Abstract
Objective(s):
The use of doxorubicin (DOX) is limited by its dose-dependent cardio toxicity in which reactive Oxygen Species (ROS) play an important role in the pathological process. The aim of this study was to evaluate the protective effect of three medicinal plants, Nigella sativa (N), Glycyrrhiza glabra (G) and Zingiber officinale (Z), and their combination (NGZ), against DOX-induced apoptosis and death in H9c2 cells.
Materials and Methods:
The cells were incubated with different concentrations of each extract or NGZ for 4 hr which continued in the presence or absence of 5µM doxorubicin for 24 hr. Cell viability and the apoptotic rate were determined using 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyl tetrazolium (MTT) and propidium iodide (PI) staining assays, respectively. The level of ROS and lipid peroxidation were measured by fluorimetric methods.
Results:
Treatment with doxorubicin increased ROS generation, enhanced malondialdehyde (MDA) formation, and induced apoptosis. Co-treatment of the cells with each herb extract increased viability of cells dose-dependently with a maximum protection effect of about 30%, and their potencies were N>G>Z. The combination of the threshold dose of each extract (NGZ) produced a similar effect, which was increased dose-dependently to a maximum protection of 70%. These effects were correlated with the effects of NGZ on ROS and MDA.
Conclusion:
All of the extracts have some protective effects against DOX-induced toxicity in cardiomyocytes with similar efficacies, but with different potencies. However, NGZ produced much higher protective effect via reducing oxidative stress and inhibiting of apoptotic induction processes. Further investigations are needed to determine the effects of NGZ on DOX chemotherapy.
Keywords: Doxorubicin, Nigella sativa, Glycyrrhiza glabra, Zingiber officinale
Introduction
Doxorubicin (DOX) is an important component of multimodality therapy for several antineoplastic combined chemotherapy protocols. It is used to treat a variety of malignacies such as leukemias, Hodgkin and non-Hodgkin lymphoma, and solid tumors (1). However, despite high efficacy, the major side effect of DOX is cardio-toxicity, which has dramatically hindered its clinical usage for a prolonged period of time. The mechanisms of DOX-induced cardio toxicity are not completely understood, but most evidences indicate the generation of reactive oxygen species (ROS) involvement (2). Interestingly, some natural foods have been reported to contain substantial amounts of antioxidants and free radical scavenging agents. These compounds diminish some side effects of chemotherapeutic agents on normal cells by reducing their genotoxicity (2). There are many evidences showing the protective effects for different herbs against oxidative injury-related cardio toxicity, and among which N. sativa, Zingiber officinale and Glycyrrhiza glabra showed the highest protective effect.
N. sativa is used as spice and food preservative (3). Its seeds have long been used in traditional medicine for a wide range of disorders including bronchial asthma, headache, infections, obesity, back pain, hypertension and gastrointestinal problems (4). These effects are mediated via different mechanism such as antioxidant (5), anti-inflammatory (6), anticancer (7), and antihistaminic effects (8). It has been shown that N. sativa attenuates isoproterenol induced myocardial infarction (MI) (9). Also, N. sativa oil supplementation reduces lead-induced cardio-toxicity by mechanisms related to its ability to decrease the pro-inflammatory cytokines; oxidative stress and cardiac tissue damage, and preserve the activity of antioxidant enzymes (10). Ebru et al (2008) showed that pretreatment with N. sativa oil decreased the subsequent cyclosporine-A injury in rat heart (11), Recent studies revealed a protective effect of thymoquinone against doxorubicin-induced cardio toxicity (12-14).
Z. officinale, commonly known as ginger belongs to the Zingiberaceae family. The rhizome of ginger has an aromatic pungent taste. It is consumed worldwide as a spice and flavoring agent, and is attributed to have many medicinal properties (15). It is used in traditional medicine as carminative, and antipyretic, and in the treatment of pain, rheumatism and bronchitis (15). It has different pharmacological activities such as hepatoprotective (16), antiparasitic (17), antimalarial (18), antimicrobial (19), antidiabetic (20), and radioprotective effects (21). Ginger also is a potential remedy for cardiovascular diseases (22), and can prevent the development of morphine analgesic tolerance and physical dependence in rats (23). It is also used for the treatment of gastrointestinal disorders including gastric ulcerogenesis (24). The plant contains high level of phenolic and flavonoid compounds, responsible for its high antioxidant activities (25).
G. glabra (licorice) is used widely as a flavoring and sweetening agent in tobacco products, chewing gum, candy, toothpaste and beverages. In addition, licorice is frequently prescribed as a treatment in oriental herbal medicine. Licorice root has cancer chemopreventive effect (26).
