Iron overload alters glucose homeostasis, causes liver ...€¦ · Iron overload alters glucose...

8
Iron overload alters glucose homeostasis, causes liver steatosis, and increases serum triacylglycerols in rats Maísa Silva a , Marcelo E. Silva a,b , Heberth de Paula a , Cláudia Martins Carneiro c , Maria Lucia Pedrosa a,d, a Núcleo de Pesquisas em Ciências Biológicas, Universidade Federal de Ouro Preto, 35400-000 Ouro Preto, MG, Brazil b Departamento de Alimentos, Universidade Federal de Ouro Preto, 35400-000 Ouro Preto, MG, Brazil c Departamento de Análises Clínicas, Universidade Federal de Ouro Preto, 35400-000 Ouro Preto, MG, Brazil d Departamento de Ciências Biológicas, Universidade Federal de Ouro Preto, 35400-000 Ouro Preto, MG, Brazil Received 17 July 2007; revised 24 December 2007; accepted 5 February 2008 Abstract The objective of this study was to investigate the effect of iron overload with a hyperlipidemic diet on the histologic feature of hepatic tissue, the lipid and glycemic serum profiles, and the markers of oxidative damage and stress in a rat model. Twenty-four male Fischer rats, purchased from Experimental Nutrition Laboratory, Federal University of Ouro Preto, were assigned to 4 equal groups, 2 were fed a standard cholesterol-free diet (group C or control and CI or control with iron) containing 8.0% soybean oil and 2 were fed a hyperlipidemic diet (group H or hyperlipidemic and HI or hyperlipidemic with iron) containing 1.0% cholesterol and 25.0% soybean oil. A total of 50 mg of iron was administered to rats in groups CI and HI in 5 equal doses (1 every 3 weeks for a 16-week period) by intraperitoneal injections of 0.1 mL of iron dextran solution (100 g Fe 2+ /L; Sigma, St Louis, Mo). The other rats in groups C and H were treated in a similar manner but with sterile saline (0.1 mL). Irrespective of the diet, iron excess enhanced serum triacylglycerols (P b .05) and reduced serum glucose and glycated hemoglobin levels (P b .05) but did not affect serum cholesterol concentration. Histologic analysis showed steatosis in groups H and to a lesser extent in HI. No significant differences (P N .05) were observed in paraoxonase activities or in serum levels of free or total sulfhydryl radicals, malondialdehyde, or total antioxidants. The findings suggest that iron excess in the rat probably modifies lipid metabolism and, as a consequence, alters glucose homeostasis and increases the level of serum triacylglycerols but not of cholesterol. © 2008 Published by Elsevier Inc. Keywords: Rat; Iron overload; Hyperlipemic diet; Hepatic steatosis; Serum glucose and lipids Abbreviations: ALT, alanine aminotransferase; ANOVA, analysis of variance; AOAC, Association of Official Analytical Che- mists; HI, hyperlipemic iron group; MDA, malondialdehyde; PON, paraoxonase; SH, sulfhydryl; TIBC, total iron- binding capacity; TS, transferrin saturation; UIBC, unsaturable iron-binding capacity. 1. Introduction There is considerable evidence indicating that a reduction in antioxidant defense capacity, and subsequent damage to macromolecules (ie, proteins and lipids), is related to the development of disease and ageing [1-3]. Endogenous mechanisms of antioxidant defense, involving both enzy- matic and nonenzymatic processes, play important roles in transforming oxygen-reactive species into inert molecules [4]. More important, a number of dietary components can also modify the redox status by increasing or decreasing the antioxidant capacity in the body. In this context, high intracellular iron concentrations are associated with an increase in free radicals that can cause oxidative damage and trigger pathologic processes [4]. Iron metabolism is regulated by a sophisticated and complex mechanism, the purpose of which is to maintain a sufficient supply of the element to fulfill the demands of the Available online at www.sciencedirect.com Nutrition Research 28 (2008) 391 398 www.nrjournal.com Corresponding author. Departamento de Ciências Biológicas, Uni- versidade Federal de Ouro Preto, Ouro Preto, MG, Brazil. Tel.: +55 31 35591686; fax: +55 31 35591680. E-mail address: [email protected] (M.L. Pedrosa). 0271-5317/$ see front matter © 2008 Published by Elsevier Inc. doi:10.1016/j.nutres.2008.02.009

Transcript of Iron overload alters glucose homeostasis, causes liver ...€¦ · Iron overload alters glucose...

Page 1: Iron overload alters glucose homeostasis, causes liver ...€¦ · Iron overload alters glucose homeostasis, causes liver steatosis, and increases serum triacylglycerols in rats Maísa

Available online at www.sciencedirect.com

(2008) 391–398www.nrjournal.com

Nutrition Research 28

Iron overload alters glucose homeostasis, causes liver steatosis, andincreases serum triacylglycerols in rats

Maísa Silvaa, Marcelo E. Silvaa,b, Heberth de Paulaa,Cláudia Martins Carneiroc, Maria Lucia Pedrosaa,d,⁎

aNúcleo de Pesquisas em Ciências Biológicas, Universidade Federal de Ouro Preto, 35400-000 Ouro Preto, MG, BrazilbDepartamento de Alimentos, Universidade Federal de Ouro Preto, 35400-000 Ouro Preto, MG, Brazil

cDepartamento de Análises Clínicas, Universidade Federal de Ouro Preto, 35400-000 Ouro Preto, MG, BrazildDepartamento de Ciências Biológicas, Universidade Federal de Ouro Preto, 35400-000 Ouro Preto, MG, Brazil

Received 17 July 2007; revised 24 December 2007; accepted 5 February 2008

Abstract

The objective of this study was to investigate the effect of iron overload with a hyperlipidemic diet

