1
|
Loughran ST, Walls D. Tagging Recombinant Proteins to Enhance Solubility and Aid Purification. Methods Mol Biol 2023; 2699:97-123. [PMID: 37646996 DOI: 10.1007/978-1-0716-3362-5_7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/01/2023]
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 a long history, and there is a considerable repertoire of these that can be used to address 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 become indispensable tools for structural and functional proteomic initiatives that involve the expression of many proteins in parallel. In this chapter, 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 Life and Health Sciences, School of Health and Science, Dundalk Institute of Technology, Dundalk, Louth, Ireland.
| | - Dermot Walls
- School of Biotechnology, Dublin City University, Dublin, Ireland
| |
Collapse
|
2
|
Mahmoodi S, Pourhassan-Moghaddam M, Wood DW, Majdi H, Zarghami N. Current affinity approaches for purification of recombinant proteins. ACTA ACUST UNITED AC 2019. [DOI: 10.1080/23312025.2019.1665406] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Affiliation(s)
- Sahar Mahmoodi
- Department of Medical Biotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mohammad Pourhassan-Moghaddam
- Department of Medical Biotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
- Department of Medical Nanotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - David W. Wood
- Department of Chemical and Biomolecular Engineering, The Ohio State University, Columbus, OH 43210, USA
| | - Hasan Majdi
- Department of Medical Biotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
- Department of Medical Nanotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Nosratollah Zarghami
- Department of Medical Biotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
3
|
Matsuo Y, Maurer SP, Surrey T, Toda T. Purification and characterisation of the fission yeast Ndc80 complex. Protein Expr Purif 2017; 135:61-69. [PMID: 28502666 PMCID: PMC5489075 DOI: 10.1016/j.pep.2017.05.002] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2017] [Revised: 05/07/2017] [Accepted: 05/08/2017] [Indexed: 12/02/2022]
Abstract
The Ndc80 complex is a conserved outer kinetochore protein complex consisting of Ndc80 (Hec1), Nuf2, Spc24 and Spc25. This complex comprises a major, if not the sole, platform with which the plus ends of the spindle microtubules directly interact. In fission yeast, several studies indicate that multiple microtubule-associated proteins including the Dis1/chTOG microtubule polymerase and the Mal3/EB1 microtubule plus-end tracking protein directly or indirectly bind Ndc80, thereby ensuring stable kinetochore-microtubule attachment. However, the purification of the Ndc80 complex from this yeast has not been achieved, which hampers the in-depth investigation as to how the outer kinetochore attaches to the plus end of the spindle microtubule. Here we report the two-step purification of the fission yeast Ndc80 holo complex from bacteria. First, we purified separately two sub-complexes consisting of Ndc80-Nuf2 and Spc24-Spc25. Then, these two sub-complexes were mixed and applied to size-exclusion chromatography. The reconstituted Ndc80 holo complex is composed of four subunits with equal stoichiometry. The complex possesses microtubule-binding activity, and Total Internal Reflection Fluorescence (TIRF)-microscopy assays show that the complex binds the microtubule lattice. Interestingly, unlike the human complex, the fission yeast complex does not track depolymerising microtubule ends. Further analysis shows that under physiological ionic conditions, the Ndc80 holo complex does not detectably bind Dis1, but instead it interacts with Mal3/EB1, by which the Ndc80 complex tracks the growing microtubule plus end. This result substantiates the notion that the Ndc80 complex plays a crucial role in establishment of the dynamic kinetochore-microtubule interface by cooperating with chTOG and EB1. The Ndc80 complex is a conserved outer kinetochore complex. The fission yeast Ndc80 complex is purified from bacteria through a two-step purification scheme. The Ndc80 complex possesses microtubule-binding activity similar to those from other species. TIRF-microscopy assays show that the complex binds the microtubule lattice. The Ndc80 complex tracks the microtubule plus end when mixed with Mal3/EB1.
Collapse
Affiliation(s)
- Yuzy Matsuo
- Cell Regulation Laboratory, The Francis Crick Institute, 44 Lincoln's Inn Fields, London WC2A 3LY, UK; Synthetic and Systems Biochemistry of the Microtubule Cytoskeleton Laboratory, The Francis Crick Institute, 1 Midland Road, London, NW1 1AT, UK
| | - Sebastian P Maurer
- Synthetic and Systems Biochemistry of the Microtubule Cytoskeleton Laboratory, The Francis Crick Institute, 1 Midland Road, London, NW1 1AT, UK; Centre for Genomic Regulation (CRG), Barcelona Institute of Science and Technology (BIST), Dr. Aiguader 88, 08003 Barcelona, Spain; Universitat Pompeu Fabra (UPF), Barcelona, Spain
| | - Thomas Surrey
- Synthetic and Systems Biochemistry of the Microtubule Cytoskeleton Laboratory, The Francis Crick Institute, 1 Midland Road, London, NW1 1AT, UK.
