Research Article
BibTex RIS Cite

DNA Tamir Mekanizması İlişkili Genlerin Biyoinformatik Yöntemlerle Glioblastomda Tanımlanması

Year 2022, Volume: 8 Issue: 2, 117 - 124, 31.05.2022
https://doi.org/10.30934/kusbed.1003777

Abstract

Amaç: DNA tamir mekanizmalarında (DTM) görev alan genlerin ifade değişimleri glioblastomda (GBM) radyoterapi direnci ile ilişkilendirilmiştir. DTM’de rol oynayan genlerin biyoinformatik yöntemlerle tanımlanması GBM tedavisinde kullanılabilecek potansiyel yeni hedeflerin belirlenmesine yardımcı olabilir. Bu çalışmanın amacı, DNA tamir mekanizmalarında rol oynayan genlerin biyoinformatik yöntemler kullanılarak GBM tümörlerinde tanımlanmasıdır.
Yöntem: DNA tamiri mekanizmaları ile ilişkili genler “Reactome” ve “KEGG” veri tabanları üzerinde tanımlandı. GBM tümörlerinde genlere ait mRNA ifade profilleri GEO GDS1813 ve GDS2853 veri setlerinde “Orange Canvas” yazılımı kullanılarak incelendi. Genlerdeki genetik değişimler cBioPortal veri tabanı kullanılarak GBM TCGA olgularında tanımlandı. GEPIA2, değişen gen ifadelerinin TCGA GBM hasta sağ kalım süreleri üzerindeki etkisini göstermek için kullanıldı.
Bulgular: ERCC6, FAN1, MBD4, PARP1 ve UNG genlerinin mRNA ifade profillerinin GBM tümörlerinde değişime uğradığı bulundu. Tanımlanan genler için farklı tipte mutasyonlar ve kopya sayı değişimleri TCGA GBM olgularında gözlendi. Yüksek ve düşük gen ifade profillerinin GBM hastalarının genel ve hastalıksız sağ kalım süreleri üzerinde etkisi olmadığı saptandı.
Sonuç: Bu çalışmada tanımlanan ERCC6, PARP1 ve UNG genleri baskılanması durumunda GBM’de radyoterapi etkinliğini arttırabilecek potansiyel birer terapötik hedef olabilir.

