1
|
Abstract
The ability to manipulate the chemical composition of proteins and peptides has been central to the development of improved polypeptide-based therapeutics and has enabled researchers to address fundamental biological questions that would otherwise be out of reach. Protein ligation, in which two or more polypeptides are covalently linked, is a powerful strategy for generating semisynthetic products and for controlling polypeptide topology. However, specialized tools are required to efficiently forge a peptide bond in a chemoselective manner with fast kinetics and high yield. Fortunately, nature has addressed this challenge by evolving enzymatic mechanisms that can join polypeptides using a diverse set of chemical reactions. Here, we summarize how such nature-inspired protein ligation strategies have been repurposed as chemical biology tools that afford enhanced control over polypeptide composition.
Collapse
Affiliation(s)
- Rasmus Pihl
- Chemical Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Department of Biomedicine, Aarhus University, Aarhus C, Denmark
| | - Qingfei Zheng
- Department of Radiation Oncology, College of Medicine, The Ohio State University, Columbus, OH, USA.
- Center for Cancer Metabolism, James Comprehensive Cancer Center, The Ohio State University, Columbus, OH, USA.
- Department of Biological Chemistry and Pharmacology, College of Medicine, The Ohio State University, Columbus, OH, USA.
| | - Yael David
- Chemical Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
- Department of Pharmacology, Weill Cornell Medicine, New York, NY, USA.
- Department of Physiology, Biophysics and Systems Biology, Weill Cornell Medicine, New York, NY, USA.
| |
Collapse
|
2
|
Liu S, Duan X, Peng F, Wang Y, Liu Y, Wan X, Zhang J, Li X, Sun X. A tunable genome editing system of the prime editor mediated by dihydrofolate reductase. J Genet Genomics 2023; 50:204-207. [PMID: 36055523 DOI: 10.1016/j.jgg.2022.08.004] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Revised: 08/19/2022] [Accepted: 08/22/2022] [Indexed: 01/28/2023]
Affiliation(s)
- Shu Liu
- Department of Ophthalmology, Shanghai General Hospital (Shanghai First People's Hospital), Shanghai Jiao Tong University School of Medicine, Shanghai 200080, China; Department of Ophthalmology, Shanghai Changhai Hospital, Shanghai 200043, China
| | - Xiaoyue Duan
- Department of Ophthalmology, Shanghai General Hospital (Shanghai First People's Hospital), Shanghai Jiao Tong University School of Medicine, Shanghai 200080, China
| | - Feng Peng
- Department of Ophthalmology, Shanghai General Hospital (Shanghai First People's Hospital), Shanghai Jiao Tong University School of Medicine, Shanghai 200080, China
| | - Yafang Wang
- Department of Ophthalmology, Shanghai General Hospital (Shanghai First People's Hospital), Shanghai Jiao Tong University School of Medicine, Shanghai 200080, China
| | - Yang Liu
- Department of Ophthalmology, Shanghai General Hospital (Shanghai First People's Hospital), Shanghai Jiao Tong University School of Medicine, Shanghai 200080, China
| | - Xiaoling Wan
- Department of Ophthalmology, Shanghai General Hospital (Shanghai First People's Hospital), Shanghai Jiao Tong University School of Medicine, Shanghai 200080, China; Shanghai Key Laboratory of Ocular Fundus Diseases, 100 Haining Road, Shanghai 200080, China
| | - Jingfa Zhang
- Department of Ophthalmology, Shanghai General Hospital (Shanghai First People's Hospital), Shanghai Jiao Tong University School of Medicine, Shanghai 200080, China; Shanghai Key Laboratory of Ocular Fundus Diseases, 100 Haining Road, Shanghai 200080, China
| | - Xiaosa Li
- Department of Ophthalmology, Shanghai General Hospital (Shanghai First People's Hospital), Shanghai Jiao Tong University School of Medicine, Shanghai 200080, China; Shanghai Key Laboratory of Ocular Fundus Diseases, 100 Haining Road, Shanghai 200080, China.
| | - Xiaodong Sun
- Department of Ophthalmology, Shanghai General Hospital (Shanghai First People's Hospital), Shanghai Jiao Tong University School of Medicine, Shanghai 200080, China; Shanghai Key Laboratory of Ocular Fundus Diseases, 100 Haining Road, Shanghai 200080, China
| |
Collapse
|
3
|
Kupatt C, Windisch A, Moretti A, Wolf E, Wurst W, Walter MC. Genome editing for Duchenne muscular dystrophy: a glimpse of the future? Gene Ther 2021; 28:542-548. [PMID: 33531685 PMCID: PMC8455335 DOI: 10.1038/s41434-021-00222-4] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2020] [Revised: 12/01/2020] [Accepted: 01/15/2021] [Indexed: 12/11/2022]
Abstract
Mutations in Dystrophin, one of the largest proteins in the mammalian body, are causative for a severe form of muscle disease, Duchenne Muscular Dystrophy (DMD), affecting not only skeletal muscle, but also the heart. In particular, exons 45–52 constitute a hotspot for DMD mutations. A variety of molecular therapies have been developed, comprising vectors encoding micro- and minidystrophins as well as utrophin, a protein with partially overlapping functions. With the advent of the CRISPR-Cas9-nuclease, genome editing offers a novel option of correction of the disease-cuasing mutations. Full restoration of the healthy gene by homology directed repair is a rare event. However, non-homologous end-joining (NHEJ) may restore the reading frame by causing exon excision. This approach has first been demonstrated in mice and then translated to large animals (dogs, pigs). This review discusses the potential opportunities and limitations of genome editing in DMD, including the generation of appropriate animal models as well as new developments in genome editing tools.
Collapse
Affiliation(s)
- Christian Kupatt
- Klinik und Poliklinik für Innere Medizin I, Klinikum rechts der Isar, Technical University Munich, Munich, Germany. .,DZHK (German Center for Cardiovascular Research), Munich Heart Alliance, Munich, Germany.
| | - Alina Windisch
- Klinik und Poliklinik für Innere Medizin I, Klinikum rechts der Isar, Technical University Munich, Munich, Germany.,DZHK (German Center for Cardiovascular Research), Munich Heart Alliance, Munich, Germany
| | - Alessandra Moretti
- Klinik und Poliklinik für Innere Medizin I, Klinikum rechts der Isar, Technical University Munich, Munich, Germany.,DZHK (German Center for Cardiovascular Research), Munich Heart Alliance, Munich, Germany
| | - Eckhard Wolf
- Chair for Molecular Animal Breeding and Biotechnology, Gene Center and Department of Veterinary Sciences, and Center for Innovative Medical Models (CiMM), LMU Munich, Munich, Germany
| | - Wolfgang Wurst
- Institute of Development Genetics, Helmholtz-Centre Munich, Munich, Germany.,German Center for Neurodegenerative Diseases, Munich, Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - Maggie C Walter
- Friedrich Baur Institute, Department of Neurology, LMU Munich, Munich, Germany
| |
Collapse
|
4
|
Bhagawati M, Hoffmann S, Höffgen KS, Piehler J, Busch KB, Mootz HD. In Cellulo Protein Semi‐Synthesis from Endogenous and Exogenous Fragments Using the Ultra‐Fast Split Gp41‐1 Intein. Angew Chem Int Ed Engl 2020. [DOI: 10.1002/ange.202006822] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Affiliation(s)
- Maniraj Bhagawati
- Department of Chemistry and Pharmacy Institute of Biochemistry, University of Münster Corrensstrasse 36 48149 Münster Germany
| | - Simon Hoffmann
- Department of Chemistry and Pharmacy Institute of Biochemistry, University of Münster Corrensstrasse 36 48149 Münster Germany
| | - Katharina S. Höffgen
- Department of Chemistry and Pharmacy Institute of Biochemistry, University of Münster Corrensstrasse 36 48149 Münster Germany
| | - Jacob Piehler
- Department of Biology and Center for Cellular Nanoanalytics University of Osnabrück Barbarastrasse 11 49076 Osnabrück Germany
| | - Karin B. Busch
- Institute of Molecular Cell Biology University of Münster Schlossplatz 5 48149 Münster Germany
| | - Henning D. Mootz
- Department of Chemistry and Pharmacy Institute of Biochemistry, University of Münster Corrensstrasse 36 48149 Münster Germany
| |
Collapse
|
5
|
Bhagawati M, Hoffmann S, Höffgen KS, Piehler J, Busch KB, Mootz HD. In Cellulo Protein Semi-Synthesis from Endogenous and Exogenous Fragments Using the Ultra-Fast Split Gp41-1 Intein. Angew Chem Int Ed Engl 2020; 59:21007-21015. [PMID: 32777124 PMCID: PMC7693240 DOI: 10.1002/anie.202006822] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2020] [Revised: 07/15/2020] [Indexed: 12/19/2022]
Abstract
Protein semi-synthesis inside live cells from exogenous and endogenous parts offers unique possibilities for studying proteins in their native context. Split-intein-mediated protein trans-splicing is predestined for such endeavors and has seen some successes, but a much larger variety of established split inteins and associated protocols is urgently needed. We characterized the association and splicing parameters of the Gp41-1 split intein, which favorably revealed a nanomolar affinity between the intein fragments combined with the exceptionally fast splicing rate. Following bead-loading of a chemically modified intein fragment precursor into live mammalian cells, we fluorescently labeled target proteins on their N- and C-termini with short peptide tags, thus ensuring minimal perturbation of their structure and function. In combination with a nuclear-entrapment strategy to minimize cytosolic fluorescence background, we applied our technique for super-resolution imaging and single-particle tracking of the outer mitochondrial protein Tom20 in HeLa cells.
