You are on page 1of 27

1

Curcumin Protects Liver and Spleen from Oxidative Stress induced by

Excessive Dietary Iron Intake in a Rat Model of Nutritional Iron

Overload

Farid A. Badria1*, Ahmed S Ibrahim2 , and Adel F. Badria3

35516, and 3Medical Technology Center, Alexandria University, Egypt

*Corresponding authors: Farid A Badria, PhD, Pharmacognosy Department, Mansoura University,

Mansoura 35516 Egypt, email: faridbadria@yahoo.com;

Running Title: Curcumin As Anti-Oxidative Stress In Iron overload-Induced Toxicity.

10
11

Pharmacognosy Department, 2Department of Biochemistry, Faculty of Pharmacy, Mansoura University

Keywords: Curcumin, Oxidative Stress, iron overload, liver toxicity, spleen toxicity.

2
12

ABASTRACT

13

Background: Iron overload (IO) is now being recognized as a health problem in industrialized countries

14

where cultural practices encourage acquisition of excess iron from diets. Excess iron is highly toxic for

15

vital organs especially liver and spleen. Curcumin, a superb nature's medicine with still unfolding health

16

benefits, has been shown to possess iron chelation property that has not been studied in nutritional IO.

17

Objective: A three-year field study was conducted by our team on the impact of environmental excess

18

iron in drinking water and its health hazard on liver diseases. Here, we evaluate the efficacy of curcumin

19

as nutritional supplement to alleviate the hepatospleno-disorders and their sequelae on IO-induced rats.

20

Design/Results: Initial studies revealed that the treatment of iron-overloaded rats with curcumin

21

resulted in marked decreases in iron accumulation within liver/spleen. To further assess the role of

22

curcumin, we studied its effects on iron overload-induced oxidative stress, protein loss, and anti-oxidant

23

depletion. Iron-overloaded rats had significantly more TBARS, NO and OH radicals in liver and spleen

24

compared with control group. These effects of IO were all reduced by intervention treatment with

25

curcumin. Following this further, the bulk protein content in liver and spleen as well as their endogenous

26

anti-oxidants (SOD, CAT, GSH, and ASA) were highly significantly decreased in chronic iron

27

intoxication. Inversely, the administration of curcumin improved the alterations in these biochemical

28

parameters.

29

Conclusions: These findings implicate the therapeutic potential of the multifunctional, nutritional iron

30

chelator, curcumin, in dampening IO-induced hepatosplenic disorders. This beneficial effect of

31

curcumin represents a new horizon in managing nutritional iron overload.

32

3
33

INTRODUCTION

34

Iron, one of the most abundant metals on earth, is an integral part of many proteins and enzymes that

35

maintain good health (1). However, excess amounts of iron has a destructive nature if not properly

36

diagnosed and treated (2). Undiagnosed iron overload can lead to hemochromatosis, in which the excess

37

iron stored in body organs causing serious tissue damage that leads to life-threatening conditions.

38

Nevertheless, the majority of public are unaware of the problem and unconcerned due to the mistaken

39

idea that the majority of hemochromatosis cases are genetic in origin, which affects only about 1 million

40

of the estimated 275 million Americans. In fact, the potential threat of iron overload is universal and

41

comes with advancing age regardless of genetic factors (3). Badria et al. reported the impact of

42

environmental excess iron on human health among Egyptians who are inhabitants an area with high iron

43

content in drinking water (4).

44

The content of body iron is regulated primarily by absorption since humans have no

45

physiological mechanism by which excess iron is excreted. The quantity of dietary iron and the

46

composition of the diet significantly influence iron absorption. Prolonged intake of high doses of iron

47

leads to the accumulation of excess iron (5). This phenomenon has been well demonstrated in the

48

episode of siderosis among the Bantu tribe in Africa who ingested excessive amounts of iron from their

49

diet and beer. Their use of iron pots for cooking and brewing beer increased the iron content (6).

50

Another important factor in iron absorption relates to the form of iron present in a diet. Herne iron and

51

nonheme iron are the two major sources of iron. Heme iron, that is found in meat, fish, and poultry, is

52

more effectively absorbed because this iron is associated with the porphyrin ring (7). A large population

53

survey from Australia showed that normal volunteers have average iron storage is about twice as much

54

as the optimal iron store for normal adults. Heavy ethanol intake and high meat consumption are

55

believed to be the critical factors (8). Such practices have occurred in industrialized countries where

4
56

alcohol intake as well as red meat consumption are high, and the use of iron fortification is widespread

57

(9). Hepatotoxicity and spleen dysfunction are the most common findings in patients with iron

58

overloading and in severe cases, iron is found within the Kupffer cells in the liver and the macrophage

59

cells of the spleen (10). The etiology of these multiple organ dysfunctions has been attributed to the

60

presence of excess non-protein bound free iron released to spark uncontrolled oxidation via generation

61

of reactive oxygen species (ROS) through Fenton and Haber-Weiss reactions (11). ROS react directly

62

with proteins, lipids, and nucleic acids and induce oxidative stress by depleting cellular stores of

63

antioxidants (12). Consequently, it is of utmost importance to maintain iron homeostasis to ensure iron

64

supply but to prevent accumulation of excess iron.

