Brain Tumor Res Treat.  2020 Apr;8(1):43-52. 10.14791/btrt.2020.8.e1.

Inhibition of the Spectraplakin Protein Microtubule ActinCrosslinking Factor 1 Sensitizes Glioblastomas to Radiation

Affiliations
  • 1Department of Biological Sciences, Tennessee State University, Nashville, TN, USA

Abstract

Background
: Microtubule actin crosslinking factor 1 (MACF1) is a spectraplakin cytoskeletal crosslinking protein whose function and role in cancer biology has lacked investigation. Recent studies have identified MACF1 as a novel target in glioblastomas expressed in tissue from tumor patient explants but not normal brain tissue and when silenced has an antitumorigenic impact on these tumors. Radiation as a single agent therapy to treat glioblastomas has been used for decades and has done little to improve survival of individuals diagnosed with this disease. However, contemporary clinical radiotherapy protocols have provided evidence that combinatorial radiotherapy approaches confer a therapeutic benefit in glioblastoma patients. In this study MACF1 was investigated as a radiosensitization target in glioblastomas.
Methods
: To provide context of MACF1 in glioblastomas, The Cancer Genome Atlas expression analyses were performed in conjunction with genes associated with glioblastoma evolution, while a genetic inhibitory approach, cell migratory assays, and immunofluorescence procedures were used to evaluate responses to MACF1 suppression with radiation. Additionally, expression analyses were conducted to assess co-expression of mTOR signaling pathway regulators and MACF1 in glioblastoma patient samples.
Results
: Our amalgamation approach demonstrated that negative regulation of MACF1, which was positively correlated with epidermal growth factor receptor and p70s6k expression, enhanced the sensitivity of glioblastoma cells to radiation as a consequence of reducing glioblastoma cell viability and migration. Mechanistically, the antitumorigenic effects on glioblastoma cell behaviors after radiation and impairing MACF1 function were associated with decreased expression of ribosomal protein S6, a downstream effector of p70s6k.
Conclusion
: MACF1 represents a diagnostic marker with target specificity in glioblastomas that can enhance the efficacy of radiation while minimizing normal tissue toxicity. This approach could potentially expand combinatorial radiation strategies for glioblastoma treatments via impairment of translational regulatory processes that contribute to poor patient survival.

Keyword

MACF1; Glioblastoma; Radiation; Ribosomal protein S6

Figure

  • Fig. 1 Correlative expression of MACF1 to patient survival and glioblastoma associated genes. Comparative survival curve analysis of glioblastoma patients with low MACF1 expression (<1.048; n=74) and high MACF1 mRNA expression (>1.048; n=83). MACF1 co-expression with EGFR, PTEN, p53, and CDKN2A.

  • Fig. 2 Down-regulation of MACF1 in genetically silenced glioblastoma cells. U251 glioblastoma cells treated with non-targeting shRNAs (A–C) and shRNAs targeting MACF1 (D–F); A172 glioblastoma cells treated with non-targeting shRNAs (G–I) and shRNAs targeting MACF1 (J–L); MACF1 expression (green); DAPI (blue); cytoplasmic MACF1 expression (arrows). Displayed are immunofluorescent images representative of three separate independent experiments with comparable results. Image J was used to measure the fluorescence intensity from a total of 50 individual cells from three separate independent experiments (n=3) and averaged. Student's t-test was used to determine statistical significance (**p<0.01; error bars: standard error of the means). Scale bar=100 µm; total magnification ×200; RFU: relative fluorescence units.

  • Fig. 3 MACF1 inhibition sensitizes glioblastoma cells to radiation. Impairing MACF1 function reduces cell viability in non-irradiated cells and enhances the efficacy of radiation. Data displayed is of a single experiment performed in duplicate and equivalent to three separate independent experiments performed in duplicate with comparable outcomes. ANOVA analysis followed by Tukey's post-hoc analysis revealed statistical significant differences between means of experimental conditions (*p<0.05; **p<0.01; error bars: standard error of the means).

  • Fig. 4 Radiation and suppression of MACF1 impairs glioblastoma cell migration. U251 (A–D) and A172 (E–H) cells treated with shMACF1 and irradiated with a single 5 Gy dose. Displayed are stained migrating cells in Boyden chambers and data from a single experiment performed in duplicate representative of at least three separate independent experiments performed in duplicate with comparable results 24 hours after radiation exposure. ANOVA analysis followed by Tukey's post-hoc analysis revealed statistical significant differences between means of experimental conditions (*p<0.05; **p<0.01; error bars: standard error of the means). Scale bar=200 µm; stained with crystal violet; total magnification ×100.

