Discovery of a Therapeutic Agent for Glioblastoma Using a Systems Biology-Based Drug Repositioning Approach
Abstract
:1. Introduction
2. Results
2.1. Survival Analysis Identifies Prognostic Genes for GBM
2.2. DEG Analysis Supports Survival Results
2.3. WGCNA Identify Mostly Connected Genes in GBM
2.4. Discovery of Target Genes for Effective Treatment of GBM
2.5. Drug Repositioning for Treatment of the GBM
2.6. In Vitro Validation of Drug Candidate
3. Discussion
4. Materials and Method
4.1. Data Acquisition and Pre-Processing
4.2. Survival Analysis
4.3. DEG Analysis
4.4. Co-Expression Network Analysis
4.5. Functional Enrichment Analysis
4.6. Drug Target Identification
4.7. Drug Repositioning
4.8. Cell Culture and Drug Treatment
4.9. Western Blot Analysis
4.10. Cell Viability Assay
4.11. Wound Healing Assay
5. Conclusions
Supplementary Materials
Author Contributions
Funding
Institutional Review Board Statement
Informed Consent Statement
Data Availability Statement
Conflicts of Interest
References
- Ceccarelli, M.; Barthel, F.P.; Malta, T.M.; Sabedot, T.S.; Salama, S.R.; Murray, B.A.; Morozova, O.; Newton, Y.; Radenbaugh, A.; Pagnotta, S.M.; et al. Molecular Profiling Reveals Biologically Discrete Subsets and Pathways of Progression in Diffuse Glioma. Cell 2016, 164, 550–563. [Google Scholar] [CrossRef] [PubMed]
- Ma, H.; Zhao, C.; Zhao, Z.; Hu, L.; Ye, F.; Wang, H.; Fang, Z.; Wu, Y.; Chen, X. Specific glioblastoma multiforme prognostic-subtype distinctions based on DNA methylation patterns. Cancer Gene Ther. 2020, 27, 702–714. [Google Scholar] [CrossRef] [PubMed]
- Wu, Y.; Fletcher, M.; Gu, Z.; Wang, Q.; Costa, B.; Bertoni, A.; Man, K.-H.; Schlotter, M.; Felsberg, J.; Mangei, J.; et al. Glioblastoma epigenome profiling identifies SOX10 as a master regulator of molecular tumour subtype. Nat. Commun. 2020, 11, 6434. [Google Scholar] [CrossRef] [PubMed]
- Ostrom, Q.T.; Patil, N.; Cioffi, G.; Waite, K.; Kruchko, C.; Barnholtz-Sloan, J.S. CBTRUS Statistical Report: Primary Brain and Other Central Nervous System Tumors Diagnosed in the United States in 2013–2017. Neuro-Oncology 2020, 22 (Suppl. S2), iv1–iv96. [Google Scholar] [CrossRef] [PubMed]
- Kotecha, R.; Odia, Y.; Khosla, A.A.; Ahluwalia, M.S. Key Clinical Principles in the Management of Glioblastoma. JCO Oncol. Pract. 2023, 19, 180–189. [Google Scholar] [CrossRef] [PubMed]
- Tamimi, A.F.; Juweid, M. Epidemiology and Outcome of Glioblastoma. In Glioblastoma; De Vleeschouwer, S., Ed.; Exon Publications: Brisbane City, QLD, Australia, 2017. [Google Scholar] [CrossRef]
- Eder, K.; Kalman, B. Molecular heterogeneity of glioblastoma and its clinical relevance. Pathol. Oncol. Res. 2014, 20, 777–787. [Google Scholar] [CrossRef] [PubMed]
- Verhaak, R.G.W.; Hoadley, K.A.; Purdom, E.; Wang, V.; Wilkerson, M.D.; Miller, C.R.; Ding, L.; Golub, T.; Jill, P.; Alexe, G.; et al. Integrated genomic analysis identifies clinically relevant subtypes of glioblastoma characterized by abnormalities in PDGFRA, IDH1, EGFR, and NF1. Cancer Cell 2010, 17, 98–110. [Google Scholar] [CrossRef] [PubMed]
