210
Views
4
CrossRef citations to date
0
Altmetric
Research Article

Naja sumatrana venom cytotoxin, sumaCTX exhibits concentration-dependent cytotoxicity via caspase-activated mitochondrial-mediated apoptosis without transitioning to necrosis

& ORCID Icon
Pages 886-900 | Received 08 Feb 2020, Accepted 15 Jul 2020, Published online: 28 Jul 2020

References

  • Attarde, S. and Pandit, S., 2017. Cytotoxic activity of NN-32 toxin from Indian spectacled cobra venom on human breast cancer cell lines. BMC complementary and alternative medicine, 17 (1), 503.
  • Banerjee, A., et al., 2014. The DNA intercalators ethidium bromide and propidium iodide also bind to core histones. FEBS open bio, 4, 251–259.
  • Bell, C.W., et al., 2006. The extracellular release of HMGB1 during apoptotic cell death. American journal of physiology. Cell physiology, 291 (6), C1318–C1325.
  • Brady, H. J. M., 2004. Apoptosis methods and protocols. New Jersey: Humana Press.
  • Brentnall, M., et al., 2013. Caspase-9, caspase-3 and caspase-7 have distinct roles during intrinsic apoptosis. BMC cell biology, 14, 32.
  • Butterick, T.A., et al., 2014. Use of a caspase multiplexing assay to determine apoptosis in a hypothalamic cell model. Journal of visualized experiments, 86:51305.
  • Chan, F.K.M., Moriwaki, K., and De Rosa, M.J., 2013. Detection of necrosis by release of lactate dehydrogenase activity. Methods in molecular biology, 979, 65–70.
  • Chen, K.C., et al., 2007. The mechanism of cytotoxicity by Naja naja atra cardiotoxin 3 is physically distant from its membrane-damaging effect. Toxicon, 50 (6), 816–824.
  • Chen, X., et al., 2009. Apoptosis of human hepatocellular carcinoma cell (HepG2) induced by cardiotoxin III through S-phase arrest. Experimental and toxicologic pathology, 61 (4), 307–315.
  • Chien, C.M., et al., 2010. Taiwan cobra cardiotoxin III inhibits Src kinase leading to apoptosis and cell cycle arrest of oral squamous cell carcinoma Ca9-22 cells. Toxicon, 56 (4), 508–520.
  • Chien, K.Y., et al., 1994. Two distinct types of cardiotoxin as revealed by the structure and activity relationship of their interaction with zwitterionic phospholipid dispersions. Journal of biological chemistry, 266, 3252–3259.
  • Chiou, J.T., et al., 2019. Naja atra cardiotoxin 3 elicits autophagy and apoptosis in U937 human leukemia cells through the Ca2+/PP2A/AMPK axis. Toxins, 11 (9), 527.
  • Christofferson, D.E. and Yuan, J., 2010. Cyclophilin A release as a biomarker of necrotic cell death. Cell death and differentiation, 17 (12), 1942–1943.
  • Das, T., et al., 2013. Inhibition of leukemic U937 cell growth by induction of apoptosis, cell cycle arrest and suppression of VEGF, MMP-2 and MMP-9 activities by cytotoxin protein NN-32 purified from Indian spectacled cobra (Naja naja) venom. Toxicon, 65, 1–4.
  • Das, T., et al., 2011. Cytotoxic and antioxidant property of a purified fraction (NN-32) of Indian Naja naja venom on Ehrlich ascites carcinoma in BALB/c mice. Toxicon, 57 (7–8), 1065–1072.
  • Debnath, A., et al., 2010. A lethal cardiotoxic-cytotoxic protein from the Indian monocellate cobra (Naja kaouthia) venom. Toxicon, 56 (4), 569–579.
  • Denecker, G., et al., 2001. Death receptor-induced apoptotic and necrotic cell death: differential role of caspases and mitochondria. Cell death and differentiation, 8 (8), 829–840.
  • Dorn, G.W., 2013. Molecular mechanisms that differentiate apoptosis from programmed necrosis. Toxicologic pathology, 41 (2), 227–234.
  • Dubovskii, P. and Utkin, Y., 2015. Antiproliferative activity of cobra venom cytotoxins. Current topics in medicinal chemistry, 15 (7), 638–648.
  • Dubovskii, P.V. and Utkin, Y.N., 2014. Cobra cytotoxins: structural organization and antibacterial activity. Acta naturae, 6 (3), 11–18.