⁎ Correspondingversidade Federal de35591686; fax: +55 3

E-mail address: lp

0271-5317/$ – see frodoi:10.1016/j.nutres.2

on the histologic feature of hepatic tissue, the lipid and glycemic serum profiles, and the markers ofoxidative damage and stress in a rat model. Twenty-four male Fischer rats, purchased fromExperimental Nutrition Laboratory, Federal University of Ouro Preto, were assigned to 4 equal groups,2 were fed a standard cholesterol-free diet (group C or control and CI or control with iron) containing8.0% soybean oil and 2 were fed a hyperlipidemic diet (group H or hyperlipidemic and HI orhyperlipidemic with iron) containing 1.0% cholesterol and 25.0% soybean oil. A total of 50 mg of ironwas administered to rats in groups CI and HI in 5 equal doses (1 every 3 weeks for a 16-week period) byintraperitoneal injections of 0.1 mL of iron dextran solution (100 g Fe2+/L; Sigma, St Louis, Mo). Theother rats in groups C and H were treated in a similar manner but with sterile saline (0.1 mL).Irrespective of the diet, iron excess enhanced serum triacylglycerols (P b .05) and reduced serumglucose and glycated hemoglobin levels (P b .05) but did not affect serum cholesterol concentration.Histologic analysis showed steatosis in groups H and to a lesser extent in HI. No significant differences(P N .05) were observed in paraoxonase activities or in serum levels of free or total sulfhydryl radicals,malondialdehyde, or total antioxidants. The findings suggest that iron excess in the rat probablymodifies lipid metabolism and, as a consequence, alters glucose homeostasis and increases the level ofserum triacylglycerols but not of cholesterol.© 2008 Published by Elsevier Inc.

Keywords: Rat; Iron overload; Hyperlipemic diet; Hepatic steatosis; Serum glucose and lipids

Abbreviations: ALT, alanine aminotransferase; ANOVA, analysis of variance; AOAC, Association of Official Analytical Che-

mists; HI, hyperlipemic iron group; MDA, malondialdehyde; PON, paraoxonase; SH, sulfhydryl; TIBC, total iron-binding capacity; TS, transferrin saturation; UIBC, unsaturable iron-binding capacity.

1. Introduction

There is considerable evidence indicating that a reductionin antioxidant defense capacity, and subsequent damage tomacromolecules (ie, proteins and lipids), is related to thedevelopment of disease and ageing [1-3]. Endogenousmechanisms of antioxidant defense, involving both enzy-

author. Departamento de Ciências Biológicas, Uni-Ouro Preto, Ouro Preto, MG, Brazil. Tel.: +55 311 [email protected] (M.L. Pedrosa).

nt matter © 2008 Published by Elsevier Inc.008.02.009

matic and nonenzymatic processes, play important roles intransforming oxygen-reactive species into inert molecules[4]. More important, a number of dietary components canalso modify the redox status by increasing or decreasing theantioxidant capacity in the body. In this context, highintracellular iron concentrations are associated with anincrease in free radicals that can cause oxidative damageand trigger pathologic processes [4].

Iron metabolism is regulated by a sophisticated andcomplex mechanism, the purpose of which is to maintain asufficient supply of the element to fulfill the demands of the

Page 2: Iron overload alters glucose homeostasis, causes liver ...€¦ · Iron overload alters glucose homeostasis, causes liver steatosis, and increases serum triacylglycerols in rats Maísa

392 M. Silva et al. / Nutrition Research 28 (2008) 391–398

cell while ensuring that the potentially toxic levels do notaccumulate. However, iron excess can occur under certaincircumstances, especially in individuals with diseases such ashereditary hemochromatosis, the most common inheriteddisorder among European descendents and a widespreaddisease in sub-Saharan populations [5]. Dietary ironsupplementation can also lead to iron excess in otherwisehealthy subjects. For instance in Sweden, where the diet hasbeen supplemented with iron for some 50 years, 5% of menexhibited augmented levels of the mineral, whereas in 2% ofthe subjects, the levels were similar to those found in theinitial stages of hemochromatosis [6,7]. Moreover, amongelderly subjects, the intake of highly bioavailable forms ofiron (ie, supplemental iron, red meat, and fruit) may promotehigh iron stores [8].

Iron excess has been linked to risks of development ofcertain chronic disorders such as diabetes [9], glucoseintolerance [10], and cardiovascular diseases [11-13]. Ironexcess induces cellular injury and functional abnormalities inhepatocytes by the process of lipid peroxidation [14].

Liver plays a central role in iron metabolism and in themaintenance of glucose and lipid homeostasis. Thus, hepaticinjury triggered by iron excess may increase the concentra-tion of secondary serum metabolites, such as cholesterol,triacylglycerols, and glucose, which are considered riskfactors for cardiovascular diseases. The association betweeniron and a lipid-rich diet may exacerbate this situationbecause lipids also cause oxidative stress and steatohepatitis[15]. Various experimental models involving iron excesshave been applied to assess the risk factors of cardiovasculardiseases and alterations in liver histologic conditions [16-19].

In the present study, nontoxic doses of iron have beenused to assess the effects of the metal for a long period and inassociation with a hyperlipidic diet. To investigate theinteraction between hyperlipidemic diet and the effects ofiron overload, we have targeted serum markers of anti-oxidant defenses and oxidative stress as well as glucose andlipid levels in rats receiving iron treatment and cholesterol-free or lipid-rich diets. Furthermore, we evaluated the effectof these treatments on the markers of hepatic function andhistologic conditions.

2. Methods and materials

This study was approved by the Ethical Committee onResearch of the Universidade Federal de Ouro Preto, OuroPreto-MG,Brazil. All experimental procedureswere approvedand performed in accordance with the principles defined bythe Brazilian School of Animal Experimentation [20].

