309
Views
32
CrossRef citations to date
0
Altmetric
Research Article

Doxorubicin-induced neurotoxicity is attenuated by a 43-kD protein from the leaves of Cajanus indicus L. via NF-κB and mitochondria dependent pathways

, &
Pages 785-798 | Received 30 Dec 2011, Accepted 20 Mar 2012, Published online: 10 Apr 2012

References

  • Calbresi P, Clark J, Hananske AR, Wiemann MC. Pharmacology of antineoplastic agents, In: Calabresi P, Schein PS (eds). Medical oncology. 2nd ed. New York: McGraw-Hill; 1993. pp. 294–297.
  • Beretta GL, Zunino F. Molecular mechanisms of anthracycline activity. Top Curr Chem 2008;283:1–19.
  • Park ES, Kim SD, Lee MH, Lee HS, Lee IS, Sung JK, . Protective effects of N-acetylcysteine and selenium against doxorubicin toxicity in rats. J Vet Sci 2003;4:129–136.
  • Olson RD, Mushlin PS. Doxorubicin cardiotoxicity: analysis of prevailing hypotheses. FASEB J. 1990;4:3076–3086.
  • Rajagopalan S, Politi PM, Sinha BK, Myers E. Adriamycin-induced free radical formation in the perfused rat heart: implications for cardiotoxicity. Cancer Res 1988;48:4766–4769.
  • Kalender Y, Yel M, Kalender S. Doxorubicin hepatotoxicity and hepatic free radical metabolism in rats: the effects of vitamin E and catechin. Toxicology 2005;209:39–45.
  • Yagmurca M, Bas O, Mollaoglu H, Sahin O, Nacar A, Karaman O, . Protective effects of erdosteine on doxorubicin-induced hepatotoxicity in rats. Arch Med Res 2007;38:380–385.
  • Das J, Ghosh J, Manna P, Sil PC. Taurine suppresses doxorubicin-triggered oxidative stress and cardiac apoptosis in rat via up-regulation of PI3-K/Akt and inhibition of p53, p38-JNK. Biochem Pharmacol 2011;81:891–909.
  • Singal PK, Siveski-Iliskovic N, Hill M, Thomas TP, Li T. Combination therapy with probucol prevents adriamycin-induced cardiomyopathy. J Mol Cell Cardiol 1995;27:1055–1063.
  • Ghosh J, Das J, Manna P, Sil PC. The protective role of arjunolic acid against doxorubicin induced intracellular ROS dependent JNK-p38 and p53 mediated cardiac apoptosis. Biomaterials 2011;32:4857–4866.
  • Sugimoto K, Tamayose K, Sasaki M, Hayashi K, Oshimi K. Low-dose doxorubicin-induced necrosis in Jurkat cells and its acceleration and conversion to apoptosis by antioxidants. British J Haematol 2002;118:229–238.
  • Dahl JE. Influence of doxorubicin on rat incisor mesenchymal cells. Eur J Oral Sci 1984;92:6–13
  • Flora SJS, Mittal M, Mishra D. Co-exposure to arsenic and fluoride on oxidative stress, glutathione linked enzymes, biogenic amines and DNA damage in mouse brain. J Neurol Sci 2009;285:198–205.
  • Tsakiris S, Angelogianni P, Schulpis KH, Stavridis JC. Protective effect of l-phenyl alanine on rat brain acetylcholinesterase inhibition induced by radicals. Clin Biochem 2000;33:103–106.
  • Eckenstein F, Sofroniew MV. Identification of central cholinergic neurons containing both choline acetyltransferase and acetylcholinesterase and of central neurons containing only acetylcholinesterase. J Neurosci 1983;3:2286–2291.
  • Bhattacharjee R, Sil PC. The protein fraction of Phyllanthus niruri plays a protective role against acetaminophen induced hepatic disorder via its antioxidant properties. Phytother Res 2006;20:595–601.
  • Bhattacharjee R, Sil PC. Protein isolate from the herb, Phyllanthus niruri L. (Euphorbiaceae), plays hepatoprotective role against carbon tetrachloride induced liver damage via its antioxidant properties. Food Chem Toxicol 2007;45:817–826.
  • Chatterjee M, Sil PC. Hepatoprotective effect of aqueous extract of Phyllanthus niruri on nimesulide-induced oxidative stress in vivo. Indian J Biochem Biophys 2006;43:299–305.
  • Chatterjee M, Sarkar K, Sil PC. Herbal (Phyllanthus niruri) protein isolate protects liver from nimesulide induced oxidative stress. Pathophysiology 2006;13:95–102.
  • Ghosh J, Das J, Manna P, Sil PC. Protective effect of the fruits of Terminalia arjuna against cadmium-induced oxidant stress and hepatic cell injury via MAPK activation and mitochondria dependent pathway. Food Chem 2010;123: 1062–1073.
  • Manna P, Sinha M, Sil PC. A 43 kD protein isolated from the herb Cajanus indicus L attenuates sodium fluoride- induced hepatic and renal disorders in vivo. J Biochem Mol Biol 2007;40:382–395.
