1
|
Zhou J, Ridderbeek K, Zou P, Naden AB, Gaussmann S, Song F, Falter-Braun P, Kay ER, Sattler M, Cui J. Modular Nanoparticle Platform for Solution-Phase Optical Sensing of Protein-Protein Interactions. ACS APPLIED OPTICAL MATERIALS 2025; 3:676-688. [PMID: 40176919 PMCID: PMC11959585 DOI: 10.1021/acsaom.4c00486] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/21/2024] [Revised: 02/12/2025] [Accepted: 02/13/2025] [Indexed: 04/05/2025]
Abstract
Protein-protein interactions regulate essentially all cellular processes. Understanding these interactions, including the quantification of binding parameters, is crucial for unraveling the molecular mechanisms underlying cellular pathways and, ultimately, their roles in cellular physiology and pathology. Current methods for measuring protein-protein interactions in vitro generally require amino acid conjugation of fluorescent tags, complex instrumentation, large amounts of purified protein, or measurement at extended surfaces. Here, we present an elegant nanoparticle-based platform for the optical detection of protein-protein interactions in the solution phase. We synthesized gold-coated silver decahedral nanoparticles possessing high chemical stability and exceptional optical sensing properties. The nanoparticle surface is then tailored for specific binding to commonly used polyhistidine tags of recombinant proteins. Sequential addition of proteins to the nanoparticle suspension results in spectral shifts of the localized surface plasmon resonance that can be monitored by conventional UV-vis spectrophotometry. With this approach, we demonstrate both the qualitative detection of specific protein-protein interactions and the quantification of equilibrium and kinetic binding parameters between small globular proteins. Requiring minimal protein quantities and basic laboratory equipment, this technique offers a simple, economical, and modular approach to characterizing protein-protein interactions, holds promise for broad use in future studies, and may serve as a template for future biosensing technologies.
Collapse
Affiliation(s)
- Jieying Zhou
- Helmholtz
Pioneer Campus, Helmholtz Munich, Neuherberg 85764, Germany
| | | | - Peijian Zou
- Institute
of Structural Biology, Molecular Targets and Therapeutics Center, Helmholtz Munich, Neuherberg 85764, Germany
- Bavarian
NMR Center, Department of Bioscience, School of Natural Sciences, Technical University of Munich, Garching 85748, Germany
| | - Aaron B. Naden
- EaStCHEM
School of Chemistry, University of St. Andrews, St. Andrews KY16 9ST, U.K.
| | - Stefan Gaussmann
- Institute
of Structural Biology, Molecular Targets and Therapeutics Center, Helmholtz Munich, Neuherberg 85764, Germany
- Bavarian
NMR Center, Department of Bioscience, School of Natural Sciences, Technical University of Munich, Garching 85748, Germany
| | - Fangyuan Song
- Helmholtz
Pioneer Campus, Helmholtz Munich, Neuherberg 85764, Germany
| | - Pascal Falter-Braun
- Institute
of Network Biology (INET), Molecular Targets and Therapeutics Center
(MTTC), Helmholtz Munich, Neuherberg 85764 Germany
- Microbe-Host
Interactions, Faculty of Biology, Ludwig-Maximilians-Universität
(LMU) München, Planegg-Martinsried 82152, Germany
| | - Euan R. Kay
- EaStCHEM
School of Chemistry, University of St. Andrews, St. Andrews KY16 9ST, U.K.
| | - Michael Sattler
- Institute
of Structural Biology, Molecular Targets and Therapeutics Center, Helmholtz Munich, Neuherberg 85764, Germany
- Bavarian
NMR Center, Department of Bioscience, School of Natural Sciences, Technical University of Munich, Garching 85748, Germany
| | - Jian Cui
- Helmholtz
Pioneer Campus, Helmholtz Munich, Neuherberg 85764, Germany
- Department
of Bioscience, School of Natural Sciences, Technical University of Munich, Garching 85748, Germany
| |
Collapse
|
2
|
Zhang Y, Li Y, Ren T, Xiao P, Duan JA. Novel and efficient techniques in the discovery of antioxidant peptides. Crit Rev Food Sci Nutr 2024; 64:11934-11948. [PMID: 37585700 DOI: 10.1080/10408398.2023.2245052] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/18/2023]
Abstract
As a research hotspot in food science and nutrition, antioxidant peptides can function by scavenging free radicals, inhibiting peroxides, and chelating metal ions. Therefore, how to efficiently discover and screen antioxidant peptides has become a key issue in research and production. Traditional discovery methods are time-consuming and costly, but also challenging to resolve the quantitative structure-activity relationship of antioxidant peptides. Several novel techniques, including artificial intelligence, molecular docking, bioinformatics, quantum chemistry, phage display, switchSENSE, surface plasmon resonance, and fluorescence polarization, are emerging rapidly as solutions. These techniques possess efficient capability for the discovery of antioxidant peptides, even with the potential for high-throughput screening. In addition, the quantitative structure-activity relationship can be resolved. Notably, combining these novel techniques can overcome the drawbacks of a single one, thus improving efficiency and expanding the discovery horizon. This review has summarized eight novel and efficient techniques for discovering antioxidant peptides and the combination of techniques. This review aims to provide scientific evidence and perspectives for antioxidant peptide research.
Collapse
Affiliation(s)
- Yuhao Zhang
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, National and Local Collaborative Engineering Center of Chinese Medicinal Resources Industrialization and Formulae Innovative Medicine, and Jiangsu Key Laboratory for High Technology Research of TCM Formulae, Nanjing University of Chinese Medicine, Nanjing, China
| | - Yun Li
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, National and Local Collaborative Engineering Center of Chinese Medicinal Resources Industrialization and Formulae Innovative Medicine, and Jiangsu Key Laboratory for High Technology Research of TCM Formulae, Nanjing University of Chinese Medicine, Nanjing, China
| | - Tianyi Ren
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, National and Local Collaborative Engineering Center of Chinese Medicinal Resources Industrialization and Formulae Innovative Medicine, and Jiangsu Key Laboratory for High Technology Research of TCM Formulae, Nanjing University of Chinese Medicine, Nanjing, China
| | - Ping Xiao
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, National and Local Collaborative Engineering Center of Chinese Medicinal Resources Industrialization and Formulae Innovative Medicine, and Jiangsu Key Laboratory for High Technology Research of TCM Formulae, Nanjing University of Chinese Medicine, Nanjing, China
| | - Jin-Ao Duan
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, National and Local Collaborative Engineering Center of Chinese Medicinal Resources Industrialization and Formulae Innovative Medicine, and Jiangsu Key Laboratory for High Technology Research of TCM Formulae, Nanjing University of Chinese Medicine, Nanjing, China
| |
Collapse
|
3
|
Reusch J, Andersen JT, Rant U, Schlothauer T. Insight into the avidity-affinity relationship of the bivalent, pH-dependent interaction between IgG and FcRn. MAbs 2024; 16:2361585. [PMID: 38849969 PMCID: PMC11164218 DOI: 10.1080/19420862.2024.2361585] [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: 10/01/2023] [Accepted: 05/24/2024] [Indexed: 06/09/2024] Open
Abstract
Monoclonal antibodies (mAbs) as therapeutics necessitate favorable pharmacokinetic properties, including extended serum half-life, achieved through pH-dependent binding to the neonatal Fc receptor (FcRn). While prior research has mainly investigated IgG-FcRn binding kinetics with a focus on single affinity values, it has been shown that each IgG molecule can engage two FcRn molecules throughout an endosomal pH gradient. As such, we present here a more comprehensive analysis of these interactions with an emphasis on both affinity and avidity by taking advantage of switchSENSE technology, a surface-based biosensor where recombinant FcRn was immobilized via short DNA nanolevers, mimicking the membranous orientation of the receptor. The results revealed insight into the avidity-to-affinity relationship, where assessing binding through a pH gradient ranging from pH 5.8 to 7.4 showed that the half-life extended IgG1-YTE has an affinity inflection point at pH 7.2, reflecting its engineering for improved FcRn binding compared with the wild-type counterpart. Furthermore, IgG1-YTE displayed a pH switch for the avidity enhancement factor at pH 6.2, reflecting strong receptor binding to both sides of the YTE-containing Fc, while avidity was abolished at pH 7.4. When compared with classical surface plasmon resonance (SPR) technology and complementary methods, the use of switchSENSE demonstrated superior capabilities in differentiating affinity from avidity within a single measurement. Thus, the methodology provides reliable kinetic rate parameters for both binding modes and their direct relationship as a function of pH. Also, it deciphers the potential effect of the variable Fab arms on FcRn binding, in which SPR has limitations. Our study offers guidance for how FcRn binding properties can be studied for IgG engineering strategies.
Collapse
Affiliation(s)
- Johannes Reusch
- Dynamic Biosensors GmbH, Munich, Germany
- Roche Pharma Research and Early Development, Therapeutic Modalities, Roche Innovation Center Munich, Roche Diagnostics GmbH, Penzberg, Germany
| | - Jan Terje Andersen
- Department of Immunology, Oslo University Hospital Rikshospitalet, Oslo, Norway
- Institute of Clinical Medicine, Department of Pharmacology, University of Oslo, Oslo, Norway
- Precision Immunotherapy Alliance (PRIMA), University of Oslo, Oslo, Norway
| | | | - Tilman Schlothauer
- Roche Pharma Research and Early Development, Therapeutic Modalities, Roche Innovation Center Munich, Roche Diagnostics GmbH, Penzberg, Germany
| |
Collapse
|
4
|
Vogt M, List J, Langecker M, Santiago I, Simmel FC, Kopperger E. Electrokinetic Torque Generation by DNA Nanorobotic Arms Studied via Single-Molecule Fluctuation Analysis. J Phys Chem B 2023; 127:10710-10722. [PMID: 38060372 DOI: 10.1021/acs.jpcb.3c05959] [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: 12/22/2023]
Abstract
DNA nanotechnology has enabled the creation of supramolecular machines, whose shape and function are inspired from traditional mechanical engineering as well as from biological examples. As DNA inherently is a highly charged biopolymer, the external application of electric fields provides a versatile, computer-programmable way to control the movement of DNA-based machines. However, the details of the electrohydrodynamic interactions underlying the electrical manipulation of these machines are complex, as the influence of their intrinsic charge, the surrounding cloud of counterions, and the effect of electrokinetic fluid flow have to be taken into account. In this work, we identify the relevant effects involved in this actuation mechanism by determining the electric response of an established DNA-based nanorobotic arm to varying design and operation parameters. Borrowing an approach from single-molecule biophysics, we determined the electrical torque exerted on the nanorobotic arms by analyzing their thermal fluctuations when oriented in an electric field. We analyze the influence of various experimental and design parameters on the "actuatability" of the nanostructures and optimize the generated torque according to these parameters. Our findings give insight into the physical processes involved in the actuation mechanism and provide general guidelines that aid in designing and efficiently operating electrically driven nanorobotic devices made from DNA.
Collapse
Affiliation(s)
- Matthias Vogt
- Physics of Synthetic Biological Systems─E14, Department of Bioscience, TUM School of Natural Science, Technical University of Munich, 85748 Garching, Germany
| | - Jonathan List
- Physics of Synthetic Biological Systems─E14, Department of Bioscience, TUM School of Natural Science, Technical University of Munich, 85748 Garching, Germany
| | - Martin Langecker
- Physics of Synthetic Biological Systems─E14, Department of Bioscience, TUM School of Natural Science, Technical University of Munich, 85748 Garching, Germany
| | - Ibon Santiago
- CIC nanoGUNE BRTA, Donostia-San Sebastián 20018, Spain
| | - Friedrich C Simmel
- Physics of Synthetic Biological Systems─E14, Department of Bioscience, TUM School of Natural Science, Technical University of Munich, 85748 Garching, Germany
| | - Enzo Kopperger
- Physics of Synthetic Biological Systems─E14, Department of Bioscience, TUM School of Natural Science, Technical University of Munich, 85748 Garching, Germany
| |
Collapse
|
5
|
Schedler B, Yukhnovets O, Lindner L, Meyer A, Fitter J. The Thermodynamic Fingerprints of Ultra-Tight Nanobody-Antigen Binding Probed via Two-Color Single-Molecule Coincidence Detection. Int J Mol Sci 2023; 24:16379. [PMID: 38003569 PMCID: PMC10671529 DOI: 10.3390/ijms242216379] [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: 10/11/2023] [Revised: 11/07/2023] [Accepted: 11/13/2023] [Indexed: 11/26/2023] Open
Abstract
Life on the molecular scale is based on a versatile interplay of biomolecules, a feature that is relevant for the formation of macromolecular complexes. Fluorescence-based two-color coincidence detection is widely used to characterize molecular binding and was recently improved by a brightness-gated version which gives more accurate results. We developed and established protocols which make use of coincidence detection to quantify binding fractions between interaction partners labeled with fluorescence dyes of different colors. Since the applied technique is intrinsically related to single-molecule detection, the concentration of diffusing molecules for confocal detection is typically in the low picomolar regime. This makes the approach a powerful tool for determining bi-molecular binding affinities, in terms of KD values, in this regime. We demonstrated the reliability of our approach by analyzing very strong nanobody-EGFP binding. By measuring the affinity at different temperatures, we were able to determine the thermodynamic parameters of the binding interaction. The results show that the ultra-tight binding is dominated by entropic contributions.
