Multiple Faces of the Glioblastoma Microenvironment
Abstract
:1. Introduction
2. Genes and Epigenetic Factors
3. Neuroinflammation: Cytokines and Oxidative Stress
4. Hypoxia
4.1. Genetic Alterations That Increase HIF Proteins
- -
- -
- PTEN (phosphatase and tensin homolog) deletions are found in 20–40% of GBM. PTEN is the main inhibitor of the PI3K/AKT/mTOR signaling pathway [91]; the result is an increase in HIF-1α via the PI3K/AKT pathway.
- -
- -
- FAT1 (FAT atypical Cadherin 1) is linked to HIF-1α’s fate, being an upstream regulator of HIF-1α expression [94]. Endogenous depletion of FAT1 under hypoxic conditions was accompanied by a decrease in HIF-1α expression, along with its downstream target genes (CA9, GLUT1, VEGFA, MCT4, HK2, BNIP3, and REDD1) [95]; the result was an important diminution in the GBM’s aggressiveness. Loss of FAT1 heterozygosity has been found in astrocytic tumors [96].
4.2. Maintenance of Glioma Stem-like Cells
4.3. Hypoxic Metabolic Adaptation
4.4. Epithelial-Mesenchymal Transition
5. The Tumor Microenvironment and Vascular Modifications
6. Conclusions
Author Contributions
Funding
Institutional Review Board Statement
Informed Consent Statement
Data Availability Statement
Conflicts of Interest
References
- Mostofa, A.G.; Punganuru, S.R.; Madala, H.R.; Al-Obaide, M.; Srivenugopal, K.S. The Process and Regulatory Components of Inflammation in Brain Oncogenesis. Biomolecules 2017, 7, 34. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Li, Q.J.; Cai, J.Q.; Liu, C.Y. Evolving Molecular Genetics of Glioblastoma. Chin. Med. J. 2016, 129, 464–471. [Google Scholar] [CrossRef]
- Buruiană, A.; Florian, Ș.I.; Florian, A.I.; Timiș, T.L.; Mihu, C.M.; Miclăuș, M.; Oșan, S.; Hrapșa, I.; Cataniciu, R.C.; Farcaș, M.; et al. The Roles of miRNA in Glioblastoma Tumor Cell Communication: Diplomatic and Aggressive Negotiations. Int. J. Mol. Sci. 2020, 21, 1950. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Deutschman, C.S. Acute-phase responses and SIRS/MODS: The good, the bad, and the nebulous. Crit. Care Med. 1998, 26, 1630–1631. [Google Scholar] [CrossRef] [PubMed]
- 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]
- Heddleston, J.M.; Li, Z.; McLendon, R.E.; Hjelmeland, A.B.; Rich, J.N. The hypoxic microenvironment maintains glioblastoma stem cells and promotes reprogramming towards a cancer stem cell phenotype. Cell Cycle 2009, 8, 3274–3284. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Molina, M.L.; García-Bernal, D.; Martinez, S.; Valdor, R. Autophagy in the Immunosuppressive Perivascular Microenvironment of Glioblastoma. Cancers 2019, 12, 102. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Chen, Z.; Hambardzumyan, D. Immune Microenvironment in Glioblastoma Subtypes. Front. Immunol. 2018, 9, 1004. [Google Scholar] [CrossRef] [Green Version]
- Perrin, S.L.; Samuel, M.S.; Koszyca, B.; Brown, M.P.; Ebert, L.M.; Oksdath, M.; Gomez, G.A. Glioblastoma heterogeneity and the tumour microenvironment: Implications for preclinical research and development of new treatments. Biochem. Soc. Trans. 2019, 47, 625–638. [Google Scholar] [CrossRef]
- Qiu, R.; Zhong, Y.; Li, Q.; Li, Y.; Fan, H. Metabolic Remodeling in Glioma Immune Microenvironment: Intercellular Interactions Distinct From Peripheral Tumors. Front. Cell Dev. Biol. 2021, 9, 693215. [Google Scholar] [CrossRef]
- Haksoyler, V.; Besen, A.A.; Koseci, T.; Olgun, P.; Bayram, E.; Topkan, E. Neutrophil-to-lymphocyte ratio is prognostic in recurrent glioblastoma multiforme treated with bevacizumab plus irinotecan. Biomark. Med. 2021, 15, 851–859. [Google Scholar] [CrossRef]
- Zhang, H.; Zhou, Y.; Cui, B.; Liu, Z.; Shen, H. Novel insights into astrocyte-mediated signaling of proliferation, invasion and tumor immune microenvironment in glioblastoma. Biomed. Pharmacother. 2020, 126, 110086. [Google Scholar] [CrossRef] [PubMed]
- Wang, R.; Chadalavada, K.; Wilshire, J.; Kowalik, U.; Hovinga, K.E.; Geber, A.; Fligelman, B.; Leversha, M.; Brennan, C.; Tabar, V. Glioblastoma stem-like cells give rise to tumour endothelium. Nature 2010, 468, 829–833. [Google Scholar] [CrossRef] [PubMed]
- Ricci-Vitiani, L.; Pallini, R.; Biffoni, M.; Todaro, M.; Invernici, G.; Cenci, T.; Maira, G.; Parati, E.A.; Stassi, G.; Larocca, L.M.; et al. Tumour vascularization via endothelial differentiation of glioblastoma stem-like cells. Nature 2010, 468, 824–828. [Google Scholar] [CrossRef]
- Carmeliet, P.; Jain, R.K. Molecular mechanisms and clinical applications of angiogenesis. Nature 2011, 473, 298–307. [Google Scholar] [CrossRef] [Green Version]
- Aldape, K.; Zadeh, G.; Mansouri, S.; Reifenberger, G.; von Deimling, A. Glioblastoma: Pathology, molecular mechanisms and markers. Acta Neuropathol. 2015, 129, 829–848. [Google Scholar] [CrossRef] [PubMed]
- 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] [Green Version]
- Costello, J.F.; Plass, C.; Arap, W.; Chapman, V.M.; Held, W.A.; Berger, M.S.; Su Huang, H.J.; Cavenee, W.K. Cyclin-dependent kinase 6 (CDK6) amplification in human gliomas identified using two-dimensional separation of genomic DNA. Cancer Res. 1997, 57, 1250–1254. [Google Scholar] [PubMed]