Collapse
Affiliation(s)
- Maniraj Bhagawati
- Department of Chemistry and PharmacyInstitute of Biochemistry, University of MünsterCorrensstrasse 3648149MünsterGermany
| | - Simon Hoffmann
- Department of Chemistry and PharmacyInstitute of Biochemistry, University of MünsterCorrensstrasse 3648149MünsterGermany
| | - Katharina S. Höffgen
- Department of Chemistry and PharmacyInstitute of Biochemistry, University of MünsterCorrensstrasse 3648149MünsterGermany
| | - Jacob Piehler
- Department of Biology and Center for Cellular NanoanalyticsUniversity of OsnabrückBarbarastrasse 1149076OsnabrückGermany
| | - Karin B. Busch
- Institute of Molecular Cell BiologyUniversity of MünsterSchlossplatz 548149MünsterGermany
| | - Henning D. Mootz
- Department of Chemistry and PharmacyInstitute of Biochemistry, University of MünsterCorrensstrasse 3648149MünsterGermany
| |
Collapse
|
6
|
Li R, Chen Y, Du K, Feng W. Peptide Bond Formation Between the Hetrosubunits of ω-Transaminase, Alanine Dehydrogenase, and Formate Dehydrogenase Through Subunit Splicing Promoted by Heterodimerization of Leucine Zipper Motifs. Front Bioeng Biotechnol 2020; 8:686. [PMID: 32695764 PMCID: PMC7338344 DOI: 10.3389/fbioe.2020.00686] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2020] [Accepted: 06/02/2020] [Indexed: 11/13/2022] Open
Abstract
For the multimeric enzymes R-ω-transaminase (RTA), alanine dehydrogenase (AlaDH), and formate dehydrogenase (FDH), peptide bond formation between the hetrosubunits has been achieved by the intein-mediated in vivo subunit splicing. The subunit ligation is triggered by the heterodimerization of an arginine rich leucine zipper motif with a glutamic acid rich leucine zipper motif. The one-by-one ligation of hetrosubunits constructs the pairing enzymes RTA&AlaDH and AlaDH&FDH. The ligation modes were analyzed based on blue native polyacrylamide gel electrophoresis (BN-PAGE). The spectra of circular dichroism (CD), fluorescence, and two-dimensional FTIR provide information on the secondary structures and stability of the pairing enzymes. The enzyme-substrate interaction was analyzed based on microscale thermophoresis analysis. In contrast to the mixed three enzymes RTA + AlaDH + FDH, the ligated enzymes RTA&AlaDH + AlaDH&FDH exhibited a much larger substrate affinity, higher stability, and significantly enhanced activity.
Collapse
Affiliation(s)
- Rong Li
- Department of Biological Engineering, Beijing University of Chemical Technology, Beijing, China
| | - Yao Chen
- Department of Biological Engineering, Beijing University of Chemical Technology, Beijing, China
| | - Kun Du
- Department of Biological Engineering, Beijing University of Chemical Technology, Beijing, China
| | - Wei Feng
- Department of Biological Engineering, Beijing University of Chemical Technology, Beijing, China
| |
Collapse
|
7
|
Beyer HM, Mikula KM, Li M, Wlodawer A, Iwaï H. The crystal structure of the naturally split gp41-1 intein guides the engineering of orthogonal split inteins from cis-splicing inteins. FEBS J 2020; 287:1886-1898. [PMID: 31665813 PMCID: PMC7190452 DOI: 10.1111/febs.15113] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2019] [Revised: 10/01/2019] [Accepted: 10/29/2019] [Indexed: 01/09/2023]
Abstract
Protein trans-splicing catalyzed by split inteins has increasingly become useful as a protein engineering tool. We solved the 1.0 Å-resolution crystal structure of a fused variant from the naturally split gp41-1 intein, previously identified from environmental metagenomic sequence data. The structure of the 125-residue gp41-1 intein revealed a compact pseudo-C2-symmetry commonly found in the Hedgehog/Intein superfamily with extensive charge-charge interactions between the split N- and C-terminal intein fragments that are common among naturally occurring split inteins. We successfully created orthogonal split inteins by engineering a similar charge network into the same region of a cis-splicing intein. This strategy could be applicable for creating novel natural-like split inteins from other, more prevalent cis-splicing inteins. DATABASE: Structural data are available in the RCSB Protein Data Bank under the accession number 6QAZ.
Collapse
Affiliation(s)
- Hannes Michael Beyer
- Research Program in Structural Biology and Biophysics, Institute of Biotechnology, University of Helsinki, 00014 Helsinki, Finland
| | - Kornelia Malgorzata Mikula
- Research Program in Structural Biology and Biophysics, Institute of Biotechnology, University of Helsinki, 00014 Helsinki, Finland
| | - Mi Li
- Macromolecular Crystallography Laboratory, National Cancer Institute, Frederick, MD 21702, USA
- Basic Science Program, Leidos Biomedical Research, Frederick National Laboratory for Cancer Research, Frederick, MD 21702, USA
| | - Alexander Wlodawer
- Macromolecular Crystallography Laboratory, National Cancer Institute, Frederick, MD 21702, USA
| | - Hideo Iwaï
- Research Program in Structural Biology and Biophysics, Institute of Biotechnology, University of Helsinki, 00014 Helsinki, Finland
| |
Collapse
|
8
|
Seo HN, Bang D. Promiscuous Trans-splicing Activities Revealed by Next Generation Sequencing-based Analysis of 298 Split Inteins. BIOTECHNOL BIOPROC E 2020. [DOI: 10.1007/s12257-019-0394-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
9
|
Abstract
Protein semisynthesis-defined herein as the assembly of a protein from a combination of synthetic and recombinant fragments-is a burgeoning field of chemical biology that has impacted many areas in the life sciences. In this review, we provide a comprehensive survey of this area. We begin by discussing the various chemical and enzymatic methods now available for the manufacture of custom proteins containing noncoded elements. This section begins with a discussion of methods that are more chemical in origin and ends with those that employ biocatalysts. We also illustrate the commonalities that exist between these seemingly disparate methods and show how this is allowing for the development of integrated chemoenzymatic methods. This methodology discussion provides the technical foundation for the second part of the review where we cover the great many biological problems that have now been addressed using these tools. Finally, we end the piece with a short discussion on the frontiers of the field and the opportunities available for the future.
Collapse
Affiliation(s)
| | - Tom W. Muir
- Department of Chemistry, Princeton University, Frick Laboratory, Princeton, New Jersey 08544, United States
| |
Collapse
|
10
|
Abstract
In recent years, split inteins have seen widespread use as molecular platforms for the design of a variety of peptide and protein chemistry technologies, most notably protein ligation. The development of these approaches is dependent on the identification and/or design of split inteins with robust activity, stability, and solubility. Here, we describe two approaches to characterize and compare the activities of newly identified or engineered split inteins. The first assay employs an E. coli-based selection system to rapidly screen the activities of many inteins and can be repurposed for directed evolution. The second assay utilizes reverse-phase high-performance liquid chromatography (RP-HPLC) to provide insights into individual chemical steps in the protein splicing reaction, information that can guide further engineering efforts. These techniques provide useful alternatives to common assays that utilize SDS-PAGE to analyze splicing reaction progress.
Collapse
|
11
|
Windsor IW, Graff CJ, Raines RT. Circular zymogens of human ribonuclease 1. Protein Sci 2019; 28:1713-1719. [PMID: 31306518 PMCID: PMC6699097 DOI: 10.1002/pro.3686] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2019] [Revised: 07/11/2019] [Accepted: 07/11/2019] [Indexed: 12/16/2022]
Abstract
The endogenous production of enzymes as zymogens provides a means to control catalytic activities. Here, we describe the heterologous production of ribonuclease 1 (RNase 1), which is the most prevalent secretory ribonuclease in humans, as a zymogen. In folded RNase 1, the N and C termini flank the enzymic active site. By using intein-mediated cis-splicing, we created circular proteins in which access to the active site of RNase 1 is obstructed by an amino-acid sequence that is recognized by the HIV-1 protease. Installing a sequence that does not perturb the RNase 1 fold led to only modest inactivation. In contrast, the ancillary truncation of residues from each terminus led to a substantial decrease in the catalytic activity of the zymogen with the maintenance of thermostability. For optimized zymogens, activation by HIV-1 protease led to a > 104 -fold increase in ribonucleolytic activity at a rate comparable to that for the cleavage of endogenous viral substrates. Molecular modeling indicated that these zymogens are inactivated by conformational distortion in addition to substrate occlusion. Because protease levels are elevated in many disease states and ribonucleolytic activity can be cytotoxic, RNase 1 zymogens have potential as generalizable prodrugs.
Collapse
Affiliation(s)
- Ian W. Windsor
- Department of BiochemistryUniversity of Wisconsin−MadisonMadisonWisconsin
- Department of ChemistryMassachusetts Institute of TechnologyCambridgeMassachusetts
| | - Crystal J. Graff
- Department of BiochemistryUniversity of Wisconsin−MadisonMadisonWisconsin
| | - Ronald T. Raines
- Department of BiochemistryUniversity of Wisconsin−MadisonMadisonWisconsin
- Department of ChemistryMassachusetts Institute of TechnologyCambridgeMassachusetts
- Department of ChemistryUniversity of Wisconsin−MadisonMadisonWisconsin
| |
Collapse
|
12
|
Wright JN, Wong WL, Harvey JA, Garnett JA, Itzhaki LS, Main ERG. Scalable Geometrically Designed Protein Cages Assembled via Genetically Encoded Split Inteins. Structure 2019; 27:776-784.e4. [PMID: 30879889 DOI: 10.1016/j.str.2019.02.005] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2018] [Revised: 12/21/2018] [Accepted: 02/15/2019] [Indexed: 01/20/2023]
Abstract
Engineering proteins to assemble into user-defined structures is key in their development for biotechnological applications. However, designing generic rather than bespoke solutions is challenging. Here we describe an expandable recombinant assembly system that produces scalable protein cages via split intein-mediated native chemical ligation. Three types of component are used: two complementary oligomeric "half-cage" protein fusions and an extendable monomeric "linker" fusion. All are composed of modular protein domains chosen to fulfill the required geometries, with two orthogonal pairs of split intein halves to drive assembly when mixed. This combination enables both one-pot construction of two-component cages and stepwise assembly of larger three-component scalable cages. To illustrate the system's versatility, trimeric half-cages and linker constructs comprising consensus-designed repeat proteins were ligated in one-pot and stepwise reactions. Under mild conditions, rapid high-yielding ligations were obtained, from which discrete proteins cages were easily purified and shown to form the desired trigonal bipyramidal structures.