Collapse
Affiliation(s)
- Benno Schedler
- AG Biophysik, I. Physikalisches Institut (IA), RWTH Aachen University, 52074 Aachen, Germany; (B.S.); (O.Y.); (L.L.); (A.M.)
| | - Olessya Yukhnovets
- AG Biophysik, I. Physikalisches Institut (IA), RWTH Aachen University, 52074 Aachen, Germany; (B.S.); (O.Y.); (L.L.); (A.M.)
| | - Lennart Lindner
- AG Biophysik, I. Physikalisches Institut (IA), RWTH Aachen University, 52074 Aachen, Germany; (B.S.); (O.Y.); (L.L.); (A.M.)
| | - Alida Meyer
- AG Biophysik, I. Physikalisches Institut (IA), RWTH Aachen University, 52074 Aachen, Germany; (B.S.); (O.Y.); (L.L.); (A.M.)
| | - Jörg Fitter
- AG Biophysik, I. Physikalisches Institut (IA), RWTH Aachen University, 52074 Aachen, Germany; (B.S.); (O.Y.); (L.L.); (A.M.)
- ER-C-3 Structural Biology & IBI-6 Cellular Structural Biology, Forschungszentrum Jülich, 52425 Jülich, Germany
| |
Collapse
|
6
|
Bingham M, Pesnot T, Scott AD. Biophysical screening and characterisation in medicinal chemistry. PROGRESS IN MEDICINAL CHEMISTRY 2023; 62:61-104. [PMID: 37981351 DOI: 10.1016/bs.pmch.2023.10.002] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/21/2023]
Abstract
In the last two decades the use of biophysical assays and methods in medicinal chemistry has increased significantly, to meet the demands of the novel targets and modalities that drug discoverers are looking to tackle. The desire to obtain accurate affinities, kinetics, thermodynamics and structural data as early as possible in the drug discovery process has fuelled this innovation. This review introduces the principles underlying the techniques in common use and provides a perspective on the weaknesses and strengths of different methods. Case studies are used to further illustrate some of the applications in medicinal chemistry and a discussion of the emerging biophysical methods on the horizon is presented.
Collapse
|
7
|
Freitag JS, Möser C, Belay R, Altattan B, Grasse N, Pothineni BK, Schnauß J, Smith DM. Integration of functional peptides into nucleic acid-based nanostructures. NANOSCALE 2023; 15:7608-7624. [PMID: 37042085 DOI: 10.1039/d2nr05429a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/05/2023]
Abstract
In many applications such as diagnostics and therapy development, small peptide fragments consisting of only a few amino acids are often attractive alternatives to bulky proteins. This is due to factors such as the ease of scalable chemical synthesis and numerous methods for their discovery. One drawback of using peptides is that their activity can often be negatively impacted by the lack of a rigid, 3D stabilizing structure provided by the rest of the protein. In many cases, this can be alleviated by different methods of rational templating onto nanomaterials, which provides additional possibilities to use concepts of multivalence or rational nano-engineering to enhance or even create new types of function or structure. In recent years, nanostructures made from the self-assembly of DNA strands have been used as scaffolds to create functional arrangements of peptides, often leading to greatly enhanced biological activity or new material properties. This review will give an overview of nano-templating approaches based on the combination of DNA nanotechnology and peptides. This will include both bioengineering strategies to control interactions with cells or other biological systems, as well as examples where the combination of DNA and peptides has been leveraged for the rational design of new functional materials.
Collapse
Affiliation(s)
- Jessica S Freitag
- Fraunhofer Institute for Cell Therapy and Immunology, 04103 Leipzig, Germany.
| | - Christin Möser
- Fraunhofer Institute for Cell Therapy and Immunology, 04103 Leipzig, Germany.
| | - Robel Belay
- Fraunhofer Institute for Cell Therapy and Immunology, 04103 Leipzig, Germany.
| | - Basma Altattan
- Fraunhofer Institute for Cell Therapy and Immunology, 04103 Leipzig, Germany.
| | - Nico Grasse
- Fraunhofer Institute for Cell Therapy and Immunology, 04103 Leipzig, Germany.
| | | | - Jörg Schnauß
- Fraunhofer Institute for Cell Therapy and Immunology, 04103 Leipzig, Germany.
- Peter Debye Institute for Soft Matter Physics, Leipzig University, 04103 Leipzig, Germany
- Unconventional Computing Lab, UWE, Bristol, BS16 1QY, UK
| | - David M Smith
- Fraunhofer Institute for Cell Therapy and Immunology, 04103 Leipzig, Germany.
- Peter Debye Institute for Soft Matter Physics, Leipzig University, 04103 Leipzig, Germany
- Institute of Clinical Immunology, University of Leipzig Medical Faculty, 04103 Leipzig, Germany
| |
Collapse
|
8
|
Zhu L, Chang Y, Li Y, Qiao M, Liu L. Biosensors Based on the Binding Events of Nitrilotriacetic Acid-Metal Complexes. BIOSENSORS 2023; 13:bios13050507. [PMID: 37232868 DOI: 10.3390/bios13050507] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/25/2023] [Revised: 04/21/2023] [Accepted: 04/24/2023] [Indexed: 05/27/2023]
Abstract
Molecular immobilization and recognition are two key events for the development of biosensors. The general ways for the immobilization and recognition of biomolecules include covalent coupling reactions and non-covalent interactions of antigen-antibody, aptamer-target, glycan-lectin, avidin-biotin and boronic acid-diol. Tetradentate nitrilotriacetic acid (NTA) is one of the most common commercial ligands for chelating metal ions. The NTA-metal complexes show high and specific affinity toward hexahistidine tags. Such metal complexes have been widely utilized in protein separation and immobilization for diagnostic applications since most of commercialized proteins have been integrated with hexahistidine tags by synthetic or recombinant techniques. This review focused on the development of biosensors with NTA-metal complexes as the binding units, mainly including surface plasmon resonance, electrochemistry, fluorescence, colorimetry, surface-enhanced Raman scattering spectroscopy, chemiluminescence and so on.
Collapse
Affiliation(s)
- Lin Zhu
- College of Chemistry and Chemical Engineering, Anyang Normal University, Anyang 455000, China
| | - Yong Chang
- College of Chemistry and Chemical Engineering, Anyang Normal University, Anyang 455000, China
| | - Yingying Li
- College of Chemistry and Chemical Engineering, Anyang Normal University, Anyang 455000, China
| | - Mingyi Qiao
- College of Chemistry and Chemical Engineering, Anyang Normal University, Anyang 455000, China
| | - Lin Liu
- College of Chemistry and Chemical Engineering, Anyang Normal University, Anyang 455000, China
| |
Collapse
|
9
|
Khuda N, Somasundaram S, Urgunde AB, Easley CJ. Ionic Strength and Hybridization Position near Gold Electrodes Can Significantly Improve Kinetics in DNA-Based Electrochemical Sensors. ACS APPLIED MATERIALS & INTERFACES 2023; 15:5019-5027. [PMID: 36661270 PMCID: PMC10370289 DOI: 10.1021/acsami.2c22741] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/16/2023]
Abstract
A variety of electrochemical (EC) biosensors play critical roles in disease diagnostics. More recently, DNA-based EC sensors have been established as promising for detecting a wide range of analyte classes. Since most of these sensors rely on the high specificity of DNA hybridization for analyte binding or structural control, it is crucial to understand the kinetics of hybridization at the electrode surface. In this work, we have used methylene blue-labeled DNA strands to monitor the kinetics of DNA hybridization at the electrode surface with square-wave voltammetry. By varying the position of the double-stranded DNA segment relative to the electrode surface as well as the bulk solution's ionic strength (0.125-1.00 M), we observed significant interferences with DNA hybridization closer to the surface, with more substantial interference at lower ionic strength. As a demonstration of the effect, toehold-mediated strand displacement reactions were slowed and diminished close to the surface, while strategic placement of the DNA binding site improved reaction rates and yields. This work manifests that both the salt concentration and DNA hybridization site relative to the electrode are important factors to consider when designing DNA-based EC sensors that measure hybridization directly at the electrode surface.
Collapse
Affiliation(s)
- Niamat Khuda
- Department of Chemistry and Biochemistry, Auburn University, Auburn, AL 36849, USA
| | | | - Ajay B. Urgunde
- Department of Chemistry and Biochemistry, Auburn University, Auburn, AL 36849, USA
| | | |
Collapse
|
10
|
The Impact of Nε-Acryloyllysine Piperazides on the Conformational Dynamics of Transglutaminase 2. Int J Mol Sci 2023; 24:ijms24021650. [PMID: 36675164 PMCID: PMC9865645 DOI: 10.3390/ijms24021650] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Revised: 01/09/2023] [Accepted: 01/11/2023] [Indexed: 01/17/2023] Open
Abstract
In addition to the classic functions of proteins, such as acting as a biocatalyst or binding partner, the conformational states of proteins and their remodeling upon stimulation need to be considered. A prominent example of a protein that undergoes comprehensive conformational remodeling is transglutaminase 2 (TGase 2), the distinct conformational states of which are closely related to particular functions. Its involvement in various pathophysiological processes, including fibrosis and cancer, motivates the development of theranostic agents, particularly based on inhibitors that are directed toward the transamidase activity. In this context, the ability of such inhibitors to control the conformational dynamics of TGase 2 emerges as an important parameter, and methods to assess this property are in great demand. Herein, we describe the application of the switchSENSE® principle to detect conformational changes caused by three irreversibly binding Nε-acryloyllysine piperazides, which are suitable radiotracer candidates of TGase 2. The switchSENSE® technique is based on DNA levers actuated by alternating electric fields. These levers are immobilized on gold electrodes with one end, and at the other end of the lever, the TGase 2 is covalently bound. A novel computational method is introduced for describing the resulting lever motion to quantify the extent of stimulated conformational TGase 2 changes. Moreover, as a complementary biophysical method, native polyacrylamide gel electrophoresis was performed under similar conditions to validate the results. Both methods prove the occurrence of an irreversible shift in the conformational equilibrium of TGase 2, caused by the binding of the three studied Nε-acryloyllysine piperazides.
Collapse
|
11
|
Schulte C, Soldà A, Spänig S, Adams N, Bekić I, Streicher W, Heider D, Strasser R, Maric HM. Multivalent binding kinetics resolved by fluorescence proximity sensing. Commun Biol 2022; 5:1070. [PMID: 36207490 PMCID: PMC9546861 DOI: 10.1038/s42003-022-03997-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2022] [Accepted: 09/14/2022] [Indexed: 11/12/2022] Open
Abstract
Multivalent protein interactors are an attractive modality for probing protein function and exploring novel pharmaceutical strategies. The throughput and precision of state-of-the-art methodologies and workflows for the effective development of multivalent binders is currently limited by surface immobilization, fluorescent labelling and sample consumption. Using the gephyrin protein, the master regulator of the inhibitory synapse, as benchmark, we exemplify the application of Fluorescence proximity sensing (FPS) for the systematic kinetic and thermodynamic optimization of multivalent peptide architectures. High throughput synthesis of +100 peptides with varying combinatorial dimeric, tetrameric, and octameric architectures combined with direct FPS measurements resolved on-rates, off-rates, and dissociation constants with high accuracy and low sample consumption compared to three complementary technologies. The dataset and its machine learning-based analysis deciphered the relationship of specific architectural features and binding kinetics and thereby identified binders with unprecedented protein inhibition capacity; thus, highlighting the value of FPS for the rational engineering of multivalent inhibitors.
