Abstract

Glucose and lipid metabolism disorder in diabetes mellitus often causes damage to multiple tissues and organs. Diabetes mellitus is beneficially affected by quercetin. However, its concrete mechanisms are yet to be fully elucidated. In our study, diabetes was induced in Sprague-Dawley rats by STZ injection. The rats were randomly divided into normal control, diabetic model, low-dose quercetin treatment, high-dose quercetin treatment, and pioglitazone treatment groups. Fasting blood glucose was collected to evaluate diabetes. Immunohistochemistry and fluorometric assay were performed to explore SIRT1. Akt levels were measured through immunoprecipitation and Western blot. After 12 weeks of quercetin treatment, the biochemical parameters of glucose and lipid metabolism improved to varying degrees. Hepatic histomorphological injury was alleviated, and hepatic glycogen content was increased. The expression and activity of hepatic SIRT1 were enhanced, and Akt was activated by phosphorylation and deacetylation. These results suggested that the beneficial effects of quercetin on glucose and lipid metabolism disorder are probably associated with the upregulated activity and protein level of SIRT1 and its influence on Akt signaling pathway. Hence, quercetin shows potential for the treatment of glucose and lipid metabolism disorder in diabetes mellitus.

1. Introduction

Flavonoids beneficially affect human and animal health. As a typical flavonol-type flavonoid, quercetin is ubiquitously present in fruits and vegetables and is considered a strong antioxidant because of its ability to scavenge free radicals and bind to transition metal ions; as such, this flavonoid participates in multiple pharmacological activities, including anti-inflammation, platelet aggregation prevention, and antitumor effects [15]. Quercetin protects organisms against several types of liver diseases [6, 7] and elicits beneficial effects on glucose and lipid metabolism [8]. Euonymus alatus as a folk medicine in China has been clinically used to treat type 2 diabetes and to alleviate hyperglycemia in animals with diabetes. Interestingly, quercetin is one of the active components of E. alatus. This flavonoid can effectively attenuate hyperglycemia in alloxan-induced diabetic mice [9]; however, its concrete mechanisms remain unclear. Considering that glucose and lipid metabolism disorder is the leading cause of diabetes-related complications, we aimed to explore the protective effect of quercetin against glucose and lipid metabolism disorder in diabetes and further clarify the underlying molecular mechanisms.

Akt is a serine/threonine protein kinase, which is also known as PKB. Its phosphorylation is closely associated with glucose and lipid metabolism. After metformin treatment is administered, Akt phosphorylation is restored in the pancreas and visceral adipose tissues [10]. Hepatic Dapper1 overexpression increases the phosphorylated Akt levels by fourfold and suppresses gluconeogenesis and lipogenesis in db/db mouse livers [11]. MiR-423-5p promotes hepatic gluconeogenesis and lipid deposition and induces glucose intolerance, insulin resistance, and hyperglycemia by inhibiting the FAM3A-ATP-Akt pathway in the liver [12]. Dysfunctional Akt causes glucose and lipid metabolism disorder because this form of Akt changes the functions of downstream kinases that regulate glucose and lipid metabolism or other related signal molecules [13].

Sirtuin type 1 (SIRT1), a nicotinamide adenine dinucleotide-positive-dependent protein deacetylase belonging to the mammalian sirtuin family, regulates gene transcription and chromosome instability and targets protein functions and activities by deacetylating histone, transcription factors, transcriptional regulators, and lysine residues in intracellular functional proteins [14]. Therefore, SIRT1 participates in a series of energy metabolism and pathological and physiological processes, such as gluconeogenesis and lipid accumulation, aging, development, inflammation, and tumorigenesis [1517]. SIRT1 gene mutation may induce type 1 diabetes mellitus [18]. In SIRT1 transgenic mice, hepatic insulin sensitivity is increased, body energy demands are decreased, and insulin resistance and diabetes are further alleviated [19]. SIRT1 also protects pancreatic β-cells against cytokine toxicity [20]. In our previous studies, SIRT1 elicits protective effects on diabetic nephropathy [21]. Recent studies also showed that SIRT1 was involved in the regulation of Akt activity [22, 23]. SIRT1-induced deacetylation of K14 and K20 of Akt is necessary to promote the binding of Akt to PIP3 and cause membrane localization and activation [23]. Lysine acetylation inhibits IRS2 activity, and SIRT1-dependent deacetylation increases its activity, thereby enhancing Akt activity [24].

