Combining Sense and Nonsense Codon Reassignment for Site

Dec 14, 2016 - Incorporation of unnatural amino acids (uAAs) via codon reassignment is a powerful approach for introducing novel chemical and biologic...
0 downloads 18 Views 1MB Size
Subscriber access provided by GAZI UNIV

Article

Combining sense and nonsense codon reassignment for siteselective protein modification with unnatural amino acids Zhenling Cui, Zakir Tnimov, Zhong Guo, Mark Polinkovsky, Thomas Durek, Alun Jones, Sergey Mureev, and Kirill Alexandrov ACS Synth. Biol., Just Accepted Manuscript • DOI: 10.1021/acssynbio.6b00245 • Publication Date (Web): 14 Dec 2016 Downloaded from http://pubs.acs.org on December 18, 2016

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 free 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 accessible to all readers and 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.

ACS Synthetic Biology 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 15

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

ACS Synthetic Biology

Combining sense and nonsense codon reassignment for siteselective protein modification with unnatural amino acids Zhenling Cui1, Zakir Tnimov2, Zhong Guo1, Mark Polinkovsky3, Thomas Durek1, Alun Jones1, Sergey Mureev1‡* and Kirill Alexandrov1‡* 1

Institute for Molecular Bioscience, The University of Queensland, St Lucia, QLD, 4072, Australia.

2

Medical Research Council Laboratory of Molecular Biology, Francis Crick Avenue, Cambridge CB2 0QH, United Kingdom.

3

StemProtein (Stemedica Cell Technologies), LevorWood, Inc., 6350 Nancy Ridge Dr, San Diego, CA 92121.

‡These authors contributed equally.

E-mail: [email protected] or [email protected] KEYWORDS: in vitro protein translation, sense codon reassignment, synthetic tRNA, site-specific protein modification, dual protein labelling, single molecular FRET ABSTRACT: Incorporation of unnatural amino acids (uAAs) via codon reassignment is a powerful approach for introducing novel chemical and biological properties to synthesized polypeptides. However, the site-selective incorporation of multiple uAAs into polypeptides is hampered by the limited number of re-assignable nonsense codons. This challenge is addressed in the current work by developing Escherichia coli in vitro translation system depleted of specific endogenous tRNAs. The translational activity in this system is dependent on the addition of synthetic tRNAs for the chosen sense codon. This allows site-selective uAA incorporation via addition of tRNAs pre- or co-translationally charged with uAA. We demonstrate the utility of this system by incorporating the BODIPY fluorophore into the unique AGG codon of the calmodulin(CaM) open reading frame using in vitro pre-charged BODIPY-tRNACysCCU. The deacylated tRNACysCCU is a poor substrate for Cysteinyl-tRNA synthetase which ensures low background incorporation of Cys into the chosen codon. Simultaneously, p-azidophenylalanine mediated amber-codon suppression and its post-translational conjugation to tetramethylrhodamine dibenzocyclooctyne (TAMRA-DIBO) were performed on the same polypeptide. This simple and robust approach takes advantage of the compatibility of BODIPY fluorophore with the translational machinery and thus requires only one post-translational derivatization step to introduce two fluorescent labels. Using this approach, we obtained CaM nearly homogeneously labelled with two FRET-forming fluorophores. The single molecule FRET analysis revealed dramatic changes in the conformation of the CaM probe upon its exposure to Ca2+ or a chelating agent. The presented approach is applicable to other sense codons and can be directly transferred to the eukaryotic cell-free systems.

thetase (o-tRNA/aaRS) pairs derived from Introduction: Tyr tRNA /tyrosyl-tRNA synthetase of Methanocaldococcus The universal nature of the genetic code’s translation into jannaschii (tRNATyr/TyrRS(Mj)) or tRNAPyl/pyrrolysylprotein sequence was considered indisputable after its tRNA synthetase (tRNAPyl/PylRS) of Methanosarcina famielucidation in 1960s1. However, the discovery that selenoly2-4. Yet the efficient codon reassignment still poses a cysteine2 and pyrrolysine3 are genetically encoded by UGA significant challenge as out of three termination codons and UAG codons respectively demonstrated plasticity of naturally devoid of cognate tRNAs only the amber-codon the genetic code and inspired the use of nonsense codons (UAG) is efficient and displays low level of aminoacid for amino acid encoding. Subsequent work by several misincorporation. Suppression of UGA opal, UAA ochre groups established genetic code reassignment techniques codons as well as of quadruplet codons were explored for which enabled incorporation of unnatural amino acids directing co-translational uAA incorporation5-7 but high (uAAs) into polypeptides endowing the latter with novel levels of Trp misincorporation at opal codon5 and poor chemical and biological activities2, 3. To date more than overall suppression of ochre codon6 significantly limit 150 uAAs have been co-translationally incorporated into their practical use. Effective frameshift suppression repolypeptides in Escherichia coli mainly through engiquires modification of the ribosomal decoding center as neered archaeal orthogonal tRNA/aminoacyl tRNA synACS Paragon Plus Environment

ACS Synthetic Biology

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

otherwise the yield of polypeptide with uAA is significantly reduced due to competition with endogenous tRNAs7. Several approaches to achieve multisite specific uAA incorporation by either using two quadruplets suppression8 or by combining amber suppression with quadruplet or ochre suppression9, 10 have been reported. However, the limited number of available nonsense codons and the low efficiency of quadruplet suppression call for the development of new strategies for multisite installation of uAAs. The degeneracy of the genetic code potentially enables orthogonalisation of the redundant sense codons and their reassignment to uAAs3. This was attempted by several groups with partial success6, 11-14. Early example demonstrated reassignment of UUU codons with 80% efficiency to naphthylalanine(Nal) by using engineered yeast tRNAPheAAA and PheRS mutant in Phe-auxotrophic strain15. It is expected that the codons decoded through wobble recognition would be easier to reassign by an otRNA with Watson-Crick base-pairing. The reassignment of UUU (Phe), AAG (Lys), CAU (His), AAU (Asn) codons through wobble recognition was evaluated using GFP reporters only achieving the maximum of 6% suppression efficiency for UUU codon16. The attempt to reassign AGU (Ser) to 3-iodo-phenylalanine led to its 65% incorporation into the position 208 of GFP but no incorporation into the position 72 pointing to strong contextuality17. Due to the low abundance of the corresponding tRNA isoacceptors in E. coli, AGG(Arg) codon was assumed to be the easiest one to reassign. However, an early attempt of AGG-codon reassignment to Nε-tert-butoxycarbonyl-Llysine (BocK) using tRNAPylCUU in the BL21(DE3) strain was unsuccessful6. Subsequently AGG reassignment to various uAAs mediated by TyrRS(Mj)12 or PylRS13, 14 aminoacylation systems were achieved through multiple genetic knockouts, gene complementation, promoter tuning, codon replacements as well as Arg-depleted media to counteract the competing endogenous Arg-tRNA. This points to a strong competition of native aa-tRNAs with the uAA-(o-tRNA) for codon recognition with even the least abundant Arg-tRNA thus representing a major hurdle for quantitative uAA incorporation. The latter becomes particularly important for multi-site protein labelling where otherwise partial suppression at each codon results in a mixed protein population. Hence, effective suppression of competition with the endogenous tRNA is critical. Recently, Suga’s group demonstrated the replacement of redundant isoacceptors for three wobble-restricted codon boxes with synonymous orthogonal counterparts charged with uAAs in the context of a fully reconstituted E. coli in vitro translation system. This enabled production of a peptide containing a library of 20 canonical AAs extended with 3 uAAs18. Favorable competition of o-tRNAs for the reassigned codons with their endogenous counterparts that co-purify with the cognate aaRSs and result in incorporation of native AAs19, is achieved by maintaining ultrahigh o-tRNA/aaRS ratio ranging from 103 to 5x103. If ap-

