The “Superoncogene” Myc at the Crossroad between Metabolism and Gene Expression in Glioblastoma Multiforme
Abstract
:1. Introduction
2. Glioblastoma Multiforme
2.1. GBM Classification
2.1.1. Genetic Classification
2.1.2. Molecular Classification
Transcriptional/Transcriptomic Classification
Epigenetic Classification
Classification Based on BUB1B Inhibition Sensitivity
2.2. Glioblastoma Stem Cells (GSCs)
2.2.1. GSCs and the Tumor Microenvironment
2.2.2. GSC Metabolism
3. GBM Metabolism
3.1. Glucose Metabolism
3.2. Glutamine Addiction
3.3. Lipid Metabolism
4. Myc
4.1. Brief Overview on Myc Structure, Post-Transcriptional Regulation, and Control of Gene Expression
4.1.1. Myc Structure
4.1.2. Myc Protein Network
4.1.3. Myc Post-Translational Regulation
4.1.4. Control of Gene Expression by Myc
4.1.5. Myc-Dependent Regulation of Apoptosis
4.2. Myc and the Control of Gene Expression in Glioblastoma
4.3. Myc and the Maintenance of GSCs
4.4. Myc-Targeted Therapies in GBM
5. Myc and Metabolism in Cancer
5.1. Metabolic Control of Myc Activity in Glioblastoma
5.2. Myc-Dependent Regulation of Metabolic Pathways in Glioblastoma
Crosstalk between Myc-Dependent Angiogenesis and Metabolism in GBM
5.3. Targeting Myc-Dependent Metabolic Pathways in GBM
6. Conclusions: Postulating a Myc-Centered Metabolic Circuit in Glioblastoma
- I.
- Coordinating a plethora of transcriptional coregulators, especially RNAP I, II, and III, as well as controlling RNA splicing and capping, Myc activity is not limited to the activation or repression of directly bound genes at promoters. Furthermore, by binding to superenhancers and increasing chromatin contacts at transcriptionally active domains, Myc also governs chromatin topology [330]. Taking into account all this information, Myc may be defined as a “superoncogene”, orchestrating chromatin architecture, modulating RNAP activity, and directly inducing or repressing transcription, thus ruling the expression of gene expression programs and, ultimately, determining cell fate.
- II.
- Myc involvement in metabolic processes further underpins its importance in the maintenance of cellular homeostasis and in the metabolic rewiring of cancer cells where it is largely overexpressed. In GBM, multiple metabolic signals control Myc expression, which, in turn, governs the activation of cancer metabolic routes and the shunting from one pathway to another (Figure 3), suggesting a Myc-centered, self-sustaining metabolic circuit fulfilling GBM cell and, more importantly, GSC energy demand, fostering tumor growth.
- III.
- The launching and completion of many clinical trials targeting cancer cell metabolism in a variety of cancer types using small molecules that hit pivotal Myc-dependent metabolic enzymes and pathways, including astrocytomas and gliomas, highlights the importance of providing an in-depth characterization of this Myc-centered circuit in GBM as a way to design novel therapeutic strategies aimed at increasing the pool of weapons against this deadly type of tumor.
Author Contributions
Funding
Institutional Review Board Statement
Informed Consent Statement
Data Availability Statement
Conflicts of Interest
References
- Preusser, M.; de Ribaupierre, S.; Wohrer, A.; Erridge, S.C.; Hegi, M.; Weller, M.; Stupp, R. Current concepts and management of glioblastoma. Ann. Neurol. 2011, 70, 9–21. [Google Scholar] [CrossRef]
- Patel, A.P.; Tirosh, I.; Trombetta, J.J.; Shalek, A.K.; Gillespie, S.M.; Wakimoto, H.; Cahill, D.P.; Nahed, B.V.; Curry, W.T.; Martuza, R.L.; et al. Single-cell RNA-seq highlights intratumoral heterogeneity in primary glioblastoma. Science 2014, 344, 1396–1401. [Google Scholar] [CrossRef] [Green Version]
- Plate, K.H.; Breier, G.; Weich, H.A.; Risau, W. Vascular endothelial growth factor is a potential tumour angiogenesis factor in human gliomas in vivo. Nature 1992, 359, 845–848. [Google Scholar] [CrossRef]
- Frei, K.; Gramatzki, D.; Tritschler, I.; Schroeder, J.J.; Espinoza, L.; Rushing, E.J.; Weller, M. Transforming growth factor-beta pathway activity in glioblastoma. Oncotarget 2015, 6, 5963–5977. [Google Scholar] [CrossRef] [Green Version]
- Verhaak, R.G.; Hoadley, K.A.; Purdom, E.; Wang, V.; Qi, Y.; Wilkerson, M.D.; Miller, C.R.; Ding, L.; Golub, T.; Mesirov, J.P.; 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] [Green Version]
- Gimple, R.C.; Bhargava, S.; Dixit, D.; Rich, J.N. Glioblastoma stem cells: Lessons from the tumor hierarchy in a lethal cancer. Genes Dev. 2019, 33, 591–609. [Google Scholar] [CrossRef] [PubMed]
- Friedmann-Morvinski, D.; Bushong, E.A.; Ke, E.; Soda, Y.; Marumoto, T.; Singer, O.; Ellisman, M.H.; Verma, I.M. Dedifferentiation of neurons and astrocytes by oncogenes can induce gliomas in mice. Science 2012, 338, 1080–1084. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Liu, C.; Sage, J.C.; Miller, M.R.; Verhaak, R.G.; Hippenmeyer, S.; Vogel, H.; Foreman, O.; Bronson, R.T.; Nishiyama, A.; Luo, L.; et al. Mosaic analysis with double markers reveals tumor cell of origin in glioma. Cell 2011, 146, 209–221. [Google Scholar] [CrossRef] [Green Version]
- Wang, Q.; Hu, B.; Hu, X.; Kim, H.; Squatrito, M.; Scarpace, L.; deCarvalho, A.C.; Lyu, S.; Li, P.; Li, Y.; et al. Tumor Evolution of Glioma-Intrinsic Gene Expression Subtypes Associates with Immunological Changes in the Microenvironment. Cancer Cell 2017, 32, 42–56.e46. [Google Scholar] [CrossRef] [Green Version]
- Epstein, T.; Gatenby, R.A.; Brown, J.S. The Warburg effect as an adaptation of cancer cells to rapid fluctuations in energy demand. PLoS ONE 2017, 12, e0185085. [Google Scholar] [CrossRef] [Green Version]
- Hoang-Minh, L.B.; Siebzehnrubl, F.A.; Yang, C.; Suzuki-Hatano, S.; Dajac, K.; Loche, T.; Andrews, N.; Schmoll Massari, M.; Patel, J.; Amin, K.; et al. Infiltrative and drug-resistant slow-cycling cells support metabolic heterogeneity in glioblastoma. EMBO J. 2018, 37, e98772. [Google Scholar] [CrossRef]
- Lin, H.; Patel, S.; Affleck, V.S.; Wilson, I.; Turnbull, D.M.; Joshi, A.R.; Maxwell, R.; Stoll, E.A. Fatty acid oxidation is required for the respiration and proliferation of malignant glioma cells. Neuro Oncol. 2017, 19, 43–54. [Google Scholar] [CrossRef] [Green Version]
- Meyer, N.; Penn, L.Z. Reflecting on 25 years with MYC. Nat. Rev. Cancer 2008, 8, 976–990. [Google Scholar] [CrossRef]
- Zhu, S.; Cheng, X.; Wang, R.; Tan, Y.; Ge, M.; Li, D.; Xu, Q.; Sun, Y.; Zhao, C.; Chen, S.; et al. Restoration of microRNA function impairs MYC-dependent maintenance of MLL leukemia. Leukemia 2020, 34, 2484–2488. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Mao, A.; Zhao, Q.; Zhou, X.; Sun, C.; Si, J.; Zhou, R.; Gan, L.; Zhang, H. MicroRNA-449a enhances radiosensitivity by downregulation of c-Myc in prostate cancer cells. Sci. Rep. 2016, 6, 27346. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Zhu, Y.; Zhou, B.; Hu, X.; Ying, S.; Zhou, Q.; Xu, W.; Feng, L.; Hou, T.; Wang, X.; Zhu, L.; et al. LncRNA LINC00942 promotes chemoresistance in gastric cancer by suppressing MSI2 degradation to enhance c-Myc mRNA stability. Clin. Transl. Med. 2022, 12, e703. [Google Scholar] [CrossRef] [PubMed]
- Zhu, P.; He, F.; Hou, Y.; Tu, G.; Li, Q.; Jin, T.; Zeng, H.; Qin, Y.; Wan, X.; Qiao, Y.; et al. A novel hypoxic long noncoding RNA KB-1980E6.3 maintains breast cancer stem cell stemness via interacting with IGF2BP1 to facilitate c-Myc mRNA stability. Oncogene 2021, 40, 1609–1627. [Google Scholar] [CrossRef]
- Farrell, A.S.; Sears, R.C. MYC degradation. Cold Spring Harb. Perspect. Med. 2014, 4, a014365. [Google Scholar] [CrossRef] [Green Version]
- Zhao, K.; Wang, Q.; Wang, Y.; Huang, K.; Yang, C.; Li, Y.; Yi, K.; Kang, C. EGFR/c-myc axis regulates TGFbeta/Hippo/Notch pathway via epigenetic silencing miR-524 in gliomas. Cancer Lett. 2017, 406, 12–21. [Google Scholar] [CrossRef]
- Xu, Q.; Ahmed, A.K.; Zhu, Y.; Wang, K.; Lv, S.; Li, Y.; Jiang, Y. Oncogenic MicroRNA-20a is downregulated by the HIF-1alpha/c-MYC pathway in IDH1 R132H-mutant glioma. Biochem. Biophys. Res. Commun. 2018, 499, 882–888. [Google Scholar] [CrossRef]
- Luo, H.; Chen, Z.; Wang, S.; Zhang, R.; Qiu, W.; Zhao, L.; Peng, C.; Xu, R.; Chen, W.; Wang, H.W.; et al. c-Myc-miR-29c-REV3L signalling pathway drives the acquisition of temozolomide resistance in glioblastoma. Brain 2015, 138, 3654–3672. [Google Scholar] [CrossRef] [Green Version]
- Zhang, G.; Zhu, Q.; Fu, G.; Hou, J.; Hu, X.; Cao, J.; Peng, W.; Wang, X.; Chen, F.; Cui, H. TRIP13 promotes the cell proliferation, migration and invasion of glioblastoma through the FBXW7/c-MYC axis. Br. J. Cancer 2019, 121, 1069–1078. [Google Scholar] [CrossRef]
- Ding, Z.; Liu, X.; Liu, Y.; Zhang, J.; Huang, X.; Yang, X.; Yao, L.; Cui, G.; Wang, D. Expression of far upstream element (FUSE) binding protein 1 in human glioma is correlated with c-Myc and cell proliferation. Mol. Carcinog. 2015, 54, 405–415. [Google Scholar] [CrossRef] [PubMed]
- Wang, J.; Wang, H.; Li, Z.; Wu, Q.; Lathia, J.D.; McLendon, R.E.; Hjelmeland, A.B.; Rich, J.N. c-Myc is required for maintenance of glioma cancer stem cells. PLoS ONE 2008, 3, e3769. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Galardi, S.; Savino, M.; Scagnoli, F.; Pellegatta, S.; Pisati, F.; Zambelli, F.; Illi, B.; Annibali, D.; Beji, S.; Orecchini, E.; et al. Resetting cancer stem cell regulatory nodes upon MYC inhibition. EMBO Rep. 2016, 17, 1872–1889. [Google Scholar] [CrossRef] [Green Version]
- Dhanasekaran, R.; Deutzmann, A.; Mahauad-Fernandez, W.D.; Hansen, A.S.; Gouw, A.M.; Felsher, D.W. The MYC oncogene—The grand orchestrator of cancer growth and immune evasion. Nat. Rev. Clin. Oncol. 2022, 19, 23–36. [Google Scholar] [CrossRef] [PubMed]
- Stine, Z.E.; Walton, Z.E.; Altman, B.J.; Hsieh, A.L.; Dang, C.V. MYC, Metabolism, and Cancer. Cancer Discov. 2015, 5, 1024–1039. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Miller, C.R.; Perry, A. Glioblastoma. Arch. Pathol. Lab. Med. 2007, 131, 397–406. [Google Scholar] [CrossRef]
- Nagy, A.; Garzuly, F.; Padanyi, G.; Szucs, I.; Feldmann, A.; Murnyak, B.; Hortobagyi, T.; Kalman, B. Molecular Subgroups of Glioblastoma- an Assessment by Immunohistochemical Markers. Pathol. Oncol. Res. 2019, 25, 21–31. [Google Scholar] [CrossRef]
- Yonekura, A.; Kawanaka, H.; Prasath, V.B.S.; Aronow, B.J.; Takase, H. Automatic disease stage classification of glioblastoma multiforme histopathological images using deep convolutional neural network. BioMed. Eng. Lett. 2018, 8, 321–327. [Google Scholar] [CrossRef]
- Ostrom, Q.T.; 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 2014–2018. Neuro. Oncol. 2021, 23, iii1–iii105. [Google Scholar] [CrossRef] [PubMed]
- Sturm, D.; Witt, H.; Hovestadt, V.; Khuong-Quang, D.A.; Jones, D.T.; Konermann, C.; Pfaff, E.; Tonjes, M.; Sill, M.; Bender, S.; et al. Hotspot mutations in H3F3A and IDH1 define distinct epigenetic and biological subgroups of glioblastoma. Cancer Cell 2012, 22, 425–437. [Google Scholar] [CrossRef] [Green Version]
- Pollen, A.A.; Nowakowski, T.J.; Chen, J.; Retallack, H.; Sandoval-Espinosa, C.; Nicholas, C.R.; Shuga, J.; Liu, S.J.; Oldham, M.C.; Diaz, A.; et al. Molecular identity of human outer radial glia during cortical development. Cell 2015, 163, 55–67. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Burger, P.C.; Kleihues, P. Cytologic composition of the untreated glioblastoma with implications for evaluation of needle biopsies. Cancer 1989, 63, 2014–2023. [Google Scholar] [CrossRef]
