Protective effects of Ficus carica leaves on glucose and lipids levels, carbohydrate metabolism enzymes and β-cells in type 2 diabetic rats.
Journal: 2017/March - Pharmaceutical Biology
ISSN: 1744-5116
Abstract:
BACKGROUND
The decoctions of Ficus carica Linn. (Moraceae) leaves are used in the folklore treatment of diabetes.
OBJECTIVE
To evaluate the effect of F. carica on glucose and lipids levels, carbohydrate metabolism enzymes and β-cells protective effects in type 2 diabetes.
METHODS
Diabetes was induced in 15 days high-fat diet (HFD)-fed Wistar rats by intraperitoneal injection of streptozotocin (STZ) (40 mg/kg). The ethyl acetate extract (250 and 500 mg/kg) of F. carica leaves was administered for 28 days. Oral glucose tolerance (OGTT) and intraperitoneal insulin tolerance tests (ITT) were evaluated on 15th and 25th days, respectively.
RESULTS
The ethyl acetate extract (250 and 500 mg/kg) of n F. carica leaves showed significant effect (p < 0.005) in the levels of blood glucose, total cholesterol (TC), triglycerides (TG), body weight and hepatic glycogen. In OGTT, F. carica (250 and 500 mg/kg) significantly (p < 0.005) detained the increase in blood glucose levels at 60 and 120 min and in ITT, F. carica enhanced the glucose utilization significantly (p < 0.005) over 30 and 60 min compared to diabetic control. Further, the altered activities of key carbohydrate metabolizing enzymes such as glucose-6-phosphatase, fructose-1,6-bisphosphatase and hexokinase in the liver tissue of diabetic rats were significantly (p < 0.005) reverted to near normal levels upon treatment with F. carica. Immumohistochemical studies of islets substantiated the cytoprotective effect on pancreatic β-cells.
CONCLUSIONS
F. carica leaves exerted significant effect on carbohydrate metabolism enzymes with promising hypoglycemic and hypolipidemic activities in type 2 diabetic rats.
Relations:
Content
Citations
(2)
Diseases
(3)
Drugs
(3)
Chemicals
(8)
Organisms
(4)
Processes
(3)
Anatomy
(3)
Affiliates
(2)
Similar articles
Articles by the same authors
Discussion board
Pharmaceutical Biology. Dec/31/2016; 55(1): 1074-1081
Published online Feb/12/2017

Protective effects of Ficus carica leaves on glucose and lipids levels, carbohydrate metabolism enzymes and β-cells in type 2 diabetic rats

Abstract

Context: The decoctions of Ficus carica Linn. (Moraceae) leaves are used in the folklore treatment of diabetes.

Objective: To evaluate the effect of F. carica on glucose and lipids levels, carbohydrate metabolism enzymes and β-cells protective effects in type 2 diabetes.

Material and methods: Diabetes was induced in 15 days high-fat diet (HFD)-fed Wistar rats by intraperitoneal injection of streptozotocin (STZ) (40 mg/kg). The ethyl acetate extract (250 and 500 mg/kg) of F. carica leaves was administered for 28 days. Oral glucose tolerance (OGTT) and intraperitoneal insulin tolerance tests (ITT) were evaluated on 15th and 25th days, respectively.

Results: The ethyl acetate extract (250 and 500 mg/kg) of n F. carica leaves showed significant effect (p < 0.005) in the levels of blood glucose, total cholesterol (TC), triglycerides (TG), body weight and hepatic glycogen. In OGTT, F. carica (250 and 500 mg/kg) significantly (p < 0.005) detained the increase in blood glucose levels at 60 and 120 min and in ITT, F. carica enhanced the glucose utilization significantly (p < 0.005) over 30 and 60 min compared to diabetic control. Further, the altered activities of key carbohydrate metabolizing enzymes such as glucose-6-phosphatase, fructose-1,6-bisphosphatase and hexokinase in the liver tissue of diabetic rats were significantly (p < 0.005) reverted to near normal levels upon treatment with F. carica. Immumohistochemical studies of islets substantiated the cytoprotective effect on pancreatic β-cells.

Discussion and conclusions:F. carica leaves exerted significant effect on carbohydrate metabolism enzymes with promising hypoglycemic and hypolipidemic activities in type 2 diabetic rats.

