1
|
Ptaszek AL, Kratzwald S, Sagan F, Migotti M, Sánchez-Murcia PA, Konrat R, Platzer G. From Weak Interactions to High Stability: Deciphering the Streptavidin-Biotin Interaction through NMR and Computational Analysis. J Phys Chem B 2025; 129:4917-4928. [PMID: 40356290 DOI: 10.1021/acs.jpcb.5c00155] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/15/2025]
Abstract
Understanding weak interactions in protein-ligand complexes is essential for advancing drug design. Here, we combine experimental and quantum mechanical approaches to study the streptavidin-biotin complex, one of the strongest interacting protein-ligand systems. Using a monomeric streptavidin mutant, we analyze 1H NMR chemical shift perturbations (CSPs) of biotin upon binding, identifying remarkable upfield shifts of up to -3.2 ppm. Quantum chemical calculations attribute these shifts primarily to aromatic ring currents, with additional contributions from charge transfer effects linked to weak interactions. The agreement between experimental and computed chemical shifts validated the X-ray structure as a reliable basis for detailed computational analyses. Energy decomposition analysis reveals that electrostatics dominate the biotin-streptavidin interaction, complemented by significant orbital and dispersion contributions. Notably, weak noncovalent interactions, such as CH···S, CH···π, and CH···HC contacts, driven by London dispersion forces, contribute ∼44% to the complex's stability.
Collapse
Affiliation(s)
- Aleksandra L Ptaszek
- Christian Doppler Laboratory for High-Content Structural Biology and Biotechnology, Department of Structural and Computational Biology, Max Perutz Laboratories, University of Vienna, Campus Vienna Biocenter 5, 1030 Vienna, Austria
- Laboratory of Computer-Aided Molecular Design, Division of Medicinal Chemistry, Otto-Loewi Research Center, Medical University of Graz, Neue Stiftintalstr. 6/III, A-8010 Graz, Austria
- Department of Structural and Computational Biology, Max Perutz Laboratories, University of Vienna, Campus Vienna Biocenter 5, 1030 Vienna, Austria
| | - Sarah Kratzwald
- Department of Structural and Computational Biology, Max Perutz Laboratories, University of Vienna, Campus Vienna Biocenter 5, 1030 Vienna, Austria
- CIC bioGUNE, Precision Medicine and Metabolism Lab, Bizkaia Science and Technology Park, Building 800, Derio (Bizkaia), Derio 48160, Spain
- MAG-LAB GmbH, Karl-Farkas-Gasse 22, 1030 Vienna, Austria
- Institute of Organic Chemistry, Faculty of Chemistry, University of Vienna, Währingerstr. 38, Vienna 1090, Austria
| | - Filip Sagan
- Faculty of Chemistry, Jagiellonian University, Gronostajowa 2, 30-387 Cracow, Poland
| | - Mario Migotti
- Department of Structural and Computational Biology, Max Perutz Laboratories, University of Vienna, Campus Vienna Biocenter 5, 1030 Vienna, Austria
| | - Pedro A Sánchez-Murcia
- Laboratory of Computer-Aided Molecular Design, Division of Medicinal Chemistry, Otto-Loewi Research Center, Medical University of Graz, Neue Stiftintalstr. 6/III, A-8010 Graz, Austria
- BioTechMed-Graz, Mozartgasse 12/II, A-8010 Graz, Austria
| | - Robert Konrat
- Christian Doppler Laboratory for High-Content Structural Biology and Biotechnology, Department of Structural and Computational Biology, Max Perutz Laboratories, University of Vienna, Campus Vienna Biocenter 5, 1030 Vienna, Austria
- Department of Structural and Computational Biology, Max Perutz Laboratories, University of Vienna, Campus Vienna Biocenter 5, 1030 Vienna, Austria
- MAG-LAB GmbH, Karl-Farkas-Gasse 22, 1030 Vienna, Austria
| | - Gerald Platzer
- Christian Doppler Laboratory for High-Content Structural Biology and Biotechnology, Department of Structural and Computational Biology, Max Perutz Laboratories, University of Vienna, Campus Vienna Biocenter 5, 1030 Vienna, Austria
- MAG-LAB GmbH, Karl-Farkas-Gasse 22, 1030 Vienna, Austria
| |
Collapse
|
2
|
Rhym LH, Manan RS, Koller A, Stephanie G, Anderson DG. Peptide-encoding mRNA barcodes for the high-throughput in vivo screening of libraries of lipid nanoparticles for mRNA delivery. Nat Biomed Eng 2023; 7:901-910. [PMID: 37127709 DOI: 10.1038/s41551-023-01030-4] [Citation(s) in RCA: 50] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2022] [Accepted: 03/26/2023] [Indexed: 05/03/2023]
Abstract
Developing safe and effective nanoparticles for the delivery of messenger RNA (mRNA) is slow and expensive, partly due to the lack of predictive power of in vitro screening methods and the low-throughput nature of in vivo screening. While DNA barcoding and batch analysis present methods for increasing in vivo screening throughput, they can also result in incomplete or misleading measures of efficacy. Here, we describe a high-throughput and accurate method for the screening of pooled nanoparticle formulations within the same animal. The method uses liquid chromatography with tandem mass spectrometry to detect peptide barcodes translated from mRNAs in nanoparticle-transfected cells. We show the method's applicability by evaluating a library of over 400 nanoparticle formulations with 384 unique ionizable lipids using only nine mice to optimize the formulation of a biodegradable lipid nanoparticle for mRNA delivery to the liver. Barcoding lipid nanoparticles with peptide-encoding mRNAs may facilitate the rapid development of nanoparticles for mRNA delivery to specific cells and tissues.
Collapse
Affiliation(s)
- Luke H Rhym
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Rajith S Manan
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Antonius Koller
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Georgina Stephanie
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Daniel G Anderson
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA.
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA.
| |
Collapse
|
3
|
Iida S, Kameda T. Dissociation Rate Calculation via Constant-Force Steered Molecular Dynamics Simulation. J Chem Inf Model 2023. [PMID: 37188657 DOI: 10.1021/acs.jcim.2c01529] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/17/2023]
Abstract
Steered molecular dynamics (SMD) simulations are used to study molecular dissociation events by applying a harmonic force to the molecules and pulling them at a constant velocity. Instead of constant-velocity pulling, we use a constant force: the constant-force SMD (CF-SMD) simulation. The CF-SMD simulation employs a constant force to reduce the activation barrier of molecular dissociation, thereby enhancing the dissociation event. Here, we present the capability of the CF-SMD simulation to estimate the dissociation time at equilibrium. We performed all-atom CF-SMD simulations for NaCl and protein-ligand systems, producing dissociation time at various forces. We extrapolated these values to the dissociation rate without a constant force using Bell's model or the Dudko-Hummer-Szabo model. We demonstrate that the CF-SMD simulations with the models predicted the dissociation time in equilibrium. A CF-SMD simulation is a powerful tool for estimating the dissociation rate in a direct and computationally efficient manner.
Collapse
Affiliation(s)
- Shinji Iida
- Artificial Intelligence Research Center, National Institute of Advanced Industrial Science and Technology (AIST), 2-4-7 Aomi, Koto-ku, Tokyo 135-0064, Japan
| | - Tomoshi Kameda
- Artificial Intelligence Research Center, National Institute of Advanced Industrial Science and Technology (AIST), 2-4-7 Aomi, Koto-ku, Tokyo 135-0064, Japan
| |
Collapse
|
4
|
Sarathy JP, Aldrich CC, Go ML, Dick T. PROTAC antibiotics: the time is now. Expert Opin Drug Discov 2023; 18:363-370. [PMID: 37027333 PMCID: PMC10540314 DOI: 10.1080/17460441.2023.2178413] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Accepted: 02/06/2023] [Indexed: 02/24/2023]
Abstract
INTRODUCTION Novel antibiotics are needed to keep antibiotic resistance at bay and to improve treatment of the many drug-susceptible infections for which current therapies achieve poor cure rates. While revolutionizing human therapeutics, the concept of targeted protein degradation (TPD) by bifunctional proteolysis targeting chimeras (PROTACs) has not yet been applied to the discovery of antibiotics. A major obstacle precluding successful translation of this strategy to antibiotic development is that bacteria lack the E3 ligase-proteasome system exploited by human PROTACs to facilitate target degradation. AREAS COVERED The authors describe the serendipitous discovery of the first monofunctional target-degrading antibiotic pyrazinamide, supporting TPD as a viable and novel approach in antibiotic discovery. They then discuss the rational design, mechanism, and activity of the first bifunctional antibacterial target degrader BacPROTAC, enabling a generalizable approach to TPD in bacteria. EXPERT OPINION BacPROTACs demonstrate that linking a target directly to a bacterial protease complex can promote target degradation. BacPROTACs successfully bypass the 'middleman' E3 ligase, providing an entry strategy for the generation of antibacterial PROTACs. We speculate that antibacterial PROTACs will not only expand the target space but may also improve treatment by allowing dosage reduction, stronger bactericidal activity and activity against drug-tolerant 'persisters.'
