RESEARCH ARTICLE Protective Effects of Essential Oils as Natural Antioxidants against Hepatotoxicity Induced by Cyclophosphamide in Mice

Salah A. Sheweita1*, Lobna S. El-Hosseiny2, Munther A. Nashashibi1

1 Department of Biotechnology, Institute of Graduate Studies and Research, Alexandria University, Alexandria, Egypt, 2 Department of Environmental Studies, Institute of Graduate Studies and Research, Alexandria University, Alexandria, Egypt

a11111 * [email protected]

Abstract

Clinical application of cyclophosphamide (CP) as an anticancer drug is often limited due to its toxicity. CP is metabolized mainly in the liver by cytochrome P450 system into OPEN ACCESS which is the proximate toxic metabolite. Many different natural antioxidants were found to Citation: Sheweita SA, El-Hosseiny LS, Nashashibi MA (2016) Protective Effects of Essential Oils as alleviate the toxic effects of various toxic agents via different mechanisms. Therefore, the Natural Antioxidants against Hepatotoxicity present study aimed at investigating the role of essential oils extracted from fennel, cumin Induced by Cyclophosphamide in Mice. PLoS ONE and clove as natural antioxidants in the alleviation of hepatotoxicity induced by CP through 11(11): e0165667. doi:10.1371/journal. assessment of hepatotoxicity biomarkers (AST, ALT, ALP), histopathology of liver tissues pone.0165667 as well as other biochemical parameters involved in the metabolism of CP. The data of the Editor: Hans-Joachim Lehmler, University of Iowa, present study showed that treatment of male mice with cyclophosphamide (2.5 mg/Kg BW) UNITED STATES as repeated dose for 28 consecutive days was found to induce hepatotoxicity through the Received: July 2, 2016 elevation in the activities of AST, ALT, and ALP. Combined administration of any of these Accepted: October 14, 2016 oils with CP to mice partially normalized the altered hepatic biochemical markers caused by Published: November 1, 2016 CP, whereas administration of fennel, clove or cumin essential oils alone couldn't change

Copyright: © 2016 Sheweita et al. This is an open liver function indices. Moreover, CP caused histological changes in livers of mice including access article distributed under the terms of the swelling and dilation in sinusoidal space, inflammation in portal tract and hepatocytes, as Creative Commons Attribution License, which well as, hyperplasia in Kuppfer cells. However, co-administration of any of the essential oils permits unrestricted use, distribution, and with CP alleviated to some extent the changes caused by CP but not as the normal liver. reproduction in any medium, provided the original author and source are credited. CP was also found to induce free radical levels (measured as thiobarbituric acid reactive substances) and inhibited the activities of superoxide dismutase, glutathione reductase, Data Availability Statement: All relevant data are within the paper and its Supporting Information and catalase as well as activities and protein expressions of both glutathione S-transferase files. (GSTπ) and glutathione peroxidase. Essential oils restored changes in activities of antioxi-

Funding: The author(s) received no specific dant enzymes (SOD, CAT, GR, GST, and GPx) caused by CP to their normal levels com- funding for this work. pared to control group. In addition, treatment of mice with CP was found to induce the

Competing Interests: The authors have declared protein expression of CYP 3A4, 2B1/2, 2C6, 2C23. Moreover, the present study showed that no competing interests exist. that essential oils reduced the expression of CYPs 2E1, 3A4 but could not restore the

Abbreviations: CP, Cyclophosphamide; CYP, expression of CYP 2C6 and 2C23 compared to CP-treated mice. Interestingly, pretreat- Cytochrome P450; GST, glutathione S-transferase; ment of mice with essential oil of clove was found to restore activities of DMN-dI, AHH, and GPx, glutathione peroxidase; GR, glutathione ECOD which were induced by CP to their normal control levels. It is concluded that EOs reductase; CAT, catalase; SOD, superoxide

PLOS ONE | DOI:10.1371/journal.pone.0165667 November 1, 2016 1 / 17 Protective Effects of Essential Oils against Hepatotoxicity Induced by Cyclophosphamide

dismutase; TBARS, thiobarbituric acid reactive showed a marked hepatoprotective effect against hepatotoxicity induced by CP. In addition, substances; ALT, alanine aminotransferase; AST, co-administration of CP with any of these oils might be used as a new strategy for cancer aspartate aminotransferase; ALP, Alkaline phosphatase; GSH, glutathione; GSSG, oxidized treatment to alleviate the hepatotoxicity induced by CP. gluathione; MDA, malondialdehyde; GC-MS, Gas chromatography-Mass spectroscopy (GC-MS); EO, Essential oil; DMN-dI, dimethylnitrosamine N- demethylase I; ECOD, 7-ethoxycoumarin-O- deethylase; BSA, bovine serum albumin. Introduction For more than 50 years cyclophosphamide (CP) has been widely used to treat various forms of cancers, including lymphoma, breast cancer and leukemia [1]. However, clinical application of CP is often restricted due to its deleterious side effects [2], especially hepatotoxicity [3]. The toxic effects of CP are mainly due to the generation of two major metabolites namely phos- phoramide mustard which is the antineoplastic moiety and acrolein metabolite which is the most toxic agent. These metabolites are generated by cytochrome P450 isozymes including CYP 3A4, 2B6, 2C9, and 2C19 [4]. Acrolein is a highly reactive α, β- unsaturated aldehyde, and was identified as the initiator of lipid peroxidation. This reactivity is the main reason of the cytotoxicity in all cells exposed to acrolein [5] which limits using CP in clinical practice. CP- induced oxidative stress through the generation of free radicals leading to biochemical and physiological disturbances in animal models [6]. Protection of cells from the lethal effects of toxic compounds was observed due to the pres- ence of abundant amounts of glutathione which is an important determinant of cellular sensi- tivity to various drugs and other toxic compounds [7,8]. Depletion of GSH levels in the cells could promote tumor development in different animal species [9]. Supporting this suggestion, depletion of GSH level and inhibition of GST activity were found to reduce the covalent bind-

ing of the ultimate metabolites of both aflatoxin B1 and benzo[a]pyrene, with DNA [10], and decreasing of hepatocarcinogenesis caused by these compounds was correlated with low the level of DNA adducts [10]. Several studies suggested that dietary antioxidants supplementation can reduce the develop- ment of adverse effects associated with anticancer drugs including CP [11,12]. It has been found that some plants contain a wide variety of antioxidant phytochemicals or bioactive mole- cules which can neutralize free radicals and decreased the oxidative stress that plays a critical role in the incidence of adverse toxic effects associated with chemotherapies [13]. Fennel (Foe- niculum vulgare Miller) is a medicinal and aromatic plant with a diverse pharmacological spec- trum and of considerable importance in food industries. Previous studies have shown that various extracts of fennel possess a range of pharmacological actions ranging from antispas- modic and anxiolytic to cytoprotective and anti-inflammatory effects due to the presence of terpenes and terpenoids [14]. Cumin (Cuminum Cyminum L.) is belonging to the family of Apiaceous. Cumin seeds are used in folklore therapy [15,16]. Clove (Syzygiumaromaticum L.) is one of the most valuable spices that have been used for centuries as a food preservative and for many medicinal purposes including antimicrobial, anti-inflammatory, antioxidant and anticancer activities [17]. Phenolic compounds (, eugenol acetate, and gallic acid) which possess pharmaceutical, cosmetic, food and agricultural applications are the main con- stituent of clove oil. Recently, the protective effects of Foeniculum vulgare (fennel) essential oil (FEO) and Syzy- gium cumini extract against genotoxicity and oxidative stress induced by CP was investigated [18,19]. It has been shown that CP produced a significant increase in the average percentage of aberrant metaphases and chromosomal aberrations, excluding gap and micronuclei formation in polychromatic erythrocytes,produced cytotoxicity in mouse bone marrow cells [18,19]. CP

