Design of a Split Intein with Exceptional Protein Splicing Activity
Abstract

Protein trans-splicing (PTS) by split inteins has found widespread use in chemical biology and biotechnology. Herein, we describe the use of a consensus design approach to engineer a split intein with enhanced stability and activity that make it more robust than any known PTS system. Using batch mutagenesis, we first conduct a detailed analysis of the difference in splicing rates between the Npu (fast) and Ssp (slow) split inteins of the DnaE family and find that most impactful residues lie on the second shell of the protein, directly adjacent to the active site. These residues are then used to generate an alignment of 73 naturally occurring DnaE inteins that are predicted to be fast. The consensus sequence from this alignment (Cfa) demonstrates both rapid protein splicing and unprecedented thermal and chaotropic stability. Moreover, when fused to various proteins including antibody heavy chains, the N-terminal fragment of Cfa exhibits increased expression levels relative to other N-intein fusions. The durability and efficiency of Cfa should improve current intein based technologies and may provide a platform for the development of new protein chemistry techniques.
Protein splicing is a post-translational autoprocessing event in which an intervening protein domain called an intein excises itself from a host protein in a traceless manner such that the flanking polypeptide sequences (exteins) are ligated together via a normal peptide bond (Figure S1A). (1) While protein splicing typically occurs spontaneously following translation of a contiguous polypeptide, some inteins exist naturally in a split form. (1) The two pieces of the split intein are separately expressed and remain inactive until encountering their complementary partner, upon which they cooperatively fold and undergo splicing in trans (Figure S1B). This activity has been harnessed in a host of protein engineering methods that provide control over the structure and activity of proteins both in vitro and in vivo. (1) The first two split inteins to be characterized, from the cyanobacteria Synechocystis species PCC6803 (Ssp) and Nostoc punctiforme PCC73102 (Npu), are orthologs naturally found inserted in the α subunit of DNA Polymerase III (DnaE). (2-4) Npu is especially notable due its remarkably fast rate of protein trans-splicing (PTS) (t1/2 = 50 s at 30 °C). (5) This half-life is significantly shorter than that of Ssp (t1/2 = 80 min at 30 °C), (5) an attribute that has expanded the range of applications open to PTS. (1)
Despite the ongoing discovery of new fast inteins, (6, 7) little is known about what separates them from their slower homologues. Such an understanding should help identify new inteins that are likely to splice rapidly and potentially allow for the engineering of split inteins with superior PTS properties. We sought to investigate the basis of rapid protein splicing through a comparative study of Npu and Ssp. The substantial difference in splicing rate between these two proteins is especially puzzling given their highly similar sequences (63% identity) and near-superimposable active site structures (Figure S2). Previous mutagenesis studies on Npu and Ssp suggest that the difference in activity between the two is likely due to the combined effects of several residues, rather than a single site. (6, 8) However, it remains unclear just how many residues are responsible for the fast versus slow reaction rates and, by extension, whether these “accelerator” residues contribute equally to the individual chemical steps in the overall protein splicing process. Consequently, we began our study by exploring these questions, in the hope that this would provide a starting point for developing an improved PTS system.
The high level of conservation within the active sites of Npu and Ssp suggests that differences in distal amino acids account for the disparity in splicing rate between the two. Thus, we focused our attention on “second shell” residues, those directly adjacent to the active site. To simplify this analysis, we employed a batch mutagenesis strategy in conjunction with a previously reported in vitro PTS assay. (5) This assay uses split intein constructs with short native extein sequences and allows the rates of branched intermediate formation (k1,k2) and its resolution to final splice products (k3) to be determined using a three-state kinetic model (Figure S3).
The known cross-reactivity of Npu and Ssp intein fragments served as a convenient platform to assess which half of the split intein contributes most significantly to the difference in activity. (3) Both the SspN-NpuC (chimera 1) and NpuN-SspC (chimera 2) chimeras show a decrease in the rates of branch formation and resolution compared to that of native Npu (Figure S4C, S4D). This indicates that residues on both the N- and C-intein fragments of Npu and Ssp contribute to the difference in their splicing rate. Next, four groups of second shell positions on each of these chimeras were chosen based on their proximity to key catalytic residues, and the corresponding Ssp residues were mutated to those in Npu (Figure S4A, S4B). From the chimera 1 mutants, Batch 2 (L56F, S70K, A83P, E85D) completely restored branch formation activity to that of native Npu (Figure S4C), while Batch 1 (R73K, L75M, Y79G, L81M) restored the majority of branch resolution activity (Figure S4D). The effects of mutations on the chimera 2 background were more prosaic, with no single batch able to restore splicing activity to that of native Npu (Figures S4C, S4D). Lastly, the A136S mutation on SspC has previously been shown to accelerate protein splicing and was examined separately. (8) This A136S mutation increases the rate of branch resolution 2-fold, but has no impact on branch formation (Figures S4C, S4D).
Next we sought to better understand the individual contributions of residues within batch mutants 1 and 2, since these had the most profound effect on splicing activity. For Batch 2, further mutagenesis shows that the interaction between F56, K70, and D85 is likely responsible for the increased rate of branch formation in NpuN (Figure S5A). Structural evidence supports these data, as K70 is a part of the highly conserved TXXH block B loop in NpuN (residues 69–72) that catalyzes the initial N-to-S acyl shift in protein splicing. (9) Thus, the position and dynamics of K70 (packed against F56 and D85) should directly impact the catalytic residues T69 and H72 (Figure S5B). (10-12) From Batch 1, K73, M75, and M81 are responsible for the faster rate of branch resolution in NpuN (Figure S6A). These residues pack against the terminal asparagine of the C-intein, which must undergo succinimide formation in the final step of protein splicing (Figure S6B). Taken together, the mutagenesis data point to the key role that second shell “accelerator” residues play in tuning the activity of split inteins.
The “accelerator” residues found to affect the splicing rate allow for an activity-guided approach to engineer a consensus DnaE intein. Consensus protein engineering is a tool applied to a homologous set of proteins in order to create a thermostable variant derived from the parent family. (13, 14) A multiple sequence alignment (MSA) is first generated from homologues of a particular protein, from which the most frequent residue at each position is chosen as the representative in the consensus sequence. For the DnaE inteins, 105 sequences were identified through a BLAST (15) search of the JGI (16) and NCBI (17) databases (Figure S7A). Next, the alignment was filtered to only contain sequences bearing the second shell indicators of fast splicing: K70, M75, M81, and S136. The 73 theoretically fast inteins left in the MSA (Figure S7B) were then used to generate a consensus fast DnaE intein sequence (Cfa) (Figure 1). The Cfa intein has high sequence similarity to Npu (82%), and the nonidentical residues are spread throughout the 3D structure of the protein.
Figure 1

Figure 1. Design of the Cfa split intein. (a) Sequence alignment of Npu DnaE and Cfa DnaE. The sequences share 82% identity with the differences (cyan) evenly distributed through the primary sequence. Catalytic residues and second shell “accelerator” residues are shown in orange and green, respectively. (b) The same residues highlighted in panel a mapped on to the Npu structure (PDB ID 4Kl5).
We generated the Cfa intein fragments fused to model exteins and measured their PTS activity using the aforementioned in vitro assay (Figure 2). This revealed that the Cfa intein splices 2.5-fold faster at 30 °C than Npu (t1/2 20 s vs 50 s), a notable enhancement in activity since the latter is the fastest characterized DnaE split intein (Figure 2A). Cfa demonstrates faster equilibrium rates of branch formation (3-fold) and rate branch resolution (2-fold) (Figure S8). We note that, in line with parent DnaE inteins, Cfa retains the preference for a bulky hydrophobic residue at the +2 position of the C-extein (Figure S9). Strikingly, Cfa shows an increased splicing rate as a function of temperature and is consistently faster than Npu (Figure 2A). The Cfa intein even maintains activity at 80 °C, albeit with reduced yield of splice products, while Npu is inactive at this temperature. These results demonstrate that consensus engineering is effective in producing an intein that is highly active across a broad range of temperatures.
Figure 2

Figure 2. Characterization of the Cfa intein. (a) Splicing rates for Cfa and Npu as a function of temperature. Npu is inactive at 80 °C. Error = SD (n = 3). (b, c) Splicing rates for Cfa and Npu as a function of added chaotrope. Npu is inactive in 3 M GuHCl or 8 M Urea. Note: Cfa has residual activity in 4 M GuHCl (k = 7 × 10–5). Error = SD (n = 3).
Applications of PTS typically require fission of a target protein and fusion of the resulting fragments to the appropriate split intein segments. (1) As a consequence, the solubility of these fusion proteins can sometimes be poor. Because protein denaturants such as guanidine hydrochloride (GuHCl) and urea are frequently used to keep these less soluble fragments in solution, we tested the ability of Cfa to splice in the presence of these chaotropic agents. We found that the Cfa intein can splice in the presence of up to 4 M GuHCl (with little decrease in activity seen up to 3 M), while no activity was observed for Npu in ≥3 M GuHCl (Figure 2B). Remarkably, the splicing of Cfa is largely unaffected up to 8 M urea, while splicing of Npu falls off dramatically above 4 M urea (Figure 2C).
The unprecedented tolerance of Cfa to high concentrations of GuHCl and urea suggests the intein might retain activity directly following chaotropic extraction of insoluble proteins from bacterial inclusion bodies, thereby expediting PTS-based studies. Accordingly, we overexpressed the model fusion protein, His6-Sumo-CfaN, in E. coli cells and extracted the protein from inclusion bodies with 6 M urea. The protein was purified from this extract by nickel affinity chromatography and then directly, and efficiently, modified by PTS under denaturing conditions, i.e. without the need for any intervening refolding steps (Figure S10). In general, we expect that the robust activity of Cfa in the presence of chaotropic agents will prove useful when working with protein fragments that demonstrate poor solubility under native conditions.
