SciELO - Scientific Electronic Library Online

 
vol.37 número5La suplementación con BCAA no mejora el salto vertical en los jugadores de voleibol profesionales. Estudio doble ciego, controlado y aleatorizadoEfectos de una dieta baja en hidratos de carbono sobre la composición corporal y el rendimiento en el ciclismo de carretera: estudio aleatorizado y controlado índice de autoresíndice de materiabúsqueda de artículos
Home Pagelista alfabética de revistas  

Servicios Personalizados

Revista

Articulo

Indicadores

Links relacionados

  • En proceso de indezaciónCitado por Google
  • No hay articulos similaresSimilares en SciELO
  • En proceso de indezaciónSimilares en Google

Compartir


Nutrición Hospitalaria

versión On-line ISSN 1699-5198versión impresa ISSN 0212-1611

Nutr. Hosp. vol.37 no.5 Madrid sep./oct. 2020  Epub 04-Ene-2021

https://dx.doi.org/10.20960/nh.03051 

Original Papers

Antidiabetic effect of olive leaf extract on streptozotocin-induced diabetes mellitus in experimental animals

Efecto antidiabético del extracto de hoja de olivo sobre la diabetes mellitus inducida por estreptozotocina en animales de experimentación

Murat Gürbüz1  , Serdal Öğüt2 

1Department of Nutrition and Dietetics. Trakya University. Edirne, Turkey

2Department of Nutrition and Dietetics. Adnan Menderes University. Aydın, Turkey

Abstract

Introduction:

recently, a relationship between diabetic complications and oxidative stress has been emphasized. There have been some studies showing the effect of olive leaf on hyperglycemia and diabetic complications due to its antioxidant properties. In many studies the effect of olive leaf on plasma total antioxidant level has been measured by different methods. Our study represents the first time it has been measured by a new method of total thiol disulfide homeostasis.

Objective:

chronic exposure to hyperglycemia and hyperlipidemia contributes to the pathogenesis of diabetic complications through oxidative stress mediators. Thiol is one of the most important antioxidant barriers in humans, and thiol disulfide homeostasis is a new oxidative stress marker. We aimed to investigate the effect of olive leaf extract (OLE) obtained from fresh leaves of Olea europaea, var oleaster on diabetic complications through their hypoglycemic and antioxidant effect in diabetic rats.

Methods:

twenty-eight Wistar albino rats aged 12-13 weeks were used in the study. The rats were divided into a control group (C), a diabetic control group (DC), a diabetic group treated with 200 mg/kg OLE (D+200), and a diabetic group treated with 400 mg/kg OLE (D+400), having 7 rats in each group. The treatment groups received OLE by the gavage method for 21 days. At the end of the study, all rats were sacrificed by cervical dislocation. Blood samples collected from the heart were centrifuged and glucose, total cholesterol, triglyceride, urea, uric acid, creatinine, alanine aminotransferase (ALT), aspartate aminotransferase (AST), lipid hydroperoxide (LOOH) level, and thiol-disulfide homeostasis were determined. The hemoglobin A1c (HbA1c) analysis was performed on complete blood. In addition, a tail flick test and hot plate modeling were performed to indicate pain perception loss.

Results:

it was observed that OLE had no effect on serum glucose and HbA1c levels. On the contrary, OLE reduced the levels of total cholesterol (p < 0.01), urea (p < 0.01) and hot plate latency (p < 0.01) in a significant manner. Also, OLE showed a tendency to reduce LOOH levels and to increase thiol levels in a dose-dependent manner (p > 0.05).

Conclusion:

OLE supplementation for 21 days, at the amounts used, cannot protect against hyperglycemia but may be protective against hypercholesterolemia and tissue damage as caused by diabetes mellitus in rats.

Key words: Olive leaf; Oleuropein; Antidiabetic activity; Olea europaea L

Resumen

Introducción:

recientemente se ha resaltado la relación entre las complicaciones diabéticas y el estrés oxidativo. Se han realizado algunos estudios que muestran el efecto de la hoja de olivo sobre la hiperglucemia y las complicaciones diabéticas debido a sus propiedades antioxidantes. En muchos estudios, el efecto de la hoja de olivo sobre el nivel de antioxidantes totales en plasma se ha medido mediante diferentes métodos. En nuestro estudio se ha medido por primera vez mediante un nuevo método de homeostasis total de disulfuro de tiol.

Objetivo:

la exposición crónica a la hiperglucemia y la hiperlipidemia contribuye a la patogénesis de las complicaciones diabéticas a través de mediadores del estrés oxidativo. El tiol es una de las barreras antioxidantes más importantes de los seres humanos y la homeostasis del disulfuro de tiol es un nuevo marcador de estrés oxidativo. El objetivo fue0 investigar el efecto del extracto de hoja de olivo (OLE), obtenido de hojas frescas de Olea europaea var. Oleaster, sobre las complicaciones diabéticas a través del efecto hipoglucémico y antioxidante en ratas diabéticas.

