1
|
Streit M, Budiarta M, Jungblut M, Beliu G. Fluorescent labeling strategies for molecular bioimaging. BIOPHYSICAL REPORTS 2025; 5:100200. [PMID: 39947326 PMCID: PMC11914189 DOI: 10.1016/j.bpr.2025.100200] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/20/2024] [Revised: 01/28/2025] [Accepted: 02/06/2025] [Indexed: 03/16/2025]
Abstract
Super-resolution microscopy (SRM) has transformed biological imaging by circumventing the diffraction limit of light and enabling the visualization of cellular structures and processes at the molecular level. Central to the capabilities of SRM is fluorescent labeling, which ensures the precise attachment of fluorophores to biomolecules and has direct impact on the accuracy and resolution of imaging. Continuous innovation and optimization in fluorescent labeling are essential for the successful application of SRM in cutting-edge biological research. In this review, we discuss recent advances in fluorescent labeling strategies for molecular bioimaging, with a special focus on protein labeling. We compare different approaches, highlight technological breakthroughs, and address challenges such as linkage error and labeling density. By evaluating both established and emerging methods, we aim to guide researchers through all aspects that should be considered before opting for any labeling technique.
Collapse
Affiliation(s)
- Marcel Streit
- Rudolf Virchow Center, Research Center for Integrative and Translational Bioimaging, University of Würzburg, 97080 Würzburg, Germany
| | - Made Budiarta
- Rudolf Virchow Center, Research Center for Integrative and Translational Bioimaging, University of Würzburg, 97080 Würzburg, Germany
| | - Marvin Jungblut
- Rudolf Virchow Center, Research Center for Integrative and Translational Bioimaging, University of Würzburg, 97080 Würzburg, Germany
| | - Gerti Beliu
- Rudolf Virchow Center, Research Center for Integrative and Translational Bioimaging, University of Würzburg, 97080 Würzburg, Germany.
| |
Collapse
|
2
|
Taper M, Carrington G, Peckham M, Lal S, Hume RD. A comparison of fixation and immunofluorescence protocols for successful reproducibility and improved signal in human left ventricle cardiac tissue. J Microsc 2024; 296:34-47. [PMID: 38856969 DOI: 10.1111/jmi.13336] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2024] [Revised: 04/14/2024] [Accepted: 05/17/2024] [Indexed: 06/11/2024]
Abstract
Immunohistochemistry (IHC) and immunofluorescence (IF) are crucial techniques for studying cardiac physiology and disease. The accuracy of these techniques is dependent on various aspects of sample preparation and processing. However, standardised protocols for sample preparation of tissues, particularly for fresh-frozen human left ventricle (LV) tissue, have yet to be established and could potentially lead to differences in staining and interpretation. Thus, this study aimed to optimise the reproducibility and quality of IF staining in fresh-frozen human LV tissue by systematically investigating crucial aspects of the sample preparation process. To achieve this, we subjected fresh-frozen human LV tissue to different fixation protocols, primary antibody incubation temperatures, antibody penetration reagents, and fluorescent probes. We found that neutral buffered formalin fixation reduced image artefacts and improved antibody specificity compared to both methanol and acetone fixation. Additionally, incubating primary antibodies at 37°C for 3 h improved fluorescence intensity compared to the commonly practised 4°C overnight incubation. Furthermore, we found that DeepLabel, an antibody penetration reagent, and smaller probes, such as fragmented antibodies and Affimers, improved the visualisation depth of cardiac structures. DeepLabel also improved antibody penetration in CUBIC cleared thick LV tissue fragments. Thus, our data underscores the importance of standardised protocols in IF staining and provides various means of improving staining quality. In addition to contributing to cardiac research by providing methodologies for IF, the findings and processes presented herein also establish a framework by which staining of other tissues may be optimised.
Collapse
Affiliation(s)
- Matthew Taper
- Faculty of Medicine and Health, School of Medical Sciences, The University of Sydney, Sydney, Australia
- Centre for Heart Failure and Diseases of the Aorta, The Baird Institute, Sydney, Australia
| | - Glenn Carrington
- Faculty of Biological Sciences, Astbury Centre for Structural Biology and the School of Molecular and Cellular Biology, University of Leeds, Leeds, UK
| | - Michelle Peckham
- Faculty of Biological Sciences, Astbury Centre for Structural Biology and the School of Molecular and Cellular Biology, University of Leeds, Leeds, UK
| | - Sean Lal
- Faculty of Medicine and Health, School of Medical Sciences, The University of Sydney, Sydney, Australia
- Centre for Heart Failure and Diseases of the Aorta, The Baird Institute, Sydney, Australia
- Department of Cardiology, Royal Prince Alfred Hospital, Sydney, Australia
| | - Robert D Hume
- Faculty of Medicine and Health, School of Medical Sciences, The University of Sydney, Sydney, Australia
- Centre for Heart Failure and Diseases of the Aorta, The Baird Institute, Sydney, Australia
| |
Collapse
|
3
|
Hellmeier J, Strauss S, Xu S, Masullo LA, Unterauer EM, Kowalewski R, Jungmann R. Quantification of absolute labeling efficiency at the single-protein level. Nat Methods 2024; 21:1702-1707. [PMID: 38658647 PMCID: PMC11399078 DOI: 10.1038/s41592-024-02242-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2023] [Accepted: 03/11/2024] [Indexed: 04/26/2024]
Abstract
State-of-the-art super-resolution microscopy allows researchers to spatially resolve single proteins in dense clusters. However, accurate quantification of protein organization and stoichiometries requires a general method to evaluate absolute binder labeling efficiency, which is currently unavailable. Here we introduce a universally applicable approach that uses a reference tag fused to a target protein of interest. By attaching high-affinity binders, such as antibodies or nanobodies, to both the reference tag and the target protein, and then employing DNA-barcoded sequential super-resolution imaging, we can correlate the location of the reference tag with the target molecule binder. This approach facilitates the precise quantification of labeling efficiency at the single-protein level.
Collapse
Affiliation(s)
| | | | - Shuhan Xu
- Max Planck Institute of Biochemistry, Planegg, Germany
| | | | | | - Rafal Kowalewski
- Max Planck Institute of Biochemistry, Planegg, Germany
- Faculty of Physics and Center for Nanoscience, Ludwig Maximilian University, Munich, Germany
| | - Ralf Jungmann
- Max Planck Institute of Biochemistry, Planegg, Germany.
- Faculty of Physics and Center for Nanoscience, Ludwig Maximilian University, Munich, Germany.
| |
Collapse
|
4
|
Callaghan KL, Sherrell PC, Ellis AV. The Impact of Activating Agents on Non-Enzymatic Nucleic Acid Extension Reactions. Chembiochem 2024; 25:e202300859. [PMID: 38282207 DOI: 10.1002/cbic.202300859] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Revised: 01/21/2024] [Accepted: 01/28/2024] [Indexed: 01/30/2024]
Abstract
Non-enzymatic template-directed primer extension is increasingly being studied for the production of RNA and DNA. These reactions benefit from producing RNA or DNA in an aqueous, protecting group free system, without the need for expensive enzymes. However, these primer extension reactions suffer from a lack of fidelity, low reaction rates, low overall yields, and short primer extension lengths. This review outlines a detailed mechanistic pathway for non-enzymatic template-directed primer extension and presents a review of the thermodynamic driving forces involved in entropic templating. Through the lens of entropic templating, the rate and fidelity of a reaction are shown to be intrinsically linked to the reactivity of the activating agent used. Thus, a strategy is discussed for the optimization of non-enzymatic template-directed primer extension, providing a path towards cost-effective in vitro synthesis of RNA and DNA.
Collapse
Affiliation(s)
- Kimberley L Callaghan
- Department of Chemical Engineering, The University of Melbourne, Parkville, Victoria, 3010, Australia
| | - Peter C Sherrell
- Department of Chemical Engineering, The University of Melbourne, Parkville, Victoria, 3010, Australia
- School of Science, RMIT University, Melbourne, Victoria, 3000, Australia
| | - Amanda V Ellis
- Department of Chemical Engineering, The University of Melbourne, Parkville, Victoria, 3010, Australia
| |
Collapse
|
5
|
Banerjee A, Anand M, Ganji M. Labeling approaches for DNA-PAINT super-resolution imaging. NANOSCALE 2023; 15:6563-6580. [PMID: 36942769 DOI: 10.1039/d2nr06541j] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/18/2023]
Abstract
Super-resolution imaging is becoming a commonly employed tool to visualize biological targets in unprecedented detail. DNA-PAINT is one of the single-molecule localization microscopy-based super-resolution imaging modalities allowing the ultra-high-resolution imaging with superior multiplexing capabilities. We discuss the importance of patterned DNA nanostructures in demonstrating the capabilities of DNA-PAINT and the design of various combinations of imager-docking strand pairs for imaging. Central to the implementation of DNA-PAINT imaging in a biological context is the generation of docking strand-conjugated binders against the target molecules. Several researchers have developed a variety of labelling probes for improving resolution while also providing multiplexing capabilities for the broader application of DNA-PAINT. This review provides a comprehensive summary of the repertoire of labelling probes used for DNA-PAINT in cells and the strategies implemented to chemically modify them with a docking strand.
Collapse
Affiliation(s)
- Abhinav Banerjee
- Department of Biochemistry, Indian Institute of Science, Malleshwaram, Bengaluru 560012, India.
| | - Micky Anand
- Department of Biochemistry, Indian Institute of Science, Malleshwaram, Bengaluru 560012, India.
| | - Mahipal Ganji
- Department of Biochemistry, Indian Institute of Science, Malleshwaram, Bengaluru 560012, India.
| |
Collapse
|
6
|
Eklund AS, Jungmann R. Optimized Coiled-Coil Interactions for Multiplexed Peptide-PAINT. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2023; 19:e2206347. [PMID: 36642829 DOI: 10.1002/smll.202206347] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/15/2022] [Revised: 12/09/2022] [Indexed: 06/17/2023]
Abstract
Super-resolution microscopy has revolutionized how researchers characterize samples in the life sciences in the last decades. Amongst methods employing single-molecule localization microscopy, DNA points accumulation for imaging in nanoscale topography (DNA-PAINT) is a relatively easy-to-implement method that uses the programmable and repetitive binding of dye-labeled DNA imager strands to their respective docking strands. Recently developed Peptide-PAINT replaces the interaction of oligonucleotides by short coiled-coil peptide sequences leading to an improved labeling scheme by reducing linkage errors to target proteins. However, only one coiled-coil pair is currently available for Peptide-PAINT, preventing multiplexed imaging. In this study, the initial Peptide-PAINT E/K coil is improved by modifying its length for optimized binding kinetics leading to improved localization precisions. Additionally, an orthogonal P3/P4 coil pair is introduced, enabling 2-plex Peptide-PAINT imaging and benchmarking its performance and orthogonality using single-molecule and DNA origami assays. Finally, the P3/P4 peptide pair is used to image the human epidermal growth factor receptors 2 (ErbB2/Her2) in 2D and 3D at the single receptor level using genetically encoded peptide tags.
Collapse
Affiliation(s)
- Alexandra S Eklund
- Max Planck Institute of Biochemistry, Am Klopferspitz 18, 82152, Planegg, Germany
- Faculty of Physics and Center for Nanoscience, Ludwig Maximilian University, Geschwister-Scholl-Platz 1, 80539, Munich, Germany
| | - Ralf Jungmann
- Max Planck Institute of Biochemistry, Am Klopferspitz 18, 82152, Planegg, Germany
- Faculty of Physics and Center for Nanoscience, Ludwig Maximilian University, Geschwister-Scholl-Platz 1, 80539, Munich, Germany
| |
Collapse
|
7
|
Chen Z, Yin G, Wei J, Qi T, Qian Z, Wang Z, Zong S, Cui Y. Quantitative analysis of multiple breast cancer biomarkers using DNA-PAINT. ANALYTICAL METHODS : ADVANCING METHODS AND APPLICATIONS 2022; 14:3671-3679. [PMID: 36063064 DOI: 10.1039/d2ay00670g] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Immunotherapy has become an efficient treatment method of breast cancer. Detection of proteins such as PD-L1 and CTLA-4, which are important immune checkpoint molecules, is attracting more and more attention as they play key roles in immunotherapy. Here, by combining the high resolution of DNA-PAINT (DNA points accumulation for imaging in nanoscale topography) with the qPAINT quantitative analysis method, accurate spatial localization and absolute quantification of PD-L1 and CTLA-4 on the membrane of breast cancer cells could be achieved. Meanwhile, exchange-PAINT was also conducted to count three other biomarkers (EpCAM, EGFR, and HER2). Simultaneous analysis of these biomarkers can greatly facilitate the differentiation of different kinds of breast cancer. Such a simple quantitative analysis method holds great potential in diagnosis and immunotherapy of cancers.