PLOS ONE | DOI:10.1371/journal.pone.0165667 November 1, 2016 2 / 17 Protective Effects of Essential Oils against Hepatotoxicity Induced by Cyclophosphamide

also markedly inhibited the activities of SOD, CAT, and GSH and increased MDA content. Pretreatments with FEO and Syzygium cumini extract significantly inhibited the frequencies of aberrant metaphases, chromosomal abreactions, micronuclei formation, and cytotoxicity in mouse bone marrow cells induced by CP and antagonized the reduction of CP-induced SOD, CAT, and GSH activities and inhibited increased MDA content in the liver of mice [18,19]. In addition, FEO and Syzygium cumini extract inhibited genotoxicity and oxidative stress induced by CP (Tripathi et al., 2013a&b). In another study, the lyophilized ethanolic leaf of Eugenia dysenterica exhibited protection against CP induced genotoxic and cytotoxic actions at all doses tested [20]. CP-induced hepatotoxicity is critical issues during the clinical application of CP. Oxidative stress, caused by the active metabolites of CP, exhibited deleterious effects on hepatocellular membranes and other tissues. Therefore, finding a strategy to reduce the oxidative stress might be a key, at least in part, to alleviate the CP-induced hepatotoxicity. To the best of our knowl- edge, no previous study investigating the protective effects of essential oils of cumin and/or clove either alone or in combination with CP has been conducted. However, the influence of Foeniculum vulgare (fennel) essential oil either alone or in combination with CP was tested in the livers of mice on the activities of antioxidant enzymes but not on the protein expression of CYP450 [19]. Therefore, the present study aimed at investigating the extracted essential oils from fennel, clove and cumin and their roles in alleviating the toxic effects exerted by CP administration to mice through determination of activities and protein expressions of cyto- chrome P450 isozymes. In addition, the present study showed the influence of essential oils either alone or in combination with CP on levels of free radicals and activities and protein expressions of antioxidant enzymes including glutathione peroxidase and glutathione S-trans- ferase after treatment of mice.

Materials and Methods Chemicals Cyclophosphamide was obtained from Baxter Oncology, Germany. Nicotinamide adenine dinucleotide phosphate (NADP) sodium salt, nicotinamide adenine dinucleotide phosphate reduced (NADPH) sodium salt, hydroperoxide, 1-chloro-2,4-dinitrobenzene (CDNB) and all other chemicals were purchased from Sigma Chemical Co., UK. Anti-cytochrome P450 3A4, 2B1/2, 2C6, 2C23, 2E1 and NADPH-cytochrome c reductase were purchased from ABCAM, UK. Cytochrome P450 Western blotting detection kits were purchased from Amer- sham, UK. Diagnostic kits for aspartate aminotransferase, alanine amino transferase, and alka- line phosphatase were obtained Spectrum Diagnostics, Hannover, Germany.

Dimethylsulfoxide (DMSO) pharmaceutical grade (C2H6OS, purity 99%) was obtained from Gaylord Chemical Co., LA, USA. All other agents and solvents were of analytical grade.

Essential oil extraction and characterization by GC-MS Dried seeds of fennel, cumin and flower buds of clove authenticated by Prof. Salama El Darier, Professor of Plant Ecology, Botany and Microbiology Department, Faculty of Science, Alexan- dria University were used for the extraction of essential oils. 100 gm of the respective dried seeds or flower buds were subjected to hydrodistillation for 4 hr using Clevenger type appara- tus. The distillate oil was collected, dried over anhydrous sodium sulfate and stored in dark sealed vials at 4°C prior to characterization of their chemical composition. Compositional analysis of the extracted essential oils was assessed using Thermo scientific GC-MS version 5 system. Qualitative identification of the oil constituents was carried out on the basis of their retention indices and matching their mass spectral fragmentation patterns

PLOS ONE | DOI:10.1371/journal.pone.0165667 November 1, 2016 3 / 17 Protective Effects of Essential Oils against Hepatotoxicity Induced by Cyclophosphamide

with NIST library data base [21]. Quantitative analysis of the oil components expressed as rela- tive percentage of peak area, was estimated using peak area normalization measurements.

Animal treatments Sixty-four Swiss male albino mice, weighing 25±2 g were obtained from the animal house of the Faculty of Medicine, Alexandria University. This study was carried out in strict accordance with the recommendations in the Guide for the Care and Use of Laboratory Animals of the College of Medicine, Alexandria University. The protocol was approved by the committee of postgraduate studies & research on the Ethics of Animal Experiments of the University of Alex- andria. All surgery was performed under diethyl ether anesthesia, and all efforts were made to minimize suffering. Mice were housed in a stainless steel wire bottom cages placed in a well- ventilated animal house, maintained for one week for acclimatization period on food and water ad libitum, and subjected to the natural photoperiod of 12 hours light:dark cycle. The animals were assigned into 8 groups comprising 8 mice/cage in each group and fed the same diet throughout the experimental period (28 days). Group I served as the control and received 0.1 ml of DMSO orally. Group II (CP) was orally administered with the aqueous solution of CP at a dose of 2.5 mg/Kg BW. Groups III, IV and V were orally treated with the essential oils of fen-

nel, cumin and clove dissolved in DMSO at doses of 1/50 their LD50 corresponding to 0.12 ml/ Kg BW, 0.10 ml/Kg BW and 0.106 ml/Kg BW respectively. Groups VI, VII, and VII were administered orally by a combination of CP with fennel, cumin or clove essential oils at the same doses scheduled in groups II, III, IV and V. At the end of the experimental period, mice were starved overnight and then sacrificed. Blood and liver samples were collected. Serum was obtained by centrifugation of coagulated blood at 3000 xg for 5 min. Livers were washed with cold phosphate buffer saline and the S9 fraction was prepared by homogenizing the livers in 33% W/V of 0.1 M phosphate buffered solution (pH 7.4) and centrifuging at 10000 xg for 20 min. Liver microsomal fractions were prepared by centrifugation of the S9 fraction supernatant at 105,000 xg for 1 hour at 4°C, then the microsomal pellets were suspended in 0.1 M phosphate buffer (pH 7.4) and stored in ali- quots at -80°C [22].