  • Dubovskii, P.V., et al., 2003. Interaction of the P-type cardiotoxin with phospholipid membranes. European journal of biochemistry, 270 (9), 2038–2046.
  • Ebrahim, K., et al., 2015. Cobra venom cytotoxins; apoptotic or necrotic agents? Toxicon, 108, 134–140.
  • Ebrahim, K., et al., 2014. Anticancer activity of cobra venom polypeptide, cytotoxin-II, against human breast adenocarcinoma cell line (MCF-7) via the induction of apoptosis. Journal of breast cancer, 17 (4), 314–322.
  • Elefantova, K., et al., 2018. Detection of the mitochondrial membrane potential by the cationic dye JC-1 in L1210 cells with massive overexpression of the plasma membrane ABCB1 drug transporter. International journal of molecular science, 19, 1–14.
  • Elmore, S., 2007. Apoptosis: a review of programmed cell death. Toxicologic pathology, 35 (4), 495–516.
  • Esmekaya, M.A., et al., 2017. Mitochondrial hyperpolarization and cytochrome-c release in microwave-exposed MCF-7 cells. General physiology and biophysics, 36 (2), 211–218.
  • Feofanov, A.V., et al., 2004. Comparative study of structure and activity of cytotoxins from venom of the cobras Naja oxiana, Naja kaouthia, and Naja haje. Biochemistry. Biokhimiia, 69 (10), 1148–1157.
  • Florentin, A. and Arama, E., 2012. Caspase levels and execution efficiencies determine the apoptotic potential of the cell. The journal of cell biology, 196 (4), 513–527.
  • Gasanov, S.E., Dagda, R.K., and Rael, E.D., 2014. Snake venom cytotoxins, phospholipase A2s, and Zn2+-dependent metalloproteinases: mechanisms of action and pharmacological relevance. Journal of clinical toxicology, 4 (1), 1000181.
  • Gasanov, S.E., et al., 1990. The fusogenic properties of the cytotoxins of cobra venom in a model membrane system. Nauchnye doki vyss shkoly biol nauki, 2, 42–50.
  • Gasanov, S.E., et al., 2015. Naja naja oxiana cobra venom cytotoxins CTI and CTII disrupt mitochondrial membrane integrity: implications for basic three-fingered cytotoxins. PLoS one, 10 (6), e0129248.
  • Gergely, P., et al., 2002. Mitochondrial hyperpolarization and ATP depletion in patients with systemic lupus erythematosus. Arthritis & rheumatism, 46 (1), 175–190.
  • Gergely, P., et al., 2002. Persistent mitochondrial hyperpolarization, increased reactive oxygen intermediate production, and cytoplasmic alkalinization characterize altered IL-10 signaling in patients with systemic lupus erythematosus. Journal of immunology, 169 (2), 1092–1101.
  • Giovannini, C., et al., 2002. Mitochondria hyperpolarization is an early event in oxidized low-density lipoprotein-induced apoptosis in Caco-2 intestinal cells. FEBS letters, 523 (1–3), 200–206.
  • Gurunanselage Don, R.A.S. and Yap, M.K.K., 2019. Arctium lappa L. root extract induces cell death via mitochondrial-mediated caspase-dependent apoptosis in Jurkat human leukemic T cells. Biomedicine & pharmacotherapy = biomedecine & pharmacotherapie, 110, 918–929.
  • Hakim, A.E.E., et al., 2011. Purification and characterization of a cytotoxic neurotoxin-like protein from Naja haje haje venom that induces mitochondrial apoptosis pathway. Archives of toxicology, 85 (8), 941–952.
  • He, Y.F., et al., 2018. Changes in apoptosis factors and activation of caspase-3 in tilapia muscle during storage. International journal of food properties, 21 (1), 1800–1810.
  • Henry-Mowatt, J., et al., 2004. Role of mitochondrial membrane permeabilization in apoptosis and cancer. Oncogene, 23 (16), 2850–2860.
  • Hodges, S.J., et al., 1987. Cobra cardiotoxins. Purification, effects on skeletal muscle and structure/activity relationships [published erratum appears in Eur J Biochem 1988 Feb 1;171(3):727]. European journal of biochemistry, 165 (2), 373–383.
  • Hou, L., et al., 2016. Necrotic pyknosis is a morphologically and biochemically distinct event from apoptotic pyknosis. Journal of cell science, 129 (16), 3084–3090.