Introduction

Type 2 diabetes mellitus (T2DM) or non-insulin-dependent diabetes mellitus accounts for more than 90–95% of world human diabetic population (Naik et al. 2013). Insulin resistance, defective insulin secretion and pancreatic β-cell damage were observed as metabolic features of T2DM. It is predicted that by 2030 the total number of people having diabetes will reach 366 million (Shaw et al. 2010). Along with high blood glucose level (hyperglycemia) and variation in the levels of serum lipids, diabetes will lead to micro- and macro-vascular complications. These complications are the major cause of morbidity and death in diabetic subjects (Sunil et al. 2012a). Insulin, oral hypoglycemic drugs like sulphonylurea derivatives, biguanides, thiazolidinediones and α-glucosidase inhibitors are currently used to treat type 2 diabetes. These agents have undesirable side effects: thiazolidinediones induce obesity, osteoporosis and sodium retention; sulphonylurea derivatives produce hypoglycemia and the biguanide (metformin) puts patients at the risk of developing lactic acidosis (Surya et al. 2014). Medicinal plants are used traditionally in many countries to control diabetes. Consequently, many medicinal plants have been investigated with the aim of discovering potential hypoglycemic agents (Sunil et al. 2012b). Because of its activity, nontoxic, with little or no side effects, World Health Organization (WHO) has recommended that traditional plants can be an excellent candidate for the treatment of diabetes (Stephen Irudayaraj et al. 2012). Plants used in traditional medicine to treat diabetes mellitus represent a valuable alternative for the control of this disease.

Ficus carica Linn. (Moraceae), commonly referred as Figs or Anjir, is cultivated in tropical and subtropical regions of India and worldwide for their nutritive and medicinal properties; it has been widely used in indigenous systems of medicine, such as ayurveda, siddha and homoeopathy (Manda et al. 1997). F. carica leaves, bark, tender shoots, fruits, seeds and latex are traditionally used in the treatment of jaundice, diarrhea, nutritional anemia and as an anti-inflammatory agent (Canal et al. 2000). Decoctions of F. carica leaves are used as folk medicine for the treatment of diabetes (Perez et al. 1996). Hepatoprotective, hypoglycemic, antifungal, antispasmodic, antipyretic, anthelmintic, antioxidant and antimutagenic activities have been reported (Vikas & Vijay 2011). The antidiabetic property of F. carica has been reported in alloxan-induced insulin-deficient diabetic rat model (Stalin et al. 2012). The antihyperlipidemic effect of F. carica in high-fat diet and Triton X-100-induced hyperlipidemic animal models are reported (Dayanand & Kishanchandra 2014; Joerin et al. 2014). Therefore, this study was undertaken to find the effect of F. carica on glucose and lipids levels, carbohydrate metabolism enzymes and β-cells protective effects in HFD-fed STZ-induced type 2 diabetic rats.

Materials and methods

Chemicals and reagents

Streptozotocin (STZ) and all fine chemicals were purchased from Sigma-Aldrich Chemical Company (St. Louis, MO). Ultrasensitive rat insulin ELISA kit was obtained from Crystal Chem, Inc. (Downers Grove, IL). Glibenclamide and all other chemicals of analytical grade were purchased from local firm (Chennai, Tamil Nadu, India).

Plant material

F. carica leaves were collected from Western Ghats, Tamil Nadu, India, during January 2015 and were duly authenticated by Ayyanar, the taxonomist of Entomology Research Institute, Loyola College (Voucher no. ERI/ETHPH/FC/234).

Extraction of crude extracts of F. carica

F. carica leaves were rinsed with water, shade-dried and powdered. Fine powder (3 kg) was soaked in 9 L of hexane, ethyl acetate and methanol, sequentially for 72 h at room temperature. The extrats were filtered and concentrated using rotary evaporator at 40–55 °C. 42, 36 and 19 g were yielded from the hexane, ethyl acetate and methanol extracts, respectively, and they were stored in refrigerator at 4 °C. In our preliminary studies among hexane, ethyl acetate and methonal extracts, the ethyl acetate extract of F. carica showed highly significant glucose lowering activity (data not shown). Hence, the study was designed to investigate the hypoglycemic, hypolipidaemic and β-cells protective effects of the ethyl acetate extract of F. carica leaves in HFD-fed STZ-induced type 2 diabetic rats.

HPLC-DAD analysis

Qualitative and quantitative analyses of major constituent/constituents in F. carica were carried out on Waters Alliance 2695 separations Module HPLC system with photodiode array detector (Waters, Milford, MA, 2996), with the reversed phase LC column, YMC pack ODS A (150 mm ×4.6 mm, 3.5 μm) was eluted with 0.1% orthophosphoric acid (A) and acetonitrile (B) in the ratio of 1:1 (A:B, v/v) as mobile phase system with a flow rate of 1 mL/min. The samples were injected using a 20 μL loop (Rheodyne, Rohnet Park, CA) and the separations were monitored with PDA signal at 270 nm. Peak purity was checked and the quantification was done. Ficusin (HPLC purity 98.6%) was used as standard and run at five concentrations (3–150 μg/mL) and should be linear in the range with a correlation coefficient (r2) of 0.99700. Using linear regression analysis, the ficusin content was estimated using the calibration cure appearing in the standard chromatogram using the following equation:C(c)=A(c)A(st)×C(st)where C(c) is the concentration of the constituent in the sample, A(c) is the peak area of the constituent in the sample chromatogram, C(st) is the concentration of the standard in the reference solution and A (st) is the area of the peak for the standard in the reference chromatogram (Vinholes et al. 2011).

