Proximity Induced Splicing Utilizing Caged Split Inteins | Journal of the

6 days ago - The Supporting Information is available free of charge on the ACS Publications ... Full methods and experimental data (PDF) ... Download...
0 downloads 0 Views 1MB Size
Subscriber access provided by Nottingham Trent University

Communication

Proximity Induced Splicing Utilizing Caged Split Inteins Josef A. Gramespacher, Antony J. Burton, Luis F. Guerra, and Tom W. Muir J. Am. Chem. Soc., Just Accepted Manuscript • DOI: 10.1021/jacs.9b05721 • Publication Date (Web): 16 Aug 2019 Downloaded from pubs.acs.org on August 16, 2019

Just Accepted “Just Accepted” manuscripts have been peer-reviewed and accepted for publication. They are posted online prior to technical editing, formatting for publication and author proofing. The American Chemical Society provides “Just Accepted” as a service to the research community to expedite the dissemination of scientific material as soon as possible after acceptance. “Just Accepted” manuscripts appear in full in PDF format accompanied by an HTML abstract. “Just Accepted” manuscripts have been fully peer reviewed, but should not be considered the official version of record. They are citable by the Digital Object Identifier (DOI®). “Just Accepted” is an optional service offered to authors. Therefore, the “Just Accepted” Web site may not include all articles that will be published in the journal. After a manuscript is technically edited and formatted, it will be removed from the “Just Accepted” Web site and published as an ASAP article. Note that technical editing may introduce minor changes to the manuscript text and/or graphics which could affect content, and all legal disclaimers and ethical guidelines that apply to the journal pertain. ACS cannot be held responsible for errors or consequences arising from the use of information contained in these “Just Accepted” manuscripts.

is published by the American Chemical Society. 1155 Sixteenth Street N.W., Washington, DC 20036 Published by American Chemical Society. Copyright © American Chemical Society. However, no copyright claim is made to original U.S. Government works, or works produced by employees of any Commonwealth realm Crown government in the course of their duties.

Page 1 of 9 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Journal of the American Chemical Society

Proximity Induced Splicing Utilizing Caged Split Inteins Josef A. Gramespacher, Antony J. Burton, Luis F. Guerra and Tom W. Muir* Department of Chemistry, Princeton University, Frick Laboratory, Princeton, New Jersey 08544, United States

Supporting Information Placeholder

ABSTRACT: Naturally split inteins drive the ligation of separately expressed polypeptides through a process called protein trans splicing (PTS). The ability to control PTS – so-called conditional protein splicing (CPS) - has led to the development of tools to modulate protein structure and function at the post-translational level. CPS applications that utilize proximity as a trigger are especially intriguing as they afford the possibility to activate proteins in both a temporal and spatially targeted manner. In this study, we present the first proximity triggered CPS method that utilizes a naturally split fast splicing intein, Npu. We show that this method is amenable to diverse proximity triggers and capable of reconstituting and locally activating the acetyltransferase p300 in mammalian cells. This technology opens up a range of possibilities for the use of proximity triggered CPS.

Inteins, or intervening proteins, spontaneously remove themselves from the host protein in which they are embedded.1 While this protein splicing reaction is performed in cis by the more common contiguous inteins, a subset of these autocatalytic proteins are expressed as two separate polypeptides and undergo protein trans splicing (PTS) upon association.2 Because protein splicing results in a significant change in protein primary structure - the intein is removed and the flanking protein sequences (exteins) are joined through a native peptide bond – there has long been interest in harnessing the process to modulate protein function at the posttranslational level.3 A key challenge in this area relates to the autocatalytic nature of the splicing reaction, which makes temporal control of activity difficult. To address this issue, a variety of conditional protein splicing (CPS) methods have been developed in which intein activity is activated only upon addition of a specified trigger. Generally, these triggers induce splicing by introducing either a splicing favorable conformational change in an otherwise structurally locked intein,4-10 deprotecting chemically caged residues that inhibit catalytic activity,11-

