Attenuation effect of Abnormal Savda Munziq on liver and heart toxicity caused by chemotherapy in mice

  • Authors:
    • Ainiwaer Aikemu
    • Nurmuhamat Amat
    • Abdiryim Yusup
    • Lianlian Shan
    • Xinwei Qi
    • Halmurat Upur
  • View Affiliations

  • Published online on: May 10, 2016     https://doi.org/10.3892/etm.2016.3328
  • Pages: 384-390
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Abnormal Savda Munziq (ASMq), an Uighur medicine formula commonly used in the treatment of cancer, has been speculated to possess antioxidative and antiproliferative effects, and to regulate immune activity. The present study was designed to systematically elucidate the toxicity-reducing activity of ASMq in mice undergoing combination chemotherapy with doxorubicin and 5-fluorouracil (5-FU). The mice were divided into normal (saline, 10 ml/kg) and doxorubicin + 5‑FU groups (doxorubicin, 2.5 mg/kg; 5‑FU, 10 mg/kg on alternate days). In addition, three groups received different doses of ASMq (2, 4 and 8 g/kg), in addition to doxorubicin (2.5 mg/kg) and 5‑FU (10 mg/kg) treatment on alternate days. The histology of the heart and liver, superoxide dismutase (SOD), glutathione peroxidase (GSH‑Px) activity, malondialdehyde (MDA) concentrations in heart homogenate, and various biochemical parameters of the liver were evaluated. Compared with the normal control group, ASMq dose‑dependently improved a number of variables, including body weight, liver index, transaminase and total protein, and partially normalized liver and cardiac pathology. ASMq restored activities of defense antioxidant enzymes SOD and GSH‑Px towards normal levels, and decreased MDA concentration in dose‑dependent manner. These results demonstrated that ASMq provides significant protection against doxorubicin + 5‑FU combination induced hepatotoxicity and cardiotoxicity. Further studies are required to determine the effects of ASMq against doxorubicin + 5‑FU-induced toxicity during chemotherapy in vivo.

Introduction

The radiotherapy and chemotherapy are the primary treatment options for the majority of types of cancer (1). However, a variety of side effects are associated with chemotherapeutic drugs, including immunosuppression, discomfort of the gastrointestinal tract, vomiting and inappetence, which may substantially impact patient health and quality of life (2). The majority of the anticancer drugs currently used in chemotherapy are cytotoxic to normal cells, leading to unwanted side effects (3). Therapeutically effective doses of numerous types of anticancer drug may produce irreversible changes in normal tissues (4). Therefore, there is a required for cancer treatments which are able to reduce the harmful side effects of anticancer drugs in normal tissues. Combining herbal medicinal herbs with chemotherapy may improve quality of life, tumor response and performance status, as well as reduce the toxicity of chemotherapy (5). At present, certain medicinal herbs have already attracted a attention due to their low toxicity and purported curative effects.

Doxorubicin (Adriamycin®) is a widely used anthraquinone anticancer drug, with significant dose-limiting cardiac toxicity (68). In combination with other anticancer drugs, it is used as first line therapy in malignant lymphoma, sarcomas, cancer of the breast, lung, bladder and various other cancer types (9). 5-Fluorouracil (5-FU), a thymidylate synthetase enzyme inhibitor, is metabolized intracellularly to 5-FU deoxynucleotide (5F-dUMP), which inhibits deoxythymidylic acid synthetase, then prevents deoxyuridylic acid (dUMP) from methylating to deoxythymidine monophosphate (dTMP), which affects DNA synthesis (10). 5-FU is effective against cancers of the digestive system (esophageal, stomach, intestinal cancer, carcinoma of pancreas, liver cancer) and breast cancer (11). Furthermore, 5-FU may be effective in treating cancer of the cervix, ovaries, bladder, head and neck, in addition to chorioepithelioma (11). Clinically doxorubicin and 5-FU (AF) are often combined; however, their use is limited by cardiac and liver toxicity (12).

Abnormal Savda Munziq (ASMq) is an Uighur medicinal herbal preparation that is widely used in the Xinjiang region of China (13). ASMq consists of ten medicinal herbs: Adiantum capillus-veneris L. Alhagi pseudalhagi (Bieb.) Desv., Anchusa italica Retz., Cordia dichotoma G. Forst., Euphorbia maculata L., Foeniculum vulgare Mill., Glycyrrhiza glabra L., Lavandula angustifolia Mill., Melissa officinalis L., and Ziziphus jujuba Mill (14). ASMq has been applied to the prevention and treatment of numerous chronic diseases, including cancer, hypertension, diabetes mellitus and memory dysfunction (15). ASMq has been used as a traditional remedy for the prevention or treatment of digestive cancer (16).

Previous pharmacological and clinical studies have demonstrated that ASMq exhibits antioxidant effects, reducing oxidative damage by free radicals (17), anti-DNA oxidative damage, and inhibiting radiation-induced damage in mice (1821). In addition, ASMq has been shown to modulate cellular and humoral immunity in a combined stress mouse model, protecting mitochondria and DNA against damage induced by OH in a cell-free system (17,22). Furthermore, ASMq is able to inhibit cancer cell proliferation and viability in vitro (15,23,24), and has exhibited anti-tumor properties in vitro (15,2224) and in rats (25). However, its potential protective effects against toxicity induced by doxorubicin and 5-FU have not been systematically evaluated. Therefore, the aim of the present study was to investigate the protective effect of ASMq against doxorubicin- and 5-FU-induced toxicity in mice.