Collapse
Affiliation(s)
- James N Wright
- School of Biological and Chemical Sciences, Queen Mary, University of London, Mile End Road, London E1 4NS, UK
| | - Wan Ling Wong
- School of Biological and Chemical Sciences, Queen Mary, University of London, Mile End Road, London E1 4NS, UK
| | - Joseph A Harvey
- School of Biological and Chemical Sciences, Queen Mary, University of London, Mile End Road, London E1 4NS, UK
| | - James A Garnett
- School of Biological and Chemical Sciences, Queen Mary, University of London, Mile End Road, London E1 4NS, UK
| | - Laura S Itzhaki
- Department of Pharmacology, University of Cambridge, Tennis Court Road, Cambridge CB2 1PD, UK
| | - Ewan R G Main
- School of Biological and Chemical Sciences, Queen Mary, University of London, Mile End Road, London E1 4NS, UK.
| |
Collapse
|
13
|
Du K, Li R, Zhang D, Feng W. Covalent Linkage of an R-ω-Transaminase to a d-Amino Acid Oxidase through Protein Splicing to Enhance Enzymatic Catalysis of Transamination. Chembiochem 2019; 20:701-709. [PMID: 30447031 DOI: 10.1002/cbic.201800646] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2018] [Indexed: 11/07/2022]
Abstract
R-ω-Transaminases (RTAs) catalyse the conversion of R-configured amines [e.g., (R)-1-phenylethylamine] into the corresponding ketones (e.g., acetophenone), by transferring an amino group from an amino donor [e.g., (R)-1-phenylethylamine] onto an amino acceptor (e.g., pyruvate), resulting in a co-product (e.g., d-alanine). d-Alanine can be deaminated back to pyruvate by d-amino acid oxidase (DAAOs). Here, through in vivo subunit splicing, the N terminus of an RTA subunit (RTAS ) was specifically ligated to the C terminus of a DAAO subunit (DAAOS ) through native peptide bonds (RTA&DAAO). RTAS is in close proximity to DAAOS , at a molecular-scale distance. Thus the transfer of pyruvate and d-alanine between RTA and DAAO can be directional and efficient. Pyruvate→d-alanine→pyruvate cycles are efficiently formed, thus promoting the forward transamination reaction. In a different, in vitro noncovalent approach, based on coiled-coil association, the RTAS N terminus was specifically associated with the DAAOS C terminus (RTA#DAAO). In addition, the two mixed individual enzymes (RTA+DAAO) were also studied. RTA&DAAO has a shorter distance between the paired subunits (RTAS -DAAOS ) than RTA#DAAO, and the number of the paired subunits is higher than in the case of RTA#DAAO, whereas RTA+DAAO cannot form the paired subunits. RTA&DAAO exhibited a transamination catalysis efficiency higher than that of RTA#DAAO and much higher than that of RTA+DAAO.
Collapse
Affiliation(s)
- Kun Du
- Department of Biochemical Engineering, Beijing University of Chemical Technology, Beisanhuandonglu 15, Beijing, 100029, China
| | - Rong Li
- Department of Biochemical Engineering, Beijing University of Chemical Technology, Beisanhuandonglu 15, Beijing, 100029, China
| | - Dongrui Zhang
- Department of Biochemical Engineering, Beijing University of Chemical Technology, Beisanhuandonglu 15, Beijing, 100029, China
| | - Wei Feng
- Department of Biochemical Engineering, Beijing University of Chemical Technology, Beisanhuandonglu 15, Beijing, 100029, China
| |
Collapse
|
14
|
Chen Y, Chu H, Liu W, Feng W. Simultaneous synthesis of l-DOPA and oxidation of d-amino acid by specific coupling of a peroxidase to d-amino acid oxidase. Enzyme Microb Technol 2019; 121:8-16. [PMID: 30554648 DOI: 10.1016/j.enzmictec.2018.10.013] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2018] [Revised: 10/14/2018] [Accepted: 10/26/2018] [Indexed: 12/21/2022]
Abstract
3,4 Dihydroxy phenyl l-alanine (L-DOPA) is the gold standard Parkinson's disease therapy. A heme-dependent peroxidase (HDP) catalyzes the ortho-hydroxylation of l-tyrosine to l-DOPA using H2O2 as the co-substrate. d-amino acid oxidase (DAAO) catalyzes the oxidative deamination of d-amino acids (e.g. d-alanine), and H2O2 is evolved. However, both the enzymes DAAO and HDP can be inactivated by H2O2 during the catalysis. In situ generation and utilization of H2O2 can siginificatly reduce the inactivation by H2O2. HDP exists as a monomer and DAAO is a dimeric enzyme. Herein, the C-terminus of HDP was specifically ligated to the N-terminus of the DAAO subunit with native peptide through the in vivo monomer-subunit splicing. In the splicing product HDP&DAAO, HDP is close to the DAAO subunit at a molecular distance, and the transfer of H2O2 from DAAO to HDP is facilitated. In addition, HDP&DAAO exhibited a higher stability than HDP. Kinetics analysis showed that both the substrates l-tyrosine and d-alanine obey the Michaelis-Menten kinetics. For the deamination of d-alanine, the catalytic efficiency of HDP&DAAO is 3.05 times that of DAAO. For the sybthesis of l-DOPA from l-tyrosine, the catalytic efficiency of HDP&DAAO is 1.58 times that of HDP. Furthermore, HDP&DAAO was encapsulated within a Znic-based coordination polymer (Zn-CP). The morphorogy of HDP&DAAO/Zn-CP can be regulated by the enzyme concentration, the catalytic efficiency of the conjugates was found to be dependent on the morphorogy. The conjugates HDP&DAAO/Zn-CP exhibited a higher catalytic efficiency than free HDP&DAAO.
Collapse
Affiliation(s)
- Yao Chen
- Department of Biochemical Engineering, Beijing University of Chemical Technology, Beijing, China
| | - Hao Chu
- Department of Biochemical Engineering, Beijing University of Chemical Technology, Beijing, China
| | - Wei Liu
- Department of Biochemical Engineering, Beijing University of Chemical Technology, Beijing, China
| | - Wei Feng
- Department of Biochemical Engineering, Beijing University of Chemical Technology, Beijing, China.
| |
Collapse
|
15
|
Reduction of non-specific toxicity of immunotoxin by intein mediated reconstitution on target cells. Int Immunopharmacol 2019; 66:288-295. [DOI: 10.1016/j.intimp.2018.11.039] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2018] [Revised: 11/19/2018] [Accepted: 11/23/2018] [Indexed: 12/13/2022]
|
16
|
Li R, Zhou X, Liu D, Feng W. Enhancing the activity and stability of Mn-superoxide dismutase by one-by-one ligation to catalase. Free Radic Biol Med 2018; 129:138-145. [PMID: 30227270 DOI: 10.1016/j.freeradbiomed.2018.09.018] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/17/2018] [Revised: 09/09/2018] [Accepted: 09/14/2018] [Indexed: 02/07/2023]
Abstract
Dismutation of superoxide by superoxide dismutase (SOD) generates hydrogen peroxide, which may be reduced to hydroxyl radical. The generated H2O2 during the catalysis can have an oxidative damage to SOD. Hydrogen peroxide decomposition by catalase (CAT) can help circumvent the problem. Mn-superoxide dismutase (herein referred to as SOD) and CAT are dimeric and tetrameric proteins, respectively. Herein, through intein-mediated in vivo subunit splicing, the C-terminus of the CAT subunit (CATS) has been specifically ligated to the N-terminus of the SOD subunit (SODS) with a peptide bond. Thus, the splicing product SOD&CAT combines the superoxide anion (•O2-) scavenging ability and the ability of decomposing H2O2. The in vivo subunit splicing has little effect on the secondary structures of the enzymes as confirmed by circular dichroism (CD) spectra. Fluorescence spectra showed that the splicing product SOD&CAT has a higher stability than SOD. In the splicing product SOD&CAT, the SOD subunits are in close proximity to the CAT subunits, facilitating immediate transfer of H2O2 between the enzymes and enabling efficient decomposition of H2O2. SOD&CAT exhibited a superoxide anion (•O2-) scavenging ability 244% higher than that of SOD and 46% higher than that of the mixed enzymes SOD+CAT.
