Capped nascent RNA sequencing reveals novel therapy-responsive enhancers in prostate cancer

Mounting evidence suggests that enhancer RNA (eRNA) transcription start sites (TSSs) provide higher sensitivity and specificity for enhancer identification than histone modifications and chromatin accessibility. The extent to which changes in eRNA transcription correspond to changes in enhancer activity, however, remains unclear. Here, we used precision run-on and capped RNA sequencing (PRO-cap) to assess changes in enhancer activity in response to treatment with the androgen receptor signaling inhibitor, enzalutamide (ENZ). We identified 6,189 high-confidence candidate enhancers in the human prostate cancer cell line, LNCaP; 853 of which demonstrated significant changes in activity in response to drug treatment. Notably, we found that 67% and 54% of drug-responsive enhancers did not show similar changes in activity in previous studies that utilized ChIP-seq and ATAC-seq, respectively. Strikingly, 79% of regions with increased eRNA transcription showed no other biochemical alterations, implying that PRO-cap can capture a set of precise changes in enhancer activity that classical approaches lack the sensitivity to detect. We performed in vivo functional validations of candidate enhancers and found that CRISPRi targeting of PRO-cap-specific drug-responsive enhancers impaired ENZ regulation of downstream target genes, suggesting that changes in eRNA TSSs mark true biological changes in enhancer activity with high sensitivity. Our study highlights the utility of using PRO-cap as a complementary approach to canonical biochemical methods for detecting precise changes in enhancer activity and, in particular, for better understanding disease progression and responses to treatment.

[1]  Haiyuan Yu,et al.  A comparison of experimental assays and analytical methods for genome-wide identification of active enhancers , 2022, Nature Biotechnology.

[2]  K. D. Sørensen,et al.  Trans-ancestry genome-wide association meta-analysis of prostate cancer identifies new susceptibility loci and informs genetic risk prediction , 2021, Nature genetics.

[3]  Sylvan C. Baca,et al.  H3k27ac-HiChIP in prostate cell lines identifies risk genes for prostate cancer susceptibility , 2020 .

[4]  Han Liang,et al.  A High-Resolution Map of Human Enhancer RNA Loci Characterizes Super-enhancer Activities in Cancer. , 2020, Cancer cell.

[5]  H. G. van der Poel,et al.  Functional mapping of androgen receptor enhancer activity , 2020, Genome Biology.

[6]  Fidencio J. Neri,et al.  Index and biological spectrum of human DNase I hypersensitive sites , 2020, Nature.

[7]  Henry W. Long,et al.  Prostate cancer reactivates developmental epigenomic programs during metastatic progression , 2020, Nature Genetics.

[8]  Michael Q. Zhang,et al.  The landscape of RNA polymerase II associated chromatin interactions in prostate cancer. , 2020, The Journal of clinical investigation.

[9]  Castration Resistance , 2020, Definitions.

[10]  J. Shendure,et al.  Towards a comprehensive catalogue of validated and target-linked human enhancers , 2020, Nature Reviews Genetics.

[11]  Runan Yao,et al.  ShinyGO: a graphical gene-set enrichment tool for animals and plants , 2019, Bioinform..

[12]  B. Győrffy,et al.  TLE3 loss confers AR inhibitor resistance by facilitating GR-mediated human prostate cancer cell growth , 2019, eLife.

[13]  Henry W. Long,et al.  CREB5 Promotes Resistance to Androgen-Receptor Antagonists and Androgen Deprivation in Prostate Cancer , 2019, Cell reports.

[14]  Shayne D. Wierbowski,et al.  Transcription imparts architecture, function, and logic to enhancer units , 2019, Nature Genetics.

[15]  P. Farnham,et al.  A high-resolution 3D epigenomic map reveals insights into the creation of the prostate cancer transcriptome , 2019, Nature Communications.

[16]  I. Asangani,et al.  CDK7 inhibition suppresses Castration-Resistant Prostate Cancer through MED1 inactivation. , 2019, Cancer discovery.