Materials and methods

Chemicals and reagents

ASMq was provided by Qikang Habo pharmaceutical Co., Ltd. (Xinjiang, China). Doxorubicin hydrochloride for injection (adriamycin) was purchased from Pfizer Italia Srl Co., Ltd. (Rome, Italy). 5-FU was purchased from JinYao Amino Acid Co., Ltd. (batch no. 0912302; Tianjin, China). Kits for determining serum aspartate aminotransferase (AST) and alanine aminotransferase (ALT) activities, and total protein (TP), were obtained from the Xiamen Jiaxing Biotechnology Co., Ltd. (Xiamen, China; cat. nos. C0010-2, C009-2 and A045-2, respectively). Assay kits used for determination of malondialdehyde (MDA) content, superoxide dismutase (SOD) activity and glutathione peroxidase (GSH-Px) activity were obtained from the Nanjing Jiancheng Institute of Biological Engineering Institute (Nanjing, China; cat. nos. 201304010, 201004010 and 201304010, respectively).

Animals and treatment

A total of 50 Kunming mice (age, 4–6 weeks; weight, 20±2 g), including 25 male and 25 female mice, were supplied by the Experimental Animal Centre of Xinjiang Medical University (Urumqi, China). The present study was approved by the Ethics Committee of the Xinjiang Medical University. Mice were housed in plastic cages at room temperature (22±1°C) under a 12-h light/dark cycle and provided with rodent chow and water ad libitum. A total of 50 mice were randomly divided into five groups (n=10 per group): i) Normal control, orally received saline 0.2 ml/10 g for 14 days, then intraperitoneally injected with 0.4 ml/10 g body weight saline (normal group); ii) doxorubicin + 5-FU toxicity control mice were intraperitoneally injected with doxorubicin (2.5 mg/kg) and 5-FU (10 mg/kg) once in two days (doxorubicin + 5-FU group); and iii-v) three groups of animals were treated with the 2, 4 or 8 g/kg ASMq per day (ASMq.L, ASMq.M and ASMq.H groups, respectively) for 14 days, then intraperitoneally administered doxorubicin (2.5 mg/kg) and 5-FU (10 mg/kg) once in two days (in 0.4 ml/10 g saline). Mice were weighed and sacrificed by cervical dislocation following intraperitoneal injection with 10% chloral hydrate (4 ml/kg; Tianjin Fucheng Chemical Reagents Factory, Tianjin, China). Blood and kidney, spleen, liver and heart tissue samples were collected. Serum was separated for the hematological and biochemical assays. Spleen and body weights were also measured, and the heart, liver, kidney and spleen index was calculated as organ weight divided by body weight. Subsequently, heart, liver, kidney and spleen tissues were fixed in 10% buffered formalin (Sichuan Xilong Chemical Industry Co., Ltd., Chengdu, China) for histopathological analysis. The experiments were performed in accordance with local institutional and governmental regulations on the use of experimental animals.

Biochemical determinations

Serum biochemical markers of hepatic injury ALT, AST and TP were assayed using commercial kits. The activities of AST and ALT are expressed as an international unit (U/l).

Measurement of SOD, MDA and GSH-Px levels in heart homogenate

Heart samples were homogenized in Tris-HCl buffer (5 mM, containing 2 mM EDTA; pH 7.4) resulting in 10% (w/v) liver homogenate. Homogenates were then centrifuged at 191 × g for 10 min at 4°C and the supernatants were used immediately for the determination of antioxidant status. Activities of SOD and GSH-Px, as well as the level of MDA, as an index of the extent of lipid peroxidation in liver tissue, were determined using commercial kits, according to the manufacturer's instructions. All samples were assayed in triplicate. The content of MDA is expressed in nmol, whereas SOD and GSH-Px activities are expressed as U/mg protein. The protein content of the homogenates was determined using a standard commercial kit (Nanjing Jiancheng Institute of Biological Engineering Institute; cat. no. 20100420).

Histological investigation

After removal, samples of heart and liver tissue were fixed in 10% buffered formalin. Samples were embedded in paraffin and at least four 4–5 µm sections were produced from each heart and liver and stained with hematoxylin and eosin (Beijing SUOLAIBAO Technology, Co., Ltd., Beijing, China). Two changes (2 min xylene treatment each) were performed, and finally tissue sections were mounted with DPX. The slides were observed for histopathological changes and microphotographs were captured using a BX50 microscope system (Olympus Corporation, Tokyo, Japan).

Statistical analysis

All data are presented as the mean ± standard error. Statistical analyses were performed using SPSS 17.0 software (SPSS, Inc., Chicago, IL, USA). Differences between groups were assessed using Student's t-test and general linear model univariate analysis of variance. P<0.05 was considered to indicate a statistically significant difference.

Results

Effects of ASMq on heart, liver, kidney and spleen indices

The body weight and heart, liver, kidney and spleen weights were examined on the day of sacrifice, and organ indices were calculated using the ratio of body weight to organ weight (Tables I and II). Doxorubicin + 5-FU treatment significantly reduced mouse body weight compared with the normal group (P<0.05). In addition, treatment with low-, intermediate-, and high-dose ASMq did not significantly increase the heart, liver or kidney weights, as compared with the doxorubicin + 5-FU treatment group (P>0.05). However, there was a significant difference in the spleen and liver indices of the ASMq-treated groups, as compared with the doxorubicin + 5-FU treatment group (P<0.05), thus suggesting that ASMq showed a little therapeutic effects to spleen and liver shrinking during doxorubicin and 5-FU treatment. ASMq can restore the liver and spleen indices proportionately, but it decreases kidney and heart indexes compared with doxorubicin and 5-FU treatment, its reason still unknown (Tables I and II).