References

  • Louis DN, Perry A, Reifenberger G, von Deimling A, Figarella-Branger D, Cavenee WK, et al. The 2016 World Health Organization Classification of Tumors of the Central Nervous System: a summary. Acta Neuropathol 2016; 131(6):803-820. doi: 10.1007/s00401-016-1545-1.
  • Stupp R, Hegi ME, Mason WP, van den Bent MJ, Taphoorn MJ, Janzer RC, et al. Effects of radiotherapy with concomitant and adjuvant temozolomide versus radiotherapy alone on survival in glioblastoma in a randomised phase III study: 5-year analysis of the EORTC-NCIC trial. Lancet Oncol 2009; 10(5):459-466. doi: 10.1016/S1470-2045(09)70025-7.
  • Henson JW. Treatment of glioblastoma multiforme: a new standard. Arch Neurol. 2006; 63(3): 337-341. doi: 10.1001/archneur.63.3.337.
  • Stupp R, Mason WP, van den Bent MJ, Weller M, Fisher B, Taphoorn MJ, et al. Radiotherapy plus concomitant and adjuvant temozolomide for glioblastoma. N Engl J Med 2005; 352(10): 987-996. doi: 10.1056/NEJMoa043330.
  • Palanichamy K, Chakravarti A. Combining drugs and radiotherapy: from the bench to the bedside. Curr Opin Neurol 2009; 22(6): 625-632. doi: 10.1097/WCO.0b013e3283327d33.
  • Erasimus H, Gobin M, Niclou S, Van Dyck E. DNA repair mechanisms and their clinical impact in glioblastoma. Mutat Res Rev Mutat Res 2016; 769: 19-35. doi: 10.1016/j.mrrev.2016.05.005.
  • Meng W, Palmer JD, Siedow M, Haque SJ, Chakravarti A. Overcoming Radiation Resistance in Gliomas by Targeting Metabolism and DNA Repair Pathways. Int. J. Mol. Sci 2022; 23(4): 2246. doi: 10.3390/ijms23042246.
  • Biau J, Chautard E, Verrelle P, Dutreix M. Altering DNA Repair to Improve Radiation Therapy: Specific and Multiple Pathway Targeting. Front Oncol 2019; 9: 1009. doi: 10.3389/fonc.2019.01009.
  • Kesari S, Advani SJ, Lawson JD, Kahle KT, Ng K, Carter B, et al. DNA damage response and repair: insights into strategies for radiation sensitization of gliomas. Future Oncol 2011; 7(11): 1335-1346. doi: 10.2217/fon.11.111.
  • Riballo E, Kuhne M, Rief N, Doherty A, Smith GC, Recio MJ, et al. A pathway of double-strand break rejoining dependent upon ATM, Artemis, and proteins locating to gamma-H2AX foci. Mol Cell 2004; 16(5): 715-724. doi: 10.1016/j.molcel.2004.10.029.
  • Lee JH, Paull TT. Activation and regulation of ATM kinase activity in response to DNA double-strand breaks. Oncogene 2007; 26(56): 7741-7748. doi: 10.1038/sj.onc.1210872.
  • Vecchio D, Daga A, Carra E, Marubbi D, Baio G, Neumaier CE, et al. Predictability, efficacy and safety of radiosensitization of glioblastoma-initiating cells by the ATM inhibitor KU-60019. Int J Cancer 2014; 135(2): 479-491. doi: 10.1002/ijc.28680.
  • Seol HJ, Yoo HY, Jin J, Joo KM, Kong DS, Yoon SJ, et al. Prognostic implications of the DNA damage response pathway in glioblastoma. Oncol Rep 2011; 26(2): 423-430. doi: 10.3892/or.2011.1325.
  • Hashimoto T, Urushihara Y, Murata Y, Fujishima Y, Hosoi Y. AMPK increases expression of ATM through transcriptional factor Sp1 and induces radioresistance under severe hypoxia in glioblastoma cell lines. Biochem Biophys Res Commun 2022; 590: 82-88. doi: 10.1016/j.bbrc.2021.12.076.
  • Short SC, Giampieri S, Worku M, Alcaide-German M, Sioftanos G, Bourne S, et al. Rad51 inhibition is an effective means of targeting DNA repair in glioma models and CD133+ tumor-derived cells. Neuro Oncol 2011; 13(5): 487-499. doi: 10.1093/neuonc/nor010.
  • Lees-Miller SP, Godbout R, Chan DW, Weinfeld M, Day RS 3rd, Barron GM, et al. Absence of p350 subunit of DNA-activated protein kinase from a radiosensitive human cell line. Science 1995; 267(5201): 1183-1185. doi: 10.1126/science.7855602.
  • Li L, Lei Q, Zhang S, Kong L, Qin B. Screening and identification of key biomarkers in hepatocellular carcinoma: Evidence from bioinformatic analysis. Oncol Rep 2017; 38(5): 2607-2618. doi: 10.3892/or.2017.5946.
  • Yan P, He Y, Xie K, Kong S, Zhao W. In silico analyses for potential key genes associated with gastric cancer. PeerJ 2018; 6:e6092. doi: 10.7717/peerj.6092.
  • Zhang Y, Li Y, Chachad D, Liu B, Godavarthi JD, Williams-Villalobo A, et al. In silico analysis of DND1 and its co-expressed genes in human cancers. Biochem Biophys Rep 2022; 29: 101206. doi: 10.1016/j.bbrep.2022.101206.
  • Fabregat A, Sidiropoulos K, Garapati P, Gillespie M, Hausmann K, Haw R, et al. The Reactome pathway Knowledgebase. Nucleic Acids Res 2016; 44(D1): D481-D487. doi: 10.1093/nar/gkv1351.
  • Kanehisa M, Goto S. KEGG: kyoto encyclopedia of genes and genomes. Nucleic Acids Res 2000; 28(1): 27-30. doi: 10.1093/nar/28.1.27.
  • Heberle H, Meirelles GV, da Silva FR, Telles GP, Minghim R. InteractiVenn: a web-based tool for the analysis of sets through Venn diagrams. Bmc Bioinformatics 2015; 16: 169. doi: 10.1186/s12859-015-0611-3.
  • Demšar J, Curk T, Erjavec A, Gorup Č, Hočevar T, Milutinovič M, et al. Orange: data mining toolbox in Python. the Journal of machine Learning research 2013; 14(1): 2349-2353.
  • Bredel M, Bredel C, Juric D, Harsh GR, Vogel H, Recht LD, et al. Functional network analysis reveals extended gliomagenesis pathway maps and three novel MYC-interacting genes in human gliomas. Cancer Res 2005; 65(19): 8679-8689. doi: 10.1158/0008-5472.CAN-05-1204.
  • Khatua S, Peterson KM, Brown KM, Lawlor C, Santi MR, LaFleur B. et al. Overexpression of the EGFR/FKBP12/HIF-2alpha pathway identified in childhood astrocytomas by angiogenesis gene profiling. Cancer Res 2003; 63(8): 1865-1870.
  • Gao J, Aksoy BA, Dogrusoz U, Dresdner G, Gross B, Sumer SO, et al. Integrative analysis of complex cancer genomics and clinical profiles using the cBioPortal. Sci Signal 2013; 6(269): pl1. doi: 10.1126/scisignal.2004088.