Biochemical analyses Activities of biomarker enzymes for hepatocellular damage, including alanine aminotransfer- ase (ALT), aspartate aminotransferase (AST) and alkaline phosphatase (ALP) were determined in serum of different groups according to the well-established spectrophotometric methods according to the manuals supplied with the commercial kits obtained from Spectrum Diagnos- tics, Hannover, Germany. A substrate concentration of 4 mM dimethylnitrosamine, which represents the saturation level for dimethylnitrosamine N-demethylase I (DMN-dI) activity, was used [23,24]. Also, the microsomal 7-ethoxycoumarin-O-deethylase (ECOD) activity was measured as described by Greenle and Poland, 1978 [25] and the intensity of 7-hydroxy-coumarin was measured fluoro- metrically at excitation and emission wavelengths of 338 and 458 nm respectively. NADPH- cytochrome c reductase activity in hepatic microsomal fraction was assayed by measuring the reduction of oxidized cytochrome cat 550nm using an extinction coefficient of 21 mM-1 Cm-1 [26]. The activity of GPx was determined in the S9 fraction of cell homogenates according to the method of Chiu et al., 1976 [27]. The principle of determination is based on the decrease in absorbance of NADPH at 340 nm. The specific activity was expressed in terms of the amount of enzyme that oxidizes 1 μmol of NADPH per min. Glutathione reductase activity in hepatic

PLOS ONE | DOI:10.1371/journal.pone.0165667 November 1, 2016 4 / 17 Protective Effects of Essential Oils against Hepatotoxicity Induced by Cyclophosphamide

S9 fraction was monitored by the oxidation of NADPH in the presence of GSSG according to the method of David and Richard, 1983 [28]. Glutathione-S-transferase activity was assayed according to the method of Habig et al., 1974 [29], which is based on the measurement of 1-chloro-2,4-dinitrobenzene(CDNB) conjugate formed at 340 nm. A molar extinction coeffi- cient of 9.6 mM-1 Cm-1 was used for the calculation of GST activity. One μmole of CDNB con- jugate/mg protein/min is expressed as a unit/mg protein. Catalase activity was determined in the hepatic S9 fraction according to the method of Beers

and Sizer, 1952 [30], which is based on decreasing in absorbance of H2O2 solution decomposed by the catalase enzyme (assay kit was obtained from Randox Laboratory, UK). The quantity of

H2O2 was measured at 240 nm and catalase activity was expressed as U/mg protein. Superoxide dismutase (SOD) was assayed according to the method of Misra and Fridovich, 1972 [31]. SOD quantitation was dependent on the generation of superoxide radicals by xanthine and xanthine oxidase, which react with nitro blue tetrazolium to form the formazin dye. SOD activ- ity was measured at 560 nm and expressed as U/mg protein. The protein content was measured in both S9 fraction and microsomal pellets according to the method of Lowry et al., 1950 [32] using bovine serum albumin as standard. Quantitative measurement of lipid peroxidation was performed in cell homogenates accord- ing to the methods of Tappel and Zalkin, 1959 [33], which is based on the formation of thio- barbituric acid reactive substances (TBARS) and expressed as the extent of malondialdehyde (MDA) production. TBARS was expressed as μmole MDA per gram tissue. Glutathione level was measured in the S9 fractions according to the method of Mitchell et al., 1973 [34], after protein precipitation with sulfosalicylic acid. After centrifugation at 3000 xg for 20 min, one ml of clear supernatant was added to bis-(3-carboxy-4-nitrophenyl)-disulphide for color develop- ment. The developed color solution was spectrophotometrically at 412 nm. Glutathione levels were expressed as μmole GSH per gram tissue.

Western Immunoblotting From each pooled group (10 mice each), forty microgram of the S9 and the microsomal frac- tions were mixed with the sample application buffer (SAB), then boiled for 3 minutes and loaded on a 10% SDS-polyacrylamide gel. After electrophoresis, proteins were transferred to nitrocellulose membranes using a semidry trans blotter. Each membrane was blocked in 0.5% BSA for 2 hours with gentle shaking at room temperature prior to antibody incubations. Pri- mary antibodies of GSTπ, glutathione peroxidase, NADPH-cytochrome c reductase, CYP2E1, CYP3A4, CYP2B1/2, CYP2C23 and CYP2C6 in 0.5% bovine serum albumin were used in the present study. The secondary antibody (anti-mouse conjugated-peroxidase) was used after incubation with the membrane for a minimum of 30 minutes at room temperature. Signals of immune-reactive proteins were detected using X-ray films according to the manufacturer's instructions [35]. The band intensity was measured using quantity one software program.

Histological examination of the liver Formalin fixed paraffin embedded hepatic tissues were sectioned and stained routinely with hematoxylin and eosin (H&E). Hepatic damage in H&E-stained sections was examined from a digital image captured at 200x magnification [36].

Statistical Analysis Data presented in the study were shown as mean ± standard error. Mean of different treatment groups were tested for significance using one-way analysis of variance (ANOVA) and were

PLOS ONE | DOI:10.1371/journal.pone.0165667 November 1, 2016 5 / 17 Protective Effects of Essential Oils against Hepatotoxicity Induced by Cyclophosphamide

compared using posthoc Duncan's Multiple Range Test (DMRT). Difference were considered significant at P<0.05. SPSS 17 statistical software package was used in statistical evaluation.

Results Compositional analysis of the extracted clove, cumin and fennel essential oils Compositional analysis of the extracted essential oils by GC-MS demonstrated that eugenol (82.84%), acetyl eugenol (6.29%), β-caryophyllene (8.13%) and α-caryophyllene (1.05%) were the major chemical constituents of the hydrodistilled clove essential oil (Fig 1A, Table 1). Fen- nel essential oil was constituted mainly by estragole, limonene, fenchone, α-pinene and trans- anethole comprising 80.75%, 10.47%, 4.64%, 1.10% and 1.02% respectively (Fig 1B, Table 2). The terpene hydrocarbons; γ-terpinene (23.91%) and 3-carene (7.66%), as well as, the terpe- noids; cumin aldehyde (19.43%), 2-caren-10-al (15.22%) and cuminyl alcohol (9.08%) were the main components constituting the extracted cumin essential oil (Fig 1C and Table 3).

Cyclophosphamide and/or essential oils induced alterations in hepatic biochemical indices and hepatic oxidative response The oral administration of CP to mice significantly increased hepatocellular injury biomarkers (AST, ALT, and ALP) in serum as demonstrated in Table 4. Meanwhile, administration of fen- nel, clove or cumin essential oils alone insignificantly changed liver function indices, whereas their combined administration with CP partially normalized the altered hepatic biochemical markers. As presented in Table 4, hepatic oxidative response in different treatment groups revealed that CP-treated mice exhibited a significant elevation in TBARS level, significant depletion in GSH level in addition to a significant reduction in the activity of antioxidant enzymes (GST, GR, GPx, SOD and CAT). Administration of essential oils alone resulted in a favorable effect on hepatic antioxidant status, where they significantly increased the activity of endogenous antioxidant enzymes (GST, GR, GPx, SOD and CAT) meanwhile neither inducing lipid peroxidation nor depleting GSH levels. The Co-administration of essential oils with CP resulted in significant reduction of the escalated lipid peroxidation and compensated the deple- tion in endogenous enzymatic (GST, GR, GPx, SOD and CAT) and non-enzymatic (GSH) antioxidants. Supporting the present perturbations in antioxidant defense parameters, protein expression of both GSTπ and GPx was inhibited after treatment of mice with cyclophospha- mide. Furthermore, treatment of mice with fennel, cumin or clove essential oils increased the protein expression of both enzymes compared to control group (Fig 2).

