91,92,93]
Allium cepaBulbSTZ-induced diabetic rats↓ Blood glucose
↓ Triglycerides, LDL, VLDL, total cholesterol
↑ HDL cholesterol
↑ Insulin secretion
100–200 mg/kg6 weeksLowers blood glucose
Improves glucose tolerance
[94,95,96]
Prunus aviumFruitSTZ-induced diabetic rats↓ Blood glucose
↑ Insulin secretion
↓ LDL and VLDL cholesterol
50–80 mg/kg45 daysReduces blood glucose levels
Improves oxidative stress
[97,98,99]
Capparis spinosaFruitAlloxan-induced diabetic mice↓ Fasting blood glucose
↑ Insulin secretion
↓ Liver damage
50 mg/kg7 daysDecreases fasting blood glucose
Reduces ALT and AST levels
[100,101,102]
Brassica oleracea var. ItalicaFlowerSTZ-induced diabetic rats↑ Insulin secretion
↓ Blood glucose
10 mg/kg4 weeksDecreases blood glucose levels
Reduces creatinine and blood urea nitrogen levels
[103,104,105]
Lactuca sativaLeafAlloxan-induced diabetic rats↓ Fasting blood glucose
↑ Insulin secretion
↓ Inflammation
50 mg/kg4 weeksReduces blood glucose levels
Decreases creatinine, ALT, AST, and cholesterol levels
[106,107,108]
Asparagus officinalisStemSTZ-induced diabetic rats↑ Insulin secretion
↓ Blood glucose
↓ Inflammation
50 mg/kg12 weeksReduces fasting blood glucose
Decreases the production of reactive oxygen species (ROS)
Improves glucose tolerance
[109,110]
Acacia arabicaBarkHFF-induced obese diabetic rats↑ Insulin secretion
Inhibits DPP-IV activity
↓ Protein glycation
30 mg/kg8 weeksReduces fasting blood glucose
Decreases LDL and TG levels
Increases HDL levels
[15,111]
Solanum lycopersicumFruitSTZ-induced diabetic rats↓ Blood glucose
↑ Insulin secretion
10 mg/kg28 daysDecreases blood glucose levels
Increases insulin secretion
Inhibits apoptosis
[112,113]
Piper nigrumFlowerAlloxan-induced diabetic mice↑ Insulin secretion
↓ Blood glucose
↓ Inflammation
50 mg/kg7 daysReduces blood glucose levels[114,115]
Toona sinensisHFF-induced obese diabetic rats↑ Insulin secretion
↓ Blood glucose
↓ Inflammation
200 mg/kg4 weeksImproves glucose tolerance
Decreases TG and TC levels
[116]
Symbols. ↑: Increase; ↓: Decrease.

5. Mechanisms of Action of Quercetin

Quercetin maintains glucose homeostasis by interacting with molecular targets in the small intestine, pancreas, skeletal muscle, adipose tissue, and liver. Studies carried out on STZ-induced diabetic rats have revealed that quercetin could restore the impaired protein expression of insulin signaling molecules, such as phosphatidylinositol 3 kinases (PI3K) and insulin receptor substrate-1 (IRS-1), resulting in increased insulin-mediated glucose uptake [117]. Quercetin has also been shown to activate adenosine monophosphate-activated protein kinase (AMPK) in the livers of rats, which reduces glucose synthesis primarily via downregulating glycogenic isoenzymes, such as phosphoenolpyruvate carboxylase (PEPCK) and glucose-6-phosphatase (G6Pase) [52,118]. In mouse skeletal muscle cells, it has been reported to enhance glucose uptake by promoting the translocation of GLUT4 to the cell membrane [119]. These findings indicate that quercetin regulates the metabolism of glucose, increasing glycolysis while decreasing gluconeogenesis [120]. In healthy individuals, around 80% of the absorbed glucose is stored in the form of glycogen in skeletal muscles upon the action of insulin. A reduction in this uptake has been shown to contribute to the etiology of T2DM as irregularities in the expression of the GLUT4 transporter lower the rate of glucose entering the cells, leading to a rise in blood glucose levels [121]. In skeletal muscles, quercetin activates AMPK, which in turn stimulates GLUT4 receptors and Akt (protein kinase B) in the cell membrane [122]. This allows glucose to enter the cells via the GLUT4 transporter, thereby regulating glycaemia [117]. Similarly, exercise is a potent activator of GLUT4 expression, which increases insulin activity and muscle glycogen storage. Defective activation of AMPK leads to insulin resistance, which causes T2DM [123]. Quercetin-induced AMPK activation in hepatocytes inhibits glucose-6 phosphatases [118]. Treatment with quercetin decreases GLUT2 expression and the intestinal sodium-dependent glucose uptake, in turn reducing glucose absorption in the gastrointestinal tract and controlling glycaemia (Figure 3) [119].
In addition, quercetin has been reported to enhance the AMP/ATP ratio and scavenge reactive oxygen species (ROS) in clonal pancreatic β-cells, reducing oxidative stress (Figure 3) [124]. The increased AMP/ATP ratio activates the mitochondrial target of rapamycin (mTOR), which induces mitogenesis (via transduction signaling pathways activation) and stimulates insulin secretion [125]. mTOR plays a significant role in the regulation of transcription, protein synthesis, and cell nutrition [126]. Under hyperglycaemic conditions, proteins, lipids, and nucleic acids undergo non-enzymatic glycation to form Advanced Glycation End (AGE)-products. The latter cause diabetic complications such as cardiovascular diseases, nephropathy, retinopathy, and neuropathy. Quercetin has been found to inhibit protein glycation more potently than the synthetic drug aminoguanidine [122,123]. It can reduce the formation of AGE-products by trapping methylglyoxal and glyoxal [127] and improve diabetic complications due to its antioxidant, anti-inflammatory, and antihyperglycemic properties [47].
In STZ-induced rats, quercetin improved retinopathy by down-regulating matrix metalloproteinase-9 (MMP-9), monocyte chemo-attractant protein-1 (MCP-1), and vascular endothelial growth factor (VEGF) [128]. In hypercholesterolemic mice, it reduced diabetic nephropathy by lowering triglycerides and blood glucose levels [129]. Moreover, it also showed neuroprotective effects on enteric neurons in the cecum of DM rats [130].
Previous studies have revealed that fat accumulation in the liver and muscles activates the Jun N-terminal kinases (JNK) and the nuclear transcription factor Kappa-B (NF-κB) inflammatory pathways, leading to obesity-associated T2DM (Figure 3) [131]. Quercetin suppresses both these pathways, which in turn improves glycaemia [132]. It also suppresses the FcεRI receptor by inhibiting the phosphorylation of several kinases like PKC (protein kinase C), Syk (spleen tyrosine kinase), and p38 mitogen-activated protein kinase (MAPK) in mast cells and basophils [122].
The release of pro-inflammatory mediators such as IL-1, IL-6, IL-8, IL-4, TNF-α, and histamine in brown adipose tissue has been linked with increased insulin resistance and high blood glucose levels (Figure 3) [133]. Quercetin inhibits these mediators and prevents oxidative stress [134]. In the kidneys, quercetin reduces DPP-IV and cyclooxygenase-2 (COX-2) activity, leading to a decrease in blood glucose reabsorption (Figure 3) [135]. Quercetin also activates leukocytes and targets various enzymes such as kinases, membrane proteins, and phosphatases to control inflammation and the immune response [129]. It suppresses lipoxygenase and cyclooxygenase enzymes, which suppresses the release of pro-inflammatory mediators including leukotrienes and prostaglandins [136]. Quercetin also inhibits TNF-α, a cytokine that plays a vital role in leukocyte formation, proliferation, and differentiation, specifically in the liver and gastrointestinal tract [69,125,135]. This causes a reduction in gluconeogenesis, glucose reabsorption, and α-glucosidase activity (Figure 3) [135]. Pancreatic β-cell apoptosis may occur due to hyperglycaemia-induced oxidative stress, and this can lead to diabetes mellitus. Glutathione peroxidase 4 (GPX4), an enzyme that protects cells against lipid peroxidation, suppresses the ferroptosis or apoptosis of pancreatic β-cells [137]. It has been demonstrated that quercetin can increase GPX4 activity in the pancreas, reducing oxidative stress, increased β-cell production, and insulin secretion [138]. Quercetin has also been reported to reduce the intestinal absorption of cholesterol by reducing the expression of the epithelial cholesterol transporter Niemann-Pick C1-Like 1 (NPC1L1) [100] and it has been suggested that the consumption of quercetin in the diet could lower systolic, diastolic, and mean arterial pressure in hypertensive individuals [139]. Quercetin can also lower blood pressure by reducing oxidative stress, enhancing the renin-angiotensin-aldosterone system (RAAS), and increasing vascular activity [133].