- Ohgaki, H.; Dessen, P.; Jourde, B.; Horstmann, S.; Nishikawa, T.; Di Patre, P.L.; Burkhard, C.; Schuler, D.; Probst-Hensch, N.M.; Maiorka, P.C.; et al. Genetic pathways to glioblastoma: A population-based study. Cancer Res. 2004, 64, 6892–6899. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Hartmann, C.; Meyer, J.; Balss, J.; Capper, D.; Mueller, W.; Christians, A.; Felsberg, J.; Wolter, M.; Mawrin, C.; Wick, W.; et al. Type and frequency of IDH1 and IDH2 mutations are related to astrocytic and oligodendroglial differentiation and age: A study of 1,010 diffuse gliomas. Acta Neuropathol. 2009, 118, 469–474. [Google Scholar] [CrossRef] [Green Version]
- Brennan, C.W.; Verhaak, R.G.; 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]
- Yamazaki, H.; Ohba, Y.; Tamaoki, N.; Shibuya, M. A deletion mutation within the ligand binding domain is responsible for activation of epidermal growth factor receptor gene in human brain tumors. Jpn. J. Cancer Res. 1990, 81, 773–779. [Google Scholar] [CrossRef]
- Appin, C.L.; Brat, D.J. Molecular pathways in gliomagenesis and their relevance to neuropathologic diagnosis. Adv. Anat. Pathol. 2015, 22, 50–58. [Google Scholar] [CrossRef]
- Cancer Genome Atlas Research, N. Comprehensive genomic characterization defines human glioblastoma genes and core pathways. Nature 2008, 455, 1061–1068. [Google Scholar] [CrossRef] [Green Version]
- Rickman, D.S.; Bobek, M.P.; Misek, D.E.; Kuick, R.; Blaivas, M.; Kurnit, D.M.; Taylor, J.; Hanash, S.M. Distinctive molecular profiles of high-grade and low-grade gliomas based on oligonucleotide microarray analysis. Cancer Res. 2001, 61, 6885–6891. [Google Scholar]
- Phillips, H.S.; Kharbanda, S.; Chen, R.; Forrest, W.F.; Soriano, R.H.; Wu, T.D.; Misra, A.; Nigro, J.M.; Colman, H.; Soroceanu, L.; et al. Molecular subclasses of high-grade glioma predict prognosis, delineate a pattern of disease progression, and resemble stages in neurogenesis. Cancer Cell 2006, 9, 157–173. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Kastrup, J. Can YKL-40 be a new inflammatory biomarker in cardiovascular disease? Immunobiology 2012, 217, 483–491. [Google Scholar] [CrossRef] [PubMed]
- Johansen, J.S.; Schultz, N.A.; Jensen, B.V. Plasma YKL-40: A potential new cancer biomarker? Future Oncol. 2009, 5, 1065–1082. [Google Scholar] [CrossRef]
- Nutt, C.L.; Betensky, R.A.; Brower, M.A.; Batchelor, T.T.; Louis, D.N.; Stemmer-Rachamimov, A.O. YKL-40 is a differential diagnostic marker for histologic subtypes of high-grade gliomas. Clin. Cancer Res. 2005, 11, 2258–2264. [Google Scholar] [CrossRef] [Green Version]
- Pelloski, C.E.; Mahajan, A.; Maor, M.; Chang, E.L.; Woo, S.; Gilbert, M.; Colman, H.; Yang, H.; Ledoux, A.; Blair, H.; et al. YKL-40 expression is associated with poorer response to radiation and shorter overall survival in glioblastoma. Clin. Cancer Res. 2005, 11, 3326–3334. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Pegg, A.E.; Dolan, M.E.; Moschel, R.C. Structure, function, and inhibition of O6-alkylguanine-DNA alkyltransferase. Prog. Nucleic Acid Res. Mol. Biol. 1995, 51, 167–223. [Google Scholar] [CrossRef]
- Esteller, M.; Garcia-Foncillas, J.; Andion, E.; Goodman, S.N.; Hidalgo, O.F.; Vanaclocha, V.; Baylin, S.B.; Herman, J.G. Inactivation of the DNA-repair gene MGMT and the clinical response of gliomas to alkylating agents. N. Engl. J. Med. 2000, 343, 1350–1354. [Google Scholar] [CrossRef]
- Stommel, J.M.; Kimmelman, A.C.; Ying, H.; Nabioullin, R.; Ponugoti, A.H.; Wiedemeyer, R.; Stegh, A.H.; Bradner, J.E.; Ligon, K.L.; Brennan, C.; et al. Coactivation of receptor tyrosine kinases affects the response of tumor cells to targeted therapies. Science 2007, 318, 287–290. [Google Scholar] [CrossRef]
- Kouzarides, T. Chromatin modifications and their function. Cell 2007, 128, 693–705. [Google Scholar] [CrossRef] [Green Version]
- Ng, R.K.; Gurdon, J.B. Epigenetic memory of an active gene state depends on histone H3.3 incorporation into chromatin in the absence of transcription. Nat. Cell Biol. 2008, 10, 102–109. [Google Scholar] [CrossRef]
- Pasque, V.; Halley-Stott, R.P.; Gillich, A.; Garrett, N.; Gurdon, J.B. Epigenetic stability of repressed states involving the histone variant macroH2A revealed by nuclear transfer to Xenopus oocytes. Nucleus 2011, 2, 533–539. [Google Scholar] [CrossRef] [Green Version]
- Lejeune, E.; Allshire, R.C. Common ground: Small RNA programming and chromatin modifications. Curr. Opin. Cell Biol. 2011, 23, 258–265. [Google Scholar] [CrossRef]
- Ong, C.T.; Corces, V.G. CTCF: An architectural protein bridging genome topology and function. Nat. Rev. Genet. 2014, 15, 234–246. [Google Scholar] [CrossRef] [Green Version]
- Neems, D.S.; Garza-Gongora, A.G.; Smith, E.D.; Kosak, S.T. Topologically associated domains enriched for lineage-specific genes reveal expression-dependent nuclear topologies during myogenesis. Proc. Natl. Acad. Sci. USA 2016, 113, E1691–E1700. [Google Scholar] [CrossRef] [Green Version]
- Jenuwein, T.; Allis, C.D. Translating the histone code. Science 2001, 293, 1074–1080. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- 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] [Green Version]
- Mackay, A.; Burford, A.; Carvalho, D.; Izquierdo, E.; Fazal-Salom, J.; Taylor, K.R.; Bjerke, L.; Clarke, M.; Vinci, M.; Nandhabalan, M.; et al. Integrated Molecular Meta-Analysis of 1,000 Pediatric High-Grade and Diffuse Intrinsic Pontine Glioma. Cancer Cell 2017, 32, 520–537.e525. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Lewis, P.W.; Muller, M.M.; Koletsky, M.S.; Cordero, F.; Lin, S.; Banaszynski, L.A.; Garcia, B.A.; Muir, T.W.; Becher, O.J.; Allis, C.D. Inhibition of PRC2 activity by a gain-of-function H3 mutation found in pediatric glioblastoma. Science 2013, 340, 857–861. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Portela, A.; Esteller, M. Epigenetic modifications and human disease. Nat. Biotechnol. 2010, 28, 1057–1068. [Google Scholar] [CrossRef] [PubMed]
- Lambiv, W.L.; Vassallo, I.; Delorenzi, M.; Shay, T.; Diserens, A.C.; Misra, A.; Feuerstein, B.; Murat, A.; Migliavacca, E.; Hamou, M.F.; et al. The Wnt inhibitory factor 1 (WIF1) is targeted in glioblastoma and has a tumor suppressing function potentially by induction of senescence. Neuro Oncol. 2011, 13, 736–747. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Gotze, S.; Wolter, M.; Reifenberger, G.; Muller, O.; Sievers, S. Frequent promoter hypermethylation of Wnt pathway inhibitor genes in malignant astrocytic gliomas. Int. J. Cancer 2010, 126, 2584–2593. [Google Scholar] [CrossRef] [PubMed]
- Horiguchi, K.; Tomizawa, Y.; Tosaka, M.; Ishiuchi, S.; Kurihara, H.; Mori, M.; Saito, N. Epigenetic inactivation of RASSF1A candidate tumor suppressor gene at 3p21.3 in brain tumors. Oncogene 2003, 22, 7862–7865. [Google Scholar] [CrossRef] [Green Version]
- Noushmehr, H.; Weisenberger, D.J.; Diefes, K.; Phillips, H.S.; Pujara, K.; Berman, B.P.; Pan, F.; Pelloski, C.E.; Sulman, E.P.; Bhat, K.P.; et al. Identification of a CpG island methylator phenotype that defines a distinct subgroup of glioma. Cancer Cell 2010, 17, 510–522. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Ichimura, K. Molecular pathogenesis of IDH mutations in gliomas. Brain Tumor Pathol. 2012, 29, 131–139. [Google Scholar] [CrossRef]
- Dang, L.; White, D.W.; Gross, S.; Bennett, B.D.; Bittinger, M.A.; Driggers, E.M.; Fantin, V.R.; Jang, H.G.; Jin, S.; Keenan, M.C.; et al. Cancer-associated IDH1 mutations produce 2-hydroxyglutarate. Nature 2009, 462, 739–744. [Google Scholar] [CrossRef] [Green Version]
- Xu, W.; Yang, H.; Liu, Y.; Yang, Y.; Wang, P.; Kim, S.H.; Ito, S.; Yang, C.; Wang, P.; Xiao, M.T.; et al. Oncometabolite 2-hydroxyglutarate is a competitive inhibitor of alpha-ketoglutarate-dependent dioxygenases. Cancer Cell 2011, 19, 17–30. [Google Scholar] [CrossRef] [Green Version]
- Yuan, F.; Wang, Y.; Cai, X.; Du, C.; Zhu, J.; Tang, C.; Yang, J.; Ma, C. N6-methyladenosine-related microRNAs risk model trumps the isocitrate dehydrogenase mutation status as a predictive biomarker for the prognosis and immunotherapy in lower grade gliomas. Explor. Target. Antitumor Ther. 2022, 3, 553–569. [Google Scholar] [CrossRef]
- Pan, T. N6-methyl-adenosine modification in messenger and long non-coding RNA. Trends Biochem. Sci. 2013, 38, 204–209. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Ma, S.; Chen, C.; Ji, X.; Liu, J.; Zhou, Q.; Wang, G.; Yuan, W.; Kan, Q.; Sun, Z. The interplay between m6A RNA methylation and noncoding RNA in cancer. J. Hematol. Oncol. 2019, 12, 121. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Wang, Y.; Gao, M.; Zhu, F.; Li, X.; Yang, Y.; Yan, Q.; Jia, L.; Xie, L.; Chen, Z. METTL3 is essential for postnatal development of brown adipose tissue and energy expenditure in mice. Nat. Commun. 2020, 11, 1648. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Ping, X.L.; Sun, B.F.; Wang, L.; Xiao, W.; Yang, X.; Wang, W.J.; Adhikari, S.; Shi, Y.; Lv, Y.; Chen, Y.S.; et al. Mammalian WTAP is a regulatory subunit of the RNA N6-methyladenosine methyltransferase. Cell Res. 2014, 24, 177–189. [Google Scholar] [CrossRef] [Green Version]
- Meyer, K.D.; Jaffrey, S.R. Rethinking m(6)A Readers, Writers, and Erasers. Annu. Rev. Cell Dev. Biol. 2017, 33, 319–342. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Schwartz, S.; Mumbach, M.R.; Jovanovic, M.; Wang, T.; Maciag, K.; Bushkin, G.G.; Mertins, P.; Ter-Ovanesyan, D.; Habib, N.; Cacchiarelli, D.; et al. Perturbation of m6A writers reveals two distinct classes of mRNA methylation at internal and 5′ sites. Cell Rep. 2014, 8, 284–296. [Google Scholar] [CrossRef] [Green Version]
- Jia, G.; Fu, Y.; Zhao, X.; Dai, Q.; Zheng, G.; Yang, Y.; Yi, C.; Lindahl, T.; Pan, T.; Yang, Y.G.; et al. N6-methyladenosine in nuclear RNA is a major substrate of the obesity-associated FTO. Nat. Chem. Biol. 2011, 7, 885–887. [Google Scholar] [CrossRef] [PubMed]
- Zheng, G.; Dahl, J.A.; Niu, Y.; Fedorcsak, P.; Huang, C.M.; Li, C.J.; Vagbo, C.B.; Shi, Y.; Wang, W.L.; Song, S.H.; et al. ALKBH5 is a mammalian RNA demethylase that impacts RNA metabolism and mouse fertility. Mol. Cell 2013, 49, 18–29. [Google Scholar] [CrossRef] [Green Version]
- Chen, X.Y.; Zhang, J.; Zhu, J.S. The role of m(6)A RNA methylation in human cancer. Mol. Cancer 2019, 18, 103. [Google Scholar] [CrossRef] [Green Version]
- Musacchio, A.; Salmon, E.D. The spindle-assembly checkpoint in space and time. Nat. Rev. Mol. Cell Biol. 2007, 8, 379–393. [Google Scholar] [CrossRef]
- Ding, Y.; Hubert, C.G.; Herman, J.; Corrin, P.; Toledo, C.M.; Skutt-Kakaria, K.; Vazquez, J.; Basom, R.; Zhang, B.; Risler, J.K.; et al. Cancer-Specific requirement for BUB1B/BUBR1 in human brain tumor isolates and genetically transformed cells. Cancer Discov. 2013, 3, 198–211. [Google Scholar] [CrossRef] [Green Version]
