Skip to content
Publicly Available Published by De Gruyter August 6, 2015

Effects of long-term administration of aspartame on biochemical indices, lipid profile and redox status of cellular system of male rats

  • Oluwatosin A. Adaramoye EMAIL logo and Olubukola O. Akanni

Abstract

Background: Aspartame (N-L-α-aspartyl-L-phenylalanine-1-methyl ester) (ASP) is a synthetic sweetener used in foods and its safety remains controversial. The study was designed to investigate the effects of long-term administration of aspartame on redox status, lipid profile and biochemical indices in tissues of male Wistar rats.

Methods: Rats were assigned into four groups and given distilled water (control), aspartame at doses of 15 mg/kg (ASP 1), 35 mg/kg (ASP 2) and 70 mg/kg (ASP 3) daily by oral gavage for consecutive 9 weeks.

Results: Administration of ASP 2 and ASP 3 significantly increased the weight of liver and brain, and relative weight of liver of rats. Lipid peroxidation products significantly increased in the kidney, liver and brain of rats at all doses of ASP with concomitant depletion of antioxidant parameters, viz. glutathione-s-transferase, glutathione peroxidase, superoxide dismutase, catalase and reduced glutathione. Furthermore, ASP 2 and ASP 3 significantly increased the levels of gamma glutamyl transferase by 70% and 85%; alanine aminotransferase by 66% and 117%; aspartate aminotransferase by 21% and 48%; urea by 72% and 58% and conjugated bilirubin by 63% and 64%, respectively. Also, ASP 2 and ASP 3 significantly increased the levels of total cholesterol, triglycerides and low-density lipoprotein cholesterol in the rats. Histological findings showed that ASP 2 and ASP 3 caused cyto-architectural changes such as degeneration, monocytes infiltration and necrotic lesions in brain, kidney and liver of rats.

Conclusions: Aspartame may induce redox and lipid imbalance in rats via mechanism that involves oxidative stress and depletion of glutathione-dependent system.

Introduction

Aspartame (ASP) is an artificial sweetener used in food products as alternative to sugar. Because aspartame is approximately 200 times sweeter than sugar, it is used in low-calorie soft drinks and foods [1]. Aspartame consists of two amino acids (aspartic acid and phenylalanine) and methanol. It was approved by Food Drug Administration (FDA) for use in dry applications in 1981, followed by approval for use in carbonated soft drinks in 1983 and as a general sweetener in 1996 [2]. It is now known that aspartame represents 62% of the value of the intense sweetener market in terms of its world consumption [3], for example, in the US, more than 70% of aspartame sales are attributed to soft drinks [4]. Upon ingestion, aspartame is metabolized by digestive esterases and peptidases in the intestinal lumen to methanol and its constituent amino acids [5]. Subsequent to aspartame consumption, the concentrations of its metabolites are increased in the blood [6]. Ever since its approval by the FDA for use as an artificial sweetener, aspartame has been the subject of much debate with respect to its health effects such as increasing brain cancer rates [7]. It has also been reported that high doses of aspartame can also generate major neurochemical changes in rats [8, 9]. The toxicity of aspartame has been linked to its pro-oxidative effects in animal studies [10]. Generation of excessive reactive oxygen species (ROS) by aspartame has led to a marked increase in pro-apoptotic marker (Bax), whereas decrease in anti-apoptotic marker (Bcl-2) in rats’ brain, indicating that aspartame is harmful at cellular level [11]. ROS are generated continuously during oxidative metabolism in cells. Because of their metabolic rate, vital biochemical functions and high content of oxidizable substrates, some vital organs in the body are very vulnerable to ROS. To prevent the effects of ROS, organisms have evolved multiple systems of antioxidant defence that are essential for cellular metabolism and functions [12]. On the basis of the aforementioned, this study was undertaken to investigate the effects of long-term administration of aspartame on redox status, lipid profile and biochemical indices in kidney, liver and brain of male Wistar rats.

Materials and methods

Chemicals

Aspartame was purchased from a pharmacy store in Berlin, Germany. Thiobarbituric acid (TBA) was procured from Aldrich Chemical Co. (Milwaukee, WI, USA). Glutathione, hydrogen peroxide, 5,5′-dithios-bis-2-nitrobenzoic acid (DNTB) and epinephrine were purchased from Sigma Chemical Co. (Saint Louis, MO USA). Other reagents were of analytical grade and the purest quality available.

Animals

Inbred male Wistar rats weighing between 185 and 193 g were purchased from the animal house of the Department of Veterinary Physiology, Biochemistry and Pharmacology, University of Ibadan, Nigeria. These animals were kept in ventilated cages at room temperature (28 °C–30 °C) and maintained on normal laboratory chow (Ladokun Feeds, Ibadan, Nigeria) and water ad libitum. Rats handling and treatments conform to guidelines of the National Institute of Health (NIH publication 85-23, 1985) for laboratory animal care and use. The study was approved by the Animal Ethics Committee, Faculty of Basic Medical Sciences, University of Ibadan, Nigeria.

