Acessibilidade / Reportar erro

Conus vexillum venom induces oxidative stress in Ehrlich's ascites carcinoma cells: an insight into the mechanism of induction

Abstract

Background

It is estimated that venoms of marine cone snails (genus Conus) contain more than 100,000 different small peptides with a wide range of pharmacological and biological actions. Some of these peptides were developed into potential therapeutic agents and as molecular tools to understand biological functions of nervous and cardiovascular systems. In this study we examined the cytotoxic and anticancer properties of the marine vermivorous cone snail Conus vexillum (collected from Hurgada and Sharm El-Shaikh, Red Sea, Egypt) and suggest the possible mechanisms involved. The in vitro cytotoxic effects of Conus venom were assessed against Ehrlich’s ascites carcinoma (EAC) cells.

Results

Conus venom treatment resulted in concentration-dependent cytotoxicity as indicated by a lactate dehydrogenase leakage assay. Apoptotic effects were measured in vivo by measuring levels of reactive oxygen species and oxidative defense agents in albino mice injected with EAC cells. Conus venom (1.25 mg/kg) induced a significant increase ( p  < 0.05) in several oxidative stress biomarkers (lipid peroxidation, protein carbonyl content and reactive nitrogen intermediates) of EAC cells after 3, 6, 9 and 12 hours of venom injection. Conus venom significantly reduced ( p  < 0.05) the activities of oxidative defense enzymes (catalase and superoxide dismutase) as well as the total antioxidant capacity of EAC cells, as evidenced by lowered levels of reduced glutathione.

Conclusions

These results demonstrate the cytotoxic potential of C. vexillum venom by inducing oxidative stress mediated mechanisms in tumor cells and suggest that the venom contains novel molecules with potential anticancer activity.

Conus vexillum venom; Ehrlich’s cells; Oxidative stress; Cancer; Egypt


Background

It is acknowledged that natural products are one of the major sources for drug discovery and used to treat several life-threading diseases including cancer [ 1Mann J. Natural products in cancer chemotherapy: past, present and future. Nat Rev Cancer. 2002;19(2):143–148. doi: 10.1038/nrc723.
https://doi.org/10.1038/nrc723...
]. Venoms of animal species (such as snakes, scorpions, spiders and cone snails) are a combination of unique bioactive molecules that display a plethora of molecular targets and functions. Some of these molecules are currently being developed as candidate or to provide a critical template for the design of others [ 2Lewis RJ, Garcia ML. Therapeutic potential of venom peptides. Nat Rev Drug Discov.2003;19(10):790–802. doi: 10.1038/nrd1197.
https://doi.org/10.1038/nrd1197...
].

Cancer is defined as a disturbance in mechanisms involved in the control of cell growth and division [ 3Renan MJ. How many mutations are required for tumorigenesis? Implications from human cancer data. Mol Carcinog. 1993;19(3):139–146. doi: 10.1002/mc.2940070303.
https://doi.org/10.1002/mc.2940070303...
, 4Wang Z. Roles of K+ channels in regulating tumour cell proliferation and apoptosis.Pflugers Arch. 2004;19(3):274–286. doi: 10.1007/s00424-004-1258-5.
https://doi.org/10.1007/s00424-004-1258-...
]. It is well known that resistance to apoptotic signals is one of the characteristic features of cancer cells [ 5Hanahan D, Weinberg RA. The hallmarks of cancer. Cell. 2000;19(1):57–70. doi: 10.1016/S0092-8674(00)81683-9.
https://doi.org/10.1016/S0092-8674(00)81...
]. Chemotherapy is the usage of conventional antitumor drugs to treat several types of cancerous cells. While chemotherapy can be quite effective in treating advanced or metastatic diseases, it often accompanied with many harmful side-effects through inducing severe damage to normal cells and tissues [ 6Espinosa E, Zamora P, Feliu J, Barón MG. Classification of anticancer drugs – a new system based on therapeutic targets. Cancer Treat Rev. 2003;19(6):515–523. doi: 10.1016/S0305-7372(03)00116-6.
https://doi.org/10.1016/S0305-7372(03)00...
- 8Kalyanaraman B, Joseph J, Kalivendi S, Wang S, Konorev E, Kotamraju S. Doxorubicin-induced apoptosis: implications in cardiotoxicity. Mol Cell Biochem. 2002;19(1–4):119–124. ]. Moreover through particular cellular and molecular changes, cancer cells eventually become not susceptible to chemotherapeutic agents. These changes include up-regulation the enzymes of drug detoxification and drug transporters and alteration in the molecular targets of drugs. Also, drug tolerance may be due to increase the ability of cancer cells to evade apoptosis through suppression apoptotic pathways and improve the machinery of DNA repair [ 9Naumov GN, Towson JL, MacDonald IC, Wilson SM, Bramwell VH, Groom AC. et al. Ineffectiveness of doxorubicin treatment on solitary dormant mammary carcinoma cells or late-developing metastasis. Breast Cancer Res Treat. 2003;19(3):199–206. doi: 10.1023/B:BREA.0000004377.12288.3c.
https://doi.org/10.1023/B:BREA.000000437...
- 11Gatti L, Zunino F. Overview of tumor cell chemoresistance mechanisms. Methods Mol Med. 2005;19:127–148. ]. Thus, development of alternative anticancer drugs with minimal side-effects is urgently required.

The scientific literature on the antitumor activities of snake venoms extends back many decades [ 12Abu-Sinna G, Esmat AY, Al-Zahaby AS, Soliman NA, Ibrahim TM. Fractionation and characterization of Cerastes cerastes cerastes snake venom and the antitumor action of its lethal and non-lethal fractions. Toxicon. 2003;19(2):207–215. doi: 10.1016/S0041-0101(03)00138-7.
https://doi.org/10.1016/S0041-0101(03)00...
, 13El-Refael MF, Sarkar NH. Snake venom inhibits the growth of mouse mammary tumor cells in vitro and in vivo. Toxicon. 2009;19(1):33–41. doi: 10.1016/j.toxicon.2009.03.017.
https://doi.org/10.1016/j.toxicon.2009.0...
]. More recently, peptides with specific antitumor activity have been isolated, such as cystatin [ 14Xie Q, Tang N, Wan R, Qi Y, Lin X, Lin J. Recombinant snake venom cystatin inhibits the growth, invasion and metastasis of B16F10 cells and MHCC97H cells in vitro and in vivo. Toxicon. 2011;19(5):704–711. doi: 10.1016/j.toxicon.2011.02.002.
https://doi.org/10.1016/j.toxicon.2011.0...
]. However the high toxicity of snake venoms often halts their therapeutic properties and in most cases, the cytotoxic doses for malignant and normal cells are the same. In addition to snake venom, anti-cancer efficacy of bee venom has been reported using both in vitro and in vivo approaches [ 15Orsolić N, Sver L, Verstovsek S, Terzić S, Basić I. Inhibition of mammary carcinoma cell proliferation in vitro and tumor growth in vivo by bee venom. Toxicon. 2003;19(7):861–870. doi: 10.1016/S0041-0101(03)00045-X.
https://doi.org/10.1016/S0041-0101(03)00...
, 16Huh JE, Baek YH, Lee MH, Choi DY, Park DS, Lee JD. Bee venom inhibits tumor angiogenesis and metastasis by inhibiting tyrosine phosphorylation of VEGFR-2 in LLC-tumor-bearing mice. Cancer Lett. 2010;19(1):98–110. doi: 10.1016/j.canlet.2009.11.013.
https://doi.org/10.1016/j.canlet.2009.11...
]. Bee venom has been reported to be cytotoxic and induce apoptosis in various tumour cell lines [ 17Chu ST, Cheng HH, Huang CJ, Chang HC, Chi CC, Su HH. et al. Phospholipase A2-independent Ca2+ entry and subsequent apoptosis induced by melittin in human MG63 osteosarcoma cells. Life Sci. 2007;19(4):364–369. doi: 10.1016/j.lfs.2006.09.024.
https://doi.org/10.1016/j.lfs.2006.09.02...
, 18Son DJ, Lee JW, Lee YH, Song HS, Lee CK, Hong JT. Therapeutic application of anti-arthritis, pain-releasing, and anti-cancer effects of bee venom and its constituent compounds. Pharmacol Ther. 2007;19(2):246–270. doi: 10.1016/j.pharmthera.2007.04.004.
https://doi.org/10.1016/j.pharmthera.200...
]. The promotion of apoptosis through several cancer cell death mechanisms is essential for bee venom-induced anticancer effects (and the bee venom peptide, melittin) [ 19Bhoopathi P, Chetty C, Kunigal S, Vanamala SK, Rao JS, Lakka SS. Blockade of tumor growth due to matrix metalloproteinase-9 inhibition is mediated by sequential activation of beta1-integrin, ERK, and NF-kappaB. J Biol Chem. 2008;19(3):1545–1552. ]. Scorpion venom also possess the property of inhibiting growth of various types of cancers [ 20Zargan J, Sajad M, Umar S, Naime M, Ali S, Khan H. Scorpion (Androctonus crassicauda) venom limits growth of transformed cells (SH-SY5Y and MCF-7) by cytotoxicity and cell cycle arrest. Exp Mol Pathol. 2011;19(1):447–454. doi: 10.1016/j.yexmp.2011.04.008.
https://doi.org/10.1016/j.yexmp.2011.04....
]. Several cytotoxic antitumor peptides have been characterized from scorpion venom, which exert their action by distinct mechanisms: as an ion channel blocker, as a matrix metalloproteinase (MMP-2) inhibitor and as an apoptosis inducer, activating intracellular pathways [ 21Jäger H, Dreker T, Buck A, Giehl K, Gress T, Grissmer S. Blockage of intermediate-conductance Ca2+-activated K+ channels inhibit human pancreatic cancer cell growth in vitro. Mol Pharmacol. 2004;19(3):630–638. doi: 10.1124/mol.65.3.630.
https://doi.org/10.1124/mol.65.3.630...
- 23Gupta SD, Gomes A, Debnath A, Saha A, Gomes A. Apoptosis induction in human leukemic cells by a novel protein Bengalin, isolated from Indian black scorpion: through mitochondrial pathway and inhibition of heat shock proteins. Chem Biol Interact.2010;19(2):293–303. doi: 10.1016/j.cbi.2009.11.006.
https://doi.org/10.1016/j.cbi.2009.11.00...
].