2.1. Animals and experimental design

Male Fischer rats (n = 24), weighing approximately 100 g,were maintained in metabolic cages at 24°C under a constantrelative humidity of 55% and a 12-hour light/dark cycle. Allrats were provided food and water ad libitum. The rats weredivided into 4 groups as follows: the control group (C) was

fed a standard cholesterol-free diet containing 8.0% soybeanoil; the control iron group (CI) was fed the same standard dietand given iron dextran by intraperitoneal injections; thehyperlipemic group (H) was fed a diet containing 1.0%cholesterol and 25.0% soybean oil in accordance withTurbino-Ribeiro et al [18]; and the hyperlipemic iron group(HI) was fed the same high-cholesterol diet and given irondextran injections.

A total of 50 mg of iron was administered to rats in groupsCI and HI in 5 equal doses (1 every 3 weeks for a 16-weekperiod) by injection of iron dextran solution (100 g Fe2+/L;Sigma, St Louis, Mo). Rats in groups C and H were similarlyinjected with 0.1 mL of sterile saline during the same period.At the end of the treatment period, rats were subjected toovernight food deprivation and euthanized by an intraper-itoneal injection of pentobarbital (Sigma) at a dose of 60 mg/kg body weight. For the determination of the levels of serumcomponents, blood samples were collected from the brachialplexus in 5-mL test tubes (containing EDTA for thedetermination of glucose) and centrifuged at 12 800g for15 minutes. The livers were removed, weighed, and portionsfrozen for subsequent determination of iron and fat content;the remaining liver samples, together with the aorta (thoracicand abdominal), were stored in 3.7% buffered formaldehydefor histologic analysis.

2.2. Dietary composition

The control diet contained contains the following (g/kg):cornstarch (686; Unilever Bestfoods, Mogi-Guaçu, SãoPaulo, Brazil), casein (150; Isofar, Duque de Caxias, Rio deJaneiro, Brazil), soybean oil (80; Sadia, São Paulo, SãoPaulo, Brazil), salt mixture (50), cellulose (20; Merck,Darmstadt, Germany), vitamin mixture (10), and choline (4;Labsynth, Diadema, São Paulo, Brazil), with a total energy of4064 kJ/kg. The hyperlipemic diet contained the following(in g/kg: cornstarch (311), cellulose (200), soybean oil (250),casein (125), salt mixture (50), cholesterol (10), vitaminmixture (10), and choline (4), with a total energy of4084 kJ/kg [21]. The salt mixture contained the following(g/kg): NaCl (139.3), KI (0.79), MgSO4.7H2O (57.3),CaCO3 (381.4), MnSO4.H2O (4.01), FeSO4.7H2O (27.0),ZnSO4.7H2O (0.548), CuSO4.5H2O (0.477), CoCl2.6H2O(0.023), and KH2PO4 (389.0) [21]. The vitamin mixturecontained (g/kg unless otherwise stated) the following:retinyl acetate (2 000 000 IU), cholecalciferol (200 000IU), p-aminobenzoic acid (10.00), I-inositol (10.00), niacin(4.00), calcium pantothenate (4.00), riboflavin (0.80),thiamine hydrochloride (0.50), pyridoxine hydrochloride(0.50), folic acid (0.20), biotin (0.04), vitamin B12 (0.003),sucrose (qsp 1000), choline (200.0), and dl α-tocopherol(10 000 IU) [21].

2.3. Assay methods

Serum iron concentration and total iron-binding capacity(TIBC) were determined in nonhemolyzed serum samples byspectrophotometric analysis using Labtest kits no. 38 and 41

Page 3: Iron overload alters glucose homeostasis, causes liver ...€¦ · Iron overload alters glucose homeostasis, causes liver steatosis, and increases serum triacylglycerols in rats Maísa

393M. Silva et al. / Nutrition Research 28 (2008) 391–398

(Labtest, Lagoa Santa, MG, Brazil), respectively, with astandard iron solution (89.5 μmol/L). Transferrin saturation(TS) was estimated from the ratio of serum iron/TIBC.Serum triacylglycerols and cholesterol were measuredenzymatically using Labtest kits no. 59-4/50 and 60-2/100,respectively, with glycerol or cholesterol standards asappropriate. After precipitation of low-density lipoproteinand very low-density lipoprotein with phosphotungstic acid/MgCl2, high-density lipoprotein (HDL-C) was determined inthe supernatant (Labtest kit no. 13). Glucose, hemoglobin,alanine aminotransferase (ALT), and aspartate aminotrans-ferase (AST) levels were measured with Labtest kits no. 84,43, 53, and 52, respectively. To determine enzymatic activity,a calibration curve (supplied by the manufacturer) was usedto correlate the obtained readings with the standardizedvalues (U/L). The percentage of glycated hemoglobin wasassayed using Labtest kit no. 17.

The total antioxidant status was determined with aCalbiochem kit no. 615700 (Calbiochem, San Diego,Calif). Lipid peroxidation was estimated spectrophotome-trically (Calbiochem kit no. 437634) via assay of 4-hydroxyalkenals (HAE) and malondialdehyde (MDA) with1,1,3,3-tetramethoxypropane in Tris-hydrochloride as theexternal standard. The concentrations of free and totalsulfhydryl (SH) radicals were determined using the Ellmantest according to the method of Sedlak et al [22].

Paraoxonase (PON) activity toward phenyl acetate(arylesterase activity, AE) was determined by measuring theinitial rate of substrate hydrolysis (monitored at 270 nm for3 minutes) in an assay mixture (3 mL) containing 2 mmol/Lof substrate, 2 mmol/L of CaCl2, and 10 μL of plasma in50 mmol/L of Tris-HCl (pH 8.0). The background rateassociated with the nonenzymatic hydrolysis of substrate,determined in a blank sample prepared as described above butwithout plasma, was subtracted, and the enzymatic activitywas calculated assuming the molar extinction coefficient ofphenyl acetate to be 1310 mol/L·cm. The results wereexpressed in U/mL, where 1 U of arylesterase hydrolyzes1 μmol of phenyl acetate per minute [23].

