AI Chat Paper
Note: Please note that the following content is generated by AMiner AI. SciOpen does not take any responsibility related to this content.
{{lang === 'zh_CN' ? '文章概述' : 'Summary'}}
{{lang === 'en_US' ? '中' : 'Eng'}}
Chat more with AI
PDF (24.6 MB)
Collect
Submit Manuscript AI Chat Paper
Show Outline
Outline
Show full outline
Hide outline
Outline
Show full outline
Hide outline
Original Article | Open Access

Omics-based integrated analysis identified ATRX as a biomarker associated with glioma diagnosis and prognosis

Yingbin Xie1,2,*Yanli Tan3,4,*Chao Yang5Xuehao Zhang2Can Xu2Xiaoxia Qiao1Jianglong Xu1Shaohui Tian1Chuan Fang1( )Chunsheng Kang5,6( )
Department of Neurosurgery, Affiliated Hospital of Hebei University, Baoding 071000, China
Department of Neurosurgery, Hebei University Medical College, Baoding 071000, China
Department of Pathology, Hebei University Medical College, Baoding 071000, China
Department of Pathology, Affiliated Hospital of Hebei University, Baoding 071000, China
Lab of Neuro-oncology, Tianjin Neurological Institute, Key Laboratory of Post-Neuroinjury Neuro-repair and Regeneration in Central Nervous System, Tianjin Medical University General Hospital, Tianjin 300052, China
Department of Oncology, Affiliated Cancer Hospital & Institute of Guangzhou Medical University, Guangzhou 510095, China

*These authors contributed equally to this work.

Show Author Information

Abstract

Objective

ATRX is a multifunctional protein that is tightly regulated by and implicated in transcriptional regulation and chromatin remodeling. Numerous studies have shown that genetic alterations in ATRX play a significant role in gliomas. This study aims to further determine the relationship between ATRX and glioma prognosis and identify possible mechanisms for exploring the biological significance of ATRX using large data sets.

Methods

We used The Cancer Genome Atlas (TCGA) database and 130 immunohistochemical results to confirm the difference in ATRX mutations in high- and low-grade gliomas. An online analysis of the TCGA glioma datasets using the cBioPortal platform was performed to study the relationship between ATRX mutations and IDH1, TP53, CDKN2A and CDKN2B mutations in the corresponding TCGA glioma dataset. In combination with clinical pathology data, the biological significance of the relationships were analyzed. Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analyses and annotations of all adjacent genes in the network were performedin the Database for Annotation, Visualization and Integrated Discovery (DAVID) and R language. A protein-protein interaction (PPI) network was constructed, and the interactions of all adjacent nodes were analyzed by the String database and using Cytoscape software.

Results

In the selected TCGA glioma datasets, a total of 2,228 patients were queried, 21% of whom had ATRX alterations, which co-occurred frequently with TP53 and IDH1 mutations. ATRX alterations are associated with multiple critical molecular events, which results in a significantly improved overall survival (OS) rate. In low-grade gliomas, ATRX mutations are significantly associated with multiple important molecular events, such as ZNF274 and FDXR at mRNA and protein levels. A functional cluster analysis revealed that these genes played a role in chromatin binding and P53, and a link was observed between ATRX and IDH1 and TP53 in the interaction network. ATRX and TP53 are important nodes in the network and have potential links with the blood oxygen imbalance.

Conclusions

ATRX mutations have clinical implications for the molecular diagnosis of gliomas and can provide diagnostic and prognostic information for gliomas. ATRX is expected to serve as a new therapeutic target.

References

1

Suzuki H, Aoki K, Chiba K, Sato Y, Shiozawa Y, Shiraishi Y, et al. Mutational landscape and clonal architecture in grade Ⅱ and Ⅲ gliomas. Nat Genet. 2015; 47: 458-68.

2

Castro-Gamero AM, Pezuk JA, Brassesco MS, Tone LG. G2/M inhibitors as pharmacotherapeutic opportunities for glioblastoma: the old, the new, and the future. Cancer Biol Med. 2018; 15: 354-74.

3

Han L, Liu CY, Qi HZ, Zhou JH, Wen J, Wu D, et al. Systemic delivery of monoclonal antibodies to the central nervous system for brain tumor therapy. Adv Mater. 2019; 31: e1805697.

