Skip to main content
Brain Pathology logoLink to Brain Pathology
. 2013 Feb 25;23(2):200–205. doi: 10.1111/bpa.12021

Rhabdoid Tumors: An Initial Clue to the Role of Chromatin Remodeling in Cancer

Ryan S Lee 1,2,3, Charles W M Roberts 1,2,3
PMCID: PMC8029057  PMID: 23432645

Abstract

The discovery of biallelic, inactivating SMARCB1 mutations in rhabdoid tumors (RTs) over a decade ago represented the first recognized link between chromatin remodeling and tumor suppression. SMARCB1 is a core subunit of the SWI/SNF chromatin remodeling complex, and the recent emergence of frequent mutations in genes that encode subunits of this complex across a wide variety of cancers suggests that perturbation of this chromatin remodeling complex constitutes a key driver of cancer formation. Despite the highly aggressive nature of RTs, they are genetically simple cancers that appear to lack chromosomal instability and contain very few mutations. Indeed, the mutation rate in RTs is among the lowest of all cancers sequenced, with loss of SMARCB1 as essentially the sole recurrent event. Given the genetic simplicity of this disease, understanding the chromatin dysregulation caused by SMARCB1 loss may provide more general insight into how epigenetic alterations can contribute to oncogenic transformation and may reveal opportunities for targeted therapy not only of RT but also the variety of other SWI/SNF mutant cancers.

Keywords: AT/RT, cancer genomics, epigenetics, rhabdoid, SMARCB1, SWI/SNF

Chromatin and Cancer

History has revealed a theme that genes mutated in early onset pediatric cancers and familial cancer predisposition syndromes are often of fundamental importance to cancer in general. For example, retinoblastoma is a rare pediatric cancer but served as the initial clue to the importance of the RB gene in tumor suppression. In 1998, the causative mutation of rhabdoid tumors (RTs) was identified as biallelic inactivation of the gene SMARCB1 (SNF5/INI1/BAF47) 5, 6, 71. SMARCB1 encodes a core subunit of SWI/SNF chromatin remodeling complexes 53, 76. This finding represented the first identified link between ATPase chromatin remodeling complexes and tumor suppression—a link that has since blossomed with findings of SWI/SNF subunit mutations in many types of cancer, both pediatric and adult. Indeed, recent cancer genome sequencing efforts have further highlighted the role of chromatin regulators in cancer as a variety of genes whose products covalently modify histones and chromatin have also been found to be mutant in cancer. Here we review and discuss RT and describe mechanistic and potential therapeutic insights that have resulted from subsequent studies.

RTs are aggressive pediatric cancers that arise most frequently in the brain, where they are referred to as atypical teratoid/rhabdoid tumor (AT/RT), and the kidney, but can also arise in soft tissues throughout the body 3, 8, 59, 78. These tumors tend to have a very early onset, with roughly half of cases arising in children less than 1 year of age and the large majority of cases occurring in children under the age of 3 3. These tumors are highly malignant and confer a particularly poor prognosis, although the recent use of highly intensive multimodality therapeutic regimens may offer some improvement in outcome 14. Although an origin from neural progenitors has been hypothesized, analyses of gene expression have yielded conflicting conclusions, and the cell(s) of origin remains unclear 11, 20.

After cytogenetic analysis identified the deletion of chromosome 22q11.2 region as the only frequent copy number change in RT cell lines and primary tumors, further mapping showed that the gene SMARCB1 was almost universally missing in these cancers 4, 6, 31, 71. These findings provided the first clear evidence that AT/RT, renal RT and soft‐tissue RT are all genetically related 6, 64. Analogous to retinoblastoma, germ line mutations in SMARCB1 strongly predispose to RTs and the rare cases of familial rhabdoid predisposition syndrome are predominantly due to inherited heterozygous mutations in SMARCB1 65, 68. Indeed, biallelic loss of SMARCB1, particularly as evidenced by negative immunostaining with the INI1 antibody, is now used in the diagnosis of RT 3. It should be noted, however, that in a very small percentage of RT cases, the tumors lack SMARCB1 mutations but carry mutations in another SWI/SNF subunit, SMARCA4 (BRG1/SNF2‐beta) 24, 61, further establishing mutation of the SWI/SNF complex as a signature event of RT.

SWI/SNF Complex Subunits are Frequently and Widely Mutated in Cancer

SWI/SNF complexes were originally characterized in yeast as factors being essential for mating type switching (SWI) or for sucrose metabolism (mutants were sucrose non‐fermenting, SNF). Mammalian versions of these complexes consist of 10–12 subunits and are transcriptional modulators that possess ATP‐dependent nucleosome remodeling activity 9, 16. SWI/SNF complexes are comprised of one of two ATPase subunits (SMARCA4 or SMARCA2/BRM), a set of highly conserved “core” subunits (SMARCB1, SMARCC1/BAF155 and SMARCC2/BAF170), and variant subunits thought to contribute to lineage‐specific functions of the complexes 26, 27, 36, 51, 53, 75. While SMARCB1 is constitutively present in SWI/SNF complexes, its biochemical contribution remains unclear.