Métodos:

se utilizaron en el estudio veintiocho ratas albinas Wistar de 12-13 semanas de edad. Las ratas se agruparon en un grupo de control (C), un grupo de control diabético (DC), un grupo diabético tratado con 200 mg/kg de OLE (D+200) y un grupo diabético tratado con 400 mg/kg de OLE (D+400), teniendo 7 ratas en cada grupo. Los grupos de tratamiento recibieron OLE por el método del “gavage” durante 21 días. Al final del estudio, todas las ratas fueron sacrificadas por dislocación cervical. Las muestras de sangre recogidas del corazón se centrifugaron y se determinaron los niveles de glucosa, colesterol total, triglicéridos, urea, ácido úrico, creatinina, alanina-aminotransferasa (ALT), aspartato-aminotransferasa (AST), hidroperóxido de lípidos (LOOH) y homeostasis de tiol disulfuro. El análisis de la hemoglobina A1c (HbA1c) se realizó en sangre entera. Además, se realizaron pruebas de movimiento de la cola y modelado de placa caliente para indicar la pérdida de percepción del dolor.

Resultados:

se observó que el OLE no tuvo efecto sobre los niveles de glucosa y HbA1c en el suero. Por el contrario, el OLE redujo los niveles de colesterol total (p < 0,01) y urea (p < 0,01), y la latencia de la placa caliente (p < 0,01) de manera significativa. Además, el OLE mostró tendencia a reducir el nivel de LOOH y a aumentar el nivel de tiol de manera dependiente de la dosis (p > 0,05).

Conclusión:

la suplementación con OLE durante 21 días en las cantidades usadas no puede proteger contra la hiperglucemia pero sí puede proteger contra la hipercolesterolemia y el daño tisular causado por la diabetes mellitus en las ratas.

Palabras clave: Hoja de olivo; Oleuropeína; Actividad antidiabética; Olea europaea L

INTRODUCTION

The International Diabetes Federation (IDF) defines diabetes mellitus (DM) as a chronic condition that occurs when insulin cannot be produced or used, and is diagnosed by the elevation of blood glucose. The IDF recognizes DM as one of the most important public health problems of the next millennium. According to the IDF's report, the number of diabetic patients worldwide is approximately 463 million, and this number is projected to reach 578 million by 2030, and 700 million by 2045. Also, DM and its complications caused the death of 4,2 million people in 2019, and it is of late one of the costliest chronic diseases in the world (1).

Many drugs have been used to treat diabetes and delay diabetes complications. However, the high costs and adverse effects of treatment have increased interest in complementary and alternative therapies. Therefore, the number of studies on alternative treatments using medicinal plants has increased recently (2). Among these medicinal plants, olive tree leaves, which are an important source of antioxidants, have been used in traditional treatments for centuries in regions where olives grow (3). Moreover, there are numerous academic studies reporting the benefits of olive leaf in terms of health (4 5-6). However, there are many bioactive molecules in the composition of olive leaves, with the most widely known and recognized compounds in OLE being secoiridoids (oleuropein, ligstrosid, I-methyloleuropein), flavonoids (kaempferol, apigenin, and luteloin), and phenolic compounds (hdyroxytyrosol, tyrosol, and caffeic acid) (7). Several experimental studies have reported the health benefits of these bioactive compounds, and these studies have focused on the antioxidant, antihypertensive, hypoglycemic, hypolipidemic, anti-inflammatory, and antiviral effects of these compounds (8 9 10 11 12-13).

Diabetes is a serious syndrome that may cause chronic complications such as neuropathy, diabetic kidney disease, retinopathy, cardiomyopathy, peripheral artery disease, coronary artery disease, and stroke. Oxidative stress has been an important part of the complex mechanism that leads to the development or aggravation of diabetes and its complications (14). Recently, the relationship between diabetic complications and oxidative stress has been emphasized (3,14,15). Glycemic control is considered the primary therapeutic intervention in DM because hyperglycemia causes an increase in lipid peroxidation, a change in glutathione redox state, and a reduction in antioxidant enzyme activity (8). However, interest in the use of natural antioxidants is increasing considerably to alleviate oxidative damage in diabetes. It is suggested that the negative effects of oxidative stress are reduced by supplementation with natural antioxidants. Therefore, we investigated the effect of olive leaf extract on oxidative damage and the progression of diabetes in a diabetic rat model.

MATERIALS AND METHODS

ANIMALS

Twenty-eight male Wistar albino rats weighing between 180 and 220 g were used in the study, and four rats were housed per cage. Rats were maintained under a 12: 12 light/dark cycle. The laboratory temperature was kept between 20 and 22 oC. The experimental protocol was certified by the Local Animal Ethics Committee in accordance with the Guide for the Care and Use of Experimental Animals. All rats were fed a standard rat diet and tap water ad libitum.

PREPARATION OF OLE AND CONTENT ANALYSIS BY HPLC

The olive leaves used in the study were obtained from the Izmir province, Turkey. The leaves were collected during the Fall, in November. Before the drying process, leaves were washed with distilled water at room temperature (25 oC), and then were stored until the removal of washing water. Then, they were dried at room temperature and granulated by a grinder until a powdered form was obtained. The dried and granulated leaves were treated with 70 % ethanol solution. To achieve fifty grams of dried and ground olive leaves, these were refluxed at 55 °C for 5 hours with 250 mL of 70 % ethanol. After the extraction process, the resolvents were filtrated and evaporated at 60 oC. The prepared samples were stored in the refrigerator at 4 °C during the experiment. The total amount of oleuropein and its equivalent antioxidant capacity in 1 mL of OLE are listed in table I.