Collapse
Affiliation(s)
- Rong Li
- Department of Biochemical Engineering, Beijing University of Chemical Technology, Beijing 100029, China
| | - Xiaoqi Zhou
- Department of Biochemical Engineering, Beijing University of Chemical Technology, Beijing 100029, China
| | - Dan Liu
- Department of Biochemical Engineering, Beijing University of Chemical Technology, Beijing 100029, China
| | - Wei Feng
- Department of Biochemical Engineering, Beijing University of Chemical Technology, Beijing 100029, China.
| |
Collapse
|
17
|
Kelley DS, Lennon CW, Li Z, Miller MR, Banavali NK, Li H, Belfort M. Mycobacterial DnaB helicase intein as oxidative stress sensor. Nat Commun 2018; 9:4363. [PMID: 30341292 PMCID: PMC6195587 DOI: 10.1038/s41467-018-06554-x] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2018] [Accepted: 09/10/2018] [Indexed: 11/09/2022] Open
Abstract
Inteins are widespread self-splicing protein elements emerging as potential post-translational environmental sensors. Here, we describe two inteins within one protein, the Mycobacterium smegmatis replicative helicase DnaB. These inteins, DnaBi1 and DnaBi2, have homology to inteins in pathogens, splice with vastly varied rates, and are differentially responsive to environmental stressors. Whereas DnaBi1 splicing is reversibly inhibited by oxidative and nitrosative insults, DnaBi2 is not. Using a reporter that measures splicing in a native intein-containing organism and western blotting, we show that H2O2 inhibits DnaBi1 splicing in M. smegmatis. Intriguingly, upon oxidation, the catalytic cysteine of DnaBi1 forms an intramolecular disulfide bond. We report a crystal structure of the class 3 DnaBi1 intein at 1.95 Å, supporting our findings and providing insight into this splicing mechanism. We propose that this cysteine toggle allows DnaBi1 to sense stress, pausing replication to maintain genome integrity, and then allowing splicing immediately when permissive conditions return.
Collapse
Affiliation(s)
- Danielle S Kelley
- Department of Biomedical Sciences, School of Public Health, University at Albany, Albany, NY, 12222, USA
| | - Christopher W Lennon
- Department of Biological Sciences and RNA Institute, University at Albany, Albany, NY, 12222, USA
| | - Zhong Li
- Wadsworth Center, New York State Department of Health, 120 New Scotland Ave, Albany, NY, 12208, USA
| | - Michael R Miller
- Department of Chemistry, University at Albany, Albany, NY, 12222, USA
| | - Nilesh K Banavali
- Department of Biomedical Sciences, School of Public Health, University at Albany, Albany, NY, 12222, USA
- Wadsworth Center, New York State Department of Health, 120 New Scotland Ave, Albany, NY, 12208, USA
| | - Hongmin Li
- Department of Biomedical Sciences, School of Public Health, University at Albany, Albany, NY, 12222, USA.
- Wadsworth Center, New York State Department of Health, 120 New Scotland Ave, Albany, NY, 12208, USA.
| | - Marlene Belfort
- Department of Biomedical Sciences, School of Public Health, University at Albany, Albany, NY, 12222, USA.
- Department of Biological Sciences and RNA Institute, University at Albany, Albany, NY, 12222, USA.
| |
Collapse
|
18
|
Di Ventura B, Mootz HD. Switchable inteins for conditional protein splicing. Biol Chem 2018; 400:467-475. [DOI: 10.1515/hsz-2018-0309] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2018] [Accepted: 08/20/2018] [Indexed: 12/19/2022]
Abstract
Abstract
Synthetic biologists aim at engineering controllable biological parts such as DNA, RNA and proteins in order to steer biological activities using external inputs. Proteins can be controlled in several ways, for instance by regulating the expression of their encoding genes with small molecules or light. However, post-translationally modifying pre-existing proteins to regulate their function or localization leads to faster responses. Conditional splicing of internal protein domains, termed inteins, is an attractive methodology for this purpose. Here we discuss methods to control intein activity with a focus on those compatible with applications in living cells.
Collapse
Affiliation(s)
- Barbara Di Ventura
- Faculty of Biology, University of Freiburg , 79104 Freiburg , Germany
- BIOSS – Centre for Biological Signalling Studies, University of Freiburg , 79104 Freiburg , Germany
| | - Henning D. Mootz
- Department Chemistry and Pharmacy , Institute of Biochemistry, University of Münster , Münster D-48149 , Germany
| |
Collapse
|
19
|
Gordo V, Aparicio D, Pérez-Luque R, Benito A, Vilanova M, Usón I, Fita I, Ribó M. Structural Insights into Subunits Assembly and the Oxyester Splicing Mechanism of Neq pol Split Intein. Cell Chem Biol 2018; 25:871-879.e2. [PMID: 29754955 DOI: 10.1016/j.chembiol.2018.04.008] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2017] [Revised: 02/28/2018] [Accepted: 04/08/2018] [Indexed: 11/28/2022]
Abstract
Split inteins are expressed as two separated subunits (N-intein and C-intein) fused to the corresponding exteins. The specific association of both intein subunits precedes protein splicing, which results in excision of the intein subunits and in ligation, by a peptide bond, of the concomitant exteins. Catalytically active intein precursors are typically too reactive for crystallization or even isolation. Neq pol is the trans-intein of the B-type DNA polymerase I split gene from hyperthermophile Nanoarchaeum equitans. We have determined the crystal structures of both the isolated NeqN and the complex of NeqN and NeqC subunits carrying the wild-type sequences, including the essential catalytic residues Ser1 and Thr+1, in addition to seven and three residues of the N- and C-exteins, respectively. These structures provide detailed information on the unique oxyester chemistry of the splicing mechanism of Neq pol and of the extensive rearrangements that occur in NeqN during the association step.
Collapse
Affiliation(s)
- Verónica Gordo
- Laboratori d'Enginyeria de Proteïnes, Departament de Biologia, Facultat de Ciències, Universitat de Girona, C/ Maria Aurèlia Capmany 40, 17003 Girona, Spain; IdIBGi Hospital Universitari Josep Trueta, Girona, Spain
| | - David Aparicio
- Instituto de Biología Molecular de Barcelona (IBMB-CSIC), Parc Cientific de Barcelona, Baldiri i Reixac 10, 08028 Barcelona, Spain
| | - Rosa Pérez-Luque
- Instituto de Biología Molecular de Barcelona (IBMB-CSIC), Parc Cientific de Barcelona, Baldiri i Reixac 10, 08028 Barcelona, Spain
| | - Antoni Benito
- Laboratori d'Enginyeria de Proteïnes, Departament de Biologia, Facultat de Ciències, Universitat de Girona, C/ Maria Aurèlia Capmany 40, 17003 Girona, Spain; IdIBGi Hospital Universitari Josep Trueta, Girona, Spain
| | - Maria Vilanova
- Laboratori d'Enginyeria de Proteïnes, Departament de Biologia, Facultat de Ciències, Universitat de Girona, C/ Maria Aurèlia Capmany 40, 17003 Girona, Spain; IdIBGi Hospital Universitari Josep Trueta, Girona, Spain
| | - Isabel Usón
- Instituto de Biología Molecular de Barcelona (IBMB-CSIC), Parc Cientific de Barcelona, Baldiri i Reixac 10, 08028 Barcelona, Spain; ICREA Lluís Companys 23, 08003 Barcelona, Spain
| | - Ignacio Fita
- Instituto de Biología Molecular de Barcelona (IBMB-CSIC), Parc Cientific de Barcelona, Baldiri i Reixac 10, 08028 Barcelona, Spain.
| | - Marc Ribó
- Laboratori d'Enginyeria de Proteïnes, Departament de Biologia, Facultat de Ciències, Universitat de Girona, C/ Maria Aurèlia Capmany 40, 17003 Girona, Spain; IdIBGi Hospital Universitari Josep Trueta, Girona, Spain.
| |
Collapse
|
20
|
Abstract
Exciting new technological developments have pushed the boundaries of structural biology, and have enabled studies of biological macromolecules and assemblies that would have been unthinkable not long ago. Yet, the enhanced capabilities of structural biologists to pry into the complex molecular world have also placed new demands on the abilities of protein engineers to reproduce this complexity into the test tube. With this challenge in mind, we review the contents of the modern molecular engineering toolbox that allow the manipulation of proteins in a site-specific and chemically well-defined fashion. Thus, we cover concepts related to the modification of cysteines and other natural amino acids, native chemical ligation, intein and sortase-based approaches, amber suppression, as well as chemical and enzymatic bio-conjugation strategies. We also describe how these tools can be used to aid methodology development in X-ray crystallography, nuclear magnetic resonance, cryo-electron microscopy and in the studies of dynamic interactions. It is our hope that this monograph will inspire structural biologists and protein engineers alike to apply these tools to novel systems, and to enhance and broaden their scope to meet the outstanding challenges in understanding the molecular basis of cellular processes and disease.
Collapse
|
21
|
Li G, Yuan S, Zheng S, Liu Y, Huang G. In Situ Living Cell Protein Analysis by Single-Step Mass Spectrometry. Anal Chem 2018; 90:3409-3415. [DOI: 10.1021/acs.analchem.7b05055] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
|
22
|
Du K, Zhao J, Sun J, Feng W. Specific Ligation of Two Multimeric Enzymes with Native Peptides and Immobilization with Controlled Molar Ratio. Bioconjug Chem 2017; 28:1166-1175. [DOI: 10.1021/acs.bioconjchem.7b00043] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Affiliation(s)
- Kun Du
- Department of Biochemical
Engineering, Beijing University of Chemical Technology, Beijing, 100029, China
| | - Jinjin Zhao
- Department of Biochemical
Engineering, Beijing University of Chemical Technology, Beijing, 100029, China
| | - Jian Sun
- Department of Biochemical
Engineering, Beijing University of Chemical Technology, Beijing, 100029, China
| | - Wei Feng
- Department of Biochemical
Engineering, Beijing University of Chemical Technology, Beijing, 100029, China
| |
Collapse
|
23
|
Abstract
On a past volume of this monograph we have reviewed general aspects of the varied technologies available to generate peptide arrays. Hallmarks in the development of the technology and a main sketch of preparative steps and applications in binding assays were used to walk the reader through details of peptide arrays. In this occasion, we resume from that work and bring in some considerations on quantitative evaluation of measurements as well as on selected reports applying the technology.