Collapse
Affiliation(s)
- Clemens Schulte
- Rudolf Virchow Center, Center for Integrative and Translational Bioimaging, University of Wuerzburg, Josef-Schneider-Str. 2, Germany, 97080, Wuerzburg, Germany
| | - Alice Soldà
- Dynamic Biosensors GmbH Germany, Lochhamer Strasse 15, 82152, Martinsried/Planegg, Germany
| | - Sebastian Spänig
- Department of Bioinformatics, Faculty of Mathematics and Computer Science, Philipps-University of Marburg, Hans-Meerwein-Strasse 6, 35043, Marburg, Germany
| | - Nathan Adams
- Nanotemper Technologies GmbH, Flößergasse 4, 81369, Munich, Germany
| | - Ivana Bekić
- Nanotemper Technologies GmbH, Flößergasse 4, 81369, Munich, Germany
| | - Werner Streicher
- Nanotemper Technologies GmbH, Flößergasse 4, 81369, Munich, Germany
| | - Dominik Heider
- Department of Bioinformatics, Faculty of Mathematics and Computer Science, Philipps-University of Marburg, Hans-Meerwein-Strasse 6, 35043, Marburg, Germany
| | - Ralf Strasser
- Dynamic Biosensors GmbH Germany, Lochhamer Strasse 15, 82152, Martinsried/Planegg, Germany
| | - Hans Michael Maric
- Rudolf Virchow Center, Center for Integrative and Translational Bioimaging, University of Wuerzburg, Josef-Schneider-Str. 2, Germany, 97080, Wuerzburg, Germany.
| |
Collapse
|
12
|
Zargartalebi H, Yousefi H, Flynn CD, Gomis S, Das J, Young TL, Chien E, Mubareka S, McGeer A, Wang H, Sargent EH, Nezhad AS, Kelley SO. Capillary-Assisted Molecular Pendulum Bioanalysis. J Am Chem Soc 2022; 144:18338-18349. [PMID: 36173381 DOI: 10.1021/jacs.2c06192] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
The development of robust biosensing strategies that can be easily implemented in everyday life remains a challenge for the future of modern biosensor research. While several reagentless approaches have attempted to address this challenge, they often achieve user-friendliness through sacrificing sensitivity or universality. While acceptable for certain applications, these trade-offs hinder the widespread adoption of reagentless biosensing technologies. Here, we report a novel approach to reagentless biosensing that achieves high sensitivity, rapid detection, and universality using the SARS-CoV-2 virus as a model target. Universality is achieved by using nanoscale molecular pendulums, which enables reagentless electrochemical biosensing through a variable antibody recognition element. Enhanced sensitivity and rapid detection are accomplished by incorporating the coffee-ring phenomenon into the sensing scheme, allowing for target preconcentration on a ring-shaped electrode. Using this approach, we obtained limits of detection of 1 fg/mL and 20 copies/mL for the SARS-CoV-2 nucleoproteins and viral particles, respectively. In addition, clinical sample analysis showed excellent agreement with Ct values from PCR-positive SARS-CoV-2 patients.
Collapse
Affiliation(s)
- Hossein Zargartalebi
- Department of Mechanical and Manufacturing Engineering, University of Calgary, Calgary, Alberta T2N 1N4, Canada.,Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, ON M5S 3M2, Canada
| | - Hanie Yousefi
- Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, ON M5S 3M2, Canada
| | - Connor D Flynn
- Department of Chemistry, Northwestern University, Evanston, Illinois 60208 United States.,Department of Chemistry, University of Toronto, Toronto, ON M5S 3M2, Canada
| | - Surath Gomis
- The Edward S. Rogers Sr. Department of Electrical and Computer Engineering, University of Toronto, Toronto, ON M5S 3G4, Canada
| | - Jagotamoy Das
- Department of Chemistry, Northwestern University, Evanston, Illinois 60208 United States
| | - Tiana L Young
- Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, ON M5S 3M2, Canada
| | - Emily Chien
- Sunnybrook Research Institute, Toronto, ON M4N 3N5, Canada
| | | | - Allison McGeer
- Department of Microbiology, Sinai Health System, Toronto, ON M5G 1X5, Canada
| | - Hansen Wang
- Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, ON M5S 3M2, Canada
| | - Edward H Sargent
- The Edward S. Rogers Sr. Department of Electrical and Computer Engineering, University of Toronto, Toronto, ON M5S 3G4, Canada
| | - Amir Sanati Nezhad
- Department of Mechanical and Manufacturing Engineering, University of Calgary, Calgary, Alberta T2N 1N4, Canada
| | - Shana O Kelley
- Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, ON M5S 3M2, Canada.,Department of Chemistry, Northwestern University, Evanston, Illinois 60208 United States.,Department of Biomedical Engineering, Northwestern University, Evanston, Illinois 60208, United States.,Department of Biochemistry and Molecular Genetics, Northwestern University, Chicago, Illinois 60611, United States
| |
Collapse
|
13
|
Ramotowska S, Spisz P, Brzeski J, Ciesielska A, Makowski M. Application of the SwitchSense Technique for the Study of Small Molecules’ (Ethidium Bromide and Selected Sulfonamide Derivatives) Affinity to DNA in Real Time. J Phys Chem B 2022; 126:7238-7251. [PMID: 36106569 PMCID: PMC9527753 DOI: 10.1021/acs.jpcb.2c03138] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
![]()
The discovery and introduction of the switchSense technique
in
the chemical laboratory have drawn well-deserved interest owing to
its wide range of applications. Namely, it can be used to determine
the diameter of proteins, alterations in their tertiary structures
(folding), and many other conformational changes that are important
from a biological point of view. The essence of this technique is
based on its ability to study of the interactions between an analyte
and a ligand in real time (in a buffer flow). Its simplicity, on the
other hand, is based on the use of a signaling system that provides
information about the ongoing interactions based on the changes in
the fluorescence intensity. This technique can be extremely advantageous
in the study of new pharmaceuticals. The design of compounds with
biological activity, as well as the determination of their molecular
targets and modes of interactions, is crucial in the search for new
drugs and the fight against drug resistance. This article presents
another possible application of the switchSense technique for the
study of the binding kinetics of small model molecules such as ethidium
bromide (EB) and selected sulfonamide derivatives with DNA in the
static and dynamic modes at three different temperatures (15, 25,
and 37 °C) each. The experimental results remain in very good
agreement with the molecular dynamics docking ones. These physicochemical
insights and applications obtained from the switchSense technique
allow for the design of an effective strategy for molecular interaction
assessments of small but pharmaceutically important molecules with
DNA.
Collapse
Affiliation(s)
- Sandra Ramotowska
- Faculty of Chemistry, University of Gdańsk, Wita Stwosza 63, Gdańsk 80-308, Poland
| | - Paulina Spisz
- Faculty of Chemistry, University of Gdańsk, Wita Stwosza 63, Gdańsk 80-308, Poland
| | - Jakub Brzeski
- Faculty of Chemistry, University of Gdańsk, Wita Stwosza 63, Gdańsk 80-308, Poland
- Department of Chemistry, University of Pittsburgh, Pittsburgh, Pennsylvania 15260, United States
| | | | - Mariusz Makowski
- Faculty of Chemistry, University of Gdańsk, Wita Stwosza 63, Gdańsk 80-308, Poland
| |
Collapse
|
14
|
Ren J, Xiong H, Huang C, Ji F, Jia L. An engineered peptide tag-specific nanobody for immunoaffinity chromatography application enabling efficient product recovery at mild conditions. J Chromatogr A 2022; 1676:463274. [DOI: 10.1016/j.chroma.2022.463274] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2022] [Revised: 06/15/2022] [Accepted: 06/22/2022] [Indexed: 10/17/2022]
|
15
|
Chu W, Prodromou R, Moore B, Elhanafi D, Kilgore R, Shastry S, Menegatti S. Development of Peptide Ligands for the Purification of α-1 Antitrypsin from Cell Culture Fluids. J Chromatogr A 2022; 1679:463363. [DOI: 10.1016/j.chroma.2022.463363] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2022] [Revised: 07/19/2022] [Accepted: 07/20/2022] [Indexed: 11/16/2022]
|
16
|
A Robust Assay to Monitor Ataxin-3 Amyloid Fibril Assembly. Cells 2022; 11:cells11121969. [PMID: 35741099 PMCID: PMC9222203 DOI: 10.3390/cells11121969] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Revised: 06/14/2022] [Accepted: 06/16/2022] [Indexed: 02/05/2023] Open
Abstract
Spinocerebellar ataxia type 3 (SCA3) is caused by the expansion of a glutamine repeat in the protein ataxin-3, which is deposited as intracellular aggregates in affected brain regions. Despite the controversial role of ataxin-3 amyloid structures in SCA3 pathology, the identification of molecules with the capacity to prevent aberrant self-assembly and stabilize functional conformation(s) of ataxin-3 is a key to the development of therapeutic solutions. Amyloid-specific kinetic assays are routinely used to measure rates of protein self-assembly in vitro and are employed during screening for fibrillation inhibitors. The high tendency of ataxin-3 to assemble into oligomeric structures implies that minor changes in experimental conditions can modify ataxin-3 amyloid assembly kinetics. Here, we determine the self-association rates of ataxin-3 and present a detailed study of the aggregation of normal and pathogenic ataxin-3, highlighting the experimental conditions that should be considered when implementing and validating ataxin-3 amyloid progress curves in different settings and in the presence of ataxin-3 interactors. This assay provides a unique and robust platform to screen for modulators of the first steps of ataxin-3 aggregation—a starting point for further studies with cell and animal models of SCA3.
Collapse
|
17
|
Tao Y, Chen L, Pan M, Zhu F, Zhu D. Tailored Biosensors for Drug Screening, Efficacy Assessment, and Toxicity Evaluation. ACS Sens 2021; 6:3146-3162. [PMID: 34516080 DOI: 10.1021/acssensors.1c01600] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Biosensors have been flourishing in the field of drug discovery with pronounced developments in the past few years. They facilitate the screening and discovery of innovative drugs. However, there is still a lack of critical reviews that compare the merits and shortcomings of these biosensors from a pharmaceutical point of view. This contribution presents a critical and up-to-date overview on the recent progress of tailored biosensors, including surface plasmon resonance, fluorescent, photoelectrochemical, and electrochemical systems with emphasis on their mechanisms and applications in drug screening, efficacy assessment, and toxicity evaluation. Multiple functional nanomaterials have also been incorporated into the biosensors. Representative examples of each type of biosensors are discussed in terms of design strategy, response mechanism, and potential applications. In the end, we also compare the results and summarize the major insights gained from the works, demonstrating the challenges and prospects of biosensors-assisted drug discovery.
Collapse
Affiliation(s)
- Yi Tao
- College of Pharmaceutical Science, Zhejiang University of Technology, Hangzhou 310014, China
| | - Lin Chen
- College of Pharmaceutical Science, Zhejiang University of Technology, Hangzhou 310014, China
| | - Meiling Pan
- College of Pharmaceutical Science, Zhejiang University of Technology, Hangzhou 310014, China
| | - Fei Zhu
- College of Pharmaceutical Science, Zhejiang University of Technology, Hangzhou 310014, China
| | - Dong Zhu
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210023, China
| |
Collapse
|
18
|
Soltermann F, Struwe WB, Kukura P. Label-free methods for optical in vitro characterization of protein-protein interactions. Phys Chem Chem Phys 2021; 23:16488-16500. [PMID: 34342317 PMCID: PMC8359934 DOI: 10.1039/d1cp01072g] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2021] [Accepted: 07/23/2021] [Indexed: 12/13/2022]
Abstract
Protein-protein interactions are involved in the regulation and function of the majority of cellular processes. As a result, much effort has been aimed at the development of methodologies capable of quantifying protein-protein interactions, with label-free methods being of particular interest due to the associated simplified workflows and minimisation of label-induced perturbations. Here, we review recent advances in optical technologies providing label-free in vitro measurements of affinities and kinetics. We provide an overview and comparison of existing techniques and their principles, discussing advantages, limitations, and recent applications.