The therapeutic potential of G. glabra as a hypoglycemic (27), hypocholesteremic (28), anti-ulcer (29), anti-inflammatory (30), renoprotective (31), and antiatherogenic (32) have been demonstrated. In traditional medicine, it is useful in the treatment of agent renovascular and cardiovascular diseases, and hence constitutes as the major ingredient of polyherbal formulations indicated for cardiovascular diseases (31). The investigations have shown that G. glabra has protective effects against ischemic damages of the several body organs due to its potent antioxidant and free radical scavenging activity (27, 28, 30-32). Also, a recent study has shown that pretreatment with G. glabra significantly attenuates ischemic–reperfusion induced myocardial injury by improving antioxidant status in heart (33).
In addition, other species of G. uralensis, suppressed doxorubicin-induced apoptosis in H9c2, rat cardiac myoblast cells (34). Treatment with licorice extract significantly protected the mice against DOX-induced cardio toxicity (35), and improved cardiac performance (36).
Based on these findings, we hypothesized that the combination of these medicinal plants may produce a higher protective effect against doxorubicin-induced cardiomyopathy. Therefore, the present study was designed to investigate the potential effects of Nigella sativa with Glycyrrhiza glabra and Zingiber officinale (NGZ) extracts on cell viability, lipid peroxidation level, ROS content and apoptotic induction in H9c2 cardiomyocytes.
Materials and Methods
Reagents and chemicals
3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyl tetrazolium (MTT), Thiobarbituric acid (TBA), 2,7-dichlorofluorescin diacetate (DCFH-DA), Propidium iodide (PI), sodium citrate and Triton X-100 were purchased from Sigma (St. Louis, MO, USA). High glucose Dulbecco’s Modified Eagles Medium (DMEM), penicillin-streptomycin and fetal bovine serum were purchased from Gibco. Trichloroacetic acid (TCA) and malondialdehyde bis-(dimethyl acetal) (MDA) were obtained from Merck (Darmstadt, Germany). H9c2 cells were obtained from Pasteur Institute (Tehran, Iran).
Preparation of extracts
N. sativa seeds were collected from Gonabad region (northeast of Iran) and authenticated by Herbarium of the Ferdowsi University of Mashhad (FUMH) (voucher specimen = 293-0303-1). G. glabra rhizomes was purchased from local market and authenticated by FUMH (voucher specimen number = 25947). Whereas Z. officinale is not native to Iran, afforded from a local herb market in Mashhad and confirmed by plant specialist of FUMH. The seeds of N. sativa were washed, dried, and crushed to a powder with an electric micronizer. The G. glabra and Z. officinale rhizomes were peeled chopped into tiny bits, air-dried and were ground with a mechanical grinder. Each herbal sample was extracted separately in a Soxhlet extractor with ethanol (70%) and the resulting extract was dried and kept at -20°C until use.
Cell culture and treatment
H9c2 Cells were maintained at 37°C in a humidified atmosphere containing 5% CO2. The cells were cultured in Dulbecco’s Modified Eagles Medium (DMEM) supplemented with 10% fetal bovine serum, 100 Units/ml penicillin and 100 µg/ml streptomycin. For the experiments, they were seeded in 96-well and 24-well culture plates for MTT/ROS and MDA assays, respectively. For apoptosis assay, cells were seeded at 100,000 cell/well in a 24-well plate. All treatments were carried out in triplicate. The cells were pretreated with each herbal extract alone (6 to 200 µg/ml) for 4 hr and then incubation was continued in the presence of the herbal extract with or without 5 µM doxorubicin for 24 hr. For assaying the effect of three extract in combination, the minimum effective concentration of each extract was chosen (N=12.5, G=25 and Z=25 μg/ml sum=62.5 μg/ml) as the basis and one (62.5 μg/ml), two (125 μg/ml), four (250 μg/ml) and eight (500 μg/ml) folds of this concentration were applied.
Cell viability assay
Cell viability was determined using a modified MTT assay as described previously (37). Briefly, MTT solution in phosphate-buffered saline (5 mg/ml) was added to each well at final concentration of 0.05%. After 3 hr, the formazan precipitate was dissolved in DMSO. The absorbance at 570 and 620 nm (background) was measured using a StatFAX303 plate reader.
Lipid peroxidation assay
The level of lipid peroxidation was estimated by measuring MDA, which is the end product of lipid peroxidation (38). At the end of incubation, the cells were scraped and centrifuged for 30 min. Then, 400 µl of TCA (15%) and 800 µl of TBA (0.7%) were added to 500 µl of cell samples. The mixture was vortexed and heated for 40 min in a boiling water bath. Then, 200 µl of the sample was transferred to 96-well plate and the fluorescence intensity was read with excitation/emission of 480/530 nm. The experiment was carried out in triplicate.
