7
Evaluation of the Effects of Fructose on Oxidative Stress and Inflammatory Parameters in Rat Brain Abigail Lopes & Thais Ceresér Vilela & Luciane Taschetto & Franciele Vuolo & Fabricia Petronilho & Felipe Dal-Pizzol & Emilio Luiz Streck & Gustavo Costa Ferreira & Patrícia Fernanda Schuck Received: 10 January 2014 /Accepted: 11 March 2014 # Springer Science+Business Media New York 2014 Abstract Hereditary fructose intolerance is an autosomal reces- sive disorder characterized by the accumulation of fructose in tissues and biological fluids of patients. The disease results from a deficiency of aldolase B, responsible for metabolizing fructose in the liver, kidney, and small intestine. We investigated the effect of acute fructose administration on oxidative stress and neuroinflammatory parameters in the cerebral cortex of 30-day- old Wistar rats. Animals received subcutaneous injection of sodium chloride (0.9 %) (control group) or fructose solution (5 μmol/g) (fructose group). One hour later, the animals were euthanized and the cerebral cortex was isolated. Oxidative stress (levels of thiobarbituric acid-reactive substances (TBA-RS), carbonyl content, nitrate and nitrite levels, 2,7- dihydrodichlorofluorescein (DCFH) oxidation, glutathione (GSH) levels, as well as the activities of catalase (CAT) and superoxide dismutase (SOD)) and neuroinflammatory parame- ters (TNF-α, IL-1β, and IL-6 levels and myeloperoxidase (MPO) activity) were investigated. Acute fructose administration increased levels of TBA-RS and carbonyl content, indicating lipid peroxidation and protein damage. Furthermore, SOD activ- ity increased, whereas CAT activity was decreased. The levels of GSH, nitrate, and nitrite and DCFH oxidation were not altered by acute fructose administration. Finally, cytokines IL-1β, IL-6, and TNF-α levels, as well as MPO activity, were not altered. Our present data indicate that fructose provokes oxidative stress in the cerebral cortex, which induces oxidation of lipids and proteins and changes of CAT and SOD activities. It seems therefore reasonable to propose that antioxidants may serve as an adjuvant therapy to diets or to other pharmacological agents used for these patients, to avoid oxidative damage to the brain. Keywords Brain . Fructosemia . Oxidative damage . Neuroinflammation . Hereditary fructose intolerance Introduction Hereditary fructose intolerance (HFI, OMIM 229600) is an autosomal-recessive disorder with an average incidence of 1:40,000 newborns [1]. The disease arises from a deficiency of aldolase B (EC 4.1.2.13), an enzyme responsible for me- tabolizing fructose in the liver, kidney, and small intestine, and it is biochemically characterized by the accumulation of fruc- tose and fructose-1-phosphate in biological fluids and tissues of affected patients [2, 3]. The main clinical and biochemical presentation of patients includes hypoglycemia, vomiting, jaundice, liver failure, hepatomegaly, metabolic acidosis, sei- zures, coma, and eventually death [38]. Furthermore, A. Lopes : T. C. Vilela : L. Taschetto : P. F. Schuck (*) Laboratório de Erros Inatos do Metabolismo, Programa de Pós-Graduação em Ciências da Saúde, Unidade Acadêmica de Ciências da Saúde, Universidade do Extremo Sul Catarinense, Avenida Universitária, 1105, Bloco S, Sala 6, 88806-000 Criciúma, SC, Brazil e-mail: [email protected] F. Vuolo : F. Dal-Pizzol Laboratório de Fisiopatologia Experimental, Programa de Pós-Graduação em Ciências da Saúde, Unidade Acadêmica de Ciências da Saúde, Universidade do Extremo Sul Catarinense, Criciúma, SC, Brazil F. Petronilho Laboratório de Fisiopatologia Clínica e Experimental, Programa de Pós-Graduação em Ciências da Saúde, Universidade do Sul de Santa Catarina, Tubarão, SC, Brazil E. L. Streck Laboratório de Bioenergética, Programa de Pós-Graduação em Ciências da Saúde, Unidade Acadêmica de Ciências da Saúde, Universidade do Extremo Sul Catarinense, Criciúma, SC, Brazil G. C. Ferreira Universidade Federal do Rio de Janeiro, Instituto de Biofísica Carlos Chagas Filho, Avenida Carlos Chagas Filho, 373, Bloco G, Cidade Universitária, Ilha do Fundão 21941-902, RJ, Brazil Mol Neurobiol DOI 10.1007/s12035-014-8676-y

Evaluation of the Effects of Fructose on Oxidative Stress and Inflammatory Parameters in Rat Brain

Embed Size (px)

Citation preview

Page 1: Evaluation of the Effects of Fructose on Oxidative Stress and Inflammatory Parameters in Rat Brain

Evaluation of the Effects of Fructose on Oxidative Stressand Inflammatory Parameters in Rat Brain

Abigail Lopes & Thais Ceresér Vilela & Luciane Taschetto & Franciele Vuolo &

Fabricia Petronilho & Felipe Dal-Pizzol & Emilio Luiz Streck & Gustavo Costa Ferreira &