Study design

Twenty adult male Wistar rats were randomly divided into four groups of five rats each. The 1st group (control) received drug vehicle (distilled water), whereas the 2nd, 3rd and 4th groups tagged ASP 1, ASP 2 and ASP 3 received ASP at doses of 15, 35 and 70 mg/kg body weight, respectively. ASP was given once daily by oral gavage for consecutive 9 weeks.

Preparation of tissues

The rats were fasted overnight and sacrificed 24 h after the last dose of ASP. Kidney, liver and brain were quickly removed and washed in ice-cold 1.15% KCl solution to remove blood stains, dried and weighed. The tissues were homogenized in four volumes of 50 mM phosphate buffer, pH 7.4 and centrifuged at 10,000 g for 15 min to obtain post-mitochondrial supernatant fraction (PMF). Procedures were carried out at temperature of 4 °C.

Preparation of serum

Blood was collected from the heart of the animals into plain centrifuge tubes and was allowed to stand for 1 h. Serum was prepared by centrifugation at 3000 g for 15 min in a Beckman bench centrifuge (Beckman and Hirsch, Burlington, IO, USA). The clear supernatant was used for the estimation of serum lipids and enzymes.

Biochemical assays

Protein contents of the samples were assayed by the method of Lowry et al. [13], using bovine serum albumin as standard. The activities of alanine and aspartate aminotransferases (ALT and AST) were assayed by the combined methods of Mohun and Cook [14], and Reitman and Frankel [15]. Serum gamma-glutamyl transferase (GGT) and lactate dehydrogenase (LDH) activities were determined by the methods of Fossati et al. [16], and Zimmerman and Weinstein [17], respectively. Serum total cholesterol level was assayed by the method of Richmond et al. [18]. The method involved enzymatic hydrolysis and oxidation of cholesterol with the formation of quinone imine (an indicator) from hydrogen peroxide and 4-aminoantipyrine in the presence of phenol and peroxide. The serum level of triglyceride was determined by Jacobs and Van Demark [19], and Koditschek and Umbreit [20]; this was based on the hydrolysis of triglycerides with the formation of glycerol which is substrate for other enzymes with the subsequent formation of hydrogen peroxide. This then reacts with 4-aminophenazone and 4-chlorophenol in the presence of peroxidase to give quinoneimine which is measured spectrophotometrically at 500 nm. The lipoproteins (measured using the enzymatic colorimetric method), very low-density lipoprotein (VLDL) and low-density lipoprotein (LDL) were precipitated by the addition of phosphotungstic acid and magnesium chloride. After centrifugation at 3000 g for 10 min at 25 °C, the clear supernatant contained HDL fraction, which was assayed for cholesterol with the Randox diagnostic kit. The LDL was calculated using the formula of Friedewald et al. [21]. Lipid peroxidation level was assayed by the reaction between 2-thiobarbituric acid (TBA) and malondialdehyde (MDA), an end product of lipid peroxides as described by Buege and Aust [22]. Superoxide dismutase (SOD) activity was measured by the nitro blue tetrazolium (NBT) reduction method of McCord and Fridovich [23]. Catalase (CAT) activity was assayed spectrophotometrically by measuring the rate of decomposition of hydrogen peroxide at 240 nm as described by Aebi [24]. Reduced glutathione level was measured by the method of Beutler et al. [25]; this method is based on the development of a relatively stable (yellow) colour when 5′,5′-dithiobis-(2-nitrobenzoic acid) (Ellman’s reagent) is added to sulfhydryl compounds. The chromophoric product resulting from the reaction of Ellman’s reagent with the reduced glutathione (2-nitro-5-thiobenzoic acid) possesses a molar absorption at 412 nm which is proportion to the level of reduced glutathione in the test sample. The glutathione peroxidase (GPx) activity was assessed by the method of Rotruck et al. [26], whereas glutathione-S-transferase (GST) activity was determined according to Habig et al. [27]; the principle is based on the fact that all known GST demonstrate a relatively high activity with 1-chloro-2,4-dinitrobenzene as the 2nd substrate. When this substance is conjugated with reduced glutathione, its absorption maximum shifts to a longer wavelength 340 nm and the absorption increase at this wavelength provides a direct measurement of the enzymatic reaction.

Statistical analysis

All values were expressed as the mean±SD of five animals per group. Data were analysed using one-way ANOVA followed by the post-hoc Duncan multiple range test for analysis of biochemical data using SPSS (10.0; SPSS Inc., Chicago, IL, USA). Values were considered statistically significant at p<0.05.