| | - Takashi Toda
- Cell Regulation Laboratory, The Francis Crick Institute, 44 Lincoln's Inn Fields, London WC2A 3LY, UK; Hiroshima Research Center for Healthy Aging (HiHA), Department of Molecular Biotechnology, Graduate School of Advanced Science of Matter, Hiroshima University, 1-3-1 Kagamiyama, Higashi-Hiroshima 739-8530, Japan.
| |
Collapse
|
4
|
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
|
5
|
Friberg A, Thumann S, Hennig J, Zou P, Nössner E, Ling PD, Sattler M, Kempkes B. The EBNA-2 N-Terminal Transactivation Domain Folds into a Dimeric Structure Required for Target Gene Activation. PLoS Pathog 2015; 11:e1004910. [PMID: 26024477 PMCID: PMC4449002 DOI: 10.1371/journal.ppat.1004910] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2015] [Accepted: 04/24/2015] [Indexed: 11/18/2022] Open
Abstract
Epstein-Barr virus (EBV) is a γ-herpesvirus that may cause infectious mononucleosis in young adults. In addition, epidemiological and molecular evidence links EBV to the pathogenesis of lymphoid and epithelial malignancies. EBV has the unique ability to transform resting B cells into permanently proliferating, latently infected lymphoblastoid cell lines. Epstein-Barr virus nuclear antigen 2 (EBNA-2) is a key regulator of viral and cellular gene expression for this transformation process. The N-terminal region of EBNA-2 comprising residues 1-58 appears to mediate multiple molecular functions including self-association and transactivation. However, it remains to be determined if the N-terminus of EBNA-2 directly provides these functions or if these activities merely depend on the dimerization involving the N-terminal domain. To address this issue, we determined the three-dimensional structure of the EBNA-2 N-terminal dimerization (END) domain by heteronuclear NMR-spectroscopy. The END domain monomer comprises a small fold of four β-strands and an α-helix which form a parallel dimer by interaction of two β-strands from each protomer. A structure-guided mutational analysis showed that hydrophobic residues in the dimer interface are required for self-association in vitro. Importantly, these interface mutants also displayed severely impaired self-association and transactivation in vivo. Moreover, mutations of solvent-exposed residues or deletion of the α-helix do not impair dimerization but strongly affect the functional activity, suggesting that the EBNA-2 dimer presents a surface that mediates functionally important intra- and/or intermolecular interactions. Our study shows that the END domain is a novel dimerization fold that is essential for functional activity. Since this specific fold is a unique feature of EBNA-2 it might provide a novel target for anti-viral therapeutics. Epstein-Barr virus is an oncogenic γ-herpesvirus that may cause infectious mononucleosis in young adults and fatal lymphoproliferative disorders in immunocompromised patients and is associated with the pathogenesis of Burkitt's lymphoma, nasopharyngeal and gastric carcinoma. Epstein-Barr virus nuclear antigen 2 (EBNA-2) is a key regulator of viral and cellular gene expression which initiates and maintains a specific transcription program that promotes proliferation and differentiation of the infected B cell. EBNA-2 is a transcriptional activator that is recruited to DNA by cellular adaptor proteins, carries two transactivation domains, and has the capacity to form dimers or multimers. This study provides the first three-dimensional structure of the EBNA-2 N-terminal Dimerization (END) domain. Two END domain monomers, each consisting of four β-strands and a single α-helix, assemble into a dimer by interaction of two β-strands from each monomer in a parallel fashion. The dimer surface exposes residues that are critical for transactivation of target genes by EBNA-2. The dimeric fold of the EBNA-2 END domain has not been observed for any cellular protein and thus could provide a novel target for anti-viral therapeutics.
Collapse
Affiliation(s)
- Anders Friberg
- Institute of Structural Biology, Helmholtz Zentrum München, National Research Center for Environmental Health, Neuherberg, Germany
- Center for Integrated Protein Science Munich at Chair Biomolecular NMR Spectroscopy, Department Chemie, Technische Universität München, Garching, Germany
| | - Sybille Thumann
- Department of Gene Vectors, Hematologikum, Helmholtz Zentrum München, National Research Center for Environmental Health, München, Germany
| | - Janosch Hennig
- Institute of Structural Biology, Helmholtz Zentrum München, National Research Center for Environmental Health, Neuherberg, Germany
- Center for Integrated Protein Science Munich at Chair Biomolecular NMR Spectroscopy, Department Chemie, Technische Universität München, Garching, Germany
| | - Peijian Zou
- Institute of Structural Biology, Helmholtz Zentrum München, National Research Center for Environmental Health, Neuherberg, Germany
- Center for Integrated Protein Science Munich at Chair Biomolecular NMR Spectroscopy, Department Chemie, Technische Universität München, Garching, Germany
- Industrial Enzymes National Engineering Laboratory, Tianjin Institute of Industrial Biotechnology, Chinese Academy of Sciences, Tianjin, China
| | - Elfriede Nössner
- Institute of Molecular Immunology, Hematologikum, Helmholtz Zentrum München, National Research Center for Environmental Health, München, Germany