Cyclophosphamide and/or essential oils induced alteration in expression/activity of CYPs Treatment of mice with CP-induced the protein expressions of cytochrome P450 isozymes (CYP 3A4, 2B1/2, 2C6, 2C23) involved in CP metabolism (Fig 3). The protein expression of CYP2B1/2 was inhibited when mice were treated individually by clove EO and was induced upon treatment of mice with fennel or cumin essential oils. Synchronically, the corresponding activity of ECOD correlated to the inhibition or induction of CYP2B1/2 inferred by the three tested essential oils. Fennel and clove essential oils reduced the expression of CYP3A4, whereas cumin induced its expression compared to the control group. The expression of CYP2C6 and CYP2C23 was induced indifferently in all treatment groups comparative to the control. Cyclo- phosphamide administration also induced the protein expression of CYP2E1 and increased its corresponding DMN-dI activity (Fig 3, Table 4). Fennel, cumin or clove essential oils reduced

PLOS ONE | DOI:10.1371/journal.pone.0165667 November 1, 2016 6 / 17 Protective Effects of Essential Oils against Hepatotoxicity Induced by Cyclophosphamide

PLOS ONE | DOI:10.1371/journal.pone.0165667 November 1, 2016 7 / 17 Protective Effects of Essential Oils against Hepatotoxicity Induced by Cyclophosphamide

Fig 1. (A) GC-MS chromatogram and major components of essential oil of Clove, (B) Fennel, and (C) Cumin respectively. doi:10.1371/journal.pone.0165667.g001

the expression of CYP2E1 and significantly reduced the activity of DMN-dI, whereas their combined administration with CP normalized the expression of CYP2E1 and the correspond- ing DMN-dI activity (Fig 3, Table 4). Both the protein expression and activity of NADPH-cyto- chrome c reductase was induced in CP or essential oils' treated groups. However, the combined treatment of mice with CP and either of the investigated essential oils did not significantly change the activity or expression of NADPH-cytochrome c reductase comparative to groups treated with either CP or the tested EOs individually (Fig 3, Table 4).

Cyclophosphamide and/or essential oils induced pathomorphology Histological examination of liver sections of different treatment groups revealed that mice treated with CP demonstrated distortion in hepatic architecture manifested as swelling and dilation in sinusoidal space, hemorrhage in central vein, mild inflammatory changes in portal tract and hepatocytes in addition to hyperplasia in Kuppfer cells (Fig 4B). Conversely, liver sec- tion of fennel, cumin or clove essential oil-treated mice exhibited normal architecture with nor- mal hepatocytes and sinusoidal space (Fig 4C, 4D and 4E). In liver section of mice treated concomitantly with essential oils and CP, the hydropic, necrotic and inflammatory changes induced by CP regressed to some extent (Fig 4F, 4G and 4H).

Discussion Cyclophosphamide (CP), a chemotherapeutic agent, is restricted due to its side effects, espe- cially hepatotoxicity [37]. CYP2E1 is the major enzyme that catalyzes the biotransformation of numerous xenobiotics and anticancer drugs [8,38]. There is an association between CP expo- sure and induction of various types of neoplasms in experimental animals [39]. The present study may provide a new possible mechanism of cancer induction by CP through enhancement of protein expression of CYP2E1 since CP-induced protein expression of CYP2E1 in the liver of male mice. Supporting our suggestion, several investigations have shown that repeated dose of CP induces its own metabolism through the generation of acrolein (the toxic metabolite of CP) in excessive amounts [40,41]. Interestingly, administration of fennel, cumin, and/or clove essential oils in combination with CP to mice inhibited the induced protein expression of CYP2E1, 3A4, 2B1/2 and their associated enzymatic activities caused by CP which may protect the liver against toxicity and carcinogenicity induced by CP. Changes in hepatic cytochrome P450 isozymes are mainly due to the presence of active compounds which have been isolated and identified from herbs and medicinal plants. The protective effects exerted by these oils could be related to their chemical components. For example, ß-myrcene, a constituent of fennel essential oil, has been reported to up-regulate the protein expression of CYP2B1/2 [42]. These compounds act as substrates, inhibitors and/or inducers of various CYP isozymes. Eugenol, the major constituent of clove essential oil, was reported to inhibit CYP2B1/2 and CYP1A1 that

Table 1. Major components of clove essential oil. Component Eugenol β-caryophyllene α-caryophyllene Acetyl eugenol Retention time 21.13 22.73 23.74 26.14 Relative % 82.84 8.13 1.05 6.29 Molecular formula (chemical class) C10H12O2 (terpenoid) C15H24 (terpene) C15H24 (terpene) C12H14O3 (terpenoid) doi:10.1371/journal.pone.0165667.t001

PLOS ONE | DOI:10.1371/journal.pone.0165667 November 1, 2016 8 / 17 Protective Effects of Essential Oils against Hepatotoxicity Induced by Cyclophosphamide

Table 2. Major components of fennel essential oil. Component α-pinene Limonene Fenchone Estragole trans-anethole Retention time 5.99 9.04 11.07 15.63 18.10 Relative % 1.10 10.47 4.64 80.75 1.02 Molecular formula (chemical class) C10H16 (terpene) C10H16 (terpene) C10H160 (terpenoid) C10H12O (terpenoid) C10H12O (terpenoid) doi:10.1371/journal.pone.0165667.t002

catalyze the biotransformation of 7,12-dimethylbenz[α]anthracene to more carcinogenic metabolites [43]. On the other hand, it has been demonstrated that cumin significantly increased the activity of CYP P450 and NADPH-cytochrome c reductase activity [44]. The mechanism of up-regulation of cytochrome P450 isozymes could be due to the presence of lim- onene and myrcene as major constituents of essential oils, which were found to induce delta- aminolaevulinic acid synthetase activity, the key enzyme involved in the synthesis of haem moiety of cytochrome P450 isozymes [45,46]. Moreover, the protective effects of essential oils are dependent on the presence of double bonds in the chemical structures of terpene hydrocar- bons [47], limonene, terpinolene, and β-caryophyllene [48–50]. Recently, it has been found that blocking of these double bonds was found to decrease the antioxidant activities of monoterpenes and other components [51]. In clinical applications, oxidative stress induced by CP play a significant role in the inci- dence of hepatotoxicity [52,53]. The present study showed that increment of oxidative stress in liver of mice after administration of CP could be due to inhibition of antioxidant enzymes activities (GST, GPx, GST, SOD, CAT) and reduction of GSH levels. In agreement with the present study, oxidative stress was increased after CP exposure in both humans and mice [52,53]. The mechanism of induction of oxidative stress could be due to metabolic conversion of CP to several types of toxic metabolites [54]. For example, acrolein, a highly reactive metabo- lite of CP with short biological half-life, could readily react with glutathione (GSH), one of thiol-containing proteins, which serves several vital functions, including detoxifying electro- philes and relieving oxidative stress, with the presence of glutathione S-transferase (GST) [55]. Acrolein, the highly reactive α, β–unsaturated aldehyde, was found to react with cellular nucle- ophiles such as the thiol groups of cysteine residues in proteins and nitrogen atoms in lysine and histidine groups, leading to the loss of protein function, which could induce the oxidative stress and finally give rise to the disastrous effects on hepatocyte including inhibition of antiox- idant enzymes [55,56]. On the other hand, pretreatment of mice with essential oil of Cumin, Fennel and/or Clove before administration of CP restored the activities of antioxidant enzymes, free radicals, and GSH levels to their normal levels compared to control mice. Moreover, treatment of mice with essential oils of Cumin, Fennel, and/or Clove induced activities of GST, CAT, and GPx com- pared to the control mice. Supporting the data of enzymatic activities, Western immunoblot data showed that the protein expressions of both GPx and GSTπ were markedly reduced after treatment of mice with CP, and potentially induced after treatment of mice with either essential oils alone or in combination with CP. From the present study, induction of free radical levels