Collapse
Affiliation(s)
- Zengwei Chen
- Advanced Photonics Center, Southeast University, Nanjing 210096, Jiangsu, China.
| | - Gaoqiang Yin
- Advanced Photonics Center, Southeast University, Nanjing 210096, Jiangsu, China.
| | - Jinxiu Wei
- Advanced Photonics Center, Southeast University, Nanjing 210096, Jiangsu, China.
| | - Tongsheng Qi
- Advanced Photonics Center, Southeast University, Nanjing 210096, Jiangsu, China.
| | - Ziting Qian
- Advanced Photonics Center, Southeast University, Nanjing 210096, Jiangsu, China.
| | - Zhuyuan Wang
- Advanced Photonics Center, Southeast University, Nanjing 210096, Jiangsu, China.
| | - Shenfei Zong
- Advanced Photonics Center, Southeast University, Nanjing 210096, Jiangsu, China.
| | - Yiping Cui
- Advanced Photonics Center, Southeast University, Nanjing 210096, Jiangsu, China.
| |
Collapse
|
8
|
Cordell P, Carrington G, Curd A, Parker F, Tomlinson D, Peckham M. Affimers and nanobodies as molecular probes and their applications in imaging. J Cell Sci 2022; 135:276020. [PMID: 35848463 PMCID: PMC9450889 DOI: 10.1242/jcs.259168] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Antibodies are the most widely used, traditional tool for labelling molecules in cells. In the past five to ten years, many new labelling tools have been developed with significant advantages over the traditional antibody. Here, we focus on nanobodies and the non-antibody binding scaffold proteins called Affimers. We explain how they are generated, selected and produced, and we describe how their small size, high binding affinity and specificity provides them with many advantages compared to antibodies. Of particular importance, their small size enables them to better penetrate dense cytoskeletal regions within cells, as well as tissues, providing them with specific advantage for super-resolution imaging, as they place the fluorophore with a few nanometres of the target protein being imaged. We expect these novel tools to be of broad interest to many cell biologists and anticipate them becoming the tools of choice for super-resolution imaging.
Collapse
|
9
|
Yang Q, Chang X, Lee JY, Olivera TR, Saji M, Wisniewski H, Kim S, Zhang F. Recent Advances in Self-Assembled DNA Nanostructures for Bioimaging. ACS APPLIED BIO MATERIALS 2022; 5:4652-4667. [PMID: 35559619 DOI: 10.1021/acsabm.2c00128] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
DNA nanotechnology has been proven to be a powerful platform to assist the development of imaging probes for biomedical research. The attractive features of DNA nanostructures, such as nanometer precision, controllable size, programmable functions, and biocompatibility, have enabled researchers to design and customize DNA nanoprobes for bioimaging applications. However, DNA probes with low molecular weights (e.g., 10-100 nt) generally suffer from low stability in physiological buffer environments. To improve the stability of DNA nanoprobes in such environments, DNA nanostructures can be designed with relatively larger sizes and defined shapes. In addition, the established modification methods for DNA nanostructures are also essential in enhancing their properties and performances in a physiological environment. In this review, we begin with a brief recap of the development of DNA nanostructures including DNA tiles, DNA origami, and multifunctional DNA nanostructures with modifications. Then we highlight the recent advances of DNA nanostructures for bioimaging, emphasizing the latest developments in probe modifications and DNA-PAINT imaging. Multiple imaging modules for intracellular biomolecular imaging and cell membrane biomarkers recognition are also summarized. In the end, we discuss the advantages and challenges of applying DNA nanostructures in bioimaging research and speculate on its future developments.
Collapse
Affiliation(s)
- Qi Yang
- Department of Chemistry, Rutgers University, Newark, New Jersey 07102, United States
| | - Xu Chang
- Department of Chemistry, Rutgers University, Newark, New Jersey 07102, United States
| | - Jung Yeon Lee
- Department of Chemistry, Rutgers University, Newark, New Jersey 07102, United States
| | - Tiffany R Olivera
- Department of Chemistry, Rutgers University, Newark, New Jersey 07102, United States
| | - Minu Saji
- Department of Chemistry, Rutgers University, Newark, New Jersey 07102, United States
| | - Henry Wisniewski
- Department of Chemistry, Rutgers University, Newark, New Jersey 07102, United States
| | - Suchan Kim
- Department of Chemistry, Rutgers University, Newark, New Jersey 07102, United States
| | - Fei Zhang
- Department of Chemistry, Rutgers University, Newark, New Jersey 07102, United States
| |
Collapse
|
10
|
Tagliatti E, Cortese K. Imaging Endocytosis Dynamics in Health and Disease. MEMBRANES 2022; 12:membranes12040393. [PMID: 35448364 PMCID: PMC9028293 DOI: 10.3390/membranes12040393] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Revised: 03/16/2022] [Accepted: 03/29/2022] [Indexed: 02/06/2023]
Abstract
Endocytosis is a critical process for cell growth and viability. It mediates nutrient uptake, guarantees plasma membrane homeostasis, and generates intracellular signaling cascades. Moreover, it plays an important role in dead cell clearance and defense against external microbes. Finally, endocytosis is an important cellular route for the delivery of nanomedicines for therapeutic treatments. Thus, it is not surprising that both environmental and genetic perturbation of endocytosis have been associated with several human conditions such as cancer, neurological disorders, and virus infections, among others. Over the last decades, a lot of research has been focused on developing advanced imaging methods to monitor endocytosis events with high resolution in living cells and tissues. These include fluorescence imaging, electron microscopy, and correlative and super-resolution microscopy. In this review, we outline the major endocytic pathways and briefly discuss how defects in the molecular machinery of these pathways lead to disease. We then discuss the current imaging methodologies used to study endocytosis in different contexts, highlighting strengths and weaknesses.
Collapse
Affiliation(s)
- Erica Tagliatti
- Laboratory of Pharmacology and Brain Pathology, Humanitas Clinical and Research Center, Via Manzoni 56, 20089 Milano, Italy
- Department of Clinical and Experimental Epilepsy, UCL Queen Square Institute of Neurology, University College London, London WC1E 6BT, UK
- Correspondence: (E.T.); (K.C.)
| | - Katia Cortese
- Cellular Electron Microscopy Laboratory, Department of Experimental Medicine (DIMES), Human Anatomy, Università di Genova, Via Antonio de Toni 14, 16132 Genova, Italy
- Correspondence: (E.T.); (K.C.)
| |
Collapse
|
11
|
Abstract
Super-resolution microscopy techniques, and specifically single-molecule localization microscopy (SMLM), are approaching nanometer resolution inside cells and thus have great potential to complement structural biology techniques such as electron microscopy for structural cell biology. In this review, we introduce the different flavors of super-resolution microscopy, with a special emphasis on SMLM and MINFLUX (minimal photon flux). We summarize recent technical developments that pushed these localization-based techniques to structural scales and review the experimental conditions that are key to obtaining data of the highest quality. Furthermore, we give an overview of different analysis methods and highlight studies that used SMLM to gain structural insights into biologically relevant molecular machines. Ultimately, we give our perspective on what is needed to push the resolution of these techniques even further and to apply them to investigating dynamic structural rearrangements in living cells. Expected final online publication date for the Annual Review of Biophysics, Volume 51 is May 2022. Please see http://www.annualreviews.org/page/journal/pubdates for revised estimates.
Collapse
Affiliation(s)
- Sheng Liu
- Cell Biology & Biophysics Unit, European Molecular Biology Laboratory, Heidelberg, Germany;
| | - Philipp Hoess
- Cell Biology & Biophysics Unit, European Molecular Biology Laboratory, Heidelberg, Germany;
| | - Jonas Ries
- Cell Biology & Biophysics Unit, European Molecular Biology Laboratory, Heidelberg, Germany;
| |
Collapse
|
12
|
Farrell MV, Nunez AC, Yang Z, Pérez-Ferreros P, Gaus K, Goyette J. Protein-PAINT: Superresolution microscopy with signaling proteins. Sci Signal 2022; 15:eabg9782. [PMID: 35104163 DOI: 10.1126/scisignal.abg9782] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Superresolution techniques have advanced our understanding of complex cellular structures and processes but require the attachment of fluorophores to targets through tags or antibodies, which can be bulky and result in underlabeling. To overcome these limitations, we developed a technique to visualize the nanoscale binding locations of signaling proteins by taking advantage of their native interaction domains. Here, we demonstrated that pPAINT (protein point accumulation in nanoscale topography) is a new, single-molecule localization microscopy (SMLM) technique and used it to investigate T cell signaling by visualizing the Src homology 2 (SH2) domain, which is common in signaling molecules. When SH2 domain-containing proteins relocate to the plasma membrane, the domains selectively, transiently, and reversibly bind to preferred phosphorylated tyrosine residues on receptors. This transient binding yields the stochastic blinking events necessary for SMLM when observed with total internal reflection microscopy and enables quantification of binding coefficients in intact cells. We used pPAINT to reveal the binding sites of several T cell receptor-proximal signaling molecules, including Zap70, PI3K, Grb2, Syk, Eat2, and SHP2, and showed that the probes could be multiplexed. We showed that the binding half-life of the tandem SH2 domain of PI3K correlated with binding site cluster size at the immunological synapses of T cells, but that longer binding lifetimes were associated with smaller clusters for the monovalent SH2 domain of Eat2. These results demonstrate the potential of pPAINT for investigating phosphotyrosine-mediated signaling processes at the plasma membrane.
Collapse
Affiliation(s)
- Megan V Farrell
- EMBL Australia Node in Single Molecule Science, School of Medical Sciences, University of New South Wales, Sydney, Australia
| | - Andrea C Nunez
- EMBL Australia Node in Single Molecule Science, School of Medical Sciences, University of New South Wales, Sydney, Australia
| | - Zhengmin Yang
- EMBL Australia Node in Single Molecule Science, School of Medical Sciences, University of New South Wales, Sydney, Australia
| | - Pablo Pérez-Ferreros
- EMBL Australia Node in Single Molecule Science, School of Medical Sciences, University of New South Wales, Sydney, Australia
| | - Katharina Gaus
- EMBL Australia Node in Single Molecule Science, School of Medical Sciences, University of New South Wales, Sydney, Australia
| | - Jesse Goyette
- EMBL Australia Node in Single Molecule Science, School of Medical Sciences, University of New South Wales, Sydney, Australia
| |
Collapse
|
13
|
Dey A, Maiti S. Determining the Stoichiometry of Amyloid Oligomers by Single-Molecule Photobleaching. Methods Mol Biol 2022; 2538:55-74. [PMID: 35951293 DOI: 10.1007/978-1-0716-2529-3_5] [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: 06/15/2023]
Abstract
Small oligomers are the initial intermediates in the pathway to amyloid fibril formation. They have a distinct identity from the monomers as well as from the protofibrils and the fibrils, both in their structure and in their properties. In many cases, they play a crucial biological role. However, due to their transient nature, they are difficult to characterize. "Oligomer" is a diffuse definition, encompassing aggregates of many different sizes, and this lack of precise definition causes much confusion and disagreement between different research groups. Here, we define the small oligomers as "n"-mers with n < 10, which is the size range in which the amyloid proteins typically exist at the initial phase of the aggregation process. Since the oligomers dynamically interconvert into each other, a solution of aggregating amyloid proteins will contain a distribution of sizes. A precise characterization of an oligomeric solution will, therefore, require quantification of the relative population of each size. Size-based separation methods, such as size-exclusion chromatography, are typically used to characterize this distribution. However, if the interconversion between oligomers of different sizes is fast, this would not yield reliable results. Single-molecule photobleaching (smPB) is a direct method to evaluate this size distribution in a heterogeneous solution without separation. In addition, understanding the mechanism of action of amyloid oligomers requires knowing the affinity of each oligomer type to different cellular components, such as the cell membrane. These measurements are also amenable to smPB. Here we show how to perform smPB, both for oligomers in solution and for oligomers attached to the membrane.
Collapse
Affiliation(s)
- Arpan Dey
- Department of Chemical Sciences, Tata Institute of Fundamental Research, Mumbai, India
| | - Sudipta Maiti
- Department of Chemical Sciences, Tata Institute of Fundamental Research, Mumbai, India.
| |
Collapse
|
14
|
Kuhlemann A, Beliu G, Janzen D, Petrini EM, Taban D, Helmerich DA, Doose S, Bruno M, Barberis A, Villmann C, Sauer M, Werner C. Genetic Code Expansion and Click-Chemistry Labeling to Visualize GABA-A Receptors by Super-Resolution Microscopy. Front Synaptic Neurosci 2021; 13:727406. [PMID: 34899260 PMCID: PMC8664562 DOI: 10.3389/fnsyn.2021.727406] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2021] [Accepted: 11/02/2021] [Indexed: 01/15/2023] Open
Abstract
Fluorescence labeling of difficult to access protein sites, e.g., in confined compartments, requires small fluorescent labels that can be covalently tethered at well-defined positions with high efficiency. Here, we report site-specific labeling of the extracellular domain of γ-aminobutyric acid type A (GABA-A) receptor subunits by genetic code expansion (GCE) with unnatural amino acids (ncAA) combined with bioorthogonal click-chemistry labeling with tetrazine dyes in HEK-293-T cells and primary cultured neurons. After optimization of GABA-A receptor expression and labeling efficiency, most effective variants were selected for super-resolution microscopy and functionality testing by whole-cell patch clamp. Our results show that GCE with ncAA and bioorthogonal click labeling with small tetrazine dyes represents a versatile method for highly efficient site-specific fluorescence labeling of proteins in a crowded environment, e.g., extracellular protein domains in confined compartments such as the synaptic cleft.