- Lee, E.; Pain, M.; Wang, H.; Herman, J.A.; Toledo, C.M.; DeLuca, J.G.; Yong, R.L.; Paddison, P.; Zhu, J. Sensitivity to BUB1B Inhibition Defines an Alternative Classification of Glioblastoma. Cancer Res. 2017, 77, 5518–5529. [Google Scholar] [CrossRef] [Green Version]
- Bonnet, D.; Dick, J.E. Human acute myeloid leukemia is organized as a hierarchy that originates from a primitive hematopoietic cell. Nat. Med. 1997, 3, 730–737. [Google Scholar] [CrossRef]
- Vlashi, E.; Pajonk, F. Cancer stem cells, cancer cell plasticity and radiation therapy. Semin. Cancer Biol. 2015, 31, 28–35. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Ignatova, T.N.; Kukekov, V.G.; Laywell, E.D.; Suslov, O.N.; Vrionis, F.D.; Steindler, D.A. Human cortical glial tumors contain neural stem-like cells expressing astroglial and neuronal markers in vitro. Glia 2002, 39, 193–206. [Google Scholar] [CrossRef]
- Guo, G.; Yang, J.; Nichols, J.; Hall, J.S.; Eyres, I.; Mansfield, W.; Smith, A. Klf4 reverts developmentally programmed restriction of ground state pluripotency. Development 2009, 136, 1063–1069. [Google Scholar] [CrossRef] [Green Version]
- Akberdin, I.R.; Omelyanchuk, N.A.; Fadeev, S.I.; Leskova, N.E.; Oschepkova, E.A.; Kazantsev, F.V.; Matushkin, Y.G.; Afonnikov, D.A.; Kolchanov, N.A. Pluripotency gene network dynamics: System views from parametric analysis. PLoS ONE 2018, 13, e0194464. [Google Scholar] [CrossRef] [PubMed]
- Ignatova, T.N.; Chaitin, H.J.; Kukekov, N.V.; Suslov, O.N.; Dulatova, G.I.; Hanafy, K.A.; Vrionis, F.D. Gliomagenesis is orchestrated by the Oct3/4 regulatory network. J. Neurosurg. Sci. 2021. [Google Scholar] [CrossRef]
- Kanwar, S.S.; Yu, Y.; Nautiyal, J.; Patel, B.B.; Majumdar, A.P. The Wnt/beta-catenin pathway regulates growth and maintenance of colonospheres. Mol. Cancer 2010, 9, 212. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Liu, Y.; Luan, Y.; Ma, K.; Zhang, Z.; Liu, Y.; Chen, X. ISL1 Promotes Human Glioblastoma-Derived Stem Cells’ Self-Renewal by Activation of Sonic Hedgehog/GLI1 Function. Stem Cells Dev. 2022, 31, 258–268. [Google Scholar] [CrossRef]
- Wang, J.; Wakeman, T.P.; Lathia, J.D.; Hjelmeland, A.B.; Wang, X.F.; White, R.R.; Rich, J.N.; Sullenger, B.A. Notch promotes radioresistance of glioma stem cells. Stem Cells 2010, 28, 17–28. [Google Scholar] [CrossRef] [Green Version]
- Adams, J.M.; Strasser, A. Is tumor growth sustained by rare cancer stem cells or dominant clones? Cancer Res. 2008, 68, 4018–4021. [Google Scholar] [CrossRef] [Green Version]
- Tamura, K.; Aoyagi, M.; Ando, N.; Ogishima, T.; Wakimoto, H.; Yamamoto, M.; Ohno, K. Expansion of CD133-positive glioma cells in recurrent de novo glioblastomas after radiotherapy and chemotherapy. J. Neurosurg. 2013, 119, 1145–1155. [Google Scholar] [CrossRef] [Green Version]
- Diehn, M.; Cho, R.W.; Clarke, M.F. Therapeutic implications of the cancer stem cell hypothesis. Semin. Radiat. Oncol. 2009, 19, 78–86. [Google Scholar] [CrossRef] [Green Version]
- Bao, S.; Wu, Q.; McLendon, R.E.; Hao, Y.; Shi, Q.; Hjelmeland, A.B.; Dewhirst, M.W.; Bigner, D.D.; Rich, J.N. Glioma stem cells promote radioresistance by preferential activation of the DNA damage response. Nature 2006, 444, 756–760. [Google Scholar] [CrossRef]
- Bhat, K.P.L.; Balasubramaniyan, V.; Vaillant, B.; Ezhilarasan, R.; Hummelink, K.; Hollingsworth, F.; Wani, K.; Heathcock, L.; James, J.D.; Goodman, L.D.; et al. Mesenchymal differentiation mediated by NF-kappaB promotes radiation resistance in glioblastoma. Cancer Cell 2013, 24, 331–346. [Google Scholar] [CrossRef] [Green Version]
- Venere, M.; Hamerlik, P.; Wu, Q.; Rasmussen, R.D.; Song, L.A.; Vasanji, A.; Tenley, N.; Flavahan, W.A.; Hjelmeland, A.B.; Bartek, J.; et al. Therapeutic targeting of constitutive PARP activation compromises stem cell phenotype and survival of glioblastoma-initiating cells. Cell Death Differ. 2014, 21, 258–269. [Google Scholar] [CrossRef] [Green Version]
- Barazzuol, L.; Jena, R.; Burnet, N.G.; Meira, L.B.; Jeynes, J.C.; Kirkby, K.J.; Kirkby, N.F. Evaluation of poly (ADP-ribose) polymerase inhibitor ABT-888 combined with radiotherapy and temozolomide in glioblastoma. Radiat. Oncol. 2013, 8, 65. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Gage, F.H. Mammalian neural stem cells. Science 2000, 287, 1433–1438. [Google Scholar] [CrossRef] [PubMed]
- Eyme, K.M.; Sammarco, A.; Badr, C.E. Orthotopic brain tumor models derived from glioblastoma stem-like cells. Methods Cell Biol. 2022, 170, 1–19. [Google Scholar] [CrossRef] [PubMed]
- Safa, A.R.; Saadatzadeh, M.R.; Cohen-Gadol, A.A.; Pollok, K.E.; Bijangi-Vishehsaraei, K. Glioblastoma stem cells (GSCs) epigenetic plasticity and interconversion between differentiated non-GSCs and GSCs. Genes Dis. 2015, 2, 152–163. [Google Scholar] [CrossRef] [Green Version]
- De Bacco, F.; Casanova, E.; Medico, E.; Pellegatta, S.; Orzan, F.; Albano, R.; Luraghi, P.; Reato, G.; D’Ambrosio, A.; Porrati, P.; et al. The MET oncogene is a functional marker of a glioblastoma stem cell subtype. Cancer Res. 2012, 72, 4537–4550. [Google Scholar] [CrossRef] [Green Version]
- Marziali, G.; Signore, M.; Buccarelli, M.; Grande, S.; Palma, A.; Biffoni, M.; Rosi, A.; D’Alessandris, Q.G.; Martini, M.; Larocca, L.M.; et al. Metabolic/Proteomic Signature Defines Two Glioblastoma Subtypes With Different Clinical Outcome. Sci. Rep. 2016, 6, 21557. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Wang, L.; Babikir, H.; Muller, S.; Yagnik, G.; Shamardani, K.; Catalan, F.; Kohanbash, G.; Alvarado, B.; Di Lullo, E.; Kriegstein, A.; et al. The Phenotypes of Proliferating Glioblastoma Cells Reside on a Single Axis of Variation. Cancer Discov. 2019, 9, 1708–1719. [Google Scholar] [CrossRef] [Green Version]
- Park, N.I.; Guilhamon, P.; Desai, K.; McAdam, R.F.; Langille, E.; O’Connor, M.; Lan, X.; Whetstone, H.; Coutinho, F.J.; Vanner, R.J.; et al. ASCL1 Reorganizes Chromatin to Direct Neuronal Fate and Suppress Tumorigenicity of Glioblastoma Stem Cells. Cell Stem Cell 2017, 21, 411. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Varn, F.S.; Johnson, K.C.; Martinek, J.; Huse, J.T.; Nasrallah, M.P.; Wesseling, P.; Cooper, L.A.D.; Malta, T.M.; Wade, T.E.; Sabedot, T.S.; et al. Glioma progression is shaped by genetic evolution and microenvironment interactions. Cell 2022, 185, 2184–2199.e2116. [Google Scholar] [CrossRef]
- Snuderl, M.; Fazlollahi, L.; Le, L.P.; Nitta, M.; Zhelyazkova, B.H.; Davidson, C.J.; Akhavanfard, S.; Cahill, D.P.; Aldape, K.D.; Betensky, R.A.; et al. Mosaic amplification of multiple receptor tyrosine kinase genes in glioblastoma. Cancer Cell 2011, 20, 810–817. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Piccirillo, S.G.M.; Colman, S.; Potter, N.E.; van Delft, F.W.; Lillis, S.; Carnicer, M.J.; Kearney, L.; Watts, C.; Greaves, M. Genetic and functional diversity of propagating cells in glioblastoma. Stem Cell Rep. 2015, 4, 7–15. [Google Scholar] [CrossRef] [Green Version]
- Sottoriva, A.; Spiteri, I.; Piccirillo, S.G.; Touloumis, A.; Collins, V.P.; Marioni, J.C.; Curtis, C.; Watts, C.; Tavare, S. Intratumor heterogeneity in human glioblastoma reflects cancer evolutionary dynamics. Proc. Natl. Acad. Sci. USA 2013, 110, 4009–4014. [Google Scholar] [CrossRef] [Green Version]
- Johnson, B.E.; Mazor, T.; Hong, C.; Barnes, M.; Aihara, K.; McLean, C.Y.; Fouse, S.D.; Yamamoto, S.; Ueda, H.; Tatsuno, K.; et al. Mutational analysis reveals the origin and therapy-driven evolution of recurrent glioma. Science 2014, 343, 189–193. [Google Scholar] [CrossRef] [Green Version]
- Lan, X.; Jorg, D.J.; Cavalli, F.M.G.; Richards, L.M.; Nguyen, L.V.; Vanner, R.J.; Guilhamon, P.; Lee, L.; Kushida, M.M.; Pellacani, D.; et al. Fate mapping of human glioblastoma reveals an invariant stem cell hierarchy. Nature 2017, 549, 227–232. [Google Scholar] [CrossRef] [Green Version]
- Gallo, M.; Coutinho, F.J.; Vanner, R.J.; Gayden, T.; Mack, S.C.; Murison, A.; Remke, M.; Li, R.; Takayama, N.; Desai, K.; et al. MLL5 Orchestrates a Cancer Self-Renewal State by Repressing the Histone Variant H3.3 and Globally Reorganizing Chromatin. Cancer Cell 2015, 28, 715–729. [Google Scholar] [CrossRef] [Green Version]
- Hensler, M.; Kasikova, L.; Fiser, K.; Rakova, J.; Skapa, P.; Laco, J.; Lanickova, T.; Pecen, L.; Truxova, I.; Vosahlikova, S.; et al. M2-like macrophages dictate clinically relevant immunosuppression in metastatic ovarian cancer. J. Immunother. Cancer 2020, 8, e000979. [Google Scholar] [CrossRef]
- Arlauckas, S.P.; Garren, S.B.; Garris, C.S.; Kohler, R.H.; Oh, J.; Pittet, M.J.; Weissleder, R. Arg1 expression defines immunosuppressive subsets of tumor-associated macrophages. Theranostics 2018, 8, 5842–5854. [Google Scholar] [CrossRef] [PubMed]
- Wu, L.; Wu, W.; Zhang, J.; Zhao, Z.; Li, L.; Zhu, M.; Wu, M.; Wu, F.; Zhou, F.; Du, Y.; et al. Natural Coevolution of Tumor and Immunoenvironment in Glioblastoma. Cancer Discov. 2022, 12, 2820–2837. [Google Scholar] [CrossRef]
- Spehalski, E.I.; Lee, J.A.; Peters, C.; Tofilon, P.; Camphausen, K. The Quiescent Metabolic Phenotype of Glioma Stem Cells. J. Proteom. Bioinform. 2019, 12, 96–103. [Google Scholar] [CrossRef] [Green Version]
- Gunther, H.S.; Schmidt, N.O.; Phillips, H.S.; Kemming, D.; Kharbanda, S.; Soriano, R.; Modrusan, Z.; Meissner, H.; Westphal, M.; Lamszus, K. Glioblastoma-derived stem cell-enriched cultures form distinct subgroups according to molecular and phenotypic criteria. Oncogene 2008, 27, 2897–2909. [Google Scholar] [CrossRef] [PubMed]
- Schulte, A.; Gunther, H.S.; Phillips, H.S.; Kemming, D.; Martens, T.; Kharbanda, S.; Soriano, R.H.; Modrusan, Z.; Zapf, S.; Westphal, M.; et al. A distinct subset of glioma cell lines with stem cell-like properties reflects the transcriptional phenotype of glioblastomas and overexpresses CXCR4 as therapeutic target. Glia 2011, 59, 590–602. [Google Scholar] [CrossRef] [PubMed]
- Oizel, K.; Chauvin, C.; Oliver, L.; Gratas, C.; Geraldo, F.; Jarry, U.; Scotet, E.; Rabe, M.; Alves-Guerra, M.C.; Teusan, R.; et al. Efficient Mitochondrial Glutamine Targeting Prevails Over Glioblastoma Metabolic Plasticity. Clin. Cancer Res. 2017, 23, 6292–6304. [Google Scholar] [CrossRef] [Green Version]
- Ye, F.; Zhang, Y.; Liu, Y.; Yamada, K.; Tso, J.L.; Menjivar, J.C.; Tian, J.Y.; Yong, W.H.; Schaue, D.; Mischel, P.S.; et al. Protective properties of radio-chemoresistant glioblastoma stem cell clones are associated with metabolic adaptation to reduced glucose dependence. PLoS ONE 2013, 8, e80397. [Google Scholar] [CrossRef] [Green Version]
- Garnier, D.; Renoult, O.; Alves-Guerra, M.C.; Paris, F.; Pecqueur, C. Glioblastoma Stem-Like Cells, Metabolic Strategy to Kill a Challenging Target. Front. Oncol. 2019, 9, 118. [Google Scholar] [CrossRef]
- Badr, C.E.; Silver, D.J.; Siebzehnrubl, F.A.; Deleyrolle, L.P. Metabolic heterogeneity and adaptability in brain tumors. Cell Mol. Life Sci. 2020, 77, 5101–5119. [Google Scholar] [CrossRef]
- Virtuoso, A.; Giovannoni, R.; De Luca, C.; Gargano, F.; Cerasuolo, M.; Maggio, N.; Lavitrano, M.; Papa, M. The Glioblastoma Microenvironment: Morphology, Metabolism, and Molecular Signature of Glial Dynamics to Discover Metabolic Rewiring Sequence. Int. J. Mol. Sci. 2021, 22, 3301. [Google Scholar] [CrossRef] [PubMed]
