ArticlePublisher preview available

SWI/SNF remains localized to chromatin in the presence of SCHLAP1

Authors:
To read the full-text of this research, you can request a copy directly from the authors.

Abstract and Figures

SCHLAP1 is a long noncoding RNA that is reported to function by depleting the SWI/SNF complex from the genome. We investigated the hypothesis that SCHLAP1 affects only specific compositions of SWI/SNF. Using several assays, we found that SWI/SNF is not depleted from the genome by SCHLAP1 and that SWI/SNF is associated with many coding and noncoding RNAs, suggesting that SCHLAP1 may function in a SWI/SNF-independent manner. © 2018, The Author(s), under exclusive licence to Springer Nature America, Inc.
This content is subject to copyright. Terms and conditions apply.
Brief CommuniCation
https://doi.org/10.1038/s41588-018-0272-z
1Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA. 2Department of Pharmacology and Lineberger Comprehensive
Cancer Center, University of North Carolina, Chapel Hill, NC, USA. 3Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill,
Chapel Hill, NC, USA. *e-mail: tmagnuson@unc.edu
SCHLAP1 is a long noncoding RNA that is reported to func-
tion by depleting the SWI/SNF complex from the genome.
We investigated the hypothesis that SCHLAP1 affects only
specific compositions of SWI/SNF. Using several assays,
we found that SWI/SNF is not depleted from the genome by
SCHLAP1 and that SWI/SNF is associated with many coding
and noncoding RNAs, suggesting that SCHLAP1 may function
in a SWI/SNF-independent manner.
The long noncoding RNA (lncRNA) second chromosome locus
associated with prostate cancer 1 (SCHLAP1) is a promising bio-
marker for metastatic prostate cancer1,2. SCHLAP1 is proposed to
function by antagonizing the SWI/SNF complex through direct
interaction, leading to complete disruption of SWI/SNF genomic
occupancy1. Evidence for this mechanism comes from the reported
loss of SMARCB1 occupancy measured by chromatin immunopre-
cipitation followed by sequencing (ChIP–seq)1. SWI/SNF is a large
multi-subunit chromatin remodeling complex that can be combina-
torially assembled to yield hundreds to thousands of biochemically
unique complexes35. We investigated the alternative hypothesis that
distinct forms of SWI/SNF are affected by SCHLAP1 expression.
However, using a varriety of biochemical and genomics assays, we
demonstrate that SWI/SNF occupancy is unaffected by SCHLAP1
expression, in contrast to results reported previously1. We show that
SWI/SNF binds coding and noncoding RNA, raising the possibility
that SCHLAP1 function is SWI/SNF-independent.
Consistent with the report by Prensner et al.1, we observed an
interaction between SMARCB1 and SCHLAP1 (Fig. 1a). We next
generated SCHLAP1-overexpressing benign prostate epithelial cells
(RWPE1;SCHLAP1 cells) or control cells (RWPE1;LACZ cells)1
(SCHLAP1 gift of A. Chinnaiyan). This model is the same as
that originally used to suggest global depletion of SMARCB1 by
SCHLAP1 (ref. 1). We then confirmed the phenotype of the cells
with respect to SWI/SNF expression and growth (Fig. 1b and
Supplementary Figs. 1,2). In addition, we confirmed the key result
that SCHLAP1 increased cell invasion (Fig. 1c).
To investigate which SWI/SNF subunits were depleted from chro-
matin upon SCHLAP1 expression, we fractionated RWPE1;LACZ
and RWPE1;SCHLAP1 cells based on subcellular localization or salt
extraction. Surprisingly, all SWI/SNF subunits assayed remained
strongly enriched in the chromatin or high salt fractions (Fig. 1d
and Supplementary Figs. 3–5). Consistent with these biochemical
experiments, we found that SMARCA4 and SMARCB1 localization
was not affected in RWPE1;SCHLAP1 cells by immunofluorescence
(Supplementary Fig. 3b). Immunoprecipitation of SMARCA4 or
SMARCB1 demonstrated that the SWI/SNF complex remains intact
in the presence of SCHLAP1 (Supplementary Figs. 3c,6). Finally, we
used a malignant rhabdoid tumor cell line with inducible SMARCB1
that, when expressed, causes growth arrest (gift of B. Weissman)6.