Collapse
Affiliation(s)
| | - Rudolf Volkmer
- Institute of Medical Immunology, Charité-Universitätsmedizin zu Berlin, Hessische Str. 3-4, Berlin, 10115, Germany
| |
Collapse
|
24
|
Braner M, Kollmannsperger A, Wieneke R, Tampé R. 'Traceless' tracing of proteins - high-affinity trans-splicing directed by a minimal interaction pair. Chem Sci 2015; 7:2646-2652. [PMID: 28660037 PMCID: PMC5477019 DOI: 10.1039/c5sc02936h] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2015] [Accepted: 12/18/2015] [Indexed: 12/26/2022] Open
Abstract
Using a minimal lock-and-key element the affinity between the intein fragments for N-terminal protein trans-splicing was significantly increased, allowing for site-specific, ‘traceless’ covalent protein labeling in living mammalian cells at nanomolar probe concentrations.
Protein trans-splicing mediated by split inteins is a powerful technique for site-specific protein modification. Despite recent developments there is still an urgent need for ultra-small high-affinity intein tags for in vitro and in vivo approaches. To date, only very few in-cell applications of protein trans-splicing have been reported, all limited to C-terminal protein modifications. Here, we developed a strategy for covalent N-terminal intein-mediated protein labeling at (sub) nanomolar probe concentrations. Combined with a minimal synthetic lock-and-key element, the affinity between the intein fragments was increased more than 50-fold to 10 nM. Site-specific and efficient ‘traceless’ protein modification by high-affinity trans-splicing is demonstrated at nanomolar concentrations in living mammalian cells.
Collapse
Affiliation(s)
- M Braner
- Institute of Biochemistry, Biocenter, and Cluster of Excellence - Macromolecular Complexes , Goethe-University Frankfurt , Max-von-Laue-Str. 9 , 60438 Frankfurt/M. , Germany .
| | - A Kollmannsperger
- Institute of Biochemistry, Biocenter, and Cluster of Excellence - Macromolecular Complexes , Goethe-University Frankfurt , Max-von-Laue-Str. 9 , 60438 Frankfurt/M. , Germany .
| | - R Wieneke
- Institute of Biochemistry, Biocenter, and Cluster of Excellence - Macromolecular Complexes , Goethe-University Frankfurt , Max-von-Laue-Str. 9 , 60438 Frankfurt/M. , Germany .
| | - R Tampé
- Institute of Biochemistry, Biocenter, and Cluster of Excellence - Macromolecular Complexes , Goethe-University Frankfurt , Max-von-Laue-Str. 9 , 60438 Frankfurt/M. , Germany .
| |
Collapse
|
25
|
Postsynthetic Domain Assembly with NpuDnaE and SspDnaB Split Inteins. Appl Biochem Biotechnol 2015; 177:1137-51. [DOI: 10.1007/s12010-015-1802-0] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2015] [Accepted: 08/04/2015] [Indexed: 10/23/2022]
|
26
|
Chamoun-Emanuelli AM, Wright G, Roger S, Münch RC, Buchholz CJ, Chen Z. In vitro incorporation of a cell-binding protein to a lentiviral vector using an engineered split intein enables targeted delivery of genetic cargo. Biotechnol Bioeng 2015; 112:2611-7. [PMID: 26108964 DOI: 10.1002/bit.25685] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2015] [Revised: 05/28/2015] [Accepted: 06/15/2015] [Indexed: 01/22/2023]
Abstract
Gene therapy represents a promising therapeutic paradigm for addressing many disorders, but the absence of a vector that can be robustly and reproducibly functionalized with cell-homing functionality to mediate the delivery of genetic cargo specifically to target cells following systemic administration has stood as a major impediment. In this study, a high-affinity protein-protein pair comprising a splicing-deficient naturally split intein was used as molecular Velcro to append a HER2/neu-binding protein (DARPin) onto the surface of a binding-deficient, fusion-competent lentivirus. HER2/neu-specific lentiviruses created using this in vitro pseudotyping approach were able to deliver their genetic reporter cargo specifically to cells that express the target receptor at high levels in a co-culture. We envision that the described technology could provide a powerful, broadly applicable platform for the incorporation of cell-targeting functionality onto viral vectors.
Collapse
Affiliation(s)
- Ana M Chamoun-Emanuelli
- Department of Microbial and Molecular Pathogenesis, Texas A&M Health Science Center, College Station, Texas, 77843
| | - Gus Wright
- Department of Veterinary Physiology and Pharmacology, Texas A&M University, College Station, Texas
| | - Smith Roger
- Department of Veterinary Physiology and Pharmacology, Texas A&M University, College Station, Texas
| | - Robert C Münch
- Division of Medical Biotechnology, Paul-Ehrlich-Institut, Langen, Germany
| | | | - Zhilei Chen
- Department of Microbial and Molecular Pathogenesis, Texas A&M Health Science Center, College Station, Texas, 77843.
| |
Collapse
|
27
|
Truong DJJ, Kühner K, Kühn R, Werfel S, Engelhardt S, Wurst W, Ortiz O. Development of an intein-mediated split-Cas9 system for gene therapy. Nucleic Acids Res 2015; 43:6450-8. [PMID: 26082496 PMCID: PMC4513872 DOI: 10.1093/nar/gkv601] [Citation(s) in RCA: 242] [Impact Index Per Article: 26.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2014] [Accepted: 05/26/2015] [Indexed: 11/14/2022] Open
Abstract
Using CRISPR/Cas9, it is possible to target virtually any gene in any organism. A major limitation to its application in gene therapy is the size of Cas9 (>4 kb), impeding its efficient delivery via recombinant adeno-associated virus (rAAV). Therefore, we developed a split–Cas9 system, bypassing the packaging limit using split-inteins. Each Cas9 half was fused to the corresponding split-intein moiety and, only upon co-expression, the intein-mediated trans-splicing occurs and the full Cas9 protein is reconstituted. We demonstrated that the nuclease activity of our split-intein system is comparable to wild-type Cas9, shown by a genome-integrated surrogate reporter and by targeting three different endogenous genes. An analogously designed split-Cas9D10A nickase version showed similar activity as Cas9D10A. Moreover, we showed that the double nick strategy increased the homologous directed recombination (HDR). In addition, we explored the possibility of delivering the repair template accommodated on the same dual-plasmid system, by transient transfection, showing an efficient HDR. Most importantly, we revealed for the first time that intein-mediated split–Cas9 can be packaged, delivered and its nuclease activity reconstituted efficiently, in cells via rAAV.
Collapse
Affiliation(s)
- Dong-Jiunn Jeffery Truong
- Institute of Developmental Genetics, Helmholtz Zentrum München, German Research Center for Environmental Health, Munich 85764, Germany Institute of Developmental Genetics,Technische Universität München, Freising-Weihenstephan 85354, Germany
| | - Karin Kühner
- Institute of Developmental Genetics, Helmholtz Zentrum München, German Research Center for Environmental Health, Munich 85764, Germany
| | - Ralf Kühn
- Institute of Developmental Genetics, Helmholtz Zentrum München, German Research Center for Environmental Health, Munich 85764, Germany Max Delbrück Center for Molecular Medicine (MDC), Berlin 13125, Germany
| | - Stanislas Werfel
- Institute of Pharmacology and Toxicology. Technische Universität München, Munich 80802, Germany
| | - Stefan Engelhardt
- Institute of Pharmacology and Toxicology. Technische Universität München, Munich 80802, Germany German Center for Cardiovascular Research (DZHK), partner site Munich Heart Alliance, Munich 80802, Germany
| | - Wolfgang Wurst
- Institute of Developmental Genetics, Helmholtz Zentrum München, German Research Center for Environmental Health, Munich 85764, Germany Institute of Developmental Genetics,Technische Universität München, Freising-Weihenstephan 85354, Germany Deutsches Zentrum für Neurodegenerative Erkrankungen (DZNE), Munich 80336, Germany Munich Cluster for Systems Neurology (SyNergy) Adolf-Butenandt-Institut Ludwig-Maximilians-Universität München, Munich 81377, Germany
| | - Oskar Ortiz
- Institute of Developmental Genetics, Helmholtz Zentrum München, German Research Center for Environmental Health, Munich 85764, Germany
| |
Collapse
|
28
|
Abstract
Protein splicing in trans by split inteins has increasingly become a powerful protein-engineering tool for protein ligation, both in vivo and in vitro. Over 100 naturally occurring and artificially engineered split inteins have been reported for protein ligation using protein trans-splicing. Here, we review the current status of the reported split inteins in order to delineate an empirical or rational strategy for constructing new split inteins suitable for various applications in biotechnology and chemical biology.
Collapse
Affiliation(s)
- A Sesilja Aranko
- Research Program in Structural Biology and Biophysics, Institute of Biotechnology, University of Helsinki, P.O. Box 65, Helsinki FIN-00014, Finland
| | - Alexander Wlodawer
- Macromolecular Crystallography Laboratory, National Cancer Institute-Frederick, MD 21702, USA
| | - Hideo Iwaï
- Research Program in Structural Biology and Biophysics, Institute of Biotechnology, University of Helsinki, P.O. Box 65, Helsinki FIN-00014, Finland
| |
Collapse
|
29
|
Bachmann AL, Matern JCJ, Schütz V, Mootz HD. Chemical-tag labeling of proteins using fully recombinant split inteins. Methods Mol Biol 2015; 1266:145-159. [PMID: 25560073 DOI: 10.1007/978-1-4939-2272-7_10] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/04/2023]
Abstract
Chemical-tag labeling of proteins involving split inteins is an approach for the selective chemical modification of proteins without the requirement of any chemical synthesis to be performed. In a two-step protocol, a very short tag fused to a split intein auxiliary protein is first labeled in a bioconjugation reaction with a synthetic moiety either at its N-terminus (amine-tag) or at the side chain of an unnatural amino acid (click-tag). The labeled protein is then mixed with the protein of interest fused to the complementary intein fragment. In the resulting spontaneous protein trans-splicing reaction the split intein fragments remove themselves and ligate the tag to the protein of interest in a virtually traceless fashion. The reaction can be performed either using a purified protein of interest or to label a protein in the context of a living cell. All protein components are recombinantly expressed and all chemical reagents are commercially available.