4

Cai HQ, Wang PF, Zhang HP, Cheng ZJ, Li SW, He J, et al. Phosphorylated Hsp27 is mutually exclusive with ATRX loss and the IDH1R132H mutation and may predict better prognosis among glioblastomas without the IDH1 mutation and ATRX loss. J Clin Pathol. 2018; 71: 702-7.

5

Synhaeve NE, Van Den Bent MJ, French PJ, Dinjens WNM, Atmodimedjo PN, Kros JM, et al. Clinical evaluation of a dedicated next generation sequencing panel for routine glioma diagnostics. Acta Neuropathol Commun. 2018; 6: 126.

6

Godek KM, Venere M, Wu Q, Mills KD, Hickey WF, Rich JN, et al. Chromosomal instability affects the tumorigenicity of glioblastoma tumor-initiating cells. Cancer Discov. 2016; 6: 532-45.

7

Stanislaw C, Xue Y, Wilcox WR. Genetic evaluation and testing for hereditary forms of cancer in the era of next-generation sequencing. Cancer Biol Med. 2016; 13: 55-67.

8

Nandakumar P, Mansouri A, Das S. The role of ATRX in glioma biology. Front Oncol. 2017; 7: 236.

9

Pekmezci M, Rice T, Molinaro AM, Walsh KM, Decker PA, Hansen H, et al. Adult infiltrating gliomas with WHO 2016 integrated diagnosis: additional prognostic roles of ATRX and TERT. Acta Neuropathol. 2017; 133: 1001-16.

10

Van Den Bent MJ, Weller M, Wen PY, Kros JM, Aldape K, Chang SS. A clinical perspective on the 2016 WHO brain tumor classification and routine molecular diagnostics. Neuro Oncol. 2017; 19: 614-24.

11

Núñez FJ, Mendez FM, Kadiyala P, Alghamri MS, Savelieff MG, Garcia-Fabiani MB, et al. IDH1-R132H acts as a tumor suppressor in glioma via epigenetic up-regulation of the DNA damage response. Sci Transl Med. 2019; 11: eaaq1427.

12

Danussi C, Bose P, Parthasarathy PT, Silberman PC, Van Arnam JS, Vitucci M, et al. Atrx inactivation drives disease-defining phenotypes in glioma cells of origin through global epigenomic remodeling. Nat Commun. 2018; 9: 1057.

13

Nguyen DT, Voon HPJ, Xella B, Scott C, Clynes D, Babbs C, et al. The chromatin remodelling factor ATRX suppresses R-loops in transcribed telomeric repeats. EMBO Rep. 2017; 18: 914-28.

14

Mukherjee J, Johannessen TC, Ohba S, Chow TT, Jones L, Pandita A, et al. Mutant IDH1 cooperates with ATRX loss to drive the alternative lengthening of telomere phenotype in glioma. Cancer Res. 2018; 78: 2966-77.

15

Koschmann C, Lowenstein PR, Castro MG. ATRX mutations and glioblastoma: impaired DNA damage repair, alternative lengthening of telomeres, and genetic instability. Mol Cell Oncol. 2016; 3: e1167158.

16

Haase S, Garcia-Fabiani MB, Carney S, Altshuler D, Núñez FJ, Méndez FM, et al. Mutant ATRX: uncovering a new therapeutic target for glioma. Expert Opin Ther Targets. 2018; 22: 599-613.

17

Cai JQ, Zhang CB, Zhang W, Wang GZ, Yao K, Wang ZL, et al. ATRX, IDH1-R132H and Ki-67 immunohistochemistry as a classification scheme for astrocytic tumors. Oncoscience. 2016; 3: 258-65.

18

Modrek AS, Golub D, Khan T, Bready D, Prado J, Bowman C, et al. Low-grade astrocytoma mutations in IDH1, P53, and ATRX cooperate to block differentiation of human neural stem cells via repression of SOX2. Cell Rep. 2017; 21: 1267-80.

19

Reinhardt A, Stichel D, Schrimpf D, Sahm F, Korshunov A, Reuss DE, et al. Anaplastic astrocytoma with piloid features, a novel molecular class of IDH wildtype glioma with recurrent MAPK pathway, CDKN2A/B and ATRX alterations. Acta Neuropathol. 2018; 136: 273-91.

