Skip to main content

Ameliorative activity of standardized Coccoloba uvifera leaves extract against streptozotocin-induced diabetic rats via activation of IRS-1/PI3K/AKT/GLUT2 pathway in liver

Abstract

Background

Coccoloba uvifera L. (Family: Polygonaceae) known as sea grape is natively distributed in middle and south America. The aqueous leaf extract showed inhibitory activities against α-glucosidase and α-amylase in previous reports. Moreover, the hydroalcoholic leaves extract ameliorated hyperglycemia in the oral glucose tolerance test. Despite these promising results, the extracts used in these studies were not standardized, nor was their mechanism of action elucidated. The current study aims to standardize the ethanolic C. uvifera leaves extract (CU) using markers, and assess its ameliorative activity against diabetes and its hepatoprotective activity against diabetic complications.

Results

Standardized leaves’ ethanolic extract contained 0.09 ± 0.00057 and 0.23 ± 0.0011 mg/g gallic acid and rutin, respectively, as estimated by HPLC. Administration of CU (100, 200 and 400 mg/kg) for 6 weeks ameliorated DM manifestations in STZ-induced diabetic rats in a dose-dependent manner. The ethanolic extract reduced fasting blood glucose, increased serum insulin and reduced elevated liver enzymes. CU counteracted oxidative stress, promoted glucose metabolizing enzymes and reduced gluconeogenesis enzymes. The underlying mechanism involved increased expression of IR, IRS-1, IRS-2 and GLUT2 in liver tissue through activation of PI3K/AKT signaling. The histopathological study demonstrated reduced inflammation and hepatocyte degeneration.

Conclusion

CU could be used as a promising antidiabetic drug with hepatoprotective activity in diabetes hepatic complications. The standardized CU ethanolic extract should be further assessed clinically alone or in combination with other antidiabetic remedies.

Background

Combating diabetes mellitus (DM) and its complications is one of the major challenges facing the healthcare system worldwide. DM has a high prevalence among all ages. It afflicted 463 million people in 2019, projected to increase to 578 million (10.2%) by 2030 [1]. DM complications due to persistent hyperglycemia harmfully affect vital physiological systems involving the Kidney, neurons, heart and liver [2]. One of these complications is hepatotoxicity [3].

DM is associated with multiple liver abnormalities such as abnormal glycogen deposition, non-alcoholic fatty liver disease, fibrosis, cirrhosis, abnormal elevated hepatic enzymes, acute liver disease and viral hepatitis [4]. Excessive accumulation of fats in the liver with persistent hyperglycemia may worsen insulin resistance leading to severe metabolic dysfunction. Consequently, this would increase the mortality and morbidity within diabetic patients through the damage of hepatocytes. The biochemical alterations induced by DM are comparable to those observed in serious liver diseases, e.g., the formation of hepatocellular carcinomas (HCCs) and even end-stage liver failure [5].

A combined pharmacological therapy approach is applied to have a better control on the serum glucose level and to limit diabetes complications, e.g., metformin and pioglitazone (thiazolidinediones), together with other drugs such as atorvastatin, betaine, losartan and orlistat [6, 7]. However, these synthetic drugs possess not only undesirable side effects but also high cost. Herbal plants are safe and cost-effective complementary therapy for diabetic patients [8, 9].

Coccoloba uvifera L. (Family: Polygonaceae) known as sea grape which is natively distributed in Middle and South America [10]. The sea grape was traditionally used by the Native Americans to make medicinal teas from its leaves, bark and roots. The sea grape's astringent juice and decoctions of wood, bark and roots were used to treat diarrhea, dysentery, hemorrhages and venereal diseases; they were also applied externally for rashes and other skin afflictions. A tea made from the leaves was used to treat hoarseness and asthma, as well as to bathe wounds. The resinous gum of the bark was also used against throat ailments, and the root decoction was used to treat dysentery [11].

Leaves of sea grape have many biological activities such as antidiabetic, antioxidant, anti-inflammatory, antimicrobial and cytotoxic activities [12,13,14]. The aqueous extract of leaves showed inhibitory activities against α-glucosidase and α-amylase enzymes [15]. Moreover, the leaf’s hydroalcoholic extract ameliorated hyperglycemia in the oral glucose tolerance test [14]. In addition, a patent application demonstrated effective treatment of a diabetic condition by the daily ingestion of a tea brewed from Coccoloba uvifera leaves ethanolic extract [16]. Several compounds, such as flavonoids, anthraquinones, anthocyanins and terpenoids, were isolated from the plant [17, 18]. Moreover, different phenolic acids were quantified from C. uvifera leaves ethanolic extract which have been reported to control DM and its complications [12, 19]. Despite these promising results, the ethanolic extracts were not standardized, nor was their mechanism of action elucidated. The current study aims to (i) standardize the ethanolic C. uvifera leaves extract (CU) using markers, (ii) assess the ameliorative activity of the standardized extract against diabetes and (iii) assess its hepatoprotective activity against diabetic complications.

Materials and methods

Chemical and reagents

For extraction and HPLC analysis, respectively, solvents of analytical and HPLC grades were used. They were purchased from Sigma-Aldrich (St. Louis, MO, USA).

Plant material and extraction

Leaves (2 kg) were air-dried and grinded to coarse powder. Powdered leaves were exhaustively extracted with 95% ethanol by maceration (5 X 10L). Under reduced pressure, the solvents were distilled off using Buchi Rotavapor (Switzerland) to yield 75 g of greenish-black extract of CU, and it was stored in closed airtight container at 4 °C. Aliquots were used for the chemical analysis and the pharmacological study.

Chemical standardization of Coccoloba uvifera L. extract

Determination of total phenolic and flavonoid content

Total phenolics was assessed using Folin–Ciocalteu reagent and it was expressed as mg/g gallic acid equivalents (GAE) [20]. Gallic acid standard curve was set up at concentration range (250–15.6 µg/mL). The assay was constituted by mixing 10% Folin–Ciocalteu reagent (200 µL) with CU extract, gallic acid or methanol (100 µL) to form sample, standard and blank assay mixtures. Next, saturated Na2CO3 solution (7%, 800 µL) was added and mixed. The mixture was incubated for an hour at room temperature in a dark place. Aliquots of the assays (200 µL) were transferred to a transparent 96-well plate to measure the absorbance at 630 nm using plate reader (ELx 808, BIO-TEK Instruments, US).

Besides, the total flavonoid content was investigated using aluminum chloride colorimetric assay [21]. The flavonoid–aluminum complexes were measured at absorbance 403 nm. Quercetin (100–10 µg/mL) was chosen as a reference standard. 250 µL AlCl3 aqueous solution 10% was combined and vortexed with 500 µl extracts, methanol or quercetin and 250 µL of water. Following a 10-min incubation period at room temperature, 250 µL of the aliquots was transferred to a transparent 96-well plate, water was replaced with the same volume as AlCl3 in the blank. The amount of flavonoid compounds in the dry extract was represented as milligrams equivalents of quercetin per gram (mg/g QE).

High-performance liquid chromatography (HPLC) analysis

Aliquots of CU (121 mg) were dissolved in methanol:water (50:50) at final concentration 50 mg/ml, filtered using Millex-HV 0.45-mm membrane filters (Millipore, Bedford, MA, USA). HPLC analysis was carried out on waters 2690 Alliance HPLC system equipped with a Waters 996 photodiode array detector PDA (Milford, MA, USA). The flow rate was maintained at 1.0 mL/min; 10 µL sample was injected; and the chromatogram was traced at 280 nm. Chromatographic separations were performed on a Kromasil C-18 guard column (5 µm, 4.6 × 250 mm) (Eka Chemicals, Sweden). For the standardization process, rutin and gallic acid were selected as markers for the standardization of CU. A standard calibration curve was performed in the detector linear range from 10 to 50 µg/mL for both standards (supplementary data, Figures S1, S2). The mobile phase comprises of mixture A (0.1% phosphoric acid in water) and B (methanol) run in gradient mode. The following gradient elution was used: 95/5% A/B for 3 min, then increased to 50% B at 50 min, then elevated to 30/70% A/B at 55 min, followed by 10/90% A/B in 75 min. Under the previous conditions, gallic acid and rutin were eluted at 13.60 and 51.16 min, respectively (Fig. 1).

Fig. 1
figure 1

HPLC standardization of CU leaves extract. A Chromatogram of total ethanolic leaves extract of C. uvifera (CU) traced at 280 nm. B Authentic gallic acid and rutin chromatogram eluted at 13.6 and 51.16 min at 280 nm, respectively

The pharmacological study

Animal

Adult male Wistar albino rats weighing 150–200 g were used. Rats were kept under controlled environmental conditions, humidity (60 ± 10%), a light/dark cycle of 12/12 h and room temperature. All animals had free access to food and water throughout the study.

Experimental design

For diabetes induction, rats were fasted overnight then injected with STZ (40 mg/kg, i.p.) (Sigma-Aldrich Chemicals (St. Louis, MO, USA), which was freshly prepared in cold 0.1 M citrate buffer (pH 4.5) [22]. STZ-injected rats were allowed to drink 5% glucose solution for 24 h to overcome initial hypoglycemic mortality induced by STZ. After 48 h of injection, diabetic animals were identified by measuring blood glucose level using an analyzer (Roche Diagnostic Accu-Check test strips, Germany). For the experiment, rats with blood glucose levels more than 250 mg/dl were selected.

