Site-Specifically-Labeled Antibodies for Super-Resolution Microscopy Reveal In Situ Linkage Errors
- Susanna M. FrühSusanna M. FrühHahn-Schickard, Georges-Koehler-Allee 103, 79110 Freiburg, GermanyLaboratory for MEMS Applications, IMTEK, Department of Microsystems Engineering, University of Freiburg, 79110 Freiburg, GermanyMore by Susanna M. Früh
- ,
- Ulf MattiUlf MattiCell Biology and Biophysics Unit, European Molecular Biology Laboratory (EMBL), 69117 Heidelberg, GermanyMore by Ulf Matti
- ,
- Philipp R. SpycherPhilipp R. SpycherCenter for Radiopharmaceutical Sciences, Paul Scherrer Institute, 5232 Villigen, SwitzerlandMore by Philipp R. Spycher
- ,
- Marina RubiniMarina RubiniSchool of Chemistry, University College Dublin, Belfield, Dublin 4, IrelandMore by Marina Rubini
- ,
- Sebastian LickertSebastian LickertDepartment of Health Sciences and Technology, ETH Zurich, 8093 Zurich, SwitzerlandMore by Sebastian Lickert
- ,
- Thomas SchlichthaerleThomas SchlichthaerleFaculty of Physics and Center for Nanoscience, Ludwig Maximilian University, 80539 Munich, GermanyMax Planck Institute of Biochemistry, 82152 Martinsried, GermanyMore by Thomas Schlichthaerle
- ,
- Ralf JungmannRalf JungmannFaculty of Physics and Center for Nanoscience, Ludwig Maximilian University, 80539 Munich, GermanyMax Planck Institute of Biochemistry, 82152 Martinsried, GermanyMore by Ralf Jungmann
- ,
- Viola VogelViola VogelDepartment of Health Sciences and Technology, ETH Zurich, 8093 Zurich, SwitzerlandMore by Viola Vogel
- ,
- Jonas RiesJonas RiesCell Biology and Biophysics Unit, European Molecular Biology Laboratory (EMBL), 69117 Heidelberg, GermanyMore by Jonas Ries
- , and
- Ingmar Schoen*Ingmar Schoen*Email: [email protected]School of Pharmacy and Biomolecular Sciences, Irish Centre for Vascular Biology, Royal College of Surgeons in Ireland (RCSI), Dublin 2, IrelandMore by Ingmar Schoen
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.
This publication is licensed under
License Summary*
You are free to share (copy and redistribute) this article in any medium or format and to adapt (remix, transform, and build upon) the material for any purpose, even commercially within the parameters below:
Creative Commons (CC): This is a Creative Commons license.
Attribution (BY): Credit must be given to the creator.
*Disclaimer
This summary highlights only some of the key features and terms of the actual license. It is not a license and has no legal value. Carefully review the actual license before using these materials.
License Summary*
You are free to share (copy and redistribute) this article in any medium or format and to adapt (remix, transform, and build upon) the material for any purpose, even commercially within the parameters below:
Creative Commons (CC): This is a Creative Commons license.
Attribution (BY): Credit must be given to the creator.
*Disclaimer
This summary highlights only some of the key features and terms of the actual license. It is not a license and has no legal value. Carefully review the actual license before using these materials.
License Summary*
You are free to share (copy and redistribute) this article in any medium or format and to adapt (remix, transform, and build upon) the material for any purpose, even commercially within the parameters below:
Creative Commons (CC): This is a Creative Commons license.
Attribution (BY): Credit must be given to the creator.
*Disclaimer
This summary highlights only some of the key features and terms of the actual license. It is not a license and has no legal value. Carefully review the actual license before using these materials.
Results/Discussion
Site-Specific Antibody–Reporter Conjugation Strategy
Scheme 1

Modification of Different IgG Subtypes from Different Species
Figure 1

Figure 1. Sequence alignment and evaluation of potential modification sites for different IgG subclasses from different host species. Sequences were sourced from the UniProt database and aligned with respect to the N-linked glycosylation sites (gray background). The glutamine at position −2 (black background) is preserved across the majority of IgG subtypes and species for which sequence information was available. *Potential conflict with additional glutamine marked in red; Fl = Florida.
Microtubule Labeling Shell Dimensions in STORM
Figure 2

Figure 2. 3D SMLM imaging of microtubule networks and determination of labeling shell dimensions for indirect and direct immunolabeling. (a–d) 3D STORM using Alexa Fluor 647 labeled antibodies in fixed U2OS cells. (a) Schematic (not to scale). (b) Indirect immunolabeling using a randomly labeled secondary donkey-anti-mouse (NHS 2°). (c) Indirect immunolabeling using a site-specifically-labeled secondary donkey-anti-mouse (2°). (d) Direct immunostaining using a site-specifically-labeled primary mouse-anti-α-tubulin (1°). (e–h) 3D DNA-PAINT using DNA-labeled antibodies in U2OS cells (f) and human platelets (g, h). (e) Schematic (not to scale). (f, g) Indirect immunolabeling using a site-specifically-labeled donkey-anti-mouse secondary (2°). (h) Direct labeling using a site-specifically-labeled primary mouse-anti-α-tubulin (1°). For all conditions, representative 3D SMLM images are shown (left). Labeling shell dimensions around microtubules were determined from averaged experimental yz cross-sections and fitted label distributions (right). The fitted label distribution was a Gaussian ring kernel (r: radius; w: full width at half-maximum) convolved with the localization precisions in y and z, respectively. (i) Comparison of center positions and widths of labeling shell dimensions for the different labeling strategies in b–d and f–h.
cells | antibody labeling | shell radius r (nm)b | thickness w (nm)b | minimum linkage errorc (nm) | maximum linkage errord (nm) |
---|---|---|---|---|---|
U2OS | αTub + αMs-647 NHS 2° | 22.5 ± 0.18 | 18.1 ± 0.27 | 1.0 ± 0.32 | 19.1 ± 0.32 |
U2OS | αTub + αMs-647 2° | 22.5 ± 0.22 | 16.9 ± 0.34 | 1.6 ± 0.41 | 18.5 ± 0.41 |
U2OS | αTub-647 1° | 20.2 ± 0.24 | 10.0 ± 0.48 | 2.7 ± 0.54 | 12.7 ± 0.54 |
U2OS | αTub + αMs-DNA 2° | 31.8 ± 0.14 | 21.6 ± 0.19 | 8.5 ± 0.24 | 30.1 ± 0.24 |
human platelets | αTub + αMs-DNA 2° | 29.2 ± 0.16 | 21.4 ± 0.21 | 6.0 ± 0.26 | 27.4 ± 0.26 |
human platelets | αTub-DNA 1° | 23.5 ± 0.20 | 16.7 ± 0.28 | 2.7 ± 0.34 | 19.4 ± 0.34 |
Microtubule Labeling Shell Dimensions in DNA-PAINT
Linkage Errors in Monte Carlo Simulations of Immunolabeled Microtubules
Figure 3

Figure 3. Monte Carlo simulations of antibody conformations at microtubules. (a) Location of the epitope of the anti-α-tubulin antibody in our geometric model (right) based on the cryo-EM structure of a microtubule (left; PDB 5SYF, ref (27); molecular surface rendered using Mol*, ref (33)). (b) Each IgG molecule was modeled by two segments corresponding to Fab and Fc fragments connected by a flexible hinge region. Right: Parametrization of Fab, Fc, and linker segments of the labeled primary antibody. Left: Dimensions were based on the crystallographic structure of IgG2a (PDB 1IGT, ref (30), visualization by Mol*). While the location of the modification is precisely known (Glu), the binding sites of polyclonal secondary antibodies are assumed to be evenly distributed over the Fc region. For a schematic of indirect immunolabeling, see Supplementary Figure S5c). Simulated reporter distribution for primary antibodies. Left: yz cross-section. Right: Radial distribution. (d) Simulated reporter distribution for primary plus secondary antibody complexes. Panels as in (c). (e) Comparison of reporter distributions for primary antibodies between simulations (left; convolved with the localization and experimental imprecision) and experiments (middle: pooled cross-sections as in Figure 2). Right: Normalized residuals of the difference between experiment and model. Top: STORM. Bottom: DNA-PAINT. (f) Comparison of reporter distributions for secondary antibodies between simulations and experiments. Panels as in (d).
Benchmarking of SMLM Results against Simulations
Conclusions
Methods/Experimental
Antibody Labeling
DOL Characterization
Sample Preparation for SMLM
Quantification of Apparent Microtubule Dimensions
Monte Carlo Simulations
Statistical Analysis

Supporting Information
The Supporting Information is available free of charge at https://pubs.acs.org/doi/10.1021/acsnano.1c03677.