| | - Paul D. Ling
- Department of Molecular Virology & Microbiology, Baylor College of Medicine, Houston, Texas, United States of America
| | - Michael Sattler
- Institute of Structural Biology, Helmholtz Zentrum München, National Research Center for Environmental Health, Neuherberg, Germany
- Center for Integrated Protein Science Munich at Chair Biomolecular NMR Spectroscopy, Department Chemie, Technische Universität München, Garching, Germany
- Industrial Enzymes National Engineering Laboratory, Tianjin Institute of Industrial Biotechnology, Chinese Academy of Sciences, Tianjin, China
- * E-mail: (MS); (BK)
| | - Bettina Kempkes
- Department of Gene Vectors, Hematologikum, Helmholtz Zentrum München, National Research Center for Environmental Health, München, Germany
- * E-mail: (MS); (BK)
| |
Collapse
|
6
|
Purification antibody by thiophilic magnetic sorbent modified with 2-mercapto-1-methylimidazol. Colloids Surf B Biointerfaces 2013; 108:72-9. [DOI: 10.1016/j.colsurfb.2013.02.037] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2012] [Revised: 02/21/2013] [Accepted: 02/22/2013] [Indexed: 01/28/2023]
|
7
|
Xu N, Yuan H, Liu W, Li S, Liu Y, Wan J, Li X, Zhang R, Chang Y. Activation of RAW264.7 mouse macrophage cells in vitro through treatment with recombinant ricin toxin-binding subunit B: involvement of protein tyrosine, NF-κB and JAK-STAT kinase signaling pathways. Int J Mol Med 2013; 32:729-35. [PMID: 23820591 DOI: 10.3892/ijmm.2013.1426] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2013] [Accepted: 06/18/2013] [Indexed: 11/06/2022] Open
Abstract
Ricin toxin-binding subunit B (RTB) is a galactose-binding lectin protein. In the present study, we investigated the effects of RTB on inducible nitric oxide (NO) synthase (iNOS), interleukin (IL)-6 and tumor necrosis factor (TNF)-α, as well as the signal transduction mechanisms involved in recombinant RTB-induced macrophage activation. RAW264.7 macrophages were treated with RTB. The results revealed that the mRNA and protein expression of iNOS was increased in the recombinant RTB-treated macrophages. TNF-α production was observed to peak at 20 h, whereas the production of IL-6 peaked at 24 h. In another set of cultures, the cells were co-incubated with RTB and the tyrosine kinase inhibitor, genistein, the phosphatidylinositol 3-kinase (PI3K) inhibitor, LY294002, the p42/44 inhibitor, PD98059, the p38 inhibitor, SB203580, the JNK inhibitor, SP600125, the protein kinase C (PKC) inhibitor, staurosporine, the JAK2 inhibitor, tyrphostin (AG490), or the NOS inhibitor, L-NMMA. The recombinant RTB-induced production of NO, TNF-α and IL-6 was inhibited in the macrophages treated with the pharmacological inhibitors genistein, LY294002, staurosporine, AG490, SB203580 and BAY 11-7082, indicating the possible involvement of protein tyrosine kinases, PI3K, PKC, JAK2, p38 mitogen-activated protein kinase (MAPK) and nuclear factor (NF)-κB in the above processes. A phosphoprotein analysis identified tyrosine phosphorylation targets that were uniquely induced by recombinant RTB and inhibited following treatment with genistein; some of these proteins are associated with the downstream cascades of activated JAK-STAT and NF-κB receptors. Our data may help to identify the most important target molecules for the development of novel drug therapies.
Collapse
Affiliation(s)
- Na Xu
- Department of Immunology, Norman Bethune College of Medical Science, Jilin University, Jilin, P.R. China
| | | | | | | | | | | | | | | | | |
Collapse
|
8
|
Lin X, Xie J, Chen X. Protein-based tumor molecular imaging probes. Amino Acids 2011; 41:1013-36. [PMID: 20232092 PMCID: PMC3617487 DOI: 10.1007/s00726-010-0545-z] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2010] [Accepted: 02/24/2010] [Indexed: 12/30/2022]
Abstract
Molecular imaging is an emerging discipline which plays critical roles in diagnosis and therapeutics. It visualizes and quantifies markers that are aberrantly expressed during the disease origin and development. Protein molecules remain to be one major class of imaging probes, and the option has been widely diversified due to the recent advances in protein engineering techniques. Antibodies are part of the immunosystem which interact with target antigens with high specificity and affinity. They have long been investigated as imaging probes and were coupled with imaging motifs such as radioisotopes for that purpose. However, the relatively large size of antibodies leads to a half-life that is too long for common imaging purposes. Besides, it may also cause a poor tissue penetration rate and thus compromise some medical applications. It is under this context that various engineered protein probes, essentially antibody fragments, protein scaffolds, and natural ligands have been developed. Compared to intact antibodies, they possess more compact size, shorter clearance time, and better tumor penetration. One major challenge of using protein probes in molecular imaging is the affected biological activity resulted from random labeling. Site-specific modification, however, allows conjugation happening in a stoichiometric fashion with little perturbation of protein activity. The present review will discuss protein-based probes with focus on their application and related site-specific conjugation strategies in tumor imaging.
