Skip to main content

Pre-administration of turmeric prevents methotrexate-induced liver toxicity and oxidative stress

Abstract

Background

Methotrexate (MTX) is an antimetabolite broadly used in treatment of cancer and autoimmune diseases. MTX-induced hepatotoxicity limits its application. We investigated hepatoprotective effects of turmeric in MTX-induced liver toxicity.

Methods

All experiments were performed on male Wistar albino rats that were randomly divided into six groups. Group one received saline orally for 30 days (control group), groups two and three received turmeric extract (100, 200 mg/kg respectively) orally for 30 days, group four received single dose, of MTX IP at day 30, groups five and six received turmeric extract 100 and 200 mg/kg orally respectively for 30 days and single dose of methoterxate IP (20 mg/kg) at day 30. Four days after MTX injection animals were sacrificed and evaluated. Blood ALT and AST (indicators of hepatocyte injury), ALP and bilirubin (markers of biliary function), albumin (reflect liver synthetic function) as well as the plasma TAS concentration (antioxidant defenses) were determined. The cellular antioxidant defense activities were examined in liver tissue samples using SOD, CAT, and GSH-Px for the oxidative stress, and MDA for lipid peroxidation. In addition, liver damage was evaluated histopathologically.

Results

MTX significantly induced liver damage (P < 0.05) and decreased its antioxidant capacity, while turmeric was hepatoprotective. Liver tissue microscopic evaluation showed that MTX treatment induced severe centrilobular and periportal degeneration, hyperemia of portal vein, increased artery inflammatory cells infiltration and necrosis, while all of histopathological changes were attenuated by turmeric (200 mg/kg).

Conclusion

Turmeric extract can successfully attenuate MTX-hepatotoxicity. The effect is partly mediated through extract’s antinflammatory activity.

Peer Review reports

Background

MTX has is a versatile drug that has been clinically applied in a wide range of diseases including systemic lupus erythematosus, rheumatoid arthritis, psoriasis, and neoplastic diseases [14]. The therapeutic applications of MTX is usually limited by its severe hepatotoxicity which causes malabsorption and diarrhea in the patients [5, 6]. The underlying mechanism of MTX-induced hepatotoxicity has not been fully identified, however, several distinct mechanisms have been proposed including cellular antioxidant defense deregulation which cause more oxidative stress-induced damages to the liver cells [7]. Mitochondria are the primary intracellular target for oxidative stress [8], however reactive oxygen metabolites scavenger systems like superoxide dismutase (SOD), glutathione peroxidase (GPx), catalase and glutathione-S-transferrase (GST), continuously remove ROS and maintain mitochondria function in an steady state [9, 10]. MTX is indirectly involved in liver mitochondria damage via depletion of mitochondria enzymatic and non-enzymatic antioxidants machinery [1113]. MTX administration may also cause the accumulation of reactive oxygen species (ROS), targeting macromolecules and many pathological processes [14, 15]. The first line of defense against the cell damaging effects of oxidative stress for liver is antioxidant defense system which neutralizes the deleterious effects of free radicals [16]. Therefore, the level of ROS in liver tissue may be estimated by the analysis of the antioxidant enzymes activity including superoxide dismutase (SOD), catalase (CAT), and glutathione-S-transferase (GST) [17, 18].

Turmeric (Curcuma longa L., zingiberaceae), is a yellow-colored spice derived from the perennial herb, which has been widely used for centuries in traditional therapies for different diseases including cancer, inflammatory disorders, hepatitis and other liver disorders, skin diseases, Alzheimer’s disease, rheumatoid arthitis [1922]. Besides curcumin, turmeric contains more than 300 other components, including, notably phenolics and terpenoids which may more widely and strongly affect various molecular and biochemical cascades [23, 24]. Turmeric provides protection against hepatic damage induced by free radicals and attenuates hepatic lipid peroxidation [25, 26]. Previously, it has been shown in a short term-trial (five days), that curcumin, a main integrant of turmeric, could alleviate hepatic injury induced by MTX, when applied after MTX treatment [13]. However, Gupta and colleagues concluded that the effect of whole turmeric may markedly differ from curcumin itself [27]. The effect of long term administration of whole turmeric on hepatic injury induced by MTX are yet completely unknown. In the current study, using a rat model, we have investigated the effect of pre-administration of turmeric in MTX-induced liver injury. The observed effects were assessed by measuring the levels of antioxidant enzymes, lipid peroxidation marker, total antioxidant status, as well as biochemical and histopathologic evaluations.

Methods

Animals and study protocol

Experiments were performed on male Wistar Albino rats weighing between 220–280 g purchased from Drug Applied Research Center, Tabriz University of Medicine Science. Rats were kept in 12 h light and 12 h dark cycle at the temperature of 21 ± 2 °C, food and water were available ad libitum throughout the study. Investigations using experimental animals were conducted in accordance with the internationally accepted principles for laboratory animal use and care as found in the United States guidelines (United States National Institutes for Health publication # 85–23, revised in 1985) and the Institutional Animal Ethics Committee of the Ardabil University of Medical Sciences approved the protocol (# 90374).

42 rats were randomly divided into six groups, group I received normal saline orally for 30 days (control group), group II and III received oral turmeric extract (100 and 200 mg/kg respectively) by gastric gavages for 30 days, group IV received single IP MTX (20 mg/kg) on day 30, group V received oral turmeric extract (100 mg/kg) for 30 days and IP single dose of MTX (20 mg/kg) on day 30, and group VI received oral turmeric extract (200 mg/kg) for 30 days and IP single dose of MTX (20 mg/kg) on day 30. Four days after MTX injection animals were anesthetized; blood and tissue samples were collected for different experiments. Study protocol is summarized in Table 1.

Table 1 Schematic diagram of the study protocol

Chemicals

MTX was purchased from EBEWE Pharma Ges, Austria. All kits were provided by Randox® (Cat No. NX2332, Randox laboratories, Ltd, Crumlin, UK). Dimethyl sulfoxide (DMSO), ethylene diamine tetracetic acid (EDTA) 3-(4,5-dimethylthiazol-2-yl), ethanol, ethyl acetate, Tris HCl, 2-thiobarbituric acid (TBA) and other chemicals were obtained from Sigma Chemical Co. (St Louis, MO, USA). All chemicals used were analytical grade. Turmeric (Curcuma longa L., Zingiberaceae) was purchased from the local market and identified by the Department of Cultivation and Development of Institute of Medical Plants, Tehran, Iran. The plant was then thoroughly washed, air dried, and grinded to a uniform powder. Turmeric powder (10 g) was mixed in distilled ethanol (100 ml) for 3 h at room temperature and was then centrifuged at 2500 rpm for 15 min at 4 °C. This mixture was three times filtered and then the supernatant was evaporated by employing rotary under reduced pressure at 40 °C. Such prepared turmeric extract was used at two different concentrations: 100 mg/0.2 mL, 200 mg/0.2 mL [28]. The voucher specimen (no. 12215) was deposited at the herbarium of Ardabil University of Medical Science.