This work aimed to determine whether quercetin can improve the glucose and lipid metabolism of rats with diabetes and to explore its underlying mechanisms, especially those associated with Akt and SIRT1.

2. Materials and Methods

2.1. Experimental Design

Male Sprague-Dawley (SD) rats (, 6 w, 130 ± 15 g) were purchased from the Laboratory Animal Center, Sun Yat-sen University, Guangzhou, China. (Animal quality certificate number: 0112395). The rats were maintained under standard conditions with free access to water and chow. All experimental procedures conformed to the China Animal Welfare Legislation and approved by the Ethics Committee on the Care and Use of Laboratory Animals of Sun Yat-sen University. After high-fat and sugar diet (contained 0.25% bile salt, 1% cholesterol, 10% lard, and 20% sucrose) for 4 weeks, SD rats received a low dose of streptozocin (STZ) intraperitoneal injection (30 mg/kg, Sigma-Aldrich, USA) to induce diabetes. Diabetes was regarded as successfully induced in rats with fasting blood glucose (FBG) levels above 11.1 mM. Diabetic rats were randomly assigned to STZ-induced diabetic group (), low-dose quercetin treatment (QU-low, 10 mg/kg, , gavage, purity > 98%, HPLC; Zelang, Nanjing, China), high-dose quercetin treatment (QU-high, 50 mg/kg, , gavage), and pioglitazone treatment (5 mg/kg, , gavage, Takeda, Japan, subpackaged by Tianjin Takeda) groups. The doses of quercetin chosen were based on other previous preclinical experiment [2527]. Both normal control rats and STZ -induced diabetic rats were also fed equal volumes of 0.5% (w/v) CMC-Na. Rats in all group were administered by gavage 6 days every week at 9:00–10:00 am for 12 weeks.

2.2. Biochemical Analysis

At the age of 16 weeks, rats were fasted for 12 h and killed after anesthesia. FBG levels were analyzed by a OneTouch glucometer (Johnson and Johnson, USA). Glycosylated hemoglobin (HbA1c) detection was carried out following the manufacturer’s instruction (Jiancheng, Nanjing, China). Glycosylated serum protein (GSP), triglyceride (TG), total cholesterol (TC), low-density lipoprotein cholesterol (LDL-C), and high-density lipoprotein cholesterol (HDL-C) in serum were measured by commercial available kits purchased from Nanjing Jiancheng Bioengineering Institute (Nanjing, China). Liver tissues were stored at −80°C until analysis. Liver samples were rinsed with physiological saline, dried, and immediately weighed for hepatic glycogen measurement (Jiancheng, Nanjing, China).

2.3. Liver Histological Examination

Paraffin-embedded liver tissue samples were cut into 5 μm thick sections for periodic acid-Schiff pas staining, and the sections were then examined by light microscopy. Rat livers were fixed with 10% formalin, cut into 10 μm sections with a cryostat for Sudan IV staining, and then examined by light microscopy.

2.4. Immunohistochemistry

Liver specimens fixed in 10% buffered formalin were embedded in paraffin blocks. Sections (5 μm thick) of the liver were processed using a standard immunostaining protocol. After routine deparaffinization, hydration, and blockage of endogenous peroxidase, sections were pretreated by microwave for 20 min in 10 mmol/L sodium citrate buffer (pH 6.0) for antigen retrieval and followed by incubation sequentially with blocking agent, rabbit anti-SIRT1 antibody (1 : 100, Abcam, Cambridge, MA, USA) and secondary antibody (1 : 200, Promega, Madison, WI, USA).

Slides were counterstained with hematoxylin after 3 min of diaminobenzidine reaction, and cover slipped using Vectashield (Vector Labs, Burlingame, CA, USA), then photographed and converted to a digital image using light microscopy equipped with a camera (Olympus CX31, NY, USA). Negative control was carried out by omitting the primary antibody. Positive staining (dark brown) for SIRT1 in livers was quantified by two investigators in a blinded manner at a magnification of 400x using image analysis software Image-Pro Plus, and the results were expressed as the ratio of the mean of normal SD rats. Three slides were chosen at random in each animal for quantification (total 24 slides for each group).