6. Effects of Quercetin on Diabetic Complications

Hyperglycemia over an extended period of time can increase the risk of macro- and microvascular problems including retinopathy, nephropathy, neuropathy, and cardiovascular diseases. A serious complication of DM, diabetic retinopathy (DR) is one of the leading causes of adult blindness and visual impairment [140]. Recent studies have indicated that quercetin at 150 mg/kg improved retinopathy in STZ-induced rats by reducing the expression of monocyte chemoattractant protein-1 (MCP-1), matrix metalloproteinase-9 (MMP-9), and vascular endothelial growth factor (VEGF), and decreased protein damage caused by oxidative stress [49,141]. Another major complication of DM, diabetic nephropathy is caused by long-term hyperglycemia which leads renal cells to secrete an array of pro-inflammatory and pro-fibrotic substances, resulting in cell hypertrophy and proliferation and the development of renal interstitial fibrosis [142]. Previous reports have shown that quercetin deactivated the SphK1-S1P (sphingosine kinase-1) signaling pathway, and hence inhibited the development of renal fibrosis [48].
Oxidative stress due to chronic hyperglycaemia induces complications related to the central nervous system and thus, may lead to neurodegenerative disorders such as Parkinson’s and Alzheimer’s diseases [143]. Recent studies have shown that the administration of quercetin improved memory impairment and reduced brain energy metabolism in STZ-induced rats by reducing ATP content in a dose-dependent manner [144]. Chronic insulin resistance and hyperglycaemia increase the risk of macrovascular complications such as hypertension, cardiomyopathy, and coronary artery diseases [145,146]. In STZ-induced rats, quercetin, with or without glibenclamide, was seen to decrease damage due to cardiomyopathy, in a dose-dependent manner [146]. Recent studies have revealed that quercetin increased cardioprotection via increasing endothelium cell receptors and nitric oxide production in STZ-induced rats [147]. Another study performed with Type 2 diabetic women reported that quercetin supplements could significantly reduce systolic blood pressure [148].
In recent years, various studies have reported the positive effects of quercetin on diabetes and its complications, and it is likely that these are alleviated following quercetin administration. However, few of these studies have been performed on humans. In recent years, only a few antidiabetic mechanisms of action have been reported for quercetin. More clinical trials including those using quercetin supplementation are required to better understand the mechanism(s) of action of quercetin in humans. It has also been discovered that defective iron metabolism in diabetic patients leads to increased severity of diabetic complications [149]. However, the impact of quercetin on iron regulation in diabetic complications has rarely been reported. Further investigation is required to understand the role of quercetin in iron regulation to develop new potential drugs for nephropathy, neuropathy, retinopathy, and other diabetic complications.

7. Conclusions

Studies have revealed that quercetin displays a wide range of pharmacological properties, including antihyperglycaemic effects. It can alleviate hyperglycaemia, hyperlipidemia, hypertension, and oxidative stress, contributing to lowering the risk of cardiovascular diseases emerging. Quercetin decreases blood glucose levels, improves glucose tolerance, and enhances pancreatic β-cell function via various mechanistic pathways such as AMPK which regulates GLUT4 expression in adipose tissue and muscles. It also regulates glycaemia by reducing GLUT2 expression and sodium-dependent glucose uptake in the gut, as well as lowering glucose absorption. It also inhibits the release of pro-inflammatory mediators, such as TNF-α, IL-1, -4, -6, and -8, preventing pancreatic β-cell damage. Quercetin has been shown to improve insulin sensitivity, glucose metabolism, and insulin secretion in diabetic animal models by promoting pancreatic β-cell proliferation. Due to its numerous benefits, quercetin has been identified to play a vital role in the treatment of T2DM. Various studies are currently underway to determine the potential of quercetin as a future antidiabetic medicine. However, only a few clinical trials have been performed to understand how quercetin works in humans. Therefore, the correct dose and duration of quercetin treatment are still unknown. It is necessary to address these limitations in current and future studies in order to confirm the true effects of quercetin on diabetic patients. The studies presented in this review support the conclusion that quercetin is a promising template for the development of new antidiabetic drugs. These would offer an alternative to current synthetic drugs that have undesirable side effects. However, further studies, ranging from animal models to clinical trials, are warranted to investigate the effects, including the mechanism(s) of action at the molecular level, of quercetin on lowering blood glucose levels and increasing insulin release in T2DM.

Author Contributions

Conceptualisation, P.A.; formal Analysis, P.A., V.S. and S.T.C.; funding acquisition, J.M.A.H. and P.A.; investigation, resources, writing, and editing, P.A., V.S., S.T.C., A.B.R., M.A.A., A.E.R., A.R. and U.H.J.; Visualization, P.A., V.S. and J.M.A.H.; supervision and reviewing, P.A., V.S. and Y.H.A.A.-W. All authors have read and agreed to the published version of the manuscript.

Funding

This study was not supported by any external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Acknowledgments

We would like to acknowledge Independent University, Bangladesh, Ulster University, UK and University of Strathclyde, UK for providing us access to their libraries, which enabled us to gather the research data required to write this paper.

Conflicts of Interest

The authors declare that there are no conflict of interest associated with this manuscript.

Abbreviations

DMDiabetes Mellitus
T2DMType 2 Diabetes Mellitus
DPP-IVDipeptidyl peptidase 4
GLP-1Glucagon-like peptide-1
GIPGlucose-dependent insulinotropic polypeptide
ILInterleukin
TNFTumor necrosis factor
SGLT2Sodium-glucose cotransporter-2
PPARPeroxisome proliferator-activated receptors
VEGFVascular endothelial growth factor
TGTriglycerides
HDLHigh-density lipoprotein
LDLLow-density lipoprotein
VLDLVery low-density lipoprotein
HMG-CoA3-hydroxy-3-methylglutaryl coenzyme A
HFDHigh fat fed
CTGFConnective tissue growth factor
TGF-β1Transforming growth factor-β1
CYP2E1Cytochrome P450 2E1
BMIBody mass index
NAFLDNon-alcoholic fatty liver disease
NANicotinamide
ALTAlanine transaminase
ASTAspartate transaminase
STZStreptozotocin
AGEAdvanced Glycation End Products
IRS-1Insulin receptor substrate 1
AMPKAMP-activated protein kinase
PEPCKPhosphoenolpyruvate carboxykinase
G6PaseGlucose 6 Phosphate
AktProtein kinase B
AMPAdenosine monophosphate
ATPAdenosine triphosphate
ROSReactive oxygen species
MMP-9Matrix metalloproteinase 9
MCP-1Monocyte chemoattractant protein-1
JNKc-Jun N-terminal kinase
NF-κBNuclear transcription factor kappa-B
PKCProtein kinase C
SykSpleen tyrosine kinase
MAPKp38 mitogen-activated protein kinase
COX-2Cyclooxygenase-2
GPX4Glutathione peroxidase 4
NPC1L1Niemann-Pick C1-Like 1
RAASRenin-angiotensin-aldosterone system

