Abstract

•Genetically encoded lysine-selective protein photo-crosslinker in living cells•Capturing protein interactions with lysine-selectivity in a temporal-resolved manner•Producing residue-predictable photo-crosslinking site and crosslinked peptides•Capture of elusive enzyme-substrate interaction and validation of lysine PTM site Protein-protein interactions play pivotal roles in virtually all cellular processes. The identification of transient and weak protein interactions in living cells still remains challenging with traditional strategies. Combining advantages of temporal control and the unique ability of residue selectivity, photo-crosslinkers offer are a powerful tool for identification of dynamic, weak, and transient protein interactions in living cells with specificity, accuracy, and reliability, thus, for better understanding of biological processes and pathophysiology. Investigating photo-crosslinking with residue selectivity may lead to the discovery of new photo-crosslinkers and help in expanding the repertoire of current protein crosslinking technology and interactive proteomics. Consequently, this approach will have an effect on a wide range of fields involving protein-protein and ligand-receptor interactions such as basic biological studies, interactive proteomics, protein structural studies, and chemical biology. The genetically encoded non-selective photo-crosslinkers suffer from unpredictable crosslinking sites and false positive interacting proteins, thus, the identification of protein interactions and mapping accurate interaction interfaces remain largely elusive in living systems. Here, we report genetically encoded, residue-selective protein photo-crosslinker enabling covalent crosslinking of the interacting proteins with proximal lysine, upon UV light activation in vitro and in living cells. Using this photo-crosslinker, we demonstrate a crosslinking strategy to capture protein-protein interactions with residue selectivity in a temporal-resolved manner. In addition, this crosslinking strategy produces the lysine-selective crosslinking site and predictable crosslinked peptide, which serve as the direct evidence for protein-protein interactions and facilitate mass spectrometry analysis. More importantly, this photo-crosslinker enables the capture of elusive enzyme-substrate interaction with directly interacting lysine and validation of acetylation site of the substrate. This strategy represents higher spatiotemporal resolution, accuracy, and reliability for investigating dynamic, transient, and weak protein-protein interactions in living systems. The genetically encoded non-selective photo-crosslinkers suffer from unpredictable crosslinking sites and false positive interacting proteins, thus, the identification of protein interactions and mapping accurate interaction interfaces remain largely elusive in living systems. Here, we report genetically encoded, residue-selective protein photo-crosslinker enabling covalent crosslinking of the interacting proteins with proximal lysine, upon UV light activation in vitro and in living cells. Using this photo-crosslinker, we demonstrate a crosslinking strategy to capture protein-protein interactions with residue selectivity in a temporal-resolved manner. In addition, this crosslinking strategy produces the lysine-selective crosslinking site and predictable crosslinked peptide, which serve as the direct evidence for protein-protein interactions and facilitate mass spectrometry analysis. More importantly, this photo-crosslinker enables the capture of elusive enzyme-substrate interaction with directly interacting lysine and validation of acetylation site of the substrate. This strategy represents higher spatiotemporal resolution, accuracy, and reliability for investigating dynamic, transient, and weak protein-protein interactions in living systems. Protein-protein interactions play crucial roles in regulation of many biological processes. However, the identification of dynamic, transient, and weak protein-protein interactions in living cells still remains highly challenging with traditional strategies.1Berggård T. Linse S. James P. Methods for the detection and analysis of protein-protein interactions.Proteomics. 