Serum biochemical factors evaluation

Blood samples collected from the retro-orbital plexus were centrifuged at 2500 rpm for 15 min at 30 °C for sera preparation. The sera were then stored at −30 °C and later the following parameters were measured: alanine aminotransferase (ALT), aspartate aminotransferase (AST), alkaline phosphatase (ALP), Albumin (ALB), and total protein (TP). ALT and AST are markers for hepatocyte injury, ALP and bilirubin are sensitive markers to investigate about biliary function, while albumin reflects liver synthetic function [29]. Measurements were done according to the corresponding kits instructions using a colorimetric autoanalyzer [30].

Measurement of antioxidants

Measurement of total antioxidant status (TAS)

Blood samples were centrifuged at 4000 rpm for 10 min. Plasma TAS was measured using an automated colorimetric of the total antioxidant response (TAR) which was described previously [31]. In this method, Fe2+–o-dianisidine reacts with hydrogen peroxide by the Fenton reaction to produce OH. The hydroxyl radical then reacts with o-dianisidine to produce yellow-brown colored dianisyl product. The color formation is increased with further oxidation reactions. By adding the samples, color formation is decreased by the antioxidant components of the plasma.

Measurements of liver superoxide dismutase (SOD)

The dissected tissues were immediately frozen in liquid nitrogen and stored at −70 °C. Later the tissues were homogenized in 1.15 % KCl solution and 20 % (W/V) homogenate, which was later, centrifuged and then used to measure SOD activity using spectrophotometric method. The method was based on the inhibition of a superoxide-induced nicotinamide adenine dinucleotide (NADH) oxidation by a chemical reaction in the presence of EDTA, manganese (II) chloride and mercaptoethanol as previously described [32].

Liver glutathione peroxidase (GSH-Px) activity

GSH-Px activity was determined in liver tissues [33]. Briefly this measurement is based on the conversion between the oxidized glutathione (NADPH) to the reduced form (NADP). The rate of absorbance during this conversion was measured using spectrophotometer at 340 nm for 3 min. The activity of GSH peroxidase was expressed as nmol of NADPH oxidized per min per mg DNA.

Liver catalase (CAT) activities

The method was previously described by Claiborne group [34]. Briefly 50 μL of liver tissue lysate was added to of H2O2 (10 mM, 0.95 mL) and later to reaction buffer (60 mM phosphate buffer; pH 7.0). Catalase activity was measured as micromoles of H2O2 consumed per minute at 25 °C. The changes in absorbance were recorded at 240 nm using spectrophotometer.

Liver lipid peroxidation assay

Oxidative stress was evaluated using Malondialdehyde (MDA) equivalents which were assessed utilizing the thiobarbituric acid (TBARS) as reactive substrate [35]. The colorimetric reaction between MDA and TBARS) were assayed (pH 2–3, 90 °C) for 15 min. The maximum absorption was recorded at 532 nm.

Histopathological analysis

Liver tissue samples were taken from each animal, fixed in 10 % neutral formalin and after histological process of dehydration, tissue embedded in paraffin, section with a microtome set at a thickness of 5–6 μm. The tissue sections were stained with hematoxylin and eosin (H&E) for histopathological analysis and examined with a light microscope. The degree of liver tissue injury was evaluated semiquantitatively according to the method reported by Jamshidzadeh et al. [36]. The stained 5 μm sections were graded as follows: 0, absent; 1, minimal; 2, mild; 3, modest; 4, severe. The histological changes were evaluated in nonconsecutive, randomly chosen × 200 histological fields using light microscope, NIKON ECLIPSE E200. Neutrophils were counted in 50 high-power fields (×400). The pathologist performing the histologic evaluation had no knowledge about the treatment of the animals, from which the samples originated.

Data analysis

The Statistical Package for Social Sciences (SPSS Inc., Chicago, IL, USA), version 17.0, was used for statistical analysis. All data are presented as mean ± SD. Before statistical analysis, all variables were checked for normality and homogeneity of variance by using the Kolmogorov-Smirnoff and Levene tests, respectively. The data obtained were tested by ANOVA followed by Tukey’s posthoc multiple comparison test. The Kruskal-Wallis test, followed by Mann–Whitney U posttest, was used for the analysis of degree of histopathological liver injury. P < 0.05 was considered statistically significant.

Results

Effect of turmeric on antioxidant status in rat liver

The liver antioxidant markers (SOD, GSH-Px and CAT) are shown in Table 2 for different groups. MTX significantly (P < 0.05) decreased SOD, GSH-Px and CAT activities in liver tissue compared to control, turmeric (100 mg/kg) and turmeric (200 mg/kg) groups. However, the groups, which were treated with MTX in presence of TUR, (200 mg/kg) showed significant increase in SOD, GSH-Px and CAT activity in liver tissue compared to MTX group (P < 0.05). On the other hand turmeric (100 mg/kg) and turmeric (200 mg/kg) control groups did not show any significant changes in the activities of SOD and GSH-Px, while CAT level were significantly increased compared to control group in the liver tissue (P < 0.05).

Table 2 Effect of turmeric on antioxidant status in rat liver

Effect of turmeric on lipid proxidation and total antioxidant status in liver and plasma

The hallmarks of lipid peroxidation and total antioxidant (MDA and TAS) are shown in Table 3. Liver’s MDA content was significantly increased in MTX group compared to control group (P < 0.05). Whereas, the concentration of MDA in liver tissue was significantly decreased in group VI treated with turmeric 200 and MTX when compared to MTX group (P < 0.05). TAS was significantly decreased in MTX group while turmeric 200 mg/kg induce an increase in plasma TAS compared to control group (P < 0.05). In the group which TUR 200 and MTX was used, plasma TAS was significantly elevated compared to MTX group (P < 0.05).

Table 3 Effect of turmeric on lipid proxidation and total antioxidant status in liver and plasma

Effect of turmeric on the serum ALT, AST, ALP, Bilirubin, ALB and TP

The results of serum liver biochemical hallmarks are presented in Table 4. MTX treatment significantly increased serum ALT, AST, ALP activity and bilirubin concentration (P < 0.05) while induced a significant decrease in serum ALB and TP (P < 0.05) compared to control group. Also, the same trends were observed in comparison between the group, which was treated with turmeric 100 + MTX and the control group. The group, which was treated with turmeric + MTX, significant had lower ALT, AST, ALP serum activity, and bilirubin concentration compared to the MTX group (P < 0.05). Likewise, significant increases were observed in ALB and TP concentrations in turmeric 200 + MTX group, compared to MTX group (P < 0.05).

Table 4 Effect of turmeric on the serum levels of ALT, AST, ALP, Bilirubin, ALB and TP

Effect of turmeric on liver to body weight ratio

The liver/body weight ratios in different groups are shown in Fig. 1. The body weight loss was observed for rats receiving MTX (P < 0.01) and MTX + turmeric 100 mg/kg (group V) (P < 0.05) compared to control (Fig. 1a). The body weight loss in-group treated with MTX + turmeric 200 mg/kg was also observed, as compared with the control group. However, the body weight loss was not significant (P > 0.05) in this group. The liver weights of groups IV, V, VI were significantly higher than the control group, suggesting hypertrophy of the liver (Fig. 1b). This is further emphasized by the fact that liver weight/body weight ratio of group IV was significantly (P < 0.01) higher than the groups I, II, and III (Fig. 1c). In addition, a significant decrease in liver to body weight ratio was observed in animals treated with MTX + turmeric 200 mg/kg compared to group receiving MTX (P < 0.05). The liver/body weight ratio was higher in the group given MTX + turmeric 100 mg/kg in comparison with the control group (P < 0.05) (Fig. 1c).

