Skip to main content
Top
Published in: BMC Cancer 1/2015

Open Access 01-12-2015 | Research article

TRIM8 downregulation in glioma affects cell proliferation and it is associated with patients survival

Authors: Lucia Micale, Carmela Fusco, Andrea Fontana, Raffaela Barbano, Bartolomeo Augello, Pasquelena De Nittis, Massimiliano Copetti, Maria Teresa Pellico, Barbara Mandriani, Dario Cocciadiferro, Paola Parrella, Vito Michele Fazio, Lucia Maria Cecilia Dimitri, Vincenzo D’Angelo, Chiara Novielli, Lidia Larizza, Antonio Daga, Giuseppe Merla

Published in: BMC Cancer | Issue 1/2015

Login to get access

Abstract

Background

Human gliomas are a heterogeneous group of primary malignant brain tumors whose molecular pathogenesis is not yet solved. In this regard, a major research effort has been directed at identifying novel specific glioma-associated genes. Here, we investigated the effect of TRIM8 gene in glioma.

Methods

TRIM8 transcriptional level was profiled in our own glioma cases collection by qPCR and confirmed in the independent TCGA glioma cohort. The association between TRIM8 expression and Overall Survival and Progression-free Survival in TCGA cohort was determined by using uni-multivariable Cox regression analysis. The effect of TRIM8 on patient glioma cell proliferation was evaluated by performing MTT and clonogenic assays. The mechanisms causing the reduction of TRIM8 expression were explored by using qPCR and in vitro assays.

Results

We showed that TRIM8 expression correlates with unfavorable clinical outcome in glioma patients. We found that a restored TRIM8 expression induced a significant reduction of clonogenic potential in U87MG and patient’s glioblastoma cells. Finally we provide experimental evidences showing that miR-17 directly targets the 3′ UTR of TRIM8 and post-transcriptionally represses the expression of TRIM8.

