Recurrent inactivation of STAG2 in bladder cancer is not associated with aneuploidy

Cristina Balbás-Martínez, Ana Sagrera, Enrique Carrillo-de-Santa-Pau, Julie Earl, Mirari Márquez, Miguel Vazquez, Eleonora Lapi, Francesc Castro-Giner, Sergi Beltran, Mònica Bayés, Alfredo Carrato, Juan C Cigudosa, Orlando Domínguez, Marta Gut, Jesús Herranz, Núria Juanpere, Manolis Kogevinas, Xavier Langa, Elena López-Knowles, José A Lorente, Josep Lloreta, David G Pisano, Laia Richart, Daniel Rico, Rocío N Salgado, Adonina Tardón, Stephen Chanock, Simon Heath, Alfonso Valencia, Ana Losada, Ivo Gut, Núria Malats, Francisco X Real, Cristina Balbás-Martínez, Ana Sagrera, Enrique Carrillo-de-Santa-Pau, Julie Earl, Mirari Márquez, Miguel Vazquez, Eleonora Lapi, Francesc Castro-Giner, Sergi Beltran, Mònica Bayés, Alfredo Carrato, Juan C Cigudosa, Orlando Domínguez, Marta Gut, Jesús Herranz, Núria Juanpere, Manolis Kogevinas, Xavier Langa, Elena López-Knowles, José A Lorente, Josep Lloreta, David G Pisano, Laia Richart, Daniel Rico, Rocío N Salgado, Adonina Tardón, Stephen Chanock, Simon Heath, Alfonso Valencia, Ana Losada, Ivo Gut, Núria Malats, Francisco X Real

Abstract

Urothelial bladder cancer (UBC) is heterogeneous at the clinical, pathological and genetic levels. Tumor invasiveness (T) and grade (G) are the main factors associated with outcome and determine patient management. A discovery exome sequencing screen (n = 17), followed by a prevalence screen (n = 60), identified new genes mutated in this tumor coding for proteins involved in chromatin modification (MLL2, ASXL2 and BPTF), cell division (STAG2, SMC1A and SMC1B) and DNA repair (ATM, ERCC2 and FANCA). STAG2, a subunit of cohesin, was significantly and commonly mutated or lost in UBC, mainly in tumors of low stage or grade, and its loss was associated with improved outcome. Loss of expression was often observed in chromosomally stable tumors, and STAG2 knockdown in bladder cancer cells did not increase aneuploidy. STAG2 reintroduction in non-expressing cells led to reduced colony formation. Our findings indicate that STAG2 is a new UBC tumor suppressor acting through mechanisms that are different from its role in preventing aneuploidy.

Figures

Figure 1
Figure 1
Distribution of single nucleotide variants identified in the discovery screen through exome sequencing: total (a), according to type of nucleotide substitution (b), and according to the predicted effect (c).
Figure 2
Figure 2
Distribution of mutations in genes recurrently mutated in UBC that are expressed in >30% of tumors; joint analysis of the discovery and prevalence screens. In 22/77 tumors (28.6%), none of the genes listed in this Figure was found to be mutated.
Figure 3
Figure 3
Immunohistochemical analysis of STAG2 expression in bladder tumors of different stage and grade: 2 of the tumors show lack of STAG2 expression (a, c) and one tumor shows strong expression (b). Of note the strong STAG2 protein expression in normal urothelium of a patient with a STAG2-negative tumor (a) and in the stroma of all tumor samples. (T, tumor; S, stroma; NU, normal urothelium). Scale bar: 200 µm.
Figure 4
Figure 4
STAG2 loss is not associated with aneuploidy in primary tumors and in 639V bladder cancer cells. Effects of STAG2 reconstitution on cell growth. (a) SNP array genomic plots show lack of chromosomal changes (aneuploidy) in a tumor lacking STAG2 expression (note strong STAG2 labeling of normal stroma). Scale bar: 200 µm. (b) Western blotting analysis of STAG2 in UBC lines shows undetectable expression in 4 of 11 lines used for functional studies. (c) STAG2 overexpression in UM-UC-6 cells leads to reduced colony formation efficiency. (d) Efficient STAG2 knockdown in 639V cells demonstrated by western blotting does not lead to consistent changes in chromosome number (quantification shown in Supplementary Table 12).
Figure 5
Figure 5
Kaplan-Meier plots of the association of STAG2 expression with outcome in patients with UBC. (a) Recurrence in patients with STAG2-high (n=309) vs. STAG2-low (n=171) NMIBC. (b) Progression in patients with STAG2-high (n=309) vs. STAG2-low (n=171) NMIBC. (c) Progression in patients with STAG2-high (n=158) vs. STAG2-low (n=24) MIBC. (d) Cancer-specific survival of patients with STAG2-high (n=158) vs. STAG2-low (n=24) MIBC. P-values correspond to the results of the multivariable analysis. Details on results and variables used for adjustment are shown in Supplementary Tables 17–20.