Fig. 1
figure 1

Effect of turmeric and methotrexate on body weight, liver weight, and liver weight/body weight ratio. a The body weight, (b) liver weight, and (c) body weight/liver weight ratio are shown. Values of body weight and liver weight are shown as mean ± S.D.* P < 0.01 compared with control, ** P < 0.05 compared with control, *** P < 0.05 compared with MTX group

Histopathology study

Histopathology examination of liver sections of the normal group were stained with H&E, showed in low magnification with normal appearance of central vein (cv, indicated by arrow) and portal triad structures (arrowhead) (Fig. 2ab). The cord-like structure of distinct polyhedral-shaped large hepatocytes well-preserved cytoplasm and rounded euchromatic nucleus with prominent nuclei showed around the central vein (pericentral zone) (Fig. 2c). The normal appearance of portal vein (v), artery (a), and bile duct (d) showed in portal triad that embedded in hepatocytes cords in periportal zone (Fig. 2d).

Fig. 2
figure 2

Histopathology examination of liver sections of the control group. a, b Microscopic view from the liver tissue of a rat belonged to healthy/control group stained with H&E (group I). c Microscopic view from the pericentral zone of rat liver stained with H&E (group I). d Microscopic view from the periportal zone of rat liver stained with H&E. Portal triad structures comprise of portal vein (v), hepatic artery (a), and bile duct (d) (group I). (central vein = cv), Scale bars = 200 μm in A; 100 μm in B; 20 μm in C and D

In turmeric (100 mg/kg) treatment group, no obvious histopathological changes were observed in the liver section of this group in pericentral zone (Fig. 3a) and periportal zone (Fig. 3b). In examination of hepatic tissue from turmeric (200 mg/kg) treatment group, liver tissue had normal appearance and there were no histopathological changes as shown in pericentral zone (Fig. 3c) and periportal zone (Fig. 3d).

Fig. 3
figure 3

Histopathology examination of liver sections of the group treated with tumeric. a Microscopic view from the pericentral zone of a rat liver belonged to turmeric (100 mg/kg) treatment group stained with H&E (groups II and III). b Microscopic view from the periportal zone of a rat liver belonged to turmeric (100 mg/kg) treatment group stained with H&E. c Microscopic view from the pericentral zone of a rat liver belonged to turmeric (200 mg/kg) treatment group stained with H&E (groups II and III). d Microscopic view from the periportal zone of a rat liver belonged to turmeric (200 mg/kg) treatment group stained with H&E (groups II and III). Portal triad structures comprise of portal vein (v), hepatic artery (a), and bile duct (d) (central vein = cv, Scale bars = 20 μm in A, B, C and D)

Examination of liver tissues from MTX treatment group showed severe histopathological changes that prominent around dilation and congestion of central vein (cv) and portal vein (v) (Fig. 4). In pericentral zone of liver tissues from MTX treatment group showed inflammation and degenerating hepatocytes with condense nuclei (Fig. 4ab). The severe histopathological changes were prominent in periportal zone with appearance of hyperemia of portal vein, and inflammatory cells infiltration (Fig. 4cd). The necrotic cells and degenerating hepatocytes with condense nuclei and lack of polygonal-shaped outline were obvious in periportal zone that indicate severe toxicity effects of MTX to liver and hepatocyte (Fig. 4d).

Fig. 4
figure 4

Histopathology examination of liver sections of the group treated with MTX. a, b Microscopic view from the pericental zone rat liver belonged to MTX-group stained with H&E (group IV). c, d Microscopic view from the periportal zone of rat liver belonged to MTX-group stained with H&E. Portal triad structures comprise of portal vein (v), hepatic artery (a), and bile duct (d) (group IV) (central vein = cv). Scale bars = 20 μm in A and D; 100 μm in B

To determine whether pretreatment of turmeric reduces the toxicity of MTX, we have treated animals with turmeric (100 and 200 mg/kg) and followed by administration of MTX. The histopathological studies of liver section from showed an improvement in damages caused by MTX to the hepatic tissues in a dose dependent manner (Fig. 5). Microscopic image from liver tissues of animals which have been treated with MTX in the presence of turmeric (100 mg/kg) (group IV) showed moderate pericentral zone hepatocytes degeneration with clear nuclei and nucleoli (Fig. 5a). A section from portal triad area showed that periportal zone has a moderate degeneration while mild hyperemia was still retained in portal vein (Fig. 5b). MTX treatment in presence of turmeric (200 mg/kg) treatment (group VI), showed degeneration significantly decreased in pericentral zone (Fig. 5c) and Periportal zone (Fig. 5d). Histopathological examination of livers form group VI shows that most of hepatocytes appeared with concentric arrangement and prominent nuclei and nucleoli. Quantitative assessment of the samples shown in Fig. 6 (microscopic evaluation) are presented in Table 5.

Fig. 5
figure 5

Histopathology examination of liver sections of the group treated with tumeric and MTX. a Microscopic view from the pericentral zone of a rat liver belonged to turmeric (100 mg/kg) + MTX treatment group stained with H&E (groups V and VI). b Microscopic view from the periportal zone of a rat liver belonged to turmeric (100 mg/kg) + MTX treatment group stained witch H&E (groups V and VI). c Microscopic view from the pericentral zone of a rat liver belonged to turmeric (200 mg/kg) + MTX treatment group stained with H&E (groups V and VI). d Microscopic view from the periportal zone of a rat liver belonged to turmeric (200 mg/kg) + MTX treatment group stained with H&E (groups V and VI). Portal triad structures comprise of portal vein (v), hepatic artery (a), and bile duct (d) (central vein = cv). Scale bars = 20 μm in A, B, C and D

Fig. 6
figure 6

Effect of turmeric on PMN content of MTX-induced hepatic injuries. The number of neutrophils was obtained by counting these cells in 50 high-power fields. In control livers, relatively few neutrophils were found at sites of erosion. Similar results were obtained in group II and III. There was a significant increase in the number of neutrophils in the liver of rats treated with MTX (P < 0.05). There was no statistically significant difference between number of neutrophils in rats treated with MTX + turmeric (100 mg/kg) as compared to livers obtained from rats injected with MTX (group IV). However, the number of neutrophils accumulating in the liver in group (MTX + turmeric 200 mg/kg) has been significantly different compared to MTX treatment group (P < 0.05). Values are shown as mean ± S.D. * P < 0.05 compared with control, ** P < 0.05 compared with MTX group

Table 5 Effect of turmeric on hepatic injuries

We have next assessed the liver infiltration by neutrophils. In control livers, relatively few neutrophils were observed in sinusoids (7 ± 2 PMN/50 HPF). The turmeric treatment alone (100 and 200 mg/kg) had no significant effect on PMN level in the treated groups (7 ± 1 & 6 ± 1 PMN/50 HPF). The number of neutrophils increased significantly (P < 0.05) in MTX treated groups (237 ± 8 PMN/50 HPF). In general, turmeric at 100 mg/kg, had no significant effect on hepatic neutrophile counts (210 ± 5 PMN/50 HPF) regardless of the group. In contrast, turmeric at the dose of 200 mg/kg (21 ± 3 PMN/50 HPF) had significant effect (P < 0.05) when compared between MTX treated groups (Fig. 6).