Collapse
Affiliation(s)
- Alexander Kuhlemann
- Department of Biotechnology and Biophysics, University of Würzburg, Biocenter, Würzburg, Germany
| | - Gerti Beliu
- Department of Biotechnology and Biophysics, University of Würzburg, Biocenter, Würzburg, Germany.,Rudolf Virchow Center for Integrative and Translational Bioimaging, University of Wuerzburg, Würzburg, Germany
| | - Dieter Janzen
- Institute of Clinical Neurobiology, University of Würzburg, Würzburg, Germany
| | - Enrica Maria Petrini
- Neuroscience and Brain Technologies Department, Istituto Italiano Di Tecnologia, Genova, Italy
| | - Danush Taban
- Department of Biotechnology and Biophysics, University of Würzburg, Biocenter, Würzburg, Germany
| | - Dominic A Helmerich
- Department of Biotechnology and Biophysics, University of Würzburg, Biocenter, Würzburg, Germany
| | - Sören Doose
- Department of Biotechnology and Biophysics, University of Würzburg, Biocenter, Würzburg, Germany
| | - Martina Bruno
- Neuroscience and Brain Technologies Department, Istituto Italiano Di Tecnologia, Genova, Italy
| | - Andrea Barberis
- Neuroscience and Brain Technologies Department, Istituto Italiano Di Tecnologia, Genova, Italy
| | - Carmen Villmann
- Institute of Clinical Neurobiology, University of Würzburg, Würzburg, Germany
| | - Markus Sauer
- Department of Biotechnology and Biophysics, University of Würzburg, Biocenter, Würzburg, Germany
| | - Christian Werner
- Department of Biotechnology and Biophysics, University of Würzburg, Biocenter, Würzburg, Germany
| |
Collapse
|
15
|
Konč J, Brown L, Whiten DR, Zuo Y, Ravn P, Klenerman D, Bernardes GJL. A Platform for Site‐Specific DNA‐Antibody Bioconjugation by Using Benzoylacrylic‐Labelled Oligonucleotides. Angew Chem Int Ed Engl 2021. [DOI: 10.1002/ange.202109713] [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)
- Juraj Konč
- Yusuf Hamied Department of Chemistry University of Cambridge Lensfield Road Cambridge CB2 1EW UK
| | - Libby Brown
- Yusuf Hamied Department of Chemistry University of Cambridge Lensfield Road Cambridge CB2 1EW UK
| | - Daniel R. Whiten
- Yusuf Hamied Department of Chemistry University of Cambridge Lensfield Road Cambridge CB2 1EW UK
| | - Yukun Zuo
- Yusuf Hamied Department of Chemistry University of Cambridge Lensfield Road Cambridge CB2 1EW UK
| | - Peter Ravn
- AstraZeneca R&D BioPharmaceuticals Unit
- Antibody Discovery & Protein Engineering (ADPE) Milstein Building, Granta Park Cambridge CB21 6GH UK
- Current address: Department of Biotherapeutic Discovery H. Lundbeck A/S Ottiliavej 9, 2500 Valby Denmark
| | - David Klenerman
- Yusuf Hamied Department of Chemistry University of Cambridge Lensfield Road Cambridge CB2 1EW UK
- UK Dementia Research Institute University of Cambridge Cambridge CB2 0XY UK
| | - Gonçalo J. L. Bernardes
- Yusuf Hamied Department of Chemistry University of Cambridge Lensfield Road Cambridge CB2 1EW UK
- Instituto de Medicina Molecular João Lobo Antunes Faculdade de Medicina Universidade de Lisboa Avenida Professor Egas Moniz 1649-028 Lisboa Portugal
| |
Collapse
|
16
|
Konč J, Brown L, Whiten DR, Zuo Y, Ravn P, Klenerman D, Bernardes GJL. A Platform for Site-Specific DNA-Antibody Bioconjugation by Using Benzoylacrylic-Labelled Oligonucleotides. Angew Chem Int Ed Engl 2021; 60:25905-25913. [PMID: 34555238 PMCID: PMC9297960 DOI: 10.1002/anie.202109713] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2021] [Indexed: 12/27/2022]
Abstract
Many bioconjugation strategies for DNA oligonucleotides and antibodies suffer limitations, such as site-specificity, stoichiometry and hydrolytic instability of the conjugates, which makes them unsuitable for biological applications. Here, we report a new platform for the preparation of DNA-antibody bioconjugates with a simple benzoylacrylic acid pentafluorophenyl ester reagent. Benzoylacrylic-labelled oligonucleotides prepared with this reagent can be site-specifically conjugated to a range of proteins and antibodies through accessible cysteine residues. The homogeneity of the prepared DNA-antibody bioconjugates was confirmed by a new LC-MS protocol and the bioconjugate probes were used in fluorescence or super-resolution microscopy cell imaging experiments. This work demonstrates the versatility and robustness of our bioconjugation protocol that gives site-specific, well-defined and plasma-stable DNA-antibody bioconjugates for biological applications.
Collapse
Affiliation(s)
- Juraj Konč
- Yusuf Hamied Department of ChemistryUniversity of CambridgeLensfield RoadCambridgeCB2 1EWUK
| | - Libby Brown
- Yusuf Hamied Department of ChemistryUniversity of CambridgeLensfield RoadCambridgeCB2 1EWUK
| | - Daniel R. Whiten
- Yusuf Hamied Department of ChemistryUniversity of CambridgeLensfield RoadCambridgeCB2 1EWUK
| | - Yukun Zuo
- Yusuf Hamied Department of ChemistryUniversity of CambridgeLensfield RoadCambridgeCB2 1EWUK
| | - Peter Ravn
- AstraZenecaR&D BioPharmaceuticals Unit|Antibody Discovery & Protein Engineering (ADPE)Milstein Building, Granta ParkCambridgeCB21 6GHUK
- Current address: Department of Biotherapeutic DiscoveryH. Lundbeck A/SOttiliavej 9, 2500ValbyDenmark
| | - David Klenerman
- Yusuf Hamied Department of ChemistryUniversity of CambridgeLensfield RoadCambridgeCB2 1EWUK
- UK Dementia Research InstituteUniversity of CambridgeCambridgeCB2 0XYUK
| | - Gonçalo J. L. Bernardes
- Yusuf Hamied Department of ChemistryUniversity of CambridgeLensfield RoadCambridgeCB2 1EWUK
- Instituto de Medicina Molecular João Lobo AntunesFaculdade de MedicinaUniversidade de LisboaAvenida Professor Egas Moniz1649-028LisboaPortugal
| |
Collapse
|
17
|
Masullo LA, Szalai AM, Lopez LF, Stefani FD. Fluorescence nanoscopy at the sub-10 nm scale. Biophys Rev 2021; 13:1101-1112. [PMID: 35059030 PMCID: PMC8724505 DOI: 10.1007/s12551-021-00864-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2021] [Accepted: 10/20/2021] [Indexed: 12/14/2022] Open
Abstract
Fluorescence nanoscopy represented a breakthrough for the life sciences as it delivers 20-30 nm resolution using far-field fluorescence microscopes. This resolution limit is not fundamental but imposed by the limited photostability of fluorophores under ambient conditions. This has motivated the development of a second generation of fluorescence nanoscopy methods that aim to deliver sub-10 nm resolution, reaching the typical size of structural proteins and thus providing true molecular resolution. In this review, we present common fundamental aspects of these nanoscopies, discuss the key experimental factors that are necessary to fully exploit their capabilities, and discuss their current and future challenges.
Collapse
Affiliation(s)
- Luciano A. Masullo
- Centro de Investigaciones en Bionanociencias (CIBION), Consejo Nacional de Investigaciones Científicas Y Técnicas (CONICET), Godoy Cruz 2390, C1425FQD Ciudad Autónoma de Buenos Aires, Argentina
- Departamento de Física, Facultad de Ciencias Exactas Y Naturales, Universidad de Buenos Aires, Güiraldes 2620, C1428EHA Ciudad Autónoma de Buenos Aires, Argentina
| | - Alan M. Szalai
- Centro de Investigaciones en Bionanociencias (CIBION), Consejo Nacional de Investigaciones Científicas Y Técnicas (CONICET), Godoy Cruz 2390, C1425FQD Ciudad Autónoma de Buenos Aires, Argentina
| | - Lucía F. Lopez
- Departamento de Física, Facultad de Ciencias Exactas Y Naturales, Universidad de Buenos Aires, Güiraldes 2620, C1428EHA Ciudad Autónoma de Buenos Aires, Argentina
| | - Fernando D. Stefani
- Centro de Investigaciones en Bionanociencias (CIBION), Consejo Nacional de Investigaciones Científicas Y Técnicas (CONICET), Godoy Cruz 2390, C1425FQD Ciudad Autónoma de Buenos Aires, Argentina
- Departamento de Física, Facultad de Ciencias Exactas Y Naturales, Universidad de Buenos Aires, Güiraldes 2620, C1428EHA Ciudad Autónoma de Buenos Aires, Argentina
| |
Collapse
|
18
|
Tas RP, Albertazzi L, Voets IK. Small Peptide-Protein Interaction Pair for Genetically Encoded, Fixation Compatible Peptide-PAINT. NANO LETTERS 2021; 21:9509-9516. [PMID: 34757759 PMCID: PMC8631740 DOI: 10.1021/acs.nanolett.1c02895] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/27/2021] [Revised: 11/01/2021] [Indexed: 05/08/2023]
Abstract
Super-resolution microscopy via PAINT has been widely adopted in life sciences to interrogate the nanoscale architecture of many cellular structures. However, obtaining quantitative information in fixed cellular samples remains challenging because control of labeling stoichiometry is hampered in current approaches due to click-chemistry and additional targeting probes. To overcome these challenges, we have identified a small, PDZ-based, peptide-protein interaction pair that is genetically encodable and compatible with super-resolution imaging upon cellular fixation without additional labeling. Stoichiometric labeling control by genetic incorporation of this probe into the cellular vimentin network and mitochondria resulted in super-resolved 3D reconstructions with high specificity and spatial resolution. Further characterization reveals that this peptide-protein interaction is compatible with quantitative PAINT and that its binding kinetics remains unaltered upon fixation. Finally, by fusion of our probe to nanobodies against conventional expression markers, we show that this approach provides a versatile addition to the super-resolution toolbox.
Collapse
Affiliation(s)
- Roderick P. Tas
- Laboratory
of Self-Organizing Soft Matter, Institute for Complex Molecular Systems
and Department of Chemical Engineering and Chemistry, Eindhoven University of Technology (TUE), Eindhoven 5612 AP, The Netherlands
| | - Lorenzo Albertazzi
- Laboratory
of Nanoscopy for Nanomedicine, Institute for Complex Molecular Systems
and Department of Biomedical Engineering, Eindhoven University of Technology (TUE), Eindhoven 5612 AP, The Netherlands
| | - Ilja K. Voets
- Laboratory
of Self-Organizing Soft Matter, Institute for Complex Molecular Systems
and Department of Chemical Engineering and Chemistry, Eindhoven University of Technology (TUE), Eindhoven 5612 AP, The Netherlands
| |
Collapse
|
19
|
Kocabey S, Ekim Kocabey A, Schneiter R, Rüegg C. Membrane-Interacting DNA Nanotubes Induce Cancer Cell Death. NANOMATERIALS 2021; 11:nano11082003. [PMID: 34443832 PMCID: PMC8397952 DOI: 10.3390/nano11082003] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/28/2021] [Revised: 07/25/2021] [Accepted: 07/30/2021] [Indexed: 12/31/2022]
Abstract
DNA nanotechnology offers to build nanoscale structures with defined chemistries to precisely position biomolecules or drugs for selective cell targeting and drug delivery. Owing to the negatively charged nature of DNA, for delivery purposes, DNA is frequently conjugated with hydrophobic moieties, positively charged polymers/peptides and cell surface receptor-recognizing molecules or antibodies. Here, we designed and assembled cholesterol-modified DNA nanotubes to interact with cancer cells and conjugated them with cytochrome c to induce cancer cell apoptosis. By flow cytometry and confocal microscopy, we observed that DNA nanotubes efficiently bound to the plasma membrane as a function of the number of conjugated cholesterol moieties. The complex was taken up by the cells and localized to the endosomal compartment. Cholesterol-modified DNA nanotubes, but not unmodified ones, increased membrane permeability, caspase activation and cell death. Irreversible inhibition of caspase activity with a caspase inhibitor, however, only partially prevented cell death. Cytochrome c-conjugated DNA nanotubes were also efficiently taken up but did not increase the rate of cell death. These results demonstrate that cholesterol-modified DNA nanotubes induce cancer cell death associated with increased cell membrane permeability and are only partially dependent on caspase activity, consistent with a combined form of apoptotic and necrotic cell death. DNA nanotubes may be further developed as primary cytotoxic agents, or drug delivery vehicles, through cholesterol-mediated cellular membrane interactions and uptake.