2.5. Western Blotting

Liver tissues were lysed and protein was extracted as previously described [28]. Equal amounts of supernatant of liver lysates were separated on 8% SDS-PAGE gels and transferred onto a PVDF or a NC membrane. After washing, the membranes were blocked with 5% nonfat milk at room temperature and then incubated overnight at 4°C with primary antibodies. After further washing, the membranes were exposed to the secondary antibodies (anti-rabbit IgG or anti-mouse IgG, 1 : 10,000; Promega, USA) for 2 h at room temperature before visualizing by enhanced chemiluminescence (ECL) (Thermo Fisher Scientific, Rockford, IL, USA). The signals were visualized by GE ImageQuant LAS 4000 mini quantified by densitometry using Gel Doc XR System (Bio-Rad Laboratories, Hercules, CA, USA). The primary antibodies used were phospho-Akt (Ser473) (rabbit, 1 : 1000 dilution, Cell Signaling Technology), phospho-Akt (Thr308) (rabbit, 1 : 1000 dilution, Cell Signaling Technology), phospho-GSK-3β (Ser9) (rabbit, 1 : 1000 dilution, Cell Signaling Technology), GSK-3 β (rabbit, 1 : 1000 dilution, Cell Signaling Technology), GCK (rabbit, 1 : 1000 dilution, Abcam Company), SREBP-1 (rabbit, 1 : 1000 dilution, Abcam Company), LDLR (rabbit, 1 : 1000 dilution, Abcam Company), SIRT1 (rabbit, 1 : 1000 dilution, Abcam Company), Akt (rabbit, 1 : 1000 dilution, Cell Signaling Technology), acetylated-lysine (rabbit, 1 : 1000 dilution, Cell Signaling Technology), and tubulin (mouse, 1 : 10000 dilution, Sigma). The secondary antibodies used were anti-rabbit IgG (H + L) (1 : 1000 dilution, Cell Signaling Technology) and anti-mouse IgG (H + L) (1 : 1000 dilution, Cell Signaling Technology).

2.6. Deacetylase Activity Assay

The deacetylase activity of SIRT1 was determined with a fluorometric assay kit (CycLex, Ina, Nagano, Japan). Liver nuclear extracts (100 mg) were immunoprecipitated with rabbit anti-SIRT1 polyclonal antibody (Sigma, USA). Precipitates (15 mL) were mixed with a reaction mixture (35 mL) containing 50 mM Tris-HCl (pH 8.8), 0.25 mAU·mL-1 lysyl-endopeptidase, 1 mM trichostatin A, 800 mM NAD, and 20 mM fluorosubstrate peptide and followed by incubation for 30 min at room temperature. The fluorescence intensity was measured using a microtiter plate fluorimeter with excitation at 490 ± 10 nm and emission at 530 ± 10 nm and normalized to the protein concentration. All tests were performed in triplicate.

2.7. Immunoprecipitation

Immunoprecipitation was performed by incubating 0.5 mg liver tissue lysate protein which was determined by bicinchoninic acid assay (BCA) according to the manufacturer’ s instructions (Thermo Fisher Scientific, Rockford, IL) with 1 μg of corresponding antibody and protein G/A agarose bead (Merk, Darmstadt, Germany) at 4°C overnight. Western blotting was performed as previously described [28].

2.8. Statistical Analysis

All experiments were performed at least in triplicate. Values were expressed as means ± S.D. All data were assessed by SPSS 11.5. Statistical analyses of data were performed by one-way ANOVA using post hoc multiple comparisons. was considered to be statistically significant.

3. Results

3.1. Biochemical Parameters of STZ-Induced Diabetic Rats after Quercetin Treatment

In Table 1, the biochemical parameters were notably deteriorated in the STZ-induced diabetic rats compared with those in the normal control group at the end of the experimental period (). Quercetin (10 and 50 mg/kg) and pioglitazone treatments evidently ameliorated the biochemical parameters compared with those of the STZ-induced diabetic rats (). No evident difference in HDL-C levels was observed between these groups.