Discussion

Several histopathology and serology markers have been previously used to investigate MTX-induced liver toxicity, including increased relative fatty acid changes in hepatocytes and sinusoidal lining cells, necrosis, inflammation and elevated activities of serum ALT and AST [13, 37, 38]. In the current study, histology outcomes confirm MTX-induced hepatotoxicity, and biochemical results are in agreement with the histological findings. As MTX mechanism of action is involved in folate antagonism, some of its toxicity effects such as anemia, neutropenia and stomatitis could be prevented or alleviated by folate supplementation. But there are other effects, which are unrelated to suppression of folate metabolism, including hepatic fibrosis, pulmonary fibrosis, nodulosis, lethargy and renal insufficiency [3941]. Several studies have shown that MTX-induced hepatic fibrosis is mediated through oxidation and adenosine pathways (Fig. 7). Adenosine is a potent endogenous regulator of inflammation and tissue repair. Adenosine, which might be released from injured and hypoxic tissue in response to toxins and medications, may induce hepatic fibrosis in mice, presumably via adenosine receptor [4244].

Fig. 7
figure 7

Schematic representation of the beneficial effects of turmeric on MTX induced liver toxicity. Turmeric plays an important role in modification of liver injury caused by MTX. The turmeric mostly exerts its effects by regulation of antioxidant capacity including enzymatic (SOD and CAT) and non-enzymatic antioxidants (GSH) as well as lipid peroxidation. It also modulates some liver related biochemical parameters causing in improvement of biliary and hepatic synthetic functions. Interestingly turmeric can normalize the histological changes in hepatocytes induced by MTX such as decrease in periportal degeneration, hyperemia, necrosis, and prevention of inflammatory cells infiltration

Our results showed prominent pathological changes in the liver tissue of MTX-treated animals including inflammatory cells infiltration, severe centrilobular/intermediate zone/ periportal degeneration, bile duct hyperplasia, hyperemia and necrosis observed compared to control and turmeric groups. Interestingly turmeric ameliorated the histological alteration in hepatocytes induced by MTX including decrease in periportal degeneration, hyperemia, necrosis, and prevention of inflammatory cells infiltration. The histology results were later confirmed by biochemical findings as have been mentioned in the previous sections.

Curcumin is the prevalent ingredient of turmeric and has been shown to possess several pleiotropic beneficial effects in many iatrogenic organ maladies [45, 46]. Animal studies have shown that curcumin might have positive effect against a wide range of human diseases, including diabetes, obesity, neurologic and psychiatric disorders, and cancer, as well as chronic illnesses affecting the eyes, lungs, liver, kidneys, and gastrointestinal and cardiovascular systems [4749]. Curcumin has anti-inflammatory, anti-oxidant, pro-apoptotic, chemopreventive, chemotherapeutic, anti-proliferative, wound healing, anti-nociceptive, anti-parasitic, and anti-malarial effects as well [5052]. However, besides curcumin, Tumeric extracts cointain many other active constituents such as turmerin, demethoxycurcumin, bisdemethoxycurcumin (two main active components of curcuminoids), turmeric proteins content, fats, minerals, carbohydrates, and essential oils, which could affect different biological functions and target specific organs, which depends on their molecular targets [53, 54]. Studies, over the two past decades, on turmeric extracts have indicated that its activities favorably compare to pure curcumin [27]. For example, curcumin is less potent than turmeric in delaying STZ-induced cataract [53] and in reducing blood glucose levels in type 2 diabetic [55].

Oxidative stress-induced cell damage is a major factor in the pathogenesis of many acute and chronic diseases [5659]. The cell damage is initiated by the reaction of free radicals with membrane lipids and polyunsaturated fatty acids [60]. The current study showed that a single dose of MTX (20 mg/kg) caused significant hepatotoxicity. The level of MDA in liver tissue was significantly increased in MTX-treated rats compared to control group, which suggested that the peroxidation reaction was higher than control group in MTX-treated animals and correlated with the previous research findings about MTX-induced hepatoxocity [7, 11, 61, 62]. Our findings suggest that turmeric ROS scavenging-induced protective response markedly decreased MDA concentration. Comparable antioxidant effects were reported by other researchers in pulmonary, neurological, metabolic, and cardiovascular diseases [27]. Turmeric pre-treatment (200 mg/kg) showed higher improvement and recovery of liver tissue in presence of lipid peroxidation inhibition (e.g. 30 %) compared to group treated with turmeric (100 mg/kg). Hemeida and colleagues have previously shown that curcumin post-injection could decrease MTX-induced MDA production by around 24 % [13].

Our study also indicate that MTX-induced liver injury activates neutrophiles and results in liver infiltration by PMN. Neutrophil accumulation in the liver occurred parallel to the development of hemorrhage and parenchymal cell injury. Infiltration of neutrophils of liver tissues was concomitant with the oxidant/antioxidant status and changes in MDA levels. Increased MDA levels in the rat-livers after MTX administration suggests activation of an inflammatory response. We observed increased MDA levels in the liver tissue, and this may indicate that neutrophil accumulation and lipid peroxidation contributes to MTX-induced liver injury. Likely, the activated neutrophils located in the area of inflammation, may convert hydroperoxides into free radicals, triggering lipid peroxidation and hepatocyte cell membrane damage.

In our study, SOD and CAT activities were also significantly decreased in MTX treated group compared to healthy control group. SOD and CAT are the main enzymes, which are involved in H2O2 and ROS scavenging [63]. SOD and CAT activities were increased in all groups treated with turmeric-MTX compared to groups, which received MTX alone. SOD activity was increased 27 % and 75 % in the groups, which were pre-treated with tumeric (100 mg/kg and 200 mg/kg) before MTX exposure respectively, while CAT activity were increased 32 % and 105 % respectively. Similar results, in different tissues, were reported by others [13, 6466].

The present study also shows that MTX could reduce GSH, which indicates imbalance between antioxidants and oxidants in the liver. As GSH is one of the most important parts of antioxidant defense system [67], when MTX decreases the level of GSH, cellular redox state alters and cells could be more sensitive to reactive oxygen metabolites [68]. Such findings have been observed in other experimental systems [69]. One of the possible mechanisms related to GSH depletion is based on the decrease of NADPH availability due to MTX inhibitory effects on glucose 6 phosphate dehydrogenase [70]. Glutamine is necessary for glutathione production. A study indicated that intravenous glutamine supplementation could protects liver cells from MTX-induced oxidative damage due to increasing intercellular GSH level [71] as turmeric increased GSH level with unknown mechanism. Our results show that turmeric not only affected enzymatic cellular antioxidant machinery (SOD and CAT), but also non-enzymatic defense system (GSH). Preadministration of turmeric (100 and 200 mg/kg) in groups V and VI for 30 days can significantly restore the GSH level about 34 % and 54 % respectively compared to groups that received MTX alone.