  • Cerami E, Gao J, Dogrusoz U, Gross BE, Sumer SO, Aksoy BA, et al. The cBio cancer genomics portal: an open platform for exploring multidimensional cancer genomics data. Cancer Discov 2012; 2(5): 401-404. doi: 10.1158/2159-8290.CD-12-0095.
  • Liu J, Lichtenberg T, Hoadley KA, Poisson LM, Lazar AJ, Cherniack AD, et al. An Integrated TCGA Pan-Cancer Clinical Data Resource to Drive High-Quality Survival Outcome Analytics. Cell 2018; 173(2): 400-16 e11. doi: 10.1016/j.cell.2018.02.052.
  • Cancer Genome Atlas Research N, Weinstein JN, Collisson EA, Mills GB, Shaw KR, Ozenberger BA, et al. The Cancer Genome Atlas Pan-Cancer analysis project. Nat Genet 2013; 45(10): 1113-1120. doi: 10.1038/ng.2764.
  • Hoadley KA, Yau C, Hinoue T, Wolf DM, Lazar AJ, Drill E, et al. Cell-of-Origin Patterns Dominate the Molecular Classification of 10,000 Tumors from 33 Types of Cancer. Cell 2018;173(2):291-304. doi: 10.1016/j.cell.2018.03.022.
  • Mermel CH, Schumacher SE, Hill B, Meyerson ML, Beroukhim R, Getz G. GISTIC2.0 facilitates sensitive and confident localization of the targets of focal somatic copy-number alteration in human cancers. Genome Biol 2011; 12(4): R41. doi: 10.1186/gb-2011-12-4-r41.
  • Tang Z, Kang B, Li C, Chen T, Zhang Z. GEPIA2: an enhanced web server for large-scale expression profiling and interactive analysis. Nucleic Acids Res 2019; 47(W1): W556-W60. doi: 10.1093/nar/gkz430.
  • Wang M, Chen S, Ao D. Targeting DNA repair pathway in cancer: Mechanisms and clinical application. MedComm 2021; 2(4); 654-691. doi: 10.1002/mco2.103.
  • Anindya R, Mari PO, Kristensen U, Kool H, Giglia-Mari G, Mullenders LH, et al. A ubiquitin-binding domain in Cockayne syndrome B required for transcription-coupled nucleotide excision repair. Mol Cell 2010; 38(5): 637-648. doi: 10.1016/j.molcel.2010.04.017.
  • Dabholkar MD, Berger MS, Vionnet JA, Overton L, Thompson C, Bostick-Bruton F, et al. Comparative analyses of relative ERCC3 and ERCC6 mRNA levels in gliomas and adjacent non-neoplastic brain. Mol Carcinog 1996; 17(1): 1-7. doi: 10.1002/(SICI)1098-2744(199609)17:1<1::AID-MC1>3.0.CO;2-M.
  • Grunda JM, Fiveash J, Palmer CA, Cantor A, Fathallah-Shaykh HM, Nabors LB, et al. Rationally designed pharmacogenomic treatment using concurrent capecitabine and radiotherapy for glioblastoma; gene expression profiles associated with outcome. Clin Cancer Res 2010; 16(10): 2890-2898. doi: 10.1158/1078-0432.CCR-09-3151.
  • Pascal JM. The comings and goings of PARP-1 in response to DNA damage. DNA repair 2018; 71: 177-182. doi: 10.1016/j.dnarep.2018.08.022.
  • Alemasova EE, Lavrik OI. Poly(ADP-ribosyl)ation by PARP1: reaction mechanism and regulatory proteins. Nucleic Acids Res 2019; 47(8): 3811-3827. doi: 10.1093/nar/gkz120.
  • Galia A, Calogero AE, Condorelli R, Fraggetta F, La Corte A, Ridolfo F, et al. PARP-1 protein expression in glioblastoma multiforme. Eur J Histochem 2012; 56(1): e9. doi: 10.4081/ejh.2012.e9.
  • Ghorai A, Mahaddalkar T, Thorat R, Dutt S. Sustained inhibition of PARP-1 activity delays glioblastoma recurrence by enhancing radiation-induced senescence. Cancer Lett 2020; 490: 44-53. doi: 10.1016/j.canlet.2020.06.023.
  • Pearl LH. Structure and function in the uracil-DNA glycosylase superfamily. Mutat Res 2000; 460(3-4): 165-181. doi: 10.1016/s0921-8777(00)00025-2.
  • Moon YW, Park WS, Vortmeyer AO, Weil RJ, Lee YS, Winters TA, et al. Mutation of the uracil DNA glycosylase gene detected in glioblastoma, Mutat Res 1998; 421(2): 191-196. doi: 10.1016/s0027-5107(98)00165-1.
  • Kase M, Vardja M, Lipping A, Asser T, Jaal J. Impact of PARP-1 and DNA-PK expression on survival in patients with glioblastoma multiforme. Radiother Oncol 2011; 101(1): 127-31. doi: 10.1016/j.radonc.2011.06.024.
  • Murnyák B, Kouhsari MC, Hershkovitch R, Kálmán B, Marko-Varga G, Klekner Á, et al. PARP1 expression and its correlation with survival is tumour molecular subtype dependent in glioblastoma. Oncotarget 2017; 8(28): 46348-46362. doi: 10.18632/oncotarget.18013.
  • Deshmukh AL, Porro A, Mohiuddin M, Lanni S, Panigrahi GB, Caron MC, et al. FAN1, a DNA Repair Nuclease, as a Modifier of Repeat Expansion Disorders. J Huntingtons Dis 2021; 10(1): 95-122. doi: 10.3233/JHD-200448.
  • Goold R, Hamilton J, Menneteau T, Flower M, Bunting EL, Aldous SG, et al. FAN1 controls mismatch repair complex assembly via MLH1 retention to stabilize CAG repeat expansion in Huntington's disease. Cell Rep 2021; 36(9): 109649. doi: 10.1016/j.celrep.2021.109649.
  • Wang Z, Chen N, Yang J, Wang Q, Li A. Microarray gene profiling analysis of glioblastoma cell line U87 reveals suppression of the FANCD2/Fanconi anemia pathway by the combination of Y15 and temozolomide. Arch Med Sci 2019; 15(4): 1035-1046. doi: 10.5114/aoms.2019.86063.
  • Bellacosa A, Cicchillitti L, Schepis F, Riccio A, Yeung AT, Matsumoto Y, et al. MED1, a novel human methyl-CpG-binding endonuclease, interacts with DNA mismatch repair protein MLH1. Proc Natl Acad Sci U S A 1999; 96(7): 3969-3974. doi: 10.1073/pnas.96.7.3969.
  • Schlegel J, Guneysu S, Mennel HD. Expression of the genes of methyl-binding domain proteins in human gliomas. Oncol Rep 2002; 9(2): 393-395.
  • Sasaki H, Zlatescu MC, Betensky RA, Ino Y, Cairncross JG, Louis DN. PTEN is a target of chromosome 10q loss in anaplastic oligodendrogliomas and PTEN alterations are associated with poor prognosis. Am J Pathol 2001; 159(1): 359-67. doi: 10.1016/S0002-9440(10)61702-6.
  • Jaeckle K, Zhang L, Lee P, Connor T, Ali-Osman F. ERCC6 mutations in human malignant astrocytomas. Neuro-oncology 1999; 1: 317.
  • Rodriguez-Hernandez I, Perdomo S, Santos-Briz A, Garcia JL, Gomez-Moreta JA, Cruz JJ, et al. Analysis of DNA repair gene polymorphisms in glioblastoma. Gene 2014; 536(1): 79-83. doi: 10.1016/j.gene.2013.11.077.