Table 1Iron concentration, UIBC, TIBC, and TS in the serum and the level of iron in the

Variables

C CI H

Serum iron (μmol/L) 14.54 ± 4.63b 25.45 ± 5.18a 21.30UIBC (μmol/L) 36.41 ± 7.39 38.00 ± 13.77 43.13TIBC (μmol/L) 50.96 ± 8.69 63.58 ± 11.39 64.44TS (%) 28.42 ± 8.09b 40.34 ± 7.16a 33.38Liver iron (μg/100 mg) 7.71 ± 3.96 53.36 ± 33.47 8.01

Groups of animals were fed on a cholesterol-free control (C and CI) or a hyperlipesolution (CI and HI) or sterile saline (C and H) for a 16-week period. Values aremean values/interactions were not significant; when interactions were significantdifferences between mean values. Within each row, mean values bearing differentest at P b .05.

2.4. Iron and lipid levels in the liver

Liver samples were digested at 100°C in HNO3, theexcess acid was evaporated, and the iron quantified bycolorimetric analysis using the Association of OfficialAnalytical Chemists (AOAC) o-phenanthroline assayprotocol [21] with a standard iron solution (89.5 μmol/L).The percentage of total lipids was determined after theAOAC procedure [21] in which a powdered liver sample,obtained by grinding 1 g of tissue with 12 g of clean sandin a porcelain mortar and pestle, was dried at 105°C for2 hours, placed in the cellulose cartridge of a Soxhletapparatus, and the lipids were extracted with petroleumether for 6 hours.

2.5. Histologic evaluation

Slides containing sections of formalin-fixed, paraffin-embedded liver tissue were coded (to avoid observer bias),stained with hematoxylin and eosin for routine histologicexamination, with Masson trichrome for collagen and withPerls' Prussian blue for iron, and examined under the lightmicroscopy. Steatosis, hydropic degeneration, inflammation,iron deposits, and collagen neoformation were evaluatedunder a ×40 objective in 20 random fields. Fragments of theaorta (thoracic and abdominal) were stained with hematox-ylin and eosin, Masson trichrome, Weigert, and periodicacid-Schiff stains to identify alterations characteristic ofatherosclerosis.

2.6. Statistical analysis

Statistical analyses were performed with the aid ofMinitab 13 statistical software (Minitab, State College, Pa).All data were expressed as means ± SD. Treatmentdifferences were determined using 2-way analysis ofvariance (ANOVA). Classification factors were iron anddiet as well as their interactions. When the interactions weresignificant, means of the 4 treatment groups were comparedusing the Tukey multiple range test. Differences wereconsidered significant at P b .05.

liver of rats

Experimental groups

HI ANOVA (P)

Diet Iron Iron × diet

± 4.25a 20.87 ± 1.3a,b NS b.05 b.05± 9.76 50.20 ± 6.68 b.05 NS NS± 11.02 71.07 ± 6.74 b.05 b.05 NS± 6.40a,b 29.58 ± 3.27b NS NS b.05± 7.12 47.89 ± 27.58 NS b.05 NS

mic (H and HI) diet and received 5 injections (0.1 mL each) of iron dextranmeans ± SD; n = 6. Data were tested by 2-way ANOVA; NS indicates that(P b .05), Tukey post hoc tests were performed to determine the specifict superscript letters (a and b) are significantly different according to Tukey

Page 4: Iron overload alters glucose homeostasis, causes liver ...€¦ · Iron overload alters glucose homeostasis, causes liver steatosis, and increases serum triacylglycerols in rats Maísa

394 M. Silva et al. / Nutrition Research 28 (2008) 391–398

3. Results

3.1. Iron status and histologic aspects of the liver

Serum iron concentration was 75% higher in animalsreceiving the cholesterol-free control diet together with irondextran treatment (group CI) compared with those of theuntreated control group C (Table 1). In contrast, no significant

Fig. 1. Histologic analysis of liver sections derived from rats fed a control diet andfed a hyperlipemic diet and untreated (C, G, and K) or treated with iron dextran (D,(stain) trichrome (E-H), or Perls' Prussian blue (I-L) stains and are shown at ×60intense steatosis and inflammation; D shows discreet steatosis and inflammationneoformation; J and L show iron deposition (blue); and K shows intense steatosis

differences in iron concentration could be detected betweenthe hyperlipemic groups H and HI, and the serum iron levelsin these groups were not different from that observed in thecontrol group CI. As was expected, rats fed on a control dietand treated with iron dextran (group CI) presented higher TS,but this effect was not observed in iron-treated animals fed ona hyperlipemic diet. It seems, therefore, that the hyperlipemic

untreated (A, E, and I) or treated with iron dextran (B, F, and J) and from ratsH, and L). Sections were submitted to hematoxylin and eosin (A-D), Masson0 magnification. Section B shows iron deposition (black arrows); C shows; F shows the absence of collagen neoformation; G and H show collagenand the absence of iron deposition.