2007; 7: 2833-2842Crossref PubMed Scopus (422) Google Scholar, 2Dunham W.H. Mullin M. Gingras A.C. Affinity-purification coupled to mass spectrometry: basic principles and strategies.Proteomics. 2012; 12: 1576-1590Crossref PubMed Scopus (208) Google Scholar In comparison with non-covalent binding approaches such as immunoprecipitation and affinity purification, crosslinking of protein complexes with covalent linkage irreversibly captures the interacting protein partners and tolerates stringent processing, conferring higher reliability, specificity, and accuracy.3Yu C. Huang L. Cross-linking mass spectrometry: an emerging technology for interactomics and structural biology.Anal. Chem. 2018; 90: 144-165Crossref PubMed Scopus (173) Google Scholar, 4Sinz A. Cross-linking/mass spectrometry for studying protein structures and protein-protein interactions: where are we now and where should we go from here?.Angew. Chem. Int. Ed. 2018; 57: 6390-6396Crossref PubMed Scopus (111) Google Scholar, 5Preston G.W. Wilson A.J. Photo-induced covalent cross-linking for the analysis of biomolecular interactions.Chem. Soc. Rev. 2013; 42: 3289-3301Crossref PubMed Scopus (135) Google Scholar, 6Pham N.D. Parker R.B. Kohler J.J. Photocrosslinking approaches to interactome mapping.Curr. Opin. Chem. Biol. 2013; 17: 90-101Crossref PubMed Scopus (96) Google Scholar The stable covalent linkage formation with proximal residues potentially enabling the capture and identification of dynamic, weak, and transient protein interactions. Several elegant methodologies have been developed via chemical crosslinkers.3Yu C. Huang L. Cross-linking mass spectrometry: an emerging technology for interactomics and structural biology.Anal. Chem. 2018; 90: 144-165Crossref PubMed Scopus (173) Google Scholar, 4Sinz A. Cross-linking/mass spectrometry for studying protein structures and protein-protein interactions: where are we now and where should we go from here?.Angew. Chem. Int. Ed. 2018; 57: 6390-6396Crossref PubMed Scopus (111) Google Scholar, 7Young M.M. Tang N. Hempel J.C. Oshiro C.M. Taylor E.W. Kuntz I.D. Gibson B.W. Dollinger G. High throughput protein fold identification by using experimental constraints derived from intramolecular cross-links and mass spectrometry.Proc. Natl. Acad. Sci. USA. 2000; 97: 5802-5806Crossref PubMed Scopus (392) Google Scholar, 8Herzog F. Kahraman A. Boehringer D. Mak R. Bracher A. Walzthoeni T. Leitner A. Beck M. Hartl F.U. Ban N. et al.Structural probing of a protein phosphatase 2A network by chemical cross-linking and mass spectrometry.Science. 2012; 337: 1348-1352Crossref PubMed Scopus (312) Google Scholar, 9Shi Y. Pellarin R. Fridy P.C. Fernandez-Martinez J. Thompson M.K. Li Y.Y. Wang Q.J. Sali A. Rout M.P. Chait B.T. A strategy for dissecting the architectures of native macromolecular assemblies.Nat. Methods. 