Table I.

Effect of ASMq on body and organ weights of in mice (x̅±S).

Table I.

Effect of ASMq on body and organ weights of in mice (x̅±S).

GroupLeft kidney (g)Heart (g)Spleen (g)Liver (g)Body (g)
Normal0.218±0.0610.188±0.0370.113±0.0211.718±0.325   34.3±3.17
A + 5-FU   0.107±0.015a   0.095±0.019a   0.018±0.006a   0.673±0.104a   17.61±0.85a
ASMq.H0.108±0.0130.097±0.0210.026±0.010   0.853±0.109b   19.22±1.66b
ASMq.M0.103±0.0100.095±0.0080.025±0.007   0.948±0.154b   19.15±0.96b
ASMq.L0.098±0.0090.081±0.0140.017±0.0060.673±0.08518.57±1.17

{ label (or @symbol) needed for fn[@id='tfn1-etm-0-0-3328'] } Values are presented as the mean ± standard error.

a P<0.05 vs. normal group

b P<0.05 vs. A + 5-FU group. ASMq, Abnormal Savda Munziq; A, doxorubicin; 5-FU, 5-fluorouracil.

Table II.

Effect of ASMq on A + 5-FU-induced changes in organ/body weight ratios (x̅±S).

Table II.

Effect of ASMq on A + 5-FU-induced changes in organ/body weight ratios (x̅±S).

GroupKidney/body weight (mg/10 g)Heart/body weight (mg/10 g)Spleen/body weight (mg/10 g)Liver/body weight (mg/10 g)
Normal   63±1355±733±6497±50
A + 5-FU61±8   54±10   10±3a   383±64a
ASMq.H56±650±813±5   443±37b
ASMq.M   54±3b50±5   13±3b   493±62b
ASMq.L   53±3b   44±8b   9±3363±43

{ label (or @symbol) needed for fn[@id='tfn4-etm-0-0-3328'] } Values are presented as the mean ± standard error.

a P<0.05 vs. normal group

b P<0.05 vs. A + 5-FU group. ASMq, Abnormal Savda Munziq; A, doxorubicin; 5-FU, 5-fluorouracil.

ALT, AST and TP concentrations

The concentrations of ALT and AST were significantly increased and the TP level was significantly decreased in the blood of doxorubicin + 5-FU mice compared with normal control mice, indicating the failure of liver function due to doxorubicin + 5-FU-induced hepatotoxicity (Fig. 1) (P<0.05). Treatment with ASMq significantly reduced the levels of ALT and AST (P<0.05). The ASMq.M group was most similar to the normal group. By contrast, a significant increase in TP content (P<0.05) was produced by ASMq treatment as compared to the doxorubicin + 5-FU group (Fig. 1).

Effects of ASMq on heart homogenate SOD activity, GSH-Px and MDA content in doxorubicin + 5-FU mice

As oxidative stress contributes to the development of doxorubicin + 5-FU-induced liver injury, the levels of the antioxidative enzymes SOD and GSH-Px were measured. Levels of SOD and GSH-Px were significantly decreased in the doxorubicin + 5-FU group compared with the normal group (P<0.05) (Fig. 2). Pre-treatment with ASMq significantly increased the SOD and GSH-Px levels as compared with the mice that received doxorubicin + 5-FU treatment. Results showed that the activities of SOD and GSH-Px were significantly increased (P<0.05) by ASMq at the doses of 2, 4 and 8 g/kg (Fig. 2). MDA is an end-product of the breakdown of polyunsaturated fatty acids and related esters, and its formation is an index of lipid peroxidation in numerous organ homogenates (26). Administration with doxorubicin + 5-FU resulted in a significant increase in MDA concentration when compared with the normal group (P<0.05). However, ASMq significantly reduced MDA in the heart homogenate as compared with doxorubicin + 5-FU-intoxicated group (P<0.05) (Fig. 3).

Histopathological analysis of mouse hearts

As shown in Fig. 4, histological analysis of the heart sections of normal group animals showed normal cells with well-preserved cytoplasm, prominent nucleus and nucleolus. Compared to normal group mice, extensive necrosis and mineralization of cardiomyocytes combined with cardiomyocyte vacuolation and myofibril breakage was observed in the doxorubicin + 5-FU treated group. ASMq.M and ASMq.H mice showed evidence of cardiomyocyte pathological changes, although in the absence of cardiac edema, spotty necrosis, heart vacuolar degeneration and abnormal myocardial interstitial changes (Fig. 4).

