128
Views
1
CrossRef citations to date
0
Altmetric
Review Article

Coagulation proteases and neurotransmitters in pathogenicity of glioblastoma multiforme

, , , &
Pages 398-408 | Received 22 Dec 2021, Accepted 15 Jul 2022, Published online: 08 Aug 2022

References

  • Jiang H, Cui Y, Wang J, et al. Impact of epidemiological characteristics of supratentorial gliomas in adults brought about by the 2016 world health organization classification of tumors of the Central nervous system. Oncotarget. 2017;8(12):20354–20361.
  • Mesfin FB, Al-Dhahir MA. 2020. Gliomas . [Updated 2022 Jun 4]. In: StatPearls [Internet]. Treasure Island (FL): StatPearls Publishing; 2022 Jan-. Available from: https://www.ncbi.nlm.nih.gov/books/NBK441874/.
  • Brown TJ, Brennan MC, Li M, et al. Association of the extent of resection with survival in glioblastoma: a systematic review and Meta-analysis. JAMA Oncol. 2016;2(11):1460–1469.
  • Stupp R, Taillibert S, Kanner AA, et al. Maintenance therapy with tumor-treating fields plus temozolomide vs temozolomide alone for glioblastoma: a randomized clinical trial. JAMA. 2015;314(23):2535–2543.
  • O’Sullivan T, Saddawi-Konefka R, Vermi W, et al. Cancer immunoediting by the innate immune system in the absence of adaptive immunity. J Exp Med. 2012;209(10):1869–1882.
  • Francis JM, Zhang CZ, Maire CL, et al. EGFR variant heterogeneity in glioblastoma resolved through single-nucleus sequencing. Cancer Discov. 2014;4(8):956–971.
  • Brennan CW, Verhaak RG, McKenna A, et al. The somatic genomic landscape of glioblastoma. Cell. 2013;155(2):462–477.
  • Singh D, Chan JM, Zoppoli P, et al. Transforming fusions of FGFR and TACC genes in human glioblastoma. Science. 2012;337(6099):1231–1235.
  • Pitz MW, Eisenhauer EA, MacNeil MV, et al. Phase II study of PX-866 in recurrent glioblastoma. Neuro Oncol. 2015;17(9):1270–1274.
  • Verreault M, Schmitt C, Goldwirt L, et al. Preclinical efficacy of the MDM2 inhibitor RG7112 in MDM2-Amplified and TP53 wild-type glioblastomas. Clin Cancer Res. 2016;22(5):1185–1196.
  • Vigneswaran K, Neill S, Hadjipanayis CG. Beyond the world health organization grading of infiltrating gliomas: advances in the molecular genetics of glioma classification. Ann Transl Med. 2015 May;3(7):95.
  • Amirian ES, Zhou RZ, Wrensch MR, et al. Approaching a scientific consensus on the association between allergies and glioma risk: a report from the glioma international Case-Control study. Cancer Epidemiol Biomarkers Prev. 2016;25(2):282–290.
  • Zhao H, Cai W, Su S, et al. Allergic conditions reduce the risk of glioma: a meta-analysis based on 128,936 subjects. Tumor Biol. 2014;35(4):3875–3880.
  • McCarthy BJ, Rankin KM, Aldape K, et al. Risk factors for oligodendroglial tumors: a pooled international study. Neuro Oncol. 2011;13(2):242–250.
  • Baker MJ, Goldstein AM, Gordon PL, et al. An interstitial deletion within 9p21.3 and extending beyond CDKN2A predisposes to melanoma, neural system tumours and possible haematological malignancies. J Med Genet. 2016;53(11):721–727.
  • Brain Tumor Statistics. 2017. Available from: http://www.abta.org/about-us/news/brain-tumor-statistics/.
  • National Brain Tumor Association. 2018. Quick brain tumor facts. Available from: http://braintumor.org/brain-tumor-information/brain-tumor-facts/.
  • Davis ME. Epidemiology and overview of gliomas. Semin Oncol Nurs. 2018;34(5):420–429.
  • Plotnikov SV, Waterman CM. Guiding cell migration by tugging. Curr Opin Cell Biol. 2013;25(5):619–626.
  • DuChez BJ, Doyle AD, Dimitriadis EK, et al. Durotaxis by human cancer cells. Biophys J. 2019;116(4):670–683.
  • Wormer DB, Davis KA, Henderson JH, et al. The focal adhesion-localized CdGAP regulates matrix rigidity sensing and durotaxis. PLoS One. 2014;9(3):e91815.