Collapse
Affiliation(s)
| | - Courtney C. Aldrich
- Department of Medicinal Chemistry, College of Pharmacy, University of Minnesota, Minneapolis, MN, USA
| | - Mei-Lin Go
- Department of Chemistry and Department of Pharmacy, National University of Singapore, Singapore
| | - Thomas Dick
- Center for Discovery and Innovation, Hackensack Meridian Health, Nutley, NJ, USA
- Department of Medical Sciences, Hackensack Meridian School of Medicine, Nutley, NJ, USA
- Department of Microbiology and Immunology, Georgetown University, Washington, DC, USA
| |
Collapse
|
5
|
A modular vaccine platform enabled by decoration of bacterial outer membrane vesicles with biotinylated antigens. Nat Commun 2023; 14:464. [PMID: 36709333 PMCID: PMC9883832 DOI: 10.1038/s41467-023-36101-2] [Citation(s) in RCA: 36] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2021] [Accepted: 01/15/2023] [Indexed: 01/29/2023] Open
Abstract
Engineered outer membrane vesicles (OMVs) derived from Gram-negative bacteria are a promising technology for the creation of non-infectious, nanoparticle vaccines against diverse pathogens. However, antigen display on OMVs can be difficult to control and highly variable due to bottlenecks in protein expression and localization to the outer membrane of the host cell, especially for bulky and/or complex antigens. Here, we describe a universal approach for avidin-based vaccine antigen crosslinking (AvidVax) whereby biotinylated antigens are linked to the exterior of OMVs whose surfaces are remodeled with multiple copies of a synthetic antigen-binding protein (SNAP) comprised of an outer membrane scaffold protein fused to a biotin-binding protein. We show that SNAP-OMVs can be readily decorated with a molecularly diverse array of biotinylated subunit antigens, including globular and membrane proteins, glycans and glycoconjugates, haptens, lipids, and short peptides. When the resulting OMV formulations are injected in mice, strong antigen-specific antibody responses are observed that depend on the physical coupling between the antigen and SNAP-OMV delivery vehicle. Overall, these results demonstrate AvidVax as a modular platform that enables rapid and simplified assembly of antigen-studded OMVs for application as vaccines against pathogenic threats.
Collapse
|
6
|
Boonyakida J, Khoris IM, Nasrin F, Park EY. Improvement of Modular Protein Display Efficiency in SpyTag-Implemented Norovirus-like Particles. Biomacromolecules 2023; 24:308-318. [PMID: 36475654 DOI: 10.1021/acs.biomac.2c01150] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Genetic fusion and chemical conjugation are the most common approaches for displaying a foreign protein on the surface of virus-like particles (VLPs); however, these methods may negatively affect the formation and stability of VLPs. Here, we aimed to develop a modular display platform for protein decoration on norovirus-like particles (NoV-LPs) by combining the NoV-LP scaffold with the SpyTag/SpyCatcher bioconjugation system, as the NoV-LP is an attractive protein nanoparticle to carry foreign proteins for various applications. The SpyTagged-NoV-LPs were prepared by introducing SpyTag peptide into the C-terminus of the norovirus VP1 protein. To increase surface exposure of the SpyTag peptide on the NoV-LPs, two or three repeated extension linkers (EAAAK) were inserted between the SpyTag peptide and VP1 protein. Fluorescence proteins, EGFP and mCherry, were fused to SpyCatcher and employed as SpyTag conjugation partners. These VP1-SpyTag variants and SpyCatcher-fused EGFP and mCherry were separately expressed in silkworm fat bodies and purified. This study reveals that adding an extension linker did not disrupt the VLP formation; instead, it increased the particle size by 4-6 nm. The conjugation efficiency of the VP1-SpyTag variants with the extended linker improved from ∼15-35 to ∼50-63% based on the densitometric analysis, while it was up to 77% based on an optical quantification of EGFP and mCherry. Results indicate that the linker causes the SpyTag peptides to be positioned further away from the C-termini of VP1 and potentially increases the exposure of the SpyTag to the outer surface of the NoV-LPs, allowing more SpyTag/SpyCatcher complex formation on the VLP surface. Our study provides a strategy for enhancing the conjugation efficiency of NoV-LP and demonstrates the platform's utility for developing vaccines or functional nanoparticles.
Collapse
Affiliation(s)
- Jirayu Boonyakida
- Research Institute of Green Science and Technology, Shizuoka University, 836 Ohya, Suruga ward, Shizuoka422-8529, Japan
| | - Indra Memdi Khoris
- Research Institute of Green Science and Technology, Shizuoka University, 836 Ohya, Suruga ward, Shizuoka422-8529, Japan
| | - Fahmida Nasrin
- Research Institute of Green Science and Technology, Shizuoka University, 836 Ohya, Suruga ward, Shizuoka422-8529, Japan
| | - Enoch Y Park
- Research Institute of Green Science and Technology, Shizuoka University, 836 Ohya, Suruga ward, Shizuoka422-8529, Japan
| |
Collapse
|
7
|
Morreale FE, Kleine S, Leodolter J, Junker S, Hoi DM, Ovchinnikov S, Okun A, Kley J, Kurzbauer R, Junk L, Guha S, Podlesainski D, Kazmaier U, Boehmelt G, Weinstabl H, Rumpel K, Schmiedel VM, Hartl M, Haselbach D, Meinhart A, Kaiser M, Clausen T. BacPROTACs mediate targeted protein degradation in bacteria. Cell 2022; 185:2338-2353.e18. [PMID: 35662409 PMCID: PMC9240326 DOI: 10.1016/j.cell.2022.05.009] [Citation(s) in RCA: 86] [Impact Index Per Article: 28.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2020] [Revised: 03/15/2022] [Accepted: 05/10/2022] [Indexed: 12/14/2022]
Abstract
Hijacking the cellular protein degradation system offers unique opportunities for drug discovery, as exemplified by proteolysis-targeting chimeras. Despite their great promise for medical chemistry, so far, it has not been possible to reprogram the bacterial degradation machinery to interfere with microbial infections. Here, we develop small-molecule degraders, so-called BacPROTACs, that bind to the substrate receptor of the ClpC:ClpP protease, priming neo-substrates for degradation. In addition to their targeting function, BacPROTACs activate ClpC, transforming the resting unfoldase into its functional state. The induced higher-order oligomer was visualized by cryo-EM analysis, providing a structural snapshot of activated ClpC unfolding a protein substrate. Finally, drug susceptibility and degradation assays performed in mycobacteria demonstrate in vivo activity of BacPROTACs, allowing selective targeting of endogenous proteins via fusion to an established degron. In addition to guiding antibiotic discovery, the BacPROTAC technology presents a versatile research tool enabling the inducible degradation of bacterial proteins.
Collapse
Affiliation(s)
- Francesca E Morreale
- Research Institute of Molecular Pathology, Vienna Biocenter, 1030 Vienna, Austria
| | - Stefan Kleine
- University of Duisburg-Essen, Center of Medical Biotechnology, Faculty of Biology, 45141 Essen, Germany
| | - Julia Leodolter
- Research Institute of Molecular Pathology, Vienna Biocenter, 1030 Vienna, Austria
| | - Sabryna Junker
- Research Institute of Molecular Pathology, Vienna Biocenter, 1030 Vienna, Austria
| | - David M Hoi
- Research Institute of Molecular Pathology, Vienna Biocenter, 1030 Vienna, Austria
| | - Stepan Ovchinnikov
- Research Institute of Molecular Pathology, Vienna Biocenter, 1030 Vienna, Austria
| | - Anastasia Okun
- Research Institute of Molecular Pathology, Vienna Biocenter, 1030 Vienna, Austria
| | - Juliane Kley
- Research Institute of Molecular Pathology, Vienna Biocenter, 1030 Vienna, Austria
| | - Robert Kurzbauer
- Research Institute of Molecular Pathology, Vienna Biocenter, 1030 Vienna, Austria
| | - Lukas Junk
- Saarland University, Organic Chemistry I, 66123 Saarbrücken, Germany
| | - Somraj Guha
- Saarland University, Organic Chemistry I, 66123 Saarbrücken, Germany
| | - David Podlesainski
- University of Duisburg-Essen, Center of Medical Biotechnology, Faculty of Biology, 45141 Essen, Germany
| | - Uli Kazmaier
- Saarland University, Organic Chemistry I, 66123 Saarbrücken, Germany
| | - Guido Boehmelt
- Boehringer Ingelheim RCV GmbH & Co KG, 1120 Vienna, Austria
| | | | - Klaus Rumpel
- Boehringer Ingelheim RCV GmbH & Co KG, 1120 Vienna, Austria
| | | | - Markus Hartl
- Max Perutz Laboratories, Vienna Biocenter, 1030 Vienna, Austria
| | - David Haselbach
- Research Institute of Molecular Pathology, Vienna Biocenter, 1030 Vienna, Austria
| | - Anton Meinhart
- Research Institute of Molecular Pathology, Vienna Biocenter, 1030 Vienna, Austria
| | - Markus Kaiser
- University of Duisburg-Essen, Center of Medical Biotechnology, Faculty of Biology, 45141 Essen, Germany.
| | - Tim Clausen
- Research Institute of Molecular Pathology, Vienna Biocenter, 1030 Vienna, Austria; Medical University of Vienna, 1030 Vienna, Austria.