Figure 3

Figure 3. Expression and modification of a mouse monoclonal antibody using the Cfa intein. (a) Test expression in HEK293T cells of various IntN homologues (Npu, Mcht, Ava, and Cfa) fused to the C-terminus of the heavy chain of a mouse αDec205 monoclonal antibody. Top: Western blot analysis (αMouse IgG) of antibody levels present in the medium following the 96 h expression. Bottom: α-actin Western blot of cell lysate as a loading control. (b) Quantification of normalized expression yield by densitometry of αDEC205 HC-IntN signal in panel a. Error = SD (n = 4). (c) Structure of the CfaC-dendrimer construct used in PTS reactions with the αDEC205 HC-IntN fusion. For simplicity the CfaC peptide sequence and a short peptide linker are depicted symbolically in teal. (d) Schematic of the in situ PTS approach used to modify the HC of an mAb with a multivalent cargo. (e) SDS-PAGE analysis of PTS reaction. Lane 1: Wild type mouse αDEC205 mAB. Lane 2: Mouse αDEC205-CfaN mAB fusion. Lane 3: addition of the CfaC-dendrimer to the media containing the αDEC205-CfaN mAB. The splicing reaction was analyzed after two hours by fluorescence (bottom) and Western blot (top, αMouse IgG).
Fusing a protein of interest to a split intein can result in a marked reduction in cellular expression levels compared to the protein alone. (6) This situation is more frequently encountered for fusions to N-inteins than to C-inteins, which is likely due to the larger size of the former and their partially folded state. (18) We therefore wondered whether the improved thermal and chaotropic stability of Cfa would translate to increased expression levels of CfaN fusions. Indeed, model studies in E. coli revealed a significant (30-fold) increase in soluble protein expression for a CfaN fusion compared to the corresponding NpuN fusion (Figure S11). Given this result, we wondered whether CfaN fusions would also exhibit increased protein expression levels in mammalian cells. In particular, intein fusions to the heavy chain (HC) of monoclonal antibodies (mAbs) have emerged as a powerful tool for site-specific conjugation of synthetic cargoes. (19, 20) We compared the expression levels in HEK293 cells of an mAb (αDec205) as a function of the N-intein fused to its HC. Gratifyingly, and consistent with the bacterial expression results, production of the HC-CfaN fusion was significantly higher than that for the other inteins examined; for example, the secreted levels of the mAb-Cfa construct were ∼10-fold higher than those for the corresponding Npu fusion (Figure 3A,B). Importantly, mAb-Cfa retained PTS activity and could be site-specifically modified with a synthetic peptide by splicing directly in the growth medium following the four-day expression at 37 °C (Figure S12).
Finally, to further explore the utility of the Cfa intein in the context of antibody conjugation, we asked whether the PTS system could be used to attach multiple copies of a synthetic cargo to the heavy chain of the mAb. Accordingly, we used semisynthesis to prepare a construct in which the C-terminal half of Cfa (CfaC) was fused to a C-extein containing a dendrimeric scaffold allowing multimeric attachment of cargo, in this case fluorescein (Figure 3C). This dendritic cargo was successfully linked to the αDec205 antibody via Cfa-mediated PTS, again performed directly in situ within the cellular growth medium (Figure 3D, E). To our knowledge, this represents the first time that PTS has been used to attach a branched extein construct to a target protein, highlighting the potential of the system for manipulating the payload quantity of antibody drug conjugates. (21)
The discovery of fast split inteins has revolutionized the applications of protein trans splicing. The remarkable robustness of the Cfa intein described in this study should extend the utility of many of these technologies by allowing PTS to be performed in a broader range of reaction conditions. Moreover, the ability of Cfa to increase the expression yields of N-intein fusions should encourage further use of split inteins for protein semisynthesis. The activity-guided approach we use to engineer this intein may be applied to other intein families or act as a general strategy for the refinement of multiple sequence alignments used for consensus engineering.
Supporting Information
The Supporting Information is available free of charge on the ACS Publications website at DOI: 10.1021/jacs.5b13528.
Full methods and experimental data (PDF)
Terms & Conditions
Most electronic Supporting Information files are available without a subscription to ACS Web Editions. Such files may be downloaded by article for research use (if there is a public use license linked to the relevant article, that license may permit other uses). Permission may be obtained from ACS for other uses through requests via the RightsLink permission system: http://pubs.acs.org/page/copyright/permissions.html.
Acknowledgment
The authors thank the members of the Muir laboratory for valuable discussions. This work was supported by the U.S. National Institutes of Health (R37-GM086868) and National Science Foundation Graduate Research Fellowship under Grant No. (DGE-1148900).
References
This article references 21 other publications.
- 1Shah, N. H.; Muir, T. W. Chem. Sci. 2014, 5, 446 DOI: 10.1039/C3SC52951G[Crossref], [PubMed], [CAS], Google Scholar1https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3sXitVSqtbzJ&md5=e91aafcf49109d059dbed41e363c6811Inteins: nature's gift to protein chemistsShah, Neel H.; Muir, Tom W.Chemical Science (2014), 5 (2), 446-461CODEN: CSHCCN; ISSN:2041-6520. (Royal Society of Chemistry)A review. Inteins are auto-processing domains found in organisms from all domains of life. These proteins carry out a process known as protein splicing, which is a multi-step biochem. reaction comprised of both the cleavage and formation of peptide bonds. While the endogenous substrates of protein splicing are specific essential proteins found in intein-contg. host organisms, inteins are also functional in exogenous contexts and can be used to chem. manipulate virtually any polypeptide backbone. Given this, protein chemists have exploited various facets of intein reactivity to modify proteins in myriad ways for both basic biol. research as well as potential therapeutic applications. Here, we review the intein field, first focusing on the biol. context and phylogenetic diversity of inteins, followed by a description of intein structure and biochem. function. Finally, we discuss prevalent intein-based technologies, focusing on their applications in chem. biol., followed by persistent caveats of intein chem. and approaches to alleviate these shortcomings. The findings summarized herein describe two and a half decades of research, leading from a biochem. curiosity to the development of powerful protein engineering tools.
- 2Wu, H.; Hu, Z.; Liu, X. Q. Proc. Natl. Acad. Sci. U. S. A. 1998, 95, 9226 DOI: 10.1073/pnas.95.16.9226
- 3Iwai, H.; Zuger, S.; Jin, J.; Tam, P. H. FEBS Lett. 2006, 580, 1853 DOI: 10.1016/j.febslet.2006.02.045
- 4Zettler, J.; Schutz, V.; Mootz, H. D. FEBS Lett. 2009, 583, 909 DOI: 10.1016/j.febslet.2009.02.003
- 5Shah, N. H.; Eryilmaz, E.; Cowburn, D.; Muir, T. W. J. Am. Chem. Soc. 2013, 135, 5839 DOI: 10.1021/ja401015p
- 6Shah, N. H.; Dann, G. P.; Vila-Perello, M.; Liu, Z.; Muir, T. W. J. Am. Chem. Soc. 2012, 134, 11338 DOI: 10.1021/ja303226x
- 7Carvajal-Vallejos, P.; Pallisse, R.; Mootz, H. D.; Schmidt, S. R. J. Biol. Chem. 2012, 287, 28686 DOI: 10.1074/jbc.M112.372680
- 8Wu, Q.; Gao, Z.; Wei, Y.; Ma, G.; Zheng, Y.; Dong, Y.; Liu, Y. Biochem. J. 2014, 461, 247 DOI: 10.1042/BJ20140287
- 9Aranko, A. S.; Oeemig, J. S.; Kajander, T.; Iwai, H. Nat. Chem. Biol. 2013, 9, 616 DOI: 10.1038/nchembio.1320[Crossref], [PubMed], [CAS], Google Scholar9https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3sXhtlWkurvE&md5=5cd3a3273984caeeba5201c7f3f6db50Intermolecular domain swapping induces intein-mediated protein alternative splicingAranko, A. Sesilja; Oeemig, Jesper S.; Kajander, Tommi; Iwai, HideoNature Chemical Biology (2013), 9 (10), 616-622CODEN: NCBABT; ISSN:1552-4450. (Nature Publishing Group)Protein sequences are diversified on the DNA level by recombination and mutation and can be further increased on the RNA level by alternative RNA splicing, involving introns that have important roles in many biol. processes. The protein version of introns (inteins), which catalyze protein splicing, were first reported in the 1990s. The biol. roles of protein splicing still remain elusive because inteins neither provide any clear benefits nor have an essential role in their host organisms. We now report protein alternative splicing, in which new protein sequences can be produced by protein recombination by intermol. domain swapping of inteins, as elucidated by NMR spectroscopy and crystal structures. We demonstrate that intein-mediated protein alternative splicing could be a new strategy to increase protein diversity (i.e., functions) without any modification in genetic backgrounds. We also exploited it as a post-translational protein conformation-driven switch of protein functions (for example, as highly specific protein interference).
- 10Pietrokovski, S. Protein Sci. 1994, 3, 2340 DOI: 10.1002/pro.5560031218[Crossref], [PubMed], [CAS], Google Scholar10https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADyaK2MXjslajur8%253D&md5=6c5a56795954e6fb607902b5674f2426Conserved sequence features of inteins (protein introns) and their use in identifying new inteins and related proteinsPietrokovski, ShmuelProtein Science (1994), 3 (12), 2340-50CODEN: PRCIEI; ISSN:0961-8368. (Cambridge University Press)Inteins (protein introns) are internal portions of protein sequences that are post-translationally excised while the flanking regions are spliced together, making an addnl. protein product. Inteins have been found in a no. of homologous genes in yeast, mycobacteria, and extreme thermophile archaebacteria. The inteins are probably multifunctional, auto-catalyzing their own splicing, and some were also shown to be DNA endonucleases. The splice junction regions and two regions similar to homing endonucleases were thought to be the only common sequence features of inteins. This work analyzed all published intein sequences with recently developed methods for detecting weak, conserved sequence features. The methods complemented each other in the identification and assessment of several patterns characterizing the intein sequences. New intein conserved features are discovered and the known ones are quant. described and localized. The general sequence description of all the known inteins is derived from the motifs and their relative positions. The intein sequence description is used to search the sequence data-bases for intein-like proteins. A sequence region in a mycobacterial open reading frame possessing all of the intein motifs and absent from sequences homologous to both of its flanking sequences id identified as an intein. A newly discovered putative intein in red algae chloroplasts is found not to contain the endonuclease motifs present in all other inteins. The yeast HO endonuclease is found to have an overall intein-like structure and a few viral polyprotein cleavage sites are found to be significantly similar to the inteins amino-end splice junction motif. The intein features described may serve for detection of intein sequences.