Table I.  Total oleuropein amount and antioxidant capacity equivalent per 1 mL in OLE 

EXPERIMENTAL INDUCTION OF DM IN RATS

In experimental rat studies, although higher doses of streptozotocin (STZ) are used in some of them, the common dose of streptozotocin used is in the range of 40-60 mg/kg for STZ-induced diabetic rat models [10,16]. In the present study, experimental diabetes was induced using a single intraperitoneal dose of 50 mg/kg of STZ (Sigma-Aldrich Corp, St Louis, MO, USA) after fasting for 18 hours. To prevent severe hypoglycemia, a 5 % glucose solution was given orally after two hours of the STZ injection. Standard tap water was given instead of glucose solution after eight hours of the STZ injection, and the rats had free access to food and water. DM was induced in all rats injected with STZ, and was defined by the measuring of fasting blood glucose levels three days after the STZ injection. Blood glucose levels higher than 300 mg/dL were considered to be diabetic.

EXPERIMENTAL DESIGN

The rats were allowed to adapt for 10 days before starting the research. After the adaptation period, a total of 28 rats were divided into 4 groups, having 7 rats per group, as follows: control group (C), diabetic control group (DC), diabetic group treated with 200 mg/kg of OLE (D+200), and diabetic group treated with 400 mg/kg of OLE (D+400). Except for the C group, the rats in the other groups were injected with an intraperitoneal single dose of 50 mg/kg of STZ after 18 hours of fasting. The C group was also injected with intraperitoneal citrate buffer solution. For the diabetes model groups, 5 % glucose solution was given orally to prevent severe hypoglycaemia after two hours of the STZ injection. After eight hours of the STZ injection, normal tap water was given instead of glucose solution. The levels of blood glucose were found to be 300 mg/dL or more in all STZ-induced rats at 3 days after the injection. The treatment groups D+200 and D+400 were then treated with single doses of 200 and 400 mg/kg/day of OLE using the gavage method for 21 days, respectively (Fig. 1). At the end of the study, all rats were sacrificed by the cervical dislocation method after intraperitoneal administration of 90: 10 mg/kg of ketamine/xylazine.

Figure 1.  Experimental design. 

BIOCHEMICAL ASSAYS

After 21 days of treatment, eight-hour fasting blood samples were collected from the heart. Blood samples were centrifuged at 2500 rpm for 15 minutes, and blood serum samples were separated. Serum triglycerides, total cholesterol, urea, creatinine, uric acid, AST, and ALT were determined using an automated biochemistry analyzer, and HbA1c was determined by complete blood count analysis using an automated hematology analyzer available at the laboratory of Aydın Adnan Menderes University Research Hospital in Aydın, Turkey. The blood glucose level was determined using a glucometer on blood from a tail vein. In addition, lipid LOOH levels and dynamic thiol/disulfide homeostasis were measured using commercial kits (Triogen Biotech Inch, Istanbul, Turkey). The LOOH assay kit measures the hydroperoxides directly utilizing the redox reactions with ferrous ions. The resulting ferric ions are detected using thiocyanate ion as the chromogen. Dynamic thiol/disulfide homeostasis is a novel automatic and spectrophotometric method developed by Erel and Neşelioglu (17). Reducible disulfide bonds were reduced to form free thiol groups. After reacting with DTNB (5,5p-dithiobis- (2 nitrobenzoic acid), all thiol groups were measured. The disulfide level was calculated as half of the difference between total thiol and native thiol.

STATISTICAL ANALYSIS

Statistical analyses were performed using the Prism program (version 6.01). Statistical significance and significance levels were measured using ANOVA in variables with normal distribution. Tukey's test was used for homogeneous variables, and the Games-Howell test was used for non-homogeneous variables as multiple comparisons test. For non-normal distributions, the Kruskal-Wallis H-test was used (18). Results were expressed as mean ± SEM and median (Q1-Q3) value of the measurements. The criterion for statistical significance was p < 0.05.

RESULTS

EFFECT OF DM AND OLE ON BODY WEIGHT

Body weight changes in each rat group are presented in figure 2. When the initial and final body weights of the rats in each group were compared at the end of 21 days, only the body weight changes in the rats in the C group were found to be significant (p < 0.001). The rats in the C group gained significant body weight as a requirement of growth. In contrast, there were no significant differences between the initial and final body weights in the rats included in the diabetic groups. The initial and final body weight values in all groups are shown in table II. At the beginning of the study, there were no significant differences in initial body weight between rat groups (p > 0.05). But there were significant differences in final body weight between each of the diabetic groups and C group at the end of the study (p < 0.001).

Figure 2.  Changes in body weight. C: control; DC: diabetic control; D+200: diabetes + 200 mg/kg OLE; D+400: diabetes + 400 mg/kg OLE. 

Table II.  Mean values of body weight, and glycemic, lipid profile 

C: control; DC: diabetic control; D+200: diabetes + 200 mg/kg OLE; D+400: diabetes + 400 mg/kg OLE. Values are means ± SD. ap < 0.01 (compared to C group); bp < 0.01 (compared to DC group); cp < 0.05 (compared to D+200 group).

EFFECT OF OLE ON GLYCEMIC AND LIPID PROFILE

Glucose levels in the blood samples obtained from all diabetic groups were significantly higher than in the C group (p < 0.001). However, HbA1c levels were significantly higher in the diabetic groups (p < 0.001) compared to the C group. This indicates that OLE had no effect on the glycemic profile of treated diabetic rats. Also, no significant differences were found in glucose and HbA1c levels between diabetic groups (Table II).