Total antioxidant status (TAS) reflects the total effect of all antioxidants in plasma [72]. In our study, the level of TAS in rats treated with MTX, were significantly lower compared to healthy control group. Previous studies indicated decreased antioxidant capability and increased oxidative stresses provokes hepatic damage [42, 56, 73]. In groups V and VI (turmeric + MTX), serum TAS were elevated compared to group III (MTX treated animals), but statistically significant results were obtained when we used higher concentration of turmeric (200 mg/kg). We have previously shown that dietary turmeric can markedly enhance TAS level under physilogical conditions in rats [74].

In our study, MTX increased serum activities of ALT, AST, ALP and bilirubin serum concentration and decreased the concentrations of ALB and TP, which are hepatotoxicity and impaired liver function indicators [75, 76]. ALT and AST are the major critical enzymes in the biological processes [77, 78]. Reports have suggested that their levels increase in different hepatic injures such as hepatitis and cirrhosis induced by alcohol, drugs, viruses and also under oxidative stress [79]. Also there are not similar studies about ALP, ALB, and TP levels in MTX induced liver toxicity. Our investigation revealed that in turmeric 200 + MTX group, serum activities of ALT, AST, ALP and serum bilirubin concentration were decreased, and levels of ALB and TP were increased compared to MTX group which correlates to previous reports [13, 80]. The low and high concentrations of turmeric did not induce any significant changes in ALT, AST and ALP levels compared to control group which confirms the previous finding [81]. Recently Fu et al. demonstrated that turmeric could effectively protect liver against CCL4-induced injury. They also showed that it prevented AST and ALT augmentation and improved the liver cell function [76]. In a randomized clinical trial on patients with increased levels of ALT, fermented turmeric powder decreased serum ALT and AST while there were not any significant changes in serum ALP, TB and lipids concentrations [82]. The results are varying among the investigators because of different methods of study or different population, which has been studied.

Conclusion

In conclusion turmeric could prevent MTX hepatotoxicity and it might have a great potential for application in future disease therapy strategies. Liver-, breast- and other cancers often require aggressive chemotherapy, preferably targeting cancer stem cells [8389]. The future clinical trials should be arranged to investigate the actual effect of turmeric in human patients and the possibility of considering this medical herb as a potential additive to MTX anticancer drugs.

Abbreviations

ALP:

Albumin

ALP:

Alkaline phosphatase

ALT:

Alanine transaminase

ANOVA:

Analysis of variance

AST:

Aspartate Aminotransferase

BDH:

Bile duct hyperplasia

CAT:

Catalase

CV:

Central vein

DMSO:

Dimethyl sulfoxide

EDTA:

Ethylene diamine tetracetic acid

GPx:

Glutathione peroxidase

GSH:

Glutathione

GSH-Px:

Glutathione peroxidase

GST:

Glutathione-S-transferrase

H&E:

Hematoxylin and Eosin

HPF:

High-power fields

ICI:

Inflammatory cells infiltration

LPO:

lipid proxidation

MDA:

Malondialdehyde

MTX:

Methotrexate

NADH:

Nicotinamide adenine dinucleotide

PMN:

Neutrophils

PV:

Portal vein

ROS:

Reactive oxygen species

SOD:

Superoxide dismutase

TAR:

Total antioxidant response

TAS:

Total antioxidant status

TBA:

2-thiobarbituric acid

TBARS:

Thiobarbituric acid reactive substances

TP:

Total protein

References

  1. Sakthiswary R, Suresh E. Methotrexate in systemic lupus erythematosus: a systematic review of its efficacy. Lupus. 2014;23:225–35. 0961203313519159.

    CAS  PubMed  Google Scholar 

  2. Zhu H, Deng F-Y, Mo X-B, Qiu Y-H, Lei S-F. Pharmacogenetics and pharmacogenomics for rheumatoid arthritis responsiveness to methotrexate treatment: the 2013 update. Pharmacogenomics. 2014;15(4):551–66.

    CAS  PubMed  Google Scholar 

  3. Maybury CM, Jabbar-Lopez ZK, Wong T, Dhillon AP, Barker JN, Smith CH. Methotrexate and liver fibrosis in people with psoriasis: a systematic review of observational studies. Br J Dermatol. 2014;171(1):17–29.

    CAS  PubMed  Google Scholar 

  4. Ayad MW, El Naggar AA, El Naggar M. MTHFR C677T polymorphism: association with lymphoid neoplasm and effect on methotrexate therapy. Eur J Haematol. 2014;93(1):63–9.

    CAS  PubMed  Google Scholar 

  5. Triantafyllou K, Vlachogiannakos J, Ladas SD. Gastrointestinal and liver side effects of drugs in elderly patients. Best Pract Res Clin Gastroenterol. 2010;24(2):203–15.

    CAS  PubMed  Google Scholar 

  6. Duman DG, Kumral ZNÖ, Ercan F, Deniz M, Can G, Çağlayan Yeğen B. Saccharomyces boulardii ameliorates clarithromycin-and methotrexate-induced intestinal and hepatic injury in rats. Br J Nutr. 2013;110(03):493–9.

    CAS  PubMed  Google Scholar 

  7. Ali N, Rashid S, Nafees S, Hasan SK, Sultana S. Beneficial effects of Chrysin against Methotrexate-induced hepatotoxicity via attenuation of oxidative stress and apoptosis. Mol Cell Biochem. 2014;385(1–2):215–23.

    CAS  PubMed  Google Scholar 

  8. Messarah M, Boumendjel A, Chouabia A, Klibet F, Abdennour C, Boulakoud MS, et al. Influence of thyroid dysfunction on liver lipid peroxidation and antioxidant status in experimental rats. Exp Toxicol Pathol. 2010;62(3):301–10.

    CAS  PubMed  Google Scholar 

  9. Richter C, Gogvadze V, Laffranchi R, Schlapbach R, Schweizer M, Suter M, et al. Oxidants in mitochondria: from physiology to diseases. Biochim Biophys Acta. 1995;1271(1):67–74.

    PubMed  Google Scholar 

  10. Nickel A, Kohlhaas M, Maack C. Mitochondrial reactive oxygen species production and elimination. J Mol Cell Cardiol. 2014;73:26–33.

    CAS  PubMed  Google Scholar 

  11. Tabassum H, Parvez S, Pasha ST, Banerjee BD, Raisuddin S. Protective effect of lipoic acid against methotrexate-induced oxidative stress in liver mitochondria. Food Chem Toxicol. 2010;48(7):1973–9.

    CAS  PubMed  Google Scholar 

  12. Kolli V, Natarajan K, Isaac B, Selvakumar D, Abraham P. Mitochondrial dysfunction and respiratory chain defects in a rodent model of methotrexate-induced enteritis. Hum Exp Toxicol. 2013;33(10):1051–65. 0960327113515503.

    PubMed  Google Scholar 

  13. Hemeida RA, Mohafez OM. Curcumin attenuates methotraxate-induced hepatic oxidative damage in rats. J Egypt Natl Canc Inst. 2008;20(2):141–8.