The venoms of cone snails, a family of widely distributed marine mollusks, contain a large number of small peptides (conotoxins and conopeptides) which have evolved separately in approximately 700 species of specific fish, mollusk and worm-hunting animals [ 24Davis JM, Jones A, Lewis RJ. Remarkable inter- and intraspecies complexity of conotoxins revealed by LC/MS. Peptides. 2009;19(7):11222–11227. ]. It is estimated that more than 50,000 conopeptides exist, but less than 0.1% have been functionally characterized. Those that have been examined are specific for an amazingly diverse set of important pharmacological targets, including voltage gated ion channels (Na, K, Ca), neurotransmitter receptors (acetylcholine, glutamate) and transporters (noradrenaline) [ 2Lewis RJ, Garcia ML. Therapeutic potential of venom peptides. Nat Rev Drug Discov.2003;19(10):790–802. doi: 10.1038/nrd1197.
https://doi.org/10.1038/nrd1197...
]. Their small size, relative ease of synthesis and structural stability also make them important templates for the design of novel drugs and indeed several are presently in clinical trials [ 25Scott DA, Wright CE, Angus JA. Actions of intrathecal ω-conotoxins CVID, GVIA, MVIIA, and morphine in acute and neuropathic pain in the rat. Eur J Pharmacol.2002;19(3):279–286. doi: 10.1016/S0014-2999(02)02247-1.
https://doi.org/10.1016/S0014-2999(02)02...
].

We have recently demonstrated that crude venom of the vermivorous snail C. vexillum caused an array of cytotoxic effects in mammalian systems which were attributed to the venom’s ability to induce oxidative stress [ 26Abdel-Rahman MA, Abbas OA, El-Naggar MS, Abdel-Nabi IM, Strong PN. Intraspecific variation in the venom of the vermivorous cone snail Conus vexillum. Comp Biochem Physiol C Toxicol Pharmacol. 2011;19(4):318–325. doi: 10.1016/j.cbpc.2011.06.019.
https://doi.org/10.1016/j.cbpc.2011.06.0...
]. Venoms collected from different locations in the Red Sea (Hurgada and Sharm El-Shaikh) also showed clear differences in venom potency. The present study was designed to extend our earlier work, by examining the cytotoxic activity and mechanism of action of C. vexillum venom on EAC cells, both in vitro and in vivo . To the best of our knowledge, this is the first study of its kind on this Conus species.

Methods

Venom preparation

Specimens of C. vexillum were collected from two different sites (Hurgada and Sharm El-Shaikh) in the Red Sea, Egypt [ 26Abdel-Rahman MA, Abbas OA, El-Naggar MS, Abdel-Nabi IM, Strong PN. Intraspecific variation in the venom of the vermivorous cone snail Conus vexillum. Comp Biochem Physiol C Toxicol Pharmacol. 2011;19(4):318–325. doi: 10.1016/j.cbpc.2011.06.019.
https://doi.org/10.1016/j.cbpc.2011.06.0...
]. Venom glands were dissected as described previously by Cruz et al . [ 27Cruz LJ, Ramilo CA, Corpuz GP, Olivera BM. Conus peptides: phylogenetic range of biological activity. Biol Bull. 1992;19(1):159–164. doi: 10.2307/1542418.
https://doi.org/10.2307/1542418...
] and crude venom from each location was extracted and lyophilized [ 26Abdel-Rahman MA, Abbas OA, El-Naggar MS, Abdel-Nabi IM, Strong PN. Intraspecific variation in the venom of the vermivorous cone snail Conus vexillum. Comp Biochem Physiol C Toxicol Pharmacol. 2011;19(4):318–325. doi: 10.1016/j.cbpc.2011.06.019.
https://doi.org/10.1016/j.cbpc.2011.06.0...
]. Pooled venoms were stored at −80°C until further use.

Experimental animals and ascites tumor

All animal procedures and experimental protocols were approved by the Research Ethics Committee of Suez Canal University and were carried out in accordance with the Guide for the Care and Use of Laboratory Animals ( http://www.nap.edu/catalog/12910.html ). Adult male Swiss albino mice weighing 20–25 g were purchased from the breeding unit of Theodor Bilharz Research Institute (Giza, Egypt). The animals were maintained under controlled conditions of temperature, humidity and on a 12 hour-light/dark cycle, with free access to standard pellet diet and water. The first inoculum of EAC cell line was purchased from the Department of Tumor Biology, National Cancer Institute, Cairo University. The cell line was inoculated by serial intraperitoneal (IP) passages of 106 cells per mouse. Cells were grown in the peritoneal cavity of mice and transferred every ten days to new animals. Mice were monitored daily for signs of tumor progression, including the amount of abdominal distension and signs of illness and distress. The volume of ascites fluid was determined by needle (18–22 gauge) aspiration. Withdrawal of ascites fluid was performed under aseptic conditions.

Isolation of EAC cells

Based on the method described by Mookerjee et. al. [ 28Mookerjee A, Basu JM, Dutta P, Majumder J, Bhattacharya S. et al. Overcoming drug-resistant in cancer by a newly developed copper chelate through host protective cytokine mediated apoptosis. Clin Cancer Res. 2006;19:4339–4349. doi: 10.1158/1078-0432.CCR-06-0001.
https://doi.org/10.1158/1078-0432.CCR-06...
] peritoneal fluid containing the tumor cells was withdrawn, collected in sterile petri plates and incubated at 37°C for two hours. The cells of macrophage lineage adhered to the bottom of the petri dishes. Since EAC cells do not adhere to synthetic surfaces in vitro , the non-adherent cell population was gently aspirated and washed repeatedly with phosphate buffered saline [ 29Viñuela JE, Rodriguez R, Gil J, Coll J, De La Concha EG, Subiza JL. Antigen shedding vs. development of natural suppressor cells as mechanism of tumor escape in mice bearing Ehrlich tumor. Int J Cancer. 1991;19(1):86–91. doi: 10.1002/ijc.2910470116.
https://doi.org/10.1002/ijc.2910470116...
]. More than 93% of the non-adherent cells were morphologically characterized as EAC cells by Wright staining [ 30Stuehr DJ, Marletta MA. Induction of nitrite/nitrate synthesis in murine macrophages in BCG infection, lymphokines or interferon-γ J Immunol. 1987;19(2):518–525. ] and viability was assessed by Trypan Blue dye exclusion [ 31Tavazzi B, Di Pierro D, Amorini AM, Fazzina G, Tuttobene M, Giardina B. et al. Energy metabolism and lipid peroxidation of human erythrocytes as a function of increased oxidative stress. Eur J Biochem. 2000;19(3):684–689. doi: 10.1046/j.1432-1327.2000.01042.x.
https://doi.org/10.1046/j.1432-1327.2000...
].

Lactate dehydrogenase (LDH) assay

EAC cells (1 × 106 cells/mL) were incubated in a 96-well plate with varying concentrations (10, 20 and 30 μg/mL) of C. vexillum venom for one hour at 37°C. LDH released into the medium was determined spectrophotometrically (340 nm) by measuring the rate of decrease of NAPDH, using commercial kit (Spinreact, Sant Esteve de Bas, Spain). Data was expressed as units/liter (U/L).

Antitumor activity of Conus venom

The antitumor activity of C. vexillum venom from each geographical location was evaluated. Fifty-four mice were inoculated with the EAC cells as previously mentioned. After successful establishment of the tumor (ten days after tumor inoculation), mice were randomly assorted into three groups. A Hurgada-treated group (24 animals) and a Sharm El-Shaikh-treated group (24 animals) were both injected IP with a sublethal dose of C. vexillum venom (1.25 mg/kg) [ 26Abdel-Rahman MA, Abbas OA, El-Naggar MS, Abdel-Nabi IM, Strong PN. Intraspecific variation in the venom of the vermivorous cone snail Conus vexillum. Comp Biochem Physiol C Toxicol Pharmacol. 2011;19(4):318–325. doi: 10.1016/j.cbpc.2011.06.019.
https://doi.org/10.1016/j.cbpc.2011.06.0...
] . A third control group (six animals) was injected with 100 μL of sterile saline.

Ascites fluid containing EAC cells was drawn from the peritoneal cavity of treated and control groups after 3, 6, 9 and 12 hours of venom administration. The following biochemical parameters were measured in the EAC cells of treated and control groups: the level of lipid peroxidation (LPx) and protein carbonyl contents (PCC), glutathione (reduced form) content (GSH), the activities of Cu/Zn-superoxide dismutase (Cu/Zn-SOD), catalase (CAT) and total antioxidant capacity (TAC). The level of nitric oxide (NO) was measured in the supernatant of ascites fluid.

Malondialehyde (MDA) assay

The extent of lipid peroxidation in EAC cells was determined by measuring the production of malondialdehyde, an indicator of oxidative damage and oxidative stress [ 31Tavazzi B, Di Pierro D, Amorini AM, Fazzina G, Tuttobene M, Giardina B. et al. Energy metabolism and lipid peroxidation of human erythrocytes as a function of increased oxidative stress. Eur J Biochem. 2000;19(3):684–689. doi: 10.1046/j.1432-1327.2000.01042.x.
https://doi.org/10.1046/j.1432-1327.2000...
]. Malondialdehyde was determined by incubating samples with thiobarbituric acid and measuring reaction products at 532 nm [ 32Yagi K. Lipid peroxides and human diseases. Chem Phys Lipids. 1978;19(2–4):337–351. ]. 1,1,3,3-tetramethoxypropane [malonaldehyde bis(dimethyl acetal), Sigma-Aldrich, USA] was used as an external standard and the results were expressed as μmoles of MDA/mg.