A significant increase in serum total cholesterol level was observed in the DC group when compared with the C group. Serum total cholesterol levels were significantly decreased in the diabetic rats treated with 200 mg/kg of OLE when compared to diabetic control rats (p < 0.01). But this effect was not observed in the diabetic rats treated with 400 mg/kg of OLE. In addition, serum triglycerides levels were examined at the end of the study, and no significant differences were observed in serum triglycerides levels between groups (Table II).

EFFECT OF OLE ON HEPATIC AND RENAL FUNCTION

Serum ALT, urea, and creatinine levels were significantly higher in the rats included in the DC group when compared to the rats in the C group at the end of the study. Increased ALT and AST levels by diabetes induction were reduced by OLE treatment in a dose-dependent manner, but not significantly. However, serum urea levels significantly improved in both diabetic groups treated with OLE compared to the DC group (p < 0.001). Conversely, no significant effect of OLE treatment was observed on serum creatinine levels increased by diabetes induction. In addition, neither diabetes induction nor OLE treatment caused any significant differences in serum uric acid levels (Table III).

Table III.  Mean values of liver and kidney function indicators 

C: control; DC: diabetic control; D+200: diabetes + 200 mg/kg OLE; D+400: diabetes + 400 mg/kg OLE. Values are means ± SD or medians (Q1-Q3). ap < 0.01 (compared to C group); bp < 0.01 (compared to DC group); cp < 0.05 (compared to D+200 group).

EFFECT OF OLE ON ANTIOXIDANT CAPACITY

Dynamic thiol/disulfide homesostasis and serum LOOH levels were determined to observe the effect of OLE treatment on antioxidant capacity. The results showed that OLE increased the level of native and total thiol in a dose-dependent manner, although not significantly. However, the disulfide/thiol ratio tended to decrease in the diabetic rats treated with 200 mg/kg of OLE when compared to both the control and diabetic control rats, whereas the disulfide/thiol ratio increased significantly in the D+400 group as compared to the D+200 group (p < 0.05). In addition, we observed that lipid peroxidation tended to decrease in both groups treated with OLE, although not significantly (Table IV).

Table IV.  Mean values of oxidant status and antioxidant capacity indicators 

C: control; DC: diabetic control; D+200: diabetes + 200 mg/kg OLE; D+400: diabetes + 400 mg/kg OLE. Values are means ± SD. ap < 0.01 (compared to C group); bp < 0.01 (compared to DC group); cp < 0.05 (compared to D+200 group).

EFFECT OF OLE ON PAIN RESPONSE

Loss of pain response and the effect of OLE treatment on pain response were determined using tail flick test and hot plate tests. The results indicated a loss of pain perception, determined by an increased latency time in the hot plate test, associated with neuropathy in the DC group. Treatment with OLE caused a significant decrease in latency times in the hot plate test (p < 0.001). The hot plate latency time value in group C rats was similar to that of rats in both treatment groups. However, STZ induction did not significantly increase tail flick latency time in the diabetic control rats, and OLE treatment did not significantly improve latency time in the tail flick test (Table V).

Table V.  Mean values of latency indicators 

C: control; DC: diabetic control; D+200: diabetes + 200 mg/kg OLE; D+400: diabetes + 400 mg/kg OLE. Values are means ± SD. ap < 0.01 (compared to C group); bp < 0.01 (compared to DC group); cp < 0.05 (compared to D+200 group).

DISCUSSION

DM has gone beyond the point where it can only be seen as a medical problem, and shows multi-dimensional results with acute and chronic complications. It is well known that hyperglycemia causes oxidative stress through mitochondrial dysfunction and increased production of reactive oxygen species (ROS). As a result, oxidative stress and low-grade inflammation contribute significantly to the progression and complications of DM (19). Therefore, glycemic control is considered the primary therapeutic intervention in DM. Due to the side effects and costs of antidiabetic drugs, natural plants such as olive leaves have been reported to be promising in maintaining glycemic control in DM (10). There are two basic theories that explain how olive leaves and their bioactive compounds lower serum glucose levels. The first of these is the view that these bioactive compounds enhance glucosone-dependent insulin secretion (3). The second is that they increase peripheral glucose uptake by AMP-activated protein kinases (AMPK) phosphorilation and glucose transporter type 4 (GLUT4) translocation (20).

Guex et al. (2019) compared the influence of metformin and OLE on blood glucose. They reported that 200 and 400 mg/kg of OLE were efficacious on glycemic control in a similar way to metformin. However, only metformin was reported to be effective on HbA1c (4). Nekooeian et al. (2014) reported that treatment with 20 mg/kg, 40 mg/kg, and 60 mg/kg of oleuropein significantly reduced fasting blood glucose in a type-2 diabetic rat model (21). Moreover, a systematic review and meta-analysis including 8 trials and a total of 162 rats has found that OLE increased insulin levels and decreased blood glucose levels (22). On the contrary, we found that supplementation for 21 days with OLE had no effect on serum glucose or HbA1c in treated diabetic rats (Fig. 3). This difference may be due to a difference in diabetic admission threshold values and in duration of treatment for both sets of rats. In addition, we found that the healthy rats significantly gained body weight as a requirement of growth, while the diabetic rats maintained their initial body weight (Fig. 2). The reason is an insufficient anabolic effect exerted by insulin in diabetic rats.

