DOI QR코드

DOI QR Code

An in silico Appraisal to Identify High Affinity Anti-Apoptotic Synthetic Tetrapeptide Inhibitors Targeting the Mammalian Caspase 3 Enzyme

  • Kelotra, Seema (M.B Khalsa College) ;
  • Jain, Meeta (School of Biochemistry, Devi Ahilya University, Takshashila Campus) ;
  • Kelotra, Ankit (School of Biochemistry, Devi Ahilya University, Takshashila Campus) ;
  • Jain, Ish (In silico Research Laboratory, Eminent Biosciences) ;
  • Bandaru, Srinivas (Institute of Genetics and Hospital for Genetic Diseases, Osmania University) ;
  • Nayarisseri, Anuraj (In silico Research Laboratory, Eminent Biosciences) ;
  • Bidwai, Anil (Index medical College)
  • 발행 : 2015.01.06

초록

Apoptosis is a general phenomenon of all multicellular organisms and caspases form a group of important proteins central to suicide of cells. Pathologies like cancer, Myocardial infarction, Stroke, Sepsis, Alzheimer's, Psoriasis, Parkinson and Huntington diseases are often associated with change in caspase 3 mediated apoptosis and therefore, caspases may serve as potential inhibitory targets for drug development. In the present study, two series of synthetic acetylated tetrapeptides containing aldehyde and fluromethyl keto groups respectively at the C terminus were proposed. All these compounds were evaluated for binding affinity against caspase 3 structure. In series 1 compound Ac-DEHD-CHO demonstrated appreciable and high binding affinity (Rerank Score: -138.899) against caspase 3. While in series 2 it was Ac-WEVD-FMK which showed higher binding affinity (Rerank Score: -139.317). Further these two compounds met ADMET properties and demonstrated to be non-toxic.