Patrícia Fernanda Schuck

Received: 10 January 2014 /Accepted: 11 March 2014# Springer Science+Business Media New York 2014

Abstract Hereditary fructose intolerance is an autosomal reces-sive disorder characterized by the accumulation of fructose intissues and biological fluids of patients. The disease results froma deficiency of aldolase B, responsible for metabolizing fructosein the liver, kidney, and small intestine.We investigated the effectof acute fructose administration on oxidative stress andneuroinflammatory parameters in the cerebral cortex of 30-day-old Wistar rats. Animals received subcutaneous injection ofsodium chloride (0.9 %) (control group) or fructose solution(5 μmol/g) (fructose group). One hour later, the animals wereeuthanized and the cerebral cortex was isolated. Oxidativestress (levels of thiobarbituric acid-reactive substances

(TBA-RS), carbonyl content, nitrate and nitrite levels, 2′,7′-dihydrodichlorofluorescein (DCFH) oxidation, glutathione(GSH) levels, as well as the activities of catalase (CAT) andsuperoxide dismutase (SOD)) and neuroinflammatory parame-ters (TNF-α, IL-1β, and IL-6 levels and myeloperoxidase(MPO) activity) were investigated. Acute fructose administrationincreased levels of TBA-RS and carbonyl content, indicatinglipid peroxidation and protein damage. Furthermore, SOD activ-ity increased, whereas CATactivity was decreased. The levels ofGSH, nitrate, and nitrite andDCFHoxidationwere not altered byacute fructose administration. Finally, cytokines IL-1β, IL-6, andTNF-α levels, as well as MPO activity, were not altered. Ourpresent data indicate that fructose provokes oxidative stress in thecerebral cortex, which induces oxidation of lipids and proteinsand changes of CAT and SOD activities. It seems thereforereasonable to propose that antioxidants may serve as an adjuvanttherapy to diets or to other pharmacological agents used for thesepatients, to avoid oxidative damage to the brain.

Keywords Brain . Fructosemia . Oxidative damage .

Neuroinflammation . Hereditary fructose intolerance

Introduction

Hereditary fructose intolerance (HFI, OMIM 229600) is anautosomal-recessive disorder with an average incidence of1:40,000 newborns [1]. The disease arises from a deficiencyof aldolase B (EC 4.1.2.13), an enzyme responsible for me-tabolizing fructose in the liver, kidney, and small intestine, andit is biochemically characterized by the accumulation of fruc-tose and fructose-1-phosphate in biological fluids and tissuesof affected patients [2, 3]. The main clinical and biochemicalpresentation of patients includes hypoglycemia, vomiting,jaundice, liver failure, hepatomegaly, metabolic acidosis, sei-zures, coma, and eventually death [3–8]. Furthermore,

A. Lopes : T. C. Vilela : L. Taschetto : P. F. Schuck (*)Laboratório de Erros Inatos do Metabolismo, Programa dePós-Graduação em Ciências da Saúde, Unidade Acadêmica deCiências da Saúde, Universidade do Extremo Sul Catarinense,Avenida Universitária, 1105, Bloco S, Sala 6, 88806-000 Criciúma,SC, Brazile-mail: [email protected]

F. Vuolo : F. Dal-PizzolLaboratório de Fisiopatologia Experimental, Programa dePós-Graduação em Ciências da Saúde, Unidade Acadêmica deCiências da Saúde, Universidade do Extremo Sul Catarinense,Criciúma, SC, Brazil

F. PetronilhoLaboratório de Fisiopatologia Clínica e Experimental, Programa dePós-Graduação em Ciências da Saúde, Universidade do Sul de SantaCatarina, Tubarão, SC, Brazil

E. L. StreckLaboratório de Bioenergética, Programa de Pós-Graduação emCiências da Saúde, Unidade Acadêmica de Ciências da Saúde,Universidade do Extremo Sul Catarinense, Criciúma, SC, Brazil

G. C. FerreiraUniversidade Federal do Rio de Janeiro, Instituto de Biofísica CarlosChagas Filho, Avenida Carlos Chagas Filho, 373, Bloco G, CidadeUniversitária, Ilha do Fundão 21941-902, RJ, Brazil

Mol NeurobiolDOI 10.1007/s12035-014-8676-y

Page 2: Evaluation of the Effects of Fructose on Oxidative Stress and Inflammatory Parameters in Rat Brain

neurological impairment may also be observed in HFI pa-tients, particularly during the acute phase of the disease [9].

Fructose enters hepatocytes and other cells (including tu-bular cells, adipocytes, and intestinal epithelial cells), where itis phosphorylated to fructose-1-phosphate by fructokinasewith the consumption of ATP [10]. A depletion of intracellularphosphate was observed in HFI patients after fructose admin-istration [11], which can inhibit the action of hepatic glycogenphosphorylase and, consequently, glycogenolysis [12]. Thatwould lead to hypoglycemia, which is considered the mostprominent symptom of IHF [13]. Moreover, the retroactiveinhibition of fructokinase by fructose-1-phosphate results inreduced uptake of fructose by the liver, leading to increasedfructose levels in the plasma of patients [13].