17

or bringing intein fragments with low inherent affinity into close proximity.3, 18-24 Proximity triggered splicing methods are an especially attractive variation of CPS because they afford the possibility to sense and respond to myriad cellular stimuli; in principle, any process that results in the intein fragments being brought together should be capable of triggering splicing. Despite this potential, such CPS approaches have been limited by their reliance on the artificially split VMA intein3, which suffers from poor stability25 and stringent extein dependence.26-28 In contrast, many naturally split inteins are stable over a wide range of conditions and have greater flexibility with regards to extein context.29-32 For example, the fast splicing split DnaE intein from the cyanobacterium Nostoc Punctiforme (Npu) maintains activity up to 65°C and functions in 2M Guanidine or 6M Urea31. Furthermore, a rationally engineered promiscuous version of Npu has recently been reported that requires only the native +1 cysteine to effectively splice.30 However, fast splicing inteins such as Npu have yet to be developed into a proximity triggered CPS method, as this would require overcoming the remarkably rapid and tight association of their intein fragments (Npu, Kd = 1.2 nM33). We recently reported the development of a CPS system based on inhibiting association of a variety of naturally split inteins, including Npu.9 By extending a split intein fragment with the appropriate binding region from its complementary partner, it becomes locked in an intermediate folded structure (Figure S1). As a consequence, the caged intein fragments (NpuNcage and NpuCcage) cannot associate until the caging sequences are cleaved off by a specified protease. We wondered whether this split intein zymogen system could serve as a starting point for a proximity triggered CPS system. Specifically, we imagined that the caging sequences appended to each intein fragment likely dissociate and reassociate at a given rate. Thus, forcing the caged inteins together should increase the probability of a splicing productive binding event (involving a reorganization of

ACS Paragon Plus Environment

Journal of the American Chemical Society 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

the initial complex) between the full-length intein pairs (Figure 1a, S2). To test this idea, we fused the heterodimerization domains FKBP and FRB to the C- and N-terminus of NpuNcage and NpuCcage, respectively, allowing rapamycin to be used to force the caged inteins into close proximity (Figure 1a, S3). Initial results were encouraging in that rapamycin-dependent splicing was observed at 37°C, albeit at low efficiency (Figure 1b). We reasoned that the rate-limiting step in the overall process was likely isomerization of the initial rapamycin-induced heterodimer to afford an active intein complex. With this in mind, the system was redesigned to favor this rearrangement. Informed by our previous work in which the split intein caging interface was optimized through an iterative design process9, we effectively retro-engineered the system to weaken slightly the interaction between NpuN and its caging sequence (Figure S4, S3). Gratifyingly, this exercise afforded an enhanced NpuCage system (e-NpuCage) in which efficient rapamycindependent splicing was observed at 37°C over the course of a few hours, albeit slower than either protease mediated splicing (Figure 1c) or splicing of NpuWT fragments. This represents a dramatic improvement over our previous VMA intein based system3, which while functional at lower temperatures, fails to undergo efficient CPS at the physiological relevant 37°C and, indeed, slowly precipitates out of solution (Figure 1d, Figure S5, S3).

Figure 1. Proximity triggered CPS using caged split inteins. (a) Schematic depicting CPS of NpuNCage and NpuCCage through rapamycin induced dimerization of FRB and FKBP. (b-d) Coomassie stained SDS-PAGE gels of splicing reactions (37°, 1μM each protein construct) monitored over time with or without 10μM rapamycin. SP = splice product (MBP-eGFP, MW~ 69kDa). (b-c) CC = FRB-NpuCCage-

eGFP. (b) NC = MBP-NpuNCage-FKBP. (c) NC = MBP-eNpuNCage-FKBP, Cas3 = caspase3 (cleaves off cages). (d) N = MBP-VMA-N-FKBP, C = FRB-VMA-C-eGFP. (e) Schematic depicting proximity-triggered FRET changes between Cy3 and Cy5 dyes. (f) Graph depicting observed FRET efficiencies in the presence or absence of rapamycin between FRB-NpuCCage-Cy5 and either Cy3-(C1A)NpuNCage-FKBP or Cy3-e-(C1A)-NpuNCage-FKBP. Errors = s.e.m. (n=3)