Figure 3.  Evaluation of glycemia control parameters C: control; DC: diabetic control; D+200: diabetes + 200 mg/kg OLE; D+400: diabetes + 400 mg/kg OLE. *Compared to C group (*p < 0.05, **p < 0.01, ***p < 0.001). #Compared to DC group (#p < 0.05, ##p < 0.01, ###p < 0.001). 

The long-term presence of hyperglycemia causes excessive neuronal glucose uptake, and then increased intracellular glucose drives the alternative polyol pathway. This pathway contributes to increased oxidative stress in peripheral nervous tissues. Thus, neuronal and microvascular endothelial cell damage as induced by hyperglycemia-mediated oxidative stress contributes to the pathogenesis of neuropathy with loss of protective sensation (23). It has been reported that the antioxidants that directly remove free radicals can reduce the formation or progression of ROS reactions, and thus may improve diabetic neuropathy status by reducing oxidative stress (16). However, the antioxidant effect of olive leaves in the literature is remarkable [3,8]. In our model, antioxidant status was indicated by serum LOOH and thiol levels. Thiols (RSH) are a class of organic compounds containing a sulfhydryl group, and may undergo an oxidation reaction to form disulfide (RSSR) bonds under oxidative conditions (17). We found that treatment with olive leaves increased thiol levels and decreased LOOH levels. Also, disulfide and disulfide/thiol levels decreased only with low-dose OLE treatment. Although the improvement seen in the antioxidant indicator is not significant, the loss of pain perception caused by hyperglycemia-induced oxidative stress was significantly prevented in treated diabetic rats (Fig. 6). Similarly, Ristagno et al. (2012) reported that thermal response latency and mechanical withdrawal thresholds changed significantly in parallel with our results (24). Olive leaf may have hyperalgesic effects by preventing neuronal oxidation and inflammation. Regarding this point of view, well-documented studies are needed to explain the accurate mechanism of this effect of treatment with olive leaves.

Figure 4.  Evaluation of lipid profile parameters. C: control; DC: diabetic control; D+200: diabetes + 200 mg/kg OLE; D+400: diabetes + 400 mg/kg OLE. *Compared to C group (*p < 0.05, **p < 0.01, ***p < 0.001). #Compared to DC group (#p < 0.05, ##p < 0.01, ###p < 0.001). 

Figure 5.  Mean values of liver damage indicator. C: control; DC: diabetic control; D+200: diabetes + 200 mg/kg OLE; D+400: diabetes + 400 mg/kg OLE. *Compared to C group (*p < 0.05, **p < 0.01, ***p < 0.001). #Compared to DC group (#p < 0.05, ##p < 0.01, ###p < 0.001). 

Figure 6.  Mean values of latency indicators. C: control; DC: diabetic control; D+200: diabetes + 200 mg/kg OLE; D+400: diabetes + 400 mg/kg OLE. *Compared to C group (*p < 0.05, **p < 0.01, ***p < 0.001). #Compared to DC group (#p < 0.05, ##p < 0.01, ###p < 0.001). 

Lipid profile imbalance is an expected condition in DM. Because insulin has an antilipolytic effect that inhibits the hormone-sensitive lipase in the adipose tissue, as a result, insulin reduces the secretion of free fatty acids from adipose tissue. It also significantly influences lipid metabolism by suppressing intestinal chylomicron and hepatic lipoprotein synthesis, increasing lipoprotein lipase activity and LDL receptor expression in peripheral tissues. Therefore, lipid disorders such as hypertriglyceridemia and hypercholesterolemia occur secondary to effective insulin deficiency (25). However, lipid disorders can cause vascular damage by increasing the formation of ROS, and it is well known that premature cardiovascular disease is one of the most common causes of death in diabetic patients (26,27). Similar to all these data, elevated cholesterol levels (p < 0.01) and a slightly increased lipid peroxidation (p > 0.05) were observed in our diabetic control rats. In the previous studies, oleuropein has been reported to be a peroxisome proliferator-activated receptor α (PPARα) ligand, and oleuropein treatment reduces in rat hepatocytes the activity of the rate-limiting enzyme of cholesterol synthesis, namely, hydroxymethylglutaryyl-CoA (HMG-CoA) reductase (28,29). Moreover, it was reported that treatment with olive leaves significantly decreased serum cholesterol and triglyceride levels in rats fed a high fat diet (11). Similarly, we found that treatment with low-dose olive leaf extract significantly reduced serum total cholesterol (p < 0.01), but resulted in no significant improvement in triglyceride levels (Fig. 4).