- Jiao, Y.; Killela, P.J.; Reitman, Z.J.; Rasheed, A.B.; Heaphy, C.M.; de Wilde, R.F.; Rodriguez, F.J.; Rosemberg, S.; Oba-Shinjo, S.M.; Nagahashi Marie, S.K.; et al. Frequent ATRX, CIC, FUBP1 and IDH1 mutations refine the classification of malignant gliomas. Oncotarget 2012, 3, 709–722. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Aquilanti, E.; Miller, J.; Santagata, S.; Cahill, D.P.; Brastianos, P.K. Updates in prognostic markers for gliomas. Neuro Oncol. 2018, 20 (Suppl. S7), vii17–vii26. [Google Scholar] [CrossRef] [Green Version]
- Deaton, A.M.; Bird, A. CpG islands and the regulation of transcription. Genes Dev. 2011, 25, 1010–1022. [Google Scholar] [CrossRef] [Green Version]
- 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]
- Hegi, M.E.; Diserens, A.C.; Godard, S.; Dietrich, P.Y.; Regli, L.; Ostermann, S.; Otten, P.; Van Melle, G.; de Tribolet, N.; Stupp, R. Clinical trial substantiates the predictive value of O-6-methylguanine-DNA methyltransferase promoter methylation in glioblastoma patients treated with temozolomide. Clin. Cancer Res. 2004, 10, 1871–1874. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Huang, F.W.; Hodis, E.; Xu, M.J.; Kryukov, G.V.; Chin, L.; Garraway, L.A. Highly recurrent TERT promoter mutations in human melanoma. Science 2013, 339, 957–959. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Ma, X.; Gong, R.; Wang, R.; Pan, Y.; Cai, D.; Pan, B.; Li, Y.; Xiang, J.; Li, H.; Zhang, J.; et al. Recurrent TERT promoter mutations in non-small cell lung cancers. Lung Cancer 2014, 86, 369–373. [Google Scholar] [CrossRef] [PubMed]
- Crespo, I.; Vital, A.L.; Gonzalez-Tablas, M.; Patino Mdel, C.; Otero, A.; Lopes, M.C.; de Oliveira, C.; Domingues, P.; Orfao, A.; Tabernero, M.D. Molecular and Genomic Alterations in Glioblastoma Multiforme. Am. J. Pathol. 2015, 185, 1820–1833. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Weller, M.; Pfister, S.M.; Wick, W.; Hegi, M.E.; Reifenberger, G.; Stupp, R. Molecular neuro-oncology in clinical practice: A new horizon. Lancet Oncol. 2013, 14, e370–e379, Erratum in Lancet Oncol. 2015, 16, e199. [Google Scholar] [CrossRef] [Green Version]
- Ohgaki, H.; Dessen, P.; Jourde, B.; Horstmann, S.; Nishikawa, T.; Di Patre, P.L.; Burkhard, C.; Schüler, 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] [Green Version]
- Rachakonda, P.S.; Hosen, I.; de Verdier, P.J.; Fallah, M.; Heidenreich, B.; Ryk, C.; Wiklund, N.P.; Steineck, G.; Schadendorf, D.; Hemminki, K.; et al. TERT promoter mutations in bladder cancer affect patient survival and disease recurrence through modification by a common polymorphism. Proc. Natl. Acad. Sci. USA 2013, 110, 17426–17431. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Lee, S.E.; Chang, S.H.; Kim, W.Y.; Lim, S.D.; Kim, W.S.; Hwang, T.S.; Han, H.S. Frequent somatic TERT promoter mutations and CTNNB1 mutations in hepatocellular carcinoma. Oncotarget 2016, 7, 69267–69275. [Google Scholar] [CrossRef] [Green Version]
- Labussière, M.; Boisselier, B.; Mokhtari, K.; Di Stefano, A.L.; Rahimian, A.; Rossetto, M.; Ciccarino, P.; Saulnier, O.; Paterra, R.; Marie, Y.; et al. Combined analysis of TERT, EGFR, and IDH status defines distinct prognostic glioblastoma classes. Neurology 2014, 83, 1200–1206. [Google Scholar] [CrossRef]
- Louis, D.N.; Ohgaki, H.; Wiestler, O.D.; Cavenee, W.K. WHO Classification of Tumours of the Central Nervous System, Revised, 4th ed.; IARC: Lyon, France, 2016; pp. 15–123. [Google Scholar]
- Karremann, M.; Gielen, G.H.; Hoffmann, M.; Wiese, M.; Colditz, N.; Warmuth-Metz, M.; Bison, B.; Claviez, A.; van Vuurden, D.G.; von Bueren, A.O.; et al. Diffuse high-grade gliomas with H3 K27M mutations carry a dismal prognosis independent of tumor location. Neuro Oncol. 2018, 20, 123–131. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Ryall, S.; Krishnatry, R.; Arnoldo, A.; Buczkowicz, P.; Mistry, M.; Siddaway, R.; Ling, C.; Pajovic, S.; Yu, M.; Rubin, J.B.; et al. Targeted detection of genetic alterations reveal the prognostic impact of H3K27M and MAPK pathway aberrations in paediatric thalamic glioma. Acta Neuropathol. Commun. 2016, 4, 93. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Meyronet, D.; Esteban-Mader, M.; Bonnet, C.; Joly, M.O.; Uro-Coste, E.; Amiel-Benouaich, A.; Forest, F.; Rousselot-Denis, C.; Burel-Vandenbos, F.; Bourg, V.; et al. Characteristics of H3 K27M-mutant gliomas in adults. Neuro Oncol. 2017, 19, 1127–1134. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Sochocka, M.; Diniz, B.S.; Leszek, J. Inflammatory Response in the CNS: Friend or Foe? Mol. Neurobiol. 2017, 54, 8071–8089. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Perng, P.; Lim, M. Immunosuppressive mechanisms of malignant gliomas: Parallels at non-CNS sites. Front. Oncol. 2015, 5, 153. [Google Scholar] [CrossRef] [Green Version]
- Colotta, F.; Allavena, P.; Sica, A.; Garlanda, C.; Mantovani, A. Cancer-related inflammation, the seventh hallmark of cancer: Links to genetic instability. Carcinogenesis 2009, 30, 1073–1081. [Google Scholar] [CrossRef] [Green Version]
- Mesfin, F.B.; Al-Dhahir, M.A. Gliomas; StatPearls Publishing: Treasure Island, FL, USA, 2021. Available online: https://www.ncbi.nlm.nih.gov/books/NBK441874/ (accessed on 11 December 2021).