  • Friedl P, Wolf K. Tumour-cell invasion and migration: diversity and escape mechanisms. Nat Rev Cancer. 2003;3(5):362–374.
  • Hayakawa Y, Kurimoto M, Nagai S, et al. Thrombin-induced cell proliferation and platelet-derived growth factor-AB release from A172 human glioblastoma cells. J Thromb Haemost. 2007;5(11):2219–2226.
  • Luo W, Wang Y, Reiser G. Protease-activated receptors in the brain: receptor expression, activation, and functions in neurodegeneration and neuroprotection. Brain Res Rev. 2007;56(2):331–345.
  • Chambers RC, Leoni P, Blanc-Brude OP, et al. Thrombin is a potent inducer of connective tissue growth factor production via proteolytic activation of protease-activated receptor-1. J. Biol. Chem. 2000;275(45):35584–35591.
  • Darmoul D, Gratio V, Devaud H, et al. Activation of proteinase-activated receptor 1 promotes human Colon cancer cell proliferation through epidermal growth factor receptor transactivation – PubMed. Mol Cancer Res. 2004;2(9):514–522.
  • Xie Q, Bao X, Chen ZH, et al. Role of protease-activated receptor-1 in glioma growth. Acta Neurochirurgica, Suppl. 2016;121:355–360.
  • Hayosh ZI, Stein ES, Last D, et al. Thrombin activity and thrombin receptor in rat glioblastoma model: possible markers and targets for intervention? J Mol Neurosci. 2015;56(3):644–651.
  • Jacobs VL, Valdes PA, Hickey WF, et al. Current review of in vivo GBM rodent models: emphasis on the CNS-1 tumour model. ASN Neuro. 2011;3(3):AN20110014–181.
  • Auvergne RM, Sim FJ, Wang S, et al. Transcriptional differences between normal and glioma-derived glial progenitor cells identify a core set of dysregulated genes. Cell Rep. 2013;3(6):2127–2141.
  • Vianello F, Sambado L, Goss A, et al. Dabigatran antagonizes growth, cell-cycle progression, migration, and endothelial tube formation induced by thrombin in breast and glioblastoma cell lines. Cancer Med. 2016;5(10):2886–2898.
  • Bahou WF, Nierman WC, Durkin AS, et al. Chromosomal assignment of the human thrombin receptor gene: localization to region q13 of chromosome 5. Blood. 1993;82(5):1532–1537.
  • Nystedt S, Emilsson K, Wahlestedt C, et al. Molecular cloning of a potential proteinase activated receptor. Proc Natl Acad Sci U S A. 1994;91(20):9208–9212.
  • Schmidt VA, Nierman WC, Maglott DR, et al. The human proteinase-activated receptor-3 (PAR-3) gene. Identification within a par gene cluster and characterization in vascular endothelial cells and platelets. J Biol Chem. 1998;273(24):15061–15068.
  • Kahn ML, Zeng YW, Huang W, et al. A dual thrombin receptor system for platelet activation. Nature. 1998;394(6694):690–694.
  • Vu TK, Wheaton VI, Hung DT, et al. Domains specifying thrombin-receptor interaction. Nature. 1991;353(6345):674–677.
  • Nystedt S, Emilsson K, Larsson AK, et al. Molecular cloning and functional expression of the gene encoding the human proteinase-activated receptor 2. Eur J Biochem. 1995;232(1):84–89.
  • Xu WF, Andersen H, Whitmore TE, et al. Cloning and characterization of human protease-activated receptor 4. Proc Natl Acad Sci U S A. 1998;95(12):6642–6646.
  • Heuberger DM, Schuepbach RA. Protease-activated receptors (PARs): mechanisms of action and potential therapeutic modulators in PAR-driven inflammatory diseases. Thromb J. 2019;17:1–24.
  • Gerszten RE, Chen J, Ishii M, et al. Specificity of the thrombin receptor for agonist peptide is defined by its extracellular surface. Nature. 1994;368(6472):648–651.
  • Macfarlane SR, Seatter MJ, Kanke T, et al. Proteinase- activated receptors. Pharmacol Rev. 2001;53(2):245–282.
  • Russo A, Soh UJ, Paing MM, et al. Caveolae are required for protease-selective signaling by protease-activated receptor-1. Proc Natl Acad Sci U S A. 2009;106(15):6393–6397.
  • Cretu A, Fotos JS, Little BW, et al. Human and rat glioma growth, invasion, and vascularization in a novel chick embryo brain tumor model. Clin Exp Metastasis. 2005;22(3):225–236.