Conclusions

Our study provides evidences that TRIM8 may participate in the carcinogenesis and progression of glioma and that the transcriptional repression of TRIM8 might have potential value for predicting poor prognosis in glioma patients.
Appendix
Available only for authorised users
Literature
2.
go back to reference Ohgaki H, Dessen P, Jourde B, Horstmann S, Nishikawa T, Di Patre PL, et al. Genetic pathways to glioblastoma: a population-based study. Cancer Res. 2004;64(19):6892–9.CrossRefPubMed Ohgaki H, Dessen P, Jourde B, Horstmann S, Nishikawa T, Di Patre PL, et al. Genetic pathways to glioblastoma: a population-based study. Cancer Res. 2004;64(19):6892–9.CrossRefPubMed
3.
go back to reference Furnari FB, Fenton T, Bachoo RM, Mukasa A, Stommel JM, Stegh A, et al. Malignant astrocytic glioma: genetics, biology, and paths to treatment. Genes Dev. 2007;21(21):2683–710.CrossRefPubMed Furnari FB, Fenton T, Bachoo RM, Mukasa A, Stommel JM, Stegh A, et al. Malignant astrocytic glioma: genetics, biology, and paths to treatment. Genes Dev. 2007;21(21):2683–710.CrossRefPubMed
4.
go back to reference Rasheed BK, McLendon RE, Friedman HS, Friedman AH, Fuchs HE, Bigner DD, et al. Chromosome 10 deletion mapping in human gliomas: a common deletion region in 10q25. Oncogene. 1995;10(11):2243–6.PubMed Rasheed BK, McLendon RE, Friedman HS, Friedman AH, Fuchs HE, Bigner DD, et al. Chromosome 10 deletion mapping in human gliomas: a common deletion region in 10q25. Oncogene. 1995;10(11):2243–6.PubMed
5.
go back to reference Reymond A, Meroni G, Fantozzi A, Merla G, Cairo S, Luzi L, et al. The tripartite motif family identifies cell compartments. EMBO J. 2001;20(9):2140–51.CrossRefPubMedPubMedCentral Reymond A, Meroni G, Fantozzi A, Merla G, Cairo S, Luzi L, et al. The tripartite motif family identifies cell compartments. EMBO J. 2001;20(9):2140–51.CrossRefPubMedPubMedCentral
6.
go back to reference Okumura F, Matsunaga Y, Katayama Y, Nakayama KI, Hatakeyama S. TRIM8 modulates STAT3 activity through negative regulation of PIAS3. J Cell Sci. 2010;123(Pt 13):2238–45.CrossRefPubMed Okumura F, Matsunaga Y, Katayama Y, Nakayama KI, Hatakeyama S. TRIM8 modulates STAT3 activity through negative regulation of PIAS3. J Cell Sci. 2010;123(Pt 13):2238–45.CrossRefPubMed
7.
go back to reference Caratozzolo MF, Micale L, Turturo MG, Cornacchia S, Fusco C, Marzano F, et al. TRIM8 modulates p53 activity to dictate cell cycle arrest. Cell Cycle. 2012;11(3):511–23.CrossRefPubMed Caratozzolo MF, Micale L, Turturo MG, Cornacchia S, Fusco C, Marzano F, et al. TRIM8 modulates p53 activity to dictate cell cycle arrest. Cell Cycle. 2012;11(3):511–23.CrossRefPubMed
8.
go back to reference Carinci F, Arcelli D, Lo Muzio L, Francioso F, Valentini D, Evangelisti R, et al. Molecular classification of nodal metastasis in primary larynx squamous cell carcinoma. Transl Res. 2007;150(4):233–45.CrossRefPubMed Carinci F, Arcelli D, Lo Muzio L, Francioso F, Valentini D, Evangelisti R, et al. Molecular classification of nodal metastasis in primary larynx squamous cell carcinoma. Transl Res. 2007;150(4):233–45.CrossRefPubMed
9.
go back to reference Caratozzolo MF, Valletti A, Gigante M, Aiello I, Mastropasqua F, Marzano F, et al. TRIM8 anti-proliferative action against chemo-resistant renal cell carcinoma. Oncotarget. 2014;5(17):7446–57.CrossRefPubMedPubMedCentral Caratozzolo MF, Valletti A, Gigante M, Aiello I, Mastropasqua F, Marzano F, et al. TRIM8 anti-proliferative action against chemo-resistant renal cell carcinoma. Oncotarget. 2014;5(17):7446–57.CrossRefPubMedPubMedCentral
10.
go back to reference Malzkorn B, Wolter M, Liesenberg F, Grzendowski M, Stuhler K, Meyer HE, et al. Identification and functional characterization of microRNAs involved in the malignant progression of gliomas. Brain Pathol. 2011;20(3):539–50.CrossRef Malzkorn B, Wolter M, Liesenberg F, Grzendowski M, Stuhler K, Meyer HE, et al. Identification and functional characterization of microRNAs involved in the malignant progression of gliomas. Brain Pathol. 2011;20(3):539–50.CrossRef
11.
go back to reference Bomben R, Gobessi S, Bo MD, Volinia S, Marconi D, Tissino E, Benedetti D, Zucchetto A, Rossi D, Gaidano G et al.: The miR-17 approximately 92 family regulates the response to toll-like receptor 9 triggering of CLL cells with unmutated IGHV genes. Leukemia 2012. Bomben R, Gobessi S, Bo MD, Volinia S, Marconi D, Tissino E, Benedetti D, Zucchetto A, Rossi D, Gaidano G et al.: The miR-17 approximately 92 family regulates the response to toll-like receptor 9 triggering of CLL cells with unmutated IGHV genes. Leukemia 2012.
12.
go back to reference 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):459–66.CrossRefPubMed 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):459–66.CrossRefPubMed
13.
go back to reference Roversi G, Pfundt R, Moroni RF, Magnani I, van Reijmersdal S, Pollo B, et al. Identification of novel genomic markers related to progression to glioblastoma through genomic profiling of 25 primary glioma cell lines. Oncogene. 2006;25(10):1571–83.CrossRefPubMed Roversi G, Pfundt R, Moroni RF, Magnani I, van Reijmersdal S, Pollo B, et al. Identification of novel genomic markers related to progression to glioblastoma through genomic profiling of 25 primary glioma cell lines. Oncogene. 2006;25(10):1571–83.CrossRefPubMed