Page 2 of 15

plied to protein translation such high tRNA excess may affect the yield and the fidelity of the protein synthesis. Sense codon reassignment using S30 cell extract20 would reduce the technical and economic barrier for adoption and provide a platform for rapid generation of protein probes for EPR, NMR, cryoEM and single-molecule fluorescent spectroscopy21. Förster resonance energy transfer carried out at singlemolecule resolution (smFRET) allows precise measurement of the distance between two dyes enabling rapid analysis of protein structure, folding and conformational rearrangements22. Current strategies for site-selective incorporation of a FRET-forming dye pair into proteins rely on the co-translational introduction of two uAAs that provide functionalities for the subsequent orthogonal conjugation with the dyes. One approach relies on simultaneous incorporation of azide and alkyne functionalities followed by their reciprocal and consecutive coppercatalyzed click chemistry coupling5. The main drawbacks of this approach are the utilization of reciprocally compatible reactive groups and requirement for copper (I) which often induces protein aggregation and precipitation. The former limitation requires the removal of the unreacted group before initiating the next conjugation step. To overcome the problem of cross-reactivity a number of elegant methods have been developed ranging from incorporation of mutually nonreactive uAAs such as the tetrazine-/norbornene9 or azide-/ketone- systems10, 23 to essentially perform “one-pot” sequential dual-labelling achieved by either fine-tuning of the reactivity between strain-promoted alkynes and alkenes with tetrazine derivatives24 or combining the tetrazine/alkene with azide/alkyne chemistry25. Although the “one-pot” approach minimizes purification and handling steps, it requires multiple preparatory steps and precise timing to accommodate fine differences of the conjugation kinetics. Therefore, a straightforward strategy for smFRET probe preparation is needed. In this study, we present a generic approach for reassignment of sense codons to uAA in the in vitro translation system. It is based on a generally applicable approach for removal of tRNA of choice from the in vitro translation reaction by DNA-hybridization chromatography. This approach could be potentially applied to a range of tRNAs allowing reassignment of multiple non-identical codons. In order to simplify the introduction of fluorescent uAAs into the chosen positions we developed a chemoenzymatic method that combines co-translational incorporation of a fluorogenic uAA with post translational modification of a second orthogonal group with another fluorophore. The developed approach to the production of the calmodulin proteins furnished with FRET-forming fluorescent groups.

ACS Paragon Plus Environment

Page 3 of 15

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

ACS Synthetic Biology

Results: Freeing AGG codon for uAA incorporation The main obstacle for in vitro sense codon reassignment is the presence of the native tRNA isoacceptors in the cell lysate. We recently demonstrated near-complete depletion of native arginine isoacceptors specific for the AGG codon from E. coli in vitro translation system19. Such system failed to support translation of GFP ORF harboring a single AGG-codon that could be rescued by addition of the synthetic wild type tRNAArgCCU. Semisynthetic tRNA mixtures used in this approach offer great flexibility in composition but are laborious in preparation. Therefore we decided to test an alternative approach where the target tRNA is selectively depleted from the total native tRNA mixture by DNA-hybridization chromatography. In E. coli the AGG codon is decoded by two isoacceptors, tRNAArgCCU and tRNAArgUCU19. The DNA oligonucleotides complementary to the sequence spanning the D-arm down to the anticodon loop or T-arm up to the acceptor stem of of tRNAArgCCU and tRNAArgUCU respectively were used for their chromatographic depletion from the total E. coli tRNA mixture. The translation efficiency of tRNA mixtures before and after tRNAArgCCU/UCU depletion was tested by their ability to support translation of GFP templates with one or six AGG codons (Supplementary Table S3).

depleted total E. coli tRNA at 19 µM. “Total tRNA” represents the total tRNA mixture before depletion and “Depleted tRNA” represents the total tRNA mixture after depletion of the isoacceptors. “Depleted tRNA+t7tRNA” represents depleted tRNA supplemented with 5 µM of synthetic t7tRNAArgCCU or t7tRNASerGCU. The error bars represent standard deviations of three experiments. The use of the total tRNA mixture depleted of AGGdecoding isoacceptors resulted in the negligible background translation of the 1AGG template and no detectable translation for the 6AGG template (Fig. 1A, B). Depending on the number of AGG-codons in the coding templates supplementation of the reactions with t7 polymerase-transcribed tRNAArgCCU (t7tRNAArgCCU) restored translation to the level similar or higher to that achieved with the native total tRNA mixture. According to previous report26, when the E. coli cells grow at rate of 0.4 doubling per hour the proportion of tRNAArgCCU and tRNAArgUCU in the native total mixture is 0.65% and 1.34%, respectively. This corresponds to 0.1 µM and 0.25 µM concentration of the 19 µM native total tRNA mixture. The low abundance of AGG-decoding isoacceptors of the native tRNA mixture is responsible for the lower translational level of GFPcoding template with six AGG codons, compared to the one supplemented with 5 µM t7tRNAArgCCU (Fig. 1A). We chose to use 5 μM of synthetic tRNAArgCCU in our experiments to ensure that this tRNA is present in large excess and is not limiting when templates with both 1 and 6 AGG codons are used. We defined tRNA depletion efficiency as 1-(RFU(Depleted tRNA)/RFU(Depleted tRNA +t7tRNAArgCCU)) (Equation 1). Using this assessment criteria, the depletion efficiencies for the total tRNA range from 89% to 94% (Fig. 1B) when calculated based on 1AGG- template. The depletion efficiencies were almost 100% when calculated based on 6AGG-template (Fig. 1A). In order to test whether this approach can be applied to other codons we performed depletion of the isoacceptor decoding AGC (Ser) codon (Fig. 1C). This isoacceptor is relatively abundant in E. coli accounting for approximately 2.18% of the total tRNA. We show that tRNA fraction depleted for this isoacceptor cannot support translation of templates with 2 consecutive AGC codons, while expression of templates with a single codon was strongly reduced. The depletion efficiencies of AGC-tRNA isoacceptor were approximately 100% and 60% when calculated based on 2AGC-and 1AGC-codon templates, respectively.

Figure 1. Translation efficiency of GFP-coding templates with test-codons in the tRNA-depleted E. coli lysate reconstituted with native or isoacceptor-depleted E. coli tRNA. (A) Translation efficiency of GFP-coding templates with six AGG arginine codons in the presence of native or tRNAArgCCU/UCU depleted total E. coli tRNA. The tRNA concentrations are indicated. (B) As in A but using GFP template with only one AGG codon. (C)Translation efficiency of GFP-coding templates with one or two consecutive AGC serine codons in the presence of native or tRNASerGCU

Reassigning AGG codon to AzF After demonstrating that the AGG codon can be freed of its native tRNA decoder and made available for reassignment, we set out to identify the best tRNA-charging systems that could support this process. We took advantage of the amber-codon suppression to test the ability of the available o-tRNA/aaRS pairs (MjTyr system and Pyl system) in the in vitro translation reaction (Supplementary Fig. S1 and S2). Our findings demonstrated that the orthogonal MjTyr system mediated high level of p-azido-L-

