Drug Res (Stuttg) 2020; 70(06): 265-272
DOI: 10.1055/a-1154-8672
Original Article

Trifluoperazine an Antipsychotic Drug and Inhibitor of Mitochondrial Permeability Transition Protects Cytarabine and Ifosfamide-Induced Neurotoxicity

Amir Kiani
1   Department of Pharmacology and Toxicology, School of Pharmacy, Kermanshah University of Medical Sciences, Kermanshah, Iran
,
Shadi Heydari Nik
1   Department of Pharmacology and Toxicology, School of Pharmacy, Kermanshah University of Medical Sciences, Kermanshah, Iran
,
Adineh Khodadoost
1   Department of Pharmacology and Toxicology, School of Pharmacy, Kermanshah University of Medical Sciences, Kermanshah, Iran
,
2   Department of Pharmacology and Toxicology, School of Pharmacy, Ardabil University of Medical Sciences, Ardabil, Iran
,
Jalal Pourahmad
3   Department of Pharmacology and Toxicology, Faculty of Pharmacy, Shahid Beheshti University of Medical Sciences, Tehran, Iran
› Institutsangaben

Abstract

The link between Ca2+ dysregulation, mitochondria damages, oxidative stress and cellular derangement is particularly evident in neurotoxicity induced by chemotherapeutic agents. In the current study, we investigated effects of trifluoperazine (TFP) as an inhibitor of calmodulin against the cytotoxicity induced by cytarabine (Ara-C) and Ifosfamide (IFOS) on isolated rat neurons and also the mechanisms involved in this toxicity. Isolated rat neurons were pretreated with TFP (100 µM) for 5 min at 37°C, then Ara-C (226 µM) and IFOS (290 µM) were added in separate experiments. After 3 h, the cytotoxicity, reactive oxygen species (ROS), lysosomal membrane destabilization, mitochondrial membrane potential (MMP), lipid peroxidation (LP), glutathione (GSH) and glutathione disulfide (GSSG) levels were measured. Ara-C and IFOS treatments caused a significant decrease in cellular viability, which was accompanied by ROS generation, GSSG/GSH ratio, lipid peroxidation and lysosomal and mitochondrial damages. On the other hand, TFP (100 µM) pre-treatment attenuated Ara-C and IFOS -induced decrease in cell viability. In addition, TFP (100 µM) pre-treatment significantly protected against Ara-C and IFOS -induced increase in ROS generation, lysosomal and mitochondrial damages, lipid peroxidation levels and decrease in GSH/GSSG ratio. Our data provided insights into the mechanism of protection by TFP against Ara-C and IFOS neurotoxicity, which is related, to neuronal ROS formation and mitochondrial damages.



