DOI QR코드

DOI QR Code

Comparative Reverse Screening Approach to Identify Potential Anti-neoplastic Targets of Saffron Functional Components and Binding Mode

  • Published : 2012.11.30

Abstract

Background: In the last two decades, pioneering research on anti-tumour activity of saffron has shed light on the role of crocetin, picrocrocin and safranal, as broad spectrum anti-neoplastic agents. However, the exact mechanisms have yet to be elucidated. Identification and characterization of the targets of bioactive constituents will play an imperative role in demystifying the complex anti-neoplastic machinery. Methods: In the quest of potential target identification, a dual virtual screening approach utilizing two inverse screening systems, one predicated on idTarget and the other on PharmMapper was here employed. A set of target proteins associated with multiple forms of cancer and ranked by Fit Score and Binding energy were obtained from the two independent inverse screening platforms. The validity of the results was checked by meticulously analyzing the post-docking binding pose of the picrocrocin with Hsp90 alpha in AutoDock. Results: The docking pose reveals that electrostatic and hydrogen bonds play the key role in inter-molecular interactions in ligand binding. Picrocrocin binds to the Hsp90 alpha with a definite orientation appropriate for nucleophilic attacks by several electrical residues inside the Hsp90-alpha ATPase catalytic site. Conclusion: This study reveals functional information about the anti-tumor mechanism of saffron bioactive constituents. Also, a tractable set of anti-neoplastic targets for saffron has been generated in this study which can be further authenticated by in vivo and in vitro experiments.

