Cancer Res Treat.  2017 Apr;49(2):387-398. 10.4143/crt.2016.106.

Underexpression of HOXA11 Is Associated with Treatment Resistance and Poor Prognosis in Glioblastoma

Affiliations
  • 1Department of Neurosurgery, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, Korea. gknife@snu.ac.kr
  • 2Department of Neurosurgery, Seoul National University College of Medicine, Seoul, Korea. nsckpark@snu.ac.kr
  • 3Department of Physiology, Ajou University School of Medicine, Suwon, Korea.
  • 4Department of Neurosurgery, Ajou University School of Medicine, Suwon, Korea.
  • 5Department of Neurosurgery, Korea University College of Medicine, Seoul, Korea.
  • 6Department of Radiation Oncology, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, Korea.
  • 7Department of Internal Medicine, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, Korea.
  • 8Department of Neurology, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, Korea.
  • 9Department of Radiology, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, Korea.
  • 10Department of Pathology, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, Korea.

Abstract

PURPOSE
Homeobox (HOX) genes are essential developmental regulators that should normally be in the silenced state in an adult brain. The aberrant expression of HOX genes has been associated with the prognosis of many cancer types, including glioblastoma (GBM). This study examined the identity and role of HOX genes affecting GBM prognosis and treatment resistance.
MATERIALS AND METHODS
The full series of HOX genes of five pairs of initial and recurrent human GBM samples were screened by microarray analysis to determine the most plausible candidate responsible for GBM prognosis. Another 20 newly diagnosed GBM samples were used for prognostic validation. In vitro experiments were performed to confirm the role of HOX in treatment resistance. Mediators involved in HOX gene regulation were searched using differentially expressed gene analysis, gene set enrichment tests, and network analysis.
RESULTS
The underexpression of HOXA11 was identified as a consistent signature for a poor prognosis among the HOX genes. The overall survival of the GBM patients indicated a significantly favorable prognosis in patients with high HOXA11 expression (31±15.3 months) compared to the prognoses in thosewith low HOXA11 expression (18±7.3 months, p=0.03). When HOXA11 was suppressed in the GBM cell lines, the anticancer effect of radiotherapy and/or temozolomide declined. In addition, five candidate mediators (TGFBR2, CRIM1, TXNIP, DPYSL2, and CRMP1) that may confer an oncologic effect after HOXA11 suppression were identified.
CONCLUSION
The treatment resistance induced by the underexpression of HOXA11 can contribute to a poor prognosis in GBM. Further investigation will be needed to confirm the value of HOXA11 as a potential target for overcoming the treatment resistance by developing chemo- or radiosensitizers.

Keyword

Homeobox genes; HOXA11; Glioblastoma; Treatment resistance

MeSH Terms

Adult
Brain
Cell Line
Genes, Homeobox
Glioblastoma*
Humans
In Vitro Techniques
Microarray Analysis
Prognosis*
Radiotherapy

Figure

  • Fig. 1. Relative changes in the expression of homeobox (HOX) genes among five pairs of primary and recurrent glioblastoma (GBM) samples as determined by microarray analysis. (A) Heatmap and hierarchical clustering analysis showed inconsistent results in sequential HOX gene expression changes between the primary and recurrent samples, except for HOXA11. (B) HOXA11 gene is the only HOX gene that was consistently down-regulated in the recurrent GBM samples compared to primary samples for all five sample pairs.

  • Fig. 2. (A) Results of western blot analysis for normalized HOXA11 expression. The patients were grouped according to the HOXA11 expression macroscopically, and the difference in expression level was confirmed by intensity measurement of the bands. (B) The overall survival of a separate cohort of glioblastoma patients by HOXA11 expression. The survival was significantly longer in patients with high HOXA11 expression (31±15.3 months) than in those with low HOXA11 expression (18±7.3 months, p=0.037).

  • Fig. 3. Treatment resistance of glioma cell lines (U251, U373, and LN18) assessed by cell viability tests after the inhibition of HOXA11 and presented as relative viable cell fractions. Direct inhibition of HOXA11 by small interfering RNA (siRNA) results in a significant increase in cell survival, 72 hours after single or combination applications of radiation (RT) and temozolomide (TMZ) treatments (*p < 0.05).

  • Fig. 4. Identification of the HOXA11-regulated genes in LN18 cells. (A) Reverse transcription polymerase chain reaction results from LN18 cells after HOXA11 knockdown by siRNA. (B) HOXA11-silencing siRNA versus control siRNA transduced cells compared using Affymetrix GeneChip Human Gene 1.0ST Arrays. The probe sets with fold changes greater than 2-fold are shown. After HOXA11 suppression, 11 up-regulated and 51 down-regulated genes were identified. (C) MA plots showing the distribution of regulated genes. GAPDH, glyceraldehyde 3-phosphate dehydrogenase; IQR, interquartile range; siRNA, small interfering RNA.