Results

Effects of aspartame on weight and relative weight of organs, and biochemical markers in rats

There were significant increases in the weight of liver (p=0.032; 0.037) and brain (p=0.029; 0.031) in rats treated with ASP 2 and ASP 3 relative to controls. Also, the relative weight of liver significantly increased (p=0.023; 0.027) in ASP 2- and ASP 3-treated rats when compared to controls (Table 1). Administration of ASP 2 and ASP 3 increased the activities of serum alanine and aspartate aminotransferases (ALT and AST) by 1.7-, 1.2- and 2.2-, 1.5-fold, respectively. Furthermore, activities of serum gamma glutamyl transferase (GGT) increased by 70% and 85% in ASP 2- and ASP 3-treated rats, respectively (Table 2). In Table 3, administration of ASP 2 and ASP 3 markedly elevated serum urea and conjugated bilirubin by 72%, 58% and 63%, 64%, respectively, whereas ASP 3 increased serum creatinine level by 57%. However, ASP 1 did not significantly affect the weight of organs (p=0.075–0.093), activities of ALT, AST, GGT and levels of urea, creatinine and conjugated bilirubin (p=0.061–0.11).

Table 1

Changes in the body weight and relative weight of organs of rats treated with aspartame for 9 weeks.

TreatmentBody weight, gWeight of organs, gRelative weight of organs
InitialFinalLiverKidneyBrainLiverKidneyBrain
Control194.00±3.54237.50±2.324.14±0.241.10±0.100.57±0.031.74±0.350.46±0.030.24±0.02
ASP 1193.33±2.70233.70±4.114.69±0.350.97±0.070.59±0.062.01±0.280.42±0.020.25±0.03
ASP 2190.83±3.03241.20±3.055.62±0.5a1.01±0.080.68±0.02a2.33±0.20a0.42±0.030.28±0.04
ASP 3192.09±2.31247.40±3.105.80±0.3a1.06±0.100.71±0.03a2.34±0.31a0.43±0.040.29±0.05

Values are means±SD of five animals per group; asignificantly different from control (p<0.05); ASP 1, aspartame at a dose of 15 mg/kg; ASP 2, aspartame at a dose of 35 mg/kg; ASP 3, aspartame at a dose of 70 mg/kg.

Table 2

Changes in the activities of some serum marker enzymes in rats treated with aspartame for 9 weeks.

TreatmentALT, U/LAST, U/LLDH, U/LGGT, U/L
Control38.3±3.486.7±5.850.2±6.03.55±0.94
ASP 134.5±4.189.3±5.446.0±4.72.79±0.77
ASP 263.4±3.6a105.1±7.451.1±4.56.03±1.00a
ASP 383.2±4.9a128.6±8.3a56.0±3.96.57±1.03a

Values are means±SD of five animals per group; asignificantly different from control (p<0.05); ASP 1, aspartame at a dose of 15 mg/kg; ASP 2, aspartame at a dose of 35 mg/kg; ASP 3, aspartame at a dose of 70 mg/kg; AST, aspartate aminotransferase; ALT, alanine aminotransferase, LDH, lactate dehydrogenase; GGT, gamma glutamyl transferase.

Table 3

Changes in the levels of serum biochemical indices in rats treated with aspartame for 9 weeks.

TreatmentUrea, mg/dLCreatinine, mg/dLTB, mg/dLCB, mg/dLUB, mg/dL
Control32.61±2.882.05±0.181.37±0.030.73±0.040.64±0.02
ASP 130.39±1.221.96±0.271.40±0.050.80±0.030.60±0.05
ASP 256.17±3.15a2.11±0.341.85±0.111.15±0.03a0.70±0.05
ASP 353.10±2.16a3.30±0.36a1.88±0.091.20±0.04a0.68±0.03

Values are means±SD of five animals per group; asignificantly different from control (p<0.05); ASP 1, aspartame at a dose of 15 mg/kg; ASP 2, aspartame at a dose of 35 mg/kg; ASP 3, aspartame at a dose of 70 mg/kg; TB, total bilirubin; CB, conjugated bilirubin; UB, unconjugated bilirubin.

Effects of aspartame on redox status or oxidant/antioxidant balance in rats

Administration of ASP significantly increased (p=0.018–0.025) kidney, hepatic and brain lipid peroxidation (LPO) products measured as TBARS in the rats (Figure 1) with concomitant decrease in the levels of reduced glutathione (Figure 2). Treatment of rats with ASP 2 and ASP 3 caused significant (p=0.024–0.041) reduction in the activities of kidney, hepatic and brain superoxide dismutase and glutathione-s-transferase (Figures 3 and 4), whereas only hepatic and brain catalase and glutathione peroxidase activities were adversely affected by ASP 2 and ASP 3 (p=0.031–0.045) (Figures 5 and 6).

Figure 1: Dose-dependent effects of aspartame on the levels of lipid peroxidation products in tissues of rats after 9 weeks of exposure.*Significantly different from control (p<0.05); ASP 1, aspartame at 15 mg/kg; ASP 2, aspartame at 35 mg/kg; ASP, aspartame at 70 mg/kg; LPO, lipid peroxidation.
Figure 1:

Dose-dependent effects of aspartame on the levels of lipid peroxidation products in tissues of rats after 9 weeks of exposure.