Page 5: Iron overload alters glucose homeostasis, causes liver ...€¦ · Iron overload alters glucose homeostasis, causes liver steatosis, and increases serum triacylglycerols in rats Maísa

Table 2Levels of cholesterol, HDL-C, triacylglycerols, and PON in the serum and the level of lipid in the liver of rats

Variables Experimental groups

C CI H HI ANOVA (P)

Diet Iron Iron × diet

Cholesterol (mmol/L) 1.78 ± 0.22 1.75 ± 0.21 3.84 ± 1.03 3.97 ± 0.63 b.05 NS NSHDL-C (mmol/L) 0.73 ± 0.31 0.76 ± 0.26 0.24 ± 0.07 0.25 ± 0.16 b.05 NS NSTriacylglycerols (mmol/L) 1.04 ± 0.25 2.29 ± 1.42 2.12 ± 0.71 2.64 ± 0.94 NS b.05 NSParaoxonase (U/mL) 124.43 ± 11.49a 117.68 ± 1.41a 90.88 ± 8.89b 114.63 ± 16.65a b.05 NS b.05Liver lipids (%) 13.52 ± 3.33 16.98 ± 8.03 33.19 ± 8.77 38.88 ± 3.45 b.05 NS NSLiver weight (g) 6.51 ± 0.64c 6.32 ± 0.39c 8.25 ± 0.48b 10.20 ± 1.56a b.05 NS b.05

Groups of animals were fed on a cholesterol-free control (C and CI) or a hyperlipemic (H and HI) diet and received 5 injections (0.1 mL each) of iron dextransolution (CI and HI) or sterile saline (C and H) for a 16-week period. Values are means ± SD; n = 6. Data were tested by 2-way ANOVA; when interactions weresignificant (P b .05), Tukey post hoc tests were performed to determine the specific differences between mean values. Within each row, mean values bearingdifferent superscript letters (a, b, and c) are significantly different according to Tukey test at P b .05.

395M. Silva et al. / Nutrition Research 28 (2008) 391–398

diet promotes an increase in the synthesis and/or the secretionof transferrin because the concentration of serum iron wassimilar in the CI and HI groups, but TIBC was higher in HIrats resulting in a reduction of TS for that group. Bothunsaturable iron-binding capacity (UIBC) and TIBC werehigher (P b .05) in group H compared with group C, althoughTS was lower (P b .05) in group HI compared with group CI.Liver iron concentration was significantly influenced byadministration of iron dextran, with treated rats showinglevels some 6-fold higher than that of the C group.

Group C rats presented a normal liver histologic conditionwith no iron detectable on staining with Perls' Prussian blue(Fig. 1A and I). In contrast, all control rats treated with irondextran (group CI) exhibited hepatic iron deposition,predominantly in the Kupffer cells, whereas intense hydropicdegeneration was observed in a few rats in this group(Fig. 1B and J). In the hyperlipemic group, all animalstreated with iron dextran (group HI) showed accumulation ofiron in the Kupffer cells (Fig. 1D and L), both within andaway from the inflammatory foci, and a marked reduction insteatosis (Fig. 1D) in comparison with their nontreatedcounterparts (group H; Fig. 1C). However, no differenceswere observed between the CI and HI groups with respect tothe quantity of Kupffer cells. Collagen neoformation wasobserved in all animals fed the hyperlipemic diet (groups Hand HI; Fig. 1G and H) but was not detected in thosereceiving the control diet (groups C and CI; Fig. 1E and F).

Table 3Aspartate aminotransferase and ALT activities and levels of glucose, hemoglobin,

Variables

C CI H

AST (U/L) 13.91 ± 7.67 40.55 ± 8.85 23.43ALT (U/L) 22.49 ± 4.61 39.47 ± 5.04 36.14Glucose (mmol/L) 6.37 ± 0.64 5.57 ± 0.39 5.29Hemoglobin (μmol/L) 21.95 ± 1.30 23.14 ± 1.40 19.92Glycated hemoglobin (%) 8.52 ± 3.28 6.36 ± 2.55 5.18

Groups of animals were fed on a cholesterol-free control (C and CI) or a hyperlipesolution (CI and HI) or sterile saline (C and H) for a 16-week period. Values are m

Moreover, H group animals presented intense steatosistogether with perivascular and parenchymal inflammationassociated with collagen neoformation (Fig. 1C). Noalterations were observed in the thoracic and abdominalaorta fragments from animals of any of the groups.

3.2. Lipid profiles and PON activities

Hyperlipemic rats presented serum cholesterol levels thatwere 2-fold higher than those fed the control diet (Table 2).In contrast, the hyperlipemic diet reduced the HDL-C leveland PON activity. Treatment with iron dextran increasedserum triacylglycerols but had no effect on the cholesteroland HDL-C level or on PON activity. Rats in the HI grouppresented greater levels of fat in the liver but because theirliver weights were higher than those of group H (Table 2),the fat percentage in the 2 groups was similar, and the liverwas probably hyperplasic in the iron-loaded rats.

3.3. Levels of glucose, hemoglobin, and glycatedhemoglobin and aminotransferase activity

To explore the effects of iron on glucose homeostasis, thelevels of glycated hemoglobin and serum glucose weredetermined. As shown in Table 3, iron decreased serumglucose and glycated hemoglobin levels—effects that weremore intense when associated with a hyperlipemic diet.Although administration of iron dextran increased ASTactivity, it had no effect on ALT activity. In contrast, the

and glycated hemoglobin in serum of rats

Experimental groups

HI ANOVA (P)

Diet Iron Iron × diet

± 15.01 30.47 ± 18.86 NS b.05 NS± 16.89 27.73 ± 5.29 NS NS NS± 0.99 4.78 ± 0.46 b.05 b.05 NS± 1.09 20.60 ± 2.12 b.05 NS NS± 2.21 2.25 ± 0.55 b.05 b.05 NS

mic (H and HI) diet and received 5 injections (0.1 mL each) of iron dextraneans ± SD; n = 6. Data were tested by 2-way ANOVA.

