SWI/SNF chromatin remodeling complexes are pleomorphic multisubunit cellular machines that utilize the energy of ATP hydrolysis to modulate chromatin structure. The complexes interact with transcription factors at promoters and enhancers to modulate gene expression and contribute to lineage specification, differentiation, and development. Initial clues to a role in tumor suppression for SWI/SNF complexes came over a decade ago when the gene encoding the SMARCB1/SNF5 core subunit was found specifically inactivated in nearly all pediatric rhabdoid tumors. In the last three years, cancer-genome sequencing efforts have revealed an unexpectedly high mutation rate of SWI/SNF subunit genes, which are collectively mutated in 20% of all human cancers and approach the frequency of p53 mutations. Here, we provide a background on these newly recognized tumor suppressor complexes, discuss mechanisms implicated in the tumor suppressor activity, and highlight findings that may lead to potential therapeutic targets for SWI/SNF-mutant cancers. Clin Cancer Res; 20(1); 21–27. ©2013 AACR.

Chromatin and SWI/SNF complexes

Nuclear DNA wrapped around a histone octamer constitutes the nucleosome, the basic unit of chromatin. Further compaction of DNA through progressive coiling not only provides an organizational structure for the two meters of DNA contained within each cell but also presents an access barrier to the transcriptional machinery. Numerous chromatin-modifying complexes exist in mammalian cells, and these are intimately involved in processes that require DNA access such as transcription, replication, and repair. These complexes can be grouped into two classes: those that covalently modify nucleosomes and those, like the SWI/SNF complex, that consume ATP to mobilize nucleosomes and modulate chromatin compaction.

SWI/SNF complexes are evolutionarily conserved and were originally identified in yeast. Genes encoding SWI/SNF subunits were revealed in screens of yeast for defects in mating type SWItching and in sucrose metabolism (Sucrose NonFermentable; SNF; ref. 1). Mammalian SWI/SNF complexes are also referred to as BAF (BRG1-associated factors) complexes in recognition that mammalian complexes contain additional subunits not found in the yeast complex, and therefore the extent to which activities are conserved remains unclear (2). Mammalian SWI/SNF complexes (herein referred to simply as SWI/SNF complexes) are enriched at promoters and enhancers of active genes and have been shown to contribute to the regulation of differentiation and proliferation across many lineages (3–5). SWI/SNF complexes are large, approximately 2 MDa, and composed of 12 to 15 subunits (2). These complexes are composed of one of the two mutually exclusive catalytic ATPase subunits: SMARCA2 (Brahma or BRM) or SMARCA4 (BRM/SWI2-related gene 1, or BRG1), and a set of widely expressed core subunits that include SMARCB1 (SNF5, INI-1 or BAF47), SMARCC1 (BAF155), and SMARCC2 (BAF170; ref. 2). In addition, SWI/SNF complexes also contain a large number of lineage-restricted subunits often encoded by multigene families (Table 1). Recent work has demonstrated that these complexes may further contain additional subunits not previously appreciated (6). Considering the large number of variant subunits, it has been estimated that several hundred versions of SWI/SNF complexes may exist, each with a conserved core of subunits but containing distinct combinations of variant subunits (7).

Table 1.

Summary of the SWI/SNF complex subunits and mutations in cancers

SubunitAliasesFunctionMutated in human cancer
ARID1A BAF250A Variant subunit, BAF complex only Ovarian, hepatocellular, bladder, gastric, endometrioid, pancreatic, colon, lung, neuroblastoma, Burkitt lymphoma 
ARID1B BAF250B Variant subunit, BAF complex only Melanoma, neuroblastoma, hepatocellular, pancreatic, liver 
PBRM1 BAF180 Variant subunit, PBAF complex only Renal cell carcinoma, breast, gastric, pancreatic 
ARID2 BAF200 Variant subunit, PBAF complex only Melanoma, hepatocellular, pancreatic 
SMARCA2 BRM Catalytic ATPase subunit Lung, colon, breast 
SMARCA4 BRG1 Catalytic ATPase subunit Lung, medulloblastoma, Burkitt lymphoma 
SMARCB1 SNF5, INI1 Core subunit Rhabdoid tumor, familial schwannomatosis 
SMARCC1 BAF155 Core subunit  
SMARCC2 BAF170 Core subunit  
SMARCD1/2/3 BAF60A/B/C Variant subunit Breast 
SMARCE1 BAF57 Variant subunit Spinal meningioma 
ACTL6A/B BAF53A/B Variant subunit  
PHF10 BAF45A Variant subunit  
DPF1/2/3 BAF45B/C/D Variant subunit  
ACTB β-Actin   
SubunitAliasesFunctionMutated in human cancer
ARID1A BAF250A Variant subunit, BAF complex only Ovarian, hepatocellular, bladder, gastric, endometrioid, pancreatic, colon, lung, neuroblastoma, Burkitt lymphoma 
ARID1B BAF250B Variant subunit, BAF complex only Melanoma, neuroblastoma, hepatocellular, pancreatic, liver 
PBRM1 BAF180 Variant subunit, PBAF complex only Renal cell carcinoma, breast, gastric, pancreatic 
ARID2 BAF200 Variant subunit, PBAF complex only Melanoma, hepatocellular, pancreatic 
SMARCA2 BRM Catalytic ATPase subunit Lung, colon, breast 
SMARCA4 BRG1 Catalytic ATPase subunit Lung, medulloblastoma, Burkitt lymphoma 
SMARCB1 SNF5, INI1 Core subunit Rhabdoid tumor, familial schwannomatosis 
SMARCC1 BAF155 Core subunit  
SMARCC2 BAF170 Core subunit  
SMARCD1/2/3 BAF60A/B/C Variant subunit Breast 
SMARCE1 BAF57 Variant subunit Spinal meningioma 
ACTL6A/B BAF53A/B Variant subunit  
PHF10 BAF45A Variant subunit  
DPF1/2/3 BAF45B/C/D Variant subunit  
ACTB β-Actin   