ACS Paragon Plus Environment

ACS Synthetic Biology

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

phenylalanine (AzF, 5 on Supplementary Fig. S1a) incorporation at the amber-codon. AzF features a bioorthogonal azide group that can be derivatized by a range of reagents. MjTyrRS has a shortened anticodon-binding domain27 that relies mainly on G34 for tRNA recognition28. Converting G to C in the first anticodon position in MjtRNATyr impairs its aminoacylation efficiency by approximately 100 fold29. However, it still performs well in amber-codon suppression (Supplementary Fig. S1c, d, e) and 30, 31. Mutating all three anticodon nucleotides (GUA to CAU) was reported to cause >800-fold reduction in aminoacylation efficiency29. Hence mutation of the anticodon to AGGcodon suppressor is expected to require much higher otRNA/enzyme concentrations to overcome the reduction in affinity. We set out to evaluate the possibility of reassigning AGG codon to AzF by combining the lysate lacking isoacceptors for AGG codon with MjTyr system (otRNAAzF2CCU/AzFRS(Mj)/AzF). O-tRNAAzF2CCU was constructed based on pAzPhe232. It was chosen due to the highest efficiency in supporting AzF incorporation among four tRNA variants we tested in our study (Supplementary Fig. S1c). To test the AGG-suppression under various codon contexts, we introduced a single AGG-codon at the position 1 and 151 of two codon-biased GFP templates designated A and B. In these templates the remaining arginines were encoded by the CGN family codons (Supplementary Table S3). The depletion efficiency of native AGG isoacceptors for A_151R, A_1R, B_151R, B_1R were found to be 75%, 80%, 93% and 84%, respectively (Equation 1). For 151R and 1R, supplementation of the reaction with o-tRNAAzF2CCU resulted in 4.2 and 5.2-fold increase in GFP fluorescence for A subsets and 18.4 and 7.2-fold increase for B subsets (Fig. 2). This indicates that more than 80% of GFP protein was modified with AzF. Mass spectroscopy analysis of the translation products demonstrated that most of the protein (>90%) had AzF incorporated at the AGG-defined position with a minor fraction harboring Arg at this site (Supplementary Fig. S4b-c).

Figure 2. AGG reassignment to AzF in the context of four GFP-encoding ORFs. A) Translation efficiency of GFPcoding template A with single AGG codon at the 1st or 151st positon. Template A encoding eGFP with optimised codon composition (Supplementary Table S3) were used for

Page 4 of 15

expression in the cell-free reactions containing tRNAdepleted lysate, AGG-isoacceptors depleted tRNA, AzFRS and AzF with or without o-tRNAAzF2CCU. The final concentrations for o-tRNAAzF2CCU, AzFRS, AzF were 10 µM, 10 µM and 1 mM, respectively. B) as in A but using template B with single AGG codon in the ORF (B_1R and B_151R) (Supplementary Table S3). Template B encodes sGFP with predominantly the unique codons for each type of amino acid.

Reassignment of AGG codon to BODIPY-FL uAA derivative The successful reassignment of AGG to AzF by MjTyr osystem provided us with the orthogonal codon that may be combined with UAG codon for two-site uAAs incorporation. The recently developed bioorthogonal reaction between strained alkenes/alkynes and tetrazine displays rate constants several orders of magnitude higher than earlier versions of click-reactions33. We reasoned that genetic incorporation of strained alkenes/alkynes would be an ideal strategy to achieve fast and selective protein labelling with tetrazine fluorophores. However, we failed to achieve efficient suppression with either of large cycloaliphatic alkynes or alkenes (Supplementary Fig. S1a and b). Propargyl-L-lysine (PrK) is a reasonably efficient substrate of Pyl system (Supplementary Fig. S1a and b). However, the cross-reactivity between PrK and AzF requires iterative use of copper-catalyzed click chemistry5 which would complicate the labelling procedure. In order to circumvent the problem of potential crossreactivity we decided to devise an alternative approach based on the direct incorporation of a fluorescent uAA into one reassigned codon, followed by a click-chemistry based conjugation of the second fluorophore to uAA incorporated into the second reassigned codon. This approach is expected to be simpler than the previously published dual labelling procedures5, 9. Bulky fluorophore-bearing amino acid analogues cannot be efficiently accommodated by aaRS, precluding their co-translational incorporation. Supplementation of tRNAs pre-charged with fluorescent amino acid provides an alternative approach. Although several strategies were developed for pre-charging tRNA with the uAAs all of them are technically challenging. One involves ligation of chemically prepared aminoacylated dinucleotide to a truncated tRNA lacking the 3’-CA dinucleotide34. In an alternative approach a tRNA acylation ribozyme termed flexizyme was developed to in vitro charge uAAs on tRNAs 35. Although this approach is in principle straightforward, it relies on multi-step chemical synthesis and the charging efficiency varies for different uAAs (Supplementary Fig. S3). Further approach is based on coupling of the reporter group to the enzymatically aminoacylated tRNA via the reactive sulfhydryl group of cysteine36 or εamino group of lysine37. We chose to use cysteine due to small size of the aliphatic spacer-arm as well as the fact that cysteinyl-tRNA synthetase (CysRS) is robust enzyme

ACS Paragon Plus Environment

Page 5 of 15

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

ACS Synthetic Biology

that lacks the editing domain38, 39 and, therefore, does not hydrolyze chemically precharged tRNA. We performed in silico modelling using crystal structure on EF-Tu complex with tryptophanyl tRNA40 to assess the ability of the former to accommodate a dye in the place of tryptophan side chain. The spacer separating BPFL ring from the amino-ester backbone was adjusted to 5-mer as in BPFL-tRNACys conjugate (Supplementary Fig. S5). In the resulting model BPFL occupies the pocket in a slightly different orientation as tryptophan without steric clashes (Supplementary Fig. S6). We concluded that BODIPY-FL (BPFL) was sufficiently small to fit in the available binding pocket of the EF-Tu and hence could be efficiently incorporated during protein synthesis. To test this experimentally the synthetic tRNACys variants harboring either CUA or CCU anticodons were aminoacylated in vitro and conjugated to BPFL. HPLC analysis demonstrated the formation of BPFL-tRNACys products with ~15-20% final yield (Fig. 3A, B). Next we introduced the purified BPFL-tRNACysCCU into the “parental” and the tRNA-depleted lysate. Reconstitution of the latter lysate and native total tRNA retained more than 80% activity of its parental lysate as judged either by Western blotting with anti-GFP antibody (lane 1 and 2, Fig 3C) or the total GFP fluorescence signal (Supplementary Fig. S7). The yield of labelled protein in the depleted system supplemented with BPFL-tRNACysCCU increased 1.4 times while the relative labelling efficiency (BPFL/Anti-GFP ratio) was 7-fold higher as compared to parental lysate (lane 8 and 5, Fig. 3C). The yield of labelled protein is estimated to be 18 µg/ml, which is corresponding to 20% suppression efficiency (lane 1 and 8, Fig 3C). The BPFL-tRNA less favorably competes with the native tRNA for decoding the AGG codon in “parental” lysate due to the presence of native AGG-decoding isoacceptors41. The further reduction of

BPFL/Anti-GFP ratio in the reconstituted system of depleted lysate with 19 µM native tRNAs (lane 6, Fig 3C) or 5µM synthetic tRNAArgCCU (lane 7, Fig 3C) indicate that the more abundant of the AGG-decoding tRNA isoacceptors in these systems compete effectively to the BPFL incorporation. Almost no GFP was produced in the depleted translation system supplemented with total tRNA mixture lacking isoacceptors for AGG (lane 4, Fig. 3C). The expression of GFP could be restored by the addition of BPFLtRNACysCCU (lane 8, Fig. 3C) indicating that virtually all expressed polypeptide contained BPFL-fluorophore. We concluded that BPFL-charged tRNA was compatible with the host translational machinery including both the elongation factor and the ribosome. Importantly the tRNACysCCU discharged from uAA during translation undergoes only negligible co-translational recharging with cysteine (Supplementary Fig. S8). The latter is due to the reduced kcat/Km of endogenous CysRS towards transplanted tRNACysCCU39 that fails to compete with the endogenous wt tRNACysGCA. This ensures low level of Cys misincorporation in the developed system. Next we compared amber and AGG codon-mediated BPFL incorporation efficiency using four templates, (B_1X(151X) and B_1R(151R)) that harbor either single amber- or AGGcodon at the 1st and 151st position of GFP ORF (Supplementary Table S3). The translation reactions were supplemented with purified BPFL-tRNACys and primed with the respective templates (Fig. 3D). AGG suppression with BPFL-tRNACysCCU was more robust as it gave 1.4 and 3.4 times higher BPFL incorporation efficiency (fluorescence/total protein ratio) on the 1st and 151st position respectively, than amber suppression. This indicates low competition from the native tRNA isoacceptors for the AGG-codon compared to that of release factor-mediated translation termination on amber-codon.