  • Fig. 5 Evaluation of translational regulatory proteins as mechanistic components of the radiosensitization effects of downregulating MACF1. A, B: Positive correlative expression of MACF1 with ribosomal protein S6 regulators MTOR and RPS6KB1 in clinical glioblastoma patients. C: Silencing MACF1 combined with radiation decreases p-ribosomal protein S6 expression. Immunofluorescence images displayed are from a single experiment in U251 glioblastoma cells at 24 hours and representative of three separate experiments that showed comparable results. Image J was used to measure the fluorescence intensity from a total of 50 individual cells from three separate independent experiments (n=3) and averaged. ANOVA analysis followed by Tukey's post-hoc analysis revealed statistical significant differences between means of experimental conditions (****p<0.0001; error bars: standard error of the means). Scale bar=80 µm; total magnification ×400. RFU: relative fluorescence units.


Reference

1. Bernier G, Mathieu M, De Repentigny Y, Vidal SM, Kothary R. Cloning and characterization of mouse ACF7, a novel member of the dystonin subfamily of actin binding proteins. Genomics. 1996; 38:19–29. PMID: 8954775.
2. Ka M, Jung EM, Mueller U, Kim WY. MACF1 regulates the migration of pyramidal neurons via microtubule dynamics and GSK-3 signaling. Dev Biol. 2014; 395:4–18. PMID: 25224226.
3. Hu L, Su P, Li R, et al. Knockdown of microtubule actin crosslinking factor 1 inhibits cell proliferation in MC3T3-E1 osteoblastic cells. BMB Rep. 2015; 48:583–588. PMID: 26277981.
4. Afghani N, Mehta T, Wang J, Tang N, Skalli O, Quick QA. Microtubule actin cross-linking factor 1, a novel target in glioblastoma. Int J Oncol. 2017; 50:310–316. PMID: 27959385.
5. Arai E, Sakamoto H, Ichikawa H, et al. Multilayer-omics analysis of renal cell carcinoma, including the whole exome, methylome and transcriptome. Int J Cancer. 2014; 135:1330–1342. PMID: 24504440.
6. Chang YS, Huang HD, Yeh KT, Chang JG. Identification of novel mutations in endometrial cancer patients by whole-exome sequencing. Int J Oncol. 2017; 50:1778–1784. PMID: 28339086.
7. Chen HJ, Lin CM, Lin CS, Perez-Olle R, Leung CL, Liem RK. The role of microtubule actin cross-linking factor 1 (MACF1) in the Wnt signaling pathway. Genes Dev. 2006; 20:1933–1945. PMID: 16815997.
8. McCord M, Mukouyama YS, Gilbert MR, Jackson S. Targeting WNT signaling for multifaceted glioblastoma therapy. Front Cell Neurosci. 2017; 11:318. PMID: 29081735.
9. Shapiro WR, Green SB, Burger PC, et al. Randomized trial of three chemotherapy regimens and two radiotherapy regimens and two radiotherapy regimens in postoperative treatment of malignant glioma. Brain tumor cooperative group trial 8001. J Neurosurg. 1989; 71:1–9.
10. Khosla D. Concurrent therapy to enhance radiotherapeutic outcomes in glioblastoma. Ann Transl Med. 2016; 4:54. PMID: 26904576.
11. Stupp R, Hegi ME, Mason WP, 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; 10:459–466. PMID: 19269895.
12. Kahn J, Hayman TJ, Jamal M, et al. The mTORC1/mTORC2 inhibitor AZD2014 enhances the radiosensitivity of glioblastoma stem-like cells. Neuro Oncol. 2014; 16:29–37. PMID: 24311635.
13. Gil del Alcazar CR, Hardebeck MC, Mukherjee B, et al. Inhibition of DNA double-strand break repair by the dual PI3K/mTOR inhibitor NVP-BEZ235 as a strategy for radiosensitization of glioblastoma. Clin Cancer Res. 2014; 20:1235–1248. PMID: 24366691.
14. Ahmed SU, Carruthers R, Gilmour L, Yildirim S, Watts C, Chalmers AJ. Selective inhibition of parallel DNA damage response pathways optimizes radiosensitization of glioblastoma stem-like cells. Cancer Res. 2015; 75:4416–4428. PMID: 26282173.
15. King AR, Corso CD, Chen EM, et al. Local DNA repair inhibition for sustained radiosensitization of high-grade gliomas. Mol Cancer Ther. 2017; 16:1456–1469. PMID: 28566437.
16. Shi F, Guo H, Zhang R, et al. The PI3K inhibitor GDC-0941 enhances radiosensitization and reduces chemoresistance to temozolomide in GBM cell lines. Neuroscience. 2017; 346:298–308. PMID: 28147244.
17. Sulzmaier FJ, Ramos JW. RSK isoforms in cancer cell invasion and metastasis. Cancer Res. 2013; 73:6099–6105. PMID: 24097826.
18. Cargnello M, Tcherkezian J, Roux PP. The expanding role of mTOR in cancer cell growth and proliferation. Mutagenesis. 2015; 30:169–176. PMID: 25688110.