Protein carbonyl content (PCC)

PCC was quantified using dinitrophenylhydrazide (DNPH) [ 33Reznick AZ, Packer L. Oxidative damage to proteins: spectrophotometric method for carbonyl assay. Methods Enzymol. 1994;19:357–363. ]. Protein was precipitated with an equal volume of 1% trichloroacetic acid (TCA) and the pellet was resuspended in 1 mL of 2,4- dinitrophenylhydrazide (Sigma-Aldrich, USA), 10 mM, dissolved in 2 N HCl. Based on the method described by Fulle et al. [ 34Fulle S, Mecocci P, Fano G, Vecchiet I, Vecchini A. et al. Specific oxidative alterations in vastus lateralis muscle of patients with the diagnosis of chronic fatigue syndrome. Free Radic Biol Med. 2000;19:1252–1259. doi: 10.1016/S0891-5849(00)00419-6.
https://doi.org/10.1016/S0891-5849(00)00...
], separate blanks were prepared by adding 1 mL of 2 N HCl without DNPH. Samples were left at room temperature for one hour in the dark and vortexed every 15 minutes. An equal volume of 20% TCA was added and after centrifugation (12,000 × g, 15 minutes, 4°C), pellets were washed (three times) with 1 mL of ethanol: ethyl acetate (1:1) to remove free DNPH and lipid contaminants. The final pellet was dissolved in 6 M guanidine (1 mL, 1 hour, 37°C shaking water bath). The solution was centrifuged (12,000 × g, 15 minutes) and the carbonyl content (nmol/mg), measured as protein phenylhydrazone derivatives, was determined at 370 nm using an absorption coefficient of 22,000 M−1 Cm1.

Estimation of nitric oxide (NO)

Nitric oxide was measured in the supernatant of ascites fluid by a spectrophotometric method based on the Griess reaction [ 35Green LC, Wagner DA, Glogowski J, Skipper PL, Wishnok JS, Tannenbaum SR. Analysis of nitrate, nitrite and nitrate in biological fluids. Anal Biochem. 1982;19(1):131–138. doi: 10.1016/0003-2697(82)90118-X.
https://doi.org/10.1016/0003-2697(82)901...
]. One hundred microliters of supernatant was mixed with an equal volume of Griess reagent (one part 0.1% naphthylethylendiamine dihydrochloride in distilled water plus one part 1% sulfanilamide in 5% concentrated H3PO4), at room temperature for ten minutes. The absorbance was measured at 540 nm. Sodium nitrite was used as a standard.

Reduced glutathione (GSH)

The content of glutathione in EAC cells of control and treated groups was estimated according to Beutler et al . [ 36Beutler E, Doron O, Kelly BM. Improved method for the determination of blood glutathione. J Lab Clin Med. 1963;19:882–888. ]. Aliquots of 0.2 mL of ascites fluid were added to 1.8 mL distilled water and 3 mL of precipitating solution (1.67 gm glacial metaphosphoric acid, 0.2 gm EDTA and 30 gm NaCl in 100 mL distilled water) and the mixture was centrifuged (2200 × g, 15 minutes, 4°C). To 1 mL of supernatant it was added sequentially, sodium dihydrogen phosphate (4 mL, 0.3 M) and DTNB reagent [0.5 mL, 40 mg 5,5` dithiobis-2-nitrobenzoic acid (Sigma-Aldrich, USA) dissolved in 100 mL 1% sodium citrate] and the absorbance was measured at 412 nm. Reduced glutathione (Sigma-Aldrich, USA) was used as standard.

Cu/Zn-superoxide dismutase (Cu/Zn-SOD) and catalase (CAT)

Superoxide dismutase (Cu/Zn SOD; EC 1.15.1.1) activity was estimated in the lysate of EAC cells according to Misra and Fridovich [ 37Misra HP, Fridovich I. The role of superoxide anion in the autoxidation of epinephrine and a simple assay for superoxide dismutase. J Biol Chem. 1972;19(10):3170–3175. ]. The rate of inhibition of auto oxidation was monitored at 560 nm; the amount of enzyme required to produce 50% inhibition is defined as one unit of enzyme activity. SOD activity was expressed as units/mL. Catalase (CAT; EC 1.11.1.6) activity was determined using the method of Aebi [ 38Aebi H. In: Oxygen radicals in biological systems: Meths Enzymol. Packer L, editor. Orlando: Academic; 1984. Catalase in vitro; pp. 121–126. ]. Samples were prepared in phosphate buffer (50 mM, pH 7) and Triton-X 100 (1%, v/v) was added to increase the observable CAT activity by releasing the enzyme from peroxisomes [ 39Cohen G, Dembiec D, Marcus J. Measurement of catalase activity in tissue extracts. Anal Biochem. 1970;19:30–38. doi: 10.1016/0003-2697(70)90083-7.
https://doi.org/10.1016/0003-2697(70)900...
]. CAT was measured by monitoring the decomposition of H2O2 at 240 nm. The enzyme activity was calculated using a molar extinction coefficient of 43.6 mol.

Total antioxidant capacity (TAC)

The total antioxidant capacity in EAC cell lysates was determined according to Koracevic et al . [ 40Koracevic D, Koracevic G, Djordjevic V, Andrejevic S, Cosic V. Method for the measurement of antioxidant activity in human fluids. J Clin Pathol. 2001;19(5):356–361. doi: 10.1136/jcp.54.5.356.
https://doi.org/10.1136/jcp.54.5.356...
]. As in our previous work [ 26Abdel-Rahman MA, Abbas OA, El-Naggar MS, Abdel-Nabi IM, Strong PN. Intraspecific variation in the venom of the vermivorous cone snail Conus vexillum. Comp Biochem Physiol C Toxicol Pharmacol. 2011;19(4):318–325. doi: 10.1016/j.cbpc.2011.06.019.
https://doi.org/10.1016/j.cbpc.2011.06.0...
], the ability of antioxidants to inhibit the H2O2-induced oxidation of 2,2’-azinobis (3-ethylbenzothiazoline-6-sulphonate; ABTS) was measured spectrophotometrically by the reduction in concentration of the cation radical ABTS+, absorbing at 600 nm and expressed as mmol/L.

Statistical analysis

SPSS® statistical software (v. 17.01 SPSS Inc., USA) was used in all data analyses [ 41Dancey CP, Reidy J. Statistics without maths for psychology. Harlow: Pearson Education; 2002. ]. Descriptive analyses including mean and standard error were applied to all biochemical measurements. Differences in the effects of Conus venom between control and treated groups were assessed using the Student's unpaired t -test [ 42Snedecor GW. Statistical Methods. 4. Iowa: Iowa College Press; 1956. ]. One-way analysis of variance (ANOVA) followed by a Dunnett post hoc test was performed to evaluate eventual significant differences in the biochemical measurements between control and treated groups.

Results

In vitro cytotoxicity of Conus venom

The membrane integrity of EAC cells treated with C. vexillum venom, obtained from either Hurgada or Sharm El-Shaikh, was evaluated by measuring the levels of LDH activity in cell culture supernatants. LDH levels in cells incubated with Conus venom for one hour from each location showed a significant ( p  < 0.05) dose-dependent increase in comparison to the levels in vehicle-treated cells (Table  1 ). At doses of 10, 20 and 30 μg/mL of Hurgada venom, release of LDH increased to 48.9, 131.7 and 196.7% respectively, with respect to the control. For Sharm El-Shaikh venom, analogous values were 43.6, 107.5 and 171.9%, with respect to the control. Over the course of the experiment, Hurgada venom was more potent than Sharm El-Shaikh venom in increasing LDH levels and was statistically significant ( p  < 0.05) at ≥ 20 μg/mL venom.

Table 1

Changes in LDH levels of EAC cells incubated with Conus vexillum venom collected from Hurgada and Sharm El-Shaikh (Red Sea, Egypt)


In vivo cytotoxic effects of Conus venom on EAC cells

There are several assays available to measure oxidative stress. One such indicator is the extent of lipid peroxidation (LPx) as measured by thiobarbituric acid derivatives of key metabolic markers such as malondialdehyde (MDA). Protein carbonylation (PCC) is generally recognized as a key step in the production of oxidized proteins and the conversion of NO to nitrite and nitrate is typical metabolic event in oxygenated solutions. The results in Figure  1 illustrate the effects of injection of C. vexillum venom (1.25 mg/kg) from each location into tumor-bearing mice, on the oxidative stress biomarkers (LPx, PCC and NO) at different time intervals (3, 6, 9 and 12 hours).

Figure 1

Conus vexillum venom (1.25 mg/kg) collected from Hurgada and Sharm El-Shaikh induced intracellular oxidative stress in EAC cells assessed by the level of (A) lipid peroxidation, (B) protein carbonyl content and (C) level of nitric oxide. *Significant difference between control (EAC control group) and treated group using Student’s unpaired t -test ( p  < 0.05). #Represents a significant difference between Hurgada and Sharm El-Shaikh groups using Student’s unpaired t -test ( p  < 0.05). £Significant difference between treated groups using one-way ANOVA ( p  < 0.05).


It was observed that LPx (as measured by MDA concentration) was significantly increased ( p  < 0.05) at all time intervals in both Hurgada and Sharm El-Shaikh venoms with respect to control groups (Figure  1 – A). However, levels of MDA increased more rapidly after injection of Hurgada venom (up to 235% increase over control levels after three hours); injection of Sharm El-Shaikh venom took twice as long to produce increased levels of MDA (up to 164% after six hours) which were considerably lower than that produced by Hurgada venom. One-way ANOVA revealed that differences in the concentration of LPx products between treated groups of Sharm El-Shaikh venoms was not as great (F1,3 = 35.86, p  < 0.001) as Hurgada venoms (F1,3 = 111.69, p  < 0.001). Analysis of LPx products revealed significant effects for venom (F2,72 =13.01, p  < 0.05), time (F3,72 = 71.80, p  < 0.001) and the interaction of venom and time (F6,72 = 24.16, p <  0.001) using two-way ANOVA.