Collapse
Affiliation(s)
- Xin Lin
- Laboratory of Molecular Imaging and Nanomedicine, National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, MD 20892, USA
| | | | | |
Collapse
|
9
|
Affiliation(s)
- Per-Ake Nygren
- Division of Molecular Biotechnology, School of Biotechnology, Royal Institute of Technology (KTH), SE-106 91 Stockholm, Sweden
| |
Collapse
|
10
|
Wållberg H, Löfdahl PÅ, Tschapalda K, Uhlén M, Tolmachev V, Nygren PÅ, Ståhl S. Affinity recovery of eight HER2-binding affibody variants using an anti-idiotypic affibody molecule as capture ligand. Protein Expr Purif 2011; 76:127-35. [DOI: 10.1016/j.pep.2010.10.008] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2010] [Revised: 10/19/2010] [Accepted: 10/19/2010] [Indexed: 10/18/2022]
|
11
|
Walls D, Loughran ST. Tagging recombinant proteins to enhance solubility and aid purification. Methods Mol Biol 2011; 681:151-175. [PMID: 20978965 DOI: 10.1007/978-1-60761-913-0_9] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/30/2023]
Abstract
Protein fusion technology has enormously facilitated 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 are outlined.
Collapse
Affiliation(s)
- Dermot Walls
- School of Biotechnology and National Centre for Sensor Research, Dublin City University, Dublin, Ireland.
| | | |
Collapse
|
12
|
Qian H, Li C, Lin Z, Zhang Y. Using thiophilic magnetic beads in purification of antibodies from human serum. Colloids Surf B Biointerfaces 2010; 75:342-8. [DOI: 10.1016/j.colsurfb.2009.09.007] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2009] [Revised: 07/15/2009] [Accepted: 09/09/2009] [Indexed: 10/20/2022]
|
13
|
Qian H, Li C, Zhang Y, Lin Z. Efficient isolation of immunoglobulin G by paramagnetic polymer beads modified with 2-mercapto-4-mythyl-pyrimidine. J Immunol Methods 2009; 343:119-29. [DOI: 10.1016/j.jim.2009.02.001] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
|
14
|
Grönwall C, Ståhl S. Engineered affinity proteins—Generation and applications. J Biotechnol 2009; 140:254-69. [DOI: 10.1016/j.jbiotec.2009.01.014] [Citation(s) in RCA: 89] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2008] [Revised: 12/05/2008] [Accepted: 01/26/2009] [Indexed: 12/11/2022]
|
15
|
Qian H, Lin Z, Xu H, Chen M. The efficient and specific isolation of the antibodies from human serum by thiophilic paramagnetic polymer nanospheres. Biotechnol Prog 2009; 25:376-83. [DOI: 10.1002/btpr.105] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
|
16
|
The effective and specific isolation of antibodies from human serum by using thiophilic paramagnetic polymer beads. Colloids Surf B Biointerfaces 2008; 67:224-9. [DOI: 10.1016/j.colsurfb.2008.08.023] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2008] [Revised: 08/26/2008] [Accepted: 08/29/2008] [Indexed: 11/24/2022]
|
17
|
Heterologous high-level E. coli expression, purification and biophysical characterization of the spine-associated RapGAP (SPAR) PDZ domain. Protein Expr Purif 2008; 62:9-14. [PMID: 18678258 DOI: 10.1016/j.pep.2008.07.003] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2008] [Revised: 07/10/2008] [Accepted: 07/11/2008] [Indexed: 11/22/2022]
Abstract
Spine-associated RapGAP (SPAR) is a 1783 residue, multidomain scaffolding protein which is a component of the NMDA receptor/PSD-95 complex in the post-synaptic density (PSD) of dendritic spines. Using a parallel expression screening approach, we identified a strategy to solubly express the SPAR PDZ domain in Escherichia coli. We show that maltose binding protein is required for the production of solubly expressed protein. We also show that small changes in construct length (2-5 residues) result in differential susceptibilities of the expressed proteins to proteolytic digestion, required for the expression tag removal. This has allowed us to identify a large-scale E. coli expression and purification protocol that results in the production of mg quantities of the SPAR PDZ domain. This is the first time that any of the multiple SPAR functional domains have been expressed in E. coli in quantities suitable for biophysical and biochemical studies, allowing us to investigate the role of the PDZ domain in SPAR function within the PSD.
Collapse
|
18
|
Grönwall C, Sjöberg A, Ramström M, Höidén-Guthenberg I, Hober S, Jonasson P, Ståhl S. Affibody-mediated transferrin depletion for proteomics applications. Biotechnol J 2007; 2:1389-98. [PMID: 17639529 DOI: 10.1002/biot.200700053] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
An Affibody (Affibody) ligand with specific binding to human transferrin was selected by phage display technology from a combinatorial protein library based on the staphylococcal protein A (SpA)-derived Z domain. Strong and selective binding of the selected Affibody ligand to transferrin was demonstrated using biosensor technology and dot blot analysis. Impressive specificity was demonstrated as transferrin was the only protein recovered by affinity chromatography from human plasma. Efficient Affibody-mediated capture of transferrin, combined with IgG- and HSA-depletion, was demonstrated for human plasma and cerebrospinal fluid (CSF). For plasma, 85% of the total transferrin content in the samples was depleted after only two cycles of transferrin removal, and for CSF, 78% efficiency was obtained in single-step depletion. These results clearly suggest a potential for the development of Affibody-based resins for the removal of abundant proteins in proteomics analyses.