ACS Paragon Plus Environment

ACS Synthetic Biology

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

Page 6 of 15

Figure 3. Purification of BPFL-tRNACys conjugates and analysis of their suppression efficiencies of AGG and UAG codons. (A) HPLC purification of BPFL-conjugated tRNAs. The tRNACys bearing CUA anticodons charged with Cys by CysRS subsequently reacted with the BPFL iodoacetamide group. Ribonucleic acid and BPFL were detected at 254 and 490nm respectively. (B) As in A but using tRNACys bearing CCU. (C) SDS-PAGE analysis of in vitro translated GFP labelled with BPFL. The BPFL incorporation was detected by fluorescence scanning of the gel after eliminating GFP fluorescence by boiling. The total protein was detected by Western blotting with anti-GFP antibody. The relative density of each band was calculated using ImagJ. The sGFP_T2 template harboring one AGG codon was expressed with or without addition of BPFL-tRNACysCCU in the normal lysate or tRNA depleted lysate supplemented with indicated tRNA mixtures. “Depl. lysate” denotes the tRNA-depleted lysate. “Depl. lysate+depl. tRNA” denotes tRNA depleted lysate supplemented with the total tRNA fraction after depletion for AGG-decoding isoacceptor. “t7tRNAArgCCU” denotes synthetic tRNAArgCCU. The native tRNA mixtures were added to the cell free reaction at 19 µM while synthetic tRNAArgCCU was added to at 5µM. (D) Comparison of BPFL incorporation mediated by amber or AGG codon suppression. The translation reactions for AGG suppression were reconstituted with tRNA-depleted lysate and AGG isoacceptors-depleted tRNA mixture and programmed by template B with single AGG at the 1st or 151st position of GFP ORF with or without BPFL-tRNACysCCU supplementation. Similar reactions were performed for amber suppression but using normal S30 lysate, the template harboring single amber-codon and BPFL-tRNACysCUA suppressor.

Combination of AGG and amber-codon reassignment for dual protein labelling As alluded to in the introduction, the current strategies for site-specific dual protein labelling with a FRETforming dye pair rely on incorporation of two bioorthogonal groups followed by subsequent conjugation reactions delivering the FRET forming fluorophores9, 24. The requirement for the careful control of reactivity of the groups at the uAAs and fluorophores makes these approaches technically challenging. Since BPFL could be incorporated into a protein sequence co-translationally, post-translational installation of the 2nd fluorophore through the genetically encoded uAA becomes more straightforward. We tested this approach by in vitro trans-

lating calmodulin (CaM) protein with AzF as a bioorthogonally reactive uAA and a fluorophore-bearing BPFL-Cys incorporated at 1st and 149th position via amber and AGG codon reassignment, respectively. TAMRAfluorophore (TA), that form a FRET pair with BPFL, was then introduced into the target protein via copper-free click chemistry resulting in double labelled CaM-TA1BPFL149. Fluorescence gel scanning confirmed successful installation of two FRET probes in CaM protein (Fig. 4A). Analysis of the emission spectra of the solution of the purified, double-labelled CaM revealed two distinct peaks (Fig. 4B) corresponding to emission of BPFL at 512 nm

ACS Paragon Plus Environment

Page 7 of 15

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

ACS Synthetic Biology

and TAMRA at 575 nm. To confirm formation of a FRET

pair

Figure 4. Site-specific dual labelling of calmodulin (CaM) for smFRET analysis. (A) Fluorescence scan of SDS-PAGE loaded with mono- and dual- labelled CaM. Dual labelled CaM protein was prepared in cell-free translation system reconstituted from tRNA-depleted lysate and AGG isoacceptors-depleted tRNA mixture. CaM-coding template harboring single amber and AGG codon, respectively, at the 1st and 149th position of its ORF was used (Supplementary Table S3). The gel was scanned in two emission wavelengths. (B) Fluorescence emission spectra of single- and dual- labelled CaM excited at 488 nm. The concentration of TAMRA-labelled protein was adjusted to make sure its emission fluorescence was the same as that of dual labelled protein when excited at 543nm. (C) Structural representation of CaM in Ca2+-free (PDB: 1CFC) and Ca2+-bound form (PDB: 4CLN) indicating the positions of the fluorescent labels. (D-F) smFRET histograms recorded for dual-labelled CaM under different conditions: (D) In 50mM Tris-HCl, 150mM NaCl buffer without Ca2+, (E) with 1mM Ca2+, (F) with 10mM EDTA. The solid lines represent Gaussian fits of the data in two-emission wavelength. we used the same concentration of single-labelled CaMTA1 as a reference. When both samples were excited at 488 nm, the emission fluorescence at 575 nm of double labelled protein was much stronger than that of the protein labelled with TAMRA only, indicating the energy transfer from donor fluorophore BPFL to acceptor fluorophore TAMRA in dually labelled CaM. CaM has four Ca2+-binding sites and binding of Ca2+- results in a large conformational transition that increases the distance between positions 1 and 149 by ~20 Å (Fig. 4C). When using buffers lacking Ca2+, the purified duallabelled CaM displayed two states: the first minor one, with low FRET efficiency (~0.15) and a dominating state with FRET efficiency of ~0.5. This indicates the presence of two conformations where one is presumably Ca2+bound (low FRET efficiency) and another is Ca2+-free (high FRET) (Fig. 4D). The Ca2+-bound form of CaM is present probably due to contamination of the purification and storage buffers with calcium ions42. CaM binds

four calcium ions in a cooperative manner with affinities ranging from micro- to submillimolar43, 44. This can explain the heterogeneity within the 2nd peak possibly reflecting CaM molecules with different number of bound calcium ions. Addition of Ca2+ at 1 mM eliminates the second peak corresponding to the apo-form, narrows the width of the first peak and causes an appearance of another peak centered at ~0.79 (Fig. 4E), which may reflect partial aggregation of the holo-form as was suggested previously45. When EDTA as Ca2+-chelating agent was added to the sample (10 mM), the second peak was enhanced and smoothened (Fig. 4F) representing a majority of CaM in apo-form. The presence of the first peak corresponding to holo-form is consistent to a previous report highlighting the need for additional procedure to obtain fully Ca2+-free form of CaM. Overall the smFRET data are consistent with the available structural information where the fluorophores in the 1st and 149th position of CaM are closer in Ca2+-free form than Ca2+-bound form (Fig. 4C).

ACS Paragon Plus Environment

ACS Synthetic Biology

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

Discussion In this report we describe a new strategy for reassigning sense codon to uAA in the E. coli in vitro translation system lacking selected tRNAs. This is based on a novel procedure for removing chosen tRNA isoacceptors from the total tRNA mixture by DNA-hybridization chromatography. Compared to our previously reported reconstitution approach based on semi-synthetic tRNA complement19, the tRNA-depletion method is more straightforward and efficient. We were able to achieve almost complete depletion of the native isoacceptors for AGG codon making it available for reassignment. In addition to the rare AGG-decoding isoacceptors, we achieved successful depletion of a medium abundant tRNA isoacceptor decoding AGC (Ser) codon. The depleted system for this isoacceptor could not support translation of templates with 2 consecutive AGC codons while expression of templates with a single codon was dropped by 60%. Although the reduction was not as significant as for AGG-codon, we assume the residual amount of AGCdecoding isoacceptor would not prevent efficient uAA incorporation. This assumption is based on the success of reassignment of three redundant codons (GUC, CGC, GGC) in a special designed PURE translational system18 that has higher residual amount of native tRNAs than our depleted system19. We have not proceeded further with this codon in the present study due to inefficient charging of the suppressor tRNA with AzF using AzFRS from Methanocaldococcus jannaschii.