The PCCs of EAC cells from mice injected with both venom groups were significantly increased ( p  < 0.05) at all times intervals compared with control values (Figure  1 – B). On average, Hurgada venom was approximately 1.4 times as potent as Sharm El-Shaikh venom in inducing protein oxidation. Using one-way ANOVA, significant differences in PCCs were detected between the treated groups of both Hurgada and Sharm El-Shaikh (F1,3 = 470.04, p  < 0.001 and F1,3 = 221.80, p  < 0.001, respectively). According to the post hoc comparisons, six hours of Hurgada venom treatment produced the maximum increase (+ 501%) in protein oxidation relative to control. Using two-way ANOVA, PCCs revealed significant effects for venom (F2,72 = 663.60, p  < 0.001), time (F3,72 = 214.33, p  < 0.001) and the interaction of venom and time (F6,72 = 45.54, p  < 0.001). Conus venom enhanced production of NO in Hurgada venom-injected animals (Figure  1 – C) and by six hours there was a 61% increase in levels of NO with respect to controls ( p  < 0.05). In comparison, injection of Sharm El-Shaikh venom had a negligible effect (maximum 7%) on NO levels. Differences between Hurgada and Sharm El-Shaikh were statistically significant ( p  < 0.05) up to six hours post injection. Statistical analysis of NO levels revealed significant effects for venom (F2,72 = 491.74, p  < 0.001), time (F3,72 = 77.17, p  < 0.001) and the interaction of venom and time (F6,72 = 42.09, p  < 0.001).

The in vivo effects of injecting Conus venom into EAC-tumor-bearing mice, on the activities of representative oxidative defense enzymes (CAT and Cu/Zn-SOD) was examined, together with the effects on cellular GSH (as a non-enzymatic antioxidant) and the total antioxidant capacity (TAC). The level of cellular GSH was significantly decreased ( p  < 0.05) at all time intervals in mice injected with Hurgada venom, continuing to decline to 46% of control levels at 12 hours (Figure  2 – A). By comparison, in mice injected with Sharm El-Shaikh venom, the levels of GSH declined at a slower rate and began to recover after nine hours (Figure  2 – A). Using bivariate analysis of covariance, cellular GSH brought significant effects for venom (F2,72 = 81.80, p  < 0.001), time (F3,72 = 42.00, p  < 0.001) and the interaction of venom and time (F6,72 = 4.27, p  < 0.05).

Figure 2

Changes in the antioxidant measurements GSH (panel A), CAT (panel B), Cu/Zn SOD (panel C) and TAC (panel D) of EAC cells after administration of Conus vexillum venom (1.25 mg/kg) collected from Hurgada and Sharm El-Shaikh.


There was a significant decrease ( p  < 0.05) in the activities of both CAT and Cu/Zn-SOD in mice injected with either Hurgada and Sharm El-Shaikh venoms at all time intervals as compared with the control groups. The pattern of CAT and SOD levels was the same in both Hurgada and Sharm El-Shaikh venoms, declining to a maximum of approx 56% and 47% respectively in the two locations, at nine hours (Figure  2 – B and C). Interestingly, GSH levels at the different time points mirrored CAT and Cu/Zn SOD levels in the case of Sharm El-Shaikh venom, with a partial recovery after nine hours. Unpaired t -tests showed significant decreases in the level of TAC in EAC cells from both venom groups at all time intervals as compared with the control group ( p  < 0.05). In distinction to the other antioxidant markers, there was a significant decrease in TAC levels in mice injected with Hurgada venom, in the second time period (47% decline from control levels at six hours). In comparison, TAC levels in mice injected with Sharm El-Shaikh venom declined more slowly, although the eventual amount of decline was very similar (51% decline from control levels after nine hours) (Figure  2 – D). Moreover, the activities of oxidative defense enzymes (CAT and SOD) and TAC showed significant effects for venom (F2,72 = 4.09, 0.34 and 2.61, p  < 0.05), time (F3,72 = 33.1, 42.5 and 30.1, p  < 0.001) and the interaction of venom and time (F6,72 = 1.69, 1.7 and 6.1, p  < 0.05), respectively.

Discussion

Venoms from marine cone snails ( Conus ) have received much attention over the last few decades due to their extraordinary complexity and diversity [ 43Safavi-Hemami H, Siero WA, Gorasia DG, Young ND, Macmillan D, Williamson NA. et al. Specialisation of the venom gland proteome in predatory cone snails reveals functional diversification of the conotoxin biosynthetic pathway. J Proteome Res. 2011;19(9):3904–3919. doi: 10.1021/pr1012976.
https://doi.org/10.1021/pr1012976...
]. Each Conus species synthesizes its own characteristic repertoire of ∼ 1100-1900 toxin peptides, and it has been estimated that the toxin library of the entire cone snail genus comprises as many as 500,000 different bioactive compounds [ 24Davis JM, Jones A, Lewis RJ. Remarkable inter- and intraspecies complexity of conotoxins revealed by LC/MS. Peptides. 2009;19(7):11222–11227. , 44Tayo LL, Lu BW, Cruz LJ, Yates JR 3rd. Proteomic analysis provides insights on venom processing in Conus textile. J Proteome Res. 2010;19(5):2292–2301. doi: 10.1021/pr901032r.
https://doi.org/10.1021/pr901032r...
]. The vast majority of these peptides have been shown to target various types of ion channels, both ligand-gated and voltage-gated [ 45Ekberg J, Craik DJ, Adams DJ. Conotoxin modulation of voltage-gated sodium channels.Int J Biochem Cell Biol. 2008;19(1):2363–2368. ]. In comparison, there are only a few reports describing the cytotoxic actions of Conus venoms on eukaryotic cells [ 26Abdel-Rahman MA, Abbas OA, El-Naggar MS, Abdel-Nabi IM, Strong PN. Intraspecific variation in the venom of the vermivorous cone snail Conus vexillum. Comp Biochem Physiol C Toxicol Pharmacol. 2011;19(4):318–325. doi: 10.1016/j.cbpc.2011.06.019.
https://doi.org/10.1016/j.cbpc.2011.06.0...
, 46Saminathan R, Babuji S, Sethupathy S, Viswanathan P, Balasubramanian T, Gopalakrishanakone P. Clinico-toxinological characterization of the acute effects of the venom of the marine snail, Conus loroisii. Acta Trop. 2006;19(1):75–87. doi: 10.1016/j.actatropica.2005.09.001.
https://doi.org/10.1016/j.actatropica.20...
, 47Biggs JS, Rosenfeld Y, Shai Y, Olivera BM. Conolysin-Mt: a conus peptide that disrupts cellular membranes. Biochemistry. 2007;19(44):12586–12593. doi: 10.1021/bi700775p.
https://doi.org/10.1021/bi700775p...
].

The first cytolytic peptide (conolysin-Mt) was isolated from the vermivorous cone snail venom Conus mustelinus [ 47Biggs JS, Rosenfeld Y, Shai Y, Olivera BM. Conolysin-Mt: a conus peptide that disrupts cellular membranes. Biochemistry. 2007;19(44):12586–12593. doi: 10.1021/bi700775p.
https://doi.org/10.1021/bi700775p...
]. The characterization of conolysin expanded the known repertoire of conopeptide mechanisms to include membrane perturbation. Cytolytic peptides, defined by their ability to partially or completely destroy cell membranes, are among the largest group of toxins produced by living organisms, which include bacteria, viruses, insects, scorpions, spiders, reptiles, and marine invertebrates [ 48Alouf JE. Pore-forming bacterial protein toxins: An overview. Curr Top Microbiol Immunol. 2001;19:1–14. doi: 10.1007/978-3-642-56508-3_1.
https://doi.org/10.1007/978-3-642-56508-...
]. By targeting the lipid bilayer of the cell membrane, cytolytic peptides can affect a wide range of biological processes. In our study, an attempt has been made to elucidate the cytotoxic potential of Conus venom (collected from Hurgada and Sharm El-Shaikh) and its underlying mechanism of action, using EAC cells.

The cytotoxic potential of Conus venom from these two geographical sites has been evaluated in vitro against EAC cells by measuring LDH activity. It is well documented that the in vitro release of LDH provides an accurate measure of cell membrane integrity and cell viability. This assay is based on the release of the cytosolic enzyme LDH from cells, which catalyses the conversion of lactate to pyruvate and cannot be detected extracellularly unless cell damage has occurred. The LDH levels in EAC cells incubated with Conus venom for one hour showed a significant ( p  < 0.01) dose-dependent increase in comparison to the levels in vehicle-treated cells. The release of LDH indicates a loss of cell membrane integrity and is therefore an indirect method to assess the venom-induced cytotoxicity [ 20Zargan J, Sajad M, Umar S, Naime M, Ali S, Khan H. Scorpion (Androctonus crassicauda) venom limits growth of transformed cells (SH-SY5Y and MCF-7) by cytotoxicity and cell cycle arrest. Exp Mol Pathol. 2011;19(1):447–454. doi: 10.1016/j.yexmp.2011.04.008.
https://doi.org/10.1016/j.yexmp.2011.04....
, 49Debnath A, Chatterjee U, Das M, Vedasiromoni JR, Gomes A. Venom of Indian monocellate cobra and Russell’s viper show anticancer activity in experimental models. J Ethnopharmacol. 2007;19(3):681–684. doi: 10.1016/j.jep.2006.12.027.
https://doi.org/10.1016/j.jep.2006.12.02...
]. Abdel-Rahman et al . [ 26Abdel-Rahman MA, Abbas OA, El-Naggar MS, Abdel-Nabi IM, Strong PN. Intraspecific variation in the venom of the vermivorous cone snail Conus vexillum. Comp Biochem Physiol C Toxicol Pharmacol. 2011;19(4):318–325. doi: 10.1016/j.cbpc.2011.06.019.
https://doi.org/10.1016/j.cbpc.2011.06.0...
] attributed the direct cytotoxic effects of C. vexillum on mammalian cells to the presence of proteolytic enzymes in the crude venom. Proteolytic enzymes in Conus venom are primarily responsible for venom-induced necrotic activity and could be injected by cone snails to elicit proteolytic degradation of the extracellular matrix in the prey [ 46Saminathan R, Babuji S, Sethupathy S, Viswanathan P, Balasubramanian T, Gopalakrishanakone P. Clinico-toxinological characterization of the acute effects of the venom of the marine snail, Conus loroisii. Acta Trop. 2006;19(1):75–87. doi: 10.1016/j.actatropica.2005.09.001.
https://doi.org/10.1016/j.actatropica.20...
]. Indeed, Cathepsin D and kallikrein-like proteins have recently been identified in the venom glands of C. victoriae and C. novaehollandiae, respectively [ 43Safavi-Hemami H, Siero WA, Gorasia DG, Young ND, Macmillan D, Williamson NA. et al. Specialisation of the venom gland proteome in predatory cone snails reveals functional diversification of the conotoxin biosynthetic pathway. J Proteome Res. 2011;19(9):3904–3919. doi: 10.1021/pr1012976.
https://doi.org/10.1021/pr1012976...
]. Cathepsin D (aspartyl protease) and kallikrein (trypsin and serine proteases) have also been found in other animal venoms [ 50da Silveira RB, dos Santos Filho JF, Mangili OC, Veiga SS, Gremski W, Nader HB. et al. Identification of proteases in the extract of venom glands from brown spiders. Toxicon.2002;19(6):815–822. doi: 10.1016/S0041-0101(02)00078-8.
https://doi.org/10.1016/S0041-0101(02)00...
- 52Matsui T, Fujimura Y, Titani K. Snake venom proteases affecting hemostasis and thrombosis. Biochim Biophys Acta. 2000;19(1–2):146–156. ].