    PubMed  Google Scholar 

  14. Bartsch H, Nair J. Ultrasensitive and specific detection methods for exocylic DNA adducts: markers for lipid peroxidation and oxidative stress. Toxicology. 2000;153(1):105–14.

    CAS  PubMed  Google Scholar 

  15. Stohs S, Bagchi D. Oxidative mechanisms in the toxicity of metal ions. Free Radic Biol Med. 1995;18(2):321–36.

    CAS  PubMed  Google Scholar 

  16. Padma VV, Baskaran R, Roopesh RS, Poornima P. Quercetin attenuates lindane induced oxidative stress in wistar rats. Mol Biol Rep. 2012;39(6):6895–905.

    CAS  PubMed  Google Scholar 

  17. Uz E, Söğüt S, Şahin Ş, Var A, Özyurt H, Güleç M, et al. The protective role of caffeic acid phenethyl ester (CAPE) on testicular tissue after testicular torsion and detorsion. World J Urol. 2002;20(4):264–70.

    CAS  PubMed  Google Scholar 

  18. Uzar E, Koyuncuoglu HR, Uz E, Yilmaz HR, Kutluhan S, Kilbas S, et al. The activities of antioxidant enzymes and the level of malondialdehyde in cerebellum of rats subjected to methotrexate: protective effect of caffeic acid phenethyl ester. Mol Cell Biochem. 2006;291(1–2):63–8.

    CAS  PubMed  Google Scholar 

  19. Lai CS, Wu JC, Yu SF, Badmaev V, Nagabhushanam K, Ho CT, et al. Tetrahydrocurcumin is more effective than curcumin in preventing azoxymethane-induced colon carcinogenesis. Mol Nutr Food Res. 2011;55(12):1819–28.

    CAS  PubMed  Google Scholar 

  20. Kuttan R, Bhanumathy P, Nirmala K, George M. Potential anticancer activity of turmeric (< i > Curcuma longa</i>). Cancer Lett. 1985;29(2):197–202.

    CAS  PubMed  Google Scholar 

  21. Chattopadhyay I, Biswas K, Bandyopadhyay U, Banerjee RK. Turmeric and curcumin: Biological actions and medicinal applications. Curr Sci. 2004;87(1):44–53.

    CAS  Google Scholar 

  22. Mishra S, Palanivelu K. The effect of curcumin (turmeric) on Alzheimer’s disease: An overview. Ann Indian Acad Neurol. 2008;11(1):13.

    PubMed  PubMed Central  Google Scholar 

  23. Li S, Yuan W, Deng G, Wang P, Yang P, Aggarwal BB. Chemical composition and product quality control of turmeric (Curcuma longa L.). Pharmaceutical Crops. 2011;2:28–54.

    CAS  Google Scholar 

  24. Gupta SC, Sung B, Kim JH, Prasad S, Li S, Aggarwal BB. Multitargeting by turmeric, the golden spice: from kitchen to clinic. Mol Nutr Food Res. 2013;57(9):1510–28.

    CAS  PubMed  Google Scholar 

  25. Reddy ACP, Lokesh B. Effect of dietary turmeric (Curcuma longa) on iron-induced lipid peroxidation in the rat liver. Food Chem Toxicol. 1994;32(3):279–83.

    CAS  PubMed  Google Scholar 

  26. Selvam R, Subramanian L, Gayathri R, Angayarkanni N. The anti-oxidant activity of turmeric (Curcuma longa). J Ethnopharmacol. 1995;47(2):59–67.

    CAS  PubMed  Google Scholar 

  27. Gupta SC, Kismali G, Aggarwal BB. Curcumin, a component of turmeric: from farm to pharmacy. Biofactors. 2013;39(1):2–13.

    CAS  PubMed  Google Scholar 

  28. Katiyar SK, Agarwal R, Mukhtar H. Inhibition of tumor promotion in SENCAR mouse skin by ethanol extract of Zingiber officinale rhizome. Cancer Res. 1996;56(5):1023–30.

    CAS  PubMed  Google Scholar 

  29. Pari L, Murugan P. Protective role of tetrahydrocurcumin against erythromycin estolate-induced hepatotoxicity. Pharmacol Res. 2004;49(5):481–6.

    CAS  PubMed  Google Scholar 

  30. Woreta TA, Alqahtani SA. Evaluation of abnormal liver tests. Med Clin North Am. 2014;98(1):1–16.

    PubMed  Google Scholar 

  31. Erel O. A novel automated method to measure total antioxidant response against potent free radical reactions. Clin Biochem. 2004;37(2):112–9.

    CAS  PubMed  Google Scholar 

  32. Paoletti F, Aldinucci D, Mocali A, Caparrini A. A sensitive spectrophotometric method for the determination of superoxide dismutase activity in tissue extracts. Anal Biochem. 1986;154(2):536–41.

    CAS  PubMed  Google Scholar 

  33. Paglia DE, Valentine WN. Studies on the quantitative and qualitative characterization of erythrocyte glutathione peroxidase. J Lab Clin Med. 1967;70(1):158–69.

    CAS  PubMed  Google Scholar 

  34. Claiborne A. Handbook of methods for oxygen radical research. Florida: CRC Press, Boca Raton; 1985.

    Google Scholar 

  35. Esterbauer H, Cheeseman K. Determination of aldehydic lipid peroxidation products: malonaldehyde and 4-hydroxynonenal. Methods Enzymol. 1990;186:407–21.

    CAS  PubMed  Google Scholar 

  36. Jamshidzadeh A, Baghban M, Azarpira N, Bardbori AM, Niknahad H. Effects of tomato extract on oxidative stress induced toxicity in different organs of rats. Food Chem Toxicol. 2008;46(12):3612–5.

    CAS  PubMed  Google Scholar 

  37. Tian H, Cronstein BN. Understanding the mechanisms of action of methotrexate. ull NYU Hosp Jt Dis. 2007;65(3):168–73.

    Google Scholar 

  38. van Swelm RP, Laarakkers CM, Kooijmans-Otero M, de Jong EM, Masereeuw R, Russel FG. Biomarkers for methotrexate-induced liver injury: Urinary protein profiling of psoriasis patients. Toxicol Lett. 2013;221(3):219–24.

    PubMed  Google Scholar 

  39. Tian H, Cronstein BN. Understanding the mechanisms of action of methotrexate: implications for the treatment of rheumatoid arthritis. Bull NYU Hosp Jt Dis. 2007;65(3):168–73.

    PubMed  Google Scholar 

  40. Berkun Y, Levartovsky D, Rubinow A, Orbach H, Aamar S, Grenader T, et al. Methotrexate related adverse effects in patients with rheumatoid arthritis are associated with the A1298C polymorphism of the MTHFR gene. Ann Rheum Dis. 2004;63(10):1227–31.

    CAS  PubMed  PubMed Central  Google Scholar 

  41. Sitzia J, Huggins L. Side effects of cyclophosphamide, methotrexate, and 5‐fluorouracil (CMF) chemotherapy for breast cancer. Cancer Pract. 1998;6(1):13–21.

    CAS  PubMed  Google Scholar 

  42. Tsai W-H, Yang C-C, Li P-C, Chen W-C, Chien C-T. Therapeutic Potential of Traditional Chinese Medicine on Inflammatory Diseases. J Tradit Complement Med. 2013;3(3):142.

