Author(s):
Neha Sylvia Walter, Sanchit Dora, Jasmeet Kaur
Email(s):
walterneha@gmail.com , sanchitdora5@gmail.com , jasmeet23k@gmail.com
DOI:
10.52711/0974-360X.2025.00779
Address:
Neha Sylvia Walter, Sanchit Dora, Jasmeet Kaur
Department of Biophysics, Postgraduate Institute of Medical education and Research (PGIMER), Chandigarh, India.
*Corresponding Author
Published In:
Volume - 18,
Issue - 11,
Year - 2025
ABSTRACT:
Glycogen synthase kinase 3 (GSK3) is a pivotal serine/threonine kinase regulating key process such as glycogen synthase, apoptosis and cell-cycle progression. The enzyme is a significant participant of PI3K/PTEN/AKT/GSK3/mTORC1 pathway and implicated in various human cancers. Of the two isoforms a and ß in mammals GSK3ß is extensively studied in cancer than GSK3a. The present study demonstrated expression of GSK3? mRNA to be remarkably associated with poor RFS (Relapse Free Survival) in breast cancer patients using KM plotter (Kaplan-Meier plotter). TMN plot revealed a significant (p<0.0001) difference in GSK3A gene expression among the tumour and normal tissue as determined by Wilcoxon Signed Rank Test using both gene chip and RNA sequence data. ROC analysis also revealed the predictive significance of GSK3? in patients administered endocrine therapy, anti-HER2 therapy and chemotherapy. Considering the importance of this enzyme, potential GSK3a ligands were also identified by molecular docking and ADMET analysis. Four compounds viz.(+/)-Synephrine, LTURM34, 2-Dehydro-3-deoxy-L-fuconate, 603288-22-8 (LY2090314) were found to target GSK3a with favourable pharmacological properties and were found to be non-toxic. These compounds can further be explored for their potential anticancer activity with GSK3a as the primary target.
Cite this article:
Neha Sylvia Walter, Sanchit Dora, Jasmeet Kaur. Identification of Potential GSK3α Ligands and Prediction of their Drug-like Properties. Research Journal Pharmacy and Technology. 2025;18(11):5401-0. doi: 10.52711/0974-360X.2025.00779
Cite(Electronic):
Neha Sylvia Walter, Sanchit Dora, Jasmeet Kaur. Identification of Potential GSK3α Ligands and Prediction of their Drug-like Properties. Research Journal Pharmacy and Technology. 2025;18(11):5401-0. doi: 10.52711/0974-360X.2025.00779 Available on: https://www.rjptonline.org/AbstractView.aspx?PID=2025-18-11-41
5. REFERENCES:
1. Jayashree V, Velraj M. Breast Cancer and various Prognostic Biomarkers for the diagnosis of the disease: A Review. Research J Pharm and Tech. 2017; 10: 3211–6. https://doi.org/10.5958/0974-360X.2017.00570.4.
2. Karale PA, Karale MA, Utikar MC. Advanced Molecular Targeted Therapy in Breast Cancer. Res J Pharmacology and Pharmacodynamics. 2018; 10: 29–37. https://doi.org/10.5958/2321-5836.2018.00006.X.
3. Asaad RA, Abdullah SS. Breast Cancer Subtypes (BCSs) Classification according to Hormone Receptor Status: Identification of patients at High Risk in Jableh- Syria. Research J Pharm and Tech. 2018; 11: 3703–10. https://doi.org/10.5958/0974-360X.2018.00680.7.
4. Patel P, Woodgett JR. Glycogen Synthase Kinase 3: A Kinase for All Pathways? Curr Top Dev Biol. 2017; 123: 277–302. https://doi.org/10.1016/bs.ctdb.2016.11.011.
5. Brion JP, Anderton BH, Authelet M, Dayanandan R, Leroy K, Lovestone S, et al. Neurofibrillary tangles and tau phosphorylation. Biochem Soc Symp. 2001: 81–8. https://doi.org/10.1042/bss0670081.
6. Shin K, Kim KH, Yoon MS, Suh DS, Lee JY, Kim A, et al. Expression of Interactive Genes Associated with Apoptosis and Their Prognostic Value for Ovarian Serous Adenocarcinoma. Adv Clin Exp Med. 2016; 25: 513–21. https://doi.org/10.17219/acem/62540.
7. Naruse K, Ueno M, Satoh T, Nomiyama H, Tei H, Takeda M, et al. A YAC contig of the human CC chemokine genes clustered on chromosome 17q11.2. Genomics. 1996; 34: 236–40. https://doi.org/10.1006/geno.1996.0274.
