151
|
Hu J, Chen X, Zhang X, Yuan X, Yang M, Dai H, Yang W, Zhou Q, Wen W, Wang Q, Qin W, Zhao A. A fusion-protein approach enabling mammalian cell production of tumor targeting protein domains for therapeutic development. Protein Sci 2018; 27:933-944. [PMID: 29500915 PMCID: PMC5916118 DOI: 10.1002/pro.3399] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2017] [Revised: 02/11/2018] [Accepted: 03/01/2018] [Indexed: 02/05/2023]
Abstract
A single chain Fv fragment (scFv) is a fusion of the variable regions of heavy (VH ) and light (VL ) chains of immunoglobulins. They are important elements of chimeric antigen receptors for cancer therapy. We sought to produce a panel of 16 extracellular protein domains of tumor markers for use in scFv yeast library screenings. A series of vectors comprising various combinations of expression elements was made, but expression was unpredictable and more than half of the protein domains could not be produced using any of the constructs. Here we describe a novel fusion expression system based on mouse TEM7 (tumor endothelial marker 7), which could facilitate protein expression. With this approach we could produce all but one of the tumor marker domains that could not otherwise be expressed. In addition, we demonstrated that the tumor associated antigen hFZD10 produced as a fusion protein with mTEM7 could be used to enrich scFv antibodies from a yeast display library. Collectively our study demonstrates the potential of specific fusion proteins based on mTEM7 in enabling mammalian cell production of tumor targeting protein domains for therapeutic development.
Collapse
Affiliation(s)
- Jia Hu
- Lung Cancer Research CenterWest China Hospital, Sichuan UniversityChengduChina
- Abramson Cancer Center, Perelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPennsylvania
| | - Xiang Chen
- Abramson Cancer Center, Perelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPennsylvania
| | - Xuhua Zhang
- Abramson Cancer Center, Perelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPennsylvania
- School of Life SciencesZhengzhou UniversityZhengzhouChina
| | - Xiaopeng Yuan
- Abramson Cancer Center, Perelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPennsylvania
- Zhujiang Hospital, SouthernMedical UniversityGuangzhouChina
| | - Mingjuan Yang
- Abramson Cancer Center, Perelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPennsylvania
| | - Hui Dai
- Xinjiang Karamay Central HospitalKaramay CityXinjiangChina
| | - Wei Yang
- Abramson Cancer Center, Perelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPennsylvania
| | - Qinghua Zhou
- Lung Cancer Research CenterWest China Hospital, Sichuan UniversityChengduChina
| | - Weihong Wen
- State Key Laboratory of Cancer Biology, Department of ImmunologyXijing Hospital, Fourth Military Medical UniversityChinaXi'an
| | - Qirui Wang
- Abramson Cancer Center, Perelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPennsylvania
- College of Traditional Chinese MedicineSouthernMedical UniversityGuang DongChina
| | - Weijun Qin
- Department of UrologyXijing Hospital, Fourth Military Medical UniversityChinaXi'an
| | - Aizhi Zhao
- Abramson Cancer Center, Perelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPennsylvania
| |
Collapse
|
152
|
Pane K, Verrillo M, Avitabile A, Pizzo E, Varcamonti M, Zanfardino A, Di Maro A, Rega C, Amoresano A, Izzo V, Di Donato A, Cafaro V, Notomista E. Chemical Cleavage of an Asp-Cys Sequence Allows Efficient Production of Recombinant Peptides with an N-Terminal Cysteine Residue. Bioconjug Chem 2018. [DOI: 10.1021/acs.bioconjchem.8b00083] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Affiliation(s)
| | - Mariavittoria Verrillo
- Department of Agricultural Sciences, Università degli Studi di Napoli Federico II, Via Università 100, Portici 80055, Italy
| | | | | | | | | | - Antimo Di Maro
- Department of Environmental, Biological and Pharmaceutical Sciences and Technologies, Università della Campania Luigi Vanvitelli, Via Vivaldi 43, Caserta 81100, Italy
| | - Camilla Rega
- Department of Environmental, Biological and Pharmaceutical Sciences and Technologies, Università della Campania Luigi Vanvitelli, Via Vivaldi 43, Caserta 81100, Italy
| | | | - Viviana Izzo
- Department of Medicine and Surgery, Università degli Studi di Salerno, via S. Allende, Baronissi 84081, Italy
| | | | | | | |
Collapse
|
153
|
Cooper MA, Taris JE, Shi C, Wood DW. A Convenient Split-Intein Tag Method for the Purification of Tagless Target Proteins. ACTA ACUST UNITED AC 2018. [PMID: 29516483 DOI: 10.1002/cpps.46] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
In this work, we describe a novel self-cleaving tag technology based on a highly modified split-intein cleaving element. In this system, the N-terminal segment of an engineered split intein is expressed in E. coli and covalently immobilized onto a capture resin, while the smaller C-terminal intein segment is fused to the N-terminus of the desired target protein. The tagged target can then be expressed in an appropriate expression system, without concern for premature intein cleaving. During the purification, strong association between the intein segments effectively captures the tagged target onto the capture resin while simultaneously generating a cleaving-competent intein complex. Once the complex is purified by washing the column, intein-mediated cleavage and release of the tagless target is induced with a simple shift in buffer pH from 8.5 to 6.2. The result is a convenient and effective method for the purification of traceless and tagless target proteins, which can be used in characterization and functional studies. © 2018 by John Wiley & Sons, Inc.
Collapse
Affiliation(s)
- Merideth A Cooper
- Department of Chemical and Biomolecular Engineering, The Ohio State University, Columbus, Ohio
| | - Joseph E Taris
- Department of Chemical and Biomolecular Engineering, The Ohio State University, Columbus, Ohio
| | - Changhua Shi
- Department of Chemical and Biomolecular Engineering, The Ohio State University, Columbus, Ohio
| | - David W Wood
- Department of Chemical and Biomolecular Engineering, The Ohio State University, Columbus, Ohio
| |
Collapse
|
154
|
Jia B, Jeon CO. High-throughput recombinant protein expression in Escherichia coli: current status and future perspectives. Open Biol 2017; 6:rsob.160196. [PMID: 27581654 PMCID: PMC5008019 DOI: 10.1098/rsob.160196] [Citation(s) in RCA: 194] [Impact Index Per Article: 27.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2016] [Accepted: 08/03/2016] [Indexed: 12/26/2022] Open
Abstract
The ease of genetic manipulation, low cost, rapid growth and number of previous studies have made Escherichia coli one of the most widely used microorganism species for producing recombinant proteins. In this post-genomic era, challenges remain to rapidly express and purify large numbers of proteins for academic and commercial purposes in a high-throughput manner. In this review, we describe several state-of-the-art approaches that are suitable for the cloning, expression and purification, conducted in parallel, of numerous molecules, and we discuss recent progress related to soluble protein expression, mRNA folding, fusion tags, post-translational modification and production of membrane proteins. Moreover, we address the ongoing efforts to overcome various challenges faced in protein expression in E. coli, which could lead to an improvement of the current system from trial and error to a predictable and rational design.
Collapse
Affiliation(s)
- Baolei Jia
- Department of Life Science, Chung-Ang University, Seoul 06974, Republic of Korea
| | - Che Ok Jeon
- Department of Life Science, Chung-Ang University, Seoul 06974, Republic of Korea
| |
Collapse
|
155
|
Krishnamoorthy A, Witkowski A, Tran JJ, Weers PMM, Ryan RO. Characterization of secondary structure and lipid binding behavior of N-terminal saposin like subdomain of human Wnt3a. Arch Biochem Biophys 2017; 630:38-46. [PMID: 28754322 DOI: 10.1016/j.abb.2017.07.015] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2017] [Revised: 07/14/2017] [Accepted: 07/24/2017] [Indexed: 02/06/2023]
Abstract
Wnt signaling is essential for embryonic development and adult homeostasis in multicellular organisms. A conserved feature among Wnt family proteins is the presence of two structural domains. Within the N-terminal (NT) domain there exists a motif that is superimposable upon saposin-like protein (SAPLIP) family members. SAPLIPs are found in plants, microbes and animals and possess lipid surface seeking activity. To investigate the function of the Wnt3a saposin-like subdomain (SLD), recombinant SLD was studied in isolation. Bacterial expression of this Wnt fragment was achieved only when the core SLD included 82 NT residues of Wnt3a (NT-SLD). Unlike SAPLIPs, NT-SLD required the presence of detergent to achieve solubility at neutral pH. Deletion of two hairpin loop extensions present in NT-SLD, but not other SAPLIPs, had no effect on the solubility properties of NT-SLD. Far UV circular dichroism spectroscopy of NT-SLD yielded 50-60% α-helix secondary structure. Limited proteolysis of isolated NT-SLD in buffer and detergent micelles showed no differences in cleavage kinetics. Unlike prototypical saposins, NT-SLD exhibited weak membrane-binding affinity and lacked cell lytic activity. In cell-based canonical Wnt signaling assays, NT-SLD was unable to induce stabilization of β-catenin or modulate the extent of β-catenin stabilization induced by full-length Wnt3a. Taken together, the results indicate neighboring structural elements within full-length Wnt3a affect SLD conformational stability. Moreover, SLD function(s) in Wnt proteins appear to have evolved away from those commonly attributed to SAPLIP family members.
Collapse
Affiliation(s)
- Aparna Krishnamoorthy
- Department of Nutritional Sciences and Toxicology, University of California Berkeley, Berkeley, CA 94720, USA
| | - Andrzej Witkowski
- Children's Hospital Oakland Research Institute, 5700 Martin Luther King Jr. Way, Oakland, CA 94609, USA
| | - Jesse J Tran
- Department of Chemistry and Biochemistry, California State University, Long Beach, CA 90840, USA
| | - Paul M M Weers
- Department of Chemistry and Biochemistry, California State University, Long Beach, CA 90840, USA
| | - Robert O Ryan
- Department of Nutritional Sciences and Toxicology, University of California Berkeley, Berkeley, CA 94720, USA; Children's Hospital Oakland Research Institute, 5700 Martin Luther King Jr. Way, Oakland, CA 94609, USA.
| |
Collapse
|
156
|
Petryayeva E, Jeen T, Algar WR. Optimization and Changes in the Mode of Proteolytic Turnover of Quantum Dot-Peptide Substrate Conjugates through Moderation of Interfacial Adsorption. ACS APPLIED MATERIALS & INTERFACES 2017; 9:30359-30372. [PMID: 28846381 DOI: 10.1021/acsami.7b07519] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
Abstract
Enzymes have many important roles in biology and industry, and proteases are one of the most important classes of enzymes. Semiconductor quantum dots (QDs) are attractive materials for developing protease activity probes because of their advantageous physical and optical properties; however, interactions between a protease and a QD conjugated with its substrate can affect the turnover of that substrate. Here, we study the turnover of multivalent QD-peptide substrate conjugates as a function of multiple parameters: (i) the ligand coating on the QD, including dihydrolipoic acid (DHLA), glutathione (GSH), DHLA-poly(ethylene glycol) (DHLA-PEG), and DHLA-zwitterionic sulfobetaine (DHLA-SB); (ii) the identity of the protease, including trypsin, thrombin, and plasmin; and (iii) the number of substrate and nonsubstrate biomacromolecules conjugated per QD. We show that limiting protease adsorption on QDs is critical for optimizing the turnover of conjugated peptide substrates. Protease adsorption is inhibitory, and very strong adsorption leads to an apparent "scooting" mode of activity with limited turnover. In contrast, with weaker adsorption, enhancements in the turnover rate likely result from a "hopping" mode of activity. The putative hopping mode is thought to feature processive turnover of all substrates in multivalent conjugates with a rate-limiting step of diffusion between individual conjugates, and the magnitude of such enhancements increases with decreases in adsorption. Although it was possible to passivate DHLA- and GSH-coated QDs with high densities of conjugated biomacromolecules, the most effective strategy for reducing adsorption was the substitution of these ligands. Whereas passivation incrementally increased turnover, DHLA-PEG and DHLA-SB ligands converted the mode of turnover with plasmin from scooting to hopping and the DHLA-SB enhanced the turnover rates with thrombin and trypsin by approximately an order of magnitude relative to GSH ligands. The new insights from the broad scope of this study provide an important framework for designing optimized QD conjugates as probes and sensors for enzyme activity.