Experimental methods on mass spectrometry, SMLM setups, SMLM imaging, the determination of residual experimental errors, and circular dichroism spectroscopy measurements; Supplementary Figures S1 to S8; Supplementary Tables S1 to S6 (PDF)
Terms & Conditions
Most electronic Supporting Information files are available without a subscription to ACS Web Editions. Such files may be downloaded by article for research use (if there is a public use license linked to the relevant article, that license may permit other uses). Permission may be obtained from ACS for other uses through requests via the RightsLink permission system: http://pubs.acs.org/page/copyright/permissions.html.
Acknowledgments
We thank the peer reviewers of this manuscript for their constructive critique. We would like to acknowledge R. Schibli for providing therapeutic antibodies. This work was supported by a fellowship of the Holcim Science Foundation (I.S.), the European Research Council (ERC CoG-724489, J.R.; ERC StG-680241, R.J.), the DFG (S.M.F., 397660978; R.J., JU 2957/1-1 and SFB 1032/A11), the AiF (S.M.F., 20436 N), the QBM graduate school (T.S.), the Center for Nano-Science (R.J.), the European Molecular Biology Laboratory (U.M. and J.R.), ETH Zurich (V.V.), the Max Planck Society/Foundation (J.R.), and RCSI (I.S.).
References
This article references 38 other publications.
- 1Huang, F.; Sirinakis, G.; Allgeyer, E. S.; Schroeder, L. K.; Duim, W. C.; Kromann, E. B.; Phan, T.; Rivera-Molina, F. E.; Myers, J. R.; Irnov, I.; Lessard, M.; Zhang, Y.; Handel, M. A.; Jacobs-Wagner, C.; Lusk, C. P.; Rothman, J. E.; Toomre, D.; Booth, M. J.; Bewersdorf, J. Ultra-High Resolution 3D Imaging of Whole Cells. Cell 2016, 166 (4), 1028– 1040, DOI: 10.1016/j.cell.2016.06.016[Crossref], [PubMed], [CAS], Google Scholar1https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC28XhtFCiu7rN&md5=45816ee5300c4b686789eba93ed8b28fUltra-High Resolution 3D Imaging of Whole CellsHuang, Fang; Sirinakis, George; Allgeyer, Edward S.; Schroeder, Lena K.; Duim, Whitney C.; Kromann, Emil B.; Phan, Thomy; Rivera-Molina, Felix E.; Myers, Jordan R.; Irnov, Irnov; Lessard, Mark; Zhang, Yongdeng; Handel, Mary Ann; Jacobs-Wagner, Christine; Lusk, C. Patrick; Rothman, James E.; Toomre, Derek; Booth, Martin J.; Bewersdorf, JoergCell (Cambridge, MA, United States) (2016), 166 (4), 1028-1040CODEN: CELLB5; ISSN:0092-8674. (Cell Press)Fluorescence nanoscopy, or super-resoln. microscopy, has become an important tool in cell biol. research. However, because of its usually inferior resoln. in the depth direction (50-80 nm) and rapidly deteriorating resoln. in thick samples, its practical biol. application has been effectively limited to two dimensions and thin samples. Here, the authors present the development of whole-cell 4Pi single-mol. switching nanoscopy (W-4PiSMSN), an optical nanoscope that allows imaging of three-dimensional (3D) structures at 10- to 20-nm resoln. throughout entire mammalian cells. The authors demonstrate the wide applicability of W-4PiSMSN across diverse research fields by imaging complex mol. architectures ranging from bacteriophages to nuclear pores, cilia, and synaptonemal complexes in large 3D cellular vols.
- 2Jungmann, R.; Avendaño, M. S.; Dai, M.; Woehrstein, J. B.; Agasti, S. S.; Feiger, Z.; Rodal, A.; Yin, P. Quantitative Super-Resolution Imaging with qPAINT. Nat. Methods 2016, 13 (5), 439– 442, DOI: 10.1038/nmeth.3804[Crossref], [PubMed], [CAS], Google Scholar2https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC28XkvV2jtLk%253D&md5=1ce6b7fdb5e94a4309c88942f751a172Quantitative super-resolution imaging with qPAINTJungmann, Ralf; Avendano, Maier S.; Dai, Mingjie; Woehrstein, Johannes B.; Agasti, Sarit S.; Feiger, Zachary; Rodal, Avital; Yin, PengNature Methods (2016), 13 (5), 439-442CODEN: NMAEA3; ISSN:1548-7091. (Nature Publishing Group)Counting mols. in complexes is challenging, even with super-resoln. microscopy. Here, we use the programmable and specific binding of dye-labeled DNA probes to count integer nos. of targets. This method, called quant. points accumulation in nanoscale topog. (qPAINT), works independently of dye photophysics for robust counting with high precision and accuracy over a wide dynamic range. qPAINT was benchmarked on DNA nanostructures and demonstrated for cellular applications by quantifying proteins in situ and the no. of single-mol. FISH probes bound to an mRNA target.
- 3Koch, B.; Nijmeijer, B.; Kueblbeck, M.; Cai, Y.; Walther, N.; Ellenberg, J. Generation and Validation of Homozygous Fluorescent Knock-In Cells Using CRISPR–Cas9 Genome Editing. Nat. Protoc. 2018, 13 (6), 1465– 1487, DOI: 10.1038/nprot.2018.042[Crossref], [PubMed], [CAS], Google Scholar3https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1cXhtVamurbP&md5=9a24e4b52eb37fe9f012717177b45f6bGeneration and validation of homozygous fluorescent knock-in cells using CRISPR-Cas9 genome editingKoch, Birgit; Nijmeijer, Bianca; Kueblbeck, Moritz; Cai, Yin; Walther, Nike; Ellenberg, JanNature Protocols (2018), 13 (6), 1465-1487CODEN: NPARDW; ISSN:1750-2799. (Nature Research)Gene tagging with fluorescent proteins is essential for investigations of the dynamic properties of cellular proteins. CRISPR-Cas9 technol. is a powerful tool for inserting fluorescent markers into all alleles of the gene of interest (GOI) and allows functionality and physiol. expression of the fusion protein. It is essential to evaluate such genome-edited cell lines carefully in order to preclude off-target effects caused by (i) incorrect insertion of the fluorescent protein, (ii) perturbation of the fusion protein by the fluorescent proteins or (iii) nonspecific genomic DNA damage by CRISPR-Cas9. In this protocol, we provide a step-by-step description of our systematic pipeline to generate and validate homozygous fluorescent knock-in cell lines. We have used the paired Cas9D10A nickase approach to efficiently insert tags into specific genomic loci via homol.-directed repair (HDR) with minimal off-target effects. It is time-consuming and costly to perform whole-genome sequencing of each cell clone to check for spontaneous genetic variations occurring in mammalian cell lines. Therefore, we have developed an efficient validation pipeline of the generated cell lines consisting of junction PCR, Southern blotting anal., Sanger sequencing, microscopy, western blotting anal. and live-cell imaging for cell-cycle dynamics. This protocol takes between 6 and 9 wk. With this protocol, up to 70% of the targeted genes can be tagged homozygously with fluorescent proteins, thus resulting in physiol. levels and phenotypically functional expression of the fusion proteins.
- 4Ghodke, H.; Caldas, V. E. A.; Punter, C. M.; van Oijen, A. M.; Robinson, A. Single-Molecule Specific Mislocalization of Red Fluorescent Proteins in Live Escherichia Coli. Biophys. J. 2016, 111 (1), 25– 27, DOI: 10.1016/j.bpj.2016.05.047[Crossref], [PubMed], [CAS], Google Scholar4https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC28XhtVWns7fK&md5=e4760ed7f31002c37e995eb59f14c1e8Single-Molecule Specific Mislocalization of Red Fluorescent Proteins in Live Escherichia coliGhodke, Harshad; Caldas, Victor E. A.; Punter, Christiaan M.; van Oijen, Antoine M.; Robinson, AndrewBiophysical Journal (2016), 111 (1), 25-27CODEN: BIOJAU; ISSN:0006-3495. (Cell Press)Tagging of individual proteins with genetically encoded fluorescent proteins (FPs) has been used extensively to study localization and interactions in live cells. Recent developments in single-mol. localization microscopy have enabled the dynamic visualization of individual tagged proteins inside living cells. However, tagging proteins with FPs is not without problems: formation of insol. aggregates and inhibition of native functions of the protein are well-known issues. Previously reported artifacts manifest themselves at all expression levels of the FP-tagged proteins, making the design of control expts. relatively straightforward. Here, we describe a previously uncharacterized mislocalization artifact of Entacmaea quadricolor red fluorescent protein variants that is detectable at the single-mol. level in live Escherichia coli cells.