- MacLeod, G.; Bozek, D.A.; Rajakulendran, N.; Monteiro, V.; Ahmadi, M.; Steinhart, Z.; Kushida, M.M.; Yu, H.; Coutinho, F.J.; Cavalli, F.M.G.; et al. Genome-Wide CRISPR-Cas9 Screens Expose Genetic Vulnerabilities and Mechanisms of Temozolomide Sensitivity in Glioblastoma Stem Cells. Cell Rep. 2019, 27, 971–986.e9. [Google Scholar] [CrossRef]
- Brennan, C.W.; Verhaak, R.G.W.; McKenna, A.; Campos, B.; Noushmehr, H.; Salama, S.R.; Zheng, S.; Chakravarty, D.; Sanborn, J.Z.; Berman, S.H.; et al. The somatic genomic landscape of glioblastoma. Cell 2013, 155, 462–477. [Google Scholar] [CrossRef]
- Parker, N.R.; Khong, P.; Parkinson, J.F.; Howell, V.M.; Wheeler, H.R. Molecular heterogeneity in glioblastoma: Potential clinical implications. Front. Oncol. 2015, 5, 55. [Google Scholar] [CrossRef]
- Kaynar, A.; Altay, O.; Li, X.; Zhang, C.; Turkez, H.; Uhlén, M.; Shoaie, S.; Mardinoglu, A. Systems Biology Approaches to Decipher the Underlying Molecular Mechanisms of Glioblastoma Multiforme. Int. J. Mol. Sci. 2021, 22, 13213. [Google Scholar] [CrossRef]
- Mardinoglu, A.; Boren, J.; Smith, U.; Uhlen, M.; Nielsen, J. Systems biology in hepatology: Approaches and applications. Nat. Rev. Gastroenterol. Hepatol. 2018, 15, 365–377. [Google Scholar] [CrossRef]
- Nielsen, J. Systems biology of metabolism. Annu. Rev. Biochem. 2017, 86, 245–275. [Google Scholar] [CrossRef]
- Li, X.; Shong, K.; Kim, W.; Yuan, M.; Yang, H.; Sato, Y.; Kume, H.; Ogawa, S.; Turkez, H.; Shoaie, S.; et al. Prediction of drug candidates for clear cell renal cell carcinoma using a systems biology-based drug repositioning approach. EBioMedicine 2022, 78, 103963. [Google Scholar] [CrossRef] [PubMed]
- Yuan, M.; Shong, K.; Li, X.; Ashraf, S.; Shi, M.; Kim, W.; Nielsen, J.; Turkez, H.; Shoaie, S.; Uhlen, M.; et al. A Gene Co-Expression Network-Based Drug Repositioning Approach Identifies Candidates for Treatment of Hepatocellular Carcinoma. Cancers 2022, 14, 1573. [Google Scholar] [CrossRef]
- Zhang, C.; Shi, M.; Kim, W.; Arif, M.; Klevstig, M.; Li, X.; Yang, H.; Bayram, C.; Bolat, I.; Tozlu, Ö.Ö.; et al. Discovery of therapeutic agents targeting PKLR for NAFLD using drug repositioning. EBioMedicine 2022, 83, 104214. [Google Scholar] [CrossRef]
- Rominiyi, O.; Vanderlinden, A.; Clenton, S.J.; Bridgewater, C.; Al-Tamimi, Y.; Collis, S.J. Tumour treating fields therapy for glioblastoma: Current advances and future directions. Br. J. Cancer 2021, 124, 697–709. [Google Scholar] [CrossRef] [PubMed]
- Mohiuddin, E.; Wakimoto, H. Extracellular matrix in glioblastoma: Opportunities for emerging therapeutic approaches. Am. J. Cancer Res. 2021, 11, 3742–3754. Available online: https://www.ncbi.nlm.nih.gov/pubmed/34522446 (accessed on 13 January 2023). [PubMed]