Collapse
Affiliation(s)
- Samet Kocabey
- Department of Oncology, Microbiology and Immunology, Faculty of Science and Medicine, University of Fribourg, Chemin du Musée 18, PER17, 1700 Fribourg, Switzerland
- Correspondence: (S.K.); (C.R.)
| | - Aslihan Ekim Kocabey
- Department of Biology, Faculty of Science and Medicine, University of Fribourg, Chemin du Musée 10, PER05, 1700 Fribourg, Switzerland; (A.E.K.); (R.S.)
| | - Roger Schneiter
- Department of Biology, Faculty of Science and Medicine, University of Fribourg, Chemin du Musée 10, PER05, 1700 Fribourg, Switzerland; (A.E.K.); (R.S.)
| | - Curzio Rüegg
- Department of Oncology, Microbiology and Immunology, Faculty of Science and Medicine, University of Fribourg, Chemin du Musée 18, PER17, 1700 Fribourg, Switzerland
- Correspondence: (S.K.); (C.R.)
| |
Collapse
|
20
|
Früh S, Matti U, Spycher PR, Rubini M, Lickert S, Schlichthaerle T, Jungmann R, Vogel V, Ries J, Schoen I. Site-Specifically-Labeled Antibodies for Super-Resolution Microscopy Reveal In Situ Linkage Errors. ACS NANO 2021; 15:12161-12170. [PMID: 34184536 PMCID: PMC8320235 DOI: 10.1021/acsnano.1c03677] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Accepted: 06/21/2021] [Indexed: 05/31/2023]
Abstract
The precise spatial localization of proteins in situ by super-resolution microscopy (SRM) demands their targeted labeling. Positioning reporter molecules as close as possible to the target remains a challenge in primary cells or tissues from patients that cannot be easily genetically modified. Indirect immunolabeling introduces relatively large linkage errors, whereas site-specific and stoichiometric labeling of primary antibodies relies on elaborate chemistries. In this study, we developed a simple two-step protocol to site-specifically attach reporters such as fluorophores or DNA handles to several immunoglobulin G (IgG) antibodies from different animal species and benchmarked the performance of these conjugates for 3D STORM (stochastic optical reconstruction microscopy) and DNA-PAINT (point accumulation in nanoscale topography). Glutamine labeling was restricted to two sites per IgG and saturable by exploiting microbial transglutaminase after removal of N-linked glycans. Precision measurements of 3D microtubule labeling shell dimensions in cell lines and human platelets showed that linkage errors from primary and secondary antibodies did not add up. Monte Carlo simulations of a geometric microtubule-IgG model were in quantitative agreement with STORM results. The simulations revealed that the flexible hinge between Fab and Fc segments effectively randomized the direction of the secondary antibody, while the restricted binding orientation of the primary antibody's Fab fragment accounted for most of the systematic offset between the reporter and α-tubulin. DNA-PAINT surprisingly yielded larger linkage errors than STORM, indicating unphysiological conformations of DNA-labeled IgGs. In summary, our cost-effective protocol for generating well-characterized primary IgG conjugates offers an easy route to precise SRM measurements in arbitrary fixed samples.
Collapse
Affiliation(s)
- Susanna
M. Früh
- Hahn-Schickard, Georges-Koehler-Allee 103, 79110 Freiburg, Germany
- Laboratory
for MEMS Applications, IMTEK, Department of Microsystems Engineering, University of Freiburg, 79110 Freiburg, Germany
| | - Ulf Matti
- Cell
Biology and Biophysics Unit, European Molecular
Biology Laboratory (EMBL), 69117 Heidelberg, Germany
| | - Philipp R. Spycher
- Center
for
Radiopharmaceutical Sciences, Paul Scherrer
Institute, 5232 Villigen, Switzerland
| | - Marina Rubini
- School
of Chemistry, University College Dublin, Belfield, Dublin 4, Ireland
| | - Sebastian Lickert
- Department
of Health Sciences and Technology, ETH Zurich, 8093 Zurich, Switzerland
| | - Thomas Schlichthaerle
- Faculty
of Physics and Center for Nanoscience, Ludwig
Maximilian University, 80539 Munich, Germany
- Max Planck
Institute of Biochemistry, 82152 Martinsried, Germany
| | - Ralf Jungmann
- Faculty
of Physics and Center for Nanoscience, Ludwig
Maximilian University, 80539 Munich, Germany
- Max Planck
Institute of Biochemistry, 82152 Martinsried, Germany
| | - Viola Vogel
- Department
of Health Sciences and Technology, ETH Zurich, 8093 Zurich, Switzerland
| | - Jonas Ries
- Cell
Biology and Biophysics Unit, European Molecular
Biology Laboratory (EMBL), 69117 Heidelberg, Germany
| | - Ingmar Schoen
- School
of Pharmacy and Biomolecular Sciences, Irish Centre for Vascular Biology, Royal College of Surgeons in Ireland (RCSI), Dublin 2, Ireland
| |
Collapse
|
21
|
van Wee R, Filius M, Joo C. Completing the canvas: advances and challenges for DNA-PAINT super-resolution imaging. Trends Biochem Sci 2021; 46:918-930. [PMID: 34247944 DOI: 10.1016/j.tibs.2021.05.010] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Revised: 05/16/2021] [Accepted: 05/31/2021] [Indexed: 01/02/2023]
Abstract
Single-molecule localization microscopy (SMLM) is a potent tool to examine biological systems with unprecedented resolution, enabling the investigation of increasingly smaller structures. At the forefront of these developments is DNA-based point accumulation for imaging in nanoscale topography (DNA-PAINT), which exploits the stochastic and transient binding of fluorescently labeled DNA probes. In its early stages the implementation of DNA-PAINT was burdened by low-throughput, excessive acquisition time, and difficult integration with live-cell imaging. However, recent advances are addressing these challenges and expanding the range of applications of DNA-PAINT. We review the current state of the art of DNA-PAINT in light of these advances and contemplate what further developments remain indispensable to realize live-cell imaging.
Collapse
Affiliation(s)
- Raman van Wee
- Department of BioNanoScience, Kavli Institute of Nanoscience, Delft University of Technology, Van der Maasweg 9, 2629 HZ Delft, The Netherlands
| | - Mike Filius
- Department of BioNanoScience, Kavli Institute of Nanoscience, Delft University of Technology, Van der Maasweg 9, 2629 HZ Delft, The Netherlands
| | - Chirlmin Joo
- Department of BioNanoScience, Kavli Institute of Nanoscience, Delft University of Technology, Van der Maasweg 9, 2629 HZ Delft, The Netherlands.
| |
Collapse
|
22
|
Ahmadi MKB, Mohammadi SA, Makvandi M, Mamouei M, Rahmati M, Dehghani H, Wood DW. Recent Advances in the Scaffold Engineering of Protein Binders. Curr Pharm Biotechnol 2021; 22:878-891. [PMID: 32838715 DOI: 10.2174/1389201021999200824101035] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2020] [Revised: 08/08/2020] [Accepted: 08/10/2020] [Indexed: 11/22/2022]
Abstract
In recent years, extensive attention has been given to the generation of new classes of ligand- specific binding proteins to supplement monoclonal antibodies. A combination of protein engineering and display technologies has been used to manipulate non-human antibodies for humanization and stabilization purposes or even the generation of new binding proteins. Engineered protein scaffolds can now be directed against therapeutic targets to treat cancer and immunological disorders. Although very few of these scaffolds have successfully passed clinical trials, their remarkable properties such as robust folding, high solubility, and small size motivate their employment as a tool for biology and applied science studies. Here, we have focused on the generation of new non-Ig binding proteins and single domain antibody manipulation, with a glimpse of their applications.
Collapse
Affiliation(s)
- Mohammad K B Ahmadi
- Department of Medical Biotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Seyed A Mohammadi
- Department of Medical Biotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Manoochehr Makvandi
- Department of Virology, School of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Morteza Mamouei
- Department of Animal Science, Ramin Agricultural and Natural Resources University, Ahvaz, Iran
| | - Mohammad Rahmati
- Department of Medical Biotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Hesam Dehghani
- Stem Cells Regenerative Research Group, Ressearch Institute of Biotechnology, Ferdowsi University of Mashhad, Azadi Square, Mashhad, Iran
| | - David W Wood
- Department of Chemical and Biomolecular Engineering, The Ohio State University, 151 W. Woodruff Ave., Columbus, OH 43210, United States
| |
Collapse
|
23
|
Danial JSH, Klenerman D. Single molecule imaging of protein aggregation in Dementia: Methods, insights and prospects. Neurobiol Dis 2021; 153:105327. [PMID: 33705938 PMCID: PMC8039184 DOI: 10.1016/j.nbd.2021.105327] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2021] [Revised: 02/21/2021] [Accepted: 03/04/2021] [Indexed: 02/06/2023] Open
Abstract
The aggregation of misfolded proteins is a fundamental pathology in neurodegeneration which remains poorly understood due to its exceptional complexity and lack of appropriate characterization tools that can probe the role of the low concentrations of heterogeneous protein aggregates formed during the progression of the disease. In this review, we explain the principles underlying the operation of single molecule microscopy, an imaging method that can resolve molecules one-by-one, its application to imaging and characterizing individual protein aggregates in human samples and in vitro as well as the important questions in neurobiology this has answered and can answer.
Collapse
Affiliation(s)
- John S H Danial
- Yusuf Hamied Department of Chemistry, University of Cambridge, Cambridge, United Kingdom; UK Dementia Research Institute, Department of Clinical Neurosciences, University of Cambridge, Cambridge, United Kingdom.
| | - David Klenerman
- Yusuf Hamied Department of Chemistry, University of Cambridge, Cambridge, United Kingdom; UK Dementia Research Institute, Department of Clinical Neurosciences, University of Cambridge, Cambridge, United Kingdom.
| |
Collapse
|
24
|
Ganji M, Schlichthaerle T, Eklund AS, Strauss S, Jungmann R. Quantitative Assessment of Labeling Probes for Super-Resolution Microscopy Using Designer DNA Nanostructures. Chemphyschem 2021; 22:911-914. [PMID: 33720501 PMCID: PMC8251534 DOI: 10.1002/cphc.202100185] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2021] [Indexed: 12/22/2022]
Abstract
Improving labeling probes for state-of-the-art super-resolution microscopy is becoming of major importance. However, there is currently a lack of tools to quantitatively evaluate probe performance regarding efficiency, precision, and achievable resolution in an unbiased yet modular fashion. Herein, we introduce designer DNA origami structures combined with DNA-PAINT to overcome this issue and evaluate labeling efficiency, precision, and quantification using antibodies and nanobodies as exemplary labeling probes. Whereas current assessment of binders is mostly qualitative, e. g. based on an expected staining pattern, we herein present a quantitative analysis platform of the antigen labeling efficiency and achievable resolution, allowing researchers to choose the best performing binder. The platform can furthermore be readily adapted for discovery and precise quantification of a large variety of additional labeling probes.
Collapse
Affiliation(s)
- Mahipal Ganji
- Faculty of Physics and Center for Nanoscience, LMU Munich, Geschwister-Scholl-Platz 1, 80539, Munich, Germany
- Max Planck Institute of Biochemistry, Am Klopferspitz 18, 82152, Martinsried, Germany
- Current Address: Department of Biochemistry, Indian Institute of Science, CV Raman Road, 560012, Bengaluru, India
| | - Thomas Schlichthaerle
- Faculty of Physics and Center for Nanoscience, LMU Munich, Geschwister-Scholl-Platz 1, 80539, Munich, Germany
- Max Planck Institute of Biochemistry, Am Klopferspitz 18, 82152, Martinsried, Germany
- Current Address: Department of Biochemistry, Institute for Protein Design, University of Washington, Seattle, WA, USA
| | - Alexandra S Eklund
- Faculty of Physics and Center for Nanoscience, LMU Munich, Geschwister-Scholl-Platz 1, 80539, Munich, Germany
- Max Planck Institute of Biochemistry, Am Klopferspitz 18, 82152, Martinsried, Germany
| | - Sebastian Strauss
- Faculty of Physics and Center for Nanoscience, LMU Munich, Geschwister-Scholl-Platz 1, 80539, Munich, Germany
- Max Planck Institute of Biochemistry, Am Klopferspitz 18, 82152, Martinsried, Germany
| | - Ralf Jungmann
- Faculty of Physics and Center for Nanoscience, LMU Munich, Geschwister-Scholl-Platz 1, 80539, Munich, Germany
- Max Planck Institute of Biochemistry, Am Klopferspitz 18, 82152, Martinsried, Germany
| |
Collapse
|
25
|
Tang AAS, Tiede C, McPherson MJ, Tomlinson DC. Isolation of Artificial Binding Proteins (Affimer Reagents) for Use in Molecular and Cellular Biology. Methods Mol Biol 2021; 2247:105-121. [PMID: 33301114 DOI: 10.1007/978-1-0716-1126-5_6] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/12/2023]
Abstract
Artificial binding proteins have been validated as alternatives to antibodies in their use as research reagents in molecular and cellular biology. For example, they have been used as inhibitors of protein-protein interactions to modulate activity, to facilitate crystallization, and as probes for cellular imaging.Phage display is a widely used approach for isolating target-specific binding reagents, and it has even been used to isolate isoform-specific binding proteins and binders that can distinguish between highly homologous protein domains.Here, we describe methods that have been employed in isolating highly specific artificial binding proteins against a wide range of target proteins.