65

In all iron-overload-induced diseases, iron removal by iron chelation therapy is an effective life-

66

saving strategy. The currently available iron-chelating agents used clinically are deferoxamine, and

67

deferasirox. However, such compounds show several side effects and limitations (13) that direct towards

68

the finding of a more effective and safe drug which may rise the therapeutic benefits for patients. As

69

there is still no safe oral iron chelator agent, the use of nutritional therapy in the area of drug discovery

70

proved to be an efficient, safe, and economic tool in the area of health care. Curcumin, Figure 1A (14),

71

has been reported to be among the most efficient and safe chemopreventive agents investigated in

72

recent years and is currently in human trials to prevent cancer (15). The mechanism of action of

73

curcumin is complex and likely multifactorial (16). Previously, Curcumin has shown antioxidant and

74

iron-chelating properties in vitro (16, 17). However, the pre-clinical effects of curcumin on hepatic and

75

splenic sequelae during nutritional iron overload toxicity have not been investigated in depth.

76

Accordingly, the present study aimed to evaluate the ability of the iron chelator, curcumin, to attenuate

77

excessive dietary iron intake-induced hepatic and splenic oxidative stress. Furthermore, this study

78

pursues to gain insight into the mechanistic basis behind this effect.

79

5
80

MATERIALS AND METHODS

81

Materials

82

Na2HPO4, KH2PO4, p-phenylenediamine.HCl, and NaNO2 were purchased from Merck Company

83

(Darmstadt, Germany). Nitroblue tetrazolium (NBT), phenazine methosulfate (PMS), NADH, 5,5\-

84

dithiobis(2-nitrobenzoic acid) [DTNB], thiobarbituric acid (TBA) were from Sigma (St Louis, MO,

85

USA).

86

Animals

87

All procedures with animals were performed in accordance with the National Institutes of Health Guide

88

for the Care and Use of Laboratory Animals and Mansoura University guideline. Adult male rats with

89

average body weight of 120 150 g were divided into the following groups (five animals each; n=5):

90

1. Control rats (no iron was administered).

91

2. Rats treated with iron (100 M FeSO4 in a drinking water for 90 days) and treated with or

92

without 100 mg/kg curcumin in a drinking water for a further 90 days.

93

The dose of curcumin (100 mg/kg, b.w.) was selected for hepatoprotective studies by referring the

94

published report (18). In various animal studies, a dose range of 100 200 mg/kg body weight exhibited

95

good antiinflammatory activity and seemed to have negligible adverse effect on human systems. Oral

96

LD50 in mice was found to be more than 2.0 g/kg body weight (18).

97

Samples Collection:

98

All these rat groups were under experiment for (3 months), then were anesthetized and scarified. Liver

99

and spleen were collected immediately, dissected, rinsed with isotonic saline and dried between two

100

o
pieces of filter papers and then stored at 20 C in plastic vials containing 0.5 ml of ice cold sterile

101

isotonic saline.

102

Preparation of homogenates

6
103

An accurately weighed piece of each organ (1g tissue in 10 ml homogenate buffer) was homogenized in

104

ice cold homogenate buffer (0.3 M sucrose and 0.1 mM potassium dihydrogen phosphate; pH 7.0) using

105

a Teflon pestle connected to a Braun Homogenizer Motor (25 strokes/min at 1000 rpm). The

106

homogenate was centrifuged at 30,000x g for 30 min at 4oC to remove cell debris and nuclei. The

107

resulting supernatant was used for biochemical analysis.

108

Methods

109

Enzymesassays

110

The activity of superoxide dismutase (SOD) was assessed spectrophotometrically by the procedure of

111

Nishikimi et al. (19). The results were expressed as percentage of inhibition of formazane formation per

112

g wet tissue. The SOD-like activity of curcumin was assessed by the same method and its activity was

113

expressed as percent of inhibition of formazan (19). Catalase (CAT) activity was measured according to

114

the method described by Bergmeyer (20). The results were expressed as unit per g wet tissue. The CAT-

115

like activity of curcumin was determined by using the same method and its activity was determined as

116

unit/min. The activity of ceruloplasmin (Cp) was assayed as described by Henry et al. (21). The results

117

were expressed as mg of oxidation products per g wet tissue.

118

Biochemical analysis

119

The concentration of reduced glutathione (GSH) was estimated spectrophotometrically at 412 nm by

120

using DTNB as a color reagent (22). The results were expressed as mg GSH per g wet tissue. The level

121

of total ascorbic acid (AsA) in tissue homogenates was determined spectrophotometrically according to

122

the method of Zebrowski et al., (23). The results were expressed as mg ascorbic acid per g wet tissue.