- Dharmajaya, R.; Sari, D.K. Role and value of inflammatory markers in brain tumors: A case-controlled study. Ann. Med. Surg. 2021, 63, 102107. [Google Scholar] [CrossRef]
- Jackson, C.M.; Choi, J.; Lim, M. Mechanisms of immunotherapy resistance: Lessons from glioblastoma. Nat. Immunol. 2019, 20, 1100–1109. [Google Scholar] [CrossRef]
- Okolie, O.; Bago, J.R.; Schmid, R.S.; Irvin, D.M.; Bash, R.E.; Miller, C.R.; Hingtgen, S.D. Reactive astrocytes potentiate tumor aggressiveness in a murine glioma resection and recurrence model. Neuro Oncol. 2016, 18, 1622–1633. [Google Scholar] [CrossRef]
- Albulescu, R.; Codrici, E.; Popescu, I.D.; Mihai, S.; Necula, L.G.; Petrescu, D.; Teodoru, M.; Tanase, C.P. Cytokine patterns in brain tumour progression. Mediat. Inflamm. 2013, 2013, 979748. [Google Scholar] [CrossRef]
- Li, T.; Chen, X.; Zhang, C.; Zhang, Y.; Yao, W. An update on reactive astrocytes in chronic pain. J. Neuroinflamm. 2019, 16, 140. [Google Scholar] [CrossRef]
- O’Brien, E.R.; Howarth, C.; Sibson, N.R. The role of astrocytes in CNS tumors: Pre-clinical models and novel imaging approaches. Front. Cell. Neurosci. 2013, 7, 40. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Mitchell, D.; Shireman, J.; Sierra Potchanant, E.A.; Lara-Velazquez, M.; Dey, M. Neuroinflammation in Autoimmune Disease and Primary Brain Tumors: The Quest for Striking the Right Balance. Front. Cell. Neurosci. 2021, 15, 716947. [Google Scholar] [CrossRef] [PubMed]
- Zhu, V.F.; Jingxuan, Y.; Drake, G.L.B.; Min, L. Understanding the role of cytokines in Glioblastoma Multiforme pathogenesis. Cancer Lett. 2012, 316, 139–150. [Google Scholar] [CrossRef] [PubMed]
- Dranoff, G. Cytokines in cancer pathogenesis and cancer therapy. Nat. Rev. Cancer 2004, 4, 11–22. [Google Scholar] [CrossRef] [PubMed]
- Bunevicius, A.; Radziunas, A.; Tamasauskas, S.; Tamasauskas, A.; Laws, E.R.; Iervasi, G.; Bunevicius, R.; Deltuva, V. Prognostic role of high sensitivity C-reactive protein and interleukin-6 in glioma and meningioma patients. J. Neurooncol. 2018, 138, 351–358. [Google Scholar] [CrossRef] [PubMed]
- Brunner, C.; Seiderer, J.; Schlamp, A.; Bidlingmaier, M.; Eigler, A.; Haimerl, W.; Lehr, H.A.; Krieg, A.M.; Hartmann, G.; Endres, S. Enhanced dendritic cell maturation by TNF-alpha or cytidine-phosphate-guanosine DNA drives T cell activation in vitro and therapeutic anti-tumor immune responses in vivo. J. Immunol. 2000, 165, 6278–6286. [Google Scholar] [CrossRef] [Green Version]
- Yoshida, S.; Ono, M.; Shono, T.; Izumi, H.; Ishibashi, T.; Suzuki, H.; Kuwano, M. Involvement of interleukin-8, vascular endothelial growth factor, and basic fibroblast growth factor in tumor necrosis factor alpha-dependent angiogenesis. Mol. Cell. Biol. 1997, 17, 4015–4023. [Google Scholar] [CrossRef] [Green Version]
- Ghosh, S.; Paul, A.; Sen, E. Tumor necrosis factor α-induced hypoxia-inducible factor 1α-β-catenin axis regulates major histocompatibility complex class I gene activation through chromatin remodeling. Mol. Cell. Biol. 2013, 33, 2718–2731. [Google Scholar] [CrossRef] [Green Version]
- Fukaya, R.; Ohta, S.; Yaguchi, T.; Matsuzaki, Y.; Sugihara, E.; Okano, H.; Saya, H.; Kawakami, Y.; Kawase, T.; Yoshida, K.; et al. MIF Maintains the Tumorigenic Capacity of Brain Tumor-Initiating Cells by Directly Inhibiting p53. Cancer Res. 2016, 76, 2813–2823. [Google Scholar] [CrossRef] [Green Version]
- Veillat, V.; Carli, C.; Metz, C.N.; Al-Abed, Y.; Naccache, P.H.; Akoum, A. Macrophage migration inhibitory factor elicits an angiogenic phenotype in human ectopic endometrial cells and triggers the production of major angiogenic factors via CD44, CD74, and MAPK signaling pathways. J. Clin. Endocrinol. Metab. 2010, 95, E403–E412. [Google Scholar] [CrossRef] [PubMed]
- Munaut, C.; Boniver, J.; Foidart, J.-M.; Deprez, M. Macrophage migration inhibitory factor (MIF) expression in human glioblastomas correlates with vascular endothelial growth factor (VEGF) expression. Neuropathol. Appl. Neurobiol. 2002, 28, 452–460. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Apte, R.N.; Dotan, S.; Elkabets, M.; White, M.R.; Reich, E.; Carmi, Y.; Song, X.; Dvozkin, T.; Krelin, Y.; Voronov, E. The involvement of IL-1 in tumorigenesis, tumor invasiveness, metastasis and tumor-host interactions. Cancer Metastasis Rev. 2006, 25, 387–408. [Google Scholar] [CrossRef] [PubMed]
- Paugh, B.S.; Bryan, L.; Paugh, S.W.; Wilczynska, K.M.; Alvarez, S.M.; Singh, S.K.; Kapitonov, D.; Rokita, H.; Wright, S.; Griswold-Prenner, I.; et al. Interleukin-1 regulates the expression of sphingosine kinase 1 in glioblastoma cells. J. Biol. Chem. 2009, 284, 3408–3417. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Wang, L.; Liu, Z.; Balivada, S.; Shrestha, T.; Bossmann, S.; Pyle, M.; Pappan, L.; Shi, J.; Troyer, D. Interleukin-1β and transforming growth factor-β cooperate to induce neurosphere formation and increase tumorigenicity of adherent LN-229 glioma cells. Stem Cell Res. Ther. 2012, 3, 5. [Google Scholar] [CrossRef] [Green Version]
- Goswami, S.; Gupta, A.; Sharma, S.K. Interleukin-6-mediated autocrine growth promotion in human glioblastoma multiforme cell line U87MG. J. Neurochem. 1998, 71, 1837–1845. [Google Scholar] [CrossRef] [Green Version]
- Wang, H.; Lathia, J.D.; Wu, Q.; Wang, J.; Li, Z.; Heddleston, J.M.; Eyler, C.E.; Elderbroom, J.; Gallagher, J.; Schuschu, J.; et al. Targeting interleukin 6 signaling suppresses glioma stem cell survival and tumor growth. Stem Cells 2009, 27, 2393–2404. [Google Scholar] [CrossRef] [Green Version]