The precise biochemical function of SWI/SNF complexes remains somewhat unclear. In vitro assays have clearly demonstrated that the complexes are capable of mobilizing and ejecting histone octamers on DNA (8). Functional studies performed to evaluate biochemical activity of SWI/SNF complexes in living cells have implicated the complexes in the establishment of nucleosome occupancy and phasing at promoters and enhancers at a subset of active genes (3–5), as well as in DNA repair processes (9–11). Recent studies have begun to demonstrate that although SWI/SNF complexes may be ubiquitously expressed, individual cells contain a selected set of variant subunits that contribute to lineage-specific targeting and determination of cell fate (3–5). Perhaps some of the clearest evidence has come from studies on neural differentiation, which showed that SWI/SNF complex composition undergoes an essential subunit switch during the progression from neural progenitors to postmitotic neurons (12). Similarly, embryonic stem cells have been shown to contain a distinctive assembly of SWI/SNF subunits essential for embryonic stem cell maintenance and pluripotency (13). Such interaction with and recruitment of lineage-specific transcriptional regulators seem to be a central mechanism by which SWI/SNF complexes contribute to lineage specification. For example, MyoD, the muscle determination factor, can be directly incorporated into SWI/SNF complexes, which then results in transcription of MyoD target genes (14). Similarly, Olig2 has been shown to physically associate with the SWI/SNF complex at oligodendrocyte-specific enhancers during differentiation (15). SWI/SNF complexes may contribute specificity to transcription factor interactions and targeting via subunits that contain DNA-binding and histone-binding domains, including high-mobility group, ARID, HSA, PHD, and bromodomains, which are found in both core and variant SWI/SNF subunits (7). Thus, regulation of gene expression and cell lineage specification via direct recruiting or interaction with key transcription factors may be a central function of SWI/SNF complexes (Fig. 1).

Figure 1.

SWI/SNF functions in chromatin modulation and tumor suppression. Two subclasses, BAF and PBAF, of many variants of SWI/SNF complexes are shown. BAF and PBAF differ in composition of the subunits that are shown in orange. Core subunits are shown in blue. Further diversity is derived from the subunits shown in green as these are all encoded by multigene families, each yielding a slightly different amino acid sequence. These diverse complexes then make specific contributions to targeting of chromatin remodeling and transcriptional control. Several of the pathways that have been implicated in tumor suppression are shown at the bottom.

Figure 1.

SWI/SNF functions in chromatin modulation and tumor suppression. Two subclasses, BAF and PBAF, of many variants of SWI/SNF complexes are shown. BAF and PBAF differ in composition of the subunits that are shown in orange. Core subunits are shown in blue. Further diversity is derived from the subunits shown in green as these are all encoded by multigene families, each yielding a slightly different amino acid sequence. These diverse complexes then make specific contributions to targeting of chromatin remodeling and transcriptional control. Several of the pathways that have been implicated in tumor suppression are shown at the bottom.

Close modal

SWI/SNF complex mutations in cancer

In 1998, specific biallelic inactivating mutations of the core SWI/SNF subunit SMARCB1 (SNF5/INI1/BAF47) were identified in the vast majority of cases of rhabdoid tumors, highly aggressive and lethal cancers arising in the kidney, brain, and soft tissues of young children (16, 17). The bona fide tumor suppressor properties of SMARCB1 were subsequently demonstrated in genetically engineered mouse models: although homozygous loss results in embryonic lethality, heterozygous mice are viable and 30% develop cancers that closely resemble human rhabdoid tumor (18–20). In these tumors, the wild-type allele has been spontaneously inactivated so, as in humans, the tumors completely lack SMARCB1. To bypass embryonic lethality associated with homozygous loss, conditional SMARCB1 alleles were generated. Conditional homozygous inactivation in postnatal mice results in cancer in 100% of mice at a median onset of only 11 weeks, a remarkably rapid rate of onset for inactivation of a single gene (21). In comparison, biallelic inactivation of p53 leads to cancer at a median of 22 weeks.

Although rhabdoid tumors served as the initial clue linking mutation of genes encoding SWI/SNF subunits to cancer, recent cancer-genome sequencing studies have revealed a much broader role (22–24). Only 3 years ago, two papers reported that more than 50% of ovarian clear cell carcinomas contain inactivating mutations in ARID1A (25, 26), which encodes a variant subunit of the complex. Subsequently, frequent mutations of SWI/SNF complex subunits have been identified in a variety of human cancers (Table 1; selected references and summary reviews: 6, 22–24, 27–46). So far, at least eight genes encoding SWI/SNF complex subunits have been found to be recurrently mutated in a variety of cancers including subsets of lung, breast, stomach, liver, pancreas, kidney, bladder, skin, and brain cancers. These mutations are predominantly loss of function (6, 22–24, 27). Interestingly, although SMARCB1 is always biallelically mutated, other subunits are often heterozygously mutated, raising the possibility of haploinsufficiency for these subunits in tumor suppression (6, 27). Thus far SMARCB1 and SMARCA4 have been validated to have bona fide tumor suppressor function using genetically engineered mouse models (18–21, 47), whereas others remain to be tested. Collectively, genes encoding SWI/SNF complex subunits have now been recognized as one of the most commonly mutated targets in human cancer, being thus far detected in 20% of human cancers (6, 48).

Genetics or epigenetics?