- 11Dearden, A. K.; Callahan, B.; Roey, P. V.; Li, Z.; Kumar, U.; Belfort, M.; Nayak, S. K. Protein Sci. 2013, 22, 557 DOI: 10.1002/pro.2236[Crossref], [PubMed], [CAS], Google Scholar11https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3sXmsFWqtr8%253D&md5=300b3e042ff7c8609b9815ad61443523A conserved threonine spring-loads precursor for intein splicingDearden, Albert K.; Callahan, Brian; Van Roey, Patrick; Li, Zhong; Kumar, Utsav; Belfort, Marlene; Nayak, Saroj K.Protein Science (2013), 22 (5), 557-563CODEN: PRCIEI; ISSN:1469-896X. (Wiley-Blackwell)Protein splicing is an autocatalytic process where an "intein" self-cleaves from a precursor and ligates the flanking N- and C-"extein" polypeptides. Inteins occur in all domains of life and have myriad uses in biotechnol. Although the reaction steps of protein splicing are known, mechanistic details remain incomplete, particularly the initial peptide rearrangement at the N-terminal extein/intein junction. Recently, we proposed that this transformation, an N-S acyl shift, is accelerated by a localized conformational strain, between the intein's catalytic cysteine (Cys1) and the neighboring glycine (Gly-1) in the N-extein. That proposal was based on the crystal structure of a catalytically competent trapped precursor. Here, we define the structural origins and mechanistic relevance of the conformational strain using a combination of quantum mech. simulations, mutational anal., and X-ray crystallog. Our results implicate a conserved, but largely unstudied, threonine residue of the Ssp DnaE intein (Thr69) as the mediator of conformational strain through hydrogen bonding. Further, the strain imposed by this residue is shown to position the splice junction in a manner that enhances the rate of the N-S acyl shift substantially. Taken together, our results not only provide fundamental understanding of the control of the first step of protein splicing but also have important implications in various biotechnol. applications that require precursor manipulation.
- 12Du, Z.; Shemella, P. T.; Liu, Y.; McCallum, S. A.; Pereira, B.; Nayak, S. K.; Belfort, G.; Belfort, M.; Wang, C. J. Am. Chem. Soc. 2009, 131, 11581 DOI: 10.1021/ja904318w
- 13Lehmann, M.; Kostrewa, D.; Wyss, M.; Brugger, R.; D’Arcy, A.; Pasamontes, L.; van Loon, A. P. Protein Eng., Des. Sel. 2000, 13, 49 DOI: 10.1093/protein/13.1.49
- 14Steipe, B. Methods Enzymol. 2004, 388, 176 DOI: 10.1016/S0076-6879(04)88016-9
- 15Altschul, S. F.; Gish, W.; Miller, W.; Myers, E. W.; Lipman, D. J. J. Mol. Biol. 1990, 215, 403 DOI: 10.1016/S0022-2836(05)80360-2[Crossref], [PubMed], [CAS], Google Scholar15https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADyaK3MXitVGmsA%253D%253D&md5=009d2323eb82f0549356880e1101db16Basic local alignment search toolAltschul, Stephen F.; Gish, Warren; Miller, Webb; Myers, Eugene W.; Lipman, David J.Journal of Molecular Biology (1990), 215 (3), 403-10CODEN: JMOBAK; ISSN:0022-2836.A new approach to rapid sequence comparison, basic local alignment search tool (BLAST), directly approximates alignments that optimize a measure of local similarity, the maximal segment pair (MSP) score. Recent math. results on the stochastic properties of MSP scores allow an anal. of the performance of this method as well as the statistical significance of alignments it generates. The basic algorithm is simple and robust; it can be implemented in a no. of ways and applied in a variety of contexts including straightforward DNA and protein sequence database searches, motif searches, gene identification searches, and in the anal. of multiple regions of similarity in long DNA sequences. In addn. to its flexibility and tractability to math. anal., BLAST is an order of magnitude faster than existing sequence comparison tools of comparable sensitivity.
- 16Grigoriev, I. V.; Nordberg, H.; Shabalov, I.; Aerts, A.; Cantor, M.; Goodstein, D.; Kuo, A.; Minovitsky, S.; Nikitin, R.; Ohm, R. A.; Otillar, R.; Poliakov, A.; Ratnere, I.; Riley, R.; Smirnova, T.; Rokhsar, D.; Dubchak, I. Nucleic Acids Res. 2012, 40, D26 DOI: 10.1093/nar/gkr947
- 17Tatusova, T.; Ciufo, S.; Fedorov, B.; O’Neill, K.; Tolstoy, I. Nucleic Acids Res. 2014, 42, D553 DOI: 10.1093/nar/gkt1274
- 18Shah, N. H.; Eryilmaz, E.; Cowburn, D.; Muir, T. W. J. Am. Chem. Soc. 2013, 135, 18673 DOI: 10.1021/ja4104364
- 19Mohlmann, S.; Bringmann, P.; Greven, S.; Harrenga, A. BMC Biotechnol. 2011, 11, 76 DOI: 10.1186/1472-6750-11-76[Crossref], [PubMed], [CAS], Google Scholar19https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BC3Mjlt1yqtQ%253D%253D&md5=0c498501b2777e84d50836f6aaa0b2ecSite-specific modification of ED-B-targeting antibody using intein-fusion technologyMohlmann Sina; Bringmann Peter; Greven Simone; Harrenga AxelBMC biotechnology (2011), 11 (), 76 ISSN:.BACKGROUND: A promising new approach in cancer therapy is the use of tumor specific antibodies coupled to cytotoxic agents. Currently these immunoconjugates are prepared by rather unspecific coupling chemistries, resulting in heterogeneous products. As the drug load is a key parameter for the antitumor activity, site-specific strategies are desired. Expressed protein ligation (EPL) and protein trans-splicing (PTS) are methods for the specific C-terminal modification of a target protein. Both include the expression as an intein fusion protein, followed by the exchange of the intein for a functionalized moiety. RESULTS: A full-length IgG specific for fibronectin ED-B was expressed as fusion protein with an intein (Mxe GyrA or Npu DnaE) attached to each heavy chain. In vitro protocols were established to site-specifically modify the antibodies in high yields by EPL or PTS, respectively. Although reducing conditions had to be employed during the process, the integrity or affinity of the antibody was not affected. The protocols were used to prepare immunoconjugates containing two biotin molecules per antibody, attached to the C-termini of the heavy chains. CONCLUSION: Full-length antibodies can be efficiently and site-specifically modified at the C-termini of their heavy chains by intein-fusion technologies. The described protocols can be used to prepare immunoconjugates of high homogeneity and with a defined drug load of two. The attachment to the C-termini is expected to retain the affinity and effector functions of the antibodies.
- 20Barbuto, S.; Idoyaga, J.; Vila-Perello, M.; Longhi, M. P.; Breton, G.; Steinman, R. M.; Muir, T. W. Nat. Chem. Biol. 2013, 9, 250 DOI: 10.1038/nchembio.1186[Crossref], [PubMed], [CAS], Google Scholar20https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3sXis1aitr0%253D&md5=460c3394e4559169746733ac91b710f5Induction of innate and adaptive immunity by delivery of poly dA:dT to dendritic cellsBarbuto, Scott; Idoyaga, Juliana; Vila-Perello, Miquel; Longhi, Maria P.; Breton, Gaelle; Steinman, Ralph M.; Muir, Tom W.Nature Chemical Biology (2013), 9 (4), 250-256CODEN: NCBABT; ISSN:1552-4450. (Nature Publishing Group)Targeted delivery of antigens to dendritic cells (DCs) is a promising vaccination strategy. However, to ensure immunity, the approach depends on coadministration of an adjuvant. Here we ask whether targeting of both adjuvant and antigen to DCs is sufficient to induce immunity. Using a protein ligation method, we develop a general approach for linking the immune stimulant, poly dA:dT (pdA:dT), to a monoclonal antibody (mAb) specific for DEC205 (DEC). We show that DEC-specific mAbs deliver pdA:dT to DCs for the efficient prodn. of type I interferon in human monocyte-derived DCs and in mice. Notably, adaptive T-cell immunity is elicited when mAbs specific for DEC-pdA:dT are used as the activation stimuli and are administered together with a DC-targeted antigen. Collectively, our studies indicate that DCs can integrate innate and adaptive immunity in vivo and suggest that dual delivery of antigen and adjuvant to DCs might be an efficient approach to vaccine development.
- 21Shah, N. D.; Parekh, H. S.; Steptoe, R. J. Pharm. Res. 2014, 31, 3150 DOI: 10.1007/s11095-014-1408-1
Cited By
This article is cited by 101 publications.