2015; 12: 1135-1138Crossref PubMed Scopus (83) Google Scholar Chemical crosslinkers coupled with mass spectrometry (MS) analysis are versatile, but the reactive small molecules used are generally incompatible with living systems, which therefore have been mainly restricted to large protein complexes with stable interactions. The highly reactive small molecules simultaneously react with all targeted residues in pairs on protein surfaces with non-selectivity and generate excess number of covalent linkages, which will complicate MS analysis and artificially alter protein interactions and protein structure conformation.10Kalkhof S. Sinz A. Chances and pitfalls of chemical cross-linking with amine-reactive N-hydroxysuccinimide esters.Anal. Bioanal. Chem. 2008; 392: 305-312Crossref PubMed Scopus (197) Google Scholar, 11Mädler S. Bich C. Touboul D. Zenobi R. Chemical cross-linking with NHS esters: a systematic study on amino acid reactivities.J. Mass Spectrom. 2009; 44: 694-706Crossref PubMed Scopus (199) Google Scholar, 12Merkley E.D. Rysavy S. Kahraman A. Hafen R.P. Daggett V. Adkins J.N. Distance restraints from crosslinking mass spectrometry: mining a molecular dynamics simulation database to evaluate lysine-lysine distances.Protein Sci. 2014; 23: 747-759Crossref PubMed Scopus (175) Google Scholar, 13Chen Z.A. Jawhari A. Fischer L. Buchen C. Tahir S. Kamenski T. Rasmussen M. Lariviere L. Bukowski-Wills J.C. Nilges M. et al.Architecture of the RNA polymerase II-TFIIF complex revealed by cross-linking and mass spectrometry.EMBO J. 2010; 29: 717-726Crossref PubMed Scopus (324) Google Scholar Alternatively, photo-crosslinking of protein complexes in a covalent manner has become a highly valuable technique for mapping dynamic or weak protein-protein interactions.5Preston G.W. Wilson A.J. Photo-induced covalent cross-linking for the analysis of biomolecular interactions.Chem. Soc. Rev. 2013; 42: 3289-3301Crossref PubMed Scopus (135) Google Scholar, 6Pham N.D. Parker R.B. Kohler J.J. Photocrosslinking approaches to interactome mapping.Curr. Opin. Chem. Biol. 2013; 17: 90-101Crossref PubMed Scopus (96) Google Scholar, 14Nguyen T.A. Cigler M. Lang K. Expanding the genetic code to study protein-protein interactions.Angew. Chem. Int. Ed. 2018; 57: 14350-14361Crossref PubMed Scopus (60) Google Scholar, 15Coin I. Application of non-canonical crosslinking amino acids to study protein-protein interactions in live cells.Curr. Opin. Chem. Biol. 2018; 46: 156-163Crossref PubMed Scopus (35) Google Scholar, 16Yang Y. Song H. Chen P.R. Genetically encoded photocrosslinkers for identifying and mapping protein-protein interactions in living cells.IUBMB Life. 2016; 68: 879-886Crossref PubMed Scopus (34) Google Scholar Specifically, site-specific incorporation of photo-crosslinkers in a protein of interest has emerged as an attractive strategy for investigation of protein-protein interactions with temporal control in living cells, with an aim to map protein interaction interfaces, to capture and identify dynamic, weak, and transient interactions.17Chin J.W. Santoro S.W. Martin A.B. King D.S. Wang L. Schultz P.G. Addition of p-azido-L-phenylalanine to the genetic code of Escherichia coli.J. Am. Chem. Soc. 2002; 124: 9026-9027Crossref PubMed Scopus (552) Google Scholar, 18Chin J.W. Martin A.B. King D.S. Wang L. Schultz P.G. Addition of a photocrosslinking amino acid to the genetic code of Escherichia coli.Proc. Natl. Acad. Sci. USA. 2002; 99: 11020-11024Crossref PubMed Scopus (526) Google Scholar, 19Hino N. Okazaki Y. Kobayashi T. Hayashi A. Sakamoto K. Yokoyama S. Protein photo-cross-linking in mammalian cells by site-specific incorporation of a photoreactive amino acid.Nat. Methods. 2005; 2: 201-206Crossref PubMed Scopus (195) Google Scholar, 20Ai H.W. Shen W. Sagi A. Chen P.R. Schultz P.G. Probing protein-protein interactions with a genetically encoded photo-crosslinking amino acid.ChemBioChem. 