ARID1A facilitates KRAS signaling-regulated enhancer activity in an AP1-dependent manner in colorectal cancer cells

Madhobi Sen, Xin Wang, Feda H Hamdan, Jacobe Rapp, Jessica Eggert, Robyn Laura Kosinsky, Florian Wegwitz, Ana Patricia Kutschat, Fereshteh S Younesi, Jochen Gaedcke, Marian Grade, Elisabeth Hessmann, Argyris Papantonis, Philipp Strӧbel, Steven A Johnsen, Madhobi Sen, Xin Wang, Feda H Hamdan, Jacobe Rapp, Jessica Eggert, Robyn Laura Kosinsky, Florian Wegwitz, Ana Patricia Kutschat, Fereshteh S Younesi, Jochen Gaedcke, Marian Grade, Elisabeth Hessmann, Argyris Papantonis, Philipp Strӧbel, Steven A Johnsen

Abstract

Background: ARID1A (AT-rich interactive domain-containing protein 1A) is a subunit of the BAF chromatin remodeling complex and plays roles in transcriptional regulation and DNA damage response. Mutations in ARID1A that lead to inactivation or loss of expression are frequent and widespread across many cancer types including colorectal cancer (CRC). A tumor suppressor role of ARID1A has been established in a number of tumor types including CRC where the genetic inactivation of Arid1a alone led to the formation of invasive colorectal adenocarcinomas in mice. Mechanistically, ARID1A has been described to largely function through the regulation of enhancer activity.

Methods: To mimic ARID1A-deficient colorectal cancer, we used CRISPR/Cas9-mediated gene editing to inactivate the ARID1A gene in established colorectal cancer cell lines. We integrated gene expression analyses with genome-wide ARID1A occupancy and epigenomic mapping data to decipher ARID1A-dependent transcriptional regulatory mechanisms.

Results: Interestingly, we found that CRC cell lines harboring KRAS mutations are critically dependent on ARID1A function. In the absence of ARID1A, proliferation of these cell lines is severely impaired, suggesting an essential role for ARID1A in this context. Mechanistically, we showed that ARID1A acts as a co-factor at enhancers occupied by AP1 transcription factors acting downstream of the MEK/ERK pathway. Consistently, loss of ARID1A led to a disruption of KRAS/AP1-dependent enhancer activity, accompanied by a downregulation of expression of the associated target genes.

Conclusions: We identify a previously unknown context-dependent tumor-supporting function of ARID1A in CRC downstream of KRAS signaling. Upon the loss of ARID1A in KRAS-mutated cells, enhancers that are co-occupied by ARID1A and the AP1 transcription factors become inactive, thereby leading to decreased target gene expression. Thus, targeting of the BAF complex in KRAS-mutated CRC may offer a unique, previously unknown, context-dependent therapeutic option in CRC.

Keywords: AP1; ARID1A; BAF complex; Colorectal cancer; Enhancers; KRAS; MEK/ERK pathway; Transcriptional regulation.

Conflict of interest statement

The authors declare that they have no competing interests.

