Skip to main content
Erschienen in: Tumor Biology 10/2016

03.09.2016 | Original Article

Ganoderic acid targeting multiple receptors in cancer: in silico and in vitro study

verfasst von: Balraj Singh Gill, Navgeet, Sanjeev Kumar

Erschienen in: Tumor Biology | Ausgabe 10/2016

Einloggen, um Zugang zu erhalten

Abstract

Receptor tyrosine kinases (RTKs) are transmembrane high-affinity surface receptors responsible for cell migration, adhesion, apoptosis, metabolism, and cell proliferation activities in various cancers. Minute aberration in the RTK signaling modulates the downstream signaling pathways that results in cancer. Ganoderic acid is a triterpene isolated from Ganoderma lucidum, which is renowned for its therapeutics effect, especially in cancer. The present study discusses receptor-based molecular docking of insulin receptor (IR), insulin-like growth factor receptor 1 (IGFR-1), vascular endothelial growth factor receptor-1 (VEGFR-1), vascular endothelial growth factor receptor-2 (VEGFR-2), and estrogen receptor (ER) with 50 isoforms of ganoderic acid along with natural inhibitors. These receptors were assessed for toxicity (ADMET) by using Maestro 9.6 (Schrödinger Inc). The calculated docking free energy yielded an excellent dock score for the ganoderic acid when docked with proteins IR, IGFR-1, VEGFR-1, VEGFR-2, and ER, suggesting its potential in combating cancer. Protein–ligand profile highlighted the binding interactions comprising lipophilic, hydrogen bonding, pi-pi stacking interactions, and noncovalent bonding which play a pivotal role in targeting cancer. In silico studies revealed structure of ganoderic acid A as best isoforms among 50 isoforms which exhibits biological activity in liver cancer cells. Ganoderic acids A significantly decrease the viability, proliferation, and oxidative stress in a dose-dependent manner in liver cancer cells.
Literatur
2.
Zurück zum Zitat Anand SS, Gill BS. Breakthroughs in epigenetics. Pharm Tutor. 2015;3(7):16–24. Anand SS, Gill BS. Breakthroughs in epigenetics. Pharm Tutor. 2015;3(7):16–24.
3.
Zurück zum Zitat Ullrich A, Schlessinger J. Signal transduction by receptors with tyrosine kinase activity. Cell. 1990;61(2):203–12.CrossRefPubMed Ullrich A, Schlessinger J. Signal transduction by receptors with tyrosine kinase activity. Cell. 1990;61(2):203–12.CrossRefPubMed
4.
Zurück zum Zitat Negi A, Gill B. Success stories of enolate form of drugs. Pharm Tutor. 2013;1(2):45–53. Negi A, Gill B. Success stories of enolate form of drugs. Pharm Tutor. 2013;1(2):45–53.
5.
Zurück zum Zitat Zwick E, Bange J, Ullrich A. Receptor tyrosine kinase signalling as a target for cancer intervention strategies. Endocr Relat Cancer. 2001;8(3):161–73.CrossRefPubMed Zwick E, Bange J, Ullrich A. Receptor tyrosine kinase signalling as a target for cancer intervention strategies. Endocr Relat Cancer. 2001;8(3):161–73.CrossRefPubMed
7.
8.
Zurück zum Zitat Gill BS, Alex JM, Kumar S. Missing link between microRNA and prostate cancer. Tumor Biol. 2016;37(5):5683–704.CrossRef Gill BS, Alex JM, Kumar S. Missing link between microRNA and prostate cancer. Tumor Biol. 2016;37(5):5683–704.CrossRef
9.
10.
Zurück zum Zitat Negi ABG, Anand SS. Tilling: versatile reverse genetic tool. Pharma Tutor. 2014;2(1):26–32. Negi ABG, Anand SS. Tilling: versatile reverse genetic tool. Pharma Tutor. 2014;2(1):26–32.