References

    1. Sylvester RJ. Natural history, recurrence, and progression in superficial bladder cancer. ScientificWorldJournal. 2006;6:2617–2625.
    1. Luis NM, Lopez-Knowles E, Real FX. Molecular biology of bladder cancer. Clin. Transl. Oncol. 2007;9:5–12.
    1. Hernández S, et al. FGFR3 mutations as a prognostic factor in non-muscle invasive urothelial bladder carcinomas: results of a prospective study. J. Clin. Oncol. 2006;24:3664–3671.
    1. López-Knowles E, et al. PIK3CA mutations are an early genetic alteration associated with FGFR3 mutations in superficial papillary bladder tumors. Cancer Res. 2006;66:7401–7404.
    1. Real FX. p53: it has it all, but will it make to the clinic as a marker in bladder cancer? J. Clin. Oncol. 2007;25:5341–5344.
    1. López-Knowles E, et al. The p53 pathway and outcome among patients with T1G3 bladder tumors. Clin. Cancer Res. 2006;12:6029–6036.
    1. Jebar AH, et al. FGFR3 and Ras gene mutations are mutually exclusive genetic events in urothelial cell carcinoma. Oncogene. 2005;24:5218–5225.
    1. Lindgren D, et al. Combined gene expression and genomic profiling define two intrinsic molecular subtypes of urothelial carcinoma and gene signatures for molecular grading and outcome. Cancer Res. 2010;70:3463–3472.
    1. Hoglund M. The bladder cancer genome; chromosomal changes as prognostic makers, opportunities, and obstacles. Urol. Oncol. 2012;30:533–540.
    1. Blaveri E, et al. Bladder cancer stage and outcome by array-based comparative genomic hybridization. Clin. Cancer Res. 2005;11:7012–7022.
    1. Gui Y, et al. Frequent mutations of chromatin remodeling genes in transitional cell carcinoma of the bladder. Nat. Genet. 2011;43:875–878.
    1. Fujimoto A, et al. Whole-genome sequencing of liver cancers identifies etiological influences on mutation patterns and recurrent mutations in chromatin regulators. Nat. Genet. 2012;44:760–764.
    1. Guichard C, et al. Integrated analysis of somatic mutations and focal copy-number changes identifies key genes and pathways in hepatocellular carcinoma. Nat. Genet. 2012;44:694–698.
    1. Varela I, et al. Exome sequencing identifies frequent mutation of the SWI/SNF complex gene PBRM1 in renal carcinoma. Nature. 2011;469:539–542.
    1. Wilson BG, Roberts CW. SWI/SNF nucleosome remodelers and cancer. Nat. Rev. Cancer. 2011;11:481–492.
    1. Solomon DA, et al. Mutational inactivation of STAG2 causes aneuploidy in human cancer. Science. 2011;333:1039–1043.
    1. Amaral AFS, et al. Plasma 25-hydroxyvitamin D3 levels and bladder cancer risk according to tumor stage and FGFR3 status: a mechanism-based epidemiological study. J. Natl. Cancer Inst. 2012;104:1897–1904.
    1. Welch JS, et al. The origin and evolution of mutations in acute myeloid leukemia. Cell. 2012;150:264–278.
    1. Walter MJ, et al. Acquired copy number alterations in adult acute myeloid leukemia genomes. Proc. Natl. Acad. Sci. USA. 2009;106:12950–12955.
    1. Walter MJ, et al. Clonal architecture of secondary acute myeloid leukemia. N. Eng. J. Med. 2012;366:1090–1098.
    1. Nasmyth K, Haering CH. Cohesin: its roles and mechanisms. Annu. Rev. Genet. 2009;43:525–558.
    1. Remeseiro S, Losada A. Cohesin, a chromatin engagement ring. Curr. Opin. Cell Biol. 2013;25:63–71.
    1. Remeseiro S, et al. Cohesin-SA1 deficiency drives aneuploidy and tumourigenesis in mice due to impaired replication of telomeres. EMBO J. 2012;31:2076–2089.
    1. Remeseiro S, et al. A unique role of cohesin-SA1 in gene regulation and development. EMBO J. 2012;31:2090–2102.
    1. Shain AH, et al. Convergent structural alterations define SWItch/Sucrose NonFermentable (SWI/SNF) chromatin remodeler as a central tumor suppressive complex in pancreatic cancer. Proc. Natl. Acad. Sci. USA. 2012;109:E252–E259.
    1. Balbás-Martínez C, et al. ARID1A alterations are associated with FGFR3 wild type, poor-prognosis, urothelial bladder tumors. PLoS ONE. 2013;8:e62483.
    1. Amaral AFS, et al. Plasma 25-hydroxyvitamin D3 levels and bladder cancer risk according to tumor stage and FGFR3 status: a mechanism-based epidemiological study. J. Natl. Cancer Inst. 2012;104:1897–1904.
    1. Reva B, et al. Predicting the functional impact of protein mutations: application to cancer genomics. Nucleic Acids Res. 2011;39:e118.
    1. Kumar P, et al. Predicting the effects of coding non-synonymous variants on protein function using the SIFT algorithm. Nat. Protoc. 2009;4:1073–1081.
    1. Gonzalez A, et al. Functional impact bias reveals cancer drivers. Nucleic Acids Res. 2012;40:e169.
    1. Vazquez M, et al. Chapter 14: cancer genome analysis. PLoS Comput. Biol. 2012;8:e1002824.
    1. Quesada V, et al. Exome sequencing identifies recurrent mutations of the splicing factor SF3B1 gene in chronic lymphocytic leukemia. Nat. Genet. 2011;44:47–52.
    1. López-Knowles E, et al. The p53 pathway and outcome among patients with T1G3 bladder tumors. Clin. Cancer Res. 2006;12:6029–6036.
    1. Rodríguez-Santiago B, et al. Mosaic uniparental disomies and aneuploidies as large structural variants of the human genome. Am. J. Hum. Genet. 2010;87:129–138.
    1. Snijders AM, et al. Assembly of microarrays for genome-wide measurement of DNA copy number. Nat. Genet. 2001;29:263–264.
    1. Blaveri E, et al. Bladder cancer stage and outcome by array-based comparative genomic hybridization. Clin. Cancer Res. 2005;11:7012–7022.

Source: PubMed

Подписаться