Experimental animals

Healthy male albino Wistar rats (170–190 g) were housed and raised at the animal housed of Entomology Research Institute in favourable environmental conditions at temperature (22 ± 2 °C), a relative humidity (45 ± 5 °C) and 12/12 h day/night cycle of 7 days. The animals were fed ad libitum with normal laboratory standard pelleted diet purchased from Sai Durga Feeds and Foods, Bangalore. The animal experiments and protocols were approved by the Institutional Animal Ethics Committee (IAEC-ERI-LC-05/13).

Fixation of doses and acute toxicity study

Overnight fasted rats were divided into five groups of six rats each. F. carica was dissolved in vehicle (0.9% sodium chloride, 0.5% carboxymethyl cellulose (CMC) and 0.2% tween 80 in distilled water) and used for treatment. Treated rats were given oral doses of F. carica (1000, 2000, 3000, 4000, 5000 mg/kg) respectively. The control group was given vehicle alone. The physical signs of toxicity, such as writhing, gasping, palpitation and decreased respiratory rate, body weight changes, food and water consumption or mortality were observered for 1 h continuously, then for 4 h and finaly after every 24 h up to 14 days.

Neither mortality nor adverse effects except for mild writhing were observed up to 5000 mg/kg of F. carica extract. Hence, 250 and 500 mg/kg doses were preferred for consequent experiments (Oliveira et al. 2008).

Development of T2DM

All the experimental rats were fed with HFD except normal control composed of standard rat pelleted diet 68%, dalda (saturated fat) 30%, and cholesterol 2% for 15 days. Single intraperitoneal injection of freshly prepared solution of STZ (40 mg/kg) in 0.1 M citrate buffer (pH 4.5) was used to induce diabetes. Rats were given 20% glucose solution for 24 h to prevent initial drug-induced hypoglycemic mortality. After seven days of STZ administration, blood samples were collected from the tail vein and plasma glucose levels were determined as described by Trinder (1969). Hyperglycemia was considered by measuring the fasting blood glucose (FBG) level. Rats with a FBG level above 250 mg/dL were categorized as diabetic and included in the study.

Experimental design

The animals were assigned into five groups of six animals in each group.

Group I: Normal control rats treated with vehicle alone.

Group II: Diabetic control rats treated with vehicle alone.

Group III: Diabetic rats treated with F. carica (250 mg/kg).

Group IV: Diabetic rats treated with F. carica (500 mg/kg).

Group V: Diabetic rats treated with glibenclamide (0.6 mg/kg).

Treatments (either vehicle or test drugs) were administered orally, once daily using intragastric tube for 28 days.

Biochemical analysis

FBG and body weights of days 0, 14, 21 and 28 were measured. Plasma insulin (Ultrasensitive rat insulin ELISA kit), TC (Henley 1957) and TG (Foster & Dunn 1973) were determined on days 0 and 28. OGTT and ITT were performed on days 15 and 25, respectively. On 28th day, the animals were anesthetized and sacrificed by cervical decapitation. Blood was collected in dry EDTA containing test tubes and the liver tissues were sliced quickly and washed in ice cold saline and stored at −70 °C in liquid nitrogen for further analysis of glycogen. The homogenate prepared in ice-chilled 10% potassium chloride in 0.1 M phosphate buffer (pH 6.5) solution was used to measure the activities of hexokinase (Brandstrup et al. 1957), glucose-6-phosphatase (Koide & Oda 1959), fructose-1,6-bisphosphatase (Gancedo & Gancedo 1971) and hepatic glycogen (Van Handel 1965), respectively.

Immunohistochemical analysis

Immunohistochemical study was done as described by Gandhi et al. (2012).

Statistical analysis

The results were presented as mean ± SEM. Statistical analyses of all the data obtained were evaluated using one-way ANOVA followed by Student’s t-test (SPSS Program; Version 11.5, Chicago, IL). The differences were considered as significant at p < 0.005.

Results

HPLC-DAD analysis

Quantification of ficusin (a furocoumarin) by HPLC analysis showed its content to be 6.46% w/w. The retention time (tR) of reference standard ficusin was observed at 1.2 min and it was found to be the same with ficusin present in the ethylacetate extract of the leaves of F. carica (Figure 1).

Figure 1.

HPLC fingerprint chromatogram of F. carica with ficusin (A) and the standard chromatogram of ficusin (B) peak. FI: ficusin.