Collapse
Affiliation(s)
- Fabian Soltermann
- Physical and Theoretical Chemistry, Department of Chemistry, University of OxfordUK
| | - Weston B. Struwe
- Physical and Theoretical Chemistry, Department of Chemistry, University of OxfordUK
| | - Philipp Kukura
- Physical and Theoretical Chemistry, Department of Chemistry, University of OxfordUK
| |
Collapse
|
19
|
Stein JAC, Ianeselli A, Braun D. Kinetic Microscale Thermophoresis for Simultaneous Measurement of Binding Affinity and Kinetics. Angew Chem Int Ed Engl 2021. [DOI: 10.1002/ange.202101261] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Affiliation(s)
- Julian A. C. Stein
- Systems Biophysics Department of Physics Ludwig-Maximilians-Universität München and Center for NanoScience Amalienstasse 54 80799 München Germany
| | - Alan Ianeselli
- Systems Biophysics Department of Physics Ludwig-Maximilians-Universität München and Center for NanoScience Amalienstasse 54 80799 München Germany
| | - Dieter Braun
- Systems Biophysics Department of Physics Ludwig-Maximilians-Universität München and Center for NanoScience Amalienstasse 54 80799 München Germany
| |
Collapse
|
20
|
Stein JAC, Ianeselli A, Braun D. Kinetic Microscale Thermophoresis for Simultaneous Measurement of Binding Affinity and Kinetics. Angew Chem Int Ed Engl 2021; 60:13988-13995. [PMID: 33793031 PMCID: PMC8251828 DOI: 10.1002/anie.202101261] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2021] [Revised: 03/16/2021] [Indexed: 12/11/2022]
Abstract
Microscale thermophoresis (MST) is a versatile technique to measure binding affinities of binder-ligand systems, based on the directional movement of molecules in a temperature gradient. We extended MST to measure binding kinetics as well as binding affinity in a single experiment by increasing the thermal dissipation of the sample. The kinetic relaxation fingerprints were derived from the fluorescence changes during thermodynamic re-equilibration of the sample after local heating. Using this method, we measured DNA hybridization on-rates and off-rates in the range 104 -106 m-1 s-1 and 10-4 -10-1 s-1 , respectively. We observed the expected exponential dependence of the DNA hybridization off-rates on salt concentration, strand length and inverse temperature. The measured on-rates showed a linear dependence on salt concentration and weak dependence on strand length and temperature. For biomolecular interactions with large enthalpic contributions, the kinetic MST technique offers a robust, cost-effective and immobilization-free determination of kinetic rates and binding affinity simultaneously, even in crowded solutions.
Collapse
Affiliation(s)
- Julian A C Stein
- Systems Biophysics, Department of Physics, Ludwig-Maximilians-Universität München and Center for NanoScience, Amalienstasse 54, 80799, München, Germany
| | - Alan Ianeselli
- Systems Biophysics, Department of Physics, Ludwig-Maximilians-Universität München and Center for NanoScience, Amalienstasse 54, 80799, München, Germany
| | - Dieter Braun
- Systems Biophysics, Department of Physics, Ludwig-Maximilians-Universität München and Center for NanoScience, Amalienstasse 54, 80799, München, Germany
| |
Collapse
|
21
|
Bioassay Development for Bispecific Antibodies-Challenges and Opportunities. Int J Mol Sci 2021; 22:ijms22105350. [PMID: 34069573 PMCID: PMC8160952 DOI: 10.3390/ijms22105350] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Revised: 05/14/2021] [Accepted: 05/15/2021] [Indexed: 12/25/2022] Open
Abstract
Antibody therapeutics are expanding with promising clinical outcomes, and diverse formats of antibodies are further developed and available for patients of the most challenging disease areas. Bispecific antibodies (BsAbs) have several significant advantages over monospecific antibodies by engaging two antigen targets. Due to the complicated mechanism of action, diverse structural variations, and dual-target binding, developing bioassays and other types of assays to characterize BsAbs is challenging. Developing bioassays for BsAbs requires a good understanding of the mechanism of action of the molecule, principles and applications of different bioanalytical methods, and phase-appropriate considerations per regulatory guidelines. Here, we review recent advances and case studies to provide strategies and insights for bioassay development for different types of bispecific molecules.
Collapse
|
22
|
Reagentless biomolecular analysis using a molecular pendulum. Nat Chem 2021; 13:428-434. [PMID: 33686229 DOI: 10.1038/s41557-021-00644-y] [Citation(s) in RCA: 66] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2020] [Accepted: 01/22/2021] [Indexed: 01/31/2023]
Abstract
The development of reagentless sensors that can detect molecular analytes in biological fluids could enable a broad range of applications in personalized health monitoring. However, only a limited set of molecular inputs can currently be detected using reagentless sensors. Here, we report a sensing mechanism that is compatible with the analysis of proteins that are important physiological markers of stress, allergy, cardiovascular health, inflammation and cancer. The sensing method is based on the motion of an inverted molecular pendulum that exhibits field-induced transport modulated by the presence of a bound analyte. We measure the sensor's electric field-mediated transport using the electron-transfer kinetics of an attached reporter molecule. Using time-resolved electrochemical measurements that enable unidirectional motion of our sensor, the presence of an analyte bound to our sensor complex can be tracked continuously in real time. We show that this sensing approach is compatible with making measurements in blood, saliva, urine, tears and sweat and that the sensors can collect data in situ in living animals.
Collapse
|
23
|
Macromolecular interactions in vitro, comparing classical and novel approaches. EUROPEAN BIOPHYSICS JOURNAL: EBJ 2021; 50:313-330. [PMID: 33792745 DOI: 10.1007/s00249-021-01517-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/09/2020] [Revised: 02/15/2021] [Accepted: 03/08/2021] [Indexed: 01/11/2023]
Abstract
Biophysical quantification of protein interactions is central to unveil the molecular mechanisms of cellular processes. Researchers can choose from a wide panel of biophysical methods that quantify molecular interactions in different ways, including both classical and more novel techniques. We report the outcome of an ARBRE-MOBIEU training school held in June 2019 in Gif-sur-Yvette, France ( https://mosbio.sciencesconf.org/ ). Twenty European students benefited from a week's training with theoretical and practical sessions in six complementary approaches: (1) analytical ultracentrifugation with or without a fluorescence detector system (AUC-FDS), (2) isothermal titration calorimetry (ITC), (3) size exclusion chromatography coupled to multi-angle light scattering (SEC-MALS), (4) bio-layer interferometry (BLI), (5) microscale thermophoresis (MST) and, (6) switchSENSE. They implemented all these methods on two examples of macromolecular interactions with nanomolar affinity: first, a protein-protein interaction between an artificial alphaRep binder, and its target protein, also an alphaRep; second, a protein-DNA interaction between a DNA repair complex, Ku70/Ku80 (hereafter called Ku), and its cognate DNA ligand. We report the approaches used to analyze the two systems under study and thereby showcase application of each of the six techniques. The workshop provided students with improved understanding of the advantages and limitations of different methods, enabling future choices concerning approaches that are most relevant or informative for specific kinds of sample and interaction.
Collapse
|
24
|
Müller-Landau H, Varela PF. Standard operation procedure for switchSENSE DRX systems. EUROPEAN BIOPHYSICS JOURNAL: EBJ 2021; 50:389-400. [PMID: 33772617 DOI: 10.1007/s00249-021-01519-3] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/08/2021] [Revised: 03/08/2021] [Accepted: 03/13/2021] [Indexed: 12/18/2022]
Abstract
There is currently a large panel of technologies available to address molecular interactions in vitro. Each technology presents individual advantages and drawbacks, and it becomes challenging to choose which technology will be best suited for a molecular interaction of interest. Approaches can be broadly categorized as either microfluidic surface-bound methods (such as Surface Plasmon Resonance (SPR) or switchSENSE) or in-solution methods (such as Isothermal Titration Calorimetry (ITC) or MicroScale Thermophoresis (MST)). In-solution methods are advantageous in terms of sample preparation and ease of use as none of the binding partners are subjected to immobilization. On the other hand, surface-based techniques require only small amounts of immobilized interaction partner and provide off-rate characterization as unbound analytes can be removed from the surface to observe analyte dissociation. Here, a standard operating procedure (SOP) for the switchSENSE method is presented, which aims to guide new users through the process of a switchSENSE measurement, covering sample preparation, instrument and biochip handling as well as data acquisition and analysis. This guide will help researchers decide whether switchSENSE is the right method for their application as well as supporting novice users to get the most information out of a switchSENSE measurement. switchSENSE technology offers the unique advantage of a controlled DNA-based ligand surface within a microfluidic channel which allows the user to distribute specifically up to two different ligand molecules on the surface at a customized density and ratio. The technology offers multi-parameter characterization of binding kinetics, affinity, enzymatic activity, and changes in protein conformation.
Collapse
Affiliation(s)
| | - Paloma Fernández Varela
- Université Paris-Saclay, CEA, CNRS, Institute for Integrative Biology of the Cell (I2BC), 91198, Gif-sur-Yvette, France.
| |
Collapse
|
25
|
Adler S, Motiei L, Mankovski N, Cohen H, Margulies D. Fluorescent Labelling of Cell Surface Proteins on a Solid Support. Isr J Chem 2021. [DOI: 10.1002/ijch.202100028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Affiliation(s)
- Sean Adler
- Department of Chemical and Structural Biology Weizmann Institute of Science Rehovot 7610001 Israel
| | - Leila Motiei
- Department of Chemical and Structural Biology Weizmann Institute of Science Rehovot 7610001 Israel
| | - Naama Mankovski
- Department of Chemical and Structural Biology Weizmann Institute of Science Rehovot 7610001 Israel
| | - Hagai Cohen
- Department of Chemical Research Support Weizmann Institute of Science Rehovot 7610001 Israel
| | - David Margulies
- Department of Chemical and Structural Biology Weizmann Institute of Science Rehovot 7610001 Israel
| |
Collapse
|
26
|
Hatai J, Prasad PK, Lahav-Mankovski N, Oppenheimer-Low N, Unger T, Sirkis YF, Dadosh T, Motiei L, Margulies D. Assessing changes in the expression levels of cell surface proteins with a turn-on fluorescent molecular probe. Chem Commun (Camb) 2021; 57:1875-1878. [PMID: 33427257 DOI: 10.1039/d0cc07095e] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Tri-nitrilotriacetic acid (NTA)-based fluorescent probes were developed and used to image His-tagged-labelled outer membrane protein C (His-OmpC) in live Escherichia coli. One of these probes was designed to light up upon binding, which provided the means to assess changes in the His-OmpC expression levels by taking a simple fluorescence spectrum.
Collapse
Affiliation(s)
- Joydev Hatai
- Department of Organic Chemistry, Weizmann Institute of Science, Rehovot 7610001, Israel.
| | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Schenckbecher E, Bec G, Sakamoto T, Meyer B, Ennifar E. Biophysical Studies of the Binding of Viral RNA with the 80S Ribosome Using switchSENSE. Methods Mol Biol 2021; 2263:341-350. [PMID: 33877606 DOI: 10.1007/978-1-0716-1197-5_15] [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: 06/12/2023]
Abstract
Translation initiation, in both eukaryotes and bacteria, requires essential elements such as mRNA, ribosome , initiator tRNA, and a set of initiation factors. For each domain of life, canonical mechanisms and signals are observed to initiate protein synthesis. However, other initiation mechanism can be used, especially in viral mRNAs. Some viruses hijack cellular machinery to translate some of their mRNAs through a noncanonical initiation pathway using internal ribosome entry site (IRES), a highly structured RNAs which can directly recruit the ribosome with a restricted set of initiation factors, and in some cases even without cap and initiator tRNA. In this chapter, we describe the use of biosensors relying on electro-switchable nanolevers using the switchSENSE® technology, to investigate kinetics of the intergenic (IGR) IRES of the cricket paralysis virus (CrPV) binding to 80S yeast ribosome . This study provides a proof of concept for the application of this method on large complexes.