Table 3. Major components of cumin essential oil. Component 3-carene γ-terpinene Cuminaldehyde 2-caren-10-al Cuminyl alcohol Retention time 7.30 10.20 16.71 18.21 18.43 Relative (%) 7.66 23.91 19.43 15.22 9.08 Molecular formula (chemical class) C10H16 (terpene) C10H16 (terpene) C10H12O (terpenoid) C10H14O (terpenoid) C10H14O (terpenoid) doi:10.1371/journal.pone.0165667.t003

PLOS ONE | DOI:10.1371/journal.pone.0165667 November 1, 2016 9 / 17 PLOS ONE | DOI:10.1 371/journ

Table 4. Biochemical parameters in serum and liver lysate of different treatment groups. al.pone.0165 Treatments Parameter Control CP Fennel EO Cumin EO Clove EO CP and Fennel EO CP and Cumin EO CP and Clove EO Serum AST (U/L) 70.1±0.32c 121±2.52a 71.4±0.37c 70.9±0.66c 70.6±0.82c 91.2±2.45b 86.7±2.78b 89.2±0.46b Serum ALT(U/L) 76.5±0.31d 123±1.63a 75.7±0.51d 74.9±0.34d 76.5±0.32d 93.8±0.57bc 90.3±3.1c 96.8±1.36b d a d d d bc c b

667 Serum ALP (U/L) 88.3±5.29 149±1.72 89.1±3.64 84.5±3.57 85.2±3.94 111±4.54 103±3.43 119±4.05 TBARS level (μmol/g tissue) 1.38 ±0.18c 2.59±0.06a 1.23±0.02d 1.11±0.02e 1.12±0.02e 1.44±0.02bc 1.34±0.03c 1.49±0.06b b c ab a ab b ab b

November GSH content (μmol/g tissue) 2.43±0.04 1.87±0.03 2.65±0.06 2.79±0.26 2.53±0.02 2.41±0.03 2.52±0.03 2.45±0.04 GPx activity (U/mg protein) 3.29±0.07cd 2.07±0.09e 4.39±0.07b 4.51±0.08b 4.97±0.18a 3.18±0.10cd 3.40±0.27c 2.98±0.06d GR activity (nmol of NADPH oxidized/ min/mg protein) 0.21±0.01c 0.12±0.01d 0.23±0.01c 0.31±0.01a 0.27±0.02b 0.21±0.01c 0.22±0.01c 0.21±0.01c GST activity(U/ mg protein) 3.55±0.04e 1.52±0.05f 5.26±0.05b 5.86±0.06a 6.01±0.11a 4.42±0.05d 4.83±0.07c 4.47±0.04d

1, SOD (U/mg protein) 11.4±1.55bc 5.38±1.38d 14.6±0.96abc 17.7±1.83a 15.6±1.46ab 10.1±1.25c 11.9±1.84bc 11.2±2.02bc 2016 CAT (U/mg protein) 0.22±0.01b 0.06±0.01c 0.31±0.01a 0.32±0.01a 0.29±0.02a 0.22±0.01b 0.24±0.02b 0.21±0.01b Protective DMN-dI (nmol HCHO/mg protein/h) 94.2±3.35b 148±4.85a 78.7±3.41cd 76.4±4.79d 75.5±3.11d 89.3±2.35bc 87.3±1.95bc 89.7±3.70b ECOD (nmol hydroxycoumarin/ mg protein/min) 0.29±0.03c 0.44±0.02a 0.41±0.01ab 0.38±0.01b 0.21±0.02d 0.38±0.01b 0.42±0.02ab 0.28±0.01c NADPH-Cytochrome c reductase (nmol cyt c reduced/mg protein/min) 9.76±2.52b 23.1±2.58a 22.8±2.39a 20.8±2.42a 21.6±2.12a 19.9±3.14a 20.2±2.48a 22.1±1.95a Effects Values are mean ± S.E., n = 8 mice/group. abcdef Mean values within a row not sharing common superscript letters were signi®cantly different, P<0.05. of

doi:10.1371/journal.pone.0165667.t004 Essentia l Oils against Hepatotoxic ity Induced by Cyclopho sphamide 10 / 17 Protective Effects of Essential Oils against Hepatotoxicity Induced by Cyclophosphamide

Fig 2. Western immune blot analysis for the protein expression of GSTπ and GPx in the pooled proteins of matched control, CP, fennel EO, cumin EO, clove EO, CP & fennel EO, CP & cumin EO and CP & clove EO treated groups. doi:10.1371/journal.pone.0165667.g002

and inhibition of antioxidant enzyme activities might be new reasons for cancer induction by CP. The inhibition of CYPs by essential oils constituents (eugenol, isoeugenol, gamma-terpi- nene, and quercetin) may decrease the formation of toxic metabolites, as CYPs play an impor- tant role in procarcinogens activation [8,57]. In the present study, the mechanism of liver protection mediated by these essential oils might be due to inhibition of the protein expression of CYP2E1 and 3A4 which were induced by cyclophosphamide. Supporting this finding, it has been reported that eugenol was found to prevent liver injury by decreasing CYP2E1 activity, lipid peroxidation indices, protein oxidation and inflammatory markers, prevented DNA strand break and abolished the expression of COX-2 gene [58]. Furthermore, eugenol increased the expression and activity of NAD(P)H:quinone oxidoreductase (QR), a major detoxifying enzyme, through NF-E2 related factor2 binding to antioxidant response element in QR gene [59]. Eugenol and isoeugenol, which are naturally found in essential oils of different spices, suppressed the growth and proliferation of HaCaT cells via aryl hydrocarbon receptor (AhR) interactions [60]. They showed a rapid and marked translocation of AhR into the nucleus of the HaCaT cells, and consequently induced the expression of the AhR target genes. In addition, these compounds reduced levels of the cyclin-dependent kinase (CDK)6 [60]. Another compo- nent of essential oils, quercetin, was found to act as scavenger of O2–, NO–, HO–, and peroxy radicals. In addition, previous studies have shown that quercetin inhibited the oxidative DNA damage induced by [61] and preventing free radical-mediated cytotoxicity [62]. Based on this finding, the present study might provide another new mechanism for essen- tial oils protection against CP-induced hepatotoxicity via induction the protein expressions and activities of GPx and GST which diminished free radicals levels, and consequently reduced cytotoxicity.