Additional studies were conducted to better understand the relationship between splicing efficiency and the presumed structural reorganization within the rapamycininduced complex. Specifically, PTS-based bioconjugation techniques were used to site-specifically install Cy3 or Cy5 dyes into the two components of the CPS system (Figure S3, S6). Importantly, a mutant version of NpuN was employed (NpuN-C1A) that cannot support PTS and that therefore afforded us the opportunity to follow the dimerization and subsequent isomerization process using FRET (Figure 1e). This biophysical experiment was conducted for both the original inefficient NpuCage system and the improved version thereof, e-NpuCage. The complementary dyelabeled constructs were mixed and FRET efficiency monitored over time as a function of added rapamycin (Figure 1f). In both cases, we observed a rapid (i.e. within 1 minute, the earliest time-point taken) rapamycindependent increase in FRET efficiency, likely due to dimerization of FRB and FKBP. Thereafter, the behavior of the two systems diverged markedly; whereas FRET efficiency increased very slightly over the course of several hours for the original NpuCage system, the more active e-NpuCage version exhibited a large increase in FRET over the course of the experiment. We interpret this second phase as reflecting the structural re-organization within the complex to give the active intein, which we note should bring the FRET pair into closer proximity based on their location. Consequently, the fluorescence data correlate well with the differing splicing activity of NpuCage and e-NpuCage and support our contention that the overall kinetics are dictated by the efficiency of the internal isomerization within the complex. An appealing feature of this CPS strategy is the potential to exchange the triggering mechanism3, 18-20. As such, we were especially interested in the idea of using DNA as a substrate20, given the availability of programmable DNA binding proteins such as the nuclease-dead version of CRISPR Cas9 (dCas9)34. A model system was designed to explore this idea in which dCas9 from Streptococcus pyogenes was fused to the Cterminus of e-NpuNcage while the NpuCcage construct was fused to the N-terminus of histone H3 within a nucleosome (Figure 2a, S3). In this way, a single stranded guide RNA (sgRNA) directed against a DNA sequence immediately upstream of the assembled nucleosome, should result in proximity triggered splicing of an extein sequence (a myc epitope) to the histone (Figure 2a).

ACS Paragon Plus Environment

Page 2 of 9

Page 3 of 9 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Journal of the American Chemical Society

Consistent with this design, we observed dose-dependent binding of the dCas9 fusion to the nucleosome in the presence of the on-target sgRNA, but not with an offtarget sgRNA (Figure 2b). Importantly, sgRNAdependent binding was accompanied by splicing to give the expected myc-tagged histone product (Figure 2c). The success of this proof-of-principle experiment illustrates the potential of using genomic targeting to mediate CPS and, more specifically, the possibility of carrying out localized protein tagging in a chromatin setting.

Figure 2. Proximity triggered CPS on a chromatin substrate. (a) Schematic depicting the dCas9-mediated CPS of an extein sequence (myc) onto the N-terminus of histone H3 within a nucleosome. sgRNA = single stranded guide RNA. (b) Gel shift assay in which increasing concentrations of myc-e-NpuNcage-dCas9, 0.1μM NpuCCage-H3 nucleosomes (containing 5’- fluorescein tagged DNA) and on-target or off-target sgRNA were incubated for 16hrs. (c) Western blot against indicated epitope tags of reactions in which 0.5μM of myc-e-NpuNcage-dCas9 and 0.1μM of NpuCCage-H3 nucleosomes were incubated with on-target or off-target sgRNA. NC = myc-e-NpuNcage-dCas9, SP = splice product (myc-H3, MW~17kDa), CC-H3 = NpuCCage-H3.