- Li, R.; Li, G.; Deng, L.; Liu, Q.; Dai, J.; Shen, J.; Zhang, J. IL-6 augments the invasiveness of U87MG human glioblastoma multiforme cells via up-regulation of MMP-2 and fascin-1. Oncol. Rep. 2010, 23, 1553–1559. [Google Scholar] [CrossRef] [Green Version]
- Graf, M.R.; Prins, R.M.; Merchant, R.E. IL-6 secretion by a rat T9 glioma clone induces a neutrophil-dependent antitumor response with resultant cellular, antiglioma immunity. J. Immunol. 2001, 166, 121–129. [Google Scholar] [CrossRef] [Green Version]
- Weissenberger, J.; Loeffler, S.; Kappeler, A.; Kopf, M.; Lukes, A.; Afanasieva, T.A.; Aguzzi, A.; Weis, J. IL-6 is required for glioma development in a mouse model. Oncogene 2004, 23, 3308–3316. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Liddelow, S.A.; Guttenplan, K.A.; Clarke, L.E.; Bennett, F.C.; Bohlen, C.J.; Schirmer, L.; Bennett, M.L.; Münch, A.E.; Chung, W.S.; Peterson, T.C.; et al. Neurotoxic reactive astrocytes are induced by activated microglia. Nature 2017, 541, 481–487. [Google Scholar] [CrossRef]
- Brat, D.J.; Bellail, A.C.; Van Meir, E.G. The role of interleukin-8 and its receptors in gliomagenesis and tumoral angiogenesis. Neuro Oncol. 2005, 7, 122–133. [Google Scholar] [CrossRef] [PubMed]
- Raychaudhuri, B.; Vogelbaum, M.A. IL-8 is a mediator of NF-κB induced invasion by gliomas. J. Neurooncol. 2011, 101, 227–235. [Google Scholar] [CrossRef]
- Carlsson, A.; Persson, O.; Ingvarsson, J.; Widegren, B.; Salford, L.; Borrebaeck, C.A.K.; Wingren, C. Plasma proteome profiling reveals biomarker patterns associated with prognosis and therapy selection in glioblastoma multiforme patients. Proteomics Clin. Appl. 2010, 4, 591–602. [Google Scholar] [CrossRef] [PubMed]
- Samaras, V.; Piperi, C.; Levidou, G.; Zisakis, A.; Kavantzas, N.; Themistocleous, M.S.; Boviatsis, E.I.; Barbatis, C.; Lea, R.W.; Kalofoutis, A.; et al. Analysis of interleukin (IL)-8 expression in human astrocytomas: Associations with IL-6, cyclooxygenase-2, vascular endothelial growth factor, and microvessel morphometry. Hum. Immunol. 2009, 70, 391–397. [Google Scholar] [CrossRef]
- De la Iglesia, N.; Konopka, G.; Lim, K.-L.; Nutt, C.L.; Bromberg, J.F.; Frank, D.A.; Mischel, P.S.; Louis, D.N.; Bonni, A. Deregulation of a STAT3-interleukin 8 signaling pathway promotes human glioblastoma cell proliferation and invasiveness. J. Neurosci. 2008, 28, 5870–5878. [Google Scholar] [CrossRef]
- 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]
- Lu, Y.; Jiang, F.; Zheng, X.; Katakowski, M.; Buller, B.; To, S.-S.T.; Chopp, M. TGF-β1 promotes motility and invasiveness of glioma cells through activation of ADAM17. Oncol. Rep. 2011, 25, 1329–1335. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Grauer, O.; Pöschl, P.; Lohmeier, A.; Adema, G.J.; Bogdahn, U. Toll-like receptor triggered dendritic cell maturation and IL-12 secretion are necessary to overcome T-cell inhibition by glioma-associated TGF-beta2. J. Neurooncol. 2007, 82, 151–161. [Google Scholar] [CrossRef]
- Llopiz, D.; Dotor, J.; Casares, N.; Bezunartea, J.; Díaz-Valdés, N.; Ruiz, M.; Aranda, F.; Berraondo, P.; Prieto, J.; Lasarte, J.J.; et al. Peptide inhibitors of transforming growth factor-β enhance the efficacy of antitumor immunotherapy. Int. J. Cancer 2009, 125, 2614–2623. [Google Scholar] [CrossRef]
- Sanchez-Perez, Y.; Soto-Reyes, E.; Garcia-Cuellar, C.M.; Cacho-Diaz, B.; Santamaria, A.; Rangel-Lopez, E. Role of Epigenetics and Oxidative Stress in Gliomagenesis. CNS Neurol. Disord. Drug Targets 2017, 16, 1090–1098. [Google Scholar] [CrossRef] [PubMed]
- Iida, T.; Furuta, A.; Kawashima, M.; Nishida, J.; Nakabeppu, Y.; Iwaki, T. Accumulation of 8-oxo-2′-deoxyguanosine and increased expression of hMTH1 protein in brain tumors. Neuro Oncol. 2001, 3, 73–81. [Google Scholar] [CrossRef] [PubMed]
- Leung, S.Y.; Yuen, S.T.; Chan, T.L.; Chan, A.S.; Ho, J.W.; Kwan, K.; Fan, Y.W.; Hung, K.N.; Chung, L.P.; Wyllie, A.H. Chromosomal instability and p53 inactivation are required for genesis of glioblastoma but not for colorectal cancer in patients with germline mismatch repair gene mutation. Oncogene 2000, 19, 4079–4083. [Google Scholar] [CrossRef] [Green Version]
- Nitta, M.; Kozono, D.; Kennedy, R.; Stommel, J.; Ng, K.; Zinn, P.O.; Kushwaha, D.; Kesari, S.; Inda, M.-M.; Wykosky, J.; et al. Targeting EGFR induced oxidative stress by PARP1 inhibition in glioblastoma therapy. PLoS ONE 2010, 5, e10767. [Google Scholar] [CrossRef]
- See, A.P.; Han, J.E.; Phallen, J.; Binder, Z.; Gallia, G.; Pan, F.; Jinasena, D.; Jackson, C.; Belcaid, Z.; Jeong, S.J.; et al. The role of STAT3 activation in modulating the immune microenvironment of GBM. J. Neurooncol. 2012, 110, 359–368. [Google Scholar] [CrossRef]
- Rink, C.; Khanna, S. Significance of brain tissue oxygenation and the arachidonic acid cascade in stroke. Antioxid. Redox Signal. 2011, 14, 1889–1903. [Google Scholar] [CrossRef] [Green Version]
- Beppu, T.; Kamada, K.; Yoshida, Y.; Arai, H.; Ogasawara, K.; Ogawa, A. Change of oxygen pressure in glioblastoma tissue under various conditions. J. Neurooncol. 2002, 58, 47–52. [Google Scholar] [CrossRef] [PubMed]
- Rong, Y.; Durden, D.L.; Van Meir, E.G.; Brat, D.J. ‘Pseudopalisading’ necrosis in glioblastoma: A familiar morphologic feature that links vascular pathology, hypoxia, and angiogenesis. J. Neuropathol. Exp. Neurol. 2006, 65, 529–539. [Google Scholar] [CrossRef]