| |
Collapse
|
8
|
Toledo A, Letellier M, Bimbi G, Tessier B, Daburon S, Favereaux A, Chamma I, Vennekens K, Vanderlinden J, Sainlos M, de Wit J, Choquet D, Thoumine O. MDGAs are fast-diffusing molecules that delay excitatory synapse development by altering neuroligin behavior. eLife 2022; 11:75233. [PMID: 35532105 PMCID: PMC9084894 DOI: 10.7554/elife.75233] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2021] [Accepted: 04/11/2022] [Indexed: 12/28/2022] Open
Abstract
MDGA molecules can bind neuroligins and interfere with trans-synaptic interactions to neurexins, thereby impairing synapse development. However, the subcellular localization and dynamics of MDGAs, or their specific action mode in neurons remain unclear. Here, surface immunostaining of endogenous MDGAs and single molecule tracking of recombinant MDGAs in dissociated hippocampal neurons reveal that MDGAs are homogeneously distributed and exhibit fast membrane diffusion, with a small reduction in mobility across neuronal maturation. Knocking-down/out MDGAs using shRNAs and CRISPR/Cas9 strategies increases the density of excitatory synapses, the membrane confinement of neuroligin-1, and the phosphotyrosine level of neuroligins associated with excitatory post-synaptic differentiation. Finally, MDGA silencing reduces the mobility of AMPA receptors, increases the frequency of miniature EPSCs (but not IPSCs), and selectively enhances evoked AMPA-receptor-mediated EPSCs in CA1 pyramidal neurons. Overall, our results support a mechanism by which interactions between MDGAs and neuroligin-1 delays the assembly of functional excitatory synapses containing AMPA receptors.
Collapse
Affiliation(s)
- Andrea Toledo
- University of Bordeaux, CNRS UMR 5297, Interdisciplinary Institute for Neuroscience
| | - Mathieu Letellier
- University of Bordeaux, CNRS UMR 5297, Interdisciplinary Institute for Neuroscience
| | - Giorgia Bimbi
- University of Bordeaux, CNRS UMR 5297, Interdisciplinary Institute for Neuroscience
| | - Béatrice Tessier
- University of Bordeaux, CNRS UMR 5297, Interdisciplinary Institute for Neuroscience
| | - Sophie Daburon
- University of Bordeaux, CNRS UMR 5297, Interdisciplinary Institute for Neuroscience
| | - Alexandre Favereaux
- University of Bordeaux, CNRS UMR 5297, Interdisciplinary Institute for Neuroscience
| | - Ingrid Chamma
- University of Bordeaux, CNRS UMR 5297, Interdisciplinary Institute for Neuroscience
| | - Kristel Vennekens
- VIB Center for Brain & Disease Research and KU Leuven, Department of Neurosciences, Leuven Brain Institute
| | - Jeroen Vanderlinden
- VIB Center for Brain & Disease Research and KU Leuven, Department of Neurosciences, Leuven Brain Institute
| | - Matthieu Sainlos
- University of Bordeaux, CNRS UMR 5297, Interdisciplinary Institute for Neuroscience
| | - Joris de Wit
- VIB Center for Brain & Disease Research and KU Leuven, Department of Neurosciences, Leuven Brain Institute
| | - Daniel Choquet
- University of Bordeaux, CNRS UMR 5297, Interdisciplinary Institute for Neuroscience
- University of Bordeaux, CNRS UAR 3420, INSERM, Bordeaux Imaging Center
| | - Olivier Thoumine
- University of Bordeaux, CNRS UMR 5297, Interdisciplinary Institute for Neuroscience
| |
Collapse
|
9
|
Lagardère M, Drouet A, Sainlos M, Thoumine O. High-Resolution Fluorescence Imaging Combined With Computer Simulations to Quantitate Surface Dynamics and Nanoscale Organization of Neuroligin-1 at Synapses. Front Synaptic Neurosci 2022; 14:835427. [PMID: 35546899 PMCID: PMC9083120 DOI: 10.3389/fnsyn.2022.835427] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Accepted: 03/09/2022] [Indexed: 11/13/2022] Open
Abstract
Neuroligins (NLGNs) form a family of cell adhesion molecules implicated in synapse development, but the mechanisms that retain these proteins at synapses are still incompletely understood. Recent studies indicate that surface-associated NLGN1 is diffusionally trapped at synapses, where it interacts with quasi-static scaffolding elements of the post-synaptic density. Whereas single molecule tracking reveals rapid diffusion and transient immobilization of NLGN1 at synapses within seconds, fluorescence recovery after photobleaching experiments indicate instead a long-term turnover of NLGN1 at synapse, in the hour time range. To gain insight into the mechanisms supporting NLGN1 anchorage at post-synapses and try to reconcile those experimental paradigms, we quantitatively analyzed here live-cell and super-resolution imaging experiments performed on NLGN1 using a newly released simulator of membrane protein dynamics for fluorescence microscopy, FluoSim. Based on a small set of parameters including diffusion coefficients, binding constants, and photophysical rates, the framework describes fairly well the dynamic behavior of extra-synaptic and synaptic NLGN1 over both short and long time ranges, and provides an estimate of NLGN1 copy numbers in post-synaptic densities at steady-state (around 50 dimers). One striking result is that the residence time of NLGN1 at synapses is much longer than what can be expected from extracellular interactions with pre-synaptic neurexins only, suggesting that NLGN1 is stabilized at synapses through multivalent interactions with intracellular post-synaptic scaffolding proteins.
Collapse
Affiliation(s)
| | | | | | - Olivier Thoumine
- CNRS, Interdisciplinary Institute for Neuroscience, IINS, UMR 5297, University of Bordeaux, Bordeaux, France
| |
Collapse
|
10
|
Santos MS, Liu H, Schittny V, Vanella R, Nash MA. Correlating single-molecule rupture mechanics with cell population adhesion by yeast display. BIOPHYSICAL REPORTS 2022; 2:None. [PMID: 35284851 PMCID: PMC8904261 DOI: 10.1016/j.bpr.2021.100035] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/13/2021] [Accepted: 11/22/2021] [Indexed: 11/20/2022]
Affiliation(s)
- Mariana Sá Santos
- Institute for Physical Chemistry, Department of Chemistry, University of Basel, Basel, Switzerland
- Department of Biosystems Science and Engineering, ETH Zurich, Basel, Switzerland
- Systems Biology PhD program, Life Science Zurich Graduate School, Zurich, Switzerland
| | - Haipei Liu
- Institute for Physical Chemistry, Department of Chemistry, University of Basel, Basel, Switzerland
- Department of Biosystems Science and Engineering, ETH Zurich, Basel, Switzerland
| | - Valentin Schittny
- Institute for Physical Chemistry, Department of Chemistry, University of Basel, Basel, Switzerland
- Department of Biosystems Science and Engineering, ETH Zurich, Basel, Switzerland
| | - Rosario Vanella
- Institute for Physical Chemistry, Department of Chemistry, University of Basel, Basel, Switzerland
- Department of Biosystems Science and Engineering, ETH Zurich, Basel, Switzerland
| | - Michael A. Nash
- Institute for Physical Chemistry, Department of Chemistry, University of Basel, Basel, Switzerland
- Department of Biosystems Science and Engineering, ETH Zurich, Basel, Switzerland
- Corresponding author
| |
Collapse
|
11
|
Pelus A, Bordes G, Barbe S, Bouchiba Y, Burnard C, Cortés J, Enjalbert B, Esque J, Estaña A, Fauré R, Henras AK, Heux S, Le Men C, Millard P, Nouaille S, Pérochon J, Toanen M, Truan G, Verdier A, Wagner C, Romeo Y, Montanier CY. A tripartite carbohydrate-binding module to functionalize cellulose nanocrystals. Biomater Sci 2021; 9:7444-7455. [PMID: 34647546 DOI: 10.1039/d1bm01156a] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
The development of protein and microorganism engineering have led to rising expectations of biotechnology in the design of emerging biomaterials, putatively of high interest to reduce our dependence on fossil carbon resources. In this way, cellulose, a renewable carbon based polysaccharide and derived products, displays unique properties used in many industrial applications. Although the functionalization of cellulose is common, it is however limited in terms of number and type of functions. In this work, a Carbohydrate-Binding Module (CBM) was used as a central core to provide a versatile strategy to bring a large diversity of functions to cellulose surfaces. CBM3a from Clostridium thermocellum, which has a high affinity for crystalline cellulose, was flanked through linkers with a streptavidin domain and an azide group introduced through a non-canonical amino acid. Each of these two extra domains was effectively produced and functionalized with a variety of biological and chemical molecules. Structural properties of the resulting tripartite chimeric protein were investigated using molecular modelling approaches, and its potential for the multi-functionalization of cellulose was confirmed experimentally. As a proof of concept, we show that cellulose can be labelled with a fluorescent version of the tripartite protein grafted to magnetic beads and captured using a magnet.