Collapse
Affiliation(s)
- Anne-Lena Bachmann
- Department of Chemistry and Pharmacy, Institute of Biochemistry, University of Muenster, Wilhelm-Klemm-Str. 2, 48149, Münster, Germany
| | | | | | | |
Collapse
|
30
|
Aranko AS, Oeemig JS, Zhou D, Kajander T, Wlodawer A, Iwaï H. Structure-based engineering and comparison of novel split inteins for protein ligation. MOLECULAR BIOSYSTEMS 2014; 10:1023-34. [PMID: 24574026 PMCID: PMC7709711 DOI: 10.1039/c4mb00021h] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Protein splicing is an autocatalytic process involving self-excision of an internal protein domain, the intein, and concomitant ligation of the two flanking sequences, the exteins, with a peptide bond. Protein splicing can also take place in trans by naturally split inteins or artificially split inteins, ligating the exteins on two different polypeptide chains into one polypeptide chain. Protein trans-splicing could work in foreign contexts by replacing the native extein sequences with other protein sequences. Protein ligation using protein trans-splicing increasingly becomes a useful tool for biotechnological applications such as semi-synthesis of proteins, segmental isotopic labeling, and in vivo protein engineering. However, only a few split inteins have been successfully applied for protein ligation. Naturally split inteins have been widely used, but they are cross-reactive to each other, limiting their applications to multiple-fragment ligation. Based on the three-dimensional structures including two newly determined intein structures, we derived 21 new split inteins from four highly efficient cis-splicing inteins, in order to develop novel split inteins suitable for protein ligation. We systematically compared trans-splicing of 24 split inteins and tested the cross-activities among them to identify orthogonal split intein fragments that could be used in chemical biology and biotechnological applications.
Collapse
Affiliation(s)
- A Sesilja Aranko
- Research Program in Structural Biology and Biophysics, Institute of Biotechnology, University of Helsinki, P.O. Box 65, Helsinki, FIN-00014, Finland.
| | | | | | | | | | | |
Collapse
|
31
|
Eryilmaz E, Shah NH, Muir TW, Cowburn D. Structural and dynamical features of inteins and implications on protein splicing. J Biol Chem 2014; 289:14506-11. [PMID: 24695731 DOI: 10.1074/jbc.r113.540302] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Protein splicing is a posttranslational modification where intervening proteins (inteins) cleave themselves from larger precursor proteins and ligate their flanking polypeptides (exteins) through a multistep chemical reaction. First thought to be an anomaly found in only a few organisms, protein splicing by inteins has since been observed in microorganisms from all domains of life. Despite this broad phylogenetic distribution, all inteins share common structural features such as a horseshoe-like pseudo two-fold symmetric fold, several canonical sequence motifs, and similar splicing mechanisms. Intriguingly, the splicing efficiencies and substrate specificity of different inteins vary considerably, reflecting subtle changes in the chemical mechanism of splicing, linked to their local structure and dynamics. As intein chemistry has widespread use in protein chemistry, understanding the structural and dynamical aspects of inteins is crucial for intein engineering and the improvement of intein-based technologies.
Collapse
Affiliation(s)
- Ertan Eryilmaz
- From the Department of Biochemistry, Albert Einstein College of Medicine, Bronx, New York 10461 and
| | - Neel H Shah
- the Department of Chemistry, Frick Laboratory, Princeton University, Princeton, New Jersey 08544
| | - Tom W Muir
- the Department of Chemistry, Frick Laboratory, Princeton University, Princeton, New Jersey 08544
| | - David Cowburn
- From the Department of Biochemistry, Albert Einstein College of Medicine, Bronx, New York 10461 and
| |
Collapse
|
32
|
Schütz V, Mootz HD. Click-tag and amine-tag: chemical tag approaches for efficient protein labeling in vitro and on live cells using the naturally split Npu DnaE intein. Angew Chem Int Ed Engl 2014; 53:4113-7. [PMID: 24615830 DOI: 10.1002/anie.201309396] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2013] [Revised: 12/16/2013] [Indexed: 11/07/2022]
Abstract
Protein labeling with synthetic moieties remains in many cases a technically challenging or unresolved task. Two new and simple concepts are presented. In both approaches, a very short tag of only a few amino acids is prepared with the desired chemical modification and, in a second step, it is transferred to the protein of interest by protein trans-splicing. For the amine-tag, a recombinant intein fragment free of lysine residues was generated such that the amine group of the N terminus could be selectively modified with regular amine-reactive reagents. Thus, standard bioconjugation procedures without any chemical synthesis could be applied without modification of lysines in the protein of interest. For the click-tag, protein trans-splicing was combined with unnatural amino acid mutagenesis and subsequent bioorthogonal side chain modification, as demonstrated for click chemistry using p-azidophenylalanine. By the two-step strategy, exposure of the protein of interest to the copper catalyst was avoided.
Collapse
Affiliation(s)
- Vivien Schütz
- Institute of Biochemistry, University of Muenster, Wilhelm-Klemm-Strasse 2, 48149 Münster (Germany)
| | | |
Collapse
|
33
|
Schütz V, Mootz HD. Click-Tag and Amine-Tag: Chemical Tag Approaches for Efficient Protein Labeling In Vitro and on Live Cells using the Naturally SplitNpuDnaE Intein. Angew Chem Int Ed Engl 2014. [DOI: 10.1002/ange.201309396] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
|
34
|
Abstract
Inteins are auto-processing domains found in organisms from all domains of life. These proteins carry out a process known as protein splicing, which is a multi-step biochemical reaction comprised of both the cleavage and formation of peptide bonds. While the endogenous substrates of protein splicing are specific essential proteins found in intein-containing host organisms, inteins are also functional in exogenous contexts and can be used to chemically manipulate virtually any polypeptide backbone. Given this, protein chemists have exploited various facets of intein reactivity to modify proteins in myriad ways for both basic biological research as well as potential therapeutic applications. Here, we review the intein field, first focusing on the biological context and phylogenetic diversity of inteins, followed by a description of intein structure and biochemical function. Finally, we discuss prevalent inteinbased technologies, focusing on their applications in chemical biology, followed by persistent caveats of intein chemistry and approaches to alleviate these shortcomings. The findings summarized herein describe two and a half decades of research, leading from a biochemical curiosity to the development of powerful protein engineering tools.
Collapse
Affiliation(s)
- Neel H Shah
- Department of Chemistry, Princeton University, Frick Laboratory, Princeton, NJ 08544, United States
| | - Tom W Muir
- Department of Chemistry, Princeton University, Frick Laboratory, Princeton, NJ 08544, United States
| |
Collapse
|
35
|
Shah NH, Eryilmaz E, Cowburn D, Muir TW. Naturally split inteins assemble through a "capture and collapse" mechanism. J Am Chem Soc 2013; 135:18673-81. [PMID: 24236406 PMCID: PMC3865799 DOI: 10.1021/ja4104364] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
![]()
Split
inteins are a class of naturally occurring proteins that
carry out protein splicing in trans. The chemical
mechanism of protein trans-splicing is well-understood
and has been exploited to develop several powerful protein engineering
technologies. Split intein chemistry is preceded by efficient molecular
recognition between two protomers that become intertwined in their
bound state. It is currently unclear how this unique topology is achieved
upon fragment association. Using biophysical techniques in conjunction
with protein engineering methods, including segmental isotopic labeling,
we show that one split intein fragment is partly folded, while the
other is completely disordered. These polypeptides capture each other
through their disordered regions and form an ordered intermediate
with native-like structure at their interface. This intermediate then
collapses into the canonical intein fold. This mechanism provides
insight into the evolutionary constraints on split intein assembly
and should enhance the development of split intein-based technologies.
Collapse
Affiliation(s)
- Neel H Shah
- Department of Chemistry, Princeton University , Frick Laboratory, Princeton, New Jersey 08544, United States
| | | | | | | |
Collapse
|
36
|
Arthur C. Cope Scholar Awards: W. R. Dichtel, M. Fujita, M. J. Krische, D. S. Matteson, B. S. Moore, T. W. Muir, J. R. Norton, S. E. Reisman, M. F. Semmelhack, S. S. Stahl. Angew Chem Int Ed Engl 2013. [DOI: 10.1002/anie.201305019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
|
37
|
Arthur C. Cope Scholar Awards: W. R. Dichtel, M. Fujita, M. J. Krische, D. S. Matteson, B. S. Moore, T. W. Muir, J. R. Norton, S. E. Reisman, M. F. Semmelhack, S. S. Stahl. Angew Chem Int Ed Engl 2013. [DOI: 10.1002/ange.201305019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
|
38
|
Sorci M, Dassa B, Liu H, Anand G, Dutta AK, Pietrokovski S, Belfort M, Belfort G. Oriented covalent immobilization of antibodies for measurement of intermolecular binding forces between zipper-like contact surfaces of split inteins. Anal Chem 2013; 85:6080-8. [PMID: 23679912 PMCID: PMC3760192 DOI: 10.1021/ac400949t] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
In order to measure the intermolecular binding forces between two halves (or partners) of naturally split protein splicing elements called inteins, a novel thiol-hydrazide linker was designed and used to orient immobilized antibodies specific for each partner. Activation of the surfaces was achieved in one step, allowing direct intermolecular force measurement of the binding of the two partners of the split intein (called protein trans-splicing). Through this binding process, a whole functional intein is formed resulting in subsequent splicing. Atomic force microscopy (AFM) was used to directly measure the split intein partner binding at 1 μm/s between native (wild-type) and mixed pairs of C- and N-terminal partners of naturally occurring split inteins from three cyanobacteria. Native and mixed pairs exhibit similar binding forces within the error of the measurement technique (~52 pN). Bioinformatic sequence analysis and computational structural analysis discovered a zipper-like contact between the two partners with electrostatic and nonpolar attraction between multiple aligned ion pairs and hydrophobic residues. Also, we tested the Jarzynski's equality and demonstrated, as expected, that nonequilibrium dissipative measurements obtained here gave larger energies of interaction as compared with those for equilibrium. Hence, AFM coupled with our immobilization strategy and computational studies provides a useful analytical tool for the direct measurement of intermolecular association of split inteins and could be extended to any interacting protein pair.