Identification of Genes Related to DNA Repair Mechanism in Glioblastoma by Bioinformatics Methods

Year 2022, Volume: 8 Issue: 2, 117 - 124, 31.05.2022
https://doi.org/10.30934/kusbed.1003777

Abstract

Objective: Aberrant expression of genes involved in DNA repair mechanisms (DRM) have been associated with radiation sensitivity of glioblastoma (GBM) cells. Identification of genes in DRM through bioinformatics methods may help identify potential novel therapeutic targets that can be used in GBM treatment. This study aims to identify genes that play a role in DRM in GBM using bioinformatics methods.
Methods: Genes associated with DRM were identified using the “Reactome” and “KEGG” databases. The mRNA expression profiles of DRM related genes were analyzed in the GEO GDS1813 and GDS2853 datasets including GBM tumor samples using the "Orange Canvas" software. Genetic changes of genes were identified in GBM TCGA cases using the cBioPortal database. The GEPIA2 was used to show the effect of altered expression profiles of these genes on patient survival.
Results: The mRNA expression profiles of ERCC6, FAN1, MBD4, PARP1 and UNG genes were found to be altered in GBM tumors. Mutations and copy number alterations for the identified genes were observed in TCGA GBM cases. The overall survival and disease-free survival of TCGA GBM patients were not significantly different between high and low expression groups.
Conclusion: ERCC6, PARP1 and UNG genes identified in the current study may be potential therapeutic targets that can increase the efficacy of radiotherapy in GBM in case of their suppression.