Encouraged by the in-vitro experiments, we next tested the functionality of our system in mammalian cells. Additionally, we were keen to take advantage of the minimal extein dependence of NpuC by reconstituting an active enzyme in a splicing-dependent manner. Previous research has shown that non-functional split fragments of the acetyl-transferase p300 can be re-ligated to restore activity using native chemical ligation (NCL)35 and that a catalytically active p300 can be targeted to promoters via dCas9 to induce gene expression36. Therefore, we wondered whether split p300 could be targeted via dCas9 to a user defined promoter and then reconstituted and activated utilizing Npucage in a proximity triggered CPS dependent manner to induce transcriptional activation. Using the p300 core domain, which has previously been shown to be sufficient for catalytic activity36, as well as the previously described split site35 (Figure S7), we fused the C-terminal fragment of p300 (p300C) to FRBNpuCcage and the N-terminal fragment of the enzyme (p300N) to e-NpuNcage-FKBP (Figure S8a, S9). Importantly, all residues flanking the intein were

maintained as native p300 residues to ensure the absence of a splicing scar in the product (Figure S7, S9). Lastly, we appended a degradation tag to the C-terminus of p300N-e-NpuNcage-FKBP to help mitigate high protein concentrations and eliminate any risk of background splicing. As was hoped, co-expression of these constructs in HEK 293T cells resulted in robust rapamycindependent generation of the p300 splice product at concentrations as low as 10nM (Figure S8b). To integrate this p300 activation system with genomic targeting, dCas9 was fused to the N-terminus of p300N-e-NpuNcageFKBP (Figure 3a, S9). Transfection of this construct along with FRB-NpuCcage-p300C and a luciferase reporter plasmid led to a robust increase in luciferase activity in the presence of rapamycin and the appropriate sgRNA (Figure 3b). Importantly, luciferase expression was dependent on both protein splicing and FRB-FKBP dimerization as constructs that were catalytically dead or missing FKBP, respectively, did not show any change in activity in the presence of rapamycin (Figure 3b). Notably, corresponding constructs containing VMA-N and VMA-C in place of e-NpuNcage and NpuCcage displayed no luciferase activation when incubated with rapamycin (Figure 3b, S9), despite equitable expression (Figure S10).

Figure 3: Proximity triggered CPS in mammalian cells. (a) Schematic depicting constructs transfected into mammalian cells for CPS mediated Luciferase expression. (b) Graph depicting luciferase activity of HEK 293T cells transfected with indicated constructs in the presence or absence of 10μM rapamycin (+rapa) for 48hrs. Relative luciferase activation represents the fold-increase in signal observed for a sample in which 3 on-target sgRNAs are used relative to when a scrambled/off target sgRNA is used. N = dCas9-p300N-eNpuNcage-FKBP-Deg, C = FRB-NpuCCage-p300C, FKBP Del

ACS Paragon Plus Environment

Journal of the American Chemical Society 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

= N with FKBP deletion, C1A = N with a NpuN-C1A mutation, VMA = N and C where e-NpuNCage and NpuCCage are replaced with VMA-N and VMA-C respectively. Errors = s.e.m. (n=3).

It has long been appreciated that protein trans splicing provides unique opportunities for controlling protein function at the post-translational level1. Progress in this area has, however, been constrained by the inability to control the activity of naturally split inteins using the proximity triggered paradigm. Herein, we provide a solution to this problem by exploiting recently developed caged versions of these proteins. Our strategy overcomes many of the limitations of previous tools in this area. Indeed, by replacing the unstable split VMA intein by the more robust naturally split Npu intein, we were able to achieve temporal control over the key transcriptional coactivator, p300, in mammalian cells. This application also illustrates the ability to integrate our strategy with other cellular control elements, in the present case genomic targeting using dCas9. While the current method relies on a caged version of Npu, it should be noted that similarly caged versions of several other naturally split inteins are available9. Since these split inteins are all functionally orthogonal to each other and to Npu, they could extend the scope of the technology, including offering the intriguing possibility of multiplexing-type applications. We imagine that the work described herein will serve as a template for such studies.

ASSOCIATED CONTENT Supporting Information. Full methods and experimental data. This material is available free of charge via the Internet at http://pubs.acs.org.

AUTHOR INFORMATION Corresponding Author

*[email protected] Notes The authors declare no competing financial interest.

ACKNOWLEDGMENT The authors thank Adam J. Stevens, Glen P. Liszczak and Robert E. Thompson, and for valuable discussions. This work was supported by the U.S. National Institutes of Health (NIH grant R37-GM086868.)