Page 8 of 15

canonical aaRS systems. This may reflect substrateinduced inter-domain dynamics of the aaRS enzymes where amino acid changes within the substrate-binding pockets may affect overall aminoacylation efficiency. Further the eight-membered ring of the strained alkynes/alkenes may cause compatibility issues with EFTu51. Comparison of four engineered o-tRNAs previously reported for the MjTyr system identified o-tRNAAzF2CUA as the most efficient one. The MjTyr system demonstrated less context-dependence in amber codon suppression while the extent of Pyl-mediated suppression was variable. This is consistent with previous findings that some gene contexts are more favorable than others52, 53. We demonstrate that AGG-codon could be reassigned to BPFL fluorophore modified cysteine by using BPFLtRNACysCCU. Preparation of this reagent involves several steps. However, once the precharged tRNA is prepared, the dual labelling procedure becomes much more straightforward and does not require balancing the selectivity of two labelling reactions. The deacylated tRNACys CCU is a poor substrate for protein translation due to the non-native anticodon and the limited concentration of endogenous CysRS and thus undergoes only negligible cotranslational recharging with cysteine preventing its misincorporation into the reassigned codon.

During preparation of our manuscript, Lee et al. demonstrated the success of free Arg-codons for uAA incorporation using Colicin D, a specific Arg-tRNAase, in the S12 lysate46. The reassignable codons accessible with this strategy are confined by the specificity and efficiency of the available tRNAases while the use of DNAhybridization allows predictable reassignment of a larger codon pool47. Both of these methods demonstrate the feasibility of reassignment of orthogonal sense codons in an in vitro translation system.

Orthogonality of sense codon reassignment can be compromised due to the prominent contribution of the anticodon to specificity of many aaRSs. Previous report11 showed that heterologous tRNAPylCCG was charged by native ArgRS and delivered Arg in to the CGG codons in Mycoplasma capricolum. In contrast, Leu tRNAPylGAG did not display unspecific charging in E. coli without the cognate uAA. This is consistent with the findings that LeuRS does not rely on the anticodon for substrate recognition54. From this perspective the tRNAs for Leu, Ser and Ala codons are the best choice for reassignment as their aaRSs does not rely on the anticodon interaction54. However, the successful reassignment of the rare AGG codon in vivo13, 14 and in vitro suggested an efficient charging system or precharged uAA-tRNACCU could bypass the hurdles to avoid mischarging by the endogenous aaRS.

We took advantage of the open nature of cell-free translation systems to test the ability of the most widely-used Pyl and MjTyr suppression systems to incorporate uAAs into nonsense and sense codons. We were particularly interested in some strained alkenes/alkynes due to the high rate constants in bio-orthogonal reaction. However, none of these uAAs demonstrated good incorporation efficiency in our study (Supplementary Fig. S1b). In contrast, n-propargyl-lysine (PrK) could be decoded by PylRSAF48, which featured two mutations (Y306A and Y384F), with relatively high efficiency. Although this result revealed an unexpected inconsistency with the in vivo data on UAG suppression where efficient incorporation of large cycloaliphatic alkynes/alkenes24, 49, earlier reports demonstrated that PylRS was less efficient, with more than 1000-fold reduction in the kcat value50 as compared to

We used the developed methodology to site-selectively dual-label CaM protein with two FRET-forming fluorophores. The successful reassignment of AGG sense codon to uAAs using our newly developed technique demonstrates the feasibility of creating a potentially large number of re-assignable orthogonal codons for reassignment with diverse chemistries in the S30 based E. coli cell-free translation system. Recently, Suga’s group showcased successful division of codon boxes for Val, Arg and Gly where G-ending codons in unsplit codon boxes were reserved for native amino acids while C-ending ones were used for uAA incorporation18. The fully reconstituted in vitro translation system (PURE) enabled manipulation of all the tRNA species. Application of our approach to division of unsplit codon family boxes in the most widely used and inexpensive S30 based cell free translation sys-

ACS Paragon Plus Environment

Page 9 of 15

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

ACS Synthetic Biology

tem, represents an alternative labeling approach applicable not only to peptides but also to full length proteins. Importantly, this approach can be directly transferred to eukaryotic cell-free systems. This sets our approach apart from those based on reconstituted translation systems that create almost unmanageable complexity when applied to eukaryotic systems. Our recent analysis demonstrates that the E.coli cell-free system could express only 10% of human cytosolic proteins in non-aggregated form while eukaryotic systems performed several fold better55. This stresses the need for eukaryotic expression systems tailored for incorporation of the uAA into defined positions.

Materials and methods Materials The unnatural amino acid, p-azido-L-phenylalanine (AzF), was purchased from SynChem. TAMRA DIBO Alkyne were purchased from Life Technologies Australia Pty Ltd. BODIPY® FL Iodoacetamide were from Molecular Probes®. Ethanolamine−sepharose was prepared by coupling ethanolamine on epoxy-activated sepharose 6B (GE Healthcare). Eight gram of epoxy-activated sepharose 6B was washed extensively by water (~200 ml per gram matrix) followed by incubation with 1 M ethanolamine at room temperature overnight with gentle agitation. Following the incubation, the matrix was washed by copious amount of water to remove the traces of ethanolamine. The resulting ethanolamine-conjugated sepharose matrix (~20 ml) was stored as 20% (v/v %) slurry in buffer A (100 mM NaOAc (pH 5.2), 0.25 mM EDTA) with 2 mM NaN3.

tRNA production The sequence of tRNAs used in this study are shown in Supplementary Table S1. The t7 transcripts for the tRNA species were prepared as described before19. In brief, each DNA template was assembled from 5 or 6 oligos (Supplementary Table S2) by 3-step PCR. The PCR products were purified by ethanol precipitation and used as templates for run-off transcription by T7 RNA polymerase. The transcribed tRNAs were purified by affinity chromatography using ethanolamine−sepharose matrix19. Specifically, the transcription reaction was terminated by addition of 0.5 volume of 10 times buffer A and equal volume of 20% matrix followed by addition of 2.5 volumes of water. The slurry was incubated for 1 h at 4° followed by extensive washing with at least 10 volumes of buffer A. The tRNA was eluted twice into one volume of buffer B, 2 M NaOAc (pH 5.2), 0.25 mM EDTA, 2.5 mM Mg(OAc)2, and precipitated by ethanol. Total tRNA preparation: Total tRNA mixture was prepared from E.coli BL21(Gold) strain by modified Zubay’s method56. In brief, the total tRNA was extracted from overnight culture. The cell pel-

let (20 g) was re-suspended in 40 ml of 10 mM Mg(OAc)2, 1 mM Tris (pH 7.4) buffer. The nucleic acids were then extracted by 34.4 ml of liquefied phenol with vigorous agitation for 2 hrs in the cold room. The extraction was repeated again by adding 10 ml liquefied phenol. The aqueous phase was then collected by centrifugation at 18,000 g for 30 min. RNA was precipitated with 0.05 volume 4 M potassium acetate and 2 volume of 100% ethanol for overnight at -20°. The precipitate was collected by a 10 min centrifugation at 5,000 g and re-suspended in 20 ml of 1 M ice-cold NaCl. After stirring the precipitate vigorously for 1 h in cold room, the supernatant was collected and extraction was repeated. The supernatant from the two NaCl extractions was combined and precipitate by addition of 2 vol ethanol. After two repeats the crude tRNA fraction was dissolved in water.