Collapse
Affiliation(s)
- Angeline Pelus
- TBI, Université de Toulouse, CNRS, INRAE, INSA, Toulouse, France.
| | - Gaëlle Bordes
- Molecular, Cellular and Developmental biology department (MCD), Centre de Biologie Integrative (CBI), Université de Toulouse, CNRS, UPS, 31062 Toulouse, France.
| | - Sophie Barbe
- TBI, Université de Toulouse, CNRS, INRAE, INSA, Toulouse, France.
| | - Younes Bouchiba
- TBI, Université de Toulouse, CNRS, INRAE, INSA, Toulouse, France.
| | - Callum Burnard
- TBI, Université de Toulouse, CNRS, INRAE, INSA, Toulouse, France.
| | - Juan Cortés
- LAAS-CNRS, Université de Toulouse, CNRS, Toulouse, France
| | - Brice Enjalbert
- TBI, Université de Toulouse, CNRS, INRAE, INSA, Toulouse, France.
| | - Jeremy Esque
- TBI, Université de Toulouse, CNRS, INRAE, INSA, Toulouse, France.
| | | | - Régis Fauré
- TBI, Université de Toulouse, CNRS, INRAE, INSA, Toulouse, France.
| | - Anthony K Henras
- Molecular, Cellular and Developmental biology department (MCD), Centre de Biologie Integrative (CBI), Université de Toulouse, CNRS, UPS, 31062 Toulouse, France.
| | - Stéphanie Heux
- TBI, Université de Toulouse, CNRS, INRAE, INSA, Toulouse, France.
| | - Claude Le Men
- TBI, Université de Toulouse, CNRS, INRAE, INSA, Toulouse, France.
| | - Pierre Millard
- TBI, Université de Toulouse, CNRS, INRAE, INSA, Toulouse, France.
| | | | - Julien Pérochon
- TBI, Université de Toulouse, CNRS, INRAE, INSA, Toulouse, France.
| | - Marion Toanen
- TBI, Université de Toulouse, CNRS, INRAE, INSA, Toulouse, France.
| | - Gilles Truan
- TBI, Université de Toulouse, CNRS, INRAE, INSA, Toulouse, France.
| | - Amandine Verdier
- TBI, Université de Toulouse, CNRS, INRAE, INSA, Toulouse, France.
| | - Camille Wagner
- TBI, Université de Toulouse, CNRS, INRAE, INSA, Toulouse, France.
| | - Yves Romeo
- Molecular, Cellular and Developmental biology department (MCD), Centre de Biologie Integrative (CBI), Université de Toulouse, CNRS, UPS, 31062 Toulouse, France.
| | | |
Collapse
|
12
|
Fredsgaard L, Goksøyr L, Thrane S, Aves KL, Theander TG, Sander AF. Head-to-Head Comparison of Modular Vaccines Developed Using Different Capsid Virus-Like Particle Backbones and Antigen Conjugation Systems. Vaccines (Basel) 2021; 9:vaccines9060539. [PMID: 34063871 PMCID: PMC8224050 DOI: 10.3390/vaccines9060539] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2021] [Revised: 05/12/2021] [Accepted: 05/17/2021] [Indexed: 01/19/2023] Open
Abstract
Capsid virus-like particles (cVLPs) are used as molecular scaffolds to increase the immunogenicity of displayed antigens. Modular platforms have been developed whereby antigens are attached to the surface of pre-assembled cVLPs. However, it remains unknown to what extent the employed cVLP backbone and conjugation system may influence the immune response elicited against the displayed antigen. Here, we performed a head-to-head comparison of antigen-specific IgG responses elicited by modular cVLP-vaccines differing by their employed cVLP backbone or conjugation system, respectively. Covalent antigen conjugation (i.e., employing the SpyTag/SpyCatcher system) resulted in significantly higher antigen-specific IgG titers compared to when using affinity-based conjugation (i.e., using biotin/streptavidin). The cVLP backbone also influenced the antigen-specific IgG response. Specifically, vaccines based on the bacteriophage AP205 cVLP elicited significantly higher antigen-specific IgG compared to corresponding vaccines using the human papillomavirus major capsid protein (HPV L1) cVLP. In addition, the AP205 cVLP platform mediated induction of antigen-specific IgG with a different subclass profile (i.e., higher IgG2a and IgG2b) compared to HPV L1 cVLP. These results demonstrate that the cVLP backbone and conjugation system can individually affect the IgG response elicited against a displayed antigen. These data will aid the understanding and process of tailoring modular cVLP vaccines to achieve improved immune responses.
Collapse
Affiliation(s)
- Laurits Fredsgaard
- Centre for Medical Parasitology, Department of Immunology and Microbiology, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark; (L.F.); (L.G.); (K.-L.A.); (T.G.T.)
| | - Louise Goksøyr
- Centre for Medical Parasitology, Department of Immunology and Microbiology, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark; (L.F.); (L.G.); (K.-L.A.); (T.G.T.)
- AdaptVac Aps, 2970 Hørsholm, Denmark;
| | | | - Kara-Lee Aves
- Centre for Medical Parasitology, Department of Immunology and Microbiology, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark; (L.F.); (L.G.); (K.-L.A.); (T.G.T.)
| | - Thor G. Theander
- Centre for Medical Parasitology, Department of Immunology and Microbiology, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark; (L.F.); (L.G.); (K.-L.A.); (T.G.T.)
| | - Adam F. Sander
- Centre for Medical Parasitology, Department of Immunology and Microbiology, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark; (L.F.); (L.G.); (K.-L.A.); (T.G.T.)
- AdaptVac Aps, 2970 Hørsholm, Denmark;
- Correspondence:
| |
Collapse
|
13
|
Jeon BJ, Kim S, Kim MS, Lee JH, Kim BS, Hwang KY. Insights into the structure of mature streptavidin C1 from Streptomyces cinnamonensis reveal the self-binding of the extension C-terminal peptide to biotin-binding sites. IUCRJ 2021; 8:168-177. [PMID: 33708394 PMCID: PMC7924230 DOI: 10.1107/s2052252520015675] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/15/2020] [Accepted: 11/30/2020] [Indexed: 06/12/2023]
Abstract
The members of the avidin protein family are well known for their high affinity towards d-biotin and their structural stability. These properties make avidins a valuable tool for various biotechnological applications. In the present study, two avidin-like biotin-binding proteins (named streptavidin C1 and C2) from Streptomyces cinnamonensis were newly identified while exploring antifungal proteins against Fusarium oxysporum f. sp. cucumerinum. Streptavidin C1 reveals a low correlation (a sequence identity of approximately 64%) with all known streptavidins, whereas streptavidin C2 shares a sequence identity of approximately 94% with other streptavidins. Here, the crystal structures of streptavidin C1 in the mature form and in complex with biotin at 2.1 and 2.5 Å resolution, respectively, were assessed. The overall structures present similar tetrameric features with D 2 symmetry to other (strept)avidin structures. Interestingly, the long C-terminal region comprises a short α-helix (C-Lid; residues 169-179) and an extension C-terminal peptide (ECP; residues 180-191) which stretches into the biotin-binding sites of the same monomer. This ECP sequence (-180VTSANPPAS188-) is a newly defined biotin-binding site, which reduces the ability to bind to (strept)avidin family proteins. The novel streptavidin C1 could help in the development of an engineered tetrameric streptavidin with reduced biotin-binding capacity as well as other biomaterial tools.
Collapse
Affiliation(s)
- Byeong Jun Jeon
- Department of Plant Biotechnology, School of Life Sciences and Biotechnology for BK21 PLUS, Institute of Life Science and Natural Resources, Korea University, 145 Anam-ro, Seongbuk-gu, Seoul 02841, South Korea
| | - Sulhee Kim
- Department of Biotechnology, School of Life Sciences and Biotechnology for BK21 PLUS, Institute of Life Science and Natural Resources, Korea University, 145 Anam-ro, Seongbuk-gu, Seoul 02841, South Korea
| | - Min-Seok Kim
- Department of Biotechnology, School of Life Sciences and Biotechnology for BK21 PLUS, Institute of Life Science and Natural Resources, Korea University, 145 Anam-ro, Seongbuk-gu, Seoul 02841, South Korea
| | - Ji-Ho Lee
- Department of Biotechnology, School of Life Sciences and Biotechnology for BK21 PLUS, Institute of Life Science and Natural Resources, Korea University, 145 Anam-ro, Seongbuk-gu, Seoul 02841, South Korea
| | - Beom Seok Kim
- Department of Plant Biotechnology, School of Life Sciences and Biotechnology for BK21 PLUS, Institute of Life Science and Natural Resources, Korea University, 145 Anam-ro, Seongbuk-gu, Seoul 02841, South Korea
| | - Kwang Yeon Hwang
- Department of Biotechnology, School of Life Sciences and Biotechnology for BK21 PLUS, Institute of Life Science and Natural Resources, Korea University, 145 Anam-ro, Seongbuk-gu, Seoul 02841, South Korea
| |
Collapse
|
14
|
Abstract
The complex of the small molecule biotin and the homotetrameric protein streptavidin is key to a broad range of biotechnological applications. Therefore, the behavior of this extraordinarily high-affinity interaction under mechanical force is intensively studied by single-molecule force spectroscopy. Recently, steered molecular dynamics simulations have identified a low force pathway for the dissociation of biotin from streptavidin, which involves partial unfolding of the N-terminal β-sheet structure of monovalent streptavidin's functional subunit. Based on these results, we now introduced two mutations (T18C,A33C) in the functional subunit of monovalent streptavidin to establish a switchable connection (disulfide bridge) between the first two β-strands to prevent this unfolding. In atomic force microscopy-based single-molecule force spectroscopy experiments, we observed unbinding forces of about 350 pN (at a force-loading rate of 10 nN s-1) for pulling a single biotin out of an N-terminally anchored monovalent streptavidin binding pocket - about 1.5-fold higher compared with what has been reported for N-terminal force loading of native monovalent streptavidin. Upon addition of a reducing agent, the unbinding forces dropped back to 200 pN, as the disulfide bridge was destroyed. Switching from reducing to oxidizing buffer conditions, the inverse effect was observed. Our work illustrates how the mechanics of a receptor-ligand system can be tuned by engineering the receptor protein far off the ligand-binding pocket.