  • Iijima, T., et al., 2003. Mitochondrial hyperpolarization after transient oxygen-glucose deprivation and subsequent apoptosis in cultured rat hippocampal neurons. Brain research, 993 (1–2), 140–145.
  • Jänicke, R.U., 2009. MCF-7 breast carcinoma cells do not express caspase-3. Breast cancer research and treatment, 117 (1), 219–221.
  • Jänicke, R.U., et al., 1998. Caspase-3 is required for DNA fragmentation and morphological changes associated with apoptosis. The journal of biological chemistry, 273 (16), 9357–9360.
  • Janko, C., et al., 2014. Redox modulation of HMGB1-related signaling. Antioxidants & redox signaling, 20 (7), 1075–1085.
  • Jayaraman, G., et al., 2000. Elucidation of the solution structure of cardiotoxin analogue V from the Taiwan cobra (Naja naja atra)—identification of structural features important for the lethal action of snake venom cardiotoxins. Protein science, 9 (4), 637–646.
  • Jiang, M.S., Fletcher, J.E., and Smith, L.A., 1989. Effects of divalent cations on snake venom cardiotoxin-induced hemolysis and 3H-deoxyglucose-6-phosphate release from human red blood cells. Toxicon, 27 (12), 1297–1305.
  • Jiang, W., Bell, C.W., and Pisetsky, D.S., 2007. The relationship between apoptosis and high-mobility group protein 1 release from murine macrophages stimulated with lipopolysaccharide or polyinosinic–polycytidylic acid. Journal of immunology (Baltimore, MD: 1950), 178 (10), 6495–6503.
  • Karch, J. and Molkentin, J.D., 2015. Regulated necrotic cell death: the passive aggressive side of Bax and Bak. Circulation research, 116 (11), 1800–1809.
  • Khaled, A.R., et al., 2001. Interleukin-3 withdrawal induces an early increase in mitochondrial membrane potential unrelated to the Bcl-2 family. Roles of intracellular pH, ADP transport, and F(0)F(1)-ATPase. The journal of biological chemistry, 276 (9), 6453–6462.
  • Kim, J.M., et al., 2003. Early mitochondrial hyperpolarization and intracellular alkalinization in lactacystin-induced apoptosis of retinal pigment epithelial cells. The journal of pharmacology and experimental therapeutics, 305 (2), 474–481.
  • Kini, R. and Evans, H.J., 1988. Mechanism of platelet effects of cardiotoxins from (spitting cobra) snake venom. Thrombosis research, 52 (3), 185–195.
  • Kini, R., Haar, N.C., and Evans, H.J., 1988. Non-enzymatic inhibitors of coagulation and platelet aggregation from Naja nigricollis venom are cardiotoxins. Biochemical and biophysical research communications, 150 (3), 1012–1016.
  • Kini, R.M., 2002. Molecular moulds with multiple missions: functional sites in three-finger toxins. Clinical and experimental pharmacology & physiology, 29 (9), 815–822.
  • Kini, R.M. and Evans, H.J., 1989. Role of cationic residues in cytolytic activity: modification of lysine residues in the cardiotoxin from Naja nigricollis venom and correlation between cytolytic and antiplatelet activity. Biochemistry, 28 (23), 9209–9215.
  • Konshina, A.G., et al., 2011. Snake cytotoxins bind to membranes via interactions with phosphatidylserine head groups of lipids. PLoS one, 6 (4), e19064.
  • Körper, S., et al., 2003. The K+ channel openers diazoxide and NS1619 induce depolarization of mitochondria and have differential effects on cell Ca2+ in CD34+ cell line KG-1a. Experimental hematology, 31 (9), 815–823.
  • Kumar, T.K.S., et al., 1997. Snake venom cardiotoxins—structure, dynamics, function and folding. Journal of biomolecular structure and dynamics, 15 (3), 431–463.
  • Kung, G., Konstantinidis, K., and Kitsis, R.N., 2011. Programmed necrosis, not apoptosis, in the heart. Circulation research, 108 (8), 1017–1036.
  • Leal, A.M.d.S., et al., 2015. Violacein induces cell death by triggering mitochondrial membrane hyperpolarization in vitro. BMC microbiology, 15, 115.
  • Lee, S., et al., 2013. Phosphatidylserine exposure during apoptosis reflects bidirectional trafficking between plasma membrane and cytoplasm. Cell death & differentiation, 20 (1), 64–76.