Measurement of reactive oxygen species
Intracellular ROS level was evaluated using a fluorescent probe, DCF-DA. At the end of incubation, the cells were treated (30 min) with DCFH-DA (10µM) at 4°C in the dark. Then, the fluorescence intensity was detected with excitation/emission of 485/530 nm. The experiment was performed in triplicate.
PI staining
Apoptotic cells were detected using PI staining of small DNA fragments followed by flow cytometry. It has been reported that a sub-G1 peak that is reflective of DNA fragmentation can be observed following the incubation of cells in a hypotonic phosphate-citrate buffer containing a quantitative DNA-binding dye such as PI. Apoptotic cells that have lost DNA will take up less stain and appear on the left side of the G1 peak in the histogram. Briefly, H9c2 cells were seeded in wells of a 24-well plate and treated according to mentioned protocol. Floating and adherent cells were then harvested and incubated at 4 °C overnight in the dark with 750 μl of a hypotonic buffer (50 μg/ml PI in 0.1% sodium citrate with 0.1% Triton X-100). Next, flow cytometry was carried out using a FACScan flow cytometer (Becton Dickinson). A total of 104 events were acquired with FACS.
Statistics
All data were expressed as mean ± SEM. Statistical analysis was performed using one way analysis of variance (ANOVA) followed by Tamhane’s T2 post-hoc test. Differences were considered significant at P<0.05.
Results
Effect of extracts alone or in combination (NGZ) on cell viability
Incubation with doxorubicin significantly decreased cell viability to 62±2% compared to control of control (P<0.001). Cell viability was assayed to determine the optimum concentrations necessary for the three extracts (N, G and Z) to protect H9c2 cells against DOX-induced cytotoxicity. The results demonstrated that, in comparison with DOX, N. sativa increased cell viability at doses of 25 to 200 μg/ml dose dependently that maximum effect was produced at effect at concentration of 50 μg/ml (75.4±0.84, P<0.01), and Z. officinale increased cell viability at doses of 50 to 200 μg/ml, its maximum effect was at dose of 100 μg/ml (75±2.5, P<0.01), while G. glabra increased cell viability at dose of 100 μg/ml (73.5±1.5, P<0.05) (Figure 1A).
Results of experiment with combination extract (NGZ) showed that all doses (62.5, 125, 250 and 500 µg/ml) protected H9c2 cells against DOX and increased cell viability significantly. The best protection was obtained at dose of 250 µg/ml (90.29±1.2, P<0.001, 70% protection).
The effect of NGZ on ROS content and lipid peroxidation
The biochemical determination of malondialdehyde (MDA) represents lipid peroxide formation. The results showed that DOX significantly increased MDA (P<0.001) and ROS (P<0.001) levels in H9c2 compared with control cells. In the presence of DOX, treatment with 125 µg/ml (P<0.05), 250 µg/ml (P<0.001) and 500 µg/ml (P<0.01) doses of NGZ significantly reduced the level of MDA. The most reduction of MDA was observed at the dose of 250 µg/ml (control 100±1.4, DOX 235±7.9 and 250 µg/ml NGZ 119±5.18). Also this combination diminished significantly ROS production in comparison with DOX, at dose of 125 µg/ml (P<0.01), 250 µg/ml (P<0.001) and 500 µg/ml (P<0.01) (Figures 2A and 2B, respectively). The lowest production of ROS was at the dose of 250 µg/ml (control 27±4, DOX 120±8 and 250 µg/ml NGZ 45±4).
The effect of NGZ on apoptosis induction
Apoptosis in H9c2 cell line was detected with flow cytometry using PI staining. Cells were pretreated for 4 hr with various concentrations of the NGZ and exposed to DOX for 24 hr. Analysis of the subG1 peak in flow cytometry histograms revealed the induction of apoptosis in cells treated with DOX (P<0.001). NGZ decreased apoptotic induction significantly at the doses of 62.5 µg/ml (P<0.05), 125 µg/ml (P<0.01), 250 µg/ml (P<0.001) and 500 µg/ml (P<0.01) (Figure 3). The most reduction of apoptotic rate was at the dose of 250 µg/ml (control 12±1.4, DOX 71±4 and 250 µg/ml NGZ 37.5±2.5)
Discussion
Cardiovascular risk factors enhance the production of ROS generated by mitochondrial electron-transport chain, xanthine oxidase, NADPH oxidase and uncoupled nitric oxide synthases in cardiomyocytes (39). Oxidative stress occurs when production of ROS exceeds the capacity of antioxidant defense systems (catalase, SOD and glutathione peroxidase) (40). Because of its deleterious effects, this stress is associated with poor outcomes in cardiovascular diseases (41). Weak antioxidant capacity in cardiomyocytes may be a risk factor responsible for their high sensitivity to oxidative damage (42), and a promising approach to cardioprotection is the use of pharmacological tools to reduce oxidative stress in the heart (37). In the present study, we used H9c2 cells as a pharmacological model to evaluate the potential cardioprotective effects of three different prevalent medicinal plants, N. sativa, Glycyrrhizin glabra and Zingiber officinale alone and in combination with all three (NGZ). The results showed that the combination of NGZ improves their protective effects against DOX-induced oxidative stress in H9c2 cells.