Diabetic kidney disease is an important cause of chronic kidney disease and end-stage renal failure worldwide (14). Controling plasma glucose and lipid concentrations in diabetic patients prevents microvascular complications, and therefore the main goal of medical treatment is metabolic control. Hyperlipidemia and increased lipid peroxidation in experimental diabetes models is a well-known and expected condition. Lipid peroxidation products such as malondialdehyde quickly react with biomolecules such as proteins, lipids, and nucleic acids, and may subsequently cause tissue damage (26). However, the synergistic effect of hypertriglyceridemia and hypercholesterolemia is reported to cause kidney damage in a short time in experimental diabetes models. A possible mechanism is that lipid molecules can induce mesangial cell proliferation and excess basement membrane material production, and thus trigger the development of glomerulosclerosis with a reduction in filtration rate (30). Increased serum creatinine levels are a significant marker of renal dysfunction, together with the less specific urea levels, and hypolipidemic agents (statins) are known to reduce urea and creatinine levels by reducing glomerular damage (31). Our findings showed that serum urea and creatinine levels increased in parallel with hyperlipidemia in diabetic control rats. Similar to the findings of Mohammed et al., we found that olive leaf therapy significantly reduced serum urea levels, although it did not reduce serum creatinine levels (Fig. 5) (32). Increased urea levels increase the risk of mortality in cardiovascu lar diseases, and OLE may possibly reduce urea reabsorption by suppressing urea transporters in collecting ducts.

It has been demonstrated that diabetes induction with STZ causes hepatic tissue damage (10,33). This situation is related to an inability to suppress lipolysis in adipose tissue, and the ectopic accumulation of fatty acids released from adipose tissue in the liver. However, increased lipid peroxides accumulate in the liver and increase oxidative stress and liver damage (26). Increased ALT and AST levels are documented as determinants of liver dysfunction, and as indicators of cell leakage, cellular damage, and ruptured cell membrane integrity in the liver (10). Also, it has been reported that triglycerides, uric acid, and HbA1c show a positive correlation with increased ALT (34). In our model, we found that ALT (p < 0.01) and AST (p > 0.05) levels were increased by diabetes induction. Fki et al. (2020) reported that olive leaf therapy exhibited hepatoprotective effects by decreasing ALT and AST levels due to its antioxidant properties (11). Although our findings are in agreement with the study by Fki et al., the decrease in ALT and AST levels was not significant in our treated diabetic rats (Fig. 5).

Recent studies on the relationship between serum uric acid levels and diabetes/diabetic complications are remarkable. Uric acid is the final product of purine metabolism, and is produced in the liver. However, the regulation of blood uric acid levels is largely carried out by the kidney (35). Many studies report that hyperuricemia causes diabetes development or a progression of diabetic complications by increasing oxidative stress (35,36). But, on the contrary, there are other studies that find that uric acid acts as an antioxidant in the early stage of the atherosclerotic process, and is the strongest determinant of plasma antioxidant capacity (37,38). Recently, pre-treatment with oleuropein has been reported to reduce uric acid levels by improving kidney function (39). In the present study, we found that serum uric acid levels were not significantly different between groups. Although there are some studies reporting that olive leaf treatment affects uric acid levels due to its effect on kidney function, further studies are needed to assess the effects on xanthine oxidase enzyme activity.

CONCLUSION

In conclusion, it may be said that OLE supplementation for 21 days does not prevent hyperglycemia in diabetic rats. However, OLE supplementation during 21 days may protect against diabetic complications by reducing the oxidative damage caused by hyperglycemia and hyperlipidemia, due to its antioxidant effect. It is, therefore, necessary to consider the “strong antioxidant” feature of olive leaf mentioned in the literature. Additional experiments are needed to explain the health effects of various treatment dosages of OLE and its bioactive compounds, one of the promising therapeutic agents for diabetic complications.

REFERENCES

1. International Diabetes Federation. IDF Diabetes Atlas (9th ed.);2019. [ Links ]

2. Furman BL, Candasamy M, Bhattamisra SK, Veettil SK. Reduction of blood glucose by plant extracts and their use in the treatment of diabetes mellitus;discrepancies in effectiveness between animal and human studies. J Ethnopharmacol 2020;247: 112264. DOI: 10.1016/j.jep.2019.112264 [ Links ]

3. Acar-Tek N, Ağagündüz D. Olive Leaf (Olea europaea L. folium): Potantial Effect on Glycemia and Lipidemia. Ann Nutr Metab 2020: 1-6. DOI: 10.1159/000505508 [ Links ]

4. Guex CG, Reginato FZ, de Jesus PR, Brondani JC, Lopes GHH, Bauermann LF. Antidiabetic effects of Olea europaea L. leaves in diabetic rats induced by high-fat diet and low-dose streptozotocin. J Ethnopharmacol 2019;235: 1-7. DOI: 10.1016/j.jep.2019.02.001 [ Links ]

5. Araki R, Fujie K, Yuine N, Watabe Y, Nakata Y, Suzuki H, et al. Olive leaf tea is beneficial for lipid metabolism in adults with prediabetes: an exploratory randomized controlled trial. Nutr Res 2019;67: 60-6. DOI: 10.1016/j.nutres.2019.05.003 [ Links ]

6. Almalki DA, Alghamdi SA, Al-Attar AM. Comparative Study on the Influence of Some Medicinal Plants on Diabetes Induced by Streptozotocin in Male Rats. Biomed Res Int 2019;2019: 3596287. DOI: 10.1155/2019/3596287 [ Links ]

7. EMA. Assessment report on Olea europaea L., folium. United Kingdom: Committee on Herbal Medicinal Products (HMPC);2017. Report No: EMA/HMPC//359236/2016. [ Links ]