Despite the prevalence of SWI/SNF complex mutations in cancer, insight into the mechanisms underlying tumor suppression and the reason for the differing cancer spectrum associated with each subunit is still in its infancy. In addition to a role in transcriptional regulation, several studies have linked the complex to DNA repair including nucleotide excision repair, double-strand break repair, and DNA decatenation (9–11, 49). Consequently, it has been a question of great interest whether the tumor suppressor activity of the complex arises via a role in the control of transcriptional programs or whether it is derived from a role for the complex in protecting genome integrity. Perhaps providing substantial insight, rhabdoid tumors are diploid, chromosomally stable, and contain a marked paucity of aberrations detectable by single-nucleotide polymorphism arrays, other than loss of SMARCB1 itself (50). More recently, exome sequencing of human rhabdoid tumors revealed that despite their highly aggressive nature, these cancers contain an extremely low rate of mutations (51), one of the lowest ever measured (52). Across 35 of these SMARCB1-deficient cancers, there were essentially no other recurrent mutations and in two of the cancers, there were no other identified mutations at all. The same conclusion was reached in other studies as well (53, 54). Given the paucity of other mutations and the extremely rapid onset of cancer in mouse models, these findings seem to suggest that SMARCB1 deficiency may not cause cancer via defects in DNA repair but rather due to epigenetic alterations such as disruption of chromatin-based contributions to control of cell fate. Given that at least eight subunits of the SWI/SNF complex are recurrently mutated in cancer, it is tempting to speculate that the general mechanism is likely shared. However, although evidence seems to suggest that SMARCB1 loss drives cancer via an epigenetic mechanism, whether a similar mechanism underlies cancer associated with ARID1A, SMARCA4, PBRM1, and other subunit mutations remains to be determined. Interestingly, mutations in more than one SWI/SNF subunit gene can occur in primary tumors, perhaps reflecting both haploinsufficiency and compound heterozygous effects (6, 47).

As increasing evidence has linked mutation of the SWI/SNF complex to a variety of human cancers, there has been a growing desire to identify vulnerabilities created by SWI/SNF mutation. Unlike some activating oncogene mutations, such as BCR-ABL fusion or EGF receptor (EGFR) mutation, which can be directly therapeutically targeted, the majority of SWI/SNF complex mutations are inactivating. Thus, the proteins are absent and cannot be directly targeted. Thus, a goal should be to achieve a better understanding of the mechanisms underlying transformation and dependencies created by mutation of SWI/SNF subunits. Indeed, early studies on the basic function of SWI/SNF complexes have revealed potential vulnerabilities currently being translated.

Pathways affected by SWI/SNF mutations

Some of the earliest studies of cancer-associated gene mutations, identification of genes activated by chromosomal translocation in acute leukemias, revealed the theme that these genes were often transcription factors that were master regulators of lineage development such that their forced overexpression resulted in perturbation of lymphoid development and predisposition to transformation. In its role of interacting with transcription factors to facilitate lineage-specific development, it is conceivable that mutation of SWI/SNF complexes predisposes to cancer in much the same way—via disruption of development, proliferation, and differentiation pathways. However, relevant target pathways are only beginning to be characterized.

One of the challenges for the field is that although SMARCB1 was recognized as a tumor suppressor 15 years ago, other subunits have only been linked to cancer recently. Consequently, mechanisms underlying the role of SMARCB1 in tumor suppression have been more extensively investigated. However, it remains to be determined the extent to which these mechanisms are shared with cancers driven by other SWI/SNF subunit mutations. Collectively, a shared mechanism of perturbation of SWI/SNF function seems likely. However, as the spectrum of cancer types associated with each subunit varies, the specific pathways affected may depend upon the subunit mutated. Ultimately, additional studies will be needed to determine the extent to which vulnerabilities caused by mutation of one SWI/SNF subunit gene extend to others.

The tumor suppressor activity of SWI/SNF complexes was first linked to the retinoblastoma tumor suppressor pathway when inactivation of SMARCB1 was shown to downregulate p16INK4A expression, a cyclin-dependent kinase (CDK) inhibitor that regulates the retinoblastoma pathway (55–59). SWI/SNF complexes can also bind to retinoblastoma itself and facilitate the repression of retinoblastoma target genes, such as E2Fs and CCND1 (60–62), suggesting a contribution of SWI/SNF complexes to cell-cycle regulation. Some, but not all, studies have found high-level cyclinD1 expression in rhabdoid tumor, and pharmacologic inhibitors of cyclin D1 have been reported effective in reducing the growth of rhabdoid tumor cell lines in vitro and in vivo (50, 63–66). Collectively, these findings have led to a phase I/II clinical trial of rhabdoid tumors using CDK4 inhibitors (http://clinicaltrials.gov/show/NCT01747876).

