1
|
Chong KHC, Liu L, Chua R, Chai YT, Lu Z, Liu R, Tan EYJ, Dong J, Khoh YH, Lin J, Zhong FL, Lescar J, Zheng P, Wu B. Structural Basis of High-Precision Protein Ligation and Its Application. J Am Chem Soc 2025. [PMID: 39745918 DOI: 10.1021/jacs.4c10689] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
Abstract
Enzyme-catalyzed protein modifications have become invaluable in diverse applications, outperforming chemical methods in terms of precision, conjugation efficiency, and biological compatibility. Despite significant advances in ligases, such as sortase A and OaAEP1, their use in heterogeneous biological environments remains constrained by limited target sequence specificity. In 2021, Lupas' group introduced Connectase, a family of repurposed archaeal proteases for protein ligations, but its low processivity and lack of structural information have impeded further engineering for practical biological and biophysical applications. Here, we present the X-ray crystallographic structures of MmConnectase (Methanococcus maripaludis, MmCET) in both apo and substrate-bound forms. Comparative analysis with its inactive paralogue, MjCET (Methanococcus janaschi), reveals the structural basis of MmCET's high-precision ligation activity. We propose modifications to the N-terminal substrate recognition motifs to suppress MmCET's reversible protease activity, enabling high-precision protein ligations in complex biological environments, such as serum-containing cell cultures. To further demonstrate the enhanced processivity and precision, single-molecule protein unfolding experiments showed that our optimized Connectase, in conjunction with OaAEP1(C247A), can perform stepwise tandem ligations of protein leading to a well-defined protein polymer.
Collapse
Affiliation(s)
- Kelvin Han Chung Chong
- School of Biological Sciences, Nanyang Technological University, 60 Nanyang Drive, Singapore 636921, Singapore
- NTU Institute of Structural Biology, Nanyang Technological University, EMB 06-01, 59 Nanyang Drive, Singapore 636921, Singapore
| | - Lichao Liu
- State Key Laboratory of Coordination Chemistry, Chemistry and Biomedicine Innovation Center (ChemBIC), School of Chemistry and Chemical Engineering, Nanjing University, Nanjing 210023, PR China
| | - Rae Chua
- NTU Institute of Structural Biology, Nanyang Technological University, EMB 06-01, 59 Nanyang Drive, Singapore 636921, Singapore
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore 308232, Singapore
| | - Yoke Tin Chai
- School of Biological Sciences, Nanyang Technological University, 60 Nanyang Drive, Singapore 636921, Singapore
| | - Zhuojian Lu
- State Key Laboratory of Coordination Chemistry, Chemistry and Biomedicine Innovation Center (ChemBIC), School of Chemistry and Chemical Engineering, Nanjing University, Nanjing 210023, PR China
| | - Renming Liu
- State Key Laboratory of Coordination Chemistry, Chemistry and Biomedicine Innovation Center (ChemBIC), School of Chemistry and Chemical Engineering, Nanjing University, Nanjing 210023, PR China
| | - Eddie Yong Jun Tan
- School of Biological Sciences, Nanyang Technological University, 60 Nanyang Drive, Singapore 636921, Singapore
| | - Jinxi Dong
- School of Biological Sciences, Nanyang Technological University, 60 Nanyang Drive, Singapore 636921, Singapore
| | - Yek How Khoh
- School of Biological Sciences, Nanyang Technological University, 60 Nanyang Drive, Singapore 636921, Singapore
| | - Jianqing Lin
- School of Biological Sciences, Nanyang Technological University, 60 Nanyang Drive, Singapore 636921, Singapore
- NTU Institute of Structural Biology, Nanyang Technological University, EMB 06-01, 59 Nanyang Drive, Singapore 636921, Singapore
| | - Franklin L Zhong
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore 308232, Singapore
- Skin Research Institute of Singapore (SRIS), Singapore 308232, Singapore
| | - Julien Lescar
- School of Biological Sciences, Nanyang Technological University, 60 Nanyang Drive, Singapore 636921, Singapore
- NTU Institute of Structural Biology, Nanyang Technological University, EMB 06-01, 59 Nanyang Drive, Singapore 636921, Singapore
| | - Peng Zheng
- State Key Laboratory of Coordination Chemistry, Chemistry and Biomedicine Innovation Center (ChemBIC), School of Chemistry and Chemical Engineering, Nanjing University, Nanjing 210023, PR China
| | - Bin Wu
- School of Biological Sciences, Nanyang Technological University, 60 Nanyang Drive, Singapore 636921, Singapore
- NTU Institute of Structural Biology, Nanyang Technological University, EMB 06-01, 59 Nanyang Drive, Singapore 636921, Singapore
| |
Collapse
|
2
|
Ding W, Gu J, Xu W, Wu J, Huang Y, Zhang S, Lin S. The Biosynthesis and Applications of Protein Lipidation. Chem Rev 2024; 124:12176-12212. [PMID: 39441663 DOI: 10.1021/acs.chemrev.4c00419] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2024]
Abstract
Protein lipidation dramatically affects protein structure, localization, and trafficking via remodeling protein-membrane and protein-protein interactions through hydrophobic lipid moieties. Understanding the biosynthesis of lipidated proteins, whether natural ones or mimetics, is crucial for reconstructing, validating, and studying the molecular mechanisms and biological functions of protein lipidation. In this Perspective, we first provide an overview of the natural enzymatic biosynthetic pathways of protein lipidation in mammalian cells, focusing on the enzymatic machineries and their chemical linkages. We then discuss strategies to biosynthesize protein lipidation in mammalian cells by engineering modification machineries and substrates. Additionally, we explore site-specific protein lipidation biosynthesis in vitro via enzyme-mediated ligations and in vivo primarily through genetic code expansion strategies. We also discuss the use of small molecule tools to modulate the process of protein lipidation biosynthesis. Finally, we provide concluding remarks and discuss future directions for the biosynthesis and applications of protein lipidation.
Collapse
Affiliation(s)
- Wenlong Ding
- Life Sciences Institute, Institute of Fundamental and Transdisciplinary Research, Zhejiang University, Hangzhou 310058, China
- Center for Oncology Medicine, the Fourth Affiliated Hospital of School of Medicine, and International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu 322000, China
| | - Jiayu Gu
- Department of Medical Oncology, State Key Laboratory of Transvascular Implantation Devices, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Wenyuan Xu
- Life Sciences Institute, Institute of Fundamental and Transdisciplinary Research, Zhejiang University, Hangzhou 310058, China
| | - Jing Wu
- Hubei Hongshan Laboratory, College of Biomedicine and Health, Huazhong Agricultural University, Wuhan 430070, China
| | - Yiwen Huang
- Hubei Hongshan Laboratory, College of Biomedicine and Health, Huazhong Agricultural University, Wuhan 430070, China
| | - Shuai Zhang
- Hubei Hongshan Laboratory, College of Biomedicine and Health, Huazhong Agricultural University, Wuhan 430070, China
| | - Shixian Lin
- Life Sciences Institute, Institute of Fundamental and Transdisciplinary Research, Zhejiang University, Hangzhou 310058, China
- Zhejiang Provincial Key Laboratory for Cancer Molecular Cell Biology, Shaoxing Institute, Zhejiang University, Shaoxing 321000, China
- Department of Medical Oncology, State Key Laboratory of Transvascular Implantation Devices, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310058, China
| |
Collapse
|
3
|
Antonenko A, Pomorski A, Singh AK, Kapczyńska K, Krężel A. Site-Specific and Fluorescently Enhanced Installation of Post-Translational Protein Modifications via Bifunctional Biarsenical Linker. ACS OMEGA 2024; 9:45127-45137. [PMID: 39554417 PMCID: PMC11561763 DOI: 10.1021/acsomega.4c05828] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/23/2024] [Revised: 09/18/2024] [Accepted: 10/21/2024] [Indexed: 11/19/2024]
Abstract
To understand how particular post-translational modifications (PTMs) affect the function of a target protein, it is essential to first prepare and investigate the target with the modification at the desired position. This drives the continuous development of site-specific protein modification technologies. Here, we present the chemical synthesis and application of the biarsenical linker SrtCrAsH-EDT2, which has a dual labeling functionality. This linker, containing a sortase A recognition motif, can be conjugated with any protein containing the LPXTG motif at the C terminus, such as ubiquitin and the SUMO tag, and then attached to a protein of interest (POI) containing a terminal or bipartite (intramolecularly placed) tetracysteine motif. This modification of the POI facilitates the straightforward and rapid incorporation of PTMs, which are further highlighted by the fluorescent biarsenical probe. Consequently, this directly correlates proteins' physical properties and cellular roles under various physiological conditions or in disease states. The proposed one-pot labeling methodology can be utilized to explore the effects of PTMs on proteins, affecting their structure, function, localization, and interactions within the cellular environment. Understanding these effects is crucial for uncovering the complex mechanisms that regulate cellular function and dysfunction.
Collapse
Affiliation(s)
- Anastasiia Antonenko
- Department
of Chemical Biology, Faculty of Biotechnology, University of Wroclaw, Joliot-Curie 14a, 50-383 Wrocław, Poland
| | - Adam Pomorski
- Department
of Chemical Biology, Faculty of Biotechnology, University of Wroclaw, Joliot-Curie 14a, 50-383 Wrocław, Poland
| | - Avinash Kumar Singh
- Department
of Chemical Biology, Faculty of Biotechnology, University of Wroclaw, Joliot-Curie 14a, 50-383 Wrocław, Poland
- Department
of Laboratory Medicine and Pathology, Mayo
Clinic, Rochester 55901, Minnesota, United States
| | - Katarzyna Kapczyńska
- Department
of Immunology of Infectious Diseases, Hirszfeld
Institute of Immunology and Experimental Therapy, Polish Academy of
Sciences, 53-114 Wrocław, Poland
| | - Artur Krężel
- Department
of Chemical Biology, Faculty of Biotechnology, University of Wroclaw, Joliot-Curie 14a, 50-383 Wrocław, Poland
| |
Collapse
|
4
|
Choi JH, Kim S, Kang OY, Choi SY, Hyun JY, Lee HS, Shin I. Selective fluorescent labeling of cellular proteins and its biological applications. Chem Soc Rev 2024; 53:9446-9489. [PMID: 39109465 DOI: 10.1039/d4cs00094c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/01/2024]
Abstract
Proteins, which are ubiquitous in cells and critical to almost all cellular functions, are indispensable for life. Fluorescence imaging of proteins is key to understanding their functions within their native milieu, as it provides insights into protein localization, dynamics, and trafficking in living systems. Consequently, the selective labeling of target proteins with fluorophores has emerged as a highly active research area, encompassing bioorganic chemistry, chemical biology, and cell biology. Various methods for selectively labeling proteins with fluorophores in cells and tissues have been established and are continually being developed to visualize and characterize proteins. This review highlights research findings reported since 2018, with a focus on the selective labeling of cellular proteins with small organic fluorophores and their biological applications in studying protein-associated biological events. We also discuss the strengths and weaknesses of each labeling approach for their utility in living systems.
Collapse
Affiliation(s)
- Joo Hee Choi
- Department of Chemistry, Yonsei University, 03722 Seoul, Republic of Korea.
| | - Sooin Kim
- Department of Chemistry, Sogang University, 04107 Seoul, Republic of Korea.
| | - On-Yu Kang
- Department of Drug Discovery, Data Convergence Drug Research Center, Korea Research Institute of Chemical Technology (KRICT), Daejeon 34114, Republic of Korea.
| | - Seong Yun Choi
- Department of Drug Discovery, Data Convergence Drug Research Center, Korea Research Institute of Chemical Technology (KRICT), Daejeon 34114, Republic of Korea.
- Pharmaceutical Chemistry, University of Science & Technology, Daejeon 34113, Republic of Korea
| | - Ji Young Hyun
- Department of Drug Discovery, Data Convergence Drug Research Center, Korea Research Institute of Chemical Technology (KRICT), Daejeon 34114, Republic of Korea.
- Pharmaceutical Chemistry, University of Science & Technology, Daejeon 34113, Republic of Korea
| | - Hyun Soo Lee
- Department of Chemistry, Sogang University, 04107 Seoul, Republic of Korea.
| | - Injae Shin
- Department of Chemistry, Yonsei University, 03722 Seoul, Republic of Korea.
| |
Collapse
|
5
|
Bondarchuk T, Vaskiv D, Zhuravel E, Shyshlyk O, Hrynyshyn Y, Nedialko O, Pokholenko O, Pohribna A, Kuchuk O, Brovarets V, Zozulya S. Synthetic Amine Linkers for Efficient Sortagging. Bioconjug Chem 2024; 35:1172-1181. [PMID: 38994647 DOI: 10.1021/acs.bioconjchem.4c00143] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/13/2024]
Abstract
Enzymatic site-specific bioconjugation techniques, in particular sortase-mediated ligation, are increasingly used to generate conjugated proteins for a wide array of applications. Extension of the utility and practicality of sortagging for diverse purposes is critically dependent on further improvement of the efficiency of sortagging reactions with a wider structural variety of substrates. We present a comprehensive comparative mass spectrometry screening study of synthetic nonpeptidic incoming amine nucleophile substrates of Staphylococcus aureus Sortase A enzyme. We have identified the optimal structural motifs among the chemically diverse set of 452 model primary and secondary amine-containing sortagging substrates, and we demonstrate the utility of representative amine linkers for efficient C-terminal biotinylation of nanobodies.