Effect of F. carica on OGTT and ITT

In oral glucose tolerance test, soon after glucose administration, the plasma glucose levels reached the maximum at 30 min in all the groups. Treatment with F. carica (250 and 500 mg/kg), significantly (p < 0.005) detained the increase of blood glucose levels at 60 and 120 min (Figure 2). In addition, F. carica treatment (250 and 500 mg/kg) enhanced the glucose utilization significantly (p < 0.005) over the complete period during the ITT compared to diabetic control rats (Figure 3).

Figure 2.

Effect of F. carica on 15th day OGTT.

Figure 3.

Effect of F. carica on 25th day ITT.

Effect of F. carica on FBG and body weight

There was a significant (p < 0.005) increase in the levels of FBG in HFD-fed STZ-treated diabetic control rats. F. carica (250 and 500 mg/kg) treatment significantly (p < 0.005) reduced the blood glucose level to near normal compared to glibenclamide-treated (0.6 mg/kg) rats (Table 1). The body weight was also increased in F. carica-treated (250 and 500 mg/kg) diabetic rats (Table 2).

Table 1.

Levels of glucose in normal and diabetic rats after 28 days of treatment with F. carica.

Fasting blood glucose levels (mg/dl)
Groups0 day14th day21st day28th day
Normal control77.82 ± 1.9382.03 ± 2.1179.21 ± 4.2480.55 ± 2.18
Diabetic control283.26 ± 7.82319.4 ± 8.42323.28 ± 3.64336.35 ± 6.14
Diabetic + F. carica (250 mg/kg)298.31 ± 6.69249.67 ± 8.12**174.81 ± 7.42**137.27 ± 7.24**
Diabetic + F. carica (500 mg/kg)274.37 ± 4.18251.6 ± 7.44**142.65 ± 8.12**129.14 ± 8.23**
Diabetic + Glibenclamide (0.6 mg/kg)283.61 ± 9.14256.1 ± 6.72**152.66 ± 7.74**131.32 ± 9.13**
Values indicate mean ± standard error of the mean (SEM) of six rats per group.

**p < 0.005, compared with diabetic control values.

Table 2.

Effect of F. carica on body weight after 28 days of treatment.

Body weight (g)
Groups0 day14th day21st day28th day
Normal control188.12 ± 11.42185. 2 ± 12.54190.20 ± 12.10190.22 ± 3.21
Diabetic control209.25 ± 18.24242.15 ± 10.71264.16 ± 10.17281.22 ± 13.52
Diabetic + F. carica (250 mg/kg)180.48 ± 9.69192.67 ± 9.12**194.65 ± 8.56**195.26 ± 9.12**
Diabetic + F. carica (500 mg/kg)190.34 ± 10.18195.4 ± 12.92**201.21 ± 9.57**204.46 ± 8.71**
Diabetic + Glibenclamide (0.6 mg/kg)202.72 ± 9.14209.8 ± 17.42**215.89 ± 9.12*219.51 ± 8.22**
Values indicate mean ± standard error of the mean (SEM) of six rats per group.

**p < 0.005, compared with diabetic control values.

Effect of F. Carica on plasma insulin, TC and TG

Plasma insulin level showed a significant increase (p < 0.005) in untreated diabetic rats. After treatment with F. carica (250 and 500 mg/kg), plasma insulin level significantly (p < 0.005) reduced to normal when compared with the diabetic control rats. Similarly, increased TC and TG levels were observed in untreated diabetic rats. After treatment with F. carica (250 and 500 mg/kg) for 28 days, TC and TG levels were significantly (p < 0.005) reverted to the near normal level (Table 3).

Table 3.

Effect of F. carica on Plasma insulin, TC and TG after 28 days of treatment.

Plasma Insulin (μU/mL)
TC (mg/dl)
TG (mg/dl)
Groups0 day28th day0 day28th day0 day28th day
Normal control16.86 ± 1.7416.28 ± 2.078.56 ± 5.8179.14 ± 3.5474.78 ± 3.4271.65 ± 4.42
Diabetic control22.45 ± 1.5425.31 ± 0.4699.24 ± 6.54126.62 ± 7.88149.74 ± 7.14169.14 ± 6.12
Diabetic + F. carica (250 mg/kg)24.52 ± 1.7820.77 ± 0.84**96.67 ± 5.3288.14 ± 1.89**163.78 ± 9.18106.84 ± 5.23**
Diabetic + F. carica (500 mg/kg)22.84 ± 1.6415.84 ± 0.76**96.98 ± 8.1271.52 ± 4.64**164.83 ± 6.2394.29 ± 11.68**
Diabetic + Glibenclamide (0.6 mg/kg)23.56 ± 1.5116.13 ± 0.53**91.26 ± 1.7668.18 ± 4.58**157.18 ± 7.1189.18 ± 8.16**
Values indicate mean ± standard error of the mean (SEM) of six rats per group.