Collapse
MESH Headings
- Biophysical Phenomena
- Biosensing Techniques/methods
- Dicistroviridae/physiology
- Internal Ribosome Entry Sites
- Kinetics
- Models, Molecular
- Proof of Concept Study
- Protein Biosynthesis
- RNA, Viral/chemistry
- RNA, Viral/metabolism
- Ribosome Subunits, Large, Eukaryotic/chemistry
- Ribosome Subunits, Large, Eukaryotic/metabolism
- Ribosome Subunits, Small, Eukaryotic/chemistry
- Ribosome Subunits, Small, Eukaryotic/metabolism
- Saccharomyces cerevisiae/metabolism
- Saccharomyces cerevisiae Proteins/chemistry
- Saccharomyces cerevisiae Proteins/metabolism
Collapse
Affiliation(s)
- Emma Schenckbecher
- Institut de Biologie Moléculaire et Cellulaire, Université de Strasbourg, CNRS, Strasbourg, France
| | - Guillaume Bec
- Institut de Biologie Moléculaire et Cellulaire, Université de Strasbourg, CNRS, Strasbourg, France
| | - Taiichi Sakamoto
- Institut de Biologie Moléculaire et Cellulaire, Université de Strasbourg, CNRS, Strasbourg, France
- Department of Life Science, Faculty of Advanced Engineering, Chiba Institute of Technology, Chiba, Japan
| | - Benoit Meyer
- Institut de Biologie Moléculaire et Cellulaire, Université de Strasbourg, CNRS, Strasbourg, France
| | - Eric Ennifar
- Institut de Biologie Moléculaire et Cellulaire, Université de Strasbourg, CNRS, Strasbourg, France.
| |
Collapse
|
28
|
Prasad PK, Motiei L, Margulies D. Steps toward enhancing the fluorescence of small-molecule-based protein labels using supramolecular hosts. RESULTS IN CHEMISTRY 2021. [DOI: 10.1016/j.rechem.2021.100134] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022] Open
|
29
|
Vicente CM, Girardet JM, Hôtel L, Aigle B. Molecular Dynamics to Elucidate the DNA-Binding Activity of AlpZ, a Member of the Gamma-Butyrolactone Receptor Family in Streptomyces ambofaciens. Front Microbiol 2020; 11:1255. [PMID: 32714286 PMCID: PMC7343708 DOI: 10.3389/fmicb.2020.01255] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2019] [Accepted: 05/18/2020] [Indexed: 11/13/2022] Open
Affiliation(s)
- Cláudia M. Vicente
- Université de Lorraine, INRAE, DynAMic, Nancy, France
- *Correspondence: Cláudia M. Vicente,
| | | | | | - Bertrand Aigle
- Université de Lorraine, INRAE, DynAMic, Nancy, France
- Bertrand Aigle,
| |
Collapse
|
30
|
Daub H, Traxler L, Ismajli F, Groitl B, Itzen A, Rant U. The trimer to monomer transition of Tumor Necrosis Factor-Alpha is a dynamic process that is significantly altered by therapeutic antibodies. Sci Rep 2020; 10:9265. [PMID: 32518229 PMCID: PMC7283243 DOI: 10.1038/s41598-020-66123-5] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2019] [Accepted: 05/12/2020] [Indexed: 11/09/2022] Open
Abstract
The cytokine tumor necrosis factor-alpha (TNF-α) readily forms homotrimers at sub-nM concentrations to promote inflammation. For the treatment of inflammatory diseases with upregulated levels of TNF-α, a number of therapeutic antibodies are currently used as scavengers to reduce the active TNF-α concentration in patients. Despite their clinical success, the mode-of-action of different antibody formats with regard to a stabilization of the trimeric state is not entirely understood. Here, we use a biosensor with dynamic nanolevers to analyze the monomeric and trimeric states of TNF-α together with the binding kinetics of therapeutic biologics. The intrinsic trimer-to-monomer decay rate k = 1.7 × 10−3 s−1 could be measured directly using a microfluidic system, and antibody binding affinities were analyzed in the pM range. Trimer stabilization effects are quantified for Adalimumab, Infliximab, Etanercept, Certolizumab, Golimumab for bivalent and monovalent binding formats. Clear differences in trimer stabilization are observed, which may provide a deeper insight into the mode-of-action of TNF-α scavengers.
Collapse
Affiliation(s)
- Herwin Daub
- Dynamic Biosensors GmbH, Lochhamer Strasse 15, 82152, Martinsried, Germany. .,Center for Integrated Protein Science Munich, Technische Universität München, Department Chemistry, Lichtenbergstrasse 4, 85748, Garching, Germany.
| | - Lukas Traxler
- Dynamic Biosensors GmbH, Lochhamer Strasse 15, 82152, Martinsried, Germany
| | - Fjolla Ismajli
- Dynamic Biosensors GmbH, Lochhamer Strasse 15, 82152, Martinsried, Germany
| | - Bastian Groitl
- Dynamic Biosensors GmbH, Lochhamer Strasse 15, 82152, Martinsried, Germany
| | - Aymelt Itzen
- Center for Integrated Protein Science Munich, Technische Universität München, Department Chemistry, Lichtenbergstrasse 4, 85748, Garching, Germany.,Department of Biochemistry and Signaltransduction, University Medical Centre Hamburg-Eppendorf (UKE), Martinistrasse 52, 20246, Hamburg, Germany
| | - Ulrich Rant
- Dynamic Biosensors GmbH, Lochhamer Strasse 15, 82152, Martinsried, Germany
| |
Collapse
|
31
|
Schiedel M, Daub H, Itzen A, Jung M. Validation of the Slow Off-Kinetics of Sirtuin-Rearranging Ligands (SirReals) by Means of Label-Free Electrically Switchable Nanolever Technology. Chembiochem 2020; 21:1161-1166. [PMID: 31692222 PMCID: PMC7217041 DOI: 10.1002/cbic.201900527] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2019] [Revised: 10/31/2019] [Indexed: 12/17/2022]
Abstract
We have discovered the sirtuin-rearranging ligands (SirReals) to be highly potent and selective inhibitors of the NAD+ -dependent lysine deacetylase Sirt2. Using a biotinylated SirReal in combination with biolayer interferometry, we previously observed a slow dissociation rate of the inhibitor-enzyme complex; this had been postulated to be the key to the high affinity and selectivity of SirReals. However, to attach biotin to the SirReal core, we introduced a triazole as a linking moiety; this was shown by X-ray co-crystallography to interact with Arg97 of the cofactor binding loop. Herein, we aim to elucidate whether the observed long residence time of the SirReals is induced mainly by triazole incorporation or is an inherent characteristic of the SirReal inhibitor core. We used the novel label-free switchSENSE® technology, which is based on electrically switchable DNA nanolevers, to prove that the long residence time of the SirReals is indeed caused by the core scaffold.
Collapse
Affiliation(s)
- Matthias Schiedel
- Department of Chemistry and Pharmacy, Medicinal Chemistry, Friedrich Alexander University Erlangen-Nürnberg, Nikolaus-Fiebiger-Strasse 10, 91058, Erlangen, Germany.,Institute of Pharmaceutical Sciences, Albert-Ludwigs-Universität Freiburg, Albertstrasse 25, 79104, Freiburg im Breisgau, Germany
| | - Herwin Daub
- Dynamic Biosensors GmbH, Lochhamer Strasse 15, 82152, Martinsried, Germany.,Center for Integrated Protein Science Munich, Technische Universität München, Department of Chemistry, Lichtenbergstrasse 4, 85748, Garching, Germany
| | - Aymelt Itzen
- Center for Integrated Protein Science Munich, Technische Universität München, Department of Chemistry, Lichtenbergstrasse 4, 85748, Garching, Germany.,Department of Biochemistry and Signal Transduction, University Medical Centre Hamburg-Eppendorf (UKE), Martinistrasse 52, 20246, Hamburg, Germany
| | - Manfred Jung
- Institute of Pharmaceutical Sciences, Albert-Ludwigs-Universität Freiburg, Albertstrasse 25, 79104, Freiburg im Breisgau, Germany
| |
Collapse
|
32
|
A DNA-Based Biosensor Assay for the Kinetic Characterization of Ion-Dependent Aptamer Folding and Protein Binding. Molecules 2019; 24:molecules24162877. [PMID: 31398834 PMCID: PMC6718989 DOI: 10.3390/molecules24162877] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2019] [Revised: 08/02/2019] [Accepted: 08/05/2019] [Indexed: 11/16/2022] Open
Abstract
Therapeutic and diagnostic nucleic acid aptamers are designed to bind tightly and specifically to their target. The combination of structural and kinetic analyses of aptamer interactions has gained increasing importance. Here, we present a fluorescence-based switchSENSE aptasensor for the detailed kinetic characterization of aptamer-analyte interaction and aptamer folding, employing the thrombin-binding aptamer (TBA) as a model system. Thrombin-binding aptamer folding into a G-quadruplex and its binding to thrombin strongly depend on the type and concentration of ions present in solution. We observed conformational changes induced by cations in real-time and determined the folding and unfolding kinetics of the aptamer. The aptamer's affinity for K+ was found to be more than one order of magnitude higher than for other cations (K+ > NH4+ >> Na+ > Li+). The aptamer's affinity to its protein target thrombin in the presence of different cations followed the same trend but differed by more than three orders of magnitude (KD = 0.15 nM to 250 nM). While the stability (kOFF) of the thrombin-TBA complex was similar in all conditions, the cation type strongly influenced the association rate (kON). These results demonstrated that protein-aptamer binding is intrinsically related to the correct aptamer fold and, hence, to the presence of stabilizing ions. Because fast binding kinetics with on-rates exceeding 108 M-1s-1 can be quantified, and folding-related phenomena can be directly resolved, switchSENSE is a useful analytical tool for in-depth characterization of aptamer-ion and aptamer-protein interactions.
Collapse
|
33
|
Häußermann K, Young G, Kukura P, Dietz H. Dissecting FOXP2 Oligomerization and DNA Binding. Angew Chem Int Ed Engl 2019; 58:7662-7667. [PMID: 30887622 PMCID: PMC6986896 DOI: 10.1002/anie.201901734] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2019] [Indexed: 01/17/2023]
Abstract
Protein–protein and protein–substrate interactions are critical to function and often depend on factors that are difficult to disentangle. Herein, a combined biochemical and biophysical approach, based on electrically switchable DNA biochips and single‐molecule mass analysis, was used to characterize the DNA binding and protein oligomerization of the transcription factor, forkhead box protein P2 (FOXP2). FOXP2 contains domains commonly involved in nucleic‐acid binding and protein oligomerization, such as a C2H2‐zinc finger (ZF), and a leucine zipper (LZ), whose roles in FOXP2 remain largely unknown. We found that the LZ mediates FOXP2 dimerization via coiled‐coil formation but also contributes to DNA binding. The ZF contributes to protein dimerization when the LZ coiled‐coil is intact, but it is not involved in DNA binding. The forkhead domain (FHD) is the key driver of DNA binding. Our data contributes to understanding the mechanisms behind the transcriptional activity of FOXP2.
Collapse
Affiliation(s)
- Katharina Häußermann
- Physik Department & Munich School of Bioengineering, Technische Universität München, Am Coulombwall 4a, 85784, Garching, Germany
| | - Gavin Young
- Department of Chemistry, Physical and Theoretical Chemistry Laboratory, University of Oxford, South Parks Road, Oxford, OX1 3QZ, UK
| | - Philipp Kukura
- Department of Chemistry, Physical and Theoretical Chemistry Laboratory, University of Oxford, South Parks Road, Oxford, OX1 3QZ, UK
| | - Hendrik Dietz
- Physik Department & Munich School of Bioengineering, Technische Universität München, Am Coulombwall 4a, 85784, Garching, Germany
| |
Collapse
|
34
|
Häußermann K, Young G, Kukura P, Dietz H. Dissecting FOXP2 Oligomerization and DNA Binding. Angew Chem Int Ed Engl 2019. [DOI: 10.1002/ange.201901734] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Affiliation(s)
- Katharina Häußermann
- Physik Department & Munich School of BioengineeringTechnische Universität München Am Coulombwall 4a 85784 Garching Germany
| | - Gavin Young
- Department of ChemistryPhysical and Theoretical Chemistry LaboratoryUniversity of Oxford South Parks Road Oxford OX1 3QZ UK
| | - Philipp Kukura
- Department of ChemistryPhysical and Theoretical Chemistry LaboratoryUniversity of Oxford South Parks Road Oxford OX1 3QZ UK
| | - Hendrik Dietz
- Physik Department & Munich School of BioengineeringTechnische Universität München Am Coulombwall 4a 85784 Garching Germany
| |
Collapse
|
35
|
Wieneke R, Tampé R. Multivalent Chelators for In Vivo Protein Labeling. Angew Chem Int Ed Engl 2019; 58:8278-8290. [PMID: 30919542 DOI: 10.1002/anie.201811293] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2018] [Indexed: 01/09/2023]
Abstract
With the advent of single-molecule methods, chemoselective and site-specific labeling of proteins evolved to become a central aspect in chemical biology as well as cell biology. Protein labeling demands high specificity, rapid as well as efficient conjugation, while maintaining low concentration and biocompatibility under physiological conditions. Generic methods that do not interfere with the function, dynamics, subcellular localization of proteins, and crosstalk with other factors are crucial to probe and image proteins in vitro and in living cells. Alternatives to enzyme-based tags or autofluorescent proteins are short peptide-based recognition tags. These tags provide high specificity, enhanced binding rates, bioorthogonality, and versatility. Here, we report on recent applications of multivalent chelator heads, recognizing oligohistidine-tagged proteins. The striking features of this system has facilitated the analysis of protein complexes by single-molecule approaches.