Keywords

References

  1. Abdullaev FI, Frenkel GD (1992). The effect of saffron on intracellular DNA, RNA and protein synthesis in malignant and non-malignant human cells. BioFactors, 4, 43-5.
  2. Abdullaev FI, Gonzalez de Mejia E (1997). Antitumor effect of plant lectins. Natural Toxins, 5, 157-3.
  3. Abdullaev FI, Gonzalez de Mejia E (1995-1996). Inhibition of colony formation of Hela cells by naturally occurring and synthetic agents. BioFactors, 5, 133-8.
  4. Abdullaev FI, Riveron NL, Rotenburd BV, et al (2000). Saffron as chemopreventive agent. In: Wenyi T, Ed. Food of 21st century: food and resource technology environment. China: Ligth Industry Press, 185-5.
  5. Abdullaev FI (1993). Biological effects of saffron. Biofactors, 4, 83-6.
  6. Abdullaev FI (2002). Cancer chemopreventive and tumoricidal properties of saffron (Crocus sativus L.). Exp Biol Med Maywood, 227, 20-5. https://doi.org/10.1177/153537020222700104
  7. Abdullaev FI (1994). Inhibitory effect of crocetin on intracellular nucleic acid and protein synthesis in malignant cells. Toxicol Lett, 40, 243-1.
  8. Amin A, Hamza AA, Bajbouj K, et al (2011). Saffron: a potential candidate for a novel anticancer drug against hepatocellular carcinoma. Hepatology, 1-46.
  9. Chang VC, Lin YL, Lee MJ, et al (1996). Inhibitory effect of crocetin on benzo(a)pyrece genotoxicity and neoplastic transformation in C3H1OT1/2 cells. Anticancer Res, 765, 3603-8.
  10. Chen YZ, Ung CY (2001). Prediction of potential toxicity and side effect protein targets of a small molecule by a ligandprotein inverse docking approach. J Mol Graph Model, 20, 199-18. https://doi.org/10.1016/S1093-3263(01)00109-7
  11. Chryssanthi DG, Dedes PG, Karamanos NK, et al (2011). Crocetin inhibits invasiveness of MDA-MB-231 breast cancer cells via downregulation of matrix metalloproteinases. Planta Med, 77, 146-1. https://doi.org/10.1055/s-0030-1250178
  12. Creighton DJ, Zheng ZB, Holewinski R, et al (2003). Glyoxalase I inhibitors in cancer chemotherapy. Biochem Soc Trans, 31, 1378-2. https://doi.org/10.1042/BST0311378
  13. Dufresne C, Cormier F, Dorion S (1997). In vitro formation of crocetin glucosyl esters by Crocus sativus callus extract. Planta Med, 63, 150-3. https://doi.org/10.1055/s-2006-957633
  14. El Daly ES (1998). Protective effect of cysteine and vitamin E, Crocus sativus and Nigella sativa extracts on cisplatininduced toxicity in rats. J Pharm Belg, 53, 93-5.
  15. Escribano J, Alonso GL, Coca-Prados M, et al (1996). Crocin, safranal and picocrocin from saffron (Crocus sativus L.) inhibit the growth of human cancer cells in vitro. Cancer Lett, 100, 23-30. https://doi.org/10.1016/0304-3835(95)04067-6
  16. Escribano J, Diaz-Guerra MJ, Riese HH, et al (1999). In vitro activation of macrophages from corms of Crocus sativus L. Cancer Lett, 144, 107-4. https://doi.org/10.1016/S0304-3835(99)00211-6
  17. Escribano J, Diaz-Guerra MJ, Riese HH, et al (2000). The cytotoxic effect of glucoconjugate extracted from corms of saffron plant (Crocus sativus) on human cell lines in culture. Planta Med, 66, 157-2. https://doi.org/10.1055/s-2000-11127
  18. Escribano J, Piqueras A, Medina J, et al (1999). Production of a cytotoxic proteoglycan using callus culture of saffron corms (Crocus sativus L.). J Biotechnol, 73, 53-9. https://doi.org/10.1016/S0168-1656(99)00125-X
  19. Escribano J, Rios J, Fernandez JA (1999). Isolation and cytotoxic properties of novel glycoconjugate from corms of saffron plant (Crocus sativus L.). Biochim Biophys Acta, 1426, 217-22. https://doi.org/10.1016/S0304-4165(98)00149-4
  20. Escribano J, Rubio A, Alvarez-Orti M, et al (2000). Purification and characterization of mannan-binding lectin specifically expressed in corms of saffron plant (Crocus sativus L.). J Agric Food Chem, 48, 457-3. https://doi.org/10.1021/jf990735r
  21. Floryk D, Thompson TC (2008). Antiproliferative effects of AVN944, a novel inosine 5-monophosphate dehydrogenase inhibitor, in prostate cancer cells. Int J Cancer, 123, 2294-2. https://doi.org/10.1002/ijc.23788
  22. Garcia-Olmo DC, Riese HH, Escribano J, et al (1999). Effects of long-term treatment of colon adenocarcinoma with crocin, a carotenoid from saffron (Crocus sativus L.): an experimental study in the rats. Nutr Cancer, 35, 120-6. https://doi.org/10.1207/S15327914NC352_4
  23. Hajduk PJ, Huth JR, Tse C (2005). Predicting protein druggability. Drug Discov Today, 10, 1675-82. https://doi.org/10.1016/S1359-6446(05)03624-X
  24. Herman SEM, Gordon AL, Hertlein E, et al (2011). Tyrosine kinase represents a promising therapeutic target for treatment of chronic lymphocytic leukemia and is effectively targeted by PCI-32765. Blood, 117, 6287-6. https://doi.org/10.1182/blood-2011-01-328484
  25. Hetschko H, Voss V, Senft C, et al (2008). BH3 mimetics reactivate autophagic cell death in anoxia-resistant malignant glioma cells. Neoplasia, 10, 873-85. https://doi.org/10.1593/neo.07842
  26. Huang CM, Elmets CA, Tang DC, et al (2004). Proteomics reveals that proteins expressed during the early stage of Bacillus anthracis infection are potential targets for the development of vaccines and drugs. Genomics Proteomics Bioinforma, 3, 143-1.
  27. Jones JO, Bolton EC, Huang Y, et al (2009). Non-competitive androgen receptor inhibition in vitro and in vivo. PNAS, 106, 7233-8. https://doi.org/10.1073/pnas.0807282106
  28. Kock N, Kasmieh R, Weissleder R, et al (2007). Tumor therapy mediated by lentiviral expression of shBcl-2 and S-TRAIL. Neoplasia, 9, 435-42. https://doi.org/10.1593/neo.07223
  29. Li H, Gao Z, Kang L, et al (2006). TarFisDock: a web server for identifying drug targets with docking approach. Nucleic Acids Res, 34, 219-24. https://doi.org/10.1093/nar/gkl114
  30. Li R, Boehm AL, Miranda MB, et al (2007). Targeting antiapoptotic Bcl-2 family members with cell-permeable BH3 peptides induces apoptosis signaling and death in head and neck squamous cell carcinoma cells. Neoplasia, 9, 801-11. https://doi.org/10.1593/neo.07394
  31. Lin JH, Lu AY (1997). Role of pharmacokinetics and metabolism in drug discovery and development. Pharmacol Rev, 49, 403-49.
  32. Liu X, Ouyang S, Yu B, et al (2010). PharmMapper Server: a web server for potential drug target identification via pharmacophore mapping approach. Nucleic Acids Res, 38, 609-14. https://doi.org/10.1093/nar/gkq300
