Genomic targeting of epigenetic probes using a chemically tailored Cas9 system

Significance Programmable DNA-binding proteins such as nuclease-deficient Cas9 (dCas9) offer a simple system for genomic localization of genetically encodable biomolecules. These tools have enabled characterization of numerous different chromatin effectors at specific genetic elements. The delivery of fully synthetic cargos to specific loci is currently limited by a lack of adequate technologies. Here we used protein trans-splicing to ligate chemical moieties to dCas9 in vitro and subsequently deliver these molecules to live cells. We demonstrate the effectiveness and versatility of this method by delivering both small-molecule and proteinaceous epigenetic probes to endogenous genomic sites where they interact with their target proteins. This facile and modular strategy will find broad use in locus-specific targeting of chemical cargos to study nuclear processes. Recent advances in the field of programmable DNA-binding proteins have led to the development of facile methods for genomic localization of genetically encodable entities. Despite the extensive utility of these tools, locus-specific delivery of synthetic molecules remains limited by a lack of adequate technologies. Here we combine the flexibility of chemical synthesis with the specificity of a programmable DNA-binding protein by using protein trans-splicing to ligate synthetic elements to a nuclease-deficient Cas9 (dCas9) in vitro and subsequently deliver the dCas9 cargo to live cells. The versatility of this technology is demonstrated by delivering dCas9 fusions that include either the small-molecule bromodomain and extra-terminal family bromodomain inhibitor JQ1 or a peptide-based PRC1 chromodomain ligand, which are capable of recruiting endogenous copies of their cognate binding partners to targeted genomic binding sites. We expect that this technology will allow for the genomic localization of a wide array of small molecules and modified proteinaceous materials.

[1]  Randall J. Platt,et al.  Optical Control of Mammalian Endogenous Transcription and Epigenetic States , 2013, Nature.

[2]  P. Cole,et al.  Chemical probes for histone-modifying enzymes. , 2008, Nature chemical biology.

[3]  Yuval Kluger,et al.  PCGF homologs, CBX proteins, and RYBP define functionally distinct PRC1 family complexes. , 2012, Molecular cell.

[4]  C. Allis,et al.  Epigenetics: A Landscape Takes Shape , 2007, Cell.

[5]  C. Bountra,et al.  Epigenetic protein families: a new frontier for drug discovery , 2012, Nature Reviews Drug Discovery.

[6]  James J. Collins,et al.  Using Targeted Chromatin Regulators to Engineer Combinatorial and Spatial Transcriptional Regulation , 2014, Cell.

[7]  Daesik Kim,et al.  Highly efficient RNA-guided genome editing in human cells via delivery of purified Cas9 ribonucleoproteins , 2014, Genome research.

[8]  A. Mapp,et al.  Transforming ligands into transcriptional regulators: building blocks for bifunctional molecules. , 2011, Chemical Society reviews.

[9]  Le Cong,et al.  Multiplex Genome Engineering Using CRISPR/Cas Systems , 2013, Science.

[10]  J. Joung,et al.  Locus-specific editing of histone modifications at endogenous enhancers using programmable TALE-LSD1 fusions , 2013, Nature Biotechnology.

[11]  William B. Smith,et al.  Selective inhibition of BET bromodomains , 2010, Nature.

[12]  P. Dervan,et al.  Recognition of the DNA minor groove by pyrrole-imidazole polyamides. , 2003, Current opinion in structural biology.

[13]  S. J. Flint,et al.  The double bromodomain proteins Brd2 and Brd3 couple histone acetylation to transcription. , 2008, Molecular cell.

[14]  William B. Smith,et al.  Genome-wide localization of small molecules , 2013, Nature Biotechnology.

[15]  Jennifer A. Smith,et al.  The Brd4 Extraterminal Domain Confers Transcription Activation Independent of pTEFb by Recruiting Multiple Proteins, Including NSD3 , 2011, Molecular and Cellular Biology.