Figures

Fig. 1
Fig. 1
ARID1A protein loss in human cancer samples is consistent with mutation rates. ARID1A is mutated in up to ~ 12% colorectal cancers from different colorectal cancer cohorts [3, 4] (a). Representative immunohistochemical staining images show clear ARID1A staining in the rectal mucosa and positive tumors while negative tumors show no staining at all (b). The University Medical Center Gӧttingen rectal cancer cohort showed 81.7% strongly positive ARID1A staining (++) and 3.7% showed weakly positive staining (+) while 14.6 % of the samples were negative for ARID1A staining (c)
Fig. 2
Fig. 2
The loss of ARID1A impairs proliferation of a subset of colorectal cancer cell lines. To mimic ARID1A-deficient colorectal cancer, we used CRISPR/Cas9-mediated gene editing to delete exon 5 of the ARID1A gene from four colorectal cancer cell lines HCT116, DLD1, COLO320DM, and HT29. Regions flanking exon 5 were targeted by two guide RNAs leading to an out of frame gene product. Genotyping PCRs show a shorter product of 761 bp in the ARID1A knockout (KO) cells (a, middle panel). This led to a complete loss of the ARID1A protein from the KO cells (a, lower panel). HSC70 is used as a loading control. Representative graphs show that the proliferation of the HCT116 and DLD1 cell lines is significantly impaired by the deletion of ARID1A, whereas the proliferation of COLO320DM and HT29 is unaffected by this knockout (b). Error bars represent standard deviation between the replicates. n for proliferation assay = 2, *p < 0.05 unpaired t test
Fig. 3
Fig. 3
Loss of ARID1A results in deregulated expression of MEK/ERK pathway target genes. Forty-eight genes are commonly downregulated in the HCT116 and DLD1 cell lines (a). Gene set enrichment analysis shows that gene sets containing targets of the MEK/ERK pathway are enriched in the parental condition (a). The genes EREG, F3, and JAG1 are highly expressed in cell lines with KRASG13D mutations as analyzed using on the Morpheus tool to analyze data from the Cancer Cell Line Encyclopedia (CCLE) [36] (b). The scale represents the minimum expression in a particular row (blue) to the maximum expression in that row (red). Treatment with 20 nM trametinib for 24 h leads to a reduction of phosphorylated ERK (pERK) (c). The expression of EREG, F3, and JAG1 was significantly reduced by trametinib treatment and ARID1A deletion. Treatment with trametinib in ARID1A-deficient cells did not lead to a further reduction of gene expression (d). ARID1A mutations are significantly mutually exclusive with KRAS mutations in the TCGA colorectal cancer patient cohort (e). The y-axis represents −log10 (p value) and the blue dots represent significantly mutually exclusive or co-occurring mutations. pERK and JUND levels remain unchanged in ARID1A-deficient cells. HSC70 is used as a loading control. The AP1 transcription network gene set [41] is enriched in the parental condition (f). qRT-PCRs were performed for biological triplicates and technical duplicates. Error bars represent the standard deviation between three biological replicates. Significance was calculated using an unpaired t test, *p < 0.05, **p < 0.01, ***p < 0.001
Fig. 4
Fig. 4
ARID1A is localized to AP1-occupied enhancers. ARID1A co-localizes with the BAF complex subunits SMARCA4 and SMARCC1 as well as the active mark H3K27ac. The heatmaps represent all ARID1A binding sites ordered in descending order of ARID1A occupancy (a). ARID1A localizes mainly to regions that are distal to transcription start sites (TSS) with the maximum number of peaks between 5 and 500 kb of TSSs (b). ARID1A-bound enhancers were defined based on overlap of ARID1A with H3K27ac and ATAC-seq. From these regions, any annotated TSSs were subtracted and 3061 ARID1A-bound enhancers were identified (c). Motif analysis on ARID1A-bound enhancers shows enrichment for the Jun-AP1(bZIP) motif (d). ReMap analysis shows that several AP1 transcription factors such as FOSL1, FOSL2, FOS, and JUND are enriched on ARID1A-occupied regions identified in HCT116 cells (e). Co-localization of JUND and FOSL1 at ARID1A-bound enhancers in HCT116 cells is shown. The heatmaps represent the 3061 ARID1A-bound enhancers in descending order of ARID1A occupancy (f)
Fig. 5
Fig. 5
The loss of ARID1A leads to a loss of enhancer activity. Six thousand seven hundred forty-one genes associated with ARID1A-bound enhancers were determined. Three hundred seventeen genes were also downregulated by the knockout of ARID1A. Among these were the EREG, F3, and JAG1 genes (a). The three MEK/ERK pathway target genes identified were downregulated by the deletion of ARID1A in both KRAS-mutated cell lines (HCT116 and DLD1) (b). At the genomic loci for these genes, we identified potential enhancers which are occupied by ARID1A, JUND, and H3K27ac within the same topologically associated domains (TAD). Vertical dotted lines represent TAD boundaries. The potential enhancers are also accessible as assessed by ATAC-seq and are transcriptionally active as assessed by PRO-seq (c). There is a significant reduction of H3K27ac at these enhancers upon the deletion of ARID1A (c, d). qRT-PCRs for gene expression and ChIP were run in biological triplicates and technical duplicates. The dotted lines represent the average ChIP-qPCR signal for the negative control IgG. Error bars represent the standard deviation between three biological replicates. Significance was calculated using unpaired t test, *p < 0.05, **p < 0.01, ***p < 0.001
Fig. 6
Fig. 6
The attenuation of the MEK/ERK pathway leads to a loss of enhancer activity at ARID1A-bound enhancers. Upon treatment with 20 nM trametinib for 24 h, the occupancy of JUND was reduced at the potential enhancers identified (a). Moreover, the same treatment also leads to the reduction of ARID1A occupancy and a loss of the H3K27ac mark on these enhancers (b, c). ChIP was carried out in biological triplicates and qRT-PCRs were run in technical duplicates. The dotted lines represent the average ChIP-qPCR signal for the negative control IgG. Error bars represent the standard deviation between three biological replicates. Significance was calculated using unpaired t test, *p < 0.05, **p < 0.01, ***p < 0.001. In summary, we show that the loss of ARID1A has context-dependent functions in colorectal cancer. Mathur et al. describe that in a wild-type background, the loss of ARID1A is tumor suppressive and leads to the formation of invasive adenocarcinomas. This function is mediated through control of enhancer activity wherein there is a loss of the active H3K27ac mark upon the loss of ARID1A [21]. We show that in colorectal cancer cells which harbor KRAS mutations, ARID1A has a tumor-supporting function by facilitating transcription at enhancers downstream of the MEK/ERK pathway also partially through the placement of the H3K27ac mark (d). In the first case, synthetic lethal targets described in the literature may provide potential therapeutic targets [, , –54]. In the case described here, targeting the BAF complex itself using PROTACS could be an option