Collapse
Affiliation(s)
- Leonard C Schendel
- Lehrstuhl für Angewandte Physik and Center for NanoScience, Ludwig-Maximilians-Universität München, Amalienstr. 54, 80799 Munich, Germany.
| | | | | |
Collapse
|
15
|
Paloni JM, Dong XH, Olsen BD. Protein-Polymer Block Copolymer Thin Films for Highly Sensitive Detection of Small Proteins in Biological Fluids. ACS Sens 2019; 4:2869-2878. [PMID: 31702912 DOI: 10.1021/acssensors.9b01020] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
In nearly all biosensors, sensitivity is greatly reduced for measurements conducted in biological matrices due to nonspecific binding from off-target molecules. One method to overcome this issue is to design a sensor that enables selective size-based uptake of proteins. Herein, a protein-polymer conjugate thin-film biosensor is fabricated that self-assembles into lamellae containing alternating domains of protein and polymer. Analyte is captured in protein regions while polymer domains restrict diffusion of large molecules. Device sensitivity and size-based exclusion properties are probed using two analytes: streptavidin (SA, 52.8 kDa) and monomeric streptavidin (mSA2, 15.6 kDa). Tuning domain spacing by adjusting polymer molecular weight allows the design of films that relatively freely uptake mSA2 and largely restrict SA diffusion. Furthermore, when detecting the smaller mSA2, no reduction in the limit of detection (LOD) is observed when transitioning from detection in the buffer to detection in biological fluids. As a result, LOD measured in fluid samples is reduced by 2 orders of magnitude compared to a traditional surface-immobilized protein monolayer.
Collapse
Affiliation(s)
- Justin M. Paloni
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, United States
| | - Xue-Hui Dong
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, United States
| | - Bradley D. Olsen
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, United States
| |
Collapse
|
16
|
Recent advances in the engineering and application of streptavidin-like molecules. Appl Microbiol Biotechnol 2019; 103:7355-7365. [DOI: 10.1007/s00253-019-10036-5] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2019] [Revised: 07/12/2019] [Accepted: 07/15/2019] [Indexed: 01/24/2023]
|
17
|
Avraham O, Bayer EA, Livnah O. Crystal structure of afifavidin reveals common features of molecular assemblage in the bacterial dimeric avidins. FEBS J 2018; 285:4617-4630. [DOI: 10.1111/febs.14685] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2018] [Revised: 10/10/2018] [Accepted: 10/24/2018] [Indexed: 01/05/2023]
Affiliation(s)
- Orly Avraham
- Department of Biological Chemistry The Alexander Silverman Institute of Life Sciences The Wolfson Centre for Applied Structural Biology The Hebrew University of Jerusalem Israel
| | - Edward A. Bayer
- Department of Biological Chemistry The Weizmann Institute of Science Rehovot Israel
| | - Oded Livnah
- Department of Biological Chemistry The Alexander Silverman Institute of Life Sciences The Wolfson Centre for Applied Structural Biology The Hebrew University of Jerusalem Israel
| |
Collapse
|
18
|
Yang X, Wei J, Wang Y, Yang C, Zhao S, Li C, Dong Y, Bai K, Li Y, Teng H, Wang D, Lyu N, Li J, Chang X, Ning X, Ouyang Q, Zhang Y, Qian L. A Genetically Encoded Protein Polymer for Uranyl Binding and Extraction Based on the SpyTag-SpyCatcher Chemistry. ACS Synth Biol 2018; 7:2331-2339. [PMID: 30261140 DOI: 10.1021/acssynbio.8b00223] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
A defining goal of synthetic biology is to develop biomaterials with superior performance and versatility. Here we introduce a purely genetically encoded and self-assembling biopolymer based on the SpyTag-SpyCatcher chemistry. We show the application of this polymer for highly efficient uranyl binding and extraction from aqueous solutions, by embedding two functional modules-the superuranyl binding protein and the monomeric streptavidin-to the polymer via genetic fusion. We further provide a modeling strategy for predicting the polymer's physical properties, and experimentally demonstrate the autosecretion of component monomers from bacterial cells. The potential of multifunctionalization, in conjunction with the genetic design and production pipeline, underscores the advantage of the SpyTag-SpyCatcher biopolymers for applications beyond trace metal enrichment and environmental remediation.
Collapse
Affiliation(s)
- Xiaoyu Yang
- Peking University Team for the International Genetically Engineered Machine Competition (iGEM), Beijing, 100871, China
| | - Jingyi Wei
- Peking University Team for the International Genetically Engineered Machine Competition (iGEM), Beijing, 100871, China
| | - Yuqing Wang
- Peking University Team for the International Genetically Engineered Machine Competition (iGEM), Beijing, 100871, China
| | - Changru Yang
- Peking University Team for the International Genetically Engineered Machine Competition (iGEM), Beijing, 100871, China
| | - Shijun Zhao
- Peking University Team for the International Genetically Engineered Machine Competition (iGEM), Beijing, 100871, China
| | - Cheng Li
- Peking University Team for the International Genetically Engineered Machine Competition (iGEM), Beijing, 100871, China
| | - Yiming Dong
- Peking University Team for the International Genetically Engineered Machine Competition (iGEM), Beijing, 100871, China
- Center for Quantitative Biology and Peking-Tsinghua Joint Center for Life Sciences, Peking University, Beijing, 100871, China
| | - Ke Bai
- Peking University Team for the International Genetically Engineered Machine Competition (iGEM), Beijing, 100871, China
| | - Yuexuan Li
- Peking University Team for the International Genetically Engineered Machine Competition (iGEM), Beijing, 100871, China
| | - Huaiyuan Teng
- Peking University Team for the International Genetically Engineered Machine Competition (iGEM), Beijing, 100871, China
| | - Dingyu Wang
- Peking University Team for the International Genetically Engineered Machine Competition (iGEM), Beijing, 100871, China
| | - Nayun Lyu
- Peking University Team for the International Genetically Engineered Machine Competition (iGEM), Beijing, 100871, China
| | - Jiamian Li
- Peking University Team for the International Genetically Engineered Machine Competition (iGEM), Beijing, 100871, China
| | - Xuyao Chang
- Peking University Team for the International Genetically Engineered Machine Competition (iGEM), Beijing, 100871, China
| | - Xin Ning
- Peking University Team for the International Genetically Engineered Machine Competition (iGEM), Beijing, 100871, China
| | - Qi Ouyang
- Center for Quantitative Biology and Peking-Tsinghua Joint Center for Life Sciences, Peking University, Beijing, 100871, China
- The State Key Laboratory for Artificial Microstructures and Mesoscopic Physics, School of Physics, Peking University, Beijing, 100871, China
| | - Yihao Zhang
- Peking University Team for the International Genetically Engineered Machine Competition (iGEM), Beijing, 100871, China
- Center for Quantitative Biology and Peking-Tsinghua Joint Center for Life Sciences, Peking University, Beijing, 100871, China
| | - Long Qian
- Center for Quantitative Biology and Peking-Tsinghua Joint Center for Life Sciences, Peking University, Beijing, 100871, China
| |
Collapse
|
19
|
Versatile targeting system for lentiviral vectors involving biotinylated targeting molecules. Virology 2018; 525:170-181. [PMID: 30290312 DOI: 10.1016/j.virol.2018.09.017] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2018] [Revised: 09/21/2018] [Accepted: 09/22/2018] [Indexed: 12/11/2022]
Abstract
Conjugating certain types of lentiviral vectors with targeting ligands can redirect the vectors to specifically transduce desired cell types. However, extensive genetic and/or biochemical manipulations are required for conjugation, which hinders applications for targeting lentiviral vectors for broader research fields. We developed envelope proteins fused with biotin-binding molecules to conjugate the pseudotyped vectors with biotinylated targeting molecules by simply mixing them. The envelope proteins fused with the monomeric, but not tetrameric, biotin-binding molecules can pseudotype lentiviral vectors and be conjugated with biotinylated targeting ligands. The conjugation is stable enough to redirect lentiviral transduction in the presence of serum, indicating their potential in in vivo . When a signaling molecule is conjugated with the vector, the conjugation facilitates transduction and signaling in a receptor-specific manner. This simple method of ligand conjugation and ease of obtaining various types of biotinylated ligands will make targeted lentiviral transduction easily applicable to broad fields of research.