A number of other target pathways and functions have been implicated in cancer driven by SWI/SNF mutations. For example, SWI/SNF complexes have been implicated in the control of cell motility via regulation of RhoA (67) and actin cytoskeletal organization (68). SWI/SNF complexes can also directly interact with MYC, an oncogene frequently overexpressed in cancer (69), and SMARCB1-deficient rhabdoid tumors show activation of MYC programs (70, 71). SWI/SNF complexes interact with nuclear hormone receptors, such as glucocorticoid receptors, estrogen receptors, and retinoic acid receptors (72). Clinical studies found that decreased levels of SWI/SNF subunits SMARCB1/SMARCA4/ARID1A correlate with steroid resistance in pediatric acute lymphoblastic leukemia, with the underlying mechanism remaining unknown (73). SWI/SNF complexes have also been implicated in the regulation of the Wnt/β-catenin pathway. BRG1 deletion results in downregulation of the Wnt receptor family and degradation of β-catenin (74). The telomerase protein component TERT (telomerase reverse transcriptase) can also interact with BRG1, to modulate the Wnt/β-catenin pathway (75), and another study reported that loss of SMARCB1 results in β-catenin hyperactivation (76). SWI/SNF complexes also directly interact with Gli1, the effector of Hedgehog signaling and in rhabdoid tumor cells, loss of SMARCB1 leads to an aberrant activation of the Hedgehog–Gli pathway (77). These results suggest that SWI/SNF complexes directly regulate the canonical Wnt/β-catenin and Hedgehog pathways. Collectively, SWI/SNF complexes have been implicated in the control of a number of cancer-related transcriptional pathways, some of which may provide therapeutic potential (Fig. 1). Via such investigations, a theme is also emerging that SWI/SNF mutations may directly affect genes and pathways at the level of chromatin, and thus uncouple pathways from upstream canonical control. For example, cancers in which the Hedgehog pathway is activated by mutation of the PTCH receptor are sensitive to blockade of SMO downstream in the signaling cascade. In contrast, SMO inhibitors are unable to block Hedgehog signaling in SMARCB1-mutant cancers. Similarly, the Wnt/β-catenin can be inhibited at multiple levels. However, none of these upstream inhibitors have an effect upon the Wnt pathway in SMARCB1-mutant cancers where a key contributor seems to be a failure of the constitutive Wnt repressor TCF to bind target DNA in the absence of SMARB1 thus resulting in spontaneous Wnt signaling uncoupled from canonical pathway control. Although still early in the understanding of mechanism, it is tempting to speculate that chromatin remodeler mutations, such as SWI/SNF, may provide an explanation for findings such as the discrepancy that EGFR-mutant cancers are susceptible to EGFR blockade, whereas cancers that show transcriptional activation of the EGFR pathway, but lack mutations in EGFR itself, are unresponsive. Consequently, disruption of chromatin structure may have the potential to directly alter pathway signaling. Therefore, gaining insight into the function of SWI/SNF complexes in transcriptional control may offer novel modulation approaches of cancer-related pathways.

Antagonism of SWI/SNF complex and polycomb complex—targeting EZH2?

High mutation rate of SWI/SNF chromatin remodelers in a large variety of cancer suggests a chromatin-based epigenetic mechanism that drives cancer development by regulating chromatin structure. Therefore, mutations of SWI/SNF complexes likely lead to an aberrant chromatin landscape, providing the driving force of oncogenesis. Recent discoveries describing an epigenetic antagonism between Polycomb and the SWI/SNF complex support this model (70, 78). Loss of SMARCB1 was found to cause elevated levels of EZH2 and H3K27me3 at PcG target genes thus leading to broad repression of these targets (70). Another study in embryonic stem cells found complex interactions between SWI/SNF and Polycomb complexes, at times antagonistic and at others cooperative (4). In a genetically engineered in vivo tumor model, inactivation of EZH2 completely blocked tumor onset driven by SMARCB1 loss (70). This discovery suggested EZH2 as a target for SMARCB1-mutant cancers. In contrast with these findings, another group found that knockdown of SMARCB1 does not affect the level of EZH2 (79), raising the possibility that antagonism between SMARCB1 and EZH2 might be complex and cell-context dependent (79, 80). Nonetheless, a recent study reported that targeted pharmacologic inhibition of EZH2 results in durable regressions in genetically altered malignant rhabdoid tumors, suggesting this as a potential new treatment for rhabdoid tumor (81), stimulating interest in therapeutic trials for rhabdoid tumor. Whether antagonism between SMARCB1 and EZH2 extends to other SWI/SNF subunits and associated cancers needs to be further investigated.

Cancer-genome sequencing studies have identified genes encoding subunits of the SWI/SNF chromatin remodeling complexes as being frequently mutated across a wide variety of cancers. However, identification of these links is largely recent and the mechanisms by which these complexes act as tumor suppressors are only just beginning to be elucidated. Although some evidence indicates that these complexes may contribute to DNA repair, involvement of these complexes in chromatin-based epigenetic regulation has been implicated in their tumor suppressor activity. Ultimately, understanding the fundamental biology of SWI/SNF complexes, such as roles in the regulation of chromatin structure/landscape, the biochemical contributions of different subunits, and activity in modulating transcription, offer promise for novel therapeutic approaches to SWI/SNF-mutant cancers.

C.W.M. Roberts has commercial research grant and is consultant/advisory board member of Novartis Institutes for Biomedical Research. No potential conflicts of interest were disclosed by the other authors.

Conception and design: X. Wang, C.W.M. Roberts

Acquisition of data (provided animals, acquired and managed patients, provided facilities, etc.):

Analysis and interpretation of data (e.g., statistical analysis, biostatistics, computational analysis): X. Wang

Writing, review, and/or revision of the manuscript: X. Wang, J.R. Haswell, C.W.M. Roberts

Study supervision: C.W.M. Roberts

The authors thank Katherine C. Helming for critical reading of the manuscript.

The work in Dr. Roberts' laboratory is partly supported by R01CA172152 (to C.W.M. Roberts) and R01CA113794 (to C.W.M. Roberts), and a U01 NCI Mouse Models of Cancer Consortium Award (C.W.M. Roberts). Alex's Lemonade Stand Foundation, Hyundai Hope on Wheels, the Cure AT/RT Now fund, Garrett B. Smith Foundation, Miles for Mary, and The Ellison Foundation and Cookies for Kids Cancer provided additional support. X. Wang was supported by St. Baldrick's Foundation, the Rally Foundation, and David Abraham Foundation.