Collapse
Affiliation(s)
- Eleonora Petryayeva
- Department of Chemistry, University of British Columbia , 2036 Main Mall, Vancouver, British Columbia V6T 1Z1, Canada
| | - Tiffany Jeen
- Department of Chemistry, University of British Columbia , 2036 Main Mall, Vancouver, British Columbia V6T 1Z1, Canada
| | - W Russ Algar
- Department of Chemistry, University of British Columbia , 2036 Main Mall, Vancouver, British Columbia V6T 1Z1, Canada
| |
Collapse
|
157
|
Cowell J, Buck M, Essa AH, Clarke R, Vollmer W, Vollmer D, Hilkens CM, Isaacs JD, Hall MJ, Gray J. Traceless Cleavage of Protein-Biotin Conjugates under Biologically Compatible Conditions. Chembiochem 2017; 18:1688-1691. [PMID: 28581639 PMCID: PMC5708275 DOI: 10.1002/cbic.201700214] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2017] [Indexed: 12/14/2022]
Abstract
Biotinylation of amines is widely used to conjugate biomolecules, but either the resulting label is non-removable or its removal leaves a tag on the molecule of interest, thus affecting downstream processes. We present here a set of reagents (RevAmines) that allow traceless, reversible biotinylation under biologically compatible, mild conditions. Release following avidin-based capture is achieved through the cleavage of a (2-(alkylsulfonyl)ethyl) carbamate linker under mild conditions (200 mm ammonium bicarbonate, pH 8, 16-24 h, room temperature) that regenerates the unmodified amine. The capture and release of biotinylated proteins and peptides from neutravidin, fluorescent labelling through reversible biotinylation at the cell surface and the selective enrichment of proteins from bacterial periplasm are demonstrated. The tags are easily prepared, stable and offer the potential for future application in proteomics, activity-based protein profiling, affinity chromatography and bio-molecule tagging and purification.
Collapse
Affiliation(s)
- Joseph Cowell
- School of ChemistryNewcastle UniversityNewcastle upon TyneNE2 7RUUK
| | - Matthew Buck
- Musculoskeletal Research GroupInstitute of Cellular MedicineNewcastle UniversityNewcastle upon TyneNE2 4HHUK
| | - Ali H. Essa
- School of ChemistryNewcastle UniversityNewcastle upon TyneNE2 7RUUK
- Department of ChemistryCollege of ScienceUniversity of BasrahBasrahIraq
| | - Rebecca Clarke
- School of ChemistryNewcastle UniversityNewcastle upon TyneNE2 7RUUK
| | - Waldemar Vollmer
- Institute for Cell and Molecular BiosciencesNewcastle UniversityNewcastle upon TyneNE2 4HHUK
| | - Daniela Vollmer
- Institute for Cell and Molecular BiosciencesNewcastle UniversityNewcastle upon TyneNE2 4HHUK
| | - Catharien M. Hilkens
- Musculoskeletal Research GroupInstitute of Cellular MedicineNewcastle UniversityNewcastle upon TyneNE2 4HHUK
| | - John D. Isaacs
- Musculoskeletal Research GroupInstitute of Cellular MedicineNewcastle UniversityNewcastle upon TyneNE2 4HHUK
| | - Michael J. Hall
- School of ChemistryNewcastle UniversityNewcastle upon TyneNE2 7RUUK
| | - Joe Gray
- Institute for Cell and Molecular BiosciencesNewcastle UniversityNewcastle upon TyneNE2 4HHUK
| |
Collapse
|
158
|
Brown AJ, Kalsi D, Fernandez-Martell A, Cartwright J, Barber NOW, Patel YD, Turner R, Bryant CL, Johari YB, James DC. Expression Systems for Recombinant Biopharmaceutical Production by Mammalian Cells in Culture. METHODS AND PRINCIPLES IN MEDICINAL CHEMISTRY 2017. [DOI: 10.1002/9783527699124.ch13] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Affiliation(s)
- Adam J. Brown
- University of Sheffield; Department of Chemical and Biological Engineering; Mappin St. Sheffield S1 3JD UK
| | - Devika Kalsi
- University of Sheffield; Department of Chemical and Biological Engineering; Mappin St. Sheffield S1 3JD UK
| | | | - Joe Cartwright
- University of Sheffield; Department of Chemical and Biological Engineering; Mappin St. Sheffield S1 3JD UK
| | - Nicholas O. W. Barber
- University of Sheffield; Department of Chemical and Biological Engineering; Mappin St. Sheffield S1 3JD UK
| | - Yash D. Patel
- University of Sheffield; Department of Chemical and Biological Engineering; Mappin St. Sheffield S1 3JD UK
| | | | - Claire L. Bryant
- University of Sheffield; Department of Chemical and Biological Engineering; Mappin St. Sheffield S1 3JD UK
| | - Yusuf B. Johari
- University of Sheffield; Department of Chemical and Biological Engineering; Mappin St. Sheffield S1 3JD UK
| | - David C. James
- University of Sheffield; Department of Chemical and Biological Engineering; Mappin St. Sheffield S1 3JD UK
| |
Collapse
|
159
|
Antos JM, Ingram J, Fang T, Pishesha N, Truttmann MC, Ploegh HL. Site-Specific Protein Labeling via Sortase-Mediated Transpeptidation. CURRENT PROTOCOLS IN PROTEIN SCIENCE 2017; 89:15.3.1-15.3.19. [PMID: 28762490 PMCID: PMC5810355 DOI: 10.1002/cpps.38] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Strategies for site-specific protein modification are highly desirable for the construction of conjugates containing non-genetically-encoded functional groups. Ideally, these strategies should proceed under mild conditions, and be compatible with a wide range of protein targets and non-natural moieties. The transpeptidation reaction catalyzed by bacterial sortases is a prominent strategy for protein derivatization that possesses these features. Naturally occurring or engineered variants of sortase A from Staphylococcus aureus catalyze a ligation reaction between a five-amino-acid substrate motif (LPXTG) and oligoglycine nucleophiles. By pairing proteins and synthetic peptides that possess these ligation handles, it is possible to install modifications onto the protein N- or C-terminus in site-specific fashion. As described in this unit, the successful implementation of sortase-mediated labeling involves straightforward solid-phase synthesis and molecular biology techniques, and this method is compatible with proteins in solution or on the surface of live cells. © 2017 by John Wiley & Sons, Inc.
Collapse
Affiliation(s)
- John M Antos
- Department of Chemistry, Western Washington University, Bellingham, Washington
| | - Jessica Ingram
- Department of Cancer Immunology and Virology, Dana Farber Cancer Institute, Boston, Massachusetts
| | - Tao Fang
- Program in Cellular and Molecular Medicine, Boston Children's Hospital, Boston, Massachusetts
| | - Novalia Pishesha
- Program in Cellular and Molecular Medicine, Boston Children's Hospital, Boston, Massachusetts
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts
| | - Matthias C Truttmann
- Program in Cellular and Molecular Medicine, Boston Children's Hospital, Boston, Massachusetts
| | - Hidde L Ploegh
- Program in Cellular and Molecular Medicine, Boston Children's Hospital, Boston, Massachusetts
| |
Collapse
|
160
|
His-FLAG Tag as a Fusion Partner of Glycosylated Human Interferon-Gamma and Its Mutant: Gain or Loss? BIOMED RESEARCH INTERNATIONAL 2017; 2017:3018608. [PMID: 28685146 PMCID: PMC5480026 DOI: 10.1155/2017/3018608] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/01/2017] [Accepted: 04/23/2017] [Indexed: 11/17/2022]
Abstract
In order to obtain glycosylated human interferon-gamma (hIFNγ) and its highly prone to aggregation mutant K88Q, a secretory expression in insect cells was employed. To facilitate recombinant proteins purification, detection, and stability the baculovirus expression vectors were constructed to bear N-terminal His6-FLAG tag. Although the obtained proteins were glycosylated, we found that their biological activity was 100 times lower than expected. Our attempts to recover the biological properties of both proteins by tag removal failed due to enterokinase resistance of the tag. Surprisingly, the tag was easily cleaved when the proteins were expressed in E. coli cells and the tag-free proteins showed fully restored activity. To shed light on this phenomenon we performed molecular dynamics simulations. The latter showed that the tags interact with the receptor binding domains and the flexible C-termini of the fusion proteins thus suppressing their complex formation with the hIFNγ receptor. We hypothesize that in the case of glycosylated proteins the tag/C-terminal interaction positions the FLAG peptide in close proximity to the glycans thus sterically impeding the enterokinase access to its recognition site.
Collapse
|
161
|
Tian Q, Zhang P, Gao Z, Li H, Bai Z, Tan S. Hirudin as a novel fusion tag for efficient production of lunasin in Escherichia coli. Prep Biochem Biotechnol 2017; 47:619-626. [PMID: 28151045 DOI: 10.1080/10826068.2017.1286600] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
Fusion expression provides an effective means for the biosynthesis of longer peptides in Escherichia coli. However, the commonly used fusion tags are primarily suitable for laboratory scale applications due to the high cost of commercial affinity resins. Herein, a novel approach exploiting hirudin as a multipurpose fusion tag in combination with tobacco etch virus (TEV) protease cleavage has been developed for the efficient and cost-effective production of a 43-amino acid model peptide lunasin in E. coli at preparative scale. A fusion gene which allows for lunasin to be N-terminally fused to the C-terminus of hirudin through a flexible linker comprising a TEV protease cleavage site was designed and cloned in a secretion vector pTASH. By cultivation in a 7-L bioreactor, the fusion protein was excreted into the culture medium at a high yield of ~380 mg/L, which was conveniently recovered and purified by inexpensive HP20 hydrophobic chromatography at a recovery rate of ~80%. After polishing and cleavage with TEV protease, the finally purified lunasin was obtained with ≥95% purity and yield of ~86 mg/L culture medium. Conclusively, this hirudin tagging strategy is powerful in the production of lunasin and could be applicable for the production of other peptides at preparative scale.