Interestingly, our toxicological data revealed significant differences in the efficacy of C. vexillum venom collected from the two Egyptian locations, Hurgada and Sharm El-Shaikh. The data showed that the venom obtained from Hurgada was more potent than that obtained from Sharm El-Shaikh. We have attributed the difference in venom potency to variation in the expression of peptides from C. vexillum collected from these two locations [ 26Abdel-Rahman MA, Abbas OA, El-Naggar MS, Abdel-Nabi IM, Strong PN. Intraspecific variation in the venom of the vermivorous cone snail Conus vexillum. Comp Biochem Physiol C Toxicol Pharmacol. 2011;19(4):318–325. doi: 10.1016/j.cbpc.2011.06.019.
https://doi.org/10.1016/j.cbpc.2011.06.0...
]. For example, the presence of the m/z peptide 2924.7 (calcium channel blocker) in the venom of Sharm El-Shaikh may explain why venom-induced oxidative stress was significantly higher in the case of Hurgada venom. The presence of this calcium channel blocker in Sharm El-Shaikh venom delays rises in intracellular calcium concentrations in venom-treated cells and hence slows the elevation of oxidative stress markers in comparison with Hurgada venom [ 26Abdel-Rahman MA, Abbas OA, El-Naggar MS, Abdel-Nabi IM, Strong PN. Intraspecific variation in the venom of the vermivorous cone snail Conus vexillum. Comp Biochem Physiol C Toxicol Pharmacol. 2011;19(4):318–325. doi: 10.1016/j.cbpc.2011.06.019.
https://doi.org/10.1016/j.cbpc.2011.06.0...
].

It is well documented that oxidative stress is extremely deleterious to cells and reactive oxygen species (ROS) are engaged in the etiology and progression of several diseases including cancer. Several environmental oxidants such as heat shock, UV irradiation, infections, and toxins [ 26Abdel-Rahman MA, Abbas OA, El-Naggar MS, Abdel-Nabi IM, Strong PN. Intraspecific variation in the venom of the vermivorous cone snail Conus vexillum. Comp Biochem Physiol C Toxicol Pharmacol. 2011;19(4):318–325. doi: 10.1016/j.cbpc.2011.06.019.
https://doi.org/10.1016/j.cbpc.2011.06.0...
, 53Ermak G, Davies KJ. Calcium and oxidative stress: from cell signaling to cell death. Mol Immunol. 2001;19(10):713–721. , 54Abdel-Rahman MA, Omran M, Abdel-Nabi IM, Nassier O, Schemerhorn B. Neurotoxic and cytotoxic effects of venom from different populations of the Egyptian Scorpio maurus palmatus. Toxicon. 2010;19(2–3):298–306. ] can induce oxidative stress that shifting the cellular redox status to a more oxidized state. In normal conditions, enzymatic and non-enzymatic antioxidants are capable of neutralizing harmful effects of ROS and protect cellular components from damage. However, under conditions of excessive oxidative stress, ROS can damage cellular components and interfere with critical cellular activities [ 55Sardar N, Singh M, Chatterjee M. Vitamin D3 as a modulator of cellular antioxidant defence in murine lymphoma. Nutr Res. 2000;19(1):91–102. doi: 10.1016/S0271-5317(99)00141-4.
https://doi.org/10.1016/S0271-5317(99)00...
]. Numerous studies have provided evidence that ROS are directly involved in oxidative damage with cellular macromolecules such as lipids, proteins and nucleic acids, leading to cell death [ 56Chan PH. Reactive oxygen radicals in signalling and damage in the ischemic brain. J Cereb Blood Flow Metab. 2001;19(1):2–14. ]. Cancer cells have active protective mechanisms to prevent lipid peroxidation. It has been demonstrated that the presence of relatively low levels of the NADPH-cytochrome-P450 electron transport chain may explain the remarkable decrease of lipid peroxidation in cancer cells when compared to normal cells [ 57Faber M, Coudray C, Hida H, Mousseau M, Favier A. Lipid peroxidation products, and vitamin and trace element status in patients with cancer before and after chemotherapy, including adriarnycin. Bio Trace Elem Res. 1995;19(1–3):117. ].

In order to examine Conus venom induced oxidative stress, the changes of intracellular ROS of EAC cells were measured. Conus venom administration (1.25 mg/kg) to tumor-bearing albino mice induced significant increases in the oxidative stress biomarkers (MDA, PCC) of the propagated EAC cells. We have recently suggested possible mechanisms by which the venom of C. vexillum induces cellular oxidative damage in murine cells [ 26Abdel-Rahman MA, Abbas OA, El-Naggar MS, Abdel-Nabi IM, Strong PN. Intraspecific variation in the venom of the vermivorous cone snail Conus vexillum. Comp Biochem Physiol C Toxicol Pharmacol. 2011;19(4):318–325. doi: 10.1016/j.cbpc.2011.06.019.
https://doi.org/10.1016/j.cbpc.2011.06.0...
]. Damage could be attributed to phospholipase A2 activity – for example an enzyme analogous to conodipine-M – on cell membranes, causing the release of arachidonic acid [ 58McIntosh JM, Ghomaschi F, Gelb MH, Dooley DJ, Stoehr SJ, Giordani AB. et al. Conodipine-M, a novel phospholipase A2 isolated from the venom of the marine snailConus magus. J Biol Chem. 1995;19(8):3518–3526. doi: 10.1074/jbc.270.8.3518.
https://doi.org/10.1074/jbc.270.8.3518...
]. Arachidonic acid can potentiate cell damage by converting apoptosis to necrosis through lipid peroxidation and the promotion of DNA fragmentation [ 59Higuchi Y, Yoshimoto T. Archidonic acid converts the glutathione depletion-induced apoptosis to necrosis by promoting lipid peroxidation in glioma cells. Arch Biochem Biophys. 2002;19(1):133–140. doi: 10.1006/abbi.2002.2784.
https://doi.org/10.1006/abbi.2002.2784...
].

NO may also play a prominent role in cytotoxicity induced by Conus venom. Our results showed that Conus venom significantly increased the production of NO in the ascites fluid of treated animals, especially in Hurgada venom. The elevation of NO levels may be due to the activation of macrophages present in the ascites fluid. It has been found that macrophages play an important role against neoplastic cells [ 60Jackson PG, Evans SR. Intraperitoneal macrophages and tumor immunity: A review. J Surg Oncol. 2000;19(2):146–154. doi: 10.1002/1096-9098(200010)75:23.0.CO;2-1.
https://doi.org/10.1002/1096-9098(200010...
]. Activated macrophages initiate cell death programs through the release of soluble immune mediators [ 61Chattopadhyay S, Das T, Sa G, Ray PK. Protein A-activated macrophages induce apoptosis in Ehrlich’s ascites carcinoma through a nitric oxide-dependent pathway.Apoptosis. 2002;19(1):49–57. doi: 10.1023/A:1013512912160.
https://doi.org/10.1023/A:1013512912160...
]. One mechanism that macrophages use to exert their cytolytic effects on target tumors is by the release of NO through activation of NADPH-dependent NO synthase [ 62Farias-Eisner R, Sherman MP, Aeberhard E, Chaudhuri G. Nitric oxide is an important mediator for tumoricidal activity in vivo. Proc Natl Acad Sci U S A. 1994;19(20):9407–9411. doi: 10.1073/pnas.91.20.9407.
https://doi.org/10.1073/pnas.91.20.9407...
, 63Bredt DS. Endogenous nitric oxide synthesis: Biological functions and pathophysiology.Free Radic Res. 1990;19(6):577–596. ]. NO can cause growth arrest and apoptosis because of its high reactivity with iron-and thiol-containing macromolecules, thereby inhibiting enzymes of the TCA cycle and those involved in mitochondrial respiration, as well as DNA synthesis and repair [ 64Cui S, Reichner JS, Mateo RB, Albina JE. Activated murine macrophages induce apoptosis in tumor cells through nitric oxide-dependent or -independent mechanisms.Cancer Res. 1994;19(9):2462–2467. , 65Stamler JS. Redox signaling: Nitrosylation and related target interactions of nitric oxide.Cell. 1994;19(6):931–936. doi: 10.1016/0092-8674(94)90269-0.
https://doi.org/10.1016/0092-8674(94)902...
]. It can be noted that our data revealed that increases in NO levels paralleled a significant increase in the level of PCC, which is an indication of protein damage of EAC cells.