Histopathological analysis of mouse livers

Liver histopathology was observed to determine the protective effects of ASMq on doxorubicin + 5-FU mice at the cellular level. Normal mice livers showed limpid central vein and hepatic cells with prominent nuclei and uniform cytoplasm. doxorubicin + 5-FU-treated mice liver sections showed vacuolization of hepatocytes, sinusoidal dilation and congestion, infiltration of cells, loss of cell boundaries and ballooning degeneration, loss of architecture and cell necrosis. The histopathological changes were prominent compared to those of mice in normal group. As demonstrated by the liver histopathological observations, ASMq at all doses showed reduced liver structure damage compared with the doxorubicin + 5-FU group (Fig. 5). Treatment with ASMq reduced the level of hepatic lesions induced by doxorubicin + 5-FU. Photomicrographs indicated reduced damage of liver tissue, on account of the absence of focal or bridging necrosis. Doxorubicin + 5-FU-induced hepatic lesions were most notably reversed by ASMq.M and ASMq.H, which appeared to be comparable to the normal group. These histopathological observations further suggest the hepatoprotective potential of ASMq as an anti-cancer agent in vitro (Fig. 5).

Discussion

At present, chemotherapy is an crucial modality in the treatment of cancer and in numerous instances may be the single best agent for treatment (27). However, toxic side effects of chemotherapy drugs may be a major limitation to their effective use, in addition to affecting quality of life (11). A key problem associated with cancer chemotherapy are the severe side effects resulting from normal tissue damage, including the heart, liver and spleen (3). Increasing studies have investigated approaches to improve the sensitivity of tumors to chemotherapeutic drugs and to reduce the associated side effects (27). Herbal drugs may offer an alternative to chemotherapeutic compounds, and have been considered to be non-toxic or less toxic, which has led to studies screen herbal drugs for their protective ability of chemotherapy drug toxicity (28,29).

The development of combination chemotherapy produced new evidence of hepatotoxicity, and more instances can be anticipated in the future (30). Combination chemotherapy uses a number of chemotherapeutic agents, each with a different mechanism of action and toxicity profile. Along with the potential for greater tumor kill, however, the possibility for enhanced toxicity occurs (31). Among combination therapies, 5-FU and doxorubicin (sold under the brand name Adriamycin) is the most prevalent combination (32). 5-FU, a pyrimidine analogue, is a cytotoxic agent which is extensively used in different solid tumors such as breast, lung and gastrointestinal tract cancers (33,34). Doxorubicin, an anthracycline glycoside, is commonly used as chemotherapeutic agent in various malignant disorders; however, the possibility of the adverse effect of irreversible cardiomyopathy limits its use (35). The hepatotoxic effects of anticancer drugs may manifest as symptoms other than liver injury, such as necrosis, steatosis, fibrosis, cholestasis and vascular injury (36). 5-FU has been shown to exhibit hepatotoxic effects, such as increased activity of aminotransferases, lactate dehydrogenase and alkaline phosphatase, which indicates hepatic damage (37).

The present results demonstrated that, following administration of doxorubicin, the levels of AST and ALT were elevated and were associated with idiosyncratic drug reactions, including focal infiltration by inflammatory cells and steatosis on liver biopsies. This was considered an idiosyncratic reaction. Chemotherapeutic agents are extensively metabolized in the liver and decrease the antioxidant capacity of the liver, including decreasing glutathione production, which protects against free radical injury (38). Doxorubicin acts via DNA intercalation, alteration of membrane function and free radical formation (39).

In the present study, injection of doxorubicin and 5-FU in mice resulted in decreased liver index and deterioration of hepatic function, as indicated by elevations in ALT and AST and by a significant reduction in total protein. In addition, the examination of liver function correlated with the histopathological changes observed by photomicroscopy. These results are consistent with the previous reports on chemotherapy induced hepatotoxicity (40,41). In the present study it was shown that ASMq protected the liver tissue against damage induced by doxorubicin and 5-FU combination. This protective effect was indicated by a reduction in serum levels of ALT and AST and by a significant elevation in total protein. However, ASMq showed controversial effects to heart and kidney indices, and these require further investigation. The results of the present study indicated that ASMq is able to exert a protective against doxorubicin and 5-FU induced hepatotoxicity. The histological observations supported the results obtained from biochemical analyses.

The clinical application of doxorubicin is complicated by its potential toxicity in heart tissue (15,42,43). The mechanism by which doxorubicin or its metabolites cause chronic cardiomyopathy is not fully understood. Hypotheses regarding the mechanism underlying this cardiac toxicity include perturbation of calcium homeostasis, formation of iron complexes, generation of radical oxygen species, mitochondrial dysfunction and damage to cell membranes (44). The heart is particularly vulnerable to injury from free radicals as it has reduced levels of protective enzymes, such as superoxide dismutase, compared with other tissues (4547).

The pathophysiology of 5-FU-induced cardiotoxicity is controversial, and conclusions are based on clinical studies and case reports to a greater extent than on experimental evidence (48). Furthermore, 5-FU cardiotoxicity is suspected to be mediated by coronary vasospasm and free radical damage to the myocardium (13,49). Prior studies showed that the administration of doxorubicin caused an increase in MDA levels, as well as reductions in GSH, SOD and glutathione-S-transferase expression in treated rats compared with a control group. Single injection of doxorubicin increased SOD, MDA, NO and xanthine oxidase and myeloperoxidase expression in kidney tissues in rats at 10 days after administration (5052). Previous approaches to reduce doxorubicin or 5-FU related toxicities have centred on the use of antioxidants to minimize the generation of reactive oxygen species.