Collapse
Affiliation(s)
- Qinghua Tian
- a Department of Molecular Biology, State Key Laboratory of Natural Medicines, School of Life Science and Technology , China Pharmaceutical University , Nanjing , P.R. China
| | - Ping Zhang
- a Department of Molecular Biology, State Key Laboratory of Natural Medicines, School of Life Science and Technology , China Pharmaceutical University , Nanjing , P.R. China
| | - Zhan Gao
- a Department of Molecular Biology, State Key Laboratory of Natural Medicines, School of Life Science and Technology , China Pharmaceutical University , Nanjing , P.R. China
| | - Hengli Li
- a Department of Molecular Biology, State Key Laboratory of Natural Medicines, School of Life Science and Technology , China Pharmaceutical University , Nanjing , P.R. China
| | - Zhengli Bai
- a Department of Molecular Biology, State Key Laboratory of Natural Medicines, School of Life Science and Technology , China Pharmaceutical University , Nanjing , P.R. China
| | - Shuhua Tan
- a Department of Molecular Biology, State Key Laboratory of Natural Medicines, School of Life Science and Technology , China Pharmaceutical University , Nanjing , P.R. China
| |
Collapse
|
162
|
Guo C, Kim H, Ovadia EM, Mourafetis CM, Yang M, Chen W, Kloxin AM. Bio-orthogonal conjugation and enzymatically triggered release of proteins within multi-layered hydrogels. Acta Biomater 2017; 56:80-90. [PMID: 28391052 PMCID: PMC5510749 DOI: 10.1016/j.actbio.2017.04.002] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2016] [Revised: 03/31/2017] [Accepted: 04/03/2017] [Indexed: 02/02/2023]
Abstract
Hydrogels are facile architectures for the controlled presentation of proteins with far-reaching applications, from fundamental biological studies in three-dimensional culture to new regenerative medicine and therapeutic delivery strategies. Here, we demonstrate a versatile approach for spatially-defined presentation of engineered proteins within hydrogels through i) immobilization using bio-orthogonal strain-promoted alkyne-azide click chemistry and ii) dynamic protease-driven protein release using exogenously applied enzyme. Model fluorescent proteins were expressed using nonsense codon replacement to incorporate azide-containing unnatural amino acids in a site-specific manner toward maintaining protein activity: here, cyan fluorescent protein (AzCFP), mCherry fluorescent protein (AzmCh), and mCh decorated with a thrombin cut-site. (AzTMBmCh). Eight-arm poly(ethylene glycol) (PEG) was modified with dibenzylcyclooctyne (DBCO) groups and reacted with azide functionalized PEG in aqueous solution for rapid formation of hydrogels. Azide functionalized full-length fluorescent proteins were successfully incorporated into the hydrogel network by reaction with PEG-DBCO prior to gel formation. Temporal release and removal of select proteins (AzTMBmCh) was triggered with the application of thrombin and monitored in real-time with confocal microscopy, providing a responsive handle for controlling matrix properties. Hydrogels with regions of different protein compositions were created using a layering technique with thicknesses of hundreds of micrometers, affording opportunities for the creation of complex geometries on size scales relevant for controlling cellular microenvironments. STATEMENT OF SIGNIFICANCE Controlling protein presentation within biomaterials is important for modulating interactions with biological systems. For example, native tissues are composed of subunits with different matrix compositions (proteins, stiffness) that dynamically interact with cells, influencing function and fate. Toward mimicking such temporally-regulated and spatially-defined microenvironments, we utilize bio-orthogonal click chemistry and protein engineering to create hydrogels with distinct regions of proteins and modify them over time. Through nonsense codon replacement, we site-specifically functionalize large proteins with i) azides for covalent conjugation and ii) an enzymatic cleavage site for user-defined release from hydrogels. Our results exemplify not only the ability to create unique bio-functionalized hydrogels with controlled mechanical properties, but also the potential for creating interesting interfaces for cell culture and tissue engineering applications.
Collapse
Affiliation(s)
- Chen Guo
- Department of Chemical and Biomolecular Engineering, University of Delaware, Newark, DE 19716, United States
| | - Heejae Kim
- Department of Chemical and Biomolecular Engineering, University of Delaware, Newark, DE 19716, United States
| | - Elisa M Ovadia
- Department of Chemical and Biomolecular Engineering, University of Delaware, Newark, DE 19716, United States
| | - Christine M Mourafetis
- Department of Chemical and Biomolecular Engineering, New York University, Brooklyn, NY 11201, United States
| | - Mingrui Yang
- Department of Chemical and Biomolecular Engineering, University of Delaware, Newark, DE 19716, United States
| | - Wilfred Chen
- Department of Chemical and Biomolecular Engineering, University of Delaware, Newark, DE 19716, United States
| | - April M Kloxin
- Department of Chemical and Biomolecular Engineering, University of Delaware, Newark, DE 19716, United States; Department of Materials Science and Engineering, University of Delaware, Newark, DE 19716, United States.
| |
Collapse
|
163
|
Affinity purification of Car9-tagged proteins on silica matrices: Optimization of a rapid and inexpensive protein purification technology. Protein Expr Purif 2017; 135:70-77. [DOI: 10.1016/j.pep.2017.05.003] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2017] [Revised: 05/11/2017] [Accepted: 05/11/2017] [Indexed: 11/21/2022]
|
164
|
Expression of the C-terminal domain of human apolipoprotein A-I using a chimeric apolipoprotein. Protein Expr Purif 2017. [PMID: 28624493 DOI: 10.1016/j.pep.2017.06.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Human apolipoprotein A-I (apoA-I) is the most abundant protein in high-density lipoprotein, an anti-atherogenic lipid-protein complex responsible for reverse cholesterol transport. The protein is composed of an N-terminal helix bundle domain, and a small C-terminal (CT) domain. To facilitate study of CT-apoA-I, a novel strategy was employed to produce this small domain in a bacterial expression system. A protein construct was designed of insect apolipophorin III (apoLp-III) and residues 179-243 of apoA-I, with a unique methionine residue positioned between the two proteins and an N-terminal His-tag to facilitate purification. The chimera was expressed in E. coli, purified by Ni-affinity chromatography, and cleaved by cyanogen bromide. SDS-PAGE revealed the presence of three proteins with masses of 7 kDa (CT-apoA-I), 18 kDa (apoLp-III), and a minor 26 kDa band of uncleaved chimera. The digest was reloaded on the Ni-affinity column to bind apoLp-III and uncleaved chimera, while CT-apoA-I was washed from the column and collected. Alternatively, CT-apoA-I was isolated from the digest by reversed-phase HPLC. CT-apoA-I was α-helical, highly effective in solubilizing phospholipid vesicles and disaggregating LPS micelles. However, CT-apoA-I was less active compared to full-length apoA-I in protecting lipolyzed low density lipoproteins from aggregating, and disrupting phosphatidylglycerol bilayer vesicles. Thus the novel expression system produced mg quantities of functional CT-apoA-I, facilitating structural and functional studies of this critical domain of apoA-I.
Collapse
|
165
|
Gabe CM, Brookes SJ, Kirkham J. Preparative SDS PAGE as an Alternative to His-Tag Purification of Recombinant Amelogenin. Front Physiol 2017; 8:424. [PMID: 28670287 PMCID: PMC5472695 DOI: 10.3389/fphys.2017.00424] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2017] [Accepted: 06/01/2017] [Indexed: 11/21/2022] Open
Abstract
Recombinant protein technology provides an invaluable source of proteins for use in structure-function studies, as immunogens, and in the development of therapeutics. Recombinant proteins are typically engineered with "tags" that allow the protein to be purified from crude host cell extracts using affinity based chromatography techniques. Amelogenin is the principal component of the developing enamel matrix and a frequent focus for biomineralization researchers. Several groups have reported the successful production of recombinant amelogenins but the production of recombinant amelogenin free of any tags, and at single band purity on silver stained SDS PAGE is technically challenging. This is important, as rigorous structure-function research frequently demands a high degree of protein purity and fidelity of protein sequence. Our aim was to generate His-tagged recombinant amelogenin at single band purity on silver stained SDS PAGE for use in functionality studies after His-tag cleavage. An acetic acid extraction technique (previously reported to produce recombinant amelogenin at 95% purity directly from E. coli) followed by repeated rounds of nickel column affinity chromatography, failed to generate recombinant amelogenin at single band purity. This was because following an initial round of nickel column affinity chromatography, subsequent cleavage of the His-tag was not 100% efficient. A second round of nickel column affinity chromatography, used in attempts to separate the cleaved His-tag free recombinant from uncleaved His-tagged contaminants, was still unsatisfactory as cleaved recombinant amelogenin exhibited significant affinity for the nickel column. To solve this problem, we used preparative SDS PAGE to successfully purify cleaved recombinant amelogenins to single band purity on silver stained SDS PAGE. The resolving power of preparative SDS PAGE was such that His-tag based purification of recombinant amelogenin becomes redundant. We suggest that acetic acid extraction of recombinant amelogenin and subsequent purification using preparative SDS PAGE provides a simple route to highly purified His-tag free amelogenin for use in structure-function experiments and beyond.
Collapse
Affiliation(s)
| | - Steven J. Brookes
- Division of Oral Biology, School of Dentistry, University of LeedsLeeds, United Kingdom
| | | |
Collapse
|
166
|
Luo J, Torres‐Kolbus J, Liu J, Deiters A. Genetic Encoding of Photocaged Tyrosines with Improved Light‐Activation Properties for the Optical Control of Protease Function. Chembiochem 2017; 18:1442-1447. [DOI: 10.1002/cbic.201700147] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2017] [Indexed: 11/09/2022]
Affiliation(s)
- Ji Luo
- Department of Chemistry University of Pittsburgh 219 Parkman Avenue Pittsburgh Pennsylvania 15260 USA
| | - Jessica Torres‐Kolbus
- Department of Chemistry University of Pittsburgh 219 Parkman Avenue Pittsburgh Pennsylvania 15260 USA
| | - Jihe Liu
- Department of Chemistry University of Pittsburgh 219 Parkman Avenue Pittsburgh Pennsylvania 15260 USA
| | - Alexander Deiters
- Department of Chemistry University of Pittsburgh 219 Parkman Avenue Pittsburgh Pennsylvania 15260 USA
| |
Collapse
|
167
|
Abud JE, Luque EH, Ramos JG, Rodriguez HA. Production of monoclonal antibodies and development of a quantitative immuno-polymerase chain reaction assay to detect and quantify recombinant Glutathione S-transferase. Protein Expr Purif 2017; 135:16-23. [PMID: 28458052 DOI: 10.1016/j.pep.2017.04.014] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2017] [Revised: 04/25/2017] [Accepted: 04/25/2017] [Indexed: 12/26/2022]
Abstract
GST-tagged proteins are important tools for the production of recombinant proteins. Removal of GST tag from its fusion protein, frequently by harsh chemical treatments or proteolytic methods, is often required. Thus, the monitoring of the proteins in tag-free form requires a significant effort to determine the remnants of GST during purification process. In the present study, we developed both a conventional enzyme-linked immunosorbent assay (ELISA) and an immuno-polymerase chain reaction (IPCR) assay, both specific for detection of recombinant GST (rGST). rGST was expressed in Escherichia coli JM109, using a pGEX4T-3 vector, and several anti-rGST monoclonal antibodies were generated using hybridoma technology. Two of these were rationally selected as capture and detection antibodies, allowing the development of a sandwich ELISA with a limit of detection (LOD) of 0.01 μg/ml. To develop the rGST-IPCR assay, we selected "Universal-IPCR" format, comprising the biotin-avidin binding as the coupling system. In addition, the rGST-IPCR was developed in standard PCR tubes, and the surface adsorption of antibodies on PCR tubes, the optimal neutravidin concentrations, the generation of a reporter DNA and the concentration effect were studied and determined. Under optimized assay conditions, the rGST-IPCR assay provided a 100-fold increase in the LOD as well as an expanded working range, in comparison with rGST-ELISA. The proposed method exhibited great potentiality for application in several fields in which measurement of very low levels of GST is necessary, and might provide a model for other IPCR assays.