REFERENCES 1. Shah, N. H.; Muir, T. W., Inteins: Nature's gift to protein chemists. Chem. Sci 2014, 5 (2), 446-461. 2. Novikova, O.; Topilina, N.; Belfort, M., Enigmatic distribution, evolution, and function of inteins. J Biol Chem 2014, 289 (21), 14490-14497.

3. Mootz, H. D.; Muir, T. W., Protein Splicing Triggered by a Small Molecule. J Am Chem Soc 2002, 124 (31), 9044-9045. 4. Buskirk, A. R.; Ong, Y. C.; Gartner, Z. J.; Liu, D. R., Directed evolution of ligand dependence: small-molecule-activated protein splicing. Proc Natl Acad Sci 2004, 101 (29), 10505-10. 5. Peck, S. H.; Chen, I.; Liu, D. R., Directed Evolution of a Small Molecule-Triggered Intein with Improved Splicing Properties in Mammalian Cells. Chem Biol 2011, 18 (5), 619–630. 6. Yuen, C. M.; Rodda, S. J.; Vokes, S. A.; McMahon, A. P.; Liu, D. R., Control of transcription factor activity and osteoblast differentiation in mammalian cells using an evolved small-moleculedependent intein. J Am Chem Soc 2006, 128 (27), 8939–8946. 7. Davis, K. M.; Pattanayak, V.; Thompson, D. B.; Zuris, J. A.; Liu, D. R., Small molecule-triggered Cas9 protein with improved genome-editing specificity. Nat Chem Biol 2015, 11 (5), 316-318. 8. Skretas, G.; Wood, D. W., Regulation of protein activity with small-molecule-controlled inteins. Protein Sci 2005, 14 (2), 523532. 9. Gramespacher, J. A.; Stevens, A. J.; Nguyen, D. P.; Chin, J. W.; Muir, T. W., Intein Zymogens: Conditional Assembly and Splicing of Split Inteins via Targeted Proteolysis. J Am Chem Soc 2017, 139 (24), 8074-8077. 10. Wong, S.; Mosabbir, A. A.; Truong, K., An Engineered Split Intein for Photoactivated Protein Trans-Splicing. PLOS One 2015, 10 (8). 11. Cook, S. N.; Jack, W. E.; Xiong, X.; Danley, L. E.; Ellman, J. A.; Schultz, P. E.; Noren, C. J., Photochemically Initiated Protein Splicing. Angew Chem Int Ed Engl 1995, 34 (15), 1629-1639. 12. Vila-Perelló, M.; Hori, Y.; Ribó, M.; Muir, T. W., Activation of Protein Splicing by Protease- or Light-Triggered O to N Acyl Migration. Angew Chem Int Ed Engl 2008, 47 (40), 7764-7767. 13. Berrade, L.; Kwon, Y.; Camarero, J. A., Photomodulation of protein trans-splicing through backbone photocaging of the DnaE split intein. Chembiochem 2010, 11 (10), 1368-1372. 14. Binschik, J.; Zettler, J.; Mootz, H. D., Photocontrol of protein activity mediated by the cleavage reaction of a split intein. Angew Chem Int Ed Engl 2011, 50 (14), 3249-3252. 15. Jung, D.; Sato, K.; Min, K.; Shigenaga, A.; Jung, J.; Otaka, A.; Kwon, Y., Photo-triggered fluorescent labelling of recombinant proteins in live cells. Chem Commun 2015, 51 (47), 9670-9673. 16. Böcker, J. K.; Friedel, K.; Matern, J. C.; Bachmann, A. L.; Mootz, H. D., Generation of a Genetically Encoded, Photoactivatable Intein for the Controlled Production of Cyclic Peptides. Angew Chem Int Ed Engl 2015, 54 (7), 2116-2120. 17. Ren, W.; Ji, A.; Ai, H. W., Light Activation of Protein Splicing with a Photocaged Fast Intein. J Am Chem Soc 2015, 137 (6), 2155-2158. 18. Tyszkiewicz, A. B.; Muir, T. W., Activation of protein splicing with light in yeast. Nat Methods 2008, 5 (4), 303-305. 19. Jeon, H.; Lee, E.; Kim, D.; Lee, M.; Ryu, J.; Kang, C.; Kim, S.; Kwon, Y., Cell-Based Biosensors Based on Intein-Mediated Protein Engineering for Detection of Biologically Active Signaling Molecules. Anal Chem 2018, 90 (16), 9779-9786. 20. Slomovic, S.; Collins, J. J., DNA sense-and-respond protein modules for mammalian cells. Nat Methods 2015, 12 (11),1085–1090. 21. Sonntag, T.; Mootz, H. D., An intein-cassette integration approach used for the generation of a split TEV protease activated by conditional protein splicing. Mol Biosyst 2011, 7 (6), 2031-2039. 22. Alford, S. C.; O'Sullivan, C.; Obst, J.; Christie, J.; Howard, P. L., Conditional protein splicing of α-sarcin in live cells. Mol Biosyst 2014, 10 (4), 831-837. 23. Schwartz, E. C.; Saez, L.; Young, M. W.; Muir, T. W., Posttranslational enzyme activation in an animal via optimized conditional protein splicing. Nat Chem Biol 2007, 3 (1), 50-54. 24. Mootz, H. D.; Blum, E. S.; Muir, T. W., Activation of an autoregulated protein kinase by conditional protein splicing. Angew Chem Int Ed Engl. 2004, 43 (39), 5189-5192. 25. Brenzel, S.; Kurpiers, T.; Mootz, H. D., Engineering Artificially Split Inteins for Applications in Protein Chemistry: Biochemical Characterization of the Split Ssp DnaB Intein and