- Jensen, R.L.; Mumert, M.L.; Gillespie, D.L.; Kinney, A.Y.; Schabel, M.C.; Salzman, K.L. Preoperative dynamic contrast-enhanced MRI correlates with molecular markers of hypoxia and vascularity in specific areas of intratumoral microenvironment and is predictive of patient outcome. Neuro Oncol. 2014, 16, 280–291. [Google Scholar] [CrossRef] [Green Version]
- Kaur, B.; Khwaja, F.W.; Severson, E.A.; Matheny, S.L.; Brat, D.J.; Van Meir, E.G. Hypoxia and the hypoxia-inducible-factor pathway in glioma growth and angiogenesis. Neuro Oncol. 2005, 7, 134–153. [Google Scholar] [CrossRef] [Green Version]
- Keith, B.; Johnson, R.S.; Simon, M.C. HIF1α and HIF2α: Sibling rivalry in hypoxic tumour growth and progression. Nat. Rev. Cancer 2011, 12, 9–22. [Google Scholar] [CrossRef] [Green Version]
- Nusblat, L.M.; Tanna, S.; Roth, C.M. Gene silencing of HIF-2α disrupts glioblastoma stem cell phenotype. Cancer Drug Resist. 2020, 3, 199–208. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Jewell, U.R.; Kvietikova, I.; Scheid, A.; Bauer, C.; Wenger, R.H.; Gassmann, M. Induction of HIF-1alpha in response to hypoxia is instantaneous. FASEB J. 2001, 15, 1312–1314. [Google Scholar] [CrossRef]
- Majmundar, A.J.; Wong, W.J.; Simon, M.C. Hypoxia-inducible factors and the response to hypoxic stress. Mol. Cell 2010, 40, 294–309. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Duan, C. Hypoxia-inducible factor 3 biology: Complexities and emerging themes. Am. J. Physiol. Cell Physiol. 2016, 310, C260–C269. [Google Scholar] [CrossRef] [Green Version]
- Holland, E.C.; Hively, W.P.; DePinho, R.A.; Varmus, H.E. A constitutively active epidermal growth factor receptor cooperates with disruption of G1 cell-cycle arrest pathways to induce glioma-like lesions in mice. Genes Dev. 1998, 12, 3675–3685. [Google Scholar] [CrossRef] [Green Version]
- Zhang, Z.; Yao, L.; Yang, J.; Wang, Z.; Du, G. PI3K/Akt and HIF-1 signaling pathway in hypoxia-ischemia (Review). Mol. Med. Rep. 2018, 18, 3547–3554. [Google Scholar] [CrossRef] [Green Version]
- Ma, J.; Sawai, H.; Matsuo, Y.; Ochi, N.; Yasuda, A.; Takahashi, H.; Wakasugi, T.; Funahashi, H.; Sato, M.; Takeyama, H. IGF-1 mediates PTEN suppression and enhances cell invasion and proliferation via activation of the IGF-1/PI3K/Akt signaling pathway in pancreatic cancer cells. J. Surg. Res. 2010, 160, 90–101. [Google Scholar] [CrossRef] [PubMed]
- Iwakuma, T.; Lozano, G. MDM2, an introduction. Mol. Cancer Res. 2003, 1, 993–1000. [Google Scholar]
- Nieminen, A.L.; Qanungo, S.; Schneider, E.A.; Jiang, B.H.; Agani, F.H. Mdm2 and HIF-1alpha interaction in tumor cells during hypoxia. J. Cell Physiol. 2005, 204, 364–369. [Google Scholar] [CrossRef]
- Li, L.C.; Zhang, M.; Feng, Y.K.; Wang, X.J. IDH1-R132H Suppresses Glioblastoma Malignancy through FAT1-ROS-HIF-1α Signaling. Neurol. India 2020, 68, 1050–1058. [Google Scholar] [CrossRef]
- Madan, E.; Dikshit, B.; Gowda, S.H.; Srivastava, C.; Sarkar, C.; Chattopadhyay, P.; Sinha, S.; Chosdol, K. FAT1 is a novel upstream regulator of HIF1α and invasion of high-grade glioma. Int. J. Cancer 2016, 139, 2570–2582. [Google Scholar] [CrossRef] [PubMed]
- Chosdol, K.; Misra, A.; Puri, S.; Srivastava, T.; Chattopadhyay, P.; Sarkar, C. Frequent loss of heterozygosity and altered expression of the candidate tumor suppressor gene ‘FAT’ in human astrocytic tumors. BMC Cancer 2009, 9, 5. [Google Scholar] [CrossRef] [Green Version]
- 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]
- Meyer, M.; Reimand, J.; Lan, X.; Head, R.; Zhu, X.; Kushida, M.; Bayani, J.; Pressey, J.C.; Lionel, A.C.; Clarke, I.D.; et al. Single cell-derived clonal analysis of human glioblastoma links functional and genomic heterogeneity. Proc. Natl. Acad. Sci. USA 2015, 112, 851–856. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Zhao, Y.; Carter, R.; Natarajan, S.; Varn, F.S.; Compton, D.A.; Gawad, C.; Cheng, C.; Godek, K.M. Single-cell RNA sequencing reveals the impact of chromosomal instability on glioblastoma cancer stem cells. BMC Med. Genom. 2019, 12, 79. [Google Scholar] [CrossRef]
- Keith, B.; Simon, M.C. Hypoxia-inducible factors, stem cells, and cancer. Cell 2007, 129, 465–472. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Li, Z.; Bao, S.; Wu, Q.; Wang, H.; Eyler, C.; Sathornsumetee, S.; Shi, Q.; Cao, Y.; Lathia, J.; McLendon, R.E.; et al. Hypoxia-inducible factors regulate tumorigenic capacity of glioma stem cells. Cancer Cell 2009, 15, 501–513. [Google Scholar] [CrossRef] [Green Version]
- Ishii, A.; Kimura, T.; Sadahiro, H.; Kawano, H.; Takubo, K.; Suzuki, M.; Ikeda, E. Histological Characterization of the Tumorigenic “Peri-Necrotic Niche” Harboring Quiescent Stem-Like Tumor Cells in Glioblastoma. PLoS ONE 2016, 11, e0147366. [Google Scholar] [CrossRef]
- Aderetti, D.A.; Hira, V.; Molenaar, R.J.; van Noorden, C. The hypoxic peri-arteriolar glioma stem cell niche, an integrated concept of five types of niches in human glioblastoma. Biochim. Biophys. Acta Rev. Cancer 2018, 1869, 346–354. [Google Scholar] [CrossRef] [PubMed]
- Zeng, X.; Wang, Q.; Tan, X.; Jia, L.; Li, Y.; Hu, M.; Zhang, Z.; Bai, X.; Zhu, Y.; Yang, X. Mild thermotherapy and hyperbaric oxygen enhance sensitivity of TMZ/PSi nanoparticles via decreasing the stemness in glioma. J. Nanobiotechnol. 2019, 17, 47. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Codrici, E.; Enciu, A.M.; Popescu, I.D.; Mihai, S.; Tanase, C. Glioma Stem Cells and Their Microenvironments: Providers of Challenging Therapeutic Targets. Stem Cells Int. 2016, 2016, 5728438. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Mergenthaler, P.; Lindauer, U.; Dienel, G.A.; Meisel, A. Sugar for the brain: The role of glucose in physiological and pathological brain function. Trends Neurosci. 2013, 36, 587–597. [Google Scholar] [CrossRef] [Green Version]