We reasoned that, if SCHLAP1 disrupted SMARCB1 chromatin
occupancy, then overexpression of SCHLAP1 should allow G401
cells to proliferate following induction of SMARCB1. However,
SMARCB1 induction led to growth arrest in a dose-dependent man-
ner (Fig. 1e and Supplementary Figs. 7,8). Together, these results
demonstrate that SCHLAP1 does not induce changes to SWI/SNF
composition or its association with chromatin.
We next performed ChIP–seq for three SWI/SNF subunits
(SMARCB1, SMARCA2, and SMARCA4) in RWPE1;SCHLAP1
cells. In contrast to a previous report1, we identify robust binding
for all three subunits in RWPE1 cells expressing SCHLAP1 (Fig. 1f).
In RWPE1;SCHLAP1 cells, we identified 6,490, 22,185, and 51,505
peaks for SMARCB1, SMARCA2, and SMARCA4, respectively
(Supplementary Table 1). This large number of peaks is in con-
trast to the previous report1, which identified approximately 6,500
SMARCB1 peaks in RWPE1;LACZ cells and close to no peaks in
the SCHLAP1-expressing cells. The numbers of peaks are consis-
tent with previous work from our laboratory, which showed 30,000–
45,000 SMARCA4 peaks4. In addition, we and others have reported
a large number of SWI/SNF peaks for a variety of subunits3,4,7,8.
The majority of SMARCA2 peaks overlapped a SMARCA4 peak
(Supplementary Fig. 9a), and SWI/SNF peaks were predominantly
located at promoters (45–75%; Supplementary Fig. 9b)4. SWI/SNF
binding was most prominent at highly expressed genes, with little to
no occupancy at non-expressed genes (Fig. 1g; expression data from
GSE98898 (ref. 9)). These results demonstrate that SCHLAP1 does
not function by disrupting SWI/SNF occupancy genome-wide, and
raises the question of how SCHLAP1 functions to promote cell
invasion and progression to metastatic disease.
To investigate whether SCHLAP1 expression induces chromatin
changes, we performed assay for transposase-accessible chromatin
with high-throughput sequencing (ATAC-seq) on RWPE1;LACZ
and RWPE1;SCHLAP1 cells10. We identified 273 and 3,167 sites
that open and close, respectively (Supplementary Table 2 and
Supplementary Fig. 10). The sites that open were more likely to
be located distally or in introns of genes (Supplementary Fig. 10).
Sites that open upon SCHLAP1 expression were enriched for dis-
tinct motifs compared to those that close (Supplementary Fig. 11
and Supplementary Tables 3, 4). Open sites were enriched in motifs
for TEAD and AP1 transcription factors, which are known to have
a role in defining oncogenic enhancers (Supplementary Fig. 11 and
Supplementary Tables 3, 4)11. To test whether these sites became
SWI/SNF remains localized to chromatin in the
presence of SCHLAP1
JesseR.Raab1, KeriaynN.Smith1, CamarieC.Spear1, CarlJ.Manner1, J.MauroCalabrese2 and
TerryMagnuson 1,3*
NATURE GENETICS | VOL 51 | JANUARY 2019 | 26–29 | www.nature.com/naturegenetics
26
Content courtesy of Springer Nature, terms of use apply. Rights reserved
... This domain is present in the SNF2 protein family, which comprises integral components of the various nucleosome rearranging SWI/SNF complexes [60]. Like the STAT family, SWI/SNF complexes are well-known players in transcription initiation, but recent RIP experiments revealed pervasive binding to protein-coding mRNAs [61]. Another interesting set of protein families are those containing bromodomains and PHD-fingers, involved in the binding to acetylated and methylated sites on histones, respectively. ...