    PubMed  PubMed Central  Google Scholar 

  43. Wang H, Guan W, Yang W, Wang Q, Zhao H, Yang F, et al. Caffeine Inhibits the Activation of Hepatic Stellate Cells Induced by Acetaldehyde via Adenosine A2A Receptor Mediated by the cAMP/PKA/SRC/ERK1/2/P38 MAPK Signal Pathway. PLoS One. 2014;9(3):e92482.

    PubMed  PubMed Central  Google Scholar 

  44. Chan ES, Cronstein BN. Methotrexate—how does it really work? Nat Rev Rheumatol. 2010;6(3):175–8.

    CAS  PubMed  Google Scholar 

  45. Arafa HM. Curcumin attenuates diet-induced hypercholesterolemia in rats. Med Sci Monit. 2005;11(7):BR228–234.

    CAS  PubMed  Google Scholar 

  46. 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(11):1545–57.

    CAS  PubMed  Google Scholar 

  47. Shehzad A, Lee YS. Molecular mechanisms of curcumin action: signal transduction. Biofactors. 2013;39(1):27–36.

    CAS  PubMed  Google Scholar 

  48. Wongcharoen W, Phrommintikul A. The protective role of curcumin in cardiovascular diseases. Int J Cardiol. 2009;133(2):145–51.

    PubMed  Google Scholar 

  49. Weisberg SP, Leibel R, Tortoriello DV. Dietary curcumin significantly improves obesity-associated inflammation and diabetes in mouse models of diabesity. Endocrinology. 2008;149(7):3549–58.

    CAS  PubMed  PubMed Central  Google Scholar 

  50. Kunnumakkara AB, Anand P, Aggarwal BB. Curcumin inhibits proliferation, invasion, angiogenesis and metastasis of different cancers through interaction with multiple cell signaling proteins. Cancer Lett. 2008;269(2):199–225.

    CAS  PubMed  Google Scholar 

  51. Lin J-K. Molecular targets of curcumin. In: The Molecular Targets and Therapeutic Uses of Curcumin in Health and Disease. Springer; 2007. p. 227–43. ISBN 978-0-387-46401-5.

  52. Gupta SC, Prasad S, Kim JH, Patchva S, Webb LJ, Priyadarsini IK, et al. Multitargeting by curcumin as revealed by molecular interaction studies. Nat Prod Rep. 2011;28(12):1937–55.

    CAS  PubMed  PubMed Central  Google Scholar 

  53. Suryanarayana P, Saraswat M, Mrudula T, Krishna TP, Krishnaswamy K, Reddy GB. Curcumin and turmeric delay streptozotocin-induced diabetic cataract in rats. Invest Ophthalmol Vis Sci. 2005;46(6):2092–9.

    PubMed  Google Scholar 

  54. Cohly HH, Taylor A, Angel MF, Salahudeen AK. Effect of Turmeric, Turmerin and Curcumin on H < sub > 2</sub > O < sub > 2</sub > −Induced Renal Epithelial (LLC-PK < sub > 1</sub>) Cell Injury. Free Radic Biol Med. 1998;24(1):49–54.

    CAS  PubMed  Google Scholar 

  55. Nishiyama T, Mae T, Kishida H, Tsukagawa M, Mimaki Y, Kuroda M, et al. Curcuminoids and sesquiterpenoids in turmeric (Curcuma longa L.) suppress an increase in blood glucose level in type 2 diabetic KK-Ay mice. J Agric Food Chem. 2005;53(4):959–63.

    CAS  PubMed  Google Scholar 

  56. Finkel T. Oxidant signals and oxidative stress. Curr Opin Cell Biol. 2003;15(2):247–54.

    CAS  PubMed  Google Scholar 

  57. Ghavami S, Shojaei S, Yeganeh B, Ande SR, Jangamreddy JR, Mehrpour M, et al. Autophagy and apoptosis dysfunction in neurodegenerative disorders. Prog Neurobiol. 2014;112:24–49.

    CAS  PubMed  Google Scholar 

  58. Ott M, Gogvadze V, Orrenius S, Zhivotovsky B. Mitochondria, oxidative stress and cell death. Apoptosis. 2007;12(5):913–22.

    CAS  PubMed  Google Scholar 

  59. Yeganeh B, Moghadam AR, Tran AT, Rahim MN, Ande SR, Hashemi M, et al. Asthma and influenza virus infection: focusing on cell death and stress pathways in influenza virus replication. Iran J Allergy Asthma Immunol. 2013;12(1):1–17.

    CAS  PubMed  Google Scholar 

  60. Naik SR, Panda VS. Antioxidant and hepatoprotective effects of Ginkgo biloba phytosomes in carbon tetrachloride‐induced liver injury in rodents. Liver Int. 2007;27(3):393–9.

    CAS  PubMed  Google Scholar 

  61. Vardi N, Parlakpinar H, Ozturk F, Ates B, Gul M, Cetin A, et al. Potent protective effect of apricot and β-carotene on methotrexate-induced intestinal oxidative damage in rats. Food Chem Toxicol. 2008;46(9):3015–22.

    CAS  PubMed  Google Scholar 

  62. Uraz S, Tahan V, Aygun C, Eren F, Unluguzel G, Yuksel M, et al. Role of ursodeoxycholic acid in prevention of methotrexate-induced liver toxicity. Dig Dis Sci. 2008;53(4):1071–7.

    CAS  PubMed  Google Scholar 

  63. Apel K, Hirt H. Reactive oxygen species: metabolism, oxidative stress, and signal transduction. Annu Rev Plant Biol. 2004;55:373–99.

    CAS  PubMed  Google Scholar 

  64. Jahovic N, Cevik H, Sehirli AO, Yegen BC, Sener G. Melatonin prevents methotrexate-induced hepatorenal oxidative injury in rats. J Pineal Res. 2003;34(4):282–7.

    CAS  PubMed  Google Scholar 

  65. Cetinkaya A, Bulbuloglu E, Kurutas EB, Kantarceken B. N-acetylcysteine ameliorates methotrexate-induced oxidative liver damage in rats. Med Sci Monit. 2006;12(8):BR274–278.

    CAS  PubMed  Google Scholar 

  66. Jahovic N, Sener G, Cevik H, Ersoy Y, Arbak S, Yegen BC. Amelioration of methotrexate-induced enteritis by melatonin in rats. Cell Biochem Funct. 2004;22(3):169–78.

    CAS  PubMed  Google Scholar 

  67. Babiak RM, Campello AP, Carnieri EG, Oliveira MB. Methotrexate: pentose cycle and oxidative stress. Cell Biochem Funct. 1998;16(4):283–93.

    CAS  PubMed  Google Scholar 

  68. Fiocchi C. Inflammatory bowel disease: new insights into mechanisms of inflammation and increasingly customized approaches to diagnosis and therapy. Curr Opin Gastroenterol. 2004;20(4):309–10.

    PubMed  Google Scholar 

  69. Los M, Khazaie K, Schulze-Osthoff K, Baeuerle PA, Schirrmacher V, Chlichlia K. Human T cell leukemia virus-I (HTLV-I) Tax-mediated apoptosis in activated T cells requires an enhanced intracellular prooxidant state. J Immunol (Baltimore, Md : 1950). 1998;161(6):3050–5.