Although SWI/SNF complexes were first linked to cancer when SMARCB1 was found to be recurrently mutated in RT, recent findings from cancer genome sequencing studies reveal that at least seven SWI/SNF subunits are mutated, often at high frequency, across a wide variety of cancers (Table 1) 82. For example, the ARID1A (BAF250a) subunit is mutated in 50% of ovarian clear cell carcinomas, 30% of endometrioid carcinomas, 27% of gastric cancers, 13% of bladder carcinomas, 10% of colorectal and lung adenocarcinomas, 10% of hepatocellular carcinomas, and occasionally in breast, pancreatic, and prostate cancers and neuroblastomas 10, 15, 19, 29, 30, 34, 35, 54, 62, 66, 80, 84. The PBRM1 (BAF180) subunit is mutated in 40% of renal carcinomas, as well as occasionally in breast, gastric and pancreatic cancers 18, 52, 66, 70, 85. The BRD7 subunit is lost in up to 20% of p53 wild‐type breast cancers 17, and the ARID2 (BAF200) subunit is mutated in 18% of hepatitis C‐associated hepatocellular carcinomas, 7% of melanomas, and occasionally in some pancreatic cancers 19, 28, 43, 46. The SMARCA4 subunit is mutated in 35% of non‐small cell lung cancers and in medulloblastomas, particularly the WNT (26%) and group 3 (11%) subtypes 44, 49, 50, 54, 55, 58. Finally, mutations in SWI/SNF subunits were identified in one‐third of pancreatic cancers 2, 66. It remains unclear why different SWI/SNF subunits are mutated in different cancers, although it raises the possibility of lineage‐specific contributions of individual subunits. Collectively, a broad role for the SWI/SNF complex as a tumor suppressor has recently emerged, suggesting substantial relevance for this complex in tumor suppression beyond RT.

Table 1.

SWI/SNF subunits are mutated in a wide variety of cancers. Selected studies revealing SWI/SNF subunit mutations in cancer

Common SWI/SNF subunit mutations discovered by cancer genome sequencing
SWI/SNF complex subunit Cancer type Cell line or primary tumor Mutation type % Mutated Reference
SMARCB1 Rhabdoid tumor Primary tumors Deletions and copy neutral loss of heterozygosity 94 25
Primary tumors Homozygous inactivation: deletions, nonsense and other null mutations 100 42
Primary tumors Homozygous inactivation: deletions, nonsense and other null mutations 98 31
SMARCA4 Medulloblastoma Primary tumors Missense mutations 4 54
Primary tumors Null and missense mutations 5 33
Lung adenocarcinoma Both Unknown point mutation 32 44
Primary tumors Null and missense mutations 11 30
Pancreatic ductal adenocarcinoma Both Heterozygous deletions 9.6 66
SMARCA2 Pancreatic ductal adenocarcinoma Both Heterozygous deletions 2.6 66
PBRM1 Renal clear cell carcinoma Primary tumors Null mutations 53 52
Both Null mutations 60 18
Primary tumors Null and missense mutations 21 23
Primary tumors Truncating mutations 41 70
Pancreatic ductal adenocarcinoma Both Null mutations and deletions 9.6 66
Cell lines Truncating mutations 27 74
ARID2 Hepatocellular carcinoma Primary tumors Indel, nonsense and null mutations 5.8 19
Primary tumors Truncating mutations 14–18 43
Non‐small cell lung carcinoma Both Homozygous deletions, null mutations 7.3 46
Pancreatic ductal adenocarcinoma Primary tumors Null and missense mutations, copy number changes 12 2
Melanoma Primary tumors Null and missense mutations 9 28
ARID1A Ovarian clear cell carcinoma Both Null and missense mutations 46 80
Primary tumors Null mutations 57 35
Endometrioid carcinoma Primary tumors Null and missense mutations 30 80
Gastric adenocarcinoma Primary tumors Null mutations 8 86
Both Indel, missense and nonsense mutations 10 34
Primary tumors Truncating mutations 29 73
Bladder transitional cell carcinoma Primary tumors Null and missense mutations 13 21
Colorectal carcinoma Primary tumors Indel and nonsense mutations 10 10
Both Indel and nonsense mutations 10 34
Hepatocellular carcinoma Primary tumors Indel, missense and nonsense mutations 13 29
Primary tumors Indel, missense and nonsense mutations 10 19
Pancreatic ductal adenocarcinoma Primary tumors Indel and nonsense mutations 10 2
Both Null mutations and deletions 8.3 66
Both Indel and nonsense mutations 8 34
Neuroblastoma Primary tumors Null and missense mutations 6 60
Lung adenocarcinoma Primary tumors Indel, missense and nonsense mutations 8 30
Both Indels 2 34
Breast cancer Primary tumors Indel, missense and nonsense mutations 4 67
Primary tumors Deletions (point mutations) 37 (3) 15
Both Indel, missense and nonsense mutations 4 34
Primary tumors Deletion 13 45
Prostate cancer Both Indels 8.7 34
ARID1B Neuroblastoma Primary tumors Deletions, null and missense mutations 7 60
Breast cancer Primary tumors Indel, missense and nonsense mutations 5 67
Pancreatic ductal adenocarcinoma Both Null mutations and deletions 3.9 66
Hepatocellular carcinoma Primary tumors Indel, missense and nonsense mutations 6.7 19