H9c2 cells are morphologically similar to immature embryonic cardiomyocytes. Considering that these cells preserve electrical and hormonal signal pathways found in adult cardiac cells (43), they are a useful model for studying oxidative stress-induced cardiomyocyte damage (44). In this model, DOX significantly increased the level of ROS and lipid peroxidation and induced the rate of apoptosis. These changes are similar to the DOX-induced deleterious effects on normal cardiac cells, which lead to the loss of cardiomyocytes viability (2).
In this study, pretreatment with each of the three herb extracts alone produced some degree of protection in a concentration dependent manner. These findings are in accordance with previous studies which show the capability of cadioprotective effects of N. sativa, G. glabra and Z. officinale (10-14).
In mice, thymoquinone, the pharmacologically active component of N. sativa, reduces DOX-induced cardio toxicity without reducing the serum concentration of DOX (12). Also, studies have shown that thymoquinone potentiates cisplatin antitumor activity and protects against cisplatin-induced nephrotoxicity in mice and rats (45). A recent investigation has shown that N. sativa oil supplementation attenuates lead-induced cardio toxicity by mechanisms related to its ability to decrease the pro-inflammatory cytokines; oxidative stress and cardiac tissue damage, and preserve the activity of antioxidant enzymes (10). Ebru et al (2008) showed that pretreatment with N. sativa oil decreased cyclosporine-A injury in rat heart (11), and later studies revealed a protective effect of thymoquinone against doxorubicin-induced cardio toxicity (12-14). Recently, it was shown that gingerol protected cardiomyocytes against DOX-induced toxicity through its antioxidative effect and modulation of NF-κB as well as apoptosis (46).
Glycyrrhizin, the chief sweet-tasting constituent of G. glabra (liquorice) root and its metabolites (glycyrrhizic acid or glycyrrhizinic acid) induce apoptosis and cell growth inhibition in various cancer cells, including human stomach cancer cells, promyelotic leukemia HL-60 cells and hepatoma cells (47, 48). Moreover, experimental results showed that the combination of antibiotic anti-cancer drug with 18β-glycyrrhetinic acid (derivative of the beta-amyrin type obtained from the hydrolysis of glycyrrhizic acid) exhibits a synergistic toxic effect on cancer cell lines (49). The extract of Glycyrrhiza uralensis suppresses doxorubicin-induced apoptosis in H9c2 rat cardiac myoblasts (34).
Recent experimental observations reported that Z. officinale is an effective anticancer agent (50). Also, in a polyherbal preparation, ginger was one of the components and reported to be effective against DOX-induced cardio toxicity without interfering its antineoplastic activity (51).
Thus, it is of great interest to investigate the effect of combination of these extracts on DOX-induced disorders. In this study, the combination of extracts (NGZ) augmented their protective effects, suggesting the presence of a positive interaction between components. In this experiment, a maximum effect about 30 percent protection was produced by each extracts. However, the maximum protective effect was achieved with combination of the threshold concentrations of the extracts and the protection increased in concentration dependent manner to a maximum protection of more than 2.5 fold. These observations do not show an additive effect, but implicates the presence of a positive interaction between extracts against DOX-induced apoptosis in H9c2 cells.
In high concentrations, N. sativa did not produce any further protective effect, while the effects of the two other extracts were decreased in high concentrations (Figure 1A). The decreasing effect of G. glabra or Z. officinale at concentration of 200 µg/ml could not be explained by a non-specific effect such as ionic disturbances. The combination at concentration of 250 µg/ml increased their protective effect. However, at higher concentration (500 µg/ml) in mixed extracts, the effect was reduced. This may suggest the presence of some components in crude extracts, which may have toxic effect and reduce the protection.