Collapse
Affiliation(s)
- Caroline Grönwall
- Department of Molecular Biotechnology, School of Biotechnology, AlbaNova University Center, Royal Institute of Technology (KTH), Stockholm, Sweden
| | | | | | | | | | | | | |
Collapse
|
19
|
Renberg B, Nordin J, Merca A, Uhlén M, Feldwisch J, Nygren PA, Karlström AE. Affibody molecules in protein capture microarrays: evaluation of multidomain ligands and different detection formats. J Proteome Res 2007; 6:171-9. [PMID: 17203961 DOI: 10.1021/pr060316r] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
The importance of the ligand presentation format for the production of protein capture microarrays was evaluated using different Affibody molecules, produced either as single 6 kDa monomers or genetically linked head-to-tail multimers containing up to four domains. The performances in terms of selectivity and sensitivity of the monomeric and the multidomain Affibody molecules were compared by immobilization of the ligands on microarray slides, followed by incubation with fluorescent-labeled target protein. An increase in signal intensities for the multimers was demonstrated, with the most pronounced difference observed between monomers and dimers. A protein microarray containing six different dimeric Affibody ligands with specificity for IgA, IgE, IgG, TNF-alpha, insulin, or Taq DNA polymerase was characterized for direct detection of fluorescent-labeled analytes. No cross-reactivity was observed and the limits of detection were 600 fM for IgA, 20 pM for IgE, 70 fM for IgG, 20 pM for TNF-alpha, 60 pM for insulin, and 10 pM for Taq DNA polymerase. Also, different sandwich formats for detection of unlabeled protein were evaluated and used for selective detection of IgA or TNF-alpha in human serum or plasma samples, respectively. Finally, the presence of IgA was determined using detection of directly Cy5-labeled normal or IgA-deficient serum samples.
Collapse
Affiliation(s)
- Björn Renberg
- School of Biotechnology, Division of Molecular Biotechnology, Royal Institute of Technology, AlbaNova University Center, SE - 106 91 Stockholm, Sweden
| | | | | | | | | | | | | |
Collapse
|
20
|
Affiliation(s)
- Johannes M Herrmann
- Institut für Zellbiologie, Universität Kaiserslautern, 67663 Kaiserslautern, Germany
| | | |
Collapse
|
21
|
Tolmachev V, Orlova A, Nilsson FY, Feldwisch J, Wennborg A, Abrahmsén L. Affibody molecules: potential for in vivo imaging of molecular targets for cancer therapy. Expert Opin Biol Ther 2007; 7:555-68. [PMID: 17373906 DOI: 10.1517/14712598.7.4.555] [Citation(s) in RCA: 101] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
Targeting radionuclide imaging of tumor-associated antigens may help to select patients who will benefit from a particular biological therapy. Affibody molecules are a novel class of small (approximately 7 kDa) phage display-selected affinity proteins, based on the B-domain scaffold of staphylococcal protein A. A large library (3 x 10(9) variants) has enabled selection of high-affinity (up to 22 pM) binders for a variety of tumor-associated antigens. The small size of Affibody molecules provides rapid tumor localization and fast clearance from nonspecific compartments. Preclinical studies have demonstrated the potential of Affibody molecules for specific and high-contrast radionuclide imaging of HER2 in vivo, and pilot clinical data using indium-111 and gallium-68 labeled anti-HER2 Affibody tracer have confirmed its utility for radionuclide imaging in cancer patients.
Collapse
|
22
|
Lundberg E, Höidén-Guthenberg I, Larsson B, Uhlén M, Gräslund T. Site-specifically conjugated anti-HER2 Affibody molecules as one-step reagents for target expression analyses on cells and xenograft samples. J Immunol Methods 2006; 319:53-63. [PMID: 17196217 DOI: 10.1016/j.jim.2006.10.013] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2006] [Revised: 10/13/2006] [Accepted: 10/21/2006] [Indexed: 01/21/2023]
Abstract
Affibody molecules are a class of small and robust affinity proteins that can be generated to interact with a variety of antigens, thus having the potential to provide useful tools for biotechnological research and diagnostic applications. In this study, we have investigated Affibody-based reagents interacting specifically with the tyrosine kinase receptor HER2. A head-to-tail dimeric construct was site-specifically conjugated with different fluorescent and enzymatic groups resulting in reagents that were used for detection and quantification. The amount of cell surface expressed HER2 on eleven (11) well characterized cell lines was quantified relative to each other by flow cytometry and shown to correlate well with results from parallel analyses of HER2 mRNA levels measured by real-time PCR. Further, immunofluorescence microscopy studies of the cell lines and immunohistochemical analyses of cryosections of HER2 expressing SKOV-3 xenografts showed strong staining of the plasma membrane of tumor cells with little background staining. Full-length HER2 protein could also be efficiently recovered from a cell extract by an immunoprecipitation procedure, using an Affibody ligand-based resin. These novel non-IgG derived reagents could be used to detect and quantify HER2 expression. By adapting the methods for use with Affibody molecules binding to other cell surface receptors, it is anticipated that also these receptors can be detected and quantified in a similar manner.
Collapse
Affiliation(s)
- Emma Lundberg
- Department of Biotechnology, Albanova University Center, Royal Institute of Technology, SE-106 91, Stockholm, Sweden
| | | | | | | | | |
Collapse
|
23
|
Grönwall C, Jonsson A, Lindström S, Gunneriusson E, Ståhl S, Herne N. Selection and characterization of Affibody ligands binding to Alzheimer amyloid beta peptides. J Biotechnol 2006; 128:162-83. [PMID: 17088007 DOI: 10.1016/j.jbiotec.2006.09.013] [Citation(s) in RCA: 92] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2006] [Revised: 09/05/2006] [Accepted: 09/11/2006] [Indexed: 11/19/2022]
Abstract
Affibody (Affibody) ligands specific for human amyloid beta (Abeta) peptides (40 or 42 amino acid residues in size), involved in the progress of Alzheimer's disease, were selected by phage display technology from a combinatorial protein library based on the 58-amino acid residue staphylococcal protein A-derived Z domain. Post-selection screening of 384 randomly picked clones, out of which 192 clones were subjected to DNA sequencing and clustering, resulted in the identification of 16 Affibody variants that were produced and affinity purified for ranking of their binding properties. The two most promising Affibody variants were shown to selectively and efficiently bind to Abeta peptides, but not to the control proteins. These two Affibody ligands were in dimeric form (to gain avidity effects) coupled to affinity resins for evaluation as affinity devices for capture of Abeta peptides from human plasma and serum. It was found that both ligands could efficiently capture Abeta that were spiked (100 microgml(-1)) to plasma and serum samples. A ligand multimerization problem that would yield suboptimal affinity resins, caused by a cysteine residue present at the binding surface of the Affibody ligands, could be circumvented by the generation of second-generation Affibody ligands (having cysteine to serine substitutions). In an epitope mapping effort, the preferred binding site of selected Affibody ligands was mapped to amino acids 30-36 of Abeta, which fortunately would indicate that the Affibody molecules should not bind the amyloid precursor protein (APP). In addition, a significant effort was made to analyze which form of Abeta (monomer, dimer or higher aggregates) that was most efficiently captured by the selected Affibody ligand. By using Western blotting and a dot blot assay in combination with size exclusion chromatography, it could be concluded that selected Affibody ligands predominantly bound a non-aggregated form of analyzed Abeta peptide, which we speculate to be dimeric Abeta. In conclusion, we have successfully selected Affibody ligands that efficiently capture Abeta peptides from human plasma and serum. The potential therapeutic use of these optimized ligands for extracorporeal capture of Abeta peptides in order to slow down or reduce amyloid plaque formation, is discussed.
Collapse
Affiliation(s)
- Caroline Grönwall
- Department of Molecular Biotechnology, School of Biotechnology, AlbaNova University Center, Royal Institute of Technology (KTH), SE-106 91 Stockholm, Sweden
| | | | | | | | | | | |
Collapse
|
24
|
Peti W, Page R. Strategies to maximize heterologous protein expression in Escherichia coli with minimal cost. Protein Expr Purif 2006; 51:1-10. [PMID: 16904906 DOI: 10.1016/j.pep.2006.06.024] [Citation(s) in RCA: 169] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2006] [Revised: 06/20/2006] [Accepted: 06/26/2006] [Indexed: 11/23/2022]
Abstract
Automation and miniaturization are key issues of high-throughput research projects in the post-genomic era. The implementation of robotics and parallelization has enabled researchers to process large numbers of protein targets for structural studies in a short time with reasonable cost efficiency. However, the cost of implementing the robotics and parallelization often prohibit their use in the traditional academic laboratory. Fortunately, multiple groups have made significant efforts to minimize the cost of heterologous protein expression for the production of protein samples in quantities suitable for high resolution structural studies. In this review, we describe recent efforts to continue to minimize the cost for the parallel processing of multiple protein targets and focus on those materials and strategies that are highly suitable for the traditional academic laboratory.
Collapse
Affiliation(s)
- Wolfgang Peti
- Brown University, Department of Molecular Pharmacology, Physiology, and Biotechnology, Box G-E3, Providence, RI 02912, USA
| | | |
Collapse
|
25
|
Graumann K, Premstaller A. Manufacturing of recombinant therapeutic proteins in microbial systems. Biotechnol J 2006; 1:164-86. [PMID: 16892246 DOI: 10.1002/biot.200500051] [Citation(s) in RCA: 108] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Recombinant therapeutic proteins have gained enormous importance for clinical applications. The first recombinant products have been produced in E. coli more than 20 years ago. Although with the advent of antibody-based therapeutics mammalian expression systems have experienced a major boost, microbial expression systems continue to be widely used in industry. Their intrinsic advantages, such as rapid growth, high yields and ease of manipulation, make them the premier choice for expression of non-glycosylated peptides and proteins. Innovative product classes such as antibody fragments or alternative binding molecules will further expand the use of microbial systems. Even more, novel, engineered production hosts and integrated technology platforms hold enormous potential for future applications. This review summarizes current applications and trends for development, production and analytical characterization of recombinant therapeutic proteins in microbial systems.
Collapse
Affiliation(s)
- Klaus Graumann
- Novartis Biopharmaceutical Operations, Sandoz GmbH, Biochemiestrasse 10, 6250 Kundl, Austria.
| | | |
Collapse
|
26
|
Arnau J, Lauritzen C, Petersen GE, Pedersen J. Current strategies for the use of affinity tags and tag removal for the purification of recombinant proteins. Protein Expr Purif 2005; 48:1-13. [PMID: 16427311 DOI: 10.1016/j.pep.2005.12.002] [Citation(s) in RCA: 449] [Impact Index Per Article: 23.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2005] [Revised: 11/22/2005] [Accepted: 12/02/2005] [Indexed: 10/25/2022]
Abstract
Affinity tags are highly efficient tools for protein purification. They allow the purification of virtually any protein without any prior knowledge of its biochemical properties. The use of affinity tags has therefore become widespread in several areas of research e.g., high throughput expression studies aimed at finding a biological function to large numbers of yet uncharacterized proteins. In some cases, the presence of the affinity tag in the recombinant protein is unwanted or may represent a disadvantage for the projected application of the protein, like for clinical use. Therefore, an increasing number of approaches are available at present that are designed for the removal of the affinity tag from the recombinant protein. Most of these methods employ recombinant endoproteases that recognize a specific sequence. These process enzymes can subsequently be removed from the process by affinity purification, since they also include a tag. Here, a survey of the most common affinity tags and the current methods for tag removal is presented, with special emphasis on the removal of N-terminal histidine tags using TAGZyme, a system based on exopeptidase cleavage. In the quest to reduce the significant costs associated with protein purification at large scale, relevant aspects involved in the development of downstream processes for pharmaceutical protein production that incorporate a tag removal step are also discussed. A comparison of the yield of standard vs. affinity purification together with an example of tag removal using TAGZyme is also included.
Collapse
Affiliation(s)
- José Arnau
- Unizyme Laboratories A/S, Dr. Neergaards vej 17, DK-2970 Hørsholm, Denmark.
| | | | | | | |
Collapse
|
27
|
Hedhammar M, Gräslund T, Hober S. Protein Engineering Strategies for Selective Protein Purification. Chem Eng Technol 2005. [DOI: 10.1002/ceat.200500144] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
|
28
|
Renberg B, Shiroyama I, Engfeldt T, Nygren PK, Karlström AE. Affibody protein capture microarrays: Synthesis and evaluation of random and directed immobilization of affibody molecules. Anal Biochem 2005; 341:334-43. [PMID: 15907880 DOI: 10.1016/j.ab.2005.03.039] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2005] [Indexed: 11/21/2022]
Abstract
Affibody molecules, 58-amino acid three-helix bundle proteins directed to different targets by combinatorial engineering of staphylococcal protein A, were used as capture ligands on protein microarrays. An evaluation of slide types and immobilization strategies was performed to find suitable conditions for microarray production. Two affibody molecules, Z(Taq) and Z(IgA), binding Taq DNA polymerase and human IgA, respectively, were synthesized by solid phase peptide synthesis using an orthogonal protection scheme, allowing incorporation of selective immobilization handles. The resulting affibody variants were used for random surface immobilization (through amino groups) or oriented surface immobilization (through cysteine or biotin coupled to the side chain of Lys58). Evaluation of the immobilization techniques was carried out using both a real-time surface plasmon resonance biosensor system and a microarray system using fluorescent detection of Cy3-labeled target protein. The results from the biosensor analyses showed that directed immobilization strategies significantly improved the specific binding activity of affibody molecules. However, in the microarray system, random immobilization onto carboxymethyl dextran slides and oriented immobilization onto thiol dextran slides resulted in equally good signal intensities, whereas biotin-mediated immobilization onto streptavidin-coated slides produced slides with lower signal intensities and higher background staining. For the best slides, the limit of detection was 3 pM for IgA and 30 pM for Taq DNA polymerase.
Collapse
Affiliation(s)
- Björn Renberg
- Department of Biotechnology, Royal Institute of Technology, AlbaNova University Center, S-10691 Stockholm, Sweden
| | | | | | | | | |
Collapse
|
29
|
Faye L, Boulaflous A, Benchabane M, Gomord V, Michaud D. Protein modifications in the plant secretory pathway: current status and practical implications in molecular pharming. Vaccine 2005; 23:1770-8. [PMID: 15734039 DOI: 10.1016/j.vaccine.2004.11.003] [Citation(s) in RCA: 133] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Plants have become, over the last ten years, a suitable alternative to microbial and animal cell factories for the production of clinically-useful, therapeutic proteins. Besides the well known advantage of low-cost and large-scale production of safe and biologically active mammalian proteins, plants also are able to perform most post-translational maturations required for biological activity and suitable pharmacokinetics of recombinant therapeutic proteins. In this short review we focus on glycosylation and proteolytic processing of plant-made pharmaceuticals during their transport through the plant cell's secretory pathway. We also address the practical implications of these important processes on the effectiveness of plant molecular pharming systems.
Collapse
Affiliation(s)
- Loïc Faye
- CNRS UMR 6037, IFRMP 23, GDR 2590, Université de Rouen-Bâtiment de biologie (extension), Blvd de Broglie, 76821 Mont Saint Aignan Cedex, France.
| | | | | | | | | |
Collapse
|
30
|
Lendel C, Dincbas-Renqvist V, Flores A, Wahlberg E, Dogan J, Nygren PA, Härd T. Biophysical characterization of Z(SPA-1)--a phage-display selected binder to protein A. Protein Sci 2004; 13:2078-88. [PMID: 15238637 PMCID: PMC2279809 DOI: 10.1110/ps.04728604] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
Affibodies are a novel class of binding proteins selected from phagemid libraries of the Z domain from staphylococcal protein A. The Z(SPA-1) affibody was selected as a binder to protein A, and it binds the parental Z domain with micromolar affinity. In earlier work we determined the structure of the Z:Z(SPA-1) complex and noted that Z(SPA-1) in the free state exhibits several properties characteristic of a molten globule. Here we present a more detailed biophysical investigation of Z(SPA-1) and four Z(SPA-1) mutants with the objective to understand these properties. The characterization includes thermal and chemical denaturation profiles, ANS binding assays, size exclusion chromatography, isothermal titration calorimetry, and an investigation of structure and dynamics by NMR. The NMR characterization of Z(SPA-1) was facilitated by the finding that trimethylamine N-oxide (TMAO) stabilizes the molten globule conformation in favor of the fully unfolded state. All data taken together lead us to conclude the following: (1) The topology of the molten globule conformation of free Z(SPA-1) is similar to that of the fully folded structure in the Z-bound state; (2) the extensive mutations in helices 1 and 2 destabilize these without affecting the intrinsic stability of helix 3; (3) stabilization and reduced aggregation can be achieved by replacing mutated residues in Z(SPA-1) with the corresponding wild-type Z residues. This stabilization is better correlated to changes in helix propensity than to an expected increase in polar versus nonpolar surface area of the fully folded state.
Collapse
Affiliation(s)
- Christofer Lendel
- KTH Biotechnology, Royal Institute of Technology (KTH), SE-106 91 Stockholm, Sweden.
| | | | | | | | | | | | | |
Collapse
|
31
|
Eklund M, Sandström K, Teeri TT, Nygren PA. Site-specific and reversible anchoring of active proteins onto cellulose using a cellulosome-like complex. J Biotechnol 2004; 109:277-86. [PMID: 15066765 DOI: 10.1016/j.jbiotec.2004.01.008] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2003] [Revised: 12/22/2003] [Accepted: 01/16/2004] [Indexed: 10/26/2022]
Abstract
Protein engineering strategies facilitating controlled and spontaneous assembly of macromolecular complexes are of great interest for the design of artificial multi-enzyme systems of pre-defined composition. Here we have combined affinity proteins from different sources to achieve specific and reversible anchoring of affinity domain-tagged reporter proteins to a cellulose-anchored fusion protein. The design principle mimics the architecture of macromolecular cellulosome complexes produced by some cellulolytic microbes. A fusion protein between a cellulose-binding module (CBM1Cel6A) of the Trichoderma reesei cellobiohydrolase Cel6A and a five-domain staphylococcal protein A (SPA) was constructed to serve as platform for docking of easily detectable reporter proteins onto cellulose surfaces. In turn, the reporter proteins were produced as fusions to two copies of a SPA-binding affinity protein (an affibody denoted Z(SPA-1)), selected from a phage display library constructed by combinatorial protein engineering. In a series of experiments, involving repeated washing and low pH elution, affinity-tagged Enhanced Green Fluorescent Protein (EGFP) and Fusarium solani pisi lipase cutinase reporter proteins were both found to be specifically directed from solution to the same region of a cellulose filter paper where SPA-CBM1Cel6A fusion protein had been previously applied. This showed that the SPA-CBM1Cel6A fusion protein had been stably anchored to the cellulose surface without loss of binding capacity and that the interaction between SPA and the Z(SPA-1) affibody domains was selective. The generality of this biospecificity-driven system for assembly applications is discussed.
Collapse
Affiliation(s)
- Malin Eklund
- Department of Biotechnology, Royal Institute of Technology (KTH), AlbaNova University Center, Roslagstullsbacken 21, SE-106 91 Stockholm, Sweden
| | | | | | | |
Collapse
|