Summary

Enforced Activation of Enhancer RNAs In Situ through the dCas9 Synergistic Activation Mediator System

Published: June 05, 2020
doi:

Summary

Enhancer RNAs (eRNAs) are non-coding RNAs produced from active enhancers. An optimal approach to study eRNA functions is to manipulate their levels in the native chromatin regions. Here we introduce a robust system for eRNA studies by using CRISPR-dCas9-fused transcriptional activators to induce the expression of eRNAs of interest.

Abstract

Enhancers are pivotal genomic elements scattered through the mammalian genome and dictate tissue-specific gene expression programs. Increasing evidence has shown that enhancers not only provide DNA binding motifs for transcription factors (TFs) but also generate non-coding RNAs that are referred to as eRNAs. Studies have demonstrated that eRNA transcripts can play significant roles in gene regulation in both physiology and disease. Commonly used methods to investigate the function of eRNAs are constrained to “loss-of-function” approaches by knockdown of eRNAs, or by chemical inhibition of the enhancer transcription. There has not been a robust method to conduct “gain-of-function” studies of eRNAs to mimic specific disease conditions such as human cancer, where eRNAs are often overexpressed. Here, we introduce a method for precisely and robustly activating eRNAs for functional interrogation of their roles by applying the dCas9 mediated Synergistic Activation Mediators (SAM) system. We present the entire workflow of eRNA activation, from the selection of eRNAs, the design of gRNAs to the validation of eRNA activation by RT-qPCR. This method represents a unique approach to study the roles of a particular eRNA in gene regulation and disease development. In addition, this system can be employed for unbiased CRISPR screening to identify phenotype-driving eRNA targets in the context of a specific disease.

Introduction

The human genome contains a constellation of regulatory elements1,2,3. Among these, enhancers emerge to be one of the most critical categories4,5,6. Enhancers play essential roles in regulating development, and are responsible for generating spatial-temporal gene expression programs to determine cell identity5,6,7. Conventionally, enhancers are only considered to be DNA elements that provide binding motifs for transcription factors (TFs), which then control target gene expression6,8. However, a series of studies found that many active enhancers also transcribe non-coding enhancer RNAs (i.e., eRNAs)4,9,10.

The level of eRNA transcription was found to correlate with the activity of an enhancer4,10. Active enhancers produce more eRNA transcripts and show higher levels of epigenome markers associated with active transcription, such as H3K27ac and H3K4me19,11,12. Some studies have demonstrated that eRNA transcripts can play important roles in transcriptional activation of target genes10,12. A large number of eRNAs were identified to be deregulated in human cancers13,14,15,16, many of which exhibited high cancer type specificity and clinical relevance. These findings bring opportunities that the elucidation of eRNAs that can drive/promote tumorigenesis may offer novel targets for therapeutic intervention13,15

Current methods to study eRNA functions are almost exclusively based on knockdown strategies that used small interference RNAs (siRNA), short hairpin RNAs (shRNAs), or antisense oligonucleotides (ASOs, of which locked nucleic acids (LNAs) are the commonly used type in research)10,12,17. However, human diseases such as cancer predominantly show overexpression of eRNAs as compared to their adjacent normal tissue15, demanding tools to “overexpress” eRNAs to mimic their disease-relevant expression patterns for functional studies. To achieve this, a plasmid-based ectopic overexpression system is not optimal because the exact transcription start and termination sites of eRNAs remain largely unclear. In addition, a plasmid expression system may alter the location of eRNAs, causing potential artifacts of their functions18. Here we provide a detailed protocol to facilitate the functional characterization of eRNAs by enforcing their “overexpression” in the native genomic locus of their production (i.e., in situ), which is based on the CRISPR/dCas9-Synergistic Activation Mediators System (SAM).

The SAM system was initially developed for activating coding genes and long intergenic non-coding RNAs (lincRNAs) associated with BRAF inhibitor resistance in melanoma cells19. Unlike other CRISPR activation (CRISPRa) technologies, the SAM system consists of a combination of transcription activators to confer robust transcriptional activation of target regions. These activators include: an enzymatically dead Cas9 (dCas9) fused with VP64 (i.e., dCas9-VP64); a guide RNA containing two MS2 RNA aptamers, and an MS2-p65-HSF1 fusion activator protein. The presence of the MS2 aptamers in the gRNA can recruit the MS2-p65-HSF1 fusion protein to the vicinity of dCas9/gRNA binding sites. Among these, VP64 is an engineered tetramer of the herpes simplex VP16 transcriptional activator domain, which has been shown to strongly activate gene transcription by recruiting general transcription factors20,21,22. The MS2-p65-HSF1 fusion protein consists of three parts. The first part, the MS2-N55K, is a mutant form of MS2 binding protein that has a stronger affinity23; the other two parts of this fusion protein are the transactivation domain of p65 and heat shock factor 1 (HSF1), both of which are transcription factors that possess strong transactivation domains and can induce robust transcription programs24,25. Therefore, the SAM system essentially created a highly potent activator complex to activate transcription of designated coding genes and lincRNAs19.