- Polyzos, A.A.; Lee, D.Y.; Datta, R.; Hauser, M.; Budworth, H.; Holt, A.; Mihalik, S.; Goldschmidt, P.; Frankel, K.; Trego, K.; et al. Metabolic Reprogramming in Astrocytes Distinguishes Region-Specific Neuronal Susceptibility in Huntington Mice. Cell Metab. 2019, 29, 1258–1273.e1211. [Google Scholar] [CrossRef] [PubMed]
- De Luca, C.; Colangelo, A.M.; Virtuoso, A.; Alberghina, L.; Papa, M. Neurons, Glia, Extracellular Matrix and Neurovascular Unit: A Systems Biology Approach to the Complexity of Synaptic Plasticity in Health and Disease. Int. J. Mol. Sci. 2020, 21, 1539. [Google Scholar] [CrossRef] [Green Version]
- Dong, Z.; Cui, H. Epigenetic modulation of metabolism in glioblastoma. Semin. Cancer Biol. 2019, 57, 45–51. [Google Scholar] [CrossRef]
- El Khayari, A.; Bouchmaa, N.; Taib, B.; Wei, Z.; Zeng, A.; El Fatimy, R. Metabolic Rewiring in Glioblastoma Cancer: EGFR, IDH and Beyond. Front. Oncol. 2022, 12, 901951. [Google Scholar] [CrossRef] [PubMed]
- Quinones, A.; Le, A. The Multifaceted Glioblastoma: From Genomic Alterations to Metabolic Adaptations. Adv. Exp. Med. Biol. 2021, 1311, 59–76. [Google Scholar] [CrossRef] [PubMed]
- Flavahan, W.A.; Wu, Q.; Hitomi, M.; Rahim, N.; Kim, Y.; Sloan, A.E.; Weil, R.J.; Nakano, I.; Sarkaria, J.N.; Stringer, B.W.; et al. Brain tumor initiating cells adapt to restricted nutrition through preferential glucose uptake. Nat. Neurosci. 2013, 16, 1373–1382. [Google Scholar] [CrossRef] [PubMed]
- Bhowmick, R.; Subramanian, A.; Sarkar, R.R. Exploring the differences in metabolic behavior of astrocyte and glioblastoma: A flux balance analysis approach. Syst. Synth. Biol. 2015, 9, 159–177. [Google Scholar] [CrossRef] [Green Version]
- Lin, H.; Muramatsu, R.; Maedera, N.; Tsunematsu, H.; Hamaguchi, M.; Koyama, Y.; Kuroda, M.; Ono, K.; Sawada, M.; Yamashita, T. Extracellular Lactate Dehydrogenase A Release From Damaged Neurons Drives Central Nervous System Angiogenesis. EBioMedicine 2018, 27, 71–85. [Google Scholar] [CrossRef] [Green Version]
- Brand, A.; Singer, K.; Koehl, G.E.; Kolitzus, M.; Schoenhammer, G.; Thiel, A.; Matos, C.; Bruss, C.; Klobuch, S.; Peter, K.; et al. LDHA-Associated Lactic Acid Production Blunts Tumor Immunosurveillance by T and NK Cells. Cell Metab. 2016, 24, 657–671. [Google Scholar] [CrossRef] [Green Version]
- Toyonaga, T.; Yamaguchi, S.; Hirata, K.; Kobayashi, K.; Manabe, O.; Watanabe, S.; Terasaka, S.; Kobayashi, H.; Hattori, N.; Shiga, T.; et al. Hypoxic glucose metabolism in glioblastoma as a potential prognostic factor. Eur. J. Nucl. Med. Mol. Imaging 2017, 44, 611–619. [Google Scholar] [CrossRef] [Green Version]
- Zhang, Y.; Yu, G.; Chu, H.; Wang, X.; Xiong, L.; Cai, G.; Liu, R.; Gao, H.; Tao, B.; Li, W.; et al. Macrophage-Associated PGK1 Phosphorylation Promotes Aerobic Glycolysis and Tumorigenesis. Mol. Cell 2018, 71, 201–215.e207. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Babic, I.; Anderson, E.S.; Tanaka, K.; Guo, D.; Masui, K.; Li, B.; Zhu, S.; Gu, Y.; Villa, G.R.; Akhavan, D.; et al. EGFR mutation-induced alternative splicing of Max contributes to growth of glycolytic tumors in brain cancer. Cell Metab. 2013, 17, 1000–1008. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Wolf, A.; Agnihotri, S.; Micallef, J.; Mukherjee, J.; Sabha, N.; Cairns, R.; Hawkins, C.; Guha, A. Hexokinase 2 is a key mediator of aerobic glycolysis and promotes tumor growth in human glioblastoma multiforme. J. Exp. Med. 2011, 208, 313–326. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Guo, D.; Tong, Y.; Jiang, X.; Meng, Y.; Jiang, H.; Du, L.; Wu, Q.; Li, S.; Luo, S.; Li, M.; et al. Aerobic glycolysis promotes tumor immune evasion by hexokinase2-mediated phosphorylation of IkappaBalpha. Cell Metab. 2022, 34, 1312–1324.e1316. [Google Scholar] [CrossRef] [PubMed]
- Maddocks, O.D.; Vousden, K.H. Metabolic regulation by p53. J. Mol. Med. 2011, 89, 237–245. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Schwartzenberg-Bar-Yoseph, F.; Armoni, M.; Karnieli, E. The tumor suppressor p53 down-regulates glucose transporters GLUT1 and GLUT4 gene expression. Cancer Res. 2004, 64, 2627–2633. [Google Scholar] [CrossRef] [Green Version]
- Struys, E.A. 2-Hydroxyglutarate is not a metabolite; D-2-hydroxyglutarate and L-2-hydroxyglutarate are! Proc. Natl. Acad. Sci. USA 2013, 110, E4939. [Google Scholar] [CrossRef] [Green Version]
- Newsholme, P.; Lima, M.M.; Procopio, J.; Pithon-Curi, T.C.; Doi, S.Q.; Bazotte, R.B.; Curi, R. Glutamine and glutamate as vital metabolites. Braz. J. Med. Biol. Res. 2003, 36, 153–163. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Nicklin, P.; Bergman, P.; Zhang, B.; Triantafellow, E.; Wang, H.; Nyfeler, B.; Yang, H.; Hild, M.; Kung, C.; Wilson, C.; et al. Bidirectional transport of amino acids regulates mTOR and autophagy. Cell 2009, 136, 521–534. [Google Scholar] [CrossRef] [Green Version]
- Marin-Valencia, I.; Yang, C.; Mashimo, T.; Cho, S.; Baek, H.; Yang, X.L.; Rajagopalan, K.N.; Maddie, M.; Vemireddy, V.; Zhao, Z.; et al. Analysis of tumor metabolism reveals mitochondrial glucose oxidation in genetically diverse human glioblastomas in the mouse brain in vivo. Cell Metab. 2012, 15, 827–837. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Rosati, A.; Poliani, P.L.; Todeschini, A.; Cominelli, M.; Medicina, D.; Cenzato, M.; Simoncini, E.L.; Magrini, S.M.; Buglione, M.; Grisanti, S.; et al. Glutamine synthetase expression as a valuable marker of epilepsy and longer survival in newly diagnosed glioblastoma multiforme. Neuro Oncol. 2013, 15, 618–625. [Google Scholar] [CrossRef] [PubMed]
- Oppermann, H.; Ding, Y.; Sharma, J.; Berndt Paetz, M.; Meixensberger, J.; Gaunitz, F.; Birkemeyer, C. Metabolic response of glioblastoma cells associated with glucose withdrawal and pyruvate substitution as revealed by GC-MS. Nutr. Metab. 2016, 13, 70. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- DeBerardinis, R.J.; Mancuso, A.; Daikhin, E.; Nissim, I.; Yudkoff, M.; Wehrli, S.; Thompson, C.B. Beyond aerobic glycolysis: Transformed cells can engage in glutamine metabolism that exceeds the requirement for protein and nucleotide synthesis. Proc. Natl. Acad. Sci. USA 2007, 104, 19345–19350. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Phang, J.M. Proline Metabolism in Cell Regulation and Cancer Biology: Recent Advances and Hypotheses. AntiOxid. Redox Signal 2019, 30, 635–649. [Google Scholar] [CrossRef] [Green Version]
- Panosyan, E.H.; Lin, H.J.; Koster, J.; Lasky, J.L., 3rd. In search of druggable targets for GBM amino acid metabolism. BMC Cancer 2017, 17, 162. [Google Scholar] [CrossRef] [Green Version]
- Rocha, C.R.; Kajitani, G.S.; Quinet, A.; Fortunato, R.S.; Menck, C.F. NRF2 and glutathione are key resistance mediators to temozolomide in glioma and melanoma cells. Oncotarget 2016, 7, 48081–48092. [Google Scholar] [CrossRef] [Green Version]
- Lv, H.; Zhen, C.; Liu, J.; Yang, P.; Hu, L.; Shang, P. Unraveling the Potential Role of Glutathione in Multiple Forms of Cell Death in Cancer Therapy. Oxid. Med. Cell Longev. 2019, 2019, 3150145. [Google Scholar] [CrossRef] [Green Version]
- Chinopoulos, C.; Seyfried, T.N. Mitochondrial Substrate-Level Phosphorylation as Energy Source for Glioblastoma: Review and Hypothesis. ASN Neuro 2018, 10, 1759091418818261. [Google Scholar] [CrossRef] [Green Version]
- Fahy, E.; Subramaniam, S.; Murphy, R.C.; Nishijima, M.; Raetz, C.R.; Shimizu, T.; Spener, F.; van Meer, G.; Wakelam, M.J.; Dennis, E.A. Update of the LIPID MAPS comprehensive classification system for lipids. J. Lipid Res. 2009, 50, S9–S14. [Google Scholar] [CrossRef] [Green Version]
- Taib, B.; Aboussalah, A.M.; Moniruzzaman, M.; Chen, S.; Haughey, N.J.; Kim, S.F.; Ahima, R.S. Lipid accumulation and oxidation in glioblastoma multiforme. Sci. Rep. 2019, 9, 19593. [Google Scholar] [CrossRef] [Green Version]
- Ledwozyw, A.; Lutnicki, K. Phospholipids and fatty acids in human brain tumors. Acta Physiol. Hung 1992, 79, 381–387. [Google Scholar] [PubMed]
- Guo, D.; Bell, E.H.; Chakravarti, A. Lipid metabolism emerges as a promising target for malignant glioma therapy. CNS Oncol. 2013, 2, 289–299. [Google Scholar] [CrossRef]
- Mashimo, T.; Pichumani, K.; Vemireddy, V.; Hatanpaa, K.J.; Singh, D.K.; Sirasanagandla, S.; Nannepaga, S.; Piccirillo, S.G.; Kovacs, Z.; Foong, C.; et al. Acetate is a bioenergetic substrate for human glioblastoma and brain metastases. Cell 2014, 159, 1603–1614. [Google Scholar] [CrossRef] [Green Version]
- Park, J.K.; Coffey, N.J.; Limoges, A.; Le, A. The Heterogeneity of Lipid Metabolism in Cancer. Adv. Exp. Med. Biol. 2021, 1311, 39–56. [Google Scholar] [CrossRef]
- Guo, D.; Prins, R.M.; Dang, J.; Kuga, D.; Iwanami, A.; Soto, H.; Lin, K.Y.; Huang, T.T.; Akhavan, D.; Hock, M.B.; et al. EGFR signaling through an Akt-SREBP-1-dependent, rapamycin-resistant pathway sensitizes glioblastomas to antilipogenic therapy. Sci. Signal. 2009, 2, ra82. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Duman, C.; Yaqubi, K.; Hoffmann, A.; Acikgoz, A.A.; Korshunov, A.; Bendszus, M.; Herold-Mende, C.; Liu, H.K.; Alfonso, J. Acyl-CoA-Binding Protein Drives Glioblastoma Tumorigenesis by Sustaining Fatty Acid Oxidation. Cell Metab. 2019, 30, 274–289.e275. [Google Scholar] [CrossRef] [PubMed]
- Huang, R.; Dong, R.; Wang, N.; He, Y.; Zhu, P.; Wang, C.; Lan, B.; Gao, Y.; Sun, L. Adaptive Changes Allow Targeting of Ferroptosis for Glioma Treatment. Cell Mol. Neurobiol. 2022, 42, 2055–2074. [Google Scholar] [CrossRef] [PubMed]
- Cheng, X.; Geng, F.; Guo, D. DGAT1 protects tumor from lipotoxicity, emerging as a promising metabolic target for cancer therapy. Mol. Cell Oncol. 2020, 7, 1805257. [Google Scholar] [CrossRef] [PubMed]
- Lautenberger, J.A.; Schulz, R.A.; Garon, C.F.; Tsichlis, P.N.; Papas, T.S. Molecular cloning of avian myelocytomatosis virus (MC29) transforming sequences. Proc. Natl. Acad. Sci. USA 1981, 78, 1518–1522. [Google Scholar] [CrossRef] [Green Version]
- Colby, W.W.; Chen, E.Y.; Smith, D.H.; Levinson, A.D. Identification and nucleotide sequence of a human locus homologous to the v-myc oncogene of avian myelocytomatosis virus MC29. Nature 1983, 301, 722–725. [Google Scholar] [CrossRef] [PubMed]
- Dalla-Favera, R.; Wong-Staal, F.; Gallo, R.C. Onc gene amplification in promyelocytic leukaemia cell line HL-60 and primary leukaemic cells of the same patient. Nature 1982, 299, 61–63. [Google Scholar] [CrossRef]
- Crews, S.; Barth, R.; Hood, L.; Prehn, J.; Calame, K. Mouse c-myc oncogene is located on chromosome 15 and translocated to chromosome 12 in plasmacytomas. Science 1982, 218, 1319–1321. [Google Scholar] [CrossRef] [PubMed]
- Steffen, D. Proviruses are adjacent to c-myc in some murine leukemia virus-induced lymphomas. Proc. Natl. Acad. Sci. USA 1984, 81, 2097–2101. [Google Scholar] [CrossRef] [Green Version]
- Kohl, N.E.; Kanda, N.; Schreck, R.R.; Bruns, G.; Latt, S.A.; Gilbert, F.; Alt, F.W. Transposition and amplification of oncogene-related sequences in human neuroblastomas. Cell 1983, 35, 359–367. [Google Scholar] [CrossRef] [PubMed]
- Nau, M.M.; Brooks, B.J.; Battey, J.; Sausville, E.; Gazdar, A.F.; Kirsch, I.R.; McBride, O.W.; Bertness, V.; Hollis, G.F.; Minna, J.D. L-myc, a new myc-related gene amplified and expressed in human small cell lung cancer. Nature 1985, 318, 69–73. [Google Scholar] [CrossRef]