19. Ronellenfitsch MW, Zeiner PS, Mittelbronn M, et al. Akt and mTORC1 signaling as predictive biomarkers for the EGFR antibody nimotuzumab in glioblastoma. Acta Neuropathol Commun. 2018; 6:81. PMID: 30129426.
20. Sekhar KR, Benamar M, Venkateswaran A, et al. Targeting nucleophosmin 1 represents a rational strategy for radiation sensitization. Int J Radiat Oncol Biol Phys. 2014; 89:1106–1114. PMID: 25035215.
21. The cBioPortal for Cancer Genomics. The Cancer Genome Atlas. Accessed December 15, 2019. at https://www.cbioportal.org/.
22. Kao GD, Jiang Z, Fernandes AM, Gupta AK, Maity A. Inhibition of phosphatidylinositol-3-OH kinase/Akt signaling impairs DNA repair in glioblastoma cells following ionizing radiation. J Biol Chem. 2007; 282:21206–21212. PMID: 17513297.
23. Kil WJ, Cerna D, Burgan WE, et al. In vitro and in vivo radiosensitization induced by the DNA methylating agent temozolomide. Clin Cancer Res. 2008; 14:931–938. PMID: 18245557.
24. Sheng Y, Xu M, Li C, et al. Nm23-H1 is involved in the repair of ionizing radiation-induced DNA double-strand breaks in the A549 lung cancer cell line. BMC Cancer. 2018; 18:710. PMID: 29970055.
25. Martinou M, Giannopoulou E, Malatara G, Argyriou AA, Kalofonos HP, Kardamakis D. Ionizing radiation affects epidermal growth factor receptor signalling and metalloproteinase secretion in glioma cells. Cancer Genomics Proteomics. 2011; 8:33–38. PMID: 21289335.
26. Contessa JN, Hampton J, Lammering G, et al. Ionizing radiation activates Erb-B receptor dependent Akt and p70 S6 kinase signaling in carcinoma cells. Oncogene. 2002; 21:4032–4041. PMID: 12037685.
27. Kim Y, Kim KH, Lee J, et al. Wnt activation is implicated in glioblastoma radioresistance. Lab Invest. 2012; 92:466–473. PMID: 22083670.
28. Wild-Bode C, Weller M, Rimner A, Dichgans J, Wick W. Sublethal irradiation promotes migration and invasiveness of glioma cells: implications for radiotherapy of human glioblastoma. Cancer Res. 2001; 61:2744–2750. PMID: 11289157.
29. Lammering G, Hewit TH, Valerie K, et al. EGFRvIII-mediated radioresistance through a strong cytoprotective response. Oncogene. 2003; 22:5545–5553. PMID: 12944901.
30. Pelloski CE, Lin E, Zhang L, et al. Prognostic associations of activated mitogen-activated protein kinase and Akt pathways in glioblastoma. Clin Cancer Res. 2006; 12:3935–3941. PMID: 16818690.
31. Li B, Yuan M, Kim IA, Chang CM, Bernhard EJ, Shu HK. Mutant epidermal growth factor receptor displays increased signaling through the phosphatidylinositol-3 kinase/AKT pathway and promotes radioresistance in cells of astrocytic origin. Oncogene. 2004; 23:4594–4602. PMID: 15077177.
32. Liu Q, Nguyen DH, Dong Q, et al. Molecular properties of CD133+ glioblastoma stem cells derived from treatment-refractory recurrent brain tumors. J Neurooncol. 2009; 94:1–19. PMID: 19468690.
33. Shih HA, Betensky RA, Dorfman MV, Louis DN, Loeffler JS, Batchelor TT. Genetic analyses for predictors of radiation response in glioblastoma. Int J Radiat Oncol Biol Phys. 2005; 63:704–710. PMID: 15978739.
34. Barker FG 2nd, Simmons ML, Chang SM, et al. EGFR overexpression and radiation response in glioblastoma multiforme. Int J Radiat Oncol Biol Phys. 2001; 51:410–418. PMID: 11567815.
35. Yang P, Zhang W, Wang Y, et al. IDH mutation and MGMT promoter methylation in glioblastoma: results of a prospective registry. Oncotarget. 2015; 6:40896–40906. PMID: 26503470.
36. Tini P, Nardone V, Pastina P, et al. Patients affected by unmethylated O(6)-methylguanine-DNA methyltransferase glioblastoma undergoing radiochemotherapy may benefit from moderately dose-escalated radiotherapy. Biomed Res Int. 2017; 2017:9461402. PMID: 29159183.
37. Miki S, Imamichi S, Fujimori H, et al. Concomitant administration of radiation with eribulin improves the survival of mice harboring intracerebral glioblastoma. Cancer Sci. 2018; 109:2275–2285. PMID: 29758120.
38. Wang Y, Mei H, Shao Q, Wang J, Lin Z. Association of ribosomal protein S6 kinase 1 with cellular radiosensitivity of non-small lung cancer. Int J Radiat Biol. 2017; 93:581–589. PMID: 28276898.
39. Lee S, Nahm M, Lee M, et al. The F-actin-microtubule crosslinker Shot is a platform for Krasavietz-mediated translational regulation of midline axon repulsion. Development. 2007; 134:1767–1777. PMID: 17409115.
Full Text Links
  • BTRT
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