3.2. Quercetin Ameliorated the Hepatic Histomorphological Injury and Enhanced the Hepatic Glycogen Content in the STZ-Induced Diabetic Rats

The liver is an important organ for glucose and lipid metabolism. Excessive lipid accumulation in the liver may cause liver dysfunction and hepatic insulin resistance that affect gluconeogenesis, thereby promoting the conversion of glycogen to glucose in this organ. In our study, the structure of the hepatic lobule from the control group was normal (Figure 1(a)). The hepatocytes showed a distinct outline, distributed evenly, and arranged radically around the central vein in a streak. The structure of the hepatic tissues from the model group was significantly disrupted compared with that from the control group. However, quercetin and pioglitazone evidently alleviated the morphological damage to the hepatocytes of the diabetic rats. These results suggested that quercetin elicited protective effects against liver injury in STZ-induced diabetic rats. The hepatic glycogen content in STZ-induced diabetic rats largely decreased compared with that in the control group (). By contrast, the hepatic glycogen content in the STZ-induced diabetic rats remarkably increased in the quercetin- and pioglitazone-treated groups (Figure 1(b), ).

3.3. Quercetin Treatment Enhanced the Expression and Activity of Hepatic SIRT1 in STZ-Induced Diabetic Rats

SIRT1 activation shows therapeutic potential for type 2 diabetes and obesity [15]. Immunohistochemistry was conducted to investigate SIRT1 expression, which is involved in glucose and lipid metabolism in diabetes, in the livers of diabetic rats (Figure 2(a)). The results showed that SIRT1 was expressed in the nuclei and cytoplasm of the hepatocytes from the control group. Conversely, the positively stained SIRT1 was markedly reduced by fivefold in the hepatocytes, especially in the cytoplasm of the diabetic group compared with that of the control group (). The hepatic SIRT1 expression substantially increased by threefold in the quercetin-treated group compared with that in the diabetic group (, Figure 2(b)). The hepatic SIRT1 expression in the pioglitazone-treated group increased to a certain extent, but this increase was not as evident as that in the quercetin-treated group. No visible staining was also found in the negative group. We further confirmed the SIRT1 protein expression in the livers through Western blot (Figure 2(c), ). Our results showed that the protein level of SIRT1 severely decreased in the diabetic group compared with that in the control group. The SIRT1 protein in the pioglitazone-treated group was slightly upregulated compared with that in the diabetic group. However, the SIRT1 expression was increased in the quercetin-treated diabetic rats. Immunohistochemistry and Western blot results suggested that the protective effect of quercetin on glucose and lipid metabolism dysfunction in diabetes might partly depend on the upregulated SIRT1 protein level. We also explored the deacetylase activity of SIRT1 in the livers of the diabetic rats (Figure 2(d), ). The deacetylase activity of SIRT1 in the diabetic group was severely inhibited compared with that in the control group. Conversely, this activity was slightly enhanced by pioglitazone and was significantly improved by high-dose quercetin treatment.

3.4. Quercetin-Activated Hepatic Akt Signaling in STZ-Induced Diabetic Rats

Akt activity is related to phosphorylation and deacetylation. The phosphorylated Akt expression levels in the livers were examined through Western blot, and our results showed that these levels significantly decreased in the livers of the diabetic rats. Pioglitazone and quercetin markedly enhanced the expression of phosphorylated Akt (Figure 3(a), ). Akt as a substrate of SITR1 can be deacetylated by SIRT1, thereby upregulating Akt activity [22]. The acetylated Akt levels were also explored through immunoprecipitation and Western blot. Our findings demonstrated that acetylated Akt levels were enhanced in the livers of the diabetic rats but were slightly reduced by pioglitazone. By comparison, its levels were markedly attenuated after quercetin was administered (Figure 3(b), ). These observations implied that deacetylated Akt levels corresponded to hepatic SIRT1 protein levels.