Publikationsverlauf

Eingereicht: 24. Februar 2020
Eingereicht: 18. März 2020

Angenommen: 23. März 2020

Artikel online veröffentlicht:
04. Mai 2020

© Georg Thieme Verlag KG
Stuttgart · New York

 
  • References

  • 1 Bhagra A, Rao RD. Chemotherapy-induced neuropathy. Current Oncology Reports 2007; 9: 290-299
  • 2 Park SB, Goldstein D, Krishnan AV. et al. Chemotherapy-induced peripheral neurotoxicity: a critical analysis. CA: a Cancer Journal for Clinicians 2013; 63: 419-437
  • 3 Areti A, Yerra VG, Naidu V. et al. Oxidative stress and nerve damage: role in chemotherapy induced peripheral neuropathy. Redox Biology 2014; 2: 289-295
  • 4 Tong L, Chuang C-C, Wu S. et al. Reactive oxygen species in redox cancer therapy. Cancer Letters 2015; 367: 18-25
  • 5 Ye Z-W, Zhang J, Townsend DM. et al. Oxidative stress, redox regulation and diseases of cellular differentiation. Biochimica et Biophysica Acta (BBA)-General Subjects 2015; 1850: 1607-1621
  • 6 Gogvadze V, Orrenius S, Zhivotovsky B. Mitochondria as targets for chemotherapy. Apoptosis 2009; 14: 624-640
  • 7 Bozza FA, D’Avila JC, Ritter C. et al. Bioenergetics, mitochondrial dysfunction, and oxidative stress in the pathophysiology of septic encephalopathy. Shock 2013; 39: 10-16
  • 8 Figueira TR, Barros MH, Camargo AA. et al. Mitochondria as a source of reactive oxygen and nitrogen species: from molecular mechanisms to human health. Antioxidants & Redox Signaling 2013; 18: 2029-2074
  • 9 Shipley JL, Butera JN. Acute myelogenous leukemia. Experimental Hematology 2009; 37: 649-658
  • 10 Zhuo M, Gorgun MF, Englander EW. Neurotoxicity of cytarabine (Ara-C) in dorsal root ganglion neurons originates from impediment of mtDNA synthesis and compromise of mitochondrial function. Free Radical Biology and Medicine 2018; 121: 9-19
  • 11 Lee BS, Lee JH, Kang H-G. et al. Ifosfamide nephrotoxicity in pediatric cancer patients. Pediatric Nephrology 2001; 16: 796-799
  • 12 Hamadani M, Awan F. Role of thiamine in managing ifosfamide-induced encephalopathy. Journal of Oncology Pharmacy Practice 2006; 12: 237-239
  • 13 Salimi A, Pirhadi R, Jamali Z. et al. Mitochondrial and lysosomal protective agents ameliorate cytotoxicity and oxidative stress induced by cyclophosphamide and methotrexate in human blood lymphocytes. Human & Experimental Toxicology 2019; 38: 1266-1274
  • 14 Lee CS, Park SY, Ko HH. et al. Inhibition of MPP+-induced mitochondrial damage and cell death by trifluoperazine and W-7 in PC12 cells. Neurochemistry International 2005; 46: 169-178
  • 15 Han JH, Kim YJ, Han ES. et al. Prevention of 7-ketocholesterol-induced mitochondrial damage and cell death by calmodulin inhibition. Brain Research 2007; 1137: 11-19
  • 16 Rodrigues T, Santos AC, Pigoso AA. et al. Thioridazine interacts with the membrane of mitochondria acquiring antioxidant activity toward apoptosis–potentially implicated mechanisms. British Journal of Pharmacology 2002; 136: 136-142
  • 17 Liu S, Han Y, Zhang T. et al. Protective effect of trifluoperazine on hydrogen peroxide-induced apoptosis in PC12 cells. Brain Research Bulletin 2011; 84: 183-188
  • 18 Pereira RS, Bertocchi APF, Vercesi AE. Protective effect of trifluoperazine on the mitochondrial damage induced by Ca2+ plus prooxidants. Biochemical Pharmacology 1992; 44: 1795-1801
  • 19 Yan L, Zeng Q, Wang J. et al. Dexmedetomidine reduces propofol-induced apoptosis of neonatal rat hippocampal neurons via up-regulating Bcl-2 expression. Biomedical Research 2018; 29: 1199-1204
  • 20 Faizi M, Salimi A, Rasoulzadeh M. Schizophrenia induces oxidative stress and cytochrome c release in isolated rat brain mitochondria. Iranian Journal of Pharmaceutical Research 2014; 13: 93-100