**p < 0.005, compared with diabetic control values.

Effect of F. Carica on hexokinase, glucose-6-phosphatase, fructose-1,6-bisphosphatase and hepatic glycogen

The activities of hexokinase, glucose-6-phosphatase and fructose-1,6-bisphosphatase and the level of glycogen content in the liver of normal and diabetic rats are shown in Table 4. A significant (p < 0.005) increase in the activities of glucose-6-phosphatase and fructose- 1,6-bisphosphatase with significant (p < 0.005) decrease in hexokinase and glycogen were observed in diabetic rats when compared to normal control rats. Oral administration of F. carica (250 and 500 mg/kg) for 28 days significantly decreased the glucose-6-phosphatase and fructose-1,6-bisphosphatase, and significantly increased the hexokinase and glycogen to near normal.

Table 4.

Effect of F. carica on hexokinase, glucose-6-phosphatase, fructose-1, 6-bisphosphatase and hepatic glycogen after 28 days of treatment.

GroupsHexokinase (U/mg protein /min)Glucose-6- phosphatase (U/mg protein/min)Fructose-1,6- bisphosphatase (U/mg protein/min)Hepatic glycogen (mg/g tissue)
Normal control8.13 ± 1.740.29 ± 0.120.39 ± 0.0724.74 ± 2.84
Diabetic control4.98 ± 0.980.61 ± 0.120.69 ± 0.159.54 ± 1.82
Diabetic + F. carica (250 mg/kg)6.28 ± 1.67*0.37 ± 0.08**0.53 ± 0.08*18.58 ± 2.92**
Diabetic + F. carica (500 mg/kg)7.74 ± 1.93**0.26 ± 0.16**0.42 ± 0.06**20.69 ± 1.74**
Diabetic + Glibenclamide (0.6 mg/kg)7.18 ± 1.24**0.22 ± 0.05**0.41 ± 0.07**18.27 ± 1.22**
Values indicate mean ± standard error of the mean (SEM) of six rats per group.

*p < 0.05,

**p < 0.005, compared with diabetic control values.

Immunohistochemical observations

The results of the intensity of insulin-immunostaining expression is shown in Figure 4. Diabetic control rats showed a significant (p < 0.005) reduction in the level of insulin-immunostaining expression as compared to normal control rats. F. carica-treated (250 and 500 mg/kg) diabetic groups exhibited significant (p < 0.005) increase in the insulin-immunostaining expression compared to the vehicle-received diabetic control group. The immumohistochemical examinations showed islets with positive insulin-immunoreactive expression in the pancreas of normal rats (Figure 5(A)). Diabetic rats showed low amount of insulin-immunoreactive expression (Figure 5(B)). There was a relative rise in the expression of insulin-immunoreactivity with reliable dark brown insulin-immunostaining in the F. carica- (250 and 500 mg/kg) and glibenclamide-treated (0.6 mg/kg) diabetic groups compared with the diabetic control group (Figure 5(C–E)).

Figure 4.

Histogram showing intensity of insulin-immunostaining expression of β-cells in islet of Langerhans. (A) Normal control. (B) Diabetic control. (C and D) Diabetic + F. carica (250 and 500 mg/kg b wt.). (E) Diabetic + glibenclamide (0.6 mg/kg). Values represent the mean ± standard error of the mean (SEM) of six rats per group. **p < 0.005, compared with diabetic control values.

Figure 5.

Light photo micrographs of rat pancreas showing insulin-immunoreative expression of β-cells in islet of Langerhans. (A) Islet of normal control rat. (B) Islet of diabetic control rat. (C and D) Islet of F. carica-treated (250 and 500 mg/kg) diabetic rats. (E) Islet of glibenclamide-treated (0.6 mg/kg) diabetic rats.

Discussion

T2DM is a progressive and complex disorder that is emerging as a crucial threat to human population (Steppan et al. 2001). We observed in our study that HFD treatment along with intraperitoneal administration of STZ (40 mg/kg) damaged the insulin-secreting β-cells of the pancreas by breaking the DNA strand and caused depressed β-cell function, decreased endogenous insulin release and defects in insulin action (Koning et al. 2008). Other investigators have also used similar experimental T2DM animal model to explore drug discoveries (Zhang et al. 2010; Veerapur et al. 2012).

OGTT and ITT were performed on 15th and 25th day, respectively. The in vivo peripheral insulin action and insulin resistance in animals can be assed by OGTT, hence it is a simple and widely accepted method (Liou et al. 2002). In our study, F. carica stimulated the glucose uptake into peripheral tissues of the diabetic rats in a dose-dependent manner. ITT is useful to assess insulin sensitivity by exogenous administration of insulin. F. carica (250 and 500 mg/kg) reduced the blood glucose significantly at 30 and 60 min, suggesting imporved insulin sensitivity possibly by improving one or more defects, namely insulin receptor, insulin receptor substrate, glucose transporters or enzymes involved in phosphorylation of glucose (Benwahhoud et al. 2001; El Hilaly & Lyoussi 2002). These results from ITT and OGTT tests clearly established that F. carica administration reduced the glucose level by the mechanism of insulin-sensitized glucose uptake. Menaka et al. (2010) has also been reported in his study that, the similar mechanism of insulin-sensitized glucose uptake has been observed in ITT and OGTT of Sida rhomboidea extract on the C57BL/6J mice model.