Specific tRNA depletion in total tRNA mixture: The specific tRNA species were depleted by DNAhybridization chromatography from the total mixture as described19. The DNA oligos complementary to the sequence spanning the D-arm down to the anticodon loop (tRNAArgUCU) or variable loop up to the acceptor stem (tRNAArgCCU) or anticodon stem to variable loop (tRNASerGCU) of the target tRNAs (Supplementary Table S2) were designed and synthesized together with 3’-amine group by Integrated DNA Technologies IDT. DNA oligos were immobilized on NHS-activated sepharose (GE) according to the manufacturer’s protocol with 15 nmole of oligonucleotide used per 100 µl of settled resin. 200 µl of 19 µM unfractionated tRNA total mixture was mixed with the same volume of 2× hybridization buffer (20 mM TrisHCl (pH 7.6), 1.8 M NaCl, 0.2 mM EDTA) and then subjected for hybridization with the resin-immobilized oligonucleotides. For each target tRNA, 125 µl of settled resin was used. The tRNA mixture suspended with the resin was heat-denatured at 65° for 10 min, slowly cooled down to room temperature with agitation followed by collection of the non-hybridized tRNA-fraction devoid of targeted isoacceptors by centrifugation. 20 µl of 1× hybridization buffer used to wash the resin were combined with the main fraction of tRNAs and ethanol precipitated. tRNA aminoacylation and fluorophore conjugation The synthetic cysteine tRNAs with CCU or CUA anticodons were prepared by t7 transcription as described above. Cysteine (Cys) was charged on these tRNAs by recombinant cysteinyl-tRNA synthetase (Supplementary methods). Firstly, 50 mM Cys was pre-incubated with 50 mM TCEP at 37° for 15 min in order to convert it to fully reduced form. Meantime, tRNAs at 50 µM concentration were denatured at 78° and refolded in the presence of 5 mM Mg2+ slowly at room temperature to maintain its maximal activity. The aminoacylation reaction was then performed in 100 mM Hepes-KOH (pH 8.0), 2 mM Cys, 2 mM TCEP, 10 mM MgCl2, 5 mM KCl, 5 mM KOAc, 0.1 mM CTP, 4 mM ATP, 10 µM t7tRNACys variant, 5 µM CysRS, 25% DMSO at 37° for 1 h.

ACS Paragon Plus Environment

ACS Synthetic Biology

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

After aminoacylation, the reaction mixture was diluted into 1× buffer A for ethanolamine-sepharose purification followed by conjugation reaction with BODIPY® FL Iodoacetamide (BPFL-IA). The conjugation reaction was performed in dark in 50 mM Tris-HCl (pH 8.5), 80 µM cysteinylated-tRNA, 75% DMSO, 1 mM BPFL-IA and 100 mM NaCl. Conjugated product (BPFL-tRNA) was precipitated by ethanol and dissolved in a minimal volume of tRNA buffer (0.5 mM MgCl2, 0.5 mM NaOAc pH 5.0). The sample was then adjusted to 1× HPLC buffer (0.1 M TEAA, 1% ACN) followed purification by HPLC on POROS® R1 10 µm column (Applied Biosystems) using buffers both containing 0.1M TEAA and either 1% (buffer C) or 90% of ACN (buffer D); 9 min run duration with 1-20% linear gradient was applied. After HPLC purification, the BPFL-tRNA fraction was precipitated by ethanol, re-suspended in tRNA buffer and stored at -80°.

In vitro protein translation assay The tRNA-depleted lysate is prepared by affinity chromatography as described19. DNA templates corresponding to GFP and CaM-coding ORFs were constructed as described in Supplementary methods. The sequence information of all the open reading frames used here is provided in Supplementary Table S3. The cell-free translation reactions for GFP production were performed following the standard protocol20 with the Mg2+ concentration optimized at 10 mM and 0.35 volume of depleted lysate (same as that for the standard S30 lysate). Any additional modifications to the reaction setup were given in the description of the corresponding experiments. GFP production was monitored on a fluorescence spectrometer with automatic plate reading module (Synergy) at 30° for 3-5 h at 485 nm excitation and 528 nm emission wavelengths. Protein labelling Expression of CaM with AzF and BPFL in cell-free translation reaction: The pOPINE-CaM template, with UAG and AGG codon located at position 1 and 149 of CaM ORF respectively (Supplementary Table S3), were supplemented at 80 ng/µl in the cell-free translation reaction containing 19 µM depleted tRNA mixture, 10 µM o-tRNAAzF2CUA, 10 µM AzFRS, 1 mM AzF, 10 µM BPFL-tRNACysCCU in addition to the standard reagents20. Two separate reactions were performed to prepare single labelled CaM protein. To express CaM-BPFL, 10 µM BPFL-tRNACysCCU was supplemented to the corresponding translation reaction while 10 µM tRNATyrCUA was added for decoding UAG codon to Tyr. To obtain CaM-TAMRA, AzF was incorporated into protein sequence through the amber codon with 10 µM otRNAAzF2CUA, 10 µM AzFRS and 1 mM AzF while 5 µM synthetic tRNAArgCCU was supplemented for decoding AGG codon to Arg. The AzF was then modified by TAMRA DIBO through copper-free click chemistry as described below. The translation reaction was performed at 32° for 4h. Purification of labelled CaM on Affinity Clamp resin: 20 µl 50% affinity clamp matrix (Supplementary methods) pre-

Page 10 of 15

blocked with 0.1 mg/ml of BSA in PBS was added to the clarified translation sample. The resin was incubated with the sample for 30 min at room temperature, washed with PBS-Triton (0.1%) buffer following overnight incubation with 60 µM TAMRA-DIBO in PBS at room temperature to allow TAMRA conjugation with the protein. After conjugation the resin was extensively washed with 20 mM Hepes-KOH pH 7.6, 0.5 M NaCl, 0.1 mg/ml BSA, 0.1% Triton buffer alternated by washes with just 0.1 mg/ml BSA, 0.1% Triton in water. Before elution via cleavage with PreScission protease the resin was equilibrated with the cleavage buffer containing 50mM Tris-HCl pH 8, 150 mM NaCl, 1 mM DTT. Subsequently 40 µl PreScission protease (0.1 mg/ml) in cleavage buffer was added to the matrix followed by overnight incubation at 16°. The protein was eluted by centrifugation and the beads were additionally eluted with the same volume of cleavage buffer. The two eluates were combined.

smFRET measurements smFRET measurements were carried out using a LSM710 microscope equipped with the ConfoCor3 module and BiG detectors. Excitation was achieved by focusing 488 nm Argon-ion laser light into 16 µl sample solution using a water immersion objective (Apochromat 1.2 NA, 40X; Zeiss). The fluorescence emission was separated from excitation light using a 488/561 dichroic mirror, collected using the same objective, spatially filtered through the 35 µm pinhole followed by splitting into donor and acceptor detectors using a second dichroic mirror and band-path 500–550 nm and 570-610 nm filters for donor and acceptor emissions, respectively, mounted into the same filter cube unit (Zeiss). Experiments were performed in 50 mM Tris-HCl (pH 7.5), 150 mM NaCl, 50 µg/ml BSA, either in the presence of 1 mM Ca2+ or 10 mM EDTA, loaded on a home-made silicone plate (SYLGARD®) held on a 70× 80 mm Coverglass No.1.5 (ProSciTech). Double labelled CaM was used at ~100 pM concentrations to ensure that the detected signal would be sourced by a single molecule. The FRET efficiency histograms were generated by using Zen10 software where a two-channel data collection mode was used for simultaneous recording of donor and acceptor signals for 30 min (in 10 rounds by 3 min each) with a bin time of 1 ms. The leakage of donor emission into the acceptor channel was estimated in a separate experiment with 1 µM of donor fluorophore and calculated as 12% to correct the signals before FRET analysis. A threshold was set at 20 counts for the sum of donor and acceptor signals to filter the background noise out. The FRET efficiency was calculated as (IA – 0.12*ID)/ (IA + 0.88*ID) and plotted as a histogram. IA and ID are light intensities in acceptor and donor channel, respectively. 0.12 is the leakage fraction from Donor emission in the acceptor channel. Gaussian function was used to fit the data by Origin software (OriginLab Corp.).