  • Manna P, Sinha M, Sil PC. Galactosamine-induced hepatotoxic effect and hepatoprotective role of a protein isolated from the herb Cajanus indicus L in vivo. J Biochem Mol Toxicol 2007;21:13–23.
  • Sarkar MK, Kinter M, Mazumder B, Sil PC. Purification and characterization of a novel antioxidant protein molecule from Phyllanthus niruri. Food Chem 2009;114:1405–1412.
  • Sarkar K, Ghosh A, Kinter A, Mazumder B, Sil PC. Purification and characterization of a 43 kD hepatoprotective protein from the herb Cajanus indicus L. Protein J 2006;25:411–421.
  • Ghosh A, Sarkar K, Sil PC. Protective effect of a 43 kD protein from the leaves of the herb, Cajanus indicus L on chloroform induced hepatic-disorder. J Biochem Mol Biol 2006;39:197–207.
  • Ghosh A, Sil PC. Anti-oxidative effect of a protein from Cajanus indicus L. against acetaminophen-induced hepato-nephro toxicity. J Biochem Mol Biol 2007;39:1039–1049.
  • Ghosh A, Sil PC. A protein from Cajanus indicus spreng protects liver and kidney against mercuric chloride-induced oxidative stress. Biol Pharm Bull 2008;31:1651–1658.
  • Ghosh A, Sil PC. Protection of acetaminophen induced mitochondrial dysfunctions and hepatic necrosis via Akt-NF-kappaB pathway: role of a novel plant protein. Chem-Biol Interact 2009;177:96–106.
  • Sarkar K, Ghosh A, Sil PC. Preventive and curative role of a 43 kDa protein from the leaves of the herb Cajanus indicus L on thioacetamide-induced hepatotoxicity in vivo. Hepatol Res 2005;33:39–49.
  • Sarkar K, Sil PC. A 43 kDa protein from the herb Cajanus indicus L. protects thioacetamide induced cytotoxicity in hepatocytes. Toxicol In Vitro 2006;20:634–640.
  • Sarkar K, Sil PC. Attenuation of acetaminophen-induced hepatotoxicity in vivo and in vitro by a 43 kDa protein from the herb Cajanus indicus L. Toxicol Mech Methods 2007; 17:305–315.
  • Sinha M, Manna P, Sil PC. Attenuation of cadmium chloride induced cytotoxicity in murine hepatocytes by a protein isolated from the leaves of the herb Cajanus indicus L. Arch Toxicol 2007;81:397–406.
  • Sarkar K, Sil PC. Cajanus indicus leaf protein: beneficial role in experimental organ pathophysiology. A review. Pathophysiology 2011;18:295–303.
  • Ellman GL, Courtney KD, Andres VJ, Featherstone RMA. A new and rapid colorimetric determination of acetylcholinesterase activity. Biochem Pharmacol 1961;7:88–95.
  • Bradford MM. A rapid and sensitive method for the quantitation of microgram quantities of protein utilizing the principle of protein-dye binding. Anal Biochem 1976;72: 248–254.
  • LeBel CP, Bondy SC. Sensitive and rapid quantitation of oxygen reactive species formation in rat synaptosomes. Neurochem Int 1990;17:435–440.
  • Kim JD, McCarter RJM, Yu BP. Influence of age, exercise and dietary restriction oxidative stress in rats. Aging Clin Exp Res 1996;8:123–129.
  • Wurtman RJ, Axelrod J. A sensitive and specific assay for the estimation of monoamine oxidase. Biochem Pharmacol 1963;12:1439–1445.
  • Jacobwitz DW, Richardson JS. Method for rapid determination of norepinephrine, dopamine and serotonin in the same brain region. Pharmacol Biochem Behav 1978;8:515–519.
  • Esterbauer H, Cheeseman KH. Determination of aldehydic lipid peroxidation products: malonaldehyde and 4-hydroxynonenal. Methods Enzymol 1990;186:407–421.
  • Uchida K, Stadtman ER. Covalent attachment of 4-hydroxynonenal to glyceraldehydes-3-phosphate dehydrogenase. J Biol Chem 1993;268:6388–6393.
  • Nishikimi M, Rao NA, Yagi K. The occurrence of superoxide anion in the reaction of reduced phenazine methosulfate and molecular oxygen. Biochem Biophys Res Commun 1972;46: 849–854.
  • Kakkar P, Das B, Viswanathan PN. A modified spectrophotometric assay of superoxide dismutase. Ind J Biochem Biophys 1984;21:130–132.
  • Habig WH, PabstMJ Jakoby WB, Glutathione S-transferases. The first enzymatic step in mercapturic acid formation. J Biol Chem 1974;249:7130–7139.
  • Smith IK, Vierheller TL, Thorne CA. Assay of glutathione reductase in crude tissue homogenates using 5,5′-dithiobis (2-nitrobenzoic acid). Anal Biochem 1988;175:408–413.