References

    1. GLOBOCAN 2018 . Global Health Observatory. Geneva: World Health Organization; 2018. World Health Organization.
    1. Fearon Eric R., Vogelstein Bert. A genetic model for colorectal tumorigenesis. Cell. 1990;61(5):759–767. doi: 10.1016/0092-8674(90)90186-I.
    1. Cerami E, Gao J, Dogrusoz U, Gross BE, Sumer SO, Aksoy BA, et al. The cBio cancer genomics portal: an open platform for exploring multidimensional cancer genomics data. Cancer Discov. 2012;2.
    1. Gao J., Aksoy B. A., Dogrusoz U., Dresdner G., Gross B., Sumer S. O., Sun Y., Jacobsen A., Sinha R., Larsson E., Cerami E., Sander C., Schultz N. Integrative Analysis of Complex Cancer Genomics and Clinical Profiles Using the cBioPortal. Science Signaling. 2013;6(269):pl1–pl1. doi: 10.1126/scisignal.2004088.
    1. Karnoub AE, Weinberg RA. Ras oncogenes: split personalities. Nat Rev Mol Cell Biol. 2008;9.
    1. Whitmarsh A. J., Davis R. J. Transcription factor AP-1 regulation by mitogen-activated protein kinase signal transduction pathways. Journal of Molecular Medicine. 1996;74(10):589–607. doi: 10.1007/s001090050063.
    1. Whitmarsh Alan J. Regulation of gene transcription by mitogen-activated protein kinase signaling pathways. Biochimica et Biophysica Acta (BBA) - Molecular Cell Research. 2007;1773(8):1285–1298. doi: 10.1016/j.bbamcr.2006.11.011.
    1. Nie Z, Xue Y, Yang D, Zhou S, Deroo BJ, Archer TK, et al. A specificity and targeting subunit of a human SWI/SNF family-related chromatin-remodeling complex. Mol Cell Biol 2000; 20.
    1. Dallas Peter B., Cheney Ian Wayne, Liao Da-Wei, Bowrin Valerie, Byam Whitney, Pacchione Stephen, Kobayashi Ryuji, Yaciuk Peter, Moran Elizabeth. p300/CREB Binding Protein-Related Protein p270 Is a Component of Mammalian SWI/SNF Complexes. Molecular and Cellular Biology. 1998;18(6):3596–3603. doi: 10.1128/MCB.18.6.3596.
    1. Kwon Hyockman, Imbalzano Anthony N., Khavari Paul A., Kingston Robert E., Green Michael R. Nucleosome disruption and enhancement of activator binding by a human SW1/SNF complex. Nature. 1994;370(6489):477–481. doi: 10.1038/370477a0.
    1. Imbalzano Anthony N., Kwon Hyockman, Green Michael R., Kingston Robert E. Facilitated binding of TATA-binding protein to nucleosomal DNA. Nature. 1994;370(6489):481–485. doi: 10.1038/370481a0.
    1. Phelan Michael L, Sif Saïd, Narlikar Geeta J, Kingston Robert E. Reconstitution of a Core Chromatin Remodeling Complex from SWI/SNF Subunits. Molecular Cell. 1999;3(2):247–253. doi: 10.1016/S1097-2765(00)80315-9.
    1. Chi Tian H., Wan Mimi, Zhao Keji, Taniuchi Ichiro, Chen Lei, Littman Dan R., Crabtree Gerald R. Reciprocal regulation of CD4/CD8 expression by SWI/SNF-like BAF complexes. Nature. 2002;418(6894):195–199. doi: 10.1038/nature00876.
    1. Wang Xi, Werneck Miriam B.F., Wilson Boris G., Kim Hye-Jung, Kluk Michael J., Thom Christopher S., Wischhusen Jonathan W., Evans Julia A., Jesneck Jonathan L., Nguyen Phuong, Sansam Courtney G., Cantor Harvey, Roberts Charles W.M. TCR-dependent transformation of mature memory phenotype T cells in mice. Journal of Clinical Investigation. 2011;121(10):3834–3845. doi: 10.1172/JCI37210.