20

Reis GF, Pekmezci M, Hansen HM, Rice T, Marshall RE, Molinaro AM, et al. CDKN2A loss is associated with shortened overall survival in lower-grade (World Health Organization Grades Ⅱ-Ⅲ) astrocytomas. J Neuropathol Exp Neurol. 2015; 74: 442-52.

21

Patil V, Mahalingam K. Comprehensive analysis of reverse phase protein array data reveals characteristic unique proteomic signatures for glioblastoma subtypes. Gene. 2019; 685: 85-95.

22

Borodovsky A, Meeker AK, Kirkness EF, Zhao Q, Eberhart CG, Gallia GL, et al. A model of a patient-derived IDH1 mutant anaplastic astrocytoma with alternative lengthening of telomeres. J Neurooncol. 2015; 121: 479-87.

23

Liu JY, Zhang XB, Yan XL, Sun M, Fan YS, Huang Y. Significance of TERT and ATRX mutations in glioma. Oncol Lett. 2019; 17: 95-102.

24

Cerami E, Gao JJ, 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: 401-4.

25

Gao JJ, 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: pl1.

26

Nguyen DN, Heaphy CM, De Wilde RF, Orr BA, Odia Y, Eberhart CG, et al. Molecular and morphologic correlates of the alternative lengthening of telomeres phenotype in high-grade astrocytomas. Brain Pathol. 2013; 23: 237-43.

27

Chaurasia A, Park SH, Seo JW, Park CK. Immunohistochemical analysis of ATRX, IDH1 and p53 in glioblastoma and their correlations with patient survival. J Korean Med Sci. 2016; 31: 1208-14.

28

Louis DN, Perry A, Burger P, Ellison DW, Reifenberger G, Von Deimling A, et al. International society of neuropathology-Haarlem consensus guidelines for nervous system tumor classification and grading. Brain Pathol. 2014; 24: 429-35.

29

Louis DN, Perry A, Reifenberger G, Von Deimling A, FigarellaBranger D, Cavenee WK, et al. The 2016 World Health Organization classification of tumors of the central nervous system: a summary. Acta Neuropathol. 2016; 131: 803-20.

30

Zhou LN, Wang ZC, Hu CX, Zhang CQ, Kovatcheva-Datchary P, Yu D, et al. Integrated metabolomics and lipidomics analyses reveal metabolic reprogramming in human glioma with IDH1 mutation. J Proteome Res. 2019; 18: 960-9.

31

Ebrahimi A, Skardelly M, Bonzheim I, Ott I, Muhleisen H, Eckert F, et al. ATRX immunostaining predicts IDH and H3F3A status in gliomas. Acta Neuropathol Commun. 2016; 4: 60.

32

Deng L, Xiong PJ, Luo YH, Bu X, Qian SK, Zhong WZ, et al. Association between IDH1/2 mutations and brain glioma grade. Oncol Lett. 2018; 16: 5405-9.

33

Liu XY, Gerges N, Korshunov A, Sabha N, Khuong-Quang DA, Fontebasso AM, et al. Frequent ATRX mutations and loss of expression in adult diffuse astrocytic tumors carrying IDH1/IDH2 and TP53 mutations. Acta Neuropathol. 2012; 124: 615-25.

34

Olar A, Sulman EP. Molecular markers in low-grade glioma—toward tumor reclassification. Semin Radiat Oncol. 2015; 25: 155-63.

35

Eckel-Passow JE, Lachance DH, Molinaro AM, Walsh KM, Decker PA, Sicotte H, et al. Glioma groups based on 1p/19q, IDH, and TERT promoter mutations in tumors. N Engl J Med. 2015; 372: 2499-508.

36

Ballester LY, Fuller GN, Powell SZ, Sulman EP, Patel KP, Luthra R, et al. Retrospective analysis of molecular and immunohistochemical characterization of 381 primary brain tumors. J Neuropathol Exp Neurol. 2017; 76: 179-88.

37

Johnson BE, Mazor T, Hong CB, Barnes M, Aihara K, McLean CY, et al. Mutational analysis reveals the origin and therapy-driven evolution of recurrent glioma. Science. 2014; 343: 189-93.

38

Chen R, Cohen AL, Colman H. Targeted therapeutics in patients with high-grade gliomas: past, present, and future. Curr Treat Options Oncol. 2016; 17: 42.