Collapse
Affiliation(s)
- J E Abud
- Instituto de Salud y Ambiente del Litoral (ISAL, CONICET-UNL), Ciudad Universitaria, Paraje El Pozo s/n, Facultad de Bioquímica y Ciencias Biológicas, Universidad Nacional del Litoral, CP3000 Santa Fe, Argentina
| | - E H Luque
- Instituto de Salud y Ambiente del Litoral (ISAL, CONICET-UNL), Ciudad Universitaria, Paraje El Pozo s/n, Facultad de Bioquímica y Ciencias Biológicas, Universidad Nacional del Litoral, CP3000 Santa Fe, Argentina
| | - J G Ramos
- Instituto de Salud y Ambiente del Litoral (ISAL, CONICET-UNL), Ciudad Universitaria, Paraje El Pozo s/n, Facultad de Bioquímica y Ciencias Biológicas, Universidad Nacional del Litoral, CP3000 Santa Fe, Argentina; Departamento de Bioquímica Clínica y Cuantitativa, Facultad de Bioquímica y Ciencias Biológicas, Universidad Nacional del Litoral, Santa Fe, Argentina
| | - H A Rodriguez
- Instituto de Salud y Ambiente del Litoral (ISAL, CONICET-UNL), Ciudad Universitaria, Paraje El Pozo s/n, Facultad de Bioquímica y Ciencias Biológicas, Universidad Nacional del Litoral, CP3000 Santa Fe, Argentina.
| |
Collapse
|
168
|
Yu X, Sun J, Wang W, Jiang L, Wang R, Xiao W, Cheng B, Fan J. Tobacco etch virus protease mediating cleavage of the cellulose-binding module tagged colored proteins immobilized on the regenerated amorphous cellulose. Bioprocess Biosyst Eng 2017; 40:1101-1110. [DOI: 10.1007/s00449-017-1772-4] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2016] [Accepted: 04/17/2017] [Indexed: 12/12/2022]
|
169
|
Brahma RK, Blanchet G, Kaur S, Manjunatha Kini R, Doley R. Expression and characterization of haemathrins, madanin-like thrombin inhibitors, isolated from the salivary gland of tick Haemaphysalis bispinosa (Acari: Ixodidae). Thromb Res 2017; 152:20-29. [DOI: 10.1016/j.thromres.2017.01.012] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2016] [Revised: 01/26/2017] [Accepted: 01/27/2017] [Indexed: 11/28/2022]
|
170
|
Zhu K, Zhou X, Yan Y, Mo H, Xie Y, Cheng B, Fan J. Cleavage of fusion proteins on the affinity resins using the TEV protease variant. Protein Expr Purif 2017; 131:27-33. [DOI: 10.1016/j.pep.2016.02.003] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2016] [Revised: 02/04/2016] [Accepted: 02/05/2016] [Indexed: 10/22/2022]
|
171
|
Stressler T, Tanzer C, Ewert J, Claaßen W, Fischer L. Simple purification method for a recombinantly expressed native His-tag-free aminopeptidase A from Lactobacillus delbrueckii. Protein Expr Purif 2017; 131:7-15. [DOI: 10.1016/j.pep.2016.10.010] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2016] [Revised: 10/12/2016] [Accepted: 10/31/2016] [Indexed: 10/20/2022]
|
172
|
Chen T, Xu W. A purification method for tag-free human cystatin C recombinant protein expressed in Escherichia coli. Prep Biochem Biotechnol 2017; 47:123-128. [PMID: 27143404 DOI: 10.1080/10826068.2016.1181087] [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] [Indexed: 12/16/2022]
Abstract
To obtain recombinant cystatin C (CysC) protein, which can be used in immunological diagnostic kits, we focused on the preparation of tag-free CysC. The 6 × His-TF-CysC fusion protein was found to overexpress in soluble form in cells of BL21-Gold (DE3)/pCold TF-CysC, which had been induced with isopropyl-D-1-thiogalactopyranoside. Subsequently, we established a protein purification method for tag-free CysC using immobilized metal-affinity chromatography and size-exclusion chromatography. In this method, glutathione-S-transferase-human rhinovirus 3C proteases were used to remove the protein tags. High homogeneity of the purified CysC was determined by SDS-PAGE, while the purity of the tag-free CysC was ascertained to be above 95%. With a yield of 25 mg/L from bacterial culture, the biological activity of the tag-free CysC was evaluated as inhibitors like natural CysC. The performance of this purification method was successfully evaluated in the preparation of other low molecular weight heterologous proteins in Escherichia coli.
Collapse
Affiliation(s)
- Te Chen
- a Department of Laboratory Medicine , The First Affiliated Hospital of Chongqing Medical University , Chongqing , China
| | - Wenchun Xu
- b Laboratory of Medical Diagnostics (Ministry of Education of China) , Department of Key Laboratory Medicine, Chongqing Medical University , Chongqing , China
| |
Collapse
|
173
|
Biancucci M, Dolores JS, Wong J, Grimshaw S, Anderson WF, Satchell KJF, Kwon K. New ligation independent cloning vectors for expression of recombinant proteins with a self-cleaving CPD/6xHis-tag. BMC Biotechnol 2017; 17:1. [PMID: 28056928 PMCID: PMC5216533 DOI: 10.1186/s12896-016-0323-4] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2016] [Accepted: 12/09/2016] [Indexed: 12/30/2022] Open
Abstract
Background Recombinant protein purification is a crucial step for biochemistry and structural biology fields. Rapid robust purification methods utilize various peptide or protein tags fused to the target protein for affinity purification using corresponding matrices and to enhance solubility. However, affinity/solubility-tags often need to be removed in order to conduct functional and structural studies, adding complexities to purification protocols. Results In this work, the Vibrio cholerae MARTX toxin Cysteine Protease Domain (CPD) was inserted in a ligation-independent cloning (LIC) vector to create a C-terminal 6xHis-tagged inducible autoprocessing enzyme tag, called “the CPD-tag”. The pCPD and alternative pCPD/ccdB cloning vectors allow for easy insertion of DNA and expression of the target protein fused to the CPD-tag, which is removed at the end of the purification step by addition of the inexpensive small molecule inositol hexakisphosphate to induce CPD autoprocessing. This process is demonstrated using a small bacterial membrane localization domain and for high yield purification of the eukaryotic small GTPase KRas. Subsequently, pCPD was tested with 40 proteins or sub-domains selected from a high throughput crystallization pipeline. Conclusion pCPD vectors are easily used LIC compatible vectors for expression of recombinant proteins with a C-terminal CPD/6xHis-tag. Although intended only as a strategy for rapid tag removal, this pilot study revealed the CPD-tag may also increase expression and solubility of some recombinant proteins. Electronic supplementary material The online version of this article (doi:10.1186/s12896-016-0323-4) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Marco Biancucci
- Department of Microbiology-Immunology, Feinberg School of Medicine, Northwestern University, 303 E. Chicago Avenue, Ward 6-205, Chicago, IL, 60611, USA
| | - Jazel S Dolores
- Department of Microbiology-Immunology, Feinberg School of Medicine, Northwestern University, 303 E. Chicago Avenue, Ward 6-205, Chicago, IL, 60611, USA.,Present address: Northwestern Memorial Hospital, Chicago, IL, USA
| | - Jennifer Wong
- Department of Microbiology-Immunology, Feinberg School of Medicine, Northwestern University, 303 E. Chicago Avenue, Ward 6-205, Chicago, IL, 60611, USA.,Present address: Indiana University, Bloomington, IN, USA
| | - Sarah Grimshaw
- Infectious Diseases Group, J. Craig Venter Institute, 9714 Medical Center Drive, Rockville, MD, 20850, USA.,Center for Structural Genomics of Infectious Diseases, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Wayne F Anderson
- Center for Structural Genomics of Infectious Diseases, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA.,Department of Biochemistry and Molecular Genetics, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Karla J F Satchell
- Department of Microbiology-Immunology, Feinberg School of Medicine, Northwestern University, 303 E. Chicago Avenue, Ward 6-205, Chicago, IL, 60611, USA. .,Center for Structural Genomics of Infectious Diseases, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA.
| | - Keehwan Kwon
- Infectious Diseases Group, J. Craig Venter Institute, 9714 Medical Center Drive, Rockville, MD, 20850, USA. .,Center for Structural Genomics of Infectious Diseases, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA.
| |
Collapse
|
174
|
Abstract
His-tagging is the most widespread and versatile strategy used to purify recombinant proteins for biochemical and structural studies. Recombinant DNA methods are first used to engineer the addition of a short tract of poly-histidine tag (His-tag) to the N-terminus or C-terminus of a target protein. The His-tag is then exploited to enable purification of the "tagged" protein by Immobilized Metal Affinity Chromatography (IMAC). Here, we describe efficient procedures for the isolation of highly purified His-tagged target proteins from an E. coli host using IMAC.
Collapse
Affiliation(s)
- Sinéad T Loughran
- Department of Applied Sciences, School of Health and Science, Dundalk Institute of Technology, Dundalk, Louth, Ireland.
| | - Ronan T Bree
- Department of Applied Sciences, School of Health and Science, Dundalk Institute of Technology, Dundalk, Louth, Ireland
| | - Dermot Walls
- School of Biotechnology, Dublin City University, Dublin 9, Ireland.,National Centre for Sensor Research, Dublin City University, Dublin 9, Ireland
| |
Collapse
|
175
|
Going native: Complete removal of protein purification affinity tags by simple modification of existing tags and proteases. Protein Expr Purif 2017; 129:18-24. [DOI: 10.1016/j.pep.2016.09.001] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2016] [Revised: 08/18/2016] [Accepted: 09/05/2016] [Indexed: 11/17/2022]
|
176
|
Guan D, Chen Z. Affinity Purification of Proteins in Tag-Free Form: Split Intein-Mediated Ultrarapid Purification (SIRP). Methods Mol Biol 2017; 1495:1-12. [PMID: 27714606 DOI: 10.1007/978-1-4939-6451-2_1] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/06/2023]
Abstract
Proteins purified using affinity-based chromatography often exploit a recombinant affinity tag. Existing methods for the removal of the extraneous tag, needed for many applications, suffer from poor efficiency and/or high cost. Here we describe a simple, efficient, and potentially low-cost approach-split intein-mediated ultrarapid purification (SIRP)-for both the purification of the desired tagged protein from Escherichia coli lysate and removal of the tag in less than 1 h. The N- and C-fragment of a self-cleaving variant of a naturally split DnaE intein from Nostoc punctiforme are genetically fused to the N-terminus of an affinity tag and a protein of interest (POI), respectively. The N-intein/affinity tag is used to functionalize an affinity resin. The high affinity between the N- and C-fragment of DnaE intein enables the POI to be purified from the lysate via affinity to the resin, and the intein-mediated C-terminal cleavage reaction causes tagless POI to be released into the flow-through. The intein cleavage reaction is strongly inhibited by divalent ions (e.g., Zn2+) under non-reducing conditions and is significantly enhanced by reducing conditions. The POI is cleaved efficiently regardless of the identity of the N-terminal amino acid except in the cases of threonine and proline, and the N-intein-functionalized affinity resin can be regenerated for multiple cycles of use.
Collapse
Affiliation(s)
- Dongli Guan
- Department of Chemical Engineering, Texas A&M University, College Station, TX, 77843, USA
| | - Zhilei Chen
- Department of Chemical Engineering, Texas A&M University, College Station, TX, 77843, USA.