A significant decrease in blood glucose and normalization of plasma insulin levels were observed in diabetic rats treated with F. carica. This could be due to the potentiation of the extract on pancreatic secretion of insulin from regenerated β-cells, or its action to release bound insulin from regenerated β-cells by inhibiting ATP-sensitive K+ channels like glibenclamide (Sunil et al. 2012b). Yuan et al. (2001) have suggested that hyperinsulinaemia caused by insulin resistance in peripheral tissues damages the structural integrity of β-cells. In our study, the immunohistochemical analysis indicates that diabetic rats showed impaired and degranulated β-cells with the decrease of insulin-positive staining β-cells. But, F. carica treatment preserved the β-cell mass on diabetic rats. The extensive increase in insulin-immunoreactive expression in F. carica-treated diabetic rats substantiated the protective role of F. carica in the reversion of pancreatic β-cell damage caused by HFD-STZ. Coumarins are reported as potential hypoglycemic agents with insulin sensitization effect (Liang et al. 2009; Qin et al. 2010). F. carica contains good amount of furocoumarin, ficusin which could be responsible for its insulin sensitization effect. Moreover, ficusin has been reported as good antioxidant, antilipidemic and antidiabetic effects with their effects on GLUT4 translocation and PPARγ expression in type 2 diabetic rats (Stephen Irudayaraj et al. 2016).

The administration of HFD has significantly increased the TC and TG in the diabetic rats. In the present study, HFD-fed STZ-induced diabetic rats have also shown marked rise in their body weights compared to normal control rats. Diabetic condition with profound alterations in lipid profile leads to the development of atherosclerosis (Warnholtz et al. 2001). After the oral administration of F. carica for 28 days to diabetic rats, there was a significant restoration in altered lipid profile along with controlled body weight gain compared to untreated HFD-fed STZ group. Dayanand and Kishanchandra (2014) has also reported that the F. carica treatment significantly lowered the TC and TG levels in high-fat diet and Triton X-100-induced hyperlipidemic Wistar rats. Another study also confirmed the antihyperlipidemic effect of F. carica leaf extract on high-fat diet-induced Sprague–Dawley rats rats (Joerin et al. 2014).

The activities of key glycolytic enzymes, hexokinase, glucose-6-phosphatase and fructose-1,6-bisphosphatase were significantly altered during diabetic illness (Vestergard 1999). There is extensive evidence to show that elevated gluconeogenic enzymes such as glucose-6-phosphatase and fructose-1,6-bisphosphatase contribute to hyperglycemia in the diabetic state (Saxena et al. 1984). F. carica significantly normalized the activities of these gluconeogenic enzymes in the HFD-fed STZ-treated diabetic rats. F. carica has protective effect against the disturbed carbohydrate metabolic enzymes; this is probably due to enhanced insulin action. The insulin sensitization activity of F. carica was further substantiated via its improved hepatic glycogen content in the treated diabetic rats. Insulin plays a role in regulating glycogen metabolism through activation or inhibition of several mediatory enzymes and proteins (Ferrer et al. 2003).

The immunohistochemical study clearly indicated that the HFD-fed STZ-induced diabetic control rats had decreased insulin-immunoreactive expression. A defect in insulin action along the peripheral tissues resulting in hyperinsulinaemia eventually damages the structural integrity and functional status of β-cells (Yuan et al. 2001). The increase in insulin-immunoreactive expression in F. carica-treated diabetic rats proved the cytoprotective role of F. carica in preventing the deterioration of β-cells.

Conclusions

In conclusion, the ethyl acetate extract of F. carica leaves possesses antidiabetic activity by stimulating the insulin production from the regenerated beta cells of the pancreas. Increased insulin secretion after treatment with F. carica positively altered the deranged carbohydrate metabolism in the diabetic rats by decreasing gluconeogenesis and increasing glycolysis, ultimately decreasing hyperglycemia.

Acknowledgments

The authors would like to extend their sincere appreciation to the Deanship of Scientific Research at King Saud University for its funding of this research through the Research Group project No RGP-VPP-213

Disclosure statement

The authors declare that there are no conflicts of interest.