Studies have demonstrated that high-fructose intake caninduce weight gain, development of insulin resistance, andmetabolic syndrome [10, 14–19]. However, little is knownregarding the effects of fructose in the brain. Some early studiessuggested that fructose could not penetrate the blood–brainbarrier in significant amounts [20, 21]. In contrast, accumulat-ing evidence indicates that neural cells are able to metabolizefructose [22]. In this context, fructose intake was shown todisrupt plasma membranes of rat neurons impairing neuronalfunction [19]. It has been also demonstrated that a high-fructosediet has detrimental consequences for synaptic plasticity; im-pairs the cognitive function, memory, dendritic spine density,and neurogenesis in the hippocampus; as well as induces neu-ronal loss in the nucleus tractus solitaries of rats [23–28].

Therefore, considering that fructose is the main accumulatingmetabolite in HFI and that high-fructose diet can lead to oxidativestress in the heart and liver of rats [29–31], in the present work,weevaluated different oxidative stress parameters, namely thiobarbi-turic acid-reactive substances (TBA-RS), carbonyl content, nitriteand nitrate content, 2′,7′-dihydrodichlorofluorescein (DCFH) ox-idation, and reduced glutathione (GSH) levels, as well as theactivities of the antioxidant enzymes catalase (CAT) and super-oxide dismutase (SOD) in homogenates from the cerebral cortexof young rats in the hope to contribute to a better understanding ofpathomechanisms found in HFI patients. We also investigatedwhether inflammatory response is present following fructoseadministration, by evaluating TNF-α, IL-1β, and IL-6 levelsand myeloperoxidase (MPO) activity. To our knowledge, this isthe first study evaluating the effect of acute high fructose concen-trations on cell redox and inflammation in the brain of rats.

Methods

Reagents

All chemicals were purchased from Sigma-Aldrich (St. Louis,MO, USA), unless stated in the text. Fructose was dissolvedon the day of the experiments with its pH adjusted to 7.4.

Animals

For the experiments, a total of 36 30-day-old male Wistar ratsobtained from the Central Animal House of Universidade doExtremo Sul Catarinense were used. Rats were kept with damsuntil weaning at 21 days of age. The animals had free access towater and to a standard commercial chow and were main-tained on a 12:12-h light/dark cycle in an air-conditionedconstant temperature (22±1 °C) colony room. The Guide forthe Care and Use of Laboratory Animals (National ResearchCouncil, 2011) and the EC Directive 86/609/EEC werefollowed in all experiments. All efforts were made to mini-mize the number of animals used and their suffering. Thestudy was approved by the Local Ethical Committee on An-imal Use for Research under the protocol number 51/2012.

In Vivo Experiments

The animals were divided into two groups (six animals pergroup): control group, which received a single subcutaneousinjection of saline solution (0.9 %), and fructose group, whichreceived a single subcutaneous injection of 5 μmol/g of fruc-tose (body weight 0.9 mg/g) [32]. One hour after the admin-istration, the animals were euthanized by decapitation withoutanesthesia, and the brains were rapidly excised on a Petri dishplaced on ice and the cerebral cortex was isolated. The struc-ture was weighed and homogenized in 10 volumes (1:10,w/v)of 20 mM sodium phosphate buffer, pH 7.4 containing140 mM KCl. The homogenate was centrifuged at 750×gfor 10 min at 4 °C to discard nuclei and cell debris. Aliquotswere taken to measure the values of oxidative stress parame-ters (levels of TBA-RS, carbonyl content, nitrate and nitritelevels, DCFH oxidation, GSH levels, as well as the activitiesof CAT and SOD) and inflammatory parameters (TNF-α, IL-1β and IL-6 levels and MPO activity). Blood samples werealso collected in order to obtain serum aliquots for the deter-mination of fructose concentrations.

Determination of Fructose Serum Concentrations

The determination of fructose serum concentrations was per-formed by a commercial kit. Levels of fructose are expressedas milligrams per milliliter and micromoles per milliliter.

Levels of TBA-RS

TBA-RS were determined according to the method ofEsterbauer and Cheeseman [33]. A calibration curve wasperformed using 1,1,3,3-tetramethoxypropane, and eachcurve point was subjected to the same treatment as superna-tants. Values of TBA-RS were calculated as nanomoles ofTBA-RS per milligram of protein.

Mol Neurobiol

Page 3: Evaluation of the Effects of Fructose on Oxidative Stress and Inflammatory Parameters in Rat Brain

Determination of Protein Carbonyl Formation Content

Protein carbonyl content formation, a marker of oxidizedproteins, was measured spectrophotometrically according toReznick and Packer [34]. The results were calculated asnanomoles of carbonyls groups per milligram of protein, usingthe extinction coefficient of 22,000×106 nmol mL−1 for ali-phatic hydrazones.

Nitrate and Nitrite Determination

Nitrate and nitrite concentrations were determined accordingto Miranda et al. [35], using the Griess reagent. A calibrationcurve was performed using sodium nitrate, and each curvepoint was subjected to the same treatments as supernatants,and the concentrations were calculated nanomoles per milli-gram of protein.