Page 6: Iron overload alters glucose homeostasis, causes liver ...€¦ · Iron overload alters glucose homeostasis, causes liver steatosis, and increases serum triacylglycerols in rats Maísa

Table 4Total antioxidant status and levels of MDA, HAE, and free and total SH radical levels in serum of rats

Variables Experimental groups

C CI H HI ANOVA (p)

Diet Iron Iron × diet

Antioxidant status (mmol/L) 0.83 ± 0.03 0.82 ± 0.08 0.88 ± 0.10 0.83 ± 0.08 NS NS NSMDA + HAE serum (mmol/L) 2.98 ± 1.64 3.73 ± 1.37 4.27 ± 0.84 4.79 ± 0.49 b.05 NS NSFree SH (μmol/L) 50.29 ± 0.70 51.68 ± 1.49 58.24 ± 12.02 58.24 ± 12.02 NS NS NSTotal SH (μmol/L) 545.19 ± 61.08 468.34 ± 62.50 512.45 ± 40.09 521.39 ± 41.80 NS NS NS

Groups of animals were fed on a cholesterol-free control (C and CI) or a hyperlipemic (H and HI) diet and received 5 injections (0.1 mL each) of iron dextransolution (CI and HI) or sterile saline (C and H) for a 16-week period. Values are means ± SD; n = 6. Data were tested by 2-way ANOVA.

396 M. Silva et al. / Nutrition Research 28 (2008) 391–398

hyperlipemic diet did not affect the activities of either of theaminotransferases.

3.4. Antioxidant status and MDA, HAE, and SH radicallevels

Serum MDA and HAE concentrations were increased bythe hyperlipemic diet but not by administration of irondextran. Neither diet nor iron treatment affected serum totalantioxidant status or the levels of free or total SH radicals(Table 4).

4. Discussion

The model used in the present study, which involvedadministration of iron dextran by injection of rats for a16-week period, avoided any indirect mechanisms of up-regulation that could be caused by the disturbance in nutrientabsorption associated with oral loading. The increase inhepatic iron in the experimental animals was well below thecritical threshold (N22-fold increase) in which hepatocellularinjury has been reported [24] and moderate in comparisonwith that observed in patients with symptomatic hemochro-matosis (ie, 16-fold to 30-fold increase) [25].

The findings with respect to serum iron and TS suggestthat a hyperlipidemic diet affects iron metabolism in rats.These results confirm those of Dabbagh et al [26] who foundthat the levels of serum iron in rats fed a hyperlipidemic dietwere unaffected by treatment with iron pentacarbonyl. Inaddition, Du et al [27] observed that hepatic levels of metal-related genes, such as transferrin and ceruloplasmin, as wellas those related to bile acid synthesis, were up-regulated infemale Long-Evans Cinnamon rats that had been treated witha mixture of docosahexaenoic acid-rich soybean oil. In thepresent work, the intracellular iron stores were more evidentin group HI rats as compared with those in group CI,suggesting that the hyperlipidemic diet affected ironhomeostasis.

The present study demonstrated that the diet used waseffective in inducing rats to become hyperlipemic. Interest-ingly, histopathologic analysis of thoracic and abdominalaorta showed no atherosclerosis in hyperlipemic ratsconfirming the findings of other authors [28].

The data for serum cholesterol levels and iron are inagreement with those obtained by Araujo et al [29], who alsoobserved the absence of alteration in serum cholesterol inrabbits receiving iron dextran injections but contradictsprevious results from our laboratory [18]. In our earlierstudy, although the same dose of iron was used, the period ofapplication was shorter, and the distribution of iron amongthe various compartments of the liver might not have beenthe same as in the model used in the present work. Thus, thetransit pool of free iron may have been different between the2 studies and, consequently, cholesterol metabolism orcirculating cholesterol uptake could have been affected in adifferent manner. It is noteworthy, however, that the effectsof diet on serum cholesterol levels were observed to be thesame in both of our studies.

Data presented in this study indicate that iron increasedfat content, although lipid percentages in the H and HI ratswere the same. Higher liver weights, similar lipid percen-tages, and a marked reduction in steatosis (as detected in thehistologic evaluation) within the HI group probably indicateliver hyperplasia in these animals and a more efficient liverlipid metabolism. The observation that treatment with ironincreased serum triacylglycerols agrees with the findings ofRodriguez et al [30].

Because an association between hepatic iron stores andinsulin-resistance syndrome has been reported previously[5,6,31], parameters related to glycemic levels weredetermined in our experimental rat model. Interestingly, norelationship between the increase in iron stores andresistance to insulin was detected but rather iron treatment(associated or not with a hyperlipidic diet) was observed topromote a decrease in the levels of glucose and glycatedhemoglobin. The resistance of rodents to iron-induceddiabetes has recently been demonstrated in iron overloadhepcidin-deficient mice [32].

The design of our study did not allow us to propose amechanism for the observed alterations in serum glucoseand glycated hemoglobin levels. Nevertheless, it isnoteworthy that several authors have reported functionalabnormalities in mitochondrial membranes caused by ironexcess. Masini et al [33] observed that iron-inducedoxidative stress gave rise to irreversible mitochondrialdysfunctions with resultant derangement of mitochondrial

Page 7: Iron overload alters glucose homeostasis, causes liver ...€¦ · Iron overload alters glucose homeostasis, causes liver steatosis, and increases serum triacylglycerols in rats Maísa

397M. Silva et al. / Nutrition Research 28 (2008) 391–398

energy-transducing capabilities arising from a reduction inthe respiratory chain enzyme activities. Such irreversibleoxidative anomalies caused a dramatic drop in tissue ATPlevels. In addition, Ceccarelli et al [34] observed inductionof lipid peroxidation and consequential abnormalities inmitochondria and calcium transport, after feeding rats witha diet supplemented with 2.5% of iron pentacarbonyl for40 days. The present study revealed that, irrespective of thediet applied, iron treatment did not dramatically stress theantioxidant system, as indicated by the absence of alterationin the levels of free and total SH radicals and totalantioxidant status in the serum. In contrast, Lykkesfeldtet al [35] have recently demonstrated an increase in thebiochemical markers of oxidative stress and damage inresponse to increasing concentrations of iron in the liver. Inour study, we observed that iron treatment produced anincrease in AST activity, which is a marker for mitochon-drial lesions. A reduction in mitochondrial capacity andimpaired ATP production would lead to reduced levels ofgluconeogenesis, and this could explain the observedreduction in serum glucose and glycated hemoglobin levels.