- Tieu, M.T.; Lovblom, L.E.; McNamara, M.G.; Mason, W.; Laperriere, N.; Millar, B.A.; Ménard, C.; Kiehl, T.R.; Perkins, B.A.; Chung, C. Impact of glycemia on survival of glioblastoma patients treated with radiation and temozolomide. J. Neurooncol. 2015, 124, 119–126. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Bao, Z.; Chen, K.; Krepel, S.; Tang, P.; Gong, W.; Zhang, M.; Liang, W.; Trivett, A.; Zhou, M.; Wang, J.M. High Glucose Promotes Human Glioblastoma Cell Growth by Increasing the Expression and Function of Chemoattractant and Growth Factor Receptors. Transl. Oncol. 2019, 12, 1155–1163. [Google Scholar] [CrossRef]
- Hsu, P.P.; Sabatini, D.M. Cancer cell metabolism: Warburg and beyond. Cell 2008, 134, 703–707. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Liberti, M.V.; Locasale, J.W. The Warburg Effect: How Does it Benefit Cancer Cells? Trends Biochem. Sci. 2016, 41, 211–218. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Locasale, J.W.; Cantley, L.C. Metabolic flux and the regulation of mammalian cell growth. Cell Metab. 2011, 14, 443–451. [Google Scholar] [CrossRef] [Green Version]
- Chang, C.H.; Qiu, J.; O’Sullivan, D.; Buck, M.D.; Noguchi, T.; Curtis, J.D.; Chen, Q.; Gindin, M.; Gubin, M.M.; van der Windt, G.J.; et al. Metabolic Competition in the Tumor Microenvironment Is a Driver of Cancer Progression. Cell 2015, 162, 1229–1241. [Google Scholar] [CrossRef] [Green Version]
- Kucharzewska, P.; Christianson, H.C.; Belting, M. Global profiling of metabolic adaptation to hypoxic stress in human glioblastoma cells. PLoS ONE 2015, 10, e0116740. [Google Scholar] [CrossRef]
- Kim, J.W.; Tchernyshyov, I.; Semenza, G.L.; Dang, C.V. HIF-1-mediated expression of pyruvate dehydrogenase kinase: A metabolic switch required for cellular adaptation to hypoxia. Cell Metab. 2006, 3, 177–185. [Google Scholar] [CrossRef] [Green Version]
- Papandreou, I.; Cairns, R.A.; Fontana, L.; Lim, A.L.; Denko, N.C. HIF-1 mediates adaptation to hypoxia by actively downregulating mitochondrial oxygen consumption. Cell Metab. 2006, 3, 187–197. [Google Scholar] [CrossRef] [Green Version]
- Dimmer, K.S.; Friedrich, B.; Lang, F.; Deitmer, J.W.; Bröer, S. The low-affinity monocarboxylate transporter MCT4 is adapted to the export of lactate in highly glycolytic cells. Biochem. J. 2000, 350, 219–227. [Google Scholar] [CrossRef] [PubMed]
- Bartrons, R.; Simon-Molas, H.; Rodríguez-García, A.; Castaño, E.; Navarro-Sabaté, À.; Manzano, A.; Martinez-Outschoorn, U.E. Fructose 2,6-Bisphosphate in Cancer Cell Metabolism. Front. Oncol. 2018, 8, 331. [Google Scholar] [CrossRef] [PubMed]
- Schulze, A.; Harris, A.L. How cancer metabolism is tuned for proliferation and vulnerable to disruption. Nature 2012, 491, 364–373. [Google Scholar] [CrossRef]
- Kalyanaraman, B. Teaching the basics of cancer metabolism: Developing antitumor strategies by exploiting the differences between normal and cancer cell metabolism. Redox Biol. 2017, 12, 833–842. [Google Scholar] [CrossRef] [PubMed]
- Niimi, N.; Yako, H.; Takaku, S.; Chung, S.K.; Sango, K. Aldose Reductase and the Polyol Pathway in Schwann Cells: Old and New Problems. Int. J. Mol. Sci. 2021, 22, 1031. [Google Scholar] [CrossRef]
- Halliwell, B. Oxidative stress and cancer: Have we moved forward? Biochem. J. 2007, 401, 1–11. [Google Scholar] [CrossRef]
- Pereira, E.R.; Frudd, K.; Awad, W.; Hendershot, L.M. Endoplasmic reticulum (ER) stress and hypoxia response pathways interact to potentiate hypoxia-inducible factor 1 (HIF-1) transcriptional activity on targets like vascular endothelial growth factor (VEGF). J. Biol. Chem. 2014, 289, 3352–3364. [Google Scholar] [CrossRef] [Green Version]
- Krawczynski, K.; Godlewski, J.; Bronisz, A. Oxidative Stress-Part of the Solution or Part of the Problem in the Hypoxic Environment of a Brain Tumor. Antioxidants 2020, 9, 747. [Google Scholar] [CrossRef]
- Strickland, M.; Stoll, E.A. Metabolic Reprogramming in Glioma. Front. Cell Dev. Biol. 2017, 5, 43. [Google Scholar] [CrossRef] [Green Version]
- 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]
- Delikatny, E.J.; Chawla, S.; Leung, D.J.; Poptani, H. MR-visible lipids and the tumor microenvironment. NMR Biomed. 2011, 24, 592–611. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Menard, J.A.; Christianson, H.C.; Kucharzewska, P.; Bourseau-Guilmain, E.; Svensson, K.J.; Lindqvist, E.; Indira Chandran, V.; Kjellén, L.; Welinder, C.; Bengzon, J.; et al. Metastasis Stimulation by Hypoxia and Acidosis-Induced Extracellular Lipid Uptake Is Mediated by Proteoglycan-Dependent Endocytosis. Cancer Res. 2016, 76, 4828–4840. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Zaidi, N.; Lupien, L.; Kuemmerle, N.B.; Kinlaw, W.B.; Swinnen, J.V.; Smans, K. Lipogenesis and lipolysis: The pathways exploited by the cancer cells to acquire fatty acids. Prog. Lipid Res. 2013, 52, 585–589. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Pirmoradi, L.; Seyfizadeh, N.; Ghavami, S.; Zeki, A.A.; Shojaei, S. Targeting cholesterol metabolism in glioblastoma: A new therapeutic approach in cancer therapy. J. Investig. Med. 2019, 67, 715–719. [Google Scholar] [CrossRef] [Green Version]