Collapse
Affiliation(s)
- Tetiana Bondarchuk
- Enamine Ltd. (www.enamine.net), Winston Churchill Street 78, Kyiv 02094, Ukraine
| | - Diana Vaskiv
- Enamine Ltd. (www.enamine.net), Winston Churchill Street 78, Kyiv 02094, Ukraine
| | - Elena Zhuravel
- Enamine Ltd. (www.enamine.net), Winston Churchill Street 78, Kyiv 02094, Ukraine
| | - Oleh Shyshlyk
- Enamine Ltd. (www.enamine.net), Winston Churchill Street 78, Kyiv 02094, Ukraine
- V. P. Kukhar Institute of Bioorganic Chemistry and Petrochemistry, Academician Kukhar Street 1, Kyiv 02094, Ukraine
| | - Yevhenii Hrynyshyn
- Enamine Ltd. (www.enamine.net), Winston Churchill Street 78, Kyiv 02094, Ukraine
| | - Oleksandr Nedialko
- Enamine Ltd. (www.enamine.net), Winston Churchill Street 78, Kyiv 02094, Ukraine
- V. N. Karazin Kharkiv National University, 4 Svobody Square, Kharkiv 61022, Ukraine
| | - Oleksandr Pokholenko
- Enamine Ltd. (www.enamine.net), Winston Churchill Street 78, Kyiv 02094, Ukraine
- Department of Chemistry, Taras Shevchenko National University of Kyiv, Volodymyrska Street 64, Kyiv 01033, Ukraine
| | - Alla Pohribna
- Enamine Ltd. (www.enamine.net), Winston Churchill Street 78, Kyiv 02094, Ukraine
| | - Olga Kuchuk
- Enamine Ltd. (www.enamine.net), Winston Churchill Street 78, Kyiv 02094, Ukraine
| | - Volodymyr Brovarets
- V. P. Kukhar Institute of Bioorganic Chemistry and Petrochemistry, Academician Kukhar Street 1, Kyiv 02094, Ukraine
| | - Sergey Zozulya
- Enamine Ltd. (www.enamine.net), Winston Churchill Street 78, Kyiv 02094, Ukraine
| |
Collapse
|
6
|
Liu Y, Lu Z, Wu P, Liang Z, Yu Z, Ni K, Ma L. The Transpeptidase Sortase A Binds Nucleic Acids and Mediates Mammalian Cell Labeling. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2305605. [PMID: 38581131 PMCID: PMC11151058 DOI: 10.1002/advs.202305605] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/11/2023] [Revised: 03/10/2024] [Indexed: 04/08/2024]
Abstract
Wild-type sortase A is an important virulence factor displaying a diverse array of proteins on the surface of bacteria. This protein display relies on the transpeptidase activity of sortase A, which is widely engineered to allow protein ligation and protein engineering based on the interaction between sortase A and peptides. Here an unknown interaction is found between sortase A from Staphylococcus aureus and nucleic acids, in which exogenously expressed engineered sortase A binds oligonucleotides in vitro and is independent of its canonical transpeptidase activity. When incubated with mammalian cells, engineered sortase A further mediates oligonucleotide labeling to the cell surface, where sortase A attaches itself and is part of the labeled moiety. The labeling reaction can also be mediated by many classes of wild-type sortases as well. Cell surface GAG appears involved in sortase-mediated oligonucleotide cell labeling, as demonstrated by CRISPR screening. This interaction property is utilized to develop a technique called CellID to facilitate sample multiplexing for scRNA-seq and shows the potential of using sortases to label cells with diverse oligonucleotides. Together, the binding between sortase A and nucleic acids opens a new avenue to understanding the virulence of wild-type sortases and exploring the application of sortases in biotechnology.
Collapse
Affiliation(s)
- Yingzheng Liu
- College of Life SciencesZhejiang UniversityHangzhou310058China
- Westlake Laboratory of Life Sciences and Biomedicine18 Shilongshan RoadHangzhou310024China
- School of Life SciencesWestlake University600 Dunyu RoadHangzhou310030China
- Institute of BiologyWestlake Institute for Advanced Study18 Shilongshan RoadHangzhou310024China
| | - Zhike Lu
- College of Life SciencesZhejiang UniversityHangzhou310058China
- Westlake Laboratory of Life Sciences and Biomedicine18 Shilongshan RoadHangzhou310024China
- School of Life SciencesWestlake University600 Dunyu RoadHangzhou310030China
- Institute of BiologyWestlake Institute for Advanced Study18 Shilongshan RoadHangzhou310024China
| | - Panfeng Wu
- Westlake Laboratory of Life Sciences and Biomedicine18 Shilongshan RoadHangzhou310024China
- School of Life SciencesWestlake University600 Dunyu RoadHangzhou310030China
- Institute of BiologyWestlake Institute for Advanced Study18 Shilongshan RoadHangzhou310024China
| | - Zhaohui Liang
- AIdit Therapeutics1 Yunmeng Road, Building 1Hangzhou310024China
| | - Zhenxing Yu
- Westlake Laboratory of Life Sciences and Biomedicine18 Shilongshan RoadHangzhou310024China
- School of Life SciencesWestlake University600 Dunyu RoadHangzhou310030China
- Institute of BiologyWestlake Institute for Advanced Study18 Shilongshan RoadHangzhou310024China
| | - Ke Ni
- Westlake Laboratory of Life Sciences and Biomedicine18 Shilongshan RoadHangzhou310024China
- School of Life SciencesWestlake University600 Dunyu RoadHangzhou310030China
- Institute of BiologyWestlake Institute for Advanced Study18 Shilongshan RoadHangzhou310024China
- AIdit Therapeutics1 Yunmeng Road, Building 1Hangzhou310024China
| | - Lijia Ma
- Westlake Laboratory of Life Sciences and Biomedicine18 Shilongshan RoadHangzhou310024China
- School of Life SciencesWestlake University600 Dunyu RoadHangzhou310030China
- Institute of BiologyWestlake Institute for Advanced Study18 Shilongshan RoadHangzhou310024China
| |
Collapse
|
7
|
Zhou Y, Durek T, Craik DJ, Rehm FBH. Sortase-Catalyzed Protein Domain Inversion. Angew Chem Int Ed Engl 2024; 63:e202316777. [PMID: 38366985 DOI: 10.1002/anie.202316777] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2023] [Revised: 02/11/2024] [Accepted: 02/13/2024] [Indexed: 02/19/2024]
Abstract
Topological transformations and permutations of proteins have attracted significant interest as strategies to generate new protein functionalities or stability. These efforts have mainly been inspired by naturally occurring post-translational modifications, such as head-to-tail cyclization, circular permutation, or lasso-like entanglement. Such approaches can be realized experimentally via genetic encoding, in the case of circular permutation, or via enzymatic processing, in the case of cyclization. Notably, these previously described strategies leave the polypeptide backbone orientation unaltered. Here we describe an unnatural protein permutation, the protein domain inversion, whereby a C-terminal portion of a protein is enzymatically inverted from the canonical N-to-C to a C-to-C configuration with respect to the N-terminal part of the protein. The closest conceptually analogous biological process is perhaps the inversion of DNA segments as catalyzed by recombinases. We achieve these inversions using an engineered sortase A, a widely used transpeptidase. Our reactions proceed efficiently under mild conditions at 4-25 °C and are compatible with entirely heterologously-produced protein substrates.
Collapse
Affiliation(s)
- Yan Zhou
- ARC Centre of Excellence for Innovations in Peptide and Protein Science, Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD, 4072, Australia
| | - Thomas Durek
- ARC Centre of Excellence for Innovations in Peptide and Protein Science, Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD, 4072, Australia
| | - David J Craik
- ARC Centre of Excellence for Innovations in Peptide and Protein Science, Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD, 4072, Australia
| | - Fabian B H Rehm
- ARC Centre of Excellence for Innovations in Peptide and Protein Science, Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD, 4072, Australia
| |
Collapse
|
8
|
Zou Z, Ji Y, Schwaneberg U. Empowering Site-Specific Bioconjugations In Vitro and In Vivo: Advances in Sortase Engineering and Sortase-Mediated Ligation. Angew Chem Int Ed Engl 2024; 63:e202310910. [PMID: 38081121 DOI: 10.1002/anie.202310910] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Indexed: 12/23/2023]
Abstract
Sortase-mediated ligation (SML) has emerged as a powerful and versatile methodology for site-specific protein conjugation, functionalization/labeling, immobilization, and design of biohybrid molecules and systems. However, the broader application of SML faces several challenges, such as limited activity and stability, dependence on calcium ions, and reversible reactions caused by nucleophilic side-products. Over the past decade, protein engineering campaigns and particularly directed evolution, have been extensively employed to overcome sortase limitations, thereby expanding the potential application of SML in multiple directions, including therapeutics, biorthogonal chemistry, biomaterials, and biosensors. This review provides an overview of achieved advancements in sortase engineering and highlights recent progress in utilizing SML in combination with other state-of-the-art chemical and biological methodologies. The aim is to encourage scientists to employ sortases in their conjugation experiments.
Collapse
Affiliation(s)
- Zhi Zou
- DWI - Leibniz-Institute for Interactive Materials, Forckenbeckstraβe 50, 52074, Aachen, Germany
- RWTH Aachen University, Institute of Biotechnology, Worringerweg 3, 52074, Aachen, Germany
| | - Yu Ji
- RWTH Aachen University, Institute of Biotechnology, Worringerweg 3, 52074, Aachen, Germany
| | - Ulrich Schwaneberg
- DWI - Leibniz-Institute for Interactive Materials, Forckenbeckstraβe 50, 52074, Aachen, Germany
- RWTH Aachen University, Institute of Biotechnology, Worringerweg 3, 52074, Aachen, Germany
| |
Collapse
|
9
|
Wu J, Chu T, Hao J, Lin L. SpSrtA-Catalyzed Isopeptide Ligation on Lysine Residues. Microorganisms 2024; 12:179. [PMID: 38258005 PMCID: PMC10818881 DOI: 10.3390/microorganisms12010179] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2023] [Revised: 01/09/2024] [Accepted: 01/10/2024] [Indexed: 01/24/2024] Open
Abstract
Sortase-mediated ligation (SML) is widely used for protein bioconjugation. However, the sortase used in this strategy typically recognizes only the N-terminal oligoglycine, which is absent in most natural proteins. To broaden the spectrum of substrates compatible with SML, we focus on a novel sortase, sortase A from Streptococcus pneumoniae (SpSrtA), known for its expanded substrate specificity (N-terminal glycine, alanine, and serine). We present the first evidence showing that the reported SpSrtA mutant (SpSrtA*) can modify lysine residues in itself and other proteins. The modification sites of SpSrtA* were identified through LC-MS/MS analysis. Moreover, we discovered an optimal lysine-containing peptide tag by fusing it onto sfGFP, resulting in a labeling efficiency of 57%. Inspired by this, we applied the method to modify proteins on microorganism surfaces up to 13.5-fold. To enhance labeling efficiency, we fused the SpSrtA* onto a surface protein and achieved a 2.64-fold improvement. We further developed a high-throughput yeast display screening method for the directed evolution of SpSrtA*, achieving a 10-fold improvement in the labeling efficiency of this surface protein. Our study provides a novel strategy for modifying the lysine residues that will be a powerful addition to the protein bioconjugation toolbox.
Collapse
Affiliation(s)
- Jiajia Wu
- Department of Chemistry, Shanghai University, Shanghai 200444, China
- State Key Laboratory of Chemical Biology, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai 200032, China
| | - Tianyu Chu
- College of Chemistry and Molecular Engineering, Peking University, Beijing 100871, China
| | - Jian Hao
- Department of Chemistry, Shanghai University, Shanghai 200444, China
| | - Liang Lin
- State Key Laboratory of Chemical Biology, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai 200032, China
| |
Collapse
|
10
|
Obeng EM, Fulcher AJ, Wagstaff KM. Harnessing sortase A transpeptidation for advanced targeted therapeutics and vaccine engineering. Biotechnol Adv 2023; 64:108108. [PMID: 36740026 DOI: 10.1016/j.biotechadv.2023.108108] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2022] [Revised: 01/29/2023] [Accepted: 01/31/2023] [Indexed: 02/05/2023]
Abstract
The engineering of potent prophylactic and therapeutic complexes has always required careful protein modification techniques with seamless capabilities. In this light, methods that favor unobstructed multivalent targeting and correct antigen presentations remain essential and very demanding. Sortase A (SrtA) transpeptidation has exhibited these attributes in various settings over the years. However, its applications for engineering avidity-inspired therapeutics and potent vaccines have yet to be significantly noticed, especially in this era where active targeting and multivalent nanomedications are in great demand. This review briefly presents the SrtA enzyme and its associated transpeptidation activity and describes interesting sortase-mediated protein engineering and chemistry approaches for achieving multivalent therapeutic and antigenic responses. The review further highlights advanced applications in targeted delivery systems, multivalent therapeutics, adoptive cellular therapy, and vaccine engineering. These innovations show the potential of sortase-mediated techniques in facilitating the development of simple plug-and-play nanomedicine technologies against recalcitrant diseases and pandemics such as cancer and viral infections.