Collapse
Affiliation(s)
- Ralph Wieneke
- Institute of Biochemistry, Biocenter, Goethe University Frankfurt, Max-von-Laue Str. 9, 60438, Frankfurt/M., Germany
| | - Robert Tampé
- Institute of Biochemistry, Biocenter, Goethe University Frankfurt, Max-von-Laue Str. 9, 60438, Frankfurt/M., Germany
| |
Collapse
|
36
|
Wieneke R, Tampé R. Multivalent Chelators for In Vivo Protein Labeling. Angew Chem Int Ed Engl 2019. [DOI: 10.1002/ange.201811293] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Affiliation(s)
- Ralph Wieneke
- Institute of BiochemistryBiocenterGoethe University Frankfurt Max-von-Laue Str. 9 60438 Frankfurt/M. Germany
| | - Robert Tampé
- Institute of BiochemistryBiocenterGoethe University Frankfurt Max-von-Laue Str. 9 60438 Frankfurt/M. Germany
| |
Collapse
|
37
|
Aman K, Padroni G, Parkinson JA, Welte T, Burley GA. Structural and Kinetic Profiling of Allosteric Modulation of Duplex DNA Induced by DNA-Binding Polyamide Analogues. Chemistry 2019; 25:2757-2763. [PMID: 30407668 PMCID: PMC6468288 DOI: 10.1002/chem.201805338] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2018] [Indexed: 12/20/2022]
Abstract
A combined structural and quantitative biophysical profile of the DNA binding affinity, kinetics and sequence-selectivity of hairpin polyamide analogues is described. DNA duplexes containing either target polyamide binding sites or mismatch sequences are immobilized on a microelectrode surface. Quantitation of the DNA binding profile of polyamides containing N-terminal 1-alkylimidazole (Im) units exhibit picomolar binding affinities for their target sequences, whereas 5-alkylthiazole (Nt) units are an order of magnitude lower (low nanomolar). Comparative NMR structural analyses of the polyamide series shows that the steric bulk distal to the DNA-binding face of the hairpin iPr-Nt polyamide plays an influential role in the allosteric modulation of the overall DNA duplex structure. This combined kinetic and structural study provides a foundation to develop next-generation hairpin designs where the DNA-binding profile of polyamides is reconciled with their physicochemical properties.
Collapse
Affiliation(s)
- Khalid Aman
- Department of Pure and Applied ChemistryUniversity of Strathclyde, Thomas Graham Building295 Cathedral StreetGlasgowG1 1XLUK
| | - Giacomo Padroni
- Department of Pure and Applied ChemistryUniversity of Strathclyde, Thomas Graham Building295 Cathedral StreetGlasgowG1 1XLUK
| | - John A. Parkinson
- Department of Pure and Applied ChemistryUniversity of Strathclyde, Thomas Graham Building295 Cathedral StreetGlasgowG1 1XLUK
| | | | - Glenn A. Burley
- Department of Pure and Applied ChemistryUniversity of Strathclyde, Thomas Graham Building295 Cathedral StreetGlasgowG1 1XLUK
| |
Collapse
|
38
|
Yang Y, Back CR, Gräwert MA, Wahid AA, Denton H, Kildani R, Paulin J, Wörner K, Kaiser W, Svergun DI, Sartbaeva A, Watts AG, Marchbank KJ, van den Elsen JMH. Utilization of Staphylococcal Immune Evasion Protein Sbi as a Novel Vaccine Adjuvant. Front Immunol 2019; 9:3139. [PMID: 30687332 PMCID: PMC6336717 DOI: 10.3389/fimmu.2018.03139] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2018] [Accepted: 12/19/2018] [Indexed: 02/04/2023] Open
Abstract
Co-ligation of the B cell antigen receptor with complement receptor 2 on B-cells via a C3d-opsonised antigen complex significantly lowers the threshold required for B cell activation. Consequently, fusions of antigens with C3d polymers have shown great potential in vaccine design. However, these linear arrays of C3d multimers do not mimic the natural opsonisation of antigens with C3d. Here we investigate the potential of using the unique complement activating characteristics of Staphylococcal immune-evasion protein Sbi to develop a pro-vaccine approach that spontaneously coats antigens with C3 degradation products in a natural way. We show that Sbi rapidly triggers the alternative complement pathway through recruitment of complement regulators, forming tripartite complexes that act as competitive antagonists of factor H, resulting in enhanced complement consumption. These functional results are corroborated by the structure of the complement activating Sbi-III-IV:C3d:FHR-1 complex. Finally, we demonstrate that Sbi, fused with Mycobacterium tuberculosis antigen Ag85b, causes efficient opsonisation with C3 fragments, thereby enhancing the immune response significantly beyond that of Ag85b alone, providing proof of concept for our pro-vaccine approach.
Collapse
Affiliation(s)
- Yi Yang
- Department of Biology and Biochemistry, University of Bath, Bath, United Kingdom
| | - Catherine R Back
- Department of Biology and Biochemistry, University of Bath, Bath, United Kingdom
| | - Melissa A Gräwert
- Hamburg Unit, European Molecular Biology Laboratory, Deutsches Elektronen-Synchrotron, Hamburg, Germany
| | - Ayla A Wahid
- Department of Biology and Biochemistry, University of Bath, Bath, United Kingdom
| | - Harriet Denton
- Institute of Cellular Medicine, Newcastle University, Newcastle-upon-Tyne, United Kingdom
| | - Rebecca Kildani
- Institute of Cellular Medicine, Newcastle University, Newcastle-upon-Tyne, United Kingdom
| | - Joshua Paulin
- Institute of Cellular Medicine, Newcastle University, Newcastle-upon-Tyne, United Kingdom
| | | | | | - Dmitri I Svergun
- Hamburg Unit, European Molecular Biology Laboratory, Deutsches Elektronen-Synchrotron, Hamburg, Germany
| | - Asel Sartbaeva
- Department of Chemistry, University of Bath, Bath, United Kingdom
| | - Andrew G Watts
- Department of Pharmacy and Pharmacology, University of Bath, Bath, United Kingdom
| | - Kevin J Marchbank
- Institute of Cellular Medicine, Newcastle University, Newcastle-upon-Tyne, United Kingdom
| | | |
Collapse
|
39
|
Webster MW, Stowell JA, Passmore LA. RNA-binding proteins distinguish between similar sequence motifs to promote targeted deadenylation by Ccr4-Not. eLife 2019; 8:40670. [PMID: 30601114 PMCID: PMC6340701 DOI: 10.7554/elife.40670] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2018] [Accepted: 12/28/2018] [Indexed: 12/17/2022] Open
Abstract
The Ccr4-Not complex removes mRNA poly(A) tails to regulate eukaryotic mRNA stability and translation. RNA-binding proteins contribute to specificity by interacting with both Ccr4-Not and target mRNAs, but this is not fully understood. Here, we reconstitute accelerated and selective deadenylation of RNAs containing AU-rich elements (AREs) and Pumilio-response elements (PREs). We find that the fission yeast homologues of Tristetraprolin/TTP and Pumilio/Puf (Zfs1 and Puf3) interact with Ccr4-Not via multiple regions within low-complexity sequences, suggestive of a multipartite interface that extends beyond previously defined interactions. Using a two-color assay to simultaneously monitor poly(A) tail removal from different RNAs, we demonstrate that Puf3 can distinguish between RNAs of very similar sequence. Analysis of binding kinetics reveals that this is primarily due to differences in dissociation rate constants. Consequently, motif quality is a major determinant of mRNA stability for Puf3 targets in vivo and can be used for the prediction of mRNA targets. When a cell needs to make a particular protein, it first copies the instructions from the matching gene into a molecule known as a messenger RNA (or an mRNA for short). The more mRNA copies it makes, the more protein it can produce. A simple way to control protein production is to raise or lower the number of these mRNA messages, and living cells have lots of ways to make this happen. One method involves codes built into the mRNAs themselves. The mRNAs can carry short sequences of genetic letters that can trigger their own destruction. Known as “destabilising motifs”, these sequences attract the attention of a group of proteins called Ccr4-Not. Together these proteins shorten the end of the mRNAs, preparing the molecules for degradation. But how does Ccr4-Not choose which mRNAs to target? Different mRNAs carry different destabilising motifs. This means that when groups of mRNAs all carry the same motif, the cell can destroy them all together. This allows the cell to switch networks of related genes off together without affecting the mRNAs it still needs. What is puzzling is that the destabilising motifs that control different groups of mRNAs can be very similar, and scientists do not yet know how Ccr4-Not can tell the difference, or what triggers it to start breaking down groups of mRNAs. To find out, Webster et al. recreated the system in the laboratory using purified molecules. The test-tube system confirmed previous suggestions that a protein called Puf3 forms a bridge between Ccr4-Not and mRNAs. It acts as a tether, recognising a destabilising motif and linking it to Ccr4-Not. Labelling different mRNAs with two colours of fluorescent dye showed how Puf3 helps the cell to choose which to destroy. Puf3 allows Ccr4-Not to select specific mRNAs from a mixture of molecules. Puf3 could distinguish between mRNAs that differed in a single letter of genetic code. When it matched with the wrong mRNA, it disconnected much faster than when it matched with the right one, preventing Ccr4-Not from linking up. The ability to destroy specific mRNA messages is critical for cell survival. It happens when cells divide, during immune responses such as inflammation, and in early development. Understanding the targets of tethers like Puf3 could help scientists to predict which genes will switch off and when. This could reveal genes that work together, helping to unravel their roles inside cells.
Collapse
Affiliation(s)
| | | | - Lori A Passmore
- MRC Laboratory of Molecular Biology, Cambridge, United Kingdom
| |
Collapse
|
40
|
Hampel PA, Strasser R, Fischer F, Rant U. Assembly and Characterization of a Slingshot DNA Nanostructure for the Analysis of Bivalent and Bispecific Analytes with Biosensors. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2018; 34:14796-14801. [PMID: 30269507 DOI: 10.1021/acs.langmuir.8b02124] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
The characterization of novel therapeutic antibodies with multivalent or multispecific binding sites requires new measurement modalities for biosensors, to discriminate the engagement of antigens via one, two, or even more binding moieties. The presentation of antigens on a sensor surface in a well-controlled spatial arrangement is a prerequisite for the successful interpretation of binding kinetics measurements of multivalent analytes, but the adjustment of defined distances between immobilized ligands is difficult to achieve in state-of-the-art biosensor systems. Here, we introduce a simple DNA nanostructure resembling a slingshot, which can be configured with two identical or two different antigens (bivalent or bispecific), which are spaced at a defined distance. We characterize the slingshot structure with a chip-based biosensor using electrically switchable DNA nanolevers and demonstrate that bivalent and monovalent antibodies selectively interact with slingshots that have been functionalized with two identical or two different antigens, respectively. The dissociation kinetics are quantified in real-time measurements and we show that the slingshot structure enables a clear differentiation between affinity and avidity effects.
Collapse
Affiliation(s)
- Paul A Hampel
- Dynamic Biosensors GmbH , Martinsried 82152 , Germany
- Technische Universität München , Munich 80333 , Germany
| | - Ralf Strasser
- Dynamic Biosensors GmbH , Martinsried 82152 , Germany
| | - Frank Fischer
- Dynamic Biosensors GmbH , Martinsried 82152 , Germany
| | - Ulrich Rant
- Dynamic Biosensors GmbH , Martinsried 82152 , Germany
| |
Collapse
|
41
|
Preclinical Development of a Novel, Orally-Administered Anti-Tumour Necrosis Factor Domain Antibody for the Treatment of Inflammatory Bowel Disease. Sci Rep 2018; 8:4941. [PMID: 29563546 PMCID: PMC5862986 DOI: 10.1038/s41598-018-23277-7] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2018] [Accepted: 03/07/2018] [Indexed: 02/08/2023] Open
Abstract
TNFα is an important cytokine in inflammatory bowel disease. V565 is a novel anti-TNFα domain antibody developed for oral administration in IBD patients, derived from a llama domain antibody and engineered to enhance intestinal protease resistance. V565 activity was evaluated in TNFα-TNFα receptor-binding ELISAs as well as TNFα responsive cellular assays and demonstrated neutralisation of both soluble and membrane TNFα with potencies similar to those of adalimumab. Although sensitive to pepsin, V565 retained activity after lengthy incubations with trypsin, chymotrypsin, and pancreatin, as well as mouse small intestinal and human ileal and faecal supernatants. In orally dosed naïve and DSS colitis mice, high V565 concentrations were observed in intestinal contents and faeces and immunostaining revealed V565 localisation in mouse colon tissue. V565 was detected by ELISA in post-dose serum of colitis mice, but not naïve mice, demonstrating penetration of disrupted epithelium. In an ex vivo human IBD tissue culture model, V565 inhibition of tissue phosphoprotein levels and production of inflammatory cytokine biomarkers was similar to infliximab, demonstrating efficacy when present at the disease site. Taken together, results of these studies provide confidence that oral V565 dosing will be therapeutic in IBD patients where the mucosal epithelial barrier is compromised.