PLOS ONE | DOI:10.1371/journal.pone.0165667 November 1, 2016 11 / 17 Protective Effects of Essential Oils against Hepatotoxicity Induced by Cyclophosphamide

Fig 3. Immunoblot of CYP 2E1, c-reductase, 3A4, 2B1/2, 2C23 and 2C26 in the pooled proteins of matched control, CP, fennel EO, cumin EO, clove EO, CP & fennel EO, CP & cumin EO and CP & clove EO treated groups. doi:10.1371/journal.pone.0165667.g003

In the current study, levels of serum hepatic marker enzymes were elevated in the livers after treatment of mice with CP. This is in agreement with other previous reports regardless the route, duration, and dose of administration of anti-cancer drugs [63]. The hepatoprotective effects against toxicity of CP were seen since all tested essential oils recovered the hepatic marker enzymes activities to their normal levels, which are in accordance with other previous studies [64,65]. Moreover, the ethanolic extract of clove was found to restore the levels of AST, ALT, and ALP to their normal levels in -induced liver toxicity [66]. The current histopathological changes are correlated with the altered enzyme activities and are in accor- dance with the previous studies [67]. The currently observed histological disruption in CP- treated mice might be related to the generation of more toxic metabolites of CP [68] which lead to liver injury due to the generation of reactive oxygen species. Acrolein has been reported to have an inflammatory response, subepithelial edema, neutrophil infiltration, hemorrhage, and necrosis. It is concluded that administration of fennel, cumin, and clove EOs exhibited hepatoprotec- tive effects against CP-induced toxicity. Hepatoprotective effect of EOs might be due to induc- tion of antioxidant defense systems. Also, it is suggested that co-administration of fennel, cumin, and/or clove essential oils with CP can be used for cancer therapy to reduce the hepato- toxicity caused by CP.

PLOS ONE | DOI:10.1371/journal.pone.0165667 November 1, 2016 12 / 17 Protective Effects of Essential Oils against Hepatotoxicity Induced by Cyclophosphamide

Fig 4. (A) Histological photomicrographs of liver sections of control, (B) cyclophosphamide, (C) fennel EO, (D) cumin EO, (E) clove EO, (F) CP& fennel EO, (G) CP & cumin EO and (H) CP & clove EO-treated groups. Cyclophosphamide treated group exhibited distortion in hepatic architecture, dilation in sinusoidal space, central vein (CV) hemorrhage, leukocytic infiltration (L.I.) in portal tract and hepatocytes (H), as well as, hyperplasia in Kuppfer cells (B). Conversely, liver section of fennel, cumin or clove essential oil treated mice exhibited normal architecture with normal hepatocytes and sinusoidal space comparative to the control group (C, D and E). The hydropic, necrotic and inflammatory changes regressed in liver section of mice treated concomitantly with essential oils and CP (F, G and H). doi:10.1371/journal.pone.0165667.g004 Author Contributions Conceptualization: SAS LSE. Data curation: SAS LSE.

PLOS ONE | DOI:10.1371/journal.pone.0165667 November 1, 2016 13 / 17 Protective Effects of Essential Oils against Hepatotoxicity Induced by Cyclophosphamide

Formal analysis: SAS LSE. Investigation: SAS LSE. Methodology:MAN. Project administration: SAS. Resources: SAS. Software: MAN. Validation: SAS. Visualization: MAN. Writing – original draft: SAS LSE. Writing – review & editing: SAS LSE.

References 1. Moignet A, Hasanali Z, Zambello R, Pavan L, Bareau B, Tournilhac O, et al,. Cyclophosphamide as a first-line therapy in LGL leukemia. Leukemia 2014; 28:1134±1136. doi: 10.1038/leu.2013.359 PMID: 24280867 2. Tsai-Turton M., Luong BT, Tan Y, Luderer U. Cyclophosphamide-induced apoptosis in COV434 human granulosa cells involves oxidative stress and glutathione depletion. Toxicol Sci, 2007; 98, 216± 230. doi: 10.1093/toxsci/kfm087 PMID: 17434952 3. Emadi A, Jones RJ, Brodsky RA. Cyclophosphamide and cancer: golden anniversary. Nat Rev Clin Oncol. 2009; 6: 638±647. doi: 10.1038/nrclinonc.2009.146 PMID: 19786984 4. Rodriguez-AC, Ingelman-Sundberg M. Cytochrome P450 pharmacogenetics and cancer.Oncogene, 2006; 25: 1679±1691. doi: 10.1038/sj.onc.1209377 PMID: 16550168 5. Kehrer JP, Biswal SS. The molecular effects of acrolein. Toxicol Sci, 2000; 57:6±15. PMID: 10966506 6. Wahlang B, Falkner KC, Cave MC, Prough RA. Role of Cytochrome P450 Monooxygenase in Carcino- gen and Chemotherapeutic Drug Metabolism. Adv Pharmacol. 2015; 74:1±33. doi: 10.1016/bs.apha. 2015.04.004 PMID: 26233902 7. Muzio G, Marengo B, Salvo R, Semeraro A, Canuto RA, Tessitore L. Liver cancer is induced by a sub- necrogenic dose of DENA when associated with fasting/refeeding: Role of glutathione-transferase and lipid peroxidation. Free Radic Biol Med, 1999; 26(9/10):1314±1320. 8. Sheweita SA. Drug-metabolizing enzymes: Mechanisms and Functions. A review. Curr Drug Metab, 2000, 1:107±132. PMID: 11465078 9. Murray GI, Burke MD, Ewen SW. Glutathione localisation in benign and malignant human breast lesions. Br J Cancer, 1987; 55(6):605±609. PMID: 3620301 10. Gopalan P, Tsuji K, Lehmann K, Kimura M, Shinozuka H, Sato K, et al,. Modulation of aflatoxin B1- induced glutathione S-transferase placental form positive hepatic foci by pretreatment of rats with phe- nobarbital and buthionine sulfoximine. Carcinogenesis 1993; 14(7):1469±1470. PMID: 8330366 11. Gnanasekaran S, Sakthivel KM, Chandrasekaran G. Immunostimulant and chemoprotective effect of vivartana, a polyherbal formulation against cyclophosphamide induced toxicity in Swiss albino mice. J Exp Ther Oncol, 2015; 11(1):51±61 PMID: 26259390 12. Zhu H, Long MH, Wu J, Wang MM, Li XY, Shen H, et al.,. Ginseng alleviates cyclophosphamide- induced hepatotoxicity via reversing disordered homeostasis of glutathione and bile acid. Sci Rep. 2015; 5:17536. doi: 10.1038/srep17536 PMID: 26625948 13. Singh S, Lata S, Tiwari KN. Antioxidant potential of Phyllanthusfraternus Webster on cyclophospha- mide induced changes in sperm characteristics and testicular oxidative damage in mice. Indian J Exp Biol, 2015; 53(10):647±56. PMID: 26665295 14. Badgujar SB, Patel VV, Bandivdekar AH. Foeniculumvulgare Mill: A review of its botany, phytochemis- try, pharmacology, contemporary application, and Toxicology. Biomed Res Int. 2014 2014:842674. doi: 10.1155/2014/842674 PMID: 25162032 15. Johri RK. Cuminumcyminum and Carumcarvi: An update. Pharmacogn Rev, 2011; 5: 63±72. doi: 10. 4103/0973-7847.79101 PMID: 22096320