References

  • Louis DN, Perry A, Reifenberger G, von Deimling A, Figarella-Branger D, Cavenee WK, et al. The 2016 World Health Organization Classification of Tumors of the Central Nervous System: a summary. Acta Neuropathol 2016; 131(6):803-820. doi: 10.1007/s00401-016-1545-1.
  • Stupp R, Hegi ME, Mason WP, van den Bent MJ, Taphoorn MJ, Janzer RC, et al. Effects of radiotherapy with concomitant and adjuvant temozolomide versus radiotherapy alone on survival in glioblastoma in a randomised phase III study: 5-year analysis of the EORTC-NCIC trial. Lancet Oncol 2009; 10(5):459-466. doi: 10.1016/S1470-2045(09)70025-7.
  • Henson JW. Treatment of glioblastoma multiforme: a new standard. Arch Neurol. 2006; 63(3): 337-341. doi: 10.1001/archneur.63.3.337.
  • Stupp R, Mason WP, van den Bent MJ, Weller M, Fisher B, Taphoorn MJ, et al. Radiotherapy plus concomitant and adjuvant temozolomide for glioblastoma. N Engl J Med 2005; 352(10): 987-996. doi: 10.1056/NEJMoa043330.
  • Palanichamy K, Chakravarti A. Combining drugs and radiotherapy: from the bench to the bedside. Curr Opin Neurol 2009; 22(6): 625-632. doi: 10.1097/WCO.0b013e3283327d33.
  • Erasimus H, Gobin M, Niclou S, Van Dyck E. DNA repair mechanisms and their clinical impact in glioblastoma. Mutat Res Rev Mutat Res 2016; 769: 19-35. doi: 10.1016/j.mrrev.2016.05.005.
  • Meng W, Palmer JD, Siedow M, Haque SJ, Chakravarti A. Overcoming Radiation Resistance in Gliomas by Targeting Metabolism and DNA Repair Pathways. Int. J. Mol. Sci 2022; 23(4): 2246. doi: 10.3390/ijms23042246.
  • Biau J, Chautard E, Verrelle P, Dutreix M. Altering DNA Repair to Improve Radiation Therapy: Specific and Multiple Pathway Targeting. Front Oncol 2019; 9: 1009. doi: 10.3389/fonc.2019.01009.
  • Kesari S, Advani SJ, Lawson JD, Kahle KT, Ng K, Carter B, et al. DNA damage response and repair: insights into strategies for radiation sensitization of gliomas. Future Oncol 2011; 7(11): 1335-1346. doi: 10.2217/fon.11.111.
  • Riballo E, Kuhne M, Rief N, Doherty A, Smith GC, Recio MJ, et al. A pathway of double-strand break rejoining dependent upon ATM, Artemis, and proteins locating to gamma-H2AX foci. Mol Cell 2004; 16(5): 715-724. doi: 10.1016/j.molcel.2004.10.029.
  • Lee JH, Paull TT. Activation and regulation of ATM kinase activity in response to DNA double-strand breaks. Oncogene 2007; 26(56): 7741-7748. doi: 10.1038/sj.onc.1210872.
  • Vecchio D, Daga A, Carra E, Marubbi D, Baio G, Neumaier CE, et al. Predictability, efficacy and safety of radiosensitization of glioblastoma-initiating cells by the ATM inhibitor KU-60019. Int J Cancer 2014; 135(2): 479-491. doi: 10.1002/ijc.28680.
  • Seol HJ, Yoo HY, Jin J, Joo KM, Kong DS, Yoon SJ, et al. Prognostic implications of the DNA damage response pathway in glioblastoma. Oncol Rep 2011; 26(2): 423-430. doi: 10.3892/or.2011.1325.
  • Hashimoto T, Urushihara Y, Murata Y, Fujishima Y, Hosoi Y. AMPK increases expression of ATM through transcriptional factor Sp1 and induces radioresistance under severe hypoxia in glioblastoma cell lines. Biochem Biophys Res Commun 2022; 590: 82-88. doi: 10.1016/j.bbrc.2021.12.076.
  • Short SC, Giampieri S, Worku M, Alcaide-German M, Sioftanos G, Bourne S, et al. Rad51 inhibition is an effective means of targeting DNA repair in glioma models and CD133+ tumor-derived cells. Neuro Oncol 2011; 13(5): 487-499. doi: 10.1093/neuonc/nor010.
  • Lees-Miller SP, Godbout R, Chan DW, Weinfeld M, Day RS 3rd, Barron GM, et al. Absence of p350 subunit of DNA-activated protein kinase from a radiosensitive human cell line. Science 1995; 267(5201): 1183-1185. doi: 10.1126/science.7855602.
  • Li L, Lei Q, Zhang S, Kong L, Qin B. Screening and identification of key biomarkers in hepatocellular carcinoma: Evidence from bioinformatic analysis. Oncol Rep 2017; 38(5): 2607-2618. doi: 10.3892/or.2017.5946.