Collapse
|
20
|
Functional expression of monomeric streptavidin and fusion proteins in Escherichia coli: applications in flow cytometry and ELISA. Appl Microbiol Biotechnol 2018; 102:10079-10089. [PMID: 30250978 DOI: 10.1007/s00253-018-9377-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2018] [Revised: 09/03/2018] [Accepted: 09/05/2018] [Indexed: 02/06/2023]
Abstract
Monomeric streptavidin (mSA) offers a combination of structural and binding properties that are useful in many applications, including a small size and monovalent biotin binding. Because mSA contains a structurally important disulfide bond, the molecule does not fold correctly when expressed inside the cell. We show that mSA can be expressed in a functional form in Escherichia coli by fusing the OmpA signal sequence at the amino terminus. Expressed mSA is exported to the periplasm, from which the molecule leaks to the medium under vigorous shaking. Purified mSA can be conjugated with FITC and used to label microbeads and yeast cells for analysis by flow cytometry, further expanding the scope of mSA-based applications. Some applications require recombinant fusion of mSA with another protein. mSA fused to EGFP cannot be secreted to the medium but was successfully expressed in an engineered cell line that supports oxidative folding in the cytoplasm. Purified mSA-EGFP and mSA-mCherry bound biotin with high affinity and were successfully used in conventional flow cytometry and imaging flow cytometry. Finally, we demonstrate the use of mSA in ELISA, in which horseradish peroxidase-conjugated mSA and biotinylated secondary antibody are used together to detect primary antibody captured on an ELISA plate. Engineering mSA to introduce additional lysine residues can increase the reporter signal above that of wild-type streptavidin. Together, these examples establish mSA as a convenient reagent with a potentially unique role in biotechnology.
Collapse
|
21
|
Paloni JM, Miller EA, Sikes HD, Olsen BD. Improved Ordering in Low Molecular Weight Protein-Polymer Conjugates Through Oligomerization of the Protein Block. Biomacromolecules 2018; 19:3814-3824. [PMID: 30132651 DOI: 10.1021/acs.biomac.8b00928] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
The self-assembly of protein-polymer conjugates incorporating oligomers of a small, engineered high-affinity binding protein, rcSso7d.SA, is studied to determine the effect of protein oligomerization on nanoscale ordering. Oligomerization enables a systematic increase in the protein molar mass without changing its overall folded structure, leading to a higher driving force for self-assembly into well-ordered structures. Though conjugates of monomeric rcSso7d.SA are found to only exist in disordered states, oligomers of this protein linked to a poly( N-isopropylacrylamide) (PNIPAM) block self-assemble into lamellar nanostructures. Conjugates of trimeric and tetrameric rcSso7d.SA are observed to produce the strongest ordering in concentrated solution, displaying birefringent lamellae at concentrations as low as 40 wt %. In highly concentrated solution, the oligomeric rcSso7d.SA-PNIPAM block copolymers exhibit ordering and domain spacing trends atypical from that of most block copolymers. Fluorescent binding assays indicate that oligomerized protein blocks retain binding functionality and exhibit limits of detection up to three times lower than that of surface-immobilized protein sensors. Therefore, oligomerization of the protein block in these block copolymers serves as an effective method to improve both nanoscale ordering and biosensing capabilities.
Collapse
Affiliation(s)
- Justin M Paloni
- Department of Chemical Engineering , Massachusetts Institute of Technology , Cambridge , Massachusetts 02139 , United States
| | - Eric A Miller
- Department of Chemical Engineering , Massachusetts Institute of Technology , Cambridge , Massachusetts 02139 , United States
| | - Hadley D Sikes
- Department of Chemical Engineering , Massachusetts Institute of Technology , Cambridge , Massachusetts 02139 , United States
| | - Bradley D Olsen
- Department of Chemical Engineering , Massachusetts Institute of Technology , Cambridge , Massachusetts 02139 , United States
| |
Collapse
|
22
|
Liao TYA, Lau A, Sunil J, Hytönen V, Hmama Z. Expression of Exogenous Antigens in the Mycobacterium bovis BCG Vaccine via Non-genetic Surface Decoration with the Avidin-biotin System. J Vis Exp 2018. [PMID: 29443102 DOI: 10.3791/56421] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022] Open
Abstract
Tuberculosis (TB) is a serious infectious disease and the only available vaccine M. bovis bacillus Calmette-Guérin (BCG) is safe and effective for protection against children's severe TB meningitis and some forms of disseminated TB, but fails to protect against pulmonary TB, which is the most prevalent form of the disease. Promising strategies to improve BCG currently rely either on its transformation with genes encoding immunodominant M. tuberculosis (Mtb)-specific antigens and/or complementation with genes encoding co-factors that would stimulate antigen presenting cells. Major limitations to these approaches include low efficiency, low stability, and the uncertain level of safety of expression vectors. In this study, we present an alternative approach to vaccine improvement, which consists of BCG complementation with exogenous proteins of interest on the surface of bacteria, rather than transformation with plasmids encoding corresponding genes. First, proteins of interest are expressed in fusion with monomeric avidin in standard E. coli expression systems and then used to decorate the surface of biotinylated BCG. Animal experiments using BCG surface decorated with surrogate ovalbumin antigen demonstrate that the modified bacterium is fully immunogenic and capable of inducing specific T cell responses. Altogether, the data presented here strongly support a novel and efficient method for reshaping the current BCG vaccine that replaces the laborious conventional approach of complementation with exogenous nucleic acids.
Collapse
Affiliation(s)
- Ting-Yu Angela Liao
- Division of Infectious Diseases, Department of Medicine and Vancouver Costal Health Research Institute, University of British Columbia
| | - Alice Lau
- Division of Infectious Diseases, Department of Medicine and Vancouver Costal Health Research Institute, University of British Columbia
| | - Joseph Sunil
- Division of Infectious Diseases, Department of Medicine and Vancouver Costal Health Research Institute, University of British Columbia
| | - Vesa Hytönen
- Institute of Biomedical Technology, University of Tampere
| | - Zakaria Hmama
- Division of Infectious Diseases, Department of Medicine and Vancouver Costal Health Research Institute, University of British Columbia;
| |
Collapse
|
23
|
Sakamoto Y, Kikuchi K, Umeda K, Nakanishi H. Effects of various spacers between biotin and the phospholipid headgroup on immobilization and sedimentation of biotinylated phospholipid-containing liposomes facilitated by avidin-biotin interactions. J Biochem 2017; 162:221-226. [PMID: 28444248 DOI: 10.1093/jb/mvx016] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2016] [Accepted: 02/22/2017] [Indexed: 11/13/2022] Open
Abstract
Immobilization and sedimentation of liposomes (lipid vesicles) are used in liposome-protein binding assays, facilitated by avidin/streptavidin/NeutrAvidin and biotinylated phospholipid-containing liposomes. Here, we examined the effects of three spacers [six-carbon (X), polyethylene glycol (PEG) 180 (molecular weight 180) and PEG2000 (molecular weight 2,000)] between biotin and the phospholipid headgroup on the immobilization and sedimentation of small unilamellar liposomes/vesicles (SUVs). PEG180 and PEG2000 showed more efficient immobilization of biotinylated SUVs on NeutrAvidin-coated plates than X, but X and PEG180 showed more efficient sedimentation of biotinylated SUVs upon NeutrAvidin addition than PEG2000. Thus, the most appropriate spacers differed between immobilization and sedimentation. A spacer for biotinylated SUVs must be selected according to the particular liposome-protein binding assays examined.
Collapse
Affiliation(s)
- Yasuhisa Sakamoto
- Department of Molecular Pharmacology, Graduate School of Medical Sciences, Kumamoto University, 1-1-1 Honjo, Kumamoto 860-8556, Japan
| | - Koji Kikuchi
- Department of Molecular Pharmacology, Graduate School of Medical Sciences, Kumamoto University, 1-1-1 Honjo, Kumamoto 860-8556, Japan
| | - Kazuaki Umeda
- Department of Molecular Pharmacology, Graduate School of Medical Sciences, Kumamoto University, 1-1-1 Honjo, Kumamoto 860-8556, Japan
| | - Hiroyuki Nakanishi
- Department of Molecular Pharmacology, Graduate School of Medical Sciences, Kumamoto University, 1-1-1 Honjo, Kumamoto 860-8556, Japan
| |
Collapse
|
24
|
Lee SH, Jin C, Cai E, Ge P, Ishitsuka Y, Teng KW, de Thomaz AA, Nall D, Baday M, Jeyifous O, Demonte D, Dundas CM, Park S, Delgado JY, Green WN, Selvin PR. Super-resolution imaging of synaptic and Extra-synaptic AMPA receptors with different-sized fluorescent probes. eLife 2017; 6:27744. [PMID: 28749340 PMCID: PMC5779237 DOI: 10.7554/elife.27744] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2017] [Accepted: 07/26/2017] [Indexed: 12/13/2022] Open
Abstract
Previous studies tracking AMPA receptor (AMPAR) diffusion at synapses observed a large mobile extrasynaptic AMPAR pool. Using super-resolution microscopy, we examined how fluorophore size and photostability affected AMPAR trafficking outside of, and within, post-synaptic densities (PSDs) from rats. Organic fluorescent dyes (≈4 nm), quantum dots, either small (≈10 nm diameter; sQDs) or big (>20 nm; bQDs), were coupled to AMPARs via different-sized linkers. We find that >90% of AMPARs labeled with fluorescent dyes or sQDs were diffusing in confined nanodomains in PSDs, which were stable for 15 min or longer. Less than 10% of sQD-AMPARs were extrasynaptic and highly mobile. In contrast, 5-10% of bQD-AMPARs were in PSDs and 90-95% were extrasynaptic as previously observed. Contrary to the hypothesis that AMPAR entry is limited by the occupancy of open PSD 'slots', our findings suggest that AMPARs rapidly enter stable 'nanodomains' in PSDs with lifetime >15 min, and do not accumulate in extrasynaptic membranes.