Collapse
Affiliation(s)
- Eugene M Obeng
- Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, VIC 3800, Australia.
| | - Alex J Fulcher
- Monash Micro Imaging, Monash University, Clayton, VIC 3800, Australia
| | - Kylie M Wagstaff
- Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, VIC 3800, Australia.
| |
Collapse
|
11
|
Dunleavy R, Chandrasekaran S, Crane BR. Enzymatic Spin-Labeling of Protein N- and C-Termini for Electron Paramagnetic Resonance Spectroscopy. Bioconjug Chem 2023:10.1021/acs.bioconjchem.3c00029. [PMID: 36921260 PMCID: PMC10502183 DOI: 10.1021/acs.bioconjchem.3c00029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/17/2023]
Abstract
Electron paramagnetic resonance (EPR) spectroscopy is a powerful tool for investigating the structure and dynamics of proteins. The introduction of paramagnetic moieties at specific positions in a protein enables precise measurement of local structure and dynamics. This technique, termed site-directed spin-labeling, has traditionally been performed using cysteine-reactive radical-containing probes. However, large proteins are more likely to contain multiple cysteine residues and cysteine labeling at specific sites may be infeasible or impede function. To address this concern, we applied three peptide-ligating enzymes (sortase, asparaginyl endopeptidase, and inteins) for nitroxide labeling of N- and C-termini of select monomeric and dimeric proteins. Continuous wave and pulsed EPR (double electron electron resonance) experiments reveal specific attachment of nitroxide probes to ether N-termini (OaAEP1) or C-termini (sortase and intein) across three test proteins (CheY, CheA, and iLOV), thereby enabling a straightforward, highly specific, and general method for protein labeling. Importantly, the linker length (3, 5, and 9 residues for OaAEP1, intein, and sortase reactions, respectively) between the probe and the target protein has a large impact on the utility of distance measurements by pulsed EPR, with longer linkers leading to broader distributions. As these methods are only dependent on accessible N- and C-termini, we anticipate application to a wide range of protein targets for biomolecular EPR spectroscopy.
Collapse
Affiliation(s)
- Robert Dunleavy
- Department of Chemistry and Chemical Biology, Cornell University, Ithaca, NY 14853, USA
| | | | - Brian R. Crane
- Department of Chemistry and Chemical Biology, Cornell University, Ithaca, NY 14853, USA
| |
Collapse
|
12
|
Apostolos AJ, Kelly JJ, Ongwae GM, Pires MM. Structure Activity Relationship of the Stem Peptide in Sortase A Mediated Ligation from Staphylococcus aureus. Chembiochem 2022; 23:e202200412. [PMID: 36018606 PMCID: PMC9632411 DOI: 10.1002/cbic.202200412] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2022] [Revised: 08/24/2022] [Indexed: 01/11/2023]
Abstract
The surfaces of most Gram-positive bacterial cells, including that of Staphylococcus aureus (S. aureus), are heavily decorated with proteins that coordinate cellular interactions with the extracellular space. In S. aureus, sortase A is the principal enzyme responsible for covalently anchoring proteins, which display the sorting signal LPXTG, onto the peptidoglycan (PG) matrix. Considerable efforts have been made to understand the role of this signal peptide in the sortase-mediated reaction. In contrast, much less is known about how the primary structure of the other substrate involved in the reaction (PG stem peptide) could impact sortase activity. To assess the sortase activity, a library of synthetic analogs of the stem peptide that mimic naturally existing variations found in the S. aureus PG primary sequence were evaluated. Using a combination of two unique assays, we showed that there is broad tolerability of substrate variations that are effectively processed by sortase A. While some of these stem peptide derivatives are naturally found in mature PG, they are not known to be present in the PG precursor, lipid II. These results suggest that sortase A could process both lipid II and mature PG as acyl-acceptor strands that might reside near the membrane, which has not been previously described.
Collapse
Affiliation(s)
| | - Joey J. Kelly
- Department of ChemistryUniversity of VirginiaCharlottesville, VA22904USA
| | - George M. Ongwae
- Department of ChemistryUniversity of VirginiaCharlottesville, VA22904USA
| | - Marcos M. Pires
- Department of ChemistryUniversity of VirginiaCharlottesville, VA22904USA
| |
Collapse
|
13
|
Li Y, Champion JA. Self-assembling nanocarriers from engineered proteins: Design, functionalization, and application for drug delivery. Adv Drug Deliv Rev 2022; 189:114462. [PMID: 35934126 DOI: 10.1016/j.addr.2022.114462] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Revised: 06/09/2022] [Accepted: 07/15/2022] [Indexed: 01/24/2023]
Abstract
Self-assembling proteins are valuable building blocks for constructing drug nanocarriers due to their self-assembly behavior, monodispersity, biocompatibility, and biodegradability. Genetic and chemical modifications allow for modular design of protein nanocarriers with effective drug encapsulation, targetability, stimuli responsiveness, and in vivo half-life. Protein nanocarriers have been developed to deliver various therapeutic molecules including small molecules, proteins, and nucleic acids with proven in vitro and in vivo efficacy. This article reviews recent advances in protein nanocarriers that are not derived from natural protein nanostructures, such as protein cages or virus like particles. The protein nanocarriers described here are self-assembled from rationally or de novo designed recombinant proteins, as well as recombinant proteins complexed with other biomolecules, presenting properties that are unique from those of natural protein carriers. Design, functionalization, and therapeutic application of protein nanocarriers will be discussed.
Collapse
Affiliation(s)
- Yirui Li
- BioEngineering Program, Georgia Institute of Technology, United States
| | - Julie A Champion
- School of Chemical and Biomolecular Engineering, Georgia Institute of Technology, 950 Atlantic Drive NW, Atlanta, GA 30332, United States; BioEngineering Program, Georgia Institute of Technology, United States.
| |
Collapse
|
14
|
Yamaguchi S, Ikeda R, Umeda Y, Kosaka T, Yamahira S, Okamoto A. Chemoenzymatic labeling to visualize intercellular contacts using lipidated sortase A. Chembiochem 2022; 23:e202200474. [PMID: 35976800 DOI: 10.1002/cbic.202200474] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2022] [Indexed: 11/09/2022]
Abstract
Methods to label intercellular contact have attracted attention because of their potential in cell biological and medical applications for the analysis of intercellular communications. In this study, a simple and versatile method for chemoenzymatic labeling of intercellularly contacting cells is demonstrated using a cell-surface anchoring reagent of a poly(ethylene glycol)(PEG)-lipid conjugate. The surfaces of each cell in the cell pairs of interest were decorated with sortase A (SrtA) and triglycine peptide that were lipidated with PEG-lipid. In the mixture of the two cell populations, the triglycine-modified cells were enzymatically labeled with a fluorescent labeling reagent when in contact with SrtA-modified cells on a substrate. The selective labeling of the contacting cells was confirmed by confocal microscopy. The method is a promising tool for selective visualization of intercellularly contacting cells in cell mixtures for cell-cell communication analysis.
Collapse
Affiliation(s)
- Satoshi Yamaguchi
- The University of Tokyo: Tokyo Daigaku, Department of Chemistry and Biotechnology, 4-6-1 Komaba, Meguro-ku, 153-8904, Tokyo, JAPAN
| | - Ryosuke Ikeda
- The University of Tokyo: Tokyo Daigaku, Department of Chemistry and Biotechnology, JAPAN
| | - Yuki Umeda
- The University of Tokyo: Tokyo Daigaku, Department of Chemistry and Biotechnology, JAPAN
| | - Takahiro Kosaka
- The University of Tokyo: Tokyo Daigaku, Department of Chemistry and Biotechnology, JAPAN
| | - Shinya Yamahira
- St Luke's International University: Sei Roka Kokusai Daigaku, Center for Medical Sciences, JAPAN
| | - Akimitsu Okamoto
- The University of Tokyo: Tokyo Daigaku, Department of Chemistry and Biotechnology, JAPAN
| |
Collapse
|
15
|
Uchida K, Obayashi H, Minamihata K, Wakabayashi R, Goto M, Shimokawa N, Takagi M, Kamiya N. Artificial Palmitoylation of Proteins Controls the Lipid Domain-Selective Anchoring on Biomembranes and the Raft-Dependent Cellular Internalization. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2022; 38:9640-9648. [PMID: 35882009 DOI: 10.1021/acs.langmuir.2c01205] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/27/2023]
Abstract
Protein palmitoylation, a post-translational modification, is universally observed in eukaryotic cells. The localization of palmitoylated proteins to highly dynamic, sphingolipid- and cholesterol-rich microdomains (called lipid rafts) on the plasma membrane has been shown to play an important role in signal transduction in cells. However, this complex biological system is not yet completely understood. Here, we used a combined approach where an artificial lipidated protein was applied to biomimetic model membranes and plasma membranes in cells to illuminate chemical and physiological properties of the rafts. Using cell-sized giant unilamellar vesicles, we demonstrated the selective partitioning of enhanced green fluorescent protein modified with a C-terminal palmitoyl moiety (EGFP-Pal) into the liquid-ordered phase consisting of saturated phospholipids and cholesterol. Using Jurkat T cells treated with an immunostimulant (concanavalin A), we observed the vesicular transport of EGFP-Pal. Further cellular studies with the treatment of methyl β-cyclodextrin revealed the cholesterol-dependent internalization of EGFP-Pal, which can be explained by a raft-dependent, caveolae-mediated endocytic pathway. The present synthetic approach using artificial and natural membrane systems can be further extended to explore the potential utility of artificially lipidated proteins at biological and artificial interfaces.
Collapse
Affiliation(s)
- Kazuki Uchida
- Department of Applied Chemistry, Graduate School of Engineering, Kyushu University, 744 Motooka, Nishi-ku, Fukuoka 819-0395, Japan
| | - Hiroki Obayashi
- Department of Applied Chemistry, Graduate School of Engineering, Kyushu University, 744 Motooka, Nishi-ku, Fukuoka 819-0395, Japan
| | - Kosuke Minamihata
- Department of Applied Chemistry, Graduate School of Engineering, Kyushu University, 744 Motooka, Nishi-ku, Fukuoka 819-0395, Japan
| | - Rie Wakabayashi
- Department of Applied Chemistry, Graduate School of Engineering, Kyushu University, 744 Motooka, Nishi-ku, Fukuoka 819-0395, Japan
| | - Masahiro Goto
- Department of Applied Chemistry, Graduate School of Engineering, Kyushu University, 744 Motooka, Nishi-ku, Fukuoka 819-0395, Japan
- Division of Biotechnology, Center for Future Chemistry, Kyushu University, 744 Motooka, Nishi-ku, Fukuoka 819-0395, Japan
| | - Naofumi Shimokawa
- School of Materials Science, Japan Advanced Institute of Science and Technology, 1-1 Asahidai, Nomi, Ishikawa 923-1292, Japan
| | - Masahiro Takagi
- School of Materials Science, Japan Advanced Institute of Science and Technology, 1-1 Asahidai, Nomi, Ishikawa 923-1292, Japan
| | - Noriho Kamiya
- Department of Applied Chemistry, Graduate School of Engineering, Kyushu University, 744 Motooka, Nishi-ku, Fukuoka 819-0395, Japan
- Division of Biotechnology, Center for Future Chemistry, Kyushu University, 744 Motooka, Nishi-ku, Fukuoka 819-0395, Japan
| |
Collapse
|
16
|
He G, Lei H, Sun W, Gu J, Yu W, Zhang D, Chen H, Li Y, Qin M, Xue B, Wang W, Cao Y. Strong and Reversible Covalent Double Network Hydrogel Based on Force-Coupled Enzymatic Reactions. Angew Chem Int Ed Engl 2022; 61:e202201765. [PMID: 35419931 DOI: 10.1002/anie.202201765] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2022] [Indexed: 12/12/2022]
Abstract
Biological load-bearing tissues are strong, tough, and recoverable under periodic mechanical loads. However, such features have rarely been achieved simultaneously in the same synthetic hydrogels. Here, we use a force-coupled enzymatic reaction to tune a strong covalent peptide linkage to a reversible bond. Based on this concept we engineered double network hydrogels that combine high mechanical strength and reversible mechanical recovery in the same hydrogels. Specifically, we found that a peptide ligase, sortase A, can promote the proteolysis of peptides under force. The peptide bond can be re-ligated by the same enzyme in the absence of force. This allows the sacrificial network in the double-network hydrogels to be ruptured and rebuilt reversibly. Our results demonstrate a general approach for precisely controlling the mechanical and dynamic properties of hydrogels at the molecular level.