[17]  Nicola J. Rinaldi,et al.  An AR-ERG transcriptional signature defined by long-range chromatin interactomes in prostate cancer cells , 2019, Genome research.

[18]  Prashant S. Emani,et al.  Comprehensive functional genomic resource and integrative model for the human brain , 2018, Science.

[19]  Kendall R. Sanson,et al.  Optimized libraries for CRISPR-Cas9 genetic screens with multiple modalities , 2018, Nature Communications.

[20]  J. Licht,et al.  Polycomb- and Methylation-Independent Roles of EZH2 as a Transcription Activator , 2018, Cell reports.

[21]  L. Wessels,et al.  Integrative epigenetic taxonomy of primary prostate cancer , 2018, Nature Communications.

[22]  Joshua M. Stuart,et al.  Genomic Hallmarks and Structural Variation in Metastatic Prostate Cancer , 2018, Cell.

[23]  Christopher W. Whelan,et al.  Structural Alterations Driving Castration-Resistant Prostate Cancer Revealed by Linked-Read Genome Sequencing , 2018, Cell.

[24]  Henry W. Long,et al.  A Somatically Acquired Enhancer of the Androgen Receptor Is a Noncoding Driver in Advanced Prostate Cancer , 2018, Cell.

[25]  J. Weinstein,et al.  A Pan-Cancer Analysis of Enhancer Expression in Nearly 9000 Patient Samples. , 2018, Cell.

[26]  Yi Mi Wu,et al.  The long tail of oncogenic drivers in prostate cancer , 2018, Nature Genetics.

[27]  J. Lis,et al.  Enhancer transcription: what, where, when, and why? , 2018, Genes & development.

[28]  M. Rubin,et al.  Differential impact of RB status on E2F1 reprogramming in human cancer , 2017, The Journal of clinical investigation.

[29]  Brent S. Pedersen,et al.  GIGGLE: a search engine for large-scale integrated genome analysis , 2017, Nature Methods.

[30]  Christopher A. Lavender,et al.  Widespread transcriptional pausing and elongation control at enhancers , 2018, Genes & development.

[31]  M. Rubin,et al.  Aberrant Activation of a Gastrointestinal Transcriptional Circuit in Prostate Cancer Mediates Castration Resistance. , 2017, Cancer cell.

[32]  A. Gursoy,et al.  Androgen receptor binding sites are highly mutated in prostate cancer , 2017, bioRxiv.

[33]  K. White,et al.  Functional assessment of human enhancer activities using whole-genome STARR-sequencing , 2017, Genome Biology.

[34]  Takafumi N. Yamaguchi,et al.  TMPRSS2–ERG fusion co-opts master transcription factors and activates NOTCH signaling in primary prostate cancer , 2017, Nature Genetics.

[35]  J. Carroll,et al.  Novel Androgen Receptor Coregulator GRHL2 Exerts Both Oncogenic and Antimetastatic Functions in Prostate Cancer. , 2017, Cancer research.

[36]  Levi Garraway,et al.  Analysis of 100,000 human cancer genomes reveals the landscape of tumor mutational burden , 2017, Genome Medicine.

[37]  Alain Bergeron,et al.  Genomic hallmarks of localized, non-indolent prostate cancer , 2017, Nature.

[38]  Tao Liu,et al.  Cistrome Data Browser: a data portal for ChIP-Seq and chromatin accessibility data in human and mouse , 2016, Nucleic Acids Res..

[39]  Leighton J. Core,et al.  Base-pair-resolution genome-wide mapping of active RNA polymerases using precision nuclear run-on (PRO-seq) , 2016, Nature Protocols.

[40]  S. Fox,et al.  Role of the novel generation of androgen receptor pathway targeted agents in the management of castration-resistant prostate cancer: A literature based meta-analysis of randomized trials. , 2016, European journal of cancer.