Collapse
Affiliation(s)
- Sang Hak Lee
- Department of Physics, Center for Biophysics, and Quantitative Biology, and Center for the Physics of Living Cells, University of Illinois, Urbana-Champaign, Champaign, United States
| | - Chaoyi Jin
- Department of Physics, Center for Biophysics, and Quantitative Biology, and Center for the Physics of Living Cells, University of Illinois, Urbana-Champaign, Champaign, United States
| | - En Cai
- Department of Physics, Center for Biophysics, and Quantitative Biology, and Center for the Physics of Living Cells, University of Illinois, Urbana-Champaign, Champaign, United States
| | - Pinghua Ge
- Department of Physics, Center for Biophysics, and Quantitative Biology, and Center for the Physics of Living Cells, University of Illinois, Urbana-Champaign, Champaign, United States
| | - Yuji Ishitsuka
- Department of Physics, Center for Biophysics, and Quantitative Biology, and Center for the Physics of Living Cells, University of Illinois, Urbana-Champaign, Champaign, United States
| | - Kai Wen Teng
- Department of Physics, Center for Biophysics, and Quantitative Biology, and Center for the Physics of Living Cells, University of Illinois, Urbana-Champaign, Champaign, United States
| | - Andre A de Thomaz
- Department of Physics, Center for Biophysics, and Quantitative Biology, and Center for the Physics of Living Cells, University of Illinois, Urbana-Champaign, Champaign, United States
| | - Duncan Nall
- Department of Physics, Center for Biophysics, and Quantitative Biology, and Center for the Physics of Living Cells, University of Illinois, Urbana-Champaign, Champaign, United States
| | - Murat Baday
- Department of Physics, Center for Biophysics, and Quantitative Biology, and Center for the Physics of Living Cells, University of Illinois, Urbana-Champaign, Champaign, United States
| | - Okunola Jeyifous
- Department of Neurobiology, University of Chicago and the Marine Biological Laboratory, Chicago, United States
| | - Daniel Demonte
- Department of Chemical and Biological Engineering, University at Buffalo, Buffalo, United States
| | - Christopher M Dundas
- Department of Chemical and Biological Engineering, University at Buffalo, Buffalo, United States
| | - Sheldon Park
- Department of Chemical and Biological Engineering, University at Buffalo, Buffalo, United States
| | - Jary Y Delgado
- Department of Neurobiology, University of Chicago and the Marine Biological Laboratory, Chicago, United States
| | - William N Green
- Department of Neurobiology, University of Chicago and the Marine Biological Laboratory, Chicago, United States
| | - Paul R Selvin
- Department of Physics, Center for Biophysics, and Quantitative Biology, and Center for the Physics of Living Cells, University of Illinois, Urbana-Champaign, Champaign, United States
| |
Collapse
|
25
|
Structural characterization of core-bradavidin in complex with biotin. PLoS One 2017; 12:e0176086. [PMID: 28426764 PMCID: PMC5398887 DOI: 10.1371/journal.pone.0176086] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2017] [Accepted: 04/05/2017] [Indexed: 02/07/2023] Open
Abstract
Bradavidin is a tetrameric biotin-binding protein similar to chicken avidin and bacterial streptavidin, and was originally cloned from the nitrogen-fixing bacteria Bradyrhizobium diazoefficiens. We have previously reported the crystal structure of the full-length, wild-type (wt) bradavidin with 138 amino acids, where the C-terminal residues Gly129-Lys138 (“Brad-tag”) act as an intrinsic ligand (i.e. Gly129-Lys138 bind into the biotin-binding site of an adjacent subunit within the same tetramer) and has potential as an affinity tag for biotechnological purposes. Here, the X-ray structure of core-bradavidin lacking the C-terminal residues Gly114-Lys138, and hence missing the Brad-tag, was crystallized in complex with biotin at 1.60 Å resolution [PDB:4BBO]. We also report a homology model of rhodavidin, an avidin-like protein from Rhodopseudomonas palustris, and of an avidin-like protein from Bradyrhizobium sp. Ai1a-2, both of which have the Brad-tag sequence at their C-terminus. Moreover, core-bradavidin V1, an engineered variant of the original core-bradavidin, was also expressed at high levels in E. coli, as well as a double mutant (Cys39Ala and Cys69Ala) of core-bradavidin (CC mutant). Our data help us to further engineer the core-bradavidin–Brad-tag pair for biotechnological assays and chemical biology applications, and provide deeper insight into the biotin-binding mode of bradavidin.
Collapse
|
26
|
Chamma I, Rossier O, Giannone G, Thoumine O, Sainlos M. Optimized labeling of membrane proteins for applications to super-resolution imaging in confined cellular environments using monomeric streptavidin. Nat Protoc 2017; 12:748-763. [DOI: 10.1038/nprot.2017.010] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
|
27
|
Chamma I, Levet F, Sibarita JB, Sainlos M, Thoumine O. Nanoscale organization of synaptic adhesion proteins revealed by single-molecule localization microscopy. NEUROPHOTONICS 2016; 3:041810. [PMID: 27872870 PMCID: PMC5093229 DOI: 10.1117/1.nph.3.4.041810] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/04/2016] [Accepted: 10/11/2016] [Indexed: 06/06/2023]
Abstract
The advent of superresolution imaging has created a strong need for both optimized labeling strategies and analysis methods to probe the nanoscale organization of complex biological structures. We present a thorough description of the distribution of synaptic adhesion proteins at the nanoscopic scale, namely presynaptic neurexin-[Formula: see text] ([Formula: see text]), and its two postsynaptic binding partners neuroligin-1 (Nlg1) and leucine-rich-repeat transmembrane protein 2 (LRRTM2). We monitored these proteins in the membrane of neurons by direct stochastic optical reconstruction microscopy, after live surface labeling with Alexa647-conjugated monomeric streptavidin. The small probe ([Formula: see text]) efficiently penetrates into crowded synaptic junctions and reduces the distance to target. We quantified the organization of the single-molecule localization data using a tesselation-based analysis technique. We show that Nlg1 exhibits a fairly disperse organization within dendritic spines, while LRRTM2 is organized in compact domains, and [Formula: see text] in presynaptic terminals displays a dual-organization pattern intermediate between that of Nlg1 and LRRTM2. These results suggest that part of [Formula: see text] interacts transsynaptically with Nlg1 and the other part with LRRTM2.
Collapse
Affiliation(s)
- Ingrid Chamma
- Centre National de la Recherche Scientifique, Interdisciplinary Institute for Neuroscience, UMR 5297, 147 rue Léo-Saignat, Bordeaux Cedex 33077, France
- University of Bordeaux, Interdisciplinary Institute for Neuroscience, 147 rue Léo-Saignat, Bordeaux Cedex 33077, France
| | - Florian Levet
- Centre National de la Recherche Scientifique, Interdisciplinary Institute for Neuroscience, UMR 5297, 147 rue Léo-Saignat, Bordeaux Cedex 33077, France
- University of Bordeaux, Interdisciplinary Institute for Neuroscience, 147 rue Léo-Saignat, Bordeaux Cedex 33077, France
| | - Jean-Baptiste Sibarita
- Centre National de la Recherche Scientifique, Interdisciplinary Institute for Neuroscience, UMR 5297, 147 rue Léo-Saignat, Bordeaux Cedex 33077, France
- University of Bordeaux, Interdisciplinary Institute for Neuroscience, 147 rue Léo-Saignat, Bordeaux Cedex 33077, France
| | - Matthieu Sainlos
- Centre National de la Recherche Scientifique, Interdisciplinary Institute for Neuroscience, UMR 5297, 147 rue Léo-Saignat, Bordeaux Cedex 33077, France
- University of Bordeaux, Interdisciplinary Institute for Neuroscience, 147 rue Léo-Saignat, Bordeaux Cedex 33077, France
| | - Olivier Thoumine
- Centre National de la Recherche Scientifique, Interdisciplinary Institute for Neuroscience, UMR 5297, 147 rue Léo-Saignat, Bordeaux Cedex 33077, France
- University of Bordeaux, Interdisciplinary Institute for Neuroscience, 147 rue Léo-Saignat, Bordeaux Cedex 33077, France
| |
Collapse
|
28
|
Mapping the dynamics and nanoscale organization of synaptic adhesion proteins using monomeric streptavidin. Nat Commun 2016; 7:10773. [PMID: 26979420 PMCID: PMC4799371 DOI: 10.1038/ncomms10773] [Citation(s) in RCA: 115] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2015] [Accepted: 01/17/2016] [Indexed: 12/12/2022] Open
Abstract
The advent of super-resolution imaging (SRI) has created a need for optimized labelling strategies. We present a new method relying on fluorophore-conjugated monomeric streptavidin (mSA) to label membrane proteins carrying a short, enzymatically biotinylated tag, compatible with SRI techniques including uPAINT, STED and dSTORM. We demonstrate efficient and specific labelling of target proteins in confined intercellular and organotypic tissues, with reduced steric hindrance and no crosslinking compared with multivalent probes. We use mSA to decipher the dynamics and nanoscale organization of the synaptic adhesion molecules neurexin-1β, neuroligin-1 (Nlg1) and leucine-rich-repeat transmembrane protein 2 (LRRTM2) in a dual-colour configuration with GFP nanobody, and show that these proteins are diffusionally trapped at synapses where they form apposed trans-synaptic adhesive structures. Furthermore, Nlg1 is dynamic, disperse and sensitive to synaptic stimulation, whereas LRRTM2 is organized in compact and stable nanodomains. Thus, mSA is a versatile tool to image membrane proteins at high resolution in complex live environments, providing novel information about the nano-organization of biological structures. The advent of fluorescence-based super-resolution microscopy has created a need for labeling strategies relying on small probes that minimally perturb protein function. Here the authors describe a labeling method that reduces protein tag and label sizes, allowing for accurate protein targeting and measurements of protein dynamics in tight cellular spaces.