References

  1. Kharroubi, A.T.; Darwish, H.M. Diabetes Mellitus: The Epidemic of the Century. World J. Diabetes 2015, 6, 850. [Google Scholar] [CrossRef] [PubMed]
  2. American Diabetes Association. Diagnosis and Classification of Diabetes Mellitus. Diabetes Care 2013, 37, S81–S90. [Google Scholar] [CrossRef] [Green Version]
  3. Lamb, M.M.; Yin, X.; Zerbe, G.O.; Klingensmith, G.J.; Dabelea, D.; Fingerlin, T.E.; Rewers, M.; Norris, J.M. Height Growth Velocity, Islet Autoimmunity and Type 1 Diabetes Development: The Diabetes Autoimmunity Study in the Young. Diabetologia 2009, 52, 2064–2071. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  4. Robertson, R.P.; Harmon, J.; Tran, P.O.; Tanaka, Y.; Takahashi, H. Glucose Toxicity in β-Cells: Type 2 Diabetes, Good Radicals Gone Bad, and the Glutathione Connection. Diabetes 2003, 52, 581–587. [Google Scholar] [CrossRef] [Green Version]
  5. Akhtar, S.; Nasir, J.A.; Sarwar, A.; Nasr, N.; Javed, A.; Majeed, R.; Salam, M.A.; Billah, B. Prevalence of Diabetes and Pre-Diabetes in Bangladesh: A Systematic Review and Meta-Analysis. BMJ Open 2020, 10, e036086. [Google Scholar] [CrossRef]
  6. Craig, M.E.; Hattersley, A.; Donaghue, K.C. Definition, Epidemiology and Classification of Diabetes in Children and Adolescents. Pediatr. Diabetes 2009, 10, 3–12. [Google Scholar] [CrossRef]
  7. Katsarou, A.; Gudbjörnsdottir, S.; Rawshani, A.; Dabelea, D.; Bonifacio, E.; Anderson, B.J.; Jacobsen, L.M.; Schatz, D.A.; Lernmark, Å. Type 1 Diabetes Mellitus. Nat. Rev. Dis. Primers 2017, 3, 17016. [Google Scholar] [CrossRef]
  8. Kilic, G.; Alvarez-Mercado, A.I.; Zarrouki, B.; Opland, D.; Liew, C.W.; Alonso, L.C.; Myers, M.G.; Jonas, J.-C.; Poitout, V.; Kulkarni, R.N.; et al. The Islet Estrogen Receptor-α Is Induced by Hyperglycaemia and Protects against Oxidative Stress-Induced Insulin-Deficient Diabetes. PLoS ONE 2014, 9, e87941. [Google Scholar] [CrossRef] [Green Version]
  9. Devendra, D.; Liu, E.; Eisenbarth, G.S. Type 1 Diabetes: Recent Developments. BMJ 2004, 328, 750–754. [Google Scholar] [CrossRef] [Green Version]
  10. Dabelea, D.; Mayer-Davis, E.J.; Saydah, S.; Imperatore, G.; Linder, B.; Divers, J.; Bell, R.; Badaru, A.; Talton, J.W.; Crume, T.; et al. Prevalence of Type 1 and Type 2 Diabetes among Children and Adolescents from 2001 to 2009. JAMA 2014, 311, 1778–1786. [Google Scholar] [CrossRef]
  11. Chiang, J.L.; Kirkman, M.S.; Laffel, L.M.B.; Peters, A.L. Type 1 Diabetes through the Life Span: A Position Statement of the American Diabetes Association. Diabetes Care 2014, 37, 2034–2054. [Google Scholar] [CrossRef] [Green Version]
  12. Ansari, P.; Flatt, P.R.; Harriott, P.; Abdel-Wahab, Y.H.A. Insulin Secretory and Antidiabetic Actions of Heritiera Fomes Bark Together with Isolation of Active Phytomolecules. PLoS ONE 2022, 17, e0264632. [Google Scholar] [CrossRef]
  13. Ansari, P.; Flatt, P.R.; Harriott, P.; Abdel-Wahab, Y.H.A. Insulinotropic and antidiabetic properties of Eucalyptus citriodora leaves and isolation of bioactive phytomolecules. J. Pharm. Pharmacol. 2021, 73, 1049–1061. [Google Scholar] [CrossRef]
  14. Parasuraman, S.; Anand David, A.; Arulmoli, R. Overviews of Biological Importance of Quercetin: A Bioactive Flavonoid. Pharmacogn. Rev. 2016, 10, 84. [Google Scholar] [CrossRef] [Green Version]
  15. Ansari, P.; Flatt, P.R.; Harriott, P.; Hannan, J.M.A.; Abdel-Wahab, Y.H.A. Identification of Multiple Pancreatic and Extra-Pancreatic Pathways Underlying the Glucose-Lowering Actions of Acacia Arabica Bark in Type-2 Diabetes and Isolation of Active Phytoconstituents. Plants 2021, 10, 1190. [Google Scholar] [CrossRef]
  16. Diamant, M.; Heine, R.J. Thiazolidinediones in Type 2 Diabetes Mellitus. Drugs 2003, 63, 1373–1405. [Google Scholar] [CrossRef]
  17. Lin, T.-Y.; Liu, Y.-C.; Jheng, J.-R.; Tsai, H.-P.; Jan, J.-T.; Wong, W.-R.; Horng, J.-T. Anti-Enterovirus 71 Activity Screening of Chinese Herbs with Anti-Infection and Inflammation Activities. Am. J. Chin. Med. 2009, 37, 143–158. [Google Scholar] [CrossRef]
  18. Talirevic, E.; Sehovic, J. Quercetin in the Treatment of Dyslipidemia. Med. Res. Arch. 2012, 66, 87. [Google Scholar] [CrossRef] [Green Version]
  19. Ay, M.; Luo, J.; Langley, M.; Jin, H.; Anantharam, V.; Kanthasamy, A.; Kanthasamy, A.G. Molecular Mechanisms Underlying Protective Effects of Quercetin against Mitochondrial Dysfunction and Progressive Dopaminergic Neurodegeneration in Cell Culture and MitoPark Transgenic Mouse Models of Parkinson’s Disease. J. Neurochem. 2017, 141, 766–782. [Google Scholar] [CrossRef]
  20. Halban, P.A.; Polonsky, K.S.; Bowden, D.W.; Hawkins, M.A.; Ling, C.; Mather, K.J.; Powers, A.C.; Rhodes, C.J.; Sussel, L.; Weir, G.C. β-Cell Failure in Type 2 Diabetes: Postulated Mechanisms and Prospects for Prevention and Treatment. J. Clin. Endocrinol. Metab. 2014, 99, 1983–1992. [Google Scholar] [CrossRef]
  21. Usman, B.; Sharma, N.; Satija, S.; Mehta, M.; Vyas, M.; Khatik, G.L.; Khurana, N.; Hansbro, P.M.; Williams, K.; Dua, K. Recent Developments in Alpha-Glucosidase Inhibitors for Management of Type-2 Diabetes: An Update. Curr. Pharm. Des. 2019, 25, 2510–2525. [Google Scholar] [CrossRef]
  22. Kumar, S.; Kumar, V.; Prakash, O. Enzymes Inhibition and Antidiabetic Effect of Isolated Constituents from Dillenia Indica. Biomed Res. Int. 2013, 2013, e382063. [Google Scholar] [CrossRef]
  23. Wang, G.S.; Hoyte, C. Review of Biguanide (Metformin) Toxicity. J. Intensive Care Med. 2018, 34, 863–876. [Google Scholar] [CrossRef]
  24. Rena, G.; Hardie, D.G.; Pearson, E.R. The Mechanisms of Action of Metformin. Diabetologia 2017, 60, 1577–1585. [Google Scholar] [CrossRef] [Green Version]
  25. Lamos, E.M.; Levitt, D.L.; Munir, K.M. A Review of Dopamine Agonist Therapy in Type 2 Diabetes and Effects on Cardio-Metabolic Parameters. Prim. Care Diabetes 2016, 10, 60–65. [Google Scholar] [CrossRef]
  26. Andersen, I.B.; Andreassen, M.; Krogh, J. The Effect of Dopamine Agonists on Metabolic Variables in Adults with Type 2 Diabetes: A Systematic Review with Meta-Analysis and Trial Sequential Analysis of Randomized Clinical Trials. Diabetes Obes. Metab. 2020, 23, 58–67. [Google Scholar] [CrossRef]
  27. Pathak, R.; Bridgeman, M.B. Dipeptidyl Peptidase-4 (DPP-4) Inhibitors in the Management of Diabetes. Pharmacol. Ther. 2010, 35, 509–513. [Google Scholar]
  28. Capuano, A.; Sportiello, L.; Maiorino, M.I.; Rossi, F.; Giugliano, D.; Esposito, K. Dipeptidyl Peptidase-4 Inhibitors in Type 2 Diabetes therapy–Focus on Alogliptin. Drug Des. Devel. Ther. 2013, 7, 989–1001. [Google Scholar] [CrossRef] [Green Version]
  29. Gastaldelli, A.; Marchesini, G. Time for Glucagon like Peptide-1 Receptor Agonists Treatment for Patients with NAFLD? J. Hepatol. 2016, 64, 262–264. [Google Scholar] [CrossRef] [Green Version]
  30. Unger, J.R.; Parkin, C.G. Glucagon-like Peptide-1 (GLP-1) Receptor Agonists: Differentiating the New Medications. Diabetes Ther. 2011, 2, 29–39. [Google Scholar] [CrossRef] [Green Version]