Protocol

The entire workflow of this protocol is shown in Figure 1. 1. Enhancer RNA (eRNA) selection Identify a putative enhancer region of interests by using binding peaks of chromatin immunoprecipitation sequencing (ChIP-Seq) data, i.e., of histone modifications (e.g., H3K4me1 and H3K27ac), or of transcription coactivators (e.g., p300). Identify the eRNA of interest by intersecting the ChIP-Seq peak with RNA-seq signals (e.g., from total RNA-seq or …

Representative Results

Figure 1 illustrates the overall workflow of this protocol. Our focus was on a representative eRNA, NET1e15, which is overexpressed in breast cancer, for which SAM system was used to activate and study it’s biological role in regulating gene expression, cell proliferation and cancer drug response. For this NET1 enhancer, several p300 ChIP-Seq peaks, flanked by transcribed eRNA transcripts (Figure 2A,B</stro…

Discussion

Based on our data, we conclude that the SAM system is suitable for studying the role of eRNAs in regulating cellular phenotypes, e.g., cell growth or drug resistance. However, careful gRNA designing is required for robust eRNA activation, due to the following reasons. First of all, the transcription start site (TSS) of eRNA in each specific cell lines/types remains less clearly annotated. Due to this, epigenomic information (e.g., ChIP-Seq of H3K27ac, of transcription factors, or of p300), transcriptional activity depict…

Divulgazioni

The authors have nothing to disclose.

Acknowledgements

This work is supported by grants to W.L (Cancer Prevention and Research Institute of Texas, CPRIT RR160083 and RP180734; NCI K22CA204468; NIGMS R21GM132778; The University of Texas UT Stars Award; and the Welch foundation AU-2000-20190330) and a post-doctoral fellowship to J.L (UTHealth Innovation for Cancer Prevention Research Training Program Post-doctoral Fellowship, CPRIT RP160015). We acknowledge the original publicataion15 where some of our current figures were adopted from (with modifications), which follows the Creative Commons license (https://creativecommons.org/licenses/by/4.0/).

Materials

Blasticidin Goldbio B-800-100
BsmBI restriction enzyme New England BioLabs Inc. R0580S
Cas9 mAb Active Motif 61757 Lot: 10216001
Deoxynucleotide (dNTP) Solution Mix New England BioLabs Inc. N0447S
Dulbecco’s Modified Eagle Medium Corning 10-013-CM
Dynabeads Protein G Thermo Fisher Scientific 65002
EDTA Thermo Fisher Scientific BP118-500
EGTA Sigma E3889
Fetal Bovine Serum GenDEPOT F0900-050
Glycogen Thermo Fisher Scientific 10814010
Hepes-KOH Thermo Fisher Scientific BP310-100
Hexadimethrine Bromide Sigma H9268
Hygromycin B Goldbio H-270-25
IGEPAL CA630 Sigma D6750
IncuCyte live-cell imager Essen BioScience IncuCyte S3 Live-Cell Analysis System
lenti_dCAS-VP64_Blast Addgene 61425
lenti_gRNA(MS2)_zeo backbone Addgene 61427
lenti_MS2-p65-HSF1_Hygro Addgene 61426
LiCL Sigma L9650
Lipofectamine 2000 Thermo Fisher Scientific 11668-500
NaCl Sigma S3014
Na-Deoxycholate Sigma D6750
NaHCO3 Thermo Fisher Scientific BP328-500
N-lauroylsarcosine Sigma 97-78-9
Opti-MEM Reduced Serum Medium Thermo Fisher Scientific 31985070
PES syringe filter BASIX 13-1001-07
Protease Inhibitor Cocktail Tablet Roche Diagnostic 11836145001
pSpCas9(BB)-2A-Puro Addgene 62988
Q5 High-Fidelity DNA Polymerase New England BioLabs Inc. M0491S
Q5 Reaction Buffer New England BioLabs Inc. B9027S
Quick-DNA Miniprep ZYMO Research D3025
Quick-RNA Miniprep ZYMO Research R1054
Restriction enzyme buffer New England BioLabs Inc. B7203S
RT-qPCR Detection System Thermo Fisher Scientific Quant Studio3
SDS Thermo Fisher Scientific BP359-500
Sonicator Qsonica Q800R2
Sso Advanced Universal SYBR Green Supermix Bio-Rad Laboratories 1725274
Stbl3 competent cell Thermo Fisher Scientific C7373-03
Superscript IV reverse transcript Thermo Fisher Scientific 719000
Surveyor Mutation Detection Kits Integrated DNA Technologies 706020
T4 DNA Ligase New England BioLabs Inc. M0202S
T4 DNA Ligase Reaction Buffer New England BioLabs Inc. B0202S
TE buffer Thermo Fisher Scientific 46009CM
Thermal cycler Bio-Rad Laboratories T100
Thermomixer Sigma 5384000020
Zeocin Thermo Fisher Scientific ant-zn-1p