- Langfelder, P.; Horvath, S. WGCNA: An R package for weighted correlation network analysis. BMC Bioinform. 2008, 9, 559. [Google Scholar] [CrossRef]
- Das, S.; Marsden, P.A. Angiogenesis in glioblastoma. N. Engl. J. Med. 2013, 369, 1561–1563. [Google Scholar] [CrossRef]
- Ezzati, S.; Salib, S.; Balasubramaniam, M.; Aboud, O. Epidermal Growth Factor Receptor Inhibitors in Glioblastoma: Current Status and Future Possibilities. Int. J. Mol. Sci. 2024, 25, 2316. [Google Scholar] [CrossRef] [PubMed]
- Johnston, P.A.; Grandis, J.R. STAT3 signaling: Anticancer strategies and challenges. Mol. Interv. 2011, 11, 18–26. [Google Scholar] [CrossRef] [PubMed]
- Zubair, T.; Bandyopadhyay, D. Small Molecule EGFR Inhibitors as Anti-Cancer Agents: Discovery, Mechanisms of Action, and Opportunities. Int. J. Mol. Sci. 2023, 24, 2651. [Google Scholar] [CrossRef] [PubMed]
- Baselga, J.; Swain, S.M. Novel anticancer targets: Revisiting ERBB2 and discovering ERBB3. Nat. Rev. Cancer 2009, 9, 463–475. [Google Scholar] [CrossRef] [PubMed]
- Wolfson, E.; Solomon, S.; Schmukler, E.; Goldshmit, Y.; Pinkas-Kramarski, R. Nucleolin and ErbB2 inhibition reduces tumorigenicity of ErbB2-positive breast cancer. Cell Death Dis. 2018, 9, 47. [Google Scholar] [CrossRef] [PubMed]
- Díaz-Serrano, A.; Sánchez-Torre, A.; Paz-Ares, L. Necitumumab for the treatment of advanced non-small-cell lung cancer. Future Oncol. 2019, 15, 705–716. [Google Scholar] [CrossRef] [PubMed]
- Lin, B.; Ziebro, J.; Smithberger, E.; Skinner, K.R.; Zhao, E.; Cloughesy, T.F.; Binder, Z.A.; O’rourke, D.M.; Nathanson, D.A.; Furnari, F.B.; et al. EGFR, the Lazarus target for precision oncology in glioblastoma. Neuro-Oncology 2022, 24, 2035–2062. [Google Scholar] [CrossRef] [PubMed]
- Fu, Z.; Li, S.; Han, S.; Shi, C.; Zhang, Y. Antibody drug conjugate: The “biological missile” for targeted cancer therapy. Signal Transduct. Target. Ther. 2022, 7, 93. [Google Scholar] [CrossRef] [PubMed]
- Furman, O.; Zaporozhets, A.; Tobi, D.; Bazylevich, A.; Firer, M.A.; Patsenker, L.; Gellerman, G.; Lubin, B.C.R. Novel Cyclic Peptides for Targeting EGFR and EGRvIII Mutation for Drug Delivery. Pharmaceutics 2022, 14, 1505. [Google Scholar] [CrossRef]
- Li, X.; Wang, B.; Liu, W.; Gui, M.; Peng, Z.; Meng, S. Blockage of conformational changes of heat shock protein gp96 on cell membrane by a α-helix peptide inhibits HER2 dimerization and signaling in breast cancer. PLoS ONE 2015, 10, e0124647. [Google Scholar] [CrossRef]
- de Aguiar, C.B.M.; Garcez, R.C.; Alvarez-Silva, M.; Trentin, A.G. Undersulfation of proteoglycans and proteins alter C6 glioma cells proliferation, adhesion and extracellular matrix organization. Int. J. Dev. Neurosci. 2002, 20, 563–571. [Google Scholar] [CrossRef] [PubMed]
- Belousov, A.; Titov, S.; Shved, N.; Garbuz, M.; Malykin, G.; Gulaia, V.; Kagansky, A.; Kumeiko, V. The Extracellular Matrix and Biocompatible Materials in Glioblastoma Treatment. Front. Bioeng. Biotechnol. 2019, 7, 341. [Google Scholar] [CrossRef] [PubMed]