Rats were randomly divided into five groups (n = 6 per group) as follows: diabetic untreated group (STZ), diabetic CU at three dose levels (100, 200 or 400 mg/kg, p.o.) treated groups [14] and a fifth group that received a single citrate buffer i.p. and daily 0.1% tween 80 p.o and served as normal control. CU was dissolved in 0.1% tween 80 and administered for 6 weeks starting from the third day after the injection of STZ. Animals were weighed on the first and last day of treatment. The collection of blood was from the retro-orbital sinus under light anesthesia (sodium pentobarbital; 10 mg/kg, i.p), and serum was separated and stored at − 80 °C to be used for the estimation of insulin and liver enzymes. Animals were euthanized by cervical dislocation under anesthesia and the liver was removed and washed with saline. A part of liver was placed in 10% (v/v) formalin for 24 h for histopathological analysis. The other parts were stored at − 80 °C for biochemical analysis. Frozen tissues were homogenized in a buffer for biochemical testing.

Biochemical analysis

Measurement of fasting blood glucose and serum insulin levels

Blood glucose levels were determined from tail vein of overnight fasted rats using glucometer (Roche Diagnostic Accu-Check test strips, Germany). According to the manufacturer’s instructions, serum insulin levels were assessed using a commercially available rat ELISA kit (Ray Biotech, Peachtree Corners, GA, USA).

Measurement of serum aminotransferases and liver oxidative stress biomarkers

Serum aspartate aminotransferase (AST) and alanine aminotransferase (ALT) were measured by kits supplied by Biodiagnostic (Cairo, Egypt). Besides, liver malondialdehyde (MDA) as an index of lipid peroxidation and reduced glutathione (GSH) contents were measured colorimetrically according to the methods of [23] and [24], respectively. Results are expressed as nmol/g for MDA and µg/g for GSH.

Estimation of liver carbohydrate metabolic enzyme activities and glycogen content

Carbohydrate metabolic enzymes such as hexokinase, glucose-6-phosphatase, fructose-1,6-bisphosphatase and glycogen content were measured by the method of [25,26,27] and [28], respectively.

Quantitative real-time polymerase chain reaction

Using an SV Total RNA Isolation System (Promega, Madison, WI, USA), total RNA was isolated from the liver tissues, and the purity of the resulting RNA was determined spectrophotometrically at 260/280 nm. After that, the same quantities of RNA were reverse-transcribed into cDNAs using Reverse Transcription System (Promega). Quantitative RT-PCR of IR, IRS-1, IRS-2, GLUT2 and β-actin mRNAs was performed using SYBR green (iTaq Universal SYBR Green Supermix, Bio-Rad, Hercules, CA, USA). Table 1 describes the primer sequences employed. The thermal cycler protocol’s initial enzyme activation stage was performed for 10 min at 95 ◦C. This was followed by 40 cycles of 15 s denaturation at 95 ◦C and 60 s annealing/extension at 72 ◦C. Using the 2−ΔΔCT formula, the relative expression of target genes was determined. Each value was expressed as a fold change and normalized to β-actin levels.

Table 1 Primer sequences used in RT-PCR

Western blot analysis

After the total protein was extracted from the liver tissue, equal amounts of protein (10 μg) were separated by sodium dodecyl sulfate polyacrylamide gel electrophoresis. The protein was transferred to a nitrocellulose membrane (Amersham Bioscience, Piscataway, NJ, USA). Later, the membranes were washed with phosphate-buffered saline (PBS) and blocked via immersion in 5% (w/v) skim milk powder overnight. The membranes were then incubated with primary antibodies against p-PI3K (Tyr458, Tyr199) (1:1000, Cat. No. PA5-17,387), p-AKT (Ser473) (1:1000, Cat. No. 44-621G) and β-actin (1:500, Cat. No. PA1-183) obtained from Thermo Fisher Scientific Inc. (Rockford, IL, USA). Subsequently, the membranes were washed, and secondary antibodies labeled with peroxidase were added and they were incubated for one hour at 37 ◦C. The ChemiDoc™ imaging equipment with Image Lab™ software version 5.1 (Bio-Rad Laboratories Inc., Hercules, CA, USA) was used to analyze the band intensity. The results were expressed using arbitrary units following normalization to the expression of the β-actin protein.

Histopathological analysis

The fixed specimens of the liver were processed overnight for dehydration, clearing and impregnation using an automatic tissue processor. Using an embedding station, the specimens were embedded in paraffin blocks, and a microtome was used to cut serial sections with a thickness of 5 µm. Hematoxylin and eosin staining were used for routinely staining of the tissue sections. Using the light microscopy, the mounted specimens were observed and scored as previously described [29]; a semiquantitative comparison was conducted for the structural changes, and the abnormalities in the tissue sections were graded from 0 (normal structure) to 3 (severe pathological changes).

Statistical analysis

Data are expressed as mean ± standard deviations (S.D.). All results obtained were analyzed using one-way ANOVA followed by Tukey's multiple comparison test. Statistical analyses were performed using GraphPad Prism software (version 9; GraphPad Software, Inc., San Diego, CA, USA). A probability level of < 0.05 was accepted as statistically significant in all statistical tests.

Results

Characterization of Coccoloba uvifera L. ethanolic leaves extract.

It was imperative to standardize the CU extract before the pharmacological study, The total phenolic content CU was determined to be 205.26 ± 0.362 mg of GAE/g of dry extract. At the same time, the total flavonoid content assay revealed the presence of 32.756 ± 0.507 mg QE/g of dry extract. HPLC analysis revealed the presence of rutin as a major flavonol glycoside and gallic acid as a major phenolic acid in C. uvifera leaves with concentration of 0.23 mg ± 0.0011 and 0.09 ± 0.00057 mg per gram of extract, respectively (Fig. 1).

Effect of CU extract on STZ-induced changes in body weight, fasting blood glucose and serum insulin

STZ diabetic rats exhibited a significant body weight loss at the end of the 6-week experiment (Fig. 2). However, treatment with CU extract (100, 200 or 400 mg/kg) increased the body weight of animals as compared to STZ diabetic rats. Further, STZ produced a marked elevation in serum fasting blood glucose level to reach 4.9-fold along with a decline in serum insulin level to reach 48.6% of the control group values (Fig. 3A, B). Treatment with STZ increased blood glucose level to 485.20 mg/dL compared to control group showing 98.33 mg/dL. Treatment with CU extract (100, 200 or 400 mg/kg) lowered fasting blood glucose to attain 382.20, 368.70 and 195.30 mg/dL, respectively. Administration 200 or 400 mg/kg CU only increased serum insulin to reach 1.5-fold and 1.8-fold, respectively, compared to the STZ group Table S1. It was obvious that CU was effective in a dose-dependent manner.

Fig. 2
figure 2

Effect of CU extract on STZ-induced changes in body weight. Each bar with a vertical line represents the mean of experiments ± S.D. (n = 6). Statistical analyses were performed using the one-way ANOVA followed by Tukey's multiple comparison test, with the criterion for statistical significance as follows: **p < 0.01 and ****p < 0.0001

Fig. 3
figure 3

Effect of CU extract on STZ-induced changes in fasting blood glucose (A) and serum insulin (B). Each bar with a vertical line represents the mean of experiments ± S.D. (n = 6). Statistical analyses were performed using the one-way ANOVA followed by Tukey's multiple comparison tests, with the criterion for statistical significance as follows: **p < 0.01, ***p < 0.005, ****p < 0.001 and ns = no significance

Effect of CU extract on STZ-induced changes in liver enzymes and oxidative stress biomarkers

STZ diabetic rats showed markedly elevated serum activity of AST and ALT to reach 3.5-fold and 2.8-fold the control values, respectively (Fig. 4A, B). Administration of CU extract (100, 200 or 400 mg/kg) to diabetic rats resulted in a significant decrease in the serum activity of AST to reach 61.1%, 51.7% and 39.2%, and ALT to reach 88.8%, 71.3% and 44.1%, respectively, compared to the STZ group values (Table S1). Likewise, induction of diabetes using STZ increased lipid peroxidation as evidenced by elevated hepatic MDA level to reach 2.9-fold and depleted hepatic GSH content to reach 34.5% as compared to diabetic rats (Fig. 4C, D). CU extract (100, 200 or 400 mg/kg) treatment was effective in reducing MDA level to reach 76.8%, 53.7% and 44.7%, and replenishing GSH content to reach 1.6-fold, 2-fold and 2.4-fold, respectively, as compared with STZ-treated rats. As previously elucidated, the effect of CU was dose-dependent.