2011; 12: 1854-1857Crossref PubMed Scopus (90) Google Scholar, 21Zhang M. Lin S. Song X. Liu J. Fu Y. Ge X. Fu X. Chang Z. Chen P.R. A genetically incorporated crosslinker reveals chaperone cooperation in acid resistance.Nat. Chem. Biol. 2011; 7: 671-677Crossref PubMed Scopus (189) Google Scholar, 22Chou C.J. Uprety R. Davis L. Chin J.W. Deiters A. Genetically encoding an aliphatic diazirine for protein photocrosslinking.Chem. Sci. 2011; 2: 480-483Crossref Scopus (73) Google Scholar, 23Hino N. Oyama M. Sato A. Mukai T. Iraha F. Hayashi A. Kozuka-Hata H. Yamamoto T. Yokoyama S. Sakamoto K. Genetic incorporation of a photo-crosslinkable amino acid reveals novel protein complexes with GRB2 in mammalian cells.J. Mol. Biol. 2011; 406: 343-353Crossref PubMed Scopus (43) Google Scholar, 24Coin I. Perrin M.H. Vale W.W. Wang L. Photo-cross-linkers incorporated into G-protein-coupled receptors in mammalian cells: a ligand comparison.Angew. Chem. Int. Ed. 2011; 50: 8077-8081Crossref PubMed Scopus (50) Google Scholar, 25Coin I. Katritch V. Sun T. Xiang Z. Siu F.Y. Beyermann M. Stevens R.C. Wang L. Genetically encoded chemical probes in cells reveal the binding path of urocortin-I to CRF class B GPCR.Cell. 2013; 155: 1258-1269Abstract Full Text Full Text PDF PubMed Scopus (132) Google Scholar, 26Yang Y. Song H. He D. Zhang S. Dai S. Lin S. Meng R. Wang C. Chen P.R. Genetically encoded protein photocrosslinker with a transferable mass spectrometry-identifiable label.Nat. Commun. 2016; 7: 12299Crossref PubMed Scopus (58) Google Scholar, 27He D. Xie X. Yang F. Zhang H. Su H. Ge Y. Song H. Chen P.R. Quantitative and comparative profiling of protease substrates through a genetically encoded multifunctional photocrosslinker.Angew. Chem. Int. Ed. 2017; 56: 14521-14525Crossref PubMed Scopus (35) Google Scholar This strategy holds enormous potential for application to myriad normal and pathological processes, especially, where receptors dynamically signal and traffic in response to ligand-induced activation, followed by light trigger to crosslink downstream protein interactions.19Hino N. Okazaki Y. Kobayashi T. Hayashi A. Sakamoto K. Yokoyama S. Protein photo-cross-linking in mammalian cells by site-specific incorporation of a photoreactive amino acid.Nat. Methods. 2005; 2: 201-206Crossref PubMed Scopus (195) Google Scholar, 25Coin I. Katritch V. Sun T. Xiang Z. Siu F.Y. Beyermann M. Stevens R.C. Wang L. Genetically encoded chemical probes in cells reveal the binding path of urocortin-I to CRF class B GPCR.Cell. 2013; 155: 1258-1269Abstract Full Text Full Text PDF PubMed Scopus (132) Google Scholar The incorporated photo-crosslinkers can generate highly reactive intermediates under ultraviolet (UV) irradiation (Figure 1A).14Nguyen T.A. Cigler M. Lang K. Expanding the genetic code to study protein-protein interactions.Angew. Chem. Int. Ed. 2018; 57: 14350-14361Crossref PubMed Scopus (60) Google Scholar These reactive species insert into proximal C–H and heteroatom–H bonds, enabling stable covalent linkages formation with interacting protein partners. Despite widespread success of these photo-crosslinkers in chemical biology and proteomics area, the non-selective crosslinking of any nearby residue of protein complex increases background crosslinking and produces unpredictable crosslinking sites, resulting in a challenge for subsequent tandem MS analysis,3Yu C. Huang L. Cross-linking mass spectrometry: an emerging technology for interactomics and structural biology.Anal. Chem. 2018; 90: 144-165Crossref PubMed Scopus (173) Google Scholar, 4Sinz A. Cross-linking/mass spectrometry for studying protein structures and protein-protein interactions: where are we now and where should we go from here?.Angew. Chem. Int. Ed. 2018; 57: 6390-6396Crossref PubMed Scopus (111) Google Scholar, 19Hino N. Okazaki Y. Kobayashi T. Hayashi A. Sakamoto K. Yokoyama S. Protein photo-cross-linking in mammalian cells by site-specific incorporation of a photoreactive amino acid.Nat. Methods. 