  • Hanoun M, Maryanovich M, Estape AA, et al. Neural regulation of hematopoiesis, inflammation, and cancer. Neuron. 2015;86(2):360–373.
  • Entschladen F, Palm D, Niggeman B, et al. The cancer’s nervous tooth: considering the neuronal crosstalk within tumors. Semin Cancer Biol. 2008;18(3):171–175.
  • Entschladen F, Drell TL, Lang K, et al. Tumour-cell migration, invasion, and metastasis: navigation by neurotransmitters. Lancet Oncol. 2004;5(4):254–258.
  • Jiang S-H, Hu LP, Wang X, et al. Neurotransmitters: emerging targets in cancer. Oncogene. 2019;39:503–515.
  • de Groot J, Sontheimer H. Glutamate and the biology of gliomas. Glia. 2011;59(8):1181–1189.
  • Ye ZC, Sontheimer H. Glioma cells release excitotoxic concentrations of glutamate – PubMed. Cancer Res. 1999;59(17):4383–4391.
  • Takano T, Lin JH, Arcuino G, et al. Glutamate release promotes growth of malignant gliomas. Nat Med. 2001;7(9):1010–1015.
  • Choi DW. Glutamate neurotoxicity and diseases of the nervous system. Neuron. 1988;1(8):623–634.
  • Ungard R, Singh G. 2011. Oxidative Stress and Glutamate Release in Glioma. In: Ghosh A, editor. Glioma - Exploring Its Biology and Practical Relevance [Internet]. London: IntechOpen; 2011.
  • Li SC, Vu LT, Ho HW, et al. Cancer stem cells from a rare form of glioblastoma multiforme involving the neurogenic ventricular wall. Cancer Cell Int. 2012;12(1):41–19.
  • C L JS, MR M, et al. Mosaic analysis with double markers reveals tumor cell of origin in glioma. Cell. 2011;146:209–221.
  • L U MK, FK J, et al. Cell type-specific tumor suppression by Ink4a and Arf in Kras-induced mouse gliomagenesis. Cancer Res. 2005;65:2065–2069.
  • Blanchart A, Fernando R, Häring M, et al. Endogenous GAB AA receptor activity suppresses glioma growth. Oncogene. 2017;36(6):777–786.
  • Platel JC, Dave KA, Bordey A. Control of neuroblast production and migration by converging GABA and glutamate signals in the postnatal forebrain. J Physiol. 2008;586(16):3739–3743.
  • JS R, T U KI, et al. Frontal, midbrain and striatal dopaminergic function in early and advanced Parkinson’s disease a 3D [(18)F]dopa-PET study. Brain. 1999;122(Pt 9):1637–1650.
  • Zhu Y, Guignard F, Zhao D, et al. Early inactivation of p53 tumor suppressor gene cooperating with NF1 loss induces malignant astrocytoma. Cancer Cell. 2005;8(2):119–130.
  • Qin T, Wang C, Chen X, et al. Dopamine induces growth inhibition and vascular normalization through reprogramming M2-polarized macrophages in rat C6 glioma. Toxicol Appl Pharmacol. 2015;286(2):112–123.
  • Verhaak RGW, Hoadley KA, Purdom E, et al. Integrated genomic analysis identifies clinically relevant subtypes of glioblastoma characterized by abnormalities in PDGFRA, IDH1, EGFR, and NF1. Cancer Cell. 2010;17(1):98–110.
  • Hoglinger GU, Rizk P, Muriel MP, et al. Dopamine depletion impairs precursor cell proliferation in Parkinson disease. Nat Neurosci. 2004;7(7):726–735.
  • Hottinger AF, Stupp R, Homicsko K. Standards of care and novel approaches in the management of glioblastoma multiforme. Chin J Cancer. 2014;33(1):32–39.
  • Caragher SP, Hall RR, Ahsan R, et al. Monoamines in glioblastoma: complex biology with therapeutic potential. Neuro Oncol. 2018;20(8):1014–1025.
  • Lee EYHP, Muller WJ. 2010. Oncogenes and tumor suppressor genes. Cold Spring Harb Perspect Biol. 2010 Oct;2(10):a003236.
  • Noble S, Pasi J. Epidemiology and pathophysiology of cancer-associated thrombosis. Br J Cancer. 2010;102(S1):S2–S9.
  • Fuentes HE, Tafur AJ, Caprini JA. Cancer-associated thrombosis. Dis Mon. 2016;62(5):121–158.