Collapse
Affiliation(s)
- Anna A S Tang
- Astbury Centre for Structural and Molecular Biology, School of Molecular and Cellular Biology, University of Leeds, Leeds, UK
| | - Christian Tiede
- Astbury Centre for Structural and Molecular Biology, School of Molecular and Cellular Biology, University of Leeds, Leeds, UK
| | - Michael J McPherson
- Astbury Centre for Structural and Molecular Biology, School of Molecular and Cellular Biology, University of Leeds, Leeds, UK
| | - Darren C Tomlinson
- Astbury Centre for Structural and Molecular Biology, School of Molecular and Cellular Biology, University of Leeds, Leeds, UK.
| |
Collapse
|
26
|
Pushing the super-resolution limit: recent improvements in microscopy below the diffraction limit. Biochem Soc Trans 2021; 49:431-439. [PMID: 33599719 DOI: 10.1042/bst20200746] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Revised: 12/15/2020] [Accepted: 01/20/2021] [Indexed: 12/12/2022]
Abstract
Super-resolution microscopy has revolutionised the way we observe biological systems. These methods are now a staple of fluorescence microscopy. Researchers have used super-resolution methods in myriad systems to extract nanoscale spatial information on multiple interacting parts. These methods are continually being extended and reimagined to further push their resolving power and achieve truly single protein resolution. Here, we explore the most recent advances at the frontier of the 'super-resolution' limit and what opportunities remain for further improvements in the near future.
Collapse
|
27
|
Schueder F, Lara‐Gutiérrez J, Haas D, Beckwith KS, Yin P, Ellenberg J, Jungmann R. Superaufgelöste Erkennung räumlicher Nähe mit Proximity‐PAINT. Angew Chem Int Ed Engl 2021. [DOI: 10.1002/ange.202009031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Affiliation(s)
- Florian Schueder
- Faculty of Physics and Center for Nanoscience LMU Munich Geschwister-Scholl-Platz 1 80539 Munich Deutschland
- Max Planck Institute of Biochemistry Am Klopferspitz 18 82152 Martinsried Germany
| | - Juanita Lara‐Gutiérrez
- Max Planck Institute of Biochemistry Am Klopferspitz 18 82152 Martinsried Germany
- Department of Systems Biology and Wyss Institute for Biologically Inspired Engineering Harvard Medical School 3 Blackfan Circle Boston MA 02115 USA
| | - Daniel Haas
- Max Planck Institute of Biochemistry Am Klopferspitz 18 82152 Martinsried Germany
| | - Kai Sandvold Beckwith
- Cell Biology and Biophysics Unit European Molecular Biology Laboratory (EMBL) Meyerhofstraße 1 69117 Heidelberg Germany
| | - Peng Yin
- Department of Systems Biology and Wyss Institute for Biologically Inspired Engineering Harvard Medical School 3 Blackfan Circle Boston MA 02115 USA
| | - Jan Ellenberg
- Cell Biology and Biophysics Unit European Molecular Biology Laboratory (EMBL) Meyerhofstraße 1 69117 Heidelberg Germany
| | - Ralf Jungmann
- Faculty of Physics and Center for Nanoscience LMU Munich Geschwister-Scholl-Platz 1 80539 Munich Deutschland
- Max Planck Institute of Biochemistry Am Klopferspitz 18 82152 Martinsried Germany
| |
Collapse
|
28
|
Schueder F, Lara‐Gutiérrez J, Haas D, Beckwith KS, Yin P, Ellenberg J, Jungmann R. Super-Resolution Spatial Proximity Detection with Proximity-PAINT. Angew Chem Int Ed Engl 2021; 60:716-720. [PMID: 32936507 PMCID: PMC7839522 DOI: 10.1002/anie.202009031] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Revised: 08/11/2020] [Indexed: 12/21/2022]
Abstract
Visualizing the functional interactions of biomolecules such as proteins and nucleic acids is key to understanding cellular life on the molecular scale. Spatial proximity is often used as a proxy for the direct interaction of biomolecules. However, current techniques to visualize spatial proximity are either limited by spatial resolution, dynamic range, or lack of single-molecule sensitivity. Here, we introduce Proximity-PAINT (pPAINT), a variation of the super-resolution microscopy technique DNA-PAINT. pPAINT uses a split-docking-site configuration to detect spatial proximity with high sensitivity, low false-positive rates, and tunable detection distances. We benchmark and optimize pPAINT using designer DNA nanostructures and demonstrate its cellular applicability by visualizing the spatial proximity of alpha- and beta-tubulin in microtubules using super-resolution detection.
Collapse
Affiliation(s)
- Florian Schueder
- Faculty of Physics and Center for NanoscienceLMU MunichGeschwister-Scholl-Platz 180539MunichGermany
- Max Planck Institute of BiochemistryAm Klopferspitz 1882152MartinsriedGermany
| | - Juanita Lara‐Gutiérrez
- Max Planck Institute of BiochemistryAm Klopferspitz 1882152MartinsriedGermany
- Department of Systems Biology and Wyss Institute for Biologically Inspired EngineeringHarvard Medical School3 Blackfan CircleBostonMA02115USA
| | - Daniel Haas
- Max Planck Institute of BiochemistryAm Klopferspitz 1882152MartinsriedGermany
| | - Kai Sandvold Beckwith
- Cell Biology and Biophysics UnitEuropean Molecular Biology Laboratory (EMBL)Meyerhofstraße 169117HeidelbergGermany
| | - Peng Yin
- Department of Systems Biology and Wyss Institute for Biologically Inspired EngineeringHarvard Medical School3 Blackfan CircleBostonMA02115USA
| | - Jan Ellenberg
- Cell Biology and Biophysics UnitEuropean Molecular Biology Laboratory (EMBL)Meyerhofstraße 169117HeidelbergGermany
| | - Ralf Jungmann
- Faculty of Physics and Center for NanoscienceLMU MunichGeschwister-Scholl-Platz 180539MunichGermany
- Max Planck Institute of BiochemistryAm Klopferspitz 1882152MartinsriedGermany
| |
Collapse
|
29
|
Mahmoudi T, Pourhassan-Moghaddam M, Shirdel B, Baradaran B, Morales-Narváez E, Golmohammadi H. (Nano)tag-antibody conjugates in rapid tests. J Mater Chem B 2021; 9:5414-5438. [PMID: 34143173 DOI: 10.1039/d1tb00571e] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Antibodies (Abs) are naturally derived materials with favorable affinity, selectivity, and fast binding kinetics to the respective antigens, which enables their application as promising recognition elements in the development of various types of biosensors/bioassays, especially in rapid tests. These tests are low-cost and easy-to-use biosensing devices with broad applications including medical or veterinary diagnostics, environmental monitoring and industrial usages such as safety and quality analysis in food, providing on-site quick monitoring of various analytes, making it possible to save analysis costs and time. To reach such features, the conjugation of Abs with various nanomaterials (NMs) as tags is necessary, which range from conventional gold nanoparticles to other nanoparticles recently introduced, where magnetic, plasmonic, photoluminescent, or multi-modal properties play a critical role in the overall performance of the analytical device. In this context, to preserve the Ab affinity and provide a rapid response with long-term storage capability, the use of efficient bio-conjugation techniques is critical. Thanks to their prominent role in rapid tests, many studies have been devoted to the design and development of Abs-NMs conjugates with various chemistries including passive adsorption, covalent coupling, and affinity interactions. In this review, we present the state-of-the-art techniques allowing various Ab-NM conjugates with a special focus on the efficiency of the developed probes to be employed in in vitro rapid tests. Challenges and future perspectives on the development of Ab-conjugated nanotags in rapid diagnostic tests are highlighted along with a survey of the progress in commercially available Ab-NM conjugates.
Collapse
Affiliation(s)
- Tohid Mahmoudi
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
| | - Mohammad Pourhassan-Moghaddam
- ARC Research Hub for Integrated Device for End-user Analysis at Low-levels (IDEAL), Faculty of Science, University of Technology Sydney, Sydney, NSW 2007, Australia
| | - Behnaz Shirdel
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
| | - Behzad Baradaran
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
| | - Eden Morales-Narváez
- Biophotonic Nanosensors Laboratory, Centro de Investigaciones en Óptica, A. C. Loma del Bosque 115, Lomas del Campestre, 37150 León, Guanajuato, Mexico.
| | - Hamed Golmohammadi
- Nanosensors Bioplatforms Laboratory, Chemistry and Chemical Engineering Research Center of Iran, 14335-186, Tehran, Iran.
| |
Collapse
|
30
|
Lelek M, Gyparaki MT, Beliu G, Schueder F, Griffié J, Manley S, Jungmann R, Sauer M, Lakadamyali M, Zimmer C. Single-molecule localization microscopy. NATURE REVIEWS. METHODS PRIMERS 2021; 1:39. [PMID: 35663461 PMCID: PMC9160414 DOI: 10.1038/s43586-021-00038-x] [Citation(s) in RCA: 397] [Impact Index Per Article: 99.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/01/2023]
Abstract
Single-molecule localization microscopy (SMLM) describes a family of powerful imaging techniques that dramatically improve spatial resolution over standard, diffraction-limited microscopy techniques and can image biological structures at the molecular scale. In SMLM, individual fluorescent molecules are computationally localized from diffraction-limited image sequences and the localizations are used to generate a super-resolution image or a time course of super-resolution images, or to define molecular trajectories. In this Primer, we introduce the basic principles of SMLM techniques before describing the main experimental considerations when performing SMLM, including fluorescent labelling, sample preparation, hardware requirements and image acquisition in fixed and live cells. We then explain how low-resolution image sequences are computationally processed to reconstruct super-resolution images and/or extract quantitative information, and highlight a selection of biological discoveries enabled by SMLM and closely related methods. We discuss some of the main limitations and potential artefacts of SMLM, as well as ways to alleviate them. Finally, we present an outlook on advanced techniques and promising new developments in the fast-evolving field of SMLM. We hope that this Primer will be a useful reference for both newcomers and practitioners of SMLM.
Collapse
Affiliation(s)
- Mickaël Lelek
- Imaging and Modeling Unit, Department of Computational
Biology, Institut Pasteur, Paris, France
- CNRS, UMR 3691, Paris, France
| | - Melina T. Gyparaki
- Department of Biology, University of Pennsylvania,
Philadelphia, PA, USA
| | - Gerti Beliu
- Department of Biotechnology and Biophysics Biocenter,
University of Würzburg, Würzburg, Germany
| | - Florian Schueder
- Faculty of Physics and Center for Nanoscience, Ludwig
Maximilian University, Munich, Germany
- Max Planck Institute of Biochemistry, Martinsried,
Germany
| | - Juliette Griffié
- Laboratory of Experimental Biophysics, Institute of
Physics, École Polytechnique Fédérale de Lausanne (EPFL),
Lausanne, Switzerland
| | - Suliana Manley
- Laboratory of Experimental Biophysics, Institute of
Physics, École Polytechnique Fédérale de Lausanne (EPFL),
Lausanne, Switzerland
- ;
;
;
;
| | - Ralf Jungmann
- Faculty of Physics and Center for Nanoscience, Ludwig
Maximilian University, Munich, Germany
- Max Planck Institute of Biochemistry, Martinsried,
Germany
- ;
;
;
;
| | - Markus Sauer
- Department of Biotechnology and Biophysics Biocenter,
University of Würzburg, Würzburg, Germany
- ;
;
;
;
| | - Melike Lakadamyali
- Department of Physiology, Perelman School of Medicine,
University of Pennsylvania, Philadelphia, PA, USA
- Department of Cell and Developmental Biology, Perelman
School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Epigenetics Institute, Perelman School of Medicine,
University of Pennsylvania, Philadelphia, PA, USA
- ;
;
;
;
| | - Christophe Zimmer
- Imaging and Modeling Unit, Department of Computational
Biology, Institut Pasteur, Paris, France
- CNRS, UMR 3691, Paris, France
- ;
;
;
;
| |
Collapse
|
31
|
Abstract
Optical studies of single molecules have taught us much over the past three decades, but these individual quantum-mechanical objects continue to have promise as probes of nanoscale structure and dynamics in complex systems.
Collapse
Affiliation(s)
- W E Moerner
- Departments of Chemistry and of Applied Physics (courtesy), Stanford University, Stanford, California 94305-4401, United States
| |
Collapse
|
32
|
Oi C, Mochrie SGJ, Horrocks MH, Regan L. PAINT using proteins: A new brush for super-resolution artists. Protein Sci 2020; 29:2142-2149. [PMID: 32949055 PMCID: PMC7586915 DOI: 10.1002/pro.3953] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2020] [Revised: 09/11/2020] [Accepted: 09/15/2020] [Indexed: 02/06/2023]
Abstract
PAINT (points accumulation for imaging in nanoscale topography) refers to methods that achieve the sparse temporal labeling required for super-resolution imaging by using transient interactions between a biomolecule of interest and a fluorophore. There have been a variety of different implementations of this method since it was first described in 2006. Recent papers illustrate how transient peptide-protein interactions, rather than small molecule binding or DNA oligonucleotide duplex formation, can be employed to perform PAINT-based single molecule localization microscopy (SMLM). We discuss the different approaches to PAINT using peptide and protein interactions, and their applications in vitro and in vivo. We highlight the important parameters to consider when selecting suitable peptide-protein interaction pairs for such studies. We also note the opportunities for protein scientists to apply their expertise in guiding the choice of peptide and protein pairs that are used. Finally, we discuss the potential for expanding super-resolution imaging methods based on transient peptide-protein interactions, including the development of simultaneous multicolor imaging of multiple proteins and the study of very high and very low abundance proteins in live cells.