- Baluapuri, A.; Wolf, E.; Eilers, M. Target gene-independent functions of MYC oncoproteins. Nat. Rev. Mol. Cell Biol. 2020, 21, 255–267. [Google Scholar] [CrossRef]
- Kalkat, M.; Resetca, D.; Lourenco, C.; Chan, P.K.; Wei, Y.; Shiah, Y.J.; Vitkin, N.; Tong, Y.; Sunnerhagen, M.; Done, S.J.; et al. MYC Protein Interactome Profiling Reveals Functionally Distinct Regions that Cooperate to Drive Tumorigenesis. Mol. Cell 2018, 72, 836–848.e837. [Google Scholar] [CrossRef] [Green Version]
- Beaulieu, M.E.; Castillo, F.; Soucek, L. Structural and Biophysical Insights into the Function of the Intrinsically Disordered Myc Oncoprotein. Cells 2020, 9, 1038. [Google Scholar] [CrossRef]
- Rahl, P.B.; Lin, C.Y.; Seila, A.C.; Flynn, R.A.; McCuine, S.; Burge, C.B.; Sharp, P.A.; Young, R.A. c-Myc regulates transcriptional pause release. Cell 2010, 141, 432–445. [Google Scholar] [CrossRef] [Green Version]
- Cole, M.D. Myc meets its Max. Cell 1991, 65, 715–716. [Google Scholar] [CrossRef] [PubMed]
- Grandori, C.; Cowley, S.M.; James, L.P.; Eisenman, R.N. The Myc/Max/Mad network and the transcriptional control of cell behavior. Annu. Rev. Cell Dev. Biol. 2000, 16, 653–699. [Google Scholar] [CrossRef] [PubMed]
- Grandori, C.; Eisenman, R.N. Myc target genes. Trends Biochem. Sci. 1997, 22, 177–181. [Google Scholar] [CrossRef] [PubMed]
- Farhana, L.; Dawson, M.I.; Fontana, J.A. Down regulation of miR-202 modulates Mxd1 and Sin3A repressor complexes to induce apoptosis of pancreatic cancer cells. Cancer Biol. Ther. 2015, 16, 115–124. [Google Scholar] [CrossRef]
- Yang, G.; Hurlin, P.J. MNT and Emerging Concepts of MNT-MYC Antagonism. Genes 2017, 8, 83. [Google Scholar] [CrossRef]
- Link, J.M.; Ota, S.; Zhou, Z.Q.; Daniel, C.J.; Sears, R.C.; Hurlin, P.J. A critical role for Mnt in Myc-driven T-cell proliferation and oncogenesis. Proc. Natl. Acad. Sci. USA 2012, 109, 19685–19690. [Google Scholar] [CrossRef] [Green Version]
- Nair, S.K.; Burley, S.K. Structural aspects of interactions within the Myc/Max/Mad network. Curr. Top Microbiol. Immunol. 2006, 302, 123–143. [Google Scholar] [CrossRef]
- Cowling, V.H.; Cole, M.D. Mechanism of transcriptional activation by the Myc oncoproteins. Semin. Cancer Biol. 2006, 16, 242–252. [Google Scholar] [CrossRef]
- Sears, R.; Nuckolls, F.; Haura, E.; Taya, Y.; Tamai, K.; Nevins, J.R. Multiple Ras-dependent phosphorylation pathways regulate Myc protein stability. Genes Dev. 2000, 14, 2501–2514. [Google Scholar] [CrossRef] [Green Version]
- Jiang, J.; Wang, J.; Yue, M.; Cai, X.; Wang, T.; Wu, C.; Su, H.; Wang, Y.; Han, M.; Zhang, Y.; et al. Direct Phosphorylation and Stabilization of MYC by Aurora B Kinase Promote T-cell Leukemogenesis. Cancer Cell 2020, 37, 200–215.e205. [Google Scholar] [CrossRef]
- Kim, S.Y.; Herbst, A.; Tworkowski, K.A.; Salghetti, S.E.; Tansey, W.P. Skp2 regulates Myc protein stability and activity. Mol. Cell 2003, 11, 1177–1188. [Google Scholar] [CrossRef]
- von der Lehr, N.; Johansson, S.; Wu, S.; Bahram, F.; Castell, A.; Cetinkaya, C.; Hydbring, P.; Weidung, I.; Nakayama, K.; Nakayama, K.I.; et al. The F-box protein Skp2 participates in c-Myc proteosomal degradation and acts as a cofactor for c-Myc-regulated transcription. Mol. Cell 2003, 11, 1189–1200. [Google Scholar] [CrossRef] [PubMed]
- Adhikary, S.; Marinoni, F.; Hock, A.; Hulleman, E.; Popov, N.; Beier, R.; Bernard, S.; Quarto, M.; Capra, M.; Goettig, S.; et al. The ubiquitin ligase HectH9 regulates transcriptional activation by Myc and is essential for tumor cell proliferation. Cell 2005, 123, 409–421. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Patel, J.H.; Du, Y.; Ard, P.G.; Phillips, C.; Carella, B.; Chen, C.J.; Rakowski, C.; Chatterjee, C.; Lieberman, P.M.; Lane, W.S.; et al. The c-MYC oncoprotein is a substrate of the acetyltransferases hGCN5/PCAF and TIP60. Mol. Cell Biol. 2004, 24, 10826–10834. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Faiola, F.; Liu, X.; Lo, S.; Pan, S.; Zhang, K.; Lymar, E.; Farina, A.; Martinez, E. Dual regulation of c-Myc by p300 via acetylation-dependent control of Myc protein turnover and coactivation of Myc-induced transcription. Mol. Cell Biol. 2005, 25, 10220–10234. [Google Scholar] [CrossRef] [Green Version]
- Zhang, K.; Faiola, F.; Martinez, E. Six lysine residues on c-Myc are direct substrates for acetylation by p300. Biochem. Biophys. Res. Commun. 2005, 336, 274–280. [Google Scholar] [CrossRef]
- Blanc, R.S.; Richard, S. Arginine Methylation: The Coming of Age. Mol. Cell 2017, 65, 8–24. [Google Scholar] [CrossRef] [Green Version]
- Tikhanovich, I.; Zhao, J.; Bridges, B.; Kumer, S.; Roberts, B.; Weinman, S.A. Arginine methylation regulates c-Myc-dependent transcription by altering promoter recruitment of the acetyltransferase p300. J. Biol. Chem. 2017, 292, 13333–13344. [Google Scholar] [CrossRef] [Green Version]
- Favia, A.; Salvatori, L.; Nanni, S.; Iwamoto-Stohl, L.K.; Valente, S.; Mai, A.; Scagnoli, F.; Fontanella, R.A.; Totta, P.; Nasi, S.; et al. The Protein Arginine Methyltransferases 1 and 5 affect Myc properties in glioblastoma stem cells. Sci. Rep. 2019, 9, 15925. [Google Scholar] [CrossRef] [Green Version]
- Baluapuri, A.; Hofstetter, J.; Dudvarski Stankovic, N.; Endres, T.; Bhandare, P.; Vos, S.M.; Adhikari, B.; Schwarz, J.D.; Narain, A.; Vogt, M.; et al. MYC Recruits SPT5 to RNA Polymerase II to Promote Processive Transcription Elongation. Mol. Cell 2019, 74, 674–687.e611. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Koh, C.M.; Bezzi, M.; Low, D.H.; Ang, W.X.; Teo, S.X.; Gay, F.P.; Al-Haddawi, M.; Tan, S.Y.; Osato, M.; Sabo, A.; et al. MYC regulates the core pre-mRNA splicing machinery as an essential step in lymphomagenesis. Nature 2015, 523, 96–100. [Google Scholar] [CrossRef]
- Nie, Z.; Hu, G.; Wei, G.; Cui, K.; Yamane, A.; Resch, W.; Wang, R.; Green, D.R.; Tessarollo, L.; Casellas, R.; et al. c-Myc is a universal amplifier of expressed genes in lymphocytes and embryonic stem cells. Cell 2012, 151, 68–79. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Lorenzin, F.; Benary, U.; Baluapuri, A.; Walz, S.; Jung, L.A.; von Eyss, B.; Kisker, C.; Wolf, J.; Eilers, M.; Wolf, E. Different promoter affinities account for specificity in MYC-dependent gene regulation. eLife 2016, 5, e15161. [Google Scholar] [CrossRef]
- Walz, S.; Lorenzin, F.; Morton, J.; Wiese, K.E.; von Eyss, B.; Herold, S.; Rycak, L.; Dumay-Odelot, H.; Karim, S.; Bartkuhn, M.; et al. Activation and repression by oncogenic MYC shape tumour-specific gene expression profiles. Nature 2014, 511, 483–487. [Google Scholar] [CrossRef] [PubMed]
- Guccione, E.; Martinato, F.; Finocchiaro, G.; Luzi, L.; Tizzoni, L.; Dall’ Olio, V.; Zardo, G.; Nervi, C.; Bernard, L.; Amati, B. Myc-binding-site recognition in the human genome is determined by chromatin context. Nat. Cell Biol. 2006, 8, 764–770. [Google Scholar] [CrossRef]
- Psathas, J.N.; Thomas-Tikhonenko, A. MYC and the art of microRNA maintenance. Cold Spring Harb. Perspect. Med. 2014, 4. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Cowling, V.H.; Cole, M.D. The Myc transactivation domain promotes global phosphorylation of the RNA polymerase II carboxy-terminal domain independently of direct DNA binding. Mol. Cell Biol. 2007, 27, 2059–2073. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Evan, G.I.; Wyllie, A.H.; Gilbert, C.S.; Littlewood, T.D.; Land, H.; Brooks, M.; Waters, C.M.; Penn, L.Z.; Hancock, D.C. Induction of apoptosis in fibroblasts by c-myc protein. Cell 1992, 69, 119–128. [Google Scholar] [CrossRef] [PubMed]
- Shi, Y.; Glynn, J.M.; Guilbert, L.J.; Cotter, T.G.; Bissonnette, R.P.; Green, D.R. Role for c-myc in activation-induced apoptotic cell death in T cell hybridomas. Science 1992, 257, 212–214. [Google Scholar] [CrossRef] [PubMed]
- Zindy, F.; Eischen, C.M.; Randle, D.H.; Kamijo, T.; Cleveland, J.L.; Sherr, C.J.; Roussel, M.F. Myc signaling via the ARF tumor suppressor regulates p53-dependent apoptosis and immortalization. Genes Dev. 1998, 12, 2424–2433. [Google Scholar] [CrossRef] [Green Version]
- Soucie, E.L.; Annis, M.G.; Sedivy, J.; Filmus, J.; Leber, B.; Andrews, D.W.; Penn, L.Z. Myc potentiates apoptosis by stimulating Bax activity at the mitochondria. Mol. Cell Biol. 2001, 21, 4725–4736. [Google Scholar] [CrossRef] [Green Version]
- McMahon, S.B. MYC and the control of apoptosis. Cold Spring Harb. Perspect. Med. 2014, 4, a014407. [Google Scholar] [CrossRef] [Green Version]
- Hermeking, H.; Eick, D. Mediation of c-Myc-induced apoptosis by p53. Science 1994, 265, 2091–2093. [Google Scholar] [CrossRef]
- Herold, S.; Wanzel, M.; Beuger, V.; Frohme, C.; Beul, D.; Hillukkala, T.; Syvaoja, J.; Saluz, H.P.; Haenel, F.; Eilers, M. Negative regulation of the mammalian UV response by Myc through association with Miz-1. Mol. Cell 2002, 10, 509–521. [Google Scholar] [CrossRef] [PubMed]
- Gartel, A.L.; Ye, X.; Goufman, E.; Shianov, P.; Hay, N.; Najmabadi, F.; Tyner, A.L. Myc represses the p21(WAF1/CIP1) promoter and interacts with Sp1/Sp3. Proc. Natl. Acad. Sci. USA 2001, 98, 4510–4515. [Google Scholar] [CrossRef] [Green Version]
- Eischen, C.M.; Packham, G.; Nip, J.; Fee, B.E.; Hiebert, S.W.; Zambetti, G.P.; Cleveland, J.L. Bcl-2 is an apoptotic target suppressed by both c-Myc and E2F-1. Oncogene 2001, 20, 6983–6993. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Maclean, K.H.; Keller, U.B.; Rodriguez-Galindo, C.; Nilsson, J.A.; Cleveland, J.L. c-Myc augments gamma irradiation-induced apoptosis by suppressing Bcl-XL. Mol. Cell Biol. 2003, 23, 7256–7270. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Dansen, T.B.; Whitfield, J.; Rostker, F.; Brown-Swigart, L.; Evan, G.I. Specific requirement for Bax, not Bak, in Myc-induced apoptosis and tumor suppression in vivo. J. Biol. Chem. 2006, 281, 10890–10895. [Google Scholar] [CrossRef] [Green Version]
- Eischen, C.M.; Roussel, M.F.; Korsmeyer, S.J.; Cleveland, J.L. Bax loss impairs Myc-induced apoptosis and circumvents the selection of p53 mutations during Myc-mediated lymphomagenesis. Mol. Cell Biol. 2001, 21, 7653–7662. [Google Scholar] [CrossRef] [Green Version]
- Iaccarino, I.; Hancock, D.; Evan, G.; Downward, J. c-Myc induces cytochrome c release in Rat1 fibroblasts by increasing outer mitochondrial membrane permeability in a Bid-dependent manner. Cell Death Differ. 2003, 10, 599–608. [Google Scholar] [CrossRef]
- Cao, X.; Bennett, R.L.; May, W.S. c-Myc and caspase-2 are involved in activating Bax during cytotoxic drug-induced apoptosis. J. Biol. Chem. 2008, 283, 14490–14496. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Zhuang, D.; Mannava, S.; Grachtchouk, V.; Tang, W.H.; Patil, S.; Wawrzyniak, J.A.; Berman, A.E.; Giordano, T.J.; Prochownik, E.V.; Soengas, M.S.; et al. C-MYC overexpression is required for continuous suppression of oncogene-induced senescence in melanoma cells. Oncogene 2008, 27, 6623–6634. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Campaner, S.; Doni, M.; Hydbring, P.; Verrecchia, A.; Bianchi, L.; Sardella, D.; Schleker, T.; Perna, D.; Tronnersjo, S.; Murga, M.; et al. Cdk2 suppresses cellular senescence induced by the c-myc oncogene. Nat. Cell Biol. 2010, 12, 54–59. [Google Scholar] [CrossRef]