- 5Sahl, S. J.; Hell, S. W.; Jakobs, S. Fluorescence Nanoscopy in Cell Biology. Nat. Rev. Mol. Cell Biol. 2017, 18 (11), 685– 701, DOI: 10.1038/nrm.2017.71[Crossref], [PubMed], [CAS], Google Scholar5https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2sXhsVChs7rO&md5=c72d990fae52915e9a9623e2eb065cceFluorescence nanoscopy in cell biologySahl, Steffen J.; Hell, Stefan W.; Jakobs, StefanNature Reviews Molecular Cell Biology (2017), 18 (11), 685-701CODEN: NRMCBP; ISSN:1471-0072. (Nature Research)A review. Fluorescence nanoscopy uniquely combines minimally invasive optical access to the internal nanoscale structure and dynamics of cells and tissues with mol. detection specificity. While the basic phys. principles of 'super-resoln.' imaging were discovered in the 1990s, with initial exptl. demonstrations following in 2000, the broad application of super-resoln. imaging to address cell-biol. questions has only more recently emerged. Nanoscopy approaches have begun to facilitate discoveries in cell biol. and to add new knowledge. One current direction for method improvement is the ambition to quant. account for each mol. under investigation and assess true mol. colocalization patterns via multi-color analyses. In pursuing this goal, the labeling of individual mols. to enable their visualization has emerged as a central challenge. Extending nanoscale imaging into (sliced) tissue and whole-animal contexts is a further goal. In this Review we describe the successes to date and discuss current obstacles and possibilities for further development.
- 6Schlichthaerle, T.; Eklund, A. S.; Schueder, F.; Strauss, M. T.; Tiede, C.; Curd, A.; Ries, J.; Peckham, M.; Tomlinson, D. C.; Jungmann, R. Site-Specific Labeling of Affimers for DNA-PAINT Microscopy. Angew. Chem., Int. Ed. 2018, 57 (34), 11060– 11063, DOI: 10.1002/anie.201804020[Crossref], [CAS], Google Scholar6https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1cXhtlWqtL3L&md5=85559fed38b1e5ff592fb2f67975029fSite-Specific Labeling of Affimers for DNA-PAINT MicroscopySchlichthaerle, Thomas; Eklund, Alexandra S.; Schueder, Florian; Strauss, Maximilian T.; Tiede, Christian; Curd, Alistair; Ries, Jonas; Peckham, Michelle; Tomlinson, Darren C.; Jungmann, RalfAngewandte Chemie, International Edition (2018), 57 (34), 11060-11063CODEN: ACIEF5; ISSN:1433-7851. (Wiley-VCH Verlag GmbH & Co. KGaA)Optical super-resoln. techniques allow fluorescence imaging below the classical diffraction limit of light. From a technol. standpoint, recent methods are approaching mol.-scale spatial resoln. However, this remarkable achievement is not easily translated to imaging of cellular components, since current labeling approaches are limited by either large label sizes (antibodies) or the sparse availability of small and efficient binders (nanobodies, aptamers, genetically-encoded tags). In this work, we combined recently developed Affimer reagents with site-specific DNA modification for high-efficiency labeling and imaging using DNA-PAINT. We assayed our approach using an actin Affimer. The small DNA-conjugated affinity binders could provide a soln. for efficient multitarget super-resoln. imaging in the future.
- 7Shaw, A.; Hoffecker, I. T.; Smyrlaki, I.; Rosa, J.; Grevys, A.; Bratlie, D.; Sandlie, I.; Michaelsen, T. E.; Andersen, J. T.; Högberg, B. Binding to Nanopatterned Antigens Is Dominated by the Spatial Tolerance of Antibodies. Nat. Nanotechnol. 2019, 14, 1, DOI: 10.1038/s41565-018-0336-3
- 8Mikhaylova, M.; Cloin, B. M. C.; Finan, K.; van den Berg, R.; Teeuw, J.; Kijanka, M. M.; Sokolowski, M.; Katrukha, E. a; Maidorn, M.; Opazo, F.; Moutel, S.; Vantard, M.; Perez, F.; van Bergen En Henegouwen, P. M. P.; Hoogenraad, C. C.; Ewers, H.; Kapitein, L. C. Resolving Bundled Microtubules Using Anti-Tubulin Nanobodies. Nat. Commun. 2015, 6 (May), 7933, DOI: 10.1038/ncomms8933[Crossref], [PubMed], [CAS], Google Scholar8https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2MXhsVKhtbjJ&md5=ccb0d03ce3408f23623cfa0049540572Resolving bundled microtubules using anti-tubulin nanobodiesMikhaylova, Marina; Cloin, Bas M. C.; Finan, Kieran; van den Berg, Robert; Teeuw, Jalmar; Kijanka, Marta M.; Sokolowski, Mikolaj; Katrukha, Eugene A.; Maidorn, Manuel; Opazo, Felipe; Moutel, Sandrine; Vantard, Marylin; Perez, Frank; van Bergen en Henegouwen, Paul M. P.; Hoogenraad, Casper C.; Ewers, Helge; Kapitein, Lukas C.Nature Communications (2015), 6 (), 7933CODEN: NCAOBW; ISSN:2041-1723. (Nature Publishing Group)Microtubules are hollow biopolymers of 25-nm diam. and are key constituents of the cytoskeleton. In neurons, microtubules are organized differently between axons and dendrites, but their precise organization in different compartments is not completely understood. Super-resoln. microscopy techniques can detect specific structures at an increased resoln., but the narrow spacing between neuronal microtubules poses challenges because most existing labeling strategies increase the effective microtubule diam. by 20-40 nm and will thereby blend neighboring microtubules into one structure. Here we develop single-chain antibody fragments (nanobodies) against tubulin to achieve super-resoln. imaging of microtubules with a decreased apparent diam. To test the resolving power of these novel probes, we generate microtubule bundles with a known spacing of 50-70 nm and successfully resolve individual microtubules. Individual bundled microtubules can also be resolved in different mammalian cells, including hippocampal neurons, allowing novel insights into fundamental mechanisms of microtubule organization in cell- and neurobiol.
- 9Ries, J.; Kaplan, C.; Platonova, E.; Eghlidi, H.; Ewers, H. A Simple, Versatile Method for GFP-Based Super-Resolution Microscopy via Nanobodies. Nat. Methods 2012, 9 (6), 582– 584, DOI: 10.1038/nmeth.1991[Crossref], [PubMed], [CAS], Google Scholar9https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC38Xmt1Gksbs%253D&md5=678f32bb3f032e786fb8ca42d893030aA simple, versatile method for GFP-based super-resolution microscopy via nanobodiesRies, Jonas; Kaplan, Charlotte; Platonova, Evgenia; Eghlidi, Hadi; Ewers, HelgeNature Methods (2012), 9 (6), 582-584CODEN: NMAEA3; ISSN:1548-7091. (Nature Publishing Group)We developed a method to use any GFP-tagged construct in single-mol. super-resoln. microscopy. By targeting GFP with small, high-affinity antibodies coupled to org. dyes, we achieved nanometer spatial resoln. and minimal linkage error when analyzing microtubules, living neurons and yeast cells. We show that in combination with libraries encoding GFP-tagged proteins, virtually any known protein can immediately be used in super-resoln. microscopy and that simplified labeling schemes allow high-throughput super-resoln. imaging.
- 10Opazo, F.; Levy, M.; Byrom, M.; Schäfer, C.; Geisler, C.; Groemer, T. W.; Ellington, A. D.; Rizzoli, S. O. Aptamers as Potential Tools for Super-Resolution Microscopy. Nat. Methods 2012, 9 (10), 938– 939, DOI: 10.1038/nmeth.2179[Crossref], [PubMed], [CAS], Google Scholar10https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC38XhsVWntb%252FE&md5=854e68813111f273803bc0b79db829d4Aptamers as potential tools for super-resolution microscopyOpazo, Felipe; Levy, Matthew; Byrom, Michelle; Schaefer, Christina; Geisler, Claudia; Groemer, Teja W.; Ellington, Andrew D.; Rizzoli, Silvio O.Nature Methods (2012), 9 (10), 938-939CODEN: NMAEA3; ISSN:1548-7091. (Nature Publishing Group)This article shows that aptamers, which are single-stranded DNA or RNA oligonucleotides, are potential probes that would complement nanobodies for super-resoln. imaging. Three aptamers were tested for mols. involved in endosomal trafficking: the transferrin receptor (TfnR; aptamer size ∼15 kDa), the prostate-specific membrane antigen (PSMA; ∼15 kDa) and the epidermal growth factor receptor (EGFR; ∼36 kDa). Epitope recognition with aptamers was higher than with conventional antibodies, and this allowed the aptamers to provide superior super-resoln. images, just as the nanobodies did, when investigated using stimulated emission depletion (STED) microscopy. The high epitope recognition also permitted rapid live super-resoln. imaging.