... ChIP-seq in A549 cells [108,109], murine macrophages [110], 3134 cells [111] RIP-seq with UV crosslinking in U2OS cells [112] RIP in BEAS-2B cells [77], rat smooth muscle cells [113] Important MPEC formation [81] Drives the dysfunctional phenotype in tumor-infiltrating T cells [82] STAT family STAT1 ChIP-seq in HeLa S3 cells [114] STAT1 ChIP in murine CD8 + T cells [85], STAT4 and STAT6 ChIP-seq in murine CD4 + T cells [115] OOPS in human and murine CD4 + T cells [59] STAT1 is important for T cell quiescence [85] STAT1 and STAT3 drive memory formation [85][86][87][88] STAT4 facilitates effector T cell differentiation [89] STAT3 and STAT5 regulate differentiation during chronic stimulation of T cells [90][91][92] SWI/SNF complex SMARCA4, SS18, ARID1A and PBRM1 CUT&Tag in human CD8 + T cells [60] SMARCA4 and ARID1A CUT&Run in murine CD8 + T cells [94] SMARCB1, SMARCA2, and SMARCA4 ChIP-seq in RWPE1 cells [61] SMARCA4 RIP-seq with UV crosslinking in 22Rv1 and LNCaP cells [61] ARID1A important for effector T cell [94] and terminally dysfunctional T cell differentiation [95] ARID2 drives pre-dysfunctional T cell formation during chronic activation [97] 7 ...
... ChIP-seq in A549 cells [108,109], murine macrophages [110], 3134 cells [111] RIP-seq with UV crosslinking in U2OS cells [112] RIP in BEAS-2B cells [77], rat smooth muscle cells [113] Important MPEC formation [81] Drives the dysfunctional phenotype in tumor-infiltrating T cells [82] STAT family STAT1 ChIP-seq in HeLa S3 cells [114] STAT1 ChIP in murine CD8 + T cells [85], STAT4 and STAT6 ChIP-seq in murine CD4 + T cells [115] OOPS in human and murine CD4 + T cells [59] STAT1 is important for T cell quiescence [85] STAT1 and STAT3 drive memory formation [85][86][87][88] STAT4 facilitates effector T cell differentiation [89] STAT3 and STAT5 regulate differentiation during chronic stimulation of T cells [90][91][92] SWI/SNF complex SMARCA4, SS18, ARID1A and PBRM1 CUT&Tag in human CD8 + T cells [60] SMARCA4 and ARID1A CUT&Run in murine CD8 + T cells [94] SMARCB1, SMARCA2, and SMARCA4 ChIP-seq in RWPE1 cells [61] SMARCA4 RIP-seq with UV crosslinking in 22Rv1 and LNCaP cells [61] ARID1A important for effector T cell [94] and terminally dysfunctional T cell differentiation [95] ARID2 drives pre-dysfunctional T cell formation during chronic activation [97] 7 ...
Article
Full-text available
Cellular differentiation allows cells to transition between different functional states and adapt to various environmental cues. The diversity and plasticity of this process is beautifully exemplified by T cells responding to pathogens, which undergo highly specialized differentiation tailored to the ongoing infection. Such antigen‐induced T cell differentiation is regulated at the transcriptional level by DNA‐binding proteins and at the post‐transcriptional level by RNA‐binding proteins. Although traditionally defined as separate protein classes, a growing body of evidence indicates an overlap between these two groups of proteins, collectively coined DNA/RNA‐binding proteins (DRBPs). In this review, we describe how DRBPs might bind both DNA and RNA, discuss the putative functional relevance of this dual binding, and provide an exploratory analysis into characteristics that are associated with DRBPs. To exemplify the significance of DRBPs in T cell biology, we detail the activity of several established and putative DRBPs during the T cell response. Finally, we highlight several methodologies that allow untangling of the distinct functionalities of DRBPs at the DNA and RNA level, including key considerations to take into account when applying such methods.
... While all of these mechanisms likely function together, they fail to adequately address the issue of cell type-specific and genome-wide SWI/SNF targeting. Various studies including our own have explored the roles of individual long non-coding RNAs (lncRNAs) in SWI/SNF genome targeting [21][22][23][24][25][26] . LncRNAs are increasingly understood to control gene expression, often through interactions with DNA-binding proteins and chromatin itself [27][28][29][30] . ...