Collapse
Affiliation(s)
- Guangxiao He
- Collaborative Innovation Center of Advanced Microstructures, National Laboratory of Solid State Microstructure, Key Laboratory of Intelligent Optical Sensing and Manipulation, Ministry of Education, Department of Physics, Nanjing University, Nanjing, 210093, China.,Jinan Microecological Biomedicine Shandong Laboratory, Jinan, 250021, China.,School of Public Health and Management, Hubei University of Medicine, Shiyan, 442000, China
| | - Hai Lei
- Collaborative Innovation Center of Advanced Microstructures, National Laboratory of Solid State Microstructure, Key Laboratory of Intelligent Optical Sensing and Manipulation, Ministry of Education, Department of Physics, Nanjing University, Nanjing, 210093, China
| | - Wenxu Sun
- School of Public Health and Management, Hubei University of Medicine, Shiyan, 442000, China
| | - Jie Gu
- Collaborative Innovation Center of Advanced Microstructures, National Laboratory of Solid State Microstructure, Key Laboratory of Intelligent Optical Sensing and Manipulation, Ministry of Education, Department of Physics, Nanjing University, Nanjing, 210093, China
| | - Wenting Yu
- Collaborative Innovation Center of Advanced Microstructures, National Laboratory of Solid State Microstructure, Key Laboratory of Intelligent Optical Sensing and Manipulation, Ministry of Education, Department of Physics, Nanjing University, Nanjing, 210093, China
| | - Di Zhang
- Collaborative Innovation Center of Advanced Microstructures, National Laboratory of Solid State Microstructure, Key Laboratory of Intelligent Optical Sensing and Manipulation, Ministry of Education, Department of Physics, Nanjing University, Nanjing, 210093, China
| | - Huiyan Chen
- Collaborative Innovation Center of Advanced Microstructures, National Laboratory of Solid State Microstructure, Key Laboratory of Intelligent Optical Sensing and Manipulation, Ministry of Education, Department of Physics, Nanjing University, Nanjing, 210093, China
| | - Ying Li
- School of Science, Nantong University, Nantong, 226019, China
| | - Meng Qin
- Collaborative Innovation Center of Advanced Microstructures, National Laboratory of Solid State Microstructure, Key Laboratory of Intelligent Optical Sensing and Manipulation, Ministry of Education, Department of Physics, Nanjing University, Nanjing, 210093, China
| | - Bin Xue
- Collaborative Innovation Center of Advanced Microstructures, National Laboratory of Solid State Microstructure, Key Laboratory of Intelligent Optical Sensing and Manipulation, Ministry of Education, Department of Physics, Nanjing University, Nanjing, 210093, China
| | - Wei Wang
- Collaborative Innovation Center of Advanced Microstructures, National Laboratory of Solid State Microstructure, Key Laboratory of Intelligent Optical Sensing and Manipulation, Ministry of Education, Department of Physics, Nanjing University, Nanjing, 210093, China
| | - Yi Cao
- Collaborative Innovation Center of Advanced Microstructures, National Laboratory of Solid State Microstructure, Key Laboratory of Intelligent Optical Sensing and Manipulation, Ministry of Education, Department of Physics, Nanjing University, Nanjing, 210093, China.,Institute of Advanced Materials and Flexible Electronics (IAMFE), School of Chemistry and Materials Science, Nanjing University of Information Science & Technology, Nanjing, 210044, China.,Institute for Brain Sciences, Nanjing University, Nanjing, 210093, China
| |
Collapse
|
17
|
Negi S, Hamori M, Sato A, Shimizu K, Kawahara-Nakagawa Y, Manabe T, Shibata N, Kitagishi H, Mashimo M, Sugiura Y. Transpeptidation reaction mediated by ligand- and metal cofactor-substituted Sortase A from Staphylococcus aureus. BULLETIN OF THE CHEMICAL SOCIETY OF JAPAN 2022. [DOI: 10.1246/bcsj.20220098] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Affiliation(s)
- Shigeru Negi
- Faculty of Pharmaceutical Science, Doshisha Women's University, Koudo, Kyotanabe, Kyoto 610-0395
| | - Mami Hamori
- Faculty of Pharmaceutical Science, Doshisha Women's University, Koudo, Kyotanabe, Kyoto 610-0395
| | - Ayaka Sato
- Faculty of Pharmaceutical Science, Doshisha Women's University, Koudo, Kyotanabe, Kyoto 610-0395
| | - Kyoko Shimizu
- Faculty of Pharmaceutical Science, Doshisha Women's University, Koudo, Kyotanabe, Kyoto 610-0395
| | - Yuka Kawahara-Nakagawa
- Graduate School of Faculty of Science, University of Hyogo, 3-2-1 Kouto, Kamigori-cho, Ako-gun, Hyogo, 678-1297
| | - Takayuki Manabe
- Clinical Research Support Center, Asahikawa Medical University Hospital, 2-1-1-1 Midorigaoka Higashi, Asahikawa 078-8510
| | - Nobuhito Shibata
- Faculty of Pharmaceutical Science, Doshisha Women's University, Koudo, Kyotanabe, Kyoto 610-0395
| | - Hiroaki Kitagishi
- Department of Molecular Chemistry and Biochemistry, Faculty of Science and Engineering, Doshisha University, Kyotanabe, Kyoto 610-0321
| | - Masato Mashimo
- Faculty of Pharmaceutical Science, Doshisha Women's University, Koudo, Kyotanabe, Kyoto 610-0395
| | - Yukio Sugiura
- Faculty of Pharmaceutical Science, Doshisha Women's University, Koudo, Kyotanabe, Kyoto 610-0395
| |
Collapse
|
18
|
Morgan HE, Turnbull WB, Webb ME. Challenges in the use of sortase and other peptide ligases for site-specific protein modification. Chem Soc Rev 2022; 51:4121-4145. [PMID: 35510539 PMCID: PMC9126251 DOI: 10.1039/d0cs01148g] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Site-specific protein modification is a widely-used biochemical tool. However, there are many challenges associated with the development of protein modification techniques, in particular, achieving site-specificity, reaction efficiency and versatility. The engineering of peptide ligases and their substrates has been used to address these challenges. This review will focus on sortase, peptidyl asparaginyl ligases (PALs) and variants of subtilisin; detailing how their inherent specificity has been utilised for site-specific protein modification. The review will explore how the engineering of these enzymes and substrates has led to increased reaction efficiency mainly due to enhanced catalytic activity and reduction of reversibility. It will also describe how engineering peptide ligases to broaden their substrate scope is opening up new opportunities to expand the biochemical toolkit, particularly through the development of techniques to conjugate multiple substrates site-specifically onto a protein using orthogonal peptide ligases. We highlight chemical and biochemical strategies taken to optimise peptide and protein modification using peptide ligases.![]()
Collapse
Affiliation(s)
- Holly E Morgan
- School of Chemistry and Astbury Centre for Structural Molecular Biology, University of Leeds, Woodhouse Lane, Leeds, LS2 9JT, UK.
| | - W Bruce Turnbull
- School of Chemistry and Astbury Centre for Structural Molecular Biology, University of Leeds, Woodhouse Lane, Leeds, LS2 9JT, UK.
| | - Michael E Webb
- School of Chemistry and Astbury Centre for Structural Molecular Biology, University of Leeds, Woodhouse Lane, Leeds, LS2 9JT, UK.
| |
Collapse
|
19
|
He G, Lei H, Sun W, Gu J, Yu W, Zhang D, Chen H, Li Y, Qin M, Xue B, Wang W, Cao Y. Strong and Reversible Covalent Double Network Hydrogel Based on Force‐coupled Enzymatic Reactions. Angew Chem Int Ed Engl 2022. [DOI: 10.1002/ange.202201765] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Affiliation(s)
| | - Hai Lei
- Nanjing University Physics CHINA
| | - Wenxu Sun
- Nantong University School of Science CHINA
| | - Jie Gu
- Nanjing University Physics CHINA
| | | | - Di Zhang
- Nanjing University Physics CHINA
| | | | - Ying Li
- Nanjing University of Information Science and Technology School of Environmental Science and Engineering CHINA
| | - Meng Qin
- Nanjing University Physics CHINA
| | - Bin Xue
- Nanjing University Physics CHINA
| | - Wei Wang
- Nanjing University Physics CHINA
| | - Yi Cao
- Nanjing University Department of Physics 22 Hankou Road 210093 Nanjing CHINA
| |
Collapse
|
20
|
Rehm FBH, Tyler TJ, de Veer SJ, Craik DJ, Durek T. Enzymatic C-to-C Protein Ligation. Angew Chem Int Ed Engl 2022; 61:e202116672. [PMID: 35018698 PMCID: PMC9303898 DOI: 10.1002/anie.202116672] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2021] [Indexed: 01/11/2023]
Abstract
Transpeptidase-catalyzed protein and peptide modifications have been widely utilized for generating conjugates of interest for biological investigation or therapeutic applications. However, all known transpeptidases are constrained to ligating in the N-to-C orientation, limiting the scope of attainable products. Here, we report that an engineered asparaginyl ligase accepts diverse incoming nucleophile substrate mimetics, particularly when a means of selectively quenching the reactivity of byproducts released from the recognition sequence is employed. In addition to directly catalyzing formation of l-/d- or α-/β-amino acid junctions, we find C-terminal Leu-ethylenediamine (Leu-Eda) motifs to be bona fide mimetics of native N-terminal Gly-Leu sequences. Appending a C-terminal Leu-Eda to synthetic peptides or, via an intein-splicing approach, to recombinant proteins enables direct transpeptidase-catalyzed C-to-C ligations. This work significantly expands the synthetic scope of enzyme-catalyzed protein transpeptidation reactions.
Collapse
Affiliation(s)
- Fabian B. H. Rehm
- Institute for Molecular BioscienceAustralian Research Council Centre of Excellence for Innovations in Peptide and Protein ScienceThe University of QueenslandBrisbaneQLD 4072Australia
| | - Tristan J. Tyler
- Institute for Molecular BioscienceAustralian Research Council Centre of Excellence for Innovations in Peptide and Protein ScienceThe University of QueenslandBrisbaneQLD 4072Australia
| | - Simon J. de Veer
- Institute for Molecular BioscienceAustralian Research Council Centre of Excellence for Innovations in Peptide and Protein ScienceThe University of QueenslandBrisbaneQLD 4072Australia
| | - David J. Craik
- Institute for Molecular BioscienceAustralian Research Council Centre of Excellence for Innovations in Peptide and Protein ScienceThe University of QueenslandBrisbaneQLD 4072Australia
| | - Thomas Durek
- Institute for Molecular BioscienceAustralian Research Council Centre of Excellence for Innovations in Peptide and Protein ScienceThe University of QueenslandBrisbaneQLD 4072Australia
| |
Collapse
|
21
|
Hori K, Yoshimoto S, Yoshino T, Zako T, Hirao G, Fujita S, Nakamura C, Yamagishi A, Kamiya N. Recent advances in research on biointerfaces: From cell surfaces to artificial interfaces. J Biosci Bioeng 2022; 133:195-207. [PMID: 34998688 DOI: 10.1016/j.jbiosc.2021.12.004] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2021] [Revised: 11/16/2021] [Accepted: 12/06/2021] [Indexed: 12/16/2022]
Abstract
Biointerfaces are regions where biomolecules, cells, and organic materials are exposed to environmental media or come in contact with other biomaterials, cells, and inorganic/organic materials. In this review article, six research topics on biointerfaces are described to show examples of state-of-art research approaches. First, biointerface design of nanoparticles for molecular detection is described. Functionalized gold nanoparticles can be used for sensitive detection of various target molecules, including chemical compounds and biomolecules, such as DNA, proteins, cells, and viruses. Second, the interaction between bacterial cell surfaces and material surfaces, including the introduction of advances in analytical methods and theoretical calculations, are explained as well as their applications to bioprocesses. Third, bioconjugation technologies for localizing functional proteins at biointerfaces are introduced, in particular, by focusing the potential of enzymes as a catalytic tool for designing different types of bioconjugates that function at biointerfaces. Forth topics is focusing on lipid-protein interaction in cell membranes as natural biointerfaces. Examples of membrane lipid engineering are introduced, and it is mentioned how their compositional profiles affect membrane protein functions. Fifth topic is the physical method for molecular delivery across the biointerface being developed currently, such as highly efficient nanoinjection, electroporation, and nanoneedle devices, in which the key is how to perforate the cell membrane. Final topic is the chemical design of lipid- or polymer-based RNA delivery carriers and their behavior on the cell interface, which are currently attracting attention as RNA vaccine technologies targeting COVID-19. Finally, future directions of biointerface studies are presented.