ACS Paragon Plus Environment

Page 4 of 9

Page 5 of 9 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Journal of the American Chemical Society Comparison to the Split Sce VMA Intein. Biochem 2006, 45 (6), 15711578. 26. Chong, S.; Montello, G. E.; Zhang, A.; Cantor, E. J.; Liao, W.; Xu, M. Q.; J, B., Utilizing the C-terminal cleavage activity of a protein splicing element to purify recombinant proteins in a single chromatographic step. Nucleic Acids Res 1998, 26 (22), 5109–5115. 27. Chong, S.; Mersha, F. B.; Comb, D. G.; Scott, M. E.; Landry, D.; Vence, L. M.; Perler, F. B.; Benner, J.; Kucera, R. B.; Hirvonen, C. A.; Pelletier, J. J.; Paulus, H.; Xu, M. Q., Single-column purification of free recombinant proteins using a self-cleavable affinity tag derived from a protein splicing element. Gene 1997, 192 (2), 271-281. 28. Chong, S.; Williams, K. S.; Wotkowicz, C.; Xu, M. Q., Modulation of protein splicing of the Saccharomyces cerevisiae vacuolar membrane ATPase intein. Chem 1998, 273 (17), 1056710577. 29. Carvajal-Vallejos, P.; Pallisse, R.; Mootz, H. D.; Schmidt, S. R., Unprecedented Rates and Efficiencies Revealed for New Natural Split Inteins from Metagenomic Sources. J Biol Chem 2012, 287 (34), 28686-28696. 30. Stevens, A. J.; Sekar, G.; Shah, N. H.; Mostafavi, A. Z.; Cowburn, D.; Muir, T. W., A promiscuous split intein with expanded protein engineering applications. Proc Natl Acad Sci 2017, 114 (32), 8538-8543. 31. Stevens, A. J.; Brown, Z. Z.; Shah, N. H.; Sekar, G.; Cowburn, D.; Muir, T. W., Design of a Split Intein with Exceptional Protein Splicing Activity. J. Am. Chem. Soc. 2016, 138 (7), 2162–2165. 32. Stevens, A. J.; Sekar, G.; Gramespacher, J. A.; Cowburn, A.; Muir, T. W., An Atypical Mechanism of Split Intein Molecular Recognition and Folding. J Am Chem Soc 2018, 140 (37), 1179111799. 33. Shah, N. H.; Muir, T. W., Naturally Split Inteins Assemble through a “Capture and Collapse” Mechanism. J Am Chem Soc 2013, 135 (49), 18673-18681. 34. Qi, L. S.; Larson, M. H.; Gilbert, L. A.; Doudna, J. A.; Weissman, J. S.; Arkin, A. P.; Lim, W. A., Repurposing CRISPR as an RNA-Guided Platform for Sequence-Specific Control of Gene Expression. Cell 2013, 152 (52), 1173-1183. 35. Thompson, P. R.; Wang, D.; Wang, L.; Fulco, M.; Pediconi, N.; Zhang, D.; An, W.; Ge, Q.; Roeder. R.G.; Wong, J.; Levrero, M.; Sartorelli, V.; Cotter, R. J.; Cole, P. A., Regulation of the p300 HAT domain via a novel activation loop. Nat Struct Mol Biol 2004, 11 (4), 308-315. 36. Hilton, I. B.; D'Ippolito, A. M.; Vockley, C. M.; Thakore, P. I.; Crawford, G. E.; Reddy, T. E.; Gersbach, C. A., Epigenome editing by a CRISPR-Cas9-based acetyltransferase activates genes from promoters and enhancers. Nat Biotechnol 2015, 33 (5), 510-517.