  • Fig. 5. Coexpression network incorporating selected gene sets drawn from the functional gene annotation enrichment analysis of differentially expressed genes after HOXA11 suppression. The network was constructed based on coexpression interactions using GeneMANIA software (ver. 3.1.2.8). The key hub regulators associated with HOXA11 suppression are expressed as black solid circles.


Reference

References

1. Shah N, Sukumar S. The Hox genes and their roles in oncogenesis. Nat Rev Cancer. 2010; 10:361–71.
Article
2. Takahashi Y, Hamada J, Murakawa K, Takada M, Tada M, Nogami I, et al. Expression profiles of 39 HOX genes in normal human adult organs and anaplastic thyroid cancer cell lines by quantitative real-time RT-PCR system. Exp Cell Res. 2004; 293:144–53.
Article
3. Abate-Shen C. Deregulated homeobox gene expression in cancer: cause or consequence? Nat Rev Cancer. 2002; 2:777–85.
Article
4. Abdel-Fattah R, Xiao A, Bomgardner D, Pease CS, Lopes MB, Hussaini IM. Differential expression of HOX genes in neoplastic and non-neoplastic human astrocytes. J Pathol. 2006; 209:15–24.
5. Bodey B, Bodey B Jr, Siegel SE, Kaiser HE. Immunocytochemical detection of the homeobox B3, B4, and C6 gene products in childhood medulloblastomas/primitive neuroectodermal tumors. Anticancer Res. 2000; 20:1769–80.
6. Buccoliero AM, Castiglione F, Rossi Degl'Innocenti D, Ammanati F, Giordano F, Sanzo M, et al. Hox-D genes expression in pediatric low-grade gliomas: real-time-PCR study. Cell Mol Neurobiol. 2009; 29:1–6.
Article
7. Ferrando AA, Armstrong SA, Neuberg DS, Sallan SE, Silverman LB, Korsmeyer SJ, et al. Gene expression signatures in MLL-rearranged T-lineage and B-precursor acute leukemias: dominance of HOX dysregulation. Blood. 2003; 102:262–8.
Article
8. Miao J, Wang Z, Provencher H, Muir B, Dahiya S, Carney E, et al. HOXB13 promotes ovarian cancer progression. Proc Natl Acad Sci U S A. 2007; 104:17093–8.
Article
9. Murat A, Migliavacca E, Gorlia T, Lambiv WL, Shay T, Hamou MF, et al. Stem cell-related "self-renewal" signature and high epidermal growth factor receptor expression associated with resistance to concomitant chemoradiotherapy in glioblastoma. J Clin Oncol. 2008; 26:3015–24.
Article
10. Wang Z, Dahiya S, Provencher H, Muir B, Carney E, Coser K, et al. The prognostic biomarkers HOXB13, IL17BR, and CHDH are regulated by estrogen in breast cancer. Clin Cancer Res. 2007; 13:6327–34.
Article
11. Gaspar N, Marshall L, Perryman L, Bax DA, Little SE, Viana-Pereira M, et al. MGMT-independent temozolomide resistance in pediatric glioblastoma cells associated with a PI3-kinase-mediated HOX/stem cell gene signature. Cancer Res. 2010; 70:9243–52.
12. Kim JW, Kim JY, Kim JE, Kim SK, Chung HT, Park CK. HOXA10 is associated with temozolomide resistance through regulation of the homologous recombinant DNA repair pathway in glioblastoma cell lines. Genes Cancer. 2014; 5:165–74.
Article
13. Kim KJ, Moon SM, Kim SA, Kang KW, Yoon JH, Ahn SG. Transcriptional regulation of MDR-1 by HOXC6 in multidrug-resistant cells. Oncogene. 2013; 32:3339–49.
Article
14. Bao S, Wu Q, McLendon RE, Hao Y, Shi Q, Hjelmeland AB, et al. Glioma stem cells promote radioresistance by preferential activation of the DNA damage response. Nature. 2006; 444:756–60.
Article
15. Chen J, Li Y, Yu TS, McKay RM, Burns DK, Kernie SG, et al. A restricted cell population propagates glioblastoma growth after chemotherapy. Nature. 2012; 488:522–6.
Article
16. Atkinson SP, Koch CM, Clelland GK, Willcox S, Fowler JC, Stewart R, et al. Epigenetic marking prepares the human HOXA cluster for activation during differentiation of pluripotent cells. Stem Cells. 2008; 26:1174–85.