... To corroborate the RedChIP findings and identify high-confidence SWI/SNF-RNA interactions for further stratification, we performed both RNA-immunoprecipitation followed by sequencing (RIP-seq) and individual-nucleotide resolution crosslinking and immunoprecipitation (iCLIP) 39,40 . In line with previous observations 26 , BRG1 RIP-seq identified many RNAs interacting with SWI/SNF. BRG1 iCLIP was performed to more accurately evaluate the RNA interactions. ...
Preprint
The coordination of chromatin remodeling is essential for DNA accessibility and gene expression control ¹ . The highly conserved and ubiquitously expressed SWItch/Sucrose Non-Fermentable (SWI/SNF) chromatin remodeling complex plays a central role in cell type- and context-dependent gene expression ² . Despite the absence of a defined DNA recognition motif, SWI/SNF binds lineage specific enhancers genome-wide where it actively maintains open chromatin state 2–5 . It does so while retaining the ability to respond dynamically to cellular signals ⁴ . However, the mechanisms that guide SWI/SNF to specific genomic targets have remained elusive. Here we demonstrate that trans -acting long non-coding RNAs (lncRNAs) direct the SWI/SNF complex to cell type-specific enhancers. SWI/SNF preferentially binds lncRNAs and these predominantly bind DNA targets in trans . Together they localize to enhancers, many of which are cell type-specific. Knockdown of SWI/SNF- and enhancer-bound lncRNAs causes the genome-wide redistribution of SWI/SNF away from enhancers and a concomitant differential expression of spatially connected target genes. These lncRNA-SWI/SNF-enhancer networks support an enhancer hub model of SWI/SNF genomic targeting. Our findings reveal a competitive recruitment of SWI/SNF by lncRNAs which provide a specific and dynamic layer of control in chromatin accessibility and gene expression.
... SCHLAP1 has been hypothesized to increase prostate cancer invasion and metastasis by a variety of pathways, including altering the SWI/ SNF complex (SWItch/Sucrose Non-Fermentable) complex's metastasis inhibiting action, which functions as a chromatin remodeler and tumor suppressor (12) . However, Schap1 tumorigenic effects were also found to be independent of SWI/SNF in a recent study (19) . Furthermore, by binding miR-198 and activating the MAPK signaling pathway, SChLAP1 might promote PCa cell proliferation and metastasis (20) . ...
... While all of these mechanisms likely function together, they fail to adequately address the issue of cell type-specific and genome-wide SWI/SNF targeting. Various studies including our own have explored the roles of individual long non-coding RNAs (lncRNAs) in SWI/SNF genome targeting [21][22][23][24][25][26] . LncRNAs are increasingly understood to control gene expression, often through interactions with DNA-binding proteins and chromatin itself [27][28][29][30] . ...
Article
Full-text available
The coordination of chromatin remodeling is essential for DNA accessibility and gene expression control. The highly conserved and ubiquitously expressed SWItch/Sucrose Non-Fermentable (SWI/SNF) chromatin remodeling complex plays a central role in cell type- and context-dependent gene expression. Despite the absence of a defined DNA recognition motif, SWI/SNF binds lineage specific enhancers genome-wide where it actively maintains open chromatin state. It does so while retaining the ability to respond dynamically to cellular signals. However, the mechanisms that guide SWI/SNF to specific genomic targets have remained elusive. Here we demonstrate that trans-acting long non-coding RNAs (lncRNAs) direct the SWI/SNF complex to cell type-specific enhancers. SWI/SNF preferentially binds lncRNAs and these predominantly bind DNA targets in trans. Together they localize to enhancers, many of which are cell type-specific. Knockdown of SWI/SNF- and enhancer-bound lncRNAs causes the genome-wide redistribution of SWI/SNF away from enhancers and a concomitant differential expression of spatially connected target genes. These lncRNA-SWI/SNF-enhancer networks support an enhancer hub model of SWI/SNF genomic targeting. Our findings reveal that lncRNAs competitively recruit SWI/SNF, providing a specific and dynamic layer of control over chromatin accessibility, and reinforcing their role in mediating enhancer activity and gene expression.