RT: Remarkably Simple Genomes

Since first being linked to RT, studies have sought to identify additional genetic mechanisms and mutations that cooperate with SMARCB1 loss to drive RT. While SMARCB1 was initially implicated in DNA repair 72, a subsequent evaluation found no clear role and further found that 16 out of 18 primary RT samples were diploid and indistinguishable from normal peripheral blood DNA by SNP array, other than SMARCB1 loss, suggesting an unusual degree of genomic stability in RT 47. To characterize the genomes of RT and search for genetic mutations that cooperate with SMARCB1 loss to drive RT formation, SNP analysis and exome sequencing were recently performed on 32 frozen primary RT samples paired with matched normal genomic DNA. RT genomes were found to be remarkably simple with no recurrent copy number variations other than focal deletions of the SMARCB1 locus or monosomy of chromosome 22, where SMARCB1 is located 42, 47. Whole exome sequencing revealed a particularly low mutation rate, with an average of four non‐synonymous somatic mutations detected per tumor exome 42. This mutation rate is among the lowest of all sequenced cancer genomes, and indeed two tumors contained no somatic mutations other than biallelic loss of SMARCB1. Across the 32 samples, SMARCB1 was the only gene found to have significant recurrent mutations. Two additional studies focused upon known cancer‐related genes also found an essential absence of cooperating mutations 25, 39. Collectively, these data raise the possibility that, at least within the protein coding exome, SMARCB1 loss might be the sole genetic driving event in RT, although it remains possible that germ line variants or mutations located outside the exome could be required to cooperate with SMARCB1 loss to drive RT tumor formation.

The surprisingly low mutation rate seen in RT suggests that widespread genomic instability might not be a necessary hallmark for the genesis of even highly aggressive cancers such as RT. Rather, at least in certain contexts, perhaps only a few, or one, genetic perturbation can be sufficient to drive cancer formation. Other pediatric cancers such as infant leukemia, neuroblastoma and retinoblastoma have also been found to possess mutation rates several log‐fold lower than highly mutagenic adult cancers such as melanoma 28, 60, 87. Some adult cancers, such as acute myeloid leukemia (AML), have also been found to have low mutation rates, and occasional individual cases of most common cancer types also have very low mutation rates 79. While 5–15 somatic driver mutations had been predicted as being essential for the genesis of cancer 7, it now appears that substantially lower numbers are required, at least in some contexts. But how could a single genetic lesion be sufficient for cancer? The mathematics would seem to suggest, were this true, that everyone would develop cancer at a young age. This raises the possibility that cooperating events, rather than being genetic mutations, might occur as epigenetic chromatin alterations.

Functional Insights into SMARCB1

Insights come from studies regarding the normal function of SMARCB1 and the mechanisms by which its loss drives cancer formation. Several mouse models of Smarcb1 inactivation have been developed 22, 40, 56, 57, 69. Homozygous deletion of Smarcb1 results in early embryonic lethality while heterozygous knockout mice develop tumors comparable to human RT 22, 40, 56. Analysis revealed that these tumors have spontaneously lost the second Smarcb1 allele, a requirement that likely explains the 30% penetrance and 11‐month median onset. In comparison, in conditional mice in which Smarcb1 is inducibly inactivated in some cells, 100% of mice develop lymphomas or RTs with a median onset of only 11 weeks, markedly faster than that occurs following inactivation of tumor suppressors such as p53, p16 or p14 37, 57, 63, 81. Of note, as cancers driven by Smarcb1 loss occur in multiple genetic backgrounds, this could suggest that a specific germ line variant is not essential for the genesis of cancers driven by Smarcb1 loss. Collectively, these experiments demonstrated a potent and bona fide role for Smarcb1 as a tumor suppressor.

Providing important insight into transformation driven by Smarcb1 loss, while Smarcb1 inactivation results in rapid cancer onset, its loss is detrimental to the vast majority of cells. Inactivation in embryonic stem cells blocks development and causes arrest at the blastocyst stage 40. Deletion of Smarcb1 in primary fibroblasts results in cell cycle arrest and a block in proliferation 41. Indeed, even in a T‐cell lymphoma model where Smarcb1 deletion in vivo results in rapid cancer onset in all mice 57, the vast majority of T cells do not tolerate Smarcb1 loss, resulting in markedly reduced T‐cell numbers in these animals. From this aberrant T‐cell environment, cancer arises like a phoenix from the ashes. As the human tumor data suggest that this phenotypic variation is not explained by the acquisition of genetic mutations, epigenetic differences and alterations likely play a key role in determining susceptibility to transformation upon SMARCB1 loss. Notably, while SWI/SNF subunit mutations are collectively found in a wide variety of cancers, SMARCB1 loss itself has only been associated with RT and a few other rare cancers—it is not generally mutated in common adult cancers. In mouse lymphomas, Smarcb1 loss drives lymphoma formation from mature CD8+ memory‐like T cells, but not from immature T cells or B cells 77. Collectively, this suggests that only certain cell types, or epigenetic states, are susceptible to SMARCB1 loss. Other cell types/states might be susceptible to the loss of other specific SWI/SNF subunits.