39

Cho SY, Park C, Na D, Han JY, Lee J, Park OK, et al. High prevalence of TP53 mutations is associated with poor survival and an EMT signature in gliosarcoma patients. Exp Mol Med. 2017; 49: e317.

40

Clynes D, Jelinska C, Xella B, Ayyub H, Scott C, Mitson M, et al. Suppression of the alternative lengthening of telomere pathway by the chromatin remodelling factor ATRX. Nat Commun. 2015; 6: 7538.

41

Brennan CW, Verhaak RGW, McKenna A, Campos B, Noushmehr H, Salama SR, et al. The somatic genomic landscape of glioblastoma. Cell. 2013; 155: 462-77.

42

Bettegowda C, Agrawal N, Jiao YC, Sausen M, Wood LD, Hruban RH, et al. Mutations in CIC and FUBP1 contribute to human oligodendroglioma. Science. 2011; 333: 1453-5.

43

Di Stefano AL, Enciso-Mora V, Marie Y, Desestret V, Labussière M, Boisselier B, et al. Association between glioma susceptibility loci and tumour pathology defines specific molecular etiologies. Neuro-Oncol. 2013; 15: 542-7.

44

McNulty SN, Cottrell CE, Vigh-Conrad KA, Carter JH, Heusel JW, Ansstas G, et al. Beyond sequence variation: assessment of copy number variation in adult glioblastoma through targeted tumor somatic profiling. Hum Pathol. 2019; 86: 170-81.

45

Han B, Cai JQ, Gao WD, Meng XQ, Gao F, Wu PF, et al. Loss of ATRX suppresses ATM dependent DNA damage repair by modulating H3K9me3 to enhance temozolomide sensitivity in glioma. Cancer Lett. 2018; 419: 280-90.

46

Valle-García D, Qadeer ZA, McHugh DS, Ghiraldini FG, Chowdhury AH, Hasson D, et al. ATRX binds to atypical chromatin domains at the 3' exons of zinc finger genes to preserve H3K9me3 enrichment. Epigenetics. 2016; 11: 398-414.

47

Ratnakumar K, Duarte LF, LeRoy G, Hasson D, Smeets D, Vardabasso C, et al. ATRX-mediated chromatin association of histone variant macroH2A1 regulates α-globin expression. Genes Dev. 2012; 26: 433-8.

48

Law MJ, Lower KM, Voon HPJ, Hughes JR, Garrick D, Viprakasit V, et al. ATR-X syndrome protein targets tandem repeats and influences allele-specific expression in a size-dependent manner. Cell. 2010; 143: 367-78.

49

Mitson M, Kelley LA, Sternberg MJE, Higgs DR, Gibbons RJ. Functional significance of mutations in the Snf2 domain of ATRX. Hum Mol Genet. 2011; 20: 2603-10.

50

Zhang J, Chen XB. p53 tumor suppressor and iron homeostasis. FEBS J. 2019; 286: 620-9.

51

Zhang YH, Qian YJ, Zhang J, Yan WS, Jung YS, Chen MY, et al. Ferredoxin reductase is critical for p53-dependent tumor suppression via iron regulatory protein 2. Genes Dev. 2017; 31: 1243-56.

52

Wang HY, Tang K, Liang TY, Zhang WZ, Li JY, Wang W, et al. The comparison of clinical and biological characteristics between IDH1 and IDH2 mutations in gliomas. J Exp Clin Cancer Res. 2016; 35: 86.

53

Inoue M, Nakashima R, Enomoto M, Koike Y, Zhao X, Yip K, et al. Plasma redox imbalance caused by albumin oxidation promotes lung-predominant NETosis and pulmonary cancer metastasis. Nat Commun. 2018; 9: 5116.

Cancer Biology & Medicine
Pages 784-796
Cite this article:
Xie Y, Tan Y, Yang C, et al. Omics-based integrated analysis identified ATRX as a biomarker associated with glioma diagnosis and prognosis. Cancer Biology & Medicine, 2019, 16(4): 784-796. https://doi.org/10.20892/j.issn.2095-3941.2019.0143

17

Views

0

Downloads

19

Crossref

N/A

Web of Science

28

Scopus

Altmetrics

Received: 08 April 2019
Accepted: 19 July 2019
Published: 01 November 2019
© 2019 by Cancer Biology & Medicine
Return