ASSOCIATED CONTENT

ACS Paragon Plus Environment

Page 11 of 15

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

ACS Synthetic Biology

Supporting Information Details on plasmid construction, recombinant production of aaRSs, chemical synthesis of uAAs and precharging on tRNAs, mass spectrum, structural modelling are included. This material is available free of charge via the Internet at http://pubs.acs.org.

AUTHOR INFORMATION Corresponding Author *Kirill Alexandrov or Sergey Mureev [email protected] or [email protected]

Author Contributions The manuscript was written through contributions of all authors. / All authors have given approval to the final version of the manuscript. / ‡These authors contributed equally.

ACKNOWLEDGMENT We thank Dejan Gagoski for discussion on protein labelling and microscopy setup. We are grateful to Hadi Ahmad Fuaad for allowing access to their HPLC facility. We acknowledge the support from ARC grant DP1094080, and NHMRC program grant APP1037320 to K.A.

REFERENCES

ACS Paragon Plus Environment

ACS Synthetic Biology

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

Page 12 of 15

[1] Crick, F. H. (1968) The origin of the genetic code, J Mol Biol 38, 367-379. [2] Liu, C. C., and Schultz, P. G. (2010) Adding New Chemistries to the Genetic Code, Annu Rev Biochem 79, 413-444. [3] O'Donoghue, P., Ling, J., Wang, Y. S., and Soll, D. (2013) Upgrading protein synthesis for synthetic biology, Nat Chem Biol 9, 594-598. [4] Dumas, A., Lercher, L., Spicer, C. D., and Davis, B. G. (2015) Designing logical codon reassignment – Expanding the chemistry in biology, Chem. Sci. 6, 50-69. [5] Wan, W., Huang, Y., Wang, Z. Y., Russell, W. K., Pai, P. J., Russell, D. H., and Liu, W. R. (2010) A Facile System for Genetic Incorporation of Two Different Noncanonical Amino Acids into One Protein in Escherichia coli, Angew Chem Int Edit 49, 3211-3214. [6] Odoi, K. A., Huang, Y., Rezenom, Y. H., and Liu, W. R. (2013) Nonsense and sense suppression abilities of original and derivative Methanosarcina mazei pyrrolysyl-tRNA synthetasetRNA(Pyl) pairs in the Escherichia coli BL21(DE3) cell strain, Plos One 8, e57035. [7] Neumann, H., Wang, K. H., Davis, L., Garcia-Alai, M., and Chin, J. W. (2010) Encoding multiple unnatural amino acids via evolution of a quadruplet-decoding ribosome, Nature 464, 441-444. [8] Kajihara, D., Abe, R., Iijima, I., Komiyama, C., Sisido, M., and Hohsaka, T. (2006) FRET analysis of protein conformational change through position-specific incorporation of fluorescent amino acids, Nat Methods 3, 923-929. [9] Wang, K. H., Sachdeva, A., Cox, D. J., Wilf, N. W., Lang, K., Wallace, S., Mehl, R. A., and Chin, J. W. (2014) Optimized orthogonal translation of unnatural amino acids enables spontaneous protein double-labelling and FRET, Nat Chem 6, 393-403. [10] Wu, B., Wang, Z., Huang, Y., and Liu, W. R. (2012) Catalyst-free and site-specific one-pot duallabeling of a protein directed by two genetically incorporated noncanonical amino acids, Chembiochem 13, 1405-1408. [11] Krishnakumar, R., Prat, L., Aerni, H. R., Ling, J. Q., Merryman, C., Glass, J. I., Rinehart, J., and Soll, D. (2013) Transfer RNA Misidentification Scrambles Sense Codon Recoding, Chembiochem 14, 1967-1972. [12] Lee, B. S., Shin, S., Jeon, J. Y., Jang, K. S., Lee, B. Y., Choi, S., and Yoo, T. H. (2015) Incorporation of Unnatural Amino Acids in Response to the AGG Codon, ACS Chem Biol 10, 1648-1653. [13] Mukai, T., Yamaguchi, A., Ohtake, K., Takahashi, M., Hayashi, A., Iraha, F., Kira, S., Yanagisawa, T., Yokoyama, S., Hoshi, H., Kobayashi, T., and Sakamoto, K. (2015) Reassignment of a rare sense codon to a non-canonical amino acid in Escherichia coli, Nucleic Acids Res 43, 8111-8122. [14] Zeng, Y., Wang, W., and Liu, W. S. R. (2014) Towards Reassigning the Rare AGG Codon in Escherichia coli, Chembiochem 15, 1750-1754. [15] Kwon, I., Kirshenbaum, K., and Tirrell, D. A. (2003) Breaking the degeneracy of the genetic code, J Am Chem Soc 125, 7512-7513. [16] Biddle, W., Schmitt, M. A., and Fisk, J. D. (2015) Evaluating Sense Codon Reassignment with a Simple Fluorescence Screen, Biochemistry 54, 7355-7364. [17] Ho, J. M., Reynolds, N. M., Rivera, K., Connolly, M., Guo, L. T., Ling, J., Pappin, D. J., Church, G. M., and Soll, D. (2016) Efficient Reassignment of a Frequent Serine Codon in Wild-Type Escherichia coli, ACS Synth Biol 5, 163-171. [18] Iwane, Y., Hitomi, A., Murakami, H., Katoh, T., Goto, Y., and Suga, H. (2016) Expanding the amino acid repertoire of ribosomal polypeptide synthesis via the artificial division of codon boxes, Nat Chem. [19] Cui, Z., Stein, V., Tnimov, Z., Mureev, S., and Alexandrov, K. (2015) Semisynthetic tRNA complement mediates in vitro protein synthesis, J Am Chem Soc 137, 4404-4413. [20] Schwarz, D., Junge, F., Durst, F., Frolich, N., Schneider, B., Reckel, S., Sobhanifar, S., Dotsch, V., and Bernhard, F. (2007) Preparative scale expression of membrane proteins in Escherichia colibased continuous exchange cell-free systems, Nat Protoc 2, 2945-2957. [21] Quast, R. B., Mrusek, D., Hoffmeister, C., Sonnabend, A., and Kubick, S. (2015) Cotranslational incorporation of non-standard amino acids using cell-free protein synthesis, Febs Lett 589, 17031712. [22] Michalet, X., Weiss, S., and Jager, M. (2006) Single-molecule fluorescence studies of protein folding and conformational dynamics, Chem Rev 106, 1785-1813. [23] Chatterjee, A., Sun, S. B., Furman, J. L., Xiao, H., and Schultz, P. G. (2013) A Versatile Platform for Single- and Multiple-Unnatural Amino Acid Mutagenesis in Escherichia coli, Biochemistry 52, 1828-1837. [24] Nikic, I., Plass, T., Schraidt, O., Szymanski, J., Briggs, J. A. G., Schultz, C., and Lemke, E. A. (2014) Minimal Tags for Rapid Dual-Color Live-Cell Labeling and Super-Resolution Microscopy, Angew Chem Int Edit 53, 2245-2249. [25] Sachdeva, A., Wang, K. H., Elliott, T., and Chin, J. W. (2014) Concerted, Rapid, Quantitative, and Site-Specific Dual Labeling of Proteins, J Am Chem Soc 136, 7785-7788. 12 ACS Paragon Plus Environment