Collapse
Affiliation(s)
- Katsutoshi Hori
- Department of Biomolecular Engineering, Graduate School of Engineering, Nagoya University, Furo-cho, Chikusa-ku, Nagoya, Aichi 464-8603, Japan.
| | - Shogo Yoshimoto
- Department of Biomolecular Engineering, Graduate School of Engineering, Nagoya University, Furo-cho, Chikusa-ku, Nagoya, Aichi 464-8603, Japan
| | - Tomoko Yoshino
- Division of Biotechnology and Life Science, Institute of Engineering, Tokyo University of Agriculture and Technology, 2-24-16, Naka-cho, Koganei, Tokyo 184-8588, Japan
| | - Tamotsu Zako
- Faculty of Science, Ehime University, 2-5 Bunkyo, Matsuyama, Ehime 790-8577, Japan
| | - Gen Hirao
- Faculty of Science, Ehime University, 2-5 Bunkyo, Matsuyama, Ehime 790-8577, Japan
| | - Satoshi Fujita
- Photo BIO-OIL, National Institute of Advanced Industrial Science and Technology, Suita, Osaka 565-0871, Japan; Department of Applied Physics, Osaka University, 2-1 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Chikashi Nakamura
- DAILAB, Cellular and Molecular Biotechnology Research Institute, National Institute of Advanced Industrial Science and Technology, Central 5 1-1-1 Higashi, Tsukuba, Ibaraki 305-8565, Japan; Department of Biotechnology and Life Science, Tokyo University of Agriculture and Technology, 2-24-16 Naka-cho, Koganei, Tokyo 184-8588, Japan
| | - Ayana Yamagishi
- DAILAB, Cellular and Molecular Biotechnology Research Institute, National Institute of Advanced Industrial Science and Technology, Central 5 1-1-1 Higashi, Tsukuba, Ibaraki 305-8565, Japan; Department of Biotechnology and Life Science, Tokyo University of Agriculture and Technology, 2-24-16 Naka-cho, Koganei, Tokyo 184-8588, Japan
| | - Noriho Kamiya
- Department of Applied Chemistry, Graduate School of Engineering, Kyushu University, 744 Motooka, Nishi-ku, Fukuoka 819-0395, Japan; Division of Biotechnology, Center for Future Chemistry, Kyushu University, 744 Motooka, Nishi-ku, Fukuoka 819-0395, Japan
| |
Collapse
|
22
|
Obeng EM, Dzuvor CKO, Danquah MK. Anti-SARS-CoV-1 and -2 nanobody engineering towards avidity-inspired therapeutics. NANO TODAY 2022; 42:101350. [PMID: 34840592 PMCID: PMC8608585 DOI: 10.1016/j.nantod.2021.101350] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Revised: 09/22/2021] [Accepted: 11/18/2021] [Indexed: 05/15/2023]
Abstract
In the past two decades, the emergence of coronavirus diseases has been dire distress on both continental and global fronts and has resulted in the search for potent treatment strategies. One crucial challenge in this search is the recurrent mutations in the causative virus spike protein, which lead to viral escape issues. Among the current promising therapeutic discoveries is the use of nanobodies and nanobody-like molecules. While these nanobodies have demonstrated high-affinity interaction with the virus, the unpredictable spike mutations have warranted the need for avidity-inspired therapeutics of potent inhibitors such as nanobodies. This article discusses novel approaches for the design of anti-SARS-CoV-1 and -2 nanobodies to facilitate advanced innovations in treatment technologies. It further discusses molecular interactions and suggests multivalent protein nanotechnology and chemistry approaches to translate mere molecular affinity into avidity.
Collapse
Affiliation(s)
- Eugene M Obeng
- Biomedicine Discovery Institute, Monash University, Clayton, VIC 3800, Australia
- Bioengineering Laboratory, Department of Chemical and Biological Engineering, Monash University, Clayton, VIC 3800, Australia
| | - Christian K O Dzuvor
- Bioengineering Laboratory, Department of Chemical and Biological Engineering, Monash University, Clayton, VIC 3800, Australia
| | - Michael K Danquah
- Department of Chemical Engineering, University of Tennessee, Chattanooga 615 McCallie Ave, Chattanooga, TN 37403, United States
| |
Collapse
|
23
|
Rehm FBH, Tyler TJ, de Veer SJ, Craik DJ, Durek T. Enzymatic C‐to‐C Protein Ligation. Angew Chem Int Ed Engl 2022. [DOI: 10.1002/ange.202116672] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Affiliation(s)
- Fabian B. H. Rehm
- The University of Queensland Institute for Molecular Bioscience Chemistry and Structural Biology AUSTRALIA
| | - Tristan J. Tyler
- The University of Queensland Institute for Molecular Bioscience Chemistry and Structural Biology AUSTRALIA
| | - Simon J. de Veer
- The University of Queensland Institute for Molecular Bioscience Chemistry and Structural Biology AUSTRALIA
| | - David J. Craik
- The University of Queensland Institute for Molecular Bioscience Chemistry and Structural Biology AUSTRALIA
| | - Thomas Durek
- The University of Queensland Institute for Molecular Bioscience 306 Carmody RdLvl 7 North 4072 Brisbane AUSTRALIA
| |
Collapse
|
24
|
Alberti D, Guarniero M, Maciola AK, Dotta E, Pasqual G. Engineering Ligand and Receptor Pairs with LIPSTIC to Track Cell-Cell Interactions. Curr Protoc 2021; 1:e311. [PMID: 34870906 PMCID: PMC7613713 DOI: 10.1002/cpz1.311] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Interactions between different cell types are critical for a plethora of biological processes, such as the immune response. We recently developed a novel technology, called LIPSTIC (labeling of immune partnership by SorTagging intercellular contacts), that allows for identifying cells undergoing specific interactions thanks to an enzymatic labeling reaction. Our work demonstrated the use of this technology to monitor interactions between immune cells, both in vitro and in vivo, by the genetic engineering of CD40 and CD40L, an essential costimulatory axis between antigen-presenting cells and T cells. Here we describe protocols to design novel LIPSTIC-engineered ligand and receptor pairs, clone constructs into retroviral expression vector, perform their initial validation, and use them to measure interactions ex vivo. This information will be useful to investigators interested in exploiting the LIPSTIC technology to track their favorite immune interaction. © 2021 The Authors. Current Protocols published by Wiley Periodicals LLC. Basic Protocol 1: Design of LIPSTIC-engineered ligand and receptor pairs Basic Protocol 2: Cloning of LIPSTIC-engineered ligand and receptor pairs Basic Protocol 3: Validation of LIPSTIC-engineered ligand and receptor pairs in 293T cells Basic Protocol 4: Measuring interaction with LIPSTIC in immune cells ex vivo.
Collapse
Affiliation(s)
- Dafne Alberti
- Laboratory of Synthetic Immunology, Department of Surgery, Oncology and Gastroenterology, University of Padova, Padova, Italy
| | - Michelle Guarniero
- Laboratory of Synthetic Immunology, Department of Surgery, Oncology and Gastroenterology, University of Padova, Padova, Italy
| | - Agnieszka K. Maciola
- Laboratory of Synthetic Immunology, Department of Surgery, Oncology and Gastroenterology, University of Padova, Padova, Italy
| | - Enrico Dotta
- Laboratory of Synthetic Immunology, Department of Surgery, Oncology and Gastroenterology, University of Padova, Padova, Italy
| | - Giulia Pasqual
- Laboratory of Synthetic Immunology, Department of Surgery, Oncology and Gastroenterology, University of Padova, Padova, Italy
- Veneto Institute of Oncology IOV-IRCCS, Padua, Italy
| |
Collapse
|
25
|
Kumari P, Bowmik S, Paul SK, Biswas B, Banerjee SK, Murty US, Ravichandiran V, Mohan U. Sortase A: A chemoenzymatic approach for the labeling of cell surfaces. Biotechnol Bioeng 2021; 118:4577-4589. [PMID: 34491580 DOI: 10.1002/bit.27935] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Revised: 06/20/2021] [Accepted: 08/27/2021] [Indexed: 01/31/2023]
Abstract
Sortase A, a transpeptidase enzyme is present in many Gram-positive bacteria and helps in the recruitment of the cell surface proteins. Over the last two decades, Sortase A has become an attractive tool for performing in vivo and in vitro ligations. Sortase A-mediated ligation has continuously been used for its specificity, robustness, and highly efficient nature. These properties make it a popular choice among protein engineers as well as researchers from different fields. In this review, we give an overview of Sortase A-mediated ligation of various molecules on the cell surfaces, which can have diverse applications in interdisciplinary fields.
Collapse
Affiliation(s)
- Poonam Kumari
- Department of Biotechnology, National Institute of Pharmaceutical Education & Research (NIPER), Guwahati, Assam, India
| | - Sujoy Bowmik
- Department of Biotechnology, National Institute of Pharmaceutical Education & Research (NIPER), Guwahati, Assam, India
| | - Sudipto Kumar Paul
- Department of Biotechnology, National Institute of Pharmaceutical Education & Research (NIPER), Guwahati, Assam, India
| | - Bidisha Biswas
- Department of Biotechnology, National Institute of Pharmaceutical Education & Research (NIPER), Guwahati, Assam, India
| | - Sanjay K Banerjee
- Department of Biotechnology, National Institute of Pharmaceutical Education & Research (NIPER), Guwahati, Assam, India
| | | | - Velayutham Ravichandiran
- Department of Medicinal Chemistry, National Institute of Pharmaceutical Education & Research (NIPER), Kolkata, West Bengal, India
| | - Utpal Mohan
- Department of Medicinal Chemistry, National Institute of Pharmaceutical Education & Research (NIPER), Kolkata, West Bengal, India
| |
Collapse
|
26
|
Tong Y, Lee M, Drenth J, Fraaije MW. Flavin-tag: A Facile Method for Site-Specific Labeling of Proteins with a Flavin Fluorophore. Bioconjug Chem 2021; 32:1559-1563. [PMID: 34304568 DOI: 10.1021/acs.bioconjchem.1c00306] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Site-specific protein labeling methods are highly valuable tools for research and applications. We present a new protein labeling method that allows covalent attachment of a chromo- and fluorogenic flavin (FMN) to any targeted protein using a short flavinylation peptide-tag. We show that this peptide can be as short as 7 residues and can be located at the N-terminus, C-terminus, or in internal regions of the target protein. Analogous to kinase-catalyzed phosphorylation, the flavin is covalently attached via a stable phosphothreonyl linkage. The site-specific covalent tethering of FMN is accomplished by using a bacterial flavin transferase. The covalent coupling of FMN was shown to work in Escherichia coli and Saccharomyces cerevisiae cells and could be performed in vitro, rendering the "Flavin-tag" method a powerful tool for the selective decoration of proteins with a biocompatible redox-active fluorescent chromophore.
Collapse
Affiliation(s)
- Yapei Tong
- Molecular Enzymology Group, University of Groningen, Nijenborgh 4, Groningen 9747AG, The Netherlands
| | - Misun Lee
- Molecular Enzymology Group, University of Groningen, Nijenborgh 4, Groningen 9747AG, The Netherlands
| | - Jeroen Drenth
- Molecular Enzymology Group, University of Groningen, Nijenborgh 4, Groningen 9747AG, The Netherlands
| | - Marco W Fraaije
- Molecular Enzymology Group, University of Groningen, Nijenborgh 4, Groningen 9747AG, The Netherlands
| |
Collapse
|
27
|
De Rosa L, Di Stasi R, Romanelli A, D’Andrea LD. Exploiting Protein N-Terminus for Site-Specific Bioconjugation. Molecules 2021; 26:3521. [PMID: 34207845 PMCID: PMC8228110 DOI: 10.3390/molecules26123521] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2021] [Revised: 06/07/2021] [Accepted: 06/07/2021] [Indexed: 11/29/2022] Open
Abstract
Although a plethora of chemistries have been developed to selectively decorate protein molecules, novel strategies continue to be reported with the final aim of improving selectivity and mildness of the reaction conditions, preserve protein integrity, and fulfill all the increasing requirements of the modern applications of protein conjugates. The targeting of the protein N-terminal alpha-amine group appears a convenient solution to the issue, emerging as a useful and unique reactive site universally present in each protein molecule. Herein, we provide an updated overview of the methodologies developed until today to afford the selective modification of proteins through the targeting of the N-terminal alpha-amine. Chemical and enzymatic strategies enabling the selective labeling of the protein N-terminal alpha-amine group are described.