Fig. 4
figure 4

Effect of CU extract on STZ-induced changes in serum AST (A), serum ALT (B) activities, hepatic MDA (C) and hepatic GSH (D) contents. Each bar with a vertical line represents the mean of experiments ± S.D. (n = 6). Statistical analyses were performed using the one-way ANOVA followed by Tukey's multiple comparison test, with the criterion for statistical significance as follows: *p < 0.05, **p < 0.01, ***p < 0.005, ****p < 0.001 and ns = no significance

Effect of CU extract on STZ-induced changes in hepatic carbohydrate metabolic enzyme activity and glycogen content

The liver tissue of STZ diabetic rats revealed a significant inhibition in the activity of hexokinase and glycogen content to reach 61.4% and 45.1%, respectively, along with a substantial increase in the activity of glucose-6-phosphatase and fructose-1,6-bisphosphatase to reach 2.6-fold and 3.1-fold, respectively, in comparison with the control group (Fig. 5). On the other hand, treatment with CU extract (200 or 400 mg/kg) inversed the decrease in hexokinase to reach 1.3-fold and 1.5-fold, as well as glycogen to reach 1.7-fold and 1.9-fold, respectively, as compared to STZ group (Fig. 5A, B). Similarly, administration of CU extract (100, 200 or 400 mg/kg) to diabetic rats decreased the glucose-6-phosphatase activity to reach 75.8%, 58.7% and 42.4%, respectively, and fructose-1,6-bisphosphatase to reach 80.9%, 69.8% and 53.8%, respectively, as compared to STZ group (Fig. 5C, D).

Fig. 5
figure 5

Effect of CU extract on STZ-induced changes in the hepatic activity of hexokinase (A), glycogen content (B), fructose-1,6-bisphosphatase (C) and glucose-6-phosphatase (D). Each bar with a vertical line represents the mean of experiments ± S.D. (n = 6). Statistical analyses were performed using the one-way ANOVA followed by Tukey's multiple comparison test, with the criterion for statistical significance as follows: *p < 0.05, **p < 0.01, ***p < 0.005, ****p < 0.001 and ns = no significance

Effect of CU extract on STZ-induced changes in mRNA expression of IR, IRS-1, IRS-2 and GLUT2 in liver tissue

Induction of diabetes by STZ significantly downregulated the mRNA expression of hepatic IR, IRS-1, IRS-2 and GLUT2 to reach 28.1%, 16.1%, 38.4% and 30.7%, respectively, in comparison with control rats (Fig. 6). Treatment of diabetic rats with CU extract (100, 200 or 400 mg/kg) significantly improved the expression of IR to reach 2.2-fold, 2.3-fold and 2.8-fold, respectively, IRS-1 to reach 3.3-fold, 3.4-fold and 4.3-fold, respectively, IRS-2 to reach 1.9-fold, 1.9-fold and 2.3-fold, and GLUT2 to reach 2.2-fold, 2.2-fold and 2.7-fold, respectively, compared to STZ group values.

Fig. 6
figure 6

Effect of CU extract on STZ-induced changes in mRNA expression of IR (A), IRS-1 (B), IRS-2 (C) and GLUT2 (D) in liver tissue. Each bar with a vertical line represents the mean of experiments ± S.D. (n = 3). Statistical analyses were performed using the one-way ANOVA followed by Tukey's multiple comparison test, with the criterion for statistical significance as follows: *p < 0.05, **p < 0.01, ***p < 0.005, ****p < 0.001 and ns = no significance

Effect of CU extract on STZ-induced changes in PI3K/AKT signaling in liver tissue

STZ diabetic rats demonstrated a decreased expression of p-PI3K and p-AKT to reach 30.3% and 16.8%, respectively, compared to normal rats (Fig. 7). In contrast, CU extract (100, 200 or 400 mg/kg) administration effectively ameliorated STZ-induced downregulation of p-PI3K to reach 2.6-fold, 2.5-fold and 2.5-fold, respectively, and p-AKT to reach 3.6-fold, 3.6-fold and 4.7-fold, respectively, compared to STZ group.

Fig. 7
figure 7

Effect of CU extract on STZ-induced changes in p-PI3K (A) and p-AKT (B) expression in liver tissue. Each bar with a vertical line represents the mean of experiments ± S.D. (n = 3). Statistical analyses were performed using the one-way ANOVA followed by Tukey's multiple comparison tests, with the criterion for statistical significance as follows: *p < 0.05, **p < 0.01, ****p < 0.001 and ns = no significance

Effect of CU extract on STZ-induced histopathological changes in liver tissue

Control group (Fig. 8A, B) showed normal hepatic parenchyma without any detectable histopathological alteration. On the other hand, examination of hepatic tissue of STZ group (Fig. 8C, D) showed serious injuries in the hepatic parenchyma. Multifocal random and portal mononuclear inflammation infiltration were detected in the several examined sections accompanied by increased portal fibroplasia and necrobiotic changes in the surrounding hepatocytes. Marked oval cells hyperplasia were commonly observed among several affected sections. Mild improvement was detected in CU (100 mg/kg) group (Fig. 8E, F) that revealed moderate to high number of inflammatory cells infiltration accompanied by mild oval cells hyperplasia and limited degeneration of the hepatocytes. Comparable results were detected in CU (200 mg/kg) group (Fig. 8G, H), except for fewer sections that exhibited fewer inflammatory cells infiltration when compared to CU (100 mg/kg) group. Meanwhile, marked improvement was reported in CU (400 mg/kg) group (Fig. 8I, J) revealing apparently normal hepatic parenchyma in several examined sections. A sporadic case in CU (400 mg/kg) group showed limited area of sinusoidal dilation. The statistical analysis of lesion score of STZ-induced hepatic injuries was conducted in different experimental groups. STZ group exhibited a significant increase in lesion score compared with other groups. Meanwhile, all groups of CU treatment showed a significant decrease in lesion score when compared to STZ group (Fig. 8K).

Fig. 8
figure 8

Effect of CU extract on STZ-induced histopathological changes. H&E staining of the liver tissues from different experimental groups. A, B The control group showed normal hepatic parenchyma with healthy hepatocytes. C, D STZ group showing portal fibroplasia with mononuclear inflammatory cell infiltration (arrow). E, F CU (100 mg/kg) group showing limited portal inflammatory cell infiltration (arrow) with congested blood vessels. G, H CU (200 mg/kg) group showing mild portal hepatitis (arrow) with limited oval cell hyperplasia. I, J CU (400 mg/kg) group normal hepatic parenchyma. K Liver lesion score in different groups represented as mean ± S.D. (n = 3). Statistical analyses were performed using the one-way ANOVA followed by Tukey's multiple comparison tests, with the criterion for statistical significance as follows: *p < 0.05, **p < 0.01, ****p < 0.001 and ns = no significance

Discussion

Globally, diabetes has become a serious health concern, with over 90% of cases of DM being type 2 diabetes [30]. About one-third of patients with cirrhosis also have diabetes, and people with type 2 diabetes have a higher risk of developing chronic liver disease, including steatohepatitis and non-alcoholic fatty liver disease [31]. This study assessed the CU’s in vivo antidiabetic and hepatoprotective effect in STZ-induced diabetic mice. STZ is a cell-specific toxin that causes DNA damage and free radical production in pancreatic islets, which results in permanent damage [32]. Prior to the pharmacological study, the extract was chemically characterized. The HPLC chromatogram of the CU showed that gallic acid and rutin are among the major constituents of the extract. They were selected as markers because of their wide availability, cost-effectiveness and efficacy in clinical trials, which are important factors for QC protocols. The biochemical, molecular and histopathological levels evidenced the CU efficacy. Notably, the extract showed a dose-related activity, with 400 mg/kg being the most active dose. The study disclosed the possible underlying molecular mechanisms and assessed the histopathological changes.

The ameliorative CU activity against diabetes and diabetes liver complications.

CU had effectively restored serum insulin, resulting in a significant decrease in serum glucose. These effects were reflected in increased body mass. These effects suggest CU-protected Langerhans cells against STZ toxicity, maintaining insulin secretion. CU would be beneficial for type 2 DM patients regarding these aspects. Elevated liver enzymes are correlated with higher odds of diabetes [33]. This could be related to oxidative stress induced by DM [34]. It was reflected in the current study by low GSH and high MDA levels (Fig. 4). CU phenolics had counteracted this oxidative stress leading to restoration of GSH levels and reduction in the oxidative stress marker MDA.

CU normalized the hepatic carbohydrates metabolic enzymes and replenished the glycogen content

The liver is involved in glycogen formation and controls postprandial hyperglycemia. It is well recognized that diabetes mellitus affects the liver's normal ability to synthesize glycogen [35]. Furthermore, partial or complete insufficient amounts of insulin in DM disrupt the metabolism of carbohydrates and lower the activity of the enzymes phosphofructokinase, hexokinase and glucokinase, which depletes muscle and liver glycogen. These enzymes provide an approach for evaluating the peripheral utilization of glucose [36]. CU effectively normalized the hepatic carbohydrate metabolic enzyme hexokinase and restored the glycogen content almost to the normal level. These activities in turn revealed the CU insulin-mimetic activity and its ability to facilitate the glucose uptake by the cells. On the other hand, glucose-6-phosphatase and fructose-1,6-bisphosphatase are crucial enzymes in gluconeogenesis [37]. The activity of these enzymes was reduced significantly by CU. Consequently, CU will result in the inhibition of gluconeogenesis and, hence, lower endogenous glucose synthesis.