- Department of Microbial and Molecular Pathogenesis, Texas A&M University Health Science Center, College Station, TX, 77843, USA.
| |
Collapse
|
177
|
Peleg Y, Prabahar V, Bednarczyk D, Unger T. Harnessing the Profinity eXact™ System for Expression and Purification of Heterologous Proteins in E. coli. Methods Mol Biol 2017; 1586:33-43. [PMID: 28470597 DOI: 10.1007/978-1-4939-6887-9_3] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
Abstract
Highly purified recombinant proteins in large quantities are valuable material for biochemical and structural studies. To achieve this goal, versatile tools were developed to increase the expression of the recombinant proteins and to facilitate the purification process. Fusion tags are commonly used for enhancing expression and solubility and some can be used in the purification process. However, these tags may need to be removed by treatment with specific proteases in order to obtain the tag-free protein. The Profinity eXact™ system provides an alternative system for a fusion tag, enhancing expression and purification in one-step. Here we describe a set of new vectors in which the Profinity eXact™ tag, in addition to a 6× His-tag, with or without additional expression-enhancing sequences, could be used in the Profinity eXact™ system. We show that the solubility enhancing tags (Trx, GST, GB1) increase the yield of the purified tested protein compared to the vector containing only a His-tag upstream of the Profinity eXact™ fusion tag.
Collapse
Affiliation(s)
- Yoav Peleg
- The Israel Structural Proteomics Center (ISPC), Weizmann Institute of Science, 234 Herzl Street, Rehovot, Israel
| | - Vadivel Prabahar
- Migal-Galilee Research Institute, S. Industrial Zone, Kiryat Shmona, Israel
| | - Dominika Bednarczyk
- Department of Bimolecular Sciences, Weizmann Institute of Science, Rehovot, 7610001, Israel
| | - Tamar Unger
- The Israel Structural Proteomics Center (ISPC), Weizmann Institute of Science, 234 Herzl Street, Rehovot, Israel.
| |
Collapse
|
178
|
Abstract
Protein fusion technology has had a major impact on the efficient production and purification of individual recombinant proteins. The use of genetically engineered affinity and solubility-enhancing polypeptide "tags" has increased greatly in recent years and there now exists a considerable repertoire of these that can be used to solve issues related to the expression, stability, solubility, folding, and purification of their fusion partner. In the case of large-scale proteomic studies, the development of purification procedures tailored to individual proteins is not practicable, and affinity tags have therefore become indispensable tools for structural and functional proteomic initiatives that involve the expression of many proteins in parallel. Here, the rationale and applications of a range of established and more recently developed solubility-enhancing and affinity tags is described.
Collapse
Affiliation(s)
- Sinéad T Loughran
- Department of Applied Sciences, Dundalk Institute of Technology, Dundalk, Ireland
| | - Dermot Walls
- School of Biotechnology, Dublin City University, Glasnevin, Dublin 9, Ireland.
- National Centre for Sensor Research, Dublin City University, Glasnevin, Dublin 9, Ireland.
| |
Collapse
|
179
|
Coolbaugh M, Shakalli Tang M, Wood D. High-throughput purification of recombinant proteins using self-cleaving intein tags. Anal Biochem 2017; 516:65-74. [DOI: 10.1016/j.ab.2016.10.016] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2016] [Revised: 10/16/2016] [Accepted: 10/18/2016] [Indexed: 12/13/2022]
|
180
|
Assessment of the Fusion Tags on Increasing Soluble Production of the Active TEV Protease Variant and Other Target Proteins in E. coli. Appl Biochem Biotechnol 2016; 182:769-781. [DOI: 10.1007/s12010-016-2360-9] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2016] [Accepted: 12/05/2016] [Indexed: 10/20/2022]
|
181
|
Abstract
Affinity capture is an effective technique for isolating endogenous protein complexes for further study. When used in conjunction with an antibody, this technique is also frequently referred to as immunoprecipitation. Affinity capture can be applied in a bench-scale and in a high-throughput context. When coupled with protein mass spectrometry, affinity capture has proven to be a workhorse of interactome analysis. Although there are potentially many ways to execute the numerous steps involved, the following protocols implement our favored methods. Two features are distinctive: the use of cryomilled cell powder to produce cell extracts, and antibody-coupled paramagnetic beads as the affinity medium. In many cases, we have obtained superior results to those obtained with more conventional affinity capture practices. Cryomilling avoids numerous problems associated with other forms of cell breakage. It provides efficient breakage of the material, while avoiding denaturation issues associated with heating or foaming. It retains the native protein concentration up to the point of extraction, mitigating macromolecular dissociation. It reduces the time extracted proteins spend in solution, limiting deleterious enzymatic activities, and it may reduce the non-specific adsorption of proteins by the affinity medium. Micron-scale magnetic affinity media have become more commonplace over the last several years, increasingly replacing the traditional agarose- and Sepharose-based media. Primary benefits of magnetic media include typically lower non-specific protein adsorption; no size exclusion limit because protein complex binding occurs on the bead surface rather than within pores; and ease of manipulation and handling using magnets.
Collapse
Affiliation(s)
- John LaCava
- Laboratory of Cellular and Structural Biology, The Rockefeller University; Institute for Systems Genetics, Department of Biochemistry and Molecular Pharmacology, New York University School of Medicine;
| | - Hua Jiang
- Laboratory of Cellular and Structural Biology, The Rockefeller University
| | - Michael P Rout
- Laboratory of Cellular and Structural Biology, The Rockefeller University
| |
Collapse
|
182
|
Yadav DK, Yadav N, Yadav S, Haque S, Tuteja N. An insight into fusion technology aiding efficient recombinant protein production for functional proteomics. Arch Biochem Biophys 2016; 612:57-77. [DOI: 10.1016/j.abb.2016.10.012] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2016] [Revised: 10/15/2016] [Accepted: 10/18/2016] [Indexed: 11/27/2022]
|
183
|
Chen Y, Li Q, Yang J, Xie H. Promoting Tag Removal of a MBP-Fused Integral Membrane Protein by TEV Protease. Appl Biochem Biotechnol 2016; 181:939-947. [PMID: 27696139 DOI: 10.1007/s12010-016-2260-z] [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: 05/27/2016] [Accepted: 09/19/2016] [Indexed: 11/30/2022]
Abstract
Tag removal is a prerequisite issue for structural and functional analysis of affinity-purified membrane proteins. The present study took a MBP-fused membrane protein, MrpF, as a model to investigate the tag removal by TEV protease. Influences of the linking sequence between TEV cleavage site and MrpF on protein expression and predicted secondary structure were investigated. The steric accessibility of TEV protease to cleavage site of MBP-fused MrpF was explored. It was found that reducing the size of hydrophilic group of detergents and/or extending the linking sequence between cleavage site and target protein can significantly improve the accessibility of the cleavage site and promote tag removal by TEV protease.
Collapse
Affiliation(s)
- Yanke Chen
- Key Laboratory of Magnetic Resonance in Biological Systems, State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, Wuhan Center for Magnetic Resonance, Wuhan Institute of Physics and Mathematics, Chinese Academy of Sciences, Wuhan, 430071, China
| | - Qichang Li
- School of Chemistry, Chemical Engineering and Life Sciences, Wuhan University of Technology, Wuhan, 430070, China
| | - Jun Yang
- Key Laboratory of Magnetic Resonance in Biological Systems, State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, Wuhan Center for Magnetic Resonance, Wuhan Institute of Physics and Mathematics, Chinese Academy of Sciences, Wuhan, 430071, China.
| | - Hao Xie
- School of Chemistry, Chemical Engineering and Life Sciences, Wuhan University of Technology, Wuhan, 430070, China.
| |
Collapse
|
184
|
Cruz-Teran CA, Carlin KB, Efimenko K, Genzer J, Rao BM. Targeted Mutagenesis and Combinatorial Library Screening Enables Control of Protein Orientation on Surfaces and Increased Activity of Adsorbed Proteins. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2016; 32:8660-8667. [PMID: 27490089 DOI: 10.1021/acs.langmuir.6b01446] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/06/2023]
Abstract
While nonspecific adsorption is widely used for immobilizing proteins on solid surfaces, the random nature of protein adsorption may reduce the activity of immobilized proteins due to occlusion of the active site. We hypothesized that the orientation a protein assumes on a given surface can be controlled by systematically introducing mutations into a region distant from its active site, thereby retaining activity of the immobilized protein. To test this hypothesis, we generated a combinatorial protein library by randomizing six targeted residues in a binding protein derived from highly stable, nonimmunoglobulin Sso7d scaffold; mutations were targeted in a region that is distant from the binding site. This library was screened to isolate binders that retain binding to its cognate target (chicken immunoglobulin Y, cIgY) as well as exhibit adsorption on unmodified silica at pH 7.4 and high ionic strength conditions. A single mutant, Sso7d-2B5, was selected for further characterization. Sso7d-2B5 retained binding to cIgY with an apparent dissociation constant similar to that of the parent protein; both mutant and parent proteins saturated the surface of silica with similar densities. Strikingly, however, silica beads coated with Sso7d-2B5 could achieve up to 7-fold higher capture of cIgY than beads coated with the parent protein. These results strongly suggest that mutations introduced in Sso7d-2B5 alter its orientation relative to the parent protein, when adsorbed on silica surfaces. Our approach also provides a generalizable strategy for introducing mutations in proteins so as to improve their activity upon immobilization, and has direct relevance to development of protein-based biosensors and biocatalysts.
Collapse
Affiliation(s)
- Carlos A Cruz-Teran
- Department of Chemical and Biomolecular Engineering, North Carolina State University , Raleigh, North Carolina 27695, United States
| | - Kevin B Carlin
- Department of Chemical and Biomolecular Engineering, North Carolina State University , Raleigh, North Carolina 27695, United States
| | - Kirill Efimenko
- Department of Chemical and Biomolecular Engineering, North Carolina State University , Raleigh, North Carolina 27695, United States
| | - Jan Genzer
- Department of Chemical and Biomolecular Engineering, North Carolina State University , Raleigh, North Carolina 27695, United States
| | - Balaji M Rao
- Department of Chemical and Biomolecular Engineering, North Carolina State University , Raleigh, North Carolina 27695, United States
| |
Collapse
|
185
|
A simple method for recombinant protein purification using self-assembling peptide-tagged tobacco etch virus protease. Protein Expr Purif 2016; 128:86-92. [PMID: 27546453 DOI: 10.1016/j.pep.2016.08.013] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2016] [Revised: 08/15/2016] [Accepted: 08/17/2016] [Indexed: 11/22/2022]
Abstract
Recombinant protein purification remains to be a major challenge in biotechnology and medicine. In this paper we report a simple method for recombinant protein purification using self-assembling peptide-tagged tobacco etch virus protease (TEVp). After construction of an N-terminal ELK16 peptide fusion expression vector, we expressed ELK16-TEVp fusion protein in E. coli. SDS-PAGE analysis showed that ELK16-TEVp was expressed as active protein aggregates which could be purified to 91% purity with 92% recovery by centrifugation in the presence 0.5% Triton X-100. By using His-tagged bovine interferon-γ (His-BoIFN-γ) as the substrate, we demonstrated that EKL16-TEVp had a protease activity of 1.3 × 10(4) units/mg protein with almost 100% cleavage efficiency under the optimized conditions. More importantly, EKL16-TEVp could be removed from the cleavage reaction by single-step centrifugation. After removing the His-tag by nickel-conjugated agarose bead absorption, the recombinant BoIFN-γ (rBoIFN-γ) was purified to 98.3% purity with 63% recovery. The rBoIFN-γ had an antiviral activity of 1.6 × 10(3) units/mg protein against vesicular stomatitis virus. These data suggest that ELK16-TEVp may become a universal tool for recombinant protein purification.