  • Yan P, He Y, Xie K, Kong S, Zhao W. In silico analyses for potential key genes associated with gastric cancer. PeerJ 2018; 6:e6092. doi: 10.7717/peerj.6092.
  • Zhang Y, Li Y, Chachad D, Liu B, Godavarthi JD, Williams-Villalobo A, et al. In silico analysis of DND1 and its co-expressed genes in human cancers. Biochem Biophys Rep 2022; 29: 101206. doi: 10.1016/j.bbrep.2022.101206.
  • Fabregat A, Sidiropoulos K, Garapati P, Gillespie M, Hausmann K, Haw R, et al. The Reactome pathway Knowledgebase. Nucleic Acids Res 2016; 44(D1): D481-D487. doi: 10.1093/nar/gkv1351.
  • Kanehisa M, Goto S. KEGG: kyoto encyclopedia of genes and genomes. Nucleic Acids Res 2000; 28(1): 27-30. doi: 10.1093/nar/28.1.27.
  • Heberle H, Meirelles GV, da Silva FR, Telles GP, Minghim R. InteractiVenn: a web-based tool for the analysis of sets through Venn diagrams. Bmc Bioinformatics 2015; 16: 169. doi: 10.1186/s12859-015-0611-3.
  • Demšar J, Curk T, Erjavec A, Gorup Č, Hočevar T, Milutinovič M, et al. Orange: data mining toolbox in Python. the Journal of machine Learning research 2013; 14(1): 2349-2353.
  • Bredel M, Bredel C, Juric D, Harsh GR, Vogel H, Recht LD, et al. Functional network analysis reveals extended gliomagenesis pathway maps and three novel MYC-interacting genes in human gliomas. Cancer Res 2005; 65(19): 8679-8689. doi: 10.1158/0008-5472.CAN-05-1204.
  • Khatua S, Peterson KM, Brown KM, Lawlor C, Santi MR, LaFleur B. et al. Overexpression of the EGFR/FKBP12/HIF-2alpha pathway identified in childhood astrocytomas by angiogenesis gene profiling. Cancer Res 2003; 63(8): 1865-1870.
  • Gao J, Aksoy BA, Dogrusoz U, Dresdner G, Gross B, Sumer SO, et al. Integrative analysis of complex cancer genomics and clinical profiles using the cBioPortal. Sci Signal 2013; 6(269): pl1. doi: 10.1126/scisignal.2004088.
  • Cerami E, Gao J, Dogrusoz U, Gross BE, Sumer SO, Aksoy BA, et al. The cBio cancer genomics portal: an open platform for exploring multidimensional cancer genomics data. Cancer Discov 2012; 2(5): 401-404. doi: 10.1158/2159-8290.CD-12-0095.
  • Liu J, Lichtenberg T, Hoadley KA, Poisson LM, Lazar AJ, Cherniack AD, et al. An Integrated TCGA Pan-Cancer Clinical Data Resource to Drive High-Quality Survival Outcome Analytics. Cell 2018; 173(2): 400-16 e11. doi: 10.1016/j.cell.2018.02.052.
  • Cancer Genome Atlas Research N, Weinstein JN, Collisson EA, Mills GB, Shaw KR, Ozenberger BA, et al. The Cancer Genome Atlas Pan-Cancer analysis project. Nat Genet 2013; 45(10): 1113-1120. doi: 10.1038/ng.2764.
  • Hoadley KA, Yau C, Hinoue T, Wolf DM, Lazar AJ, Drill E, et al. Cell-of-Origin Patterns Dominate the Molecular Classification of 10,000 Tumors from 33 Types of Cancer. Cell 2018;173(2):291-304. doi: 10.1016/j.cell.2018.03.022.
  • Mermel CH, Schumacher SE, Hill B, Meyerson ML, Beroukhim R, Getz G. GISTIC2.0 facilitates sensitive and confident localization of the targets of focal somatic copy-number alteration in human cancers. Genome Biol 2011; 12(4): R41. doi: 10.1186/gb-2011-12-4-r41.
  • Tang Z, Kang B, Li C, Chen T, Zhang Z. GEPIA2: an enhanced web server for large-scale expression profiling and interactive analysis. Nucleic Acids Res 2019; 47(W1): W556-W60. doi: 10.1093/nar/gkz430.
  • Wang M, Chen S, Ao D. Targeting DNA repair pathway in cancer: Mechanisms and clinical application. MedComm 2021; 2(4); 654-691. doi: 10.1002/mco2.103.
  • Anindya R, Mari PO, Kristensen U, Kool H, Giglia-Mari G, Mullenders LH, et al. A ubiquitin-binding domain in Cockayne syndrome B required for transcription-coupled nucleotide excision repair. Mol Cell 2010; 38(5): 637-648. doi: 10.1016/j.molcel.2010.04.017.
  • Dabholkar MD, Berger MS, Vionnet JA, Overton L, Thompson C, Bostick-Bruton F, et al. Comparative analyses of relative ERCC3 and ERCC6 mRNA levels in gliomas and adjacent non-neoplastic brain. Mol Carcinog 1996; 17(1): 1-7. doi: 10.1002/(SICI)1098-2744(199609)17:1<1::AID-MC1>3.0.CO;2-M.