The molecular mechanisms for protective effect of each extract and augmenting effect of their combination (NGZ) are not clear. However, it has been shown that Glycyrrhizate could ameliorate rabbit myocardial ischemia-reperfusion injury through P38MAPK pathway (52). Also, treatment with Glycyrrhiza uralensis extract significantly protected mice from DOX-induced cardio toxicity, by decreasing levels of serum LDH and Creatin Kinase-MB improving heart morphology and increasing GSH-P(X) activity and GSH level (35). Also, results revealed that glycyrrhizin and glycyrrhetinic acid directly affected cardiac performance (36). In this paper, we showed that the protective effects may be mediated by reducing oxidative stress, which could decrease the production of ROS, lipid peroxidation and diminished apoptotic induction.
It is interesting that in comparison with the effect of each extract alone, the combination produced a stronger protective effect. Therefore, it can be concluded that cardioprotective effect of NGZ is mediated at least in part by its active ingredients. However, further investigations are needed to reveal molecular mechanisms to increase the possible positive interactions and diminish negative interactions in combination. These must be included in fractionation of the extracts to assess the effect of each fraction in combination. These may lead to find purified active ingredients, which could be used in a clinical trial.
Conclusion
It was shown that all of three extract produced protective effect on H9c2 cell against DOX-induced toxicity with different potencies, but similar efficacy with a maximum effect of about 30 percent. The protective effect of extracts was increased by using them in combination. This effect is correlated with reducing oxidative stress and inhibition of apoptosis induction. The effects of extracts in combination showed a positive interaction between components of different extracts, which augments their protective effect. These observations could suggest new experiments that can be performed to test the roles of each extract components in the combination. However, potential interactions of extracts with DOX chemotherapy should be assessed. These may lead to the development of a new class of drugs to prevent cardio toxicity of DOX and improve its anticancer effect.
Acknowledgment
The authors declare that they have no conflict of interest. This work was supported by Mashhad University of Medical Sciences (grant number: 930033), Mashhad, Iran.
References
- 1.Wu S, Ko YS, Teng MS, Ko YL, Hsu LA, Hsueh C, et al. Adriamycin-induced cardiomyocyte and endothelial cell apoptosis In vitro and in vivo studies. J Mol Cell Cardiol. 2002;34:1595–1607. doi: 10.1006/jmcc.2002.2110. [DOI] [PubMed] [Google Scholar]
- 2.Bryant J, Picot J, Levitt G, Sullivan I, Baxter L, Clegg A. Cardioprotection against the toxic effects of anthracyclines given to children with cancer: a systematic review. Health Technol Assess. 2007;11:1–84. doi: 10.3310/hta11270. [DOI] [PubMed] [Google Scholar]
- 3.Goreja WG. New York: Amazing Herbs Press; 2003. Black Seed: Nature's Miracle Remedy. [Google Scholar]
- 4.Al-Rowais NA. Herbal medicine in the treatment of diabetes mellitus. Saudi Med J. 2002;23:1327–1331. [PubMed] [Google Scholar]
- 5.Burits M, Bucar F. Antioxidant activity of Nigella sativa essential oil. Phytother Res. 2000;14:323–328. doi: 10.1002/1099-1573(200008)14:5<323::aid-ptr621>3.0.co;2-q. [DOI] [PubMed] [Google Scholar]
- 6.Houghton PJ, Zarka R, de las Heras B, Hoult JR. Fixed oil of Nigella sativa and derived thymoquinone inhibit eicosanoid generation in leukocytes and membrane lipid peroxidation. Planta Med. 1995;61:33–36. doi: 10.1055/s-2006-957994. [DOI] [PubMed] [Google Scholar]