2005; 2: 201-206Crossref PubMed Scopus (195) Google Scholar, 28Kleiner P. Heydenreuter W. Stahl M. Korotkov V.S. Sieber S.A. A whole proteome inventory of background photocrosslinker binding.Angew. Chem. Int. Ed. 2017; 56: 1396-1401Crossref PubMed Scopus (69) Google Scholar dramatically making it complicated to map accurate interaction interfaces and leading to false positive interacting partners.28Kleiner P. Heydenreuter W. Stahl M. Korotkov V.S. Sieber S.A. A whole proteome inventory of background photocrosslinker binding.Angew. Chem. Int. Ed. 2017; 56: 1396-1401Crossref PubMed Scopus (69) Google Scholar In addition, the reactive intermediates also can be readily quenched or convert to the less reactive species, resulting in short half-life (nanosecond to microsecond range),14Nguyen T.A. Cigler M. Lang K. Expanding the genetic code to study protein-protein interactions.Angew. Chem. Int. Ed. 2018; 57: 14350-14361Crossref PubMed Scopus (60) Google Scholar which will lead to lower crosslinking efficiency and increase the incidence of nonspecific crosslinking of interacting proteins. Clearly, residue-selective photo-crosslinkers are highly in demand, which could confer greater decoding precision of protein-protein interactions and facilitate structural studies of protein complexes.26Yang Y. Song H. He D. Zhang S. Dai S. Lin S. Meng R. Wang C. Chen P.R. Genetically encoded protein photocrosslinker with a transferable mass spectrometry-identifiable label.Nat. Commun. 2016; 7: 12299Crossref PubMed Scopus (58) Google Scholar, 29Yang B. Wu H. Schnier P.D. Liu Y. Liu J. Wang N. DeGrado W.F. Wang L. Proximity-enhanced SuFEx chemical cross-linker for specific and multitargeting cross-linking mass spectrometry.Proc. Natl. Acad. Sci. USA. 2018; 115: 11162-11167Crossref PubMed Scopus (42) Google Scholar Recently, tetrazole derived unnatural amino acid (Uaa) was genetically encoded as an excellent photo-crosslinker (Figure 1B) (mPyTK) to achieve site-selective protein photo-crosslinking in living cells.30Tian Y. Jacinto M.P. Zeng Y. Yu Z. Qu J. Liu W.R. Lin Q. Genetically encoded 2-aryl-5-carboxytetrazoles for site-selective protein photo-cross-linking.J. Am. Chem. Soc. 2017; 139: 6078-6081Crossref PubMed Scopus (47) Google Scholar, 31Tian Y.L. Lin Q. Genetic encoding of 2-aryl-5-carboxytetrazole-based protein photo-cross-linkers.Chem. Commun. (Camb.). 2018; 54: 4449-4452Crossref PubMed Google Scholar The tetrazoles are previously designed for photoclick reaction with alkenes,32Lim R.K. Lin Q. Photoinducible bioorthogonal chemistry: a spatiotemporally controllable tool to visualize and perturb proteins in live cells.Acc. Chem. Res. 2011; 44: 828-839Crossref PubMed Scopus (181) Google Scholar while the nitrile imine intermediates photogenerated from tetrazoles could react with several nucleophilic groups (e.g., thiol groups,33Feng W. Li L. Yang C. Welle A. Trapp O. Levkin P.A. UV-induced tetrazole-thiol reaction for polymer conjugation and surface functionalization.Angew. Chem. Int. Ed. 2015; 54: 8732-8735Crossref PubMed Scopus (51) Google Scholar, 34An P. Lewandowski T.M. Erbay T.G. Liu P. Lin Q. Sterically shielded, stabilized nitrile imine for rapid bioorthogonal protein labeling in live cells.J. Am. Chem. Soc. 2018; 140: 4860-4868Crossref PubMed Scopus (69) Google Scholar carboxylic acids,35Li Z. Qian L. Li L. Bernhammer J.C. Huynh H.V. Lee J.S. Yao S.Q. Tetrazole photoclick chemistry: reinvestigating its suitability as a bioorthogonal reaction and potential applications.Angew. Chem. Int. Ed. 2016; 55: 2002-2006Crossref PubMed Scopus (132) Google Scholar, 36Zhao S. Dai J. Hu M. Liu C. Meng R. Liu X. Wang C. Luo T. Photo-induced coupling reactions of tetrazoles with carboxylic acids in aqueous solution: application in protein labelling.Chem. Commun. (Camb.). 