Alterations of the epigenetic landscape caused by SMARCB1 loss affect several pathways that might be important in the genesis of RT in susceptible cell types. Studies of mouse models and human tumors revealed the existence of epigenetic antagonism between SMARCB1 and the Polycomb repressor complex subunit EZH2, such that the loss of SMARCB1 results in unopposed activity of EZH2 that serves an essential role in tumor formation 38, 83. In addition, the loss of SMARCB1 in RT has been found to have effects upon a wide spectrum of cellular processes. SMARCB1 and SWI/SNF complexes appear to modulate the cell cycle through transcriptional regulation of the p16INK4a‐cyclinD/CDK4‐pRb‐E2F mitotic checkpoint 1, 13, 72. Similarly, the absence of SMARCB1 increases cell motility by posttranslational deregulation of RhoA, possibly through epigenetically driven changes in expression of G‐protein guanine exchange factors and activating proteins 12. These various effects might be modulated by adaptor proteins such as BIN1 48. Evidence has also linked SMARCB1 directly to GLI1, an effector in the hedgehog signaling pathway 32. While it remains unknown whether these same pathways are activated in cancers linked to mutations in other SWI/SNF subunits, the understanding gleaned from studying RT suggests that SMARCB1 loss results in the deregulation of multiple pathways that contribute to oncogenesis in a lineage‐specific fashion.

Understanding the mechanisms by which epigenetic changes contribute to oncogenesis offers the promise of informing development of therapeutics. Recent sequencing efforts have revealed a theme that genes encoding chromatin modifiers, not just SWI/SNF, are recurrently mutated across a wide spectrum of cancer types, suggesting that changes in chromatin structure might be important in oncogenesis. Indeed, epigenetic alterations have been implicated in many cancer types, although it has often been challenging to define potential epigenetic contributions when they occur in the setting of high mutation rates and genomic instability. Perhaps in part due to a low incidence of age‐related and environmentally induced passenger mutations, early onset pediatric cancers such as RT often have simpler genomes. Consequently, these cancers might constitute outstanding models with which to mechanistically characterize the roles of chromatin‐based changes in driving cancer growth and, potentially, with which to identify specific mutation‐driven therapeutic susceptibilities.