- 7.Khalife KH, Lupidi G. Non enzymatic reduction of thymoquinone in physiological conditions. Free Radic Res. 2007;41:153–161. doi: 10.1080/10715760600978815. [DOI] [PubMed] [Google Scholar]
- 8.Kanter M, Coskun O, Uysal H. The antioxidative and antihistaminic effect of Nigella sativa and its major constituent, thymoquinone on ethanol-induced gastric mucosal damage. Arch Toxicol. 2006;80:217–224. doi: 10.1007/s00204-005-0037-1. [DOI] [PubMed] [Google Scholar]
- 9.Murugesan M, Ragunath M, Prabu T, Nadanasabapathi S, Sakthivel M, Manju V. Protective role of black cumin (Nigella sativa) on isoproterenol induced myocardial infarction in rats. Int J Pharmacol and Clin Sci. 2012;1:45–53. [Google Scholar]
- 10.Ahmed MA, Hassanein KMA. Cardio protective effects of Nigella sativa oil on lead induced cardio toxicity: Anti-inflammatory and antioxidant mechanism. J Physiol Pathophysiol. 2013;4:72–80. [Google Scholar]
- 11.Ebru U, Burak U, Yusuf S, Reyhan B, Arif K, Faruk TH, et al. Cardioprotective effects of Nigella sativa oil on cyclosporine A-induced cardio toxicity in rats. Basic Clin Pharmacol Toxicol. 2008;103:574–580. doi: 10.1111/j.1742-7843.2008.00313.x. [DOI] [PubMed] [Google Scholar]
- 12.al-Shabanah OA, Badary OA, Nagi MN, al-Gharably NM, al-Rikabi AC, al-Bekairi AM. Thymoquinone protects against doxorubicin-induced cardio toxicity without compromising its antitumor activity. J Experimen Clin Cancer Res. 1998;17:193–198. [PubMed] [Google Scholar]
- 13.Effenberger-Neidnicht K, Schobert R. Combinatorial effects of thymoquinone on the anti-cancer activity of doxorubicin. Cancer Chemother Pharmacol. 2011;67:867–874. doi: 10.1007/s00280-010-1386-x. [DOI] [PubMed] [Google Scholar]
- 14.Nagi MN, Mansour MA. Protective effect of thymoquinone against doxorubicin-induced cardio toxicity in rats: A possible mechanism of protection. Pharmacol Res. 2000;41:283–289. doi: 10.1006/phrs.1999.0585. [DOI] [PubMed] [Google Scholar]
- 15.Afzal M, Al-Hadidi D, Menon M, Pesek J, Dhami MSI. Ginger: an ethnomedical, chemical and Pharmacological review. Drug Metabol Drug Interact. 2001;18:159–190. doi: 10.1515/dmdi.2001.18.3-4.159. [DOI] [PubMed] [Google Scholar]
- 16.Ezeonu C, Egbuna P, Ezeanyika US, Nkwonta CG, Idoko ND. Antihepatotoxicity studies of crude extract of Zingiber officinale on CCl4 induced toxicityand comparison of the extract's fraction D hepatoprotective capacity. Res J Med Sci. 2011;5:102–107. [Google Scholar]
- 17.Forouzan S, Bahmani M, Parsaei P, Mohsenzadegan A, Gholami-Ahangaran M, Sadeghi E, et al. Anti-parasitic activites of Zingiber officinale methanolic extract on Limnatis Nilotica. Global Veterinaria. 2012;9:144–148. [Google Scholar]
- 18.Mostafa OM, Eid RA, Adly MA. Antischistosomal activity of ginger (Zingiber officinale) against Schistosoma mansoni harbored in C57BL/6 mice. Parasitology Res. 2011;109:395–403. doi: 10.1007/s00436-011-2267-x. [DOI] [PubMed] [Google Scholar]
- 19.Karuppiah P, Rajaram S. Antibacterial effect of Allium sativum cloves and Zingiber officinale rhizomes against multiple-drug resistant clinical pathogens. Asian Pacific J Tropical Biomed. 2012;2:597–601. doi: 10.1016/S2221-1691(12)60104-X. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Al-Amin ZM, Thomson M, Al-Qattan KK, Peltonen-Shalaby R, Ali M. Anti-diabetic and hypolipidaemic properties of ginger (Zingiber officinale) in streptozotocin-induced diabetic rats. Br J Nutrition. 2006;96:660–666. doi: 10.1079/bjn20061849. [DOI] [PubMed] [Google Scholar]
- 21.Baliga MS, Haniadka R, Pereira MM, Thilakchand KR, Rao S, Arora R. Radioprotective effects of Zingiber officinale (ginger): past, present and future. Food Funct. 2012;3:714–723. doi: 10.1039/c2fo10225k. [DOI] [PubMed] [Google Scholar]
- 22.Nicoll R, Henein MY. Ginger (Zingiber officinale): a hot remedy for cardiovascular disease? Int J Cardiol. 2009;131:408–409. doi: 10.1016/j.ijcard.2007.07.107. [DOI] [PubMed] [Google Scholar]