*Significantly different from control (p<0.05); ASP 1, aspartame at 15 mg/kg; ASP 2, aspartame at 35 mg/kg; ASP, aspartame at 70 mg/kg; LPO, lipid peroxidation.

Figure 2: Dose-dependent effects of aspartame on the levels of reduced glutathiones tissues of rats after 9 weeks of exposure.*Significantly different from control (p<0.06). ASP, aspartame at 16 mg/kg; ASP 2, aspartame at 35 mg/kg; ASP 3, aspartame at 70 mg/kg; GSH, reduced glutathione.
Figure 2:

Dose-dependent effects of aspartame on the levels of reduced glutathiones tissues of rats after 9 weeks of exposure.

*Significantly different from control (p<0.06). ASP, aspartame at 16 mg/kg; ASP 2, aspartame at 35 mg/kg; ASP 3, aspartame at 70 mg/kg; GSH, reduced glutathione.

Figure 3: Dose-dependent effects of aspartame on the activities of glutathlone-s-transferase in tissues of rats after 9 weeks of exposure.*Significantly different from control (p<0.05); ASP 1, aspartame at 15 mg/kg; ASP 2, aspartame at 35 mg/kg; ASP 3, aspartame at 70 mg/kg; G3T, glutathione-s-transferase.
Figure 3:

Dose-dependent effects of aspartame on the activities of glutathlone-s-transferase in tissues of rats after 9 weeks of exposure.

*Significantly different from control (p<0.05); ASP 1, aspartame at 15 mg/kg; ASP 2, aspartame at 35 mg/kg; ASP 3, aspartame at 70 mg/kg; G3T, glutathione-s-transferase.

Figure 4: Dose-dependent effects of aspartame on the activities of superoxide dismutase in tissues of rats after 9 weeks of exposure.*Significantly different from control (p<0.05); ASP 1, aspartame at 15 mg/kg; ASP 2, aspartame at 35 mg/kg; ASP 3, aspartame at 70 mg/kg; SOD, superoxide dismutase.
Figure 4:

Dose-dependent effects of aspartame on the activities of superoxide dismutase in tissues of rats after 9 weeks of exposure.

*Significantly different from control (p<0.05); ASP 1, aspartame at 15 mg/kg; ASP 2, aspartame at 35 mg/kg; ASP 3, aspartame at 70 mg/kg; SOD, superoxide dismutase.

Figure 5: Dose-dependent effects of aspartame on the activities of glutathione peroxidase in tissues of rats after 9 weeks of exposure.*Significantly different from control (p<0.05); ASP 1, aspartame at 15 mg/kg; ASP 2, aspartame at 36 mg/kg; ASP 3, aspartame at 70 mg/kg; GPx, glutathione peroxidase.
Figure 5:

Dose-dependent effects of aspartame on the activities of glutathione peroxidase in tissues of rats after 9 weeks of exposure.

*Significantly different from control (p<0.05); ASP 1, aspartame at 15 mg/kg; ASP 2, aspartame at 36 mg/kg; ASP 3, aspartame at 70 mg/kg; GPx, glutathione peroxidase.

Figure 6: Dose-dependent effects of aspartame on the activities of catalase in tissues of rats after 9 weeks of exposure.*Significantly different from control (p<0.05); ASP 1, aspartame at 15 mg/kg; ASP 2, aspartame at 35 mg/kg; ASP 3, aspartame at 70 mg/kg; CAT, catalase.
Figure 6:

Dose-dependent effects of aspartame on the activities of catalase in tissues of rats after 9 weeks of exposure.

*Significantly different from control (p<0.05); ASP 1, aspartame at 15 mg/kg; ASP 2, aspartame at 35 mg/kg; ASP 3, aspartame at 70 mg/kg; CAT, catalase.

Effects of aspartame on lipid profile of rats

Administration of aspartame at doses of 35 and 70 mg/kg/day (ASP 2 and ASP 3) for consecutive 9 weeks significantly increased the levels of serum total cholesterol, triglycerides and LDL-cholesterol by 1.3-, 1.5-, 2.4-fold and 1.8-, 1.4-, 2.5-fold, respectively (p=0.031–0.042). In addition, rats fed on aspartame at a dose of 70 mg/kg/day had their serum HDL-C levels decreased by 33% relative to control. In contrast, aspartame at a dose of 15 mg/kg/day (ASP 1) did not elicit adverse affects on the lipid profile of the rats (Table 4).

Table 4

Changes in the levels of serum lipid parameters in rats treated with aspartame for 9 weeks.

TreatmentTC, U/LTG, U/LHDL-C, U/LLDL-C, U/L
Control205.1±11.6162.0±9.562.3±7.3110.4±6.3
ASP 1197.8±13.3183.2±13.058.1±5.5103.1±8.0
ASP 2375.0±15.0a247.6±10.1a59.3±7.0266.2±11.4a
ASP 3361.3±12.2a231.0±9.7a42.1±4.7a273.0±10.7a

Values are means±SD of five animals per group; asignificantly different from Control (p<0.05); ASP 1, aspartame at a dose of 15 mg/kg; ASP 2, aspartame at a dose of 35 mg/kg; ASP 3, aspartame at a dose of 70 mg/kg; TC, total cholesterol; TG, triglycerides; HDL-C, high-density lipoprotein cholesterol; LDL-C, low-density lipoprotein cholesterol.