- Barber, C.N.; Raben, D.M. Lipid Metabolism Crosstalk in the Brain: Glia and Neurons. Front. Cell. Neurosci. 2019, 13, 212. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Kambach, D.M.; Halim, A.S.; Cauer, A.G.; Sun, Q.; Tristan, C.A.; Celiku, O.; Kesarwala, A.H.; Shankavaram, U.; Batchelor, E.; Stommel, J.M. Disabled cell density sensing leads to dysregulated cholesterol synthesis in glioblastoma. Oncotarget 2017, 8, 14860–14875. [Google Scholar] [CrossRef]
- Ahmad, F.; Sun, Q.; Patel, D.; Stommel, J.M. Cholesterol Metabolism: A Potential Therapeutic Target in Glioblastoma. Cancers 2019, 11, 146. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Villa, G.R.; Hulce, J.J.; Zanca, C.; Bi, J.; Ikegami, S.; Cahill, G.L.; Gu, Y.; Lum, K.M.; Masui, K.; Yang, H.; et al. An LXR-Cholesterol Axis Creates a Metabolic Co-Dependency for Brain Cancers. Cancer Cell 2016, 30, 683–693. [Google Scholar] [CrossRef] [Green Version]
- Lieu, E.L.; Nguyen, T.; Rhyne, S.; Kim, J. Amino acids in cancer. Exp. Mol. Med. 2020, 52, 15–30. [Google Scholar] [CrossRef]
- Reeds, P.J. Dispensable and indispensable amino acids for humans. J. Nutr. 2000, 130, 1835S–1840S. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Zhang, Y.; Commisso, C. Macropinocytosis in Cancer: A Complex Signaling Network. Trends Cancer 2019, 5, 332–334. [Google Scholar] [CrossRef] [PubMed]
- Muir, A.; Danai, L.V.; Gui, D.Y.; Waingarten, C.Y.; Lewis, C.A.; Vander Heiden, M.G. Environmental cystine drives glutamine anaplerosis and sensitizes cancer cells to glutaminase inhibition. eLife 2017, 6, e27713. [Google Scholar] [CrossRef] [PubMed]
- Mayers, J.R.; Torrence, M.E.; Danai, L.V.; Papagiannakopoulos, T.; Davidson, S.M.; Bauer, M.R.; Lau, A.N.; Ji, B.W.; Dixit, P.D.; Hosios, A.M.; et al. Tissue of origin dictates branched-chain amino acid metabolism in mutant Kras-driven cancers. Science 2016, 353, 1161–1165. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Ananieva, E.A.; Wilkinson, A.C. Branched-chain amino acid metabolism in cancer. Curr. Opin. Clin. Nutr. Metab. Care 2018, 21, 64–70. [Google Scholar] [CrossRef] [Green Version]
- Zhang, B.; Chen, Y.; Shi, X.; Zhou, M.; Bao, L.; Hatanpaa, K.J.; Patel, T.; DeBerardinis, R.J.; Wang, Y.; Luo, W. Regulation of branched-chain amino acid metabolism by hypoxia-inducible factor in glioblastoma. Cell. Mol. Life Sci. 2021, 78, 195–206. [Google Scholar] [CrossRef]
- Srivastava, C.; Irshad, K.; Dikshit, B.; Chattopadhyay, P.; Sarkar, C.; Gupta, D.K.; Sinha, S.; Chosdol, K. FAT1 modulates EMT and stemness genes expression in hypoxic glioblastoma. Int. J. Cancer 2018, 142, 805–812. [Google Scholar] [CrossRef] [Green Version]
- Gu, Z.; Cui, X.; Sun, P.; Wang, X. Regulatory Roles of Tumor Necrosis Factor-α-Induced Protein 8 Like-Protein 2 in Inflammation, Immunity and Cancers: A Review. Cancer Manag. Res. 2020, 12, 12735–12746. [Google Scholar] [CrossRef]
- Bhagat, M.; Palanichamy, J.K.; Ramalingam, P.; Mudassir, M.; Irshad, K.; Chosdol, K.; Sarkar, C.; Seth, P.; Goswami, S.; Sinha, S.; et al. HIF-2α mediates a marked increase in migration and stemness characteristics in a subset of glioma cells under hypoxia by activating an Oct-4/Sox-2-Mena (INV) axis. Int. J. Biochem. Cell Biol. 2016, 74, 60–71. [Google Scholar] [CrossRef]
- Sun, H.; Gong, S.; Carmody, R.J.; Hilliard, A.; Li, L.; Sun, J.; Kong, L.; Xu, L.; Hilliard, B.; Hu, S.; et al. TIPE2, a negative regulator of innate and adaptive immunity that maintains immune homeostasis. Cell 2008, 133, 415–426. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Cao, X.; Zhang, L.; Shi, Y.; Sun, Y.; Dai, S.; Guo, C.; Zhu, F.; Wang, Q.; Wang, J.; Wang, X.; et al. Human tumor necrosis factor (TNF)-alpha-induced protein 8-like 2 suppresses hepatocellular carcinoma metastasis through inhibiting Rac1. Mol. Cancer 2013, 12, 149. [Google Scholar] [CrossRef] [Green Version]
- Li, Y.; Li, X.; Liu, G.; Sun, R.; Wang, L.; Wang, J.; Wang, H. Downregulated TIPE2 is associated with poor prognosis and promotes cell proliferation in non-small cell lung cancer. Biochem. Biophys. Res. Commun. 2015, 457, 43–49. [Google Scholar] [CrossRef] [PubMed]
- Liu, Z.J.; Liu, H.L.; Zhou, H.C.; Wang, G.C. TIPE2 Inhibits Hypoxia-Induced Wnt/β-Catenin Pathway Activation and EMT in Glioma Cells. Oncol. Res. 2016, 24, 255–261. [Google Scholar] [CrossRef] [PubMed]
- Tsai, Y.P.; Wu, K.J. Hypoxia-regulated target genes implicated in tumor metastasis. J. Biomed. Sci. 2012, 19, 102. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Busuioc, C.; Ciocan-Cartita, C.A.; Braicu, C.; Zanoaga, O.; Raduly, L.; Trif, M.; Muresan, M.S.; Ionescu, C.; Stefan, C.; Crivii, C.; et al. Epithelial-Mesenchymal Transition Gene Signature Related to Prognostic in Colon Adenocarcinoma. J. Pers. Med. 2021, 11, 476. [Google Scholar] [CrossRef]
- Weathers, S.P.; de Groot, J. VEGF Manipulation in Glioblastoma. Oncology 2015, 29, 720–727. [Google Scholar] [PubMed]
- Yi, Y.; Hsieh, I.Y.; Huang, X.; Li, J.; Zhao, W. Glioblastoma Stem-Like Cells: Characteristics, Microenvironment, and Therapy. Front. Pharmacol. 2016, 7, 477. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Caffo, M.; Barresi, V.; Caruso, G.; La Fata, G.; Pino, M.A.; Raudino, G.; Alafaci, C.; Tomasello, F. Gliomas Biology: Angiogenesis and Invasion. In Evolution of the Molecular Biology of Brain Tumors and the Therapeutic Implications; Lichtor, T., Ed.; In Tech Open: London, UK, 2013; pp. 37–103. [Google Scholar]