- Katanasaka, Y.; Kodera, Y.; Kitamura, Y.; Morimoto, T.; Tamura, T.; Koizumi, F. Epidermal growth factor receptor variant type III markedly accelerates angiogenesis and tumor growth via inducing c-myc mediated angiopoietin-like 4 expression in malignant glioma. Mol. Cancer 2013, 12, 31. [Google Scholar] [CrossRef] [Green Version]
- Lee, D.H.; Kim, G.W.; Yoo, J.; Lee, S.W.; Jeon, Y.H.; Kim, S.Y.; Kang, H.G.; Kim, D.H.; Chun, K.H.; Choi, J.; et al. Histone demethylase KDM4C controls tumorigenesis of glioblastoma by epigenetically regulating p53 and c-Myc. Cell Death Dis. 2021, 12, 89. [Google Scholar] [CrossRef]
- Ke, X.X.; Zhang, R.; Zhong, X.; Zhang, L.; Cui, H. Deficiency of G9a Inhibits Cell Proliferation and Activates Autophagy via Transcriptionally Regulating c-Myc Expression in Glioblastoma. Front. Cell Dev. Biol. 2020, 8, 593964. [Google Scholar] [CrossRef] [PubMed]
- Hou, J.; Liu, Y.; Huang, P.; Wang, Y.; Pei, D.; Tan, R.; Zhang, Y.; Cui, H. RANBP10 promotes glioblastoma progression by regulating the FBXW7/c-Myc pathway. Cell Death Dis. 2021, 12, 967. [Google Scholar] [CrossRef]
- Wang, F.; Zhang, J.; Ke, X.; Peng, W.; Zhao, G.; Peng, S.; Xu, J.; Xu, B.; Cui, H. WDR5-Myc axis promotes the progression of glioblastoma and neuroblastoma by transcriptional activating CARM1. Biochem. Biophys. Res. Commun. 2020, 523, 699–706. [Google Scholar] [CrossRef]
- Liu, N.; Wang, Z.; Liu, D.; Xie, P. HOXC13-AS-miR-122-5p-SATB1-C-Myc feedback loop promotes migration, invasion and EMT process in glioma. Onco Targets Ther. 2019, 12, 7165–7173. [Google Scholar] [CrossRef] [Green Version]
- Huang, L.; Hu, C.; Chao, H.; Wang, R.; Lu, H.; Li, H.; Chen, H. miR-29c regulates resistance to paclitaxel in nasopharyngeal cancer by targeting ITGB1. Exp. Cell Res. 2019, 378, 1–10. [Google Scholar] [CrossRef]
- Pyko, I.V.; Nakada, M.; Sabit, H.; Teng, L.; Furuyama, N.; Hayashi, Y.; Kawakami, K.; Minamoto, T.; Fedulau, A.S.; Hamada, J. Glycogen synthase kinase 3beta inhibition sensitizes human glioblastoma cells to temozolomide by affecting O6-methylguanine DNA methyltransferase promoter methylation via c-Myc signaling. Carcinogenesis 2013, 34, 2206–2217. [Google Scholar] [CrossRef] [Green Version]
- Wang, J.; Li, Y.; Xu, B.; Dong, J.; Zhao, H.; Zhao, D.; Wu, Y. ALYREF Drives Cancer Cell Proliferation Through an ALYREF-MYC Positive Feedback Loop in Glioblastoma. Onco Targets Ther. 2021, 14, 145–155. [Google Scholar] [CrossRef] [PubMed]
- Chen, X.; Xu, H.; Yuan, P.; Fang, F.; Huss, M.; Vega, V.B.; Wong, E.; Orlov, Y.L.; Zhang, W.; Jiang, J.; et al. Integration of external signaling pathways with the core transcriptional network in embryonic stem cells. Cell 2008, 133, 1106–1117. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Kim, J.; Woo, A.J.; Chu, J.; Snow, J.W.; Fujiwara, Y.; Kim, C.G.; Cantor, A.B.; Orkin, S.H. A Myc network accounts for similarities between embryonic stem and cancer cell transcription programs. Cell 2010, 143, 313–324. [Google Scholar] [CrossRef] [Green Version]
- Zhang, G.; Dong, Z.; Prager, B.C.; Kim, L.J.; Wu, Q.; Gimple, R.C.; Wang, X.; Bao, S.; Hamerlik, P.; Rich, J.N. Chromatin remodeler HELLS maintains glioma stem cells through E2F3 and MYC. JCI Insight 2019, 4, e126140. [Google Scholar] [CrossRef] [Green Version]
- Dixit, D.; Prager, B.C.; Gimple, R.C.; Poh, H.X.; Wang, Y.; Wu, Q.; Qiu, Z.; Kidwell, R.L.; Kim, L.J.Y.; Xie, Q.; et al. The RNA m6A Reader YTHDF2 Maintains Oncogene Expression and Is a Targetable Dependency in Glioblastoma Stem Cells. Cancer Discov. 2021, 11, 480–499. [Google Scholar] [CrossRef] [PubMed]
- Huang, H.; Yu, X.; Han, X.; Hao, J.; Zhao, J.; Bebek, G.; Bao, S.; Prayson, R.A.; Khalil, A.M.; Jankowsky, E.; et al. Piwil1 Regulates Glioma Stem Cell Maintenance and Glioblastoma Progression. Cell Rep. 2021, 34, 108522. [Google Scholar] [CrossRef] [PubMed]
- Fang, X.; Zhou, W.; Wu, Q.; Huang, Z.; Shi, Y.; Yang, K.; Chen, C.; Xie, Q.; Mack, S.C.; Wang, X.; et al. Deubiquitinase USP13 maintains glioblastoma stem cells by antagonizing FBXL14-mediated Myc ubiquitination. J. Exp. Med. 2017, 214, 245–267. [Google Scholar] [CrossRef] [Green Version]
- Fukasawa, K.; Kadota, T.; Horie, T.; Tokumura, K.; Terada, R.; Kitaguchi, Y.; Park, G.; Ochiai, S.; Iwahashi, S.; Okayama, Y.; et al. CDK8 maintains stemness and tumorigenicity of glioma stem cells by regulating the c-MYC pathway. Oncogene 2021, 40, 2803–2815. [Google Scholar] [CrossRef] [PubMed]
- Li, D.; Zhang, Q.; Li, L.; Chen, K.; Yang, J.; Dixit, D.; Gimple, R.C.; Ci, S.; Lu, C.; Hu, L.; et al. beta2-Microglobulin Maintains Glioblastoma Stem Cells and Induces M2-like Polarization of Tumor-Associated Macrophages. Cancer Res. 2022, 82, 3321–3334. [Google Scholar] [CrossRef] [PubMed]
- Chen, H.; Liu, H.; Qing, G. Targeting oncogenic Myc as a strategy for cancer treatment. Signal. Transduct. Target. Ther. 2018, 3, 5. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Llombart, V.; Mansour, M.R. Therapeutic targeting of “undruggable” MYC. EBioMedicine 2022, 75, 103756. [Google Scholar] [CrossRef] [PubMed]
- Le Rhun, E.; von Achenbach, C.; Lohmann, B.; Silginer, M.; Schneider, H.; Meetze, K.; Szabo, E.; Weller, M. Profound, durable and MGMT-independent sensitivity of glioblastoma cells to cyclin-dependent kinase inhibition. Int. J. Cancer 2019, 145, 242–253. [Google Scholar] [CrossRef]
- Devaiah, B.N.; Mu, J.; Akman, B.; Uppal, S.; Weissman, J.D.; Cheng, D.; Baranello, L.; Nie, Z.; Levens, D.; Singer, D.S. MYC protein stability is negatively regulated by BRD4. Proc. Natl. Acad. Sci. USA 2020, 117, 13457–13467. [Google Scholar] [CrossRef] [PubMed]
- Delmore, J.E.; Issa, G.C.; Lemieux, M.E.; Rahl, P.B.; Shi, J.; Jacobs, H.M.; Kastritis, E.; Gilpatrick, T.; Paranal, R.M.; Qi, J.; et al. BET bromodomain inhibition as a therapeutic strategy to target c-Myc. Cell 2011, 146, 904–917. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Kato, F.; Fiorentino, F.P.; Alibes, A.; Perucho, M.; Sanchez-Cespedes, M.; Kohno, T.; Yokota, J. MYCL is a target of a BET bromodomain inhibitor, JQ1, on growth suppression efficacy in small cell lung cancer cells. Oncotarget 2016, 7, 77378–77388. [Google Scholar] [CrossRef] [Green Version]
- Zhou, S.; Zhang, S.; Wang, L.; Huang, S.; Yuan, Y.; Yang, J.; Wang, H.; Li, X.; Wang, P.; Zhou, L.; et al. BET protein inhibitor JQ1 downregulates chromatin accessibility and suppresses metastasis of gastric cancer via inactivating RUNX2/NID1 signaling. Oncogenesis 2020, 9, 33. [Google Scholar] [CrossRef] [Green Version]
- Falchook, G.; Rosen, S.; LoRusso, P.; Watts, J.; Gupta, S.; Coombs, C.C.; Talpaz, M.; Kurzrock, R.; Mita, M.; Cassaday, R.; et al. Development of 2 Bromodomain and Extraterminal Inhibitors With Distinct Pharmacokinetic and Pharmacodynamic Profiles for the Treatment of Advanced Malignancies. Clin. Cancer Res. 2020, 26, 1247–1257. [Google Scholar] [CrossRef] [Green Version]
- Leal, A.S.; Liu, P.; Krieger-Burke, T.; Ruggeri, B.; Liby, K.T. The Bromodomain Inhibitor, INCB057643, Targets Both Cancer Cells and the Tumor Microenvironment in Two Preclinical Models of Pancreatic Cancer. Cancers 2020, 13, 96. [Google Scholar] [CrossRef]
- Cole, M.D.; Nikiforov, M.A. Transcriptional activation by the Myc oncoprotein. Curr. Top. Microbiol. Immunol. 2006, 302, 33–50. [Google Scholar] [CrossRef]
- Wall, M.; Poortinga, G.; Hannan, K.M.; Pearson, R.B.; Hannan, R.D.; McArthur, G.A. Translational control of c-MYC by rapamycin promotes terminal myeloid differentiation. Blood 2008, 112, 2305–2317. [Google Scholar] [CrossRef] [Green Version]
- Cianfanelli, V.; Fuoco, C.; Lorente, M.; Salazar, M.; Quondamatteo, F.; Gherardini, P.F.; De Zio, D.; Nazio, F.; Antonioli, M.; D’Orazio, M.; et al. AMBRA1 links autophagy to cell proliferation and tumorigenesis by promoting c-Myc dephosphorylation and degradation. Nat. Cell Biol. 2015, 17, 20–30. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Delpuech, O.; Griffiths, B.; East, P.; Essafi, A.; Lam, E.W.; Burgering, B.; Downward, J.; Schulze, A. Induction of Mxi1-SR alpha by FOXO3a contributes to repression of Myc-dependent gene expression. Mol. Cell Biol. 2007, 27, 4917–4930. [Google Scholar] [CrossRef] [Green Version]
- Jensen, K.S.; Binderup, T.; Jensen, K.T.; Therkelsen, I.; Borup, R.; Nilsson, E.; Multhaupt, H.; Bouchard, C.; Quistorff, B.; Kjaer, A.; et al. FoxO3A promotes metabolic adaptation to hypoxia by antagonizing Myc function. EMBO J. 2011, 30, 4554–4570. [Google Scholar] [CrossRef] [PubMed]
- Okuyama, H.; Endo, H.; Akashika, T.; Kato, K.; Inoue, M. Downregulation of c-MYC protein levels contributes to cancer cell survival under dual deficiency of oxygen and glucose. Cancer Res. 2010, 70, 10213–10223. [Google Scholar] [CrossRef] [Green Version]
- Gordan, J.D.; Thompson, C.B.; Simon, M.C. HIF and c-Myc: Sibling rivals for control of cancer cell metabolism and proliferation. Cancer Cell 2007, 12, 108–113. [Google Scholar] [CrossRef] [Green Version]
- Shim, H.; Dolde, C.; Lewis, B.C.; Wu, C.S.; Dang, G.; Jungmann, R.A.; Dalla-Favera, R.; Dang, C.V. c-Myc transactivation of LDH-A: Implications for tumor metabolism and growth. Proc. Natl. Acad. Sci. USA 1997, 94, 6658–6663. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Gao, P.; Tchernyshyov, I.; Chang, T.C.; Lee, Y.S.; Kita, K.; Ochi, T.; Zeller, K.I.; De Marzo, A.M.; Van Eyk, J.E.; Mendell, J.T.; et al. c-Myc suppression of miR-23a/b enhances mitochondrial glutaminase expression and glutamine metabolism. Nature 2009, 458, 762–765. [Google Scholar] [CrossRef] [Green Version]
- Bello-Fernandez, C.; Packham, G.; Cleveland, J.L. The ornithine decarboxylase gene is a transcriptional target of c-Myc. Proc. Natl. Acad. Sci. USA 1993, 90, 7804–7808. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Sabo, A.; Kress, T.R.; Pelizzola, M.; de Pretis, S.; Gorski, M.M.; Tesi, A.; Morelli, M.J.; Bora, P.; Doni, M.; Verrecchia, A.; et al. Selective transcriptional regulation by Myc in cellular growth control and lymphomagenesis. Nature 2014, 511, 488–492. [Google Scholar] [CrossRef] [Green Version]
- Kim, J.W.; Zeller, K.I.; Wang, Y.; Jegga, A.G.; Aronow, B.J.; O’Donnell, K.A.; Dang, C.V. Evaluation of myc E-box phylogenetic footprints in glycolytic genes by chromatin immunoprecipitation assays. Mol. Cell Biol. 2004, 24, 5923–5936. [Google Scholar] [CrossRef] [Green Version]
- Lin, C.Y.; Loven, J.; Rahl, P.B.; Paranal, R.M.; Burge, C.B.; Bradner, J.E.; Lee, T.I.; Young, R.A. Transcriptional amplification in tumor cells with elevated c-Myc. Cell 2012, 151, 56–67. [Google Scholar] [CrossRef] [Green Version]
- Venkateswaran, N.; Lafita-Navarro, M.C.; Hao, Y.H.; Kilgore, J.A.; Perez-Castro, L.; Braverman, J.; Borenstein-Auerbach, N.; Kim, M.; Lesner, N.P.; Mishra, P.; et al. MYC promotes tryptophan uptake and metabolism by the kynurenine pathway in colon cancer. Genes Dev. 2019, 33, 1236–1251. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- David, C.J.; Chen, M.; Assanah, M.; Canoll, P.; Manley, J.L. HnRNP proteins controlled by c-Myc deregulate pyruvate kinase mRNA splicing in cancer. Nature 2010, 463, 364–368. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Kaadige, M.R.; Elgort, M.G.; Ayer, D.E. Coordination of glucose and glutamine utilization by an expanded Myc network. Transcription 2010, 1, 36–40. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Wang, R.; Dillon, C.P.; Shi, L.Z.; Milasta, S.; Carter, R.; Finkelstein, D.; McCormick, L.L.; Fitzgerald, P.; Chi, H.; Munger, J.; et al. The transcription factor Myc controls metabolic reprogramming upon T lymphocyte activation. Immunity 2011, 35, 871–882. [Google Scholar] [CrossRef] [Green Version]