Lipid peroxidation is an crucial mechanism in the pathogenesis of cell damage (53). MDA is the final product of cell membrane lipid peroxidation, and thus its concentration may reflect the intensity of lipid peroxidation (26). SOD and GSH-Px are indicators of resistance to oxidative processes (54). SOD is able to specifically eliminate free oxygen radicals by transforming surplus oxygen free radicals into hydrogen peroxide, which is transformed by catalase and GSH-Px into water, thus reducing free radical-mediated cell damage (54). Furthermore, GSH-Px is an important antioxidase (1921), and may cause lipid peroxide to change into a alcohol type fatty acid, to complement SOD.

Chemicals such as doxorubicin + 5-FU may induce massive production of free radicals, which accumulate causing cell toxicity and leading to lipid peroxidation of the cell membrane, resulting in cell damage or death (55). The present study found that the administration of doxorubicin and 5-FU was accompanied by signs of oxidative damage, including reduced SOD and GSH-Px activity and increased MDA. These results were consistent with previous studies (56,57), and may be primary to the organ toxicity, or a result of organ toxicity and cell death. ASMq reduced the indicators of oxidative damage, increased SOD and GSH-Px levels to values similar to normal, while partially normalizing the increased levels of MDA. Furthermore, ASMq was found to reverse or oppose the negative effects of doxorubicin + 5-FU on liver function and liver and heart morphology, which was consistent with previous studies (20,21,5860). In the present study, significant differences were detected in the heart homogenate levels of MDA, GSH-Px and SOD between normal groups and doxorubicin + 5-FU treatment groups. ASMq exhibited a significant effect (P<0.05) by increasing levels of SOD, GSH-Px and by reducing MDA levels. However, the histological findings show that of doxorubicin + 5-FU induced a degree of cardiotoxicity. The severity of the histological change was notably reduced in sections from animals treated with ASMq. The most marked effects were observed in the ASMq.M (4 mg/kg) group, with moderate efficacy shown by ASMq.H (8 mg/kg).

Future studies may be warranted involving whole organisms treated with toxic chemotherapy, to assess the capacity of ASMq to reduce the adverse reactions of chemotherapeutics that may be dose- and treatment-limiting. As ASMq also has anti-tumoral effects, it may be useful to monitor the beneficial and toxic effects of this treatment. The presently observed reversal of a number of the signs of toxicity associated with doxorubicin + 5-FU by ASMq are consistent with its known anti-oxidative properties in vitro, and warrant further exploratory in vivo studies.

Acknowledgements

This study was supported by the Ministry of Education Changjiang Scholar and Innovative Team Development Program of China (grant no. IRT0977) and the National Natural Science Foundation of China (grant no. 30260128).

References

1 

Powathil GG, Adamson DJ and Chaplain MA: Towards predicting the response of a solid tumour to chemotherapy and radiotherapy treatments: Clinical insights from a computational model. PLoS Comput Biol. 9:e10031202013. View Article : Google Scholar : PubMed/NCBI

2 

Hirose A, Sato E, Fujii H, Sun B, Nishioka H and Aruoma OI: The influence of active hexose correlated compound (AHCC) on cisplatin-evoked chemotherapeutic and side effects in tumor-bearing mice. Toxicol Appl Pharmacol. 222:152–158. 2007. View Article : Google Scholar : PubMed/NCBI

3 

Dantzer R, Meagher MW and Cleeland CS: Translational approaches to treatment-induced symptoms in cancer patients. Nat Rev Clin Oncol. 9:414–426. 2012. View Article : Google Scholar : PubMed/NCBI

4 

Cleeland CS, Allen JD, Roberts SA, Brell JM, Giralt SA, Khakoo AY, Kirch RA, Kwitkowski VE, Liao Z and Skillings J: Reducing the toxicity of cancer therapy: Recognizing needs, taking action. Nat Rev Clin Oncol. 9:471–478. 2012. View Article : Google Scholar : PubMed/NCBI

5 

Dong J, Su S, Wang M and Zhan Z: Shenqi fuzheng, an injection concocted from Chinese medicinal herbs, combined with platinum-based chemotherapy for advanced non-small cell lung cancer: A systematic review. J Exp Clin Cancer Res. 29:1372010. View Article : Google Scholar : PubMed/NCBI

6 

Arcamone F, Penco S and Vigevani A: Adriamycin (NSC-123127): New chemical developments and analogs. Cancer Chemother. 6:123–129. 1975.

7 

Huang CH and Chiang CP: Bladder instillation of adriamycin in the treatment of bladder cancer. Cancer Chemother Pharmacol. 11(Suppl): S91–S93. 1983.PubMed/NCBI

8 

Matsumura Y, Tsushima T, Ozaki Y, Yoshimoto J, Akagi T, Obama T, Nash Y and Ohmori H: Intravesical chemotherapy with 4′-epi-Adriamycin in patients with superficial bladder tumors. Cancer Chemother Pharmacol. 16:176–177. 1986. View Article : Google Scholar : PubMed/NCBI

9 

Maksimenko A, Dosio F, Mougin J, Ferrero A, Wack S, Reddy LH, Weyn AA, Lepeltier E, Bourgaux C, Stella B, et al: A unique squalenoylated and nonpegylated doxorubicin nanomedicine with systemic long-circulating properties and anticancer activity. Proc Natl Acad Sci USA. 111:E217–E226. 2014. View Article : Google Scholar : PubMed/NCBI

10 

Longley DB, Harkin DP and Johnston PG: 5-fluorouracil: Mechanisms of action and clinical strategies. Nat Rev Cancer. 3:330–338. 2003. View Article : Google Scholar : PubMed/NCBI