Collapse
Affiliation(s)
- Curran Oi
- Department of Molecular Biophysics and BiochemistryYale UniversityNew HavenConnecticutUSA
- Integrated Graduate Program in Physical and Engineering BiologyYale UniversityNew HavenConnecticutUSA
| | - Simon G. J. Mochrie
- Integrated Graduate Program in Physical and Engineering BiologyYale UniversityNew HavenConnecticutUSA
- Department of PhysicsYale UniversityNew HavenConnecticutUSA
| | | | - Lynne Regan
- Institute of Quantitative Biology, Biochemistry and Biotechnology, Centre for Synthetic and Systems Biology, School of Biological SciencesUniversity of EdinburghEdinburghUK
| |
Collapse
|
33
|
Thiele JC, Helmerich DA, Oleksiievets N, Tsukanov R, Butkevich E, Sauer M, Nevskyi O, Enderlein J. Confocal Fluorescence-Lifetime Single-Molecule Localization Microscopy. ACS NANO 2020; 14:14190-14200. [PMID: 33035050 DOI: 10.1021/acsnano.0c07322] [Citation(s) in RCA: 51] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/22/2023]
Abstract
Fluorescence lifetime imaging microscopy is an important technique that adds another dimension to intensity and color acquired by conventional microscopy. In particular, it allows for multiplexing fluorescent labels that have otherwise similar spectral properties. Currently, the only super-resolution technique that is capable of recording super-resolved images with lifetime information is stimulated emission depletion microscopy. In contrast, all single-molecule localization microscopy (SMLM) techniques that employ wide-field cameras completely lack the lifetime dimension. Here, we combine fluorescence-lifetime confocal laser-scanning microscopy with SMLM for realizing single-molecule localization-based fluorescence-lifetime super-resolution imaging. Besides yielding images with a spatial resolution much beyond the diffraction limit, it determines the fluorescence lifetime of all localized molecules. We validate our technique by applying it to direct stochastic optical reconstruction microscopy and points accumulation for imaging in nanoscale topography imaging of fixed cells, and we demonstrate its multiplexing capability on samples with two different labels that differ only by fluorescence lifetime but not by their spectral properties.
Collapse
Affiliation(s)
- Jan Christoph Thiele
- III. Institute of Physics-Biophysics, Georg August University, Göttingen 37077, Germany
| | - Dominic A Helmerich
- Department of Biotechnology and Biophysics, Biocenter, University of Würzburg, Am Hubland, Würzburg 97074, Germany
| | - Nazar Oleksiievets
- III. Institute of Physics-Biophysics, Georg August University, Göttingen 37077, Germany
| | - Roman Tsukanov
- III. Institute of Physics-Biophysics, Georg August University, Göttingen 37077, Germany
| | - Eugenia Butkevich
- III. Institute of Physics-Biophysics, Georg August University, Göttingen 37077, Germany
| | - Markus Sauer
- Department of Biotechnology and Biophysics, Biocenter, University of Würzburg, Am Hubland, Würzburg 97074, Germany
| | - Oleksii Nevskyi
- III. Institute of Physics-Biophysics, Georg August University, Göttingen 37077, Germany
| | - Jörg Enderlein
- III. Institute of Physics-Biophysics, Georg August University, Göttingen 37077, Germany
- Cluster of Excellence "Multiscale Bioimaging: from Molecular Machines to Networks of Excitable Cells" (MBExC), Georg August University, Göttingen 37077, Germany
| |
Collapse
|
34
|
Möckl L, Moerner WE. Super-resolution Microscopy with Single Molecules in Biology and Beyond-Essentials, Current Trends, and Future Challenges. J Am Chem Soc 2020; 142:17828-17844. [PMID: 33034452 PMCID: PMC7582613 DOI: 10.1021/jacs.0c08178] [Citation(s) in RCA: 105] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2020] [Indexed: 12/31/2022]
Abstract
Single-molecule super-resolution microscopy has developed from a specialized technique into one of the most versatile and powerful imaging methods of the nanoscale over the past two decades. In this perspective, we provide a brief overview of the historical development of the field, the fundamental concepts, the methodology required to obtain maximum quantitative information, and the current state of the art. Then, we will discuss emerging perspectives and areas where innovation and further improvement are needed. Despite the tremendous progress, the full potential of single-molecule super-resolution microscopy is yet to be realized, which will be enabled by the research ahead of us.
Collapse
Affiliation(s)
- Leonhard Möckl
- Department of Chemistry, Stanford University, Stanford, California 94305, United States
| | - W. E. Moerner
- Department of Chemistry, Stanford University, Stanford, California 94305, United States
| |
Collapse
|
35
|
Delcanale P, Porciani D, Pujals S, Jurkevich A, Chetrusca A, Tawiah KD, Burke DH, Albertazzi L. Aptamers with Tunable Affinity Enable Single-Molecule Tracking and Localization of Membrane Receptors on Living Cancer Cells. Angew Chem Int Ed Engl 2020; 59:18546-18555. [PMID: 32627326 PMCID: PMC7590183 DOI: 10.1002/anie.202004764] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2020] [Indexed: 12/31/2022]
Abstract
Tumor cell-surface markers are usually overexpressed or mutated protein receptors for which spatiotemporal regulation differs between and within cancers. Single-molecule fluorescence imaging can profile individual markers in different cellular contexts with molecular precision. However, standard single-molecule imaging methods based on overexpressed genetically encoded tags or cumbersome probes can significantly alter the native state of receptors. We introduce a live-cell points accumulation for imaging in nanoscale topography (PAINT) method that exploits aptamers as minimally invasive affinity probes. Localization and tracking of individual receptors are based on stochastic and transient binding between aptamers and their targets. We demonstrated single-molecule imaging of a model tumor marker (EGFR) on a panel of living cancer cells. Affinity to EGFR was finely tuned by rational engineering of aptamer sequences to define receptor motion and/or native receptor density.
Collapse
Affiliation(s)
- Pietro Delcanale
- Institute for Bioengineering of Catalonia (IBEC)The Barcelona Institute of Science and Technology (BIST)Baldiri Reixac 15–2108028BarcelonaSpain
| | - David Porciani
- Department of Molecular Microbiology & ImmunologySchool of MedicineUniversity of Missouri-Columbia1 Hospital DrColumbiaMO65212USA
- MU Bond Life Sciences CenterUniversity of Missouri-Columbia1201 Rollins StreetColumbiaMO65211-7310USA
| | - Silvia Pujals
- Institute for Bioengineering of Catalonia (IBEC)The Barcelona Institute of Science and Technology (BIST)Baldiri Reixac 15–2108028BarcelonaSpain
- Department of Electronics and Biomedical EngineeringFaculty of PhysicsUniversitat de BarcelonaMartí i Franquès 108028BarcelonaSpain
| | - Alexander Jurkevich
- Molecular Cytology Core at MU Bond Life Sciences CenterUniversity of Missouri-ColumbiaUSA
| | - Andrian Chetrusca
- Institute for Bioengineering of Catalonia (IBEC)The Barcelona Institute of Science and Technology (BIST)Baldiri Reixac 15–2108028BarcelonaSpain
| | - Kwaku D. Tawiah
- Department of BiochemistryUniversity of Missouri-Columbia117 Schweitzer HallColumbiaMO65211USA
| | - Donald H. Burke
- Department of Molecular Microbiology & ImmunologySchool of MedicineUniversity of Missouri-Columbia1 Hospital DrColumbiaMO65212USA
- MU Bond Life Sciences CenterUniversity of Missouri-Columbia1201 Rollins StreetColumbiaMO65211-7310USA
- Department of BiochemistryUniversity of Missouri-Columbia117 Schweitzer HallColumbiaMO65211USA
| | - Lorenzo Albertazzi
- Institute for Bioengineering of Catalonia (IBEC)The Barcelona Institute of Science and Technology (BIST)Baldiri Reixac 15–2108028BarcelonaSpain
- Department of Biomedical EngineeringInstitute for Complex Molecular Systems (ICMS)Eindhoven University of Technology5612AZEindhovenThe Netherlands
| |
Collapse
|
36
|
Schueder F, Unterauer EM, Ganji M, Jungmann R. DNA-Barcoded Fluorescence Microscopy for Spatial Omics. Proteomics 2020; 20:e1900368. [PMID: 33030780 DOI: 10.1002/pmic.201900368] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2020] [Revised: 07/24/2020] [Indexed: 12/18/2022]
Abstract
Innovation in genomics, transcriptomics, and proteomics research has created a plethora of state-of-the-art techniques such as nucleic acid sequencing and mass-spectrometry-based proteomics with paramount impact in the life sciences. While current approaches yield quantitative abundance analysis of biomolecules on an almost routine basis, coupling this high content to spatial information in a single cell and tissue context is challenging. Here, current implementations of spatial omics are discussed and recent developments in the field of DNA-barcoded fluorescence microscopy are reviewed. Light is shed on the potential of DNA-based imaging techniques to provide a comprehensive toolbox for spatial genomics and transcriptomics and discuss current challenges, which need to be overcome on the way to spatial proteomics using high-resolution fluorescence microscopy.
Collapse
Affiliation(s)
- Florian Schueder
- Department of Physics and Center for Nanoscience, Ludwig Maximilian University, Geschwister-Scholl-Platz 1, Munich, 80539, Germany.,Max Planck Institute of Biochemistry, Am Klopferspitz 18, Martinsried, 82152, Germany
| | - Eduard M Unterauer
- Department of Physics and Center for Nanoscience, Ludwig Maximilian University, Geschwister-Scholl-Platz 1, Munich, 80539, Germany.,Max Planck Institute of Biochemistry, Am Klopferspitz 18, Martinsried, 82152, Germany
| | - Mahipal Ganji
- Department of Physics and Center for Nanoscience, Ludwig Maximilian University, Geschwister-Scholl-Platz 1, Munich, 80539, Germany.,Max Planck Institute of Biochemistry, Am Klopferspitz 18, Martinsried, 82152, Germany
| | - Ralf Jungmann
- Department of Physics and Center for Nanoscience, Ludwig Maximilian University, Geschwister-Scholl-Platz 1, Munich, 80539, Germany.,Max Planck Institute of Biochemistry, Am Klopferspitz 18, Martinsried, 82152, Germany
| |
Collapse
|
37
|
Eklund A, Ganji M, Gavins G, Seitz O, Jungmann R. Peptide-PAINT Super-Resolution Imaging Using Transient Coiled Coil Interactions. NANO LETTERS 2020; 20:6732-6737. [PMID: 32787168 PMCID: PMC7496730 DOI: 10.1021/acs.nanolett.0c02620] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/24/2020] [Revised: 07/26/2020] [Indexed: 05/24/2023]
Abstract
Super-resolution microscopy is transforming research in the life sciences by enabling the visualization of structures and interactions on the nanoscale. DNA-PAINT is a relatively easy-to-implement single-molecule-based technique, which uses the programmable and transient interaction of dye-labeled oligonucleotides with their complements for super-resolution imaging. However, similar to many imaging approaches, it is still hampered by the subpar performance of labeling probes in terms of their large size and limited labeling efficiency. To overcome this, we here translate the programmability and transient binding nature of DNA-PAINT to coiled coil interactions of short peptides and introduce Peptide-PAINT. We benchmark and optimize its binding kinetics in a single-molecule assay and demonstrate its super-resolution capability using self-assembled DNA origami structures. Peptide-PAINT outperforms classical DNA-PAINT in terms of imaging speed and efficiency. Finally, we prove the suitability of Peptide-PAINT for cellular super-resolution imaging by visualizing the microtubule and vimentin network in fixed cells.
Collapse
Affiliation(s)
- Alexandra
S. Eklund
- Faculty
of Physics and Center for Nanoscience, Ludwig
Maximilian University, Munich, Germany
- Max
Planck Institute of Biochemistry, Martinsried, Germany
| | - Mahipal Ganji
- Faculty
of Physics and Center for Nanoscience, Ludwig
Maximilian University, Munich, Germany
- Max
Planck Institute of Biochemistry, Martinsried, Germany
| | - Georgina Gavins
- Institute
of Chemistry, Humboldt University, Berlin, Germany
| | - Oliver Seitz
- Institute
of Chemistry, Humboldt University, Berlin, Germany
| | - Ralf Jungmann
- Faculty
of Physics and Center for Nanoscience, Ludwig
Maximilian University, Munich, Germany
- Max
Planck Institute of Biochemistry, Martinsried, Germany
| |
Collapse
|
38
|
Delcanale P, Porciani D, Pujals S, Jurkevich A, Chetrusca A, Tawiah KD, Burke DH, Albertazzi L. Aptamers with Tunable Affinity Enable Single‐Molecule Tracking and Localization of Membrane Receptors on Living Cancer Cells. Angew Chem Int Ed Engl 2020. [DOI: 10.1002/ange.202004764] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Affiliation(s)
- Pietro Delcanale
- Institute for Bioengineering of Catalonia (IBEC) The Barcelona Institute of Science and Technology (BIST) Baldiri Reixac 15–21 08028 Barcelona Spain
| | - David Porciani
- Department of Molecular Microbiology & Immunology School of Medicine University of Missouri-Columbia 1 Hospital Dr Columbia MO 65212 USA
- MU Bond Life Sciences Center University of Missouri-Columbia 1201 Rollins Street Columbia MO 65211-7310 USA
| | - Silvia Pujals
- Institute for Bioengineering of Catalonia (IBEC) The Barcelona Institute of Science and Technology (BIST) Baldiri Reixac 15–21 08028 Barcelona Spain
- Department of Electronics and Biomedical Engineering Faculty of Physics Universitat de Barcelona Martí i Franquès 1 08028 Barcelona Spain
| | - Alexander Jurkevich
- Molecular Cytology Core at MU Bond Life Sciences Center University of Missouri-Columbia USA
| | - Andrian Chetrusca
- Institute for Bioengineering of Catalonia (IBEC) The Barcelona Institute of Science and Technology (BIST) Baldiri Reixac 15–21 08028 Barcelona Spain
| | - Kwaku D. Tawiah
- Department of Biochemistry University of Missouri-Columbia 117 Schweitzer Hall Columbia MO 65211 USA
| | - Donald H. Burke
- Department of Molecular Microbiology & Immunology School of Medicine University of Missouri-Columbia 1 Hospital Dr Columbia MO 65212 USA
- MU Bond Life Sciences Center University of Missouri-Columbia 1201 Rollins Street Columbia MO 65211-7310 USA
- Department of Biochemistry University of Missouri-Columbia 117 Schweitzer Hall Columbia MO 65211 USA
| | - Lorenzo Albertazzi
- Institute for Bioengineering of Catalonia (IBEC) The Barcelona Institute of Science and Technology (BIST) Baldiri Reixac 15–21 08028 Barcelona Spain
- Department of Biomedical Engineering Institute for Complex Molecular Systems (ICMS) Eindhoven University of Technology 5612AZ Eindhoven The Netherlands
| |
Collapse
|
39
|
Strauss S, Jungmann R. Up to 100-fold speed-up and multiplexing in optimized DNA-PAINT. Nat Methods 2020; 17:789-791. [PMID: 32601424 PMCID: PMC7610413 DOI: 10.1038/s41592-020-0869-x] [Citation(s) in RCA: 115] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2020] [Accepted: 05/18/2020] [Indexed: 12/12/2022]
Abstract
DNA-PAINT's imaging speed has recently been significantly enhanced by optimized sequence design and buffer conditions. However, this implementation has not reached an ultimate speed limit and is only applicable to imaging of single targets. To further improve acquisition speed, we introduce concatenated, periodic DNA sequence motifs, yielding up to 100-fold-faster sampling in comparison to traditional DNA-PAINT. We extend this approach to six orthogonal sequence motifs, now enabling speed-optimized multiplexed imaging.
Collapse
Affiliation(s)
- Sebastian Strauss
- Faculty of Physics and Center for Nanoscience, Ludwig Maximilian University, Munich, Germany.,Max Planck Institute of Biochemistry, Martinsried, Germany
| | - Ralf Jungmann
- Faculty of Physics and Center for Nanoscience, Ludwig Maximilian University, Munich, Germany. .,Max Planck Institute of Biochemistry, Martinsried, Germany.
| |
Collapse
|
40
|
Carrington G, Tomlinson D, Peckham M. Exploiting nanobodies and Affimers for superresolution imaging in light microscopy. Mol Biol Cell 2020; 30:2737-2740. [PMID: 31609674 PMCID: PMC6789155 DOI: 10.1091/mbc.e18-11-0694] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Antibodies have long been the main approach used for localizing proteins of interest by light microscopy. In the past 5 yr or so, and with the advent of superresolution microscopy, the diversity of tools for imaging has rapidly expanded. One main area of expansion has been in the area of nanobodies, small single-chain antibodies from camelids or sharks. The other has been the use of artificial scaffold proteins, including Affimers. The small size of nanobodies and Affimers compared with the traditional antibody provides several advantages for superresolution imaging.
Collapse
Affiliation(s)
- Glenn Carrington
- School of Molecular and Cellular Biology, University of Leeds, Leeds LS2 9JT, United Kingdom
| | - Darren Tomlinson
- School of Molecular and Cellular Biology, University of Leeds, Leeds LS2 9JT, United Kingdom
| | - Michelle Peckham
- School of Molecular and Cellular Biology, University of Leeds, Leeds LS2 9JT, United Kingdom
| |
Collapse
|
41
|
Clowsley AH, Kaufhold WT, Lutz T, Meletiou A, Di Michele L, Soeller C. Detecting Nanoscale Distribution of Protein Pairs by Proximity-Dependent Super-resolution Microscopy. J Am Chem Soc 2020; 142:12069-12078. [DOI: 10.1021/jacs.9b03418] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Affiliation(s)
- Alexander H. Clowsley
- Living Systems Institute & Biomedical Physics, University of Exeter, Exeter EX4 4QD, U.K
| | - William T. Kaufhold
- Cavendish Laboratory, University of Cambridge, Cambridge CB3 0HE, U.K
- Department of Chemistry, Molecular Sciences Research Hub, Imperial College London, London SW7 2AZ, U.K
| | - Tobias Lutz
- Living Systems Institute & Biomedical Physics, University of Exeter, Exeter EX4 4QD, U.K
| | - Anna Meletiou
- Living Systems Institute & Biomedical Physics, University of Exeter, Exeter EX4 4QD, U.K
| | - Lorenzo Di Michele
- Cavendish Laboratory, University of Cambridge, Cambridge CB3 0HE, U.K
- Department of Chemistry, Molecular Sciences Research Hub, Imperial College London, London SW7 2AZ, U.K
| | - Christian Soeller
- Living Systems Institute & Biomedical Physics, University of Exeter, Exeter EX4 4QD, U.K
| |
Collapse
|
42
|
Sograte-Idrissi S, Schlichthaerle T, Duque-Afonso CJ, Alevra M, Strauss S, Moser T, Jungmann R, Rizzoli SO, Opazo F. Circumvention of common labelling artefacts using secondary nanobodies. NANOSCALE 2020; 12:10226-10239. [PMID: 32356544 DOI: 10.1039/d0nr00227e] [Citation(s) in RCA: 56] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
A standard procedure to study cellular elements is via immunostaining followed by optical imaging. This methodology typically requires target-specific primary antibodies (1.Abs), which are revealed by secondary antibodies (2.Abs). Unfortunately, the antibody bivalency, polyclonality, and large size can result in a series of artifacts. Alternatively, small, monovalent probes, such as single-domain antibodies (nanobodies) have been suggested to minimize these limitations. The discovery and validation of nanobodies against specific targets are challenging, thus only a minimal amount of them are currently available. Here, we used STED, DNA-PAINT, and light-sheet microscopy, to demonstrate that secondary nanobodies (1) increase localization accuracy compared to 2.Abs; (2) allow direct pre-mixing with 1.Abs before staining, reducing experimental time, and enabling the use of multiple 1.Abs from the same species; (3) penetrate thick tissues more efficiently; and (4) avoid probe-induced clustering of target molecules observed with conventional 2.Abs in living or poorly fixed samples. Altogether, we show how secondary nanobodies are a valuable alternative to 2.Abs.
Collapse
Affiliation(s)
- Shama Sograte-Idrissi
- Institute of Neuro- and Sensory Physiology, University Medical Center Göttingen, 37073 Göttingen, Germany. and Center for Biostructural Imaging of Neurodegeneration (BIN), University of Göttingen Medical Center, 37075 Göttingen, Germany and International Max Planck Research School for Molecular Biology, Göttingen, Germany
| | - Thomas Schlichthaerle
- Faculty of Physics and Center for Nanoscience, LMU Munich, 80539, Munich, Germany and Max Planck Institute of Biochemistry, 82152, Martinsried, Germany
| | - Carlos J Duque-Afonso
- Institute for Auditory Neuroscience and InnerEarLab, University Medical Center Göttingen, 37075 Göttingen, Germany and Max Planck Institute for Experimental Medicine, 37075 Göttingen, Germany and Multiscale Bioimaging Cluster of Excellence (MBExC), University of Göttingen, 37075 Göttingen, Germany and University of Göttingen, 37075, Göttingen, Germany
| | - Mihai Alevra
- Institute of Neuro- and Sensory Physiology, University Medical Center Göttingen, 37073 Göttingen, Germany.
| | - Sebastian Strauss
- Faculty of Physics and Center for Nanoscience, LMU Munich, 80539, Munich, Germany and Max Planck Institute of Biochemistry, 82152, Martinsried, Germany
| | - Tobias Moser
- Institute for Auditory Neuroscience and InnerEarLab, University Medical Center Göttingen, 37075 Göttingen, Germany and Max Planck Institute for Experimental Medicine, 37075 Göttingen, Germany and Multiscale Bioimaging Cluster of Excellence (MBExC), University of Göttingen, 37075 Göttingen, Germany and University of Göttingen, 37075, Göttingen, Germany
| | - Ralf Jungmann
- Faculty of Physics and Center for Nanoscience, LMU Munich, 80539, Munich, Germany and Max Planck Institute of Biochemistry, 82152, Martinsried, Germany
| | - Silvio O Rizzoli
- Institute of Neuro- and Sensory Physiology, University Medical Center Göttingen, 37073 Göttingen, Germany. and Center for Biostructural Imaging of Neurodegeneration (BIN), University of Göttingen Medical Center, 37075 Göttingen, Germany and Multiscale Bioimaging Cluster of Excellence (MBExC), University of Göttingen, 37075 Göttingen, Germany
| | - Felipe Opazo
- Institute of Neuro- and Sensory Physiology, University Medical Center Göttingen, 37073 Göttingen, Germany. and Center for Biostructural Imaging of Neurodegeneration (BIN), University of Göttingen Medical Center, 37075 Göttingen, Germany and NanoTag Biotechnologies GmbH, 37079, Göttingen, Germany
| |
Collapse
|
43
|
Zhang L, Wang F, Li Q, Wang L, Fan C, Li J, Zhu Y. Classifying Cell Types with DNA-Encoded Ligand-Receptor Interactions on the Cell Membrane. NANO LETTERS 2020; 20:3521-3527. [PMID: 32223268 DOI: 10.1021/acs.nanolett.0c00445] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Clustering, endocytosis, and intracellular transport of molecules on the cell membrane are critically dependent on the type of cells. However, the membrane-associated redistribution of molecules has not been exploited to realize cell classification for diagnostic purposes. Here, we develop a set of DNA-encoded artificial receptors and ligands to monitor the cell membrane redistribution. In this system, a cholesterol-modified single-stranded DNA strand serves as the receptor localized on the membrane, and a tetrahedral DNA framework (TDF) nanostructure with a complementary overhang serves as the ligand. The DNA-encoded receptor-ligand interaction is highly orthogonal, mimicking the dynamics of natural receptors and ligands on cells. We demonstrate that the dynamics of membrane redistribution can be resolved by the dual-color fluorescent patterns of the receptor-ligand interactions in a single image, which can be exploited to classify cell lines with high fidelity. This DNA-encoded method thus holds great promise for cell typing and diagnosis.
Collapse
Affiliation(s)
- Luhao Zhang
- Division of Physical Biology, CAS Key Laboratory of Interfacial Physics and Technology, Shanghai Institute of Applied Physics, Chinese Academy of Sciences, Shanghai 201800, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Fei Wang
- Frontiers Science Center for Transformative Molecules, School of Chemistry and Chemical Engineering, Institute of Molecular Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200024, China
| | - Qian Li
- Frontiers Science Center for Transformative Molecules, School of Chemistry and Chemical Engineering, Institute of Molecular Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200024, China
| | - Lihua Wang
- Division of Physical Biology, CAS Key Laboratory of Interfacial Physics and Technology, Shanghai Institute of Applied Physics, Chinese Academy of Sciences, Shanghai 201800, China
- Bioimaging Center, Shanghai Synchrotron Radiation Facility, Zhangjiang Laboratory, Shanghai Advanced Research Institute, Chinese Academy of Sciences, Shanghai 201204, China
| | - Chunhai Fan
- Frontiers Science Center for Transformative Molecules, School of Chemistry and Chemical Engineering, Institute of Molecular Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200024, China
| | - Jiang Li
- Division of Physical Biology, CAS Key Laboratory of Interfacial Physics and Technology, Shanghai Institute of Applied Physics, Chinese Academy of Sciences, Shanghai 201800, China
- Bioimaging Center, Shanghai Synchrotron Radiation Facility, Zhangjiang Laboratory, Shanghai Advanced Research Institute, Chinese Academy of Sciences, Shanghai 201204, China
| | - Ying Zhu
- Division of Physical Biology, CAS Key Laboratory of Interfacial Physics and Technology, Shanghai Institute of Applied Physics, Chinese Academy of Sciences, Shanghai 201800, China
- Bioimaging Center, Shanghai Synchrotron Radiation Facility, Zhangjiang Laboratory, Shanghai Advanced Research Institute, Chinese Academy of Sciences, Shanghai 201204, China
| |
Collapse
|
44
|
Pereira PM, Gustafsson N, Marsh M, Mhlanga MM, Henriques R. Super-beacons: Open-source probes with spontaneous tuneable blinking compatible with live-cell super-resolution microscopy. Traffic 2020; 21:375-385. [PMID: 32170988 PMCID: PMC7643006 DOI: 10.1111/tra.12728] [Citation(s) in RCA: 6] [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: 02/07/2020] [Revised: 03/10/2020] [Accepted: 03/11/2020] [Indexed: 11/28/2022]
Abstract
Localization-based super-resolution microscopy relies on the detection of individual molecules cycling between fluorescent and non-fluorescent states. These transitions are commonly regulated by high-intensity illumination, imposing constrains to imaging hardware and producing sample photodamage. Here, we propose single-molecule self-quenching as a mechanism to generate spontaneous photoswitching. To demonstrate this principle, we developed a new class of DNA-based open-source super-resolution probes named super-beacons, with photoswitching kinetics that can be tuned structurally, thermally and chemically. The potential of these probes for live-cell compatible super-resolution microscopy without high-illumination or toxic imaging buffers is revealed by imaging interferon inducible transmembrane proteins (IFITMs) at sub-100 nm resolutions.