123

Lipid peroxidation (LPO) in liver and spleen homogenates was estimated by measuring the formation of

124

thiobarbituric acid reactive substance (TBARS) (24). Nitric oxide (NO) concentration was determined

125

spectrophotometrically according to the method of Hortelano et al., (25). The results were expressed as

7
126

nmol nitrite level per g wet tissue. The level of hydroxyl radicals (.OH) was determined by Abou-Seif

127

and Elgendy (26). The results were expressed as nmol of thiobarbituric acid reactive substance

128

(TBARS) formed per g wet tissue.

129

Quantitative determination of total proteins

130

Protein content of homogenized samples was performed according to Lowry et al., method (27). The

131

results were expressed as g protein per g wet tissue.

132

Estimation of iron and copper levels

133

Iron and copper concentrations in liver and spleen homogenates were determined using an Atomic

134

Absorption Spectrophotometer ( Perkin-Elmer 2380, Norwalk, CT 06859-0012, USA) using an air

135

acetylene flame.

136

Statistical analysis

137

All analyses were performed with the GraphPad Prism 3 package. Data were checked for skewness, and

138

an unpaired t-test was performed if the distribution of the values was Gaussian. If the distribution was

139

not normal, a Mann-Whitney test was used. P-values less than 0.05 were considered to be statistically

140

significant.

8
141

RESULTS

142

Iron overload-induced oxidative stress in the liver as well as in the spleen and the protective effect

143

of curcumin as an iron chelator

144

Numerous pathological alterations in liver and spleen have been reported in cases with primary or

145

secondary iron overload. Furthermore, it has been well established that iron-catalyzed free radical-

146

mediated lipid peroxidation plays a significant role in these abnormal events. Therefore, the iron-

147

overloaded rats were used in this study to investigate whether curcumin, a potential iron chelator, could

148

ameliorate hepatic and splenic oxidative stress induced by excess dietary iron intake and the underlying

149

mechanisms involved therein. To test this, we first sought to verify the potential of curcumin as an iron

150

chelator in this preclinical study. This necessarily entails to get a more exact measure of iron overload.

151

Measurement of hepatic or splenic iron concentration is the most quantitative, specific, and sensitive

152

method for determining the body iron burden (28). Accordingly, these tissues were removed to

153

determine how much iron is present. Employing this measurement, the organ iron concentrations for the

154

liver and spleen are shown in Figure 1 B and C, where the chronic supplementation with iron led to

155

about a 2.6- and 3.9-fold, respectively, elevation of total hepatic and splenic iron content over control

156

pair-fed values. Treatment with curcumin resulted in a highly significant decrease in iron concentration

157

in the liver as well as in the spleen (P < 0.001 for each).

158

After

having shown the iron chelating properties of curcumin, we proposed its natural

159

implementation in preventing metal-catalyzed prooxidative effects. Thereby we next aimed to assess the

160

effect of curcumin on iron overloadinduced MDA, a terminal compound of lipid peroxidation that is

161

commonly used as an index of oxidative stress status. As expected from excessive iron deposition in the

162

liver, lipid peroxidation was induced locally as reflected by the significant increase in MDA in the

9
163

untreated iron-overloaded group compared with the normal group, and that increase in tissue MDA level

164

was reduced significantly by the curcumin treatment (Figure 2A and B).

165

Furthermore, the generation of hydroxyl radical (HO), the highly reactive and cytotoxic free

166

radical, is facilitated by iron, which catalyzes the interaction of superoxide (O2) and hydrogen peroxide

167

(H2O2) by Fenton or Haber-Weiss reactions (11). Consequently, hepatic or splenic hydroxyl radical

168

level was evaluated in iron-overloaded rats. The results showed that hepatic and splenic hydroxyl radical

169

levels were (11.75-fold and 10.5-fold, respectively) higher in iron-overloaded rats compared to non-

170

iron-overloaded. The curcumin treatment resulted in a significant decrease in hydroxyl radical level

171

(approximately 62%) compared to untreated iron-overloaded rats (Figure 2C and D). Following this

172

further, the hepatic and splenic iron overload was accompanied by a marked elevation of tissue nitric

173

oxide (NO) concentrations up to 3.5- fold in both liver and spleen (Figure 2E and F). NO is an

174

inorganic free-radical gaseous molecule that contributes to the oxidative burden and its concentrations

175

were reduced significantly by the curcumin treatment (Figure 2E and F). These observations are

176

consistent with the less severe lipid peroxidation phenotype observed in curcumin-treated rats, as free

177

radicals play a critical role in the regulation of oxidative stress.

178

Given the oxidative stress nature of iron overload-induced toxicity and the fact that free radicals

179

signaling cascade plays a pivotal role in initiating protein loss (12), we finally examined the bulk

180

degradation of proteins in liver and spleen under the chronic iron intake condition. As the continuation

181

of the work, we hypothesized that curcumin may also be effective in attenuation of iron overload-

182

induced hepatic and splenic oxidative damage and protein loss. To test this, the effect of oral ingested

183

curcumin on hepatic and splenic protein levels were determined. Iron-overload significantly (P< 0.001)

184

promoted protein loss in liver and spleen to be 137.174.1 g/wet liver tissue versus 164 8.28 g/wet

185

liver tissue in control rats and 1504.1 g/wet spleen tissue versus 185.32 3.65 g/wet spleen tissue in

10
186

control rats. Curcumin intervention resulted in a significant inhibition of iron overload-induced hepatic

187

and splenic protein loss to be 1504.6 g/wet tissue and 171.56.76 g/wet tissue, respectively.