ACS Paragon Plus Environment

Journal of the American Chemical Society 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Insert Table of Contents artwork here

ACS Paragon Plus Environment

Page 6 of 9

Page 7 of 9 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Journal of the American Chemical Society

Proximity triggered CPS using caged split inteins. (a) Schematic depicting CPS of NpuNCage and NpuCCage through rapamycin induced dimerization of FRB and FKBP. (b-d) Coomassie stained SDS-PAGE gels of splicing reactions (37°, 1μM each protein construct) monitored over time with or without 10μM rapamycin. SP = splice product (MBP-eGFP, MW~ 69kDa). (b-c) CC = FRB-NpuCCage-eGFP. (b) NC = MBP-NpuNCageFKBP. (c) NC = MBP-e-NpuNCage-FKBP, Cas3 = caspase3 (cleaves off cages). (d) N = MBP-VMA-N-FKBP, C = FRB-VMA-C-eGFP. (e) Schematic depicting proximity-triggered FRET changes between Cy3 and Cy5 dyes. (f) Graph depicting observed FRET efficiencies in the presence or absence of rapamycin between FRBNpuCCage-Cy5 and either Cy3-(C1A)-NpuNCage-FKBP or Cy3-e-(C1A)-NpuNCage-FKBP. Errors = s.e.m. (n=3)

ACS Paragon Plus Environment

Journal of the American Chemical Society 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Proximity triggered CPS on a chromatin substrate. (a) Schematic depicting the dCas9-mediated CPS of an extein sequence (myc) onto the N-terminus of histone H3 within a nucleosome. sgRNA = single stranded guide RNA. (b) Gel shift assay in which increasing concentrations of myc-e-NpuNcage-dCas9, 0.1μM NpuCCage-H3 nucleosomes (containing 5’- fluorescein tagged DNA) and on-target or off-target sgRNA were incubated for 16hrs. (c) Western blot against indicated epitope tags of reactions in which 0.5μM of myc-eNpuNcage-dCas9 and 0.1μM of NpuCCage-H3 nucleosomes were incubated with on-target or off-target sgRNA. NC = myc-e-NpuNcage-dCas9, SP = splice product (myc-H3, MW~17kDa), CC-H3 = NpuCCage-H3.

ACS Paragon Plus Environment

Page 8 of 9

Page 9 of 9 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Journal of the American Chemical Society

Proximity triggered CPS in mammalian cells. (a) Schematic depicting constructs transfected into mammalian cells for CPS mediated Luciferase expression. (b) Graph depicting luciferase activity of HEK 293T cells transfected with indicated constructs in the presence or absence of 10μM rapamycin (+rapa) for 48hrs. Relative luciferase activation represents the fold-increase in signal observed for a sample in which 3 ontarget sgRNAs are used relative to when a scrambled/off target sgRNA is used. N = dCas9-p300N-eNpuNcage-FKBP-Deg, C = FRB-NpuCCage-p300C, FKBP Del = N with FKBP deletion, C1A = N with a NpuNC1A mutation, VMA = N and C where e-NpuNCage and NpuCCage are replaced with VMA-N and VMA-C respectively. Errors = s.e.m. (n=3).

ACS Paragon Plus Environment