- 11Agarwal, P.; Bertozzi, C. R. Site-Specific Antibody-Drug Conjugates: The Nexus of Bioorthogonal Chemistry, Protein Engineering, and Drug Development. Bioconjugate Chem. 2015, 26 (2), 176– 192, DOI: 10.1021/bc5004982[ACS Full Text
], [CAS], Google Scholar
11https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2cXitFCrsrfK&md5=c3d822b0e53e5cdb074e5b63b8060ccfSite-Specific Antibody-Drug Conjugates: The Nexus of Bioorthogonal Chemistry, Protein Engineering, and Drug DevelopmentAgarwal, Paresh; Bertozzi, Carolyn R.Bioconjugate Chemistry (2015), 26 (2), 176-192CODEN: BCCHES; ISSN:1043-1802. (American Chemical Society)A review. Antibody-drug conjugates (ADCs) combine the specificity of antibodies with the potency of small mols. to create targeted drugs. Despite the simplicity of this concept, generation of clin. successful ADCs has been very difficult. Over the past several decades, scientists have learned a great deal about the constraints on antibodies, linkers, and drugs as they relate to successful construction of ADCs. Once these components are in hand, most ADCs are prepd. by nonspecific modification of antibody lysine or cysteine residues with drug-linker reagents, which results in heterogeneous product mixts. that cannot be further purified. With advances in the fields of bioorthogonal chem. and protein engineering, there is growing interest in producing ADCs by site-specific conjugation to the antibody, yielding more homogeneous products that have demonstrated benefits over their heterogeneous counterparts in vivo. Here, we chronicle the development of a multitude of site-specific conjugation strategies for assembly of ADCs and provide a comprehensive account of key advances and their roots in the fields of bioorthogonal chem. and protein engineering. - 12Beck, A.; Goetsch, L.; Dumontet, C.; Corvaïa, N. Strategies and Challenges for the Next Generation of Antibody–Drug Conjugates. Nat. Rev. Drug Discovery 2017, 16 (5), 315– 337, DOI: 10.1038/nrd.2016.268[Crossref], [PubMed], [CAS], Google Scholar12https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2sXkt1Onsbc%253D&md5=7358b6080dd1aed1d21e98c078cd9ef1Strategies and challenges for the next generation of antibody-drug conjugatesBeck, Alain; Goetsch, Liliane; Dumontet, Charles; Corvaia, NathalieNature Reviews Drug Discovery (2017), 16 (5), 315-337CODEN: NRDDAG; ISSN:1474-1776. (Nature Publishing Group)A review. Antibody-drug conjugates (ADCs) are one of the fastest growing classes of oncol. therapeutics. After half a century of research, the approvals of brentuximab vedotin (in 2011) and trastuzumab emtansine (in 2013) have paved the way for ongoing clin. trials that are evaluating more than 60 further ADC candidates. The limited success of first-generation ADCs (developed in the early 2000s) informed strategies to bring second-generation ADCs to the market, which have higher levels of cytotoxic drug conjugation, lower levels of naked antibodies and more-stable linkers between the drug and the antibody. Furthermore, lessons learned during the past decade are now being used in the development of third-generation ADCs. In this Review, we discuss strategies to select the best target antigens as well as suitable cytotoxic drugs; the design of optimized linkers; the discovery of bioorthogonal conjugation chemistries; and toxicity issues. The selection and engineering of antibodies for site-specific drug conjugation, which will result in higher homogeneity and increased stability, as well as the quest for new conjugation chemistries and mechanisms of action, are priorities in ADC research.
- 13Li, X.; Fang, T.; Boons, G. J. Preparation of Well-Defined Antibody-Drug Conjugates through Glycan Remodeling and Strain-Promoted Azide-Alkyne Cycloadditions. Angew. Chem., Int. Ed. 2014, 53 (28), 7179– 7182, DOI: 10.1002/anie.201402606[Crossref], [CAS], Google Scholar13https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2cXos1aks7w%253D&md5=d59a17169fb0891a63e88960285c4f55Preparation of well-defined antibody-drug conjugates through glycan remodeling and strain-promoted azide-alkyne cycloadditionsLi, Xiuru; Fang, Tao; Boons, Geert-JanAngewandte Chemie, International Edition (2014), 53 (28), 7179-7182CODEN: ACIEF5; ISSN:1433-7851. (Wiley-VCH Verlag GmbH & Co. KGaA)Antibody-drug conjugates hold considerable promise as anticancer agents, however, producing them remains a challenge and there is a need for mild, broadly applicable, site-specific conjugation methods that yield homogenous products. It was envisaged that enzymic remodeling of the oligosaccharides of an antibody would enable the introduction of reactive groups that can be exploited for the site-specific attachment of cytotoxic drugs. This is based on the observation that glycosyltransferases often tolerate chem. modifications in their sugar nucleotide substrates, thus allowing the installation of reactive functionalities. An azide was incorporated because this functional group is virtually absent in biol. systems and can be reacted by strain-promoted alkyne-azide cycloaddn. This method, which does not require genetic engineering, was used to produce an anti-CD22 antibody modified with doxorubicin to selectively target and kill lymphoma cells.
- 14Van Geel, R.; Wijdeven, M. A.; Heesbeen, R.; Verkade, J. M. M.; Wasiel, A. A.; Van Berkel, S. S.; Van Delft, F. L. Chemoenzymatic Conjugation of Toxic Payloads to the Globally Conserved N-Glycan of Native mAbs Provides Homogeneous and Highly Efficacious Antibody-Drug Conjugates. Bioconjugate Chem. 2015, 26 (11), 2233– 2242, DOI: 10.1021/acs.bioconjchem.5b00224[ACS Full Text
], [CAS], Google Scholar
14https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2MXhtVShtLjL&md5=3473e419ded9df111414a8788bcdce43Chemoenzymatic conjugation of toxic payloads to the globally conserved N-glycan of native mAbs provides homogeneous and highly efficacious antibody-drug conjugatesvan Geel, Remon; Wijdeven, Marloes A.; Heesbeen, Ryan; Verkade, Jorge M. M.; Wasiel, Anna A.; van Berkel, Sander S.; van Delft, Floris L.Bioconjugate Chemistry (2015), 26 (11), 2233-2242CODEN: BCCHES; ISSN:1043-1802. (American Chemical Society)A robust, generally applicable, nongenetic technol. is presented to convert monoclonal antibodies into stable and homogeneous ADCs. Starting from a native (nonengineered) mAb, a chemoenzymic protocol allows for the highly controlled attachment of any given payload to the N-glycan residing at asparagine-297, based on a two-stage process: first, enzymic remodeling (trimming and tagging with azide), followed by ligation of the payload based on copper-free click chem. The technol., termed GlycoConnect, is applicable to any IgG isotype irresp. of glycosylation profile. Application to trastuzumab and maytansine, both components of the marketed ADC Kadcyla, demonstrate a favorable in vitro and in vivo efficacy for GlycoConnect ADC. Moreover, the superiority of the native glycan as attachment site was demonstrated by in vivo comparison to a range of trastuzumab-based glycosylation mutants. A side-by-side comparison of the copper-free click probes bicyclononyne (BCN) and a dibenzoannulated cyclooctyne (DBCO) showed a surprising difference in conjugation efficiency in favor of BCN, which could be even further enhanced by introduction of electron-withdrawing fluoride substitutions onto the azide. The resulting mAb-conjugates were in all cases found to be highly stable, which in combination with the demonstrated efficacy warrants ADCs with a superior therapeutic index. - 15Rosen, C. B.; Kodal, A. L. B.; Nielsen, J. S.; Schaffert, D. H.; Scavenius, C.; Okholm, A. H.; Voigt, N. V.; Enghild, J. J.; Kjems, J.; Tørring, T.; Gothelf, K. V. Template-Directed Covalent Conjugation of DNA to Native Antibodies, Transferrin and Other Metal-Binding Proteins. Nat. Chem. 2014, 6 (9), 804– 809, DOI: 10.1038/nchem.2003[Crossref], [PubMed], [CAS], Google Scholar15https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2cXhtFygs7bL&md5=8a402d92b83e7d605e841cba7fb72b27Template-directed covalent conjugation of DNA to native antibodies, transferrin and other metal-binding proteinsRosen, Christian B.; Kodal, Anne L. B.; Nielsen, Jesper S.; Schaffert, David H.; Scavenius, Carsten; Okholm, Anders H.; Voigt, Niels V.; Enghild, Jan J.; Kjems, Joergen; Toerring, Thomas; Gothelf, Kurt V.Nature Chemistry (2014), 6 (9), 804-809CODEN: NCAHBB; ISSN:1755-4330. (Nature Publishing Group)DNA-protein conjugates are important in bioanal. chem., mol. diagnostics and bionanotechnol., as the DNA provides a unique handle to identify, functionalize or otherwise manipulate proteins. To maintain protein activity, conjugation of a single DNA handle to a specific location on the protein is often needed. However, prepg. such high-quality site-specific conjugates often requires genetically engineered proteins, which is a laborious and tech. challenging approach. Here we demonstrate a simpler method to create site-selective DNA-protein conjugates. Using a guiding DNA strand modified with a metal-binding functionality, we directed a second DNA strand to the vicinity of a metal-binding site of His6-tagged or wild-type metal-binding proteins, such as serotransferrin, where it subsequently reacted with lysine residues at that site. This method, DNA-templated protein conjugation, facilitates the prodn. of site-selective protein conjugates, and also conjugation to IgG1 antibodies via a histidine cluster in the const. domain.