  • Grunda JM, Fiveash J, Palmer CA, Cantor A, Fathallah-Shaykh HM, Nabors LB, et al. Rationally designed pharmacogenomic treatment using concurrent capecitabine and radiotherapy for glioblastoma; gene expression profiles associated with outcome. Clin Cancer Res 2010; 16(10): 2890-2898. doi: 10.1158/1078-0432.CCR-09-3151.
  • Pascal JM. The comings and goings of PARP-1 in response to DNA damage. DNA repair 2018; 71: 177-182. doi: 10.1016/j.dnarep.2018.08.022.
  • Alemasova EE, Lavrik OI. Poly(ADP-ribosyl)ation by PARP1: reaction mechanism and regulatory proteins. Nucleic Acids Res 2019; 47(8): 3811-3827. doi: 10.1093/nar/gkz120.
  • Galia A, Calogero AE, Condorelli R, Fraggetta F, La Corte A, Ridolfo F, et al. PARP-1 protein expression in glioblastoma multiforme. Eur J Histochem 2012; 56(1): e9. doi: 10.4081/ejh.2012.e9.
  • Ghorai A, Mahaddalkar T, Thorat R, Dutt S. Sustained inhibition of PARP-1 activity delays glioblastoma recurrence by enhancing radiation-induced senescence. Cancer Lett 2020; 490: 44-53. doi: 10.1016/j.canlet.2020.06.023.
  • Pearl LH. Structure and function in the uracil-DNA glycosylase superfamily. Mutat Res 2000; 460(3-4): 165-181. doi: 10.1016/s0921-8777(00)00025-2.
  • Moon YW, Park WS, Vortmeyer AO, Weil RJ, Lee YS, Winters TA, et al. Mutation of the uracil DNA glycosylase gene detected in glioblastoma, Mutat Res 1998; 421(2): 191-196. doi: 10.1016/s0027-5107(98)00165-1.
  • Kase M, Vardja M, Lipping A, Asser T, Jaal J. Impact of PARP-1 and DNA-PK expression on survival in patients with glioblastoma multiforme. Radiother Oncol 2011; 101(1): 127-31. doi: 10.1016/j.radonc.2011.06.024.
  • Murnyák B, Kouhsari MC, Hershkovitch R, Kálmán B, Marko-Varga G, Klekner Á, et al. PARP1 expression and its correlation with survival is tumour molecular subtype dependent in glioblastoma. Oncotarget 2017; 8(28): 46348-46362. doi: 10.18632/oncotarget.18013.
  • Deshmukh AL, Porro A, Mohiuddin M, Lanni S, Panigrahi GB, Caron MC, et al. FAN1, a DNA Repair Nuclease, as a Modifier of Repeat Expansion Disorders. J Huntingtons Dis 2021; 10(1): 95-122. doi: 10.3233/JHD-200448.
  • Goold R, Hamilton J, Menneteau T, Flower M, Bunting EL, Aldous SG, et al. FAN1 controls mismatch repair complex assembly via MLH1 retention to stabilize CAG repeat expansion in Huntington's disease. Cell Rep 2021; 36(9): 109649. doi: 10.1016/j.celrep.2021.109649.
  • Wang Z, Chen N, Yang J, Wang Q, Li A. Microarray gene profiling analysis of glioblastoma cell line U87 reveals suppression of the FANCD2/Fanconi anemia pathway by the combination of Y15 and temozolomide. Arch Med Sci 2019; 15(4): 1035-1046. doi: 10.5114/aoms.2019.86063.
  • Bellacosa A, Cicchillitti L, Schepis F, Riccio A, Yeung AT, Matsumoto Y, et al. MED1, a novel human methyl-CpG-binding endonuclease, interacts with DNA mismatch repair protein MLH1. Proc Natl Acad Sci U S A 1999; 96(7): 3969-3974. doi: 10.1073/pnas.96.7.3969.
  • Schlegel J, Guneysu S, Mennel HD. Expression of the genes of methyl-binding domain proteins in human gliomas. Oncol Rep 2002; 9(2): 393-395.
  • Sasaki H, Zlatescu MC, Betensky RA, Ino Y, Cairncross JG, Louis DN. PTEN is a target of chromosome 10q loss in anaplastic oligodendrogliomas and PTEN alterations are associated with poor prognosis. Am J Pathol 2001; 159(1): 359-67. doi: 10.1016/S0002-9440(10)61702-6.
  • Jaeckle K, Zhang L, Lee P, Connor T, Ali-Osman F. ERCC6 mutations in human malignant astrocytomas. Neuro-oncology 1999; 1: 317.
  • Rodriguez-Hernandez I, Perdomo S, Santos-Briz A, Garcia JL, Gomez-Moreta JA, Cruz JJ, et al. Analysis of DNA repair gene polymorphisms in glioblastoma. Gene 2014; 536(1): 79-83. doi: 10.1016/j.gene.2013.11.077.
There are 52 citations in total.