DCFH Oxidation

Reactive species production was assessed according to Lebelet al. [36] by using 2′,7′-dihydrodichlorofluorescein diacetate(DCF-DA). A calibration curve was performed with standardDCF (0.25–10 mM), and the levels of reactive species werecalculated as picomoles of DCF formed per milligram ofprotein.

GSH Concentrations

GSH concentrations were measured according to Browne andArmstrong [37]. Calibration curve was prepared with standardGSH (0.01–1 mM) and the concentrations were calculated asnanomoles per milligram of protein.

CATActivity

CAT (EC 1.11.1.6) activity was assayed according to Aebi[38] by measuring the H2O2 absorbance decrease at 240 nm.The specific activity was expressed as nanomoles per minuteper milligram of protein.

SOD Activity

SOD (EC 1.15.1.1) activity was determined according toBannister and Calabrese [39] using a spectrophotometric as-say based on superoxide-dependent oxidation of epinephrineto adrenochrome at 32 °C. SOD specific activity is represent-ed as nanomoles per minute per milligram of protein.

Inflammatory Parameters

Determination of the dosage of TNF-α, IL-1β, and IL-6 wasperformed using enzyme-linked immunosorbent assay

(ELISA) by commercial kits. Levels of cytokines areexpressed as picograms per milliliter.

MPO (EC 1.11.2.2) activity was determined according toLiaudet et al. [40]. Results are expressed as milliunits permilligram of protein.

Statistical Analysis

Results are presented as mean ± standard error of the mean.Data were analyzed using Student’s t test for independentsamples. Differences between groups were rated significantat p<0.05. All analyses were carried out in an IBM-compatible PC computer using the Statistical Package forthe Social Sciences (SPSS) software for Windows 20.0.

Results

The serum fructose concentrations in the animals submitted tothe model of fructosemia described by Monteiro et al. [32]were first determined. It was found that these concentrationsreached approximately 3.05 μmol/mL (0.55 mg/mL, n=3animals).

Our next step was to evaluate whether acute fructose ad-ministration elicits oxidative damage to the biological com-pounds in the cerebral cortex (Fig. 1). It is observed in Fig. 1that animals receiving fructose acutely showed increased

Fig. 1 Effect of acute administration of fructose on the levels of TBA-RS(a) and carbonyl content (b) in the cerebral cortex of 30-day-old rats.Values are means ± standard deviation for six independent experimentsperformed in duplicate or triplicate and are expressed as nanomoles permilligram of protein. *p<0.05, **p<0.01 compared to control group(Student’s t test for independent samples)

Mol Neurobiol

Page 4: Evaluation of the Effects of Fructose on Oxidative Stress and Inflammatory Parameters in Rat Brain

levels of TBA-RS [p=0.013] (Fig. 1a) and carbonyl content[p=0.021] (Fig. 1b) in the cerebral cortex.

Next, we assessed the influence of acute fructose adminis-tration on DCF production and observed that this carbohy-drate did not influence DCFH oxidation (Table 1). We thenevaluated the effects of acute fructose administration on ni-trate and nitrite production, in order to verify if this metabolitecould induce reactive nitrogen species generation, and thesame lack of interference was observed.

Our next step was to investigate the in vivo effects of acutefructose administration on important cellular antioxidant de-fenses (Fig. 2). It was observed that the levels of GSH, themain nonenzymatic antioxidant cellular defense, were notaltered by the administration of this carbohydrate (Fig. 2a).Regarding the enzymatic antioxidant defenses, it was verifiedthat acute fructose administration inhibited CAT activity [p=0.0449] (Fig. 2b), whereas SOD activity was increased by thistreatment [p=0.0165] in the cerebral cortex (Fig. 2c).

Finally, aiming to verify whether rats receiving fructoseacutely develop neuroinflammation, the levels of pro-inflammatory mediators (TNF-α, IL-1β, IL-6) and markerof neutrophil activation, MPO activity, in rat (Fig. 3) werealso investigated. It was observed that the levels of TNF-α,IL-1β, and IL-6, as well as MPO activity, were not altered inthe fructose group, as compared to the control group.

Discussion

Liver, kidney, and small intestine are the main sites of fructosemetabolism [41]. Toxicity of fructose, after intravenous ad-ministration at high doses, results in hyperuricemia,hyperlactatemia, and ultrastructural alterations in liver andintestinal cells [42–44]. Furthermore, animal studies haveshown that a high-fructose diet induces well-characterizedmetabolic syndrome resulting in hyperinsulinemia, insulinresistance, glucose tolerance reduction, hypertension,hypertriglyceridemia, and decreased high-density lipoproteincholesterol [45, 46].

Despite the number of studies investigating the peripheraleffects of fructose, the understanding of brain complications

related to this monosaccharide is still poor. Since oxidativestress and neuroinflammation are involved in the pathophys-iology of common neurodegenerative disorders [47, 48] andof some inborn errors of metabolism [49, 50] and that high-fructose diet can lead to oxidative stress in peripheral tissues[29–31], in the present study, we evaluated the in vivo influ-ence of acute fructose administration on several parameters ofoxidative stress, as well as neuroinflammation, in the cerebralcortex of young rats.