- Angara, K.; Rashid, M.H.; Shankar, A.; Ara, R.; Iskander, A.; Borin, T.F.; Jain, M.; Achyut, B.R.; Arbab, A.S. Vascular mimicry in glioblastoma following anti-angiogenic and anti-20-HETE therapies. Histol. Histopathol. 2017, 32, 917–928. [Google Scholar] [CrossRef]
- Melincovici, C.S.; Boşca, A.B.; Şuşman, S.; Mărginean, M.; Mihu, C.; Istrate, M.; Moldovan, I.M.; Roman, A.L.; Mihu, C.M. Vascular endothelial growth factor (VEGF)—Key factor in normal and pathological angiogenesis. Rom. J. Morphol. Embryol. 2018, 59, 455–467. [Google Scholar] [PubMed]
- Tena-Suck, M.L.; Celis-Lopez, M.A.; Collado-Ortiz, M.A.; Castillejos-Lopez, M.; Tenorio-Serralta, M. Glioblastoma Multiforme and Angiogenesis: A Clinicopathological and Immunohistochemistry Approach. J. Neurol. Res. 2015, 5, 199–206. [Google Scholar] [CrossRef] [Green Version]
- Hambardzumyan, D.; Bergers, G. Glioblastoma: Defining Tumor Niches. Trends Cancer 2015, 1, 252–265. [Google Scholar] [CrossRef] [Green Version]
- Ahir, B.K.; Engelhard, H.H.; Lakka, S.S. Tumor Development and Angiogenesis in Adult Brain Tumor: Glioblastoma. Mol. Neurobiol. 2020, 57, 2461–2478. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Kioi, M.; Vogel, H.; Schultz, G.; Hoffman, R.M.; Harsh, G.R.; Brown, J.M. Inhibition of vasculogenesis, but not angiogenesis, prevents the recurrence of glioblastoma after irradiation in mice. J. Clin. Investig. 2010, 120, 694–705. [Google Scholar] [CrossRef]
- Mao, J.M.; Liu, J.; Guo, G.; Mao, X.G.; Li, C.X. Glioblastoma vasculogenic mimicry: Signaling pathways progression and potential anti-angiogenesis targets. Biomark. Res. 2015, 3, 8. [Google Scholar] [CrossRef] [Green Version]
- Pearson, J.R.D.; Regad, T. Targeting cellular pathways in glioblastoma multiforme. Signal Transduct. Target Ther. 2017, 2, 17040. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Brat, D.J.; Castellano-Sanchez, A.; Kaur, B.; Van Meir, E.G. Genetic and Biologic Progression in Astrocytomas and Their Relation to Angiogenic Dysregulation. Adv. Anat. Pathol. 2002, 9, 24–36. [Google Scholar] [CrossRef]
- Atzori, M.G.; Tentori, L.; Ruffini, F.; Ceci, C.; Bonanno, E.; Scimeca, M.; Lacal, P.M.; Graziani, G. The Anti-Vascular Endothelial Growth Factor Receptor-1 Monoclonal Antibody D16F7 Inhibits Glioma Growth and Angiogenesis In Vivo. J. Pharmacol. Exp. Ther. 2018, 364, 77–86. [Google Scholar] [CrossRef]
- Plate, K.H. Mechanisms of Angiogenesis in the Brain. J. Neuropathol. Exp. Neurol. 1999, 58, 313–320. [Google Scholar] [CrossRef] [Green Version]
- Hundsberger, T.; Reardon, D.A.; Wen, P.Y. Angiogenesis inhibitors in tackling recurrent glioblastoma. Expert Rev. Anticancer Ther. 2017, 17, 507–515. [Google Scholar] [CrossRef]
- Marcus, H.J.; Carpenter, K.L.; Price, S.J.; Hutchinson, P.J. In vivo assessment of high-grade glioma biochemistry using microdialysis: A study of energy-related molecules, growth factors and cytokines. J. Neurooncol. 2010, 97, 11–23. [Google Scholar] [CrossRef] [PubMed]
- Du, R.; Petritsch, C.; Lu, K.; Liu, P.; Haller, A.; Ganss, R.; Song, H.; Vandenberg, S.; Bergers, G. Matrix metalloproteinase-2 regulates vascular patterning and growth affecting tumor cell survival and invasion in GBM. Neuro Oncol. 2008, 10, 254–264. [Google Scholar] [CrossRef] [Green Version]
- Xue, Q.; Cao, L.; Chen, X.Y.; Zhao, J.; Gao, L.; Li, S.Z.; Fei, Z. High expression of MMP9 in glioma affects cell proliferation and is associated with patient survival rates. Oncol. Lett. 2017, 13, 1325–1330. [Google Scholar] [CrossRef] [Green Version]
- Das, S.; Marsden, P.A. Angiogenesis in Glioblastoma. N. Engl. J. Med. 2013, 369, 1561–1563. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Batchelor, T.T.; Sorensen, A.G.; di Tomaso, E.; Zhang, W.T.; Duda, D.G.; Cohen, K.S.; Kozak, K.R.; Cahill, D.P.; Chen, P.J.; Zhu, M.; et al. AZD2171, a pan-VEGF receptor tyrosine kinase inhibitor, normalizes tumor vasculature and alleviates edema in glioblastoma patients. Cancer Cell 2007, 11, 83–95. [Google Scholar] [CrossRef] [PubMed] [Green Version]
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations. |
© 2022 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
Șovrea, A.S.; Boșca, B.; Melincovici, C.S.; Constantin, A.-M.; Crintea, A.; Mărginean, M.; Dronca, E.; Jianu, M.E.; Suflețel, R.; Gonciar, D.; et al. Multiple Faces of the Glioblastoma Microenvironment. Int. J. Mol. Sci. 2022, 23, 595. https://doi.org/10.3390/ijms23020595
Șovrea AS, Boșca B, Melincovici CS, Constantin A-M, Crintea A, Mărginean M, Dronca E, Jianu ME, Suflețel R, Gonciar D, et al. Multiple Faces of the Glioblastoma Microenvironment. International Journal of Molecular Sciences. 2022; 23(2):595. https://doi.org/10.3390/ijms23020595
Chicago/Turabian StyleȘovrea, Alina Simona, Bianca Boșca, Carmen Stanca Melincovici, Anne-Marie Constantin, Andreea Crintea, Mariana Mărginean, Eleonora Dronca, Mihaela Elena Jianu, Rada Suflețel, Diana Gonciar, and et al. 2022. "Multiple Faces of the Glioblastoma Microenvironment" International Journal of Molecular Sciences 23, no. 2: 595. https://doi.org/10.3390/ijms23020595
APA StyleȘovrea, A. S., Boșca, B., Melincovici, C. S., Constantin, A. -M., Crintea, A., Mărginean, M., Dronca, E., Jianu, M. E., Suflețel, R., Gonciar, D., Bungărdean, M., & Crivii, C. -B. (2022). Multiple Faces of the Glioblastoma Microenvironment. International Journal of Molecular Sciences, 23(2), 595. https://doi.org/10.3390/ijms23020595