References

  • 1. Betz BL, Strobeck MW, Reisman DN, Knudsen ES, Weissman BE (2002) Re‐expression of hSNF5/INI1/BAF47 in pediatric tumor cells leads to G1 arrest associated with induction of p16ink4a and activation of RB. Oncogene 21:5193–5203. [DOI] [PubMed] [Google Scholar]
  • 2. Biankin AV, Waddell N, Kassahn KS, Gingras M‐C, Muthuswamy LB, Johns AL et al (2012) Pancreatic cancer genomes reveal aberrations in axon guidance pathway genes. Nature 491:399–405. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3. Biegel JA, Kalpana G, Knudsen ES, Packer RJ, Roberts CWM, Thiele CJ et al (2002) The role of INI1 and the SWI/SNF complex in the development of rhabdoid tumors: meeting summary from the workshop on childhood atypical teratoid/rhabdoid tumors. Cancer Res 62:323–328. [PubMed] [Google Scholar]
  • 4. Biegel JA, Rorke LB, Packer RJ, Emanuel BS (1990) Monosomy 22 in rhabdoid or atypical tumors of the brain. J Neurosurg 73:710–714. [DOI] [PubMed] [Google Scholar]
  • 5. Biegel JA, Tan L, Zhang F, Wainwright L, Russo P, Rorke LB (2002) Alterations of the hSNF5/INI1 gene in central nervous system atypical teratoid/rhabdoid tumors and renal and extrarenal rhabdoid tumors. Clin Cancer Res 8:3461–3467. [PubMed] [Google Scholar]
  • 6. Biegel JA, Zhou JY, Rorke LB, Stenstrom C, Wainwright LM, Fogelgren B (1999) Germ‐line and acquired mutations of INI1 in atypical teratoid and rhabdoid tumors. Cancer Res 59:74–79. [PubMed] [Google Scholar]
  • 7. Bozic I, Antal T, Ohtsuki H, Carter H, Kim D, Chen S et al (2010) Accumulation of driver and passenger mutations during tumor progression. Proc Natl Acad Sci USA 107:18545–18550. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8. Burger PC, Yu IT, Tihan T, Friedman HS, Strother DR, Kepner JL et al (1998) Atypical teratoid/rhabdoid tumor of the central nervous system: a highly malignant tumor of infancy and childhood frequently mistaken for medulloblastoma: a Pediatric Oncology Group study. Am J Surg Pathol 22:1083–1092. [DOI] [PubMed] [Google Scholar]
  • 9. Cairns BR, Kim YJ, Sayre MH, Laurent BC, Kornberg RD (1994) A multisubunit complex containing the SWI1/ADR6, SWI2/SNF2, SWI3, SNF5, and SNF6 gene products isolated from yeast. Proc Natl Acad Sci USA 91:1950–1954. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10. Cancer Genome Atlas Network (2012) Comprehensive molecular characterization of human colon and rectal cancer. Nature 487:330–337. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11. Caramel J, Medjkane S, Quignon F, Delattre O (2008) The requirement for SNF5/INI1 in adipocyte differentiation highlights new features of malignant rhabdoid tumors. Oncogene 27:2035–2044. [DOI] [PubMed] [Google Scholar]
  • 12. Caramel J, Quignon F, Delattre O (2008) RhoA‐dependent regulation of cell migration by the tumor suppressor hSNF5/INI1. Cancer Res 68:6154–6161. [DOI] [PubMed] [Google Scholar]
  • 13. Chai J, Charboneau AL, Betz BL, Weissman BE (2005) Loss of the hSNF5 gene concomitantly inactivates p21CIP/WAF1 and p16INK4a activity associated with replicative senescence in A204 rhabdoid tumor cells. Cancer Res 65:10192–10198. [DOI] [PubMed] [Google Scholar]
  • 14. Chi SN, Zimmerman MA, Yao X, Cohen KJ, Burger P, Biegel JA et al (2009) Intensive multimodality treatment for children with newly diagnosed CNS atypical teratoid rhabdoid tumor. J Clin Oncol 27:385–389. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15. Cornen S, Adelaide J, Bertucci F, Finetti P, Guille A, Birnbaum DJ et al (2012) Mutations and deletions of ARID1A in breast tumors. Oncogene 31:4255–4256. [DOI] [PubMed] [Google Scholar]
  • 16. Côté J, Quinn J, Workman JL, Peterson CL (1994) Stimulation of GAL4 derivative binding to nucleosomal DNA by the yeast SWI/SNF complex. Science 265:53–60. [DOI] [PubMed] [Google Scholar]
  • 17. Drost J, Mantovani F, Tocco F, Elkon R, Comel A, Holstege H et al (2010) BRD7 is a candidate tumour suppressor gene required for p53 function. Nat Cell Biol 12:380–389. [DOI] [PubMed] [Google Scholar]
  • 18. Duns G, Hofstra RMW, Sietzema JG, van Hollema H, Duivenbode I, Kuik A et al (2012) Targeted exome sequencing in clear cell renal cell carcinoma tumors suggests aberrant chromatin regulation as a crucial step in ccRCC development. Hum Mutat 33:1059–1062. [DOI] [PubMed] [Google Scholar]
  • 19. Fujimoto A, Totoki Y, Abe T, Boroevich KA, Hosoda F, Nguyen HH et al (2012) Whole‐genome sequencing of liver cancers identifies etiological influences on mutation patterns and recurrent mutations in chromatin regulators. Nat Genet 44:760–764. [DOI] [PubMed] [Google Scholar]
  • 20. Gadd S, Sredni ST, Huang C‐C, Perlman EJ; Group RTCOTCO (2010) Rhabdoid tumor: gene expression clues to pathogenesis and potential therapeutic targets. Lab Invest 90:724–738. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21. Gui Y, Guo G, Huang Y, Hu X, Tang A, Gao S et al (2011) Frequent mutations of chromatin remodeling genes in transitional cell carcinoma of the bladder. Nat Genet 43:875–878. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22. Guidi CJ, Sands AT, Zambrowicz BP, Turner TK, Demers DA, Webster W et al (2001) Disruption of Ini1 leads to peri‐implantation lethality and tumorigenesis in mice. Mol Cell Biol 21:3598–3603. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23. Guo G, Gui Y, Gao S, Tang A, Hu X, Huang Y et al (2012) Frequent mutations of genes encoding ubiquitin‐mediated proteolysis pathway components in clear cell renal cell carcinoma. Nature Genetics 44:17–19. [DOI] [PubMed] [Google Scholar]