- Morrish, F.; Isern, N.; Sadilek, M.; Jeffrey, M.; Hockenbery, D.M. c-Myc activates multiple metabolic networks to generate substrates for cell-cycle entry. Oncogene 2009, 28, 2485–2491. [Google Scholar] [CrossRef] [Green Version]
- Locasale, J.W. Serine, glycine and one-carbon units: Cancer metabolism in full circle. Nat. Rev. Cancer 2013, 13, 572–583. [Google Scholar] [CrossRef] [Green Version]
- Agarwal, S.; Chakravarthi, B.; Behring, M.; Kim, H.G.; Chandrashekar, D.S.; Gupta, N.; Bajpai, P.; Elkholy, A.; Balasubramanya, S.A.H.; Hardy, C.; et al. PAICS, a Purine Nucleotide Metabolic Enzyme, is Involved in Tumor Growth and the Metastasis of Colorectal Cancer. Cancers 2020, 12, 772. [Google Scholar] [CrossRef] [Green Version]
- Liu, Y.C.; Li, F.; Handler, J.; Huang, C.R.; Xiang, Y.; Neretti, N.; Sedivy, J.M.; Zeller, K.I.; Dang, C.V. Global regulation of nucleotide biosynthetic genes by c-Myc. PLoS ONE 2008, 3, e2722. [Google Scholar] [CrossRef]
- Cunningham, J.T.; Moreno, M.V.; Lodi, A.; Ronen, S.M.; Ruggero, D. Protein and nucleotide biosynthesis are coupled by a single rate-limiting enzyme, PRPS2, to drive cancer. Cell 2014, 157, 1088–1103. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Liu, T.; Han, Y.; Yu, C.; Ji, Y.; Wang, C.; Chen, X.; Wang, X.; Shen, J.; Zhang, Y.; Lang, J.Y. MYC predetermines the sensitivity of gastrointestinal cancer to antifolate drugs through regulating TYMS transcription. EBioMedicine 2019, 48, 289–300. [Google Scholar] [CrossRef] [Green Version]
- Yue, M.; Jiang, J.; Gao, P.; Liu, H.; Qing, G. Oncogenic MYC Activates a Feedforward Regulatory Loop Promoting Essential Amino Acid Metabolism and Tumorigenesis. Cell Rep. 2017, 21, 3819–3832. [Google Scholar] [CrossRef] [Green Version]
- Hattori, A.; Tsunoda, M.; Konuma, T.; Kobayashi, M.; Nagy, T.; Glushka, J.; Tayyari, F.; McSkimming, D.; Kannan, N.; Tojo, A.; et al. Cancer progression by reprogrammed BCAA metabolism in myeloid leukaemia. Nature 2017, 545, 500–504. [Google Scholar] [CrossRef] [Green Version]
- Cervenka, I.; Agudelo, L.Z.; Ruas, J.L. Kynurenines: Tryptophan’s metabolites in exercise, inflammation, and mental health. Science 2017, 357. [Google Scholar] [CrossRef] [Green Version]
- Gouw, A.M.; Margulis, K.; Liu, N.S.; Raman, S.J.; Mancuso, A.; Toal, G.G.; Tong, L.; Mosley, A.; Hsieh, A.L.; Sullivan, D.K.; et al. The MYC Oncogene Cooperates with Sterol-Regulated Element-Binding Protein to Regulate Lipogenesis Essential for Neoplastic Growth. Cell Metab. 2019, 30, 556–572.e555. [Google Scholar] [CrossRef]
- Priolo, C.; Pyne, S.; Rose, J.; Regan, E.R.; Zadra, G.; Photopoulos, C.; Cacciatore, S.; Schultz, D.; Scaglia, N.; McDunn, J.; et al. AKT1 and MYC induce distinctive metabolic fingerprints in human prostate cancer. Cancer Res. 2014, 74, 7198–7204. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Morrish, F.; Noonan, J.; Perez-Olsen, C.; Gafken, P.R.; Fitzgibbon, M.; Kelleher, J.; VanGilst, M.; Hockenbery, D. Myc-dependent mitochondrial generation of acetyl-CoA contributes to fatty acid biosynthesis and histone acetylation during cell cycle entry. J. Biol. Chem. 2010, 285, 36267–36274. [Google Scholar] [CrossRef] [Green Version]
- Edmunds, L.R.; Sharma, L.; Kang, A.; Lu, J.; Vockley, J.; Basu, S.; Uppala, R.; Goetzman, E.S.; Beck, M.E.; Scott, D.; et al. c-Myc programs fatty acid metabolism and dictates acetyl-CoA abundance and fate. J. Biol. Chem. 2014, 289, 25382–25392. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Carroll, P.A.; Diolaiti, D.; McFerrin, L.; Gu, H.; Djukovic, D.; Du, J.; Cheng, P.F.; Anderson, S.; Ulrich, M.; Hurley, J.B.; et al. Deregulated Myc requires MondoA/Mlx for metabolic reprogramming and tumorigenesis. Cancer Cell 2015, 27, 271–285. [Google Scholar] [CrossRef] [Green Version]
- Zhong, C.; Fan, L.; Yao, F.; Shi, J.; Fang, W.; Zhao, H. HMGCR is necessary for the tumorigenecity of esophageal squamous cell carcinoma and is regulated by Myc. Tumour Biol. 2014, 35, 4123–4129. [Google Scholar] [CrossRef] [PubMed]
- Cao, Z.; Fan-Minogue, H.; Bellovin, D.I.; Yevtodiyenko, A.; Arzeno, J.; Yang, Q.; Gambhir, S.S.; Felsher, D.W. MYC phosphorylation, activation, and tumorigenic potential in hepatocellular carcinoma are regulated by HMG-CoA reductase. Cancer Res. 2011, 71, 2286–2297. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Camarda, R.; Zhou, A.Y.; Kohnz, R.A.; Balakrishnan, S.; Mahieu, C.; Anderton, B.; Eyob, H.; Kajimura, S.; Tward, A.; Krings, G.; et al. Inhibition of fatty acid oxidation as a therapy for MYC-overexpressing triple-negative breast cancer. Nat. Med. 2016, 22, 427–432. [Google Scholar] [CrossRef]
- Casciano, J.C.; Perry, C.; Cohen-Nowak, A.J.; Miller, K.D.; Vande Voorde, J.; Zhang, Q.; Chalmers, S.; Sandison, M.E.; Liu, Q.; Hedley, A.; et al. MYC regulates fatty acid metabolism through a multigenic program in claudin-low triple negative breast cancer. Br. J. Cancer 2020, 122, 868–884. [Google Scholar] [CrossRef] [Green Version]
- Mossmann, D.; Park, S.; Hall, M.N. mTOR signalling and cellular metabolism are mutual determinants in cancer. Nat. Rev. Cancer 2018, 18, 744–757. [Google Scholar] [CrossRef]
- DeHaven, J.E.; Robinson, K.A.; Nelson, B.A.; Buse, M.G. A novel variant of glutamine: Fructose-6-phosphate amidotransferase-1 (GFAT1) mRNA is selectively expressed in striated muscle. Diabetes 2001, 50, 2419–2424. [Google Scholar] [CrossRef]
- Liu, B.; Huang, Z.B.; Chen, X.; See, Y.X.; Chen, Z.K.; Yao, H.K. Mammalian Target of Rapamycin 2 (MTOR2) and C-MYC Modulate Glucosamine-6-Phosphate Synthesis in Glioblastoma (GBM) Cells Through Glutamine: Fructose-6-Phosphate Aminotransferase 1 (GFAT1). Cell Mol. Neurobiol. 2019, 39, 415–434. [Google Scholar] [CrossRef] [PubMed]
- Masui, K.; Tanaka, K.; Akhavan, D.; Babic, I.; Gini, B.; Matsutani, T.; Iwanami, A.; Liu, F.; Villa, G.R.; Gu, Y.; et al. mTOR complex 2 controls glycolytic metabolism in glioblastoma through FoxO acetylation and upregulation of c-Myc. Cell Metab. 2013, 18, 726–739. [Google Scholar] [CrossRef] [Green Version]
- Nguyen, T.T.T.; Shang, E.; Shu, C.; Kim, S.; Mela, A.; Humala, N.; Mahajan, A.; Yang, H.W.; Akman, H.O.; Quinzii, C.M.; et al. Aurora kinase A inhibition reverses the Warburg effect and elicits unique metabolic vulnerabilities in glioblastoma. Nat. Commun. 2021, 12, 5203. [Google Scholar] [CrossRef]
- Yang, W.; Xia, Y.; Hawke, D.; Li, X.; Liang, J.; Xing, D.; Aldape, K.; Hunter, T.; Alfred Yung, W.K.; Lu, Z. PKM2 phosphorylates histone H3 and promotes gene transcription and tumorigenesis. Cell 2012, 150, 685–696. [Google Scholar] [CrossRef] [Green Version]
- Lee, J.H.; Shao, F.; Ling, J.; Lu, S.; Liu, R.; Du, L.; Chung, J.W.; Koh, S.S.; Leem, S.H.; Shao, J.; et al. Phosphofructokinase 1 Platelet Isoform Promotes beta-Catenin Transactivation for Tumor Development. Front. Oncol. 2020, 10, 211. [Google Scholar] [CrossRef]
- Haynes, H.R.; Scott, H.L.; Killick-Cole, C.L.; Shaw, G.; Brend, T.; Hares, K.M.; Redondo, J.; Kemp, K.C.; Ballesteros, L.S.; Herman, A.; et al. shRNA-mediated PPARalpha knockdown in human glioma stem cells reduces in vitro proliferation and inhibits orthotopic xenograft tumour growth. J. Pathol. 2019, 247, 422–434. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Li, J.; Liu, Q.; Liu, Z.; Xia, Q.; Zhang, Z.; Zhang, R.; Gao, T.; Gu, G.; Wang, Y.; Wang, D.; et al. KPNA2 promotes metabolic reprogramming in glioblastomas by regulation of c-myc. J. Exp. Clin. Cancer Res. 2018, 37, 194. [Google Scholar] [CrossRef] [PubMed]
- Rothhammer, V.; Quintana, F.J. The aryl hydrocarbon receptor: An environmental sensor integrating immune responses in health and disease. Nat. Rev. Immunol. 2019, 19, 184–197. [Google Scholar] [CrossRef]
- Denison, M.S.; Nagy, S.R. Activation of the aryl hydrocarbon receptor by structurally diverse exogenous and endogenous chemicals. Annu. Rev. Pharmacol. Toxicol. 2003, 43, 309–334. [Google Scholar] [CrossRef] [PubMed]
- Lafita-Navarro, M.C.; Perez-Castro, L.; Zacharias, L.G.; Barnes, S.; DeBerardinis, R.J.; Conacci-Sorrell, M. The transcription factors aryl hydrocarbon receptor and MYC cooperate in the regulation of cellular metabolism. J. Biol. Chem. 2020, 295, 12398–12407. [Google Scholar] [CrossRef]
- Mair, R.; Wright, A.J.; Ros, S.; Hu, D.E.; Booth, T.; Kreis, F.; Rao, J.; Watts, C.; Brindle, K.M. Metabolic Imaging Detects Low Levels of Glycolytic Activity That Vary with Levels of c-Myc Expression in Patient-Derived Xenograft Models of Glioblastoma. Cancer Res. 2018, 78, 5408–5418. [Google Scholar] [CrossRef] [Green Version]
- Nguyen, T.T.T.; Zhang, Y.; Shang, E.; Shu, C.; Torrini, C.; Zhao, J.; Bianchetti, E.; Mela, A.; Humala, N.; Mahajan, A.; et al. HDAC inhibitors elicit metabolic reprogramming by targeting super-enhancers in glioblastoma models. J. Clin. Investig. 2020, 130, 3699–3716. [Google Scholar] [CrossRef]
- Szeliga, M.; Albrecht, J. Opposing roles of glutaminase isoforms in determining glioblastoma cell phenotype. Neurochem. Int. 2015, 88, 6–9. [Google Scholar] [CrossRef]
- Yachi, K.; Tsuda, M.; Kohsaka, S.; Wang, L.; Oda, Y.; Tanikawa, S.; Ohba, Y.; Tanaka, S. miR-23a promotes invasion of glioblastoma via HOXD10-regulated glial-mesenchymal transition. Signal. Transduct. Target. Ther. 2018, 3, 33. [Google Scholar] [CrossRef] [Green Version]
- Xin, R.; Qu, D.; Su, S.; Zhao, B.; Chen, D. Downregulation of miR-23b by transcription factor c-Myc alleviates ischemic brain injury by upregulating Nrf2. Int. J. Biol. Sci. 2021, 17, 3659–3671. [Google Scholar] [CrossRef]
- Chang, T.C.; Yu, D.; Lee, Y.S.; Wentzel, E.A.; Arking, D.E.; West, K.M.; Dang, C.V.; Thomas-Tikhonenko, A.; Mendell, J.T. Widespread microRNA repression by Myc contributes to tumorigenesis. Nat. Genet. 2008, 40, 43–50. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Winter-Vann, A.M.; Casey, P.J. Post-prenylation-processing enzymes as new targets in oncogenesis. Nat. Rev. Cancer 2005, 5, 405–412. [Google Scholar] [CrossRef] [PubMed]
- Wang, X.; Huang, Z.; Wu, Q.; Prager, B.C.; Mack, S.C.; Yang, K.; Kim, L.J.Y.; Gimple, R.C.; Shi, Y.; Lai, S.; et al. MYC-Regulated Mevalonate Metabolism Maintains Brain Tumor-Initiating Cells. Cancer Res. 2017, 77, 4947–4960. [Google Scholar] [CrossRef] [Green Version]
- Wang, X.; Yang, K.; Xie, Q.; Wu, Q.; Mack, S.C.; Shi, Y.; Kim, L.J.Y.; Prager, B.C.; Flavahan, W.A.; Liu, X.; et al. Purine synthesis promotes maintenance of brain tumor initiating cells in glioma. Nat. Neurosci. 2017, 20, 661–673. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Rosinska, S.; Gavard, J. Tumor Vessels Fuel the Fire in Glioblastoma. Int. J. Mol. Sci. 2021, 22, 6514. [Google Scholar] [CrossRef] [PubMed]
- Domenech, M.; Hernandez, A.; Plaja, A.; Martinez-Balibrea, E.; Balana, C. Hypoxia: The Cornerstone of Glioblastoma. Int. J. Mol. Sci. 2021, 22, 12608. [Google Scholar] [CrossRef] [PubMed]