188

In vitro antioxidant properties of Curcumin per se in scavenging harmful free radicals seen in

189

iron overload

190

In light of the aforementioned curcumins iron chelating property and anti-lipid peroxidation effects,

191

interest in its mechanisms of action has been expanded to investigate whether curcumin per se has a free

192

radical scavenging capability against the most potent reactive species, which has been implicated in the

193

pathogenesis of liver and spleen dysfunction seen in iron overload. To address this point, the capacity of

194

curcumin to scavenge hydroxyl, H2O2, and superoxide radicals was evaluated, respectively, by

195

thiobarbituric acid reactive substance (TBARS), catalase-like, and the superoxide dismutase-like

196

reaction in vitro assays. The efficiency curcumin in inhibition of hydroxyl radical generation was

197

quantified as the amount of TBARS formed, and the results are depicted in Figure 3A. Furthermore,

198

The ability of curcumin to scavenge H2O2 (catalase-like activity) and its ability to scavenge O2- (SOD-

199

like activity) were measured. As shown in Figure 3B and C, respectively, the rate of H2O2 and

200

superoxide degradation was significantly enhanced by curcumin in all doses tested. Collectively, these

201

results implied that curcumin limits iron-overload induced oxidative stress both by chelating excess iron

202

and by simultaneously functioning as an efficient radical scavenger .

203

Curcumin boosts levels of endogenous antioxidants that excessive dietary iron-intake depletes

204

The induction of oxidative stress in iron overloaded model can be caused by increased free-radical

205

generation, antioxidant depletion, or both. Given the fact that cells are armed with a variety of

206

antioxidants acting as a multilevel defenses against free radical damage, we next sought to determine

207

whether curcumin alleviation of the oxidative stress associated with iron accumulation is also mediated

11
208

by restoring endogenous antioxidant defenses. These defense systems include glutathione (GSH),

209

ascorbic acid (ASA), and enzymes such as SOD, and catalase (29).

210

As shown in Figure 4A and B, chronic iron intake was clearly associated with a significant

211

depletion of hepatic and splenic GSH when compared to non-iron overloaded rats. The curcumin

212

supplementation resulted in a great improvement of liver and spleen levels of GSH by approximately

213

65% and 90%, respectively. Moreover, the storage of ASA in livers of rats fed with excessive iron

214

intake for 12 weeks was 66% less, and spleen ascorbic acid was 70% less than that of the controls fed a

215

diet with normal iron level (Figure 4C and D). The administration of curcumin to rats receiving

216

excessive dietary iron intake has been shown to increase liver and spleen content of ascorbic acid as it

217

did with glutathione by approximately 1.5-fold.

218

Furthermore, excess iron in liver lowers the activity of catalase in homogenates of both liver and

219

spleen of chronic iron intoxication group by 40 and 50%, respectively, compared to the corresponding

220

values of normal control group, whereas the treatment with curcumin arrested the iron-induced depletion

221

of CAT enzyme activity significantly (Figure 5A and B).

222

Similarly, treatment with curcumin dramatically boosted activity of hepatic and splenic Cu-

223

SOD activity by approximately 2.4- and 2.2-fold, respectively, in chronic iron overloaded rats, whereas

224

a significant decrease in SOD activity has been observed in chronic iron overloaded rats (Figure 5C and

225

D). In practice, the most common cause of poor SOD activity in rats with chronic dietary iron overload

226

appears to be copper deficiency. At this stage, it is unclear whether the enhancement of liver and splenic

227

SOD activity seen after curcumin treatment was also achieved through modulation of organ

228

copper content and its antioxidant protein (ceruloplasmin). To delineate this preclinical association,

229

Figure 6 demonstrates the average levels of copper and ceruloplasmin in both liver and spleen

230

homogenates of iron intoxication rats either were treated with curcumin or served as non-treated controls

12
231

group. Compared to the corresponding values of the control rat group, the administration of curcumin

232

significantly restored the average of copper (Figure 6A and B) as well as ceruloplasmin levels (Figure

233

6C and D) in both liver and spleen homogenates.

234

13
235

DISCUSSION

236

Iron overload is seen in increased absorption from diets, hereditary hemochromatosis, iron loading

237

anemias, chronic liver disease, porphyria cutanea, congenital defects associated with iron overload,

238

transfusions and therapeutic injections (30). In this study, we put forward a new pre-clinical evidence for

239

introducing curcumin, an abundant and cheap micronutrient, as a multi-target therapeutic agent for

240

treating hepatosplenic complications associated with excessive dietary iron intake. Curcumin has a wide

241

therapeutic index with very low toxicity (31) and its efficacy has been demonstrated in different animal

242

models of multiple diseases associated with oxidative injury (32, 33). The most interesting feature of

243

curcumin is the lack of gastrointestinal side effects despite being an anti-inflammatory agent (18).