  31. Black, C.; Donnelly, P.; McIntyre, L.; Royle, P.; Shepherd, J.J.; Thomas, S. Meglitinide Analogues for Type 2 Diabetes Mellitus. Cochrane Database Syst. Rev. 2007, 2010, CD004654. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  32. Ekanayake, P.; Hupfeld, C.; Mudaliar, S. Sodium-Glucose Cotransporter Type 2 (SGLT-2) Inhibitors and Ketogenesis: The Good and the Bad. Curr. Diab. Rep. 2020, 20, 74. [Google Scholar] [CrossRef] [PubMed]
  33. Scholtes, R.A.; Baar, M.J.B.; Lytvyn, Y.; Bjornstad, P.; Nieuwdorp, M.; Cherney, D.Z.I.; Raalte, D.H. Sodium Glucose Cotransporter (SGLT)-2 Inhibitors: Do We Need Them for Glucose-Lowering, for Cardiorenal Protection or Both? Diabetes Obes. Metab. 2019, 21, 24–33. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  34. Sola, D.; Rossi, L.; Schianca, G.P.C.; Maffioli, P.; Bigliocca, M.; Mella, R.; Corlianò, F.; Fra, G.P.; Bartoli, E.; Derosa, G. State of the Art Paper Sulfonylureas and Their Use in Clinical Practice. Arch. Med. Sci. 2015, 4, 840–848. [Google Scholar] [CrossRef] [PubMed]
  35. Kalra, S.; Aamir, A.H.; Raza, A.; Das, A.K.; Khan, A.A.; Shrestha, D.; Qureshi, M.F.; Fariduddin, M.; Pathan, M.F.; Jawad, F.; et al. Place of Sulfonylureas in the Management of Type 2 Diabetes Mellitus in South Asia: A Consensus Statement. Indian J. Endocrinol. Metab. 2015, 19, 577. [Google Scholar] [CrossRef]
  36. Rizos, C.V.; Kei, A.; Elisaf, M.S. The Current Role of Thiazolidinediones in Diabetes Management. Arch. Toxicol. 2016, 90, 1861–1881. [Google Scholar] [CrossRef]
  37. Shebeko, S.K.; Zupanets, I.A.; Popov, O.S.; Tarasenko, O.O.; Shalamay, A.S. Effects of Quercetin and Its Combinations on Health. In Polyphenols: Mechanisms of Action in Human Health and Disease; Academic Press: Cambridge, MA, USA, 2018; pp. 373–394. [Google Scholar] [CrossRef]
  38. Williams, C.A.; Grayer, R.J. Anthocyanins and Other Flavonoids. Nat. Prod. Rep. 2004, 21, 539. [Google Scholar] [CrossRef]
  39. Cao, G.; Sofic, E.; Prior, R.L. Antioxidant and Prooxidant Behavior of Flavonoids: Structure-Activity Relationships. Free Radic. Biol. Med. 1997, 22, 749–760. [Google Scholar] [CrossRef]
  40. Metodiewa, D.; Jaiswal, A.K.; Cenas, N.; Dickancaité, E.; Segura-Aguilar, J. Quercetin May Act as a Cytotoxic Prooxidant after Its Metabolic Activation to Semiquinone and Quinoidal Product. Free Radic. Biol. Med. 1999, 26, 107–116. [Google Scholar] [CrossRef]
  41. Oboh, G.; Ademosun, A.; Ayeni, P.O.; Omojokun, O.S.; Bello, F.O. Comparative Effect of Quercetin and Rutin on α-Amylase, α-Glucosidase, and Some Pro-Oxidant-Induced Lipid Peroxidation in Rat Pancreas. Comp. Clin. Path. 2014, 24, 1103–1110. [Google Scholar] [CrossRef]
  42. Spínola, V.; Llorent-Martínez, E.J.; Castilho, P.C. Inhibition of α-Amylase, α-Glucosidase and Pancreatic Lipase by Phenolic Compounds of Rumex Maderensis (Madeira Sorrel). Influence of Simulated Gastrointestinal Digestion on Hyperglycaemia-Related Damage Linked with Aldose Reductase Activity and Protein Glycation. LWT 2020, 118, 108727. [Google Scholar] [CrossRef]
  43. Gong, L.; Feng, D.; Wang, T.; Ren, Y.; Liu, Y.; Wang, J. Inhibitors of α-Amylase and α-Glucosidase: Potential Linkage for Whole Cereal Foods on Prevention of Hyperglycemia. Food Sci. Nutr. 2020, 8, 6320–6337. [Google Scholar] [CrossRef]
  44. Yang, D.K.; Kang, H.-S. Anti-Diabetic Effect of Cotreatment with Quercetin and Resveratrol in Streptozotocin-Induced Diabetic Rats. Biomol. Ther. 2018, 26, 130–138. [Google Scholar] [CrossRef] [Green Version]
  45. Chen, S.; Jiang, H.; Wu, X.; Fang, J. Therapeutic Effects of Quercetin on Inflammation, Obesity, and Type 2 Diabetes. Mediators Inflamm. 2016, 2016, 9340637. [Google Scholar] [CrossRef]
  46. Adewole, S.; Caxton-Martins, E.; Ojewole, J. Protective Effect of Quercetin on the Morphology of Pancreatic β-Cells of Streptozotocin-Treated Diabetic Rats. Afr. J. Tradit. Complement Altern. Med. 2007, 4, 64–74. [Google Scholar] [CrossRef] [Green Version]
  47. Dhanya, R.; Arya, A.D.; Nisha, P.; Jayamurthy, P. Quercetin, a Lead Compound against Type 2 Diabetes Ameliorates Glucose Uptake via AMPK Pathway in Skeletal Muscle Cell Line. Front. Pharmacol. 2017, 8, 336. [Google Scholar] [CrossRef]
  48. Chen, P.; Chen, J.; Zheng, Q.; Chen, W.; Wang, Y.; Xu, X. Pioglitazone, Extract of Compound Danshen Dripping Pill, and Quercetin Ameliorate Diabetic Nephropathy in Diabetic Rats. J. Endocrinol. Investig. 2013, 36, 422–427. [Google Scholar] [CrossRef]
  49. Thomas, A.A.; Feng, B.; Chakrabarti, S. ANRIL: A Regulator of VEGF in Diabetic Retinopathy. Investig. Ophthalmol. Vis. Sci. 2017, 58, 470. [Google Scholar] [CrossRef]
  50. Vessal, M.; Hemmati, M.; Vasei, M. Antidiabetic Effects of Quercetin in Streptozocin-Induced Diabetic Rats. Comp. Biochem. Physiol. Part C Toxicol. Pharmacol. 2003, 135, 357–364. [Google Scholar] [CrossRef]
  51. Shi, G.-J.; Li, Y.; Cao, Q.-H.; Wu, H.-X.; Tang, X.-Y.; Gao, X.-H.; Yu, J.-Q.; Chen, Z.; Yang, Y. In Vitro and in Vivo Evidence That Quercetin Protects against Diabetes and Its Complications: A Systematic Review of the Literature. Biomed. Pharmacother. 2019, 109, 1085–1099. [Google Scholar] [CrossRef]
  52. Zhou, M.; Wang, S.; Zhao, A.; Wang, K.; Fan, Z.; Yang, H.; Liao, W.; Bao, S.; Zhao, L.; Zhang, Y.; et al. Transcriptomic and Metabonomic Profiling Reveal Synergistic Effects of Quercetin and Resveratrol Supplementation in High Fat Diet Fed Mice. J. Proteome Res. 2012, 11, 4961–4971. [Google Scholar] [CrossRef]
  53. Saisho, Y.; Kou, K.; Tanaka, K.; Abe, T.; Kurosawa, H.; Shimada, A.; Meguro, S.; Kawai, T.; Itoh, H. Postprandial Serum C-Peptide to Plasma Glucose Ratio as a Predictor of Subsequent Insulin Treatment in Patients with Type 2 Diabetes. Endocr. J. 2011, 58, 315–322. [Google Scholar] [CrossRef] [Green Version]
  54. Kulkarni, C.R.; Joglekar, M.M.; Patil, S.B.; Arvindekar, A.U. Antihyperglycemic and Antihyperlipidemic Effect OfSantalum Albumin Streptozotocin Induced Diabetic Rats. Pharm. Biol. 2011, 50, 360–365. [Google Scholar] [CrossRef]
  55. Yim, S.; Malhotra, A.; Veves, A. Antioxidants and CVD in Diabetes: Where Do We Stand Now? Curr. Diab. Rep. 2007, 7, 8–13. [Google Scholar] [CrossRef] [Green Version]
  56. Spencer, J.P.E.; Vauzour, D.; Rendeiro, C. Flavonoids and Cognition: The Molecular Mechanisms Underlying Their Behavioural Effects. Arch. Biochem. Biophys. 2009, 492, 1–9. [Google Scholar] [CrossRef]
  57. Sikder, K.; Das, N.; Kesh, S.B.; Dey, S. Quercetin and Beta-Sitosterol Prevent High Fat Diet Induced Dyslipidemia and Hepatotoxicity in Swiss Albino Mice. Indian J. Exp. Biol. 2014, 52, 60–66. [Google Scholar]
  58. Mazloom, Z.; Abdollahzadeh, S.M.; Dabbaghmanesh, M.H.; Rezaianzadeh, A. The Effect of Quercetin Supplementation on Oxidative Stress, Glycemic Control, Lipid Profile, and Insulin Resistance in Type 2 Diabetes: A Randomized Clinical Trial. J. Health Sci. Surveill. Sys. 2014, 2, 8–14. [Google Scholar]