In this work, our first step was to reproduce the animalmodel described by Monteiro et al. [32], used for fructosemiastudies. We measured serum fructose concentrations in this ratmodel and found that this sugar reached a level as high as0.55 mg/mL (control values≈0.14 mg/mL). It is important tomention that brain fructose concentrations are approximatelyseven times lower than plasma levels [20]. These concentra-tions are in line with the fructose levels of patients [51],indicating that this animal model is suitable for preclinicalstudies of fructosemia effects.

Klein et al. [20] evaluated the distribution of intravenouslyinjected fructose between blood and brain in cats and observedthat the highest level of brain fructose was obtained about 1 hafter injection of this sugar and that appreciable levels wereobtained in some experiments about 30 min after injection.We then evaluate biochemical parameters 1 h after the fructoseadministration.

We chose 30-day-old rats in this study since this age in ratscorresponds to 2.5 years of life in humans [52], a life periodthat usually comprises the onset of clinical symptoms inpatients affected by fructosemia due to a higher content offructose in their diet. Herein, we demonstrate that after acutefructose administration, the levels of TBA-RS and carbonylcontent were increased. TBA-RS reflect the content ofmalondialdehyde, the most abundant individual aldehyderesulting from lipid breakdown due to lipid peroxidationprocess [53]. On the other hand, the level of carbonyl groupsof proteins is widely used as a marker of oxidative proteindamage [53]. Thus, our data indicate that acute fructose ad-ministration elicits oxidative damage to lipids and proteins inthe cerebral cortex of rats.

It was also observed in this study that the concentration offructose used was not able to increase the DCF levels. DCFHoxidation is widely used as an indicator of the formation ofreactive species, particularly H2O2, RO·, OH·, HOCl, andONOO− [54]. However, there are limitations to the interpre-tation of DCF production as a specific marker for quantitativeintracellular H2O2 formation. For example, the H2O2-depen-dent oxidation of DCFH to DCF occurs slowly, if at all, in theabsence of ferrous iron [55]. DCF formation is greatly en-hanced in the presence of heme-containing substances, suchas hematin, peroxidases, or cytochrome c [55, 56]. Regardingthe effects of fructose on reactive nitrogen species generation,it was observed that nitrate and nitrite production was

Table 1 Effects of the acute fructose administration on DCFH oxidationand nitrate and nitrite production

DCFH oxidation Nitrate and nitrite content

Control 170.56±20.29 15.05±1.87

Fructose 159.71±17.08 12.54±1.12

The experiments were performed in triplicate, and data represent mean ±standard deviation of the mean (n=6 animals/group). Any differencebetween groups (Student’s t test for independent samples) was not ob-served. DCFH oxidation is in picomoles per minute per milligram ofprotein; nitrate and nitrite content is nanomoles per milligram of protein

Mol Neurobiol

Page 5: Evaluation of the Effects of Fructose on Oxidative Stress and Inflammatory Parameters in Rat Brain

unaffected by acute fructose administration, therefore indicat-ing that RNS production was not altered by fructose.

Although the above results show oxidant activity, fructosedid not affect GSH concentrations in the brain. Since thisparameter is used to evaluate a tissue’s capacity to prevent orrespond to damage associated to reactive species [57]. Inter-estingly, similar findings were previously reported by Kundeet al. [31] in the liver of mice receiving a high-fructose diet,which showed increased levels of TBA-RS without howeveraltering GSH levels.

On the other hand, we observed that CAT activity wasinhibited in the cerebral cortex of rats receiving fructoseacutely, while SOD activity was increased. CAT is a ferricheme protein that directly catalyzes the decomposition ofhydrogen peroxide. It has been suggested that CAT activitymay be decreased secondarily to CAT saturation during thebreakdown of free radicals and hydrogen peroxide or theinhibition of CAT by these radicals [58]. SOD removes the

anion superoxide by accelerating the rate of its dismutation tohydrogen peroxide [53]. In this context, elevated SOD/CATratio may result in an increase of H2O2 concentration orincreased rate of H2O2 production and may lead to lipid andprotein oxidation, resulting in increased neuronal damage[59]. This finding corroborates the increased levels of TBA-RS and carbonyl content, however, without affecting DCFformation.

Considering that fructose may activate the inflammatorycascade in hepatocytes of mice [60] and hamsters [61] byincreasing the concentrations of TNF-α and Jun amino termi-nal kinase (JNK) [18], we assessed whether neuroinflamma-tion could also play a role in the brain following fructoseexposure. We did not observe alterations in the levels ofinflammatory markers (TNF-α, IL-1β, and IL-6 levels andMPO activity) in the cerebral cortex of rats receiving fructoseacutely, as compared to control animals. Exposure of rathepatocytes to fructose for 2 to 4 h caused the activation of

Fig. 2 Effect of acuteadministration of fructose onGSH levels (a), catalase activity(b), and superoxide dismutaseactivity (c) in the cerebral cortexof 30-day-old rats. Values aremeans ± standard deviation for sixindependent experimentsperformed in duplicate and areexpressed as nanomoles permilligram of protein (GSH levels)and nanomoles per minute permilligram of protein (catalase andsuperoxide dismutase activities).*p<0.05 compared to controlgroup (Student’s t test forindependent samples)