11.
Zurück zum Zitat Hollosy F, Keri G. Plant-derived protein tyrosine kinase inhibitors as anticancer agents. Curr Med Chem Anticancer Agents. 2004;4(2):173–97.CrossRefPubMed Hollosy F, Keri G. Plant-derived protein tyrosine kinase inhibitors as anticancer agents. Curr Med Chem Anticancer Agents. 2004;4(2):173–97.CrossRefPubMed
12.
Zurück zum Zitat Gill BS, Sharma P, Kumar R, Kumar S. Misconstrued versatility of Ganoderma lucidum: a key player in multi-targeted cellular signaling. Tumor Biol. 2015;37(3):2789–804.CrossRef Gill BS, Sharma P, Kumar R, Kumar S. Misconstrued versatility of Ganoderma lucidum: a key player in multi-targeted cellular signaling. Tumor Biol. 2015;37(3):2789–804.CrossRef
13.
Zurück zum Zitat Yuen JW, Gohel MDI. Anticancer effects of Ganoderma lucidum: a review of scientific evidence. Nutr Cancer. 2005;53(1):11–7.CrossRefPubMed Yuen JW, Gohel MDI. Anticancer effects of Ganoderma lucidum: a review of scientific evidence. Nutr Cancer. 2005;53(1):11–7.CrossRefPubMed
14.
Zurück zum Zitat Gill BS, Kumar S. Differential algorithms-assisted molecular modeling-based identification of mechanistic binding of ganoderic acids. Med Chem Res. 2015;24(9):3483–93.CrossRef Gill BS, Kumar S. Differential algorithms-assisted molecular modeling-based identification of mechanistic binding of ganoderic acids. Med Chem Res. 2015;24(9):3483–93.CrossRef
15.
Zurück zum Zitat Liu J, Shiono J, Shimizu K, Kukita A, Kukita T, Kondo R. Ganoderic acid DM: anti-androgenic osteoclastogenesis inhibitor. Bioorg Med Chem Lett. 2009;19(8):2154–7.CrossRefPubMed Liu J, Shiono J, Shimizu K, Kukita A, Kukita T, Kondo R. Ganoderic acid DM: anti-androgenic osteoclastogenesis inhibitor. Bioorg Med Chem Lett. 2009;19(8):2154–7.CrossRefPubMed
16.
Zurück zum Zitat Yao X, Li G, Xu H, Lü C. Inhibition of the JAK-STAT3 signaling pathway by ganoderic acid A enhances chemosensitivity of HepG2 cells to cisplatin. Planta Med. 2012;78(16):1740–8.CrossRefPubMed Yao X, Li G, Xu H, Lü C. Inhibition of the JAK-STAT3 signaling pathway by ganoderic acid A enhances chemosensitivity of HepG2 cells to cisplatin. Planta Med. 2012;78(16):1740–8.CrossRefPubMed
17.
Zurück zum Zitat Chen N-H, Liu J-W, Zhong J-J. Ganoderic acid Me inhibits tumor invasion through down-regulating matrix metalloproteinases 2/9 gene expression. J Pharmacol Sci. 2008;108(2):212–6.CrossRefPubMed Chen N-H, Liu J-W, Zhong J-J. Ganoderic acid Me inhibits tumor invasion through down-regulating matrix metalloproteinases 2/9 gene expression. J Pharmacol Sci. 2008;108(2):212–6.CrossRefPubMed
18.
Zurück zum Zitat Hong R-L, Spohn WH, Hung M-C. Curcumin inhibits tyrosine kinase activity of p185neu and also depletes p185neu. Clin Cancer Res. 1999;5(7):1884–91.PubMed Hong R-L, Spohn WH, Hung M-C. Curcumin inhibits tyrosine kinase activity of p185neu and also depletes p185neu. Clin Cancer Res. 1999;5(7):1884–91.PubMed
19.
Zurück zum Zitat Huang YT, Hwang JJ, Lee PP, Ke FC, Huang JH, Huang CJ, et al. Effects of luteolin and quercetin, inhibitors of tyrosine kinase, on cell growth and metastasis‐associated properties in A431 cells overexpressing epidermal growth factor receptor. Br J Pharmacol. 1999;128(5):999–1010.CrossRefPubMedPubMedCentral Huang YT, Hwang JJ, Lee PP, Ke FC, Huang JH, Huang CJ, et al. Effects of luteolin and quercetin, inhibitors of tyrosine kinase, on cell growth and metastasis‐associated properties in A431 cells overexpressing epidermal growth factor receptor. Br J Pharmacol. 1999;128(5):999–1010.CrossRefPubMedPubMedCentral