... RIPs were performed similar to Schertzer et al. (2019a), which is a protocol originally adapted from Hendrickson et al. (2016) and Raab et al. (2019). An amount of 25 µL protein A/G agarose beads (Santa Cruz sc-2003) were washed three times in blocking buffer (0.5% BSA in 1× PBS) and incubated overnight at 4°C with 10 µg antibody (anti-SAFB; Bethyl 812-300A; FLAG, Sigma F1804; V5, Sigma V8012; mouse IgG, Invitrogen 02-6502). ...
Article
Full-text available
Scaffold Attachment Factor B (SAFB) is a conserved RNA-binding protein that is essential for early mammalian development. However, the functions of SAFB in mouse embryonic stem cells (ESCs) have not been characterized. Using RNA immunoprecipitation followed by RNA-seq (RIP-seq), we examined the RNAs associated with SAFB in wild-type and SAFB/SAFB2 double-knockout ESCs. SAFB predominantly associated with introns of protein-coding genes through purine-rich motifs. The transcript most enriched in SAFB association was the lncRNA Malat1, which also contains a purine-rich region in its 5' end. Knockout of SAFB/SAFB2 led to differential expression of approximately 1,000 genes associated with multiple biological processes, including apoptosis, cell division, and cell migration. Knockout of SAFB/SAFB2 also led to splicing changes in a set of genes that were largely distinct from those that exhibited changes in expression level. The spliced and nascent transcripts of many genes whose expression levels were positively regulated by SAFB also associated with high levels of SAFB, implying that SAFB binding promotes their expression. Reintroduction of SAFB into double-knockout cells restored gene expression towards wild-type levels, an effect again observable at the level of spliced and nascent transcripts. Proteomics analysis revealed a significant enrichment of nuclear speckle-associated and RS domain-containing proteins among SAFB interactors. Neither Xist nor Polycomb function were dramatically altered in SAFB/2 knockout ESCs. Our findings suggest that among other potential functions in ESCs, SAFB promotes the expression of certain genes through its ability to bind nascent RNA.
... RIPs were performed similar to Schertzer et al. (2019a), which is a protocol originally adapted from Hendrickson et al. (2016); Raab et al. (2019). 25 μl protein A/G agarose beads (Santa Cruz Biotechnology, #sc-2003) were washed three times in blocking buffer (0.5% BSA in 1xPBS) and incubated overnight at 4°C with 10 μl antibody (anti-SAFB, Bethyl 812-300A; EZH2, Cell Signaling Technology, #5246) or 10μg IgG (Invitrogen, #02-6502). ...
Article
Full-text available
The Polycomb Repressive Complex 2 (PRC2) is a conserved enzyme that tri-methylates Lysine 27 on Histone 3 (H3K27me3) to promote gene silencing. PRC2 is remarkably responsive to the expression of certain long noncoding RNAs (lncRNAs). In the most notable example, PRC2 is recruited to the X-chromosome shortly after expression of the lncRNA Xist begins during X-chromosome inactivation. However, the mechanisms by which lncRNAs recruit PRC2 to chromatin are not yet clear. We report that a broadly used rabbit monoclonal antibody raised against human EZH2, a catalytic subunit of PRC2, cross-reacts with an RNA-binding protein called Scaffold Attachment Factor B (SAFB) in mouse embryonic stem cells (ESCs) under buffer conditions that are commonly used for chromatin immunoprecipitation (ChIP). Knockout of EZH2 in ESCs demonstrated that the antibody is specific for EZH2 by western blot (no cross-reactivity). Likewise, comparison to previously published datasets confirmed that the antibody recovers PRC2-bound sites by ChIP-Seq. However, RNA-IP from formaldehyde-crosslinked ESCs using ChIP wash conditions recovers distinct peaks of RNA association that co-localize with peaks of SAFB and whose enrichment disappears upon knockout of SAFB but not EZH2. IP and mass spectrometry-based proteomics in wild-type and EZH2 knockout ESCs confirm that the EZH2 antibody recovers SAFB in an EZH2-independent manner. Our data highlight the importance of orthogonal assays when studying interactions between chromatin-modifying enzymes and RNA.