1.
Neigeborn
L
,
Carlson
M
. 
Genes affecting the regulation of SUC2 gene expression by glucose repression in Saccharomyces cerevisiae
.
Genetics
1984
;
108
:
845
58
.
2.
Wang
W
,
Cote
J
,
Xue
Y
,
Zhou
S
,
Khavari
PA
,
Biggar
SR
, et al
Purification and biochemical heterogeneity of the mammalian SWI-SNF complex
.
EMBO J
1996
;
15
:
5370
82
.
3.
Tolstorukov
MY
,
Sansam
CG
,
Lu
P
,
Koellhoffer
EC
,
Helming
KC
,
Alver
BH
, et al
Swi/Snf chromatin remodeling/tumor suppressor complex establishes nucleosome occupancy at target promoters
.
Proc Natl Acad Sci U S A
2013
;
110
:
10165
70
.
4.
Ho
L
,
Jothi
R
,
Ronan
JL
,
Cui
K
,
Zhao
K
,
Crabtree
GR
. 
An embryonic stem cell chromatin remodeling complex, esBAF, is an essential component of the core pluripotency transcriptional network
.
Proc Natl Acad Sci U S A
2009
;
106
:
5187
91
.
5.
Hu
G
,
Schones
DE
,
Cui
K
,
Ybarra
R
,
Northrup
D
,
Tang
Q
, et al
Regulation of nucleosome landscape and transcription factor targeting at tissue-specific enhancers by BRG1
.
Genome Res
2011
;
21
:
1650
8
.
6.
Kadoch
C
,
Hargreaves
DC
,
Hodges
C
,
Elias
L
,
Ho
L
,
Ranish
J
, et al
Proteomic and bioinformatic analysis of mammalian SWI/SNF complexes identifies extensive roles in human malignancy
.
Nat Genet
2013
;
45
:
592
601
.
7.
Wu
JI
,
Lessard
J
,
Crabtree
GR
. 
Understanding the words of chromatin regulation
.
Cell
2009
;
136
:
200
6
.
8.
Phelan
ML
,
Sif
S
,
Narlikar
GJ
,
Kingston
RE
. 
Reconstitution of a core chromatin remodeling complex from SWI/SNF subunits
.
Mol Cell
1999
;
3
:
247
53
.
9.
Hara
R
,
Sancar
A
. 
The SWI/SNF chromatin-remodeling factor stimulates repair by human excision nuclease in the mononucleosome core particle
.
Mol Cell Biol
2002
;
22
:
6779
87
.
10.
Park
JH
,
Park
EJ
,
Lee
HS
,
Kim
SJ
,
Hur
SK
,
Imbalzano
AN
, et al
Mammalian SWI/SNF complexes facilitate DNA double-strand break repair by promoting gamma-H2AX induction
.
EMBO J
2006
;
25
:
3986
97
.
11.
Gong
F
,
Fahy
D
,
Smerdon
MJ
. 
Rad4-Rad23 interaction with SWI/SNF links ATP-dependent chromatin remodeling with nucleotide excision repair
.
Nat Struct Mol Biol
2006
;
13
:
902
7
.
12.
Lessard
J
,
Wu
JI
,
Ranish
JA
,
Wan
M
,
Winslow
MM
,
Staahl
BT
, et al
An essential switch in subunit composition of a chromatin remodeling complex during neural development
.
Neuron
2007
;
55
:
201
15
.
13.
Ho
L
,
Ronan
JL
,
Wu
J
,
Staahl
BT
,
Chen
L
,
Kuo
A
, et al
An embryonic stem cell chromatin remodeling complex, esBAF, is essential for embryonic stem cell self-renewal and pluripotency
.
Proc Natl Acad Sci U S A
2009
;
106
:
5181
6
.
14.
de la Serna
IL
,
Carlson
KA
,
Imbalzano
AN
. 
Mammalian SWI/SNF complexes promote MyoD-mediated muscle differentiation
.
Nat Genet
2001
;
27
:
187
90
.
15.
Yu
Y
,
Chen
Y
,
Kim
B
,
Wang
H
,
Zhao
C
,
He
X
, et al
Olig2 targets chromatin remodelers to enhancers to initiate oligodendrocyte differentiation
.
Cell
2013
;
152
:
248
61
.
16.
Versteege
I
,
Sevenet
N
,
Lange
J
,
Rousseau-Merck
MF
,
Ambros
P
,
Handgretinger
R
, et al
Truncating mutations of hSNF5/INI1 in aggressive paediatric cancer
.
Nature
1998
;
394
:
203
6
.
17.
Biegel
JA
,
Zhou
JY
,
Rorke
LB
,
Stenstrom
C
,
Wainwright
LM
,
Fogelgren
B
. 
Germ-line and acquired mutations of INI1 in atypical teratoid and rhabdoid tumors
.
Cancer Res
1999
;
59
:
74
9
.
18.
Roberts
CW
,
Galusha
SA
,
McMenamin
ME
,
Fletcher
CD
,
Orkin
SH
. 
Haploinsufficiency of Snf5 (integrase interactor 1) predisposes to malignant rhabdoid tumors in mice
.
Proc Natl Acad Sci U S A
2000
;
97
:
13796
800
.
19.
Klochendler-Yeivin
A
,
Fiette
L
,
Barra
J
,
Muchardt
C
,
Babinet
C
,
Yaniv
M
. 
The murine SNF5/INI1 chromatin remodeling factor is essential for embryonic development and tumor suppression
.
EMBO Rep
2000
;
1
:
500
6
.
20.
Guidi
CJ
,
Sands
AT
,
Zambrowicz
BP
,
Turner
TK
,
Demers
DA
,
Webster
W
, et al