Collapse
Affiliation(s)
- Lucia De Rosa
- Istituto di Biostrutture e Bioimmagini, CNR, Via Mezzocannone 16, 80134 Napoli, Italy; (L.D.R.); (R.D.S.)
| | - Rossella Di Stasi
- Istituto di Biostrutture e Bioimmagini, CNR, Via Mezzocannone 16, 80134 Napoli, Italy; (L.D.R.); (R.D.S.)
| | - Alessandra Romanelli
- Dipartimento di Scienze Farmaceutiche, Università Degli Studi di Milano, Via Venezian 21, 20133 Milano, Italy;
| | - Luca Domenico D’Andrea
- Istituto di Scienze e Tecnologie Chimiche “Giulio Natta”, CNR Via M. Bianco 9, 20131 Milano, Italy
| |
Collapse
|
28
|
Kordbacheh S, Kasko AM. Peptide and protein engineering by modification of backbone and sidechain functional groups. POLYM INT 2021. [DOI: 10.1002/pi.6208] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Affiliation(s)
- Shadi Kordbacheh
- Department of Bioengineering University of California Los Angeles CA USA
| | - Andrea M Kasko
- Department of Bioengineering University of California Los Angeles CA USA
- California Nanosystems Institute Los Angeles CA USA
| |
Collapse
|
29
|
Takahara M, Mochizuki S, Wakabayashi R, Minamihata K, Goto M, Sakurai K, Kamiya N. Extending the Half-Life of a Protein in Vivo by Enzymatic Labeling with Amphiphilic Lipopeptides. Bioconjug Chem 2021; 32:655-660. [PMID: 33689283 DOI: 10.1021/acs.bioconjchem.1c00027] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Synthesis of lipid-protein conjugates is one of the significant techniques in drug delivery systems of proteins; however, the intact conjugation of a lipid and protein is yet challenging due to the hydrophobicity of lipid molecules. In order to facilitate easy handling of the lipid moiety in conjugation, we have focused on a microbial transglutaminase (MTG) that can ligate specific lysine (K) and glutamine (Q) residues in lipopeptides and a protein of interest. As MTG substrates, monolipid- and dilipid-fused amphiphilic short lipopeptide substrates (lipid-G3S-RHK or lipid2-KG3S-RHK) were designed. These amphiphilic lipopeptides and a model protein (enhanced green fluorescent protein, EGFP) fused with LLQG (LQ-EGFP) were both water-soluble, and thus lipid-protein conjugates were efficiently obtained through the MTG reaction with a >80% conversion rate of LQ-EGFP even using cholesterol-G3S-RHK. In vitro cell adhesion and in vivo half-life stability of the successfully obtained lipid-protein conjugates were evaluated, showing that the monocholesterol-G3S-RHK modification of a protein gave the highest cell adhesion efficiency and longest half-life time by formation of a stable albumin/lipid-protein complex.
Collapse
Affiliation(s)
- Mari Takahara
- Department of Materials Science & Chemical Engineering, National Institute of Technology, Kitakyushu College, 5-20-1 Shii, Kokuraminamiku, Kitakyushu 802-0985, Japan
| | - Shinichi Mochizuki
- Department of Chemistry and Biochemistry, the University of Kitakyushu, 1-1 Hibikino, Wakamatsuku, Kitakyushu 808-0135, Japan
| | - Rie Wakabayashi
- Department of Applied Chemistry, Graduate School of Engineering, Kyushu University, 744 Motooka, Fukuoka 819-0395, Japan
| | - Kosuke Minamihata
- Department of Applied Chemistry, Graduate School of Engineering, Kyushu University, 744 Motooka, Fukuoka 819-0395, Japan
| | - Masahiro Goto
- Department of Applied Chemistry, Graduate School of Engineering, Kyushu University, 744 Motooka, Fukuoka 819-0395, Japan.,Division of Biotechnology, Center for Future Chemistry, Kyushu University, 744 Motooka, Fukuoka 819-0395, Japan
| | - Kazuo Sakurai
- Department of Chemistry and Biochemistry, the University of Kitakyushu, 1-1 Hibikino, Wakamatsuku, Kitakyushu 808-0135, Japan
| | - Noriho Kamiya
- Department of Applied Chemistry, Graduate School of Engineering, Kyushu University, 744 Motooka, Fukuoka 819-0395, Japan.,Division of Biotechnology, Center for Future Chemistry, Kyushu University, 744 Motooka, Fukuoka 819-0395, Japan
| |
Collapse
|
30
|
Lieser RM, Yur D, Sullivan MO, Chen W. Site-Specific Bioconjugation Approaches for Enhanced Delivery of Protein Therapeutics and Protein Drug Carriers. Bioconjug Chem 2020; 31:2272-2282. [DOI: 10.1021/acs.bioconjchem.0c00456] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Affiliation(s)
- Rachel M. Lieser
- Department of Chemical and Biomolecular Engineering, University of Delaware, 150 Academy Street, Newark, Delaware 19716, United States of America
| | - Daniel Yur
- Department of Chemical and Biomolecular Engineering, University of Delaware, 150 Academy Street, Newark, Delaware 19716, United States of America
| | - Millicent O. Sullivan
- Department of Chemical and Biomolecular Engineering, University of Delaware, 150 Academy Street, Newark, Delaware 19716, United States of America
| | - Wilfred Chen
- Department of Chemical and Biomolecular Engineering, University of Delaware, 150 Academy Street, Newark, Delaware 19716, United States of America
| |
Collapse
|
31
|
Hossain MS, Maller C, Dai Y, Nangia S, Mozhdehi D. Non-canonical lipoproteins with programmable assembly and architecture. Chem Commun (Camb) 2020; 56:10281-10284. [PMID: 32734969 DOI: 10.1039/d0cc03271a] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The substrate promiscuity of an acyltransferase is leveraged to synthesize artificial lipoproteins bearing a non-canonical PTM (ncPTM). The non-canonical functionality of these lipoproteins results in a distinctive hysteretic assembly-absent from the canonical lipoproteins-and is used to prepare hybrid multiblock materials with precise and programmable patterns of amphiphilicity. This study demonstrates the promise of expanding the repertoire of PTMs for the development of nanomaterials with a unique assembly and function.
Collapse
Affiliation(s)
- Md Shahadat Hossain
- Department of Chemistry, 1-014 Center for Science and Technology, Syracuse University, Syracuse, NY 13244, USA.
| | | | | | | | | |
Collapse
|
32
|
Sortase-A mediated chemoenzymatic lipidation of single-domain antibodies for cell membrane engineering. Eur J Pharm Biopharm 2020; 153:121-129. [DOI: 10.1016/j.ejpb.2020.05.019] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2020] [Revised: 05/11/2020] [Accepted: 05/19/2020] [Indexed: 12/24/2022]
|
33
|
Takahara M, Kamiya N. Synthetic Strategies for Artificial Lipidation of Functional Proteins. Chemistry 2020; 26:4645-4655. [DOI: 10.1002/chem.201904568] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2019] [Revised: 11/29/2019] [Indexed: 11/08/2022]
Affiliation(s)
- Mari Takahara
- Department of Materials Science & Chemical EngineeringNational Institute of TechnologyKitakyushu College 5-20-1 Shii Kokuraminamiku Kitakyushu 802-0985 Japan
| | - Noriho Kamiya
- Department of Applied ChemistryGraduate School of Engineering 744 Motooka Nishiku Fukuoka 819-0395 Japan
- Division of Biotechnology, Center for Future ChemistryKyushu University 744 Motooka Nishiku Fukuoka 819-0395 Japan
| |
Collapse
|
34
|
Scheibel DM, Hossain MS, Smith AL, Lynch CJ, Mozhdehi D. Post-Translational Modification Mimicry for Programmable Assembly of Elastin-Based Protein Polymers. ACS Macro Lett 2020; 9:371-376. [PMID: 35648543 DOI: 10.1021/acsmacrolett.0c00041] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Post-translational modification (PTM) of protein polymers is emerging as a powerful bioinspired strategy to create protein-based hybrid materials with molecularly encoded assembly and function for applications in nanobiotechnology and medicine. While these modifications can be accomplished by harnessing native biological machinery (i.e., enzymes), the evolutionarily programmed specificity of these enzymes (recognition of select substrates and the limited repertoire of ligation chemistries catalyzed by these enzymes) can limit the type and linkage of PTMs appended to proteins. One approach to overcome this limitation is to leverage advances in site-selective biomolecular modification to prepare synthetic mimics of naturally occurring PTMs that are absent in nature. As a proof of concept, we used scalable bio-orthogonal reactions to prepare synthetic mimics of lipidated proteins with tunable assembly and disassembly. Additionally, we demonstrated that our PTM mimicry regulates the stimuli-responsive phase behavior of intrinsically disordered biopolymers, modulates their self-assembly at the nanoscale, and can be used for programmable disassembly of these materials in acidic environments. Synthetic PTM mimicry opens a path to encode new assembly and disassembly capabilities into hybrid materials that cannot be produced via biosynthesis.
Collapse
Affiliation(s)
- Dieter M. Scheibel
- Department of Chemistry, 1-014 Center for Science and Technology, Syracuse University, Syracuse, New York 13244, United States
| | - Md. Shahadat Hossain
- Department of Chemistry, 1-014 Center for Science and Technology, Syracuse University, Syracuse, New York 13244, United States
| | - Amy L. Smith
- Department of Chemistry, 1-014 Center for Science and Technology, Syracuse University, Syracuse, New York 13244, United States
| | - Christopher J. Lynch
- Department of Chemistry, 1-014 Center for Science and Technology, Syracuse University, Syracuse, New York 13244, United States
| | - Davoud Mozhdehi
- Department of Chemistry, 1-014 Center for Science and Technology, Syracuse University, Syracuse, New York 13244, United States
| |
Collapse
|
35
|
Li J, Zhang Y, Soubias O, Khago D, Chao FA, Li Y, Shaw K, Byrd RA. Optimization of sortase A ligation for flexible engineering of complex protein systems. J Biol Chem 2020; 295:2664-2675. [PMID: 31974162 DOI: 10.1074/jbc.ra119.012039] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2019] [Revised: 01/22/2020] [Indexed: 01/06/2023] Open
Abstract
Engineering and bioconjugation of proteins is a critically valuable tool that can facilitate a wide range of biophysical and structural studies. The ability to orthogonally tag or label a domain within a multidomain protein may be complicated by undesirable side reactions to noninvolved domains. Furthermore, the advantages of segmental (or domain-specific) isotopic labeling for NMR, or deuteration for neutron scattering or diffraction, can be realized by an efficient ligation procedure. Common methods-expressed protein ligation, protein trans-splicing, and native chemical ligation-each have specific limitations. Here, we evaluated the use of different variants of Staphylococcus aureus sortase A for a range of ligation reactions and demonstrate that conditions can readily be optimized to yield high efficiency (i.e. completeness of ligation), ease of purification, and functionality in detergents. These properties may enable joining of single domains into multidomain proteins, lipidation to mimic posttranslational modifications, and formation of cyclic proteins to aid in the development of nanodisc membrane mimetics. We anticipate that the method for ligating separate domains into a single functional multidomain protein reported here may enable many applications in structural biology.
Collapse
Affiliation(s)
- Jess Li
- Structural Biophysics Laboratory, Center for Cancer Research, NCI, National Institutes of Health, Frederick, Maryland 21702-1201
| | - Yue Zhang
- Structural Biophysics Laboratory, Center for Cancer Research, NCI, National Institutes of Health, Frederick, Maryland 21702-1201
| | - Olivier Soubias
- Structural Biophysics Laboratory, Center for Cancer Research, NCI, National Institutes of Health, Frederick, Maryland 21702-1201
| | - Domarin Khago
- Structural Biophysics Laboratory, Center for Cancer Research, NCI, National Institutes of Health, Frederick, Maryland 21702-1201
| | - Fa-An Chao
- Structural Biophysics Laboratory, Center for Cancer Research, NCI, National Institutes of Health, Frederick, Maryland 21702-1201
| | - Yifei Li
- Structural Biophysics Laboratory, Center for Cancer Research, NCI, National Institutes of Health, Frederick, Maryland 21702-1201
| | - Katherine Shaw
- Structural Biophysics Laboratory, Center for Cancer Research, NCI, National Institutes of Health, Frederick, Maryland 21702-1201
| | - R Andrew Byrd
- Structural Biophysics Laboratory, Center for Cancer Research, NCI, National Institutes of Health, Frederick, Maryland 21702-1201.
| |
Collapse
|
36
|
Rehm FBH, Harmand TJ, Yap K, Durek T, Craik DJ, Ploegh HL. Site-Specific Sequential Protein Labeling Catalyzed by a Single Recombinant Ligase. J Am Chem Soc 2019; 141:17388-17393. [PMID: 31573802 DOI: 10.1021/jacs.9b09166] [Citation(s) in RCA: 55] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Protein ligases of defined substrate specificity are versatile tools for protein engineering. Upon completion of the reaction, the products of currently reported protein ligases contain the amino acid sequence that is recognized by that same ligase, resulting in repeated cycles of ligation and hydrolysis as competing reactions. Thus, previous efforts to sequentially label proteins at distinct positions required ligases of orthogonal specificity. A recombinant Oldenlandia affinis asparaginyl endopeptidase, OaAEP1, is promiscuous for incoming nucleophiles. This promiscuity enabled us to define a nucleophile composed of natural amino acids that is ligated efficiently to the substrate yet yields a product that is poorly recognized by OaAEP1. Proteins modified with an efficient recognition module could be readily modified to yield a defined product bearing a cleavage-resistant motif, whereas proteins containing this inferior recognition motif remained essentially unmodified. We demonstrate the versatility of the N- or C-terminal protein modifications obtainable with this approach and modify the N- and C-termini of a single substrate protein in a sequential, site-specific manner in excellent yield.