  • 24. Hasselblatt M, Gesk S, Oyen F, Rossi S, Viscardi E, Giangaspero F et al (2011) Nonsense mutation and inactivation of SMARCA4 (BRG1) in an atypical teratoid/rhabdoid tumor showing retained SMARCB1 (INI1) expression. Am J Surg Pathol 35:933–935. [DOI] [PubMed] [Google Scholar]
  • 25. Hasselblatt M, Isken S, Linge A, Eikmeier K, Jeibmann A, Oyen F et al (2012) High‐resolution genomic analysis suggests the absence of recurrent genomic alterations other than SMARCB1 aberrations in atypical teratoid/rhabdoid tumors. Genes Chromosomes Cancer 52:185–190. [DOI] [PubMed] [Google Scholar]
  • 26. Ho L, Jothi R, Ronan JL, Cui K, Zhao K, Crabtree GR (2009) An embryonic stem cell chromatin remodeling complex, esBAF, is an essential component of the core pluripotency transcriptional network. Proc Natl Acad Sci USA 106:5187–5191. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27. Ho L, Ronan JL, Wu J, Staahl BT, Chen L, Kuo A et al (2009) An embryonic stem cell chromatin remodeling complex, esBAF, is essential for embryonic stem cell self‐renewal and pluripotency. Proc Natl Acad Sci USA 106:5181–5186. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28. Hodis E, Watson IR, Kryukov GV, Arold ST, Imielinski M, Theurillat J‐P et al (2012) A landscape of driver mutations in melanoma. Cell 150:251–263. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29. Huang J, Deng Q, Wang Q, Li K‐Y, Dai J‐H, Li N et al (2012) Exome sequencing of hepatitis B virus‐associated hepatocellular carcinoma. Nat Genet 44:1117–1121. [DOI] [PubMed] [Google Scholar]
  • 30. Imielinski M, Berger AH, Hammerman PS, Hernandez B, Pugh TJ, Hodis E et al (2012) Mapping the hallmarks of lung adenocarcinoma with massively parallel sequencing. Cell 150:1107–1120. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31. Jackson EM, Sievert AJ, Gai X, Hakonarson H, Judkins AR, Tooke L et al (2009) Genomic analysis using high‐density single nucleotide polymorphism‐based oligonucleotide arrays and multiplex ligation‐dependent probe amplification provides a comprehensive analysis of INI1/SMARCB1 in malignant rhabdoid tumors. Clin Cancer Res 15:1923–1930. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32. Jagani Z, Mora‐Blanco EL, Sansam CG, McKenna ES, Wilson B, Chen D et al (2010) Loss of the tumor suppressor Snf5 leads to aberrant activation of the Hedgehog‐Gli pathway. Nat Med 16:1429–1433. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33. Jones DTW, Jäger N, Kool M, Zichner T, Hutter B, Sultan M et al (2012) Dissecting the genomic complexity underlying medulloblastoma. Nature 488:100–105. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34. Jones S, Li M, Parsons DW, Zhang X, Wesseling J, Kristel P et al (2012) Somatic mutations in the chromatin remodeling gene ARID1A occur in several tumor types. Hum Mutat 33:100–103. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35. Jones S, Wang T‐L, Shih I‐M, Mao T‐L, Nakayama K, Roden R et al (2010) Frequent mutations of chromatin remodeling gene ARID1A in ovarian clear cell carcinoma. Science 330:228–231. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36. Kaeser MD, Aslanian A, Dong M‐Q, Yates JR, Emerson BM (2008) BRD7, a novel PBAF‐specific SWI/SNF subunit, is required for target gene activation and repression in embryonic stem cells. J Biol Chem 283:32254–32263. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37. Kamijo T, Zindy F, Roussel MF, Quelle DE, Downing JR, Ashmun RA et al (1997) Tumor suppression at the mouse INK4a locus mediated by the alternative reading frame product p19ARF. Cell 91:649–659. [DOI] [PubMed] [Google Scholar]
  • 38. Kia SK, Gorski MM, Giannakopoulos S, Verrijzer CP (2008) SWI/SNF mediates polycomb eviction and epigenetic reprogramming of the INK4b‐ARF‐INK4a locus. Mol Cell Biol 28:3457–3464. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39. Kieran MW, Roberts CWM, Chi SN, Ligon KL, Rich BE, Macconaill LE et al (2012) Absence of oncogenic canonical pathway mutations in aggressive pediatric rhabdoid tumors. Pediatr Blood Cancer 59:1155–1157. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40. Klochendler‐Yeivin A, Fiette L, Barra J, Muchardt C, Babinet C, Yaniv M (2000) The murine SNF5/INI1 chromatin remodeling factor is essential for embryonic development and tumor suppression. EMBO Rep 1:500–506. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41. Klochendler‐Yeivin A, Picarsky E, Yaniv M (2006) Increased DNA damage sensitivity and apoptosis in cells lacking the Snf5/Ini1 subunit of the SWI/SNF chromatin remodeling complex. Mol Cell Biol 26:2661–2674. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42. Lee RS, Stewart C, Carter SL, Ambrogio L, Cibulskis K, Sougnez C et al (2012) A remarkably simple genome underlies highly malignant pediatric rhabdoid cancers. J Clin Invest 122:2983–2988. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43. Li M, Zhao H, Zhang X, Wood LD, Anders RA, Choti MA et al (2011) Inactivating mutations of the chromatin remodeling gene ARID2 in hepatocellular carcinoma. Nat Genet 43:828–829. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44. Liu J, Lee W, Jiang Z, Chen Z, Jhunjhunwala S, Haverty PM et al (2012) Genome and transcriptome sequencing of lung cancers reveal diverse mutational and splicing events. Genome Res 22:2315–2327. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45. Mamo A, Cavallone L, Tuzmen S, Chabot C, Ferrario C, Hassan S et al (2012) An integrated genomic approach identifies ARID1A as a candidate tumor‐suppressor gene in breast cancer. Oncogene 31:2090–2100. [DOI] [PubMed] [Google Scholar]