- Pintér, P.; Alpár, A. The Role of Extracellular Matrix in Human Neurodegenerative Diseases. Int. J. Mol. Sci. 2022, 23, 11085. [Google Scholar] [CrossRef] [PubMed]
- Siddiqui, N.; Oshima, K.; Hippensteel, J.A. Proteoglycans and glycosaminoglycans in central nervous system injury. Am. J. Physiol. Physiol. 2022, 323, C46–C55. [Google Scholar] [CrossRef] [PubMed]
- Bratulic, S.; Limeta, A.; Dabestani, S.; Birgisson, H.; Enblad, G.; Stålberg, K.; Hesselager, G.; Häggman, M.; Höglund, M.; Simonson, O.E.; et al. Noninvasive detection of any-stage cancer using free glycosaminoglycans. Proc. Natl. Acad. Sci. USA 2022, 119, e2115328119. [Google Scholar] [CrossRef] [PubMed]
- Gallagher, J. Multiprotein signalling complexes: Regional assembly on heparan sulphate. Biochem. Soc. Trans. 2006, 34 Pt 3, 438–441. [Google Scholar] [CrossRef] [PubMed]
- Sugaya, N.; Habuchi, H.; Nagai, N.; Ashikari-Hada, S.; Kimata, K. 6-O-Sulfation of Heparan Sulfate Differentially Regulates Various Fibroblast Growth Factor-dependent Signalings in Culture *. J. Biol. Chem. 2008, 283, 10366–10376. [Google Scholar] [CrossRef] [PubMed]
- Nandini, C.D.; Sugahara, K. Role of the sulfation pattern of chondroitin sulfate in its biological activities and in the binding of growth factors. Adv. Pharmacol. 2006, 53, 253–279. [Google Scholar] [CrossRef] [PubMed]
- Knelson, E.H.; Nee, J.C.; Blobe, G.C. Heparan sulfate signaling in cancer. Trends Biochem. Sci. 2014, 39, 277–288. [Google Scholar] [CrossRef] [PubMed]
- Pomin, V.H. Keratan sulfate: An up-to-date review. Int. J. Biol. Macromol. 2015, 72, 282–289. [Google Scholar] [CrossRef]
- An, Z.; Aksoy, O.; Zheng, T.; Fan, Q.-W.; Weiss, W.A. Epidermal growth factor receptor and EGFRvIII in glioblastoma: Signaling pathways and targeted therapies. Oncogene 2018, 37, 1561–1575. [Google Scholar] [CrossRef]
- Xiong, Y.; Wang, Q. STC1 regulates glioblastoma migration and invasion via the TGF-β/SMAD4 signaling pathway. Mol. Med. Rep. 2019, 20, 3055–3064. [Google Scholar] [CrossRef]
- Zhao, F.; Yang, G.; Feng, M.; Cao, Z.; Liu, Y.; Qiu, J.; You, L.; Zheng, L.; Zhang, T.; Zhao, Y. Expression, function and clinical application of stanniocalcin-1 in cancer. J. Cell. Mol. Med. 2020, 24, 7686–7696. [Google Scholar] [CrossRef]
- Hwangbo, C.; Tae, N.; Lee, S.; Kim, O.; Park, O.K.; Kim, J.; Kwon, S.-H.; Lee, J.-H. Syntenin regulates TGF-β1-induced Smad activation and the epithelial-to-mesenchymal transition by inhibiting caveolin-mediated TGF-β type I receptor internalization. Oncogene 2016, 35, 389–401. [Google Scholar] [CrossRef]
- Wu, F.; Zhang, Y.; Sun, B.; McMahon, A.P.; Wang, Y. Hedgehog Signaling: From Basic Biology to Cancer Therapy. Cell Chem. Biol. 2017, 24, 252–280. [Google Scholar] [CrossRef] [PubMed]
- Zhang, J.; Liu, Z.; Jia, J. Mechanisms of Smoothened Regulation in Hedgehog Signaling. Cells 2021, 10, 2138. [Google Scholar] [CrossRef]