[16]  J. Keith Joung,et al.  Efficient Delivery of Genome-Editing Proteins In Vitro and In Vivo , 2014, Nature Biotechnology.

[17]  Anne E Carpenter,et al.  The Bromodomain Protein Brd4 Insulates Chromatin from DNA Damage Signaling , 2013, Nature.

[18]  David R Corey,et al.  Recognition of chromosomal DNA by PNAs. , 2004, Chemistry & biology.

[19]  Neville E. Sanjana,et al.  High-throughput functional genomics using CRISPR–Cas9 , 2015, Nature Reviews Genetics.

[20]  C. Barbas,et al.  Synthetic Zinc Finger Proteins: The Advent of Targeted Gene Regulation and Genome Modification Technologies , 2014, Accounts of chemical research.

[21]  Data production leads,et al.  An integrated encyclopedia of DNA elements in the human genome , 2012 .

[22]  Benjamin L. Oakes,et al.  Programmable RNA recognition and cleavage by CRISPR/Cas9 , 2014, Nature.

[23]  M. Dawson,et al.  Cancer Epigenetics: From Mechanism to Therapy , 2012, Cell.

[24]  P. Dervan,et al.  Design of Sequence-Specific DNA Binding Molecules for DNA Methyltransferase Inhibition , 2014, Journal of the American Chemical Society.

[25]  K. Helin,et al.  Transcriptional regulation by Polycomb group proteins , 2013, Nature Structural &Molecular Biology.

[26]  James E. DiCarlo,et al.  RNA-Guided Human Genome Engineering via Cas9 , 2013, Science.

[27]  N. Stephanopoulos,et al.  Choosing an effective protein bioconjugation strategy. , 2011, Nature chemical biology.

[28]  ENCODEConsortium,et al.  An Integrated Encyclopedia of DNA Elements in the Human Genome , 2012, Nature.

[29]  J. Workman,et al.  Signals and combinatorial functions of histone modifications. , 2011, Annual review of biochemistry.

[30]  L. Rajendran,et al.  Subcellular targeting strategies for drug design and delivery , 2010, Nature Reviews Drug Discovery.

[31]  Yang Shi,et al.  Emerging roles for chromatin as a signal integration and storage platform , 2013, Nature Reviews Molecular Cell Biology.

[32]  Christopher M. Vockley,et al.  Epigenome editing by a CRISPR/Cas9-based acetyltransferase activates genes from promoters and enhancers , 2015, Nature Biotechnology.

[33]  T. Muir,et al.  Ultrafast protein splicing is common among cyanobacterial split inteins: implications for protein engineering. , 2012, Journal of the American Chemical Society.

[34]  Pilar Blancafort,et al.  Epigenetic reprogramming of cancer cells via targeted DNA methylation , 2012, Epigenetics.

[35]  Namritha Ravinder,et al.  Rapid and highly efficient mammalian cell engineering via Cas9 protein transfection. , 2015, Journal of biotechnology.

[36]  B. Fierz,et al.  Chromatin as an expansive canvas for chemical biology. , 2012, Nature chemical biology.

[37]  Christopher M. Vockley,et al.  RNA-guided gene activation by CRISPR-Cas9-based transcription factors , 2013, Nature Methods.

[38]  Robert Tjian,et al.  CASFISH: CRISPR/Cas9-mediated in situ labeling of genomic loci in fixed cells , 2015, Proceedings of the National Academy of Sciences.

[39]  Peter E. Nielsen Peptide Nucleic Acids (PNA) in Chemical Biology and Drug Discovery , 2010, Chemistry & biodiversity.

[40]  Robert E. Kingston,et al.  Mechanisms of Polycomb gene silencing: knowns and unknowns , 2009, Nature Reviews Molecular Cell Biology.

[41]  Nancy Cheng,et al.  A cellular chemical probe targeting the chromodomains of Polycomb Repressive Complex 1 , 2015, Nature chemical biology.