244

Excess iron in liver is potentially detrimental to the cell because of its propensity to participate in

245

oxidation-reduction reactions that promote tissue injury by catalyzing lipid peroxidation (34, 35). Iron

246

initiates peroxidation by producing highly reactive hydroxyl radicals from hydrogen peroxide via Fenton

247

type reactions or by complexing with oxygen directly to yield reactive perferryl and ferryl ions (12, 36).

248

The role of iron in lipid peroxidation has been well studied both in vivo and in vitro (37, 38). In

249

addition, it is also thought that a chronic increase in tissue iron level leads to depletion of antioxidants

250

which, if not restored, can by itself lead to further ROS elevation and provide a mechanism for further

251

dysfunction (12). Given the crucial participation of oxidative stress in liver and spleen failure seen in

252

chronic iron overload, an iron chelator represents an important player in modulating these pathogeneses.

253

This input has originated partly from histopathologic studies that show collagen fibrils were

254

demonstrated immediately adjacent to the hepatocytes in the iron-overloaded animals but not the

255

controls (39). These initial observations have been supported in postmortem human liver

256

specimens

257

mitochondrial and microsomal lipid peroxidation

(40) and reinforced by additional studies showing that oral iron loading induced
that was associated with selective decreases

14
258

in microsomal hemoprotein concentrations and cytochrome P450-dependent enzymes (41, 42). The

259

treatment with curcumin or antioxidants has demonstrated beneficial effects in the prevention of iron

260

induced liver toxicity (43, 44). Moreover, the in vitro studies on cultured hepatocytes with high dose of

261

iron have shown that iron induced free radical generation that kills liver cells (45).

262

All of this seemingly overwhelming evidence has sculpted the concept of iron overload-mediated

263

oxidative stress as a major contributing factor in the pathogenesis of its associated liver injury.

264

Consistent with these previous studies, we observed similar findings regarding the oxidative stress and

265

bulk protein degradation of liver and spleen under the chronic iron intake condition. However, this study

266

is the first to show the preclinical utility and translational relevance of the specific curcumins iron-

267

chelating potency during excessive iron intake. This notion was strengthen by the observation that the

268

decrease in liver and spleen iron deposition induced by curcumin treatment was associated with

269

remission of the aforementioned oxidative stress and protein loss.

270

The specific metal chelator effect of curcumin against iron which was established previously in

271

vitro (46) might be attributed to the presence of chemical groups such as -diketonate group. This

272

assertion is supported by using spectrophotometry to quantify curcumin affinity for copper, zinc, and

273

iron ions. Zn2+ showed little binding, but each Cu2+ or Fe2+ appeared to bind at least two curcumin

274

molecules. The interaction of curcumin with copper exhibited positive cooperativity, with Kd

275

approximately 10-60 M, while curcumin-iron interaction exhibited negative cooperativity, with Kd

276

approximately 0.5-1.6 M (47). A recent study reported with the intracellular free copper tracer

277

indicates that curcumin brings copper into the cells and releases it thereafter, resulting in an elevated

278

intracellular free copper level. The dissociation of copper ions from curcumin in the cells supports the

279

previous finding that the binding affinity of curcumin for copper is relatively moderate (48).

15
280

Because excess iron is known to be responsible for the functional abnormalities associated with

281

chronic iron overload, any beneficial effects of curcumin might be secondary to its action as a an iron

282

chelator. Nevertheless, the demonstration that curcumin reduces the oxidative stress in other diseases

283

(49, 50) has raised the possibility of multi-target properties beyond its role as an iron-chelating agent.

284

An alternative mechanism of these effects is the possession of antioxidant properties. These properties

285

were endorsed by the presence of chemical groups like hydroxyl, methoxy and 1,3-diketone conjugated

286

diene system (51). In the context of chronic iron overload, these two effects of curcumin cannot be

287

separated. Therefore, we studied the redox properties of curcumin against different reactive species

288

generated in vitro. From these experimental results it can be derived that curcumin is also able to reduce

289

oxidative stress during iron overload by scavenging, and/or neutralizing free radicals (such as hydroxyl

290

radical, superoxide anions and H2O2 (52) known to participate in the pathogenesis of liver and spleen

291

dysfunctions during iron overload. Accordingly, the hepato- and splenic-protective potential of

292

curcumin comes from its iron chelating property as well as its antioxidant effect.

293

Further, besides curcumins ability to chelate iron and its antioxidant potential other reported

294

effects might have contributed to our finding of curcumins salutary effects, such as restoring activities

295

of endogenous antioxidants (53). In agreement, the oxidative stress in our experimental model due to

296

chronic iron intake resulted in the depletion of putative non-enzymatic (GSH, ASA) and enzymatic

297

(CAT and SOD) antioxidants in target tissues. This depletion of antioxidants was significantly restored

298

after curcumin treatment. Thus, curcumin significantly helping revival from hepatic and splenic

299

oxidative stress associated with excessive dietary iron intake both directly or indirectly via mending the

300

levels of endogenous antioxidants.