Article
The mechanisms and biological roles of Polycomb repressive complex (PRC) recruitment by long noncoding RNAs (lncRNAs) remain unclear. To gain insight, we expressed two lncRNAs that recruit PRCs to multi-megabase domains, Airn and Xist, from an ectopic locus in mouse stem cells and compared effects. Unexpectedly, ectopic Airn recruited PRC1 and PRC2 to chromatin with a potency resembling Xist yet did not repress genes. Compared with PRC2, PRC1 was more proximal to Airn and Xist, where its enrichment over C-rich elements required the RNA-binding protein HNRNPK. Fusing Airn to Repeat A, the domain required for gene silencing by Xist, enabled gene silencing and altered local patterns but not relative levels of PRC-directed modifications. Our data suggest that, endogenously, Airn recruits PRCs to maintain rather than initiate gene silencing, that PRC recruitment occurs independently of Repeat A, and that protein-bridged interactions, not direct RNA contacts, underlie PRC recruitment by Airn, Xist, and other lncRNAs. Temporary link: https://authors.elsevier.com/c/1koB83vVUPVZhl
Article
This study aimed to investigate the expression and clinical significance of the long chain non-coding RNA SCHLAP1 in breast cancer tissues. The research included 60 breast cancer patients treated between June 2017 and September 2019. Cancer and adjacent tissues were collected for analysis. Furthermore, breast cancer cell lines MCF-7 and HCC1937, along with normal breast epithelial cell line MCF10A, were used to study the impact of LncRNA SCHLAP1 on breast cancer cell phenotypes. qRT-PCR was employed to measure LncRNA SCHLAP1 expression levels in cells and tissues. The results demonstrated that LncRNA SCHLAP1 was significantly up-regulated in breast cancer cells and patient tissues ( P <0.01). Moreover, differences in LncRNA SCHLAP1 expression were observed in patients with varying age, lymph node invasion, TNM staging, HER-2, and Ki-67 expression levels ( P <0.01). Patients with high LncRNA SCHLAP1 expression had a significantly lower two-year survival rate ( P <0.01). In vitro experiments revealed that down-regulated LncRNA SCHLAP1 inhibited the proliferation, migration, and invasion of MCF-7 cells, while promoting apoptosis ( P <0.01). This study suggests that LncRNA SCHLAP1 is associated with breast cancer progression and patient survival, serving as an independent predictor for breast cancer progression.
Article
Full-text available
Thoracic aortic aneurysm (TAA) has been associated with mutations affecting members of the TGF-β signaling pathway, or components and regulators of the vascular smooth muscle cell (VSMC) actomyosin cytoskeleton. Although both clinical groups present similar phenotypes, the existence of potential common mechanisms of pathogenesis remain obscure. Here we show that mutations affecting TGF-β signaling and VSMC cytoskeleton both lead to the formation of a ternary complex comprising the histone deacetylase HDAC9, the chromatin-remodeling enzyme BRG1, and the long noncoding RNA MALAT1. The HDAC9-MALAT1-BRG1 complex binds chromatin and represses contractile protein gene expression in association with gain of histone H3-lysine 27 trimethylation modifications. Disruption of Malat1 or Hdac9 restores contractile protein expression, improves aortic mural architecture, and inhibits experimental aneurysm growth. Thus, we highlight a shared epigenetic pathway responsible for VSMC dysfunction in both forms of TAA, with potential therapeutic implication for other known HDAC9-associated vascular diseases.
Article
Full-text available
BackgroundSWI/SNF is a large heterogeneous multi-subunit chromatin remodeling complex. It consists of multiple sets of mutually exclusive components. Understanding how loss of one sibling of a mutually exclusive pair affects the occupancy and function of the remaining complex is needed to understand how mutations in a particular subunit might affect tumor formation. Recently, we showed that the members of the ARID family of SWI/SNF subunits (ARID1A, ARID1B and ARID2) had complex transcriptional relationships including both antagonism and cooperativity. However, it remains unknown how loss of the catalytic subunit(s) affects the binding and genome-wide occupancy of the remainder complex and how changes in occupancy affect transcriptional output. ResultsWe addressed this gap by depleting BRG1 and BRM, the two ATPase subunits in SWI/SNF, and characterizing the changes to chromatin occupancy of the remaining subunit and related this to transcription changes induced by loss of the ATPase subunits. We show that depletion of one subunit frequently leads to loss of the remaining subunit. This could cause either positive or negative changes in gene expression. At a subset of sites, the sibling subunit is either retained or gained. Additionally, we show genome-wide that BRG1 and BRM have both cooperative and antagonistic interactions with respect to transcription. Importantly, at genes where BRG1 and BRM antagonize one another we observe a nearly complete rescue of gene expression changes in the combined BRG/BRM double knockdown. Conclusion This series of experiments demonstrate that mutually exclusive SWI/SNF complexes have heterogeneous functional relationships and highlight the importance of considering the role of the remaining SWI/SNF complexes following loss or depletion of a single subunit.