  • 46. Manceau G, Letouzé E, Guichard C, Didelot A, Cazes A, Corté H et al (2012) Recurrent inactivating mutations of ARID2 in non‐small cell lung carcinoma. Int J Cancer [Epub ahead of print; doi:10.1002/ijc.27900]. [DOI] [PubMed] [Google Scholar]
  • 47. McKenna ES, Sansam CG, Cho Y‐J, Greulich H, Evans JA, Thom CS et al (2008) Loss of the epigenetic tumor suppressor SNF5 leads to cancer without genomic instability. Mol Cell Biol 28:6223–6233. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48. McKenna ES, Tamayo P, Cho Y‐J, Tillman EJ, Mora‐Blanco EL, Sansam CG et al (2012) Epigenetic inactivation of the tumor suppressor BIN1 drives proliferation of SNF5‐deficient tumors. Cell Cycle 11:1956–1965. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49. Medina PP, Romero OA, Kohno T, Montuenga LM, Pio R, Yokota J et al (2008) Frequent BRG1/SMARCA4‐inactivating mutations in human lung cancer cell lines. Hum Mutat 29:617–622. [DOI] [PubMed] [Google Scholar]
  • 50. Northcott PA, Jones DTW, Kool M, Robinson GW, Gilbertson RJ, Cho Y‐J et al (2012) Medulloblastomics: the end of the beginning. Nat Rev Cancer 12:818–834. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51. Olave I, Wang W, Xue Y, Kuo A, Crabtree GR (2002) Identification of a polymorphic, neuron‐specific chromatin remodeling complex. Genes Dev 16:2509–2517. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52. Peña‐Llopis S, Vega‐Rubín‐de‐Celis S, Liao A, Leng N, Pavía‐Jiménez A, Wang S et al (2012) BAP1 loss defines a new class of renal cell carcinoma. Nat Genet 44:751–759. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53. Phelan ML, Sif S, Narlikar GJ, Kingston RE (1999) Reconstitution of a core chromatin remodeling complex from SWI/SNF subunits. Mol Cell 3:247–253. [DOI] [PubMed] [Google Scholar]
  • 54. Pugh TJ, Weeraratne SD, Archer TC, Pomeranz Krummel DA, Auclair D, Bochicchio J et al (2012) Medulloblastoma exome sequencing uncovers subtype‐specific somatic mutations. Nature 488:106–110. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55. Reisman DN, Sciarrotta J, Wang W, Funkhouser WK, Weissman BE (2003) Loss of BRG1/BRM in human lung cancer cell lines and primary lung cancers: correlation with poor prognosis. Cancer Res 63:560–566. [PubMed] [Google Scholar]
  • 56. Roberts CW, Galusha SA, McMenamin ME, Fletcher CD, Orkin SH (2000) Haploinsufficiency of Snf5 (integrase interactor 1) predisposes to malignant rhabdoid tumors in mice. Proc Natl Acad Sci USA 97:13796–13800. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57. Roberts CWM, Leroux MM, Fleming MD, Orkin SH (2002) Highly penetrant, rapid tumorigenesis through conditional inversion of the tumor suppressor gene Snf5. Cancer Cell 2:415–425. [DOI] [PubMed] [Google Scholar]
  • 58. Rodriguez‐Nieto S, Cañada A, Pros E, Pinto AI, Torres‐Lanzas J, Lopez‐Rios F et al (2010) Massive parallel DNA pyrosequencing analysis of the tumor suppressor BRG1/SMARCA4 in lung primary tumors. Hum Mutat 32:E1999–E2017. [DOI] [PubMed] [Google Scholar]
  • 59. Rorke LB, Packer RJ, Biegel JA (1996) Central nervous system atypical teratoid/rhabdoid tumors of infancy and childhood: definition of an entity. J Neurosurg 85:56–65. [DOI] [PubMed] [Google Scholar]
  • 60. Sausen M, Leary RJ, Jones SAN, Wu J, Reynolds CP, Liu X et al (2012) Integrated genomic analyses identify ARID1A and ARID1B alterations in the childhood cancer neuroblastoma. Nat Genet 45:12–17. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61. Schneppenheim R, Frühwald MC, Gesk S, Hasselblatt M, Jeibmann A, Kordes U et al (2010) Germline nonsense mutation and somatic inactivation of SMARCA4/BRG1 in a family with rhabdoid tumor predisposition syndrome. Am J Hum Genet 86:279–284. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62. Seo J‐S, Ju YS, Lee W‐C, Shin J‐Y, Lee JK, Bleazard T et al (2012) The transcriptional landscape and mutational profile of lung adenocarcinoma. Genome Res 22:2109–2119. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63. Serrano M, Lee H, Chin L, Cordon‐Cardo C, Beach D, DePinho RA (1996) Role of the INK4a locus in tumor suppression and cell mortality. Cell 85:27–37. [DOI] [PubMed] [Google Scholar]
  • 64. Sévenet N, Lellouch‐Tubiana A, Schofield D, Hoang‐Xuan K, Gessler M, Birnbaum D et al (1999) Spectrum of hSNF5/INI1 somatic mutations in human cancer and genotype‐phenotype correlations. Hum Mol Genet 8:2359–2368. [DOI] [PubMed] [Google Scholar]