PLOS ONE | DOI:10.1371/journal.pone.0165667 November 1, 2016 14 / 17 Protective Effects of Essential Oils against Hepatotoxicity Induced by Cyclophosphamide

16. Nadeem M, Riaz A. Cumin (Cuminumcyminum) as a potential source of antioxidants. Pak J Food Sci. 2012; 22: 101±107. 17. Cortes-Rojas DF, Souza CR, Oliveira WP. Clove (Syzygiumaromaticum): a precious spice. Asian Pac J Trop Biomed. 2014; 2: 90±96. 18. Tripathi P, Patel RK, Tripathi R, Kanzariya NR Investigation of antigenotoxic potential of Syzygium cumini extract (SCE) on cyclophosphamide-induced genotoxicity and oxidative stress in mice. Drug Chem Toxicol, 2013; 36(4):396±402. doi: 10.3109/01480545.2012.749271 PMID: 23298251 19. Tripathi P1, Tripathi R, Patel RK, Pancholi SS. Investigation of antimutagenic potential of Foeniculum vulgare essential oil on cyclophosphamide induced genotoxicity and oxidative stress in mice. Drug Chem Toxicol. 2013; 36(1):35±41 doi: 10.3109/01480545.2011.648328 PMID: 22264205 20. Vieira PM, Veronezi E, Silva CR, Chen-Chen L. Detection of genotoxic, cytotoxic, and protective activi- ties of Eugenia dysenterica DC. (Myrtaceae) in mice. J Med Food. 2012; 15(6):563±567. doi: 10.1089/ jmf.2011.0270 PMID: 22404573 21. Adams RR. Identification of essential oil components by gas chromatography/mass spectrometry. 4th ed. Illinois: Allured Publishing Corporation; 2007 22. Rayan D, Lu A, Levin W. Preparation of rat hepatic microsomes. Methods Enzymol.1978; 752: 118± 122. 23. Venkatesan N, Arcos JC, Argus MF. Differential effect of polycyclic hydrocarbons on the demethylation of the carcinogenic dimethylnistrosamine by rat tissues. Life Sci, 1968; 7:1111±1119. 24. Mostafa M, Sheweita SA. Modification of the oxidative N-demethylation of dimethylnistrosamine by various anti-inflammatory drugs. Ramaz Newslett, 1992; 2:15±22. 25. Greenle W, Poland A. An improved assay of 7-ethoxycoumarin O-deethylase activity: induction of hepatic enzyme activity in C57BL/6J and DBA/2J mice by Phenobarbital, 3-methylcholanthrene, and 2,3,7,8-tetrachlorodibenzo-p-dioxin. J Pharmacol Exp Ther, 1978; 205:596±605. PMID: 660532 26. Willimias CH, Kamin H. Microsomal triphosphopyridine nucleotide cytochrome C reductase of liver. J Biol Chem, 1962; 237:587±590. PMID: 14007123 27. Chiu DT, Stults FH, Tappel AL. Purification and properties of rat lung soluble glutathione peroxidase. BiochimBiophysActa,1976; 445: 558±566. 28. David M and Richard JS. Methods of enzymatic analysis. In: J. Bergmeyer and G. B. Marianna (eds.), Basel; 1983; 26: 358±359. 29. Habig WH, Pabst MJ, Jacoby WB. Glutathione S-transferases: the first enzymatic step in mercapturic acid formation. J Biol Chem,1974; 249:7130±7139. PMID: 4436300 30. Beers RF, Sizer IW. A spectrophotometric method for measuring the breakdown of hydrogen peroxide by catalase. J Biol Chem, 1952; 195:133±140. PMID: 14938361 31. Misra HP, Fridovich I. the role of superoxide anion in the autoxidation of epinephrine and a simple assay for superoxide dismutase. J Biol Chem, 1972; 247:3170±3175. PMID: 4623845 32. Lowry OH, Rosbrough NJ, Farr AL, Randall RJ. Protein measurement with the Folin phenol reagent. J Biol Chem, 1951; 193: 265±275. PMID: 14907713 33. Tappel AL, Zalkin H. Inhibition of lipid peroxidation in mitochondria by vitamin E. Arch Biochem Bio- phys,1959; 80: 333±336. 34. Mitchell JR, Jollow DJ, Potter WZ, Davis DC, Gillette JR, Brodie BB. Acetaminophen-induced hepatic necrosis. I. Role of drug metabolism. J Pharmacol Exp Ther,1973; 187: 185±194. PMID: 4746326 35. Matsudaira P. Sequence from picomole quantities of proteins electroblotted onto polyviny idenedifluor- ide membranes. J Biol Chem, 1987; 262: 10035±10038. PMID: 3611052 36. Drury AA, Wallington EA. Carleton's histological technique. 5th ed. New York, Toronto: Oxford Uni- versity press; 1980. 37. Maor Y, Malnick, S. Liver injury induced by anticancer chemotherapy and radiation therapy. Int J Hepa- tol. 2013, 2013:815105 doi: 10.1155/2013/815105 PMID: 23970972 38. Sheweita SA, Tilmisany AK. Cancer and phase II drug-metabolizing enzymes. Curr Drug Metab, 2003; 4(1):45±58. PMID: 12570745 39. IARC, IARC monographs on the evaluation of carcinogenic risks to humans, a review of human carcin- ogens: An Updating of IARC Monographs Volumes 100 A, 2012., Lyon, France. 40. Chang TK, Yu L, Maurel P, Waxman DJ. Enhanced cyclophosphamide and ifosfamide activation in pri- mary human hepatocyte cultures: response to cytochrome P-450 inducers and autoinduction by oxa- zaphosphorines. Cancer Res, 1997; 57: 1946±1954. PMID: 9157990 41. Xie HJ, Yasar U, Lundgren S, Griskevicius L, Terelius Y, Hassan M, et al. Role of polymorphic human CYP2B6 in cyclophosphamide bioactivation. Pharmacogenomics J, 2003; 2: 53±61.