3.5. Effects of Quercetin on the Related Downstream Effector Protein Levels in the Livers of Diabetic Rats

Western blot results showed that the GSK-3β phosphorylation level was markedly reduced in the diabetic rats, compared with that in the normal control (Figure 4(a), ). No evident difference in the GSK-3β phosphorylation was detected between the pioglitazone group and the diabetic group (Figure 4(a), ). The GSK-3β phosphorylation level in the quercetin-treated diabetic rats significantly increased (Figure 4(a), ). The hepatic GCK protein expression was markedly downregulated in the diabetic rats compared with that in the normal control (Figure 4(b), ). By comparison, the protein expression of GCK in the pioglitazone-treated group was upregulated (Figure 4(b), ). The protein expression of GCK was remarkably increased by quercetin (Figure 4(b), ). Likewise, the protein expression of SREBP-1c in the liver was increased in the diabetic group, compared with that in the control group (Figure 4(c), ). Quercetin and pioglitazone treatment evidently attenuated SREBP-1c protein level (Figure 4(c), ). The hepatic LDLR expression in the diabetic group was markedly downregulated compared with that in the control group (Figure 4(d), ) but was evidently upregulated after quercetin and pioglitazone treatments were administered (Figure 4(d), ).

4. Discussion

Most patients with type 2 diabetes suffer not only from hyperglycemia but also from lipid metabolism dysfunction. Pioglitazone is a prescription drug belonging to the thiazolidinedione class with a hypoglycemic action to treat diabetes. It modulates the transcription of genes involved in the control of glucose and lipid metabolism in the muscles, adipose tissues, and livers [29, 30]. In clinical trials, pioglitazone as monotherapy or in combination with other oral antidiabetic drugs or insulin enhances blood glucose levels, long-term glucose control, and lipid profiles [31]. Patients with prediabetes or T2DM treated with pioglitazone experience a remarkable improvement in hepatic steatosis, and steatohepatitis can be cured within 6 months [32]. Therefore, pioglitazone was chosen as a positive control drug in the analysis of glucose and lipid metabolism in our research. Our results revealed that FBG, HbA1c, GSP, TC, TG, and LDL-C levels significantly increased in STZ-induced diabetic rats. These biochemical parameters were also improved in diabetic rats treated with quercetin and pioglitazone, thereby suggesting that quercetin ameliorated the dysfunction of glucose and lipid metabolism in STZ-induced diabetic rats. However, the exact mechanism remains unclear and needs further investigations.

The hypoglycemic effect mediated by quercetin in diabetic rats is closely related to changes in hepatic Akt activity. Quercetin and pioglitazone effectively increase Akt phosphorylation levels at sites T308 and S473, which are pivotal sites for activity Akt [33], in the livers of diabetic rats, and thus contribute to the improvement of glucose metabolism.

GSK-3β is a downstream factor regulated by Akt and Akt-induced GSK-3β phosphorylation at the site of serine 9 which inhibits GSK-3β [13]. Increased GSK-3β activity also prevents IRS activity under diabetic conditions, thereby leading to dysfunction in insulin-induced signaling pathway and consequently aggravating damage to the Akt signaling pathway [34]. In our study, the downregulation of phosphorylated GSK-3β (Ser9) in the diabetic liver was significantly increased by quercetin. However, no significant changes were observed between the pioglitazone-treated group and the diabetic group. Based on our data, our conclusion is that quercetin could effectively prevent the abnormal increase in GSK-3β activity by regulating Akt in the livers of diabetic rats, relieve the inhibitory effect on downstream glycogen synthase, and consequently promote the synthesis of liver glycogen.

Glucokinase (GCK), also known as hexokinase type IV expressed mainly in pancreatic β-cells and hepatocytes and affected by Akt, is an important regulatory factor in glucose metabolism [35, 36]. GCK serves as a glucose sensor to predict the variability of blood glucose concentrations and plays a crucial role in the regulation of insulin secretion in pancreatic β-cells [13]. Glucose is initially transported into cells by glucose transporters in the liver, subsequently converted to glucose-6-phosphate (G6P) by GCK and stored as glycogen. Consistent with previous findings [13], our results indicated that the GCK protein expression and activity in the livers of the diabetic rats significantly decreased. However, quercetin and pioglitazone promoted the GCK protein expression in the livers of the diabetic rats, suggesting that quercetin and pioglitazone enhanced the synthetic function of hepatic glycogen and alleviated the glucose metabolism disorder by regulating GCK.

Akt activation is involved not only in the regulation of glucose metabolism but also in other processes. For example, Akt controls sterol regulatory element-binding proteins (SREBPs), which are the main transcriptional regulators of lipid metabolism [37]. Akt also regulates lipogenesis by modulating LDLR expression [13]. Thus, Akt signaling pathway is closely implicated in the control of lipid metabolism. On the basis of our data, we concluded that quercetin and pioglitazone improved the blood lipid parameters of diabetic rats by activating Akt and upregulating SREBPs and LDLR.