  33. Majewski S, Skopinska M, Marczak M, et al (1996). Vitamin D3 is a potent inhibitor of tumor cell-induced angiogenesis. J Investig Dermatol Symp Proc, 1, 97-1.
  34. Martin G, Goh E, Neff AW (2002). Evaluation of the developmental toxicity of crocetin on Xenopus. Food Chem Toxicol, 40, 959-4. https://doi.org/10.1016/S0278-6915(02)00040-6
  35. Martin SA, Lord CJ, Ashworth A (2010). Therapeutic Targeting of the DNA Mismatch Repair Pathway. Clin. Cancer Res, 16, 5107. https://doi.org/10.1158/1078-0432.CCR-10-0821
  36. Molnar J, Szabo D, Pusztai R, et al (2000). Membrane associated antitumor effects of crocine-, ginseniside-and cannabinoid derivates. Anticancer Res, 20, 861-7.
  37. Morjani H, Tarantilis P, Polissiou M, et al (1990). Growth inhibition and induction of crythroid differentiation activity by crocin, dimethylcrocetine and carotene on K562 tumor cells. Anticancer Res, 10, 1398-6.
  38. Morris GM, Goodsell DS, Halliday RS, et al (1998). Automated docking using a lamarckian genetic algorithm and and empirical binding free energy function. J Comput Chem, 19, 1639-62. https://doi.org/10.1002/(SICI)1096-987X(19981115)19:14<1639::AID-JCC10>3.0.CO;2-B
  39. Nair SC, Kurumboor SK, Hasegawa JH (1995). Saffron chemoprevention in biology and medicine: a review. Cancer Biother, 10, 257-4. https://doi.org/10.1089/cbr.1995.10.257
  40. Nair SC, Panikkar B, Panikkar KR (1991). Antitumor activity of saffron (Crocus sativus). Cancer Lett, 57, 109-14. https://doi.org/10.1016/0304-3835(91)90203-T
  41. Nair SC, Salomi MJ, Pannikar. B, et al (1991). Modulatory effects of the extracts of saffron and Nigela sativa against cisplatinum induced toxicity in mice. J Ethnopharmacol, 31, 75-3. https://doi.org/10.1016/0378-8741(91)90146-5
  42. Nair SC, Salomi MJ, Varghese CD, et al (1992). Effect of saffron on thymocute proliferation, intracellular gluthation levels and its antitumor activity. BioFactors, 1, 51-4.
  43. Nair SC, Varghese CD, Pannikar KR, et al (1994). Effects of saffron on vitamin A levels and its antitumor activity on the growth of solid tumors in mice. Int J Pharmacog, 32, 105-14. https://doi.org/10.3109/13880209409082981
  44. Neckers L (2006). Chaperoning oncogenes: Hsp90 as a target of geldanamycin. Handb Exp Pharmacol, 172, 259-77. https://doi.org/10.1007/3-540-29717-0_11
  45. Oda Y, Tatsumi Y (1993). New lectins from bulbs of Crocus sativus. Biol Pharm Bull, 16, 978-1. https://doi.org/10.1248/bpb.16.978
  46. Persidis A (1998). Proteomics: an ambitious drug development platform attempts to link gene sequence to expressed phenotype under various physiological states. Nat Biotechnol, 16, 393-4. https://doi.org/10.1038/nbt0498-393
  47. Pons JL, Labesse G (2009). -TOME-2: a new pipeline for comparative modeling of protein-ligand complexes. Nucl Acids Res, 37, 485-91. https://doi.org/10.1093/nar/gkp368
  48. Premkumar K, Abraham SK, Santhiya ST, et al (2001). Inhibition of genotoxicity by saffron (Crocus sativus L.) in mice. Drug Chem Toxicol, 24, 421-8. https://doi.org/10.1081/DCT-100106266
  49. Reed JC (2003). Apoptosis-targeted therapies for cancer. Cancer Cell, 1, 17-2.
  50. Salomi MJ, Nair SC, Panikkar PR (1991). Cytotoxicity and nonmutagenicity of Nigela sativa and saffron (Crocus sativus) in vitro. Proc Ker Sci Congr, 5, 244.
  51. Salomi MJ, Nair SC, Panikkar PR (1990). Inhibitory effects of Nigela sativa and saffron (Crocus sativus) on chemical carcinogenesis in mice and its non-mutagenic activity. Proc Ker Sci Congr, 3, 125-6.
  52. Salomi MJ, Nair SC, Panikkar PR (1991). Inhibitory effects of Nigela sativa and saffron (Crocus sativus) on chemical carcinogenesis in mice. Nutr Cancer, 16, 67-2. https://doi.org/10.1080/01635589109514142
  53. Smith TAD (1998). Carotenoids and cancer: prevention and potential therapy. Br J Biomed Sci, 55, 268-5.
  54. Takashi I (1992). Antioxidative property of the anthraquinonepigment from the cultured cells of saffron, and enzymatic comparison between some cultured cells. Shokubutsu Soshiki Baiyo, 9, 51-3.
  55. Tanaka T, De Luca LM (2009). Therapeutic Potential of "Rexinoids" in cancer prevention and treatment. Cancer Res, 69, 4945. https://doi.org/10.1158/0008-5472.CAN-08-4407
  56. Tang XH, Gudas LJ (2011). Retinoids-Retinoic Acid Receptors and Cancer. Annual Review of Pathology: Mechanisms of Disease, 6, 345-4. https://doi.org/10.1146/annurev-pathol-011110-130303
  57. Tarantilis PA, Morjani H, Polissiou M, et al (1994). Inhibition of growth and induction of differentiation promyelocytic leukemia (HL-60) by carotenoids from Crocus sativus L. Anticancer Res, 14, 1913-8.
  58. Thornalley PJ (2003). Glyoxalase I - structure, function and a critical role in the enzymatic defence against glycation. Biochem Soc Trans, 31, 1343-8. https://doi.org/10.1042/BST0311343
  59. Trott O, Olson AJ (2010). AutoDock Vina: improving the speed and accuracy of docking with a new scoring function, efficient optimization and multithreading. J Comput Chem, 31, 455-1.
  60. Wang JC, Chu PY, Chen CM, et al (2012). idTarget: a webserver for identifying protein targets of small chemical molecules with robust scoring functions and a divide-and-conquer docking approach. Nucleic Acids Research, 40, 393-9. https://doi.org/10.1093/nar/gks496
  61. Yang R, Vernon K, Thomas A, et al (2011). Crocetin Reduces Activation of Hepatic Apoptotic Pathways and Improves Survival in Experimental Hemorrhagic Shock. JPEN J Parenter Enteral Nutr, 35, 107-3. https://doi.org/10.1177/0148607110374058
  62. Yanyan L, Tao Z, Duxin S (2009). New developments in Hsp90 inhibitors as anti-cancer therapeutics: mechanisms, clinical perspective and more potential. Drug Resist Updat, 12, 17-7. https://doi.org/10.1016/j.drup.2008.12.002
  63. Yu XC, Miranda M, Liu Z, et al (2010). Novel potent inhibitors of deoxycytidine kinase identified and compared by multiple assays. J Biomol Screen, 15, 72-9. https://doi.org/10.1177/1087057109353604
  64. Zheng R, Chen TS, Lu T (2011). A Comparative Reverse Docking Strategy to Identify Potential Antineoplastic Targets of Tea Functional Components and Binding Mode. Int J Mol Sci, 12, 5200-12. https://doi.org/10.3390/ijms12085200
  65. Zhou CH, Xiang M, He SY, et al (2010). Protein kinase C pathway is involved in the inhibition by crocetin of vascular smooth muscle cells proliferation. Phytotherapy Res, 24, 1680-6. https://doi.org/10.1002/ptr.3194