- Katoh, M. Fibroblast growth factor receptors as treatment targets in clinical oncology. Nat. Rev. Clin. Oncol. 2019, 16, 105–122. [Google Scholar] [CrossRef] [PubMed]
- De Boeck, A.; Ahn, B.Y.; D’mello, C.; Lun, X.; Menon, S.V.; Alshehri, M.M.; Szulzewsky, F.; Shen, Y.; Khan, L.; Dang, N.H.; et al. Glioma-derived IL-33 orchestrates an inflammatory brain tumor microenvironment that accelerates glioma progression. Nat. Commun. 2020, 11, 4997. [Google Scholar] [CrossRef] [PubMed]
- Zhu, V.F.; Yang, J.; LeBrun, D.G.; Li, M. Understanding the role of cytokines in Glioblastoma Multiforme pathogenesis. Cancer Lett. 2012, 316, 139–150. [Google Scholar] [CrossRef]
- Li, X.; Tu, L.; Murphy, P.G.; Kadono, T.; Steeber, D.A.; Tedder, T.F. CHST1 and CHST2 sulfotransferase expression by vascular endothelial cells regulates shear-resistant leukocyte rolling via L-selectin. J. Leukoc. Biol. 2001, 69, 565–574. Available online: https://www.ncbi.nlm.nih.gov/pubmed/11310842 (accessed on 13 January 2023). [CrossRef]
- Huang, L.; Wang, Z.; Chang, Y.; Wang, K.; Kang, X.; Huang, R.; Zhang, Y.; Chen, J.; Zeng, F.; Wu, F.; et al. EFEMP2 indicates assembly of M0 macrophage and more malignant phenotypes of glioma. Aging 2020, 12, 8397–8412. [Google Scholar] [CrossRef] [PubMed]
- Li, F.; Li, Y.; Zhang, K.; Li, Y.; He, P.; Liu, Y.; Yuan, H.; Lu, H.; Liu, J.; Che, S.; et al. FBLN4 as candidate gene associated with long-term and short-term survival with primary glioblastoma. OncoTargets Ther. 2017, 10, 387–395. [Google Scholar] [CrossRef] [PubMed]
- Shen, X.; Jin, X.; Fang, S.; Chen, J. EFEMP2 upregulates PD-L1 expression via EGFR/ERK1/2/c-Jun signaling to promote the invasion of ovarian cancer cells. Cell. Mol. Biol. Lett. 2023, 28, 53. [Google Scholar] [CrossRef] [PubMed]
- Grossman, R.L.; Heath, A.P.; Ferretti, V.; Varmus, H.E.; Lowy, D.R.; Kibbe, W.A.; Staudt, L.M. Toward a Shared Vision for Cancer Genomic Data. N. Engl. J. Med. 2016, 375, 1109–1112. [Google Scholar] [CrossRef] [PubMed]
- Colaprico, A.; Silva, T.C.; Olsen, C.; Garofano, L.; Cava, C.; Garolini, D.; Sabedot, T.S.; Malta, T.M.; Pagnotta, S.M.; Castiglioni, I.; et al. TCGAbiolinks: An R/Bioconductor package for integrative analysis of TCGA data. Nucleic Acids Res. 2016, 44, e71. [Google Scholar] [CrossRef] [PubMed]
- Zhao, Z.; Zhang, K.-N.; Wang, Q.; Li, G.; Zeng, F.; Zhang, Y.; Wu, F.; Chai, R.; Wang, Z.; Zhang, C.; et al. Chinese Glioma Genome Atlas (CGGA): A Comprehensive Resource with Functional Genomic Data from Chinese Glioma Patients. Genom. Proteom. Bioinform. 2021, 19, 1–12. [Google Scholar] [CrossRef] [PubMed]
- Love, M.I.; Huber, W.; Anders, S. Moderated estimation of fold change and dispersion for RNA-seq data with DESeq2. Genome Biol. 2014, 15, 550. [Google Scholar] [CrossRef]
- R Core Team. R: A Language and Environment for Statistical Computing, R Foundation for Statistical Computing. 2020. Available online: https://www.R-project.org (accessed on 14 July 2024).