    1. Gresh L, Bourachot B, Reimann A, Guigas B, Fiette L, Garbay S, et al. The SWI/SNF chromatin-remodeling complex subunit SNF5 is essential for hepatocyte differentiation. EMBO J. 2005;24.
    1. Gao X, Tate P, Hu P, Tjian R, Skarnes WC, Wang Z. ES cell pluripotency and germ-layer formation require the SWI/SNF chromatin remodeling component BAF250a. Proc Natl Acad Sci USA. 2008;105.
    1. Ho L., Jothi R., Ronan J. L., Cui K., Zhao K., Crabtree G. R. An embryonic stem cell chromatin remodeling complex, esBAF, is an essential component of the core pluripotency transcriptional network. Proceedings of the National Academy of Sciences. 2009;106(13):5187–5191. doi: 10.1073/pnas.0812888106.
    1. Kadoch Cigall, Hargreaves Diana C, Hodges Courtney, Elias Laura, Ho Lena, Ranish Jeff, Crabtree Gerald R. Proteomic and bioinformatic analysis of mammalian SWI/SNF complexes identifies extensive roles in human malignancy. Nature Genetics. 2013;45(6):592–601. doi: 10.1038/ng.2628.
    1. Shain A. Hunter, Pollack Jonathan R. The Spectrum of SWI/SNF Mutations, Ubiquitous in Human Cancers. PLoS ONE. 2013;8(1):e55119. doi: 10.1371/journal.pone.0055119.
    1. Guan B., Wang T.-L., Shih I.-M. ARID1A, a Factor That Promotes Formation of SWI/SNF-Mediated Chromatin Remodeling, Is a Tumor Suppressor in Gynecologic Cancers. Cancer Research. 2011;71(21):6718–6727. doi: 10.1158/0008-5472.CAN-11-1562.
    1. Mathur R, Alver BH, San Roman AK, Wilson BG, Wang X, Agoston AT, et al. ARID1A loss impairs enhancer-mediated gene regulation and drives colon cancer in mice. Nat Genet. 2017;49.
    1. Chandler RL, Damrauer JS, Raab JR, Schisler JC, Wilkerson MD, Didion JP, et al. Coexistent ARID1A-PIK3CA mutations promote ovarian clear-cell tumorigenesis through pro-tumorigenic inflammatory cytokine signalling. Nat Commun. 2015;6.
    1. Livshits G, Alonso-curbelo D, Iv JPM, Koche R, Saborowski M, Wilkinson JE, et al. Arid1a restrains Kras-dependent changes in acinar cell identity. eLife. 2018;7.
    1. Huang Jianmin, Zhao Yan-Ling, Li Yu, Fletcher Jonathan A., Xiao Sheng. Genomic and functional evidence for anARID1A tumor suppressor role. Genes, Chromosomes and Cancer. 2007;46(8):745–750. doi: 10.1002/gcc.20459.
    1. Lowery William J., Schildkraut Joellen M., Akushevich Liudmila, Bentley Rex, Marks Jeffrey R., Huntsman David, Berchuck Andrew. Loss of ARID1A-Associated Protein Expression is a Frequent Event in Clear Cell and Endometrioid Ovarian Cancers. International Journal of Gynecologic Cancer. 2012;22(1):9–14. doi: 10.1097/IGC.0b013e318231f140.
    1. Shen J., Peng Y., Wei L., Zhang W., Yang L., Lan L., Kapoor P., Ju Z., Mo Q., Shih I.-M., Uray I. P., Wu X., Brown P. H., Shen X., Mills G. B., Peng G. ARID1A Deficiency Impairs the DNA Damage Checkpoint and Sensitizes Cells to PARP Inhibitors. Cancer Discovery. 2015;5(7):752–767. doi: 10.1158/-14-0849.
    1. Wang X, Lee RS, Alver BH, Haswell JR, Wang S, Mieczkowski J, et al. SMARCB1-mediated SWI/SNF complex function is essential for enhancer regulation. Nat Genet. 2017;49.
    1. Vierbuchen Thomas, Ling Emi, Cowley Christopher J., Couch Cameron H., Wang Xiaofeng, Harmin David A., Roberts Charles W.M., Greenberg Michael E. AP-1 Transcription Factors and the BAF Complex Mediate Signal-Dependent Enhancer Selection. Molecular Cell. 2017;68(6):1067-1082.e12. doi: 10.1016/j.molcel.2017.11.026.