  • 21 Assadian E, Zarei MH, Gilani AG. Toxicity of Copper Oxide (CuO) Nanoparticles on Human Blood Lymphocytes. Biological Trace Element Research; 2018. 184 350-357
  • 22 Boya P, Kroemer G. Lysosomal membrane permeabilization in cell death. Oncogene 2008; 27: 6434
  • 23 Beach DC, Giroux E. Inhibition of lipid peroxidation promoted by iron (III) and ascorbate. Archives of Biochemistry and Biophysics 1992; 297: 258-264
  • 24 Hissin PJ, Hilf R. A fluorometric method for determination of oxidized and reduced glutathione in tissues. Analytical Biochemistry 1976; 74: 214-226
  • 25 Lushchak VI. Free radicals, reactive oxygen species, oxidative stress and its classification. Chemico-Biological Interactions 2014; 224: 164-175
  • 26 Deavall DG, Martin EA, Horner JM et al. Drug-induced oxidative stress and toxicity. J Toxicol 2012; 2012: 645460. doi: 10.1155/2012/645460
  • 27 Sharma P, Jha AB, Dubey RS, Pessarakli M Reactive oxygen species, oxidative damage, and antioxidative defense mechanism in plants under stressful conditions. Journal of Botany 2012; 2012: doi: 10.1155/2012/217037
  • 28 Froklage FE, Reijneveld JC, Heimans JJ. Central neurotoxicity in cancer chemotherapy: pharmacogenetic insights. Pharmacogenomics 2011; 12: 379-395
  • 29 Yokoyama C, Sueyoshi Y, Ema M. et al. Induction of oxidative stress by anticancer drugs in the presence and absence of cells. Oncology Letters 2017; 14: 6066-6070
  • 30 Zhang J, Lei W, Chen X. et al. Oxidative stress response induced by chemotherapy in leukemia treatment. Molecular and Clinical Oncology 2018; 8: 391-399
  • 31 Chan K, Truong D, Shangari N. et al. Drug-induced mitochondrial toxicity. Expert Opinion on Drug Metabolism & Toxicology 2005; 1: 655-669
  • 32 Schapira AH. Mitochondrial disease. The Lancet 2006; 368: 70-82
  • 33 Will Y, Shields JE, Wallace KB. Drug-induced mitochondrial toxicity in the geriatric population: Challenges and future directions. Biology 2019; 8: 32
  • 34 Aits S, Jäättelä M Lysosomal Cell Death at a Glance. The Company of Biologists Ltd: 2013
  • 35 Woldemichael T, Rosania GR. The physiological determinants of drug-induced lysosomal stress resistance. PLoS One 2017; 12: e0187627
  • 36 Moustapha A, Pérétout P, Rainey N. et al. Curcumin induces crosstalk between autophagy and apoptosis mediated by calcium release from the endoplasmic reticulum, lysosomal destabilization and mitochondrial events. Cell Death Discovery 2015; 1: 15017
  • 37 Mut-Salud N, Álvarez PJ, Garrido JM et al. Antioxidant intake and antitumor therapy: toward nutritional recommendations for optimal results. Oxid Med Cell Longev 2016; 2016: 6719534. doi: 10.1155/2016/6719534
  • 38 Guo C, Sun L, Chen X. et al. Oxidative stress, mitochondrial damage and neurodegenerative diseases. Neural Regeneration Research 2013; 8: 2003
  • 39 Shevtsova FE, Vinogradova VD, Neganova EM. et al. Mitochondrial permeability transition pore as a suitable target for neuroprotective agents against Alzheimer’s disease. CNS & Neurological Disorders-Drug Targets (Formerly Current Drug Targets-CNS & Neurological Disorders) 2017; 16: 677-685
  • 40 Gorini S, De Angelis A, Berrino L, et al. Chemotherapeutic drugs and mitochondrial dysfunction: focus on doxorubicin, trastuzumab, and sunitinib. Oxidative Medicine and Cellular Longevity 2018, 2018
  • 41 Morota S, Månsson R, Hansson MJ. et al. Evaluation of putative inhibitors of mitochondrial permeability transition for brain disorders—Specificity vs. toxicity. Experimental Neurology 2009; 218: 353-362
  • 42 Banerjee S, Melnyk SB, Krager KJ. et al. Trifluoperazine inhibits acetaminophen-induced hepatotoxicity and hepatic reactive nitrogen formation in mice and in freshly isolated hepatocytes. Toxicology Reports 2017; 4: 134-142