Several previous studies have clarified the influence of ROS overproduction on intracellular Ca2+ levels. Chandra et al . [ 66Chandra J, Samali A, Orrenius S. Triggering and modulation of apoptosis by 612 oxidative stress. Free Radic Biol Med. 2000;19(3–4):323–333. ] found that increases in the concentrations of intracellular ROS were a sign of the onset of apoptotic processes; they cited ROS production as a critical determinant of the toxicity associated with exposure to chemotherapeutic drugs. It has been shown that oxidative stress increases intracellular Ca2+ concentrations, leading to activation of endonucleases which degrade DNA and, ultimately, contribute to cell damage [ 67Cantoni O, Sestili F, Cattabeni F, Bellomo G, Pou S, Cohen M. et al. Calcium Chelator Quin-2 prevents hydrogenperoxide induced DNA breakage and cytotoxicity. Eur J Biochem. 1982;19(2):209–212. ]. Orrenius et al . [ 68Orrenius S, Burkitt MJ, Kass GE, Dybukt GM, Nicotera P. Calcium ions and oxidative cell injury. Ann Neurol. 1992;19:S33–S42. doi: 10.1002/ana.410320708.
https://doi.org/10.1002/ana.410320708...
] found that free radical overproduction may inhibit Ca2+ ATPases and this leads to altered regulation of Ca2+ levels and cell death. Ermak et al. [ 53Ermak G, Davies KJ. Calcium and oxidative stress: from cell signaling to cell death. Mol Immunol. 2001;19(10):713–721. ] clearly demonstrated the influence of rising intracellular Ca2+ concentration induced by oxidative stress. They found that oxidative stress increases the process of Ca2+ diffusion from both the endoplasmic reticulum and extracellular environment into the cytoplasm. Consequently, the elevation of cytoplasmic Ca2+ concentrations causes Ca2+ influx into both mitochondria and nuclei. High concentration of Ca2+ in mitochondria leads to cell damage through disruption normal cellular metabolism. In nuclei, high Ca2+ levels modulate transcription factors and nucleases that regulate cell apoptosis. Moreover, the enzymes (e.g. nitric oxide synthase) that produce free radicals can be activated by the elevation of Ca2+ level [ 69Lynch DR, Dawson TM. Secondary mechanisms in neuronal trauma. Curr Opin Neurol.1994;19(6):510–516. doi: 10.1097/00019052-199412000-00007.
https://doi.org/10.1097/00019052-1994120...
]. Therefore, by increasing cytoplasmic Ca2+ concentrations, oxidants can also indirectly cause more oxidant production and further raise Ca2+ levels.

In the present work, increased levels of cellular oxidative stress in EAC cells treated with Conus venom were also accompanied by a remarkable decrease in the levels of antioxidants in treated cells. The results clearly demonstrated that Conus venom significantly reduced the content of GSH of tumor cells. Modifications of GSH metabolism have been postulated as being useful in cancer therapy [ 70Estrela JM, Ortega A, Obrador E. Glutathione in cancer biology and therapy. Crit Rev Clin Lab Sci. 2006;19(2):143–181. doi: 10.1080/10408360500523878.
https://doi.org/10.1080/1040836050052387...
]. Indeed, the elevation of intracellular GSH content has been associated with mitogenic stimulation [ 71Shaw JP, Chou IN. Elevation of intracellular glutathione content associated with mitogenic stimulation of quiescent fibroblasts. J Cell Physiol. 1986;19(2):193–198. doi: 10.1002/jcp.1041290210.
https://doi.org/10.1002/jcp.1041290210...
, 72Ortega A, Mena S, Estrela JM. Glutathione in cancer cell death. Cancers.2011;19(1):1285–1310. ]. GSH has been suggested as a potential regulator of protein synthesis, DNA synthesis and cell proliferation [ 72Ortega A, Mena S, Estrela JM. Glutathione in cancer cell death. Cancers.2011;19(1):1285–1310. , 73Suthanthiran M, Anderson ME, Sharma VK, Meister A. Glutathione regulates activation-dependent DNA synthesis in highly purified normal human T lymphocytes stimulated via the CD2 and CD3 antigens. Proc Natl Acad Sci USA. 1990;19(9):3343–3347. doi: 10.1073/pnas.87.9.3343.
https://doi.org/10.1073/pnas.87.9.3343...
]. Several previous in vivo and in vitro studies have revealed that the amino acid precursors (glutamine and methionine) for GSH synthesis play a crucial role in cancer metabolism. Tumour cells use the amino acid glutamine as a main respiratory fuel [ 74Moreadith RW, Lehninger AL. The pathways of glutamate and glutamine oxidation by tumor cell mitochondria. Role of mitochondrial NAD (P)+-dependent malic enzyme. J Biol Chem. 1984;19(10):6215–6221. ] and methionine in cell growth [ 75Hoffman RM. Altered methionine metabolism and transmethylation in cancer.Anticancer Res. 1985;19(1):1–30. ]. Moreover, our data showed that the depletion of GSH content was concomitant with a reduction in the activity of enzymatic antioxidants (CAT and SOD) as well as a reduction in the level of TAC. The observed reduction in the level of antioxidants of tumor cells from venom-treated animals may explain the role of oxidative stress in cell damage. It is well established that antioxidant defense enzymes such as SOD and CAT play a crucial role in maintaining cellular homeostasis by detoxifying the generated ROS such as superoxide radicals and H2O2[ 76Halliwell B, Gutteridge JM. Free radicals in biology and medicine. 3. Oxford: University Press; 2000. ]. Our findings are corroborate those of Sun et al . [ 77Sun LK, Yoshii Y, Hyodo A, Tsurushima H, Saito A, Harakuni T. et al. Apoptotic effect in the glioma cells induced by specific protein extracted from Okinawa Habu (Trimeresurus flavoviridis) venom in relation to oxidative stress. Toxicology in Vitro. 2003;19(2):169–177. doi: 10.1016/S0887-2333(03)00010-9.
https://doi.org/10.1016/S0887-2333(03)00...
], who reported that the inhibition of cell growth and apoptotic activity of the snake venom fraction OHAP-1 ( Okinawa Habu apoxin protein-1) was reversed by the addition of GSH and catalase to rat and human malignant glioma cell lines.

Conclusions

Conus venoms have provided extensive libraries of toxins active against a diverse range of ion channel proteins, belonging to both voltage-gated and neurotransmitter-activated gene families. The recognition that cone snails also have cytolytic peptides in their venom arsenals should not be surprising, given that similarly acting peptides in snake venom have been known for thirty years but it was only a few years ago that the first Conus venom cytolytic peptide, conolysin MT was discovered [ 47Biggs JS, Rosenfeld Y, Shai Y, Olivera BM. Conolysin-Mt: a conus peptide that disrupts cellular membranes. Biochemistry. 2007;19(44):12586–12593. doi: 10.1021/bi700775p.
https://doi.org/10.1021/bi700775p...
]. Our present study aims at contributing to the study of the mechanism of action of these cytolytic toxins from Cone snails that interact with and disrupt cell membranes. Our data indicate that C. vexillum venom contains components which induce oxidative stress mechanisms in Erhlich’s ascites carcninoma cells and suggest that the venom contains novel molecules with potential anticancer activity.

Ethics committee approval

The present study was approved by the Research Ethics Committee of Suez Canal University and was carried out in accordance with the Guide for the Care and Use of Laboratory Animals (by the Committee for the Update of the Guide for the Care and Use of Laboratory Animals of the National Research Council, Washington, USA, 2011).

Acknowledgments

We would like to thank The Red Sea Protectorate Management Unit (RSPMU) in Hurgada and Sharm El-Shaikh for giving us the logistical support and permission to collect Conus species from these locations.