- Karthik Nadendla, Grant G. Simpson, Julie Becher, Toby Journeaux, Mar Cabeza-Cabrerizo, Gonçalo J. L. Bernardes. Strategies for Conditional Regulation of Proteins. JACS Au 2023, 3 (2) , 344-357. https://doi.org/10.1021/jacsau.2c00654
- Giridhar Sekar, Adam J. Stevens, Anahita Z. Mostafavi, Pulikallu Sashi, Tom W. Muir, David Cowburn. A Conserved Histidine Residue Drives Extein Dependence in an Enhanced Atypically Split Intein. Journal of the American Chemical Society 2022, 144 (41) , 19196-19203. https://doi.org/10.1021/jacs.2c08985
- Xiaofan Jia, Yan Zhang, Ting Wang, Yuan Fu. Highly Efficient Method for Intracellular Delivery of Proteins Mediated by Cholera Toxin-Induced Protein Internalization. Molecular Pharmaceutics 2021, 18 (11) , 4067-4078. https://doi.org/10.1021/acs.molpharmaceut.1c00479
- Michelle M. Mitchener, Tom W. Muir. Janus Bioparticles: Asymmetric Nucleosomes and Their Preparation Using Chemical Biology Approaches. Accounts of Chemical Research 2021, 54 (16) , 3215-3227. https://doi.org/10.1021/acs.accounts.1c00313
- Hyunseok Oh, Younghun Jung, Seokoh Moon, Jaehyeon Hwang, Choongjin Ban, Jinhyo Chung, Woo-Jae Chung, Dae-Hyuk Kweon. Development of End-Spliced Dimeric Nanodiscs for the Improved Virucidal Activity of a Nanoperforator. ACS Applied Materials & Interfaces 2021, 13 (31) , 36757-36768. https://doi.org/10.1021/acsami.1c06364
- Tomoharu Motoyama, Nozomi Hiramatsu, Yasuhisa Asano, Shogo Nakano, Sohei Ito. Protein Sequence Selection Method That Enables Full Consensus Design of Artificial l-Threonine 3-Dehydrogenases with Unique Enzymatic Properties. Biochemistry 2020, 59 (40) , 3823-3833. https://doi.org/10.1021/acs.biochem.0c00570
- Robert E. Thompson, Tom W. Muir. Chemoenzymatic Semisynthesis of Proteins. Chemical Reviews 2020, 120 (6) , 3051-3126. https://doi.org/10.1021/acs.chemrev.9b00450
- Christopher Hyde Bowen, Tavis J. Reed, Cameron Jay Sargent, Beryl Mpamo, Jonathan M. Galazka, Fuzhong Zhang. Seeded Chain-Growth Polymerization of Proteins in Living Bacterial Cells. ACS Synthetic Biology 2019, 8 (12) , 2651-2658. https://doi.org/10.1021/acssynbio.9b00362
- Josef A. Gramespacher, Antony J. Burton, Luis F. Guerra, Tom W. Muir. Proximity Induced Splicing Utilizing Caged Split Inteins. Journal of the American Chemical Society 2019, 141 (35) , 13708-13712. https://doi.org/10.1021/jacs.9b05721
- Eugene Kim, Bin Dai, James B. Qiao, Wenlu Li, John D. Fortner, Fuzhong Zhang. Microbially Synthesized Repeats of Mussel Foot Protein Display Enhanced Underwater Adhesion. ACS Applied Materials & Interfaces 2018, 10 (49) , 43003-43012. https://doi.org/10.1021/acsami.8b14890
- Adam J. Stevens, Giridhar Sekar, Josef A. Gramespacher, David Cowburn, Tom W. Muir. An Atypical Mechanism of Split Intein Molecular Recognition and Folding. Journal of the American Chemical Society 2018, 140 (37) , 11791-11799. https://doi.org/10.1021/jacs.8b07334
- Julian C. J. Matern, Kristina Friedel, Jens Binschik, Kira-Sophie Becher, Zahide Yilmaz, Henning D. Mootz. Altered Coordination of Individual Catalytic Steps in Different and Evolved Inteins Reveals Kinetic Plasticity of the Protein Splicing Pathway. Journal of the American Chemical Society 2018, 140 (36) , 11267-11275. https://doi.org/10.1021/jacs.8b04794
- Christopher H. Bowen, Bin Dai, Cameron J. Sargent, Wenqin Bai, Pranay Ladiwala, Huibao Feng, Wenwen Huang, David L. Kaplan, Jonathan M. Galazka, Fuzhong Zhang. Recombinant Spidroins Fully Replicate Primary Mechanical Properties of Natural Spider Silk. Biomacromolecules 2018, 19 (9) , 3853-3860. https://doi.org/10.1021/acs.biomac.8b00980
- Thomas Pirzer, Kira-Sophie Becher, Marcel Rieker, Tobias Meckel, Henning D. Mootz, Harald Kolmar. Generation of Potent Anti-HER1/2 Immunotoxins by Protein Ligation Using Split Inteins. ACS Chemical Biology 2018, 13 (8) , 2058-2066. https://doi.org/10.1021/acschembio.8b00222
- Shogo Nakano, Tomoharu Motoyama, Yurina Miyashita, Yuki Ishizuka, Naoya Matsuo, Hiroaki Tokiwa, Suguru Shinoda, Yasuhisa Asano, Sohei Ito. Benchmark Analysis of Native and Artificial NAD+-Dependent Enzymes Generated by a Sequence-Based Design Method with or without Phylogenetic Data. Biochemistry 2018, 57 (26) , 3722-3732. https://doi.org/10.1021/acs.biochem.8b00339
- Stanislav Anastassov, Maurice Filo, Ching-Hsiang Chang, Mustafa Khammash. A cybergenetic framework for engineering intein-mediated integral feedback control systems. Nature Communications 2023, 14 (1) https://doi.org/10.1038/s41467-023-36863-9
- Risa Yamada, Ishin Nakahara, Izumi Kumagai, Ryutaro Asano, Takeshi Nakanishi, Koki Makabe. Construction of IgG–Fab2 bispecific antibody via intein-mediated protein trans-splicing reaction. Scientific Reports 2023, 13 (1) https://doi.org/10.1038/s41598-023-43110-0
- Baochang Zhang, Yupeng Zheng, Guochao Chu, Xiangyu Deng, Tongyue Wang, Weiwei Shi, Yongkang Zhou, Shan Tang, Ji‐Shen Zheng, Lei Liu. Backbone‐Installed Split Intein‐Assisted Ligation for the Chemical Synthesis of Mirror‐Image Proteins. Angewandte Chemie International Edition 2023, 62 (33) https://doi.org/10.1002/anie.202306270
- Baochang Zhang, Yupeng Zheng, Guochao Chu, Xiangyu Deng, Tongyue Wang, Weiwei Shi, Yongkang Zhou, Shan Tang, Ji‐Shen Zheng, Lei Liu. Backbone‐Installed Split Intein‐Assisted Ligation for the Chemical Synthesis of Mirror‐Image Proteins. Angewandte Chemie 2023, 135 (33) https://doi.org/10.1002/ange.202306270
- Eduard Ebberink, Simon Fernandes, Georgios Hatzopoulos, Ninad Agashe, Po-Han Chang, Nora Guidotti, Timothy M. Reichart, Luc Reymond, Marie-Claire Velluz, Fabian Schneider, Cédric Pourroy, Carsten Janke, Pierre Gönczy, Beat Fierz, Charlotte Aumeier. Tubulin engineering by semi-synthesis reveals that polyglutamylation directs detyrosination. Nature Chemistry 2023, 15 (8) , 1179-1187. https://doi.org/10.1038/s41557-023-01228-8
- Qin Zhan, Changhua Shi, Yu Jiang, Xianling Gao, Ying Lin. Efficient splicing of the CPE intein derived from directed evolution of the Cryptococcus neoformans PRP8 intein. Acta Biochimica et Biophysica Sinica 2023, 55 (8) , 1310-1318. https://doi.org/10.3724/abbs.2023135
- Jia Sen Tan, Muhamad Norizwan Bin Jaffar Ali, Bee Koon Gan, Wen Siang Tan. Next-generation viral nanoparticles for targeted delivery of therapeutics: Fundamentals, methods, biomedical applications, and challenges. Expert Opinion on Drug Delivery 2023, 20 (7) , 955-978. https://doi.org/10.1080/17425247.2023.2228202
- Tingting Lin, Shuncheng Zhang, Di Zhang, Xiaofeng Chen, Yuanyuan Ge, Yafang Hu, Jun Fan. Use of the redox-dependent intein system for enhancing production of the cyclic green fluorescent protein. Protein Expression and Purification 2023, 207 , 106272. https://doi.org/10.1016/j.pep.2023.106272
- Zhixi Liu, Lu Huang, Han Deng, Yan Chen, Hongtao Xiao. Characteristics, Recombination Methods, and Applications Progresses of Split-Cas9 System. Human Gene Therapy 2023, 34 (13-14) , 594-604. https://doi.org/10.1089/hum.2022.223
- Ciaran P. Seath, Antony J. Burton, Xuemeng Sun, Gihoon Lee, Ralph E. Kleiner, David W. C. MacMillan, Tom W. Muir. Tracking chromatin state changes using nanoscale photo-proximity labelling. Nature 2023, 616 (7957) , 574-580. https://doi.org/10.1038/s41586-023-05914-y
- Rasmus Pihl, Qingfei Zheng, Yael David. Nature-inspired protein ligation and its applications. Nature Reviews Chemistry 2023, 7 (4) , 234-255. https://doi.org/10.1038/s41570-023-00468-z
- Qun Zhou. Site-Specific Antibody Conjugation with Payloads beyond Cytotoxins. Molecules 2023, 28 (3) , 917. https://doi.org/10.3390/molecules28030917
- Jiahe Li, Chao Ma, Hongjie Zhang, Kai Liu. Engineering Mechanical Strong Biomaterials Inspired by Structural Building Blocks in Nature. Chemical Research in Chinese Universities 2023, 39 (1) , 92-106. https://doi.org/10.1007/s40242-023-2357-1
- Raymond F. Berkeley, Galia T. Debelouchina. Chemical tools for study and modulation of biomolecular phase transitions. Chemical Science 2022, 13 (48) , 14226-14245. https://doi.org/10.1039/D2SC04907D
- Rory L. Williams, Richard M. Murray. Integrase-mediated differentiation circuits improve evolutionary stability of burdensome and toxic functions in E. coli. Nature Communications 2022, 13 (1) https://doi.org/10.1038/s41467-022-34361-y
- Rosana S. Molina, Gordon Rix, Amanuella A. Mengiste, Beatriz Álvarez, Daeje Seo, Haiqi Chen, Juan E. Hurtado, Qiong Zhang, Jorge Donato García-García, Zachary J. Heins, Patrick J. Almhjell, Frances H. Arnold, Ahmad S. Khalil, Andrew D. Hanson, John E. Dueber, David V. Schaffer, Fei Chen, Seokhee Kim, Luis Ángel Fernández, Matthew D. Shoulders, Chang C. Liu. In vivo hypermutation and continuous evolution. Nature Reviews Methods Primers 2022, 2 (1) https://doi.org/10.1038/s43586-022-00119-5