키워드

참고문헌

  1. An FF, Liu YC, Zhang WW, Liang, L (2013). Dihydroartemisinine enhances dictamnine-induced apoptosis via a caspase dependent pathway in human lung adenocarcinoma A549 cells. Asian Pac J Cancer Prev, 14, 5895-900. https://doi.org/10.7314/APJCP.2013.14.10.5895
  2. Arockiaraj J, Gnanam AJ, Muthukrishnan D, et al (2013). An upstream initiator caspase 10 of snakehead murrel Channa striatus, containing DED, p20 and p10 subunits: Molecular cloning, gene expression and proteolytic activity. Fish Shellfish Immunol, 34, 505-13. https://doi.org/10.1016/j.fsi.2012.11.040
  3. Becker JW, Rotonda J, Soisson SM, et al (2004). Reducing the peptidyl features of caspase-3 inhibitors: a structural analysis. J Med Chem, 47, 2466-74. https://doi.org/10.1021/jm0305523
  4. Belizario JE, Alves J, Garay-Malpartida M, Occhiucci JM (2008). Coupling caspase cleavage and proteasomal degradation of proteins carrying PEST motif. Curr Protein Pept Sci, 9, 210-20. https://doi.org/10.2174/138920308784534023
  5. Chai J, Shiozaki E, Srinivasula SM, et al (2001). Structural basis of caspase-7 inhibition by XIAP. Cell, 104, 769-80. https://doi.org/10.1016/S0092-8674(01)00272-0
  6. Concha NO, Abdel-Meguid SS (2002). Controlling apoptosis by inhibition of caspases. Curr Med Chem, 9, 713-26. https://doi.org/10.2174/0929867023370761
  7. Danial NN, Korsmeyer SJ (2004). Cell death: critical control points. Cell, 116, 205-19. https://doi.org/10.1016/S0092-8674(04)00046-7
  8. Denault JB, Salvesen GS (2002). Caspases: keys in the ignition of cell death. Chem Rev, 102, 4489-500. https://doi.org/10.1021/cr010183n
  9. Fischer U, Schulze-Osthoff K (2005). Apoptosis-based therapies and drug targets. Cell Death Differ, 12, 942-61. https://doi.org/10.1038/sj.cdd.4401556
  10. Gomez-Vicente V, Donovan M, Cotter TG (2005). Multiple death pathways in retina-derived 661W cells following growth factor deprivation: crosstalk between caspases and calpains. Cell Death Differ, 12, 796-804. https://doi.org/10.1038/sj.cdd.4401621
  11. Grawert MA, Groll M (2012). Exploiting nature's rich source of proteasome inhibitors as starting points in drug development. Chem Commun (Camb), 48, 1364-78. https://doi.org/10.1039/C1CC15273D
  12. Hardy B, Douglas N, Helma C, et al (2010). Collaborative development of predictive toxicology applications. J Cheminform, 2, 1-29. https://doi.org/10.1186/1758-2946-2-1
  13. Hengartner MO (2000). The biochemistry of apoptosis. Nature, 407, 770-6. https://doi.org/10.1038/35037710
  14. Huang Y, Park YC, Rich RL, et al (2001). Structural basis of caspase inhibition by XIAP: differential roles of the linker versus the BIR domain. Cell, 104, 781-90.
  15. Kuhn D, Orlowski R, Bjorklund C (2011). Second generation proteasome inhibitors: carfilzomib and immunoproteasomespecific inhibitors (IPSIs). Curr Cancer Drug Targets, 11, 285-95. https://doi.org/10.2174/156800911794519725
  16. Lavrik IN, Golks A, Krammer PH (2005). Caspases: pharmacological manipulation of cell death. J Clin Invest, 115, 2665-72. https://doi.org/10.1172/JCI26252
  17. Le GT, Abbenante G, Madala PK, Hoang HN, Fairlie DP (2006). Organic azide inhibitors of cysteine proteases. J Am Chem Soc, 128, 12396-7. https://doi.org/10.1021/ja0637649
  18. Leist M, Jaattela M (2001). Four deaths and a funeral: from caspases to alternative mechanisms. Nat Rev Mol Cell Biol, 2, 589-98.
  19. Li SX, Chai L, Cai ZG, et al (2012). Expression of survivin and caspase 3 in oral squamous cell carcinoma and peritumoral tissue. Asian Pac J Cancer Prev, 13, 5027-31. https://doi.org/10.7314/APJCP.2012.13.10.5027
  20. Liu DD, Ye YL, Zhang J, et al (2014). Distinct pro-apoptotic properties of Zhejiang saffron against human lung cancer via a caspase-8-9-3 cascade. Asian Pac J Cancer Prev, 15, 6075. https://doi.org/10.7314/APJCP.2014.15.15.6075
  21. Miao EA, Rajan JV, Aderem A (2011). Caspase-1-induced pyroptotic cell death. Immunol Rev, 243, 206-14. https://doi.org/10.1111/j.1600-065X.2011.01044.x
  22. Nayarisseri A (2013). In silico investigations on HSP90 and its inhibition for the therapeutic prevention of breast cancer. J Pharm Res, 7, 150-6. https://doi.org/10.1016/j.jopr.2013.02.020
  23. Noy N (2010). Between death and survival: retinoic acid in regulation of apoptosis. Annu Rev Nutr, 30, 201-17. https://doi.org/10.1146/annurev.nutr.28.061807.155509
  24. Powers JC, Asgian JL, Ekici OD, James KE (2002). Irreversible inhibitors of serine, cysteine, and threonine proteases. Chem Rev, 102, 4639-750. https://doi.org/10.1021/cr010182v
  25. Sankari SL, Masthan KM, Babu NA, Bhattacharjee T, Elumalai M (2012). Apoptosis in cancer - an update. Asian Pac J Cancer Prev, 13, 4873-8. https://doi.org/10.7314/APJCP.2012.13.10.4873
  26. Shi Y (2002). Mechanisms of caspase activation and inhibition during apoptosis. Mol Cell, 9, 459-70. https://doi.org/10.1016/S1097-2765(02)00482-3
  27. Shiozaki EN, Shi Y (2004). Caspases, IAPs and Smac/DIABLO: mechanisms from structural biology. Trends Biochem Sci, 29, 486-94. https://doi.org/10.1016/j.tibs.2004.07.003
  28. Takai N, Kira N, Ishii T, et al (2012). Bufalin, a traditional oriental medicine, induces apoptosis in human cancer cells. Asian Pac J Cancer Prev, 13, 399-402. https://doi.org/10.7314/APJCP.2012.13.1.399
  29. Turk B, Stoka V (2007). Protease signalling in cell death: caspases versus cysteine cathepsins. FEBS Lett, 581, 2761-7. https://doi.org/10.1016/j.febslet.2007.05.038
  30. Vuree S (2013). Pharmacogenomics of drug resistance in Breast Cancer Resistance Protein (BCRP) and its mutated variants. J Pharm Res, 7, 791-8.
  31. Zou X, Liu SL, Zhou JY, et al (2012). Beta-asarone induces lovo colon cancer cell apoptosis by up-regulation of caspases through a mitochondrial pathway in vitro and in vivo. Asian Pac J Cancer Prev, 13, 5291. https://doi.org/10.7314/APJCP.2012.13.10.5291