301

Taken together, because curcumin has an attractive safety profile and appreciable iron chelating

302

property as well as anti-oxidant activity, it may represent a potential therapeutic agent for managing

16
303

excessive dietary iron intake sequelae long before the occurrence of hepatic and splenic failure among

304

those at risk for iron accumulation.

305

Disclosures

306

No conflicts of interest are declared by authors.

307

Author contributions: All the authors contributed to the researched data as well as the discussion,

308

reviewed and edited manuscript.

17
309

REFERENCES

310
311
312
313
314
315
316
317
318
319
320
321
322
323
324
325
326
327
328
329
330
331
332
333
334
335
336
337
338
339
340
341
342
343
344
345
346
347
348
349
350
351
352

1.

2.
3.
4.

5.
6.
7.
8.

9.
10.
11.
12.
13.
14.

15.
16.
17.
18.
19.
20.
21.
22.

Trumbo P, Yates AA, Schlicker S, Poos M. Dietary reference intakes: vitamin A, vitamin K,
arsenic, boron, chromium, copper, iodine, iron, manganese, molybdenum, nickel, silicon,
vanadium, and zinc. J Am Diet Assoc 2001;101:294-301.
Corbett JV. Accidental poisoning with iron supplements. MCN Am J Matern Child Nurs
1995;20:234.
Arthur CK, Isbister JP. Iron deficiency. Misunderstood, misdiagnosed and mistreated. Drugs
1987;33:171-82.
Badria F, Mandour R, Ghanem A. Impact of iron overload in drinking water on animal and
human health in Dakahlyia governorate and role of catechins as iron chelator. Journal of
Environmental Science 2007;33:25-45.
Kang JO. Chronic iron overload and toxicity: clinical chemistry perspective. Clin Lab Sci
2001;14:209-19; quiz 222.
Aisen P, Cohen G, Kang JO. Iron toxicosis. Int Rev Exp Pathol 1990;31:1-46.
Cook JD. Adaptation in iron metabolism. Am J Clin Nutr 1990;51:301-8.
Leggett BA, Brown NN, Bryant SJ, Duplock L, Powell LW, Halliday JW. Factors affecting the
concentrations of ferritin in serum in a healthy Australian population. Clin Chem 1990;36:13505.
Martins JM. Universal iron fortification of foods: the view of a hematologist. Rev Bras Hematol
Hemoter 2013;34:459-63.
Cork SC. Iron storage diseases in birds. Avian Pathol 2000;29:7-12.
Valko M, Morris H, Cronin MT. Metals, toxicity and oxidative stress. Curr Med Chem
2005;12:1161-208.
Eaton JW, Qian M. Molecular bases of cellular iron toxicity. Free Radic Biol Med 2002;32:83340.
Richardson DR. The therapeutic potential of iron chelators. Expert Opin Investig Drugs
1999;8:2141-2158.
Anand P, Thomas SG, Kunnumakkara AB, et al. Biological activities of curcumin and its
analogues (Congeners) made by man and Mother Nature. Biochem Pharmacol 2008;76:1590611.
Sharma RA, Euden SA, Platton SL, et al. Phase I clinical trial of oral curcumin: biomarkers of
systemic activity and compliance. Clin Cancer Res 2004;10:6847-54.
Aggarwal BB, Sundaram C, Malani N, Ichikawa H. Curcumin: the Indian solid gold. Adv Exp
Med Biol 2007;595:1-75.
Messner DJ, Sivam G, Kowdley KV. Curcumin reduces the toxic effects of iron loading in rat
liver epithelial cells. Liver Int 2009;29:63-72.
Kohli K, J. Ali, Ansari MJ, Rahaman Z. Curcumin: a natural anti-inflammatory agent. Ind J
Pharmacol 2005;37 141-147.
Nishikimi M, Appaji N, Yagi K. The occurrence of superoxide anion in the reaction of reduced
phenazine methosulfate and molecular oxygen. Biochem Biophys Res Commun 1972;46:849-54.
Bergmeyer H. Methods of Enzymatic Analysis. 2nd ed. New York: Academic Press, 1974.
Henry R, Chaimori N, Jacob S, Segalove M. Estimation of ceruloplasmin concentrations in
biological fluids. Proc Soc Exp Biol Med 1960;104: 620-625.
Beutler E, ed. A manual of Biochemical Methods. 3rd ed. Philadelphia: Grune & Stratton, 1984.

18
353
354
355
356
357
358
359
360
361
362
363
364
365
366
367
368
369
370
371
372
373
374
375
376
377
378
379
380
381
382
383
384
385
386
387
388
389
390
391
392
393
394
395
396
397
398

23.
24.

25.
26.
27.
28.
29.
30.
31.
32.
33.
34.

35.
36.
37.
38.

39.
40.
41.
42.
43.
44.