- 23.Darvishzadeh-Mahani F, Esmaeili-Mahani S, Komeili G, Sheibani V, Zare L. Ginger (Zingiber officinale) prevents the development of morphine analgesic tolerance and physical dependence in rats. J Ethnopharmacol. 2012;141:901–907. doi: 10.1016/j.jep.2012.03.030. [DOI] [PubMed] [Google Scholar]
- 24.Agrawal AK, Rao CV, Sairam K, Joshi VK, Goel RK. Effect of Piper longum Linn Zingiber officinale Linn and Ferula species on gastric ulceration and secretion in rats. Indian J Exp Biol. 2000;38:994–998. [PubMed] [Google Scholar]
- 25.Ghasemzadeh A, Jaafar HZ, Rahmat A. Antioxidant activities, total phenolics and flavonoids content in two varieties of Malaysia young ginger (Zingiber officinale Roscoe) Molecules. 2010;15:4324–4333. doi: 10.3390/molecules15064324. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Asl MN, Hosseinzadeh H. Review of pharmacological effects of Glycyrrhiza sp. and its bioactive compounds. Phytother Res. 2008;22:709–724. doi: 10.1002/ptr.2362. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Nakagawa K, Kishida H, Arai N, Nishiyama T, Mae T. Licorice flavonoids suppress abdominal fat accumulation and increase in blood glucose level in obese diabetic KK-A(y) mice. Biol Pharm Bull. 2004;27:1775–1778. doi: 10.1248/bpb.27.1775. [DOI] [PubMed] [Google Scholar]
- 28.Lim WY, Chia YY, Liong SY, Ton SH, Kadir KA, Husain SN. Lipoprotein lipase expression, serum lipid and tissue lipid deposition in orally administered glycyrrhizic acid reated rats. Lipids Health Dis. 2009;8:31. doi: 10.1186/1476-511X-8-31. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Wittschier N, Faller G, Hensel A. Aqueous extracts and polysaccharides from liquorice roots (Glycyrrhiza glabra L.) inhibit adhesion of helicobacter pylori to human gastric mucosa. J Ethnopharmacol. 2009;125:218–223. doi: 10.1016/j.jep.2009.07.009. [DOI] [PubMed] [Google Scholar]
- 30.Racková L, Jancinová V, Petríková M, Drábiková K, Noslál’ R, Stefek M, et al. Mechanism of anti-inflammatory action of liquorice extract and glycyrrhizin. Nat Prod Res. 2007;21:1234–1241. doi: 10.1080/14786410701371280. [DOI] [PubMed] [Google Scholar]
- 31.Bafna PA, Balaraman R. Antioxidant activity of DHC-1, an herbal formulation, in experimentally-induced cardiac and renal damage. Phytother Res. 2005;19:216–221. doi: 10.1002/ptr.1659. [DOI] [PubMed] [Google Scholar]
- 32.Visavadiya NP, Soni B, Dalwadi N. Evaluation of antioxidant and antiatherogenic properties of Glycyrrhiza glabra root using in vitro models. Int J Food Sci Nutr. 2009;60:135–149. doi: 10.1080/09637480902877998. [DOI] [PubMed] [Google Scholar]
- 33.Ojha Sh, Golechha M, Kumari S, Bhatia J, Arya DS. Glycyrrhiza glabra protects from myocardial ischemia–reperfusion injury by improving hemodynamic, biochemical, histopathological and ventricular function. Exp Toxicol Pathol. 2013;65:219–227. doi: 10.1016/j.etp.2011.08.011. [DOI] [PubMed] [Google Scholar]
- 34.Choi HJ, Seon MR, Lim SS, Kim JS, Chun HS, Park JHY. Hexane/ethanol extract of Glycyrrhiza uralensis licorice suppresses doxorubicin-induced apoptosis in H9C2 rat cardiac myoblasts. Exp Biol Med (Maywood) 2008;233:1554–1560. doi: 10.3181/0807-RM-221. [DOI] [PubMed] [Google Scholar]
- 35.Zhang L, Yang Y, Yu L, Wang Y, Liu L, Fan X. Cardioprotective effects of Glycyrrhiza uralensis extract against doxorubicin-induced toxicity. Int J Toxicol. 2011;30:181–189. doi: 10.1177/1091581810393033. [DOI] [PubMed] [Google Scholar]
- 36.Parisella ML, Angelone T, Gattuso A, Cerra MC, Pellegrino D. Glycyrrhizin and glycyrrhetinic acid directly modulate rat cardiac performance. J Nutrition Biochem. 2012;23:69–75. doi: 10.1016/j.jnutbio.2010.10.011. [DOI] [PubMed] [Google Scholar]