Details

Primary Language Turkish
Subjects Biochemistry and Cell Biology (Other)
Journal Section Original Article / Medical Sciences
Authors

Hasan Onur Çağlar 0000-0002-3637-4755

Publication Date May 31, 2022
Submission Date October 2, 2021
Acceptance Date May 6, 2022
Published in Issue Year 2022 Volume: 8 Issue: 2

Cite

APA Çağlar, H. O. (2022). DNA Tamir Mekanizması İlişkili Genlerin Biyoinformatik Yöntemlerle Glioblastomda Tanımlanması. Kocaeli Üniversitesi Sağlık Bilimleri Dergisi, 8(2), 117-124. https://doi.org/10.30934/kusbed.1003777
AMA Çağlar HO. DNA Tamir Mekanizması İlişkili Genlerin Biyoinformatik Yöntemlerle Glioblastomda Tanımlanması. KOU Sag Bil Derg. May 2022;8(2):117-124. doi:10.30934/kusbed.1003777
Chicago Çağlar, Hasan Onur. “DNA Tamir Mekanizması İlişkili Genlerin Biyoinformatik Yöntemlerle Glioblastomda Tanımlanması”. Kocaeli Üniversitesi Sağlık Bilimleri Dergisi 8, no. 2 (May 2022): 117-24. https://doi.org/10.30934/kusbed.1003777.
EndNote Çağlar HO (May 1, 2022) DNA Tamir Mekanizması İlişkili Genlerin Biyoinformatik Yöntemlerle Glioblastomda Tanımlanması. Kocaeli Üniversitesi Sağlık Bilimleri Dergisi 8 2 117–124.
IEEE H. O. Çağlar, “DNA Tamir Mekanizması İlişkili Genlerin Biyoinformatik Yöntemlerle Glioblastomda Tanımlanması”, KOU Sag Bil Derg, vol. 8, no. 2, pp. 117–124, 2022, doi: 10.30934/kusbed.1003777.
ISNAD Çağlar, Hasan Onur. “DNA Tamir Mekanizması İlişkili Genlerin Biyoinformatik Yöntemlerle Glioblastomda Tanımlanması”. Kocaeli Üniversitesi Sağlık Bilimleri Dergisi 8/2 (May 2022), 117-124. https://doi.org/10.30934/kusbed.1003777.
JAMA Çağlar HO. DNA Tamir Mekanizması İlişkili Genlerin Biyoinformatik Yöntemlerle Glioblastomda Tanımlanması. KOU Sag Bil Derg. 2022;8:117–124.
MLA Çağlar, Hasan Onur. “DNA Tamir Mekanizması İlişkili Genlerin Biyoinformatik Yöntemlerle Glioblastomda Tanımlanması”. Kocaeli Üniversitesi Sağlık Bilimleri Dergisi, vol. 8, no. 2, 2022, pp. 117-24, doi:10.30934/kusbed.1003777.
Vancouver Çağlar HO. DNA Tamir Mekanizması İlişkili Genlerin Biyoinformatik Yöntemlerle Glioblastomda Tanımlanması. KOU Sag Bil Derg. 2022;8(2):117-24.