    1. Lakshminarasimhan Ranjani, Andreu-Vieyra Claudia, Lawrenson Kate, Duymich Christopher E., Gayther Simon A., Liang Gangning, Jones Peter A. Down-regulation of ARID1A is sufficient to initiate neoplastic transformation along with epigenetic reprogramming in non-tumorigenic endometriotic cells. Cancer Letters. 2017;401:11–19. doi: 10.1016/j.canlet.2017.04.040.
    1. Hu G., Schones D. E., Cui K., Ybarra R., Northrup D., Tang Q., Gattinoni L., Restifo N. P., Huang S., Zhao K. Regulation of nucleosome landscape and transcription factor targeting at tissue-specific enhancers by BRG1. Genome Research. 2011;21(10):1650–1658. doi: 10.1101/gr.121145.111.
    1. 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 Inc. 2012:152.
    1. Alver BH, Kim KH, Lu P, Wang X, Manchester HE, Wang W, et al. The SWI/SNF chromatin remodelling complex is required for maintenance of lineage specific enhancers. Nat Commun. 2017;8.
    1. Heinz Sven, Romanoski Casey E., Benner Christopher, Glass Christopher K. The selection and function of cell type-specific enhancers. Nature Reviews Molecular Cell Biology. 2015;16(3):144–154. doi: 10.1038/nrm3949.
    1. Raab Jesse R., Resnick Samuel, Magnuson Terry. Genome-Wide Transcriptional Regulation Mediated by Biochemically Distinct SWI/SNF Complexes. PLOS Genetics. 2015;11(12):e1005748. doi: 10.1371/journal.pgen.1005748.
    1. Shema-Yaacoby Efrat, Nikolov Miroslav, Haj-Yahya Mahmood, Siman Peter, Allemand Eric, Yamaguchi Yuki, Muchardt Christian, Urlaub Henning, Brik Ashraf, Oren Moshe, Fischle Wolfgang. Systematic Identification of Proteins Binding to Chromatin-Embedded Ubiquitylated H2B Reveals Recruitment of SWI/SNF to Regulate Transcription. Cell Reports. 2013;4(3):601–608. doi: 10.1016/j.celrep.2013.07.014.
    1. 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.
    1. Jones Siân, Li Meng, Parsons D. Williams, Zhang Xiaosong, Wesseling Jelle, Kristel Petra, Schmidt Marjanka K., Markowitz Sanford, Yan Hai, Bigner Darell, Hruban Ralph H., Eshleman James R., Iacobuzio-Donahue Christine A., Goggins Michael, Maitra Anirban, Malek Sami N., Powell Steve, Vogelstein Bert, Kinzler Kenneth W., Velculescu Victor E., Papadopoulos Nickolas. Somatic mutations in the chromatin remodeling gene ARID1A occur in several tumor types. Human Mutation. 2011;33(1):100–103. doi: 10.1002/humu.21633.
    1. Wang Xiaomei, Nagl Norman G., Flowers Stephen, Zweitzig Daniel, Dallas Peter B., Moran Elizabeth. Expression of p270(ARID1A), a component of human SWI/SNF complexes, in human tumors. International Journal of Cancer. 2004;112(4):636–642. doi: 10.1002/ijc.20450.
    1. MORPHEUS, .
    1. Toyoda Hitoshi, Komurasaki Toshi, Uchida Daisuke, Takayama Yasuko, Isobe Toshiaki, Okuyama Tuneo, Hanada Kazunori. Epiregulin. Journal of Biological Chemistry. 1995;270(13):7495–7500. doi: 10.1074/jbc.270.13.7495.
    1. Kuramochi H, Nakajima G, Kaneko Y, Nakamura A, Inoue Y, Yamamoto M. Amphiregulin and Epiregulin mRNA expression in primary colorectal cancer and corresponding liver metastases. BMC Cancer. 2012;12:88.