  • 65. Sévenet N, Sheridan E, Amram D, Schneider P, Handgretinger R, Delattre O (1999) Constitutional mutations of the hSNF5/INI1 gene predispose to a variety of cancers. Am J Hum Genet 65:1342–1348. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66. Shain AH, Giacomini CP, Matsukuma K, Karikari CA, Bashyam MD, Hidalgo M et al (2012) 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 109:E252–E259. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67. Stephens PJ, Tarpey PS, Davies H, Van Loo P, Greenman C, Wedge DC et al (2012) The landscape of cancer genes and mutational processes in breast cancer. Nature 486:400–404. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68. Taylor MD, Gokgoz N, Andrulis IL, Mainprize TG, Drake JM, Rutka JT (2000) Familial posterior fossa brain tumors of infancy secondary to germline mutation of the hSNF5 gene. Am J Hum Genet 66:1403–1406. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69. Tsikitis M, Zhang Z, Edelman W, Zagzag D, Kalpana GV (2005) Genetic ablation of Cyclin D1 abrogates genesis of rhabdoid tumors resulting from Ini1 loss. Proc Natl Acad Sci USA 102:12129–12134. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70. Varela I, Tarpey P, Raine K, Huang D, Ong CK, Stephens P et al (2011) Exome sequencing identifies frequent mutation of the SWI/SNF complex gene PBRM1 in renal carcinoma. Nature 469:539–542. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71. Versteege I, Sévenet N, Lange J, Rousseau‐Merck MF, Ambros P, Handgretinger R et al (1998) Truncating mutations of hSNF5/INI1 in aggressive paediatric cancer. Nature 394:203–206. [DOI] [PubMed] [Google Scholar]
  • 72. Vries RGJ, Bezrookove V, Zuijderduijn LMP, Kia SK, Houweling A, Oruetxebarria I et al (2005) Cancer‐associated mutations in chromatin remodeler hSNF5 promote chromosomal instability by compromising the mitotic checkpoint. Genes Dev 19:665–670. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73. Wang K, Kan J, Yuen ST, Shi ST, Chu KM, Law S et al (2011) Exome sequencing identifies frequent mutation of ARID1A in molecular subtypes of gastric cancer. Nat Genet 43:1219–1223. [DOI] [PubMed] [Google Scholar]
  • 74. Wang L, Tsutsumi S, Kawaguchi T, Nagasaki K, Tatsuno K, Yamamoto S et al (2012) Whole‐exome sequencing of human pancreatic cancers and characterization of genomic instability caused by MLH1 haploinsufficiency and complete deficiency. Genome Res 22:208–219. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75. Wang W, Côté J, Xue Y, Zhou S, Khavari PA, Biggar SR et al (1996) Purification and biochemical heterogeneity of the mammalian SWI‐SNF complex. EMBO J 15:5370–5382. [PMC free article] [PubMed] [Google Scholar]
  • 76. Wang W, Xue Y, Zhou S, Kuo A, Cairns BR, Crabtree GR (1996) Diversity and specialization of mammalian SWI/SNF complexes. Genes Dev 10:2117–2130. [DOI] [PubMed] [Google Scholar]
  • 77. Wang X, Werneck MBF, Wilson BG, Kim H‐J, Kluk MJ, Thom CS et al (2011) TCR‐dependent transformation of mature memory phenotype T cells in mice. J Clin Invest 121:3834–3845. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78. Weeks DA, Beckwith JB, Mierau GW, Luckey DW (1989) Rhabdoid tumor of kidney. A report of 111 cases from the National Wilms' Tumor Study Pathology Center. Am J Surg Pathol 13:439–458. [PubMed] [Google Scholar]
  • 79. Welch JS, Ley TJ, Link DC, Miller CA, Larson DE, Koboldt DC et al (2012) The origin and evolution of mutations in acute myeloid leukemia. Cell 150:264–278. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80. Wiegand KC, Shah SP, Al‐Agha OM, Zhao Y, Tse K, Zeng T et al (2010) ARID1A mutations in endometriosis‐associated ovarian carcinomas. N Engl J Med 363:1532–1543. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81. Williams BO, Remington L, Albert DM, Mukai S, Bronson RT, Jacks T (1994) Cooperative tumorigenic effects of germline mutations in Rb and p53. Nat Genet 7:480–484. [DOI] [PubMed] [Google Scholar]
  • 82. Wilson BG, Roberts CWM (2011) SWI/SNF nucleosome remodellers and cancer. Nat Rev Cancer 11:481–492. [DOI] [PubMed] [Google Scholar]
  • 83. Wilson BG, Wang X, Shen X, McKenna ES, Lemieux ME, Cho Y‐J et al (2010) Epigenetic antagonism between polycomb and SWI/SNF complexes during oncogenic transformation. Cancer Cell 18:316–328. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84. Wu JN, Roberts CWM (2012) ARID1A mutations in cancer: another epigenetic tumor suppressor? Cancer Discov 3:35–43. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85. Xia W, Nagase S, Montia AG, Kalachikov SM, Keniry M, Su T et al (2008) BAF180 is a critical regulator of p21 induction and a tumor suppressor mutated in breast cancer. Cancer Res 68:1667–1674. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86. Zang ZJ, Cutcutache I, Poon SL, Zhang SL, McPherson JR, Tao J et al (2012) Exome sequencing of gastric adenocarcinoma identifies recurrent somatic mutations in cell adhesion and chromatin remodeling genes. Nat Genet 44:570–574. [DOI] [PubMed] [Google Scholar]
  • 87. Zhang J, Benavente CA, Mcevoy J, Flores‐Otero J, Ding L, Chen X et al (2012) ) A novel retinoblastoma therapy from genomic and epigenetic analyses. Nature 481:329–334. [DOI] [PMC free article] [PubMed] [Google Scholar]

Articles from Brain Pathology are provided here courtesy of Wiley

RESOURCES