[41]  Fidel Ramírez,et al.  deepTools2: a next generation web server for deep-sequencing data analysis , 2016, Nucleic Acids Res..

[42]  Aaron T. L. Lun,et al.  Three-dimensional disorganization of the cancer genome occurs coincident with long-range genetic and epigenetic alterations , 2016, Genome research.

[43]  Meagan E. Sullender,et al.  Optimized sgRNA design to maximize activity and minimize off-target effects of CRISPR-Cas9 , 2015, Nature Biotechnology.

[44]  Henry W. Long,et al.  The androgen receptor cistrome is extensively reprogrammed in human prostate tumorigenesis , 2015, Nature Genetics.

[45]  Lan T M Dao,et al.  High-throughput and quantitative assessment of enhancer activity in mammals by CapStarr-seq , 2015, Nature Communications.

[46]  S. Dhanasekaran,et al.  Targeting the MLL complex in castration resistant prostate cancer , 2015, Nature Medicine.

[47]  André L. Martins,et al.  Analysis of nascent RNA identifies a unified architecture of initiation regions at mammalian promoters and enhancers , 2014, Nature Genetics.

[48]  Kevin Y. Yip,et al.  FunSeq2: a framework for prioritizing noncoding regulatory variants in cancer , 2014, Genome Biology.

[49]  B. Cohen,et al.  High-throughput functional testing of ENCODE segmentation predictions , 2014, Genome research.

[50]  T. Mikkelsen,et al.  Systematic dissection of regulatory motifs in 2000 predicted human enhancers using a massively parallel reporter assay. , 2013, Genome research.

[51]  A. Sivachenko,et al.  Punctuated Evolution of Prostate Cancer Genomes , 2013, Cell.

[52]  Łukasz M. Boryń,et al.  Genome-Wide Quantitative Enhancer Activity Maps Identified by STARR-seq , 2013, Science.

[53]  I. Mills,et al.  The androgen receptor induces a distinct transcriptional program in castration-resistant prostate cancer in man. , 2013, Cancer cell.

[54]  A. Sivachenko,et al.  Exome sequencing identifies recurrent SPOP, FOXA1 and MED12 mutations in prostate cancer , 2012, Nature Genetics.

[55]  Wing-Kin Sung,et al.  Integration of Regulatory Networks by NKX3-1 Promotes Androgen-Dependent Prostate Cancer Survival , 2012, Molecular and Cellular Biology.

[56]  Eric S. Lander,et al.  The genomic complexity of primary human prostate cancer , 2010, Nature.

[57]  G. Kreiman,et al.  Widespread transcription at neuronal activity-regulated enhancers , 2010, Nature.

[58]  Mark D. Robinson,et al.  edgeR: a Bioconductor package for differential expression analysis of digital gene expression data , 2009, Bioinform..

[59]  Arul M Chinnaiyan,et al.  Prevalence of TMPRSS2-ERG Fusion Prostate Cancer among Men Undergoing Prostate Biopsy in the United States , 2009, Clinical Cancer Research.

[60]  J. Tchinda,et al.  Recurrent fusion of TMPRSS2 and ETS transcription factor genes in prostate cancer. , 2006, Science.

[61]  H. Scher,et al.  Biology of progressive, castration-resistant prostate cancer: directed therapies targeting the androgen-receptor signaling axis. , 2005, Journal of clinical oncology : official journal of the American Society of Clinical Oncology.

[62]  J. Trapman,et al.  The androgen receptor in prostate cancer. , 1996, Pathology, research and practice.

[63]  G. Bubley,et al.  Mutation of the androgen-receptor gene in metastatic androgen-independent prostate cancer. , 1995, The New England journal of medicine.

[64]  Jorma Isola,et al.  In vivo amplification of the androgen receptor gene and progression of human prostate cancer , 1995, Nature Genetics.

[65]  G. Murphy,et al.  LNCaP model of human prostatic carcinoma. , 1983, Cancer research.