Zebrowski EJ, Bhatnagar PK. Urinary excretion pattern of ascorbic acid in streptozotocin
diabetic and insulin treated rats. Pharmacol Res Commun 1979;11:95-103.
Erdincler DS, Seven A, Inci F, Beger T, Candan G. Lipid peroxidation and antioxidant status in
experimental animals: effects of aging and hypercholesterolemic diet. Clin Chim Acta
1997;265:77-84.
Hortelano S, Dewez B, Genaro AM, Diaz-Guerra MJ, Bosca L. Nitric oxide is released in
regenerating liver after partial hepatectomy. Hepatology 1995;21:776-86.
Abou-Seif MA, Elgendy EM. Photolysis and membrane lipid peroxidation of human
erythrocytes by m-chloroperbenzoic acid. Clin Chim Acta 1998;277:1-11.
Lowry OH, Rosebrough NJ, Farr AL, Randall RJ. Protein measurement with the Folin phenol
reagent. J Biol Chem 1951;193:265-75.
Overmoyer BA, McLaren CE, Brittenham GM. Uniformity of liver density and nonheme
(storage) iron distribution. Arch Pathol Lab Med 1987;111:549-54.
Beckman KB, Ames BN. The free radical theory of aging matures. Physiol Rev 1998;78:547-81.
Gordeuk VR, Bacon BR, Brittenham GM. Iron overload: causes and consequences. Annu Rev
Nutr 1987;7:485-508.
Lao CD, Ruffin MTt, Normolle D, et al. Dose escalation of a curcuminoid formulation. BMC
Complement Altern Med 2006;6:10.
Epstein J, Sanderson IR, Macdonald TT. Curcumin as a therapeutic agent: the evidence from in
vitro, animal and human studies. Br J Nutr 2010;103:1545-57.
Rivera-Espinoza Y, Muriel P. Pharmacological actions of curcumin in liver diseases or damage.
Liver Int 2009;29:1457-66.
Pardo-Andreu GL, Barrios MF, Curti C, et al. Protective effects of Mangifera indica L extract
(Vimang), and its major component mangiferin, on iron-induced oxidative damage to rat serum
and liver. Pharmacol Res 2008;57:79-86.
Gao Y, Wang N, Zhang Y, et al. Mechanism of protective effects of Danshen against iron
overload-induced injury in mice. J Ethnopharmacol 2012;145:254-60.
Awai M. [Pathogenesis and mechanism of iron overload: ferric nitrilotriacetate, hemosiderin,
active oxygen, and carcinogenesis]. Rinsho Ketsueki 1989;30:1115-27.
Ryan TP, Aust SD. The role of iron in oxygen-mediated toxicities. Crit Rev Toxicol
1992;22:119-41.
Thephinlap C, Phisalaphong C, Fucharoen S, Porter JB, Srichairatanakool S. Efficacy of
curcuminoids in alleviation of iron overload and lipid peroxidation in thalassemic mice. Med
Chem 2009;5:474-82.
Weintraub LR, Goral A, Grasso J, Franzblau C, Sullivan A, Sullivan S. Pathogenesis of hepatic
fibrosis in experimental iron overload. Br J Haematol 1985;59:321-31.
Pestaner JP, Ishak KG, Mullick FG, Centeno JA. Ferrous sulfate toxicity: a review of autopsy
findings. Biol Trace Elem Res 1999;69:191-8.
Bacon BR, Healey JF, Brittenham GM, et al. Hepatic microsomal function in rats with chronic
dietary iron overload. Gastroenterology 1986;90:1844-53.
Bacon BR, Tavill AS, Brittenham GM, Park CH, Recknagel RO. Hepatic lipid peroxidation in
vivo in rats with chronic iron overload. J Clin Invest 1983;71:429-39.
Reddy AC, Lokesh BR. Effect of curcumin and eugenol on iron-induced hepatic toxicity in rats.
Toxicology 1996;107:39-45.
Inan C, Kilinc K, Kotiloglu E, Akman HO, Kilic I, Michl J. Antioxidant therapy of cobalt and
vitamin E in hemosiderosis. J Lab Clin Med 1998;132:157-65.

19
399
400
401
402
403
404
405
406
407
408
409
410
411
412
413
414
415
416
417
418
419
420
421
422
423

45.
46.

47.
48.

49.
50.
51.
52.

53.

Stal P. Iron as a hepatotoxin. Dig Dis 1995;13:205-22.