11 

Nair KL, Jagadeeshan S, Nair SA and Kumar GS: Biological evaluation of 5-fluorouracil nanoparticles for cancer chemotherapy and its dependence on the carrier, PLGA. Int J Nanomedicine. 6:1685–1697. 2011.PubMed/NCBI

12 

Mitchell MS and Deconti RC: Immunosuppression by 5-fluorouracil. Cancer. 26:884–889. 1970. View Article : Google Scholar : PubMed/NCBI

13 

Upur H, Yusup A, Umar A and Moore N: Uighur traditional medicine syndrome of Abnormal Savda in men is associated with oxidative stress, which can be improved by Munziq and Mushil of Abnormal Savda. Therapie. 59:483–484. 2004. View Article : Google Scholar : PubMed/NCBI

14 

Yusup A, Upur H, Umar A, Berke B, Yimit D, Lapham JC, Moore N and Cassand P: Abnormal Savda Munziq, an Herbal Preparation of Traditional Uighur Medicine, May Prevent 1,2-dimethylhydrazine-Induced Rat Colon Carcinogenesis. Evid Based Complement Alternat Med. 2011:1520152011. View Article : Google Scholar : PubMed/NCBI

15 

Yusup A, Upur H, Baudrimont I, Umar A, Kader T, Begaud B, Creppy EE and Moore N: Cytotoxicity of abnormal Savda Munziq aqueous extract in human hepatoma (HepG2) cells. Fundam Clin Pharmacol. 19:465–472. 2005. View Article : Google Scholar : PubMed/NCBI

16 

Abliz G, Mijit F, Hua L, Abdixkur G, Ablimit T, Amat N and Upur H: Anti-carcinogenic effects of the phenolic-rich extract from abnormal Savda Munziq in association with its cytotoxicity, apoptosis-inducing properties and telomerase activity in human cervical cancer cells (SiHa). BMC Complement Altern Med. 15:232015. View Article : Google Scholar : PubMed/NCBI

17 

Yusup A, Upur H, Umar A and Moore N: Protective effects of Munziq and Mushil of abnormal Savda to mitochondrial oxidative damage. Fundam Clin Pharmacol. 18:471–476. 2004. View Article : Google Scholar : PubMed/NCBI

18 

Parekh H, Advani S and Chitnis M: Bepridil enhances adriamycin-induced DNA biosynthesis inhibition in human myeloid leukemia cells. Sel Cancer Ther. 6:183–191. 1990. View Article : Google Scholar : PubMed/NCBI

19 

Gupta V, Lahiri S, Sultana S, Tulsawani R and Kumar R: Anti-oxidative effect of Rhodiola imbricata root extract in rats during cold, hypoxia and restraint (C-H-R) exposure and post-stress recovery. Food Chem Toxicol. 48:1019–1025. 2010. View Article : Google Scholar : PubMed/NCBI

20 

Ku SK, Seo BI, Park JH, Park GY, Seo YB, Kim JS, Lee HS and Roh SS: Effect of Lonicerae Flos extracts on reflux esophagitis with antioxidant activity. World J Gastroenterol. 15:4799–4805. 2009. View Article : Google Scholar : PubMed/NCBI

21 

Salo DC, Lin SW, Pacifici RE and Davies KJ: Superoxide dismutase is preferentially degraded by a proteolytic system from red blood cells following oxidative modification by hydrogen peroxide. Free Radic Biol Med. 5:335–339. 1988. View Article : Google Scholar : PubMed/NCBI

22 

Upur H, Yusup A, Baudrimont I, Umar A, Berke B, Yimit D, Lapham JC, Creppy EE and Moore N: Inhibition of cell growth and cellular protein, DNA and RNA synthesis in human hepatoma (HepG2) cells by ethanol extract of abnormal Savda Munziq of traditional Uighur medicine. Evid Based Complement Alternat Med. 2011:2514242011. View Article : Google Scholar : PubMed/NCBI

23 

Yusup A, Upur H, Tursun X, Berke B, Baudrimont I and Moore N: Study on mechanism of abnormal savda munziq flavonoids in induction of apoptosis of Hep2 cells. Zhongguo Zhong Yao Za Zhi. 32:1068–1071. 2007.(In Chinese). PubMed/NCBI

24 

Yusup A, Upur H, Umar A, Berke B and Moore N: Ethanol extract of abnormal Savda Munziq, a herbal preparation of traditional Uighur medicine, inhibits Caco-2 cells proliferation via cell cycle arrest and apoptosis. Evid Based Complement Alternat Med. 2012:9263292012. View Article : Google Scholar : PubMed/NCBI

25 

Yusup A, Upur H, Umar A, Berke B, Yimit D, Lapham JC, Moore N and Cassand P: Abnormal Savda Munziq, an herbal preparation of traditional Uighur medicine, may prevent 1,2-dimethylhydrazine-induced rat colon carcinogenesis. Evid Based Complement Alternat Med. 2011:1520152011. View Article : Google Scholar : PubMed/NCBI

26 

Sim AS, Salonikas C, Naidoo D and Wilcken DE: Improved method for plasma malondialdehyde measurement by high performance liquid chromatography using methyl malondialdehyde as an internal standard. J Chromatogr B Analyt Technol Biomed Life Sci. 758:337–344. 2003. View Article : Google Scholar