Collapse
Affiliation(s)
- Fabian B H Rehm
- Program in Cellular and Molecular Medicine , Boston Children's Hospital, Harvard Medical School , Boston , Massachussets 02115 , United States.,Institute for Molecular Bioscience , The University of Queensland , Brisbane , QLD 4072 , Australia
| | - Thibault J Harmand
- Program in Cellular and Molecular Medicine , Boston Children's Hospital, Harvard Medical School , Boston , Massachussets 02115 , United States
| | - Kuok Yap
- Institute for Molecular Bioscience , The University of Queensland , Brisbane , QLD 4072 , Australia
| | - Thomas Durek
- Institute for Molecular Bioscience , The University of Queensland , Brisbane , QLD 4072 , Australia
| | - David J Craik
- Institute for Molecular Bioscience , The University of Queensland , Brisbane , QLD 4072 , Australia
| | - Hidde L Ploegh
- Program in Cellular and Molecular Medicine , Boston Children's Hospital, Harvard Medical School , Boston , Massachussets 02115 , United States
| |
Collapse
|
37
|
Takahara M, Wakabayashi R, Fujimoto N, Minamihata K, Goto M, Kamiya N. Enzymatic Cell‐Surface Decoration with Proteins using Amphiphilic Lipid‐Fused Peptide Substrates. Chemistry 2019; 25:7315-7321. [DOI: 10.1002/chem.201900370] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2019] [Revised: 03/03/2019] [Indexed: 12/17/2022]
Affiliation(s)
- Mari Takahara
- Department of Materials Science & Chemical EngineeringNational Institute of Technology, Kitakyushu College 5-20-1 Shii, Kokuraminamiku Kitakyushu 802-0985 Japan
| | - Rie Wakabayashi
- Department of Applied ChemistryGraduate School of EngineeringKyushu University 744 Motooka, Nishiku Fukuoka 819-0395 Japan
| | - Naoki Fujimoto
- Department of Applied ChemistryGraduate School of EngineeringKyushu University 744 Motooka, Nishiku Fukuoka 819-0395 Japan
| | - Kosuke Minamihata
- Department of Applied ChemistryGraduate School of EngineeringKyushu University 744 Motooka, Nishiku Fukuoka 819-0395 Japan
| | - Masahiro Goto
- Department of Applied ChemistryGraduate School of EngineeringKyushu University 744 Motooka, Nishiku Fukuoka 819-0395 Japan
- Division of Biotechnology, Center for Future ChemistryKyushu University 744 Motooka, Nishiku Fukuoka 819-0395 Japan
| | - Noriho Kamiya
- Department of Applied ChemistryGraduate School of EngineeringKyushu University 744 Motooka, Nishiku Fukuoka 819-0395 Japan
- Division of Biotechnology, Center for Future ChemistryKyushu University 744 Motooka, Nishiku Fukuoka 819-0395 Japan
| |
Collapse
|
38
|
Dai X, Böker A, Glebe U. Broadening the scope of sortagging. RSC Adv 2019; 9:4700-4721. [PMID: 35514663 PMCID: PMC9060782 DOI: 10.1039/c8ra06705h] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2019] [Accepted: 01/31/2019] [Indexed: 01/20/2023] Open
Abstract
Sortases are enzymes occurring in the cell wall of Gram-positive bacteria. Sortase A (SrtA), the best studied sortase class, plays a key role in anchoring surface proteins with the recognition sequence LPXTG covalently to oligoglycine units of the bacterial cell wall. This unique transpeptidase activity renders SrtA attractive for various purposes and motivated researchers to study multiple in vivo and in vitro ligations in the last decades. This ligation technique is known as sortase-mediated ligation (SML) or sortagging and developed to a frequently used method in basic research. The advantages are manifold: extremely high substrate specificity, simple access to substrates and enzyme, robust nature and easy handling of sortase A. In addition to the ligation of two proteins or peptides, early studies already included at least one artificial (peptide equipped) substrate into sortagging reactions - which demonstrates the versatility and broad applicability of SML. Thus, SML is not only a biology-related technique, but has found prominence as a major interdisciplinary research tool. In this review, we provide an overview about the use of sortase A in interdisciplinary research, mainly for protein modification, synthesis of protein-polymer conjugates and immobilization of proteins on surfaces.
Collapse
Affiliation(s)
- Xiaolin Dai
- Fraunhofer Institute for Applied Polymer Research IAP Geiselbergstr. 69 14476 Potsdam-Golm Germany
- Lehrstuhl für Polymermaterialien und Polymertechnologie, Universität Potsdam 14476 Potsdam-Golm Germany
| | - Alexander Böker
- Fraunhofer Institute for Applied Polymer Research IAP Geiselbergstr. 69 14476 Potsdam-Golm Germany
- Lehrstuhl für Polymermaterialien und Polymertechnologie, Universität Potsdam 14476 Potsdam-Golm Germany
| | - Ulrich Glebe
- Fraunhofer Institute for Applied Polymer Research IAP Geiselbergstr. 69 14476 Potsdam-Golm Germany
| |
Collapse
|
39
|
Wöll S, Bachran C, Schiller S, Schröder M, Conrad L, Scherließ R, Swee LK. Sortagging of liposomes with a murine CD11b-specific VHH increases in vitro and in vivo targeting specificity of myeloid cells. Eur J Pharm Biopharm 2019; 134:190-198. [DOI: 10.1016/j.ejpb.2018.11.014] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2018] [Revised: 11/15/2018] [Accepted: 11/16/2018] [Indexed: 10/27/2022]
|
40
|
Zhang Y, Park KY, Suazo KF, Distefano MD. Recent progress in enzymatic protein labelling techniques and their applications. Chem Soc Rev 2018; 47:9106-9136. [PMID: 30259933 PMCID: PMC6289631 DOI: 10.1039/c8cs00537k] [Citation(s) in RCA: 174] [Impact Index Per Article: 24.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Protein-based conjugates are valuable constructs for a variety of applications. Conjugation of proteins to fluorophores is commonly used to study their cellular localization and the protein-protein interactions. Modification of therapeutic proteins with either polymers or cytotoxic moieties greatly enhances their pharmacokinetics or potency. To label a protein of interest, conventional direct chemical reaction with the side-chains of native amino acids often yields heterogeneously modified products. This renders their characterization complicated, requires difficult separation steps and may impact protein function. Although modification can also be achieved via the insertion of unnatural amino acids bearing bioorthogonal functional groups, these methods can have lower protein expression yields, limiting large scale production. As a site-specific modification method, enzymatic protein labelling is highly efficient and robust under mild reaction conditions. Significant progress has been made over the last five years in modifying proteins using enzymatic methods for numerous applications, including the creation of clinically relevant conjugates with polymers, cytotoxins or imaging agents, fluorescent or affinity probes to study complex protein interaction networks, and protein-linked materials for biosensing. This review summarizes developments in enzymatic protein labelling over the last five years for a panel of ten enzymes, including sortase A, subtiligase, microbial transglutaminase, farnesyltransferase, N-myristoyltransferase, phosphopantetheinyl transferases, tubulin tyrosin ligase, lipoic acid ligase, biotin ligase and formylglycine generating enzyme.
Collapse
Affiliation(s)
- Yi Zhang
- Department of Chemistry, University of Minnesota, Minneapolis, Minnesota 55455, USA.
| | | | | | | |
Collapse
|
41
|
Sortaggable liposomes: Evaluation of reaction conditions for single-domain antibody conjugation by Sortase-A and targeting of CD11b+ myeloid cells. Eur J Pharm Biopharm 2018; 133:138-150. [DOI: 10.1016/j.ejpb.2018.09.017] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2018] [Revised: 08/31/2018] [Accepted: 09/22/2018] [Indexed: 10/28/2022]
|
42
|
Takahara M, Wakabayashi R, Minamihata K, Goto M, Kamiya N. Design of Lipid–Protein Conjugates Using Amphiphilic Peptide Substrates of Microbial Transglutaminase. ACS APPLIED BIO MATERIALS 2018; 1:1823-1829. [DOI: 10.1021/acsabm.8b00271] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Affiliation(s)
- Mari Takahara
- Department of Materials Science & Chemical Engineering, National Institute of Technology, Kitakyushu College, 5-20-1 Shii, Kokuraminamiku, Kitakyushu, Fukuoka, Japan
| | | | | | | | | |
Collapse
|
43
|
Harmand TJ, Bousbaine D, Chan A, Zhang X, Liu DR, Tam JP, Ploegh HL. One-Pot Dual Labeling of IgG 1 and Preparation of C-to-C Fusion Proteins Through a Combination of Sortase A and Butelase 1. Bioconjug Chem 2018; 29:3245-3249. [PMID: 30231608 DOI: 10.1021/acs.bioconjchem.8b00563] [Citation(s) in RCA: 61] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Site-specific chemical modification of proteins can assist in the study of their function. Furthermore, these methods are essential to develop biologicals for diagnostic and therapeutic use. Standard protein engineering protocols and recombinant expression enable the production of proteins with short peptide tags recognized by enzymes capable of site-specific modification. We report here the application of two enzymes of orthogonal specificity, sortase A and butelase 1, to prepare non-natural C-to-C fusion proteins. Using these enzymes, we further demonstrate site-selective installation of different chemical moieties at two sites in a full-size antibody molecule.
Collapse
Affiliation(s)
- Thibault J Harmand
- Program in Cellular and Molecular Medicine , Boston Children's Hospital, Harvard Medical School , Boston , Massachussets 02115 , United States
| | - Djenet Bousbaine
- Program in Cellular and Molecular Medicine , Boston Children's Hospital, Harvard Medical School , Boston , Massachussets 02115 , United States.,Department of Microbiology , Massachusetts Institute of Technology , Cambridge , Massachussets 02139 , United States
| | - Alix Chan
- The Broad Institute of Harvard and MIT, Howard Hughes Medical Institute, and the Department of Chemistry and Chemical Biology , Harvard University , Cambridge , Massachussets 02142 , United States
| | - Xiaohong Zhang
- School of Biological Sciences , Nanyang Technological University , 637551 , Singapore
| | - David R Liu
- The Broad Institute of Harvard and MIT, Howard Hughes Medical Institute, and the Department of Chemistry and Chemical Biology , Harvard University , Cambridge , Massachussets 02142 , United States
| | - James P Tam
- School of Biological Sciences , Nanyang Technological University , 637551 , Singapore
| | - Hidde L Ploegh
- Program in Cellular and Molecular Medicine , Boston Children's Hospital, Harvard Medical School , Boston , Massachussets 02115 , United States
| |
Collapse
|
44
|
Pishesha N, Ingram JR, Ploegh HL. Sortase A: A Model for Transpeptidation and Its Biological Applications. Annu Rev Cell Dev Biol 2018; 34:163-188. [PMID: 30110557 DOI: 10.1146/annurev-cellbio-100617-062527] [Citation(s) in RCA: 89] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Molecular biologists and chemists alike have long sought to modify proteins with substituents that cannot be installed by standard or even advanced genetic approaches. We here describe the use of transpeptidases to achieve these goals. Living systems encode a variety of transpeptidases and peptide ligases that allow for the enzyme-catalyzed formation of peptide bonds, and protein engineers have used directed evolution to enhance these enzymes for biological applications. We focus primarily on the transpeptidase sortase A, which has become popular over the past few years for its ability to perform a remarkably wide variety of protein modifications, both in vitro and in living cells.
Collapse
Affiliation(s)
- Novalia Pishesha
- Program in Molecular and Cellular Medicine, Boston Children's Hospital, Boston, Massachusetts 02115, USA; .,Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, USA
| | - Jessica R Ingram
- Department of Cancer Immunology and Virology, Dana Farber Cancer Institute, Boston, Massachusetts 02215, USA
| | - Hidde L Ploegh
- Program in Molecular and Cellular Medicine, Boston Children's Hospital, Boston, Massachusetts 02115, USA;
| |
Collapse
|
45
|
McConnell SA, Amer BR, Muroski J, Fu J, Chang C, Ogorzalek Loo RR, Loo JA, Osipiuk J, Ton-That H, Clubb RT. Protein Labeling via a Specific Lysine-Isopeptide Bond Using the Pilin Polymerizing Sortase from Corynebacterium diphtheriae. J Am Chem Soc 2018; 140:8420-8423. [PMID: 29927249 PMCID: PMC6230430 DOI: 10.1021/jacs.8b05200] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Proteins that are site-specifically modified with peptides and chemicals can be used as novel therapeutics, imaging tools, diagnostic reagents and materials. However, there are few enzyme-catalyzed methods currently available to selectively conjugate peptides to internal sites within proteins. Here we show that a pilus-specific sortase enzyme from Corynebacterium diphtheriae (CdSrtA) can be used to attach a peptide to a protein via a specific lysine-isopeptide bond. Using rational mutagenesis we created CdSrtA3M, a highly activated cysteine transpeptidase that catalyzes in vitro isopeptide bond formation. CdSrtA3M mediates bioconjugation to a specific lysine residue within a fused domain derived from the corynebacterial SpaA protein. Peptide modification yields greater than >95% can be achieved. We demonstrate that CdSrtA3M can be used in concert with the Staphylococcus aureus SrtA enzyme, enabling dual, orthogonal protein labeling via lysine-isopeptide and backbone-peptide bonds.