Bernabe-Pineda M, Ramirez-Silva MT, Romero-Romo MA, Gonzalez-Vergara E, RojasHernandez A. Spectrophotometric and electrochemical determination of the formation constants
of the complexes Curcumin-Fe(III)-water and Curcumin-Fe(II)-water. Spectrochim Acta A Mol
Biomol Spectrosc 2004;60:1105-13.
Baum L, Ng A. Curcumin interaction with copper and iron suggests one possible mechanism of
action in Alzheimer's disease animal models. J Alzheimers Dis 2004;6:367-77; discussion 443-9.
Lou JR, Zhang XX, Zheng J, Ding WQ. Transient metals enhance cytotoxicity of curcumin:
potential involvement of the NF-kappaB and mTOR signaling pathways. Anticancer Res
2010;30:3249-55.
Soetikno V, Sari FR, Lakshmanan AP, et al. Curcumin alleviates oxidative stress, inflammation,
and renal fibrosis in remnant kidney through the Nrf2-keap1 pathway. Mol Nutr Food Res 2012.
Fleenor BS, Sindler AL, Marvi NK, et al. Curcumin ameliorates arterial dysfunction and
oxidative stress with aging. Exp Gerontol 2012.
Miriyala S, Panchatcharam M, Rengarajulu P. Cardioprotective effects of curcumin. Adv Exp
Med Biol 2007;595:359-77.
Motterlini R, Foresti R, Bassi R, Green CJ. Curcumin, an antioxidant and anti-inflammatory
agent, induces heme oxygenase-1 and protects endothelial cells against oxidative stress. Free
Radic Biol Med 2000;28:1303-12.
Swamy AV, Gulliaya S, Thippeswamy A, Koti BC, Manjula DV. Cardioprotective effect of
curcumin against doxorubicin-induced myocardial toxicity in albino rats. Indian J Pharmacol
2012;44:73-7.

20
424

FIGURE LEGENDS

425

Figure 1. The potential of curcumin as an iron chelator in damping iron accumulation in liver and

426

spleen. A) Structural characteristic of curcumin as an iron chelator. B, C) Level of iron (g/g wet tissue)

427

in liver and spleen, respectively, from iron-overloaded rats treated or untreated with curcumin. Data

428

shown are the meanSD, (n = 5).

429

Figure 2. Iron overload-induced oxidative stress in the liver as well as in the spleen and the

430

protective effect of curcumin. A, B) Level of MDA (N mol/g wet tissue) in liver and spleen,

431

respectively, in iron overloaded rats with or without curcumin treatment. Levels of MDA in these tissues

432

are significantly increased in iron overloaded rats than control rats. Curcumin treatment significantly

433

decreased MDA levels in iron overloaded rats. C, D) Level of hydroxyl radical (N mol/g wet tissue) in

434

liver and spleen, respectively, in iron overloaded rats with or without curcumin treatment. Levels of

435

hydroxyl radical in these tissues are significantly increased in iron overloaded rats than control rats.

436

Curcumin treatment significantly decreased hydroxyl radical levels in iron overloaded rats. E, F) Level

437

of NO (N mol/g wet tissue) in liver and spleen, respectively, in iron overloaded rats with or without

438

curcumin treatment. Levels of NO in these tissues are significantly increased in iron overloaded rats than

439

control rats. Curcumin treatment significantly NO levels in iron overloaded rats. Data shown are the

440

meanSD, (n = 5).

441

Figure 3. In vitro antioxidant properties of Curcumin per se in scavenging harmful free radicals

442

seen in iron overload. A) Hydroxyl radical (OH)-Scavenging activity of curcumin measured as % of

443

TBAR inhibition. B, C) Catalase-like and Superoxide dismutase-like activities of curcumin,

444

respectively. Data shown are the meanSD, (n = 5).

21
445

Figure 4. Curcumin improves levels of endogenous non enzymatic antioxidants that excessive

446

dietary iron-intake depletes. A, B) Levels of glutathione (GSH; mg/g wet tissue) in liver and spleen,

447

respectively; C, D) Levels of ascorbic acid (ASA; mg/g wet tissue) in liver and spleen, respectively; in

448

iron overloaded rats with or without curcumin treatment. Levels of GSH and ASA in these tissues are

449

significantly decreased in iron overloaded rats than controls. Curcumin treatment significantly increased

450

GSH and ASA levels in iron overloaded rats. Data shown are the meanSD, (n = 5).

451

Figure 5. Curcumin boosts activities of endogenous enzymatic antioxidants that excessive dietary

452

iron-intake depletes. A, B) Activity of catalase (CAT; U/Min g wet tissue) in liver and spleen,

453

respectively; C, D) Activity of Superoxide dismutase (SOD; U/Min g wet tissue) in liver and spleen,

454

respectively; in iron overloaded rats with or without curcumin treatment. Activities of CAT and SOD in

455

these tissues are significantly decreased in iron overloaded rats than controls. Curcumin treatment

456

significantly boosted activities of both CAT and SOD in iron overloaded rats. Moreover, Activities of

457

CAT and SOD of curcumin-treated iron overloaded rats remained higher than those of control rats. Data

458

shown are the meanSD, (n = 5).

459

Figure 6. Curcumins modulation of organ copper content and its antioxidant protein

460

(ceruloplasmin). Levels of copper (A, B) and ceruloplasmin (C, D) in both liver and spleen

461

homogenates of iron intoxication rats either were treated with curcumin or served as non-treated controls

462

group. Compared to the corresponding values of the control rat group, the administration of curcumin in

463

iron overloaded rats significantly enhance the average of copper as well as ceruloplasmin levels in both

464

liver and spleen homogenates. Data shown are the meanSD, (n = 5).

Figure 1
A

Figure 2
A

Figure 3
A

Figure 4
A

Figure 5
A

Figure 6
A

You might also like