    1. Morrow JJ, Bayles I, Funnell APW, Miller TE, Saiakhova A, Lizardo MM, et al. Positively selected enhancer elements endow osteosarcoma cells with metastatic competence. Nat Med. 2018;24(2):176-85.
    1. Sugiyama Masakazu, Oki Eiji, Nakaji Yu, Tsutsumi Satoshi, Ono Naomi, Nakanishi Ryota, Sugiyama Masahiko, Nakashima Yuichiro, Sonoda Hideto, Ohgaki Kippei, Yamashita Nami, Saeki Hiroshi, Okano Shinji, Kitao Hiroyuki, Morita Masaru, Oda Yoshinao, Maehara Yoshihiko. High expression of the Notch ligand Jagged-1 is associated with poor prognosis after surgery for colorectal cancer. Cancer Science. 2016;107(11):1705–1716. doi: 10.1111/cas.13075.
    1. Rouillard AD, Monteiro CD, Gundersen GW, McDermott MG, Fernandez NF, Wang Z, et al. The harmonizome: a collection of processed datasets gathered to serve and mine knowledge about genes and proteins. Database (Oxford). 2016.
    1. Zirkel Anne, Nikolic Milos, Sofiadis Konstantinos, Mallm Jan-Philipp, Brackley Chris A., Gothe Henrike, Drechsel Oliver, Becker Christian, Altmüller Janine, Josipovic Natasa, Georgomanolis Theodore, Brant Lilija, Franzen Julia, Koker Mirjam, Gusmao Eduardo G., Costa Ivan G., Ullrich Roland T., Wagner Wolfgang, Roukos Vassilis, Nürnberg Peter, Marenduzzo Davide, Rippe Karsten, Papantonis Argyris. HMGB2 Loss upon Senescence Entry Disrupts Genomic Organization and Induces CTCF Clustering across Cell Types. Molecular Cell. 2018;70(4):730-744.e6. doi: 10.1016/j.molcel.2018.03.030.
    1. Trizzino Marco, Barbieri Elisa, Petracovici Ana, Wu Shuai, Welsh Sarah A., Owens Tori A., Licciulli Silvia, Zhang Rugang, Gardini Alessandro. The Tumor Suppressor ARID1A Controls Global Transcription via Pausing of RNA Polymerase II. Cell Reports. 2018;23(13):3933–3945. doi: 10.1016/j.celrep.2018.05.097.
    1. Kelso TWR, Porter DK, Amaral ML, Shokhirev MN, Benner C, Hargreaves DC. Chromatin accessibility underlies synthetic lethality of SWI/SNF subunits in ARID1A-mutant cancers. elife. 2017;6.
    1. Zhai Yali, Kuick Rork, Tipton Courtney, Wu Rong, Sessine Michael, Wang Zhong, Baker Suzanne J, Fearon Eric R, Cho Kathleen R. Arid1ainactivation in anApc- andPten-defective mouse ovarian cancer model enhances epithelial differentiation and prolongs survival. The Journal of Pathology. 2015;238(1):21–30. doi: 10.1002/path.4599.
    1. Sun X, Wang SC, Wei Y, Yopp AC, Singal AG, Zhu H. Tumor Suppressor Functions in Liver Cancer Article Arid1a has context-dependent ncogenic and tumor suppressor functions in liver cancer. Cancer Cell2017;32.
    1. Holik Aliaksei Z., Young Madeleine, Krzystyniak Joanna, Williams Geraint T., Metzger Daniel, Shorning Boris Y., Clarke Alan R. Brg1 Loss Attenuates Aberrant Wnt-Signalling and Prevents Wnt-Dependent Tumourigenesis in the Murine Small Intestine. PLoS Genetics. 2014;10(7):e1004453. doi: 10.1371/journal.pgen.1004453.
    1. Bitler Benjamin G, Aird Katherine M, Garipov Azat, Li Hua, Amatangelo Michael, Kossenkov Andrew V, Schultz David C, Liu Qin, Shih Ie-Ming, Conejo-Garcia Jose R, Speicher David W, Zhang Rugang. Synthetic lethality by targeting EZH2 methyltransferase activity in ARID1A-mutated cancers. Nature Medicine. 2015;21(3):231–238. doi: 10.1038/nm.3799.