    CAS  Google Scholar 

  70. Walker TM, Rhodes PC, Westmoreland C. The differential cytotoxicity of methotrexate in rat hepatocyte monolayer and spheroid cultures. Toxicol In Vitro. 2000;14(5):475–85.

    CAS  PubMed  Google Scholar 

  71. Rouse K, Nwokedi E, Woodliff JE, Epstein J, Klimberg VS. Glutamine enhances selectivity of chemotherapy through changes in glutathione metabolism. Ann Surg. 1995;221(4):420–6.

    CAS  PubMed  PubMed Central  Google Scholar 

  72. Erel O. A new automated colorimetric method for measuring total oxidant status. Clin Biochem. 2005;38(12):1103–11.

    CAS  PubMed  Google Scholar 

  73. Balasubramanian T, Senthilkumar G, Karthikeyan M, Chatterjee T. Protective Effect of Ethyl Acetate Fraction of Stereospermum Suaveolens Against Hepatic Oxidative Stress in STZ Diabetic Rats. J Tradit Complement Med. 2013;3(3):175.

    PubMed  PubMed Central  Google Scholar 

  74. Rezaei-Moghadam A, Mohajeri D, Rafiei B, Dizaji R, Azhdari A, Yeganehzad M, et al. Effect of turmeric and carrot seed extracts on serum liver biomarkers and hepatic lipid peroxidation, antioxidant enzymes and total antioxidant status in rats. BioImpacts: BI. 2012;2(3):151.

    PubMed  PubMed Central  Google Scholar 

  75. Somchit MN, Sulaiman M, Noratunlina R, Ahmad Z. Hepatoprotective effects of Curcuma longa rhizomes in paracetamol-induced liver damage in rats. In: Proceedings of the Regional Symposium on Environment and Natural Resources:. 2002;2002:698–702.

    Google Scholar 

  76. Fu Y, Zheng S, Lin J, Ryerse J, Chen A. Curcumin protects the rat liver from CCl4-caused injury and fibrogenesis by attenuating oxidative stress and suppressing inflammation. Mol Pharmacol. 2008;73(2):399–409.

    CAS  PubMed  Google Scholar 

  77. Ramaiah SK. A toxicologist guide to the diagnostic interpretation of hepatic biochemical parameters. Food Chem Toxicol. 2007;45(9):1551–7.

    CAS  PubMed  Google Scholar 

  78. Shakoori A, Butt U, Riffat R, Aziz F. Haematological and Biochemical Effects of Danitol Admin stered for two Months in the Blood and Liver of Rabbits. ZEITSCHRIFT FUR ANGEWANDTE ZOOLOGIE. 1994;80:165–80.

    CAS  Google Scholar 

  79. Navarro MC, Montilla MP, Martín A, Jiménez J, Utrilla MP. Free radical scavenger and antihepatotoxic activity of Rosmarinus tomentosus. Planta Med. 1993;59(04):312–4.

    Google Scholar 

  80. Te Helen S, Schiano TD, Kuan SF, Hanauer SB, Conjeevaram HS, Baker AL. Hepatic effects of long-term methotrexate use in the treatment of inflammatory bowel disease. Am J Gastroenterol. 2000;95(11):3150–6.

    Google Scholar 

  81. Liju VB, Jeena K, Kuttan R. Acute and subchronic toxicity as well as mutagenic evaluation of essential oil from turmeric (Curcuma longa L). Food Chem Toxicol. 2013;53:52–61.

    CAS  PubMed  Google Scholar 

  82. Kim SW, Ha KC, Choi EK, Jung SY, Kim MG, Kwon DY, et al. The effectiveness of fermented turmeric powder in subjects with elevated alanine transaminase levels: a randomised controlled study. BMC Complement Altern Med. 2013;13:58.

    PubMed  PubMed Central  Google Scholar 

  83. Cieslar-Pobuda A, Back M, Magnusson K, Jain MV, Rafat M, Ghavami S, et al. Cell type related differences in staining with pentameric thiophene derivatives. Cytometry A. 2014;85(7):628–35.

    PubMed  Google Scholar 

  84. Cieslar-Pobuda A, Wiechec E. Research on liver regeneration as an answer to the shortage of donors for liver transplantation. Hepatol Res. 2014;44(9):944–6.

    PubMed  Google Scholar 

  85. Rollano Penaloza OM, Lewandowska M, Stetefeld J, Ossysek K, Madej M, Bereta J, et al. Apoptins: selective anticancer agents. Trends Mol Med. 2014;20(9):519–28.

    CAS  PubMed  Google Scholar 

  86. Wasik AM, Grabarek J, Pantovic A, Cieslar-Pobuda A, Asgari HR, Bundgaard-Nielsen C, et al. Reprogramming and carcinogenesis--parallels and distinctions. Int Rev Cell Mol Biol. 2014;308:167–203.

    CAS  PubMed  Google Scholar 

  87. Wiechec E. Implications of genomic instability in the diagnosis and treatment of breast cancer. Expert Rev Mol Diagn. 2011;11(4):445–53.

    CAS  PubMed  Google Scholar 

  88. Yeganeh B, Hashemi M, de Serres FJ, Los MJ, Ghavami S. Different faces of hepatocellular carcinoma as a health threat in 21st century. Hepat Mon. 2013;13(2), e9308.

    PubMed  PubMed Central  Google Scholar 

  89. Yeganeh B, Wiechec E, Ande SR, Sharma P, Moghadam AR, Post M, et al. Targeting the mevalonate cascade as a new therapeutic approach in heart disease, cancer and pulmonary disease. Pharmacol Ther. 2014;143(1):87–110.

    CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgment

SG was supported by University of Manitoba start-up fund. MJ Los kindly acknowledges the core/startup support from Linkoping University, from IGEN, from Cancerfonden (2013/391), and from VR-NanoVision (K2012-99X −22325-01-5).

Author information

Authors and Affiliations

Authors

Corresponding authors

Correspondence to Marek J. Łos or Saeid Ghavami.

Additional information

Competing interests

The authors declare that they have no competing interests.

Authors’ contributions

Experimental design: MM, HM, and SG. Experiments performed by: ARM, ST, AN-A-A, MY, FB, and DM. Data analysis: ARM, MM, HM, MJL, and SG. Manuscript preparation: ARM, ST, MM, HM, MJL, and SG. All authors read and approved the final manuscript.

Saeid Ghavami and Marek Łos are senior authorship.

Rights and permissions

Open Access  This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made.

The images or other third party material in this article are included in the article’s Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article’s Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder.

To view a copy of this licence, visit https://creativecommons.org/licenses/by/4.0/.

The Creative Commons Public Domain Dedication waiver (https://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Moghadam, A.R., Tutunchi, S., Namvaran-Abbas-Abad, A. et al. Pre-administration of turmeric prevents methotrexate-induced liver toxicity and oxidative stress. BMC Complement Altern Med 15, 246 (2015). https://doi.org/10.1186/s12906-015-0773-6

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s12906-015-0773-6

Keywords