Paraoxonase is a serum enzyme that is closely associatedwith HDL and prevents and inhibits lipoprotein oxidation, abasic mechanism involved in the initiation and progressionof atherosclerosis. Under certain conditions, oxidative stressis associated with the reduction of PON activity [36,37]. Inthe present study, arylesterase activity toward phenyl acetatewas decreased in rats fed a hyperlipidemic diet, which is inagreement with previous reports [37,38]. Moreover, Nguyenand Sok [39] proposed that metal-catalyzed oxidation maybe a primary cause for the decrease in HDL-associated PON1activity observed under oxidative stress. Administration ofiron might, therefore, give rise to a reduction in PON activity.This hypothesis is supported by Trudel et al [40] whoreported that PON activity was decreased in rat and humanliver microsomes in the presence of increased ironconcentrations. However, these authors found that theadministration of iron did not alter PON activity in ratserum, a result that was confirmed in the present study. It islikely that the iron dextran treatment used in our investiga-tion did not cause the oxidative stress required to bring aboutan alteration in PON activity. This possibility is supported bythe finding that MDA and HAE levels were altered by dietbut not by administration of iron dextran.

In conclusion, the present study revealed that whenhyperlipidemic rats are subjected to iron dextran therapy,there are consequential changes in glucose homeostasis andin the levels of triacylglycerols in the serum and in the liver.These aspects, together with distinct histologic featuresobserved in H and HI rats, suggest an alteration of hepaticlipid metabolism and/or liver hyperplasia when iron isadministered in association with a hyperlipemic diet. On thebasis of the model used to promote excess iron in the presentstudy, it would appear that rats do not exhibit the tendencytoward abnormal glucose tolerance and diabetes frequentlyobserved in humans with iron overload [5,6,31].

Acknowledgment

This study was supported by the Fundação de Amparo àPesquisa de Minas Gerais (FAPEMIG, Minas Gerais, Brazil)and Conselho Nacional de Desenvolvimento Científicoe Tecnológico (CNPq, Brazil; no. 475823/2007-9). Theauthors thank Rinaldo Cardoso dos Santos for suggestionsand for careful review of the manuscript.

References

[1] Tuomainen TP, Punnonen K, Nyyssonen K, Salonen JT. Associationbetween body iron stores and the risk of acute myocardial infarction inmen. Circulation 1991;97:1461-6.

[2] Jenner P. Oxidative stress in Parkinson's disease. Ann Neurol 2003;53-S3:26-36.

[3] Bunker VW. Free radicals, antioxidants and ageing. Med Lab Sci 1992;49:299-312.

[4] Halliwell B, Gutteridge JM. The antioxidants of human extracellularfluids. Arch Biochem Biophys 1990;1:1-8.

[5] Sheth S, Brittenham GM. Genetic disorders affecting proteinsof iron metabolism: clinical implications. Annu Rev Med 2000;51:443-64.

[6] Olsson KS, Heedman PA, Staugard F. Preclinical hemochromatosis ina population on a high-iron–fortified diet. J Amer Med Assoc 1978;239:1999-2000.

[7] Olsson KS, Marsell R, Ritter B, Olander B, Akerblom A, Ostergard H,et al. Iron deficiency and iron overload in Swedish male adolescents.J Intern Med 1995;237:187-94.

[8] Fleming DJ, Tucker KL, Jacques PF, Dallal GE, Wilson PW, Wood RJ.Dietary factors associated with the risk of high iron stores in the elderlyFramingham Heart Study cohort. Am J Clin Nutr 2002;6:1375-84.

[9] Britton RS, Bacon BR, Recknagel RO. Lipid peroxidation andassociated hepatic organelle dysfunction in iron overload. ChemPhys Lipids 1987;45:207-39.

[10] Jiang R, Manson JE, Meigs JB, Ma J, Rifai N, Hu FB. Body ironstores in relation to risk of type 2 diabetes in apparently healthywomen. J Amer Med Assoc 2004;291:711-7.

[11] Tuomainen TP, Nyyssonen K, Salonen R, Tervahauta A, Korpela H,Lakka T, et al. Body iron stores are associated with serum insulin andblood glucose concentrations. Population study in 1,013 easternFinnish men. Diabetes Care 1997;20:426-8.

[12] Sullivan JL. Iron therapy and cardiovascular disease. Kidney Int 1999;69:S135-7.

[13] Williams MJ, Poulton R, Williams S. Relationship of serum ferritinwith cardiovascular risk factors and inflammation in young men andwomen. Atherosclerosis 2002;165:179-84.

[14] Piperno A, Trombini P, Gelosa M, Mauri V, Pecci V, Vergani A, et al.Increased serum ferritin is common in men with essential hypertension.J Hypertens 2002;20:1513-8.

[15] Matsuzawa N, Takamura T, Kurita S, Misu H, Ota T, Ando H, et al.Lipid-induced oxidative stress causes steatohepatitis in mice fed anatherogenic diet. Hepatology 2007;46:1392-403.

[16] Brunet S, Thibault L, Delvin E, Yotov W, Bendayan M, Levy E.Dietary iron overload and induced lipid peroxidation are associatedwith impaired plasma lipid transport and hepatic sterol metabolism inrats. Hepatology 1999;29:1809-17.