  • Mann J. Natural products in cancer chemotherapy: past, present and future. Nat Rev Cancer. 2002;19(2):143–148. doi: 10.1038/nrc723.
    » https://doi.org/10.1038/nrc723
  • Lewis RJ, Garcia ML. Therapeutic potential of venom peptides. Nat Rev Drug Discov.2003;19(10):790–802. doi: 10.1038/nrd1197.
    » https://doi.org/10.1038/nrd1197
  • Renan MJ. How many mutations are required for tumorigenesis? Implications from human cancer data. Mol Carcinog. 1993;19(3):139–146. doi: 10.1002/mc.2940070303.
    » https://doi.org/10.1002/mc.2940070303
  • Wang Z. Roles of K+ channels in regulating tumour cell proliferation and apoptosis.Pflugers Arch. 2004;19(3):274–286. doi: 10.1007/s00424-004-1258-5.
    » https://doi.org/10.1007/s00424-004-1258-5
  • Hanahan D, Weinberg RA. The hallmarks of cancer. Cell. 2000;19(1):57–70. doi: 10.1016/S0092-8674(00)81683-9.
    » https://doi.org/10.1016/S0092-8674(00)81683-9
  • Espinosa E, Zamora P, Feliu J, Barón MG. Classification of anticancer drugs – a new system based on therapeutic targets. Cancer Treat Rev. 2003;19(6):515–523. doi: 10.1016/S0305-7372(03)00116-6.
    » https://doi.org/10.1016/S0305-7372(03)00116-6
  • Cassidy J, Misset JL. Oxaliplatin-related side-effects: characteristics and management.Semin Oncol. 2002;19(5 Suppl 15):11–20. doi: 10.1053/sonc.2002.35524.
    » https://doi.org/10.1053/sonc.2002.35524
  • Kalyanaraman B, Joseph J, Kalivendi S, Wang S, Konorev E, Kotamraju S. Doxorubicin-induced apoptosis: implications in cardiotoxicity. Mol Cell Biochem. 2002;19(1–4):119–124.
  • Naumov GN, Towson JL, MacDonald IC, Wilson SM, Bramwell VH, Groom AC. et al. Ineffectiveness of doxorubicin treatment on solitary dormant mammary carcinoma cells or late-developing metastasis. Breast Cancer Res Treat. 2003;19(3):199–206. doi: 10.1023/B:BREA.0000004377.12288.3c.
    » https://doi.org/10.1023/B:BREA.0000004377.12288.3c
  • Benjamin CW, Hiebsch RR, Jones DA. Caspase activation in MCF7 cells responding to etoposide treatment. Mol Pharmacol. 1998;19(3):446–450.
  • Gatti L, Zunino F. Overview of tumor cell chemoresistance mechanisms. Methods Mol Med. 2005;19:127–148.
  • Abu-Sinna G, Esmat AY, Al-Zahaby AS, Soliman NA, Ibrahim TM. Fractionation and characterization of Cerastes cerastes cerastes snake venom and the antitumor action of its lethal and non-lethal fractions. Toxicon. 2003;19(2):207–215. doi: 10.1016/S0041-0101(03)00138-7.
    » https://doi.org/10.1016/S0041-0101(03)00138-7
  • El-Refael MF, Sarkar NH. Snake venom inhibits the growth of mouse mammary tumor cells in vitro and in vivo Toxicon. 2009;19(1):33–41. doi: 10.1016/j.toxicon.2009.03.017.
    » https://doi.org/10.1016/j.toxicon.2009.03.017
  • Xie Q, Tang N, Wan R, Qi Y, Lin X, Lin J. Recombinant snake venom cystatin inhibits the growth, invasion and metastasis of B16F10 cells and MHCC97H cells in vitro and in vivo Toxicon. 2011;19(5):704–711. doi: 10.1016/j.toxicon.2011.02.002.
    » https://doi.org/10.1016/j.toxicon.2011.02.002
  • Orsolić N, Sver L, Verstovsek S, Terzić S, Basić I. Inhibition of mammary carcinoma cell proliferation in vitro and tumor growth in vivo by bee venom. Toxicon. 2003;19(7):861–870. doi: 10.1016/S0041-0101(03)00045-X.
    » https://doi.org/10.1016/S0041-0101(03)00045-X
  • Huh JE, Baek YH, Lee MH, Choi DY, Park DS, Lee JD. Bee venom inhibits tumor angiogenesis and metastasis by inhibiting tyrosine phosphorylation of VEGFR-2 in LLC-tumor-bearing mice. Cancer Lett. 2010;19(1):98–110. doi: 10.1016/j.canlet.2009.11.013.
    » https://doi.org/10.1016/j.canlet.2009.11.013
  • Chu ST, Cheng HH, Huang CJ, Chang HC, Chi CC, Su HH. et al. Phospholipase A2-independent Ca2+ entry and subsequent apoptosis induced by melittin in human MG63 osteosarcoma cells. Life Sci. 2007;19(4):364–369. doi: 10.1016/j.lfs.2006.09.024.
    » https://doi.org/10.1016/j.lfs.2006.09.024
  • Son DJ, Lee JW, Lee YH, Song HS, Lee CK, Hong JT. Therapeutic application of anti-arthritis, pain-releasing, and anti-cancer effects of bee venom and its constituent compounds. Pharmacol Ther. 2007;19(2):246–270. doi: 10.1016/j.pharmthera.2007.04.004.
    » https://doi.org/10.1016/j.pharmthera.2007.04.004
  • Bhoopathi P, Chetty C, Kunigal S, Vanamala SK, Rao JS, Lakka SS. Blockade of tumor growth due to matrix metalloproteinase-9 inhibition is mediated by sequential activation of beta1-integrin, ERK, and NF-kappaB. J Biol Chem. 2008;19(3):1545–1552.
  • Zargan J, Sajad M, Umar S, Naime M, Ali S, Khan H. Scorpion (Androctonus crassicauda) venom limits growth of transformed cells (SH-SY5Y and MCF-7) by cytotoxicity and cell cycle arrest. Exp Mol Pathol. 2011;19(1):447–454. doi: 10.1016/j.yexmp.2011.04.008.
    » https://doi.org/10.1016/j.yexmp.2011.04.008
  • Jäger H, Dreker T, Buck A, Giehl K, Gress T, Grissmer S. Blockage of intermediate-conductance Ca2+-activated K+ channels inhibit human pancreatic cancer cell growth in vitro Mol Pharmacol. 2004;19(3):630–638. doi: 10.1124/mol.65.3.630.
    » https://doi.org/10.1124/mol.65.3.630
  • Deshane J, Garner CC, Sontheimer H. Chlorotoxin inhibits glioma invasion via matrix metalloproteinase-2. J Biol Chem. 2003;19(6):4135–4144. doi: 10.1074/jbc.M205662200.
    » https://doi.org/10.1074/jbc.M205662200
  • Gupta SD, Gomes A, Debnath A, Saha A, Gomes A. Apoptosis induction in human leukemic cells by a novel protein Bengalin, isolated from Indian black scorpion: through mitochondrial pathway and inhibition of heat shock proteins. Chem Biol Interact.2010;19(2):293–303. doi: 10.1016/j.cbi.2009.11.006.
    » https://doi.org/10.1016/j.cbi.2009.11.006
  • Davis JM, Jones A, Lewis RJ. Remarkable inter- and intraspecies complexity of conotoxins revealed by LC/MS. Peptides. 2009;19(7):11222–11227.
  • Scott DA, Wright CE, Angus JA. Actions of intrathecal ω-conotoxins CVID, GVIA, MVIIA, and morphine in acute and neuropathic pain in the rat. Eur J Pharmacol.2002;19(3):279–286. doi: 10.1016/S0014-2999(02)02247-1.
    » https://doi.org/10.1016/S0014-2999(02)02247-1
  • Abdel-Rahman MA, Abbas OA, El-Naggar MS, Abdel-Nabi IM, Strong PN. Intraspecific variation in the venom of the vermivorous cone snail Conus vexillum Comp Biochem Physiol C Toxicol Pharmacol. 2011;19(4):318–325. doi: 10.1016/j.cbpc.2011.06.019.
    » https://doi.org/10.1016/j.cbpc.2011.06.019
  • Cruz LJ, Ramilo CA, Corpuz GP, Olivera BM. Conus peptides: phylogenetic range of biological activity. Biol Bull. 1992;19(1):159–164. doi: 10.2307/1542418.
    » https://doi.org/10.2307/1542418
  • Mookerjee A, Basu JM, Dutta P, Majumder J, Bhattacharya S. et al. Overcoming drug-resistant in cancer by a newly developed copper chelate through host protective cytokine mediated apoptosis. Clin Cancer Res. 2006;19:4339–4349. doi: 10.1158/1078-0432.CCR-06-0001.
    » https://doi.org/10.1158/1078-0432.CCR-06-0001
  • Viñuela JE, Rodriguez R, Gil J, Coll J, De La Concha EG, Subiza JL. Antigen shedding vs. development of natural suppressor cells as mechanism of tumor escape in mice bearing Ehrlich tumor. Int J Cancer. 1991;19(1):86–91. doi: 10.1002/ijc.2910470116.
    » https://doi.org/10.1002/ijc.2910470116
  • Stuehr DJ, Marletta MA. Induction of nitrite/nitrate synthesis in murine macrophages in BCG infection, lymphokines or interferon-γ J Immunol. 1987;19(2):518–525.
  • Tavazzi B, Di Pierro D, Amorini AM, Fazzina G, Tuttobene M, Giardina B. et al. Energy metabolism and lipid peroxidation of human erythrocytes as a function of increased oxidative stress. Eur J Biochem. 2000;19(3):684–689. doi: 10.1046/j.1432-1327.2000.01042.x.
    » https://doi.org/10.1046/j.1432-1327.2000.01042.x
  • Yagi K. Lipid peroxides and human diseases. Chem Phys Lipids. 1978;19(2–4):337–351.
  • Reznick AZ, Packer L. Oxidative damage to proteins: spectrophotometric method for carbonyl assay. Methods Enzymol. 1994;19:357–363.
  • Fulle S, Mecocci P, Fano G, Vecchiet I, Vecchini A. et al. Specific oxidative alterations in vastus lateralis muscle of patients with the diagnosis of chronic fatigue syndrome. Free Radic Biol Med. 2000;19:1252–1259. doi: 10.1016/S0891-5849(00)00419-6.
    » https://doi.org/10.1016/S0891-5849(00)00419-6
  • Green LC, Wagner DA, Glogowski J, Skipper PL, Wishnok JS, Tannenbaum SR. Analysis of nitrate, nitrite and nitrate in biological fluids. Anal Biochem. 1982;19(1):131–138. doi: 10.1016/0003-2697(82)90118-X.
    » https://doi.org/10.1016/0003-2697(82)90118-X
  • Beutler E, Doron O, Kelly BM. Improved method for the determination of blood glutathione. J Lab Clin Med. 1963;19:882–888.
  • 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;19(10):3170–3175.
  • Aebi H. In: Oxygen radicals in biological systems: Meths Enzymol. Packer L, editor. Orlando: Academic; 1984. Catalase in vitro; pp. 121–126.