Cited by

  1. Identification of Proapoptopic, Anti-Inflammatory, Anti-Proliferative, Anti-Invasive and Anti-Angiogenic Targets of Essential Oils in Cardamom by Dual Reverse Virtual Screening and Binding Pose Analysis vol.14, pp.6, 2013, https://doi.org/10.7314/APJCP.2013.14.6.3735
  2. Identification of a Potential Anticancer Target of Danshensu by Inverse Docking vol.15, pp.1, 2014, https://doi.org/10.7314/APJCP.2014.15.1.111
  3. A Potential Target of Tanshinone IIA for Acute Promyelocytic Leukemia Revealed by Inverse Docking and Drug Repurposing vol.15, pp.10, 2014, https://doi.org/10.7314/APJCP.2014.15.10.4301
  4. Hsp90 Inhibitors, Part 2: Combining Ligand-Based and Structure-Based Approaches for Virtual Screening Application vol.54, pp.3, 2014, https://doi.org/10.1021/ci400760a
  5. Identification of Anti-Cancer Targets of Eco-Friendly Waste Punica granatum Peel by Dual Reverse Virtual Screening and Binding Analysis vol.15, pp.23, 2014, https://doi.org/10.7314/APJCP.2014.15.23.10345
  6. Heat-Shock Protein 90 (Hsp90) as Anticancer Target for Drug Discovery: An Ample Computational Perspective vol.86, pp.5, 2015, https://doi.org/10.1111/cbdd.12582
  7. Anti-tumor effects of crocetin and related molecular targets vol.233, pp.3, 2017, https://doi.org/10.1002/jcp.25953
  8. Reverse Screening Methods to Search for the Protein Targets of Chemopreventive Compounds vol.6, pp.2296-2646, 2018, https://doi.org/10.3389/fchem.2018.00138