Effects of aspartame on the cyto-architecture of brain, liver and kidney of rats

Figure 7 indicates that the brain tissues from control group have normal architecture with no visible lesions. However, brain tissues from ASP 2 and ASP 3 groups contained sub-dural and focal hemorrhagic lesions, whereas mild lesions were seen in the brain of rats given ASP 1. In Figure 8, no visible lesions were seen in liver tissues from control and ASP 1 groups. Administration of ASP 2 and ASP 3 caused severe necrosis, periportal infiltration, congestion and dilatation of sinusoids of the hepatocytes. Figure 9 shows adequate, well-preserved glomeruli and normal architecture of kidney tissues in the control and ASP 1 groups. However, ASP 2 and ASP 3 caused severe congestion of vessel in the cortex and medulla as well as hyperplasia of the cuboidal cells of the renal tissues.

Figure 7: Photomicrographs of rat brain showing control (A) (no visible lesions), ASP 1 (B) (mild lesions), ASP 2 (C) (subdural haemorrhagic lesion) and ASP 3 (D) (focal area haemorrhagic lesion) × 400 ASP 1, ASP 2 and ASP 3=15, 35 and 70 mg/kg ASP, respectively.
Figure 7:

Photomicrographs of rat brain showing control (A) (no visible lesions), ASP 1 (B) (mild lesions), ASP 2 (C) (subdural haemorrhagic lesion) and ASP 3 (D) (focal area haemorrhagic lesion) × 400 ASP 1, ASP 2 and ASP 3=15, 35 and 70 mg/kg ASP, respectively.

Figure 8: Photomicrographs of rat liver showing control (A) (no visible lesions), ASP 1 (B) (mild lesions), ASP 2 (C) periportal infiltration, congestion and dilatation of sinusoid, ASP 3 (D) (severe necrosis, congestion of vessels and sinusoid) × 400 ASP 1, ASP 2 and ASP 3=15, 35 and 70 mg/kg ASP, respectively.
Figure 8:

Photomicrographs of rat liver showing control (A) (no visible lesions), ASP 1 (B) (mild lesions), ASP 2 (C) periportal infiltration, congestion and dilatation of sinusoid, ASP 3 (D) (severe necrosis, congestion of vessels and sinusoid) × 400 ASP 1, ASP 2 and ASP 3=15, 35 and 70 mg/kg ASP, respectively.

Figure 9: Photomicrographs of rat kidney showing control (A) (adequate and well-preserved glomeruli in the cortex), ASP 1 (B) (preserved glomeruli and moderate congestion of vessels), ASP 2 (C) (congestion of vessel in the cortex and medulla), ASP 3 (D) (severe congestion of vessel in cortex and medulla, and hyperplasia of the cuboidal cells) × 400 ASP 1, ASP 2 and ASP 3=15, 35 and 70 mg/kg ASP, respectively.
Figure 9:

Photomicrographs of rat kidney showing control (A) (adequate and well-preserved glomeruli in the cortex), ASP 1 (B) (preserved glomeruli and moderate congestion of vessels), ASP 2 (C) (congestion of vessel in the cortex and medulla), ASP 3 (D) (severe congestion of vessel in cortex and medulla, and hyperplasia of the cuboidal cells) × 400 ASP 1, ASP 2 and ASP 3=15, 35 and 70 mg/kg ASP, respectively.