CU elevated the expression of insulin receptors and glucose transferase

Insulin receptors (IRs) and insulin receptor substrates (IRSs), two important proteins in the insulin signaling system, are downregulated in the livers of diabetic rodents and humans [38]. The maintenance of glucose metabolism largely depends on IRSs, the major mediators of insulin signaling [39]. Tyrosine phosphorylation of the IRS protein is necessary for the metabolic effects of insulin [40]. This phosphorylation triggers a signaling cascade by activating PI3K and the serine/threonine kinase Akt/PKB. Furthermore, hepatic nutritional homeostasis is particularly dependent on IRS-2, as it mediates the anabolic effects of insulin via the PI3K-AKT cascade [41]. In the current study, CU upregulated IR, IRS-1 and IRS-2 mRNA expression.

The glucose transporter gene GLUT2 regulates the hepatocytes' ability to absorb and release glucose across the plasma membrane. It preserves the balance of glucose inside and outside liver cells. However, the expression of GLUT2 is drastically decreased in DM [42]. CU increased GLUT2 levels in the hepatocyte membrane by increasing mRNA expression.

CU upregulated PI3K and p-AKT signaling in liver tissue

Insulin initiates the PI3K/AKT signaling pathway by binding to insulin receptors on the cell membrane. The main mechanism of insulin signal transduction, which controls glucose uptake, glycogen formation and breakdown, is the PI3K/AKT signaling pathway [43]. Insulin binds to the α subunit of IR on liver cells, activating IRS in the process. Subsequently, IRS binds to PI3K's regulatory subunit, p85, and activates the catalytic subunit, p110. Phosphatidylinositol (3,4)-bisphosphate and phosphatidylinositol (3,4,5)P3, which are produced by activated PI3K, promote the activation of AKT [44].

Activated AKT results in the following: Extracellular glucose is more easily transported into cells when active AKT encourages the translocation of glucose transporter 4 to cell membranes. In addition, it facilitates the synthesis of glycogen and inhibits hepatic gluconeogenesis. Thus, the control of the liver's glucose metabolism is mostly dependent on the insulin-PI3K/AKT signaling pathway. CU effectively ameliorated STZ-induced downregulation of p-PI3K and AKT.

CU retrieved histopathological changes in liver tissues

Previous findings showed that STZ causes histological changes in the liver of diabetic mice. Liver cells of the STZ diabetic animals after 4 weeks from injection displayed severe congestion, necrotic foci, hydropic changes and aggregation of lymphocytes between the hepatocytes, among other more advanced abnormalities. Furthermore, after six weeks of STZ injection, mononuclear inflammatory cell infiltration, severe hydropic degeneration alterations and kupffer cell hyperplasia can be identified [45]. CU retrieved histopathological changes caused by STZ through inhibition of inflammatory cell infiltration and hyperplasia as well as CU limited degeneration of the hepatocytes, showing apparently normal hepatic parenchyma at 400 mg/Kg dose.

Different phenolic acids were quantified from C. uvifera aqueous, ethanolic and acetone leaves extracts such as syringic, ferulic, gallic, o/p-coumaric, ellagic, caffeic acids and others [12]. In the following sections, the antidiabetic activity of these constituents will be discussed. It is reported that high consumption of polyphenols may reduce the risk of diabetes incidence, control the postprandial glycemia and prevent the onset of glucose intolerance. These effects are mediated by facilitating the insulin response and attenuating the release of glucose-dependent insulinotropic polypeptide and glucagon-like peptide-1 [46].

Gallic acid (GA) was quantified in CU with a concentration of 0.09 mg/g extract, and also, it was isolated from C. uvifera leaf extract [47]. A study showed that GA increased glucose uptake by promoting the translocating of GLUT4 to the plasma membrane of rat adipocytes in a dose-dependent manner. Indeed, authors had concluded that the antidiabetic activity of mulberry leaves could be attributed to its gallic acid content [48]. Interestingly, GA alleviated DM detrimental effects in high-fat diet-fed streptozotocin-induced insulin resistance in diabetic rats. GA-treated rats had lower body weight gain, lower fasting blood glucose and lower insulin resistance [49]. Furthermore, the levels of glycogen content and the activity of glucose-6-phosphatase, fructose-1,6-bisphosphatase and hexokinase were dramatically restored to almost normal levels. These beneficial effects were mediated by increased expression of PPARγ (peroxisome proliferator-activated receptorγ) in adipocytes; consequently, GLUT4 was translocated and activated in PI3K- p-AKT dependent pathway [49]. Molecular docking showed that GA had promising interaction against GLUT4, GLUT1, PI3K and p-AKT. Moreover, histological examination revealed consistent distributions of pancreatic islets, adipose and hepatic cells with seemingly normal structures [49, 50]. Moreover, a study evaluated the antidiabetic activity of different hydroxybenzoic acid derivatives. GA suppressed the overexpression of microRNA-1271 generated by free fatty acids and upregulated its targets, such as p-IRS, p-PI3K, p-AKT and p-FOXO1, accompanied with the modulation of glucose metabolism genes [51].

GA was also found to be effective in clinical trials. In single-cell gel electrophoresis assays, oxidized purines are significantly reduced by 31% and pyrimidines by 2% after administration of GA, 15 mg for 7 days for patients of type 2 diabetes [52]. Moreover, following the intervention, the plasma concentrations of C-reactive protein and oxidized LDL were decreased by 39% and 24%, respectively. Consequently, a small quantity of GA (within the daily consumption range in Central Europe) lowers markers reflecting inflammation and elevated risks of CVD and cancer, as well as preventing oxidative DNA damage [52].

Several phenolic acids ameliorated DM manifestations. HbA1c and fasting plasma insulin levels were dramatically reduced by syringic acid, with improvement in liver glucose homeostasis [53, 54] Furthermore, ferulic acid (FA) has been demonstrated to increase the levels of insulin, glycogen and glucokinase, as well as lower the enzymes glucose-6-phosphatase and phosphoenolpyruvate carboxykinase in mice fed on high-fat diet [55]. Furthermore, FA was reported to regulate the gene expression of GLUT2 in the liver of diabetic rats [56]. Moreover, it raised the protein expression levels of IRS-1, PI3K and AKT in the muscles and brains of obese mice treated with FA, which could relieve obesity-related insulin-resistant case [57].

Ellagic acid (EA) enhanced the expression of IRS-1, AKT and ERK in HepG2 cells exposed to high glucose levels; simultaneously, EA downregulated the MDA level, counteracted oxidative stress and increased the glucose consumption by cells [58]. In DM-type 2 rats, EA raised GSH levels while suppressing serum MDA, TNF-α and IL-6. It also decreased liver enzymes ALT, AST and blood glucose [59].

Furthermore, caffeic acid caused upregulation of the expression of IRS-1, AKT, PI3K and GLUT4 [60]. In another study, caffeic acid lowered plasma glucose and glucose-6-phosphatase while increasing body weight and plasma insulin in diabetic rats [61]. Protocatechuic acid found in CU extract [47] has been shown to be able to counteract insulin resistance in obese volunteers by raising the levels of p-Tyr-IRS-1 and p-AKT in the visceral adipose tissue. Furthermore, protocatechuic acid reduced the inflammation and PTP1B activity [62]. In addition to lowering blood glucose levels and gluconeogenic enzymes, p-coumaric acid was reported to modify lipid and glucose metabolism via activating GLUT2.

Flavonoids are ubiquitous phenolic compounds in plants and their beverage products. Numerous in vitro and animal studies support that dietary flavonoids positively impact glucose homeostasis. In addition, through a variety of intracellular signaling mechanisms, flavonoids have been demonstrated to control carbohydrate digestion, insulin secretion, insulin signaling and glucose uptake in insulin-sensitive tissues [63]. Several flavonoids were isolated from C. uvifera leaves extract such as quercetin, myricetin and kaempferol derivatives [47, 64]. Their impact on DM and its complications would be displayed in the following paragraphs.

Rutin was quantified in this study in CU with a concentration of 0.23mg/g extract. A recent review explored the diverse mechanisms of action of rutin against DM. First, rutin reduces glucose level by reducing its absorption from the intestine, increasing the tissue glucose uptake, reducing gluconeogenesis, increasing insulin secretion and protecting islets of Langerhans against deterioration [65]. Moreover, rutin protects against DM complications by reducing sorbitol accumulation, reactive oxygen species, advanced glycation end-products and inflammatory cytokines [66]. Rutin was reported to increase the liver's antioxidant status by raising catalase, glutathione peroxidase and superoxide dismutase levels [67]. It also reduced serum levels of liver enzymes and corrected the histological damage to hepatocytes [68]. Rutin-induced insulin receptor kinase activity and GLUT4 translocation in differentiated myotubes enhance glucose uptake [69]. Furthermore, querectin, isoquercetin and rutin had α-glucosidase inhibiting activity [70].