Collapse
Affiliation(s)
- Mirco Sorci
- Howard P. Isermann Department of Chemical and Biological Engineering and Center for Biotechnology and Interdisciplinary Studies Rensselaer Polytechnic Institute, Troy, NY 12180
| | - Bareket Dassa
- Molecular Genetics Department, Weizmann Institute of Science, Rehovot 76100, Israel
| | - Hongwei Liu
- Howard P. Isermann Department of Chemical and Biological Engineering and Center for Biotechnology and Interdisciplinary Studies Rensselaer Polytechnic Institute, Troy, NY 12180
| | - Gaurav Anand
- Howard P. Isermann Department of Chemical and Biological Engineering and Center for Biotechnology and Interdisciplinary Studies Rensselaer Polytechnic Institute, Troy, NY 12180
| | - Amit K. Dutta
- Howard P. Isermann Department of Chemical and Biological Engineering and Center for Biotechnology and Interdisciplinary Studies Rensselaer Polytechnic Institute, Troy, NY 12180
| | - Shmuel Pietrokovski
- Molecular Genetics Department, Weizmann Institute of Science, Rehovot 76100, Israel
| | - Marlene Belfort
- Department of Biological Sciences, University at Albany, SUNY, Albany, NY 12222
| | - Georges Belfort
- Howard P. Isermann Department of Chemical and Biological Engineering and Center for Biotechnology and Interdisciplinary Studies Rensselaer Polytechnic Institute, Troy, NY 12180
| |
Collapse
|
39
|
Abstract
Inteins are intervening protein sequences that undergo self-excision from a precursor protein with the concomitant ligation of the flanking polypeptides. Split inteins are expressed in two separated halves, and the recognition and association of two halves are the first crucial step for initiating trans-splicing. In the present study, we carried out the structural and thermodynamic analysis on the interaction of two halves of DnaE split intein from Synechocystis sp. PCC6803. Both isolated halves (IN and IC) are disordered and undergo conformational transition from disorder to order upon association. ITC (isothermal titration calorimetry) reveals that the highly favourable enthalpy change drives the association of the two halves, overcoming the unfavourable entropy change. The high flexibility of two fragments and the marked thermodynamic preference provide a robust association for the formation of the well-folded IN/IC complex, which is the basis for reconstituting the trans-splicing activity of DnaE split intein.
Collapse
|
40
|
Shah NH, Eryilmaz E, Cowburn D, Muir TW. Extein residues play an intimate role in the rate-limiting step of protein trans-splicing. J Am Chem Soc 2013; 135:5839-47. [PMID: 23506399 PMCID: PMC3630739 DOI: 10.1021/ja401015p] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
![]()
Split
inteins play an important role in modern protein semisynthesis
techniques. These naturally occurring protein splicing domains can
be used for in vitro and in vivo protein modification, peptide and
protein cyclization, segmental isotopic labeling, and the construction
of biosensors. The most well-characterized family of split inteins,
the cyanobacterial DnaE inteins, show particular promise, as many
of these can splice proteins in less than 1 min. Despite this fact,
the activity of these inteins is context-dependent: certain peptide
sequences surrounding their ligation junction (called local N- and
C-exteins) are strongly preferred, while other sequences cause a dramatic
reduction in the splicing kinetics and yield. These sequence constraints
limit the utility of inteins, and thus, a more detailed understanding
of their participation in protein splicing is needed. Here we present
a thorough kinetic analysis of the relationship between C-extein composition
and split intein activity. The results of these experiments were used
to guide structural and molecular dynamics studies, which revealed
that the motions of catalytic residues are constrained by the second
C-extein residue, likely forcing them into an active conformation
that promotes rapid protein splicing. Together, our structural and
functional studies also highlight a key region of the intein structure
that can be re-engineered to increase intein promiscuity.
Collapse
Affiliation(s)
- Neel H Shah
- Department of Chemistry, Princeton University, Frick Laboratory, Princeton, New Jersey 08544, USA
| | | | | | | |
Collapse
|
41
|
Volkmann G, Mootz HD. Recent progress in intein research: from mechanism to directed evolution and applications. Cell Mol Life Sci 2013; 70:1185-206. [PMID: 22926412 PMCID: PMC11113529 DOI: 10.1007/s00018-012-1120-4] [Citation(s) in RCA: 77] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2011] [Revised: 07/23/2012] [Accepted: 08/06/2012] [Indexed: 10/27/2022]
Abstract
Inteins catalyze a post-translational modification known as protein splicing, where the intein removes itself from a precursor protein and concomitantly ligates the flanking protein sequences with a peptide bond. Over the past two decades, inteins have risen from a peculiarity to a rich source of applications in biotechnology, biomedicine, and protein chemistry. In this review, we focus on developments of intein-related research spanning the last 5 years, including the three different splicing mechanisms and their molecular underpinnings, the directed evolution of inteins towards improved splicing in exogenous protein contexts, as well as novel applications of inteins for cell biology and protein engineering, which were made possible by a clearer understanding of the protein splicing mechanism.
Collapse
Affiliation(s)
- Gerrit Volkmann
- Institute of Biochemistry, University of Münster, Wilhelm-Klemm-Str. 2, 48149 Münster, Germany
| | - Henning D. Mootz
- Institute of Biochemistry, University of Münster, Wilhelm-Klemm-Str. 2, 48149 Münster, Germany
| |
Collapse
|
42
|
Jagadish K, Borra R, Lacey V, Majumder S, Shekhtman A, Wang L, Camarero JA. Expression of fluorescent cyclotides using protein trans-splicing for easy monitoring of cyclotide-protein interactions. Angew Chem Int Ed Engl 2013; 52:3126-31. [PMID: 23322720 PMCID: PMC3767473 DOI: 10.1002/anie.201209219] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2012] [Indexed: 11/09/2022]
Affiliation(s)
- Krishnappa Jagadish
- Department of Pharmacology and Pharmaceutical Sciences, University of Southern California, Los Angeles, CA 90033, USA
| | - Radikha Borra
- Department of Pharmacology and Pharmaceutical Sciences, University of Southern California, Los Angeles, CA 90033, USA
| | - Vanessa Lacey
- Jack H. Skirball Center for Chemical Biology and Proteomics, The Salk Institute for Biological Studies, La Jolla, CA 92037, USA
| | - Subhabrata Majumder
- Department of Chemistry, State University of New York, Albany, NY 12222, USA
| | - Alexander Shekhtman
- Department of Chemistry, State University of New York, Albany, NY 12222, USA
| | - Lei Wang
- Jack H. Skirball Center for Chemical Biology and Proteomics, The Salk Institute for Biological Studies, La Jolla, CA 92037, USA
| | - Julio A. Camarero
- Department of Pharmacology and Pharmaceutical Sciences, University of Southern California, Los Angeles, CA 90033, USA
- Department of Chemistry, University of Southern California, Los Angeles, CA 90033, USA
| |
Collapse
|
43
|
Wong S, Mills E, Truong K. Simultaneous assembly of two target proteins using split inteins for live cell imaging. Protein Eng Des Sel 2013; 26:207-13. [PMID: 23223808 DOI: 10.1093/protein/gzs100] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Inteins are protein elements that covalently reassemble proteins from two precursor fragments in a process known as protein splicing. They are commonly used to reassemble a single target protein by protein splicing, but a second target protein can potentially reassemble by intein dimerization. Here, we use the naturally occurring split DnaE intein from Nostoc punctiforme (NpuDnaE) to demonstrate the simultaneous assembly of two target proteins in several examples studied with live cell imaging: yellow fluorescent protein (YFP) with monomeric red fluorescent protein (mRFP), dominant positive mutant of RhoA GTPase with YFP and GCaMP2 Ca(2+) indicator with mRFP. These examples showed the versatility of the strategy along with some interesting attributes: first, the two target proteins are in equal stoichiometry; second, the extent of protein splicing can be reported by a fluorescent protein. In particular, the split GCaMP2 with mRFP could find applications in tissue-specific Ca(2+) imaging in transgenic organisms, where mRFP could control for motion-related intensity changes.