Disruption of Ini1 leads to peri-implantation lethality and tumorigenesis in mice
.
Mol Cell Biol
2001
;
21
:
3598
603
.
21.
Roberts
CW
,
Leroux
MM
,
Fleming
MD
,
Orkin
SH
. 
Highly penetrant, rapid tumorigenesis through conditional inversion of the tumor suppressor gene Snf5
.
Cancer Cell
2002
;
2
:
415
25
.
22.
Garraway
LA
,
Lander
ES
. 
Lessons from the cancer genome
.
Cell
2013
;
153
:
17
37
.
23.
Wilson
BG
,
Roberts
CW
. 
SWI/SNF nucleosome remodellers and cancer
.
Nat Rev Cancer
2011
;
11
:
481
92
.
24.
Shen
H
,
Laird
PW
. 
Interplay between the cancer genome and epigenome
.
Cell
2013
;
153
:
38
55
.
25.
Wiegand
KC
,
Shah
SP
,
Al-Agha
OM
,
Zhao
Y
,
Tse
K
,
Zeng
T
, et al
ARID1A mutations in endometriosis-associated ovarian carcinomas
.
N Engl J Med
2010
;
363
:
1532
43
.
26.
Jones
S
,
Wang
TL
,
Shih Ie
M
,
Mao
TL
,
Nakayama
K
,
Roden
R
, et al
Frequent mutations of chromatin remodeling gene ARID1A in ovarian clear cell carcinoma
.
Science
2010
;
330
:
228
31
.
27.
Wu
JN
,
Roberts
CW
. 
ARID1A mutations in cancer: another epigenetic tumor suppressor?
Cancer Discov
2013
;
3
:
35
43
.
28.
Fujimoto
A
,
Totoki
Y
,
Abe
T
,
Boroevich
KA
,
Hosoda
F
,
Nguyen
HH
, 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
4
.
29.
Huang
J
,
Deng
Q
,
Wang
Q
,
Li
KY
,
Dai
JH
,
Li
N
, et al
Exome sequencing of hepatitis B virus-associated hepatocellular carcinoma
.
Nat Genet
2012
;
44
:
1117
21
.
30.
Sausen
M
,
Leary
RJ
,
Jones
S
,
Wu
J
,
Reynolds
CP
,
Liu
X
, et al
Integrated genomic analyses identify ARID1A and ARID1B alterations in the childhood cancer neuroblastoma
.
Nat Genet
2013
;
45
:
12
7
.
31.
Smith
MJ
,
O'Sullivan
J
,
Bhaskar
SS
,
Hadfield
KD
,
Poke
G
,
Caird
J
, et al
Loss-of-function mutations in SMARCE1 cause an inherited disorder of multiple spinal meningiomas
.
Nat Genet
2013
;
45
:
295
8
.
32.
Wang
K
,
Kan
J
,
Yuen
ST
,
Shi
ST
,
Chu
KM
,
Law
S
, et al
Exome sequencing identifies frequent mutation of ARID1A in molecular subtypes of gastric cancer
.
Nat Genet
2011
;
43
:
1219
23
.
33.
Comprehensive molecular characterization of human colon and rectal cancer
.
Nature
2012
;
487
:
330
7
.
34.
Kandoth
C
,
Schultz
N
,
Cherniack
AD
,
Akbani
R
,
Liu
Y
,
Shen
H
, et al
Integrated genomic characterization of endometrial carcinoma
.
Nature
2013
;
497
:
67
73
.
35.
Varela
I
,
Tarpey
P
,
Raine
K
,
Huang
D
,
Ong
CK
,
Stephens
P
, et al
Exome sequencing identifies frequent mutation of the SWI/SNF complex gene PBRM1 in renal carcinoma
.
Nature
2011
;
469
:
539
42
.
36.
Shain
AH
,
Giacomini
CP
,
Matsukuma
K
,
Karikari
CA
,
Bashyam
MD
,
Hidalgo
M
, 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 U S A
2012
;
109
:
E252
9
.
37.
Hodis
E
,
Watson
IR
,
Kryukov
GV
,
Arold
ST
,
Imielinski
M
,
Theurillat
JP
, et al
A landscape of driver mutations in melanoma
.
Cell
2012
;
150
:
251
63
.
38.
Imielinski
M
,
Berger
AH
,
Hammerman
PS
,
Hernandez
B
,
Pugh
TJ
,
Hodis
E
, et al
Mapping the hallmarks of lung adenocarcinoma with massively parallel sequencing
.
Cell
2012
;
150
:
1107
20
.
39.
Gui
Y
,
Guo
G
,
Huang
Y
,
Hu
X
,
Tang
A
,
Gao
S
, et al
Frequent mutations of chromatin remodeling genes in transitional cell carcinoma of the bladder
.
Nat Genet
2011
;
43
:
875
8
.
40.
Guichard
C
,
Amaddeo
G
,
Imbeaud
S
,
Ladeiro
Y
,
Pelletier
L
,
Maad
IB
, 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
8
.
41.
Le Gallo
M
,
O'Hara
AJ
,
Rudd
ML
,
Urick
ME
,
Hansen
NF
,
O'Neil
NJ
, et al
Exome sequencing of serous endometrial tumors identifies recurrent somatic mutations in chromatin-remodeling and ubiquitin ligase complex genes
.
Nat Genet
2012
;
44
:
1310
5
.
42.
Streppel
MM
,
Lata
S
,
Delabastide
M
,
Montgomery
EA
,
Wang
JS
,
Canto
MI
, et al
Next-generation sequencing of endoscopic biopsies identifies ARID1A as a tumor-suppressor gene in Barrett's esophagus
.
Oncogene
2013 Jan 14
.
[Epub ahead of print]
.
43.
Biankin
AV
,
Waddell
N
,