Riferimenti

  1. ENCODE Project Consortium. An integrated encyclopedia of DNA elements in the human genome. Nature. 489 (7414), 57-74 (2012).
  2. Djebali, S., et al. Landscape of transcription in human cells. Nature. 489 (7414), 101-108 (2012).
  3. Kundaje, A., et al. Integrative analysis of 111 reference human epigenomes. Nature. 518 (7539), 317-330 (2015).
  4. Andersson, R., et al. An atlas of active enhancers across human cell types and tissues. Nature. 507 (7493), 455-461 (2014).
  5. Heinz, S., Romanoski, C. E., Benner, C., Glass, C. K. The selection and function of cell type-specific enhancers. Nature Reviews Molecular Cell Biology. 16 (3), 144-154 (2015).
  6. Ong, C. T., Corces, V. G. Enhancer function: new insights into the regulation of tissue-specific gene expression. Nature Reviews Genetics. 12 (4), 283-293 (2011).
  7. Hnisz, D., et al. Super-enhancers in the control of cell identity and disease. Cell. 155 (4), 934-947 (2013).
  8. Grossman, S. R., et al. Systematic dissection of genomic features determining transcription factor binding and enhancer function. Proceedings of the National Academy of Sciences of the United States of America. 114 (7), 1291-1300 (2017).
  9. Kim, T. K., et al. Widespread transcription at neuronal activity-regulated enhancers. Nature. 465 (7295), 182-187 (2010).
  10. Li, W., Notani, D., Rosenfeld, M. G. Enhancers as non-coding RNA transcription units: recent insights and future perspectives. Nature Reviews Genetics. 17 (4), 207-223 (2016).
  11. Creyghton, M. P., et al. Histone H3K27ac separates active from poised enhancers and predicts developmental state. Proceedings of the National Academy of Sciences of the United States of America. 107 (50), 21931-21936 (2010).
  12. Li, W., et al. Functional roles of enhancer RNAs for oestrogen-dependent transcriptional activation. Nature. 498 (7455), 516-520 (2013).
  13. Lee, J. H., Xiong, F., Li, W. Enhancer RNAs in cancer: regulation, mechanisms and therapeutic potential. RNA Biology. , 1-10 (2020).
  14. Sur, I., Taipale, J. The role of enhancers in cancer. Nature Reviews Cancer. 16 (8), 483-493 (2016).
  15. Zhang, Z., et al. Transcriptional landscape and clinical utility of enhancer RNAs for eRNA-targeted therapy in cancer. Nature Communications. 10 (1), 4562 (2019).
  16. Chen, H., et al. A Pan-Cancer Analysis of Enhancer Expression in Nearly 9000 Patient Samples. Cell. 173 (2), 386-399 (2018).
  17. Kopp, F., Mendell, J. T. Functional Classification and Experimental Dissection of Long Noncoding RNAs. Cell. 172 (3), 393-407 (2018).
  18. Lubelsky, Y., Ulitsky, I. Sequences enriched in Alu repeats drive nuclear localization of long RNAs in human cells. Nature. 555 (7694), 107-111 (2018).
  19. Konermann, S., et al. Genome-scale transcriptional activation by an engineered CRISPR-Cas9 complex. Nature. 517 (7536), 583-588 (2015).
  20. Beerli, R. R., Segal, D. J., Dreier, B., Barbas, C. R. Toward controlling gene expression at will: specific regulation of the erbB-2/HER-2 promoter by using polydactyl zinc finger proteins constructed from modular building blocks. Proceedings of The National Academy of Sciences of the United States of America. 95 (25), 14628-14633 (1995).
  21. Gilbert, L. A., et al. CRISPR-mediated modular RNA-guided regulation of transcription in eukaryotes. Cell. 154 (2), 442-451 (2013).
  22. Hirai, H., Tani, T., Kikyo, N. Structure and functions of powerful transactivators: VP16, MyoD and FoxA. International Journal of Developmental Biology. 54 (11-12), 1589-1596 (2010).