Collapse
Affiliation(s)
- Pedro M. Pereira
- MRC‐Laboratory for Molecular Cell BiologyUniversity College LondonLondonUK
- The Francis Crick InstituteLondonUK
- Bacterial Cell BiologyMOSTMICRO, ITQB‐NOVAOeirasPortugal
| | - Nils Gustafsson
- MRC‐Laboratory for Molecular Cell BiologyUniversity College LondonLondonUK
- Present address:
Department für Physik and CeNSLudwig‐Maximilians‐UniversitätMunichGermany
| | - Mark Marsh
- MRC‐Laboratory for Molecular Cell BiologyUniversity College LondonLondonUK
| | - Musa M. Mhlanga
- Institute of Infectious Disease and Molecular Medicine, Faculty of Health SciencesUniversity of Cape TownCape TownSouth Africa
| | - Ricardo Henriques
- MRC‐Laboratory for Molecular Cell BiologyUniversity College LondonLondonUK
- The Francis Crick InstituteLondonUK
| |
Collapse
|
45
|
Adamson H, Nicholl A, Tiede C, Tang AA, Davidson A, Curd H, Wignall A, Ford R, Nuttall J, McPherson MJ, Johnson M, Tomlinson DC. Affimers as anti-idiotypic affinity reagents for pharmacokinetic analysis of biotherapeutics. Biotechniques 2019; 67:261-269. [DOI: 10.2144/btn-2019-0089] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
Therapeutic antibodies are the fastest growing class of drugs in the treatment of cancer, and autoimmune and inflammatory diseases that require the concomitant development of assays to monitor therapeutic antibody levels. Here, we demonstrate that the use of Affimer nonantibody binding proteins provides an advantage over current antibody-based detection systems. For four therapeutic antibodies, we used phage display to isolate highly specific anti-idiotypic Affimer reagents, which selectively bind to the therapeutic antibody idiotype. For each antibody target the calibration curves met US Food and Drug Administration criteria and the dynamic range compared favorably with commercially available reagents. Affimer proteins therefore represent promising anti-idiotypic reagents that are simple to select and manufacture, and that offer the sensitivity, specificity and consistency required for pharmacokinetic assays.
Collapse
Affiliation(s)
- Hope Adamson
- School of Molecular & Cellular Biology, Astbury Centre for Structural Molecular Biology, University of Leeds, Leeds, LS2 9JT, UK
| | - Amanda Nicholl
- Avacta Life Sciences, Ash Way, Thorp Arch Estate, Wetherby, LS23 7FA, UK
| | - Christian Tiede
- School of Molecular & Cellular Biology, Astbury Centre for Structural Molecular Biology, University of Leeds, Leeds, LS2 9JT, UK
| | - Anna A Tang
- School of Molecular & Cellular Biology, Astbury Centre for Structural Molecular Biology, University of Leeds, Leeds, LS2 9JT, UK
| | - Alex Davidson
- Avacta Life Sciences, Ash Way, Thorp Arch Estate, Wetherby, LS23 7FA, UK
| | - Helen Curd
- Avacta Life Sciences, Ash Way, Thorp Arch Estate, Wetherby, LS23 7FA, UK
| | - Alex Wignall
- Avacta Life Sciences, Ash Way, Thorp Arch Estate, Wetherby, LS23 7FA, UK
| | - Robert Ford
- Avacta Life Sciences, Ash Way, Thorp Arch Estate, Wetherby, LS23 7FA, UK
| | - James Nuttall
- Avacta Life Sciences, Ash Way, Thorp Arch Estate, Wetherby, LS23 7FA, UK
| | - Michael J McPherson
- School of Molecular & Cellular Biology, Astbury Centre for Structural Molecular Biology, University of Leeds, Leeds, LS2 9JT, UK
| | - Matt Johnson
- Avacta Life Sciences, Ash Way, Thorp Arch Estate, Wetherby, LS23 7FA, UK
| | - Darren C Tomlinson
- School of Molecular & Cellular Biology, Astbury Centre for Structural Molecular Biology, University of Leeds, Leeds, LS2 9JT, UK
| |
Collapse
|
46
|
Nieves DJ, Hilzenrat G, Tran J, Yang Z, MacRae HH, Baker MAB, Gooding JJ, Gaus K. tagPAINT: covalent labelling of genetically encoded protein tags for DNA-PAINT imaging. ROYAL SOCIETY OPEN SCIENCE 2019; 6:191268. [PMID: 31903209 PMCID: PMC6936279 DOI: 10.1098/rsos.191268] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/23/2019] [Accepted: 11/12/2019] [Indexed: 05/17/2023]
Abstract
Recently, DNA-PAINT single-molecule localization microscopy (SMLM) has shown great promise for quantitative imaging; however, labelling strategies thus far have relied on multivalent and affinity-based approaches. Here, the covalent labelling of expressed protein tags (SNAP tag and Halo tag) with single DNA-docking strands and application of SMLM via DNA-PAINT is demonstrated. tagPAINT is then used for T-cell receptor signalling proteins at the immune synapse as a proof of principle.
Collapse
Affiliation(s)
- Daniel J. Nieves
- Institute of Immunology and Immunotherapy, College of Medical and Dental Sciences, University of Birmingham, Birmingham, B15 2TT, UK
- EMBL Australia Node in Single Molecule Science, School of Medical Sciences, University of New South Wales, Sydney, NSW 2052, Australia
- ARC Centre of Excellence in Advanced Molecular Imaging, University of New South Wales, Sydney, NSW 2052, Australia
- Authors for correspondence: Daniel J. Nieves e-mail:
| | - Geva Hilzenrat
- EMBL Australia Node in Single Molecule Science, School of Medical Sciences, University of New South Wales, Sydney, NSW 2052, Australia
- ARC Centre of Excellence in Advanced Molecular Imaging, University of New South Wales, Sydney, NSW 2052, Australia
| | - Jason Tran
- EMBL Australia Node in Single Molecule Science, School of Medical Sciences, University of New South Wales, Sydney, NSW 2052, Australia
- ARC Centre of Excellence in Advanced Molecular Imaging, University of New South Wales, Sydney, NSW 2052, Australia
| | - Zhengmin Yang
- EMBL Australia Node in Single Molecule Science, School of Medical Sciences, University of New South Wales, Sydney, NSW 2052, Australia
- ARC Centre of Excellence in Advanced Molecular Imaging, University of New South Wales, Sydney, NSW 2052, Australia
| | - Hugh H. MacRae
- EMBL Australia Node in Single Molecule Science, School of Medical Sciences, University of New South Wales, Sydney, NSW 2052, Australia
- ARC Centre of Excellence in Advanced Molecular Imaging, University of New South Wales, Sydney, NSW 2052, Australia
| | - Matthew A. B. Baker
- School of Biotechnology and Biomolecular Science, University of New South Wales, Sydney, NSW 2052, Australia
- Commonwealth Scientific and Industrial Research Organisation (CSIRO), Synthetic Biology Future Science Platform, Brisbane, Australia
| | - J. Justin Gooding
- School of Chemistry, University of New South Wales, Sydney, NSW 2052, Australia
- Australian Centre for NanoMedicine and the ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, University of New South Wales, Sydney, NSW 2052, Australia
| | - Katharina Gaus
- EMBL Australia Node in Single Molecule Science, School of Medical Sciences, University of New South Wales, Sydney, NSW 2052, Australia
- ARC Centre of Excellence in Advanced Molecular Imaging, University of New South Wales, Sydney, NSW 2052, Australia
- Authors for correspondence: Katharina Gaus e-mail:
| |
Collapse
|
47
|
Gebauer M, Skerra A. Engineering of binding functions into proteins. Curr Opin Biotechnol 2019; 60:230-241. [DOI: 10.1016/j.copbio.2019.05.007] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2018] [Accepted: 05/07/2019] [Indexed: 12/13/2022]
|
48
|
Adamson H, Ajayi MO, Campbell E, Brachi E, Tiede C, Tang AA, Adams TL, Ford R, Davidson A, Johnson M, McPherson MJ, Tomlinson DC, Jeuken LJC. Affimer-Enzyme-Inhibitor Switch Sensor for Rapid Wash-free Assays of Multimeric Proteins. ACS Sens 2019; 4:3014-3022. [PMID: 31578863 DOI: 10.1021/acssensors.9b01574] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Robust technology is required to underpin rapid point-of-care and in-field diagnostics to improve timely decision making across broad sectors. An attractive strategy combines target recognition and signal generating elements into an "active" enzyme-switch that directly transduces target-binding into a signal. However, approaches that are broadly applicable to diverse targets remain elusive. Here, an enzyme-inhibitor switch sensor was developed by insertion of non-immunoglobulin Affimer binding proteins, between TEM1-β-lactamase and its inhibitor protein, such that target binding disrupts the enzyme-inhibitor complex. Design principles for a successful switch architecture are illustrated by the rapid (min), simple (wash-free), and sensitive (pM) quantification of multimeric target analytes in biological samples (serum, plasma, leaf extracts), across three application areas. A therapeutic antibody (Herceptin), protein biomarker (human C-reactive protein), and plant virus (cow pea mosaic virus) were targeted, demonstrating assays for therapeutic drug monitoring, health diagnostics, and plant pathogen detection, respectively. Batch-to-batch reproducibility, shelf-life stability, and consistency with validated enzyme-linked immunosorbent assay analysis confirm that the principle of an Affimer-enzyme-inhibitor switch provides a platform for point-of-care and in-field diagnostics.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Robert Ford
- Avacta Life Sciences Limited, Unit 20, Ash Way, Thorp Arch Estate, Wetherby LS23 7FA, U.K
| | - Alex Davidson
- Avacta Life Sciences Limited, Unit 20, Ash Way, Thorp Arch Estate, Wetherby LS23 7FA, U.K
| | - Matt Johnson
- Avacta Life Sciences Limited, Unit 20, Ash Way, Thorp Arch Estate, Wetherby LS23 7FA, U.K
| | | | | | | |
Collapse
|
49
|
Dietz MS, Heilemann M. Optical super-resolution microscopy unravels the molecular composition of functional protein complexes. NANOSCALE 2019; 11:17981-17991. [PMID: 31573593 DOI: 10.1039/c9nr06364a] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/26/2023]
Abstract
Optical super-resolution microscopy has revolutionized our understanding of cell biology. Next to visualizing cellular structures with near-molecular spatial resolution, an additional benefit is the molecular characterization of biomolecular complexes directly in an intact cell. Single-molecule localization microscopy, as one technology out of the toolbox of super-resolution methods, generates images by detecting the position of single fluorophore labels and is particularly suited for molecular quantification. We review imaging and analysis methods employing single-molecule localization microscopy and extract molecule numbers.
Collapse
Affiliation(s)
- Marina S Dietz
- Single Molecule Biophysics, Institute of Physical and Theoretical Chemistry, Goethe University Frankfurt, 60438 Frankfurt, Germany.
| | | |
Collapse
|
50
|
Chen C, Zong S, Liu Y, Wang Z, Zhang Y, Chen B, Cui Y. Profiling of Exosomal Biomarkers for Accurate Cancer Identification: Combining DNA-PAINT with Machine- Learning-Based Classification. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2019; 15:e1901014. [PMID: 31478613 DOI: 10.1002/smll.201901014] [Citation(s) in RCA: 73] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/24/2019] [Revised: 08/18/2019] [Indexed: 05/08/2023]
Abstract
Exosomes are endosome-derived vesicles enriched in body fluids such as urine, blood, and saliva. So far, they have been recognized as potential biomarkers for cancer diagnostics. However, the present single-variate analysis of exosomes has greatly limited the accuracy and specificity of diagnoses. Besides, most diagnostic approaches focus on bulk analysis using lots of exosomes and tend to be less accurate because they are vulnerable to impure extraction and concentration differences of exosomes. To address these challenges, a quantitative analysis platform is developed to implement a sequential quantification analysis of multiple exosomal surface biomarkers at the single-exosome level, which utilizes DNA-PAINT and a machine learning algorithm to automatically analyze the results. As a proof of concept, the profiling of four exosomal surface biomarkers (HER2, GPC-1, EpCAM, EGFR) is developed to identify exosomes from cancer-derived blood samples. Then, this technique is further applied to detect pancreatic cancer and breast cancer from unknown samples with 100% accuracy.
Collapse
Affiliation(s)
- Chen Chen
- Advanced Photonics Center, Southeast University, Nanjing, 210096, China
| | - Shenfei Zong
- Advanced Photonics Center, Southeast University, Nanjing, 210096, China
| | - Yun Liu
- Advanced Photonics Center, Southeast University, Nanjing, 210096, China
| | - Zhuyuan Wang
- Advanced Photonics Center, Southeast University, Nanjing, 210096, China
| | - Yizhi Zhang
- Advanced Photonics Center, Southeast University, Nanjing, 210096, China
| | - Baoan Chen
- Department of Hematology and Oncology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, 210009, China
| | - Yiping Cui
- Advanced Photonics Center, Southeast University, Nanjing, 210096, China
| |
Collapse
|