[17] Knutson MD, Walter PB, Ames BN, Viteri FE. Both iron deficiencyand daily iron supplements increase lipid peroxidation in rats. J Nutr2000;130:621-8.

[18] Turbino-Ribeiro SML, Silva ME, Chianca Jr DA, Paula H, CardosoLM, Colombari E, et al. Iron overload in hyperlipemic rats affects ironhomeostasis and serum lipids but not blood pressure. J Nutr 2003;133:15-20.

Page 8: Iron overload alters glucose homeostasis, causes liver ...€¦ · Iron overload alters glucose homeostasis, causes liver steatosis, and increases serum triacylglycerols in rats Maísa

398 M. Silva et al. / Nutrition Research 28 (2008) 391–398

[19] Brown KE, Dennery PA, Ridnour LA, Fimmel CJ, Kladney RD, BruntEM, et al. Effect of iron overload and dietary fat on indices of oxidativestress and hepatic fibrogenesis in rats. Liver Int 2003;23:232-42.

[20] Colégio Brasileiro de Experimentação Animal. Princípios Éticos naExperimentação Animal do Colégio Brasileiro de ExperimentaçãoAnimal. São Paulo: COBEA; 1991.

[21] Association of Official Analytical Chemists. Official methods ofanalysis. Washington: AOAC; 1980.

[22] Sedlak J, Lindsay RH. Estimation of total, protein-bound and non-protein sulfhydryl groups in tissue with Ellman's reagent. AnalBiochem 1968;25:192-205.

[23] Beltowski J, Wojcicka G, Jamroz A. Differential effect of 3-hydroxy-3-methylglutaryl coenzyme A reductase inhibitors on plasma paraox-onase 1 activity in the rat. Pol J Pharmacol 2002;54:661-71.

[24] Park CH, Bacon BR, Brittenham GM, Tavill AS. Pathology of dietarycarbonyl iron overload in rats. Lab Invest 1987;57:555-63.

[25] Tavill AS, Sharma BK, Bacon BR. Iron and the liver: genetichemochromatosis and other hepatic iron overload disorders. Prog LiverDis 1990;9:281-305.

[26] Dabbagh AJ, Mannion T, Lynch SM, Frei B. The effect of ironoverload on rat plasma and liver oxidant status in vivo. Biochem J1994;300:799-803.

[27] Du C, Fujii Y, Ito M, Harada M, Moriyama E, Shimada R, et al. Dietarypolyunsaturated fatty acids suppress acute hepatitis, alter geneexpression and prolong survival of female Long-Evans Cinnamonrats, a model of Wilson disease. J Nutr Biochem 2004;15:273-80.

[28] Moghadasian MH. Experimental atherosclerosis: a historical overview.Life Sci 2002;70:855-65.

[29] Araujo JA, Romano EL, Brito BE, Parthé V, Romano M, Bracho M,et al. Iron overload augments the development of atheroscleroticlesions in rabbits. Arterioscler Thromb Vasc Biol 1995;15:1172-80.

[30] Rodriguez A, Hilvo M, Kytomaki L, Fleming RE, Britton RS, BaconBR, et al. Effects of iron loading on muscle: genome-wide mRNAexpression profiling in the mouse. BMC Genomics 2007;8:379, doi:10.1186/1471-2164-8-379.

[31] Mendler MH, Turlin B, Moirand R, Jouanolle AM, Sapey T, GuyaderD, et al. Insulin resistance-associated hepatic iron overload. Gastro-enterology 1999;117:1155-63.

[32] Ramey G, Faye A, Durel B, Viollet B, Vaulont S. Iron overload inHepc1(−/−) mice is not impairing glucose homeostasis. FEBS Lett2007;581:1053-7.

[33] Masini A, Ceccarelli D, Giovannini F,Montosi G, Garuti C, PietrangeloA. Iron-induced oxidant stress leads to irreversible mitochondrialdysfunctions and fibrosis in the liver of chronic iron-dosed gerbils. Theeffect of silybin. J Bioenerg Biomembr 2000;32:175-82.

[34] Ceccarelli D, Gallesi D, Giovannini F, Ferrali M, Masini A. Relation-ship between free iron level and rat liver mitochondrial dysfunction inexperimental dietary iron overload. Biochem Biophys Res Commun1995;209:1-6.

[35] Lykkesfeldt J, Morgan E, Christen S, Skovgaard LT,Moos T. Oxidativestress and damage in liver, but not in brain, of Fischer 344 rats subjectedto dietary iron supplementation with lipid-soluble [(3,5,5-trimethylhex-anoyl)ferrocene]. J Biochem Mol Toxicol 2007;21:145-55.

[36] Ayub A, Mackness MI, Arrol S, Mackness B, Patel J, Durrington PN.Serum paraoxonase after myocardial infarction. Arterioscler ThrombVasc Biol 1999;19:330-5.

[37] AviramM, Rosenblat M, Bisgaier CL, Newton RS, Primo-Parmo S, LaDu BN. Paraoxonase inhibits high-density lipoprotein oxidation andpreserves its functions. A possible peroxidative role for paraoxonase.J Clin Invest 1998;101:1581-90.

[38] MacknessMI, Harty D, Bhatnagar D,Winocour PH, Arrol S, IsholaM,et al. Serum paraoxonase activity in familial hypercholesterolaemia andinsulin-dependent diabetes mellitus. Atherosclerosis 1991;86:193-9.

[39] Nguyen SD, Sok DE. Oxidative inactivation of paraoxonase1, anantioxidant protein and its effect on antioxidant action. Free RadicalRes 2003;37:1319-30.

[40] Trudel K, Sinnett D, James RW, Delvin E, Amre D, Seidman E, et al.Iron–ascorbic acid–induced oxidant stress and its quenching byparaoxonase 1 in HDL and the liver: comparison between humans andrats. J Cell Biochem 2005;96:404-11.