Collapse
|
29
|
Lee JM, Kim JA, Yen TC, Lee IH, Ahn B, Lee Y, Hsieh CL, Kim HM, Jung Y. A Rhizavidin Monomer with Nearly Multimeric Avidin-Like Binding Stability Against Biotin Conjugates. Angew Chem Int Ed Engl 2016. [DOI: 10.1002/ange.201510885] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Affiliation(s)
- Jeong Min Lee
- Department of Chemistry; Korea Advanced Institute of Science and Technology; Daejeon 305-701 Korea
| | - Jung A. Kim
- Graduate School of Nanoscience and Technology; Korea Advanced Institute of Science and Technology; Korea
| | - Tzu-Chi Yen
- Institute of Atomic and Molecular Sciences, Academia Sinica; Taipei 10617 Taiwan
| | - In Hwan Lee
- Department of Chemistry; Korea Advanced Institute of Science and Technology; Daejeon 305-701 Korea
| | - Byungjun Ahn
- Department of Chemistry; Korea Advanced Institute of Science and Technology; Daejeon 305-701 Korea
| | - Younghoon Lee
- Department of Chemistry; Korea Advanced Institute of Science and Technology; Daejeon 305-701 Korea
| | - Chia-Lung Hsieh
- Institute of Atomic and Molecular Sciences, Academia Sinica; Taipei 10617 Taiwan
| | - Ho Min Kim
- Graduate School of Medical Science and Engineering; Korea Advanced Institute of Science and Technology; Korea
| | - Yongwon Jung
- Department of Chemistry; Korea Advanced Institute of Science and Technology; Daejeon 305-701 Korea
| |
Collapse
|
30
|
Lee JM, Kim JA, Yen TC, Lee IH, Ahn B, Lee Y, Hsieh CL, Kim HM, Jung Y. A Rhizavidin Monomer with Nearly Multimeric Avidin-Like Binding Stability Against Biotin Conjugates. Angew Chem Int Ed Engl 2016; 55:3393-7. [DOI: 10.1002/anie.201510885] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2015] [Indexed: 12/22/2022]
Affiliation(s)
- Jeong Min Lee
- Department of Chemistry; Korea Advanced Institute of Science and Technology; Daejeon 305-701 Korea
| | - Jung A. Kim
- Graduate School of Nanoscience and Technology; Korea Advanced Institute of Science and Technology; Korea
| | - Tzu-Chi Yen
- Institute of Atomic and Molecular Sciences, Academia Sinica; Taipei 10617 Taiwan
| | - In Hwan Lee
- Department of Chemistry; Korea Advanced Institute of Science and Technology; Daejeon 305-701 Korea
| | - Byungjun Ahn
- Department of Chemistry; Korea Advanced Institute of Science and Technology; Daejeon 305-701 Korea
| | - Younghoon Lee
- Department of Chemistry; Korea Advanced Institute of Science and Technology; Daejeon 305-701 Korea
| | - Chia-Lung Hsieh
- Institute of Atomic and Molecular Sciences, Academia Sinica; Taipei 10617 Taiwan
| | - Ho Min Kim
- Graduate School of Medical Science and Engineering; Korea Advanced Institute of Science and Technology; Korea
| | - Yongwon Jung
- Department of Chemistry; Korea Advanced Institute of Science and Technology; Daejeon 305-701 Korea
| |
Collapse
|
31
|
Fogen D, Wu SC, Ng KKS, Wong SL. Engineering Streptavidin and a Streptavidin-Binding Peptide with Infinite Binding Affinity and Reversible Binding Capability: Purification of a Tagged Recombinant Protein to High Purity via Affinity-Driven Thiol Coupling. PLoS One 2015; 10:e0139137. [PMID: 26406477 PMCID: PMC4583386 DOI: 10.1371/journal.pone.0139137] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2015] [Accepted: 09/08/2015] [Indexed: 12/02/2022] Open
Abstract
To extend and improve the utility of the streptavidin-binding peptide tag (SBP-tag) in applications ranging from affinity purification to the reversible immobilization of recombinant proteins, a cysteine residue was introduced to the streptavidin mutein SAVSBPM18 and the SBP-tag to generate SAVSBPM32 and SBP(A18C), respectively. This pair of derivatives is capable of forming a disulfide bond through the newly introduced cysteine residues. SAVSBPM32 binds SBP-tag and biotin with binding affinities (Kd ~ 10-8M) that are similar to SAVSBPM18. Although SBP(A18C) binds to SAVSBPM32 more weakly than SBP-tag, the binding affinity is sufficient to bring the two binding partners together efficiently before they are locked together via disulfide bond formation–a phenomenon we have named affinity-driven thiol coupling. Under the condition with SBP(A18C) tags in excess, two SBP(A18C) tags can be captured by a tetrameric SAVSBPM32. The stoichiometry of the disulfide-bonded SAVSBPM32-SBP(A18C) complex was determined using a novel two-dimensional electrophoresis method which has general applications for analyzing the composition of disulfide-bonded protein complexes. To illustrate the application of this reversible immobilization technology, optimized conditions were established to use the SAVSBPM32-affinity matrix for the purification of a SBP(A18C)-tagged reporter protein to high purity. Furthermore, we show that the SAVSBPM32-affinity matrix can also be applied to purify a biotinylated protein and a reporter protein tagged with the unmodified SBP-tag. The dual (covalent and non-covalent) binding modes possible in this system offer great flexibility to many different applications which need reversible immobilization capability.
Collapse
Affiliation(s)
- Dawson Fogen
- Department of Biological Sciences, University of Calgary, Calgary, Alberta, Canada
| | - Sau-Ching Wu
- Department of Biological Sciences, University of Calgary, Calgary, Alberta, Canada
| | - Kenneth Kai-Sing Ng
- Department of Biological Sciences, University of Calgary, Calgary, Alberta, Canada
| | - Sui-Lam Wong
- Department of Biological Sciences, University of Calgary, Calgary, Alberta, Canada
- * E-mail:
| |
Collapse
|
32
|
Sun X, Montiel D, Li H, Yang H. “Plug-and-Go” Strategy To Manipulate Streptavidin Valencies. Bioconjug Chem 2014; 25:1375-80. [DOI: 10.1021/bc500296p] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Affiliation(s)
- Xun Sun
- Department of Chemistry, Princeton University, Princeton, New Jersey 08544, United States
| | - Daniel Montiel
- Department of Chemistry, Princeton University, Princeton, New Jersey 08544, United States
| | - Hao Li
- Department of Chemistry, Princeton University, Princeton, New Jersey 08544, United States
| | - Haw Yang
- Department of Chemistry, Princeton University, Princeton, New Jersey 08544, United States
| |
Collapse
|
33
|
Expression and purification of soluble monomeric streptavidin in Escherichia coli. Appl Microbiol Biotechnol 2014; 98:6285-95. [PMID: 24691867 DOI: 10.1007/s00253-014-5682-y] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2013] [Revised: 03/05/2014] [Accepted: 03/14/2014] [Indexed: 10/25/2022]
Abstract
We recently reported the engineering of monomeric streptavidin (mSA) for use in monomeric detection of biotinylated ligands. Although mSA can be expressed functionally on the surface of mammalian cells and yeast, the molecule does not fold correctly when expressed in Escherichia coli. Refolding from inclusion bodies is cumbersome and yields a limited amount of purified protein. Improving the final yield should facilitate its use in biotechnology. We tested the expression and purification of mSA fused to GST, MBP, thioredoxin, and sumo tags to simplify its purification and improve the yield. The fusion proteins can be expressed solubly in E. coli and increase the yield by more than 20-fold. Unmodified mSA can be obtained by proteolytically removing the fusion tag. Purified mSA can be immobilized on a solid matrix to purify biotinylated ligands. Together, expressing mSA as a fusion with a solubilization tag vastly simplifies its preparation and increases its usability in biotechnology.
Collapse
|
34
|
Streptavidin–biotin technology: improvements and innovations in chemical and biological applications. Appl Microbiol Biotechnol 2013; 97:9343-53. [DOI: 10.1007/s00253-013-5232-z] [Citation(s) in RCA: 309] [Impact Index Per Article: 25.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2013] [Revised: 08/29/2013] [Accepted: 09/02/2013] [Indexed: 12/25/2022]
|