Article
17. Kwon SM, Kang SH, Park CK, Jung S, Park ES, Lee JS, et al. Recurrent glioblastomas reveal molecular subtypes associated with mechanistic implications of drug-resistance. PLoS One. 2015; 10:e0140528.
Article
18. Reich M, Liefeld T, Gould J, Lerner J, Tamayo P, Mesirov JP. GenePattern 2.0. Nat Genet. 2006; 38:500–1.
Article
19. Huang DW, Sherman BT, Lempicki RA. Systematic and integrative analysis of large gene lists using DAVID bioinformatics resources. Nat Protoc. 2009; 4:44–57.
Article
20. Warde-Farley D, Donaldson SL, Comes O, Zuberi K, Badrawi R, Chao P, et al. The GeneMANIA prediction server: biological network integration for gene prioritization and predicting gene function. Nucleic Acids Res. 2010; 38:W214–20.
Article
21. Hueber SD, Lohmann I. Shaping segments: Hox gene function in the genomic age. Bioessays. 2008; 30:965–79.
22. Ohgo S, Itoh A, Suzuki M, Satoh A, Yokoyama H, Tamura K. Analysis of hoxa11 and hoxa13 expression during patternless limb regeneration in Xenopus. Dev Biol. 2010; 338:148–57.
Article
23. Argiropoulos B, Humphries RK. Hox genes in hematopoiesis and leukemogenesis. Oncogene. 2007; 26:6766–76.
Article
24. Bahrani-Mostafavi Z, Tickle TL, Zhang J, Bennett KE, Vachris JC, Spencer MD, et al. Correlation analysis of HOX, ErbB and IGFBP family gene expression in ovarian cancer. Cancer Invest. 2008; 26:990–8.
Article
25. Cillo C, Barba P, Freschi G, Bucciarelli G, Magli MC, Boncinelli E. HOX gene expression in normal and neoplastic human kidney. Int J Cancer. 1992; 51:892–7.
Article
26. De Vita G, Barba P, Odartchenko N, Givel JC, Freschi G, Bucciarelli G, et al. Expression of homeobox-containing genes in primary and metastatic colorectal cancer. Eur J Cancer. 1993; 29A:887–93.
Article
27. Costa BM, Smith JS, Chen Y, Chen J, Phillips HS, Aldape KD, et al. Reversing HOXA9 oncogene activation by PI3K inhibition: epigenetic mechanism and prognostic significance in human glioblastoma. Cancer Res. 2010; 70:453–62.
28. Lim JY, Yoon SO, Seol SY, Hong SW, Kim JW, Choi SH, et al. Overexpression of miR-196b and HOXA10 characterize a poor-prognosis gastric cancer subtype. World J Gastroenterol. 2013; 19:7078–88.
Article
29. Carrio M, Arderiu G, Myers C, Boudreau NJ. Homeobox D10 induces phenotypic reversion of breast tumor cells in a three-dimensional culture model. Cancer Res. 2005; 65:7177–85.
Article
30. Petrini M, Felicetti F, Bottero L, Errico MC, Morsilli O, Boe A, et al. HOXB1 restored expression promotes apoptosis and differentiation in the HL60 leukemic cell line. Cancer Cell Int. 2013; 13:101.
Article
31. Rhoads K, Arderiu G, Charboneau A, Hansen SL, Hoffman W, Boudreau N. A role for Hox A5 in regulating angiogenesis and vascular patterning. Lymphat Res Biol. 2005; 3:240–52.
Article
32. Carrera M, Bitu CC, de Oliveira CE, Cervigne NK, Graner E, Manninen A, et al. HOXA10 controls proliferation, migration and invasion in oral squamous cell carcinoma. Int J Clin Exp Pathol. 2015; 8:3613–23.
33. Cui XP, Qin CK, Zhang ZH, Su ZX, Liu X, Wang SK, et al. HOXA10 promotes cell invasion and MMP-3 expression via TGFbeta2-mediated activation of the p38 MAPK pathway in pancreatic cancer cells. Dig Dis Sci. 2014; 59:1442–51.
34. Kurscheid S, Bady P, Sciuscio D, Samarzija I, Shay T, Vassallo I, et al. Chromosome 7 gain and DNA hypermethylation at the HOXA10 locus are associated with expression of a stem cell related HOX-signature in glioblastoma. Genome Biol. 2015; 16:16.
Article