In STZ diabetic rats treated with rutin (100 mg/kg), there was a decrease in plasma glucose and a rise in insulin levels, as well as a restoration of glycogen content and the activity of carbohydrate metabolic enzymes. The pancreas' histological examination demonstrated rutin's protective function. Meanwhile, the islets became larger, and the fatty infiltration of the islets decreased. [71]. In another study, flavonoids extract from mulberry leaves containing rutin as the main ingredient (1mg/mL) remarkably increased the protein expression levels of p-IRS-1, p-PI3K, p-AKT, total GLUT4 and membrane GLUT4 in 3T3-L1 adipocytes insulin resistance model [72]. Moreover, it has been shown that rutin (23 µg/mL) reversed the high glucose-induced insulin resistance caused in hepatic FL83B cells via enhancing AKT phosphorylation, which thereby enhanced GLUT2 translocation and glucose uptake [73].

Rutin was effective in clinical trials. Consuming 1g of rutin in patients with type 2 diabetes mellitus resulted in a significant decrease in heart rates, mean arterial pressure, pulse pressure and blood pressure. Moreover, there is a substantial rise in the antioxidant enzymes catalase, glutathione peroxidase and superoxide dismutase as well as quality of life (QOL) parameters (emotional limitations, mental health, energy and freshness, social performance and general health) [74].

Rutin was effective when combined with other oral synthetic antidiabetics. When given separately, rutin effectively decreased hyperglycemia in acute assays in a way comparable to oral antidiabetic drugs (OADs). In the subchronic assay, rutin also helped to lower the HbA1c% and hyperlipidemia. In all treatments, rutin and OADs effectively reduced hyperglycemia, as seen by the decline in the percentage of HbA1c and lipid profile. consequently, rutin showed significant activity when combined with antidiabetic medications, which is a first step toward creating novel DM treatments [75].

Rutin and quercetin increased glucose uptake in insulin-resistant FL83B liver hepatocytes. This effect was mediated by the upregulation of p-AKT and GLUT2, reduction in oxidative stress and prevention of the degradation of PPARγ [76]. Quercetin aglycone and its derivatives (quercetin-3-O-glucoside and quercetin-3-O-galactoside) isolated from berry extract, enhanced insulin-independent glucose uptake and stimulated AMPK in muscle cells [77]. Over a period of 28 days, quercetin at dosages of 25 and 50 mg/kg significantly decreased blood glucose and glycosylated hemoglobin (Hb), while increasing the levels of Hb in plasma and hepatic glycogen. After receiving quercetin, the hexokinase and glucose-6-phosphatase activity in diabetic rats were also markedly restored [78].

When isoquercetin (quercetin-3-O-glucoside) was supplemented to STZ-induced diabetic rats, the insulin levels increased significantly, and the glucose levels returned to normal. Additionally, liver enzymes were reduced. Glycogen synthase GK and GLUT2 expressions were markedly upregulated, while glucose-6-phosphatase expressions were significantly downregulated. The levels of gluconeogenesis enzymes were significantly lower than those in negative control. Insulin, IR, IRS-1, IRS-2 and AKT mRNA expressions were also elevated. Indeed, isoquercetin efficacy was similar to that of glibenclamide [79].

Administration of kaempferol (K) and its glycosides rich fraction decreased the area under the curve in glucose tolerance test in genetically type 2 KK-Ay mice. Moreover, the liver had lower triglycerides level and fatty acid synthase activity [80]. By controlling mitochondrial calcium absorption, K stimulates Akt activation and enhances insulin production. Moreover, K directly restores AKT activation. Subsequently, reversing the effects of AKT inactivation causes the upregulation of gluconeogenesis, the downregulation of glycogen synthesis and the uptake of glucose. Additionally, the antioxidant K controls both apoptosis and autophagy [81].

Myricetin caused upregulation of p-IR, p-IRS-1 and p-AKT in the liver of high-fat diet-fed and STZ-induced type 2 diabetic rats. These effects were mediated by suppressing PTP1B's activity and expression; PTP1B is the tyrosine phosphatase that adversely regulates insulin signal transduction. These effects were evident when myricetin was administered alone and not in combination with horsegram protein [82]. Moreover, hexokinase, glycogen synthase, glycogen phosphorylase, glycosylated hemoglobin, glucose-6-phosphatase and plasma glucose were all inhibited from a significant increase [83]. It has been determined that myricitrin, glycosylated myricetin, activates the IRS-1/PI3K/AKT/GLUT4 pathway in the soleus muscle of type 2 DM-affected rats as well as in L6 muscle cells exposed to high glucose [84].

Conclusion

The current study provided an unequivocal experimental validation of the antidiabetic activity of standardized CU. Moreover, it deciphered the molecular mechanism of antidiabetic and hepatoprotective actions of CU for the first time. CU had ameliorated DM detrimental impact. The underlying mechanism involved the upregulation and induction of IRS isomers, GLUT2 and their upstream PI3K/AKT signaling pathway. GA and rutin were the major and key bioactive constituents of CU. Both could serve as QC markers in future analyses. As a result, C. uvifera leaves extract could be used as a promising antidiabetic drug with hepatoprotective activity in diabetes hepatic complications. The current study laid the foundation for performing further assessment and clinical testing of the antidiabetic activity CU alone or in combination with other remedies.

Availability of data and materials

All data generated or analyzed during this study are included in this published article.

Abbreviations

DM:

Diabetes mellitus

CU:

Ethanolic Coccoloba uvifera L. leaves extract

HPLC:

High-performance liquid chromatography

STZ:

Streptozotocin

AST:

Serum aspartate aminotransferase

ALT:

Alanine aminotransferase

MDA:

Liver malondialdehyde

GSH:

Reduced glutathione

RT-PCR:

Reverse transcription polymerase chain reaction

IR:

Insulin receptor

IRS-1:

Insulin receptor substrate-1

IRS-2:

Insulin receptor substrate-2

GLUT2:

Glucose transporter 2

GLUT4:

Glucose transporter 4

PI3K:

Phosphatidylinositol 3-kinase

p-AKT:

Phosphorylated protein kinase B

PPARγ:

Peroxisome proliferator-activated receptor γ

HepG2:

Hepatoblastoma cell line

ERK:

Extracellular signal-regulated kinase

TNF-α:

Tumor necrosis factor alpha

Il-6:

Interleukin-6

PTP1B:

Protein-tyrosine phosphatase 1B

FOXO1:

Forkhead box protein O1

References:

  1. Saeedi P, Petersohn I, Salpea P, Malanda B, Karuranga S, Unwin N, Colagiuri S, Guariguata L, Motala AA, Ogurtsova K (2019) Global and regional diabetes prevalence estimates for 2019 and projections for 2030 and 2045: Results from the International Diabetes Federation Diabetes Atlas. Diabetes Res Clin Pract 157:107843. https://doi.org/10.1016/j.diabres.2019.107843

    Article  PubMed  Google Scholar 

  2. Alanazi AZ, Alqahtani F, Mothana RA, Mohany M, Abuohashish HM, Ahmed MM, Al-Rejaie SS (2020) Protective role of Loranthus regularis against liver dysfunction, inflammation, and oxidative stress in streptozotocin diabetic rat model. Evid Based Complement Alternat Med. https://doi.org/10.1155/2020/5027986

    Article  PubMed  PubMed Central  Google Scholar 

  3. Al-Ghulikah HA, Mughal EU, Elkaeed EB, Naeem N, Nazir Y, Alzahrani AYA, Sadiq A, Shah SWA (2023) Discovery of chalcone derivatives as potential α-glucosidase and cholinesterase inhibitors: effect of hyperglycemia in paving a path to dementia. J Mol Struct 1275:134658. https://doi.org/10.1016/j.molstruc.2022.134658

    Article  CAS  Google Scholar 

  4. Levinthal GN, Tavill ASJCD (1999) Liver disease and diabetes mellitus. Clin Diabetes 17:73–74

    Google Scholar 

  5. Mohamed J, Nafizah AN, Zariyantey A, Budin S (2016) Mechanisms of diabetes-induced liver damage: the role of oxidative stress and inflammation. Sultan Qaboos Univ Med J 16:132. https://doi.org/10.18295/squmj.2016.16.02.002

    Article  Google Scholar 

  6. Bugianesi E, Mccullough AJ, Marchesini GJH (2005) Insulin resistance: a metabolic pathway to chronic liver disease. Hepatol 42:987–1000. https://doi.org/10.1002/hep.20920

    Article  CAS  Google Scholar 

  7. Crawford J (2005) The liver and the biliary tract. In: Kumar V, Abbas AK, Fausto N (eds) Robbins and Cotran pathologic basis of disease. Elsevier Saunders, Philadelphia; 19106, pp 877–938. https://doi.org/10.1016/b978-1-4377-0792-2.50023-7

  8. Choudhury H, Pandey M, Hua CK, Mun CS, Jing JK, Kong L, Ern LY, Ashraf NA, Kit SW, Yee TSJJOT, Medicine C (2018) An update on natural compounds in the remedy of diabetes mellitus: a systematic review. J Tradit Complement Med 8:361–376. https://doi.org/10.1016/j.jtcme.2017.08.012

    Article  PubMed  Google Scholar 

  9. Kasole R, Martin HD, Kimiywe J (2019) Traditional medicine and its role in the management of diabetes mellitus:“patients’ and herbalists’ perspectives”. Evid Based Complement Alternat Med. https://doi.org/10.1155/2019/2835691