8. Gillitzer R, Goebeler M. Chemokines in cutaneous wound healing. J Leukoc Biol. 2001; 69: 513–21.
9. Goodyear A, Jones A, Troyer R, Bielefeldt-Ohmann H, Dow S. Critical protective role for MCP-1 in pneumonic Burkholderia mallei infection. J Immunol. 2010; 184: 1445–54. https://doi.org/10.4049/jimmunol.0900411.
10. Fujimoto H, Sangai T, Ishii G, Ikehara A, Nagashima T, Miyazaki M, et al. Stromal MCP-1 in mammary tumors induces tumor-associated macrophage infiltration and contributes to tumor progression. Int J Cancer. 2009; 125: 1276–84. https://doi.org/10.1002/ijc.24378.
11. Metcalfe C, Bienz M. Inhibition of GSK3 by Wnt signalling--two contrasting models. J Cell Sci. 2011; 124: 3537–44. https://doi.org/10.1242/jcs.091991.
12. Price MA, Kalderon D. Proteolysis of the Hedgehog signaling effector Cubitus interruptus requires phosphorylation by Glycogen Synthase Kinase 3 and Casein Kinase 1. Cell. 2002; 108: 823–35. https://doi.org/10.1016/s0092-8674(02)00664-5.
13. Gotschel F, Kern C, Lang S, Sparna T, Markmann C, Schwager J, et al. Inhibition of GSK3 differentially modulates NF-κB, CREB, AP-1 and β-catenin signaling in hepatocytes, but fails to promote TNF-α-induced apoptosis. Experimental Cell Research. 2008; 314: 1351–66. https://doi.org/10.1016/j.yexcr.2007.12.015.
14. Hua F, Zhou J, Liu J, Zhu C, Cui B, Lin H, et al. Glycogen synthase kinase-3beta negatively regulates TGF-beta1 and Angiotensin II-mediated cellular activity through interaction with Smad3. Eur J Pharmacol. 2010; 644: 17–23. https://doi.org/10.1016/j.ejphar.2010.06.042.
15. Fang X, Yu SX, Lu Y, Bast RCJ, Woodgett JR, Mills GB. Phosphorylation and inactivation of glycogen synthase kinase 3 by protein kinase A. Proc Natl Acad Sci U S A 2000; 97: 11960–5. https://doi.org/10.1073/pnas.220413597.
16. Kaidanovich-Beilin O, Woodgett JR. GSK-3: Functional insights from cell biology and animal models. Front Mol Neurosci. 2011; 4.
17. Hoxhaj G, Manning BD. The PI3K–AKT network at the interface of oncogenic signalling and cancer metabolism. Nature Reviews Cancer. 2020; 20: 74–88. https://doi.org/10.1038/s41568-019-0216-7.
18. Nazio F, Strappazzon F, Antonioli M, Bielli P, Cianfanelli V, Bordi M, et al. mTOR inhibits autophagy by controlling ULK1 ubiquitylation, self-association and function through AMBRA1 and TRAF6. Nature Cell Biology. 2013; 15: 406–16. https://doi.org/10.1038/ncb2708.
19. Morita M, Gravel S-P, Chénard V, Sikström K, Zheng L, Alain T, et al. mTORC1 Controls Mitochondrial Activity and Biogenesis through 4E-BP-Dependent Translational Regulation. Cell Metabolism. 2013; 18: 698–711. https://doi.org/10.1016/j.cmet.2013.10.001.
20. Morita M, Prudent J, Basu K, Goyon V, Katsumura S, Hulea L, et al. mTOR Controls Mitochondrial Dynamics and Cell Survival via MTFP1. Mol Cell 2017; 67: 922-935.e5. https://doi.org/10.1016/j.molcel.2017.08.013.
21. Kaur J. Prognostic Effect of Glycogen Synthase Kinase 3A (GSK3A) mRNA Expression in Breast Cancer Patients. American Journal of Biomedical Sciences. 2022; 14: 58. https://doi.org/10.5099/aj220200058.
22. Bartha Á, Győrffy B. TNMplot.com: A Web Tool for the Comparison of Gene Expression in Normal, Tumor and Metastatic Tissues. Int J Mol Sci. 2021; 22. https://doi.org/10.3390/ijms22052622.