Discussion

The present results give further data to support the idea that aspartame may induce redox imbalance, altered biochemical indices and lipid profile in rats after long-term administration. The findings were supported by both biochemical and histological data. Aspartame is widely used as an artificial sweetener in low-calorie foods and soft drinks. Several tests on the safety and toxicity of aspartame have been performed and the results are conflicting. The toxicity of any agent is based on the fact that toxicity increases in a dose-dependent manner and that explains the reason for using three dose of aspartame in this study (15, 35 and 70 mg/kg). The aspartame doses used in this study are very relevant to human. The acceptable daily intake levels of aspartame established by European Food Safety Authority and US Food and Drug Administration are 40 and 50 mg/kg/day, respectively [28]. The main question is whether aspartame could be harmful at common or abuse usage levels. The first carcinogenetic effect of aspartame in an animal model was reported by Soffritti et al. [29] using bioassays at a dose of 40 mg/kg body weight/day, which falls within the doses chosen for this study. Abdel-Salam et al. [30] reported that the administration of aspartame alone or in the presence of a mild systemic inflammatory response increases oxidative stress and inflammation in the brain. This observation was consistent with the findings of the present study, in which aspartame at all doses increased lipid peroxidation (LPO) products in the kidney, liver and brain of rats. The finding derives its importance from the fact that increased oxidative stress has been linked to several ailments such as diabetes, cancer, neurodegenerative diseases or other age-related cognitive deficits [31, 32]. The prime targets for free radical reactions during aspartame metabolism are the unsaturated bonds in membrane lipids. Consequent peroxidation results in a loss in membrane fluidity and receptors alignment, suggesting oxidative damage to macromolecules such as lipids [33]. Oxidative stress can be the result of increased free radicals production or alternatively decreased endogenous antioxidants. In this study, we found that the levels of reduced glutathione and activities of antioxidant enzymes such as superoxide dismutase, catalase, glutathione-s-transferase and glutathione peroxidase were significantly reduced in the liver and brain of animals treated with ASP 2 and ASP 3. This observation is consistent with the findings of Abhilash et al. [34]. GSH is an important non-enzymatic antioxidant that plays a critical role in the maintenance of thiol redox potential in cells, keeping sulfhydryl groups of cytosolic protein in the reduced form, and plays a crucial role in the detoxification of toxic chemicals of endogenous and exogenous origin. GSH directly reacts with radicals in non-enzymatic reactions and is the electron donor of peroxides catalyzed by glutathione peroxidase. In addition to free radical produced during aspartame metabolism causing oxidative stress and depletion of antioxidant enzymes, Kruse [35] reported that among the aspartame metabolites, methanol is the toxicant that causes systemic toxicity. The primary metabolic fate of methanol is the direct oxidation to formaldehyde and then into formate. Skrzydlewska [36] revealed that cellular glutathione content and activity of glutathione-dependent enzymes were decreased in methanol intoxication which is the hall mark of aspartame toxicity. Therefore, decreased levels of GSH or activities of GSH-dependent enzymes observed in this study may be as a result of the damaging effect of free radicals produced after methanol exposure or could be a direct effect of formaldehyde formed from the oxidation of methanol on these enzymes. In this study, treatment of rats with aspartame at doses of 35 and 70 mg/kg caused significant increase in the levels of liver and kidney function markers such as creatinine, blood urea nitrogen and conjugated bilirubin relative to controls, thus indicating hepato-renal dysfunction. These injuries could be due to production of free radicals or involvement of oxidative stress during aspartame metabolism. Aspartame may influence the various metabolic pathways of liver thereby enhancing the levels of total cholesterol and triglycerides as noticed in the present study. Also, oxidative damage to kidney by aspartame may contribute to the observed high concentrations of blood urea nitrogen and creatinine in serum. Similar results have been reported in earlier studies where it was suggested that kidney dysfunction due to ASP administration occurred via oxidative stress [37]. The adverse effects of ASP on kidney as observed in this study may be detrimental to health of diabetic subjects that consume aspartame regularly in soft drinks. It is known that prolonged and untreated diabetes are characterised by high serum urea and creatinine [38]. Based on the present study, consumption of ASP by diabetic patients will further aggravate the health conditions of these individuals. Aspartame at doses of 35 mg/kg and above caused hepatotoxicity as clearly indicated by the significant increase in levels of ALT, AST and GGT. Serum levels of transaminases is generally considered as sensitive markers of liver function and their concentrations are increased in the serum because of their cytoplasmic nature and are thus released into blood due to changes in the permeability of hepatocyte membranes. Increased level of LDH in serum in the present investigation apparently indicated the toxic effects of aspartame at doses of 35 and 70 mg/kg in the rats. Histology results showed the presence of lesions in liver, kidney and brain of rats treated with aspartame at doses of 35 and 70 mg/kg, which correlate with the levels of liver and kidney function biochemical markers in the rats.

In conclusion, we believe that the metabolism of aspartame may induce oxidant/antioxidant imbalance due to free radicals generated by aspartame. Also, depletion of both enzymic and non-enzymic antioxidant parameters observed in the tissues of the rats after long-term intake of aspartame is a clear sign of oxidative stress. It has been observed that in addition to the population with diabetes mellitus, children and pregnant women are among the major consumers of aspartame; these findings may likely have important health implications because of the widespread use of aspartame in foods and beverage preparations. However, this is a single study with few animals, therefore, more studies are necessary to confirm the present effects, as well as, the mechanisms responsible for the aspartame toxicity.


Corresponding author: Dr. Oluwatosin A. Adaramoye, Department of Biochemistry, University of Ibadan, Ibadan, Nigeria, Phone: +234-808-838-2846, Fax: +234-2-810-3043, E-mail: ; ; and Drug Metabolism and Toxicology Research Laboratories, Department of Biochemistry, College of Medicine, University of Ibadan, Ibadan, Nigeria

Acknowledgments

The authors are grateful to the technical assistance of Messers E. Sabo and O.K. Ajiboye of the Department of Biochemistry, University of Ibadan, Nigeria.

Author contributions: The authors have accepted responsibility for the entire content of this submitted manuscript and approved submission.

Research funding: None declared.

Employment or leadership: None declared.