  • Omuro A, DeAngelis LM. Glioblastoma and other malignant gliomas: a clinical review. JAMA. 2013;310(17):1842–1850.
  • Fisher JL, Palmisano S, Schwartzbaum JA, et al. Comorbid conditions associated with glioblastoma. J Neurooncol. 2014;116(3):585–591.
  • Kerkhof M, Vecht CJ. Seizure characteristics and prognostic factors of gliomas. Epilepsia. 2013;54(Suppl 9):12–17.
  • Lo M, Wang YZ, Gout PW. The x(c)- cystine/glutamate antiporter: a potential target for therapy of cancer and other diseases. J Cell Physiol. 2008;215(3):593–602.
  • Takeuchi S, Wada K, Toyooka T, et al. Increased xCT expression correlates with tumor invasion and outcome in patients with glioblastomas. Neurosurgery. 2013;72(1):33–41.
  • Dührsen L, Sauvigny T, Ricklefs FL, et al. Seizures as presenting symptom in patients with glioblastoma. Epilepsia. 2019;60(1):149–154.
  • Tönjes M, Barbus S, Park YJ, et al. BCAT1 promotes cell proliferation through amino acid catabolism in gliomas carrying wild-type IDH1. Nat Med. 2013;19(7):901–908.
  • Lange F, Hörnschemeyer J, Kirschstein T. Glutamatergic mechanisms in glioblastoma and tumor-associated epilepsy. Cells. 2021;10(5):1226.
  • Lonardi S, Tosoni A. Adjuvant chemotherapy in the treatment of high grade gliomas. Cancer Treat Rev. 2005 Apr;31(2):79–89.
  • Iacob G, Dinca EB. Current data and strategy in glioblastoma multiforme. J Med Life. 2009;2(4):386–393.
  • Nourmohammadi S, Aung TN, Cui J, et al. Effect of compound kushen injection, a natural compound mixture, and its identified chemical components on migration and invasion of Colon, brain, and breast cancer cell lines. Front Oncol. 2019;9:314.
  • Park M, Song C, Yoon H, et al. Double blockade of glioma cell proliferation and migration by temozolomide conjugated with NPPB, a chloride channel blocker. ACS Chem Neurosci. 2016;7(3):275–285.
  • Lee SY. Temozolomide resistance in glioblastoma multiforme. Genes Dis. 2016;3(3):198–210.
  • Birch JL, Strathdee K, Gilmour L, et al. A novel small-molecule inhibitor of MRCK prevents radiation-driven invasion in glioblastoma. Cancer Res. 2018;78(22):6509–6522.
  • Sun C, Yu Y, Wang L, et al. Additive antiangiogenesis effect of ginsenoside Rg3 with low-dose metronomic temozolomide on rat glioma cells both in vivo and in vitro. J Exp Clin Cancer Res. 2016;35(1):12.
  • Zhang Y, Wang SX, Ma JW, et al. EGCG inhibits properties of glioma stem-like cells and synergizes with temozolomide through downregulation of P-glycoprotein inhibition. J Neurooncol. 2015;121(1):41–52.
  • Tezcan G, Taskapilioglu MO, Tunca B, et al. Olea europaea leaf extract and bevacizumab synergistically exhibit beneficial efficacy upon human glioblastoma cancer stem cells through reducing angiogenesis and invasion in vitro. Biomed Pharmacother. 2017;90:713–723.
  • Yool AJ, Ramesh S. Molecular targets for combined therapeutic strategies to limit glioblastoma cell migration and invasion. Front Pharmacol. 2020;11:358.
  • Baker NC, Lipinski MJ, Lhermusier T, et al. Overview of the 2014 Food and Drug Administration cardiovascular and renal drugs advisory committee meeting about vorapaxar. Circulation. 2014;130:1287–1294.
  • Wang A. Review of vorapaxar for the prevention of atherothrombotic events. Expert Opin Pharmacother. 2015;16:2509–2522.
  • Moschonas IC, Goudevenos JA, Tselepis AD. Protease-activated receptor-1 antagonists in long-term antiplatelet therapy. Current state of evidence and future perspectives. Int J Cardiol. 2015;185:9–18.
  • Chanakira A, Westmark PR, Ong IM, et al.. Tissue factor-factor VIIa complex triggers protease activated receptor 2-dependent growth factor release and migration in ovarian cancer. Gynecol Oncol. 2017;145:167–175.
  • Anastas JN, Shi Y.. Histone serotonylation: Can the brain have “happy” chromatin? Mol Cell. 2019;74(3):418–420.

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.