Kassahn
KS
,
Gingras
MC
,
Muthuswamy
LB
,
Johns
AL
, et al
Pancreatic cancer genomes reveal aberrations in axon guidance pathway genes
.
Nature
2012
;
491
:
399
405
.
44.
Giulino-Roth
L
,
Wang
K
,
MacDonald
TY
,
Mathew
S
,
Tam
Y
,
Cronin
MT
, et al
Targeted genomic sequencing of pediatric Burkitt lymphoma identifies recurrent alterations in antiapoptotic and chromatin-remodeling genes
.
Blood
2012
;
120
:
5181
4
.
45.
Seo
JS
,
Ju
YS
,
Lee
WC
,
Shin
JY
,
Lee
JK
,
Bleazard
T
, et al
The transcriptional landscape and mutational profile of lung adenocarcinoma
.
Genome Res
2012
;
22
:
2109
19
.
46.
Liang
H
,
Cheung
LW
,
Li
J
,
Ju
Z
,
Yu
S
,
Stemke-Hale
K
, et al
Whole-exome sequencing combined with functional genomics reveals novel candidate driver cancer genes in endometrial cancer
.
Genome Res
2012
;
22
:
2120
9
.
47.
Bultman
SJ
,
Herschkowitz
JI
,
Godfrey
V
,
Gebuhr
TC
,
Yaniv
M
,
Perou
CM
, et al
Characterization of mammary tumors from Brg1 heterozygous mice
.
Oncogene
2008
;
27
:
460
8
.
48.
Shain
AH
,
Pollack
JR
. 
The spectrum of SWI/SNF mutations, ubiquitous in human cancers
.
PloS One
2013
;
8
:
e55119
.
49.
Dykhuizen
EC
,
Hargreaves
DC
,
Miller
EL
,
Cui
K
,
Korshunov
A
,
Kool
M
, et al
BAF complexes facilitate decatenation of DNA by topoisomerase IIalpha
.
Nature
2013
;
497
:
624
7
.
50.
McKenna
ES
,
Sansam
CG
,
Cho
YJ
,
Greulich
H
,
Evans
JA
,
Thom
CS
, et al
Loss of the epigenetic tumor suppressor SNF5 leads to cancer without genomic instability
.
Mol Cell Biol
2008
;
28
:
6223
33
.
51.
Lee
RS
,
Stewart
C
,
Carter
SL
,
Ambrogio
L
,
Cibulskis
K
,
Sougnez
C
, et al
A remarkably simple genome underlies highly malignant pediatric rhabdoid cancers
.
J Clin Invest
2012
;
122
:
2983
8
.
52.
Lawrence
MS
,
Stojanov
P
,
Polak
P
,
Kryukov
GV
,
Cibulskis
K
,
Sivachenko
A
, et al
Mutational heterogeneity in cancer and the search for new cancer-associated genes
.
Nature
2013
;
499
:
214
8
.
53.
Hasselblatt
M
,
Isken
S
,
Linge
A
,
Eikmeier
K
,
Jeibmann
A
,
Oyen
F
, et al
High-resolution genomic analysis suggests the absence of recurrent genomic alterations other than SMARCB1 aberrations in atypical teratoid/rhabdoid tumors
.
Genes Chromosomes Cancer
2013
;
52
:
185
90
.
54.
Hoell
JI
,
Gombert
M
,
Bartenhagen
C
,
Ginzel
S
,
Husemann
P
,
Felsberg
J
, et al
Whole-genome paired-end analysis confirms remarkable genomic stability of atypical teratoid/rhabdoid tumors
.
Genes Chromosomes Cancer
2013
;
52
:
983
5
.
55.
Oruetxebarria
I
,
Venturini
F
,
Kekarainen
T
,
Houweling
A
,
Zuijderduijn
LM
,
Mohd-Sarip
A
, et al
P16INK4a is required for hSNF5 chromatin remodeler-induced cellular senescence in malignant rhabdoid tumor cells
.
J Biol Chem
2004
;
279
:
3807
16
.
56.
Isakoff
MS
,
Sansam
CG
,
Tamayo
P
,
Subramanian
A
,
Evans
JA
,
Fillmore
CM
, et al
Inactivation of the Snf5 tumor suppressor stimulates cell cycle progression and cooperates with p53 loss in oncogenic transformation
.
Proc Natl Acad Sci U S A
2005
;
102
:
17745
50
.
57.
Staehling-Hampton
K
,
Ciampa
PJ
,
Brook
A
,
Dyson
N
. 
A genetic screen for modifiers of E2F in Drosophila melanogaster
.
Genetics
1999
;
153
:
275
87
.
58.
Strobeck
MW
,
Knudsen
KE
,
Fribourg
AF
,
DeCristofaro
MF
,
Weissman
BE
,
Imbalzano
AN
, et al
BRG-1 is required for RB-mediated cell cycle arrest
.
Proc Natl Acad Sci U S A
2000
;
97
:
7748
53
.
59.
Betz
BL
,
Strobeck
MW
,
Reisman
DN
,
Knudsen
ES
,
Weissman
BE
. 
Re-expression of hSNF5/INI1/BAF47 in pediatric tumor cells leads to G1 arrest associated with induction of p16ink4a and activation of RB
.
Oncogene
2002
;
21
:
5193
203
.
60.
Trouche
D
,
Le Chalony
C
,
Muchardt
C
,
Yaniv
M
,
Kouzarides
T
. 
RB and hbrm cooperate to repress the activation functions of E2F1
.
Proc Natl Acad Sci U S A
1997
;
94
:
11268
73
.
61.
Versteege
I
,
Medjkane
S
,
Rouillard
D
,
Delattre
O
. 