- Arif, M.; Zhang, C.; Li, X.; Güngör, C.; Çakmak, B.; Arslantürk, M.; Tebani, A.; Özcan, B.; Subaş, O.; Zhou, W.; et al. iNetModels 2.0: An interactive visualization and database of multi-omics data. Nucleic Acids Res. 2021, 49, W271–W276. [Google Scholar] [CrossRef]
Genes | Short Information |
---|---|
ARRDC4 * | Arrestin domain containing 4 is involved in the regulation of cell growth and survival. |
CHST2 * | Carbohydrate sulfotransferase 2 is involved in the synthesis of sulfated proteoglycans and plays a role in the extracellular matrix. |
CHST6 * | Carbohydrate sulfotransferase 6 is involved in the synthesis of sulfated proteoglycans and plays a role in the extracellular matrix. |
CLU * | Clusterin is involved in the extracellular matrix and it is important for cell adhesion and migration. |
DIRAS3 * | DIRAS family GTPase 3 is involved in the regulation of cell growth and survival. |
EN1 * | Engrailed homeobox 1 is involved in the development of the nervous system and plays a role in axon guidance. |
GLIS3 * | GLIS family zinc finger 3 is involved in the regulation of gene expression and plays a role in the development of the kidney. |
GNA12 * | G protein subunit alpha 12 is involved in the regulation of cell growth and survival. |
IBSP * | Integrin-binding sialoprotein is involved in the extracellular matrix and is important for cell adhesion and migration. |
LCTL * | Lactase-like, the function of which is to hydrolyse glycosidic bonds and involved in sensory transduction. |
LZTS1 * | Leucine zipper, putative tumour suppressor 1, is involved in the regulation of cell growth and survival. |
MT1F * | Metallothionein 1F is involved in the regulation of metal ions and plays a role in the response to oxidative stress. |
SCARA3 * | Scavenger receptor class A member 3 is involved in the recognition and clearance of damaged cells and plays a role in the immune system. |
DRAXIN ** | Dorsal inhibitory axon guidance protein is involved in the development of the nervous system and plays a role in axon guidance. |
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content. |
© 2024 by the authors. Licensee MDPI, Basel, Switzerland. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license (https://creativecommons.org/licenses/by/4.0/).
Share and Cite
Kaynar, A.; Ozcan, M.; Li, X.; Turkez, H.; Zhang, C.; Uhlén, M.; Shoaie, S.; Mardinoglu, A. Discovery of a Therapeutic Agent for Glioblastoma Using a Systems Biology-Based Drug Repositioning Approach. Int. J. Mol. Sci. 2024, 25, 7868. https://doi.org/10.3390/ijms25147868
Kaynar A, Ozcan M, Li X, Turkez H, Zhang C, Uhlén M, Shoaie S, Mardinoglu A. Discovery of a Therapeutic Agent for Glioblastoma Using a Systems Biology-Based Drug Repositioning Approach. International Journal of Molecular Sciences. 2024; 25(14):7868. https://doi.org/10.3390/ijms25147868
Chicago/Turabian StyleKaynar, Ali, Mehmet Ozcan, Xiangyu Li, Hasan Turkez, Cheng Zhang, Mathias Uhlén, Saeed Shoaie, and Adil Mardinoglu. 2024. "Discovery of a Therapeutic Agent for Glioblastoma Using a Systems Biology-Based Drug Repositioning Approach" International Journal of Molecular Sciences 25, no. 14: 7868. https://doi.org/10.3390/ijms25147868
APA StyleKaynar, A., Ozcan, M., Li, X., Turkez, H., Zhang, C., Uhlén, M., Shoaie, S., & Mardinoglu, A. (2024). Discovery of a Therapeutic Agent for Glioblastoma Using a Systems Biology-Based Drug Repositioning Approach. International Journal of Molecular Sciences, 25(14), 7868. https://doi.org/10.3390/ijms25147868