Collapse
Affiliation(s)
- Stanley Wong
- Institute of Biomaterials and Biomedical Engineering, University of Toronto, Toronto, Ontario, Canada
| | | | | |
Collapse
|
44
|
Jagadish K, Borra R, Lacey V, Majumder S, Shekhtman A, Wang L, Camarero JA. Expression of Fluorescent Cyclotides using Protein Trans-Splicing for Easy Monitoring of Cyclotide-Protein Interactions. Angew Chem Int Ed Engl 2013. [DOI: 10.1002/ange.201209219] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
|
45
|
Vila-Perelló M, Liu Z, Shah NH, Willis JA, Idoyaga J, Muir TW. Streamlined expressed protein ligation using split inteins. J Am Chem Soc 2012; 135:286-92. [PMID: 23265282 PMCID: PMC3544275 DOI: 10.1021/ja309126m] [Citation(s) in RCA: 76] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
Chemically modified proteins are invaluable tools for studying the molecular details of biological processes, and they also hold great potential as new therapeutic agents. Several methods have been developed for the site-specific modification of proteins, one of the most widely used being expressed protein ligation (EPL) in which a recombinant α-thioester is ligated to an N-terminal Cys-containing peptide. Despite the widespread use of EPL, the generation and isolation of the required recombinant protein α-thioesters remain challenging. We describe here a new method for the preparation and purification of recombinant protein α-thioesters using engineered versions of naturally split DnaE inteins. This family of autoprocessing enzymes is closely related to the inteins currently used for protein α-thioester generation, but they feature faster kinetics and are split into two inactive polypeptides that need to associate to become active. Taking advantage of the strong affinity between the two split intein fragments, we devised a streamlined procedure for the purification and generation of protein α-thioesters from cell lysates and applied this strategy for the semisynthesis of a variety of proteins including an acetylated histone and a site-specifically modified monoclonal antibody.
Collapse
Affiliation(s)
- Miquel Vila-Perelló
- Department of Chemistry, Princeton University, Frick Laboratory, Princeton, New Jersey 08544, United States
| | | | | | | | | | | |
Collapse
|
46
|
Ramirez M, Valdes N, Guan D, Chen Z. Engineering split intein DnaE from Nostoc punctiforme for rapid protein purification. Protein Eng Des Sel 2012; 26:215-23. [PMID: 23223807 DOI: 10.1093/protein/gzs097] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
We report the engineering of a DnaE intein able to catalyze rapid C-terminal cleavage in the absence of N-terminal cleavage. A single mutation in DnaE intein from Nostoc punctiforme PCC73102 (NpuDnaE), Asp118Gly, was introduced based on sequence alignment with a previously engineered C-terminal cleaving intein mini-MtuRecA. This mutation was able to both suppress N-terminal cleavage and significantly elevate C-terminal cleavage efficiency. Molecular modeling suggests that in NpuDnaE Asp118 forms a hydrogen bond with the penultimate Asn, preventing its spontaneous cyclization prior to N-terminal cleavage. Mutation of Asp118 to Gly essentially abolishes this restriction leading to subsequent C-terminal cleavage in the absence of N-terminal cleavage. The Gly118 NpuDnaE mutant exhibits rapid thio-dependent C-terminal cleavage kinetics with 80% completion within 3 h at room temperature. We used this newly engineered intein to develop both column-free and chromatography-based protein purification methods utilizing the elastin-like-polypeptide and chitin-binding protein as removable purification tags, respectively. We demonstrate rapid target protein purification to electrophoretic purity at yields up to 84 mg per liter of Escherichia coli culture.
Collapse
Affiliation(s)
- Miguel Ramirez
- Artie McFerrin Department of Chemical Engineering, Texas A&M University, College Station, TX 77843, USA
| | | | | | | |
Collapse
|
47
|
Photocrosslinking approaches to interactome mapping. Curr Opin Chem Biol 2012; 17:90-101. [PMID: 23149092 DOI: 10.1016/j.cbpa.2012.10.034] [Citation(s) in RCA: 107] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2012] [Accepted: 10/22/2012] [Indexed: 11/21/2022]
Abstract
Photocrosslinking approaches can be used to map interactome networks within the context of living cells. Photocrosslinking methods rely on use of metabolic engineering or genetic code expansion to incorporate photocrosslinking analogs of amino acids or sugars into cellular biomolecules. Immunological and mass spectrometry techniques are used to analyze crosslinked complexes, thereby defining specific interactomes. Because photocrosslinking can be conducted in native, cellular settings, it can be used to define context-dependent interactions. Photocrosslinking methods are also ideally suited for determining interactome dynamics, mapping interaction interfaces, and identifying transient interactions in which intrinsically disordered proteins and glycoproteins engage. Here we discuss the application of cell-based photocrosslinking to the study of specific problems in immune cell signaling, transcription, membrane protein dynamics, nucleocytoplasmic transport, and chaperone-assisted protein folding.
Collapse
|
48
|
Circular permutation prediction reveals a viable backbone disconnection for split proteins: an approach in identifying a new functional split intein. PLoS One 2012; 7:e43820. [PMID: 22937103 PMCID: PMC3427171 DOI: 10.1371/journal.pone.0043820] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2012] [Accepted: 07/26/2012] [Indexed: 01/30/2023] Open
Abstract
Split-protein systems have emerged as a powerful tool for detecting biomolecular interactions and reporting biological reactions. However, reliable methods for identifying viable split sites are still unavailable. In this study, we demonstrated the feasibility that valid circular permutation (CP) sites in proteins have the potential to act as split sites and that CP prediction can be used to search for internal permissive sites for creating new split proteins. Using a protein ligase, intein, as a model, CP predictor facilitated the creation of circular permutants in which backbone opening imposes the least detrimental effects on intein folding. We screened a series of predicted intein CPs and identified stable and native-fold CPs. When the valid CP sites were introduced as split sites, there was a reduction in folding enthalpy caused by the new backbone opening; however, the coincident loss in entropy was sufficient to be compensated, yielding a favorable free energy for self-association. Since split intein is exploited in protein semi-synthesis, we tested the related protein trans-splicing (PTS) activities of the corresponding split inteins. Notably, a novel functional split intein composed of the N-terminal 36 residues combined with the remaining C-terminal fragment was identified. Its PTS activity was shown to be better than current reported two-piece intein with a short N-terminal segment. Thus, the incorporation of in silico CP prediction facilitated the design of split intein as well as circular permutants.
Collapse
|
49
|
Oeemig JS, Zhou D, Kajander T, Wlodawer A, Iwaï H. NMR and crystal structures of the Pyrococcus horikoshii RadA intein guide a strategy for engineering a highly efficient and promiscuous intein. J Mol Biol 2012; 421:85-99. [PMID: 22560994 PMCID: PMC3392434 DOI: 10.1016/j.jmb.2012.04.029] [Citation(s) in RCA: 55] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2012] [Revised: 04/25/2012] [Accepted: 04/27/2012] [Indexed: 11/16/2022]
Abstract
In protein splicing, an intervening protein sequence (intein) in the host protein excises itself out and ligates two split host protein sequences (exteins) to produce a mature host protein. Inteins require the involvement for the splicing of the first residue of the extein that follows the intein (which is Cys, Ser, or Thr). Other extein residues near the splicing junctions could modulate splicing efficiency even when they are not directly involved in catalysis. Mutual interdependence between this molecular parasite (intein) and its host protein (exteins) is not beneficial for intein spread but could be advantageous for intein survival during evolution. Elucidating extein-intein dependency has increasingly become important since inteins are recognized as useful biotechnological tools for protein ligation. We determined the structures of one of inteins with high splicing efficiency, the RadA intein from Pyrococcus horikoshii (PhoRadA). The solution NMR structure and the crystal structures elucidated the structural basis for its high efficiency and directed our efforts of engineering that led to rational design of a functional minimized RadA intein. The crystal structure of the minimized RadA intein also revealed the precise interactions between N-extein and the intein. We systematically analyzed the effects at the -1 position of N-extein and were able to significantly improve the splicing efficiency of a less robust splicing variant by eliminating the unfavorable extein-intein interactions observed in the structure. This work provides an example of how unveiling structure-function relationships of inteins offer a promising way of improving their properties as better tools for protein engineering.
Collapse
Affiliation(s)
- Jesper S. Oeemig
- Research Program in Structural Biology and Biophysics, Institute of Biotechnology, University of Helsinki. P.O. Box 65, Helsinki, FIN-00014, Finland
| | - Dongwen Zhou
- Macromolecular Crystallography Laboratory, National Cancer Institute at Frederick, Frederick, MD, 21702, USA
| | - Tommi Kajander
- Research Program in Structural Biology and Biophysics, Institute of Biotechnology, University of Helsinki. P.O. Box 65, Helsinki, FIN-00014, Finland
| | - Alexander Wlodawer
- Macromolecular Crystallography Laboratory, National Cancer Institute at Frederick, Frederick, MD, 21702, USA
| | - Hideo Iwaï
- Research Program in Structural Biology and Biophysics, Institute of Biotechnology, University of Helsinki. P.O. Box 65, Helsinki, FIN-00014, Finland
| |
Collapse
|
50
|
Shah NH, Dann GP, Vila-Perelló M, Liu Z, Muir TW. Ultrafast protein splicing is common among cyanobacterial split inteins: implications for protein engineering. J Am Chem Soc 2012; 134:11338-41. [PMID: 22734434 DOI: 10.1021/ja303226x] [Citation(s) in RCA: 112] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
We describe the first systematic study of a family of inteins, the split DnaE inteins from cyanobacteria. By measuring in vivo splicing efficiencies and in vitro kinetics, we demonstrate that several inteins can catalyze protein trans-splicing in tens of seconds rather than hours, as is commonly observed for this autoprocessing protein family. Furthermore, we show that when artificially fused, these inteins can be used for rapid generation of protein α-thioesters for expressed protein ligation. This comprehensive survey of split inteins provides indispensable information for the development and improvement of intein-based tools for chemical biology.
Collapse
Affiliation(s)
- Neel H Shah
- Department of Chemistry, Princeton University, 325 Frick Chemistry Laboratory, Princeton, New Jersey 08544, USA
| | | | | | | | | |
Collapse
|