20.
Zurück zum Zitat Kwon HJ, Shim JS, Kim JH, Cho HY, Yum YN, Kim SH, et al. Betulinic acid inhibits growth factor‐induced in vitro angiogenesis via the modulation of mitochondrial function in endothelial cells. Cancer Sci. 2002;93(4):417–25. Kwon HJ, Shim JS, Kim JH, Cho HY, Yum YN, Kim SH, et al. Betulinic acid inhibits growth factor‐induced in vitro angiogenesis via the modulation of mitochondrial function in endothelial cells. Cancer Sci. 2002;93(4):417–25.
21.
Zurück zum Zitat Zhang X, Mukerji R, Samadi AK, Cohen MS. Down-regulation of estrogen receptor-alpha and rearranged during transfection tyrosine kinase is associated with withaferin a-induced apoptosis in MCF-7 breast cancer cells. BMC Complement Altern Med. 2011;11(1):84.CrossRefPubMedPubMedCentral Zhang X, Mukerji R, Samadi AK, Cohen MS. Down-regulation of estrogen receptor-alpha and rearranged during transfection tyrosine kinase is associated with withaferin a-induced apoptosis in MCF-7 breast cancer cells. BMC Complement Altern Med. 2011;11(1):84.CrossRefPubMedPubMedCentral
22.
Zurück zum Zitat Zhang W, Hong D, Zhou Y, Zhang Y, Shen Q, Li J-Y, et al. Ursolic acid and its derivative inhibit protein tyrosine phosphatase 1B, enhancing insulin receptor phosphorylation and stimulating glucose uptake. BBA Gen Subjects. 2006;1760(10):1505–12.CrossRef Zhang W, Hong D, Zhou Y, Zhang Y, Shen Q, Li J-Y, et al. Ursolic acid and its derivative inhibit protein tyrosine phosphatase 1B, enhancing insulin receptor phosphorylation and stimulating glucose uptake. BBA Gen Subjects. 2006;1760(10):1505–12.CrossRef
23.
Zurück zum Zitat Mu X, Shi W, Sun L, Li H, Jiang Z, Zhang L. Pristimerin, a triterpenoid, inhibits tumor angiogenesis by targeting VEGFR2 activation. Molecules. 2012;17(6):6854–68.CrossRefPubMed Mu X, Shi W, Sun L, Li H, Jiang Z, Zhang L. Pristimerin, a triterpenoid, inhibits tumor angiogenesis by targeting VEGFR2 activation. Molecules. 2012;17(6):6854–68.CrossRefPubMed
24.
Zurück zum Zitat Jorgensen WL, Maxwell DS, Tirado-Rives J. Development and testing of the OPLS all-atom force field on conformational energetics and properties of organic liquids. J Am Chem Soc. 1996;118(45):11225–36.CrossRef Jorgensen WL, Maxwell DS, Tirado-Rives J. Development and testing of the OPLS all-atom force field on conformational energetics and properties of organic liquids. J Am Chem Soc. 1996;118(45):11225–36.CrossRef
25.
Zurück zum Zitat Jorgensen WL, Tirado-Rives J. The OPLS [optimized potentials for liquid simulations] potential functions for proteins, energy minimizations for crystals of cyclic peptides and crambin. J Am Chem Soc. 1988;110(6):1657–66.CrossRefPubMed Jorgensen WL, Tirado-Rives J. The OPLS [optimized potentials for liquid simulations] potential functions for proteins, energy minimizations for crystals of cyclic peptides and crambin. J Am Chem Soc. 1988;110(6):1657–66.CrossRefPubMed
26.