  59. Youl, E.; Bardy, G.; Magous, R.; Cros, G.; Sejalon, F.; Virsolvy, A.; Richard, S.; Quignard, J.; Gross, R.; Petit, P.; et al. Quercetin Potentiates Insulin Secretion and Protects INS-1 Pancreatic β-Cells against Oxidative Damage via the ERK1/2 Pathway. Br. J. Pharmacol. 2010, 161, 799–814. [Google Scholar] [CrossRef] [Green Version]
  60. Heřmánková, E.; Zatloukalová, M.; Biler, M.; Sokolová, R.; Bancířová, M.; Tzakos, A.G.; Křen, V.; Kuzma, M.; Trouillas, P.; Vacek, J. Redox Properties of Individual Quercetin Moieties. Free Radic. Biol. Med. 2019, 143, 240–251. [Google Scholar] [CrossRef]
  61. Sohn, E.-J.; Kim, J.M.; Kang, S.-H.; Kwon, J.; An, H.J.; Sung, J.-S.; Cho, K.A.; Jang, I.-S.; Choi, J.-S. Restoring Effects of Natural Antioxidant Quercetin on Cellular Senescent Human Dermal Fibroblasts. Am. J. Chin. Med. 2018, 46, 853–873. [Google Scholar] [CrossRef]
  62. Nekohashi, M.; Ogawa, M.; Ogihara, T.; Nakazawa, K.; Kato, H.; Misaka, T.; Abe, K.; Kobayashi, S. Luteolin and Quercetin Affect the Cholesterol Absorption Mediated by Epithelial Cholesterol Transporter Niemann–Pick C1-like 1 in Caco-2 Cells and Rats. PLoS ONE 2014, 9, e97901. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  63. Sandeep, M.S.; Nandini, C.D. Influence of Quercetin, Naringenin and Berberine on Glucose Transporters and Insulin Signalling Molecules in Brain of Streptozotocin-Induced Diabetic Rats. Biomed. Pharmacother. 2017, 94, 605–611. [Google Scholar] [CrossRef]
  64. Haddad, P.; Eid, H.; Nachar, A.; Thong, F.; Sweeney, G. The Molecular Basis of the Antidiabetic Action of Quercetin in Cultured Skeletal Muscle Cells and Hepatocytes. Pharmacogn. J. 2015, 11, 74. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  65. Alam, M.M.; Meerza, D.; Naseem, I. Protective Effect of Quercetin on Hyperglycaemia, Oxidative Stress and DNA Damage in Alloxan Induced Type 2 Diabetic Mice. Life Sci. 2014, 109, 8–14. [Google Scholar] [CrossRef] [PubMed]
  66. Fu, J.; Fu, J.; Yuan, J.; Zhang, N.; Gao, B.; Fu, G.; Tu, Y.; Zhang, Y. Anti-Diabetic Activities of Acanthopanax Senticosus Polysaccharide (ASP) in Combination with Metformin. Int. J. Biol. Macromol. 2012, 50, 619–623. [Google Scholar] [CrossRef] [PubMed]
  67. Oyama, T.; Takahashi, S.; Yoshimori, A.; Yamamoto, T.; Sato, A.; Kamiya, T.; Abe, H.; Abe, T.; Tanuma, S. Discovery of a New Type of Scaffold for the Creation of Novel Tyrosinase Inhibitors. Bioorg. Med. Chem. 2016, 24, 4509–4515. [Google Scholar] [CrossRef]
  68. Yi, H.; Peng, H.; Wu, X.; Xu, X.; Kuang, T.; Zhang, J.; Du, L.; Fan, G. The Therapeutic Effects and Mechanisms of Quercetin on Metabolic Diseases: Pharmacological Data and Clinical Evidence. Oxid. Med. Cell. Longev. 2021, 2021, e6678662. [Google Scholar] [CrossRef]
  69. D’Andrea, G. Quercetin: A Flavonol with Multifaceted Therapeutic Applications? Fitoterapia 2015, 106, 256–271. [Google Scholar] [CrossRef]
  70. Zhou, H.; Xing, J.; Liu, S.; Song, F.; Cai, Z.; Pi, Z.; Liu, Z.; Liu, S. Screening and Determination for Potential α-Glucosidase Inhibitors from Leaves of Acanthopanax Senticosus Harms by Using UF-LC/MS and ESI-MSn. Phytochem. Anal. 2011, 23, 315–323. [Google Scholar] [CrossRef]
  71. Nabi, R.K.; Abdullah, M.A. Effect of Quercetin on Parenchymatous Organ of the Alloxan Induced Diabetes in Male Rats. IJRMS 2020, 8, 3809. [Google Scholar] [CrossRef]
  72. Xiong, Y.; Zhu, G.-H.; Wang, H.-N.; Hu, Q.; Chen, L.-L.; Guan, X.-Q.; Li, H.-L.; Chen, H.-Z.; Tang, H.; Ge, G.-B. Discovery of Naturally Occurring Inhibitors against SARS-CoV-2 3Clpro from Ginkgo Biloba Leaves via Large-Scale Screening. Fitoterapia 2021, 152, 104909. [Google Scholar] [CrossRef]
  73. Lu, Q.; Hao, M.; Wu, W.; Zhang, N.; Isaac, A.T.; Yin, J.; Zhu, X.; Du, L.; Yin, X. Antidiabetic Cataract Effects of GbE, Rutin and Quercetin Are Mediated by the Inhibition of Oxidative Stress and Polyol Pathway. Acta Biochim. Pol. 2017, 65, 35–41. [Google Scholar] [CrossRef] [Green Version]
  74. Lin, C.-F.; Kuo, Y.-T.; Chen, T.-Y.; Chien, C.-T. Quercetin-Rich Guava (Psidium Guajava) Juice in Combination with Trehalose Reduces Autophagy, Apoptosis and Pyroptosis Formation in the Kidney and Pancreas of Type II Diabetic Rats. Molecules 2016, 21, 334. [Google Scholar] [CrossRef] [Green Version]
  75. Roslan, J.; Giribabu, N.; Karim, K.; Salleh, N. Quercetin Ameliorates Oxidative Stress, Inflammation and Apoptosis in the Heart of Streptozotocin-Nicotinamide-Induced Adult Male Diabetic Rats. Biomed. Pharmacother. 2017, 86, 570–582. [Google Scholar] [CrossRef]
  76. Ansari, P.; Hannon-Fletcher, M.P.; Flatt, P.R.; Abdel-Wahab, Y.H.A. Effects of 22 Traditional Anti-Diabetic Medicinal Plants on DPP-IV Enzyme Activity and Glucose Homeostasis in High-Fat Fed Obese Diabetic Rats. Biosci. Rep. 2021, 41, BSR20203824. [Google Scholar] [CrossRef]
  77. Rasheed, R.A.; Elshikh, M.S.; Mohamed, M.O.; Darweesh, M.F.; Hussein, D.S.; Almutairi, S.M.; Embaby, A.S. Quercetin Mitigates the Adverse Effects of High Fat Diet on Pancreatic and Renal Tissues in Adult Male Albino Rats. J. King Saud Univ. Sci. 2022, 34, 101946. [Google Scholar] [CrossRef]
  78. Wang, J.; Wang, Y.; Xu, C.; Wang, B.; Yu, J.; Kang, X.; Liu, X.; Zhou, L.; Qin, Y.; Liao, L.; et al. Effects of total flavonoids extracted from Polygonum perfoliatum L. on hypolipidemic and antioxidant in hyperlipidemia rats induced by high-fat diet. Int. J. Clin. Exp. Med. 2018, 11, 6758–6766. [Google Scholar]
  79. Srinivasan, P.; Vijayakumar, S.; Kothandaraman, S.; Palani, M. Anti-Diabetic Activity of Quercetin Extracted from Phyllanthus emblica, L. Fruit: In Silico and in Vivo Approaches. J. Pharm. Anal. 2018, 8, 109–118. [Google Scholar] [CrossRef]
  80. Al-Sultany, F.H.; Al-Hussaini, I.M.; Al- Saadi, A.H. Studying Hypoglycemic Activity of Cuscuta Chinesis Lam. On Type 1 Diabetes Mellitus in White Male Rats. J. Phys. Conf. Ser. 2019, 1294, 062020. [Google Scholar] [CrossRef]
  81. Mustafa, I.; Anwar, H.; Irfan, S.; Muzaffar, H.; Ijaz, M.U. Attenuation of Carbohydrate Metabolism and Lipid Profile by Methanolic Extract of Euphorbia Helioscopia and Improvement of Beta Cell Function in a Type 2 Diabetic Rat Model. BMC Complement Altern. Med. 2022, 22, 23. [Google Scholar] [CrossRef]
  82. Kim, J.-H.; Kang, M.-J.; Choi, H.-N.; Jeong, S.-M.; Lee, Y.-M.; Kim, J.-I. Quercetin Attenuates Fasting and Postprandial Hyperglycemia in Animal Models of Diabetes Mellitus. Nutr. Res. Prac. 2011, 5, 107–111. [Google Scholar] [CrossRef] [Green Version]
  83. Hassanpour Fard, M.; Naseh, G.; Lotfi, N.; Hosseini, S.M.; Hosseini, M. Effects of Aqueous Extract of Turnip Leaf (Brassica Rapa) in Alloxan-Induced Diabetic Rats. Avicenna J. Med. 2015, 5, 148–156. [Google Scholar]
  84. Abdelmoaty, M.A.; Ibrahim, M.A.; Ahmed, N.S.; Abdelaziz, M.A. Confirmatory Studies on the Antioxidant and Antidiabetic Effect of Quercetin in Rats. Indian J. Clin. Biochem. 2010, 25, 188–192. [Google Scholar] [CrossRef] [Green Version]