References

  • 1. BenwahhoudM, JouadH, EddouksM, LyoussiB2001Hypoglycemic effect of Suaeda fruticosa in streptozotocin-induced diabetic rats. J Ethnopharmacol. 76:3538.[PubMed][Google Scholar]
  • 2. BrandstrupN, KirkJE, BruniC1957The hexokinase and phosphoglucoisomerase activities of aortic and pulmonary artery tissue in individuals of various ages. J Gerontol. 12:166171.[PubMed][Google Scholar]
  • 3. CanalJR, TorresDM, RomeroA, PerezC2000A chloroform extract obtained from a decoction of Ficus carica L. leaves improve the cholesterolaemic status of rats with streptozotocin induced diabetes. Acta Vet Hung. 87:7176.[Google Scholar]
  • 4. DayanandK, KishanchandraK2014Phytochemical investigation and evaluation of antihyperlipidemic activity of Ficus carica Linn. fruit extracts. Global J Res Analysis. 3:22778160.[Google Scholar]
  • 5. El HilalyJ, LyoussiB2002Hypoglycaemic effect of the lyophilised aqueous extract of Ajuga iva in normal and streptozotocin diabetic rats. J Ethnopharmacol. 80:109113.[PubMed][Google Scholar]
  • 6. FerrerJC, FavreC, GomisRR, Fernández-NovellJM, Garcia-RochaM, De LaIglesiaN2003Control of glycogen deposition. FEBS Lett. 546:127132.[PubMed][Google Scholar]
  • 7. FosterLB, DunnRT1973Stable reagents for determination of serum triglycerides by a colorimetric Hantzsch condensation method. Clin Chem. 19:338340.[PubMed][Google Scholar]
  • 8. GancedoJM, GancedoC1971Fructose-1,6-diphosphatase, phosphofructokinase and glucose-6-phosphate dehydrogenase from fermenting and non fermenting yeasts. Arch Microbiol. 76:132138.[Google Scholar]
  • 9. GandhiGR, IgnacimuthuS, PaulrajMG2012Hypoglycemic and β-cells regenerative effects of Aegle marmelos (L.) Corr. bark extract in streptozotocin-induced diabetic rats. Food Chem Toxicol. 50:16671674.[PubMed][Google Scholar]
  • 10. HenleyAA1957The determination of serum cholesterol. Analyst. 82:286287.[Google Scholar]
  • 11. JoerinL, KauschkaM, BonnländerB, PischelI, BenedekB, ButterweckV2014Ficus carica leaf extract modulates the lipid profile of rats fed with a high-fat diet through an increase of HDL-C. Phytother Res. 28:261267.[PubMed][Google Scholar]
  • 12. KoideH, OdaT1959Pathological occurrence of glucose-6-phosphatase in serum in liver diseases. Clin Chim Acta. 4:554561.[PubMed][Google Scholar]
  • 13. KoningEJ, WeirBS, RabelinkTJ2008Preservation of beta-cell function by targeting beta-cell mass. Trends Pharmacol Sci. 29:218227.[PubMed][Google Scholar]
  • 14. LiangHJ, SukFM, WangCK, HungLF, LiuDZ, ChenNQ, ChenYC, ChangCC, LiangYC2009Osthole, a potential antidiabetic agent, alleviates hyperglycemia in db/db mice. Chem Biol Interact. 181:309315.[PubMed][Google Scholar]
  • 15. LiouSS, LiuIM, HsuJH, WuYC, HsuSF, ChenJT2002Release of acetylcholine by Die–Huang–Wan to enhance insulin secretion for lowering plasma glucose in Wistar rats. Auton Neurosci. 100:2126.[PubMed][Google Scholar]
  • 16. MandaSC, MukherjeeKP, SahaK, DasJ, PalM, SabaPB1997Hypoglycemic activity of Ficus carica L. leaves in streptozotoin-induced diabetic rats. Nat Prod Sci. 3:3841.[Google Scholar]
  • 17. MenakaCT, RavirajsinhNJ, Ansarullah, RanjitsinhVD, RamachandranAV2010Prevention of high fat diet induced insulin resistance in C57BL/6J mice by Sida rhomboidea Roxb. extract. J Health Sci. 56:9298.[Google Scholar]
  • 18. NaikSR, NitureNT, AnsariAA, ShahPD2013Anti-diabetic activity of embelin: involvement of cellular inflammatory mediators, oxidative stress and other biomarkers. Phytomedicine. 20:797804.[PubMed][Google Scholar]
  • 19. OliveiraHC, SantosMPD, GriguloR, LimaLL, MartinsDTO, LimaCS2008Antidiabetic activity of Vatairea macrocarpa extract in rats. J Ethnopharmacol. 115:515519.[PubMed][Google Scholar]
  • 20. PerezC, DominguezE, RamiroJM, RomeroA, CampilloJE, TorresMD1996A study on the glycaemic balance in streptozotocin diabetic rats treated with aqueous extract of Ficus carica (Fig tree) leaves. Phytother Res. 10:8283.[Google Scholar]