Fig. 3 Effect of acuteadministration of fructose on IL-1β (a), IL-6 (b), and TNF-α (c)levels and myeloperoxidaseactivity (d) in the cerebral cortexof 30-day-old rats. Values aremeans ± standard deviation for sixindependent experimentsperformed in duplicate and areexpressed as picomoles permilliliter (a–c) and milliunits permilligram of protein (d).Difference between groups wasnot detected (Student’s t test forindependent samples)

Mol Neurobiol

Page 6: Evaluation of the Effects of Fructose on Oxidative Stress and Inflammatory Parameters in Rat Brain

JNK [18]. Then, we suggest that only 1 h after administrationof fructose was not sufficient to activate inflammatory path-ways and further studies should be undertaken to investigatethe effects of sustained exposition to fructose in the brain.

Since oxidative stress results from an imbalance betweenthe total antioxidant defense of the tissue and the reactivespecies generated, our present data strongly indicate that fruc-tose provokes oxidative stress in vivo in the cerebral cortex,which induces oxidation of lipids and proteins and changes inthe CAT and SOD activity. It should be emphasized that thebrain has low antioxidant defenses as compared to othertissues, rendering this tissue more vulnerable to oxidativedamage [62].

At present, we cannot establish whether our data have apathophysiological significance, since brain concentrations offructose in HFI patients are unknown. It should however benoted that the significant alterations of the oxidative stressparameters elicited by fructose occurred at high micromolarlevels (5 μmol/g), and during metabolic crises in patients, theconcentrations of the accumulating metabolites dramaticallyincrease [8].

Conclusion

In conclusion, the present data indicate that oxidative stress isinduced by fructose in the brain of young rats. It seemstherefore reasonable to propose that antioxidants may serveas an adjuvant therapy to specific diets or to other pharmaco-logical agents used for these patients, especially during crises,to avoid oxidative damage to the brain.

Acknowledgments This study was supported by grants from ConselhoNacional de Pesquisa e Desenvolvimento (CNPq), Universidade doExtremo Sul Catarinense (UNESC), and Núcleo de Excelência emNeurociências Aplicadas de Santa Catarina (NENASC project,PRONEX). We thank Soliany Grassi Maravai for the determination offructose levels in the serum of the animals.

Conflict of Interest The authors declare that they have no conflict ofinterest.

References

1. Scriver CR, Beaudt AL, Sly WL, Valle D (2001) The metabolic andmolecular bases of inherited disease, 8th edn. McGraw-Hill, NewYork

2. Hommes FA (1993) Inborn errors of fructose metabolism. Am J ClinNutr 58:788–795

3. Cox TM (1994)Aldolase B and fructose intolerance. Faseb J 8:62–714. Chambers RA, Pratt RTC (1956) Idiosyncrasy to fructose. Lancet 2:

3405. Lameire N,MusscheM, Baele G, Kint J, Ringoir S (1978) Hereditary

fructose intolerance: a difficult diagnosis in the adult. Am J Med 65:416–423

6. Morris RC (1968) An experimental renal acidification defect inpatients with hereditary fructose intolerance. II. Its distinction fromclassic renal tubular acidosis; its resemblance to the renal acidifica-tion defect associated with the Fanconi syndrome of children withcystinosis. J Clin Invest 47:1648

7. Odièvre M, Gentil C, Gautier M, Alagille D (1978) Hereditaryfructose intolerance in childhood. Diagnosis, management, andcourse in 55 patients. Am J Dis Child 132:605–608

8. Steinmann B, Gitzelmann R, Van den Berghe G (2001) Disorders offructose metabolism. In: Scriver CR, Beaudt AL, Sly WL, Valle D(eds) The metabolic and molecular bases of inherited disease, 8thedn. McGraw-Hill, New York

9. Labrune P, Chatelon S, Huguet P, OdievreM (1990)Unusual cerebralmanifestations in hereditary fructose intolerance. Arch Neurol 47:1243–1244

10. Johnson RJ, Segal MS, Sautin Y, Nakagawa T, Feig DI, Kang DH,Gersch MS, Benner S, Sánchez-Lozada LG (2007) Potential role ofsugar (fructose) in the epidemic of hypertension, obesity and themetabolic syndrome, diabetes, kidney disease, and cardiovasculardisease. Am J Clin Nutr 86:899–906

11. Marthaler TM, Froesch ER (1967) Hereditary fructose intolerance.Dental status of eight patients. Br Dent J 123:597

12. Oberhaensli RD, Rajagopalan B, Taylor DJ, Radda GK, Collins JE,Leonard JV, Schwarz H, Herschkowitz N (1987) Study of hereditaryfructose intolerance by use of 31P magnetic resonance spectroscopy.Lancet 2:931–934

13. Mayatepek E, Hoffmann B, Meissner T (2010) Inborn errors ofcarbohydrate metabolism. Best Pract Res Clin Gastroenterol 24:607–618

14. Pagliassotti MJ, Prach PA, Koppenhafer TA, Pan DA (1996)Changes in insulin action, triglycerides, and lipid composition duringsucrose feeding in rats. Am J Physiol Regul Integr Comp Physiol271:1319–1326