  • Cohen G, Dembiec D, Marcus J. Measurement of catalase activity in tissue extracts. Anal Biochem. 1970;19:30–38. doi: 10.1016/0003-2697(70)90083-7.
    » https://doi.org/10.1016/0003-2697(70)90083-7
  • Koracevic D, Koracevic G, Djordjevic V, Andrejevic S, Cosic V. Method for the measurement of antioxidant activity in human fluids. J Clin Pathol. 2001;19(5):356–361. doi: 10.1136/jcp.54.5.356.
    » https://doi.org/10.1136/jcp.54.5.356
  • Dancey CP, Reidy J. Statistics without maths for psychology. Harlow: Pearson Education; 2002.
  • Snedecor GW. Statistical Methods. 4. Iowa: Iowa College Press; 1956.
  • Safavi-Hemami H, Siero WA, Gorasia DG, Young ND, Macmillan D, Williamson NA. et al. Specialisation of the venom gland proteome in predatory cone snails reveals functional diversification of the conotoxin biosynthetic pathway. J Proteome Res. 2011;19(9):3904–3919. doi: 10.1021/pr1012976.
    » https://doi.org/10.1021/pr1012976
  • Tayo LL, Lu BW, Cruz LJ, Yates JR 3rd. Proteomic analysis provides insights on venom processing in Conus textile J Proteome Res. 2010;19(5):2292–2301. doi: 10.1021/pr901032r.
    » https://doi.org/10.1021/pr901032r
  • Ekberg J, Craik DJ, Adams DJ. Conotoxin modulation of voltage-gated sodium channels.Int J Biochem Cell Biol. 2008;19(1):2363–2368.
  • Saminathan R, Babuji S, Sethupathy S, Viswanathan P, Balasubramanian T, Gopalakrishanakone P. Clinico-toxinological characterization of the acute effects of the venom of the marine snail, Conus loroisii Acta Trop. 2006;19(1):75–87. doi: 10.1016/j.actatropica.2005.09.001.
    » https://doi.org/10.1016/j.actatropica.2005.09.001
  • Biggs JS, Rosenfeld Y, Shai Y, Olivera BM. Conolysin-Mt: a conus peptide that disrupts cellular membranes. Biochemistry. 2007;19(44):12586–12593. doi: 10.1021/bi700775p.
    » https://doi.org/10.1021/bi700775p
  • Alouf JE. Pore-forming bacterial protein toxins: An overview. Curr Top Microbiol Immunol. 2001;19:1–14. doi: 10.1007/978-3-642-56508-3_1.
    » https://doi.org/10.1007/978-3-642-56508-3_1
  • Debnath A, Chatterjee U, Das M, Vedasiromoni JR, Gomes A. Venom of Indian monocellate cobra and Russell’s viper show anticancer activity in experimental models. J Ethnopharmacol. 2007;19(3):681–684. doi: 10.1016/j.jep.2006.12.027.
    » https://doi.org/10.1016/j.jep.2006.12.027
  • da Silveira RB, dos Santos Filho JF, Mangili OC, Veiga SS, Gremski W, Nader HB. et al. Identification of proteases in the extract of venom glands from brown spiders. Toxicon.2002;19(6):815–822. doi: 10.1016/S0041-0101(02)00078-8.
    » https://doi.org/10.1016/S0041-0101(02)00078-8
  • Asgari S, Zhang G, Zareie R, Schmidt O. A serine proteinase homolog venom protein from an endoparasitoid wasp inhibits melanization of the host hemolymph. Insect Biochem Mol Biol. 2003;19(10):1017–1024. doi: 10.1016/S0965-1748(03)00116-4.
    » https://doi.org/10.1016/S0965-1748(03)00116-4
  • Matsui T, Fujimura Y, Titani K. Snake venom proteases affecting hemostasis and thrombosis. Biochim Biophys Acta. 2000;19(1–2):146–156.
  • Ermak G, Davies KJ. Calcium and oxidative stress: from cell signaling to cell death. Mol Immunol. 2001;19(10):713–721.
  • Abdel-Rahman MA, Omran M, Abdel-Nabi IM, Nassier O, Schemerhorn B. Neurotoxic and cytotoxic effects of venom from different populations of the Egyptian Scorpio maurus palmatus Toxicon. 2010;19(2–3):298–306.
  • Sardar N, Singh M, Chatterjee M. Vitamin D3 as a modulator of cellular antioxidant defence in murine lymphoma. Nutr Res. 2000;19(1):91–102. doi: 10.1016/S0271-5317(99)00141-4.
    » https://doi.org/10.1016/S0271-5317(99)00141-4
  • Chan PH. Reactive oxygen radicals in signalling and damage in the ischemic brain. J Cereb Blood Flow Metab. 2001;19(1):2–14.
  • Faber M, Coudray C, Hida H, Mousseau M, Favier A. Lipid peroxidation products, and vitamin and trace element status in patients with cancer before and after chemotherapy, including adriarnycin. Bio Trace Elem Res. 1995;19(1–3):117.
  • McIntosh JM, Ghomaschi F, Gelb MH, Dooley DJ, Stoehr SJ, Giordani AB. et al. Conodipine-M, a novel phospholipase A2 isolated from the venom of the marine snailConus magus J Biol Chem. 1995;19(8):3518–3526. doi: 10.1074/jbc.270.8.3518.
    » https://doi.org/10.1074/jbc.270.8.3518
  • Higuchi Y, Yoshimoto T. Archidonic acid converts the glutathione depletion-induced apoptosis to necrosis by promoting lipid peroxidation in glioma cells. Arch Biochem Biophys. 2002;19(1):133–140. doi: 10.1006/abbi.2002.2784.
    » https://doi.org/10.1006/abbi.2002.2784
  • Jackson PG, Evans SR. Intraperitoneal macrophages and tumor immunity: A review. J Surg Oncol. 2000;19(2):146–154. doi: 10.1002/1096-9098(200010)75:23.0.CO;2-1.
    » https://doi.org/10.1002/1096-9098(200010)75:2<146::AID-JSO14>3.0.CO;2-1
  • Chattopadhyay S, Das T, Sa G, Ray PK. Protein A-activated macrophages induce apoptosis in Ehrlich’s ascites carcinoma through a nitric oxide-dependent pathway.Apoptosis. 2002;19(1):49–57. doi: 10.1023/A:1013512912160.
    » https://doi.org/10.1023/A:1013512912160
  • Farias-Eisner R, Sherman MP, Aeberhard E, Chaudhuri G. Nitric oxide is an important mediator for tumoricidal activity in vivo Proc Natl Acad Sci U S A. 1994;19(20):9407–9411. doi: 10.1073/pnas.91.20.9407.
    » https://doi.org/10.1073/pnas.91.20.9407
  • Bredt DS. Endogenous nitric oxide synthesis: Biological functions and pathophysiology.Free Radic Res. 1990;19(6):577–596.
  • Cui S, Reichner JS, Mateo RB, Albina JE. Activated murine macrophages induce apoptosis in tumor cells through nitric oxide-dependent or -independent mechanisms.Cancer Res. 1994;19(9):2462–2467.
  • Stamler JS. Redox signaling: Nitrosylation and related target interactions of nitric oxide.Cell. 1994;19(6):931–936. doi: 10.1016/0092-8674(94)90269-0.
    » https://doi.org/10.1016/0092-8674(94)90269-0
  • Chandra J, Samali A, Orrenius S. Triggering and modulation of apoptosis by 612 oxidative stress. Free Radic Biol Med. 2000;19(3–4):323–333.
  • Cantoni O, Sestili F, Cattabeni F, Bellomo G, Pou S, Cohen M. et al. Calcium Chelator Quin-2 prevents hydrogenperoxide induced DNA breakage and cytotoxicity. Eur J Biochem. 1982;19(2):209–212.
  • Orrenius S, Burkitt MJ, Kass GE, Dybukt GM, Nicotera P. Calcium ions and oxidative cell injury. Ann Neurol. 1992;19:S33–S42. doi: 10.1002/ana.410320708.
    » https://doi.org/10.1002/ana.410320708
  • Lynch DR, Dawson TM. Secondary mechanisms in neuronal trauma. Curr Opin Neurol.1994;19(6):510–516. doi: 10.1097/00019052-199412000-00007.
    » https://doi.org/10.1097/00019052-199412000-00007
  • Estrela JM, Ortega A, Obrador E. Glutathione in cancer biology and therapy. Crit Rev Clin Lab Sci. 2006;19(2):143–181. doi: 10.1080/10408360500523878.
    » https://doi.org/10.1080/10408360500523878
  • Shaw JP, Chou IN. Elevation of intracellular glutathione content associated with mitogenic stimulation of quiescent fibroblasts. J Cell Physiol. 1986;19(2):193–198. doi: 10.1002/jcp.1041290210.
    » https://doi.org/10.1002/jcp.1041290210
  • Ortega A, Mena S, Estrela JM. Glutathione in cancer cell death. Cancers.2011;19(1):1285–1310.
  • Suthanthiran M, Anderson ME, Sharma VK, Meister A. Glutathione regulates activation-dependent DNA synthesis in highly purified normal human T lymphocytes stimulated via the CD2 and CD3 antigens. Proc Natl Acad Sci USA. 1990;19(9):3343–3347. doi: 10.1073/pnas.87.9.3343.
    » https://doi.org/10.1073/pnas.87.9.3343
  • Moreadith RW, Lehninger AL. The pathways of glutamate and glutamine oxidation by tumor cell mitochondria. Role of mitochondrial NAD (P)+-dependent malic enzyme. J Biol Chem. 1984;19(10):6215–6221.
  • Hoffman RM. Altered methionine metabolism and transmethylation in cancer.Anticancer Res. 1985;19(1):1–30.
  • Halliwell B, Gutteridge JM. Free radicals in biology and medicine. 3. Oxford: University Press; 2000.
  • Sun LK, Yoshii Y, Hyodo A, Tsurushima H, Saito A, Harakuni T. et al. Apoptotic effect in the glioma cells induced by specific protein extracted from Okinawa Habu (Trimeresurus flavoviridis) venom in relation to oxidative stress. Toxicology in Vitro. 2003;19(2):169–177. doi: 10.1016/S0887-2333(03)00010-9.
    » https://doi.org/10.1016/S0887-2333(03)00010-9
  • Financial source This project has been financially supported by the Sector of Post-graduate Studies and Researches Affairs, Suez Canal University (Ismailia, Egypt).

Publication Dates

  • Publication in this collection
    2013

History

  • Received
    29 June 2012
  • Accepted
    14 Aug 2012
Centro de Estudos de Venenos e Animais Peçonhentos (CEVAP/UNESP) Av. Universitária, 3780, Fazenda Lageado, Botucatu, SP, CEP 18610-034, Brasil, Tel.: +55 14 3880-7693 - Botucatu - SP - Brazil
E-mail: editorial.jvatitd@unesp.br