27 

Gerber DE and Schiller JH: Maintenance chemotherapy for advanced non-small-cell lung cancer: New life for an old idea. J Clin Oncol. 31:1009–1020. 2013. View Article : Google Scholar : PubMed/NCBI

28 

Hedigan K: Cancer: Herbal medicine reduces chemotherapy toxicity. Nat Rev Drug Discov. 9:7652010. View Article : Google Scholar : PubMed/NCBI

29 

Lam W, Bussom S, Guan F, Jiang Z, Zhang W, Gullen EA, Liu SH and Cheng YC: The four-herb Chinese medicine PHY906 reduces chemotherapy-induced gastrointestinal toxicity. Sci Transl Med. 2:45ra592010. View Article : Google Scholar : PubMed/NCBI

30 

Joensuu H, Holli K, Heikkinen M, Suonio E, Aro AR, Hietanen P and Huovinen R: Combination chemotherapy versus single-agent therapy as first- and second-line treatment in metastatic breast cancer: A prospective randomized trial. J Clin Oncol. 16:3720–3730. 1998.PubMed/NCBI

31 

Floyd J, Mirza I, Sachs B and Perry MC: Hepatotoxicity of chemotherapy. Semin Oncol. 33:50–67. 2006. View Article : Google Scholar : PubMed/NCBI

32 

Jassem J, Pieńkowski T, Płuzańska A, Jelic S, Gorbunova V, Mrsic-Krmpotic Z, Berzins J, Nagykalnai T, Wigler N, Renard J, et al: Doxorubicin and paclitaxel versus fluorouracil, doxorubicin, and cyclophosphamide as first-line therapy for women with metastatic breast cancer: Final results of a randomized phase III multicenter trial. J Clin Oncol. 19:1707–1715. 2001.PubMed/NCBI

33 

Ciccolini J, Mercier C, Blachon MF, Favre R, Durand A and Lacarelle B: A simple and rapid high-performance liquid chromatographic (HPLC) method for 5-fluorouracil (5-FU) assay in plasma and possible detection of patients with impaired dihydropyrimidine dehydrogenase (DPD) activity. J Clin Pharm Ther. 29:307–315. 2004. View Article : Google Scholar : PubMed/NCBI

34 

Compagnon P, Thiberville L, Moore N, Thuillez C and Lacroix C: Simple high-performance liquid chromatographic method for the quantitation of 5-fluorouracil in human plasma. J Chromatogr B Biomed Appl. 677:380–383. 1996. View Article : Google Scholar : PubMed/NCBI

35 

Zhou Q and Chowbay B: Determination of doxorubicin and its metabolites in rat serum and bile by LC: Application to preclinical pharmacokinetic studies. J Pharm Biomed Anal. 30:1063–1074. 2002. View Article : Google Scholar : PubMed/NCBI

36 

Ishak KG and Zimmerman HJ: Morphologic spectrum of drug-induced hepatic disease. Gastroenterol Clin North Am. 24:759–786. 1995.PubMed/NCBI

37 

Ray S, Roy K and Sengupta C: In vitro evaluation of protective effects of ascorbic acid and water extract of Spirulina plantesis (blue green algae) on 5-fluorouracil-induced lipid peroxidation. Acta Pol Pharm. 64:335–344. 2007.PubMed/NCBI

38 

Meredith MJ and Reed DJ: Depletion in vitro of mitochondrial glutathione in rat hepatocytes and enhancement of lipid peroxidation by adriamycin and 1,3-bib(2-chloroethyl)1-nitrosurea (BCNU). Biochem Pharmacol. 32:1383–1388. 1983. View Article : Google Scholar : PubMed/NCBI

39 

Farrell GC: Drug-Induced liver disease. Edinburgh: Churchill Livingstone. 389–412. 1994.

40 

Mikalauskas S, Mikalauskiene L, Bruns H, Nickkholgh A, Hoffmann K, Longerich T, Strupas K, Büchler MW and Schemmer P: Dietary glycine protects from chemotherapy induced hepatotoxicity. Amino Acids. 40:1139–1150. 2011. View Article : Google Scholar : PubMed/NCBI

41 

Sréter I, Kiss A, Cornides A, Vereckei A, Toncsev H and Fehér J: Inhibition of doxorubicin-induced liver toxicity by a new dihydroquinoline type antioxidant. Acta Physiol Hung. 64:431–435. 1984.PubMed/NCBI

42 

Singal PK and Iliskovic N: Doxorubicin-induced cardiomyopathy. N Engl J Med. 339:900–905. 1998. View Article : Google Scholar : PubMed/NCBI

43 

Giri SN, Al-Bayati MA, Du X, Schelegle E, Mohr FC and Margolin SB: Amelioration of doxorubicin-induced cardiac and renal toxicity by pirfenidone in rats. Cancer Chemother Pharmacol. 53:141–150. 2004. View Article : Google Scholar : PubMed/NCBI

44 

Mordente A, Meucci E, Martorana GE, Giardina B and Minotti G: Human heart cytosolic reductases and anthracycline cardiotoxicity. IUBMB Life. 52:83–88. 2001. View Article : Google Scholar : PubMed/NCBI

45 

Doroshow JH, Locker GY and Myers CE: Enzymatic defenses of the mouse heart against reactive oxygen metabolites: Alterations produced by doxorubicin. J Clin Invest. 65:128–135. 1980. View Article : Google Scholar : PubMed/NCBI