    Article  PubMed  PubMed Central  Google Scholar 

  10. Põlme S, Bahram M, Kõljalg U, Tedersoo L (2017) Biogeography and specificity of ectomycorrhizal fungi of Coccoloba uvifera, in Biogeography of mycorrhizal symbiosis 345–359. https://doi.org/10.1007/978-3-319-56363-3_16

  11. Lim T (2013) Coccoloba uvifera. Edible medicinal and non-medicinal plants: Volume 5, Fruits. Springer, Dordrecht, pp 455-458. https://doi.org/10.1007/978-94-007-5653-3_24

  12. Ashmawy NA, Salem MZ, El Shanhorey N, Al-Huqail A, Ali HM, Behiry SI (2020) Eco-friendly wood-biofungicidal and antibacterial activities of various Coccoloba uvifera L. leaf extracts: HPLC analysis of phenolic and flavonoid compound. BioResources 15:4165–4187. https://doi.org/10.15376/biores.15.2.4165-4187

    Article  CAS  Google Scholar 

  13. Kaewpiboon C, Lirdprapamongkol K, Srisomsap C, Winayanuwattikun P, Yongvanich T, Puwaprisirisan P, Svasti J, Assavalapsakul W (2012) Studies of the in vitro cytotoxic, antioxidant, lipase inhibitory and antimicrobial activities of selected Thai medicinal plants. BMC Complement Altern Med 12:1–8. https://doi.org/10.1186/1472-6882-12-217

    Article  Google Scholar 

  14. Povi L-E, Batomayena B, Hode TA, Kwashie E-G, Kodjo A, Messanvi G (2015) Phytochemical screening, antioxidant and hypoglycemic activity of Coccoloba uvifera leaves and Waltheria indica roots extracts. Int J Pharm Pharm Sci 279–283

  15. Rodríguez-García CM, Ruiz-Ruiz JC, Peraza-Echeverría L, Peraza-Sánchez SR, Torres-Tapia LW, Pérez-Brito D, Tapia-Tussell R, Herrera-Chalé FG, Segura-Campos MR, Quijano-Ramayo A (2019) Antioxidant, antihypertensive, anti-hyperglycemic, and antimicrobial activity of aqueous extracts from twelve native plants of the Yucatan coast. PLoS ONE 14:e0213493. https://doi.org/10.1371/journal.pone.0213493

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Buckley W (2000) Method of controlling blood sugar levels using Coccoloba uvifera. US Patent 6,103,242, 2000.

  17. Malathi S, Masilamani P, Balasubramanian V, Rao RB, Brindha P (1995) Constituents of Coccoloba uvifera leaves. Fitoterapia. https://doi.org/10.1079/cabicompendium.14655

    Article  Google Scholar 

  18. Segura Campos MR, Ruiz Ruiz J, Chel-Guerrero L, Betancur Ancona DJ (2015) Coccoloba uvifera (L)(Polygonaceae) fruit: phytochemical screening and potential antioxidant activity. J Chem. https://doi.org/10.1155/2015/534954

    Article  Google Scholar 

  19. Vinayagam R, Jayachandran M, Xu B (2016) Antidiabetic effects of simple phenolic acids: a comprehensive review. Phytother Res 30:184–199. https://doi.org/10.1002/ptr.5528

    Article  CAS  PubMed  Google Scholar 

  20. Ainsworth EA, Gillespie KM (2007) Estimation of total phenolic content and other oxidation substrates in plant tissues using Folin–Ciocalteu reagent. Nat Protoc 2:875–877. https://doi.org/10.1038/nprot.2007.102

    Article  CAS  PubMed  Google Scholar 

  21. Pękal A, Pyrzynska K (2014) Evaluation of aluminium complexation reaction for flavonoid content assay. Food Anal Methods 7:1776–1782. https://doi.org/10.1007/s12161-014-9814-x

    Article  Google Scholar 

  22. Wadie W, El-Tanbouly DM (2017) Vinpocetine mitigates proteinuria and podocytes injury in a rat model of diabetic nephropathy. Eur J Pharmacol 814:187–195

    Article  CAS  PubMed  Google Scholar 

  23. Uchiyama M, Mihara M (1978) Determination of malonaldehyde precursor in tissues by thiobarbituric acid test. Anal Biochem 86:271–278. https://doi.org/10.1016/0003-2697(78)90342-1

    Article  CAS  PubMed  Google Scholar 

  24. Gl E (1959) Tissue sulfhydryl groups. Arch Biochem Biophys 82:70–77. https://doi.org/10.1016/0003-9861(59)90090-6

    Article  Google Scholar 

  25. Brandstrup N, Kirk J, Bruni C (1957) The hexokinase and phosphoglucoisomerase activities of aortic and pulmonary artery tissue in individuals of various ages. J Gerontol 12:166–171. https://doi.org/10.1093/geronj/12.2.166

    Article  CAS  PubMed  Google Scholar 

  26. Hikaru K, Toshitsugu OJCCA (1959) Pathological occurrence of glucose-6-phosphatase in serum in liver diseases. Clin Chim Acta 4:554–561. https://doi.org/10.1016/0009-8981(59)90165-2

    Article  Google Scholar 

  27. Gancedo JM, Gancedo C (1971) Fructose-1, 6-diphosphatase, phosphofructokinase and glucose-6-phosphate dehydrogenase from fermenting and non fermenting yeasts. Arch Microbiol 76:132–138. https://doi.org/10.1007/bf00411787

    Article  CAS  Google Scholar 

  28. Morales M, Jobaggy A, Terenzi H (1973) Mutations affecting accumulation of glycogen. Fungal Genet Rep 20:22. https://doi.org/10.4148/1941-4765.1830

    Article  Google Scholar 

  29. Ibrahim KE, Al-Mutary MG, Bakhiet AO, H K (2018) Histopathology of the liver, kidney, and spleen of mice exposed to gold nanoparticles. Molecules 23:1848. https://doi.org/10.3390/molecules23081848

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Members WG, Roger VL, Go AS, Lloyd-Jones DM, Benjamin EJ, Berry JD, Borden WB, Bravata DM, Dai S, Ford ES (2012) Heart disease and stroke statistics—2012 update: a report from the American Heart Association. Circulation 125:e2–e220. https://doi.org/10.1161/cir.0b013e31823ac046

    Article  Google Scholar 

  31. Scheen AJ (2014) Pharmacokinetics in patients with chronic liver disease and hepatic safety of incretin-based therapies for the management of type 2 diabetes mellitus. Clin Pharmacokinet 53:773–785. https://doi.org/10.1007/s40262-014-0157-y

    Article  CAS  PubMed  Google Scholar 

  32. Tan BKH, Ong KW (2014) Influence of dietary polyphenols on carbohydrate metabolism. Polyphenols in Human Health Disease. https://doi.org/10.1016/b978-0-12-398456-2.00009-8

    Article  Google Scholar 

  33. Noroozi Karimabad M, Khalili P, Ayoobi F, Esmaeili-Nadimi A, La Vecchia C, Jamali Z (2022) Serum liver enzymes and diabetes from the Rafsanjan cohort study. BMC Endocr Disord 22:127. https://doi.org/10.1186/s12902-022-01042-2

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Lucchesi AN, Freitas NTD, Cassettari LL, Marques SFG, Spadella CT (2013) Diabetes mellitus triggers oxidative stress in the liver of alloxan-treated rats: a mechanism for diabetic chronic liver disease. Acta Cir Bras 28:502–508. https://doi.org/10.1590/s0102-86502013000700005

    Article  PubMed  Google Scholar 

  35. Langdon DR, Curnow RT (1983) Impaired glycogenic substrate activation of glycogen synthase is associated with depressed synthase phosphatase activity in diabetic rat liver. Diabetes 32:1134–1140. https://doi.org/10.2337/diabetes.32.12.1134

    Article  CAS  PubMed  Google Scholar 

  36. Murphy ED, Anderson JW (1974) Tissue glycolytic and gluconeogenic enzyme activities in mildly and moderately diabetic rats: influence of tolbutamide administration. Endocrinology 94:27–34. https://doi.org/10.1210/endo-94-1-27

    Article  CAS  PubMed  Google Scholar 

  37. Wu C, Khan SA, Peng L-J, and Lange AJJaIER (2006) Roles for fructose-2, 6-bisphosphate in the control of fuel metabolism: beyond its allosteric effects on glycolytic and gluconeogenic enzymes Advances in enzyme regulation 46: 72–88. https://doi.org/10.1016/j.advenzreg.2006.01.010

  38. Rondinone CM, Wang L-M, Lonnroth P, Wesslau C, Pierce JH, Smith U (1997) Insulin receptor substrate (IRS) 1 is reduced and IRS-2 is the main docking protein for phosphatidylinositol 3-kinase in adipocytes from subjects with non-insulin-dependent diabetes mellitus. Proc Natl Acad Sci 94:4171–4175. https://doi.org/10.1073/pnas.94.8.4171