- 16Kline, T.; Steiner, A. R.; Penta, K.; Sato, A. K.; Hallam, T. J.; Yin, G. Methods to Make Homogenous Antibody Drug Conjugates. Pharm. Res. 2015, 32 (11), 3480– 3493, DOI: 10.1007/s11095-014-1596-8[Crossref], [PubMed], [CAS], Google Scholar16https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2cXitFCru7vO&md5=3e933915d0c26eac721d100dbcf28f08Methods to Make Homogenous Antibody Drug ConjugatesKline, Toni; Steiner, Alexander R.; Penta, Kalyani; Sato, Aaron K.; Hallam, Trevor J.; Yin, GangPharmaceutical Research (2015), 32 (11), 3480-3493CODEN: PHREEB; ISSN:0724-8741. (Springer)Antibody drug conjugates (ADCs) have progressed from hypothesis to approved therapeutics in less than 30 years, and the technologies available to modify both the antibodies and the cytotoxic drugs are expanding rapidly. For reasons well reviewed previously, the field is trending strongly toward homogeneous, defined antibody conjugation. In this review we present the antibody and small mol. chemistries that are currently used and being explored to develop specific, homogeneous ADCs.
- 17Jeger, S.; Zimmermann, K.; Blanc, A.; Grünberg, J.; Honer, M.; Hunziker, P.; Struthers, H.; Schibli, R. Site-Specific and Stoichiometric Modification of Antibodies by Bacterial Transglutaminase. Angew. Chem., Int. Ed. 2010, 49 (51), 9995– 9997, DOI: 10.1002/anie.201004243[Crossref], [CAS], Google Scholar17https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3cXhsFGhsLnI&md5=6c2bad043af729dcfa3a7b27a122b98fSite-Specific and Stoichiometric Modification of Antibodies by Bacterial TransglutaminaseJeger, Simone; Zimmermann, Kurt; Blanc, Alain; Gruenberg, Juergen; Honer, Michael; Hunziker, Peter; Struthers, Harriet; Schibli, RogerAngewandte Chemie, International Edition (2010), 49 (51), 9995-9997, S9995/1-S9995/46CODEN: ACIEF5; ISSN:1433-7851. (Wiley-VCH Verlag GmbH & Co. KGaA)We have shown that enzymic modification of mAbs using bacterial transglutaminase (BTG) is site-specific and versatile (with the potential to be readily scaled up), and leads to homogeneous immunoconjugates with defined stoichiometries. The in vivo characteristics of such immunoconjugates are superior to those prepd. using chem. coupling methods. Since position 295 is located in the const. Fc region, the enzymic conjugation approach is applicable not only to other human IgG1s, but also to mAbs belonging to subtypes IgG2, IgG3, and IgG4, all of which conserve the Q295 residue. Thus, the method is broadly applicable and permits the systematic assessment and improvement of immunoconjugates.
- 18Dennler, P.; Chiotellis, A.; Fischer, E.; Brégeon, D.; Belmant, C.; Gauthier, L.; Lhospice, F.; Romagne, F.; Schibli, R. Transglutaminase-Based Chemo-Enzymatic Conjugation Approach Yields Homogeneous Antibody–Drug Conjugates. Bioconjugate Chem. 2014, 25 (3), 569– 578, DOI: 10.1021/bc400574z[ACS Full Text
], [CAS], Google Scholar
18https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2cXhs1GntLs%253D&md5=7d70123f2e5cea5bbda003eb0c0d55e0Transglutaminase-Based Chemo-Enzymatic Conjugation Approach Yields Homogeneous Antibody-Drug ConjugatesDennler, Patrick; Chiotellis, Aristeidis; Fischer, Eliane; Bregeon, Delphine; Belmant, Christian; Gauthier, Laurent; Lhospice, Florence; Romagne, Francois; Schibli, RogerBioconjugate Chemistry (2014), 25 (3), 569-578CODEN: BCCHES; ISSN:1043-1802. (American Chemical Society)Most chem. techniques used to produce antibody-drug conjugates (ADCs) result in a heterogeneous mixt. of species with variable drug-to-antibody ratios (DAR) which will potentially display different pharmacokinetics, stability, and safety profiles. Here we investigated two strategies to obtain homogeneous ADCs based on site-specific modification of deglycosylated antibodies by microbial transglutaminase (MTGase), which forms isopeptidic bonds between Gln and Lys residues. We have previously shown that MTGase solely recognizes Gln295 within the heavy chain of IgGs as a substrate and can therefore be exploited to generate ADCs with an exact DAR of 2. The first strategy included the direct, one-step attachment of the antimitotic toxin monomethyl auristatin E (MMAE) to the antibody via different spacer entities with a primary amine functionality that is recognized as a substrate by MTGase. The second strategy was a chemo-enzymic, two-step approach whereby a reactive spacer entity comprising a bio-orthogonal thiol or azide function was attached to the antibody by MTGase and subsequently reacted with a suitable MMAE-deriv. To this aim, we investigated two different chem. approaches, namely, thiol-maleimide and strain-promoted azide-alkyne cycloaddn. (SPAAC). Direct enzymic attachment of MMAE-spacer derivs. at an 80 M excess of drug yielded heterogeneous ADCs with a DAR of between 1.0 to 1.6. In contrast to this, the chemo-enzymic approach only required a 2.5 M excess of toxin to yield homogeneous ADCs with a DAR of 2.0 in the case of SPAAC and 1.8 for the thiol-maleimide approach. As a proof-of-concept, trastuzumab (Herceptin) was armed with the MMAE via the chemo-enzymic approach using SPAAC and tested in vitro. Trastuzumab-MMAE efficiently killed BT-474 and SK-BR-3 cells with an IC50 of 89.0 pM and 21.7 pM, resp. Thus, the chemo-enzymic approach using MTGase is an elegant strategy to form ADCs with a defined DAR of 2. Furthermore, the approach is directly applicable to a broad variety of antibodies as it does not require prior genetic modifications of the antibody sequence. - 19Agard, N. J.; Prescher, J. A.; Bertozzi, C. R. A Strain-Promoted [3 + 2] Azide-Alkyne Cycloaddition for Covalent Modification of Biomolecules in Living Systems. J. Am. Chem. Soc. 2004, 126 (46), 15046– 15047, DOI: 10.1021/ja044996f[ACS Full Text
], [CAS], Google Scholar
19https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD2cXpt1Sks7s%253D&md5=37af3dbaa89ae4cffaba2dee30e50ec0A strain-promoted [3+2] azide-alkyne cycloaddition for covalent modification of biomolecules in living systemsAgard, Nicholas J.; Prescher, Jennifer A.; Bertozzi, Carolyn R.Journal of the American Chemical Society (2004), 126 (46), 15046-15047CODEN: JACSAT; ISSN:0002-7863. (American Chemical Society)Selective chem. reactions that are orthogonal to the diverse functionality of biol. systems have become important tools in the field of chem. biol. Two notable examples are the Staudinger ligation of azides and phosphines and the Cu(I)-catalyzed [3+2] cycloaddn. of azides and alkynes ("click chem."). The Staudinger ligation has sufficient biocompatibility for performance in living animals but suffers from phosphine oxidn. and synthetic challenges. Click chem. obviates the requirement of phosphines, but the Cu(I) catalyst is toxic to cells, thereby precluding in vivo applications. Here we present a strain-promoted [3+2] cycloaddn. between cyclooctynes and azides that proceeds under physiol. conditions without the need for a catalyst. The utility of the reaction was demonstrated by selective modification of biomols. in vitro and on living cells, with no apparent toxicity. - 20Helmerich, D. A.; Beliu, G.; Sauer, M. Multiple-Labeled Antibodies Behave Like Single Emitters in Photoswitching Buffer. ACS Nano 2020, 14 (10), 12629– 12641, DOI: 10.1021/acsnano.0c06099[ACS Full Text
], [CAS], Google Scholar
20https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BB38fmtl2ltw%253D%253D&md5=8510150e6bd05409d8c6b812609171f7Multiple-Labeled Antibodies Behave Like Single Emitters in Photoswitching BufferHelmerich Dominic A; Beliu Gerti; Sauer MarkusACS nano (2020), 14 (10), 12629-12641 ISSN:.The degree of labeling (DOL) of antibodies has so far been optimized for high brightness and specific and efficient binding. The influence of the DOL on the blinking performance of antibodies used in direct stochastic optical reconstruction microscopy (dSTORM) has so far attained limited attention. Here, we investigated the spectroscopic characteristics of IgG antibodies labeled at DOLs of 1.1-8.3 with Alexa Fluor 647 (Al647) at the ensemble and single-molecule level. Multiple-Al647-labeled antibodies showed weak and strong quenching interactions in aqueous buffer but could all be used for dSTORM imaging with spatial resolutions of ∼20 nm independent of the DOL. Single-molecule fluorescence trajectories and photon antibunching experiments revealed that individual multiple-Al647-labeled antibodies show complex photophysics in aqueous buffer but behave as single emitters in photoswitching buffer independent of the DOL. We developed a model that explains the observed blinking of multiple-labeled antibodies and can be used for the development of improved fluorescent probes for dSTORM experiments. - 21Pleiner, T.; Bates, M.; Görlich, D. A Toolbox of Anti-Mouse and Anti-Rabbit IgG Secondary Nanobodies. J. Cell Biol. 2018, 217 (3), 1143– 1154, DOI: 10.1083/jcb.201709115[Crossref], [PubMed], [CAS], Google Scholar21https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1cXhslWmsbvF&md5=47efb96d0aeb73ae07eaafad355a0277A toolbox of anti-mouse and anti-rabbit IgG secondary nanobodiesPleiner, Tino; Bates, Mark; Goerlich, DirkJournal of Cell Biology (2018), 217 (3), 1143-1159CODEN: JCLBA3; ISSN:1540-8140. (Rockefeller University Press)Polyclonal anti-IgG (anti-IgG) secondary antibodies are essential tools for many mol. biol. techniques and diagnostic tests. Their animal-based prodn. is, however, a major ethical problem. Here, we introduce a sustainable alternative, namely nanobodies against all mouse IgG subclasses and rabbit IgG. They can be produced at large scale in Escherichia coli and could thus make secondary antibody prodn. in animals obsolete. Their recombinant nature allows fusion with affinity tags or reporter enzymes as well as efficient maleimide chem. for fluorophore coupling. We demonstrate their superior performance in Western blotting, in both peroxidase- and fluorophore-linked form. Their site-specific labeling with multiple fluorophores creates bright imaging reagents for confocal and superresoln. microscopy with much smaller label displacement than traditional secondary antibodies. They also enable simpler and faster immunostaining protocols, and allow multitarget localization with primary IgGs from the same species and of the same class.