- Hiroaki Ohishi, Seiru Shimada, Satoshi Uchino, Jieru Li, Yuko Sato, Manabu Shintani, Hitoshi Owada, Yasuyuki Ohkawa, Alexandros Pertsinidis, Takashi Yamamoto, Hiroshi Kimura, Hiroshi Ochiai. STREAMING-tag system reveals spatiotemporal relationships between transcriptional regulatory factors and transcriptional activity. Nature Communications 2022, 13 (1) https://doi.org/10.1038/s41467-022-35286-2
- Hai‐Feng Xia, Jiu‐Pei Luo, Shi‐Rui Yu, Ting‐Jun Zhou. Modification of C‐segment of Cfa DnaE split intein for improving clean‐in‐place in chromatography process. Biotechnology Progress 2022, 38 (5) https://doi.org/10.1002/btpr.3266
- Xun‐Cheng Su, Ling‐Yang Zhang, Li‐Na Zhao, Bin‐Bin Pan, Ben‐Guang Chen, Jia‐Liang Chen, Cheng‐Liang Zhai, Bin Li. Efficient Protein–Protein Couplings Mediated by Small Molecules under Mild Conditions. Angewandte Chemie 2022, 134 (35) https://doi.org/10.1002/ange.202205597
- Xun‐Cheng Su, Ling‐Yang Zhang, Li‐Na Zhao, Bin‐Bin Pan, Ben‐Guang Chen, Jia‐Liang Chen, Cheng‐Liang Zhai, Bin Li. Efficient Protein–Protein Couplings Mediated by Small Molecules under Mild Conditions. Angewandte Chemie International Edition 2022, 61 (35) https://doi.org/10.1002/anie.202205597
- Seokoh Moon, Jinhyo Chung, Yuna Kim, Celab Hong, Soomin Kim, Jaehyeon Hwang, Younghun Jung, Woo-Jae Chung, Dae-Hyuk Kweon. Bifunctional hetero di-disc for broad-spectrum influenza neutralization. Nanomedicine: Nanotechnology, Biology and Medicine 2022, 44 , 102587. https://doi.org/10.1016/j.nano.2022.102587
- Evan T. Clark, Elanor E. Sievers, Galia T. Debelouchina. A Chemical Biology Primer for NMR Spectroscopists. Journal of Magnetic Resonance Open 2022, 10-11 , 100044. https://doi.org/10.1016/j.jmro.2022.100044
- Huasong Ai, Shuai Peng, Jia‐Bin Li. Chemical methods for studying the crosstalk between histone H2B ubiquitylation and H3 methylation. Journal of Peptide Science 2022, 28 (5) https://doi.org/10.1002/psc.3381
- Jaehyeon Hwang, Younghun Jung, Seokoh Moon, Seokhyeon Yu, Hyunseok Oh, Soomin Kim, Kyeong Won Kim, Jeong Hyeon Yoon, Jihwan Chun, Sang Jick Kim, Woo‐Jae Chung, Dae‐Hyuk Kweon. Nanodisc‐Mediated Conversion of Virustatic Antiviral Antibody to Disrupt Virus Envelope in Infected Cells. Small Methods 2022, 6 (4) https://doi.org/10.1002/smtd.202101516
- Hao Wang, Lin Wang, Baihua Zhong, Zhuojun Dai. Protein Splicing of Inteins: A Powerful Tool in Synthetic Biology. Frontiers in Bioengineering and Biotechnology 2022, 10 https://doi.org/10.3389/fbioe.2022.810180
- James W Bryson, Jamie Y Auxillos, Susan J Rosser. Multiplexed activation in mammalian cells using a split-intein CRISPR/Cas12a based synthetic transcription factor. Nucleic Acids Research 2022, 50 (1) , 549-560. https://doi.org/10.1093/nar/gkab1191
- Nikolai N. Sluchanko. Recent advances in structural studies of 14-3-3 protein complexes. 2022, 289-324. https://doi.org/10.1016/bs.apcsb.2021.12.004
- Li Xu, Chen Zhang, Haiwen Li, Peipei Wang, Yandi Gao, Nahush A. Mokadam, Jianjie Ma, W. David Arnold, Renzhi Han. Efficient precise in vivo base editing in adult dystrophic mice. Nature Communications 2021, 12 (1) https://doi.org/10.1038/s41467-021-23996-y
- Mary S. Morrison, Tina Wang, Aditya Raguram, Colin Hemez, David R. Liu. Disulfide-compatible phage-assisted continuous evolution in the periplasmic space. Nature Communications 2021, 12 (1) https://doi.org/10.1038/s41467-021-26279-8
- Hiroki Akiba, Tomoko Ise, Satoshi Nagata, Haruhiko Kamada, Hiroaki Ohno, Kouhei Tsumoto. Production of IgG1-based bispecific antibody without extra cysteine residue via intein-mediated protein trans-splicing. Scientific Reports 2021, 11 (1) https://doi.org/10.1038/s41598-021-98855-3
- Yi Liu, Wei Huang, Minsi Meng, Minhui Chen, Chengjian Cao. Progress in the application of spider silk protein in medicine. Journal of Biomaterials Applications 2021, 36 (5) , 859-871. https://doi.org/10.1177/08853282211003850
- Bryce E. Ackermann, Galia T. Debelouchina. Emerging Contributions of Solid-State NMR Spectroscopy to Chromatin Structural Biology. Frontiers in Molecular Biosciences 2021, 8 https://doi.org/10.3389/fmolb.2021.741581
- Minh Thuan Nguyen Tran, K.C. Rajendra, Freya M. Patterson, Guei-Sheung Liu, Anthony L. Cook, Alex W. Hewitt. Approaches for the sensitive detection of rare base and prime editing events. Methods 2021, 194 , 75-82. https://doi.org/10.1016/j.ymeth.2021.01.006
- Iacopo Galleano, Hendrik Harms, Koushik Choudhury, Keith Khoo, Lucie Delemotte, Stephan Alexander Pless. Functional cross-talk between phosphorylation and disease-causing mutations in the cardiac sodium channel Na v 1.5. Proceedings of the National Academy of Sciences 2021, 118 (33) https://doi.org/10.1073/pnas.2025320118
- Haigang Song, Antony J. Burton, Sally L. Shirran, Jūratė Fahrig‐Kamarauskaitė, Hannelore Kaspar, Tom W. Muir, Markus Künzler, James H. Naismith. Engineering of a Peptide α‐N‐Methyltransferase to Methylate Non‐Proteinogenic Amino Acids. Angewandte Chemie International Edition 2021, 60 (26) , 14319-14323. https://doi.org/10.1002/anie.202100818
- Haigang Song, Antony J. Burton, Sally L. Shirran, Jūratė Fahrig‐Kamarauskaitė, Hannelore Kaspar, Tom W. Muir, Markus Künzler, James H. Naismith. Engineering of a Peptide α‐N‐Methyltransferase to Methylate Non‐Proteinogenic Amino Acids. Angewandte Chemie 2021, 133 (26) , 14440-14444. https://doi.org/10.1002/ange.202100818
- Heather C. Hayes, Louis Y. P. Luk, Yu-Hsuan Tsai. Approaches for peptide and protein cyclisation. Organic & Biomolecular Chemistry 2021, 19 (18) , 3983-4001. https://doi.org/10.1039/D1OB00411E
- John D. Bagert, Michelle M. Mitchener, Agata L. Patriotis, Barbara E. Dul, Felix Wojcik, Benjamin A. Nacev, Lijuan Feng, C. David Allis, Tom W. Muir. Oncohistone mutations enhance chromatin remodeling and alter cell fates. Nature Chemical Biology 2021, 17 (4) , 403-411. https://doi.org/10.1038/s41589-021-00738-1
- Chakhee Kim, Myungseo Park, Jinkyeong Yang, Jonghyeok Shin, Yong-Cheol Park, Sun-Ki Kim, Dae-Hyuk Kweon. Inducible plasmid display system for high-throughput selection of proteins with improved solubility. Journal of Biotechnology 2021, 329 , 143-150. https://doi.org/10.1016/j.jbiotec.2020.12.013
- Masanobu Nakata, Bernd Kreikemeyer. Genetics, Structure, and Function of Group A Streptococcal Pili. Frontiers in Microbiology 2021, 12 https://doi.org/10.3389/fmicb.2021.616508
- Misaki Kawase, Meiko Fujioka, Tsuyoshi Takahashi. Activation of Protease and Luciferase Using Engineered Nostoc punctiforme PCC73102 DnaE Intein with Altered Split Position. ChemBioChem 2021, 22 (3) , 577-584. https://doi.org/10.1002/cbic.202000609
- Filipe Pinto, Ella Lucille Thornton, Baojun Wang. An expanded library of orthogonal split inteins enables modular multi-peptide assemblies. Nature Communications 2020, 11 (1) https://doi.org/10.1038/s41467-020-15272-2
- K. K. Khoo, I. Galleano, F. Gasparri, R. Wieneke, H. Harms, M. H. Poulsen, H. C. Chua, M. Wulf, R. Tampé, S. A. Pless. Chemical modification of proteins by insertion of synthetic peptides using tandem protein trans-splicing. Nature Communications 2020, 11 (1) https://doi.org/10.1038/s41467-020-16208-6
- Mohammad Karimi Baba ahmadi, Seyed Abolghasem Mohammadi, Manoochehr Makvandi, Morteza Mamoueie, Mohammad Rahmati, David Wood. Column-free purification and coating of SpyCatcher protein on ELISA wells generates universal solid support for capturing of SpyTag-fusion protein from the non-purified condition. Protein Expression and Purification 2020, 174 , 105650. https://doi.org/10.1016/j.pep.2020.105650
- N. Braun, Z. P. Sheikh, S. A. Pless. The current chemical biology tool box for studying ion channels. The Journal of Physiology 2020, 598 (20) , 4455-4471. https://doi.org/10.1113/JP276695
- . Cysteinyl Proteomes: Exotic Modifications. 2020, 358-381. https://doi.org/10.1039/9781839161841-00358
- Antony J. Burton, Michael Haugbro, Leah A. Gates, John D. Bagert, C. David Allis, Tom W. Muir. In situ chromatin interactomics using a chemical bait and trap approach. Nature Chemistry 2020, 12 (6) , 520-527. https://doi.org/10.1038/s41557-020-0474-8
- Han Na Seo, Duhee Bang. Promiscuous Trans-splicing Activities Revealed by Next Generation Sequencing-based Analysis of 298 Split Inteins. Biotechnology and Bioprocess Engineering 2020, 25 (2) , 293-301. https://doi.org/10.1007/s12257-019-0394-x
- Zhipeng A. Wang, Philip A. Cole. Methods and Applications of Expressed Protein Ligation. 2020, 1-13. https://doi.org/10.1007/978-1-0716-0434-2_1
- Nicholas A. Prescott, Yael David. In Vivo Histone Labeling Using Ultrafast trans-Splicing Inteins. 2020, 201-219. https://doi.org/10.1007/978-1-0716-0434-2_10
- Neel H. Shah, Adam J. Stevens. Identification, Characterization, and Optimization of Split Inteins. 2020, 31-54. https://doi.org/10.1007/978-1-0716-0434-2_3
- Byung Joon Lim, Raymond F. Berkeley, Galia T. Debelouchina. Fused Split Inteins: Tools for Introducing Multiple Protein Modifications. 2020, 163-181. https://doi.org/10.1007/978-1-0716-0434-2_8