Collapse
|
186
|
Valle A, Hervis Y, Socas L, Canet L, Faheem M, Barbosa J, Lanio M, Pazos I. The multigene families of actinoporins (part II): Strategies for heterologous production in Escherichia coli. Toxicon 2016; 118:64-81. [DOI: 10.1016/j.toxicon.2016.03.018] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2016] [Accepted: 03/24/2016] [Indexed: 11/26/2022]
|
187
|
Raran-Kurussi S, Waugh DS. A dual protease approach for expression and affinity purification of recombinant proteins. Anal Biochem 2016; 504:30-7. [PMID: 27105777 PMCID: PMC4877217 DOI: 10.1016/j.ab.2016.04.006] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2016] [Revised: 04/01/2016] [Accepted: 04/07/2016] [Indexed: 11/29/2022]
Abstract
We describe a new method for affinity purification of recombinant proteins using a dual protease protocol. Escherichia coli maltose binding protein (MBP) is employed as an N-terminal tag to increase the yield and solubility of its fusion partners. The MBP moiety is then removed by rhinovirus 3C protease, prior to purification, to yield an N-terminally His6-tagged protein. Proteins that are only temporarily rendered soluble by fusing them to MBP are readily identified at this stage because they will precipitate after the MBP tag is removed by 3C protease. The remaining soluble His6-tagged protein, if any, is subsequently purified by immobilized metal affinity chromatography (IMAC). Finally, the N-terminal His6 tag is removed by His6-tagged tobacco etch virus (TEV) protease to yield the native recombinant protein, and the His6-tagged contaminants are removed by adsorption during a second round of IMAC, leaving only the untagged recombinant protein in the column effluent. The generic strategy described here saves time and effort by removing insoluble aggregates at an early stage in the process while also reducing the tendency of MBP to "stick" to its fusion partners during affinity purification.
Collapse
Affiliation(s)
- Sreejith Raran-Kurussi
- Protein Engineering Section, Macromolecular Crystallography Laboratory, Center for Cancer Research, National Cancer Institute, Frederick, MD 21702, USA
| | - David S Waugh
- Protein Engineering Section, Macromolecular Crystallography Laboratory, Center for Cancer Research, National Cancer Institute, Frederick, MD 21702, USA.
| |
Collapse
|
188
|
Kukk K, Samel N. Enhanced expression of human prostaglandin H synthase-2 in the yeast Pichia pastoris and removal of the C-terminal tag with bovine carboxypeptidase A. J Biotechnol 2016; 231:224-231. [PMID: 27316830 DOI: 10.1016/j.jbiotec.2016.06.015] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2016] [Revised: 06/09/2016] [Accepted: 06/13/2016] [Indexed: 11/19/2022]
Abstract
Vertebrate prostaglandin H synthases (PGHSs) are membrane-bound disulphide-containing hemoglycoproteins. Therefore, eukaryotic expression systems are required for the production of recombinant PGHSs. Recently we announced the expression of human PGHS-2 (hPGHS-2) in the yeast Pichia pastoris. Here we report improved production of hPGHS-2 in P. pastoris and a convenient method for the purification and de-tagging of the protein. An affinity tag comprised of a proline, a glycine and eight histidines was introduced into the C-terminal end of hPGHS-2. The tagged hPGHS-2 was expressed intracellularly in P. pastoris under the control of a constitutive or methanol-inducible promoter. Compared to constitutive expression, methanol-induced expression yielded approximately four times more protein. The analysis of high and low gene copy number recombinants revealed a positive correlation between the gene copy number and the expression level of hPGHS-2. The recombinant hPGHS-2 was purified using immobilised metal ion affinity chromatography. A novel elution method, treatment of the affinity resin with bovine carboxypeptidase A, was employed. The yield of pure de-tagged hPGHS-2 from 1l of yeast culture was approximately 3mg. The protein purification process with simultaneous removal of the C-terminal polyhistidine tag could be easily applied for the affinity purification of other proteins.
Collapse
Affiliation(s)
- Kaia Kukk
- Department of Chemistry, Tallinn University of Technology, Akadeemia tee 15, 12618 Tallinn, Estonia
| | - Nigulas Samel
- Department of Chemistry, Tallinn University of Technology, Akadeemia tee 15, 12618 Tallinn, Estonia.
| |
Collapse
|
189
|
Cesaratto F, Burrone OR, Petris G. Tobacco Etch Virus protease: A shortcut across biotechnologies. J Biotechnol 2016; 231:239-249. [PMID: 27312702 DOI: 10.1016/j.jbiotec.2016.06.012] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2016] [Revised: 05/31/2016] [Accepted: 06/10/2016] [Indexed: 11/29/2022]
Abstract
About thirty years ago, studies on the RNA genome of Tobacco Etch Virus revealed the presence of an efficient and specific protease, called Tobacco Etch Virus protease (TEVp), that was part of the Nuclear Inclusion a (NIa) enzyme. TEVp is an efficient and specific protease of 27kDa that has become a valuable biotechnological tool. Nowadays TEVp is a unique endopeptidase largely exploited in biotechnology from industrial applications to in vitro and in vivo cellular studies. A number of TEVp mutants with different rate of cleavage, stability and specificity have been reported. Similarly, a panel of different target cleavage sites, derived from the canonical ENLYFQ-G/S site, has been established. In this review we describe these aspects of TEVp and some of its multiple applications. A particular focus is on the use and molecular biology of TEVp in living cells and organisms.
Collapse
Affiliation(s)
- Francesca Cesaratto
- International Centre for Genetic Engineering and Biotechnology, ICGEB, Trieste, Italy
| | - Oscar R Burrone
- International Centre for Genetic Engineering and Biotechnology, ICGEB, Trieste, Italy.
| | | |
Collapse
|
190
|
Fernandez-Rodriguez J, Voigt CA. Post-translational control of genetic circuits using Potyvirus proteases. Nucleic Acids Res 2016; 44:6493-502. [PMID: 27298256 PMCID: PMC5291274 DOI: 10.1093/nar/gkw537] [Citation(s) in RCA: 58] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2016] [Accepted: 06/06/2016] [Indexed: 12/25/2022] Open
Abstract
Genetic engineering projects often require control over when a protein is degraded. To this end, we use a fusion between a degron and an inactivating peptide that can be added to the N-terminus of a protein. When the corresponding protease is expressed, it cleaves the peptide and the protein is degraded. Three protease:cleavage site pairs from Potyvirus are shown to be orthogonal and active in exposing degrons, releasing inhibitory domains and cleaving polyproteins. This toolbox is applied to the design of genetic circuits as a means to control regulator activity and degradation. First, we demonstrate that a gate can be constructed by constitutively expressing an inactivated repressor and having an input promoter drive the expression of the protease. It is also shown that the proteolytic release of an inhibitory domain can improve the dynamic range of a transcriptional gate (200-fold repression). Next, we design polyproteins containing multiple repressors and show that their cleavage can be used to control multiple outputs. Finally, we demonstrate that the dynamic range of an output can be improved (8-fold to 190-fold) with the addition of a protease-cleaved degron. Thus, controllable proteolysis offers a powerful tool for modulating and expanding the function of synthetic gene circuits.
Collapse
Affiliation(s)
- Jesus Fernandez-Rodriguez
- Synthetic Biology Center, Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Christopher A Voigt
- Synthetic Biology Center, Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| |
Collapse
|
191
|
Liutkus M, Fraser SA, Caron K, Stigers DJ, Easton CJ. Peptide Synthesis through Cell-Free Expression of Fusion Proteins Incorporating Modified Amino Acids as Latent Cleavage Sites for Peptide Release. Chembiochem 2016; 17:908-12. [PMID: 26918308 DOI: 10.1002/cbic.201600091] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2016] [Indexed: 01/03/2023]
Abstract
Chlorinated analogues of Leu and Ile are incorporated during cell-free expression of peptides fused to protein, by exploiting the promiscuity of the natural biosynthetic machinery. They then act as sites for clean and efficient release of the peptides simply by brief heat treatment. Dehydro analogues of Leu and Ile are similarly incorporated as latent sites for peptide release through treatment with iodine under cold conditions. These protocols complement enzyme-catalyzed methods and have been used to prepare calcitonin, gastrin-releasing peptide, cholecystokinin-7, and prolactin-releasing peptide prohormones, as well as analogues substituted with unusual amino acids, thus illustrating their practical utility as alternatives to more traditional chemical peptide synthesis.
Collapse
Affiliation(s)
- Mantas Liutkus
- Research School of Chemistry, Australian National University, Canberra, ACT, 2601, Australia
| | - Samuel A Fraser
- Research School of Chemistry, Australian National University, Canberra, ACT, 2601, Australia
| | - Karine Caron
- Research School of Chemistry, Australian National University, Canberra, ACT, 2601, Australia
| | - Dannon J Stigers
- Research School of Chemistry, Australian National University, Canberra, ACT, 2601, Australia
| | - Christopher J Easton
- Research School of Chemistry, Australian National University, Canberra, ACT, 2601, Australia.
| |
Collapse
|
192
|
Ullah R, Shah MA, Tufail S, Ismat F, Imran M, Iqbal M, Mirza O, Rhaman M. Activity of the Human Rhinovirus 3C Protease Studied in Various Buffers, Additives and Detergents Solutions for Recombinant Protein Production. PLoS One 2016; 11:e0153436. [PMID: 27093053 PMCID: PMC4836831 DOI: 10.1371/journal.pone.0153436] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2016] [Accepted: 03/29/2016] [Indexed: 11/18/2022] Open
Abstract
Proteases are widely used to remove affinity and solubility tags from recombinant proteins to avoid potential interference of these tags with the structure and function of the fusion partner. In recent years, great interest has been seen in use of the human rhinovirus 3C protease owing to its stringent sequence specificity and enhanced activity. Like other proteases, activity of the human rhinovirus 3C protease can be affected in part by the buffer components and additives that are generally employed for purification and stabilization of proteins, hence, necessitate their removal by tedious and time-consuming procedures before proteolysis can occur. To address this issue, we examined the effect of elution buffers used for common affinity based purifications, salt ions, stability/solubility and reducing agents, and detergents on the activity of the human rhinovirus 3C protease using three different fusion proteins at 4°C, a temperature of choice for purification of many proteins. The results show that the human rhinovirus 3C protease performs better at 4°C than the frequently used tobacco etch virus protease and its activity was insensitive to most of the experimental conditions tested. Though number of fusion proteins tested is limited, we expect that these finding will facilitate the use of the human rhinovirus 3C protease in recombinant protein production for pharmaceutical and biotechnological applications.