2016; 52: 4702-4705Crossref PubMed Google Scholar tryptophan,37Siti W. Khan A.K. de Hoog H.P. Liedberg B. Nallani M. Photo-induced conjugation of tetrazoles to modified and native proteins.Org. Biomol. Chem. 2015; 13: 3202-3206Crossref PubMed Google Scholar amine,35Li Z. Qian L. Li L. Bernhammer J.C. Huynh H.V. Lee J.S. Yao S.Q. Tetrazole photoclick chemistry: reinvestigating its suitability as a bioorthogonal reaction and potential applications.Angew. Chem. Int. Ed. 2016; 55: 2002-2006Crossref PubMed Scopus (132) Google Scholar and histidine38Zhang Y. Liu W. Zhao Z.K. Nucleophilic trapping nitrilimine generated by photolysis of diaryltetrazole in aqueous phase.Molecules. 2013; 19: 306-315Crossref PubMed Scopus (28) Google Scholar ) to give ligation products. Indeed, it is a creative strategy to develop Uaa from tetrazole to capture transient protein-protein interactions in living cells, however, photogenerated intermediate potentially reacts with above mentioned nucleophiles in addition to carboxylic acid residues, providing linkages with unpredictable selectivity in complex biological environments (Figure 1B).34An P. Lewandowski T.M. Erbay T.G. Liu P. Lin Q. Sterically shielded, stabilized nitrile imine for rapid bioorthogonal protein labeling in live cells.J. Am. Chem. Soc. 2018; 140: 4860-4868Crossref PubMed Scopus (69) Google Scholar, 35Li Z. Qian L. Li L. Bernhammer J.C. Huynh H.V. Lee J.S. Yao S.Q. Tetrazole photoclick chemistry: reinvestigating its suitability as a bioorthogonal reaction and potential applications.Angew. Chem. Int. Ed. 2016; 55: 2002-2006Crossref PubMed Scopus (132) Google Scholar Therefore, methods to access photo-crosslinking capable of temporal resolution39Yang B. Tang S. Ma C. Li S.T. Shao G.C. Dang B. DeGrado W.F. Dong M.Q. Wang P.G. Ding S. et al.Spontaneous and specific chemical cross-linking in live cells to capture and identify protein interactions.Nat. Commun. 2017; 8: 2240Crossref PubMed Scopus (55) Google Scholar, 40Xiang Z. Ren H. Hu Y.S. Coin I. Wei J. Cang H. Wang L. Adding an unnatural covalent bond to proteins through proximity-enhanced bioreactivity.Nat. Methods. 2013; 10: 885-888Crossref PubMed Scopus (100) Google Scholar, 41Chen X.H. Xiang Z. Hu Y.S. Lacey V.K. Cang H. Wang L. Genetically encoding an electrophilic amino acid for protein stapling and covalent binding to native receptors.ACS Chem. Biol. 2014; 9: 1956-1961Crossref PubMed Scopus (68) Google Scholar and residue selectivity for capturing and identifying transient and weak protein-protein interactions in living cells still remain highly challenging and desirable. Herein, we have developed a genetically encoded, lysine-selective protein photo-crosslinker, which is capable of covalently crosslinking interacting protein partners with proximal lysine upon UV light activation in vitro and in living cells (Figure 1C). Using this photo-crosslinker (o-nitrobenzyl alcohol derived lysine, o-NBAK), we demonstrated a crosslinking approach enabling capture of protein-protein interactions in a temporally controlled manner with residue selectivity, producing lysine-selective crosslinking sites, and predictable crosslinked peptides. In addition, this photo-crosslinker enables capture of enzyme-substrate interaction with directly interacting lysine and validation of acetylation site of the substrate. This method represents specificity, accuracy, and reliability for investigation of protein-protein interactions in vitro as well as in living E.coli cells. o-Nitrobenzyl alcohol (o-NBA) derivatives generate corresponding aryl-nitroso compounds via photoisomerization, upon UV light activation (Figure 2A).42Klán P. Šolomek T. Bochet C.G. Blanc A. Givens R. Rubina M. Popik V. Kostikov A. Wirz J. Photoremovable protecting groups in chemistry and biology: reaction mechanisms and efficacy.Chem. Rev. 2013; 113: 119-191Crossref PubMed Scopus (1138) Google Scholar, 43Zhao H. Sterner E.S. Coughlin E.B. Theato P. o-nitrobenzyl alcohol derivatives: opportunities in polymer and materials science.Macromolecules. 