- Matt Sternke, Katherine W. Tripp, Doug Barrick. The use of consensus sequence information to engineer stability and activity in proteins. 2020, 149-179. https://doi.org/10.1016/bs.mie.2020.06.001
- Tim Hofmann, Johannes Schmidt, Elke Ciesielski, Stefan Becker, Thomas Rysiok, Mark Schütte, Lars Toleikis, Harald Kolmar, Achim Doerner. Intein mediated high throughput screening for bispecific antibodies. mAbs 2020, 12 (1) https://doi.org/10.1080/19420862.2020.1731938
- Jonathan M. Levy, Wei-Hsi Yeh, Nachiket Pendse, Jessie R. Davis, Erin Hennessey, Rossano Butcher, Luke W. Koblan, Jason Comander, Qin Liu, David R. Liu. Cytosine and adenine base editing of the brain, liver, retina, heart and skeletal muscle of mice via adeno-associated viruses. Nature Biomedical Engineering 2020, 4 (1) , 97-110. https://doi.org/10.1038/s41551-019-0501-5
- Anthony H. Keeble, Paula Turkki, Samuel Stokes, Irsyad N. A. Khairil Anuar, Rolle Rahikainen, Vesa P. Hytönen, Mark Howarth. Approaching infinite affinity through engineering of peptide–protein interaction. Proceedings of the National Academy of Sciences 2019, 116 (52) , 26523-26533. https://doi.org/10.1073/pnas.1909653116
- Femi J Olorunniji, Makeba Lawson-Williams, Arlene L McPherson, Jane E Paget, W Marshall Stark, Susan J Rosser. Control of ϕC31 integrase-mediated site-specific recombination by protein trans-splicing. Nucleic Acids Research 2019, 47 (21) , 11452-11460. https://doi.org/10.1093/nar/gkz936
- Wenqin Bai, Cameron J. Sargent, Jeong-Mo Choi, Rohit V. Pappu, Fuzhong Zhang. Covalently-assembled single-chain protein nanostructures with ultra-high stability. Nature Communications 2019, 10 (1) https://doi.org/10.1038/s41467-019-11285-8
- Shehryar Ahmad, Boyuan Wang, Matthew D. Walker, Hiu-Ki R. Tran, Peter J. Stogios, Alexei Savchenko, Robert A. Grant, Andrew G. McArthur, Michael T. Laub, John C. Whitney. An interbacterial toxin inhibits target cell growth by synthesizing (p)ppApp. Nature 2019, 575 (7784) , 674-678. https://doi.org/10.1038/s41586-019-1735-9
- Maniraj Bhagawati, Tobias M. E. Terhorst, Friederike Füsser, Simon Hoffmann, Tim Pasch, Shmuel Pietrokovski, Henning D. Mootz. A mesophilic cysteine-less split intein for protein trans -splicing applications under oxidizing conditions. Proceedings of the National Academy of Sciences 2019, 116 (44) , 22164-22172. https://doi.org/10.1073/pnas.1909825116
- Yasushi Kondo, Jana Ognjenović, Saikat Banerjee, Deepti Karandur, Alan Merk, Kayla Kulhanek, Kathryn Wong, Jeroen P. Roose, Sriram Subramaniam, John Kuriyan. Cryo-EM structure of a dimeric B-Raf:14-3-3 complex reveals asymmetry in the active sites of B-Raf kinases. Science 2019, 366 (6461) , 109-115. https://doi.org/10.1126/science.aay0543
- Anwesha Purkayastha, Taek Jin Kang. Stabilization of Proteins by Covalent Cyclization. Biotechnology and Bioprocess Engineering 2019, 24 (5) , 702-712. https://doi.org/10.1007/s12257-019-0363-4
- Robert E. Thompson, Adam J. Stevens, Tom. W. Muir. Protein engineering through tandem transamidation. Nature Chemistry 2019, 11 (8) , 737-743. https://doi.org/10.1038/s41557-019-0281-2
- Hannes M. Beyer, Hideo Iwaï. Off‐Pathway‐Sensitive Protein‐Splicing Screening Based on a Toxin/Antitoxin System. ChemBioChem 2019, 20 (15) , 1933-1938. https://doi.org/10.1002/cbic.201900139
- Matt Sternke, Katherine W. Tripp, Doug Barrick. Consensus sequence design as a general strategy to create hyperstable, biologically active proteins. Proceedings of the National Academy of Sciences 2019, 116 (23) , 11275-11284. https://doi.org/10.1073/pnas.1816707116
- James N. Wright, Wan Ling Wong, Joseph A. Harvey, James A. Garnett, Laura S. Itzhaki, Ewan R.G. Main. Scalable Geometrically Designed Protein Cages Assembled via Genetically Encoded Split Inteins. Structure 2019, 27 (5) , 776-784.e4. https://doi.org/10.1016/j.str.2019.02.005
- William Y. C. Huang, Steven Alvarez, Yasushi Kondo, Young Kwang Lee, Jean K. Chung, Hiu Yue Monatrice Lam, Kabir H. Biswas, John Kuriyan, Jay T. Groves. A molecular assembly phase transition and kinetic proofreading modulate Ras activation by SOS. Science 2019, 363 (6431) , 1098-1103. https://doi.org/10.1126/science.aau5721
- Boyuan Wang, Peng Dai, David Ding, Amanda Del Rosario, Robert A. Grant, Bradley L. Pentelute, Michael T. Laub. Affinity-based capture and identification of protein effectors of the growth regulator ppGpp. Nature Chemical Biology 2019, 15 (2) , 141-150. https://doi.org/10.1038/s41589-018-0183-4
- Francisco Castillo, Ali Tavassoli. Genetic Selections with SICLOPPS Libraries: Toward the Identification of Novel Protein–Protein Interaction Inhibitors and Chemical Tools. 2019, 317-328. https://doi.org/10.1007/978-1-4939-9504-2_15
- Igor Maksimovic, Devin Ray, Qingfei Zheng, Yael David. Utilizing intein trans-splicing for in vivo generation of site-specifically modified proteins. 2019, 203-222. https://doi.org/10.1016/bs.mie.2019.07.015
- Donghyeok Gang, Do Kim, Hee-Sung Park. Cyclic Peptides: Promising Scaffolds for Biopharmaceuticals. Genes 2018, 9 (11) , 557. https://doi.org/10.3390/genes9110557
- Dylan T. Murray, Xiaoming Zhou, Masato Kato, Siheng Xiang, Robert Tycko, Steven L. McKnight. Structural characterization of the D290V mutation site in hnRNPA2 low-complexity–domain polymers. Proceedings of the National Academy of Sciences 2018, 115 (42) https://doi.org/10.1073/pnas.1806174115
- Euiyeon Lee, Kyoungmi Min, Young‐Tae Chang, Youngeun Kwon. Efficient and wash‐free labeling of membrane proteins using engineered Npu DnaE split‐inteins. Protein Science 2018, 27 (9) , 1568-1574. https://doi.org/10.1002/pro.3455
- Anusuya Banerjee, Mark Howarth. Nanoteamwork: covalent protein assembly beyond duets towards protein ensembles and orchestras. Current Opinion in Biotechnology 2018, 51 , 16-23. https://doi.org/10.1016/j.copbio.2017.10.006
- Theetha Pavankumar. Inteins: Localized Distribution, Gene Regulation, and Protein Engineering for Biological Applications. Microorganisms 2018, 6 (1) , 19. https://doi.org/10.3390/microorganisms6010019
- Josef A. Gramespacher, Adam J. Stevens, Robert E. Thompson, Tom W. Muir. Improved protein splicing using embedded split inteins. Protein Science 2018, 27 (3) , 614-619. https://doi.org/10.1002/pro.3357
- Sebanti Gupta, Robert Tycko. Segmental isotopic labeling of HIV-1 capsid protein assemblies for solid state NMR. Journal of Biomolecular NMR 2018, 70 (2) , 103-114. https://doi.org/10.1007/s10858-017-0162-1
- Jacob Valentine, Ali Tavassoli. Genetically Encoded Cyclic Peptide Libraries: From Hit to Lead and Beyond. 2018, 117-134. https://doi.org/10.1016/bs.mie.2018.09.020
- Alexander Muik, Johanna Reul, Thorsten Friedel, Anke Muth, Karen Patricia Hartmann, Irene C. Schneider, Robert C. Münch, Christian J. Buchholz. Covalent coupling of high-affinity ligands to the surface of viral vector particles by protein trans-splicing mediates cell type-specific gene transfer. Biomaterials 2017, 144 , 84-94. https://doi.org/10.1016/j.biomaterials.2017.07.032
- Yakup Ermurat. A REVIEW ON INTEIN MEDIATED RECOMBINANT PROTEIN PURIFICATION PROCESSES WITHOUT USE OF PROTEASE. ANADOLU UNIVERSITY JOURNAL OF SCIENCE AND TECHNOLOGY –C Life Sciences and Biotechnology 2017, 6 (2) , 95-102. https://doi.org/10.18036/aubtdc.267017
- Adam J. Stevens, Giridhar Sekar, Neel H. Shah, Anahita Z. Mostafavi, David Cowburn, Tom W. Muir. A promiscuous split intein with expanded protein engineering applications. Proceedings of the National Academy of Sciences 2017, 114 (32) , 8538-8543. https://doi.org/10.1073/pnas.1701083114
- Guang-Zhong Yin, Wen-Bin Zhang, Stephen Z.D. Cheng. Giant molecules: where chemistry, physics, and bio-science meet. Science China Chemistry 2017, 60 (3) , 338-352. https://doi.org/10.1007/s11426-016-0436-x
- Galia T. Debelouchina, Tom W. Muir. A molecular engineering toolbox for the structural biologist. Quarterly Reviews of Biophysics 2017, 50 https://doi.org/10.1017/S0033583517000051
- Benjamin T. Porebski, Ashley M. Buckle. Consensus protein design. Protein Engineering Design and Selection 2016, 29 (7) , 245-251. https://doi.org/10.1093/protein/gzw015
- Heyi Zhang, Hugo van Ingen. Isotope-labeling strategies for solution NMR studies of macromolecular assemblies. Current Opinion in Structural Biology 2016, 38 , 75-82. https://doi.org/10.1016/j.sbi.2016.05.008
Abstract
Figure 1
Figure 1. Design of the Cfa split intein. (a) Sequence alignment of Npu DnaE and Cfa DnaE. The sequences share 82% identity with the differences (cyan) evenly distributed through the primary sequence. Catalytic residues and second shell “accelerator” residues are shown in orange and green, respectively. (b) The same residues highlighted in panel a mapped on to the Npu structure (PDB ID 4Kl5).