  85. Dehghani, S.; Mehri, S.; Hosseinzadeh, H. The Effects of Crataegus Pinnatifida (Chinese Hawthorn) on Metabolic Syndrome: A Review. Iran. J. Basic Med. Sci. 2019, 22, 460. [Google Scholar] [CrossRef]
  86. Iskender, H.; Dokumacioglu, E.; Sen, T.M.; Ince, I.; Kanbay, Y.; Saral, S. The Effect of Hesperidin and Quercetin on Oxidative Stress, NF-ΚB and SIRT1 Levels in a STZ-Induced Experimental Diabetes Model. Biomed. Pharmacother. 2017, 90, 500–508. [Google Scholar] [CrossRef]
  87. Wang, T.; Miao, M.; Bai, M.; Li, Y.; Li, M.; Li, C.; Xu, Y. Effect of Sophora Japonica Total Flavonoids on Pancreas, Kidney Tissue Morphology of Streptozotocin-Induced Diabetic Mice Model. Saudi J. Biol. Sci. 2017, 24, 741–747. [Google Scholar] [CrossRef]
  88. Eidi, M.; Eidi, A.; Saeidi, A.; Molanaei, S.; Sadeghipour, A.; Bahar, M.; Bahar, K. Effect of Coriander Seed (Coriandrum Sativum L.) Ethanol Extract on Insulin Release from Pancreatic Beta Cells in Streptozotocin-Induced Diabetic Rats. Phytother. Res. 2009, 23, 404–406. [Google Scholar] [CrossRef]
  89. Tang, L.; Li, K.; Zhang, Y.; Li, H.; Li, A.; Xu, Y.; Wei, B. Quercetin Liposomes Ameliorate Streptozotocin-Induced Diabetic Nephropathy in Diabetic Rats. Sci. Rep. 2020, 10, 2440. [Google Scholar] [CrossRef] [Green Version]
  90. Das, S.; Chaware, S.; Narkar, N.; Tilak, A.V.; Raveendran, S.; Rane, P. Antidiabetic Activity of Coriandrum Sativum in Streptozotocin Induced Diabetic Rats. Int. J. Basic Clin. Pharmacol. 2019, 8, 925–929. [Google Scholar] [CrossRef]
  91. Elekofehinti, O.O.; Onunkun, A.T.; Olaleye, T.M. Cymbopogon Citratus (DC.) Stapf Mitigates ER-Stress Induced by Streptozotocin in Rats via Down-Regulation of GRP78 and Up-Regulation of Nrf2 Signaling. J. Ethnopharmacol. 2020, 262, 113130. [Google Scholar] [CrossRef]
  92. Ahmed, N.Z.; Ibrahim, S.R.; Ahmed-Farid, O.A. Quercetin and Apigenin of Cymbopogon Citratus Mediate Inhibition of HCT-116 and PC-3 Cell Cycle Progression and Ameliorate Doxorubicin-Induced Testicular Dysfunction in Male Rats. Biomed. Res. Ther. 2018, 5, 2466–2479. [Google Scholar] [CrossRef] [Green Version]
  93. Wang, D.; Li, Y.; Zhai, Q.; Zhu, Y.; Liu, B.; Xu, Y. Quercetin Ameliorates Testosterone Secretion Disorder by Inhibiting Endoplasmic Reticulum Stress through the MiR-1306-5p/HSD17B7 Axis in Diabetic Rats. Bosn. J. Basic Med. Sci. 2022, 22, 191–204. [Google Scholar] [CrossRef] [PubMed]
  94. Ozougwu, C.J. Anti-Diabetic Effects of Allium Cepa (Onions) Aqueous Extracts on Alloxan-Induced Diabetic Rattus Novergicus. J. Med. Plant Res. 2011, 5, 1134–1139. [Google Scholar] [CrossRef]
  95. Khaki, A.; Fathiazad, F.; Ashtiani, H.R.; Rezazadeh, S.; Rastegar, H.; Imani, A.M. Compartments of Quercetin & Allium Cepa (Onion) on Blood Glucose in Diabetic Rats. J. Med. Plants 2010, 9, 107–112. [Google Scholar]
  96. Campos, K.E.; Diniz, Y.S.; Cataneo, A.C.; Faine, L.A.; Alves, M.J.Q.F.; Novelli, E.L.B. Hypoglycaemic and Antioxidant Effects of Onion, Allium Cepa: Dietary Onion Addition, Antioxidant Activity and Hypoglycaemic Effects on Diabetic Rats. Int. J. Food Sci. Nutr. 2003, 54, 241–246. [Google Scholar] [CrossRef] [PubMed]
  97. Vinitha, E.; Singh, H.J.C.; Kakalij, R.M.; Kshirsagar, R.P.; Kumar, B.H.; Diwan, P.V. Neuroprotective Effect of Prunus Avium on Streptozotocin Induced Neurotoxicity in Mice. Biomed. Prev. Nutr. 2014, 4, 519–525. [Google Scholar] [CrossRef]
  98. Faienza, M.F.; Corbo, F.; Carocci, A.; Catalano, A.; Clodoveo, M.L.; Grano, M.; Wang, D.Q.; D’Amato, G.; Muraglia, M.; Franchini, C.; et al. Novel Insights in Health-Promoting Properties of Sweet Cherries. J. Funct. Foods 2020, 69, 103945. [Google Scholar] [CrossRef]
  99. Mahesh, T.; Menon, V.P. Quercetin Allievates Oxidative Stress in Streptozotocin-Induced Diabetic Rats. Phytother. Res. 2004, 18, 123–127. [Google Scholar] [CrossRef]
  100. Huseini, H.F.; Hasani-Rnjbar, S.; Nayebi, N.; Heshmat, R.; Sigaroodi, F.K.; Ahvazi, M.; Alaei, B.A.; Kianbakht, S. Capparis spinosa L. (Caper) Fruit Extract in Treatment of Type 2 Diabetic Patients: A Randomized Double-Blind Placebo-Controlled Clinical Trial. Complement. Ther. Med. 2013, 21, 447–452. [Google Scholar] [CrossRef]
  101. Sirovina, D.; Oršolić, N.; Končić, M.Z.; Kovačević, G.; Benković, V.; Gregorović, G. Quercetin vs Chrysin. Hum. Exp. Toxicol. 2013, 32, 1058–1066. [Google Scholar] [CrossRef]
  102. Kalantari, H.; Foruozandeh, H.; Khodayar, M.J.; Siahpoosh, A.; Saki, N.; Kheradmand, P. Antioxidant and Hepatoprotective Effects of Capparis spinosa L. Fractions and Quercetin on Tert-Butyl Hydroperoxide- Induced Acute Liver Damage in Mice. J. Tradit. Complement. Med. 2018, 8, 120–127. [Google Scholar] [CrossRef]
  103. Gupta, S.; Burman, S.; Nair, A.B.; Chauhan, S.; Sircar, D.; Roy, P.; Dhanwat, M.; Lahiri, D.; Mehta, D.; Das, R.; et al. Brassica Oleracea Extracts Prevent Hyperglycaemia in Type 2 Diabetes Mellitus. Prev. Nutr. Food Sci. 2022, 27, 50–62. [Google Scholar] [CrossRef]
  104. Shah, M.A.; Sarker, M.M.R.; Gousuddin, M. Antidiabetic Potential of Brassica Oleracea Var. Italica in Type 2 Diabetic Sprague Dawley (sd) Rats. IJPPR 2016, 8, 462–469. [Google Scholar]
  105. Anjaneyulu, M.; Chopra, K. Quercetin, an anti-oxidant bioflavonoid, attenuates diabetic nephropathy in rats. Clin. Exp. Pharmacol. 2004, 31, 244–248. [Google Scholar] [CrossRef]
  106. Chadchan, K.S.; Jargar, J.G.; Das, S.N. Anti-Diabetic Effects of Aqueous Prickly Lettuce (Lactuca Scariola Linn.) Leaves Extract in Alloxan-Induced Male Diabetic Rats Treated with Nickel (II). J. Basic Clin. Physiol. Pharmacol. 2016, 27, 49–56. [Google Scholar] [CrossRef]
  107. Ismail, H.; Gillespie, A.L.; Calderwood, D.; Iqbal, H.; Gallagher, C.; Chevallier, O.P.; Elliott, C.T.; Pan, X.; Mirza, B.; Green, B.D. The Health Promoting Bioactivities of Lactuca Sativa Can Be Enhanced by Genetic Modulation of Plant Secondary Metabolites. Metabolites 2019, 9, 97. [Google Scholar] [CrossRef] [Green Version]
  108. Nabi, R.K.; Abdullah, M.A. Effect of Quercetin on the Biochemical Parameters of the Alloxan Induced Diabetes in Male Rats. Bas. J. Vet. Res. 2019, 18, 158–170. [Google Scholar]
  109. Hafizur, R.M.; Kabir, N.; Chishti, S. Asparagus Officinalis Extract Controls Blood Glucose by Improving Insulin Secretion and β-Cell Function in Streptozotocin-Induced Type 2 Diabetic Rats. Brit. J. Nutr. 2012, 108, 1586–1595. [Google Scholar] [CrossRef] [Green Version]
  110. Xie, J.; Song, W.; Liang, X.; Zhang, Q.; Shi, Y.; Liu, W.; Shi, X. Protective Effect of Quercetin on Streptozotocin-Induced Diabetic Peripheral Neuropathy Rats through Modulating Gut Microbiota and Reactive Oxygen Species Level. Biomed. Pharmacother. 2020, 127, 110147. [Google Scholar] [CrossRef]
  111. Zhang, F.; Feng, J.; Zhang, J.; Kang, X.; Qian, D. Quercetin Modulates AMPK/SIRT1/NF-ΚB Signaling to Inhibit Inflammatory/Oxidative Stress Responses in Diabetic High Fat Diet-Induced Atherosclerosis in the Rat Carotid Artery. Exp. Ther. Med. 2020, 20, 280. [Google Scholar] [CrossRef]