Article
Full-text available
Prostate cancer (PCa) is the leading cancer among men in the United States, with genetic factors contributing to ∼42% of the susceptibility to PCa. We analyzed a PCa risk region located at 7p15.2 to gain insight into the mechanisms by which this noncoding region may affect gene regulation and contribute to PCa risk. We performed Hi-C analysis and demonstrated that this region has long-range interactions with the HOXA locus, located ∼873 kb away. Using the CRISPR/Cas9 system, we deleted a 4-kb region encompassing several PCa risk-associated SNPs and performed RNA-seq to investigate transcriptomic changes in prostate cells lacking the regulatory element. Our results suggest that the risk element affects the expression of HOXA13 and HOTTIP, but not other genes in the HOXA locus, via a repressive loop. Forced expression of HOXA13 was performed to gain further insight into the mechanisms by which this risk element affects PCa risk.
Article
Full-text available
Perturbations to mammalian SWI/SNF (mSWI/SNF or BAF) complexes contribute to more than 20% of human cancers, with driving roles first identified in malignant rhabdoid tumor, an aggressive pediatric cancer characterized by biallelic inactivation of the core BAF complex subunit SMARCB1 (BAF47). However, the mechanism by which this alteration contributes to tumorigenesis remains poorly understood. We find that BAF47 loss destabilizes BAF complexes on chromatin, absent significant changes in complex assembly or integrity. Rescue of BAF47 in BAF47-deficient sarcoma cell lines results in increased genome-wide BAF complex occupancy, facilitating widespread enhancer activation and opposition of Polycomb-mediated repression at bivalent promoters. We demonstrate differential regulation by two distinct mSWI/SNF assemblies, BAF and PBAF complexes, enhancers and promoters, respectively, suggesting that each complex has distinct functions that are perturbed upon BAF47 loss. Our results demonstrate collaborative mechanisms of mSWI/SNF-mediated gene activation, identifying functions that are co-opted or abated to drive human cancers and developmental disorders.
Article
Full-text available
We present an update to our Galaxy-based web server for processing and visualizing deeply sequenced data. Its core tool set, deepTools, allows users to perform complete bioinformatic workflows ranging from quality controls and normalizations of aligned reads to integrative analyses, including clustering and visualization approaches. Since we first described our deepTools Galaxy server in 2014, we have implemented new solutions for many requests from the community and our users. Here, we introduce significant enhancements and new tools to further improve data visualization and interpretation. deepTools continue to be open to all users and freely available as a web service at deeptools.ie-freiburg.mpg.de. The new deepTools2 suite can be easily deployed within any Galaxy framework via the toolshed repository, and we also provide source code for command line usage under Linux and Mac OS X. A public and documented API for access to deepTools functionality is also available.
Article
Full-text available
Background Recent evidence suggests that RNA interaction can regulate the activity and localization of chromatin-associated proteins. However, it is unknown if these observations are specialized instances for a few key RNAs and chromatin factors in specific contexts, or a general mechanism underlying the establishment of chromatin state and regulation of gene expression. Results Here, we perform formaldehyde RNA immunoprecipitation (fRIP-Seq) to survey the RNA associated with a panel of 24 chromatin regulators and traditional RNA binding proteins. For each protein that reproducibly bound measurable quantities of bulk RNA (90 % of the panel), we detect enrichment for hundreds to thousands of both noncoding and mRNA transcripts. Conclusion For each protein, we find that the enriched sets of RNAs share distinct biochemical, functional, and chromatin properties. Thus, these data provide evidence for widespread specific and relevant RNA association across diverse classes of chromatin-modifying complexes. Electronic supplementary material The online version of this article (doi:10.1186/s13059-016-0878-3) contains supplementary material, which is available to authorized users.