    1. Kwan SY, Cheng X, Tsang YTM, Choi J-S, Kwan S-Y, Izaguirre DI, et al. Loss of ARID1A expression leads to sensitivity to ROS-inducing agent elesclomol in gynecologic cancer cells. Oncotarget. 2016;7.
    1. Bitler Benjamin G., Wu Shuai, Park Pyoung Hwa, Hai Yang, Aird Katherine M., Wang Yemin, Zhai Yali, Kossenkov Andrew V., Vara-Ailor Ana, Rauscher III Frank J., Zou Weiping, Speicher David W., Huntsman David G., Conejo-Garcia Jose R., Cho Kathleen R., Christianson David W., Zhang Rugang. ARID1A-mutated ovarian cancers depend on HDAC6 activity. Nature Cell Biology. 2017;19(8):962–973. doi: 10.1038/ncb3582.
    1. Samartzis EP, Gutsche K, Dedes KJ, Fink D, Stucki M, Imesch P. Loss of ARID1A expression sensitizes cancer cells to PI3K- and AKT-inhibition. Oncotarget. 2014;5.
    1. Michel Brittany C., D’Avino Andrew R., Cassel Seth H., Mashtalir Nazar, McKenzie Zachary M., McBride Matthew J., Valencia Alfredo M., Zhou Qianhe, Bocker Michael, Soares Luis M. M., Pan Joshua, Remillard David I., Lareau Caleb A., Zullow Hayley J., Fortoul Nora, Gray Nathanael S., Bradner James E., Chan Ho Man, Kadoch Cigall. A non-canonical SWI/SNF complex is a synthetic lethal target in cancers driven by BAF complex perturbation. Nature Cell Biology. 2018;20(12):1410–1420. doi: 10.1038/s41556-018-0221-1.
    1. Tian B., Zhao Y., Kalita M., Edeh C. B., Paessler S., Casola A., Teng M. N., Garofalo R. P., Brasier A. R. CDK9-Dependent Transcriptional Elongation in the Innate Interferon-Stimulated Gene Response to Respiratory Syncytial Virus Infection in Airway Epithelial Cells. Journal of Virology. 2013;87(12):7075–7092. doi: 10.1128/JVI.03399-12.
    1. Langmead Ben, Salzberg Steven L. Fast gapped-read alignment with Bowtie 2. Nature Methods. 2012;9(4):357–359. doi: 10.1038/nmeth.1923.
    1. Trapnell Cole, Roberts Adam, Goff Loyal, Pertea Geo, Kim Daehwan, Kelley David R, Pimentel Harold, Salzberg Steven L, Rinn John L, Pachter Lior. Differential gene and transcript expression analysis of RNA-seq experiments with TopHat and Cufflinks. Nature Protocols. 2012;7(3):562–578. doi: 10.1038/nprot.2012.016.
    1. Li H, Handsaker B, Wysoker A, Fennell T, Ruan J, Homer N, et al. The sequence alignment/map format and SAMtools. Bioinformatics. 2009;(16).
    1. Ramírez Fidel, Ryan Devon P, Grüning Björn, Bhardwaj Vivek, Kilpert Fabian, Richter Andreas S, Heyne Steffen, Dündar Friederike, Manke Thomas. deepTools2: a next generation web server for deep-sequencing data analysis. Nucleic Acids Research. 2016;44(W1):W160–W165. doi: 10.1093/nar/gkw257.
    1. Robinson James T, Thorvaldsdóttir Helga, Winckler Wendy, Guttman Mitchell, Lander Eric S, Getz Gad, Mesirov Jill P. Integrative genomics viewer. Nature Biotechnology. 2011;29(1):24–26. doi: 10.1038/nbt.1754.
    1. Zhang Yong, Liu Tao, Meyer Clifford A, Eeckhoute Jérôme, Johnson David S, Bernstein Bradley E, Nussbaum Chad, Myers Richard M, Brown Myles, Li Wei, Liu X Shirley. Model-based Analysis of ChIP-Seq (MACS) Genome Biology. 2008;9(9):R137. doi: 10.1186/gb-2008-9-9-r137.
    1. Subramanian A, Tamayo P, Mootha VK, Mukherjee S, Ebert BL. Gene set enrichment analysis: A knowledge-based approach for interpreting genome-wide. Proc Natl Acad Sci. 2005;102(43):15545-50.