  • Flohe L, Gunzler WA. Assay of glutathione peroxidase. Methods Enzymol 1984;105:114–121.
  • Ellman GL. Tissue sulphydryl group. Arch Biochem Biophys 1959;82:70–77.
  • Hissin PJ, Hilf RA. A fluorometric method for determination of oxidized and reduced glutathione in tissues. Anal Biochem 1976;74:214–226.
  • Bonting SL. Presence of enzyme system in mammalian tissues. In: Bilter EE (ed.), Membrane and ion transport. London: Wiley Inter Science; 1970. pp. 257–263.
  • Hjertén S, Pan H. Purification and characterization of two forms of a low-affinity Ca2 + -ATPase from erythrocyte membranes. Biochim Biophys Acta 1983;728:281–288.
  • Ohnishi T, Suzuki T, Suzuki Y, Ozawa K. A comparative study of plasma membrane Mg2 + -ATPase activities in normal, regenerating and malignant cells. Biochim Biophys Acta 1982;684:67–74.
  • Ghosh J, Das J, Manna P, Sil PC. Cytoprotective effect of arjunolic acid in response to sodium fluoride mediated oxidative stress and cell death via necrotic pathway. Toxicol In Vitro 2008;22:1918–1926.
  • Mingatto FE, Rodrigues T, Pigoso AA, Uyemura SA, Curti C, Santos AC, . The critical role of mitochondrial energetic impairment in the toxicity of nimesulide to hepatocytes. J Pharm Exp Therap 2003;303:601–617.
  • Bruton L, Chabner B, Knollman B. 2010. Goodman & Gilman's the pharmacological basis of therapeutics. 12th ed. New York: McGraw-Hill Medical.
  • Piscitelli SC, Rodvold KA, Rushing DA, Tewksbury DA. Pharmacokinetics and pharmacodynamics of doxorubicin in patients with small cell lung cancer. Clin Pharmacol Ther 1993;53:555–561.
  • Aluise CD, Miriyala S, Noel T, Sultana R, Jungsuwadee P, Taylor TJ, . 2-Mercaptoethane sulfonate prevents doxorubicin-induced plasma protein oxidation and TNF-α release: Implications for the reactive oxygen species-mediated mechanisms of chemobrain. Free Rad Biol Med 2011;50: 1630–1638.
  • Freireich EJ, Gehan EA, Rall DP, Schmidt LH, Skipper HE. Quantitative comparison of toxicity of anticancer agents in mouse, rat, hamster, dog, monkey, and man. Cancer Chemother Rep 1966;50:219–244.
  • Miller WH Jr, Schipper HM, Lee JS, Singer J, Waxman S. Mechanisms of action of arsenic trioxide. Cancer Res 2004; 62:3893–3903.
  • Bras M, Queenan B, Susin SA. Programmed cell death via mitochondria: different modes of dying. Biochemistry 2005; 70:231–239.
  • Pelicano H, Feng L, Zhou Y, Carew JS, Hileman EO, Plunkett W, . Inhibition of mitochondrial respiration: a novel strategy to enhance drug-induced apoptosis in human leukemia cells by a reactive oxygen species-mediated mechanism. J Biol Chem 2003;278:37832–37839.
  • Kroemer G, de The H. Arsenic trioxide, a novel mitochondriotoxic anticancer agent? J Natl Cancer Inst 1999;91: 743–745.
  • Booser DJ, Hortobagyi GN. Anthracycline antibiotics in cancer therapy. Drugs 1994;47:223–258.
  • Grenier MA, Lipshultz SE. Epidemiology of anthracycline cardiotoxicity in children and adults. Semin Oncol 1998;25:72–85.
  • Hortobagyi GN. Anthracyclines in the treatment of cancer. Drugs 1997;54:1–7.
  • Agapito MT, Antolín Y, del Brio MT, Lópes-Burillo S, Pablos MI, Recio JM, . Protective effect of melatonin against adriamycin toxicity in the rat. J Pineal Res 2001;31: 23–30.
  • Öz E, İlhan MN. Effects of melatonin in reducing the toxic effects of doxorubicin. Mol Cell Biochem 2006;286:11–15.
  • Sunai H, Rubin H. The role of magnesium in cell proliferation and transformation. In: Boynton AL, McKochan WL, Whitfield JP, (eds). Ions, cell proliferation and cancer. New York: Academic Press; 1982. pp. 517–537.
  • Repotto M. 1997. Toxilogia fundamental. Madrid, Spain: Diaz de Santos Editions.
  • Yan J, Hales BF. Activator protein-1 (AP-1) DNA binding activity is induced by hydroxyurea in organogenesis stage mouse embryos. Toxicol Sci 2005;85:1013–1023.
  • Sakurai H, Suzuki S, Kawasaki N, Nakano H, Okazaki T, Chino A, . Tumor necrosis factor-alpha-induced IKK phosphorylation of NF-kappaB p65 on serine 536 is mediated through the TRAF2, TRAF5, and TAK1 signaling pathway. J Biol Chem 2003;278:36916–36923.
  • Akira S, Kishimoto T. NF-IL6 and NF-kB in cytokine gene regulation, Adv Immunol 1997;65:1–46.

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.