- Lakka, S.S.; Rao, J.S. Antiangiogenic therapy in brain tumors. Expert. Rev. Neurother. 2008, 8, 1457–1473. [Google Scholar] [CrossRef] [PubMed]
- Carmeliet, P.; Jain, R.K. Principles and mechanisms of vessel normalization for cancer and other angiogenic diseases. Nat. Rev. Drug Discov. 2011, 10, 417–427. [Google Scholar] [CrossRef]
- De Bock, K.; Cauwenberghs, S.; Carmeliet, P. Vessel abnormalization: Another hallmark of cancer? Molecular mechanisms and therapeutic implications. Curr. Opin. Genet. Dev. 2011, 21, 73–79. [Google Scholar] [CrossRef]
- Nagy, J.A.; Chang, S.H.; Shih, S.C.; Dvorak, A.M.; Dvorak, H.F. Heterogeneity of the tumor vasculature. Semin. Thromb. Hemost. 2010, 36, 321–331. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Lee, J.W.; Bae, S.H.; Jeong, J.W.; Kim, S.H.; Kim, K.W. Hypoxia-inducible factor (HIF-1)alpha: Its protein stability and biological functions. Exp. Mol. Med. 2004, 36, 1–12. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Liu, Z.Z.; Tian, Y.F.; Wu, H.; Ouyang, S.Y.; Kuang, W.L. LncRNA H19 promotes glioma angiogenesis through miR-138/HIF-1alpha/VEGF axis. Neoplasma 2020, 67, 111–118. [Google Scholar] [CrossRef] [PubMed]
- Balandeh, E.; Mohammadshafie, K.; Mahmoudi, Y.; Hossein Pourhanifeh, M.; Rajabi, A.; Bahabadi, Z.R.; Mohammadi, A.H.; Rahimian, N.; Hamblin, M.R.; Mirzaei, H. Roles of Non-coding RNAs and Angiogenesis in Glioblastoma. Front. Cell Dev. Biol. 2021, 9, 716462. [Google Scholar] [CrossRef]
- Xie, T.X.; Xia, Z.; Zhang, N.; Gong, W.; Huang, S. Constitutive NF-kappaB activity regulates the expression of VEGF and IL-8 and tumor angiogenesis of human glioblastoma. Oncol. Rep. 2010, 23, 725–732. [Google Scholar]
- Yu, M.O.; Park, K.J.; Park, D.H.; Chung, Y.G.; Chi, S.G.; Kang, S.H. Reactive oxygen species production has a critical role in hypoxia-induced Stat3 activation and angiogenesis in human glioblastoma. J. Neurooncol. 2015, 125, 55–63. [Google Scholar] [CrossRef]
- Vallee, A.; Guillevin, R.; Vallee, J.N. Vasculogenesis and angiogenesis initiation under normoxic conditions through Wnt/beta-catenin pathway in gliomas. Rev. Neurosci. 2018, 29, 71–91. [Google Scholar] [CrossRef]
- O’Donnell, K.A.; Wentzel, E.A.; Zeller, K.I.; Dang, C.V.; Mendell, J.T. c-Myc-regulated microRNAs modulate E2F1 expression. Nature 2005, 435, 839–843. [Google Scholar] [CrossRef]
- Dews, M.; Fox, J.L.; Hultine, S.; Sundaram, P.; Wang, W.; Liu, Y.Y.; Furth, E.; Enders, G.H.; El-Deiry, W.; Schelter, J.M.; et al. The myc-miR-17~92 axis blunts TGFbeta signaling and production of multiple TGFbeta-dependent antiangiogenic factors. Cancer Res. 2010, 70, 8233–8246. [Google Scholar] [CrossRef] [Green Version]
- Chen, J.; Liu, G.; Wang, X.; Hong, H.; Li, T.; Li, L.; Wang, H.; Xie, J.; Li, B.; Li, T.; et al. Glioblastoma stem cell-specific histamine secretion drives pro-angiogenic tumor microenvironment remodeling. Cell Stem Cell 2022, 29, 1531–1546.e1537. [Google Scholar] [CrossRef]
- Abdel-Wahab, A.F.; Mahmoud, W.; Al-Harizy, R.M. Targeting glucose metabolism to suppress cancer progression: Prospective of anti-glycolytic cancer therapy. Pharmacol. Res. 2019, 150, 104511. [Google Scholar] [CrossRef]
- Mohanti, B.K.; Rath, G.K.; Anantha, N.; Kannan, V.; Das, B.S.; Chandramouli, B.A.; Banerjee, A.K.; Das, S.; Jena, A.; Ravichandran, R.; et al. Improving cancer radiotherapy with 2-deoxy-D-glucose: Phase I/II clinical trials on human cerebral gliomas. Int. J. Radiat. Oncol. Biol. Phys. 1996, 35, 103–111. [Google Scholar] [CrossRef]
- Coleman, M.C.; Asbury, C.R.; Daniels, D.; Du, J.; Aykin-Burns, N.; Smith, B.J.; Li, L.; Spitz, D.R.; Cullen, J.J. 2-deoxy-D-glucose causes cytotoxicity, oxidative stress, and radiosensitization in pancreatic cancer. Free Radic. Biol. Med. 2008, 44, 322–331. [Google Scholar] [CrossRef]
- Liapi, E.; Geschwind, J.F.; Vali, M.; Khwaja, A.A.; Prieto-Ventura, V.; Buijs, M.; Vossen, J.A.; Ganapathy-Kanniappan, S.; Wahl, R.L. Assessment of tumoricidal efficacy and response to treatment with 18F-FDG PET/CT after intraarterial infusion with the antiglycolytic agent 3-bromopyruvate in the VX2 model of liver tumor. J. Nucl. Med. 2011, 52, 225–230. [Google Scholar] [CrossRef] [Green Version]
- Chapiro, J.; Sur, S.; Savic, L.J.; Ganapathy-Kanniappan, S.; Reyes, J.; Duran, R.; Thiruganasambandam, S.C.; Moats, C.R.; Lin, M.; Luo, W.; et al. Systemic delivery of microencapsulated 3-bromopyruvate for the therapy of pancreatic cancer. Clin. Cancer Res. 2014, 20, 6406–6417. [Google Scholar] [CrossRef] [Green Version]
- Dunbar, E.M.; Coats, B.S.; Shroads, A.L.; Langaee, T.; Lew, A.; Forder, J.R.; Shuster, J.J.; Wagner, D.A.; Stacpoole, P.W. Phase 1 trial of dichloroacetate (DCA) in adults with recurrent malignant brain tumors. Investig. New Drugs 2014, 32, 452–464. [Google Scholar] [CrossRef] [Green Version]
- Shen, Y.C.; Ou, D.L.; Hsu, C.; Lin, K.L.; Chang, C.Y.; Lin, C.Y.; Liu, S.H.; Cheng, A.L. Activating oxidative phosphorylation by a pyruvate dehydrogenase kinase inhibitor overcomes sorafenib resistance of hepatocellular carcinoma. Br. J. Cancer 2013, 108, 72–81. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Chu, Q.S.; Sangha, R.; Spratlin, J.; Vos, L.J.; Mackey, J.R.; McEwan, A.J.; Venner, P.; Michelakis, E.D. A phase I open-labeled, single-arm, dose-escalation, study of dichloroacetate (DCA) in patients with advanced solid tumors. Investig. New Drugs 2015, 33, 603–610. [Google Scholar] [CrossRef] [PubMed]
- Le, A.; Cooper, C.R.; Gouw, A.M.; Dinavahi, R.; Maitra, A.; Deck, L.M.; Royer, R.E.; Vander Jagt, D.L.; Semenza, G.L.; Dang, C.V. Inhibition of lactate dehydrogenase A induces oxidative stress and inhibits tumor progression. Proc. Natl. Acad. Sci. USA 2010, 107, 2037–2042. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Zhou, M.; Zhao, Y.; Ding, Y.; Liu, H.; Liu, Z.; Fodstad, O.; Riker, A.I.; Kamarajugadda, S.; Lu, J.; Owen, L.B.; et al. Warburg effect in chemosensitivity: Targeting lactate dehydrogenase-A re-sensitizes taxol-resistant cancer cells to taxol. Mol. Cancer 2010, 9, 33. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Lin, S.T.; Tu, S.H.; Yang, P.S.; Hsu, S.P.; Lee, W.H.; Ho, C.T.; Wu, C.H.; Lai, Y.H.; Chen, M.Y.; Chen, L.C. Apple Polyphenol Phloretin Inhibits Colorectal Cancer Cell Growth via Inhibition of the Type 2 Glucose Transporter and Activation of p53-Mediated Signaling. J. Agric. Food Chem. 2016, 64, 6826–6837. [Google Scholar] [CrossRef] [PubMed]
- Chan, D.A.; Sutphin, P.D.; Nguyen, P.; Turcotte, S.; Lai, E.W.; Banh, A.; Reynolds, G.E.; Chi, J.T.; Wu, J.; Solow-Cordero, D.E.; et al. Targeting GLUT1 and the Warburg effect in renal cell carcinoma by chemical synthetic lethality. Sci. Transl. Med. 2011, 3, 94ra70. [Google Scholar] [CrossRef] [Green Version]
- Dalva-Aydemir, S.; Bajpai, R.; Martinez, M.; Adekola, K.U.; Kandela, I.; Wei, C.; Singhal, S.; Koblinski, J.E.; Raje, N.S.; Rosen, S.T.; et al. Targeting the metabolic plasticity of multiple myeloma with FDA-approved ritonavir and metformin. Clin. Cancer Res. 2015, 21, 1161–1171. [Google Scholar] [CrossRef] [Green Version]
- Yu, W.; Yang, X.; Zhang, Q.; Sun, L.; Yuan, S.; Xin, Y. Targeting GLS1 to cancer therapy through glutamine metabolism. Clin. Transl. Oncol. 2021, 23, 2253–2268. [Google Scholar] [CrossRef] [PubMed]
- Song, M.; Kim, S.H.; Im, C.Y.; Hwang, H.J. Recent Development of Small Molecule Glutaminase Inhibitors. Curr. Top. Med. Chem. 2018, 18, 432–443. [Google Scholar] [CrossRef]
- Matre, P.; Velez, J.; Jacamo, R.; Qi, Y.; Su, X.; Cai, T.; Chan, S.M.; Lodi, A.; Sweeney, S.R.; Ma, H.; et al. Inhibiting glutaminase in acute myeloid leukemia: Metabolic dependency of selected AML subtypes. Oncotarget 2016, 7, 79722–79735. [Google Scholar] [CrossRef] [Green Version]
- Tateishi, K.; Iafrate, A.J.; Ho, Q.; Curry, W.T.; Batchelor, T.T.; Flaherty, K.T.; Onozato, M.L.; Lelic, N.; Sundaram, S.; Cahill, D.P.; et al. Myc-Driven Glycolysis Is a Therapeutic Target in Glioblastoma. Clin. Cancer Res. 2016, 22, 4452–4465. [Google Scholar] [CrossRef] [Green Version]
- Hasmann, M.; Schemainda, I. FK866, a highly specific noncompetitive inhibitor of nicotinamide phosphoribosyltransferase, represents a novel mechanism for induction of tumor cell apoptosis. Cancer Res. 2003, 63, 7436–7442. [Google Scholar] [PubMed]
- Watson, M.; Roulston, A.; Belec, L.; Billot, X.; Marcellus, R.; Bedard, D.; Bernier, C.; Branchaud, S.; Chan, H.; Dairi, K.; et al. The small molecule GMX1778 is a potent inhibitor of NAD+ biosynthesis: Strategy for enhanced therapy in nicotinic acid phosphoribosyltransferase 1-deficient tumors. Mol. Cell Biol. 2009, 29, 5872–5888. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- See, Y.X.; Chen, K.; Fullwood, M.J. MYC overexpression leads to increased chromatin interactions at super-enhancers and MYC binding sites. Genome Res. 2022, 32, 629–642. [Google Scholar] [CrossRef]
Trial N° | Status | Study Title | Drug/Treatment | Condition | Sponsor | Collaborators |
---|---|---|---|---|---|---|
NCT03434262 | Active, not recruiting | SJDAWN: St. Jude’s Children Research Hospital Phase-I study evaluating molecularly driven doublet therapies for children and young adults with recurrent brain tumors | Ribociclib/trametinib, ribociclib/sonidegib, ribociclib/gemcitabine | Brain tumors, including medulloblstoma, ependymoma, oligodendroglioma, glioblastoma (IDHwt and IDHmut), xanthoastrocytoma, neuroblastoma, medulloepithelioma, and embryonal tumors | St. Jude’s Children Research Hospital | Novartis Pharmaceuticals |
NCT03224104 | Completed | Study of TG02 in elderly newly diagnosed or adult relapsed patients with anaplastic astrocytoma or glioblastoma | TG02, TMZ, radiation | IDH1R132H-non mutated and MGMT-promoter-unmethylated anaplastic astrocytoma or GBM; IDH1R132H-non mutated and MGMT-promoter-methylated anaplastic astrocytoma or GBM | European Organization for Research and Treatment of Cancer (EORTC) | Tragara Pharmaceuticals |
NCT02711137 | Terminated | Open-label safety and tolerability study of INCB057643 in subjects with advanced malignancies | INCB057643, gemcitabine, paclitaxel, rucaparib, abiraterone, azacitidine, ruxolitinib | Solid tumors, including brain tumors and lymphoma | Incyte Corporation | Incyte Corporation |
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. |
© 2023 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
Cencioni, C.; Scagnoli, F.; Spallotta, F.; Nasi, S.; Illi, B. The “Superoncogene” Myc at the Crossroad between Metabolism and Gene Expression in Glioblastoma Multiforme. Int. J. Mol. Sci. 2023, 24, 4217. https://doi.org/10.3390/ijms24044217
Cencioni C, Scagnoli F, Spallotta F, Nasi S, Illi B. The “Superoncogene” Myc at the Crossroad between Metabolism and Gene Expression in Glioblastoma Multiforme. International Journal of Molecular Sciences. 2023; 24(4):4217. https://doi.org/10.3390/ijms24044217
Chicago/Turabian StyleCencioni, Chiara, Fiorella Scagnoli, Francesco Spallotta, Sergio Nasi, and Barbara Illi. 2023. "The “Superoncogene” Myc at the Crossroad between Metabolism and Gene Expression in Glioblastoma Multiforme" International Journal of Molecular Sciences 24, no. 4: 4217. https://doi.org/10.3390/ijms24044217