- 22Vaughan, J. C.; Dempsey, G. T.; Sun, E.; Zhuang, X. Phosphine-Quenching of Cyanine Dyes as a Versatile Tool for Fluorescence Microscopy. J. Am. Chem. Soc. 2013, 135, 1197– 1200, DOI: 10.1021/ja3105279[ACS Full Text
], [CAS], Google Scholar
22https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3sXmvF2rug%253D%253D&md5=163fc5e390c184b71edd202f2d80ea7ePhosphine Quenching of Cyanine Dyes as a Versatile Tool for Fluorescence MicroscopyVaughan, Joshua C.; Dempsey, Graham T.; Sun, Eileen; Zhuang, XiaoweiJournal of the American Chemical Society (2013), 135 (4), 1197-1200CODEN: JACSAT; ISSN:0002-7863. (American Chemical Society)The authors report that the cyanine dye Cy5 and several of its structural relatives are reversibly quenched by the phosphine tris(2-carboxyethyl)phosphine (TCEP). Using Cy5 as a model, the quenching reaction occurs by 1,4-addn. of the phosphine to the polymethine bridge of Cy5 to form a covalent adduct. Illumination with UV light dissocs. the adduct and returns the dye to the fluorescent state. TCEP quenching can be used for super-resoln. imaging as well as for other applications, such as differentiating between mols. inside and outside the cell. - 23Olivier, N.; Keller, D.; Gönczy, P.; Manley, S. Resolution Doubling in 3D-STORM Imaging through Improved Buffers. PLoS One 2013, 8 (7), 1– 9, DOI: 10.1371/journal.pone.0069004
- 24Endesfelder, U.; Malkusch, S.; Fricke, F.; Heilemann, M. A Simple Method to Estimate the Average Localization Precision of a Single-Molecule Localization Microscopy Experiment. Histochem. Cell Biol. 2014, 141 (6), 629– 638, DOI: 10.1007/s00418-014-1192-3[Crossref], [PubMed], [CAS], Google Scholar24https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2cXisFWhs7Y%253D&md5=a804a9d46ee82438cbe478fa25034140A simple method to estimate the average localization precision of a single-molecule localization microscopy experimentEndesfelder, Ulrike; Malkusch, Sebastian; Fricke, Franziska; Heilemann, MikeHistochemistry and Cell Biology (2014), 141 (6), 629-638CODEN: HCBIFP; ISSN:0948-6143. (Springer)The localization precision is a crucial and important parameter for single-mol. localization microscopy (SMLM) and directly influences the achievable spatial resoln. It primarily depends on exptl. imaging conditions and the registration potency of the algorithm used. We propose a new and simple routine to est. the av. exptl. localization precision in SMLM, based on the nearest neighbor anal. By exploring different exptl. and simulated targets, we show that this approach can be generally used for any 2D or 3D SMLM data and that reliable values for the localization precision σSMLM are obtained. Knowing σSMLM is a prerequisite for consistent visualization or any quant. structural anal., e.g., cluster anal. or colocalization studies.
- 25Li, Y.; Mund, M.; Hoess, P.; Deschamps, J.; Matti, U.; Nijmeijer, B.; Sabinina, V. J.; Ellenberg, J.; Schoen, I.; Ries, J. Real-Time 3D Single-Molecule Localization Using Experimental Point Spread Functions. Nat. Methods 2018, 15 (5), 367– 369, DOI: 10.1038/nmeth.4661[Crossref], [PubMed], [CAS], Google Scholar25https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1cXnt1Sns7c%253D&md5=c58c4361da24656c77566dd8ee4ea120Real-time 3D single-molecule localization using experimental point spread functionsLi, Yiming; Mund, Markus; Hoess, Philipp; Deschamps, Joran; Matti, Ulf; Nijmeijer, Bianca; Sabinina, Vilma Jimenez; Ellenberg, Jan; Schoen, Ingmar; Ries, JonasNature Methods (2018), 15 (5), 367-369CODEN: NMAEA3; ISSN:1548-7091. (Nature Research)We present a real-time fitter for 3D single-mol. localization microscopy using exptl. point spread functions (PSFs) that achieves minimal uncertainty in 3D on any microscope and is compatible with any PSF engineering approach. We used this method to image cellular structures and attained unprecedented image quality for astigmatic PSFs. The fitter compensates for most optical aberrations and makes accurate 3D super-resoln. microscopy broadly accessible, even on std. microscopes without dedicated 3D optics.
- 26Auer, A.; Schlichthaerle, T.; Woehrstein, J. B.; Schueder, F.; Strauss, M. T.; Grabmayr, H.; Jungmann, R. Nanometer-Scale Multiplexed Super-Resolution Imaging with an Economic 3D-DNA-PAINT Microscope. ChemPhysChem 2018, 19 (22), 3024– 3034, DOI: 10.1002/cphc.201800630[Crossref], [PubMed], [CAS], Google Scholar26https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1cXhvVGltLzM&md5=de5d6cc8edf56158bd04e01fee92015bNanometer-scale Multiplexed Super-Resolution Imaging with an Economic 3D-DNA-PAINT MicroscopeAuer, Alexander; Schlichthaerle, Thomas; Woehrstein, Johannes B.; Schueder, Florian; Strauss, Maximilian T.; Grabmayr, Heinrich; Jungmann, RalfChemPhysChem (2018), 19 (22), 3024-3034CODEN: CPCHFT; ISSN:1439-4235. (Wiley-VCH Verlag GmbH & Co. KGaA)Optical super-resoln. microscopy is rapidly changing the way imaging studies in the biol. and biomedical sciences are conducted. Due to the unique capability of achieving mol. contrast using fluorescent labels and sub-diffraction resoln. down to a few tens of nanometers, super-resoln. is developing as an attractive imaging modality. While the increased spatial resoln. has already enabled structural studies at unprecedented mol. detail, the wide-spread use of super-resoln. approaches as a std. characterization technique in biol. labs. has thus far been prevented by mainly two issues: (1) Intricate sample prepn. and image acquisition and (2) costly and complex instrumentation. We here introduce a combination of the recently developed super-resoln. technique DNA-PAINT (DNA points accumulation for imaging in nanoscale topog.) with an easy-to-replicate, custom-built 3D single-mol. microscope (termed liteTIRF) that is an order of magnitude more economic in cost compared to most com. systems. We assay the performance of our system using synthetic two- and three-dimensional DNA origami structures and show the applicability to single- and multiplexed cellular imaging.