Figure 2
Figure 2. Characterization of the Cfa intein. (a) Splicing rates for Cfa and Npu as a function of temperature. Npu is inactive at 80 °C. Error = SD (n = 3). (b, c) Splicing rates for Cfa and Npu as a function of added chaotrope. Npu is inactive in 3 M GuHCl or 8 M Urea. Note: Cfa has residual activity in 4 M GuHCl (k = 7 × 10–5). Error = SD (n = 3).
Figure 3
Figure 3. Expression and modification of a mouse monoclonal antibody using the Cfa intein. (a) Test expression in HEK293T cells of various IntN homologues (Npu, Mcht, Ava, and Cfa) fused to the C-terminus of the heavy chain of a mouse αDec205 monoclonal antibody. Top: Western blot analysis (αMouse IgG) of antibody levels present in the medium following the 96 h expression. Bottom: α-actin Western blot of cell lysate as a loading control. (b) Quantification of normalized expression yield by densitometry of αDEC205 HC-IntN signal in panel a. Error = SD (n = 4). (c) Structure of the CfaC-dendrimer construct used in PTS reactions with the αDEC205 HC-IntN fusion. For simplicity the CfaC peptide sequence and a short peptide linker are depicted symbolically in teal. (d) Schematic of the in situ PTS approach used to modify the HC of an mAb with a multivalent cargo. (e) SDS-PAGE analysis of PTS reaction. Lane 1: Wild type mouse αDEC205 mAB. Lane 2: Mouse αDEC205-CfaN mAB fusion. Lane 3: addition of the CfaC-dendrimer to the media containing the αDEC205-CfaN mAB. The splicing reaction was analyzed after two hours by fluorescence (bottom) and Western blot (top, αMouse IgG).
References
ARTICLE SECTIONSThis article references 21 other publications.
- 1Shah, N. H.; Muir, T. W. Chem. Sci. 2014, 5, 446 DOI: 10.1039/C3SC52951G[Crossref], [PubMed], [CAS], Google Scholar1https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3sXitVSqtbzJ&md5=e91aafcf49109d059dbed41e363c6811Inteins: nature's gift to protein chemistsShah, Neel H.; Muir, Tom W.Chemical Science (2014), 5 (2), 446-461CODEN: CSHCCN; ISSN:2041-6520. (Royal Society of Chemistry)A review. Inteins are auto-processing domains found in organisms from all domains of life. These proteins carry out a process known as protein splicing, which is a multi-step biochem. reaction comprised of both the cleavage and formation of peptide bonds. While the endogenous substrates of protein splicing are specific essential proteins found in intein-contg. host organisms, inteins are also functional in exogenous contexts and can be used to chem. manipulate virtually any polypeptide backbone. Given this, protein chemists have exploited various facets of intein reactivity to modify proteins in myriad ways for both basic biol. research as well as potential therapeutic applications. Here, we review the intein field, first focusing on the biol. context and phylogenetic diversity of inteins, followed by a description of intein structure and biochem. function. Finally, we discuss prevalent intein-based technologies, focusing on their applications in chem. biol., followed by persistent caveats of intein chem. and approaches to alleviate these shortcomings. The findings summarized herein describe two and a half decades of research, leading from a biochem. curiosity to the development of powerful protein engineering tools.
- 2Wu, H.; Hu, Z.; Liu, X. Q. Proc. Natl. Acad. Sci. U. S. A. 1998, 95, 9226 DOI: 10.1073/pnas.95.16.9226
- 3Iwai, H.; Zuger, S.; Jin, J.; Tam, P. H. FEBS Lett. 2006, 580, 1853 DOI: 10.1016/j.febslet.2006.02.045
- 4Zettler, J.; Schutz, V.; Mootz, H. D. FEBS Lett. 2009, 583, 909 DOI: 10.1016/j.febslet.2009.02.003
- 5Shah, N. H.; Eryilmaz, E.; Cowburn, D.; Muir, T. W. J. Am. Chem. Soc. 2013, 135, 5839 DOI: 10.1021/ja401015p
- 6Shah, N. H.; Dann, G. P.; Vila-Perello, M.; Liu, Z.; Muir, T. W. J. Am. Chem. Soc. 2012, 134, 11338 DOI: 10.1021/ja303226x
- 7Carvajal-Vallejos, P.; Pallisse, R.; Mootz, H. D.; Schmidt, S. R. J. Biol. Chem. 2012, 287, 28686 DOI: 10.1074/jbc.M112.372680
- 8Wu, Q.; Gao, Z.; Wei, Y.; Ma, G.; Zheng, Y.; Dong, Y.; Liu, Y. Biochem. J. 2014, 461, 247 DOI: 10.1042/BJ20140287
- 9Aranko, A. S.; Oeemig, J. S.; Kajander, T.; Iwai, H. Nat. Chem. Biol. 2013, 9, 616 DOI: 10.1038/nchembio.1320[Crossref], [PubMed], [CAS], Google Scholar9https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3sXhtlWkurvE&md5=5cd3a3273984caeeba5201c7f3f6db50Intermolecular domain swapping induces intein-mediated protein alternative splicingAranko, A. Sesilja; Oeemig, Jesper S.; Kajander, Tommi; Iwai, HideoNature Chemical Biology (2013), 9 (10), 616-622CODEN: NCBABT; ISSN:1552-4450. (Nature Publishing Group)Protein sequences are diversified on the DNA level by recombination and mutation and can be further increased on the RNA level by alternative RNA splicing, involving introns that have important roles in many biol. processes. The protein version of introns (inteins), which catalyze protein splicing, were first reported in the 1990s. The biol. roles of protein splicing still remain elusive because inteins neither provide any clear benefits nor have an essential role in their host organisms. We now report protein alternative splicing, in which new protein sequences can be produced by protein recombination by intermol. domain swapping of inteins, as elucidated by NMR spectroscopy and crystal structures. We demonstrate that intein-mediated protein alternative splicing could be a new strategy to increase protein diversity (i.e., functions) without any modification in genetic backgrounds. We also exploited it as a post-translational protein conformation-driven switch of protein functions (for example, as highly specific protein interference).
- 10Pietrokovski, S. Protein Sci. 1994, 3, 2340 DOI: 10.1002/pro.5560031218[Crossref], [PubMed], [CAS], Google Scholar10https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADyaK2MXjslajur8%253D&md5=6c5a56795954e6fb607902b5674f2426Conserved sequence features of inteins (protein introns) and their use in identifying new inteins and related proteinsPietrokovski, ShmuelProtein Science (1994), 3 (12), 2340-50CODEN: PRCIEI; ISSN:0961-8368. (Cambridge University Press)Inteins (protein introns) are internal portions of protein sequences that are post-translationally excised while the flanking regions are spliced together, making an addnl. protein product. Inteins have been found in a no. of homologous genes in yeast, mycobacteria, and extreme thermophile archaebacteria. The inteins are probably multifunctional, auto-catalyzing their own splicing, and some were also shown to be DNA endonucleases. The splice junction regions and two regions similar to homing endonucleases were thought to be the only common sequence features of inteins. This work analyzed all published intein sequences with recently developed methods for detecting weak, conserved sequence features. The methods complemented each other in the identification and assessment of several patterns characterizing the intein sequences. New intein conserved features are discovered and the known ones are quant. described and localized. The general sequence description of all the known inteins is derived from the motifs and their relative positions. The intein sequence description is used to search the sequence data-bases for intein-like proteins. A sequence region in a mycobacterial open reading frame possessing all of the intein motifs and absent from sequences homologous to both of its flanking sequences id identified as an intein. A newly discovered putative intein in red algae chloroplasts is found not to contain the endonuclease motifs present in all other inteins. The yeast HO endonuclease is found to have an overall intein-like structure and a few viral polyprotein cleavage sites are found to be significantly similar to the inteins amino-end splice junction motif. The intein features described may serve for detection of intein sequences.