  112. Kermani, J.; Goodarzi, N.; Bakhtiari, M. An Experimental Study to Evaluate the Protective Effects of Solanum Lycopersicum Seed Essential Oil on Diabetes-Induced Testicular Injuries. Medicina 2019, 55, 499. [Google Scholar] [CrossRef] [Green Version]
  113. Ojo, O.O.; Olorunsogo, O.O. Quercetin and Vitamin E Attenuate Diabetes-Induced Testicular Anomaly in Wistar Rats via the Mitochondrial-Mediated Apoptotic Pathway. Andrologia 2021, 53, e14185. [Google Scholar] [CrossRef]
  114. Atal, S.; Atal, S.; Vyas, S.; Phadnis, P. Bio-Enhancing Effect of Piperine with Metformin on Lowering Blood Glucose Level in Alloxan Induced Diabetic Mice. Pharmacogn. Res. 2016, 8, 56. [Google Scholar] [CrossRef] [Green Version]
  115. Oršolić, N.; Gajski, G.; Garaj-Vrhovac, V.; Đikić, D.; Prskalo, Z.Š.; Sirovina, D. DNA-Protective Effects of Quercetin or Naringenin in Alloxan-Induced Diabetic Mice. Eur. J. Pharmacol. 2011, 656, 110–118. [Google Scholar] [CrossRef]
  116. Zhang, Y.; Dong, H.; Wang, M.; Zhang, J. Quercetin Isolated from Toona Sinensis Leaves Attenuates Hyperglycemia and Protects Hepatocytes in High-Carbohydrate/High-Fat Diet and Alloxan Induced Experimental Diabetic Mice. J. Diabetes Res. 2016, 2016, 8492780. [Google Scholar] [CrossRef] [Green Version]
  117. Fontana Pereira, D.; Cazarolli, L.H.; Lavado, C.; Mengatto, V.; Figueiredo, M.S.R.B.; Guedes, A.; Pizzolatti, M.G.; Silva, F.R.M.B. Effects of Flavonoids on α-Glucosidase Activity: Potential Targets for Glucose Homeostasis. Nutrition 2011, 27, 1161–1167. [Google Scholar] [CrossRef] [Green Version]
  118. Vafadar, A.; Shabaninejad, Z.; Movahedpour, A.; Fallahi, F.; Taghavipour, M.; Ghasemi, Y.; Akbari, M.; Shafiee, A.; Hajighadimi, S.; Moradizarmehri, S.; et al. Quercetin and Cancer: New Insights into Its Therapeutic Effects on Ovarian Cancer Cells. Cell Biosci. 2020, 10, 32. [Google Scholar] [CrossRef] [Green Version]
  119. Borghi, S.M.; Mizokami, S.S.; Pinho-Ribeiro, F.A.; Fattori, V.; Crespigio, J.; Clemente-Napimoga, J.T.; Napimoga, M.H.; Pitol, D.L.; Issa, J.P.M.; Fukada, S.Y.; et al. The Flavonoid Quercetin Inhibits Titanium Dioxide (TiO2)-Induced Chronic Arthritis in Mice. J. Nutr. Biochem. 2018, 53, 81–95. [Google Scholar] [CrossRef]
  120. Wang, S.; Yao, J.; Zhou, B.; Yang, J.; Chaudry, M.T.; Wang, M.; Xiao, F.; Li, Y.; Yin, W. Bacteriostatic Effect of Quercetin as an Antibiotic Alternative in Vivo and Its Antibacterial Mechanism in Vitro. J. Food Prot. 2018, 81, 68–78. [Google Scholar] [CrossRef]
  121. Alam, F.; Islam, A.; Khalil, I.; Hua Gan, S. Metabolic Control of Type 2 Diabetes by Targeting the GLUT4 Glucose Transporter: Intervention Approaches. Curr. Pharm. Des. 2016, 22, 3034–3049. [Google Scholar] [CrossRef]
  122. Dhanya, R.; Arun, K.B.; Syama, H.P.; Nisha, P.; Sundaresan, A.; Santhosh Kumar, T.R.; Jayamurthy, P. Rutin and Quercetin Enhance Glucose Uptake in L6 Myotubes under Oxidative Stress Induced by Tertiary Butyl Hydrogen Peroxide. Food Chem. 2014, 158, 546–554. [Google Scholar] [CrossRef] [PubMed]
  123. Richter, E.A.; Hargreaves, M. Exercise, GLUT4, and Skeletal Muscle Glucose Uptake. Physiol. Rev. 2013, 93, 993–1017. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  124. Ashraf, J.M.; Shahab, U.; Tabrez, S.; Lee, E.J.; Choi, I.; Ahmad, S. Quercetin as a Finer Substitute to Aminoguanidine in the Inhibition of Glycation Products. Int. J. Biol. Macromol. 2015, 77, 188–192. [Google Scholar] [CrossRef] [PubMed]
  125. Dhanya, R.; Kartha, C.C. Quercetin Improves Oxidative Stress-Induced Pancreatic Beta Cell Alterations via MTOR-Signaling. Mol. Cell. Biochem. 2021, 476, 3879–3887. [Google Scholar] [CrossRef]
  126. Saxton, R.A.; Sabatini, D.M. MTOR Signaling in Growth, Metabolism, and Disease. Cell 2017, 169, 361–371. [Google Scholar] [CrossRef]
  127. Li, X.; Zheng, T.; Sang, S.; Lv, L. Quercetin Inhibits Advanced Glycation End Product Formation by Trapping Methylglyoxal and Glyoxal. J. Agric. Food Chem. 2014, 62, 12152–12158. [Google Scholar] [CrossRef]
  128. Edwards, R.L.; Lyon, T.; Litwin, S.E.; Rabovsky, A.; Symons, J.D.; Jalili, T. Quercetin Reduces Blood Pressure in Hypertensive Subjects. J. Nutr. 2007, 137, 2405–2411. [Google Scholar] [CrossRef]
  129. Kalupahana, N.S.; Claycombe, K.J.; Moustaid-Moussa, N. (N-3) Fatty Acids Alleviate Adipose Tissue Inflammation and Insulin Resistance: Mechanistic Insights. Adv. Nutr. 2011, 2, 304–316. [Google Scholar] [CrossRef] [Green Version]
  130. Ferreira, P.E.B. Diabetic Neuropathy: An Evaluation of the Use of Quercetin in the Cecum of Rats. World J. Gastroenterol. 2013, 19, 6416. [Google Scholar] [CrossRef]
  131. Shoelson, S.E. Inflammation and Insulin Resistance. J. Clin. Investig. 2006, 116, 1793–1801. [Google Scholar] [CrossRef]
  132. Tsalamandris, S.; Antonopoulos, A.S.; Oikonomou, E.; Papamikroulis, G.-A.; Vogiatzi, G.; Papaioannou, S.; Deftereos, S.; Tousoulis, D. The Role of Inflammation in Diabetes: Current Concepts and Future Perspectives. Eur. Cardiol. Rev. 2019, 14, 50. [Google Scholar] [CrossRef] [Green Version]
  133. Tziomalos, K.; Athyros, V.G. Diabetic Nephropathy: New Risk Factors and Improvements in Diagnosis. Rev. Diabet. Stud. 2015, 12, 110–118. [Google Scholar] [CrossRef] [Green Version]
  134. Cermak, R.; Landgraf, S.; Wolffram, S. Quercetin Glucosides Inhibit Glucose Uptake into Brush-Border-Membrane Vesicles of Porcine Jejunum. Br. J. Nutr. 2004, 91, 849–855. [Google Scholar] [CrossRef] [Green Version]
  135. Cao, Z.; Zeng, Z.; Wang, B.; Liu, C.; Liu, C.; Wang, Z.; Li, S. Identification of Potential Bioactive Compounds and Mechanisms of GegenQinlian Decoction on Improving Insulin Resistance in Adipose, Liver, and Muscle Tissue by Integrating System Pharmacology and Bioinformatics Analysis. J. Ethnopharmacol. 2021, 264, 113289. [Google Scholar] [CrossRef]
  136. Kwon, O.; Eck, P.; Chen, S.; Corpe, C.P.; Lee, J.; Kruhlak, M.; Levine, M. Inhibition of the Intestinal Glucose Transporter GLUT2 by Flavonoids. FASEB J. 2006, 21, 366–377. [Google Scholar] [CrossRef] [Green Version]
  137. Sha, W.; Hu, F.; Xi, Y.; Chu, Y.; Bu, S. Mechanism of Ferroptosis and Its Role in Type 2 Diabetes Mellitus. J. Diabetes Res. 2021, 2021, 9999612. [Google Scholar] [CrossRef]
  138. Li, D.; Jiang, C.; Mei, G.; Zhao, Y.; Chen, L.; Liu, J.; Tang, Y.; Gao, C.; Yao, P. Quercetin Alleviates Ferroptosis of Pancreatic β Cells in Type 2 Diabetes. Nutrients 2020, 12, 2954. [Google Scholar] [CrossRef]
  139. Shoelson, S.E.; Herrero, L.; Naaz, A. Obesity, Inflammation, and Insulin Resistance. Gastroenterology 2007, 132, 2169–2180. [Google Scholar] [CrossRef]
  140. Altmann, C.; Schmidt, M.H.H. The Role of Microglia in Diabetic Retinopathy: Inflammation, Microvasculature Defects and Neurodegeneration. Int. J. Mol. Sci. 2018, 19, 110. [Google Scholar] [CrossRef] [Green Version]