Collapse
Affiliation(s)
- Raheem Ullah
- Drug Discovery and Structural Biology group, Health Biotechnology Division, National Institute for Biotechnology and Genetic Engineering (NIBGE), Faisalabad, Pakistan
- Pakistan Institute of Engineering and Applied Sciences, P.O. Nilore, Islamabad, Pakistan
| | - Majid Ali Shah
- Drug Discovery and Structural Biology group, Health Biotechnology Division, National Institute for Biotechnology and Genetic Engineering (NIBGE), Faisalabad, Pakistan
- Pakistan Institute of Engineering and Applied Sciences, P.O. Nilore, Islamabad, Pakistan
| | - Soban Tufail
- Drug Discovery and Structural Biology group, Health Biotechnology Division, National Institute for Biotechnology and Genetic Engineering (NIBGE), Faisalabad, Pakistan
| | - Fouzia Ismat
- Drug Discovery and Structural Biology group, Health Biotechnology Division, National Institute for Biotechnology and Genetic Engineering (NIBGE), Faisalabad, Pakistan
| | - Muhammad Imran
- Drug Discovery and Structural Biology group, Health Biotechnology Division, National Institute for Biotechnology and Genetic Engineering (NIBGE), Faisalabad, Pakistan
| | - Mazhar Iqbal
- Drug Discovery and Structural Biology group, Health Biotechnology Division, National Institute for Biotechnology and Genetic Engineering (NIBGE), Faisalabad, Pakistan
- Pakistan Institute of Engineering and Applied Sciences, P.O. Nilore, Islamabad, Pakistan
| | - Osman Mirza
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Moazur Rhaman
- Drug Discovery and Structural Biology group, Health Biotechnology Division, National Institute for Biotechnology and Genetic Engineering (NIBGE), Faisalabad, Pakistan
- Pakistan Institute of Engineering and Applied Sciences, P.O. Nilore, Islamabad, Pakistan
| |
Collapse
|
193
|
Wang J, Li J, Wang J, Liu L, Li J, Qin H, Ding S, Fu M, Ji J, Jiang P, Qiu L. Simultaneous monitoring of quantum dots and their assembly and disassembly with PreScission protease using capillary electrophoresis with fluorescence detection. J Sep Sci 2016; 39:1785-91. [DOI: 10.1002/jssc.201600075] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2016] [Revised: 03/01/2016] [Accepted: 03/01/2016] [Indexed: 01/23/2023]
Affiliation(s)
- Jianhao Wang
- School of Pharmaceutical Engineering and Life Science; Changzhou University; Changzhou Jiangsu P. R. China
| | - Jinchen Li
- School of Pharmaceutical Engineering and Life Science; Changzhou University; Changzhou Jiangsu P. R. China
| | - Jianpeng Wang
- School of Pharmaceutical Engineering and Life Science; Changzhou University; Changzhou Jiangsu P. R. China
| | - Li Liu
- School of Pharmaceutical Engineering and Life Science; Changzhou University; Changzhou Jiangsu P. R. China
| | - Jinping Li
- School of Pharmaceutical Engineering and Life Science; Changzhou University; Changzhou Jiangsu P. R. China
| | - Haifang Qin
- Institute of Pharmaceutical and Environmental Engineering; Changzhou Vocational Institute of Engineering; Changzhou Jiangsu P. R. China
| | - Shumin Ding
- School of Pharmaceutical Engineering and Life Science; Changzhou University; Changzhou Jiangsu P. R. China
| | - Minli Fu
- School of Pharmaceutical Engineering and Life Science; Changzhou University; Changzhou Jiangsu P. R. China
| | - Junling Ji
- Changzhou Meisheng Biomaterials Co., Ltd; Changzhou Jiangsu P. R. China
| | - Pengju Jiang
- School of Pharmaceutical Engineering and Life Science; Changzhou University; Changzhou Jiangsu P. R. China
| | - Lin Qiu
- School of Pharmaceutical Engineering and Life Science; Changzhou University; Changzhou Jiangsu P. R. China
- Changzhou Meisheng Biomaterials Co., Ltd; Changzhou Jiangsu P. R. China
- State Key Laboratory of Coordination Chemistry; Nanjing University; Nanjing Jiangsu P. R. China
| |
Collapse
|
194
|
Global characterization of in vivo enzyme catalytic rates and their correspondence to in vitro kcat measurements. Proc Natl Acad Sci U S A 2016; 113:3401-6. [PMID: 26951675 DOI: 10.1073/pnas.1514240113] [Citation(s) in RCA: 151] [Impact Index Per Article: 18.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Turnover numbers, also known as kcat values, are fundamental properties of enzymes. However, kcat data are scarce and measured in vitro, thus may not faithfully represent the in vivo situation. A basic question that awaits elucidation is: how representative are kcat values for the maximal catalytic rates of enzymes in vivo? Here, we harness omics data to calculate kmax(vivo), the observed maximal catalytic rate of an enzyme inside cells. Comparison with kcat values from Escherichia coli, yields a correlation ofr(2)= 0.62 in log scale (p < 10(-10)), with a root mean square difference of 0.54 (3.5-fold in linear scale), indicating that in vivo and in vitro maximal rates generally concur. By accounting for the degree of saturation of enzymes and the backward flux dictated by thermodynamics, we further refine the correspondence between kmax(vivo) and kcat values. The approach we present here characterizes the quantitative relationship between enzymatic catalysis in vitro and in vivo and offers a high-throughput method for extracting enzyme kinetic constants from omics data.
Collapse
|
195
|
Bhattacharyya M, Stratton MM, Going CC, McSpadden ED, Huang Y, Susa AC, Elleman A, Cao YM, Pappireddi N, Burkhardt P, Gee CL, Barros T, Schulman H, Williams ER, Kuriyan J. Molecular mechanism of activation-triggered subunit exchange in Ca(2+)/calmodulin-dependent protein kinase II. eLife 2016; 5. [PMID: 26949248 PMCID: PMC4859805 DOI: 10.7554/elife.13405] [Citation(s) in RCA: 72] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2015] [Accepted: 03/03/2016] [Indexed: 12/04/2022] Open
Abstract
Activation triggers the exchange of subunits in Ca2+/calmodulin-dependent protein kinase II (CaMKII), an oligomeric enzyme that is critical for learning, memory, and cardiac function. The mechanism by which subunit exchange occurs remains elusive. We show that the human CaMKII holoenzyme exists in dodecameric and tetradecameric forms, and that the calmodulin (CaM)-binding element of CaMKII can bind to the hub of the holoenzyme and destabilize it to release dimers. The structures of CaMKII from two distantly diverged organisms suggest that the CaM-binding element of activated CaMKII acts as a wedge by docking at intersubunit interfaces in the hub. This converts the hub into a spiral form that can release or gain CaMKII dimers. Our data reveal a three-way competition for the CaM-binding element, whereby phosphorylation biases it towards the hub interface, away from the kinase domain and calmodulin, thus unlocking the ability of activated CaMKII holoenzymes to exchange dimers with unactivated ones. DOI:http://dx.doi.org/10.7554/eLife.13405.001 How does memory outlast the lifetime of the molecules that encode it? One enzyme that is found in neurons and has been suggested to help long-term memories to form is called CaMKII. Each CaMKII assembly is typically composed of 12 to 14 protein subunits associated in a ring and can exist in either an “unactivated” or “activated” state. In 2014, researchers showed that CaMKII assemblies can exchange subunits with each other. Importantly, an active CaMKII can mix with an unactivated CaMKII and share its activation state. CaMKII may use this mechanism to spread information to the next generation of proteins – thereby allowing activation to outlast the lifespan of the initially activated proteins. However the molecular mechanism that underlies this process was not clear. Now, Bhattacharyya et al. – including some of the researchers involved in the 2014 work – address two questions about this mechanism. How do subunits exchange between CaMKII assemblies? And how does the activation of CaMKII initiate subunit exchange? A closed-ring hub ties the subunits of CaMKII together, similar to the organization of the segments in an orange. To undergo subunit exchange, the hub must open up to release and accept subunits. Bhattacharyya et al. have now uncovered an intrinsic flexibility in the hub that is triggered by a short peptide segment in CaMKII. This segment, which is exposed in activated CaMKII but not in the unactivated form, can crack open the hub ring by binding between the hub subunits, like a finger separating the segments of an orange. This allows the hub to flex and expand, and once open, the hub’s flexibility allows room for subunits to be released or accepted. Although this subunit exchange mechanism could be a powerful means for spreading the activated state throughout signaling pathways, the biological relevance of this phenomenon has not been clarified. However, the mechanistic framework provided by Bhattacharyya et al. may allow new experiments to be performed that test the consequences of subunit exchange in live cells and organisms. It could also enable investigations into the importance of subunit exchange in long-term memory. DOI:http://dx.doi.org/10.7554/eLife.13405.002
Collapse
Affiliation(s)
- Moitrayee Bhattacharyya
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, United States.,California Institute for Quantitative Biosciences, University of California, Berkeley, Berkeley, United States.,Howard Hughes Medical Institute, University of California, Berkeley, Berkeley, United States
| | - Margaret M Stratton
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, United States.,California Institute for Quantitative Biosciences, University of California, Berkeley, Berkeley, United States.,Howard Hughes Medical Institute, University of California, Berkeley, Berkeley, United States
| | - Catherine C Going
- Department of Chemistry, University of California, Berkeley, Berkeley, United States
| | - Ethan D McSpadden
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, United States.,California Institute for Quantitative Biosciences, University of California, Berkeley, Berkeley, United States.,Howard Hughes Medical Institute, University of California, Berkeley, Berkeley, United States
| | - Yongjian Huang
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, United States.,California Institute for Quantitative Biosciences, University of California, Berkeley, Berkeley, United States.,Howard Hughes Medical Institute, University of California, Berkeley, Berkeley, United States.,Biophysics Graduate Group, University of California, Berkeley, Berkeley, United States
| | - Anna C Susa
- Department of Chemistry, University of California, Berkeley, Berkeley, United States
| | - Anna Elleman
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, United States.,California Institute for Quantitative Biosciences, University of California, Berkeley, Berkeley, United States.,Howard Hughes Medical Institute, University of California, Berkeley, Berkeley, United States
| | - Yumeng Melody Cao
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, United States.,California Institute for Quantitative Biosciences, University of California, Berkeley, Berkeley, United States.,Howard Hughes Medical Institute, University of California, Berkeley, Berkeley, United States
| | - Nishant Pappireddi
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, United States.,California Institute for Quantitative Biosciences, University of California, Berkeley, Berkeley, United States.,Howard Hughes Medical Institute, University of California, Berkeley, Berkeley, United States
| | - Pawel Burkhardt
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, United States.,Howard Hughes Medical Institute, University of California, Berkeley, Berkeley, United States
| | - Christine L Gee
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, United States.,California Institute for Quantitative Biosciences, University of California, Berkeley, Berkeley, United States.,Howard Hughes Medical Institute, University of California, Berkeley, Berkeley, United States
| | - Tiago Barros
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, United States.,California Institute for Quantitative Biosciences, University of California, Berkeley, Berkeley, United States.,Howard Hughes Medical Institute, University of California, Berkeley, Berkeley, United States
| | | | - Evan R Williams
- Department of Chemistry, University of California, Berkeley, Berkeley, United States
| | - John Kuriyan
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, United States.,California Institute for Quantitative Biosciences, University of California, Berkeley, Berkeley, United States.,Howard Hughes Medical Institute, University of California, Berkeley, Berkeley, United States.,Biophysics Graduate Group, University of California, Berkeley, Berkeley, United States.,Physical Biosciences Division, Lawrence Berkeley National Laboratory, Berkeley, United States
| |
Collapse
|
196
|
Sainsbury F, Jutras PV, Vorster J, Goulet MC, Michaud D. A Chimeric Affinity Tag for Efficient Expression and Chromatographic Purification of Heterologous Proteins from Plants. FRONTIERS IN PLANT SCIENCE 2016; 7:141. [PMID: 26913045 PMCID: PMC4753422 DOI: 10.3389/fpls.2016.00141] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/23/2015] [Accepted: 01/27/2016] [Indexed: 05/23/2023]
Abstract
The use of plants as expression hosts for recombinant proteins is an increasingly attractive option for the production of complex and challenging biopharmaceuticals. Tools are needed at present to marry recent developments in high-yielding gene vectors for heterologous expression with routine protein purification techniques. In this study, we designed the Cysta-tag, a new purification tag for immobilized metal affinity chromatography (IMAC) of plant-made proteins based on the protein-stabilizing fusion partner SlCYS8. We show that the Cysta-tag may be used to readily purify proteins under native conditions, and then be removed enzymatically to isolate the protein of interest. We also show that commonly used protease recognition sites for linking purification tags are differentially stable in leaves of the commonly used expression host Nicotiana benthamiana, with those linkers susceptible to cysteine proteases being less stable then serine protease-cleavable linkers. As an example, we describe a Cysta-tag experimental scheme for the one-step purification of a clinically useful protein, human α1-antitrypsin, transiently expressed in N. benthamiana. With potential applicability to the variety of chromatography formats commercially available for IMAC-based protein purification, the Cysta-tag provides a convenient means for the efficient and cost-effective purification of recombinant proteins from plant tissues.