35. Li B, Cao X, Weng C, Wu Y, Fang X, Zhang X, et al. HoxA10 induces proliferation in human prostate carcinoma PC-3 cell line. Cell Biochem Biophys. 2014; 70:1363–8.
Article
36. Liborio-Kimura TN, Jung HM, Chan EK. miR-494 represses HOXA10 expression and inhibits cell proliferation in oral cancer. Oral Oncol. 2015; 51:151–7.
37. Oue N, Sentani K, Sakamoto N, Yasui W. Clinicopathologic and molecular characteristics of gastric cancer showing gastric and intestinal mucin phenotype. Cancer Sci. 2015; 106:951–8.
Article
38. Xiao ZD, Jiao CY, Huang HT, He LJ, Zhao JJ, Lu ZY, et al. miR-218 modulate hepatocellular carcinoma cell proliferation through PTEN/AKT/PI3K pathway and HoxA10. Int J Clin Exp Pathol. 2014; 7:4039–44.
39. Zhang HY, Li JH, Li G, Wang SR. Activation of ARK5/miR-1181/HOXA10 axis promotes epithelial-mesenchymal transition in ovarian cancer. Oncol Rep. 2015; 34:1193–202.
Article
40. Zhang L, Wan Y, Jiang Y, Ma J, Liu J, Tang W, et al. Upregulation HOXA10 homeobox gene in endometrial cancer: role in cell cycle regulation. Med Oncol. 2014; 31:52.
Article
41. Bai Y, Fang N, Gu T, Kang Y, Wu J, Yang D, et al. HOXA11 gene is hypermethylation and aberrant expression in gastric cancer. Cancer Cell Int. 2014; 14:79.
Article
42. Cui Y, Gao D, Linghu E, Zhan Q, Chen R, Brock MV, et al. Epigenetic changes and functional study of HOXA11 in human gastric cancer. Epigenomics. 2015; 7:201–13.
Article
43. Fiegl H, Windbichler G, Mueller-Holzner E, Goebel G, Lechner M, Jacobs IJ, et al. HOXA11 DNA methylation: a novel prognostic biomarker in ovarian cancer. Int J Cancer. 2008; 123:725–9.
44. Hwang JA, Lee BB, Kim Y, Park SE, Heo K, Hong SH, et al. HOXA11 hypermethylation is associated with progression of non-small cell lung cancer. Oncotarget. 2013; 4:2317–25.
Article
45. Laffaire J, Everhard S, Idbaih A, Criniere E, Marie Y, de Reynies A, et al. Methylation profiling identifies 2 groups of gliomas according to their tumorigenesis. Neuro Oncol. 2011; 13:84–98.
Article
46. Martinez R, Martin-Subero JI, Rohde V, Kirsch M, Alaminos M, Fernandez AF, et al. A microarray-based DNA methylation study of glioblastoma multiforme. Epigenetics. 2009; 4:255–64.
Article
47. Skiriute D, Vaitkiene P, Asmoniene V, Steponaitis G, Deltuva VP, Tamasauskas A. Promoter methylation of AREG, HOXA11, hMLH1, NDRG2, NPTX2 and Tes genes in glioblastoma. J Neurooncol. 2013; 113:441–9.
Article
48. Fang F, Munck J, Tang J, Taverna P, Wang Y, Miller DF, et al. The novel, small-molecule DNA methylation inhibitor SGI-110 as an ovarian cancer chemosensitizer. Clin Cancer Res. 2014; 20:6504–16.
Article
49. Matei D, Fang F, Shen C, Schilder J, Arnold A, Zeng Y, et al. Epigenetic resensitization to platinum in ovarian cancer. Cancer Res. 2012; 72:2197–205.
Article
50. Shih JY, Lee YC, Yang SC, Hong TM, Huang CY, Yang PC. Collapsin response mediator protein-1: a novel invasion-suppressor gene. Clin Exp Metastasis. 2003; 20:69–76.
51. Mukherjee J, DeSouza LV, Micallef J, Karim Z, Croul S, Siu KW, et al. Loss of collapsin response mediator protein1, as detected by iTRAQ analysis, promotes invasion of human gliomas expressing mutant EGFRvIII. Cancer Res. 2009; 69:8545–54.
Article
52. Yilmaz N, Ozaksit G, Terzi YK, Yilmaz S, Budak B, Aksakal O, et al. HOXA11 and MMP2 gene expression in uterosacral ligaments of women with pelvic organ prolapse. J Turk Ger Gynecol Assoc. 2014; 15:104–8.
Article
Full Text Links
  • CRT
Actions
Cited
CITED
export Copy
Close
Share
  • Twitter
  • Facebook
Similar articles
Copyright © 2024 by Korean Association of Medical Journal Editors. All rights reserved.     E-mail: koreamed@kamje.or.kr