  141. Kumar, B.; Gupta, S.K.; Srinivasan, B.P.; Nag, T.C.; Srivastava, S.; Saxena, R.; Jha, K.A. Hesperetin Rescues Retinal Oxidative Stress, Neuroinflammation and Apoptosis in Diabetic Rats. Microvasc. Res. 2013, 87, 65–74. [Google Scholar] [CrossRef]
  142. Sonawane, R.D.; Vishwakarma, S.L.; Lakshmi, S.; Rajani, M.; Padh, H.; Goyal, R.K. Amelioration of STZ-Induced Type 1 Diabetic Nephropathy by Aqueous Extract of Enicostemma Littorale Blume and Swertiamarin in Rats. Mol. Cell. Biochem. 2010, 340, 1–6. [Google Scholar] [CrossRef]
  143. Peeyush, K.T.; Gireesh, G.; Jobin, M.; Paulose, C.S. Neuroprotective Role of Curcumin in the Cerebellum of Streptozotocin-Induced Diabetic Rats. Life Sci. 2009, 85, 704–710. [Google Scholar] [CrossRef]
  144. Li, X.-H.; Xin, X.; Wang, Y.; Wu, J.; Jin, Z.; Ma, L.; Nie, C.; Xiao, X.; Hu, Y.; Jin, M. Pentamethylquercetin Protects against Diabetes-Related Cognitive Deficits in Diabetic Goto-Kakizaki Rats. J. Alzheimer’s Dis. 2013, 34, 755–767. [Google Scholar] [CrossRef] [Green Version]
  145. Li, C.; Zhao, X.; Zheng, H.; Cai, F. GW28-E0635 Quercetin Retards Progression of Diabetic Cardiomyopathy through Modulations of SIRT1 and AMP-Activated Protein Kinase. J. Am. Coll. Cardiol. 2017, 70, C63–C64. [Google Scholar] [CrossRef]
  146. Zhang, M.; Zhang, L.; Hu, J.; Lin, J.; Wang, T.; Duan, Y.; Man, W.; Feng, J.; Sun, L.; Jia, H.; et al. MST1 Coordinately Regulates Autophagy and Apoptosis in Diabetic Cardiomyopathy in Mice. Diabetologia 2016, 59, 2435–2447. [Google Scholar] [CrossRef] [Green Version]
  147. Bostancıeri, N.; Elbe, H.; Eşrefoğlu, M.; Vardı, N. Cardioprotective Potential of Melatonin, Quercetin and Resveratrol in an Experimental Model of Diabetes. Biotech. Histochem. 2021, 97, 152–157. [Google Scholar] [CrossRef]
  148. Zahedi, M.; Ghiasvand, R.; Feizi, A.; Asgari, G.; Darvish, L. Does Quercetin Improve Cardiovascular Risk Factors and Inflammatory Biomarkers in Women with Type 2 Diabetes: A Double-Blind Randomized Controlled Clinical Trial. Int. J. Prev. Med. 2013, 4, 777–785. [Google Scholar]
  149. Liu, J.; Li, Q.; Yang, Y.; Ma, L. Iron Metabolism and Type 2 Diabetes Mellitus: A Meta-Analysis and Systematic Review. J. Diabetes Investig. 2020, 11, 946–955. [Google Scholar] [CrossRef]
Figure 1. Chemical structure of quercetin with (A, B) and (C) representing the aromatic and γ-pyrone rings, respectively.
Figure 1. Chemical structure of quercetin with (A, B) and (C) representing the aromatic and γ-pyrone rings, respectively.
Life 12 01146 g001
Figure 2. Flow chart summarizing the pharmacological actions and therapeutic effects of quercetin.
Figure 2. Flow chart summarizing the pharmacological actions and therapeutic effects of quercetin.
Life 12 01146 g002
Figure 3. Pharmacological action of quercetin via different mechanistic pathways: Quercetin enhances pancreatic β-cell function and increases insulin release by inhibiting apoptosis, NF-κB, and JNK pathways; decreases glucose absorption in the kidney by inhibiting DPP-IV and COX-2 activity; decreases gluconeogenesis through inhibition of TNF-α and IL-4 in the liver; suppresses glucose reabsorption in the gastrointestinal tract by decreasing α-glucosidase activity; reduces blood glucose levels and oxidative stress by inhibiting IL-6 activity in the heart and blood vessels.
Figure 3. Pharmacological action of quercetin via different mechanistic pathways: Quercetin enhances pancreatic β-cell function and increases insulin release by inhibiting apoptosis, NF-κB, and JNK pathways; decreases glucose absorption in the kidney by inhibiting DPP-IV and COX-2 activity; decreases gluconeogenesis through inhibition of TNF-α and IL-4 in the liver; suppresses glucose reabsorption in the gastrointestinal tract by decreasing α-glucosidase activity; reduces blood glucose levels and oxidative stress by inhibiting IL-6 activity in the heart and blood vessels.
Life 12 01146 g003
Table 1. Pharmacological actions and side effects of antidiabetic drugs.
Table 1. Pharmacological actions and side effects of antidiabetic drugs.
Type 2 Antidiabetic AgentsPharmacological ActionsSide EffectsReferences
α-glucosidase inhibitors
(Acarbose, miglitol)
Inhibit the intestinal absorption of carbohydratesFlatulence, bloating, diarrhoea[20,21]
Biguanides
(Metformin)
Inhibit hepatic gluconeogenesis,
Reduce the liver and intestinal absorption of sugar
Increase insulin sensitivity and glucose uptake
Kidney complications, upset stomach, tiredness, and dizziness[22,23]
Dopamine agonists
(Bromocriptine, cabergoline, apomorphine)
Regulate plasma glucose, free fatty acids, and triglyceride levels in insulin-resistant patientsVisual hallucinations and confusion, edema[24,25]
Dipeptidyl peptidase-4 (DPP-4) inhibitors
(Sitagliptin, saxagliptin, linagliptin)
Increase the half-life of GLP-1 and GIPGastrointestinal problems, flu-like symptoms (headache, runny nose, sore throat)[26,27]
GLP-1 agonists
(Dulaglutide, exenatide, albiglutide)
Enhance insulin release
Reduce glucagon release
Gastrointestinal problems and nausea[28,29]
Meglitinides
(Nateglinide, repaglinide)
Stimulate the release of insulinWeight gain, hypoglycaemia, excessive sweating[30,31]
Sodium-glucose Co-transporter-2 (SGLT-2) inhibitors
(Dapagliflozin, canagliflozin, empagliflozin)
Inhibit glucose reabsorption in the renal tubuleUrinary tract infection and increased urination, upper respiratory tract infections, joint pain, nausea, and thirst[32,33]
Sulfonylureas
(Tolbutamide, tolazamide, chlorpropamide)
Inhibit ATP-sensitive potassium (KATP) channel in pancreatic β-cellsHypoglycaemia, upset stomach, skin rash, and itching[34]
Thiazolidinediones
(Rosiglitazone, pioglitazone)
Bind with the peroxisome proliferator-activated receptor (PPAR)-γ receptor resulting in the activation of several genes that regulate glucose metabolism in the liverAnaemia risk, weight gain, edema, heart failure[35,36]
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Ansari, P.; Choudhury, S.T.; Seidel, V.; Rahman, A.B.; Aziz, M.A.; Richi, A.E.; Rahman, A.; Jafrin, U.H.; Hannan, J.M.A.; Abdel-Wahab, Y.H.A. Therapeutic Potential of Quercetin in the Management of Type-2 Diabetes Mellitus. Life 2022, 12, 1146. https://doi.org/10.3390/life12081146

AMA Style

Ansari P, Choudhury ST, Seidel V, Rahman AB, Aziz MA, Richi AE, Rahman A, Jafrin UH, Hannan JMA, Abdel-Wahab YHA. Therapeutic Potential of Quercetin in the Management of Type-2 Diabetes Mellitus. Life. 2022; 12(8):1146. https://doi.org/10.3390/life12081146

Chicago/Turabian Style

Ansari, Prawej, Samara T. Choudhury, Veronique Seidel, Akib Bin Rahman, Md. Abdul Aziz, Anika E. Richi, Ayesha Rahman, Umme H. Jafrin, J. M. A. Hannan, and Yasser H. A. Abdel-Wahab. 2022. "Therapeutic Potential of Quercetin in the Management of Type-2 Diabetes Mellitus" Life 12, no. 8: 1146. https://doi.org/10.3390/life12081146

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop