1
|
Martina M, Banderali U, Yogi A, Arbabi Ghahroudi M, Liu H, Sulea T, Durocher Y, Hussack G, van Faassen H, Chakravarty B, Liu QY, Iqbal U, Ling B, Lessard E, Sheff J, Robotham A, Callaghan D, Moreno M, Comas T, Ly D, Stanimirovic D. A Novel Antigen Design Strategy to Isolate Single-Domain Antibodies that Target Human Nav1.7 and Reduce Pain in Animal Models. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2405432. [PMID: 39206821 PMCID: PMC11516162 DOI: 10.1002/advs.202405432] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/17/2024] [Revised: 08/01/2024] [Indexed: 09/04/2024]
Abstract
Genetic studies have identified the voltage-gated sodium channel 1.7 (Nav1.7) as pain target. Due to the ineffectiveness of small molecules and monoclonal antibodies as therapeutics for pain, single-domain antibodies (VHHs) are developed against the human Nav1.7 (hNav1.7) using a novel antigen presentation strategy. A 70 amino-acid peptide from the hNav1.7 protein is identified as a target antigen. A recombinant version of this peptide is grafted into the complementarity determining region 3 (CDR3) loop of an inert VHH in order to maintain the native 3D conformation of the peptide. This antigen is used to isolate one VHH able to i) bind hNav1.7, ii) slow the deactivation of hNav1.7, iii) reduce the ability of eliciting action potentials in nociceptors, and iv) reverse hyperalgesia in in vivo rat and mouse models. This VHH exhibits the potential to be developed as a therapeutic capable of suppressing pain. This novel antigen presentation strategy can be applied to develop biologics against other difficult targets such as ion channels, transporters and GPCRs.
Collapse
Affiliation(s)
- Marzia Martina
- Human Health Therapeutics Research CenterNational Research Council Canada1200 Montreal Road, Building M54OttawaONK1A 0R6Canada
| | - Umberto Banderali
- Human Health Therapeutics Research CenterNational Research Council Canada1200 Montreal Road, Building M54OttawaONK1A 0R6Canada
| | - Alvaro Yogi
- Human Health Therapeutics Research CenterNational Research Council Canada1200 Montreal Road, Building M54OttawaONK1A 0R6Canada
| | - Mehdi Arbabi Ghahroudi
- Human Health Therapeutics Research CentreNational Research Council Canada100 Sussex DriveOttawaONK1N 5A2Canada
| | - Hong Liu
- Human Health Therapeutics Research CenterNational Research Council Canada1200 Montreal Road, Building M54OttawaONK1A 0R6Canada
| | - Traian Sulea
- Human Health Therapeutics Research CentreNational Research Council Canada6100 Royalmount Avenue MontréalQuebecH4P 2R2Canada
| | - Yves Durocher
- Human Health Therapeutics Research CentreNational Research Council Canada6100 Royalmount Avenue MontréalQuebecH4P 2R2Canada
| | - Greg Hussack
- Human Health Therapeutics Research CentreNational Research Council Canada100 Sussex DriveOttawaONK1N 5A2Canada
| | - Henk van Faassen
- Human Health Therapeutics Research CentreNational Research Council Canada100 Sussex DriveOttawaONK1N 5A2Canada
| | - Balu Chakravarty
- Human Health Therapeutics Research CenterNational Research Council Canada1200 Montreal Road, Building M54OttawaONK1A 0R6Canada
| | - Qing Yan Liu
- Human Health Therapeutics Research CenterNational Research Council Canada1200 Montreal Road, Building M54OttawaONK1A 0R6Canada
| | - Umar Iqbal
- Human Health Therapeutics Research CenterNational Research Council Canada1200 Montreal Road, Building M54OttawaONK1A 0R6Canada
| | - Binbing Ling
- Human Health Therapeutics Research CenterNational Research Council Canada1200 Montreal Road, Building M54OttawaONK1A 0R6Canada
| | - Etienne Lessard
- Human Health Therapeutics Research CentreNational Research Council Canada6100 Royalmount Avenue MontréalQuebecH4P 2R2Canada
| | - Joey Sheff
- Human Health Therapeutics Research CentreNational Research Council Canada100 Sussex DriveOttawaONK1N 5A2Canada
| | - Anna Robotham
- Human Health Therapeutics Research CentreNational Research Council Canada100 Sussex DriveOttawaONK1N 5A2Canada
| | - Debbie Callaghan
- Human Health Therapeutics Research CentreNational Research Council Canada100 Sussex DriveOttawaONK1N 5A2Canada
| | - Maria Moreno
- Human Health Therapeutics Research CenterNational Research Council Canada1200 Montreal Road, Building M54OttawaONK1A 0R6Canada
| | - Tanya Comas
- Human Health Therapeutics Research CenterNational Research Council Canada1200 Montreal Road, Building M54OttawaONK1A 0R6Canada
| | - Dao Ly
- Human Health Therapeutics Research CenterNational Research Council Canada1200 Montreal Road, Building M54OttawaONK1A 0R6Canada
| | - Danica Stanimirovic
- Human Health Therapeutics Research CenterNational Research Council Canada1200 Montreal Road, Building M54OttawaONK1A 0R6Canada
| |
Collapse
|
2
|
Burgess SG, Paul NR, Richards MW, Ault JR, Askenatzis L, Claydon SG, Corbyn R, Machesky LM, Bayliss R. A nanobody inhibitor of Fascin-1 actin-bundling activity and filopodia formation. Open Biol 2024; 14:230376. [PMID: 38503329 PMCID: PMC10960945 DOI: 10.1098/rsob.230376] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2023] [Accepted: 01/16/2024] [Indexed: 03/21/2024] Open
Abstract
Fascin-1-mediated actin-bundling activity is central to the generation of plasma membrane protrusions required for cell migration. Dysregulated formation of cellular protrusions is observed in metastatic cancers, where they are required for increased invasiveness, and is often correlated with increased Fascin-1 abundance. Therefore, there is interest in generating therapeutic Fascin-1 inhibitors. We present the identification of Nb 3E11, a nanobody inhibitor of Fascin-1 actin-bundling activity and filopodia formation. The crystal structure of the Fascin-1/Nb 3E11 complex reveals the structural mechanism of inhibition. Nb 3E11 occludes an actin-binding site on the third β-trefoil domain of Fascin-1 that is currently not targeted by chemical inhibitors. Binding of Nb 3E11 to Fascin-1 induces a conformational change in the adjacent domains to stabilize Fascin-1 in an inhibitory state similar to that adopted in the presence of small-molecule inhibitors. Nb 3E11 could be used as a tool inhibitor molecule to aid in the development of Fascin-1 targeted therapeutics.
Collapse
Affiliation(s)
- Selena G. Burgess
- Astbury Centre for Structural Molecular Biology, School of Molecular and Cellular Biology, Faculty of Biological Sciences, University of Leeds, Leeds LS2 9JT, UK
| | - Nikki R. Paul
- Cancer Research UK Beatson Institute, Garscube Estate, Switchback Road, Bearsden, Glasgow G61 1BD, UK
| | - Mark W. Richards
- Astbury Centre for Structural Molecular Biology, School of Molecular and Cellular Biology, Faculty of Biological Sciences, University of Leeds, Leeds LS2 9JT, UK
| | - James R. Ault
- Astbury Centre for Structural Molecular Biology, School of Molecular and Cellular Biology, Faculty of Biological Sciences, University of Leeds, Leeds LS2 9JT, UK
| | - Laurie Askenatzis
- Cancer Research UK Beatson Institute, Garscube Estate, Switchback Road, Bearsden, Glasgow G61 1BD, UK
| | - Sophie G. Claydon
- Cancer Research UK Beatson Institute, Garscube Estate, Switchback Road, Bearsden, Glasgow G61 1BD, UK
- Institute of Cancer Sciences, University of Glasgow, Glasgow G61 1QH, UK
| | - Ryan Corbyn
- Cancer Research UK Beatson Institute, Garscube Estate, Switchback Road, Bearsden, Glasgow G61 1BD, UK
| | - Laura M. Machesky
- Cancer Research UK Beatson Institute, Garscube Estate, Switchback Road, Bearsden, Glasgow G61 1BD, UK
- Institute of Cancer Sciences, University of Glasgow, Glasgow G61 1QH, UK
- Department of Biochemistry, University of Cambridge, Cambridge CB2 1GA, UK
| | - Richard Bayliss
- Astbury Centre for Structural Molecular Biology, School of Molecular and Cellular Biology, Faculty of Biological Sciences, University of Leeds, Leeds LS2 9JT, UK
| |
Collapse
|
3
|
Rebnegger C, Coltman BL, Kowarz V, Peña DA, Mentler A, Troyer C, Hann S, Schöny H, Koellensperger G, Mattanovich D, Gasser B. Protein production dynamics and physiological adaptation of recombinant Komagataella phaffii at near-zero growth rates. Microb Cell Fact 2024; 23:43. [PMID: 38331812 PMCID: PMC10851509 DOI: 10.1186/s12934-024-02314-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2023] [Accepted: 01/23/2024] [Indexed: 02/10/2024] Open
Abstract
BACKGROUND Specific productivity (qP) in yeast correlates with growth, typically peaking at intermediate or maximum specific growth rates (μ). Understanding the factors limiting productivity at extremely low μ might reveal decoupling strategies, but knowledge of production dynamics and physiology in such conditions is scarce. Retentostats, a type of continuous cultivation, enable the well-controlled transition to near-zero µ through the combined retention of biomass and limited substrate supply. Recombinant Komagataella phaffii (syn Pichia pastoris) secreting a bivalent single domain antibody (VHH) was cultivated in aerobic, glucose-limited retentostats to investigate recombinant protein production dynamics and broaden our understanding of relevant physiological adaptations at near-zero growth conditions. RESULTS By the end of the retentostat cultivation, doubling times of approx. two months were reached, corresponding to µ = 0.00047 h-1. Despite these extremely slow growth rates, the proportion of viable cells remained high, and de novo synthesis and secretion of the VHH were observed. The average qP at the end of the retentostat was estimated at 0.019 mg g-1 h-1. Transcriptomics indicated that genes involved in protein biosynthesis were only moderately downregulated towards zero growth, while secretory pathway genes were mostly regulated in a manner seemingly detrimental to protein secretion. Adaptation to near-zero growth conditions of recombinant K. phaffii resulted in significant changes in the total protein, RNA, DNA and lipid content, and lipidomics revealed a complex adaptation pattern regarding the lipid class composition. The higher abundance of storage lipids as well as storage carbohydrates indicates that the cells are preparing for long-term survival. CONCLUSIONS In conclusion, retentostat cultivation proved to be a valuable tool to identify potential engineering targets to decouple growth and protein production and gain important insights into the physiological adaptation of K. phaffii to near-zero growth conditions.
Collapse
Affiliation(s)
- Corinna Rebnegger
- CD-Laboratory for Growth-Decoupled Protein Production in Yeast at Department of Biotechnology, University of Natural Resources and Life Sciences (BOKU), Vienna, Austria
- Department of Biotechnology, Institute of Microbiology and Microbial Biotechnology (IMMB), University of Natural Resources and Life Sciences, Vienna, Muthgasse 18, 1190, Vienna, Austria
- Austrian Centre of Industrial Biotechnology (ACIB GmbH), Muthgasse 11, 1190, Vienna, Austria
| | - Benjamin L Coltman
- CD-Laboratory for Growth-Decoupled Protein Production in Yeast at Department of Biotechnology, University of Natural Resources and Life Sciences (BOKU), Vienna, Austria
- Department of Biotechnology, Institute of Microbiology and Microbial Biotechnology (IMMB), University of Natural Resources and Life Sciences, Vienna, Muthgasse 18, 1190, Vienna, Austria
| | - Viktoria Kowarz
- CD-Laboratory for Growth-Decoupled Protein Production in Yeast at Department of Biotechnology, University of Natural Resources and Life Sciences (BOKU), Vienna, Austria
- Department of Biotechnology, Institute of Microbiology and Microbial Biotechnology (IMMB), University of Natural Resources and Life Sciences, Vienna, Muthgasse 18, 1190, Vienna, Austria
| | - David A Peña
- Department of Biotechnology, Institute of Microbiology and Microbial Biotechnology (IMMB), University of Natural Resources and Life Sciences, Vienna, Muthgasse 18, 1190, Vienna, Austria
| | - Axel Mentler
- Department of Forest- and Soil Sciences, Institute of Soil Research, University of Natural Resources and Life Sciences, Vienna, Peter-Jordan-Straße 82, 1190, Vienna, Austria
| | - Christina Troyer
- Department of Chemistry, Institute of Analytical Chemistry, University of Natural Resources and Life Sciences, Vienna, Muthgasse 18, 1190, Vienna, Austria
| | - Stephan Hann
- Department of Chemistry, Institute of Analytical Chemistry, University of Natural Resources and Life Sciences, Vienna, Muthgasse 18, 1190, Vienna, Austria
| | - Harald Schöny
- Department of Analytical Chemistry, Faculty of Chemistry, University of Vienna, Waehringer Straße 38, 1090, Vienna, Austria
| | - Gunda Koellensperger
- Department of Analytical Chemistry, Faculty of Chemistry, University of Vienna, Waehringer Straße 38, 1090, Vienna, Austria
- Vienna Metabolomics Center (VIME), University of Vienna, Althanstraße 14, 1090, Vienna, Austria
| | - Diethard Mattanovich
- CD-Laboratory for Growth-Decoupled Protein Production in Yeast at Department of Biotechnology, University of Natural Resources and Life Sciences (BOKU), Vienna, Austria
- Department of Biotechnology, Institute of Microbiology and Microbial Biotechnology (IMMB), University of Natural Resources and Life Sciences, Vienna, Muthgasse 18, 1190, Vienna, Austria
- Austrian Centre of Industrial Biotechnology (ACIB GmbH), Muthgasse 11, 1190, Vienna, Austria
| | - Brigitte Gasser
- CD-Laboratory for Growth-Decoupled Protein Production in Yeast at Department of Biotechnology, University of Natural Resources and Life Sciences (BOKU), Vienna, Austria.
- Department of Biotechnology, Institute of Microbiology and Microbial Biotechnology (IMMB), University of Natural Resources and Life Sciences, Vienna, Muthgasse 18, 1190, Vienna, Austria.