8. Rouibah Z, Ben Mensour A, Rekik O, Boumendjel M, Taibi F, Bouaziz M, et al. Chemical composition, antioxidant activities, in an allergic asthma model, of Olea europaea L. leaf extracts from Collo (Skikda, Algeria). Drug Chem Toxicol 2019: 1-12. DOI: 10.1080/01480545.2019.1679827 [ Links ]

9. Romero M, Toral M, Gómez-Guzmán M, Jiménez R, Galindo P, Sánchez M, et al. Antihypertensive effects of oleuropein-enriched olive leaf extract in spontaneously hypertensive rats. Food Funct 2016;7(1): 584-93. DOI: 10.1039/C5FO01101A [ Links ]

10. Goboza M, Aboua YG, Chegou N, Oguntibeju OO. Vindoline effectively ameliorated diabetes-induced hepatotoxicity by docking oxidative stress, inflammation and hypertriglyceridemia in type 2 diabetes-induced male Wistar rats. Biomed Pharmacother 2019;112: 108638. DOI: 10.1016/j.biopha.2019.108638 [ Links ]

11. Fki I, Sayadi S, Mahmoudi A, Daoued I, Marrekchi R, Ghorbel H. Comparative Study on Beneficial Effects of Hydroxytyrosol- and Oleuropein-Rich Olive Leaf Extracts on High-Fat Diet-Induced Lipid Metabolism Disturbance and Liver Injury in Rats. Biomed Res Int 2020;2020: 1315202. DOI: 10.1155/2020/1315202 [ Links ]

12. Qabaha K, Al-Rimawi F, Qasem A, Naser SA. Oleuropein Is Responsible for the Major Anti-Inflammatory Effects of Olive Leaf Extract. J Med Food 2018;21(3): 302-5. DOI: 10.1089/jmf.2017.0070 [ Links ]

13. Altındiş M, Aslan FG, Uzuner H, Ünal H, Köroğlu M, Kulaç S, et al. Comparison of Antiviral Effect of Olive Leaf Extract and Propolis with Acyclovir on Herpes Simplex Virus Type 1. Mikrobiyol Bul 2020;54(1): 79-94. [ Links ]

14. Dos Santos JM, Tewari S, Mendes RH. The Role of Oxidative Stress in the Development of Diabetes Mellitus and Its Complications. J Diabetes Res 2019;2019: 4189813. DOI: 10.1155/2019/4189813 [ Links ]

15. Shi Q, Wang J, Cheng Y, Dong X, Zhang M, Pei C. Palbinone alleviates diabetic retinopathy in STZ-induced rats by inhibiting NLRP3 inflammatory activity. J Biochem Mol Toxicol 2020;34(7): e22489. DOI: 10.1002/jbt.22489 [ Links ]

16. Sun Q, Wang C, Yan B, Shi X, Shi Y, Qu L, et al. Jinmaitong Ameliorates Diabetic Peripheral Neuropathy Through Suppressing TXNIP/NLRP3 Inflammasome Activation In The Streptozotocin-Induced Diabetic Rat Model. Diabetes Metab Syndr Obes 2019;12: 2145-55. DOI: 10.2147/DMSO.S223842 [ Links ]

17. Erel O, Neselioglu S. A novel and automated assay for thiol/disulfide homeostasis. Clin Biochem 2014;47: 326-32. DOI: 10.1016/j.clinbiochem.2014.09.026 [ Links ]

18. E Ostertagová, O Ostertag. Methodology and Application of One-way ANOVA. AJME 2013;1(7): 256-61. [ Links ]

19. Teodoro JS, Nunes S, Rolo AP, Reis F, Palmeira CM. Therapeutic Options Targeting Oxidative Stress, Mitochondrial Dysfunction and Inflammation to Hinder the Progression of Vascular Complications of Diabetes. Front Physiol 2019;9: 1857. DOI: 10.3389/fphys.2018.01857 [ Links ]

20. Alkhateeb H, Al-Duais M, Qnais E. Beneficial effects of oleuropein on glucose uptake and on parameters relevant to the normal homeostatic mechanisms of glucose regulation in rat skeletal muscle. Phytother Res 2018;(4): 651-6. DOI: 10.1002/ptr.6012 [ Links ]

21. Nekooeian AA, Khalili A, Khosravi MB. Effects of oleuropein in rats with simultaneous type 2 diabetes and renal hypertension: a study of antihypertensive mechanisms. J Asian Nat Prod Res 2014;16(9): 953-62. DOI: 10.1080/10286020.2014.924510 [ Links ]

22. Abunab H, Dator WL, Hawamdeh S. Effect of olive leaf extract on glucose levels in diabetes-induced rats: A systematic review and meta-analysis. J Diabetes 2017;9(10): 947-57. DOI: 10.1111/1753-0407.12508 [ Links ]

23. Xu C, Hou B, He P, Ma P, Yang X, Yang X, et al. Neuroprotective Effect of Salvianolic Acid A against Diabetic Peripheral Neuropathy through Modulation of Nrf2. Oxid Med Cell Longev 2020;2020: 6431459. DOI: 10.1155/2020/6431459 [ Links ]

24. Ristagno G, Fumagalli F, Porretta-Serapiglia C, OrrùA, Cassina C, Pesaresi M, et al. Hydroxytyrosol attenuates peripheral neuropathy in streptozotocin-induced diabetes in rats. J Agric Food Chem 2012;60(23): 5859-65. DOI: 10.1021/jf2049323 [ Links ]