2012; 45: 1723-1736Crossref Scopus (426) Google Scholar These intermediates have been successfully employed for condensation with amines to form stable indazolones (Figure 2B).44Kurth M.J. Olmstead M.M. Haddadin M.J. Claimed 2,1-benzisoxazoles are indazalones.J. Org. Chem. 2005; 70: 1060-1062Crossref PubMed Scopus (38) Google Scholar Inspired by these examples as well as the mechanism of photochemical reaction of o-NBA derivatives,42Klán P. Šolomek T. Bochet C.G. Blanc A. Givens R. Rubina M. Popik V. Kostikov A. Wirz J. Photoremovable protecting groups in chemistry and biology: reaction mechanisms and efficacy.Chem. Rev. 2013; 113: 119-191Crossref PubMed Scopus (1138) Google Scholar, 43Zhao H. Sterner E.S. Coughlin E.B. Theato P. o-nitrobenzyl alcohol derivatives: opportunities in polymer and materials science.Macromolecules. 2012; 45: 1723-1736Crossref Scopus (426) Google Scholar we envisioned that the reactive intermediate photogenerated from o-NBAK would smoothly react with proximal lysine in buffer, providing stable indazolone product upon UV light activation (Figure 2C), thus, the genetically encoded o-NBAK could be applied as a novel lysine-selective photo-crosslinker for capturing and identifying protein-protein interactions in living cells (Figures 1C and 2C). To evaluate this hypothesis, we began with o-NBAK (Figure 1C; Supplemental Experimental Procedures) to investigate the potential cross-reactivity with a variety of biological functional groups under UV light activation in aqueous media (Figures S1–S4; Table S1). To our delight, we found that o-NBAK reacted rapidly with lysine after 10 min of UV light activation, providing stable indazolone as ligation product with high yield (>70%, detected by HPLC), within 1 h incubation at 37°C, in buffer conditions (Figures S1 and S2). In contrast, no detectable stable ligation products were found from the reactions of aryl-nitroso compound and other natural amino acids with different sidechains (Table S1; Figure S3).45Lo Conte M.L. Carroll K.S. Chemoselective ligation of sulfinic acids with aryl-nitroso compounds.Angew. Chem. Int. Ed. 2012; 51: 6502-6505Crossref PubMed Scopus (53) Google Scholar The photogenerated aryl-nitroso intermediate could be detected on UPLC-MS analysis process (Figure S2) and half-life is more than 30 min in buffer conditions, which is more stable than other photogenerated reactive species from Uaas.14Nguyen T.A. Cigler M. Lang K. Expanding the genetic code to study protein-protein interactions.Angew. Chem. Int. Ed. 2018; 57: 14350-14361Crossref PubMed Scopus (60) Google Scholar, 34An P. Lewandowski T.M. Erbay T.G. Liu P. Lin Q. Sterically shielded, stabilized nitrile imine for rapid bioorthogonal protein labeling in live cells.J. Am. Chem. Soc. 2018; 140: 4860-4868Crossref PubMed Scopus (69) Google Scholar These results indicated that o-NBAK efficiently and selectively reacted with lysine upon UV light activation, providing ligation product, which could serve as stable covalent linkage for lysine-selective photo-crosslinking. The genetic incorporation of Uaas with novel structures at specific sites of proteins offers a powerful tool to explore and manipulate protein structure and function both in vitro and in vivo.46Liu C.C. Schultz P.G. Adding new chemistries to the genetic code.Annu. Rev. Biochem. 