    1. Mootha Vamsi K, Lindgren Cecilia M, Eriksson Karl-Fredrik, Subramanian Aravind, Sihag Smita, Lehar Joseph, Puigserver Pere, Carlsson Emma, Ridderstråle Martin, Laurila Esa, Houstis Nicholas, Daly Mark J, Patterson Nick, Mesirov Jill P, Golub Todd R, Tamayo Pablo, Spiegelman Bruce, Lander Eric S, Hirschhorn Joel N, Altshuler David, Groop Leif C. PGC-1α-responsive genes involved in oxidative phosphorylation are coordinately downregulated in human diabetes. Nature Genetics. 2003;34(3):267–273. doi: 10.1038/ng1180.
    1. Kuleshov Maxim V., Jones Matthew R., Rouillard Andrew D., Fernandez Nicolas F., Duan Qiaonan, Wang Zichen, Koplev Simon, Jenkins Sherry L., Jagodnik Kathleen M., Lachmann Alexander, McDermott Michael G., Monteiro Caroline D., Gundersen Gregory W., Ma'ayan Avi. Enrichr: a comprehensive gene set enrichment analysis web server 2016 update. Nucleic Acids Research. 2016;44(W1):W90–W97. doi: 10.1093/nar/gkw377.
    1. Chen EY, Tan CM, Kou Y, Duan Q, Wang Z, Meirelles GV, et al. Enrichr: interactive and collaborative HTML5 gene list enrichment analysis tool. BMC BIoinformatics. 2013.
    1. Heinz Sven, Benner Christopher, Spann Nathanael, Bertolino Eric, Lin Yin C., Laslo Peter, Cheng Jason X., Murre Cornelis, Singh Harinder, Glass Christopher K. Simple Combinations of Lineage-Determining Transcription Factors Prime cis-Regulatory Elements Required for Macrophage and B Cell Identities. Molecular Cell. 2010;38(4):576–589. doi: 10.1016/j.molcel.2010.05.004.
    1. McLean Cory Y, Bristor Dave, Hiller Michael, Clarke Shoa L, Schaar Bruce T, Lowe Craig B, Wenger Aaron M, Bejerano Gill. GREAT improves functional interpretation of cis-regulatory regions. Nature Biotechnology. 2010;28(5):495–501. doi: 10.1038/nbt.1630.
    1. Chèneby Jeanne, Gheorghe Marius, Artufel Marie, Mathelier Anthony, Ballester Benoit. ReMap 2018: an updated atlas of regulatory regions from an integrative analysis of DNA-binding ChIP-seq experiments. Nucleic Acids Research. 2017;46(D1):D267–D275. doi: 10.1093/nar/gkx1092.
    1. Ramírez F, Bhardwaj V, Arrigoni L, Lam KC, Grüning BA, Villaveces J, et al. High-resolution TADs reveal sequences.
    1. Li H., Durbin R. Fast and accurate short read alignment with Burrows-Wheeler transform. Bioinformatics. 2009;25(14):1754–1760. doi: 10.1093/bioinformatics/btp324.
    1. Chu Tinyi, Rice Edward J., Booth Gregory T., Salamanca H. Hans, Wang Zhong, Core Leighton J., Longo Sharon L., Corona Robert J., Chin Lawrence S., Lis John T., Kwak Hojoong, Danko Charles G. Chromatin run-on and sequencing maps the transcriptional regulatory landscape of glioblastoma multiforme. Nature Genetics. 2018;50(11):1553–1564. doi: 10.1038/s41588-018-0244-3.
    1. Gertz J, Savic D, Varley KE, Partridge EC, Safi A, Jain P, et al. Distinct properties of cell-type-specific and shared transcription factor binding sites. Mol Cell. 2013;52.
    1. Rao SSP, Huang S-C, Glenn St Hilaire B, Engreitz JM, Perez EM, Kieffer-Kwon KR, et al. Cohesin loss eliminates all loop domains. Cell. 2017:171.
    1. Ran F Ann, Hsu Patrick D, Wright Jason, Agarwala Vineeta, Scott David A, Zhang Feng. Genome engineering using the CRISPR-Cas9 system. Nature Protocols. 2013;8(11):2281–2308. doi: 10.1038/nprot.2013.143.

Source: PubMed

3
Sottoscrivi