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Dong X, Park S, Lin X, Copps K, Yi X, White MF (2006) Irs1 and Irs2 signaling is essential for hepatic glucose homeostasis and systemic growth. J Clin Invest 116:101–114. https://doi.org/10.1172/jci25735

    Article  CAS  PubMed  Google Scholar 

  40. Mccarthy AM, Elmendorf JS (2007) GLUT4’s itinerary in health & disease. Indian J Med Res 125:373–388

    CAS  PubMed  Google Scholar 

  41. Rother KI, Imai Y, Caruso M, Beguinot F, Formisano P, Accili D (1998) Evidence that IRS-2 phosphorylation is required for insulin action in hepatocytes. J Biol Chem 273:17491–17497. https://doi.org/10.1074/jbc.273.28.17491

    Article  CAS  PubMed  Google Scholar 

  42. Takayama H, Ohta M, Tada K, Watanabe K, Kawasaki T, Endo Y, Iwashita Y, Inomata M (2019) Additional effects of duodenojejunal bypass on glucose metabolism in a rat model of sleeve gastrectomy. Surg Today 49:637–644. https://doi.org/10.1007/s00595-019-1772-x

    Article  CAS  PubMed  Google Scholar 

  43. Gao Y-F, Zhang M-N, Wang T-X, Wu T-C, Ai R-D, Zhang Z-S (2016) Hypoglycemic effect of D-chiro-inositol in type 2 diabetes mellitus rats through the PI3K/Akt signaling pathway. Mol Cell Endocrinol 433:26–34. https://doi.org/10.1016/j.mce.2016.05.013

    Article  CAS  PubMed  Google Scholar 

  44. Hou X-LJCT, Drugs H (2015) Research progress in pharmacological effects of dihydromyricelin. Chinese Traditional Herbal Drugs 603–609.

  45. Al-Ani IM, Al-Mishadani N, Muslih RK, Hamoodi SR (2009) Histological liver changes in streptozotocin induced diabetic mice. Int Med J Malays 8.

  46. Meng S, Cao J, Feng Q, Peng J, Hu Y (2013) Roles of chlorogenic acid on regulating glucose and lipids metabolism: a review. Evid Based Complement Alternat Med. https://doi.org/10.1155/2013/801457

    Article  PubMed  PubMed Central  Google Scholar 

  47. Abu El Wafa SA, Seif-Eldein NA, Anwar Aly Taie H, Marzouk M (2023) Coccoloba uvifera leaves: polyphenolic profile, cytotoxicity, and antioxidant evaluation. ACS Omega. https://doi.org/10.1021/acsomega.3c04025

    Article  PubMed  PubMed Central  Google Scholar 

  48. Naowaboot J, Pannangpetch P, Kukongviriyapan V, Prawan A, Kukongviriyapan U, Itharat A (2012) Mulberry leaf extract stimulates glucose uptake and GLUT4 translocation in rat adipocytes. Am J Chin Med 40:163–175. https://doi.org/10.1142/s0192415x12500139

    Article  CAS  PubMed  Google Scholar 

  49. Gandhi GR, Jothi G, Antony PJ, Balakrishna K, Paulraj MG, Ignacimuthu S, Stalin A, Al-Dhabi NA (2014) Gallic acid attenuates high-fat diet fed-streptozotocin-induced insulin resistance via partial agonism of PPARγ in experimental type 2 diabetic rats and enhances glucose uptake through translocation and activation of GLUT4 in PI3K/p-Akt signaling pathway. Eur J Pharmacol 745:201–216. https://doi.org/10.1016/j.ejphar.2014.10.044

    Article  CAS  PubMed  Google Scholar 

  50. Xu Y, Tang G, Zhang C, Wang N, Feng Y (2021) Gallic acid and diabetes mellitus: its association with oxidative stress. Molecules 26:7115. https://doi.org/10.3390/molecules26237115

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  51. Lee H, Lee J (2021) Anti-diabetic effect of hydroxybenzoic acid derivatives in free fatty acid-induced HepG2 cells via miR-1271/IRS1/PI3K/AKT/FOXO1 pathway. J Food Biochem 45:e13993. https://doi.org/10.1111/jfbc.13993

    Article  CAS  PubMed  Google Scholar 

  52. Ferk F, Kundi M, Brath H, Szekeres T, Al-Serori H, Mišík M, Saiko P, Marculescu R, Wagner KH, Knasmueller S (2018) Gallic acid improves health-associated biochemical parameters and prevents oxidative damage of DNA in type 2 diabetes patients: Results of a placebo-controlled pilot study. Mol Nutr Food Res 62:1700482. https://doi.org/10.1002/mnfr.201700482

    Article  CAS  Google Scholar 

  53. Muthukumaran J, Srinivasan S, Venkatesan RS, Ramachandran V, Muruganathan U (2013) Syringic acid, a novel natural phenolic acid, normalizes hyperglycemia with special reference to glycoprotein components in experimental diabetic rats. J Acute Dis 2:304–309. https://doi.org/10.1016/s2221-6189(13)60149-3

    Article  Google Scholar 

  54. Srinivasan M, Sudheer AR, Menon VP (2007) Ferulic acid: therapeutic potential through its antioxidant property. J Clin Biochem Nutr 40:92–100. https://doi.org/10.3164/jcbn.40.92

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  55. Son MJ, Rico CW, Nam SH, Kang MY (2011) Effect of oryzanol and ferulic acid on the glucose metabolism of mice fed with a high-fat diet. J Food Sci 76:H7–H10. https://doi.org/10.1111/j.1750-3841.2010.01907.x

    Article  CAS  PubMed  Google Scholar 

  56. Narasimhan A, Chinnaiyan M, Karundevi B (2015) Ferulic acid regulates hepatic GLUT2 gene expression in high fat and fructose-induced type-2 diabetic adult male rat. Eur J Pharmacol 761:391–397. https://doi.org/10.1016/j.ejphar.2015.04.043

    Article  CAS  PubMed  Google Scholar 

  57. Naowaboot J, Piyabhan P, Tingpej P, Munkong N, Parklak W, Pannangpetch P (2018) Anti-insulin resistant effect of ferulic acid on high fat diet-induced obese mice. Asian Pac J Trop 8:604–608. https://doi.org/10.4103/2221-1691.248098

    Article  Google Scholar 

  58. Ding X, Jian T, Wu Y, Zuo Y, Li J, Lv H, Ma L, Ren B, Zhao L, Li W (2019) Ellagic acid ameliorates oxidative stress and insulin resistance in high glucose-treated HepG2 cells via miR-223/keap1-Nrf2 pathway. Biomed Pharmacother 110:85–94. https://doi.org/10.1016/j.biopha.2018.11.018

    Article  CAS  PubMed  Google Scholar 

  59. Amin MM, Arbid MS (2017) Estimation of ellagic acid and/or repaglinide effects on insulin signaling, oxidative stress, and inflammatory mediators of liver, pancreas, adipose tissue, and brain in insulin resistant/type 2 diabetic rats. Appl Physiol Nutr Metab 42:181–192. https://doi.org/10.1139/apnm-2016-0429

    Article  CAS  PubMed  Google Scholar 

  60. Chen L, Teng H, Cao H (2019) Chlorogenic acid and caffeic acid from Sonchus oleraceus Linn synergistically attenuate insulin resistance and modulate glucose uptake in HepG2 cells. Food Chem Toxicol 127:182–187. https://doi.org/10.1016/j.fct.2019.03.038

    Article  CAS  PubMed  Google Scholar 

  61. Jung UJ, Lee M-K, Park YB, Jeon S-M, Choi M-S, Therapeutics E (2006) Antihyperglycemic and antioxidant properties of caffeic acid in db/db mice. J Pharmacol Exp Ther 318:476–483. https://doi.org/10.1124/jpet.106.105163

    Article  CAS  PubMed  Google Scholar 

  62. Ormazabal P, Scazzocchio B, Varì R, Santangelo C, D’archivio M, Silecchia G, Iacovelli A, Giovannini C, Masella R (2018) Effect of protocatechuic acid on insulin responsiveness and inflammation in visceral adipose tissue from obese individuals: possible role for PTP1B. Int J Obes 42:2012–2021. https://doi.org/10.1038/s41366-018-0075-4

    Article  CAS  Google Scholar 

  63. Hanhineva K, Törrönen R, Bondia-Pons I, Pekkinen J, Kolehmainen M, Mykkänen H, Poutanen K (2010) Impact of dietary polyphenols on carbohydrate metabolism. Int J Mol Sci 11:1365–1402. https://doi.org/10.3390/ijms11041365

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  64. Kawasaki M, Kanomata T, Yoshitama K (1986) Flavonoids in the leaves of twenty-eight polygonaceous plants. Shokubutsugaku-zasshi 99:63–74. https://doi.org/10.1007/bf02488623

    Article  CAS  Google Scholar 

  65. Ahmed OM, Moneim AA, Yazid IA, Mahmoud AM (2010) Antihyperglycemic, antihyperlipidemic and antioxidant effects and the probable mechanisms of action of Ruta graveolens infusion and rutin in nicotinamide-streptozotocin-induced diabetic rats. Diabetol Croat. https://doi.org/10.2174/1871529x22666220806122012