- 37.Sadeghnia HR, Yousefsani BS, Rashidfar M, Boroushaki MT, Assadpour E, Ghorbani A. Protective effect of rutin on hexachlorobutadiene-induced nephrotoxicity. Ren Fail. 2013;35:1151–1155. doi: 10.3109/0886022X.2013.815546. [DOI] [PubMed] [Google Scholar]
- 38.Mortazavian SM, Ghorbani A, Hesari TG. Effect of hydro-alcoholic extracts of viola tricolor and its fractions on proliferation of cervix carcinoma cells. Iran J Obstet Gynecol Infertil. 2012;15:9–16. [Google Scholar]
- 39.Tsutsui H, Kinugawa S, Matsushima S. Oxidative stress and heart failure. Am J Physiol Heart Circ Physiol. 2011;301:H2181–H2190. doi: 10.1152/ajpheart.00554.2011. [DOI] [PubMed] [Google Scholar]
- 40.Li H, Horke S, Forstermann U. Oxidative stress in vascular disease and its pharmacological prevention. Trends in Pharmacol Sci. 2013;34:313–319. doi: 10.1016/j.tips.2013.03.007. [DOI] [PubMed] [Google Scholar]
- 41.Ahmed Z, Tang WH. Pharmacologic strategies to target oxidative stress in heart failure. Curr Heart Fail Rep. 2012;9:14–22. doi: 10.1007/s11897-011-0081-5. [DOI] [PubMed] [Google Scholar]
- 42.Kang YJ, Chen Y, Epstein PN. Suppression of doxorubicin cardio toxicity by overexpression of catalase in the heart of transgenic mice. J Biol Chem. 1996;271:12610–12616. doi: 10.1074/jbc.271.21.12610. [DOI] [PubMed] [Google Scholar]
- 43.Sheng R, Gu ZL, Xie ML, Zhou WX, Guo CY. Epigallocatechin gallate protects H9c2 cardiomyoblasts against hydrogen dioxides-induced apoptosis and telomere attrition. Eur J Pharmacol. 2010;641:199–206. doi: 10.1016/j.ejphar.2010.05.054. [DOI] [PubMed] [Google Scholar]
- 44.Winstead MW, Lucas KK, Dennis EA. Group IV cytosolic phospholipase A2 mediates arachidonic acid release in H9c2 rat cardiomyocyte cells in response to hydrogen peroxide. Prostaglandins Other Lipid Mediat. 2005;78:55–66. doi: 10.1016/j.prostaglandins.2005.03.004. [DOI] [PubMed] [Google Scholar]
- 45.El-Daly ES. Protective effect of cysteine and vitamin E, Crocus sativus and Nigella sativa extracts on cisplatin-induced toxicity in rats. J Pharmacol (Belgium) 1998;53:87–93. [PubMed] [Google Scholar]
- 46.El-Bakly WM, Louka ML, El-Halawany AM, Schaalan MF. 6-gingerol ameliorated doxorubicin-induced cardio toxicity: role of nuclear factor kappa B and protein glycation. Cancer Chemother Pharmacol. 2012;70:833–841. doi: 10.1007/s00280-012-1975-y. [DOI] [PubMed] [Google Scholar]
- 47.Hibasami H, Iwase H, Yoshioka K, Takahashi H. Glycyrrhizin induces apoptosis in human stomach cancer KATO III and human promyelotic leukemia HL-60 cells. Int J Mol Med. 2005;16:233–236. [PubMed] [Google Scholar]
- 48.Hibasami H, Iwase H, Yoshioka K, Takahashi H. Glycyrrhetinic acid (a metabolic substance and aglycon of glycyrrhizin) induces apoptosis in human hepatoma, promyelotic leukemia and stomach cancer cells. Int J Mol Med. 2006;17:215–219. [PubMed] [Google Scholar]
- 49.Lee CS, Kim YJ, Lee MS, Hana ES, Lee SJ. 18β-Glycyrrhetinic acid induces apoptotic cell death in SiHa cells and exhibits a synergistic effect against antibiotic anti-cancer drug toxicity. Life Sci. 2008;83:481–489. doi: 10.1016/j.lfs.2008.07.014. [DOI] [PubMed] [Google Scholar]
- 50.Shukla Y, Singh M. Cancer preventive properties of ginger: a brief review. Food Chem Toxicol. 2007;45:683–690. doi: 10.1016/j.fct.2006.11.002. [DOI] [PubMed] [Google Scholar]
- 51.Jagetia GC, Reddy TK, Malagi KJ, Nayak BS, Naidu MBR, Ravikiran PB, et al. Antarth, a polyhedral preparation protects against the doxorubin-induced toxicity without compromising its antineoplastic activity. Phytother Res. 2005;19:772–778. doi: 10.1002/ptr.1713. [DOI] [PubMed] [Google Scholar]
- 52.Liu L, Zhou HY, Ran K, Wang JB. Glycyrrhiznatis ameliorates rabbit myocardial ischemia-reperfusion injury through P38MAPK pathway. Nan Fang Yi Ke Da Xue Xue Bao. 2010;30:298–300. [PubMed] [Google Scholar]