2010; 79: 413-444Crossref PubMed Scopus (1278) Google Scholar Recently, pyrrolysyl-tRNA synthetase (PylRS) and its mutated variants have shown considerable versatility for incorporating a large number of lysine derivatives into proteins in living cells.47Wan W. Tharp J.M. Liu W.R. Pyrrolysyl-tRNA synthetase: an ordinary enzyme but an outstanding genetic code expansion tool.Biochim. Biophys. Acta. 2014; 1844: 1059-1070Crossref PubMed Scopus (255) Google Scholar In this context, we focused on tRNApylCUA/MmPylRS pair for genetic incorporation of o-NBAK as photo-crosslinker into proteins. To site-specifically incorporate the o-NBAK into proteins, the PylRS mutant should efficiently charge o-NBAK. Based on the crystal structure of Methanosarcina mazei (Mm) PylRS in complex with Pyl-AMP,48Kavran J.M. Gundllapalli S. O'Donoghue P. Englert M. Söll D. Steitz T.A. Structure of pyrrolysyl-tRNA synthetase, an archaeal enzyme for genetic code innovation.Proc. Natl. Acad. Sci. USA. 2007; 104: 11268-11273Crossref PubMed Scopus (156) Google Scholar docking models (Figure 3A) suggested that the active site of PylRS mutant (Y306A/Y384F) is capable of accommodating o-NBAK, implying that the orthogonal tRNApylCUA/MmPylRS (Y306A/Y384F) is able to recognize o-NBAK.49Yanagisawa T. Hino N. Iraha F. Mukai T. Sakamoto K. Yokoyama S. Wide-range protein photo-crosslinking achieved by a genetically encoded N(epsilon)-(benzyloxycarbonyl)lysine derivative with a diazirinyl moiety.Mol. Biosyst. 2012; 8: 1131-1135Crossref PubMed Scopus (40) Google Scholar Therefore, we examined the incorporation efficiency and specificity of o-NBAK into Schistosoma japonicum glutathione-S-transferase (GST) protein in E. coli. Guided by structural analysis of GST dimer (PDB ID: 1Y6E), we introduced a TAG codon at position 97 and a C-terminal His tag into GST sequence (GST-97TAG). A plasmid of GST mutant co-expressed with another plasmid of tRNApylCUA/MmPylRS (Y306A/Y384F) in E. coli. In absence of o-NBAK, no full-length GST protein was detected (Figure 3B), when 1 mM o-NBAK was added in growth media, full-length GST was obtained in good yield (ca. 4.0 mg L−1, Figure S14). The purified GST-97o-NBAK was analyzed by electrospray ionization time-of-flight MS (ESI-TOF MS) (Figure 3C). The masses indicate that only o-NBAK was incorporated in GST and no peaks corresponding to other amino acids were observed. Furthermore, GST-97o-NBAK protein was digested with trypsin and analyzed by tandem MS (Figure 3D), a series of y and b ions clearly indicate that o-NBAK was incorporated at position 97, specified by the TAG codon. These results demonstrate that mutated MmPylRS (Y306A/Y384F) can efficiently recognize o-NBAK (Figure S14) and only o-NBAK was incorporated at TAG-encoded position in GST, and that o-NBAK was stable during protein synthesis in E. coli and protein purification process. As proof of principle, we chose to crosslink GST homodimer as o-NBAK enabling photo-crosslinking with proximal lysine inter-molecularly (Figure 4A). Based on GST dimer crystal structure, A97 located at a distance of ∼15 Å from a lysine residue (Lys44) from the opposite monomer, which indicated that o-NBAK at position 97 would be able to generate covalent linkage with Lys44 between GST dimer.29Yang B. Wu H. Schnier P.D. Liu Y. Liu J. Wang N. DeGrado W.F. Wang L. Proximity-enhanced SuFEx chemical cross-linker for specific and multitargeting cross-linking mass spectrometry.Proc. Natl. Acad. Sci. USA. 2018; 115: 11162-11167Crossref PubMed Scopus (42) Google Scholar, 30Tian Y. Jacinto M.P. Zeng Y. Yu Z. Qu J. Liu W.R. Lin Q. Genetically encoded 2-aryl-5-carboxytetrazoles for site-selective protein photo-

Full Text
Published version (Free)

Talk to us

Join us for a 30 min session where you can share your feedback and ask us any queries you have

Schedule a call