25. Vergès B. Dyslipidemia in Type 1 Diabetes: A Masked Danger. Trends Endocrinol Metab 2020;31(6): 422-34. DOI: 10.1016/j.tem.2020.01.015 [ Links ]

26. Tang Y, Zhong Z. Obtusifolin treatment improves hyperlipidemia and hyperglycemia: possible mechanism involving oxidative stress. Cell Biochem Biophys 2014;70(3): 1751-7. DOI: 10.1007/s12013-014-0124-0 [ Links ]

27. Rawshani A, Sattar N, Franzén S, Rawshani A, Hattersley AT, Svensson AM, et al. Excess mortality and cardiovascular disease in young adults with type 1 diabetes in relation to age at onset: a nationwide, register-based cohort study. Lancet 2018;392(10146): 477-86. DOI: 10.1016/S0140-6736(18)31506-X [ Links ]

28. Malliou F, Andreadou I, Gonzalez FJ, Lazou A, Xepapadaki E, Vallianou I, et al. The olive constituent oleuropein, as a PPARαagonist, markedly reduces serum triglycerides. J Nutr Biochem 2018;59: 17-28. DOI: 10.1016/j.jnutbio.2018.05.013 [ Links ]

29. Priore P, Siculella L, Gnoni GV. Extra virgin olive oil phenols down-regulate lipid synthesis in primary-cultured rathepatocytes. J Nutr Biochem 2014;25: 683-91. DOI: 10.1016/j.jnutbio.2014.01.009 [ Links ]

30. He L, Hao L, Fu X, Huang M, Li R. Severe hypertriglyceridemia and hypercholesterolemia accelerating renal injury: a novel model of type 1 diabetic hamsters induced by short-term high-fat / high-cholesterol diet and low-dose streptozotocin. BMC Nephrol 2015;16: 51. DOI: 10.1186/s12882-015-0041-5 [ Links ]

31. El-Moselhy MA, El-Sheikh AA. Protective mechanisms of atorvastatin against doxorubicin-induced hepato-renal toxicity. Biomed Pharmacother 2014;68(1): 101-10. DOI: 10.1016/j.biopha.2013.09.001 [ Links ]

32. Mohammed HA, Okail HA, Ibrahim MA, Emam NH. Influences of olive leaf extract in the kidney of diabetic pregnant mice and their offspring. JOBAZ 2018;79: 3. DOI: 10.1186/s41936-018-0024-8 [ Links ]

33. Aldahmash BA, El-Nagar DM, Ibrahim KE. Attenuation of hepatotoxicity and oxidative stress in diabetes STZ-induced type 1 by biotin in Swiss albino mice. Saudi J Biol Sci 2016;23(2): 311-7. DOI: 10.1016/j.sjbs.2015.09.027 [ Links ]

34. Johnson RJ, Nakagawa T, Sanchez-Lozada L, Shafiu M, Sundaram S, Le M, et al. Sugar, uric acid, and the etiology of diabetes and obesity. Diabetes 2013;62: 3307-15. DOI: 10.2337/db12-1814 [ Links ]

35. Xiong Q, Liu J, Xu Y. Effects of Uric Acid on Diabetes Mellitus and Its Chronic Complications. Int J Endocrinol 2019;2019: 9691345. DOI: 10.1155/2019/9691345 [ Links ]

36. Gaita L, Timar R, Lupascu N, Roman D, Albai A, Potre O, et al. The Impact Of Hyperuricemia On Cardiometabolic Risk Factors In Patients With Diabetes Mellitus: A Cross-Sectional Study. Diabetes Metab Syndr Obes 2019;12: 2003-10. DOI: 10.2147/DMSO.S222570 [ Links ]

37. Fabbrini E, Serafini M, Colic Baric I, Hazen SL, Klein S. Effect of plasma uric acid on antioxidant capacity, oxidative stress, and insulin sensitivity in obese subjects. Diabetes 2014;63(3): 976-81. DOI: 10.2337/db13-1396 [ Links ]

38. Kuwabara M, Hisatome I, Niwa K, Bjornstad P, Roncal-Jimenez CA, Andres-Hernando A. The Optimal Range of Serum Uric Acid for Cardiometabolic Diseases: A 5-Year Japanese Cohort Study. J Clin Med 2020;9(4): E942. DOI: 10.3390/jcm9040942 [ Links ]

39. Koc K, Cerig S, Ozek NS, Aysin F, Yildirim S, Cakmak O. The efficacy of oleuropein against non-steroidal anti-inflammatory drug induced toxicity in rat kidney. Environ Toxicol 2019;34(1): 67-72. DOI: 10.1002/tox.22658 [ Links ]

Original article: This study was supported by the Adnan Menderes University Scientific Research Projects Unit, project number 17004.

Gürbüz M, Öğüt S. Antidiabetic effect of olive leaf extract on streptozotocin-induced diabetes mellitus in experimental animals. Nutr Hosp 2020;37(5):1012-1021

Received: February 19, 2020; Accepted: June 20, 2020

Correspondence: Murat Gürbüz. Department of Nutrition and Dietetics. Trakya University. Edirne, Turkey e-mail: muratgurbuz@trakya.edu.tr

Conflict of interests:

The authors declare no conflict of interests.

Creative Commons License This is an open-access article distributed under the terms of the Creative Commons Attribution License