23. Gyorffy B. Survival analysis across the entire transcriptome identifies biomarkers with the highest prognostic power in breast cancer. Computational and Structural Biotechnology Journal. 2021; 19: 4101–9. https://doi.org/10.1016/j.csbj.2021.07.014.
24. Győrffy B. Discovery and ranking of the most robust prognostic biomarkers in serous ovarian cancer. Geroscience. 2023; 45: 1889–98. https://doi.org/10.1007/s11357-023-00742-4.
25. Kovacs SA, Fekete JT, Győrffy B. Predictive biomarkers of immunotherapy response with pharmacological applications in solid tumors. Acta Pharmacol Sin. 2023; 44: 1879–89. https://doi.org/10.1038/s41401-023-01079-6.
26. Fekete JT, Győrffy B. ROCplot.org: Validating predictive biomarkers of chemotherapy/hormonal therapy/anti-HER2 therapy using transcriptomic data of 3,104 breast cancer patients. Int J Cancer. 2019; 145: 3140–51. https://doi.org/10.1002/ijc.32369.
27. Parameswari P, Devika R. In silico Molecular Docking Studies of Quercetin Compound against Anti-inflammatory and Anticancer Proteins. Research J Pharm and Tech. 2019; 12: 5305–9. https://doi.org/10.5958/0974-360X.2019.00919.3.
28. Zadorozhnii PV, Pokotylo IO, Kiselev VV, Okhtina OV, Kharchenko AV. Molecular Docking Studies of N-(((5-Aryl-1,3,4-oxadiazol-2-yl)amino)methyl)- and N-(2,2,2-Trichloro-1-((5-aryl-1,3,4-oxadiazol-2-yl)amino)ethyl)carboxamides as Potential Inhibitors of GSK-3β. Research J Pharm and Tech. 2019; 12: 523–30. https://doi.org/10.5958/0974-360X.2019.00092.1.
29. Daina A, Michielin O, Zoete V. SwissADME: a free web tool to evaluate pharmacokinetics, drug-likeness and medicinal chemistry friendliness of small molecules. Scientific Reports. 2017; 7: 42717. https://doi.org/10.1038/srep42717.
30. Yang H, Lou C, Sun L, Li J, Cai Y, Wang Z, et al. admetSAR 2.0: web-service for prediction and optimization of chemical ADMET properties. Bioinformatics. 2019; 35: 1067–9. https://doi.org/10.1093/bioinformatics/bty707.
31. Banerjee P, Eckert AO, Schrey AK, Preissner R. ProTox-II: a webserver for the prediction of toxicity of chemicals. Nucleic Acids Res. 2018; 46: W257–63. https://doi.org/10.1093/nar/gky318.
32. Siegel RL, Miller KD, Wagle NS, Jemal A. Cancer statistics, 2023. CA: A Cancer Journal for Clinicians. 2023; 73: 17–48. https://doi.org/10.3322/caac.21763.
33. Hariramani N, Jayanthi S. A Systematic Review of Intrinsic Factors and its Influence in Breast Cancer. Research J Pharm and Tech. 2018; 11: 3543–6.
34. Aloss K, Jdeed S, Alshehabi Z. Detecting the role of CCR7-CCL21/CCL19 axis in Breast Cancer Progression and lymph node Metastasis Incidence. Research J Pharm and Tech. 2018; 11: 231–5. https://doi.org/10.5958/0974-360X.2018.00043.
35. Krishnaveni K, Jose R, Sumitha S, Teena J, Shanmuga SR, Sambathkumar R. A Study on Socio Demographic and Associated Risk Factors for Cancer Patients in Private Cancer Hospital, Bangalore, India. Research J Pharm and Tech. 2018; 11: 677–80. https://doi.org/10.5958/0974-360X.2018.00127.0.
36. Yi M, Ryu YM. The Effects of Breast Health Education in Women with Breast Cancer. Research J Pharm and Tech. 2017; 10: 2295–302. https://doi.org/10.5958/0974-360X.2017.00407.3.
37. Kumar A, Nisha CM, Silakari C, Sharma I, Anusha K, Gupta N, et al. Current and novel therapeutic molecules and targets in Alzheimer’s disease. J Formos Med Assoc. 2016; 115: 3–10. https://doi.org/10.1016/j.jfma.2015.04.001.
38. Patil VS, Patil PA. Molecular Docking: A useful approach of Drug Discovery on the Basis of their Structure. Asian Journal of Pharmaceutical Research. 2023; 13: 191–5. https://doi.org/10.52711/2231-5691.2023.00036.