- 27Kellogg, E. H.; Hejab, N. M. A.; Howes, S.; Northcote, P.; Miller, J. H.; Díaz, J. F.; Downing, K. H.; Nogales, E. Insights into the Distinct Mechanisms of Action of Taxane and Non-Taxane Microtubule Stabilizers from Cryo-EM Structures. J. Mol. Biol. 2017, 429 (5), 633– 646, DOI: 10.1016/j.jmb.2017.01.001[Crossref], [PubMed], [CAS], Google Scholar27https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2sXhtFOgurk%253D&md5=2fc94e61ef199ff77990dda297020180Insights into the Distinct Mechanisms of Action of Taxane and Non-Taxane Microtubule Stabilizers from Cryo-EM StructuresKellogg, Elizabeth H.; Hejab, Nisreen M. A.; Howes, Stuart; Northcote, Peter; Miller, John H.; Diaz, J. Fernando; Downing, Kenneth H.; Nogales, EvaJournal of Molecular Biology (2017), 429 (5), 633-646CODEN: JMOBAK; ISSN:0022-2836. (Elsevier Ltd.)A no. of microtubule (MT)-stabilizing agents (MSAs) have demonstrated or predicted potential as anticancer agents, but a detailed structural basis for their mechanism of action is still lacking. We have obtained high-resoln. (3.9-4.2 Å) cryo-electron microscopy (cryo-EM) reconstructions of MTs stabilized by the taxane-site binders Taxol and zampanolide, and by peloruside, which targets a distinct, non-taxoid pocket on β-tubulin. We find that each mol. has unique distinct structural effects on the MT lattice structure. Peloruside acts primarily at lateral contacts and has an effect on the "seam" of heterologous interactions, enforcing a conformation more similar to that of homologous (i.e., non-seam) contacts by which it regularizes the MT lattice. In contrast, binding of either Taxol or zampanolide induces MT heterogeneity. In doubly bound MTs, peloruside overrides the heterogeneity induced by Taxol binding. Our structural anal. illustrates distinct mechanisms of these drugs for stabilizing the MT lattice and is of relevance to the possible use of combinations of MSAs to regulate MT activity and improve therapeutic potential.
- 28Brandt, J. P.; Patapoff, T. W.; Aragon, S. R. Construction, MD Simulation, and Hydrodynamic Validation of an All-Atom Model of a Monoclonal IgG Antibody. Biophys. J. 2010, 99 (3), 905– 913, DOI: 10.1016/j.bpj.2010.05.003[Crossref], [PubMed], [CAS], Google Scholar28https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3cXpvFWms7s%253D&md5=de25c2118b7897401ff71b6c0516cce3Construction, MD Simulation, and Hydrodynamic Validation of an All-Atom Model of a Monoclonal IgG AntibodyBrandt, J. Paul; Patapoff, Thomas W.; Aragon, Sergio R.Biophysical Journal (2010), 99 (3), 905-913CODEN: BIOJAU; ISSN:0006-3495. (Cell Press)At 150 kDa, antibodies of the IgG class are too large for their structure to be detd. with current NMR methodologies. Because of hinge-region flexibility, it is difficult to obtain at.-level structural information from the crystal, and questions regarding antibody structure and dynamics in soln. remain unaddressed. Here we describe the construction of a model of a human IgG1 monoclonal antibody (trastuzumab) from the crystal structures of fragments. We use a combination of mol.-dynamics (MD) simulation, continuum hydrodynamics modeling, and exptl. diffusion measurements to explore antibody behavior in aq. soln. Hydrodynamic modeling provides a link between the at.-level details of MD simulation and the size- and shape-dependent data provided by hydrodynamic measurements. Eight independent 40 ns MD trajectories were obtained with the AMBER program suite. The ensemble av. of the computed transport properties over all of the MD trajectories agrees remarkably well with the value of the translational diffusion coeff. obtained with dynamic light scattering at 20°C and 27°C, and the intrinsic viscosity measured at 20°C. Therefore, our MD results likely represent a realistic sampling of the conformational space that an antibody explores in aq. soln.
- 29De Michele, C.; De Los Rios, P.; Foffi, G.; Piazza, F. Simulation and Theory of Antibody Binding to Crowded Antigen-Covered Surfaces. PLoS Comput. Biol. 2016, 12 (3), 1– 17, DOI: 10.1371/journal.pcbi.1004752
- 30Harris, L. J.; Larson, S. B.; Hasel, K. W.; McPherson, A. Refined Structure of an Intact IgG2a Monoclonal Antibody. Biochemistry 1997, 36 (7), 1581– 1597, DOI: 10.1021/bi962514+[ACS Full Text
], [CAS], Google Scholar
30https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADyaK2sXht1Ohs78%253D&md5=762b87a6094c1cf0eb698314f64ebfafRefined Structure of an Intact IgG2a Monoclonal AntibodyHarris, Lisa J.; Larson, Steven B.; Hasel, Karl W.; McPherson, AlexanderBiochemistry (1997), 36 (7), 1581-1597CODEN: BICHAW; ISSN:0006-2960. (American Chemical Society)The structure of an intact, anti-canine lymphoma monoclonal antibody (Mab231) was detd. by mol. replacement and refined in a triclinic cell to an R-value of 20.9%, using synchrotron diffraction data from 2.8 to 20 Å resoln. All segments of the antibody, including the hinge region and carbohydrate component, are visible in electron d. maps. There is no overall symmetry to the antibody, as the Fc is disposed in an entirely oblique manner with respect to the Fabs. The CH2 and CH3 domains do, however, possess a nearly exact, local 2-fold relation. The Fab segments are related by a second, independent, local dyad axis, exact only with respect to const. domains. Variable domains exhibit no symmetry relation as a consequence of the 16° difference in Fab elbow angles. Variable domain pair assocns. VL:VH for the Fabs are virtually the same, and corresponding CDRs of the 2 Fabs also are nearly identical in structure. CDR-H3 displays the greatest difference. Hypervariable loops of both Fabs are involved in contacts with symmetry-related Fc segments at the CH2-CH3 switch junction, suggesting a "complex" structure. The hinge segment connecting Fabs with the Fc is quite extended and exhibits thermal factors indicative of a high degree of mobility. It consists of a well-defined upper hinge that partially maintains dyad symmetry and a fairly rigid core bounded above and below by fluid polypeptides that provide segmental flexibility. This structure represents the first visualization by x-ray anal. of a murine Fc segment, and its CH2 domains exhibit substantial rigid body conformational changes with respect to the human Fc used as an initial mol. replacement model. The oligosaccharides were found by difference Fourier syntheses to be very similar to those of the free human Fc fragment, although differences are present in the terminal residues. The detailed structure of the IgG presented here, and the distribution of effector binding sites, appears consistent with effector activation mechanisms involving translocation and/or aggregation of the Fc following antigen binding by the Fabs. - 31Bongini, L.; Fanelli, D.; Piazza, F.; De Los Rios, P.; Sandin, S.; Skoglund, U. Freezing Immunoglobulins to See Them Move. Proc. Natl. Acad. Sci. U. S. A. 2004, 101 (17), 6466– 6471, DOI: 10.1073/pnas.0400119101[Crossref], [PubMed], [CAS], Google Scholar31https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD2cXjvVyitLg%253D&md5=78a91c90a7605ec67627cd5ae249a50cFreezing immunoglobulins to see them moveBongini, L.; Fanelli, D.; Piazza, F.; De Los Rios, P.; Sandin, S.; Skoglund, U.Proceedings of the National Academy of Sciences of the United States of America (2004), 101 (17), 6466-6471CODEN: PNASA6; ISSN:0027-8424. (National Academy of Sciences)The issue of protein dynamics and its implications in the biol. function of proteins are arousing greater and greater interest in different fields of mol. biol. In cryo-electron tomog. expts. one may take several snapshots of a given biol. macromol. In principle, a large enough collection of snapshots of the mol. may then be used to calc. its equil. configuration in terms of the exptl. accessible degrees of freedom and, hence, to est. its potential energy. This information would be crucial in order to analyze the biol. functions of biomols. by directly accessing the relevant dynamical indicators. In this article, we analyze the results of cryo-electron tomog. expts. performed on monoclonal murine IgG2a antibodies. We measure the equil. distribution of the mol. in terms of the relevant angular coordinates and build a mech. model of the antibody dynamics. This approach enables us to derive an explicit expression of the IgG potential energy. Furthermore, we discuss the configuration space at equil. in relation to results from other techniques, and we set our discussion in the context of the current debate regarding conformation and flexibility of antibodies.