Honorarium: None declared.

Competing interests: The funding organization(s) played no role in the study design; in the collection, analysis and interpretation of data; in the writing of the report or in the decision to submit the report for publication.

References

1. Horio Y, Sun Y, Liu C, Saito T, Kurasaki M. Aspartame-induced apoptosis in PC12 cells. Environ Toxicol Pharmacol 2014;37: 158–65.10.1016/j.etap.2013.11.021Search in Google Scholar

2. Butchko HH, Stargel WW, Comer CP, Mayhew DA, Benninger C, Blackburn GL, et al. Aspartame: review of safety. Regul Toxicol Pharmacol 2002;35:S1–93.10.1006/rtph.2002.1542Search in Google Scholar

3. Fry J. The world market for intense sweeteners. World Rev Nutr Diet 1999;85:201–11.10.1159/000059691Search in Google Scholar

4. American Dietetic Association. Position of the American Dietetic Association: use of nutritive and non-nutritive sweeteners. J Am Diet Assoc 2004;104:225–75.10.1016/j.jada.2003.12.001Search in Google Scholar

5. Ranney RE, Opperman JA, Muldoon E, McMahon FG. Comparative metabolism of aspartame in experimental animals and humans. J Toxicol Environ Health 1976;2:441–51.10.1080/15287397609529445Search in Google Scholar

6. Stegink LD. The aspartame story: a model for the clinical testing of a food additive. Am J Clin Nutr 1987;46:204–15.10.1093/ajcn/46.1.204Search in Google Scholar

7. Olney JW, Farber NB, Spitznagel E, Robins LN. Increasing brain cancer rates: is there a link to aspartame? J Neuropathol Exp Neurol 1996;55:1115–23.10.1097/00005072-199611000-00002Search in Google Scholar

8. Yokogoshi H, Roberts CH, Caballero B, Wurtman RJ. Effects of aspartame and glucose administration on brain and plasma levels of large neutral amino acids and brain 5-hydroxyindoles. Am J Clin Nutr 1984;40:l–7.10.1093/ajcn/40.1.1Search in Google Scholar

9. Coulombe RA Jr, Sharma RP. Neurobiochemical alterations induced by the artificial sweetener aspartame (NutraSweet). Toxicol Appl Pharmacol 1986;83:79–85.10.1016/0041-008X(86)90324-8Search in Google Scholar

10. Prokic MD, Paunovic MG, Matic MM, Djordjevic NZ, Ognjanovic BI, Stajn AS, et al. Prooxidative effects of aspartame on antioxidant defense status in erythrocytes of rats. J Biosci 2014;39:859–66.10.1007/s12038-014-9487-zSearch in Google Scholar PubMed

11. Ashok I, Sheeladevi R. Biochemical responses and mitochondrial mediated activation of apoptosis on long-term effect of aspartame in rat brain. Redox Biol 2014;2:820–31.10.1016/j.redox.2014.04.011Search in Google Scholar PubMed PubMed Central

12. Poljšak B, Fink R. The protective role of antioxidants in the defence against ROS/RNS-mediated environmental pollution. Oxid Med Cell Longev 2014;2014:671539.10.1155/2014/671539Search in Google Scholar

13. Lowry OH, Rosebrough NJ, Farr AL, Randall RJ. Protein measurement with the Folin phenol reagent. J Biol Chem 1951;193: 265–75.10.1016/S0021-9258(19)52451-6Search in Google Scholar

14. Mohun AF, Cook LJ. Simple method for measuring serum level of glutamate-oxaloacetate and glutamate-pyruvate transaminases in laboratories. J Clin Pathol 1957;10:394–9.10.1136/jcp.10.4.394Search in Google Scholar

15. Reitman S, Frankel S. A colorimetric method for the determination of serum level of glutamate-oxaloacetate and pyruvate transaminases. Am J Clin Pathol 1957;28:56–63.10.1093/ajcp/28.1.56Search in Google Scholar

16. Fossati R, Melzid’Eril GV, Turenghi G, Precipe L, Berti G. A kinetic colorimetric assay of gamma-glutamyltransferase. Clin Chem 1986;32:1581–4.10.1093/clinchem/32.8.1581Search in Google Scholar

17. Zimmerman HJ, Weinstein BS. Lactic dehydrogenase activity in human serum. J Lab Clin Med 1956;48:607–9.Search in Google Scholar

18. Richmond W. Preparation and properties of a cholesterol oxidase from Nocardia sp. and its application to the enzymatic assay of total cholesterol in serum. Clin Chem 1973;19:1350–6.10.1093/clinchem/19.12.1350Search in Google Scholar

19. Jacobs NJ, Van Demark PJ. The purification and properties of the alpha-glycerophosphate-oxidizing enzyme of Streptococcus faecalis 10C1. Arch Biochem Biophys 1960;88:250–5.10.1016/0003-9861(60)90230-7Search in Google Scholar