Zurück zum Zitat Shivakumar D, Williams J, Wu Y, Damm W, Shelley J, Sherman W. Prediction of absolute solvation free energies using molecular dynamics free energy perturbation and the OPLS force field. J Chem Theory Comput. 2010;6(5):1509–19.CrossRefPubMed Shivakumar D, Williams J, Wu Y, Damm W, Shelley J, Sherman W. Prediction of absolute solvation free energies using molecular dynamics free energy perturbation and the OPLS force field. J Chem Theory Comput. 2010;6(5):1509–19.CrossRefPubMed
27.
Zurück zum Zitat Repasky MP, Shelley M, Friesner RA. Flexible ligand docking with GLIDE. Curr Protoc Bioinform. 2007;8.12.1–8.12.36 Repasky MP, Shelley M, Friesner RA. Flexible ligand docking with GLIDE. Curr Protoc Bioinform. 2007;8.12.1–8.12.36
28.
Zurück zum Zitat Friesner RA, Banks JL, Murphy RB, Halgren TA, Klicic JJ, Mainz DT, et al. GLIDE: a new approach for rapid, accurate docking and scoring. 1. Method and assessment of docking accuracy. J Med Chem. 2004;47(7):1739–49.CrossRefPubMed Friesner RA, Banks JL, Murphy RB, Halgren TA, Klicic JJ, Mainz DT, et al. GLIDE: a new approach for rapid, accurate docking and scoring. 1. Method and assessment of docking accuracy. J Med Chem. 2004;47(7):1739–49.CrossRefPubMed
29.
Zurück zum Zitat Friesner RA, Murphy RB, Repasky MP, Frye LL, Greenwood JR, Halgren TA, et al. Extra precision glide: docking and scoring incorporating a model of hydrophobic enclosure for protein-ligand complexes. J Med Chem. 2006;49(21):6177–96.CrossRefPubMed Friesner RA, Murphy RB, Repasky MP, Frye LL, Greenwood JR, Halgren TA, et al. Extra precision glide: docking and scoring incorporating a model of hydrophobic enclosure for protein-ligand complexes. J Med Chem. 2006;49(21):6177–96.CrossRefPubMed
30.
Zurück zum Zitat Jorgensen WL, Duffy EM. Prediction of drug solubility from structure. Adv Drug Deliv Rev. 2002;54(3):355–66.CrossRefPubMed Jorgensen WL, Duffy EM. Prediction of drug solubility from structure. Adv Drug Deliv Rev. 2002;54(3):355–66.CrossRefPubMed
31.
Zurück zum Zitat Singh P, Bast F. High-throughput virtual screening, identification and in vitro biological evaluation of novel inhibitors of signal transducer and activator of transcription 3. Med Chem Res. 2015;24(6):2694–708.CrossRef Singh P, Bast F. High-throughput virtual screening, identification and in vitro biological evaluation of novel inhibitors of signal transducer and activator of transcription 3. Med Chem Res. 2015;24(6):2694–708.CrossRef
32.
Zurück zum Zitat Gill BS, Navgeet, Kumar S. Ganoderic acid A targeting β-catenin in Wnt signaling pathway: in silico and in vitro study. Interdisciplinar Sci Comput Life Sci. 2016. doi:10.1007/s12539-016-0182-7. Gill BS, Navgeet, Kumar S. Ganoderic acid A targeting β-catenin in Wnt signaling pathway: in silico and in vitro study. Interdisciplinar Sci Comput Life Sci. 2016. doi:10.​1007/​s12539-016-0182-7.
Metadaten
Titel
Ganoderic acid targeting multiple receptors in cancer: in silico and in vitro study
verfasst von
Balraj Singh Gill
Navgeet
Sanjeev Kumar
Publikationsdatum
03.09.2016
Verlag
Springer Netherlands
Erschienen in
Tumor Biology / Ausgabe 10/2016
Print ISSN: 1010-4283
Elektronische ISSN: 1423-0380
DOI
https://doi.org/10.1007/s13277-016-5291-8

Weitere Artikel der Ausgabe 10/2016

Tumor Biology 10/2016 Zur Ausgabe

Update Onkologie

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.