14.
go back to reference Rozen S, Skaletsky H. Primer3 on the WWW for general users and for biologist programmers. Methods Mol Biol. 2000;132:365–86.PubMed Rozen S, Skaletsky H. Primer3 on the WWW for general users and for biologist programmers. Methods Mol Biol. 2000;132:365–86.PubMed
15.
go back to reference Ferrero GB, Howald C, Micale L, Biamino E, Augello B, Fusco C, et al. An atypical 7q11.23 deletion in a normal IQ Williams-Beuren syndrome patient. Eur J Hum Genet. 2010;18(1):33–8.CrossRefPubMed Ferrero GB, Howald C, Micale L, Biamino E, Augello B, Fusco C, et al. An atypical 7q11.23 deletion in a normal IQ Williams-Beuren syndrome patient. Eur J Hum Genet. 2010;18(1):33–8.CrossRefPubMed
16.
go back to reference Howald C, Merla G, Digilio MC, Amenta S, Lyle R, Deutsch S, et al. Two high throughput technologies to detect segmental aneuploidies identify new Williams-Beuren syndrome patients with atypical deletions. J Med Genet. 2006;43(3):266–73.CrossRefPubMed Howald C, Merla G, Digilio MC, Amenta S, Lyle R, Deutsch S, et al. Two high throughput technologies to detect segmental aneuploidies identify new Williams-Beuren syndrome patients with atypical deletions. J Med Genet. 2006;43(3):266–73.CrossRefPubMed
17.
go back to reference Micale L, Augello B, Fusco C, Selicorni A, Loviglio MN, Silengo MC, et al. Mutation spectrum of MLL2 in a cohort of Kabuki syndrome patients. Orphanet J Rare Dis. 2011;6:38.CrossRefPubMedPubMedCentral Micale L, Augello B, Fusco C, Selicorni A, Loviglio MN, Silengo MC, et al. Mutation spectrum of MLL2 in a cohort of Kabuki syndrome patients. Orphanet J Rare Dis. 2011;6:38.CrossRefPubMedPubMedCentral
18.
go back to reference Carra E, Barbieri F, Marubbi D, Pattarozzi A, Favoni RE, Florio T, et al. Sorafenib selectively depletes human glioblastoma tumor-initiating cells from primary cultures. Cell Cycle. 2013;12(3):491–500.CrossRefPubMedPubMedCentral Carra E, Barbieri F, Marubbi D, Pattarozzi A, Favoni RE, Florio T, et al. Sorafenib selectively depletes human glioblastoma tumor-initiating cells from primary cultures. Cell Cycle. 2013;12(3):491–500.CrossRefPubMedPubMedCentral
19.
go back to reference Network. CGAR: Comprehensive genomic characterization defines human glioblastoma genes and core pathways. Nature. 2008;455(7216):1061–8.CrossRef Network. CGAR: Comprehensive genomic characterization defines human glioblastoma genes and core pathways. Nature. 2008;455(7216):1061–8.CrossRef
20.
go back to reference Lu S, Wang S, Geng S, Ma S, Liang Z, Jiao B. Increased expression of microRNA-17 predicts poor prognosis in human glioma. J Biomed Biotechnol. 2012;2012:970761.PubMedPubMedCentral Lu S, Wang S, Geng S, Ma S, Liang Z, Jiao B. Increased expression of microRNA-17 predicts poor prognosis in human glioma. J Biomed Biotechnol. 2012;2012:970761.PubMedPubMedCentral
22.
go back to reference Micale L, Chaignat E, Fusco C, Reymond A, Merla G. The tripartite motif: structure and function. Adv Exp Med Biol. 2012;770:11–25.CrossRefPubMed Micale L, Chaignat E, Fusco C, Reymond A, Merla G. The tripartite motif: structure and function. Adv Exp Med Biol. 2012;770:11–25.CrossRefPubMed
23.
go back to reference Ryu YS, Lee Y, Lee KW, Hwang CY, Maeng JS, Kim JH, et al. TRIM32 protein sensitizes cells to tumor necrosis factor (TNFalpha)-induced apoptosis via its RING domain-dependent E3 ligase activity against X-linked inhibitor of apoptosis (XIAP). J Biol Chem. 2011;286(29):25729–38.CrossRefPubMedPubMedCentral Ryu YS, Lee Y, Lee KW, Hwang CY, Maeng JS, Kim JH, et al. TRIM32 protein sensitizes cells to tumor necrosis factor (TNFalpha)-induced apoptosis via its RING domain-dependent E3 ligase activity against X-linked inhibitor of apoptosis (XIAP). J Biol Chem. 2011;286(29):25729–38.CrossRefPubMedPubMedCentral
24.
go back to reference Hock A, Vousden KH. Regulation of the p53 pathway by ubiquitin and related proteins. Int J Biochem Cell Biol. 2010;42(10):1618–21.CrossRefPubMed Hock A, Vousden KH. Regulation of the p53 pathway by ubiquitin and related proteins. Int J Biochem Cell Biol. 2010;42(10):1618–21.CrossRefPubMed
Metadata
Title
TRIM8 downregulation in glioma affects cell proliferation and it is associated with patients survival
Authors
Lucia Micale
Carmela Fusco
Andrea Fontana
Raffaela Barbano
Bartolomeo Augello
Pasquelena De Nittis
Massimiliano Copetti
Maria Teresa Pellico
Barbara Mandriani
Dario Cocciadiferro
Paola Parrella
Vito Michele Fazio
Lucia Maria Cecilia Dimitri
Vincenzo D’Angelo
Chiara Novielli
Lidia Larizza
Antonio Daga
Giuseppe Merla
Publication date
01-12-2015
Publisher
BioMed Central
Published in
BMC Cancer / Issue 1/2015
Electronic ISSN: 1471-2407
DOI
https://doi.org/10.1186/s12885-015-1449-9

Other articles of this Issue 1/2015

BMC Cancer 1/2015 Go to the issue
Webinar | 19-02-2024 | 17:30 (CET)

Keynote webinar | Spotlight on antibody–drug conjugates in cancer

Antibody–drug conjugates (ADCs) are novel agents that have shown promise across multiple tumor types. Explore the current landscape of ADCs in breast and lung cancer with our experts, and gain insights into the mechanism of action, key clinical trials data, existing challenges, and future directions.

Dr. Véronique Diéras
Prof. Fabrice Barlesi
Developed by: Springer Medicine