Collapse
|
42
|
Sedlak SM, Bauer MS, Kluger C, Schendel LC, Milles LF, Pippig DA, Gaub HE. Monodisperse measurement of the biotin-streptavidin interaction strength in a well-defined pulling geometry. PLoS One 2017; 12:e0188722. [PMID: 29206886 PMCID: PMC5716544 DOI: 10.1371/journal.pone.0188722] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2017] [Accepted: 11/07/2017] [Indexed: 11/18/2022] Open
Abstract
The widely used interaction of the homotetramer streptavidin with the small molecule biotin has been intensively studied by force spectroscopy and has become a model system for receptor ligand interaction. However, streptavidin's tetravalency results in diverse force propagation pathways through the different binding interfaces. This multiplicity gives rise to polydisperse force spectroscopy data. Here, we present an engineered monovalent streptavidin tetramer with a single cysteine in its functional subunit that allows for site-specific immobilization of the molecule, orthogonal to biotin binding. Functionality of streptavidin and its binding properties for biotin remain unaffected. We thus created a stable and reliable molecular anchor with a unique high-affinity binding site for biotinylated molecules or nanoparticles, which we expect to be useful for many single-molecule applications. To characterize the mechanical properties of the bond between biotin and our monovalent streptavidin, we performed force spectroscopy experiments using an atomic force microscope. We were able to conduct measurements at the single-molecule level with 1:1-stoichiometry and a well-defined geometry, in which force exclusively propagates through a single subunit of the streptavidin tetramer. For different force loading rates, we obtained narrow force distributions of the bond rupture forces ranging from 200 pN at 1,500 pN/s to 230 pN at 110,000 pN/s. The data are in very good agreement with the standard Bell-Evans model with a single potential barrier at Δx0 = 0.38 nm and a zero-force off-rate koff,0 in the 10-6 s-1 range.
Collapse
Affiliation(s)
- Steffen M. Sedlak
- Lehrstuhl für Angewandte Physik and Center for NanoScience (CeNS), Ludwig-Maximilians-Universität München, Munich, Germany
| | - Magnus S. Bauer
- Lehrstuhl für Angewandte Physik and Center for NanoScience (CeNS), Ludwig-Maximilians-Universität München, Munich, Germany
| | - Carleen Kluger
- Lehrstuhl für Angewandte Physik and Center for NanoScience (CeNS), Ludwig-Maximilians-Universität München, Munich, Germany
| | - Leonard C. Schendel
- Lehrstuhl für Angewandte Physik and Center for NanoScience (CeNS), Ludwig-Maximilians-Universität München, Munich, Germany
| | - Lukas F. Milles
- Lehrstuhl für Angewandte Physik and Center for NanoScience (CeNS), Ludwig-Maximilians-Universität München, Munich, Germany
| | - Diana A. Pippig
- Lehrstuhl für Angewandte Physik and Center for NanoScience (CeNS), Ludwig-Maximilians-Universität München, Munich, Germany
| | - Hermann E. Gaub
- Lehrstuhl für Angewandte Physik and Center for NanoScience (CeNS), Ludwig-Maximilians-Universität München, Munich, Germany
- * E-mail:
| |
Collapse
|
43
|
Wasserberg D, Cabanas-Danés J, Prangsma J, O’Mahony S, Cazade PA, Tromp E, Blum C, Thompson D, Huskens J, Subramaniam V, Jonkheijm P. Controlling Protein Surface Orientation by Strategic Placement of Oligo-Histidine Tags. ACS NANO 2017; 11:9068-9083. [PMID: 28850777 PMCID: PMC5618149 DOI: 10.1021/acsnano.7b03717] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/26/2017] [Accepted: 08/29/2017] [Indexed: 05/24/2023]
Abstract
We report oriented immobilization of proteins using the standard hexahistidine (His6)-Ni2+:NTA (nitrilotriacetic acid) methodology, which we systematically tuned to give control of surface coverage. Fluorescence microscopy and surface plasmon resonance measurements of self-assembled monolayers (SAMs) of red fluorescent proteins (TagRFP) showed that binding strength increased by 1 order of magnitude for each additional His6-tag on the TagRFP proteins. All TagRFP variants with His6-tags located on only one side of the barrel-shaped protein yielded a 1.5 times higher surface coverage compared to variants with His6-tags on opposite sides of the so-called β-barrel. Time-resolved fluorescence anisotropy measurements supported by polarized infrared spectroscopy verified that the orientation (and thus coverage and functionality) of proteins on surfaces can be controlled by strategic placement of a His6-tag on the protein. Molecular dynamics simulations show how the differently tagged proteins reside at the surface in "end-on" and "side-on" orientations with each His6-tag contributing to binding. Also, not every dihistidine subunit in a given His6-tag forms a full coordination bond with the Ni2+:NTA SAMs, which varied with the position of the His6-tag on the protein. At equal valency but different tag positions on the protein, differences in binding were caused by probing for Ni2+:NTA moieties and by additional electrostatic interactions between different fractions of the β-barrel structure and charged NTA moieties. Potential of mean force calculations indicate there is no specific single-protein interaction mode that provides a clear preferential surface orientation, suggesting that the experimentally measured preference for the end-on orientation is a supra-protein, not a single-protein, effect.
Collapse
Affiliation(s)
- Dorothee Wasserberg
- Bioinspired
Molecular Engineering Laboratory, MIRA Biomedical Technology
and Technical Medicine Institute, Molecular nanoFabrication Group, MESA+ Institute
for Nanotechnology, and Nanobiophysics Group, MESA+ Institute for Nanotechnology,
and MIRA Biomedical Technology and Technical Medicine Institute, University of Twente, P.O. Box 217, 7500 AE Enschede, The Netherlands
| | - Jordi Cabanas-Danés
- Bioinspired
Molecular Engineering Laboratory, MIRA Biomedical Technology
and Technical Medicine Institute, Molecular nanoFabrication Group, MESA+ Institute
for Nanotechnology, and Nanobiophysics Group, MESA+ Institute for Nanotechnology,
and MIRA Biomedical Technology and Technical Medicine Institute, University of Twente, P.O. Box 217, 7500 AE Enschede, The Netherlands
| | - Jord Prangsma
- Bioinspired
Molecular Engineering Laboratory, MIRA Biomedical Technology
and Technical Medicine Institute, Molecular nanoFabrication Group, MESA+ Institute
for Nanotechnology, and Nanobiophysics Group, MESA+ Institute for Nanotechnology,
and MIRA Biomedical Technology and Technical Medicine Institute, University of Twente, P.O. Box 217, 7500 AE Enschede, The Netherlands
| | - Shane O’Mahony
- Department
of Physics, Bernal Institute, University
of Limerick, Limerick V94 T9PX, Ireland
| | - Pierre-Andre Cazade
- Department
of Physics, Bernal Institute, University
of Limerick, Limerick V94 T9PX, Ireland
| | - Eldrich Tromp
- Bioinspired
Molecular Engineering Laboratory, MIRA Biomedical Technology
and Technical Medicine Institute, Molecular nanoFabrication Group, MESA+ Institute
for Nanotechnology, and Nanobiophysics Group, MESA+ Institute for Nanotechnology,
and MIRA Biomedical Technology and Technical Medicine Institute, University of Twente, P.O. Box 217, 7500 AE Enschede, The Netherlands
| | - Christian Blum
- Bioinspired
Molecular Engineering Laboratory, MIRA Biomedical Technology
and Technical Medicine Institute, Molecular nanoFabrication Group, MESA+ Institute
for Nanotechnology, and Nanobiophysics Group, MESA+ Institute for Nanotechnology,
and MIRA Biomedical Technology and Technical Medicine Institute, University of Twente, P.O. Box 217, 7500 AE Enschede, The Netherlands
| | - Damien Thompson
- Department
of Physics, Bernal Institute, University
of Limerick, Limerick V94 T9PX, Ireland
| | - Jurriaan Huskens
- Bioinspired
Molecular Engineering Laboratory, MIRA Biomedical Technology
and Technical Medicine Institute, Molecular nanoFabrication Group, MESA+ Institute
for Nanotechnology, and Nanobiophysics Group, MESA+ Institute for Nanotechnology,
and MIRA Biomedical Technology and Technical Medicine Institute, University of Twente, P.O. Box 217, 7500 AE Enschede, The Netherlands
| | - Vinod Subramaniam
- Bioinspired
Molecular Engineering Laboratory, MIRA Biomedical Technology
and Technical Medicine Institute, Molecular nanoFabrication Group, MESA+ Institute
for Nanotechnology, and Nanobiophysics Group, MESA+ Institute for Nanotechnology,
and MIRA Biomedical Technology and Technical Medicine Institute, University of Twente, P.O. Box 217, 7500 AE Enschede, The Netherlands
- Free
University of Amsterdam, De Boelelaan 1105, 1081 HV Amsterdam, The Netherlands
| | - Pascal Jonkheijm
- Bioinspired
Molecular Engineering Laboratory, MIRA Biomedical Technology
and Technical Medicine Institute, Molecular nanoFabrication Group, MESA+ Institute
for Nanotechnology, and Nanobiophysics Group, MESA+ Institute for Nanotechnology,
and MIRA Biomedical Technology and Technical Medicine Institute, University of Twente, P.O. Box 217, 7500 AE Enschede, The Netherlands
| |
Collapse
|
44
|
Krepl M, Blatter M, Cléry A, Damberger FF, Allain FH, Sponer J. Structural study of the Fox-1 RRM protein hydration reveals a role for key water molecules in RRM-RNA recognition. Nucleic Acids Res 2017; 45:8046-8063. [PMID: 28505313 PMCID: PMC5737849 DOI: 10.1093/nar/gkx418] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2017] [Revised: 04/26/2017] [Accepted: 05/02/2017] [Indexed: 01/07/2023] Open
Abstract
The Fox-1 RNA recognition motif (RRM) domain is an important member of the RRM protein family. We report a 1.8 Å X-ray structure of the free Fox-1 containing six distinct monomers. We use this and the nuclear magnetic resonance (NMR) structure of the Fox-1 protein/RNA complex for molecular dynamics (MD) analyses of the structured hydration. The individual monomers of the X-ray structure show diverse hydration patterns, however, MD excellently reproduces the most occupied hydration sites. Simulations of the protein/RNA complex show hydration consistent with the isolated protein complemented by hydration sites specific to the protein/RNA interface. MD predicts intricate hydration sites with water-binding times extending up to hundreds of nanoseconds. We characterize two of them using NMR spectroscopy, RNA binding with switchSENSE and free-energy calculations of mutant proteins. Both hydration sites are experimentally confirmed and their abolishment reduces the binding free-energy. A quantitative agreement between theory and experiment is achieved for the S155A substitution but not for the S122A mutant. The S155 hydration site is evolutionarily conserved within the RRM domains. In conclusion, MD is an effective tool for predicting and interpreting the hydration patterns of protein/RNA complexes. Hydration is not easily detectable in NMR experiments but can affect stability of protein/RNA complexes.
Collapse
Affiliation(s)
- Miroslav Krepl
- Institute of Biophysics, Academy of Sciences of the Czech Republic, Kralovopolska 135, 612 65 Brno, Czech Republic
- Regional Centre of Advanced Technologies and Materials, Department of Physical Chemistry, Faculty of Science, Palacky University Olomouc, 17. listopadu 12, 771 46 Olomouc, Czech Republic
| | - Markus Blatter
- Institute of Molecular Biology and Biophysics, Department of Biology, ETH Zurich, CH-8093 Zurich, Switzerland
- Present address: Global Discovery Chemistry, Novartis Institute for BioMedical Research, Basel CH-4002, Switzerland
| | - Antoine Cléry
- Institute of Molecular Biology and Biophysics, Department of Biology, ETH Zurich, CH-8093 Zurich, Switzerland
| | - Fred F. Damberger
- Institute of Molecular Biology and Biophysics, Department of Biology, ETH Zurich, CH-8093 Zurich, Switzerland
| | - Frédéric H.T. Allain
- Institute of Molecular Biology and Biophysics, Department of Biology, ETH Zurich, CH-8093 Zurich, Switzerland
| | - Jiri Sponer
- Institute of Biophysics, Academy of Sciences of the Czech Republic, Kralovopolska 135, 612 65 Brno, Czech Republic
- Regional Centre of Advanced Technologies and Materials, Department of Physical Chemistry, Faculty of Science, Palacky University Olomouc, 17. listopadu 12, 771 46 Olomouc, Czech Republic
| |
Collapse
|
45
|
Franz-Oberdorf K, Langer A, Strasser R, Isono E, Ranftl QL, Wunschel C, Schwab W. Physical interaction between the strawberry allergen Fra a 1 and an associated partner FaAP: Interaction of Fra a 1 proteins and FaAP. Proteins 2017; 85:1891-1901. [DOI: 10.1002/prot.25343] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2017] [Revised: 06/21/2017] [Accepted: 06/26/2017] [Indexed: 11/11/2022]
Affiliation(s)
- Katrin Franz-Oberdorf
- Biotechnology of Natural Products, School of Life Sciences Weihenstephan Technische Universität München; 85354 Freising Germany
| | - Andreas Langer
- Dynamic Biosensors GmbH; Lochhamerstr. 15 82152 Planegg Germany
| | - Ralf Strasser
- Dynamic Biosensors GmbH; Lochhamerstr. 15 82152 Planegg Germany
| | - Erika Isono
- Department of Plant Systems Biology; Technische Universität München; 85354 Freising Germany
| | - Quirin L. Ranftl
- Department of Plant Systems Biology; Technische Universität München; 85354 Freising Germany
| | - Christian Wunschel
- Department of Botany; Technische Universität München; 85354 Freising Germany
| | - Wilfried Schwab
- Biotechnology of Natural Products, School of Life Sciences Weihenstephan Technische Universität München; 85354 Freising Germany
| |
Collapse
|
46
|
Cléry A, Sohier TJM, Welte T, Langer A, Allain FHT. switchSENSE: A new technology to study protein-RNA interactions. Methods 2017; 118-119:137-145. [PMID: 28286323 DOI: 10.1016/j.ymeth.2017.03.004] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2016] [Revised: 03/07/2017] [Accepted: 03/08/2017] [Indexed: 11/25/2022] Open
Abstract
Characterization of RNA-binding protein interactions with RNA became inevitable to properly understand the cellular mechanisms involved in gene expression regulation. Structural investigations bring information at the atomic level on these interactions and complementary methods such as Isothermal Titration Calorimetry (ITC) and Surface Plasmon Resonance (SPR) are commonly used to quantify the affinity of these RNA-protein complexes and evaluate the effect of mutations affecting these interactions. The switchSENSE technology has recently been developed and already successfully used to investigate protein interactions with different types of binding partners (DNA, protein/peptide or even small molecules). In this study, we show that this method is also well suited to study RNA binding proteins (RBPs). We could successfully investigate the binding to RNA of three different RBPs (Fox-1, SRSF1 and Tra2-β1) and obtained KD values very close to the ones determined previously by SPR or ITC for these complexes. These results show that the switchSENSE technology can be used as an alternative method to study protein-RNA interactions with KD values in the low micromolar (10-6) to nanomolar (10-7-10-9) and probably picomolar (10-10-10-12) range. The absence of labelling requirement for the analyte molecules and the use of very low amounts of protein and RNA molecules make the switchSENSE approach very attractive compared to other methods. Finally, we discuss about the potential of this approach in obtaining more sophisticated information such as structural conformational changes upon RBP binding to RNA.
Collapse
Affiliation(s)
- Antoine Cléry
- Institute of Molecular Biology and Biophysics, Department of Biology, ETH Zurich, CH-8093 Zurich, Switzerland.
| | - Thibault J M Sohier
- Institute of Molecular Biology and Biophysics, Department of Biology, ETH Zurich, CH-8093 Zurich, Switzerland
| | - Thomas Welte
- Dynamic Biosensors GmbH, Lochhamer Str. 15, 82152 Martinsried, Germany
| | - Andreas Langer
- Dynamic Biosensors GmbH, Lochhamer Str. 15, 82152 Martinsried, Germany
| | - Frédéric H T Allain
- Institute of Molecular Biology and Biophysics, Department of Biology, ETH Zurich, CH-8093 Zurich, Switzerland.
| |
Collapse
|
47
|
Feng C, Ding HM, Ren CL, Ma YQ. Designing new strategy for controlling DNA orientation in biosensors. Sci Rep 2015; 5:14415. [PMID: 26400770 PMCID: PMC4585838 DOI: 10.1038/srep14415] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2015] [Accepted: 08/27/2015] [Indexed: 01/04/2023] Open
Abstract
Orientation controllable DNA biosensors hold great application potentials in recognizing small molecules and detecting DNA hybridization. Though electric field is usually used to control the orientation of DNA molecules, it is also of great importance and significance to seek for other triggered methods to control the DNA orientation. Here, we design a new strategy for controlling DNA orientation in biosensors. The main idea is to copolymerize DNA molecules with responsive polymers that can show swelling/deswelling transitions due to the change of external stimuli, and then graft the copolymers onto an uncharged substrate. In order to highlight the responsive characteristic, we take thermo-responsive polymers as an example, and reveal multi-responsive behavior and the underlying molecular mechanism of the DNA orientation by combining dissipative particle dynamics simulation and molecular theory. Since swelling/deswelling transitions can be also realized by using other stimuli-responsive (like pH and light) polymers, the present strategy is universal, which can enrich the methods of controlling DNA orientation and may assist with the design of the next generation of biosensors.
Collapse
Affiliation(s)
- Chao Feng
- National Laboratory of Solid State Microstructures and Department of Physics, Nanjing University, Nanjing 210093, China.,Collaborative Innovation Center of Advanced Microstructures, Nanjing University, Nanjing, 210093, China
| | - Hong-ming Ding
- National Laboratory of Solid State Microstructures and Department of Physics, Nanjing University, Nanjing 210093, China.,Center for Soft Condensed Matter Physics and Interdisciplinary Research, Soochow University, Suzhou 215006, China
| | - Chun-lai Ren
- National Laboratory of Solid State Microstructures and Department of Physics, Nanjing University, Nanjing 210093, China.,Collaborative Innovation Center of Advanced Microstructures, Nanjing University, Nanjing, 210093, China
| | - Yu-qiang Ma
- National Laboratory of Solid State Microstructures and Department of Physics, Nanjing University, Nanjing 210093, China.,Collaborative Innovation Center of Advanced Microstructures, Nanjing University, Nanjing, 210093, China.,Center for Soft Condensed Matter Physics and Interdisciplinary Research, Soochow University, Suzhou 215006, China
| |
Collapse
|
48
|
Casanova-Moreno J, Bizzotto D. A Method for Determining the Actual Rate of Orientation Switching of DNA Self-Assembled Monolayers Using Optical and Electrochemical Frequency Response Analysis. Anal Chem 2015; 87:2255-63. [DOI: 10.1021/ac503919a] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Affiliation(s)
- J. Casanova-Moreno
- Department
of Chemistry, University of British Columbia, 2036 Main Mall, Vancouver, BC V6T 1Z1, Canada
- Advanced
Materials and Process Engineering Laboratory, University of British Columbia, 2355 East Mall, Vancouver, BC V6T 1Z4, Canada
| | - D. Bizzotto
- Department
of Chemistry, University of British Columbia, 2036 Main Mall, Vancouver, BC V6T 1Z1, Canada
- Advanced
Materials and Process Engineering Laboratory, University of British Columbia, 2355 East Mall, Vancouver, BC V6T 1Z4, Canada
| |
Collapse
|
49
|
Wang W, Yin L, Gonzalez-Malerva L, Wang S, Yu X, Eaton S, Zhang S, Chen HY, LaBaer J, Tao N. In situ drug-receptor binding kinetics in single cells: a quantitative label-free study of anti-tumor drug resistance. Sci Rep 2014; 4:6609. [PMID: 25312029 PMCID: PMC4196117 DOI: 10.1038/srep06609] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2014] [Accepted: 09/16/2014] [Indexed: 02/05/2023] Open
Abstract
Many drugs are effective in the early stage of treatment, but patients develop drug resistance after a certain period of treatment, causing failure of the therapy. An important example is Herceptin, a popular monoclonal antibody drug for breast cancer by specifically targeting human epidermal growth factor receptor 2 (Her2). Here we demonstrate a quantitative binding kinetics analysis of drug-target interactions to investigate the molecular scale origin of drug resistance. Using a surface plasmon resonance imaging, we measured the in situ Herceptin-Her2 binding kinetics in single intact cancer cells for the first time, and observed significantly weakened Herceptin-Her2 interactions in Herceptin-resistant cells, compared to those in Herceptin-sensitive cells. We further showed that the steric hindrance of Mucin-4, a membrane protein, was responsible for the altered drug-receptor binding. This effect of a third molecule on drug-receptor interactions cannot be studied using traditional purified protein methods, demonstrating the importance of the present intact cell-based binding kinetics analysis.
Collapse
MESH Headings
- Antibodies, Monoclonal, Humanized/administration & dosage
- Antibodies, Monoclonal, Humanized/chemistry
- Antibodies, Monoclonal, Humanized/metabolism
- Antibodies, Monoclonal, Humanized/pharmacokinetics
- Breast Neoplasms/drug therapy
- Breast Neoplasms/metabolism
- Breast Neoplasms/pathology
- Drug Interactions
- Drug Resistance, Neoplasm/genetics
- Female
- Humans
- Mucin-4/metabolism
- Protein Binding
- Receptor, ErbB-2/metabolism
- Surface Plasmon Resonance
- Trastuzumab
Collapse
Affiliation(s)
- Wei Wang
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing 210093, China
| | - Linliang Yin
- Center for Bioelectronics and Biosensors, Biodesign Institute, Arizona State University, Tempe, AZ 85287, USA
- School of Chemistry and Chemical Engineering, Chongqing University, Chongqing 400044, China
| | - Laura Gonzalez-Malerva
- Virginia G. Piper Center for Personalized Diagnostics, Biodesign Institute, Arizona State University, Tempe, AZ 85287, USA
| | - Shaopeng Wang
- Center for Bioelectronics and Biosensors, Biodesign Institute, Arizona State University, Tempe, AZ 85287, USA
| | - Xiaobo Yu
- Virginia G. Piper Center for Personalized Diagnostics, Biodesign Institute, Arizona State University, Tempe, AZ 85287, USA
| | - Seron Eaton
- Virginia G. Piper Center for Personalized Diagnostics, Biodesign Institute, Arizona State University, Tempe, AZ 85287, USA
| | - Shengtao Zhang
- School of Chemistry and Chemical Engineering, Chongqing University, Chongqing 400044, China
| | - Hong-Yuan Chen
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing 210093, China
| | - Joshua LaBaer
- Virginia G. Piper Center for Personalized Diagnostics, Biodesign Institute, Arizona State University, Tempe, AZ 85287, USA
| | - Nongjian Tao
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing 210093, China
- Center for Bioelectronics and Biosensors, Biodesign Institute, Arizona State University, Tempe, AZ 85287, USA
- Department of Electrical Engineering, Arizona State University, Tempe, AZ 85287, USA
| |
Collapse
|
50
|
Ahijado-Guzmán R, Prasad J, Rosman C, Henkel A, Tome L, Schneider D, Rivas G, Sönnichsen C. Plasmonic nanosensors for simultaneous quantification of multiple protein-protein binding affinities. NANO LETTERS 2014; 14:5528-32. [PMID: 25153997 DOI: 10.1021/nl501865p] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/24/2023]
Abstract
Most of current techniques used for the quantification of protein-protein interactions require the analysis of one pair of binding partners at a time. Herein we present a label-free, simple, fast, and cost-effective route to characterize binding affinities between multiple macromolecular partners simultaneously, using optical dark-field spectroscopy and individual protein-functionalized gold nanorods as sensing elements. Our NanoSPR method could easily become a simple and standard tool in biological, biochemical, and medical laboratories.
Collapse
Affiliation(s)
- Rubén Ahijado-Guzmán
- Institute of Physical Chemistry, University of Mainz , Duesbergweg 10-14, D-55128 Mainz, Germany
| | | | | | | | | | | | | | | |
Collapse
|