Collapse
Affiliation(s)
- Scott A. McConnell
- Department of Chemistry and Biochemistry, UCLA-DOE Institute for Genomics and Proteomics and the Molecular Biology Institute, University of California, Los Angeles, CA, USA
| | - Brendan R. Amer
- Department of Chemistry and Biochemistry, UCLA-DOE Institute for Genomics and Proteomics and the Molecular Biology Institute, University of California, Los Angeles, CA, USA
| | - John Muroski
- Department of Chemistry and Biochemistry, UCLA-DOE Institute for Genomics and Proteomics and the Molecular Biology Institute, University of California, Los Angeles, CA, USA
| | - Janine Fu
- Department of Chemistry and Biochemistry, UCLA-DOE Institute for Genomics and Proteomics and the Molecular Biology Institute, University of California, Los Angeles, CA, USA
| | - Chungyu Chang
- Department of Microbiology & Molecular Genetics, University of Texas Health Science Center, Houston, TX, USA
| | - Rachel R. Ogorzalek Loo
- Department of Chemistry and Biochemistry, UCLA-DOE Institute for Genomics and Proteomics and the Molecular Biology Institute, University of California, Los Angeles, CA, USA
| | - Joseph A. Loo
- Department of Chemistry and Biochemistry, UCLA-DOE Institute for Genomics and Proteomics and the Molecular Biology Institute, University of California, Los Angeles, CA, USA
| | - Jerzy Osipiuk
- Structural Biology Center, Argonne National Laboratory, Argonne, IL, USA
| | - Hung Ton-That
- Department of Microbiology & Molecular Genetics, University of Texas Health Science Center, Houston, TX, USA
| | - Robert T. Clubb
- Department of Chemistry and Biochemistry, UCLA-DOE Institute for Genomics and Proteomics and the Molecular Biology Institute, University of California, Los Angeles, CA, USA
| |
Collapse
|
46
|
Dougan M, Ingram JR, Jeong HJ, Mosaheb MM, Bruck PT, Ali L, Pishesha N, Blomberg O, Tyler PM, Servos MM, Rashidian M, Nguyen QD, von Andrian UH, Ploegh HL, Dougan SK. Targeting Cytokine Therapy to the Pancreatic Tumor Microenvironment Using PD-L1-Specific VHHs. Cancer Immunol Res 2018; 6:389-401. [PMID: 29459478 PMCID: PMC6079513 DOI: 10.1158/2326-6066.cir-17-0495] [Citation(s) in RCA: 66] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2017] [Revised: 11/13/2017] [Accepted: 02/07/2018] [Indexed: 12/31/2022]
Abstract
Cytokine-based therapies for cancer have not achieved widespread clinical success because of inherent toxicities. Treatment for pancreatic cancer is limited by the dense stroma that surrounds tumors and by an immunosuppressive tumor microenvironment. To overcome these barriers, we developed constructs of single-domain antibodies (VHHs) against PD-L1 fused with IL-2 and IFNγ. Targeting cytokine delivery in this manner reduced pancreatic tumor burden by 50%, whereas cytokines fused to an irrelevant VHH, or blockade of PD-L1 alone, showed little effect. Targeted delivery of IL-2 increased the number of intratumoral CD8+ T cells, whereas IFNγ reduced the number of CD11b+ cells and skewed intratumoral macrophages toward the display of M1-like characteristics. Imaging of fluorescent VHH-IFNγ constructs, as well as transcriptional profiling, demonstrated targeting of IFNγ to the tumor microenvironment. Many tumors and tumor-infiltrating myeloid cells express PD-L1, rendering them potentially susceptible to this form of targeted immunotherapy. Cancer Immunol Res; 6(4); 389-401. ©2018 AACR.
Collapse
Affiliation(s)
- Michael Dougan
- Division of Gastroenterology, Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts
| | - Jessica R Ingram
- Department of Cancer Immunology and Virology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Hee-Jin Jeong
- Department of Cancer Immunology and Virology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Munir M Mosaheb
- Department of Microbiology and Immunobiology, Harvard Medical School, Boston, Massachusetts
| | - Patrick T Bruck
- Department of Cancer Immunology and Virology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Lestat Ali
- Department of Cancer Immunology and Virology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Novalia Pishesha
- Program in Cellular and Molecular Medicine, Children's Hospital Boston, Boston, Massachusetts
| | - Olga Blomberg
- Program in Cellular and Molecular Medicine, Children's Hospital Boston, Boston, Massachusetts
| | - Paul M Tyler
- Department of Cancer Immunology and Virology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Mariah M Servos
- Department of Cancer Immunology and Virology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Mohammad Rashidian
- Program in Cellular and Molecular Medicine, Children's Hospital Boston, Boston, Massachusetts
| | - Quang-De Nguyen
- Lurie Family Imaging Center, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Ulrich H von Andrian
- Department of Microbiology and Immunobiology, Harvard Medical School, Boston, Massachusetts
| | - Hidde L Ploegh
- Program in Cellular and Molecular Medicine, Children's Hospital Boston, Boston, Massachusetts
| | - Stephanie K Dougan
- Department of Cancer Immunology and Virology, Dana-Farber Cancer Institute, Boston, Massachusetts.
- Department of Microbiology and Immunobiology, Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
47
|
Sortase ligation enables homogeneous GPCR phosphorylation to reveal diversity in β-arrestin coupling. Proc Natl Acad Sci U S A 2018; 115:3834-3839. [PMID: 29581292 DOI: 10.1073/pnas.1722336115] [Citation(s) in RCA: 46] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
The ability of G protein-coupled receptors (GPCRs) to initiate complex cascades of cellular signaling is governed by the sequential coupling of three main transducer proteins, G protein, GPCR kinase (GRK), and β-arrestin. Mounting evidence indicates these transducers all have distinct conformational preferences and binding modes. However, interrogating each transducer's mechanism of interaction with GPCRs has been complicated by the interplay of transducer-mediated signaling events. For example, GRK-mediated receptor phosphorylation recruits and induces conformational changes in β-arrestin, which facilitates coupling to the GPCR transmembrane core. Here we compare the allosteric interactions of G proteins and β-arrestins with GPCRs' transmembrane cores by using the enzyme sortase to ligate a synthetic phosphorylated peptide onto the carboxyl terminus of three different receptors. Phosphopeptide ligation onto the β2-adrenergic receptor (β2AR) allows stabilization of a high-affinity receptor active state by β-arrestin1, permitting us to define elements in the β2AR and β-arrestin1 that contribute to the receptor transmembrane core interaction. Interestingly, ligation of the identical phosphopeptide onto the β2AR, the muscarinic acetylcholine receptor 2 and the μ-opioid receptor reveals that the ability of β-arrestin1 to enhance agonist binding relative to G protein differs substantially among receptors. Furthermore, strong allosteric coupling of β-arrestin1 correlates with its ability to attenuate, or "desensitize," G protein activation in vitro. Sortase ligation thus provides a versatile method to introduce complex, defined phosphorylation patterns into GPCRs, and analogous strategies could be applied to other classes of posttranslationally modified proteins. These homogeneously phosphorylated GPCRs provide an innovative means to systematically study receptor-transducer interactions.
Collapse
|
48
|
Sortase-Mediated Ligation of Purely Artificial Building Blocks. Polymers (Basel) 2018; 10:polym10020151. [PMID: 30966187 PMCID: PMC6414994 DOI: 10.3390/polym10020151] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2017] [Revised: 01/19/2018] [Accepted: 02/02/2018] [Indexed: 01/16/2023] Open
Abstract
Sortase A (SrtA) from Staphylococcus aureus has been often used for ligating a protein with other natural or synthetic compounds in recent years. Here we show that SrtA-mediated ligation (SML) is universally applicable for the linkage of two purely artificial building blocks. Silica nanoparticles (NPs), poly(ethylene glycol) and poly(N-isopropyl acrylamide) are chosen as synthetic building blocks. As a proof of concept, NP–polymer, NP–NP, and polymer–polymer structures are formed by SrtA catalysis. Therefore, the building blocks are equipped with the recognition sequence needed for SrtA reaction—the conserved peptide LPETG—and a pentaglycine motif. The successful formation of the reaction products is shown by means of transmission electron microscopy (TEM), matrix assisted laser desorption ionization-time of flight mass spectrometry (MALDI-ToF MS), and dynamic light scattering (DLS). The sortase catalyzed linkage of artificial building blocks sets the stage for the development of a new approach to link synthetic structures in cases where their synthesis by established chemical methods is complicated.
Collapse
|
49
|
Yang M, Hong H, Liu S, Zhao X, Wu Z. Immobilization of Staphylococcus aureus Sortase A on Chitosan Particles and Its Applications in Peptide-to-Peptide Ligation and Peptide Cyclization. Molecules 2018; 23:molecules23010192. [PMID: 29351256 PMCID: PMC6017383 DOI: 10.3390/molecules23010192] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2017] [Revised: 01/11/2018] [Accepted: 01/14/2018] [Indexed: 11/16/2022] Open
Abstract
Chitosan macro-particles prepared by the neutralization method were applied to Sortase A (SrtA) immobilization using glutaraldehyde as a crosslinking agent. The particles were characterized by Fourier transform infrared spectroscopy (FTIR) and scanning electron microscopy (SEM). Response surface methodology (RSM) was employed to optimize the immobilization process. An average specific activity of 3142 U (mg protein)-1 was obtained under optimized immobilization conditions (chitosan concentration 3%, SrtA concentration 0.5 mg·mL-1, glutaraldehyde concentration 0.5%, crosslinking and immobilization at 20 °C, crosslinking for 3 h, and an immobilization time of 8 h). The transpeptidase activity of immobilized SrtA was proved by a peptide-to-peptide ligation with a conversion yield approximately at 80%, and the immobilized catalyst was successfully reused for five cycles without obvious activity loss. Moreover, the scale-up capability of using immobilized SrtA to catalyze a head-to-tail peptide cyclization was investigated in a batch reaction and the conversion yield was more than 95% when using 20 mg of peptide as a substrate.
Collapse
Affiliation(s)
- Min Yang
- Key Laboratory of Carbohydrate Chemistry & Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University, Wuxi 214122, China.
| | - Haofei Hong
- Key Laboratory of Carbohydrate Chemistry & Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University, Wuxi 214122, China.
| | - Shaozhong Liu
- Key Laboratory of Carbohydrate Chemistry & Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University, Wuxi 214122, China.
| | - Xinrui Zhao
- Key Laboratory of Carbohydrate Chemistry & Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University, Wuxi 214122, China.
| | - Zhimeng Wu
- Key Laboratory of Carbohydrate Chemistry & Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University, Wuxi 214122, China.
| |
Collapse
|
50
|
Jeong HJ, Abhiraman GC, Story CM, Ingram JR, Dougan SK. Generation of Ca2+-independent sortase A mutants with enhanced activity for protein and cell surface labeling. PLoS One 2017; 12:e0189068. [PMID: 29200433 PMCID: PMC5714338 DOI: 10.1371/journal.pone.0189068] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2017] [Accepted: 11/17/2017] [Indexed: 11/22/2022] Open
Abstract
Sortase A, a calcium-dependent transpeptidase derived from Staphylococcus aureus, is used in a broad range of applications, such as the conjugation of fluorescent dyes and other moieties to proteins or to the surface of eukaryotic cells. In vivo and cell-based applications of sortase have been somewhat limited by the large range of calcium concentrations, as well as by the often transient nature of protein-protein interactions in living systems. In order to use sortase A for cell labeling applications, we generated a new sortase A variant by combining multiple mutations to yield an enzyme that was both calcium-independent and highly active. This variant has enhanced activity for both N- and C-terminal labeling, as well as for cell surface modification under physiological conditions.
Collapse
Affiliation(s)
- Hee-Jin Jeong
- Department of Cancer Immunology and Virology, Dana-Farber Cancer Institute, Boston, Massachusetts, United States of America
- Department of Microbiology and Immunobiology, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Gita C. Abhiraman
- Department of Cancer Immunology and Virology, Dana-Farber Cancer Institute, Boston, Massachusetts, United States of America
| | - Craig M. Story
- Department of Biology, Gordon College, Wenham, Massachusetts, United States of America
| | - Jessica R. Ingram
- Department of Cancer Immunology and Virology, Dana-Farber Cancer Institute, Boston, Massachusetts, United States of America
- Department of Microbiology and Immunobiology, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Stephanie K. Dougan
- Department of Cancer Immunology and Virology, Dana-Farber Cancer Institute, Boston, Massachusetts, United States of America
- Department of Microbiology and Immunobiology, Harvard Medical School, Boston, Massachusetts, United States of America
- * E-mail:
| |
Collapse
|