15. Reaven GM (1988) Banting lecture. Role of insulin resistance inhuman disease. Diabetes 37:1595–1607

16. Elliott SS, Keim NL, Stern JS, Teff K, Havel PJ (2002) Fructose,weight gain, and the insulin resistance syndrome. Am J Clin Nutr 76:911–922

17. den Boer M, Voshol PJ, Kuipers F, Havekes LM, Romijn JA (2004)Hepatic steatosis: a mediator of the metabolic syndrome. Lessonsfrom animal models. Arterioscler Thromb Vasc Biol 24:644–649

18. Rutledge AC, Adeli K (2007) Fructose and the metabolic syndrome:pathophysiology and molecular mechanisms. Nutr Rev 65:13–23

19. Agrawal R, Pinilla FG (2012) Metabolic syndrome in the brain:deficiency in omega-3 fatty acid exacerbates dysfunctions in insulinreceptor signalling and cognition. J Physiol 590:2485–2499

20. Klein J, Hurwitz R, Olsen N (1946) Distribution of intravenouslyinjected fructose and glucose between blood and brain. J Biol Chem164:509–512

21. Thurston JH, Levy CA, Warren SK, Jones EM (1972)Permeability of the blood–brain barrier to fructose and the anaerobicuse of fructose in the brains of young mice. J Neurochem 19:1685–1696

22. Funari VA, Crandall JE, Tolan DR (2007) Fructose metabolism in thecerebellum. Cerebellum 6:130–140

23. Cao D, Lu H, Lewis TL, Li L (2007) Intake of sucrose-sweetenedwater induces insulin resistance and exacerbates memory deficits andamyloidosis in a transgenic mouse model of Alzheimer disease. JBiol Chem 282:36275–36282

24. Stranahan AM, Norman ED, Lee K, Cutler RG, Telljohann R, EganJM, Mattson MP (2008) Diet-induced insulin resistance impairshippocampal synaptic plasticity and cognition in middle-aged rats.Hippocampus 18:1085–1088

25. Ross AP, Bartness TJ, Mielke JG, Parent MB (2009) A high fructosediet impairs spatial memory in male rats. Neurobiol Learn Mem 92:410–416

Mol Neurobiol

Page 7: Evaluation of the Effects of Fructose on Oxidative Stress and Inflammatory Parameters in Rat Brain

26. Stephan BC, Wells JC, Brayne C, Albanese E, Siervo M (2010)Increased fructose intake as a risk factor for dementia. J Gerontol ABiol Sci Med Sci 65:809–814

27. Van der Borght K, Köhnke R, Göransson N, Deierborg T, Brundin P,Erlanson-Albertsson C, Lindqvist A (2011) Reduced neurogenesis inthe rat hippocampus following high fructose consumption. RegulPept 167:26–30

28. Rafati A, Anvari E, Noorafshan A (2013) High fructose solutioninduces neuronal loss in the nucleus of the solitary tract of rats.Folia Neuropathol 51:214–221

29. Busserolles J, Rock E, Gueux E, Mazur A, Grolier P, Rayssiguier Y(2002) Short-term consumption of a high sucrose diet has a pro-oxidant effect in rats. Br J Nutr 87:337–342

30. Mellor K, Ritchie RH, Meredith G, Woodman OL, Morris MJ,Delbridge LM (2010) High-fructose diet elevates myocardial super-oxide generation in mice in the absence of cardiac hypertrophy.Nutrition 26:842–848

31. Kunde SS, Roede JR, Vos MB, Orr ML, Go YM, Park Y, Ziegler TR,Jones DP (2011) Hepatic oxidative stress in fructose-induced fatty liveris not caused by sulfur amino acid insufficiency. Nutrients 3:987–1002

32. Monteiro AA, Biella MS, Bristot SF, Streck EL, Schuck PF, FerreiraGC (2012) Characterization of the biochemical profile in serum ofyoung rats submitted to high concentrations of fructose. Rev InovaSaúde 1:116–129

33. Esterbauer H, Cheeseman KH (1990) Determination of aldehydiclipid peroxidation products: malonaldehyde and 4-hydroxynonenal.Methods Enzymol 186:407–421

34. Reznick AZ, Packer L (1994) Oxidative damage to proteins: spec-trophotometric method for carbonyl assay. Methods Enzymol 233:357–363

35. Miranda KM, Espey MG, Wink DA (2001) A rapid, simple spectro-photometric method for simultaneous detection of nitrate and nitrite.Nitric Oxide 5:62–71

36. Lebel CP, Ischiropoulos H, Bondy SC (1992) Evaluation of the probe2′,7′-dichlorofluorescin as an indicator of reactive oxygen speciesformation and oxidative stress. Chem Res Toxicol 5:227–231

37. Browne RW, Armstrong D (1998) Reduced glutathione and glutathi-one disulfide. Methods Mol Biol 108:347–352

38. Aebi H (1984) Catalase in vitro. Methods Enzymol 105:121–12639. Bannister JV, Calabrese L (1987) Assays for superoxide dismutase.