  23. Lim, F., Spingola, M., Peabody, D. S. Altering the RNA binding specificity of a translational repressor. Journal of Biological Chemistry. 269 (12), 9006-9010 (1994).
  24. Schmitz, M. L., Baeuerle, P. A. The p65 subunit is responsible for the strong transcription activating potential of NF-kappa B. EMBO Journal. 10 (12), 3805-3817 (1991).
  25. Vihervaara, A., Sistonen, L. HSF1 at a glance. Journal of Cell Science. 127, 261-266 (2014).
  26. Core, L. J., et al. Analysis of nascent RNA identifies a unified architecture of initiation regions at mammalian promoters and enhancers. Nature Genetics. 46 (12), 1311-1320 (2014).
  27. Haeussler, M., et al. Evaluation of off-target and on-target scoring algorithms and integration into the guide RNA selection tool CRISPOR. Genome Biology. 17 (1), 148 (2016).
  28. . Benchling [Biology Software] Available from: https://benchling.com (2020)
  29. Labun, K., et al. CHOPCHOP v3: expanding the CRISPR web toolbox beyond genome editing. Nucleic Acids Research. 47, 171-174 (2019).
  30. Hanna, R. E., Doench, J. G. Design and analysis of CRISPR-Cas experiments. Nature Biotechnology. , (2020).
  31. Nageshwaran, S., et al. CRISPR Guide RNA Cloning for Mammalian Systems. Journal of Visualized Experiments. (140), e57998 (2018).
  32. Al-Allaf, F. A., Tolmachov, O. E., Zambetti, L. P., Tchetchelnitski, V., Mehmet, H. Remarkable stability of an instability-prone lentiviral vector plasmid in Escherichia coli Stbl3. 3 Biotech. 3 (1), 61-70 (2013).
  33. Qiu, P., et al. Mutation detection using Surveyor nuclease. Biotechniques. 36 (4), 702-707 (2004).
  34. Ran, F. A., et al. Genome engineering using the CRISPR-Cas9 system. Nature Protocols. 8 (11), 2281-2308 (2013).
  35. Brinkman, E. K., Chen, T., Amendola, M., van Steensel, B. Easy quantitative assessment of genome editing by sequence trace decomposition. Nucleic Acids Research. 42 (22), 168 (2014).
  36. Quick-DNA Miniprep Kit. ZYMO Available from: https://files.zymoresearch.com/protocols/_d3024_d3025_quick-dna_miniprep_kit.pdf (2020)
  37. SURVEYOR Mutation Detection Kit. IDT Available from: https://sfvideo.blob.core.windows.net/sitefinity/docs/default-source/user-guide-manual/surveyor-kit-for-gel-electrophoresis-user-guide.pdf?sfvrsn=a9123407_6 (2020)
  38. Quick-RNA Miniprep Kit. ZYMO Available from: https://files.zymoresearch.com/protocols/_r1054_r1055_quick_rna_miniprep_kit.pdf (2020)
  39. SuperScript IV Reverse Transcriptase Product Manual. Thermo Fisher Scientific Available from: https://assets.thermofisher.com/TFS-Assets/LSG/manuals/SSIV_Reverse_Transcriptase_UG.pdf (2020)
  40. Ounzain, S., et al. Functional importance of cardiac enhancer associated noncoding RNAs in heart development and disease. Journal of Molecular and Cellular Cardiology. 76, 55-70 (2014).
  41. McCleland, M. L., et al. CCAT1 is an enhancer-templated RNA that predicts BET sensitivity in colorectal cancer. Journal of Clinical Investigation. 126 (2), 639-652 (2016).
  42. Miao, Y., et al. Enhancer-associated long non-coding RNA LEENE regulates endothelial nitric oxide synthase and endothelial function. Nature Communications. 9 (1), 292 (2018).
This article has been published
Video Coming Soon
Keep me updated:

.

Citazione di questo articolo
Liao, Z., Lee, J., Krakowiak, J., Wang, R., Li, W. Enforced Activation of Enhancer RNAs In Situ through the dCas9 Synergistic Activation Mediator System. J. Vis. Exp. (160), e61460, doi:10.3791/61460 (2020).

View Video