46 

Keizer HG, Pinedo HM, Schuurhuis GJ and Joenje H: Doxorubicin (adriamycin): A critical review of free radical-dependent mechanisms of cytotoxicity. Pharmacol Ther. 47:219–231. 1990. View Article : Google Scholar : PubMed/NCBI

47 

Myers C: The role of iron in doxorubicin-induced cardiomyopathy. Semin Oncol. 25(4 Suppl 10): 10–40. 1998.PubMed/NCBI

48 

Polk A, Vistisen K, Vaage-Nilsen M and Nielsen DL: A systematic review of the pathophysiology of 5-fluorouracil-induced cardiotoxicity. BMC Pharmacol Toxicol. 15:472014. View Article : Google Scholar : PubMed/NCBI

49 

Ensley JF, Patel B, Kloner R, Kish JA, Wynne J and al-Sarraf M: The clinical syndrome of 5-fluorouracil cardiotoxicity. Invest New Drugs. 7:101–109. 1989. View Article : Google Scholar : PubMed/NCBI

50 

Liu LL, Li QX, Xia L, Li J and Shao L: Differential effects of dihydropyridine calcium antagonists on doxorubicin-induced nephrotoxicity in rats. Toxicology. 231:81–90. 2007. View Article : Google Scholar : PubMed/NCBI

51 

Yagmurca M, Erdogan H, Iraz M, Songur A, Ucar M and Fadillioglu E: Caffeic acid phenethyl ester as a protective agent against doxorubicin nephrotoxicity in rats. Clin Chim Acta. 348:27–34. 2004. View Article : Google Scholar : PubMed/NCBI

52 

Yilmaz S, Atessahin A, Sahna E, Karahan I and Ozer S: Protective effect of lycopene on adriamycin-induced cardiotoxicity and nephrotoxicity. Toxicology. 218:164–171. 2006. View Article : Google Scholar : PubMed/NCBI

53 

Yin H, Xu L and Porter NA: Free radical lipid peroxidation: Mechanisms and analysis. Chem Rev. 111:5944–5972. 2011. View Article : Google Scholar : PubMed/NCBI

54 

Liu Y, Zhang L and Liang J: Activation of the Nrf2 defense pathway contributes to neuroprotective effects of phloretin on oxidative stress injury after cerebral ischemia/reperfusion in rats. J Neurol Sci. 351:88–92. 2015. View Article : Google Scholar : PubMed/NCBI

55 

El-Sayyad HI, Ismail MF, Shalaby FM, Abou-El-Magd RF, Gaur RL, Fernando A, Raj MHG and Ouhtit A: Histopathological effects of cisplatin, doxorubicin and 5-flurouracil (5-FU) on the liver of male albino rats. Int J Biol Sci. 5:466–473. 2009. View Article : Google Scholar : PubMed/NCBI

56 

Andersen HR, Nielsen JB, Nielsen F and Grandjean P: Antioxidative enzyme activities in human erythrocytes. Clin Chem. 43:562–568. 1997.PubMed/NCBI

57 

Inal ME, Kanbak G and Sunal E: Antioxidant enzyme activities and malondialdehyde levels related to aging. Clin Chim Acta. 305:75–80. 2001. View Article : Google Scholar : PubMed/NCBI

58 

Gupta V, Lahiri SS, Sultana S, Tulsawani R and Kumar R: Anti-oxidative effect of Rhodiola imbricata root extract in rats during cold, hypoxia and restraint (C-H-R) exposure and post-stress recovery. Food Chem Toxicol. 48:1019–1025. 2010. View Article : Google Scholar : PubMed/NCBI

59 

Andersen HR, Nielsen JB, Nielsen F and Grandjean P: Antioxidative enzyme activities in human erythrocytes. Clinical Chem. 43:562–568. 1997.

60 

Inal ME, Kanbak G and Sunal E: Antioxidant enzyme activities and malondialdehyde levels related to aging. Clin Chim Acta. 305:75–80. 2001. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

July-2016
Volume 12 Issue 1

Print ISSN: 1792-0981
Online ISSN:1792-1015

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Aikemu A, Amat N, Yusup A, Shan L, Qi X and Upur H: Attenuation effect of Abnormal Savda Munziq on liver and heart toxicity caused by chemotherapy in mice. Exp Ther Med 12: 384-390, 2016
APA
Aikemu, A., Amat, N., Yusup, A., Shan, L., Qi, X., & Upur, H. (2016). Attenuation effect of Abnormal Savda Munziq on liver and heart toxicity caused by chemotherapy in mice. Experimental and Therapeutic Medicine, 12, 384-390. https://doi.org/10.3892/etm.2016.3328
MLA
Aikemu, A., Amat, N., Yusup, A., Shan, L., Qi, X., Upur, H."Attenuation effect of Abnormal Savda Munziq on liver and heart toxicity caused by chemotherapy in mice". Experimental and Therapeutic Medicine 12.1 (2016): 384-390.
Chicago
Aikemu, A., Amat, N., Yusup, A., Shan, L., Qi, X., Upur, H."Attenuation effect of Abnormal Savda Munziq on liver and heart toxicity caused by chemotherapy in mice". Experimental and Therapeutic Medicine 12, no. 1 (2016): 384-390. https://doi.org/10.3892/etm.2016.3328