    Article  Google Scholar 

  66. Nagasawa T, Tabata N, Ito Y, Aiba Y, Nishizawa N, Kitts DD (2003) Dietary G-rutin suppresses glycation in tissue proteins of streptozotocin-induced diabetic rats. Mol Cell Biochem 252:141–147. https://doi.org/10.1023/a:1025563519088

    Article  CAS  PubMed  Google Scholar 

  67. Niture NT, Ansari AA, Naik SR (2014) Anti-hyperglycemic activity of rutin in streptozotocin-induced diabetic rats: an effect mediated through cytokines, antioxidants and lipid biomarkers. Indian J Med Res 52(7):720–727

    Google Scholar 

  68. Ghorbani A (2017) Mechanisms of antidiabetic effects of flavonoid rutin. Biomed Pharmacother 96:305–312. https://doi.org/10.1016/j.biopha.2017.10.001

    Article  CAS  PubMed  Google Scholar 

  69. Hsu CY, Shih HY, Chia YC, Lee CH, Ashida H, Lai YK, Weng CF (2014) Rutin potentiates insulin receptor kinase to enhance insulin-dependent glucose transporter 4 translocation. Mol Nutr Food Res 58:1168–1176. https://doi.org/10.1002/mnfr.201300691

    Article  CAS  PubMed  Google Scholar 

  70. Li YQ, Zhou FC, Gao F, Bian JS, Shan F (2009) Comparative evaluation of quercetin, isoquercetin and rutin as inhibitors of α-glucosidase. J Agric Food Chem 57:11463–11468. https://doi.org/10.1021/jf903083h

    Article  CAS  PubMed  Google Scholar 

  71. Prince PSM, Kamalakkannan N (2006) Rutin improves glucose homeostasis in streptozotocin diabetic tissues by altering glycolytic and gluconeogenic enzymes. J Biochem Mol Toxicol 20:96–102. https://doi.org/10.1002/jbt.20117

    Article  CAS  Google Scholar 

  72. Meng Q, Qi X, Chao Y, Chen Q, Cheng P, Yu X, Kuai M, Wu J, Li W, Zhang Q (2020) IRS1/PI3K/AKT pathway signal involved in the regulation of glycolipid metabolic abnormalities by Mulberry (Morus alba L.) leaf extracts in 3T3-L1 adipocytes. Chin Med 15:1–11. https://doi.org/10.1186/s13020-019-0281-6

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  73. Martín MÁ, Ramos S (2021) Dietary flavonoids and insulin signaling in diabetes and obesity. Cells 10:1474. https://doi.org/10.3390/cells10061474

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  74. Bazyar H, Javid AZ, Ahangarpour A, Zaman F, Hosseini SA, Zohoori V, Aghamohammadi V, Yazdanfar S, Cheshmeh MGD (2023) The effects of rutin supplement on blood pressure markers, some serum antioxidant enzymes, and quality of life in patients with type 2 diabetes mellitus compared with placebo. Front Nutr. https://doi.org/10.3389/fnut.2023.1214420

    Article  PubMed  PubMed Central  Google Scholar 

  75. Valdes M, Calzada F, Martínez-Solís J, Martínez-Rodríguez J (2023) Antihyperglycemic effects of annona cherimola miller and the flavonoid rutin in combination with oral antidiabetic drugs on streptozocin-induced diabetic mice. Pharmaceuticals 16:112. https://doi.org/10.3390/ph16010112

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  76. Lee C-C, Hsu W-H, Shen S-R, Cheng Y-H, Wu S-C (2012) Fagopyrum tataricum (buckwheat) improved high-glucose-induced insulin resistance in mouse hepatocytes and diabetes in fructose-rich diet-induced mice. Exp Diabetes Res. https://doi.org/10.1155/2012/375673

    Article  PubMed  PubMed Central  Google Scholar 

  77. Eid HM, Martineau LC, Saleem A, Muhammad A, Vallerand D, Benhaddou-Andaloussi A, Nistor L, Afshar A, Arnason JT, Haddad PS (2010) Stimulation of AMP-activated protein kinase and enhancement of basal glucose uptake in muscle cells by quercetin and quercetin glycosides, active principles of the antidiabetic medicinal plant Vaccinium vitis-idaea. Mol Nutr Food Res 54:991–1003. https://doi.org/10.1002/mnfr.200900218

    Article  CAS  PubMed  Google Scholar 

  78. Oyedemi SO, Nwaogu G, Chukwuma CI, Adeyemi OT, Matsabisa MG, Swain SS, Aiyegoro OA (2020) Quercetin modulates hyperglycemia by improving the pancreatic antioxidant status and enzymes activities linked with glucose metabolism in type 2 diabetes model of rats: In silico studies of molecular interaction of quercetin with hexokinase and catalase. J Food Biochem 44:e13127. https://doi.org/10.1111/jfbc.13127

    Article  PubMed  Google Scholar 

  79. Jayachandran M, Zhang T, Ganesan K, Xu B, Chung SSM (2018) Isoquercetin ameliorates hyperglycemia and regulates key enzymes of glucose metabolism via insulin signaling pathway in streptozotocin-induced diabetic rats. Eur J Pharmacol 829:112–120. https://doi.org/10.1016/j.ejphar.2018.04.015

    Article  CAS  PubMed  Google Scholar 

  80. Zang Y, Sato H, Igarashi KJB (2011) Anti-diabetic effects of a kaempferol glycoside-rich fraction from unripe soybean (Edamame, Glycine max L. Merrill. ‘Jindai’) leaves on KK-Ay mice. Biosci Biotechnol Biochem 75:1677–1684. https://doi.org/10.1271/bbb.110168

    Article  CAS  PubMed  Google Scholar 

  81. Yang Y, Chen Z, Zhao X, Xie H, Du L, Gao H, Xie C (2022) Mechanisms of Kaempferol in the treatment of diabetes: A comprehensive and latest review. Front Endocrinol 13:990299. https://doi.org/10.3389/fendo.2022.990299

    Article  Google Scholar 

  82. Lalitha N, Sadashivaiah B, Talahalli RR, Singh S (2020) Lectin rich horsegram protein and myricetin activates insulin signaling–a study targeting PTP1β. J Funct Foods 67:103845. https://doi.org/10.1016/j.jff.2020.103845

    Article  CAS  Google Scholar 

  83. Kandasamy N, Ashokkumar NJT, Pharmacology A (2014) Protective effect of bioflavonoid myricetin enhances carbohydrate metabolic enzymes and insulin signaling molecules in streptozotocin–cadmium induced diabetic nephrotoxic rats. Toxicol Appl Pharmacol 279:173–185. https://doi.org/10.1016/j.taap.2014.05.014

    Article  CAS  PubMed  Google Scholar 

  84. Dua TK, Joardar S, Chakraborty P, Bhowmick S, Saha A, De Feo V, Dewanjee S (2021) Myricitrin, a glycosyloxyflavone in Myrica esculenta bark ameliorates diabetic nephropathy via improving glycemic status, reducing oxidative stress, and suppressing inflammation. Molecules 26:258. https://doi.org/10.3390/molecules26020258

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

Not applicable

Statement regarding plants

Coccoloba uvifera L., was authenticated by Mrs. Therese Labib, Botanical Specialist and consultant at Orman and Qubba Botanical Gardens. A voucher sample was kept at the Herbarium of the Department of Pharmacognosy, Faculty of Pharmacy, Cairo University number (26.7.23).

Funding

This research did not receive any specific grant from funding agencies in the public, commercial or not-for-profit sectors.

Author information

Authors and Affiliations

Authors

Contributions

Fatma Mohamed was involved in writing the original draft, methodology, formal analysis, data curation and conceptualization. Rabab Sayed contributed to biological activity, statistical analysis and writing the manuscript. Mohamed Khalil was responsible for original draft, reviewing, data analysis, chemical investigation and study design. Mohamed Salem took part in data analysis, writing-reviewing and editing, and study design. Amira El Senousy participated in data analysis, writing-reviewing and study design. Ali El-Halawany helped with supervising work and study design.

Corresponding author

Correspondence to Ali M. El-Halawany.

Ethics declarations

Ethics approval and consent to participate

The study protocol was approved by the Ethics Committee for Animal Experimentation at Faculty of Pharmacy, Cairo University MP (2964) 28/03/2022 and followed the guidelines of the US National Institutes of Health guide for the Care and Use of Laboratory Animals (NIH Publication No. 85-23, revised 2011). All protocols were followed to minimize the animals’ suffering.

Consent for publication

Not Applicable.

Competing interests

The authors declare that we have no known competing financial interests or personal relationships that could have appeared to influence the work reported in this paper.

Additional information

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Supplementary Information

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Mohamed, F.A., Sayed, R.H., Khalil, M.N.A. et al. Ameliorative activity of standardized Coccoloba uvifera leaves extract against streptozotocin-induced diabetic rats via activation of IRS-1/PI3K/AKT/GLUT2 pathway in liver. Futur J Pharm Sci 10, 132 (2024). https://doi.org/10.1186/s43094-024-00707-0

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s43094-024-00707-0

Keywords