PLOS ONE | DOI:10.1371/journal.pone.0165667 November 1, 2016 15 / 17 Protective Effects of Essential Oils against Hepatotoxicity Induced by Cyclophosphamide

42. De-Oliveira AC, Ribeiro-Pinto LF, Otto SS, GoncËalves A, Paumgartten FJ. Induction of liver monooxy- genases by beta-myrcene. Toxicol, 1997; 124(2):135±40 43. Barouki R, Han HE, Hwang PY, Jeong CT, Lee SS, Shin GJ, et al.,. Eugenol inhibit 7, 12-dimethylbenz [α]anthracene-induced genotoxicity in MCF-7 cells: Bifunctional effects on CYP1 and NAD(P)H: qui- none oxidoreductase. FEBS Lett, 2007; 581:749±756. doi: 10.1016/j.febslet.2007.01.044 PMID: 17275817 44. Gagandeep S, Dhanalakshmi E, Mendiz A, Rao R, Kale RK. Chemopreventive effects of Cuminumcy- minum in chemically induced forestomach and uterine cervix tumors in murine model systems. Nutr Cancer, 2003; 47: 171±180. doi: 10.1207/s15327914nc4702_10 PMID: 15087270 45. Ariyoshi T, Arakaki M, Ideguchi K, Ishizuka Y, Noda K. Studies on the metabolism of d-Limonene (p- Mentha-1,8-diene). III. Effects of d-Limonene on the lipids and drug-metabolizing enzymes in rat livers. Xenobiotica,1975; 5(1):33±8. PMID: 1154797 46. Austin CA1, Shephard EA, Pike SF, Rabin BR, Phillips IR. The effect of terpenoid compounds on cyto- chrome P-450 levels in rat liver. Biochem Pharmacol, 1988; 37(11):2223±9. PMID: 3377821 47. Gupta AK, Misra N. Hepatoprotective activity of aqueous ethanolic extract of Chamomile capitula in paracetamol intoxicated albino rats. Am J Pharm Toxicol, 2006; 1: 17±20. 48. Saleh MA, Clark S, Woodard B, Deolu-Sobogun SA. Antioxidant and free radical scavenging activities of essential oils. Ethn Dis, 2010; 20: 78±82. 49. Kaurinovic B, Vlaisavljevic S, Popovic M, Vastag D, Djurendic-Brenesel M. Antioxidant properties of Marrubiumperegrinum L. (Lamiaceae) essential oil. Molecules. 2010; 15:5943±5955. 50. Misharina TA, Samusenko AL. Antioxidant properties of essential oils from lemon, grapefruit, corian- der, clove and their mixtures. Appl Biochem Microbiol, 2008; 44: 438±442. 51. Huang HC, Ho YC, Lim JM, Chang TY, Ho CL, Chang TM. Investigation of the anti-melanogenic and antioxidant characteristics of eucalyptus camaldulensis flower essential oil and determination of its chemical composition. Int J Mol Sci. 2015; 16(5):10470±90 doi: 10.3390/ijms160510470 PMID: 25961954 52. Chakraborty P, Sk UH, Bhattacharya S. Chemoprotection and enhancement of cancer chemothera- peutic efficacy of cyclophosphamide in mice bearing Ehrlich ascites carcinoma by diphenyl methyl selenocyanate. Cancer Chemoth Pharm, 2009; 64: 971±980. 53. Shanmugarajan TS, Arunsundar M, Somasundaram I, Sivaraman D, Krishnakumar E, Ravichandran V. Ameliorative effect of Ficushispida Linn. leaf extract on cyclophosphamide-induced oxidative hepatic injury in rats. J Pharmacol Toxicol, 2008; 3: 363±372. 54. Zarei M, Shivanandappa T. Amelioration of cyclophosphamide-induced hepatotoxicity by the root extract of Decalepishamiltonii in mice. Food Chem. Toxicol, 2013; 57: 179±184. doi: 10.1016/j.fct. 2013.03.028 PMID: 23542512 55. Conklin DJ, Haberzettl P, Lesgards JF, Prough RA, Srivastava S, Bhatnagar A Increased Sensitivity of Glutathione S-Transferase P-Null Mice to Cyclophosphamide-Induced Urinary Bladder Toxicity. J Pharmacol Exp Ther, 2009; 331; 456±469 (). doi: 10.1124/jpet.109.156513 PMID: 19696094 56. Myers CR, Myers JM. The Effects of Acrolein on Peroxiredoxins, Thioredoxins, and Thioredoxin Reductase in Human Bronchial Epithelial Cells. Toxicology, 2009; 257, 95±104. doi: 10.1016/j.tox. 2008.12.013 PMID: 19135121 57. Zhou SF, Xue CC, Yu XQ, Wang G. Metabolic activation of herbal and dietary constituents and its clini- cal and toxicological implications: an update. Curr Drug Metab.,2007; 8(6):526±53 PMID: 17691916 58. Yogalakshmi B, Viswanathan P, Anuradha CV. Investigation of antioxidant, anti-inflammatory and DNA-protective properties of eugenol in thioacetamide-induced liver injury in rats. Toxicology. 2010; 268(3):204±12. doi: 10.1016/j.tox.2009.12.018 PMID: 20036707 59. Han EH, Hwang YP, Jeong TC, Lee SS, Shin JG, Jeong HG Eugenol inhibit 7,12-dimethylbenz[a] anthracene-induced genotoxicity in MCF-7 cells: Bifunctional effects on CYP1 and NAD(P)H:quinone oxidoreductase. FEBS Lett. 2007; 581(4):749±56. doi: 10.1016/j.febslet.2007.01.044 PMID: 17275817 60. Kalmes M, BloÈmeke B. Impact of eugenol and isoeugenol on AhR translocation, target gene expres- sion, and proliferation in human HaCaT keratinocytes. J Toxicol Environ Health A. 2012; 75(8± 10):478±91. doi: 10.1080/15287394.2012.674916 PMID: 22686307

61. Musonda CA. and Chipman J.K. ªQuercetin inhibits hydrogen peroxide (H2O2)-induced NF-κB DNA binding activity and DNA damage in HepG2 cells,º Carcinogenesis, 1998, 19(9):1583±1589. PMID: 9771928 62. Zhang YMC., ªProtective effect of quercetin on aroclor 1254-induced oxidative damage in cultured chicken spermatogonial cells,º Toxicological Sci, 2005, 88 (2): 545±550.

PLOS ONE | DOI:10.1371/journal.pone.0165667 November 1, 2016 16 / 17 Protective Effects of Essential Oils against Hepatotoxicity Induced by Cyclophosphamide

63. Nithya N, Chandrakumar K, Ganesan V, Senthilkumar S. Efficacy of Momordicacharantia in attenuat- ing hepatic abnormalities in cyclophosphamide intoxicated rats. J Pharmacol Toxicol. 2012; 7: 38±45. 64. Barakat LA, Mohamed MM. Ginger, cumin and mustard seeds modulate acetaminophen-induced acute hepatic injury in rats. J Appl Sci Res, 2011; 7: 1368±1374. 65. Rabeh NM, Aboraya AO. Hepatoprotective effect of dill (Anethumgraveolens L.) and Fennel (Foenicu- lumvulgare) oil on hepatotoxic rats. Pakistan J Nutr, 2014; 13: 303±309. 66. Abozid MM, EL-Sayed SM. Antioxidant and protective effect of clove extracts and clove essential oil on hydrogen peroxide treated rats. Int J Chem Tech Res, 2013; 5: 1470±1485. 67. Aboul-Ela EI, Omara EA. Genotoxic and histopathological aspects of treatment with grape seed extract on cancer induced with cyclophosphamide in mice. J Cell Biol, 2014; 2:18±27. 68. Bhattacharya R, Lawrence A, Krishnan A, Zaman K, Sun D, Fernandes G. Effect of dietary n-3 and n-6 oils with and without food restriction on activity of antioxidant enzymes and lipid peroxidation in livers of cyclophosphamide-treated autoimmune-prone NZB/W female mice. J Am Coll Nutr, 2003; 22: 388± 399. PMID: 14559931

PLOS ONE | DOI:10.1371/journal.pone.0165667 November 1, 2016 17 / 17