- 11Dearden, A. K.; Callahan, B.; Roey, P. V.; Li, Z.; Kumar, U.; Belfort, M.; Nayak, S. K. Protein Sci. 2013, 22, 557 DOI: 10.1002/pro.2236[Crossref], [PubMed], [CAS], Google Scholar11https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3sXmsFWqtr8%253D&md5=300b3e042ff7c8609b9815ad61443523A conserved threonine spring-loads precursor for intein splicingDearden, Albert K.; Callahan, Brian; Van Roey, Patrick; Li, Zhong; Kumar, Utsav; Belfort, Marlene; Nayak, Saroj K.Protein Science (2013), 22 (5), 557-563CODEN: PRCIEI; ISSN:1469-896X. (Wiley-Blackwell)Protein splicing is an autocatalytic process where an "intein" self-cleaves from a precursor and ligates the flanking N- and C-"extein" polypeptides. Inteins occur in all domains of life and have myriad uses in biotechnol. Although the reaction steps of protein splicing are known, mechanistic details remain incomplete, particularly the initial peptide rearrangement at the N-terminal extein/intein junction. Recently, we proposed that this transformation, an N-S acyl shift, is accelerated by a localized conformational strain, between the intein's catalytic cysteine (Cys1) and the neighboring glycine (Gly-1) in the N-extein. That proposal was based on the crystal structure of a catalytically competent trapped precursor. Here, we define the structural origins and mechanistic relevance of the conformational strain using a combination of quantum mech. simulations, mutational anal., and X-ray crystallog. Our results implicate a conserved, but largely unstudied, threonine residue of the Ssp DnaE intein (Thr69) as the mediator of conformational strain through hydrogen bonding. Further, the strain imposed by this residue is shown to position the splice junction in a manner that enhances the rate of the N-S acyl shift substantially. Taken together, our results not only provide fundamental understanding of the control of the first step of protein splicing but also have important implications in various biotechnol. applications that require precursor manipulation.
- 12Du, Z.; Shemella, P. T.; Liu, Y.; McCallum, S. A.; Pereira, B.; Nayak, S. K.; Belfort, G.; Belfort, M.; Wang, C. J. Am. Chem. Soc. 2009, 131, 11581 DOI: 10.1021/ja904318w
- 13Lehmann, M.; Kostrewa, D.; Wyss, M.; Brugger, R.; D’Arcy, A.; Pasamontes, L.; van Loon, A. P. Protein Eng., Des. Sel. 2000, 13, 49 DOI: 10.1093/protein/13.1.49
- 14Steipe, B. Methods Enzymol. 2004, 388, 176 DOI: 10.1016/S0076-6879(04)88016-9
- 15Altschul, S. F.; Gish, W.; Miller, W.; Myers, E. W.; Lipman, D. J. J. Mol. Biol. 1990, 215, 403 DOI: 10.1016/S0022-2836(05)80360-2[Crossref], [PubMed], [CAS], Google Scholar15https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADyaK3MXitVGmsA%253D%253D&md5=009d2323eb82f0549356880e1101db16Basic local alignment search toolAltschul, Stephen F.; Gish, Warren; Miller, Webb; Myers, Eugene W.; Lipman, David J.Journal of Molecular Biology (1990), 215 (3), 403-10CODEN: JMOBAK; ISSN:0022-2836.A new approach to rapid sequence comparison, basic local alignment search tool (BLAST), directly approximates alignments that optimize a measure of local similarity, the maximal segment pair (MSP) score. Recent math. results on the stochastic properties of MSP scores allow an anal. of the performance of this method as well as the statistical significance of alignments it generates. The basic algorithm is simple and robust; it can be implemented in a no. of ways and applied in a variety of contexts including straightforward DNA and protein sequence database searches, motif searches, gene identification searches, and in the anal. of multiple regions of similarity in long DNA sequences. In addn. to its flexibility and tractability to math. anal., BLAST is an order of magnitude faster than existing sequence comparison tools of comparable sensitivity.
- 16Grigoriev, I. V.; Nordberg, H.; Shabalov, I.; Aerts, A.; Cantor, M.; Goodstein, D.; Kuo, A.; Minovitsky, S.; Nikitin, R.; Ohm, R. A.; Otillar, R.; Poliakov, A.; Ratnere, I.; Riley, R.; Smirnova, T.; Rokhsar, D.; Dubchak, I. Nucleic Acids Res. 2012, 40, D26 DOI: 10.1093/nar/gkr947
- 17Tatusova, T.; Ciufo, S.; Fedorov, B.; O’Neill, K.; Tolstoy, I. Nucleic Acids Res. 2014, 42, D553 DOI: 10.1093/nar/gkt1274
- 18Shah, N. H.; Eryilmaz, E.; Cowburn, D.; Muir, T. W. J. Am. Chem. Soc. 2013, 135, 18673 DOI: 10.1021/ja4104364
- 19Mohlmann, S.; Bringmann, P.; Greven, S.; Harrenga, A. BMC Biotechnol. 2011, 11, 76 DOI: 10.1186/1472-6750-11-76[Crossref], [PubMed], [CAS], Google Scholar19https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BC3Mjlt1yqtQ%253D%253D&md5=0c498501b2777e84d50836f6aaa0b2ecSite-specific modification of ED-B-targeting antibody using intein-fusion technologyMohlmann Sina; Bringmann Peter; Greven Simone; Harrenga AxelBMC biotechnology (2011), 11 (), 76 ISSN:.BACKGROUND: A promising new approach in cancer therapy is the use of tumor specific antibodies coupled to cytotoxic agents. Currently these immunoconjugates are prepared by rather unspecific coupling chemistries, resulting in heterogeneous products. As the drug load is a key parameter for the antitumor activity, site-specific strategies are desired. Expressed protein ligation (EPL) and protein trans-splicing (PTS) are methods for the specific C-terminal modification of a target protein. Both include the expression as an intein fusion protein, followed by the exchange of the intein for a functionalized moiety. RESULTS: A full-length IgG specific for fibronectin ED-B was expressed as fusion protein with an intein (Mxe GyrA or Npu DnaE) attached to each heavy chain. In vitro protocols were established to site-specifically modify the antibodies in high yields by EPL or PTS, respectively. Although reducing conditions had to be employed during the process, the integrity or affinity of the antibody was not affected. The protocols were used to prepare immunoconjugates containing two biotin molecules per antibody, attached to the C-termini of the heavy chains. CONCLUSION: Full-length antibodies can be efficiently and site-specifically modified at the C-termini of their heavy chains by intein-fusion technologies. The described protocols can be used to prepare immunoconjugates of high homogeneity and with a defined drug load of two. The attachment to the C-termini is expected to retain the affinity and effector functions of the antibodies.
- 20Barbuto, S.; Idoyaga, J.; Vila-Perello, M.; Longhi, M. P.; Breton, G.; Steinman, R. M.; Muir, T. W. Nat. Chem. Biol. 2013, 9, 250 DOI: 10.1038/nchembio.1186[Crossref], [PubMed], [CAS], Google Scholar20https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3sXis1aitr0%253D&md5=460c3394e4559169746733ac91b710f5Induction of innate and adaptive immunity by delivery of poly dA:dT to dendritic cellsBarbuto, Scott; Idoyaga, Juliana; Vila-Perello, Miquel; Longhi, Maria P.; Breton, Gaelle; Steinman, Ralph M.; Muir, Tom W.Nature Chemical Biology (2013), 9 (4), 250-256CODEN: NCBABT; ISSN:1552-4450. (Nature Publishing Group)Targeted delivery of antigens to dendritic cells (DCs) is a promising vaccination strategy. However, to ensure immunity, the approach depends on coadministration of an adjuvant. Here we ask whether targeting of both adjuvant and antigen to DCs is sufficient to induce immunity. Using a protein ligation method, we develop a general approach for linking the immune stimulant, poly dA:dT (pdA:dT), to a monoclonal antibody (mAb) specific for DEC205 (DEC). We show that DEC-specific mAbs deliver pdA:dT to DCs for the efficient prodn. of type I interferon in human monocyte-derived DCs and in mice. Notably, adaptive T-cell immunity is elicited when mAbs specific for DEC-pdA:dT are used as the activation stimuli and are administered together with a DC-targeted antigen. Collectively, our studies indicate that DCs can integrate innate and adaptive immunity in vivo and suggest that dual delivery of antigen and adjuvant to DCs might be an efficient approach to vaccine development.
- 21Shah, N. D.; Parekh, H. S.; Steptoe, R. J. Pharm. Res. 2014, 31, 3150 DOI: 10.1007/s11095-014-1408-1
Supporting Information
Supporting Information
ARTICLE SECTIONSThe Supporting Information is available free of charge on the ACS Publications website at DOI: 10.1021/jacs.5b13528.
Full methods and experimental data (PDF)
Terms & Conditions
Most electronic Supporting Information files are available without a subscription to ACS Web Editions. Such files may be downloaded by article for research use (if there is a public use license linked to the relevant article, that license may permit other uses). Permission may be obtained from ACS for other uses through requests via the RightsLink permission system: http://pubs.acs.org/page/copyright/permissions.html.