Methods Biochem Anal 32:279–31240. Liaudet L, Mabley JG, Soriano FG, Pacher P, Marton A, Haskó G,

Szabó C (2001) Inosine reduces systemic inflammation and improvessurvival in septic shock induced by cecal ligation and puncture. Am JRespir Crit Care Med 164:1213–1220

41. Coffee EM, Yerkes L, Ewen EP, Zee T, Tolan DR (2009) Increasedprevalence of mutant null alleles that cause hereditary fructose intol-erance in the American population. J Inherit Metab Dis 33:33–42

42. Fox IH, Kelley WN (1972) Studies on the mechanism of fructose-induced hyperuricemia in man. Metabolism 21:713

43. Kogut MD, Roe TF, Won NG, Donnell GN (1975) Fructose-inducedhyperuricemia observations in normal children and in patients withhereditary fructose intolerance and galactosemia. Pediatr Res 9:774

44. Vandercammen A, van Schaftingen E (1990) The mechanism bywhich rat liver glucokinase is inhibited by the regulatory protein.Eur J Biochem 191:483

45. Yokozawa T, Kim HJ, Cho EJ (2008) Gravinol ameliorates high-fructose-induced metabolic syndrome through regulation of lipid

metabolism and proinflammatory state in rat. J Agric Food Chem56:5026–5032

46. Chan SM, Sun RQ, Zeng XY, Choong ZH,Wang H,Watt MJ, Ye JM(2013) Activation of PPARα ameliorates hepatic insulin resistanceand steatosis in high fructose-fed mice despite increased endoplasmicreticulum stress. Diabetes 62:2095–2105

47. Perez-Severiano F, Rios C, Segovia J (2000) Striatal oxidative dam-age parallels the expression of a neurological phenotype in micetransgenic for the mutation of Huntington’s disease. Brain Res 862:234–237

48. Mancuso M, Coppede F, Migliore L, Siciliano G, Murri L (2006)Mitochondrial dysfunction, oxidative stress and neurodegeneration. JAlzheimer Dis 10:59–73

49. Latini A, Scussiato K, Rosa RB, Leipnitz G, Llesuy S, Bello- KleinA, Dutra-Filho CS, Wajner M (2003) Induction of oxidative stress byL-2-hydroxyglutaric acid in rat brain. J Neurosci Res 74:103–110

50. Sgaravatti AM, Sgarbi MB, Testa CG, Durigon K, Pederzolli CD,Prestes CG, Wyse AT, Wannmacher CM, Wajner M, Dutra-Filho CS(2007) Gama-hydroxybutyric acid induces oxidative stress in cere-bral cortex of young rats. Neurochem Int 50:564–570

51. Levin B, Oberholzer VG, Snodgrass GJAI, Stimmler L, Wilmers MJ(1963) Fructosaemia, an inborn error of fructose metabolism. ArchDis Child 38:220–230

52. Andreollo NA, dos Santos EF, Araújo MR, Lopes LR (2012) Rat’sage versus human’s age: what is the relationship? Arq Bras Cir Dig 5:49–51

53. Halliwell B (2011) Role of free radicals in the neurodegenerativediseases: therapeutic implications for antioxidant treatment. DrugsAging 18:685–716

54. Cadenas E, Davies KJ (2000) Mitochondrial free radical generation,oxidative stress, and aging. Free Radic Biol Med 29:222–230

55. Rothe G, Valet G (1990) Flow cytometric analysis of respiratory burstact ivi ty in phagocytes with hydroethidine and 2 ′ ,7 ′ -dichlorofluorescin. J Leukoc Biol 47:440–448

56. Huang X, Frenkel K, Klein CB, Costa M (1993) Nickel inducesincreased oxidants in intact cultured mammalian cells as detectedby dichlorofluorescein fluorescence. Toxicol Appl Pharmacol 120:29–36

57. Halliwell B, ClementMV, Long LH (2000) Hydrogen peroxide in thehuman body. FEBS Lett 486:10–13

58. Eraslan G, Saygi S, Essìz D, Aksoy A, Gul H, Macit E (2007)Evaluation of aspect of some oxidative stress parameters using vita-min E, proanthocyanidin and N-acetylcysteine against exposure tocyfluthrin in mice. Pestic Biochem Physiol 88:43–49

59. Halliwell B (1991) Reactive oxygen species in living systems:source, biochemistry, and role in human disease. Am J Med 91:14–22

60. Spruss A, Kanuri G, Wagnerberger S, Haub S, Bischoff SC,Bergheim I (2009) Toll-like receptor 4 is involved in the develop-ment of fructose-induced hepatic steatosis in mice. Hepatology 50:1094–1104

61. Tsai J, Zhang R, Qiu W, Su Q, Naples M, Adeli K (2009)Inflammatory NF-kB activation promotes hepatic apolipoproteinB100 secretion: evidence for a link between hepatic inflammationand lipoprotein production. Am J Physiol Gastrointest Liver Physiol296:G1287–G1298

62. Halliwell B, Gutteridge JMC (1996) Oxygen radicals and nervoussystem. Trends Neurosci 8:22–26

Mol Neurobiol