- Austrian Centre of Industrial Biotechnology (ACIB GmbH), Muthgasse 11, 1190, Vienna, Austria.
| |
Collapse
|
4
|
Joest EF, Tampé R. Design principles for engineering light-controlled antibodies. Trends Biotechnol 2023; 41:1501-1517. [PMID: 37507295 DOI: 10.1016/j.tibtech.2023.06.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Revised: 06/13/2023] [Accepted: 06/14/2023] [Indexed: 07/30/2023]
Abstract
Engineered antibodies are essential tools for research and advanced pharmacy. In the development of therapeutics, antibodies are excellent candidates as they offer both target recognition and modulation. Thanks to the latest advances in biotechnology, light-activated antibody fragments can be constructed to control spontaneous antigen interaction with high spatiotemporal precision. To implement conditional antigen binding, several optogenetic and optochemical engineering concepts have recently been developed. Here, we highlight the various strategies and discuss the features of opto-conditional antibodies. Each concept offers intrinsic advantages beneficial to different applications. In summary, the novel design approaches constitute a complementary toolset to promote current and upcoming antibody technologies with ultimate precision.
Collapse
Affiliation(s)
- Eike F Joest
- Institute of Biochemistry, Biocenter, Goethe University Frankfurt, Max-von-Laue-Str. 9, 60438 Frankfurt a.M., Germany.
| | - Robert Tampé
- Institute of Biochemistry, Biocenter, Goethe University Frankfurt, Max-von-Laue-Str. 9, 60438 Frankfurt a.M., Germany.
| |
Collapse
|
5
|
Zahrl RJ, Prielhofer R, Burgard J, Mattanovich D, Gasser B. Synthetic activation of yeast stress response improves secretion of recombinant proteins. N Biotechnol 2023; 73:19-28. [PMID: 36603701 DOI: 10.1016/j.nbt.2023.01.001] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2022] [Revised: 12/22/2022] [Accepted: 01/01/2023] [Indexed: 01/03/2023]
Abstract
Yeasts, such as Pichia pastoris (syn Komagataella spp.), are particularly suitable expression systems for emerging classes of recombinant proteins. Among them, recombinant antibody fragments, such as single-chain variable fragments (scFv) and single-domain antibodies (VHH), are credible alternatives to monoclonal antibodies. The availability of powerful genetic engineering and synthetic biology tools has facilitated improvement of this cell factory to overcome certain limitations. However, cell engineering to improve secretion often remains a trial-and-error approach and improvements are often specific to the protein produced. Where multiple genetic interventions are needed to remove bottlenecks in the process of recombinant protein secretion, this leads to a high number of combinatorial possibilities for creation of new production strains. Therefore, our aim was to exploit whole transcriptional programs (stress response pathways) in order to simplify the strain engineering of new production strains. Indeed, the artificial activation of the general stress response transcription factor Msn4, as well as synthetic versions thereof, could replace the secretion enhancing effect of several cytosolic chaperones. Greater than 4-fold improvements in recombinant protein secretion were achieved by overexpression of MSN4 or synMSN4, either alone or in combination with Hac1 or ER chaperones. With this concept we were able to successfully engineer strains reaching titers of more than 2.5 g/L scFv and 8 g/L VHH in bioreactor cultivations. This increased secretion capacity of different industrially relevant model proteins indicates that MSN4 overexpression most likely represents a general concept to improve recombinant protein production in yeast.
Collapse
Affiliation(s)
- Richard J Zahrl
- ACIB GmbH, Muthgasse 11, 1190 Vienna, Austria; Department of Biotechnology, Institute of Microbiology and Microbial Biotechnology, University of Natural Resources and Life Sciences (BOKU), Muthgasse 18, 1190 Vienna, Austria
| | - Roland Prielhofer
- ACIB GmbH, Muthgasse 11, 1190 Vienna, Austria; Department of Biotechnology, Institute of Microbiology and Microbial Biotechnology, University of Natural Resources and Life Sciences (BOKU), Muthgasse 18, 1190 Vienna, Austria
| | - Jonas Burgard
- ACIB GmbH, Muthgasse 11, 1190 Vienna, Austria; Department of Biotechnology, Institute of Microbiology and Microbial Biotechnology, University of Natural Resources and Life Sciences (BOKU), Muthgasse 18, 1190 Vienna, Austria
| | - Diethard Mattanovich
- ACIB GmbH, Muthgasse 11, 1190 Vienna, Austria; Department of Biotechnology, Institute of Microbiology and Microbial Biotechnology, University of Natural Resources and Life Sciences (BOKU), Muthgasse 18, 1190 Vienna, Austria
| | - Brigitte Gasser
- ACIB GmbH, Muthgasse 11, 1190 Vienna, Austria; Department of Biotechnology, Institute of Microbiology and Microbial Biotechnology, University of Natural Resources and Life Sciences (BOKU), Muthgasse 18, 1190 Vienna, Austria.
| |
Collapse
|
6
|
O'Shea JM, Goutou A, Brydon J, Sethna CR, Wood CW, Greiss S. Generation of Photocaged Nanobodies for Intracellular Applications in an Animal Using Genetic Code Expansion and Computationally Guided Protein Engineering. Chembiochem 2022; 23:e202200321. [PMID: 35731601 PMCID: PMC9542635 DOI: 10.1002/cbic.202200321] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Indexed: 11/12/2022]
Abstract
Nanobodies are becoming increasingly popular as tools for manipulating and visualising proteins in vivo. The ability to control nanobody/antigen interactions using light could provide precise spatiotemporal control over protein function. We develop a general approach to engineer photo-activatable nanobodies using photocaged amino acids that are introduced into the target binding interface by genetic code expansion. Guided by computational alanine scanning and molecular dynamics simulations, we tune nanobody/target binding affinity to eliminate binding before uncaging. Upon photo-activation using 365 nm light, binding is restored. We use this approach to generate improved photocaged variants of two anti-GFP nanobodies that function robustly when directly expressed in a complex intracellular environment together with their antigen. We apply them to control subcellular protein localisation in the nematode worm Caenorhabditis elegans. Our approach applies predictions derived from computational modelling directly in a living animal and demonstrates the importance of accounting for in vivo effects on protein-protein interactions.
Collapse
Affiliation(s)
- Jack M O'Shea
- Centre for Discovery Brain Sciences, University of Edinburgh, Hugh Robson Building George Square, Edinburgh, EH8 9XD, UK
| | - Angeliki Goutou
- Centre for Discovery Brain Sciences, University of Edinburgh, Hugh Robson Building George Square, Edinburgh, EH8 9XD, UK
| | - Jack Brydon
- MRC Institute of Genetics & Molecular Medicine, University of Edinburgh, Western General Hospital Crewe Road South, Edinburgh, EH4 2XR, UK
| | - Cyrus R Sethna
- Centre for Discovery Brain Sciences, University of Edinburgh, Hugh Robson Building George Square, Edinburgh, EH8 9XD, UK
| | - Christopher W Wood
- Institute of Quantitative Biology, Biochemistry and Biotechnology, University of Edinburgh, Roger Land Building King's Buildings, Edinburgh, EH9 3JQ, UK
| | - Sebastian Greiss
- Centre for Discovery Brain Sciences, University of Edinburgh, Hugh Robson Building George Square, Edinburgh, EH8 9XD, UK
| |
Collapse
|
7
|
Yu D, Won Do H. Optogenetic Activation of Intracellular Nanobodies. Methods Mol Biol 2022; 2446:595-606. [PMID: 35157296 DOI: 10.1007/978-1-0716-2075-5_31] [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: 06/14/2023]
Abstract
Intracellular antibody fragments such as nanobodies and scFvs are powerful tools for imaging and for modulating and neutralizing endogenous target proteins. Optogenetically activated intracellular antibodies (optobodies) constitute a light-inducible system to directly control intrabody activities in cells, with greater spatial and temporal resolution than intracellular antibodies alone. Here, we describe optogenetic and microscopic methods to activate optobodies in cells using a confocal microscope and an automated fluorescence microscope. In the protocol, we use the examples of an optobody targeting green fluorescent protein and an optobody that inhibits the endogenous gelsolin protein.
Collapse
Affiliation(s)
- Daseuli Yu
- Life Science Research Institute, Korea Advanced Institute of Science and Technology, Daejeon, Republic of Korea
| | - Heo Won Do
- Life Science Research Institute, Korea Advanced Institute of Science and Technology, Daejeon, Republic of Korea.
- KAIST Institute for the BioCentury, Korea Advanced Institute of Science and Technology, Daejeon, Republic of Korea.
| |
Collapse
|
8
|
Wagner TR, Rothbauer U. Nanobodies Right in the Middle: Intrabodies as Toolbox to Visualize and Modulate Antigens in the Living Cell. Biomolecules 2020; 10:biom10121701. [PMID: 33371447 PMCID: PMC7767433 DOI: 10.3390/biom10121701] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Revised: 12/15/2020] [Accepted: 12/18/2020] [Indexed: 01/01/2023] Open
Abstract
In biomedical research, there is an ongoing demand for new technologies to elucidate disease mechanisms and develop novel therapeutics. This requires comprehensive understanding of cellular processes and their pathophysiology based on reliable information on abundance, localization, post-translational modifications and dynamic interactions of cellular components. Traceable intracellular binding molecules provide new opportunities for real-time cellular diagnostics. Most prominently, intrabodies derived from antibody fragments of heavy-chain only antibodies of camelids (nanobodies) have emerged as highly versatile and attractive probes to study and manipulate antigens within the context of living cells. In this review, we provide an overview on the selection, delivery and usage of intrabodies to visualize and monitor cellular antigens in living cells and organisms. Additionally, we summarize recent advances in the development of intrabodies as cellular biosensors and their application to manipulate disease-related cellular processes. Finally, we highlight switchable intrabodies, which open entirely new possibilities for real-time cell-based diagnostics including live-cell imaging, target validation and generation of precisely controllable binding reagents for future therapeutic applications.
Collapse
Affiliation(s)
- Teresa R. Wagner
- Pharmaceutical Biotechnology, Eberhard Karls University Tuebingen, 72076 Tuebingen, Germany;
- Natural and Medical Sciences Institute, University of Tuebingen, 72770 Reutlingen, Germany
| | - Ulrich Rothbauer
- Pharmaceutical Biotechnology, Eberhard Karls University Tuebingen, 72076 Tuebingen, Germany;
- Natural and Medical Sciences Institute, University of Tuebingen, 72770 Reutlingen, Germany
- Correspondence: ; Tel.: +49-7121-5153-0415; Fax: +49-7121-5153-0816
| |
Collapse
|
9
|
Merckaert T, Zwaenepoel O, Gevaert K, Gettemans J. An AKT2-specific nanobody that targets the hydrophobic motif induces cell cycle arrest, autophagy and loss of focal adhesions in MDA-MB-231 cells. Biomed Pharmacother 2020; 133:111055. [PMID: 33378961 DOI: 10.1016/j.biopha.2020.111055] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2020] [Revised: 10/21/2020] [Accepted: 11/19/2020] [Indexed: 02/06/2023] Open
Abstract
The AKT kinase family is a high-profile target for cancer therapy. Despite their high degree of homology the three AKT isoforms (AKT1, AKT2 and AKT3) are non-redundant and can even have opposing functions. Small-molecule AKT inhibitors affect all three isoforms which severely limits their usefulness as research tool or therapeutic. Using AKT2-specific nanobodies we examined the function of endogenous AKT2 in breast cancer cells. Two AKT2 nanobodies (Nb8 and Nb9) modulate AKT2 and reduce MDA-MB-231 cell viability/proliferation. Nb8 binds the AKT2 hydrophobic motif and reduces IGF-1-induced phosphorylation of this site. This nanobody also affects the phosphorylation and/or expression levels of a wide range of proteins downstream of AKT, resulting in a G0/G1 cell cycle arrest, the induction of autophagy, a reduction in focal adhesion count and loss of stress fibers. While cell cycle progression is likely to be regulated by more than one isoform, our results indicate that both the effects on autophagy and the cytoskeleton are specific to AKT2. By using an isoform-specific nanobody we were able to map a part of the AKT2 pathway. Our results confirm AKT2 and the hydrophobic motif as targets for cancer therapy. Nb8 can be used as a research tool to study AKT2 signalling events and aid in the design of an AKT2-specific inhibitor.
Collapse
Affiliation(s)
- Tijs Merckaert
- Department of Biomolecular Medicine, Faculty of Medicine and Health Sciences, Ghent University, Tech Lane Ghent Science Park 75, 9052 Ghent, Belgium; VIB-UGent Center for Medical Biotechnology, Tech Lane Ghent Science Park 75, 9052 Ghent, Belgium.
| | - Olivier Zwaenepoel
- Department of Biomolecular Medicine, Faculty of Medicine and Health Sciences, Ghent University, Tech Lane Ghent Science Park 75, 9052 Ghent, Belgium.
| | - Kris Gevaert
- Department of Biomolecular Medicine, Faculty of Medicine and Health Sciences, Ghent University, Tech Lane Ghent Science Park 75, 9052 Ghent, Belgium; VIB-UGent Center for Medical Biotechnology, Tech Lane Ghent Science Park 75, 9052 Ghent, Belgium.
| | - Jan Gettemans
- Department of Biomolecular Medicine, Faculty of Medicine and Health Sciences, Ghent University, Tech Lane Ghent Science Park 75, 9052 Ghent, Belgium.
| |
Collapse
|
10
|
Gettemans J, De Dobbelaer B. Transforming nanobodies into high-precision tools for protein function analysis. Am J Physiol Cell Physiol 2020; 320:C195-C215. [PMID: 33264078 DOI: 10.1152/ajpcell.00435.2020] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Single-domain antibodies, derived from camelid heavy antibodies (nanobodies) or shark variable new antigen receptors, have attracted increasing attention in recent years due to their extremely versatile nature and the opportunities they offer for downstream modification. Discovered more than three decades ago, these 120-amino acid (∼15-kDa) antibody fragments are known to bind their target with high specificity and affinity. Key features of nanobodies that make them very attractive include their single-domain nature, small size, and affordable high-level expression in prokaryotes, and their cDNAs are routinely obtained in the process of their isolation. This facilitates and stimulates new experimental approaches. Hence, it allows researchers to formulate new answers to complex biomedical questions. Through elementary PCR-based technologies and chemical modification strategies, their primary structure can be altered almost at leisure while retaining their specificity and biological activity, transforming them into highly tailored tools that meet the increasing demands of current-day biomedical research. In this review, various aspects of camelid nanobodies are expounded, including intracellular delivery in recombinant format for manipulation of, i.e., cytoplasmic targets, their derivatization to improve nanobody orientation as a capturing device, approaches to reversibly bind their target, their potential as protein-silencing devices in cells, the development of strategies to transfer nanobodies through the blood-brain barrier and their application in CAR-T experimentation. We also discuss some of their disadvantages and conclude with future prospects.
Collapse
Affiliation(s)
- Jan Gettemans
- Department of Biomolecular Medicine, Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium
| | - Brian De Dobbelaer
- Department of Biomolecular Medicine, Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium
| |
Collapse
|
11
|
de Beer MA, Giepmans BNG. Nanobody-Based Probes for Subcellular Protein Identification and Visualization. Front Cell Neurosci 2020; 14:573278. [PMID: 33240044 PMCID: PMC7667270 DOI: 10.3389/fncel.2020.573278] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2020] [Accepted: 10/05/2020] [Indexed: 12/14/2022] Open
Abstract
Understanding how building blocks of life contribute to physiology is greatly aided by protein identification and cellular localization. The two main labeling approaches developed over the past decades are labeling with antibodies such as immunoglobulin G (IgGs) or use of genetically encoded tags such as fluorescent proteins. However, IgGs are large proteins (150 kDa), which limits penetration depth and uncertainty of target position caused by up to ∼25 nm distance of the label created by the chosen targeting approach. Additionally, IgGs cannot be easily recombinantly modulated and engineered as part of fusion proteins because they consist of multiple independent translated chains. In the last decade single domain antigen binding proteins are being explored in bioscience as a tool in revealing molecular identity and localization to overcome limitations by IgGs. These nanobodies have several potential benefits over routine applications. Because of their small size (15 kDa), nanobodies better penetrate during labeling procedures and improve resolution. Moreover, nanobodies cDNA can easily be fused with other cDNA. Multidomain proteins can thus be easily engineered consisting of domains for targeting (nanobodies) and visualization by fluorescence microscopy (fluorescent proteins) or electron microscopy (based on certain enzymes). Additional modules for e.g., purification are also easily added. These nanobody-based probes can be applied in cells for live-cell endogenous protein detection or may be purified prior to use on molecules, cells or tissues. Here, we present the current state of nanobody-based probes and their implementation in microscopy, including pitfalls and potential future opportunities.
Collapse
Affiliation(s)
- Marit A de Beer
- Department of Biomedical Sciences of Cells and Systems, University of Groningen, University Medical Center Groningen, Groningen, Netherlands
| | - Ben N G Giepmans
- Department of Biomedical Sciences of Cells and Systems, University of Groningen, University Medical Center Groningen, Groningen, Netherlands
| |
Collapse
|
12
|
Radoman B, Grünwald-Gruber C, Schmelzer B, Zavec D, Gasser B, Altmann F, Mattanovich D. The Degree and Length of O-Glycosylation of Recombinant Proteins Produced in Pichia pastoris Depends on the Nature of the Protein and the Process Type. Biotechnol J 2020; 16:e2000266. [PMID: 32975831 DOI: 10.1002/biot.202000266] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2020] [Revised: 09/09/2020] [Indexed: 12/16/2022]
Abstract
The methylotrophic yeast Pichia pastoris is known as an efficient host for the production of heterologous proteins. While N-linked protein glycosylation is well characterized in P. pastoris there is less knowledge of the patterns of O-glycosylation. O-glycans produced by P. pastoris consist of short linear mannose chains, which in the case of recombinant biopharmaceuticals can trigger an immune response in humans. This study aims to reveal the influence of different cultivation strategies on O-mannosylation profiles in P. pastoris. Sixteen different model proteins, produced by different P. pastoris strains, are analyzed for their O-glycosylation profile. Based on the obtained data, human serum albumin (HSA) is chosen to be produced in fast and slow growth fed batch fermentations by using common promoters, PGAP and PAOX1 . After purification and protein digestion, glycopeptides are analyzed by LC/ESI-MS. In the samples expressed with PGAP it is found that the degree of glycosylation is slightly higher when a slow growth rate is used, regardless of the efficiency of the producing strain. The highest glycosylation intensity is observed in HSA produced with PAOX1 . The results indicate that the O-glycosylation level is markedly higher when the protein is produced in a methanol-based expression system.
Collapse
Affiliation(s)
- Bojana Radoman
- Austrian Centre of Industrial Biotechnology (ACIB), Vienna, 1190, Austria.,Department of Biotechnology, BOKU-University of Natural Resources and Life Sciences, Vienna, 1190, Austria
| | - Clemens Grünwald-Gruber
- Department of Chemistry, BOKU-University of Natural Resources and Life Sciences, Vienna, 1190, Austria
| | - Bernhard Schmelzer
- Austrian Centre of Industrial Biotechnology (ACIB), Vienna, 1190, Austria.,Department of Biotechnology, BOKU-University of Natural Resources and Life Sciences, Vienna, 1190, Austria
| | - Domen Zavec
- Department of Biotechnology, BOKU-University of Natural Resources and Life Sciences, Vienna, 1190, Austria
| | - Brigitte Gasser
- Austrian Centre of Industrial Biotechnology (ACIB), Vienna, 1190, Austria.,Department of Biotechnology, BOKU-University of Natural Resources and Life Sciences, Vienna, 1190, Austria
| | - Friedrich Altmann
- Austrian Centre of Industrial Biotechnology (ACIB), Vienna, 1190, Austria.,Department of Chemistry, BOKU-University of Natural Resources and Life Sciences, Vienna, 1190, Austria
| | - Diethard Mattanovich
- Austrian Centre of Industrial Biotechnology (ACIB), Vienna, 1190, Austria.,Department of Biotechnology, BOKU-University of Natural Resources and Life Sciences, Vienna, 1190, Austria
| |
Collapse
|
13
|
Uchański T, Pardon E, Steyaert J. Nanobodies to study protein conformational states. Curr Opin Struct Biol 2020; 60:117-123. [DOI: 10.1016/j.sbi.2020.01.003] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2019] [Revised: 01/08/2020] [Accepted: 01/09/2020] [Indexed: 01/07/2023]
|
14
|
Liu J, Hebbrecht T, Brans T, Parthoens E, Lippens S, Li C, De Keersmaecker H, De Vos WH, De Smedt SC, Boukherroub R, Gettemans J, Xiong R, Braeckmans K. Long-term live-cell microscopy with labeled nanobodies delivered by laser-induced photoporation. NANO RESEARCH 2020; 13:485-495. [PMID: 33154805 PMCID: PMC7116313 DOI: 10.1007/s12274-020-2633-z] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/05/2023]
Abstract
Fluorescence microscopy is the method of choice for studying intracellular dynamics. However, its success depends on the availability of specific and stable markers. A prominent example of markers that are rapidly gaining interest are nanobodies (Nbs, ~ 15 kDa), which can be functionalized with bright and photostable organic fluorophores. Due to their relatively small size and high specificity, Nbs offer great potential for high-quality long-term subcellular imaging, but suffer from the fact that they cannot spontaneously cross the plasma membrane of live cells. We have recently discovered that laser-induced photoporation is well suited to deliver extrinsic labels to living cells without compromising their viability. Being a laser-based technology, it is readily compatible with light microscopy and the typical cell recipients used for that. Spurred by these promising initial results, we demonstrate here for the first time successful long-term imaging of specific subcellular structures with labeled nanobodies in living cells. We illustrate this using Nbs that target GFP/YFP-protein constructs accessible in the cytoplasm, actin-bundling protein Fascin, and the histone H2A/H2B heterodimers. With an efficiency of more than 80% labeled cells and minimal toxicity (~ 2%), photoporation proved to be an excellent intracellular delivery method for Nbs. Time-lapse microscopy revealed that cell division rate and migration remained unaffected, confirming excellent cell viability and functionality. We conclude that laser-induced photoporation labeled Nbs can be easily delivered into living cells, laying the foundation for further development of a broad range of Nbs with intracellular targets as a toolbox for long-term live-cell microscopy.
Collapse
Affiliation(s)
- Jing Liu
- Laboratory of General Biochemistry and Physical Pharmacy, Faculty of Pharmacy, Ghent University, Ghent B-9000, Belgium
| | - Tim Hebbrecht
- Department of Biomolecular medicine, Faculty of Medicine and Health Sciences, Ghent University, Ghent B-9000, Belgium
| | - Toon Brans
- Laboratory of General Biochemistry and Physical Pharmacy, Faculty of Pharmacy, Ghent University, Ghent B-9000, Belgium
| | - Eef Parthoens
- VIB-UGent Center for Inflammation Research, VIB, Ghent B-9000, Belgium
- VIB Bioimaging Core Ghent, VIB, Ghent B-9000, Belgium
- Department of Biomedical Molecular Biology, Ghent University, Ghent B-9000, Belgium
| | - Saskia Lippens
- VIB-UGent Center for Inflammation Research, VIB, Ghent B-9000, Belgium
- VIB Bioimaging Core Ghent, VIB, Ghent B-9000, Belgium
- Department of Biomedical Molecular Biology, Ghent University, Ghent B-9000, Belgium
| | - Chengnan Li
- Univ. Lille, CNRS, Centrale Lille, ISEN, Univ. Valenciennes, UMR 8520-IEMN, Lille F-59000, France
| | - Herlinde De Keersmaecker
- Laboratory of General Biochemistry and Physical Pharmacy, Faculty of Pharmacy, Ghent University, Ghent B-9000, Belgium
- Centre for Advanced Light Microscopy, Ghent University, Ghent B-9000, Belgium
| | - Winnok H De Vos
- Laboratory of Cell Biology and Histology, Department of Veterinary Sciences, University of Antwerp, 2020 Antwerp, Belgium
| | - Stefaan C De Smedt
- Laboratory of General Biochemistry and Physical Pharmacy, Faculty of Pharmacy, Ghent University, Ghent B-9000, Belgium
- Centre for Advanced Light Microscopy, Ghent University, Ghent B-9000, Belgium
| | - Rabah Boukherroub
- Univ. Lille, CNRS, Centrale Lille, ISEN, Univ. Valenciennes, UMR 8520-IEMN, Lille F-59000, France
| | - Jan Gettemans
- Department of Biomolecular medicine, Faculty of Medicine and Health Sciences, Ghent University, Ghent B-9000, Belgium
| | - Ranhua Xiong
- Laboratory of General Biochemistry and Physical Pharmacy, Faculty of Pharmacy, Ghent University, Ghent B-9000, Belgium
| | - Kevin Braeckmans
- Laboratory of General Biochemistry and Physical Pharmacy, Faculty of Pharmacy, Ghent University, Ghent B-9000, Belgium
- Centre for Advanced Light Microscopy, Ghent University, Ghent B-9000, Belgium
| |
Collapse
|
15
|
Lee HC, Zhao YJ. Resolving the topological enigma in Ca 2+ signaling by cyclic ADP-ribose and NAADP. J Biol Chem 2019; 294:19831-19843. [PMID: 31672920 PMCID: PMC6937575 DOI: 10.1074/jbc.rev119.009635] [Citation(s) in RCA: 53] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Cyclic ADP-ribose (cADPR) and nicotinic acid adenine dinucleotide phosphate (NAADP) are two structurally distinct messengers that mobilize the endoplasmic and endolysosomal Ca2+ stores, respectively. Both are synthesized by the CD38 molecule (CD38), which has long been thought to be a type II membrane protein whose catalytic domain, intriguingly, faces to the outside of the cell. Accordingly, for more than 20 years, it has remained unresolved how CD38 can use cytosolic substrates such as NAD and NADP to produce messengers that target intracellular Ca2+ stores. The discovery of type III CD38, whose catalytic domain faces the cytosol, has now begun to clarify this topological conundrum. This article reviews the ideas and clues leading to the discovery of the type III CD38; highlights an innovative approach for uncovering its natural existence; and discusses the regulators of its activity, folding, and degradation. We also review the compartmentalization of cADPR and NAADP biogenesis. We further discuss the possible mechanisms that promote type III CD38 expression and appraise a proposal of a Ca2+-signaling mechanism based on substrate limitation and product translocation. The surprising finding of another enzyme that produces cADPR and NAADP, sterile α and TIR motif-containing 1 (SARM1), is described. SARM1 regulates axonal degeneration and has no sequence similarity with CD38 but can catalyze the same set of multireactions and has the same cytosolic orientation as the type III CD38. The intriguing finding that SARM1 is activated by nicotinamide mononucleotide to produce cADPR and NAADP suggests that it may function as a regulated Ca2+-signaling enzyme like CD38.
Collapse
Affiliation(s)
- Hon Cheung Lee
- State Key Laboratory of Chemical Oncogenomics, Key Laboratory of Chemical Genomics, Peking University Shenzhen Graduate School, Shenzhen, China, 518055
| | - Yong Juan Zhao
- State Key Laboratory of Chemical Oncogenomics, Key Laboratory of Chemical Genomics, Peking University Shenzhen Graduate School, Shenzhen, China, 518055
| |
Collapse
|
16
|
Optogenetic activation of intracellular antibodies for direct modulation of endogenous proteins. Nat Methods 2019; 16:1095-1100. [PMID: 31611691 DOI: 10.1038/s41592-019-0592-7] [Citation(s) in RCA: 87] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2018] [Accepted: 09/06/2019] [Indexed: 01/13/2023]
Abstract
Intracellular antibodies have become powerful tools for imaging, modulating and neutralizing endogenous target proteins. Here, we describe an optogenetically activated intracellular antibody (optobody) consisting of split antibody fragments and blue-light inducible heterodimerization domains. We expanded this optobody platform by generating several optobodies from previously developed intracellular antibodies, and demonstrated that photoactivation of gelsolin and β2-adrenergic receptor (β2AR) optobodies suppressed endogenous gelsolin activity and β2AR signaling, respectively.
Collapse
|
17
|
Steels A, Vannevel L, Zwaenepoel O, Gettemans J. Nb-induced stabilisation of p53 in HPV-infected cells. Sci Rep 2019; 9:12680. [PMID: 31481667 PMCID: PMC6722090 DOI: 10.1038/s41598-019-49061-9] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2019] [Accepted: 08/13/2019] [Indexed: 11/23/2022] Open
Abstract
Cervical cancer is caused by a persistent infection of the mucosal epithelia with high-risk human papilloma viruses (HPVs). The viral oncoprotein E6 is responsible for the inactivation of the tumour suppressor p53 and thus plays a crucial role in HPV-induced tumorigenesis. The viral E6 protein forms a trimeric complex with the endogenous E3 ubiquitine ligase E6AP and the DNA-binding domain (DBD) of p53, which results in the polyubiquitination and proteasomal degradation of p53. We have developed nanobodies (Nbs) against the DBD of p53, which substantially stabilise p53 in HeLa cells. The observed effect is specific for HPV-infected cells, since similar effects were not seen for U2OS cells. Despite the fact that the stabilised p53 was strongly nuclear enriched, its tumour suppressive functions were hampered. We argue that the absence of a tumour suppressive effect is caused by inhibition of p53 transactivation in both HPV-infected and HPV-negative cells. The inactivation of the transcriptional activity of p53 was associated with an increased cellular proliferation and viability of HeLa cells. In conclusion, we demonstrate that p53 DBD Nbs positively affect protein stability whilst adversely affecting protein function, attesting to their ability to modulate protein properties in a very subtle manner.
Collapse
Affiliation(s)
- Anneleen Steels
- Department of Biomolecular Medicine, Faculty of Medicine and Health Sciences, Campus Rommelaere, A. Baertsoenkaai 3, Ghent University, Ghent, Belgium
| | - Laura Vannevel
- Department of Biomolecular Medicine, Faculty of Medicine and Health Sciences, Campus Rommelaere, A. Baertsoenkaai 3, Ghent University, Ghent, Belgium
| | - Olivier Zwaenepoel
- Department of Biomolecular Medicine, Faculty of Medicine and Health Sciences, Campus Rommelaere, A. Baertsoenkaai 3, Ghent University, Ghent, Belgium
| | - Jan Gettemans
- Department of Biomolecular Medicine, Faculty of Medicine and Health Sciences, Campus Rommelaere, A. Baertsoenkaai 3, Ghent University, Ghent, Belgium.
| |
Collapse
|
18
|
Homology Modeling-Based in Silico Affinity Maturation Improves the Affinity of a Nanobody. Int J Mol Sci 2019; 20:ijms20174187. [PMID: 31461846 PMCID: PMC6747709 DOI: 10.3390/ijms20174187] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2019] [Revised: 08/21/2019] [Accepted: 08/22/2019] [Indexed: 01/08/2023] Open
Abstract
Affinity maturation and rational design have a raised importance in the application of nanobody (VHH), and its unique structure guaranteed these processes quickly done in vitro. An anti-CD47 nanobody, Nb02, was screened via a synthetic phage display library with 278 nM of KD value. In this study, a new strategy based on homology modeling and Rational Mutation Hotspots Design Protocol (RMHDP) was presented for building a fast and efficient platform for nanobody affinity maturation. A three-dimensional analytical structural model of Nb02 was constructed and then docked with the antigen, the CD47 extracellular domain (CD47ext). Mutants with high binding affinity are predicted by the scoring of nanobody-antigen complexes based on molecular dynamics trajectories and simulation. Ultimately, an improved mutant with an 87.4-fold affinity (3.2 nM) and 7.36 °C higher thermal stability was obtained. These findings might contribute to computational affinity maturation of nanobodies via homology modeling using the recent advancements in computational power. The add-in of aromatic residues which formed aromatic-aromatic interaction plays a pivotal role in affinity and thermostability improvement. In a word, the methods used in this study might provide a reference for rapid and efficient in vitro affinity maturation of nanobodies.
Collapse
|
19
|
Röth S, Fulcher LJ, Sapkota GP. Advances in targeted degradation of endogenous proteins. Cell Mol Life Sci 2019; 76:2761-2777. [PMID: 31030225 PMCID: PMC6588652 DOI: 10.1007/s00018-019-03112-6] [Citation(s) in RCA: 59] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2019] [Revised: 03/23/2019] [Accepted: 04/16/2019] [Indexed: 01/07/2023]
Abstract
Protein silencing is often employed as a means to aid investigations in protein function and is increasingly desired as a therapeutic approach. Several types of protein silencing methodologies have been developed, including targeting the encoding genes, transcripts, the process of translation or the protein directly. Despite these advances, most silencing systems suffer from limitations. Silencing protein expression through genetic ablation, for example by CRISPR/Cas9 genome editing, is irreversible, time consuming and not always feasible. Similarly, RNA interference approaches warrant prolonged treatments, can lead to incomplete protein depletion and are often associated with off-target effects. Targeted proteolysis has the potential to overcome some of these limitations. The field of targeted proteolysis has witnessed the emergence of many methodologies aimed at targeting specific proteins for degradation in a spatio-temporal manner. In this review, we provide an appraisal of the different targeted proteolytic systems and discuss their applications in understanding protein function, as well as their potential in therapeutics.
Collapse
Affiliation(s)
- Sascha Röth
- MRC Protein Phosphorylation and Ubiquitylation Unit, School of Life Sciences, University of Dundee, Dow Street, Dundee, DD1 5EH, UK
| | - Luke J Fulcher
- MRC Protein Phosphorylation and Ubiquitylation Unit, School of Life Sciences, University of Dundee, Dow Street, Dundee, DD1 5EH, UK
| | - Gopal P Sapkota
- MRC Protein Phosphorylation and Ubiquitylation Unit, School of Life Sciences, University of Dundee, Dow Street, Dundee, DD1 5EH, UK.
| |
Collapse
|
20
|
PET imaging of distinct brain uptake of a nanobody and similarly-sized PAMAM dendrimers after intra-arterial administration. Eur J Nucl Med Mol Imaging 2019; 46:1940-1951. [PMID: 31161257 DOI: 10.1007/s00259-019-04347-y] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2018] [Accepted: 04/29/2019] [Indexed: 02/06/2023]
Abstract
INTRODUCTION We have recently shown that intracerebral delivery of an anti-VEGF monoclonal antibody bevacizumab using an intra-arterial (IA) infusion is more effective than intravenous administration. While antibodies are quickly emerging as therapeutics, their disadvantages such as large size, production logistics and immunogenicity motivate search for alternatives. Thus we have studied brain uptake of nanobodies and polyamidoamine (PAMAM) dendrimers. METHODS Nanobodies were conjugated with deferoxamine (DFO) to generate NB(DFO)2. Generation-4 PAMAM dendrimers were conjugated with DFO, and subsequently primary amines were capped with butane-1,2-diol functionalities to generate G4(DFO)3(Bdiol)110. Resulting conjugates were radiolabeled with zirconium-89. Brain uptake of 89ZrNB(DFO)2 and 89ZrG4(DFO)3(Bdiol)110 upon carotid artery vs tail vein infusions with intact BBB or osmotic blood-brain barrier opening (OBBBO) with mannitol in mice was monitored by dynamic positron emission tomography (PET) over 30 min to assess brain uptake and clearance, followed by whole-body PET-CT (computed tomography) imaging at 1 h and 24 h post-infusion (pi). Imaging results were subsequently validated by ex-vivo biodistribution. RESULTS Intravenous administration of 89ZrNB(DFO)2 and 89ZrG4(DFO)3(Bdiol)110 resulted in their negligible brain accumulation regardless of BBB status and timing of OBBBO. Intra-arterial (IA) administration of 89ZrNB(DFO)2 dramatically increased its brain uptake, which was further potentiated with prior OBBBO. Half of the initial brain uptake was retained after 24 h. In contrast, IA infusion of 89ZrG4(DFO)3(Bdiol)110 resulted in poor initial accumulation in the brain, with complete clearance within 1 h of administration. Ex-vivo biodistribution results reflected those on PET-CT. CONCLUSIONS IA delivery of nanobodies might be an attractive therapeutic platform for CNS disorders where prolonged intracranial retention is necessary.
Collapse
|
21
|
Beghein E, Devriese D, Van Hoey E, Gettemans J. Cortactin and fascin-1 regulate extracellular vesicle release by controlling endosomal trafficking or invadopodia formation and function. Sci Rep 2018; 8:15606. [PMID: 30353022 PMCID: PMC6199335 DOI: 10.1038/s41598-018-33868-z] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2018] [Accepted: 10/06/2018] [Indexed: 12/21/2022] Open
Abstract
Cancer cell-derived extracellular vesicles (EVs) are increasingly being recognized as genuine invasive structures as they contribute to many aspects of invasion and metastasis. Unfortunately, the mechanisms underlying EV biogenesis or release are still poorly understood. Recent reports however indicate a role of the actin cytoskeleton in this process. In this study, we have exploited thoroughly characterized camelid nanobodies against actin binding proteins cortactin and fascin-1, a branched actin regulator and actin bundler, respectively, in order to assess their roles in EV biogenesis or release. Using this strategy, we demonstrate a role of the cortactin NTA and SH3 domains in EV release. Fascin-1 also regulates EV release, independently of its actin-bundling activity. We show a contribution of these protein domains in endosomal trafficking, a crucial step in EV biogenesis, and we confirm that EVs are preferentially released at invadopodia, the latter being actin-rich invasive cell protrusions in which cortactin and fascin-1 perform essential roles. Accordingly, EVs are enriched with invadopodial proteins such as the matrix metalloproteinase MT1-MMP and exert gelatinolytic activity. Based on our findings, we report that both cortactin and fascin-1 play key roles in EV release by regulating endosomal trafficking or invadopodia formation and function.
Collapse
Affiliation(s)
- Els Beghein
- Department of Biomolecular Medicine, Faculty of Medicine and Health Sciences, Campus Rommelaere, A. Baertsoenkaai 3, Ghent University, Ghent, Belgium
| | - Delphine Devriese
- Department of Biomolecular Medicine, Faculty of Medicine and Health Sciences, Campus Rommelaere, A. Baertsoenkaai 3, Ghent University, Ghent, Belgium
| | - Evy Van Hoey
- Department of Biomolecular Medicine, Faculty of Medicine and Health Sciences, Campus Rommelaere, A. Baertsoenkaai 3, Ghent University, Ghent, Belgium
| | - Jan Gettemans
- Department of Biomolecular Medicine, Faculty of Medicine and Health Sciences, Campus Rommelaere, A. Baertsoenkaai 3, Ghent University, Ghent, Belgium.
| |
Collapse
|
22
|
Steels A, Verhelle A, Zwaenepoel O, Gettemans J. Intracellular displacement of p53 using transactivation domain (p53 TAD) specific nanobodies. MAbs 2018; 10:1045-1059. [PMID: 30111239 DOI: 10.1080/19420862.2018.1502025] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
The tumor suppressor p53 is of crucial importance in the prevention of cellular transformation. In the presence of cellular stress signals, the negative feedback loop between p53 and Mdm2, its main negative regulator, is disrupted, which results in the activation and stabilization of p53. Via a complex interplay between both transcription-dependent and - independent functions of p53, the cell will go through transient cell cycle arrest, cellular senescence or apoptosis. However, it remains difficult to completely fathom the mechanisms behind p53 regulation and its responses, considering the presence of multiple layers involved in fine-tuning them. In order to take the next step forward, novel research tools are urgently needed. We have developed single-domain antibodies, also known as nanobodies, that specifically bind with the N-terminal transactivation domain of wild type p53, but that leave the function of p53 as a transcriptional transactivator intact. When the nanobodies are equipped with a mitochondrial-outer-membrane (MOM)-tag, we can capture p53 at the mitochondria. This nanobody-induced mitochondrial delocalization of p53 is, in specific cases, associated with a decrease in cell viability and with morphological changes in the mitochondria. These findings underpin the potential of nanobodies as bona fide research tools to explore protein function and to unravel their biochemical pathways.
Collapse
Affiliation(s)
- Anneleen Steels
- a Department of Biochemistry, Faculty of Medicine and Health Sciences , Ghent University , Ghent , Belgium
| | - Adriaan Verhelle
- a Department of Biochemistry, Faculty of Medicine and Health Sciences , Ghent University , Ghent , Belgium
| | - Olivier Zwaenepoel
- a Department of Biochemistry, Faculty of Medicine and Health Sciences , Ghent University , Ghent , Belgium
| | - Jan Gettemans
- a Department of Biochemistry, Faculty of Medicine and Health Sciences , Ghent University , Ghent , Belgium
| |
Collapse
|
23
|
Bertier L, Hebbrecht T, Mettepenningen E, De Wit N, Zwaenepoel O, Verhelle A, Gettemans J. Nanobodies targeting cortactin proline rich, helical and actin binding regions downregulate invadopodium formation and matrix degradation in SCC-61 cancer cells. Biomed Pharmacother 2018; 102:230-241. [DOI: 10.1016/j.biopha.2018.03.064] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2017] [Revised: 03/10/2018] [Accepted: 03/12/2018] [Indexed: 01/19/2023] Open
|
24
|
Soler MA, Fortuna S, de Marco A, Laio A. Binding affinity prediction of nanobody-protein complexes by scoring of molecular dynamics trajectories. Phys Chem Chem Phys 2018; 20:3438-3444. [PMID: 29328338 DOI: 10.1039/c7cp08116b] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Nanobodies offer a viable alternative to antibodies for engineering high affinity binders. Their small size has an additional advantage: it allows exploiting computational protocols for optimizing their biophysical features, such as the binding affinity. The efficient prediction of this quantity is still considered a daunting task especially for modelled complexes. We show how molecular dynamics can successfully assist in the binding affinity prediction of modelled nanobody-protein complexes. The approximate initial configurations obtained by in silico design must undergo large rearrangements before achieving a stable conformation, in which the binding affinity can be meaningfully estimated. The scoring functions developed for the affinity evaluation of crystal structures will provide accurate estimates for modelled binding complexes if the scores are averaged over long finite temperature molecular dynamics simulations.
Collapse
|
25
|
Reporter-nanobody fusions (RANbodies) as versatile, small, sensitive immunohistochemical reagents. Proc Natl Acad Sci U S A 2018; 115:2126-2131. [PMID: 29440485 DOI: 10.1073/pnas.1722491115] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Sensitive and specific antibodies are essential for detecting molecules in cells and tissues. However, currently used polyclonal and monoclonal antibodies are often less specific than desired, difficult to produce, and available in limited quantities. A promising recent approach to circumvent these limitations is to employ chemically defined antigen-combining domains called "nanobodies," derived from single-chain camelid antibodies. Here, we used nanobodies to prepare sensitive unimolecular detection reagents by genetically fusing cDNAs encoding nanobodies to enzymatic or antigenic reporters. We call these fusions between a reporter and a nanobody "RANbodies." They can be used to localize epitopes and to amplify signals from fluorescent proteins. They can be generated and purified simply and in unlimited amounts and can be preserved safely and inexpensively in the form of DNA or digital sequence.
Collapse
|
26
|
Miller PS, Scott S, Masiulis S, De Colibus L, Pardon E, Steyaert J, Aricescu AR. Structural basis for GABA A receptor potentiation by neurosteroids. Nat Struct Mol Biol 2017; 24:986-992. [PMID: 28991263 PMCID: PMC6166781 DOI: 10.1038/nsmb.3484] [Citation(s) in RCA: 137] [Impact Index Per Article: 17.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2017] [Accepted: 09/13/2017] [Indexed: 12/18/2022]
Abstract
Type A γ-aminobutyric acid receptors (GABAARs) are the principal mediators of inhibitory neurotransmission in the human brain. Endogenous neurosteroids interact with GABAARs to regulate acute and chronic anxiety and are potent sedative, analgesic, anticonvulsant and anesthetic agents. Their mode of binding and mechanism of receptor potentiation, however, remain unknown. Here we report crystal structures of a chimeric GABAAR construct in apo and pregnanolone-bound states. The neurosteroid-binding site is mechanically coupled to the helices lining the ion channel pore and modulates the desensitization-gate conformation. We demonstrate that the equivalent site is responsible for physiological, heteromeric GABAAR potentiation and explain the contrasting modulatory properties of 3a versus 3b neurosteroid epimers. These results illustrate how peripheral lipid ligands can regulate the desensitization gate of GABAARs, a process of broad relevance to pentameric ligand-gated ion channels.
Collapse
Affiliation(s)
- Paul S. Miller
- Division of Structural Biology, Wellcome Trust Centre for Human Genetics, University of Oxford, Roosevelt Drive, Oxford OX3 7BN, United Kingdom
| | - Suzanne Scott
- Division of Structural Biology, Wellcome Trust Centre for Human Genetics, University of Oxford, Roosevelt Drive, Oxford OX3 7BN, United Kingdom
- Neurobiology Division, MRC Laboratory of Molecular Biology, Cambridge CB2 0QH, United Kingdom
| | - Simonas Masiulis
- Division of Structural Biology, Wellcome Trust Centre for Human Genetics, University of Oxford, Roosevelt Drive, Oxford OX3 7BN, United Kingdom
- Neurobiology Division, MRC Laboratory of Molecular Biology, Cambridge CB2 0QH, United Kingdom
| | - Luigi De Colibus
- Division of Structural Biology, Wellcome Trust Centre for Human Genetics, University of Oxford, Roosevelt Drive, Oxford OX3 7BN, United Kingdom
| | - Els Pardon
- Structural Biology Brussels, Vrije Universiteit Brussel (VUB), Brussels, Belgium
- Structural Biology Research Center, VIB, Brussels, Belgium
| | - Jan Steyaert
- Structural Biology Brussels, Vrije Universiteit Brussel (VUB), Brussels, Belgium
- Structural Biology Research Center, VIB, Brussels, Belgium
| | - A. Radu Aricescu
- Division of Structural Biology, Wellcome Trust Centre for Human Genetics, University of Oxford, Roosevelt Drive, Oxford OX3 7BN, United Kingdom
- Neurobiology Division, MRC Laboratory of Molecular Biology, Cambridge CB2 0QH, United Kingdom
| |
Collapse
|
27
|
Verhelle A, Van Overbeke W, Peleman C, De Smet R, Zwaenepoel O, Lahoutte T, Van Dorpe J, Devoogdt N, Gettemans J. Non-Invasive Imaging of Amyloid Deposits in a Mouse Model of AGel Using 99mTc-Modified Nanobodies and SPECT/CT. Mol Imaging Biol 2017; 18:887-897. [PMID: 27130233 DOI: 10.1007/s11307-016-0960-y] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
PURPOSE Gelsolin amyloidosis (AGel), also known as familial amyloidosis, Finnish type (FAF), is an autosomal, dominant, incurable disease caused by a point mutation (G654A/T) in the gelsolin (GSN) gene. The mutation results in loss of a Ca2+-binding site in the second gelsolin domain. Subsequent incorrect folding exposes a cryptic furin cleavage site, leading to the formation of a 68-kDa C-terminal cleavage product (C68) in the trans-Golgi network. This C68 fragment is cleaved by membrane type 1-matrix metalloproteinase (MT1-MMP) during secretion into the extracellular environment, releasing 8- and 5-kDa amyloidogenic peptides. These peptides aggregate and cause disease-associated symptoms. We set out to investigate whether AGel-specific nanobodies could be used to monitor amyloidogenic gelsolin buildup. PROCEDURES Three nanobodies (FAF Nb1-3) raised against the 8-kDa fragment were screened as AGel amyloid imaging agents in WT and AGel mice using 99mTc-based single-photon emission computed tomography (SPECT)/X-ray tomography (CT), biodistribution analysis, and immunofluorescence (IF). The quantitative characteristics were analyzed in a follow-up study with a Nb11-expressing mouse model. RESULTS All three nanobodies possess the characteristics desired for a 99mTc-based SPECT/CT imaging agent, high specificity and a low background signal. FAF Nb1 was identified as the most potent, based on its superior signal-to-noise ratio and signal specificity. As a proof of concept, we implemented 99mTc-FAF Nb1 in a follow-up study of the Nb11-expressing AGel mouse model. Using biodistribution analysis and immunofluorescence, we demonstrated the validity of the data acquired via 99mTc-FAF Nb1 SPECT/CT. CONCLUSION These findings demonstrate the potential of this nanobody as a non-invasive tool to image amyloidogenic gelsolin deposition and assess the therapeutic capacity of AGel therapeutics currently under development. We propose that this approach can be extended to other amyloid diseases, thereby contributing to the development of specific therapies.
Collapse
Affiliation(s)
- Adriaan Verhelle
- Department of Biochemistry, Faculty of Medicine and Health Sciences, Ghent University, Albert Baertsoenkaai 3, B-9000, Ghent, Belgium
| | - Wouter Van Overbeke
- Department of Biochemistry, Faculty of Medicine and Health Sciences, Ghent University, Albert Baertsoenkaai 3, B-9000, Ghent, Belgium
| | - Cindy Peleman
- In Vivo Cellular and Molecular Imaging Laboratory, Vrije Universiteit Brussel, Brussels, Belgium
| | - Rebecca De Smet
- Department of Medical and Forensic Pathology, Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium
| | - Olivier Zwaenepoel
- Department of Biochemistry, Faculty of Medicine and Health Sciences, Ghent University, Albert Baertsoenkaai 3, B-9000, Ghent, Belgium
| | - Tony Lahoutte
- In Vivo Cellular and Molecular Imaging Laboratory, Vrije Universiteit Brussel, Brussels, Belgium
| | - Jo Van Dorpe
- Department of Medical and Forensic Pathology, Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium
| | - Nick Devoogdt
- In Vivo Cellular and Molecular Imaging Laboratory, Vrije Universiteit Brussel, Brussels, Belgium
| | - Jan Gettemans
- Department of Biochemistry, Faculty of Medicine and Health Sciences, Ghent University, Albert Baertsoenkaai 3, B-9000, Ghent, Belgium.
| |
Collapse
|
28
|
Hebbrecht T, Van Audenhove I, Zwaenepoel O, Verhelle A, Gettemans J. VCA nanobodies target N-WASp to reduce invadopodium formation and functioning. PLoS One 2017; 12:e0185076. [PMID: 28938008 PMCID: PMC5609757 DOI: 10.1371/journal.pone.0185076] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2017] [Accepted: 09/06/2017] [Indexed: 01/08/2023] Open
Abstract
Invasive cancer cells develop small actin-based protrusions called invadopodia, which perform a primordial role in metastasis and extracellular matrix remodelling. Neural Wiskott-Aldrich syndrome protein (N-WASp) is a scaffold protein which can directly bind to actin monomers and Arp2/3 and is a crucial player in the formation of an invadopodium precursor. Expression modulation has pointed to an important role for N-WASp in invadopodium formation but the role of its C-terminal VCA domain in this process remains unknown. In this study, we generated alpaca nanobodies against the N-WASp VCA domain and investigated if these nanobodies affect invadopodium formation. By using this approach, we were able to study functions of a selected functional/structural N-WASp protein domain in living cells, without requiring overexpression, dominant negative mutants or siRNAs which target the gene, and hence the entire protein. When expressed as intrabodies, the VCA nanobodies significantly reduced invadopodium formation in both MDA-MB-231 breast cancer and HNSCC61 head and neck squamous cancer cells. Furthermore, expression of distinct VCA Nbs (VCA Nb7 and VCA Nb14) in PC-3 prostate cancer cells resulted in reduced overall matrix degradation without affecting MMP9 secretion/activation or MT1-MMP localisation at invadopodial membranes. From these results, we conclude that we have generated nanobodies targeting N-WASp which reduce invadopodium formation and functioning, most likely via regulation of N-WASp—Arp2/3 complex interaction, indicating that this region of N-WASp plays an important role in these processes.
Collapse
Affiliation(s)
- Tim Hebbrecht
- Department of Biochemistry, Faculty of Medicine and Health Sciences, Rommelaere Campus, Ghent University, Ghent, Belgium
| | - Isabel Van Audenhove
- Department of Biochemistry, Faculty of Medicine and Health Sciences, Rommelaere Campus, Ghent University, Ghent, Belgium
| | - Olivier Zwaenepoel
- Department of Biochemistry, Faculty of Medicine and Health Sciences, Rommelaere Campus, Ghent University, Ghent, Belgium
| | - Adriaan Verhelle
- Department of Biochemistry, Faculty of Medicine and Health Sciences, Rommelaere Campus, Ghent University, Ghent, Belgium
| | - Jan Gettemans
- Department of Biochemistry, Faculty of Medicine and Health Sciences, Rommelaere Campus, Ghent University, Ghent, Belgium
- * E-mail:
| |
Collapse
|
29
|
Beghein E, Gettemans J. Nanobody Technology: A Versatile Toolkit for Microscopic Imaging, Protein-Protein Interaction Analysis, and Protein Function Exploration. Front Immunol 2017; 8:771. [PMID: 28725224 PMCID: PMC5495861 DOI: 10.3389/fimmu.2017.00771] [Citation(s) in RCA: 135] [Impact Index Per Article: 16.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2017] [Accepted: 06/16/2017] [Indexed: 01/05/2023] Open
Abstract
Over the last two decades, nanobodies or single-domain antibodies have found their way in research, diagnostics, and therapy. These antigen-binding fragments, derived from Camelid heavy chain only antibodies, possess remarkable characteristics that favor their use over conventional antibodies or fragments thereof, in selected areas of research. In this review, we assess the current status of nanobodies as research tools in diverse aspects of fundamental research. We discuss the use of nanobodies as detection reagents in fluorescence microscopy and focus on recent advances in super-resolution microscopy. Second, application of nanobody technology in investigating protein–protein interactions is reviewed, with emphasis on possible uses in mass spectrometry. Finally, we discuss the potential value of nanobodies in studying protein function, and we focus on their recently reported application in targeted protein degradation. Throughout the review, we highlight state-of-the-art engineering strategies that could expand nanobody versatility and we suggest future applications of the technology in the selected areas of fundamental research.
Collapse
Affiliation(s)
- Els Beghein
- Nanobody Laboratory, Department of Biochemistry, Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium
| | - Jan Gettemans
- Nanobody Laboratory, Department of Biochemistry, Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium
| |
Collapse
|
30
|
Verhelle A, Nair N, Everaert I, Van Overbeke W, Supply L, Zwaenepoel O, Peleman C, Van Dorpe J, Lahoutte T, Devoogdt N, Derave W, Chuah MK, VandenDriessche T, Gettemans J. AAV9 delivered bispecific nanobody attenuates amyloid burden in the gelsolin amyloidosis mouse model. Hum Mol Genet 2017; 26:1353-1364. [PMID: 28334940 DOI: 10.1093/hmg/ddx056] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2016] [Accepted: 02/07/2017] [Indexed: 12/23/2022] Open
Abstract
Gelsolin amyloidosis is a dominantly inherited, incurable type of amyloidosis. A single point mutation in the gelsolin gene (G654A is most common) results in the loss of a Ca2+ binding site in the second gelsolin domain. Consequently, this domain partly unfolds and exposes an otherwise buried furin cleavage site at the surface. During secretion of mutant plasma gelsolin consecutive cleavage by furin and MT1-MMP results in the production of 8 and 5 kDa amyloidogenic peptides. Nanobodies that are able to (partly) inhibit furin or MT1-MMP proteolysis have previously been reported. In this study, the nanobodies have been combined into a single bispecific format able to simultaneously shield mutant plasma gelsolin from intracellular furin and extracellular MT1-MMP activity. We report the successful in vivo expression of this bispecific nanobody following adeno-associated virus serotype 9 gene therapy in gelsolin amyloidosis mice. Using SPECT/CT and immunohistochemistry, a reduction in gelsolin amyloid burden was detected which translated into improved muscle contractile properties. We conclude that a nanobody-based gene therapy using adeno-associated viruses shows great potential as a novel strategy in gelsolin amyloidosis and potentially other amyloid diseases.
Collapse
Affiliation(s)
- Adriaan Verhelle
- Department of Biochemistry, Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium
| | - Nisha Nair
- Department of Gene Therapy & Regenerative Medicine, Free University of Brussels (VUB), Brussels, Belgium
| | - Inge Everaert
- Department of Movement and Sport Sciences, Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium
| | - Wouter Van Overbeke
- Department of Biochemistry, Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium
| | - Lynn Supply
- Department of Medical and Forensic Pathology, Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium
| | - Olivier Zwaenepoel
- Department of Biochemistry, Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium
| | - Cindy Peleman
- In Vivo Cellular and Molecular Imaging Laboratory, Free University of Brussels (VUB), Brussels, Belgium
| | - Jo Van Dorpe
- Department of Medical and Forensic Pathology, Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium
| | - Tony Lahoutte
- In Vivo Cellular and Molecular Imaging Laboratory, Free University of Brussels (VUB), Brussels, Belgium
| | - Nick Devoogdt
- In Vivo Cellular and Molecular Imaging Laboratory, Free University of Brussels (VUB), Brussels, Belgium
| | - Wim Derave
- Department of Movement and Sport Sciences, Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium
| | - Marinee K Chuah
- Department of Gene Therapy & Regenerative Medicine, Free University of Brussels (VUB), Brussels, Belgium.,Department of Cardiovascular Sciences, Catholic University of Leuven (KU Leuven), Leuven, Belgium
| | - Thierry VandenDriessche
- Department of Gene Therapy & Regenerative Medicine, Free University of Brussels (VUB), Brussels, Belgium.,Department of Cardiovascular Sciences, Catholic University of Leuven (KU Leuven), Leuven, Belgium
| | - Jan Gettemans
- Department of Biochemistry, Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium
| |
Collapse
|
31
|
Böldicke T. Single domain antibodies for the knockdown of cytosolic and nuclear proteins. Protein Sci 2017; 26:925-945. [PMID: 28271570 PMCID: PMC5405437 DOI: 10.1002/pro.3154] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2017] [Accepted: 03/03/2017] [Indexed: 12/12/2022]
Abstract
Single domain antibodies (sdAbs) from camels or sharks comprise only the variable heavy chain domain. Human sdAbs comprise the variable domain of the heavy chain (VH) or light chain (VL) and can be selected from human antibodies. SdAbs are stable, nonaggregating molecules in vitro and in vivo compared to complete antibodies and scFv fragments. They are excellent novel inhibitors of cytosolic/nuclear proteins because they are correctly folded inside the cytosol in contrast to scFv fragments. SdAbs are unique because of their excellent specificity and possibility to target posttranslational modifications such as phosphorylation sites, conformers or interaction regions of proteins that cannot be targeted with genetic knockout techniques and are impossible to knockdown with RNAi. The number of inhibiting cytosolic/nuclear sdAbs is increasing and usage of synthetic single pot single domain antibody libraries will boost the generation of these fascinating molecules without the need of immunization. The most frequently selected antigenic epitopes belong to viral and oncogenic proteins, followed by toxins, proteins of the nervous system as well as plant- and drosophila proteins. It is now possible to select functional sdAbs against virtually every cytosolic/nuclear protein and desired epitope. The development of new endosomal escape protein domains and cell-penetrating peptides for efficient transfection broaden the application of inhibiting sdAbs. Last but not least, the generation of relatively new cell-specific nanoparticles such as polymersomes and polyplexes carrying cytosolic/nuclear sdAb-DNA or -protein will pave the way to apply cytosolic/nuclear sdAbs for inhibition of viral infection and cancer in the clinic.
Collapse
Affiliation(s)
- Thomas Böldicke
- Helmholtz Centre for Infection Research, Structure and Function of ProteinsInhoffenstraße 7, D‐38124BraunschweigGermany
| |
Collapse
|
32
|
Bertier L, Boucherie C, Zwaenepoel O, Vanloo B, Van Troys M, Van Audenhove I, Gettemans J. Inhibitory cortactin nanobodies delineate the role of NTA- and SH3-domain-specific functions during invadopodium formation and cancer cell invasion. FASEB J 2017; 31:2460-2476. [PMID: 28235780 DOI: 10.1096/fj.201600810rr] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2016] [Accepted: 02/07/2017] [Indexed: 01/08/2023]
Abstract
Cancer cells exploit different strategies to escape from the primary tumor, gain access to the circulation, disseminate throughout the body, and form metastases, the leading cause of death by cancer. Invadopodia, proteolytically active plasma membrane extensions, are essential in this escape mechanism. Cortactin is involved in every phase of invadopodia formation, and its overexpression is associated with increased invadopodia formation, extracellular matrix degradation, and cancer cell invasion. To analyze endogenous cortactin domain function in these processes, we characterized the effects of nanobodies that are specific for the N-terminal acidic domain of cortactin and expected to target small epitopes within this domain. These nanobodies inhibit cortactin-mediated actin-related protein (Arp)2/3 activation, and, after their intracellular expression in cancer cells, decrease invadopodia formation, extracellular matrix degradation, and cancer cell invasion. In addition, one of the nanobodies affects Arp2/3 interaction and invadopodium stability, and a nanobody targeting the Src homology 3 domain of cortactin enabled comparison of 2 functional regions in invadopodium formation or stability. Given their common and distinct effects, we validate cortactin nanobodies as an instrument to selectively block and study distinct domains within a protein with unprecedented precision, aiding rational future generation of protein domain-selective therapeutic compounds.-Bertier, L., Boucherie, C., Zwaenepoel, O., Vanloo, B., Van Troys, M., Van Audenhove, I., Gettemans, J. Inhibitory cortactin nanobodies delineate the role of NTA- and SH3-domain-specific functions during invadopodium formation and cancer cell invasion.
Collapse
Affiliation(s)
- Laurence Bertier
- Department of Biochemistry, Faculty of Medicine and Health Sciences, Ghent University-Campus Rommelaere, Ghent, Belgium
| | - Ciska Boucherie
- Department of Biochemistry, Faculty of Medicine and Health Sciences, Ghent University-Campus Rommelaere, Ghent, Belgium
| | - Olivier Zwaenepoel
- Department of Biochemistry, Faculty of Medicine and Health Sciences, Ghent University-Campus Rommelaere, Ghent, Belgium
| | - Berlinda Vanloo
- Department of Biochemistry, Faculty of Medicine and Health Sciences, Ghent University-Campus Rommelaere, Ghent, Belgium
| | - Marleen Van Troys
- Department of Biochemistry, Faculty of Medicine and Health Sciences, Ghent University-Campus Rommelaere, Ghent, Belgium
| | - Isabel Van Audenhove
- Department of Biochemistry, Faculty of Medicine and Health Sciences, Ghent University-Campus Rommelaere, Ghent, Belgium
| | - Jan Gettemans
- Department of Biochemistry, Faculty of Medicine and Health Sciences, Ghent University-Campus Rommelaere, Ghent, Belgium
| |
Collapse
|
33
|
Beghein E, Van Audenhove I, Zwaenepoel O, Verhelle A, De Ganck A, Gettemans J. A new survivin tracer tracks, delocalizes and captures endogenous survivin at different subcellular locations and in distinct organelles. Sci Rep 2016; 6:31177. [PMID: 27514728 PMCID: PMC4981888 DOI: 10.1038/srep31177] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2016] [Accepted: 07/13/2016] [Indexed: 01/18/2023] Open
Abstract
Survivin, the smallest member of the inhibitor of apoptosis protein family, plays a central role during mitosis and exerts a cytoprotective function. Survivin is highly expressed in most cancer types and contributes to multiple facets of carcinogenesis. The molecular mechanisms underlying its highly diverse functions need to be extensively explored, which is crucial for rational design of future personalized therapeutics. In this study, we have generated an alpaca survivin nanobody (SVVNb8) that binds with low nanomolar affinity to its target. When expressed as an intrabody in HeLa cells, SVVNb8 faithfully tracks survivin during different phases of mitosis without interfering with survivin function. Furthermore, coupling SVVNb8 with a subcellular delocalization tag efficiently redirects endogenous survivin towards the nucleus, the cytoplasm, peroxisomes and even to the intermembrane space of mitochondria where it presumably interacts with resident mitochondrial survivin. Based on our findings, we believe that SVVNb8 is an excellent instrument to further elucidate survivin biology and topography, and can serve as a model system to investigate mitochondrial and peroxisomal (survivin) protein import.
Collapse
Affiliation(s)
- Els Beghein
- Department of Biochemistry, Faculty of Medicine and Health Sciences, Campus Rommelaere, A. Baertsoenkaai 3, Ghent University, Ghent, Belgium
| | - Isabel Van Audenhove
- Department of Biochemistry, Faculty of Medicine and Health Sciences, Campus Rommelaere, A. Baertsoenkaai 3, Ghent University, Ghent, Belgium
| | - Olivier Zwaenepoel
- Department of Biochemistry, Faculty of Medicine and Health Sciences, Campus Rommelaere, A. Baertsoenkaai 3, Ghent University, Ghent, Belgium
| | - Adriaan Verhelle
- Department of Biochemistry, Faculty of Medicine and Health Sciences, Campus Rommelaere, A. Baertsoenkaai 3, Ghent University, Ghent, Belgium
| | - Ariane De Ganck
- Department of Biochemistry, Faculty of Medicine and Health Sciences, Campus Rommelaere, A. Baertsoenkaai 3, Ghent University, Ghent, Belgium
| | - Jan Gettemans
- Department of Biochemistry, Faculty of Medicine and Health Sciences, Campus Rommelaere, A. Baertsoenkaai 3, Ghent University, Ghent, Belgium
| |
Collapse
|
34
|
Mazur AJ, Radaszkiewicz T, Makowiecka A, Malicka-Błaszkiewicz M, Mannherz HG, Nowak D. Gelsolin interacts with LamR, hnRNP U, nestin, Arp3 and β-tubulin in human melanoma cells as revealed by immunoprecipitation and mass spectrometry. Eur J Cell Biol 2016; 95:26-41. [DOI: 10.1016/j.ejcb.2015.11.001] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2015] [Revised: 10/08/2015] [Accepted: 11/04/2015] [Indexed: 01/25/2023] Open
|
35
|
Ta DT, Redeker ES, Billen B, Reekmans G, Sikulu J, Noben JP, Guedens W, Adriaensens P. An efficient protocol towards site-specifically clickable nanobodies in high yield: cytoplasmic expression in Escherichia coli combined with intein-mediated protein ligation. Protein Eng Des Sel 2015; 28:351-63. [PMID: 26243885 DOI: 10.1093/protein/gzv032] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2015] [Accepted: 07/01/2015] [Indexed: 11/13/2022] Open
Abstract
In this study, several expression strategies were investigated in order to develop a generic, highly productive and efficient protocol to produce nanobodies modified with a clickable alkyne function at their C-terminus via the intein-mediated protein ligation (IPL) technique. Hereto, the nanobody targeting the vascular cell adhesion molecule 1 (NbVCAM1) was used as a workhorse. The highlights of the protocol can be ascribed to a cytoplasmic expression of the nanobody-intein-chitin-binding domain fusion protein in the Escherichia coli SHuffle(®) T7 cells with a C-terminal extension, i.e. LEY, EFLEY or His6 spacer peptide, in the commonly used Luria-Bertani medium. The combination of these factors led to a high yield (up to 22 mg/l of culture) and nearly complete alkynation efficiency of the C-terminally modified nanobody via IPL. This yield can even be improved to ∼45 mg/l in the EnPresso(®) growth system but this method is more expensive and time-consuming. The resulting alkynated nanobodies retained excellent binding capacity towards the recombinant human VCAM1. The presented protocol benefits from time- and cost-effectiveness, which allows a feasible production up-scaling of generic alkynated nanobodies. The production of high quantities of site-specifically modified nanobodies paves the way to new biosurface applications that demand for a homogeneously oriented nanobody coupling. Prospectively, the alkynated nanobodies can be covalently coupled to a multitude of azide-containing counterparts, e.g. contrast labeling agents, particles or surfaces for numerous innovative applications.
Collapse
Affiliation(s)
- Duy Tien Ta
- Biomolecule Design Group, Institute for Materials Research (IMO), Hasselt University, Agoralaan-Building D, Diepenbeek BE-3590, Belgium Faculty of Food Technology and Biotechnology, Can Tho University of Technology, Can Tho, Vietnam
| | - Erik Steen Redeker
- Maastricht Science Programme, Maastricht University, Maastricht 6200 MD, The Netherlands
| | - Brecht Billen
- Biomolecule Design Group, Institute for Materials Research (IMO), Hasselt University, Agoralaan-Building D, Diepenbeek BE-3590, Belgium
| | - Gunter Reekmans
- Applied and Analytical Chemistry, Institute for Materials Research (IMO), Hasselt University, Agoralaan-Building D, Diepenbeek BE-3590, Belgium
| | - Josephine Sikulu
- Biomolecule Design Group, Institute for Materials Research (IMO), Hasselt University, Agoralaan-Building D, Diepenbeek BE-3590, Belgium
| | - Jean-Paul Noben
- Biomedical Research Institute (Biomed) and School of Life Sciences, Transnationale Universiteit Limburg, Hasselt University, Agoralaan-Building C, Diepenbeek BE-3590, Belgium
| | - Wanda Guedens
- Biomolecule Design Group, Institute for Materials Research (IMO), Hasselt University, Agoralaan-Building D, Diepenbeek BE-3590, Belgium
| | - Peter Adriaensens
- Biomolecule Design Group, Institute for Materials Research (IMO), Hasselt University, Agoralaan-Building D, Diepenbeek BE-3590, Belgium Applied and Analytical Chemistry, Institute for Materials Research (IMO), Hasselt University, Agoralaan-Building D, Diepenbeek BE-3590, Belgium
| |
Collapse
|
36
|
Van Overbeke W, Wongsantichon J, Everaert I, Verhelle A, Zwaenepoel O, Loonchanta A, Burtnick LD, De Ganck A, Hochepied T, Haigh J, Cuvelier C, Derave W, Robinson RC, Gettemans J. An ER-directed gelsolin nanobody targets the first step in amyloid formation in a gelsolin amyloidosis mouse model. Hum Mol Genet 2015; 24:2492-507. [PMID: 25601851 DOI: 10.1093/hmg/ddv010] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2014] [Accepted: 01/14/2015] [Indexed: 01/01/2023] Open
Abstract
Hereditary gelsolin amyloidosis is an autosomal dominantly inherited amyloid disorder. A point mutation in the GSN gene (G654A being the most common one) results in disturbed calcium binding by the second gelsolin domain (G2). As a result, the folding of G2 is hampered, rendering the mutant plasma gelsolin susceptible to a proteolytic cascade. Consecutive cleavage by furin and MT1-MMP-like proteases generates 8 and 5 kDa amyloidogenic peptides that cause neurological, ophthalmological and dermatological findings. To this day, no specific treatment is available to counter the pathogenesis. Using GSN nanobody 11 as a molecular chaperone, we aimed to protect mutant plasma gelsolin from furin proteolysis in the trans-Golgi network. We report a transgenic, GSN nanobody 11 secreting mouse that was used for crossbreeding with gelsolin amyloidosis mice. Insertion of the therapeutic nanobody gene into the gelsolin amyloidosis mouse genome resulted in improved muscle contractility. X-ray crystal structure determination of the gelsolin G2:Nb11 complex revealed that Nb11 does not directly block the furin cleavage site. We conclude that nanobodies can be used to shield substrates from aberrant proteolysis and this approach might establish a novel therapeutic strategy in amyloid diseases.
Collapse
Affiliation(s)
| | - Jantana Wongsantichon
- Institute of Molecular and Cellular Biology, A*STAR, Biopolis, Singapore 138673, Singapore
| | - Inge Everaert
- Department of Movement and Sport Sciences, Faculty of Medicine and Health Sciences
| | - Adriaan Verhelle
- Department of Biochemistry, Faculty of Medicine and Health Sciences
| | | | - Anantasak Loonchanta
- Department of Chemistry and Centre for Blood Research, Life Sciences Institute, University of British Columbia, Vancouver, British Columbia, Canada
| | - Leslie D Burtnick
- Department of Chemistry and Centre for Blood Research, Life Sciences Institute, University of British Columbia, Vancouver, British Columbia, Canada
| | - Ariane De Ganck
- Department of Biochemistry, Faculty of Medicine and Health Sciences
| | - Tino Hochepied
- Department for Molecular Biomedical Research, VIB, Ghent, Belgium, Department of Biomedical Molecular Biology and
| | - Jody Haigh
- Department of Biomedical Molecular Biology and Vascular Cell Biology Unit, VIB Inflammation Research Centre, Ghent, Belgium and Mammalian Functional Genetics Laboratory, Division of Blood Cancers, Australian Centre for Blood Diseases, Department of Clinical Haematology, Monash University and Alfred Health Centre, Melbourne, Australia
| | - Claude Cuvelier
- Department of Pathology, Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium
| | - Wim Derave
- Department of Movement and Sport Sciences, Faculty of Medicine and Health Sciences
| | - Robert C Robinson
- Institute of Molecular and Cellular Biology, A*STAR, Biopolis, Singapore 138673, Singapore, Department of Biochemistry, National University of Singapore, 8 Medical Drive, Singapore 117597, Singapore,
| | - Jan Gettemans
- Department of Biochemistry, Faculty of Medicine and Health Sciences,
| |
Collapse
|
37
|
Fascin actin bundling controls podosome turnover and disassembly while cortactin is involved in podosome assembly by its SH3 domain in THP-1 macrophages and dendritic cells. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2015; 1853:940-52. [PMID: 25601713 DOI: 10.1016/j.bbamcr.2015.01.003] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/19/2014] [Revised: 12/11/2014] [Accepted: 01/08/2015] [Indexed: 11/21/2022]
Abstract
Podosomes are dynamic degrading devices present in myeloid cells among other cell types. They consist of an actin core with associated regulators, surrounded by an adhesive ring. Both fascin and cortactin are known constituents but the role of fascin actin bundling is still unclear and cortactin research rather focuses on its homologue hematopoietic lineage cell-specific protein-1 (HS1). A fascin nanobody (FASNb5) that inhibits actin bundling and a cortactin nanobody (CORNb2) specifically targeting its Src-homology 3 (SH3) domain were used as unique tools to study the function of these regulators in podosome dynamics in both THP-1 macrophages and dendritic cells (DC). Upon intracellular FASNb5 expression, the few podosomes present were aberrantly stable, long-living and large, suggesting a role for fascin actin bundling in podosome turnover and disassembly. Fascin modulates this by balancing the equilibrium between branched and bundled actin networks. In the presence of CORNb2, the few podosomes formed show disrupted structures but their dynamics were unaffected. This suggests a role of the cortactin SH3 domain in podosome assembly. Remarkably, both nanobody-induced podosome-losses were compensated for by focal adhesion structures. Furthermore, matrix degradation capacities were altered and migratory phenotypes were lost. In conclusion, the cortactin SH3 domain contributes to podosome assembly while fascin actin bundling is a master regulator of podosome disassembly in THP-1 macrophages and DC.
Collapse
|
38
|
Bethuyne J, De Gieter S, Zwaenepoel O, Garcia-Pino A, Durinck K, Verhelle A, Hassanzadeh-Ghassabeh G, Speleman F, Loris R, Gettemans J. A nanobody modulates the p53 transcriptional program without perturbing its functional architecture. Nucleic Acids Res 2014; 42:12928-38. [PMID: 25324313 PMCID: PMC4227789 DOI: 10.1093/nar/gku962] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
The p53 transcription factor plays an important role in genome integrity. To perform this task, p53 regulates the transcription of genes promoting various cellular outcomes including cell cycle arrest, apoptosis or senescence. The precise regulation of this activity remains elusive as numerous mechanisms, e.g. posttranslational modifications of p53 and (non-)covalent p53 binding partners, influence the p53 transcriptional program. We developed a novel, non-invasive tool to manipulate endogenous p53. Nanobodies (Nb), raised against the DNA-binding domain of p53, allow us to distinctively target both wild type and mutant p53 with great specificity. Nb3 preferentially binds ‘structural’ mutant p53, i.e. R175H and R282W, while a second but distinct nanobody, Nb139, binds both mutant and wild type p53. The co-crystal structure of the p53 DNA-binding domain in complex with Nb139 (1.9 Å resolution) reveals that Nb139 binds opposite the DNA-binding surface. Furthermore, we demonstrate that Nb139 does not disturb the functional architecture of the p53 DNA-binding domain using conformation-specific p53 antibody immunoprecipitations, glutaraldehyde crosslinking assays and chromatin immunoprecipitation. Functionally, the binding of Nb139 to p53 allows us to perturb the transactivation of p53 target genes. We propose that reduced recruitment of transcriptional co-activators or modulation of selected post-transcriptional modifications account for these observations.
Collapse
Affiliation(s)
- Jonas Bethuyne
- Nanobody Lab, Department of Biochemistry, Ghent University, Albert Baertsoenkaai 3, B-9000 Ghent, Belgium
| | - Steven De Gieter
- Structural Biology Brussels, Department of Biotechnology, Vrije Universiteit Brussel and Structural Biology Research Center, Vlaams Instituut voor Biotechnologie (VIB), Pleinlaan 2, B-1050 Brussel, Belgium
| | - Olivier Zwaenepoel
- Nanobody Lab, Department of Biochemistry, Ghent University, Albert Baertsoenkaai 3, B-9000 Ghent, Belgium
| | - Abel Garcia-Pino
- Structural Biology Brussels, Department of Biotechnology, Vrije Universiteit Brussel and Structural Biology Research Center, Vlaams Instituut voor Biotechnologie (VIB), Pleinlaan 2, B-1050 Brussel, Belgium
| | - Kaat Durinck
- Center for Medical Genetics, Ghent University Hospital, De Pintelaan 185, B-9000 Ghent, Belgium
| | - Adriaan Verhelle
- Nanobody Lab, Department of Biochemistry, Ghent University, Albert Baertsoenkaai 3, B-9000 Ghent, Belgium
| | | | - Frank Speleman
- Center for Medical Genetics, Ghent University Hospital, De Pintelaan 185, B-9000 Ghent, Belgium
| | - Remy Loris
- Structural Biology Brussels, Department of Biotechnology, Vrije Universiteit Brussel and Structural Biology Research Center, Vlaams Instituut voor Biotechnologie (VIB), Pleinlaan 2, B-1050 Brussel, Belgium
| | - Jan Gettemans
- Nanobody Lab, Department of Biochemistry, Ghent University, Albert Baertsoenkaai 3, B-9000 Ghent, Belgium
| |
Collapse
|
39
|
Chaperone nanobodies protect gelsolin against MT1-MMP degradation and alleviate amyloid burden in the gelsolin amyloidosis mouse model. Mol Ther 2014; 22:1768-78. [PMID: 25023329 DOI: 10.1038/mt.2014.132] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2014] [Accepted: 07/07/2014] [Indexed: 01/11/2023] Open
Abstract
Gelsolin amyloidosis is an autosomal dominant incurable disease caused by a point mutation in the GSN gene (G654A/T), specifically affecting secreted plasma gelsolin. Incorrect folding of the mutant (D187N/Y) second gelsolin domain leads to a pathological proteolytic cascade. D187N/Y gelsolin is first cleaved by furin in the trans-Golgi network, generating a 68 kDa fragment (C68). Upon secretion, C68 is cleaved by MT1-MMP-like proteases in the extracellular matrix, releasing 8 kDa and 5 kDa amyloidogenic peptides which aggregate in multiple tissues and cause disease-associated symptoms. We developed nanobodies that recognize the C68 fragment, but not native wild type gelsolin, and used these as molecular chaperones to mitigate gelsolin amyloid buildup in a mouse model that recapitulates the proteolytic cascade. We identified gelsolin nanobodies that potently reduce C68 proteolysis by MT1-MMP in vitro. Converting these nanobodies into an albumin-binding format drastically increased their serum half-life in mice, rendering them suitable for intraperitoneal injection. A 12-week treatment schedule of heterozygote D187N gelsolin transgenic mice with recombinant bispecific gelsolin-albumin nanobody significantly decreased gelsolin buildup in the endomysium and concomitantly improved muscle contractile properties. These findings demonstrate that nanobodies may be of considerable value in the treatment of gelsolin amyloidosis and related diseases.
Collapse
|
40
|
Van Audenhove I, Boucherie C, Pieters L, Zwaenepoel O, Vanloo B, Martens E, Verbrugge C, Hassanzadeh-Ghassabeh G, Vandekerckhove J, Cornelissen M, De Ganck A, Gettemans J. Stratifying fascin and cortactin function in invadopodium formation using inhibitory nanobodies and targeted subcellular delocalization. FASEB J 2014; 28:1805-18. [PMID: 24414419 DOI: 10.1096/fj.13-242537] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Invadopodia are actin-rich protrusions arising through the orchestrated regulation of precursor assembly, stabilization, and maturation, endowing cancer cells with invasive properties. Using nanobodies (antigen-binding domains of Camelid heavy-chain antibodies) as perturbators of intracellular functions and/or protein domains at the level of the endogenous protein, we examined the specific contribution of fascin and cortactin during invadopodium formation in MDA-MB-231 breast and PC-3 prostate cancer cells. A nanobody (K(d)~35 nM, 1:1 stoichiometry) that disrupts fascin F-actin bundling emphasizes the importance of stable actin bundles in invadopodium array organization and turnover, matrix degradation, and cancer cell invasion. Cortactin-SH3 dependent WIP recruitment toward the plasma membrane was specifically inhibited by a cortactin nanobody (K(d)~75 nM, 1:1 stoichiometry). This functional domain is shown to be important for formation of properly organized invadopodia, MMP-9 secretion, matrix degradation, and cancer cell invasion. Notably, using a subcellular delocalization strategy to trigger protein loss of function, we uncovered a fascin-bundling-independent role in MMP-9 secretion. Hence, we demonstrate that nanobodies enable high resolution protein function mapping in cells.
Collapse
Affiliation(s)
- Isabel Van Audenhove
- 1Department of Biochemistry, Faculty of Medicine and Health Sciences, Ghent University, Albert Baertsoenkaai 3, B-9000 Ghent, Belgium.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Van Impe K, Bethuyne J, Cool S, Impens F, Ruano-Gallego D, De Wever O, Vanloo B, Van Troys M, Lambein K, Boucherie C, Martens E, Zwaenepoel O, Hassanzadeh-Ghassabeh G, Vandekerckhove J, Gevaert K, Fernández LÁ, Sanders NN, Gettemans J. A nanobody targeting the F-actin capping protein CapG restrains breast cancer metastasis. Breast Cancer Res 2013; 15:R116. [PMID: 24330716 PMCID: PMC3979033 DOI: 10.1186/bcr3585] [Citation(s) in RCA: 82] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2013] [Accepted: 12/06/2013] [Indexed: 12/16/2022] Open
Abstract
Introduction Aberrant turnover of the actin cytoskeleton is intimately associated with cancer cell migration and invasion. Frequently however, evidence is circumstantial, and a reliable assessment of the therapeutic significance of a gene product is offset by lack of inhibitors that target biologic properties of a protein, as most conventional drugs do, instead of the corresponding gene. Proteomic studies have demonstrated overexpression of CapG, a constituent of the actin cytoskeleton, in breast cancer. Indirect evidence suggests that CapG is involved in tumor cell dissemination and metastasis. In this study, we used llama-derived CapG single-domain antibodies or nanobodies in a breast cancer metastasis model to address whether inhibition of CapG activity holds therapeutic merit. Methods We raised single-domain antibodies (nanobodies) against human CapG and used these as intrabodies (immunomodulation) after lentiviral transduction of breast cancer cells. Functional characterization of nanobodies was performed to identify which biochemical properties of CapG are perturbed. Orthotopic and tail vein in vivo models of metastasis in nude mice were used to assess cancer cell spreading. Results With G-actin and F-actin binding assays, we identified a CapG nanobody that binds with nanomolar affinity to the first CapG domain. Consequently, CapG interaction with actin monomers or actin filaments is blocked. Intracellular delocalization experiments demonstrated that the nanobody interacts with CapG in the cytoplasmic environment. Expression of the nanobody in breast cancer cells restrained cell migration and Matrigel invasion. Notably, the nanobody prevented formation of lung metastatic lesions in orthotopic xenograft and tail-vein models of metastasis in immunodeficient mice. We showed that CapG nanobodies can be delivered into cancer cells by using bacteria harboring a type III protein secretion system (T3SS). Conclusions CapG inhibition strongly reduces breast cancer metastasis. A nanobody-based approach offers a fast track for gauging the therapeutic merit of drug targets. Mapping of the nanobody-CapG interface may provide a platform for rational design of pharmacologic compounds.
Collapse
|
42
|
De Clercq S, Boucherie C, Vandekerckhove J, Gettemans J, Guillabert A. L-plastin nanobodies perturb matrix degradation, podosome formation, stability and lifetime in THP-1 macrophages. PLoS One 2013; 8:e78108. [PMID: 24236012 PMCID: PMC3827245 DOI: 10.1371/journal.pone.0078108] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2013] [Accepted: 09/09/2013] [Indexed: 12/29/2022] Open
Abstract
Podosomes are cellular structures acting as degradation 'hot-spots' in monocytic cells. They appear as dot-like structures at the ventral cell surface, enriched in F-actin and actin regulators, including gelsolin and L-plastin. Gelsolin is an ubiquitous severing and capping protein, whereas L-plastin is a leukocyte-specific actin bundling protein. The presence of the capping protein CapG in podosomes has not yet been investigated. We used an innovative approach to investigate the role of these proteins in macrophage podosomes by means of nanobodies or Camelid single domain antibodies. Nanobodies directed against distinct domains of gelsolin, L-plastin or CapG were stably expressed in macrophage-like THP-1 cells. CapG was not enriched in podosomes. Gelsolin nanobodies had no effect on podosome formation or function but proved very effective in tracing distinct gelsolin populations. One gelsolin nanobody specifically targets actin-bound gelsolin and was effectively enriched in podosomes. A gelsolin nanobody that blocks gelsolin-G-actin interaction was not enriched in podosomes demonstrating that the calcium-activated and actin-bound conformation of gelsolin is a constituent of podosomes. THP-1 cells expressing inhibitory L-plastin nanobodies were hampered in their ability to form stable podosomes. Nanobodies did not perturb Ser5 phosphorylation of L-plastin although phosphorylated L-plastin was highly enriched in podosomes. Furthermore, nanobody-induced inhibition of L-plastin function gave rise to an irregular and unstable actin turnover of podosomes, resulting in diminished degradation of the underlying matrix. Altogether these results indicate that L-plastin is indispensable for podosome formation and function in macrophages.
Collapse
Affiliation(s)
- Sarah De Clercq
- Department of Biochemistry, Ghent University, Faculty of Medicine and Health Sciences, Ghent, Belgium
| | - Ciska Boucherie
- Department of Biochemistry, Ghent University, Faculty of Medicine and Health Sciences, Ghent, Belgium
| | - Joël Vandekerckhove
- Department of Biochemistry, Ghent University, Faculty of Medicine and Health Sciences, Ghent, Belgium
| | - Jan Gettemans
- Department of Biochemistry, Ghent University, Faculty of Medicine and Health Sciences, Ghent, Belgium
| | - Aude Guillabert
- Department of Biochemistry, Ghent University, Faculty of Medicine and Health Sciences, Ghent, Belgium
| |
Collapse
|
43
|
Van Audenhove I, Van Impe K, Ruano-Gallego D, De Clercq S, De Muynck K, Vanloo B, Verstraete H, Fernández LÁ, Gettemans J. Mapping cytoskeletal protein function in cells by means of nanobodies. Cytoskeleton (Hoboken) 2013; 70:604-22. [DOI: 10.1002/cm.21122] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2013] [Revised: 06/18/2013] [Accepted: 06/18/2013] [Indexed: 12/30/2022]
Affiliation(s)
- Isabel Van Audenhove
- Department of Biochemistry; Faculty of Medicine and Health Sciences, Ghent University; Belgium
| | - Katrien Van Impe
- Department of Biochemistry; Faculty of Medicine and Health Sciences, Ghent University; Belgium
| | - David Ruano-Gallego
- Department of Microbial Biotechnology; Centro Nacional de Biotecnología; Consejo Superior de Investigaciones Científicas (CSIC), Campus Cantoblanco Universidad Autónoma de Madrid (UAM); Madrid Spain
| | - Sarah De Clercq
- Department of Biochemistry; Faculty of Medicine and Health Sciences, Ghent University; Belgium
| | - Kevin De Muynck
- Department of Biochemistry; Faculty of Medicine and Health Sciences, Ghent University; Belgium
| | - Berlinda Vanloo
- Department of Biochemistry; Faculty of Medicine and Health Sciences, Ghent University; Belgium
| | - Hanne Verstraete
- Department of Biochemistry; Faculty of Medicine and Health Sciences, Ghent University; Belgium
| | - Luis Á. Fernández
- Department of Microbial Biotechnology; Centro Nacional de Biotecnología; Consejo Superior de Investigaciones Científicas (CSIC), Campus Cantoblanco Universidad Autónoma de Madrid (UAM); Madrid Spain
| | - Jan Gettemans
- Department of Biochemistry; Faculty of Medicine and Health Sciences, Ghent University; Belgium
| |
Collapse
|
44
|
De Clercq S, Zwaenepoel O, Martens E, Vandekerckhove J, Guillabert A, Gettemans J. Nanobody-induced perturbation of LFA-1/L-plastin phosphorylation impairs MTOC docking, immune synapse formation and T cell activation. Cell Mol Life Sci 2013; 70:909-22. [PMID: 23001012 PMCID: PMC11113607 DOI: 10.1007/s00018-012-1169-0] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2012] [Revised: 08/24/2012] [Accepted: 09/11/2012] [Indexed: 02/05/2023]
Abstract
The T cell integrin receptor LFA-1 orchestrates adhesion between T cells and antigen-presenting cells (APCs), resulting in formation of a contact zone known as the immune synapse (IS) which is supported by the cytoskeleton. L-plastin is a leukocyte-specific actin bundling protein that rapidly redistributes to the immune synapse following T cell-APC engagement. We used single domain antibodies (nanobodies, derived from camelid heavy-chain only antibodies) directed against functional and structural modules of L-plastin to investigate its contribution to formation of an immune synapse between Raji cells and human peripheral blood mononuclear cells or Jurkat T cells. Nanobodies that interact either with the EF hands or the actin binding domains of L-plastin both trapped L-plastin in an inactive conformation, causing perturbation of IS formation, MTOC docking towards the plasma membrane, T cell proliferation and IL-2 secretion. Both nanobodies delayed Ser(5) phosphorylation of L-plastin which is required for enhanced bundling activity. Moreover, one nanobody delayed LFA-1 phosphorylation, reduced the association between LFA-1 and L-plastin and prevented LFA-1 enrichment at the IS. Our findings reveal subtle mechanistic details that are difficult to attain by conventional means and show that L-plastin contributes to immune synapse formation at distinct echelons.
Collapse
Affiliation(s)
- Sarah De Clercq
- Department of Medical Protein Research, VIB, 9000 Ghent, Belgium
- Faculty of Medicine and Health Sciences, Ghent University, A. Baertsoenkaai 3, 9000 Ghent, Belgium
| | - Olivier Zwaenepoel
- Department of Medical Protein Research, VIB, 9000 Ghent, Belgium
- Faculty of Medicine and Health Sciences, Ghent University, A. Baertsoenkaai 3, 9000 Ghent, Belgium
| | - Evelien Martens
- Department of Medical Protein Research, VIB, 9000 Ghent, Belgium
- Faculty of Medicine and Health Sciences, Ghent University, A. Baertsoenkaai 3, 9000 Ghent, Belgium
| | - Joël Vandekerckhove
- Department of Medical Protein Research, VIB, 9000 Ghent, Belgium
- Faculty of Medicine and Health Sciences, Ghent University, A. Baertsoenkaai 3, 9000 Ghent, Belgium
| | - Aude Guillabert
- Department of Medical Protein Research, VIB, 9000 Ghent, Belgium
- Faculty of Medicine and Health Sciences, Ghent University, A. Baertsoenkaai 3, 9000 Ghent, Belgium
| | - Jan Gettemans
- Department of Medical Protein Research, VIB, 9000 Ghent, Belgium
- Faculty of Medicine and Health Sciences, Ghent University, A. Baertsoenkaai 3, 9000 Ghent, Belgium
| |
Collapse
|
45
|
Abstract
The discovery of naturally occurring, heavy-chain only antibodies in Camelidae, and their further development into small recombinant nanobodies, presents attractive alternatives in drug delivery and imaging. Easily expressed in microorganisms and amenable to engineering, nanobody derivatives are soluble, stable, versatile, and have unique refolding capacities, reduced aggregation tendencies, and high-target binding capabilities. This review outlines the current state of the art in nanobodies, focusing on their structural features and properties, production, technology, and the potential for modulating immune functions and for targeting tumors, toxins, and microbes.
Collapse
Affiliation(s)
- Christina G Siontorou
- Department of Industrial Management and Technology, University of Piraeus, Piraeus, Greece
| |
Collapse
|
46
|
Vanlandschoot P, Stortelers C, Beirnaert E, Ibañez LI, Schepens B, Depla E, Saelens X. Nanobodies®: New ammunition to battle viruses. Antiviral Res 2011; 92:389-407. [DOI: 10.1016/j.antiviral.2011.09.002] [Citation(s) in RCA: 83] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2011] [Revised: 08/30/2011] [Accepted: 09/06/2011] [Indexed: 01/23/2023]
|
47
|
Peddada N, Sagar A, Ashish, Garg R. Plasma gelsolin: a general prognostic marker of health. Med Hypotheses 2011; 78:203-10. [PMID: 22082609 DOI: 10.1016/j.mehy.2011.10.024] [Citation(s) in RCA: 56] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2011] [Revised: 10/19/2011] [Accepted: 10/21/2011] [Indexed: 11/25/2022]
Abstract
Plasma gelsolin (pGSN) is the only component of two member extracellular actin scavenger system capable of severing circulating actin microfilaments. Here, we put forth the hypothesis that pGSN level is an important and sensitive general prognostic biomarker for health and disease conditions in humans, urging the need for gelsolin replacement therapy to improve patient's health status. Clinical significance and the therapeutic importance of this protein have been well illustrated in animal models as well as in patients with various diseases. Patients with decreased pGSN levels were observed to have higher mortality rate, longer hospital stay and longer ventilation time in intensive care units as compared to healthy controls. pGSN levels were found to be increasing in patients recovering from diseases; furthermore, it has been confirmed that repletion with exogenous recombinant pGSN increases the survival rate in animal models of different acute insults. To be used as a biomarker of health, however, establishing the accurate levels of gelsolin in human plasma and understanding its variance with age, race, gender and health status is a prerequisite. Upon establishing the accurate levels of pGSN in healthy individuals, this biomarker would predict the prognosis/disease progression in multiple health conditions and help in prioritizing the ones in-need of gelsolin replacement therapy.
Collapse
Affiliation(s)
- Nagesh Peddada
- Institute of Microbial Technology (CSIR), Chandigarh, India
| | | | | | | |
Collapse
|
48
|
Rahbarizadeh F, Ahmadvand D, Sharifzadeh Z. Nanobody; an old concept and new vehicle for immunotargeting. Immunol Invest 2011; 40:299-338. [PMID: 21244216 DOI: 10.3109/08820139.2010.542228] [Citation(s) in RCA: 85] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
The use of antibodies in cancer therapy has come a long way since the day Paul Ehrlich described the concept and Kohler and Milstein devised the hybridoma technology to bring this theory to reality. The synthesis of murine monoclonal antibodies (mAbs) was the first success in this field, leading to the invention of chimerization, the production of variable fragments (Fv) with the progression to domain antibodies (dAb) and later humanization technologies to maximize the clinical utility of murine mAbs. It was just by chance that dAbs were found to exist in ?heavy chain? immunoglobulins from Camelidae family and cartilaginous fish. These unique antibody fragments interact with antigen by virtue of only one single variable domain, referred to as VHH or nanobody. Several characteristics make nanobody use superior to the abovementioned antibodies. They are non-immunogenic and show high thermal and chemical stability. There are several reports of raising specific nanobodies against enzymes, haptens, pathogens, toxins and tumor markers, which are outlined in this paper. All these characteristics make them strong candidates as targeting agents for cancer therapy.
Collapse
Affiliation(s)
- Fatemeh Rahbarizadeh
- Department of Medical Biotechnology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran.
| | | | | |
Collapse
|
49
|
Intrabody Expression in Mammalian Cells. ANTIBODY EXPRESSION AND PRODUCTION 2011. [PMCID: PMC7120103 DOI: 10.1007/978-94-007-1257-7_9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 10/30/2022]
Abstract
The intracellular expression of antibodies or antibody fragments (intrabodies) in different compartments of mammalian cells allows to block or modulate the function of endogenous molecules. Intrabodies can alter protein folding, protein-protein, protein-DNA, protein-RNA interactions and protein modification. They can induce a phenotypic knockout and work as neutralizing agents by direct binding to the target antigen, by diverting its intracellular traffic or by inhibiting its association with binding partners. They have been largely employed as research tools and are emerging as therapeutic molecules for the treatment of human diseases as viral pathologies, cancer and misfolding diseases. The fast growing bio-market of recombinant antibodies provides intrabodies with enhanced binding specificity, stability and solubility, together with lower immunogenicity, for their use in therapy. This chapter describes the crucial aspects required to express intrabodies in different intracellular compartments of mammalian cells, their various modes of action and gives an update on the applications of intrabodies in human diseases.
Collapse
|
50
|
Direct injection of functional single-domain antibodies from E. coli into human cells. PLoS One 2010; 5:e15227. [PMID: 21170340 PMCID: PMC2999559 DOI: 10.1371/journal.pone.0015227] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2010] [Accepted: 11/01/2010] [Indexed: 12/31/2022] Open
Abstract
Intracellular proteins have a great potential as targets for therapeutic antibodies (Abs) but the plasma membrane prevents access to these antigens. Ab fragments and IgGs are selected and engineered in E. coli and this microorganism may be also an ideal vector for their intracellular delivery. In this work we demonstrate that single-domain Ab (sdAbs) can be engineered to be injected into human cells by E. coli bacteria carrying molecular syringes assembled by a type III protein secretion system (T3SS). The injected sdAbs accumulate in the cytoplasm of HeLa cells at levels ca. 105–106 molecules per cell and their functionality is shown by the isolation of sdAb-antigen complexes. Injection of sdAbs does not require bacterial invasion or the transfer of genetic material. These results are proof-of-principle for the capacity of E. coli bacteria to directly deliver intracellular sdAbs (intrabodies) into human cells for analytical and therapeutic purposes.
Collapse
|