A key role of the hSNF5/INI1 tumour suppressor in the control of the G1-S transition of the cell cycle
.
Oncogene
2002
;
21
:
6403
12
.
62.
Biegel
JA
,
Kalpana
G
,
Knudsen
ES
,
Packer
RJ
,
Roberts
CW
,
Thiele
CJ
, et al
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
2002
;
62
:
323
8
.
63.
Alarcon-Vargas
D
,
Zhang
Z
,
Agarwal
B
,
Challagulla
K
,
Mani
S
,
Kalpana
GV
. 
Targeting cyclin D1, a downstream effector of INI1/hSNF5, in rhabdoid tumors
.
Oncogene
2006
;
25
:
722
34
.
64.
Smith
ME
,
Cimica
V
,
Chinni
S
,
Challagulla
K
,
Mani
S
,
Kalpana
GV
. 
Rhabdoid tumor growth is inhibited by flavopiridol
.
Clin Cancer Res
2008
;
14
:
523
32
.
65.
Gadd
S
,
Sredni
ST
,
Huang
CC
,
Perlman
EJ
. 
Rhabdoid tumor: gene expression clues to pathogenesis and potential therapeutic targets
.
Lab Invest
2010
;
90
:
724
38
.
66.
Venneti
S
,
Le
P
,
Martinez
D
,
Eaton
KW
,
Shyam
N
,
Jordan-Sciutto
KL
, et al
p16INK4A and p14ARF tumor suppressor pathways are deregulated in malignant rhabdoid tumors
.
J Neuropathol Exp Neurol
2011
;
70
:
596
609
.
67.
Caramel
J
,
Quignon
F
,
Delattre
O
. 
RhoA-dependent regulation of cell migration by the tumor suppressor hSNF5/INI1
.
Cancer Res
2008
;
68
:
6154
61
.
68.
Rosson
GB
,
Bartlett
C
,
Reed
W
,
Weissman
BE
. 
BRG1 loss in MiaPaCa2 cells induces an altered cellular morphology and disruption in the organization of the actin cytoskeleton
.
J Cell Physiol
2005
;
205
:
286
94
.
69.
Cheng
SW
,
Davies
KP
,
Yung
E
,
Beltran
RJ
,
Yu
J
,
Kalpana
GV
. 
c-MYC interacts with INI1/hSNF5 and requires the SWI/SNF complex for transactivation function
.
Nat Genet
1999
;
22
:
102
5
.
70.
Wilson
BG
,
Wang
X
,
Shen
X
,
McKenna
ES
,
Lemieux
ME
,
Cho
YJ
, et al
Epigenetic antagonism between polycomb and SWI/SNF complexes during oncogenic transformation
.
Cancer Cell
2010
;
18
:
316
28
.
71.
Wang
X
,
Werneck
MB
,
Wilson
BG
,
Kim
HJ
,
Kluk
MJ
,
Thom
CS
, et al
TCR-dependent transformation of mature memory phenotype T cells in mice
.
J Clin Invest
2011
;
121
:
3834
45
.
72.
Trotter
KW
,
Archer
TK
. 
The BRG1 transcriptional coregulator
.
Nucl Recept Signal
2008
;
6
:
e004
.
73.
Pottier
N
,
Yang
W
,
Assem
M
,
Panetta
JC
,
Pei
D
,
Paugh
SW
, et al
The SWI/SNF chromatin-remodeling complex and glucocorticoid resistance in acute lymphoblastic leukemia
.
J Natl Cancer Inst
2008
;
100
:
1792
803
.
74.
Griffin
CT
,
Curtis
CD
,
Davis
RB
,
Muthukumar
V
,
Magnuson
T
. 
The chromatin-remodeling enzyme BRG1 modulates vascular Wnt signaling at two levels
.
Proc Natl Acad Sci U S A
2011
;
108
:
2282
7
.
75.
Park
JI
,
Venteicher
AS
,
Hong
JY
,
Choi
J
,
Jun
S
,
Shkreli
M
, et al
Telomerase modulates Wnt signalling by association with target gene chromatin
.
Nature
2009
;
460
:
66
72
.
76.
Mora-Blanco
EL
,
Mishina
Y
,
Tillman
EJ
,
Cho
YJ
,
Thom
CS
,
Pomeroy
SL
, et al
Activation of beta-catenin/TCF targets following loss of the tumor suppressor SNF5
.
Oncogene
2013 Feb 25
.
[Epub ahead of print]
.
77.
Jagani
Z
,
Mora-Blanco
EL
,
Sansam
CG
,
McKenna
ES
,
Wilson
B
,
Chen
D
, et al
Loss of the tumor suppressor Snf5 leads to aberrant activation of the Hedgehog-Gli pathway
.
Nat Med
2010
;
16
:
1429
33
.
78.
Kia
SK
,
Gorski
MM
,
Giannakopoulos
S
,
Verrijzer
CP
. 
SWI/SNF mediates polycomb eviction and epigenetic reprogramming of the INK4b-ARF-INK4a locus
.
Mol Cell Biol
2008
;
28
:
3457
64
.
79.
You
JS
,
De Carvalho
DD
,
Dai
C
,
Liu
M
,
Pandiyan
K
,
Zhou
XJ
, et al
SNF5 is an essential executor of epigenetic regulation during differentiation
.
PLoS Genet
2013
;
9
:
e1003459
.
80.
Hargreaves
DC
,
Crabtree
GR
. 
ATP-dependent chromatin remodeling: genetics, genomics and mechanisms
.
Cell Res
2011
;
21
:
396
420
.
81.
Knutson
SK
,
Warholic
NM
,
Wigle
TJ
,
Klaus
CR
,
Allain
CJ
,
Raimondi
A
, et al
Durable tumor regression in genetically altered malignant rhabdoid tumors by inhibition of methyltransferase EZH2
.
Proc Natl Acad Sci U S A
2013
;
110
:
7922
7
.