  • 21. QinB, PanickarKS, AndersonRA2010Cinnamon: potential role in the prevention of insulin resistance, metabolic syndrome, and type 2 diabetes. J Diabetes Sci Technol. 4:685693.[PubMed][Google Scholar]
  • 22. SaxenaA, BhatnagarM, GargNK1984Enzymes changes in rat tissues during hyperglycemia. Arogya – J Health Sci. 10:3337.[Google Scholar]
  • 23. ShawJE, SicreeRA, ZimmetPZ2010Global estimates of the prevalence of diabetes for 2010 and 2030. Diabetes Res Clin Pract. 87:414.[PubMed][Google Scholar]
  • 24. StalinC, DineshkumarP, NithiyananthanK2012Evaluation of antidiabetic activity of methanolic leaf extract of Ficus carica in alloxan induced diabetic rats. Asian J Pharm Clin Res. 5:8587.[Google Scholar]
  • 25. Stephen IrudayarajS, SunilC, DuraipandiyanV, IgnacimuthuS2012Antidiabetic and antioxidant activities of Toddalia asiatica (L.) Lam. leaves in streptozotocin induced diabetic rats. J Ethnopharmacol. 143:515523.[PubMed][Google Scholar]
  • 26. Stephen IrudayarajS, StalinA, SunilC, DuraipandiyanV, Al-DhabiNA, IgnacimuthuS2016Antioxidant, antilipidemic and antidiabetic effects of ficusin with their effects on GLUT4 translocation and PPARγ expression in type 2 diabetic rats. Chem Biol Interact. 256:8593.[PubMed][Google Scholar]
  • 27. SteppanCM, BaileyST, BhatS, BrownEJ, BanerjeeRR, WrightCM2001The hormone resistin links obesity to diabetes. Nature. 409:307312.[PubMed][Google Scholar]
  • 28. SunilC, AgastianP, IgnacimuthuI2012aIn vitro antioxidant, antidiabetic and antilipidemic activities of Symplocos cochinchinensis (Lour.) S. Moore. bark. Food Chem Toxicol. 50:15471553.[PubMed][Google Scholar]
  • 29. SunilC, DuraipandiyanV, IgnacimuthuS, AgastianP2012bAntidiabetic effect of Plumbagin isolated from Plumbago zeylanica L. root and its effect on GLUT4 translocation in streptozotocin-induced diabetic rats. Food Chem Toxicol. 50:43564363.[PubMed][Google Scholar]
  • 30. SuryaS, Dhaliya SalamA, DawnVT, CarlaB, Arun KumarR, SunilC2014Diabetes mellitus and medicinal plants – a review. Asian Pac J Trop Dis. 4:337347.[Google Scholar]
  • 31. TrinderP1969Determination of glucose in blood using glucose oxidase with an alternative oxygen acceptor. Ann Clin Biochem. 6:2427.[Google Scholar]
  • 32. Van HandelE1965Estimation of glycogen in small amounts of tissue. Anal Biochem. 11:256265.[PubMed][Google Scholar]
  • 33. VeerapurVP, PrabhakarKR, ThippeswamyBS, BansalP, SrinivasanKK, UnnikrishnanMK2012Antidiabetic effect of Ficus racemosa Linn. stem bark in high-fat diet and low-dose streptozotocin-induced type 2 diabetic rats: a mechanistic study. Food Chem. 132:186193.[PubMed][Google Scholar]
  • 34. VestergardH1999Studies of gene expression and activity of hexokinase, phosphofructokinase and glycogen synthase in human skeletal muscle in states of altered insulin-stimulated glucose metabolism. Danish Med Bull. 46:1334.[PubMed][Google Scholar]
  • 35. VikasVP, VijayRP2011Ficus carica Linn – an overview. Res J Med Plant. 5:246253.[Google Scholar]
  • 36. VinholesJ, GrossoC, AndradePB, Gil-IzquierdoA, ValentaoP, PinhoPGD2011In vitro studies to assess the antidiabetic, anti-cholinesterase and antioxidant potential of Spergularia rubra. Food Chem. 129:454462.[Google Scholar]
  • 37. WarnholtzA, MollnauM, OlezeM, Wendt MunzelT2001Antioxidants and endothelial dysfunction in hyperlipidemia. Curr Hypertens Rep. 3:5360.[PubMed][Google Scholar]
  • 38. YuanM, KonstantopoulosN, LeeJ2001Reversal of obesity- and diet-induced insulin resistance with salicylates or targeted disruption of Ikkbeta. Science293:16731677.[PubMed][Google Scholar]
  • 39. ZhangL, YangJ, ChenX, ZanK, ChenH, WangQ, LaiM2010Antidiabetic and antioxidant effects of extracts from Potentilla discolor Bunge on diabetic rats induced by high fat diet and streptozotocin. J Ethnopharmacol. 132:518524.[PubMed][Google Scholar]
Collaboration tool especially designed for Life Science professionals.Drag-and-drop any entity to your messages.