In addition to phosphorylation activation, reversible acetylation is a posttranslational modification that regulates Akt activity. Under basal conditions, Akt is acetylated in various tissues, and this modification suppresses Akt activity. SIRT1, as a deacetylase, can activate Akt by inducing the Akt deacetylation [38]. Akt acetylated by SIRT1 at lysine residues in the pH domain can upregulate Akt activation to a certain extent, play a crucial role in the binding of Akt to PIP3, and consequently trigger cascades of downstream reactions [22, 23, 38]. Thus, whether quercetin improves glucose and lipid metabolism disorder in STZ-induced diabetic rats through the SIRT1/Akt pathway should be further examined. In our study, the level of lysine acetylation of Akt increased, but quercetin induced the lysine deacetylation of Akt in the livers of diabetic rats to a certain degree. As such, whether this effect results from quercetin-induced SIRT1 activation should be elucidated. To confirm this effect, we observed the expression of SIRT1 in the livers of diabetic rats and found that the reduced SIRT1 protein level and deacetylase activity in the livers of the diabetic rats were upregulated in the quercetin-treated group. One of the mechanisms to achieve SIRT1 activation in vivo is to raise intracellular levels of NAD+ either by increased synthesis or diminished degradation [39, 40]. Quercetin has been shown to activate SIRT1 in vivo by enhancing NAD+ level as an inhibitor of NAD+ nucleosidaase CD38 [41]. The change of SIRT1 in diabetic liver is consistent with the variation tendency of deacetylation on the Akt lysine site, suggesting that quercetin might induce Akt deacetylation and further activate Akt by increasing SIRT1 protein levels and activities in the livers of diabetic rats. Thus, this flavonoid might improve the glucose and lipid metabolism disorder in diabetic rats. Furthermore, SIRT1 interacted with the Akt in cytoplasm and then induced the deacetylation of Akt, promoting Akt membrane location and binding to PIP3 [23]. So, it is possible that the change of SIRT1 in the cytoplasm is more important than in the nuclei for Akt activation. Compared with quercetin treatment, pioglitazone treatment did not significantly affect the SIRT1/Akt pathway, and this finding reveals the advantage of quercetin in the treatment of glucose and lipid metabolism disorder to some degree. However, further in vitro studies should be performed to verify this hypothesis.

In our study, quercetin effectively alleviated the glucose and lipid metabolism disorder in STZ-induced diabetic rat model, and this observation was correlated with hepatic Akt activation in diabetic rats. Quercetin promoted hepatic glycogen synthesis and reduced blood glucose by increasing Akt phosphorylation, GSK-3 phosphorylation, and GCK protein expression levels. Quercetin also improved lipid metabolism by regulating SREBPs and LDLR protein expression, which are controlled by Akt. This flavonoid not only activated Akt by stimulating phosphorylation but also increased Akt activity by inducing Akt deacetylation possibly because of the effect of quercetin on SIRT1. However, further in vitro experiments should be conducted to explore the mechanisms of quercetin in diabetes.

5. Conclusion

Quercetin beneficially affects glucose and lipid metabolism disorder and is probably associated with the upregulated activity and protein level of SIRT1 and its influence on Akt signaling pathway. Hence, quercetin shows potential for the treatment of glucose and lipid metabolism disorder in diabetes.

Conflicts of Interest

The authors declare that there is no conflict of interest regarding the publication of this article.

Authors’ Contributions

Xi Xie and Jing Peng designed and performed the experiments, acquisition, and analysis of data and drafted the manuscript. Li Zhu, Keye Li, Xiaohong Lin, and Qianhui Shen helped to perform animal experiments and prepare the manuscript. Qingde Li, Xiaoding Lin, and Guoping Li have been involved in drafting the manuscript and revising it critically for important intellectual content.

Acknowledgments

This study was supported by research grants from the National Natural Science Foundation of China (Grant no. 81402984), the Medical Scientific Research Foundation of Guangdong Province, China (no. A2016591), the Hospital Pharmacy Research Foundation of Guangdong Province, China (no. 2016A02), and the Education and Teaching Research Project of Hainan University, China (no. hdjy1709).