Page 13 of 15

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

ACS Synthetic Biology

[26] Dong, H. J., Nilsson, L., and Kurland, C. G. (1996) Co-variation of tRNA abundance and codon usage in Escherichia coli at different growth rates, J Mol Biol 260, 649-663. [27] Steer, B. A., and Schimmel, P. (1999) Major anticodon-binding region missing from an archaebacterial tRNA synthetase, J Biol Chem 274, 35601-35606. [28] Kobayashi, T., Nureki, O., Ishitani, R., Yaremchuk, A., Tukalo, M., Cusack, S., Sakamoto, K., and Yokoyama, S. (2003) Structural basis for orthogonal tRNA specificities of tyrosyl-tRNA synthetases for genetic code expansion, Nat Struct Biol 10, 425-432. [29] Fechter, P., Rudinger-Thirion, J., Tukalo, M., and Giege, R. (2001) Major tyrosine identity determinants in Methanococcus jannaschii and Saccharomyces cerevisiae tRNA(Tyr) are conserved but expressed differently, Eur J Biochem 268, 761-767. [30] Young, T. S., Ahmad, I., Yin, J. A., and Schultz, P. G. (2010) An Enhanced System for Unnatural Amino Acid Mutagenesis in E. coli, J Mol Biol 395, 361-374. [31] Albayrak, C., and Swartz, J. R. (2013) Cell-free co-production of an orthogonal transfer RNA activates efficient site-specific non-natural amino acid incorporation, Nucleic Acids Res 41, 5949-5963. [32] Guo, J. T., Melancon, C. E., Lee, H. S., Groff, D., and Schultz, P. G. (2009) Evolution of Amber Suppressor tRNAs for Efficient Bacterial Production of Proteins Containing Nonnatural Amino Acids, Angew Chem Int Edit 48, 9148-9151. [33] Lang, K., and Chin, J. W. (2014) Bioorthogonal reactions for labeling proteins, ACS Chem Biol 9, 16-20. [34] Hecht, S. M., Alford, B. L., Kuroda, Y., and Kitano, S. (1978) "Chemical aminoacylation" of tRNA's, J Biol Chem 253, 4517-4520. [35] Goto, Y., Katoh, T., and Suga, H. (2011) Flexizymes for genetic code reprogramming, Nat Protoc 6, 779-790. [36] Gubbens, J., Kim, S. J., Yang, Z., Johnson, A. E., and Skach, W. R. (2010) In vitro incorporation of nonnatural amino acids into protein using tRNA(Cys)-derived opal, ochre, and amber suppressor tRNAs, RNA 16, 1660-1672. [37] Johnson, A. E., Woodward, W. R., Herbert, E., and Menninger, J. R. (1976) Nepsilon-acetyllysine transfer ribonucleic acid: a biologically active analogue of aminoacyl transfer ribonucleic acids, Biochemistry 15, 569-575. [38] Newberry, K. J., Hou, Y. M., and Perona, J. J. (2002) Structural origins of amino acid selection without editing by cysteinyl-tRNA synthetase, Embo J 21, 2778-2787. [39] Hauenstein, S., Zhang, C. M., Hou, Y. M., and Perona, J. J. (2004) Shape-selective RNA recognition by cysteinyl-tRNA synthetase, Nat Struct Mol Biol 11, 1134-1141. [40] Voorhees, R. M., Schmeing, T. M., Kelley, A. C., and Ramakrishnan, V. (2010) The Mechanism for Activation of GTP Hydrolysis on the Ribosome, Science 330, 835-838. [41] Dong, H. J., Nilsson, L., and Kurland, C. G. (1996) Co-variation of tRNA abundance and codon usage in Escherichia coli at different growth rates, J Mol Biol 260, 649-663. [42] Crouch, T. H., and Klee, C. B. (1980) Positive cooperative binding of calcium to bovine brain calmodulin, Biochemistry 19, 3692-3698. [43] Ye, Y., Lee, H. W., Yang, W., Shealy, S., and Yang, J. J. (2005) Probing site-specific calmodulin calcium and lanthanide affinity by grafting, J Am Chem Soc 127, 3743-3750. [44] Wu, G., Gao, Z., Dong, A., and Yu, S. (2012) Calcium-induced changes in calmodulin structural dynamics and thermodynamics, Int J Biol Macromol 50, 1011-1017. [45] Lafitte, D., Heck, A. J., Hill, T. J., Jumel, K., Harding, S. E., and Derrick, P. J. (1999) Evidence of noncovalent dimerization of calmodulin, Eur J Biochem 261, 337-344. [46] Lee, K. B., Hou, C. Y., Kim, C. E., Kim, D. M., Suga, H., and Kang, T. J. (2016) Genetic Code Expansion by Degeneracy Reprogramming of Arginyl Codons, Chembiochem 17, 1198-1201. [47] Miyauchi, K., Ohara, T., and Suzuki, T. (2007) Automated parallel isolation of multiple species of non-coding RNAs by the reciprocal circulating chromatography method, Nucleic Acids Res 35, e24. [48] Yanagisawa, T., Ishii, R., Fukunaga, R., Kobayashi, T., Sakamoto, K., and Yokoyama, S. (2008) Multistep Engineering of Pyrrolysyl-tRNA Synthetase to Genetically Encode N(epsilon)-(oAzidobenzyloxycarbonyl) lysine for Site-Specific Protein Modification, Chem Biol 15, 11871197. [49] Lang, K., Davis, L., Wallace, S., Mahesh, M., Cox, D. J., Blackman, M. L., Fox, J. M., and Chin, J. W. (2012) Genetic Encoding of bicyclononynes and trans-cyclooctenes for site-specific protein labeling in vitro and in live mammalian cells via rapid fluorogenic Diels-Alder reactions, J Am Chem Soc 134, 10317-10320. [50] Guo, L. T., Wang, Y. S., Nakamura, A., Eiler, D., Kavran, J. M., Wong, M., Kiessling, L. L., Steitz, T. A., O'Donoghue, P., and Soll, D. (2014) Polyspecific pyrrolysyl-tRNA synthetases from directed evolution, Proc Natl Acad Sci U S A 111, 16724-16729. [51] Ieong, K. W., Pavlov, M. Y., Kwiatkowski, M., Forster, A. C., and Ehrenberg, M. (2012) Inefficient Delivery but Fast Peptide Bond Formation of Unnatural L-Aminoacyl-tRNAs in Translation, J Am Chem Soc 134, 17955-17962. 13 ACS Paragon Plus Environment

ACS Synthetic Biology

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

Page 14 of 15

[52] Bossi, L., and Ruth, J. R. (1980) The influence of codon context on genetic code translation, Nature 286, 123-127. [53] Pott, M., Schmidt, M. J., and Summerer, D. (2014) Evolved sequence contexts for highly efficient amber suppression with noncanonical amino acids, ACS Chem Biol 9, 2815-2822. [54] Giege, R., Sissler, M., and Florentz, C. (1998) Universal rules and idiosyncratic features in tRNA identity, Nucleic Acids Res 26, 5017-5035. [55] Gagoski, D., Polinkovsky, M. E., Mureev, S., Kunert, A., Johnston, W., Gambin, Y., and Alexandrov, K. (2016) Performance benchmarking of four cell-free protein expression systems, Biotechnol Bioeng 113, 292-300. [56] Zubay, G. (1962) Isolation and Fractionation of Soluble Ribonucleic Acid, J Mol Biol 4, 347-&.

14 ACS Paragon Plus Environment

Page 15 of 15

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

ACS Synthetic Biology

For Table of Contents Use Only

Combining sense and nonsense codon reassignment for siteselective protein modification with unnatural amino acids Zhenling Cui1, Zakir Tnimov2, Zhong Guo1, Mark Polinkovsky3, Thomas Durek1, Alun Jones1, Sergey Mureev1‡* and Kirill Alexandrov1‡*

15 ACS Paragon Plus Environment