Article
Full-text available
Multiple positions within the SWI/SNF chromatin remodeling complex can be filled by mutually exclusive subunits. Inclusion or exclusion of these proteins defines many unique forms of SWI/SNF and has profound functional consequences. Often this complex is studied as a single entity within a particular cell type and we understand little about the functional relationship between these biochemically distinct forms of the remodeling complex. Here we examine the functional relationships among three complex-specific ARID (AT-Rich Interacting Domain) subunits using genome-wide chromatin immunoprecipitation, transcriptome analysis, and transcription factor binding maps. We find widespread overlap in transcriptional regulation and the genomic binding of distinct SWI/SNF complexes. ARID1B and ARID2 participate in wide-spread cooperation to repress hundreds of genes. Additionally, we find numerous examples of competition between ARID1A and another ARID, and validate that gene expression changes following loss of one ARID are dependent on the function of an alternative ARID. These distinct regulatory modalities are correlated with differential occupancy by transcription factors. Together, these data suggest that distinct SWI/SNF complexes dictate gene-specific transcription through functional interactions between the different forms of the SWI/SNF complex and associated co-factors. Most genes regulated by SWI/SNF are controlled by multiple biochemically distinct forms of the complex, and the overall expression of a gene is the product of the interaction between these different SWI/SNF complexes. The three mutually exclusive ARID family members are among the most frequently mutated chromatin regulators in cancer, and understanding the functional interactions and their role in transcriptional regulation provides an important foundation to understand their role in cancer.
Article
Full-text available
Chromatin immunoprecipitation with massively parallel sequencing (ChIP-seq) is widely used to identify binding sites for a target protein in the genome. An important scientific application is to identify changes in protein binding between different treatment conditions, i.e. to detect differential binding. This can reveal potential mechanisms through which changes in binding may contribute to the treatment effect. The csaw package provides a framework for the de novo detection of differentially bound genomic regions. It uses a window-based strategy to summarize read counts across the genome. It exploits existing statistical software to test for significant differences in each window. Finally, it clusters windows into regions for output and controls the false discovery rate properly over all detected regions. The csaw package can handle arbitrarily complex experimental designs involving biological replicates. It can be applied to both transcription factor and histone mark datasets, and, more generally, to any type of sequencing data measuring genomic coverage. csaw performs favorably against existing methods for de novo DB analyses on both simulated and real data. csaw is implemented as a R software package and is freely available from the open-source Bioconductor project.
Article
Nucleosomes are the core units of cellular chromatin and are comprised of 147 base pairs (bp) of DNA wrapped around an octamer of histone proteins. Proteins such as chromatin remodelers, transcription factors, and DNA repair proteins interact dynamically with chromatin to regulate access to DNA, control gene transcription, and maintain genome integrity. The extent of association with chromatin changes rapidly in response to stresses, such as immune activation, oxidative stress, or viral infection, resulting in downstream effects on chromatin conformation and transcription of target genes. To elucidate changes in the composition of proteins associated with chromatin under different conditions, we adapted existing protocols to isolate nuclei and fractionate cellular chromatin using a gradient of salt concentrations. The presence of specific proteins in different salt fractions can be assessed by Western blotting or mass spectrometry, providing insight into the degree to which they are associated with chromatin.
Article
ARID1A, encoding the BAF250a subunit of SWI/SNF complex, has a high mutation frequency in numerous types of cancer. LncRNAs, a type of non-coding RNAs longer than 200 nucleotides, have been reported to interplay with SWI/SNF complex during cancer progression. However, whether the interaction between ARID1A and lncRNA affects hepatocellular carcinoma (HCC) still needs to be investigated. Here, we reveal that ARID1A interacts with lncRNA MVIH through some region(s) or domain(s) including ARID domain and C-terminal ARID1A protein binding domain. ARID1A upregulates its downstream target CDKN1A and suppresses HCC cell proliferation and migration through inhibiting MVIH. Our data suggests that deficiency or loss of functional mutations of ARID1A in HCC cells might contribute to the increased activity of certain cancer-promoting lncRNAs.