  • Lin, K.L., et al., 2010. Down-regulation of the JAK2/PI3K-mediated signaling activation is involved in Taiwan cobra cardiotoxin III-induced apoptosis of human breast MDA-MB-231 cancer cells. Toxicon, 55 (7), 1263–1273.
  • Looi, C.Y., et al., 2013. Induction of apoptosis in human breast cancer cells via caspase pathway by vernodalin isolated from Centratherum anthelminticum (L.) seeds. PLoS one, 8 (2), e56643.
  • Loreto, C., et al., 2014. The role of intrinsic pathway in apoptosis activation and progression in Peyronie’s disease. BioMed research international, 2014, 616110–616149.
  • Matarrese, P., Cauda, R., and Malorni, W., 2003. Activation-associated mitochondrial hyperpolarization hijacks T cells toward an apoptosis-sensitized phenotype. Cell death and differentiation, 10 (5), 609–611.
  • Matarrese, P., et al., 2001. Expression of P-170 glycoprotein sensitizes lymphoblastoid CEM cells to mitochondria-mediated apoptosis. The biochemical journal, 355 (3), 587–595.
  • Mcilwain, D.R., Berger, T., and Mak, T.W., 2013. Caspase functions in cell death and disease. Cold spring harbor perspectives in biology, 5 (4), a008656.
  • Mcvicker, B.L., et al., 2006. Ethanol-induced apoptosis in polarized hepatic cells possibly through regulation of the Fas pathway. Alcoholism: clinical and experimental research, 30 (11), 1906–1915.
  • Mukherjee, A.K., et al., 2015. Apoptosis induction in human breast cancer (MCF-7) cells by a novel venom l-amino acid oxidase (Rusvinoxidase) is independent of its enzymatic activity and is accompanied by caspase-7 activation and reactive oxygen species production. Apoptosis, 20 (10), 1358–1372.
  • Ownby, C.L., Fletcher, J.E., and Colberg, T.R., 1993. Cardiotoxin 1 from cobra (Naja naja atra) venom causes necrosis of skeletal muscle in vivo. Toxicon, 31 (6), 697–709.
  • Panda, S., Kumari, L., and Panda, S., 2016. Structural understanding of cytotoxin 1 of Naja sputatrix: a potential anticancer agent. Journal of drug delivery and therapeutics, 6 (3), 59–63.
  • Parrish, A.B., Freel, C.D., and Kornbluth, S., 2013. Cellular mechanisms controlling caspase activation and function. Cold spring harbor perspectives in biology, 5 (6), a008672.
  • Perelman, A., et al., 2012. JC-1: alternative excitation wavelengths facilitate mitochondrial membrane potential cytometry. Cell death & disease, 3, e430.
  • Poon, I.K.H., Hulett, M.D., and Parish, C.R., 2010. Molecular mechanisms of late apoptotic/necrotic cell clearance. Cell death & differentiation, 17 (3), 381–397.
  • Pisetsky, D.S., 2014. The expression of HMGB1 on microparticles released during cell activation and cell death in vitro and in vivo. Molecular medicine (Cambridge, Mass.), 20, 158–163.
  • Proskuryakov, S.Y., Konoplyannikov, A.G., and Gabai, V.L., 2003. Necrosis: a specific form of programmed cell death? Experimental cell research, 283 (1), 1–16.
  • Qin, S., et al., 2006. Role of HMGB1 in apoptosis-mediated sepsis lethality. The journal of experimental medicine, 203 (7), 1637–1642.
  • Raucci, A., Palumbo, R., and Bianchi, M.E., 2007. HMGB1: a signal of necrosis. Autoimmunity, 40 (4), 285–289.
  • Rivel, M., et al., 2016. Pathogenesis of dermonecrosis induced by venom of the spitting cobra, Naja nigricollis: an experimental study in mice. Toxicon, 119, 171–179.
  • Sakamuru, S., et al., 2012. Application of a homogenous membrane potential assay to assess mitochondrial function. Physiological genomics, 44 (9), 495–503.
  • Sánchez-Alcázar, J., et al., 2000. Increased mitochondrial cytochrome c levels and mitochondrial hyperpolarization precede camptothecin-induced apoptosis in Jurkat cells. Cell death and differentiation, 7 (11), 1090–1100.
  • Scaffidi, P., Misteli, T., and Bianchi, M.E., 2002. Release of chromatin protein HMGB1 by necrotic cells triggers inflammation. Nature, 418 (6894), 191–195.