20. Koditschek LK, Umbreit WW. Alpha-glycerophosphate oxidase in Streptococcus faecium F 24. J Bacteriol 1969;98:1063–8.10.1128/jb.98.3.1063-1068.1969Search in Google Scholar

21. Friedewald WT, Levi RI, Fredrickson DS. Estimation of the concentration of low density lipoproteins cholesterol in plasma without use of the ultracentrifuge. Clin Chem 1972;18:499–502.10.1093/clinchem/18.6.499Search in Google Scholar

22. Buege JA, Aust SD. Microsomal lipid peroxidation. Methods Enzymol 1978;52:302–10.10.1016/S0076-6879(78)52032-6Search in Google Scholar

23. McCord JM, Fridovich I. Superoxide dismutase, an enzymatic function for erythrocuperin. J Biol Chem 1969;244:6049–55.10.1016/S0021-9258(18)63504-5Search in Google Scholar

24. Aebi H. Catalase. In: Bergmeyer HV, editor. Methods of enzymatic analysis. New York: Verlag Chemie, 1974:673–84.10.1016/B978-0-12-091302-2.50032-3Search in Google Scholar

25. Beutler E, Duron O, Kellin BM. Improved method for the determination of blood glutathione. J Lab Clin Med 1963;61:882–8.Search in Google Scholar

26. Rotruck JT, Pope AL, Ganther HE, Swanson AB, Hafeman DG, Hoekstra WG. Selenium: biochemical role as a component of glutathione peroxidase. Science 1973;179:588–90.10.1126/science.179.4073.588Search in Google Scholar

27. Habig WH, Pabst MJ, Jakoby WB. Glutathione-S-transferases. The first enzymatic step in mercapturic acid formation. J Biol Chem 1974;249:7130–9.10.1016/S0021-9258(19)42083-8Search in Google Scholar

28. Magnuson BA, Burdock GA, Doull J, Kroes RM, Marsh GM, Pariza MW, et al. Aspartame: a safety evaluation based on current use levels, regulations, and toxicological and epidemiological studies. Crit Rev Toxicol 2007;37:629–37.10.1080/10408440701516184Search in Google Scholar PubMed

29. Soffritti M, Belpoggi F, Degli Esposti D, Lambertini L, Tibaldi E, Rigano A. First experimental demonstration of the multi potential carcinogenic effects of aspartame administered in the feed to Sprague-Dawley rats. Environ Health Perspect 2006;114: 379–85.10.1289/ehp.8711Search in Google Scholar PubMed PubMed Central

30. Abdel-Salam OM, Salem NA, Hussein JS. Effect of aspartame on oxidative stress and monoamine neurotransmitter levels in lipopolysaccharide-treated mice. Neurotox Res 2012;21: 245–55.10.1007/s12640-011-9264-9Search in Google Scholar PubMed

31. Liu J, Head E, Gharib AM, Yuan W, Ingersoll RT, Hagen TM, et al. Memory loss in old rats is associated with brain mitochondrial decay and RNA-DNA oxidation: partial reversal by feeding acetyl-L-carnitine and/or R-α-lipoic acid. Proc Natl Acad Sci 2002;99:2356–61.10.1073/pnas.261709299Search in Google Scholar PubMed PubMed Central

32. Besler HT, Como Lu S. Lipoprotein oxidation, plasma total antioxidant capacity and homocysteine level in patients with multiple sclerosis. Nutr Neurosci 2006;6:189–96.10.1080/1028415031000115945Search in Google Scholar

33. Machlin LJ, Bendich A. Free radical tissue damage: protective role of antioxidant nutrients. FASEB J 1987;1:441–5.10.1096/fasebj.1.6.3315807Search in Google Scholar

34. Abhilash M, Paul MV, Varghese MV, Nair RH. Long-term consumption of aspartame and brain antioxidant defense status. Drug Chemical Toxicol 2013;36:135–40.10.3109/01480545.2012.658403Search in Google Scholar PubMed

35. Kruse JA. Methanol poisoning. Intensive Care Med 1992;18: 391–7.10.1007/BF01694340Search in Google Scholar PubMed

36. Skrzydlewska E. Toxicological and metabolic consequences of methanol poisoning. Toxicol Mech Methods 2003;13:277–93.10.1080/713857189Search in Google Scholar PubMed

37. Bilton R. Averting comfortable lifestyle crises. Sci Prog 2013;96:319–68.10.3184/003685013X13743292107915Search in Google Scholar PubMed

38. Kumar D, Agarwal S, Karoli R, Siddiqui M, Shukla V, Khanduri S. Relationship between carotid intima thickness and silent cerebral infarction in patients with type 2 diabetic nephropathy. J Assoc Physicians India 2014;62:316–22.Search in Google Scholar

Received: 2014-12-15
Accepted: 2015-5-18
Published Online: 2015-8-6
Published in Print: 2016-1-1

©2016 by De Gruyter

Downloaded on 1.5.2024 from https://www.degruyter.com/document/doi/10.1515/jbcpp-2014-0130/html
Scroll to top button