Collapse
Affiliation(s)
- Frank Sainsbury
- Département de Phytologie–Centre de Recherche et d’Innovation sur les Végétaux, Université Laval, QuébecQC, Canada
- Centre for Biomolecular Engineering, Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, BrisbaneQLD, Australia
| | - Philippe V. Jutras
- Département de Phytologie–Centre de Recherche et d’Innovation sur les Végétaux, Université Laval, QuébecQC, Canada
- Centre for Biomolecular Engineering, Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, BrisbaneQLD, Australia
| | - Juan Vorster
- Department of Plant Production and Soil Science, Forestry and Agricultural Biotechnology Institute, University of PretoriaPretoria, South Africa
| | - Marie-Claire Goulet
- Département de Phytologie–Centre de Recherche et d’Innovation sur les Végétaux, Université Laval, QuébecQC, Canada
| | - Dominique Michaud
- Département de Phytologie–Centre de Recherche et d’Innovation sur les Végétaux, Université Laval, QuébecQC, Canada
| |
Collapse
|
197
|
Li S, Yang K, Zhao B, Li X, Liu L, Chen Y, Zhang L, Zhang Y. Epitope imprinting enhanced IMAC (EI-IMAC) for highly selective purification of His-tagged protein. J Mater Chem B 2016; 4:1960-1967. [DOI: 10.1039/c5tb02505b] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Selectivity of epitope imprinted sites is introduced on the IMAC surface through epitope surface imprinting. The obtained epitope imprinting enhanced IMAC (EI-IMAC) could purify His-tagged proteins with high selectivity without any major interference from the host proteins.
Collapse
Affiliation(s)
- Senwu Li
- National Chromatographic R. & A. Center
- Key Laboratory of Separation Science for Analytical Chemistry
- Dalian Institute of Chemical Physics
- Chinese Academy of Sciences
- Dalian 116023
| | - Kaiguang Yang
- National Chromatographic R. & A. Center
- Key Laboratory of Separation Science for Analytical Chemistry
- Dalian Institute of Chemical Physics
- Chinese Academy of Sciences
- Dalian 116023
| | - Baofeng Zhao
- National Chromatographic R. & A. Center
- Key Laboratory of Separation Science for Analytical Chemistry
- Dalian Institute of Chemical Physics
- Chinese Academy of Sciences
- Dalian 116023
| | - Xiao Li
- National Chromatographic R. & A. Center
- Key Laboratory of Separation Science for Analytical Chemistry
- Dalian Institute of Chemical Physics
- Chinese Academy of Sciences
- Dalian 116023
| | - Lukuan Liu
- National Chromatographic R. & A. Center
- Key Laboratory of Separation Science for Analytical Chemistry
- Dalian Institute of Chemical Physics
- Chinese Academy of Sciences
- Dalian 116023
| | - Yuanbo Chen
- National Chromatographic R. & A. Center
- Key Laboratory of Separation Science for Analytical Chemistry
- Dalian Institute of Chemical Physics
- Chinese Academy of Sciences
- Dalian 116023
| | - Lihua Zhang
- National Chromatographic R. & A. Center
- Key Laboratory of Separation Science for Analytical Chemistry
- Dalian Institute of Chemical Physics
- Chinese Academy of Sciences
- Dalian 116023
| | - Yukui Zhang
- National Chromatographic R. & A. Center
- Key Laboratory of Separation Science for Analytical Chemistry
- Dalian Institute of Chemical Physics
- Chinese Academy of Sciences
- Dalian 116023
| |
Collapse
|
198
|
Kinna A, Tolner B, Rota EM, Titchener-Hooker N, Nesbeth D, Chester K. IMAC capture of recombinant protein from unclarified mammalian cell feed streams. Biotechnol Bioeng 2016; 113:130-40. [PMID: 26174988 PMCID: PMC4737217 DOI: 10.1002/bit.25705] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2015] [Revised: 06/30/2015] [Accepted: 07/10/2015] [Indexed: 12/25/2022]
Abstract
Fusion-tag affinity chromatography is a key technique in recombinant protein purification. Current methods for protein recovery from mammalian cells are hampered by the need for feed stream clarification. We have developed a method for direct capture using immobilized metal affinity chromatography (IMAC) of hexahistidine (His6) tagged proteins from unclarified mammalian cell feed streams. The process employs radial flow chromatography with 300-500 μm diameter agarose resin beads that allow free passage of cells but capture His-tagged proteins from the feed stream; circumventing expensive and cumbersome centrifugation and/or filtration steps. The method is exemplified by Chinese Hamster Ovary (CHO) cell expression and subsequent recovery of recombinant His-tagged carcinoembryonic antigen (CEA); a heavily glycosylated and clinically relevant protein. Despite operating at a high NaCl concentration necessary for IMAC binding, cells remained over 96% viable after passage through the column with host cell proteases and DNA detected at ∼ 8 U/mL and 2 ng/μL in column flow-through, respectively. Recovery of His-tagged CEA from unclarified feed yielded 71% product recovery. This work provides a basis for direct primary capture of fully glycosylated recombinant proteins from unclarified mammalian cell feed streams.
Collapse
Affiliation(s)
- Alexander Kinna
- Department of Oncology, University College London, UCL Cancer Institute, 72 Huntley Street, London, WC1E 6BT, UK
- Department of Biochemical Engineering, University College London, Gower Street, London, WC1E 6BT, UK
| | - Berend Tolner
- Department of Oncology, University College London, UCL Cancer Institute, 72 Huntley Street, London, WC1E 6BT, UK
| | - Enrique Miranda Rota
- Department of Oncology, University College London, UCL Cancer Institute, 72 Huntley Street, London, WC1E 6BT, UK
| | - Nigel Titchener-Hooker
- Department of Biochemical Engineering, University College London, Gower Street, London, WC1E 6BT, UK
| | - Darren Nesbeth
- Department of Biochemical Engineering, University College London, Gower Street, London, WC1E 6BT, UK
| | - Kerry Chester
- Department of Oncology, University College London, UCL Cancer Institute, 72 Huntley Street, London, WC1E 6BT, UK.
| |
Collapse
|
199
|
Last D, Müller J, Dawood AWH, Moldenhauer EJ, Pavlidis IV, Bornscheuer UT. Highly efficient and easy protease-mediated protein purification. Appl Microbiol Biotechnol 2015; 100:1945-1953. [DOI: 10.1007/s00253-015-7206-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2015] [Revised: 11/25/2015] [Accepted: 11/28/2015] [Indexed: 11/29/2022]
|
200
|
Wang HZ, Chu ZZ, Chen CC, Cao AC, Tong X, Ouyang CB, Yuan QH, Wang MN, Wu ZK, Wang HH, Wang SB. Recombinant Passenger Proteins Can Be Conveniently Purified by One-Step Affinity Chromatography. PLoS One 2015; 10:e0143598. [PMID: 26641240 PMCID: PMC4671538 DOI: 10.1371/journal.pone.0143598] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2015] [Accepted: 11/06/2015] [Indexed: 11/19/2022] Open
Abstract
Fusion tag is one of the best available tools to date for enhancement of the solubility or improvement of the expression level of recombinant proteins in Escherichia coli. Typically, two consecutive affinity purification steps are often necessitated for the purification of passenger proteins. As a fusion tag, acyl carrier protein (ACP) could greatly increase the soluble expression level of Glucokinase (GlcK), α-Amylase (Amy) and GFP. When fusion protein ACP-G2-GlcK-Histag and ACP-G2-Amy-Histag, in which a protease TEV recognition site was inserted between the fusion tag and passenger protein, were coexpressed with protease TEV respectively in E. coli, the efficient intracellular processing of fusion proteins was achieved. The resulting passenger protein GlcK-Histag and Amy-Histag accumulated predominantly in a soluble form, and could be conveniently purified by one-step Ni-chelating chromatography. However, the fusion protein ACP-GFP-Histag was processed incompletely by the protease TEV coexpressed in vivo, and a large portion of the resulting target protein GFP-Histag aggregated in insoluble form, indicating that the intracellular processing may affect the solubility of cleaved passenger protein. In this context, the soluble fusion protein ACP-GFP-Histag, contained in the supernatant of E. coli cell lysate, was directly subjected to cleavage in vitro by mixing it with the clarified cell lysate of E. coli overexpressing protease TEV. Consequently, the resulting target protein GFP-Histag could accumulate predominantly in a soluble form, and be purified conveniently by one-step Ni-chelating chromatography. The approaches presented here greatly simplify the purification process of passenger proteins, and eliminate the use of large amounts of pure site-specific proteases.
Collapse
Affiliation(s)
- Hua-zhen Wang
- College of Life Sciences, South China Agricultural University, Guangzhou, 541642, P. R. China
- Guangdong Provincial Key Laboratory of Protein Function and Regulation in Agricultural Organisms, Guangzhou, 541642, P. R. China
| | - Zhi-zhan Chu
- College of Life Sciences, South China Agricultural University, Guangzhou, 541642, P. R. China
- Guangdong Provincial Key Laboratory of Protein Function and Regulation in Agricultural Organisms, Guangzhou, 541642, P. R. China
| | - Chang-chao Chen
- College of Life Sciences, South China Agricultural University, Guangzhou, 541642, P. R. China
- Guangdong Provincial Key Laboratory of Protein Function and Regulation in Agricultural Organisms, Guangzhou, 541642, P. R. China
| | - Ao-cheng Cao
- Department of Pesticides, Institute of Plant Protection, Chinese Academy of Agricultural Sciences, Ministry of Agriculture, Beijing, 100193, China
- State Key Laboratory for Biology of Plant Diseases and Insect Pests, Beijing, 100193, China
| | - Xin Tong
- College of Life Sciences, South China Agricultural University, Guangzhou, 541642, P. R. China
- Guangdong Provincial Key Laboratory of Protein Function and Regulation in Agricultural Organisms, Guangzhou, 541642, P. R. China
| | - Can-bin Ouyang
- Department of Pesticides, Institute of Plant Protection, Chinese Academy of Agricultural Sciences, Ministry of Agriculture, Beijing, 100193, China
- State Key Laboratory for Biology of Plant Diseases and Insect Pests, Beijing, 100193, China
| | - Qi-hang Yuan
- College of Life Sciences, South China Agricultural University, Guangzhou, 541642, P. R. China
- Guangdong Provincial Key Laboratory of Protein Function and Regulation in Agricultural Organisms, Guangzhou, 541642, P. R. China
| | - Mi-nan Wang
- College of Life Sciences, South China Agricultural University, Guangzhou, 541642, P. R. China
- Guangdong Provincial Key Laboratory of Protein Function and Regulation in Agricultural Organisms, Guangzhou, 541642, P. R. China
| | - Zhong-kun Wu
- College of Life Sciences, South China Agricultural University, Guangzhou, 541642, P. R. China
- Guangdong Provincial Key Laboratory of Protein Function and Regulation in Agricultural Organisms, Guangzhou, 541642, P. R. China
| | - Hai-hong Wang
- College of Life Sciences, South China Agricultural University, Guangzhou, 541642, P. R. China
- Guangdong Provincial Key Laboratory of Protein Function and Regulation in Agricultural Organisms, Guangzhou, 541642, P. R. China
| | - Sheng-bin Wang
- College of Life Sciences, South China Agricultural University, Guangzhou, 541642, P. R. China
- Guangdong Provincial Key Laboratory of Protein Function and Regulation in Agricultural Organisms, Guangzhou, 541642, P. R. China
- * E-mail:
| |
Collapse
|