  • Schulze-Bergkamen, H., et al., 2006. The role of apoptosis versus oncotic necrosis in liver injury: facts or faith? Journal of hepatology, 44 (5), 984–993.
  • Silva, M.T., 2010. Secondary necrosis: the natural outcome of the complete apoptotic program. FEBS letters, 584 (22), 4491–4499.
  • Sivandzade, F., Bhalerao, A., and Cucullo, L., 2019. Analysis of the mitochondrial membrane potential using the cationic JC-1 dye as a sensitive fluorescent probe. Bio-protocol, 9 (1), e3128.
  • Su, J.C., et al., 2010. Concomitant inactivation of the epidermal growth factor receptor, phosphatidylinositol 3-kinase/Akt and Janus tyrosine kinase 2/signal transducer and activator of transcription 3 signaling pathways in cardiotoxin III-treated A549 cells. Clinical and experimental pharmacology & physiology, 37 (8), 833–840.
  • Talanian, R.V., et al., 1997. Substrate specificities of caspase family proteases. The journal of biological chemistry, 272 (15), 9677–9682.
  • Tan, N.H., 1982. Cardiotoxins from the venom of Malayan cobra (Naja naja sputatrix). Archives of biochemistry and biophysics, 218 (1), 51–58.
  • Tsai, P.C., et al., 2014. Cardiotoxin III suppresses hepatocyte growth factor-stimulated migration and invasion of MDA-MB-231 cells. Cell biochemistry and function, 32 (6), 485–495.
  • Twiddy, D., et al., 2006. Caspase-7 is directly activated by the approximately 700-kDa apoptosome complex and is released as a stable XIAP-caspase-7 approximately 200-kDa complex. The journal of biological chemistry, 281 (7), 3876–3888.
  • Urbonaviciute, V., et al., 2008. Induction of inflammatory and immune responses by HMGB1-nucleosome complexes: implications for the pathogenesis of SLE. The journal of experimental medicine, 205 (13), 3007–3018.
  • Walsh, J.G., et al., 2008. Executioner caspase-3 and caspase-7 are functionally distinct proteases. Proceedings of the national academy of sciences of the United States of America, 105 (35), 12815–12819.
  • Wang, C.H. and Wu, W.G., 2005. Amphiphilic beta-sheet cobra cardiotoxin targets mitochondria and disrupts its network. FEBS letters, 579 (14), 3169–3174.
  • Weiss, J.N., et al., 2003. Role of the mitochondrial permeability transition in myocardial disease. Circulation research, 93 (4), 292–301.
  • WHO, 2016. WHO guidelines for the production, control and regulation of snake antivenom immunoglobulins. Geneva, Switzerland: World Health Organization.
  • Wong, O.F., et al., 2010. Five-year experience with Chinese cobra (Naja atra)-related injuries in two acute hospitals in Hong Kong. Hong Kong medical journal, 16, 36–43.
  • Wu, W.G., 1997. Diversity of cobra cardiotoxin. Journal of Toxicology: Toxin Reviews, 16 (3), 115–134.
  • Wu, M., et al., 2013. The anticancer effect of cytotoxin 1 from Naja atra cantor venom is mediated by a lysosomal cell death pathway involving lysosomal membrane permeabilization and cathepsin B release. The American journal of Chinese medicine, 41 (3), 643–663.
  • Yang, S.H., et al., 2007. Effects of cardiotoxin III on expression of genes and proteins related to G2/M arrest and apoptosis in K562 cells. Molecular and cellular biochemistry, 300 (1–2), 185–190.
  • Yap, M.K.K., et al., 2014. Proteomic characterization of venom of the medically important Southeast Asian Naja sumatrana (Equatorial spitting cobra). Acta tropica, 133, 15–25.
  • Yap, M.K.K., Tan, N.H., and Fung, S.Y., 2011. Biochemical and toxinological characterization of Naja sumatrana (Equatorial spitting cobra) venom. Journal of venomous animals and toxins including tropical diseases, 17, 451–459.
  • Zhang, L. and Cui, L., 2007. A cytotoxin isolated from Agkistrodon acutus snake venom induces apoptosis via Fas pathway in A549 cells. Toxicology in vitro, 21 (6), 1095–1103.
  • Ziegler, U. and Groscurth, P., 2004. Morphological features of cell death. News in physiological sciences, 19, 124–128.

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.