- 32Dani, A.; Huang, B.; Bergan, J.; Dulac, C.; Zhuang, X. Superresolution Imaging of Chemical Synapses in the Brain. Neuron 2010, 68 (5), 843– 856, DOI: 10.1016/j.neuron.2010.11.021[Crossref], [PubMed], [CAS], Google Scholar32https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3cXhsFGhu73I&md5=07c554f9890e13d6767bc4205c3001f6Superresolution Imaging of Chemical Synapses in the BrainDani, Adish; Huang, Bo; Bergan, Joseph; Dulac, Catherine; Zhuang, Xiao-WeiNeuron (2010), 68 (5), 843-856CODEN: NERNET; ISSN:0896-6273. (Cell Press)Detn. of the mol. architecture of synapses requires nanoscopic image resoln. and specific mol. recognition, a task that has so far defied many conventional imaging approaches. Here, we present a superresoln. fluorescence imaging method to visualize the mol. architecture of synapses in the brain. Using multicolor, three-dimensional stochastic optical reconstruction microscopy, the distributions of synaptic proteins can be measured with nanometer precision. Furthermore, the wide-field, volumetric imaging method enables high-throughput, quant. anal. of a large no. of synapses from different brain regions. To demonstrate the capabilities of this approach, we have detd. the organization of ten protein components of the presynaptic active zone and the postsynaptic d. Variations in synapse morphol., neurotransmitter receptor compn., and receptor distribution were obsd. both among synapses and across different brain regions. Combination with optogenetics further allowed mol. events assocd. with synaptic plasticity to be resolved at the single-synapse level.
- 33Sehnal, D.; Rose, A. S.; Koča, J.; Burley, S. K.; Velankar, S. Mol*: Towards a Common Library and Tools for Web Molecular Graphics. In Workshop on Molecular Graphics and Visual Analysis of Molecular Data; Byska, J., Krone, M., Sommer, B., Eds.; The Eurographics Association, 2018; pp 29– 33 DOI: 10.2312/molva.20181103 .
- 34Zheng, K.; Bantog, C.; Bayer, R. The Impact of Glycosylation on Monoclonal Antibody Conformation and Stability. mAbs 2011, 3 (6), 568– 576, DOI: 10.4161/mabs.3.6.17922[Crossref], [PubMed], [CAS], Google Scholar34https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BC383htFGquw%253D%253D&md5=7a337ca1fb8a8e12a30b3d1a679f9849The impact of glycosylation on monoclonal antibody conformation and stabilityZheng Kai; Bantog Christopher; Bayer RobertmAbs (2011), 3 (6), 568-76 ISSN:.Antibody glycosylation is a common post-translational modification and has a critical role in antibody effector function. The use of glycoengineering to produce antibodies with specific glycoforms may be required to achieve the desired therapeutic efficacy. However, the modified molecule could have unusual behavior during development due to the alteration of its intrinsic properties and stability. In this study, we focused on the differences between glycosylated and deglycosylated antibodies, as aglycosyl antibodies are often chosen when effector function is not desired or unimportant. We selected three human IgG1 antibodies and used PNGase F to remove their oligosaccharide chains. Although there were no detected secondary or tertiary structural changes after deglycosylation, other intrinsic properties of the antibody were altered with the removal of oligosaccharide chains in the Fc region. The apparent molecular hydrodynamic radius increased after deglycosylation based on size-exclusion chromatography analysis. Deglycosylated antibodies exhibited less thermal stability for the CH2 domain and less resistance to GdnHCl induced unfolding. Susceptibility to proteolytic cleavage demonstrated that the deglycosylated version was more susceptible to papain. An accelerated stability study revealed that deglycosylated antibodies had higher aggregation rates. These changes may impact the development of aglycosyl antibody biotherapeutics.
- 35Mimura, Y.; Church, S.; Ghirlando, R.; Ashton, P. R.; Dong, S.; Goodall, M.; Lund, J.; Jefferis, R. The Influence of Glycosylation on the Thermal Stability and Effector Function Expression of Human IgG1-Fc: Properties of a Series of Truncated Glycoforms. Mol. Immunol. 2000, 37 (12–13), 697– 706, DOI: 10.1016/S0161-5890(00)00105-X[Crossref], [PubMed], [CAS], Google Scholar35https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BD3M7os1Srsw%253D%253D&md5=07eed4544a1af8d14f58c52d6a625a88The influence of glycosylation on the thermal stability and effector function expression of human IgG1-Fc: properties of a series of truncated glycoformsMimura Y; Church S; Ghirlando R; Ashton P R; Dong S; Goodall M; Lund J; Jefferis RMolecular immunology (2000), 37 (12-13), 697-706 ISSN:0161-5890.Antibodies are multifunctional molecules that following the formation of antibody antigen complexes, may activate mechanisms to effect the clearance and destruction of the antigen (pathogen). The IgG molecule is comprised of three globular protein moieties (2Fab+Fc) linked through a flexible hinge region. While the Fabs bind antigens, the Fc triggers effector mechanisms through interactions with specific ligands, e.g. cellular receptors (FcgammaR), and the C1 component of complement. Glycosylation of IgG-Fc has been shown to be essential for efficient activation of FcgammaR and C1. We report the generation of a series of truncated glycoforms of IgG-Fc, and the analysis of the contribution of the residual oligosaccharide to IgG-Fc function and thermal stability. Differential scanning microcalorimetry has been used to compare the stabilities of the homogeneous glycoforms of IgG1-Fc. The results show that all truncated oligosaccharides confer a degree of functional activity, and thermodynamic stability to the IgG1-Fc, in comparison with deglycosylated IgG1-Fc. The same truncated glycoforms of an intact IgG1 anti-MHC Class II antibody are shown to exhibit differential functional activity for FcgammaRI and C1 ligands, relative to deglycosylated IgG1. The minimal glycoform investigated had a trisaccharide attached to each heavy chain and can be expected to influence protein structure primarily in the proximity of the N-terminal region of the C(H)2 domain, implicated as a binding site for multiple effector ligands. These data provide a thermodynamic rationale for the modulation of antibody effector functions by different glycoforms.
- 36Szenczi, Á.; Kardos, J.; Medgyesi, G. A.; Závodszky, P. The Effect of Solvent Environment on the Conformation and Stability of Human Polyclonal IgG in Solution. Biologicals 2006, 34, 5– 14, DOI: 10.1016/j.biologicals.2005.06.007[Crossref], [PubMed], [CAS], Google Scholar36https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD28Xhslylsrs%253D&md5=f17aff81c43bc676ceb4fedc3e1ed7e1The effect of solvent environment on the conformation and stability of human polyclonal IgG in solutionSzenczi, Arpad; Kardos, Jozsef; Medgyesi, Gyoergy A.; Zavodszky, PeterBiologicals (2006), 34 (1), 5-14CODEN: BILSEC; ISSN:1045-1056. (Elsevier B.V.)Stability of therapeutic IgG prepns. is an important issue as adequate efficacy and safety has to be ensured throughout a long shelf life. To this end, denaturation and aggregation have to be avoided. In many cases sugars are applied for stabilizing IgG in relatively high concn. (5-10%). However, certain sugars (sucrose, maltose) are responsible for adverse effects including renal failure. In this work we reassessed the effect of pH and stabilizers to optimize the solvent environment and minimize the amt. of additives without endangering quality and stability. Since both biol. function and aggregation depend on the conformational properties of individual IgG mols., two sensitive and rapid phys. methods were introduced to assess conformational changes and structural stability as a function of pH and addn. of std. stabilizers. It was obsd. that the conformational stability decreases with decreasing pH, while the resistance against aggregation improves. The optimum pH range for storage is 5.0-6.0, as a compromise between conformational stability and the tendency for oligomerization. Intriguingly, additives in physiol. acceptable concn. have no effect on the thermal stability of IgG. On the other hand, glucose or sorbitol, even at a concn. as low as 1%, have significant effect on the tertiary structure as revealed by near-UV-CD spectroscopy, reflecting changes in the environment of arom. side-chains. Although, 0.3% leucine does not increase conformational stability, it decreases the aggregation tendency even more efficiently than 1% glucose or sorbitol. Both pH and storage temp. are decisive factors for the long-term stability of IgG solns. An increase in the dimer content was obsd. upon storage at 5°C which was partly reverted upon incubation at 37°C. Storage at temps. higher than 5°C may help to maintain an optimal proportion of dimers. Regarding the known side effects, and their limited stabilizing capacity at low concn., it is advisable to omit sugars at i.v. Ig (IVIG) formulation. Hydrophobic amino acids give promising alternatives.
- 37Vermeer, A. W. P.; Norde, W. The Thermal Stability of Immunoglobulin: Unfolding and Aggregation of a Multi-Domain Protein. Biophys. J. 2000, 78 (1), 394– 404, DOI: 10.1016/S0006-3495(00)76602-1[Crossref], [PubMed], [CAS], Google Scholar37https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD3cXktFeqsQ%253D%253D&md5=3664753139bb8ca404f4c73051cc2adeThe thermal stability of immunoglobulin: unfolding and aggregation of a multi-domain proteinVermeer, Arnoldus W. P.; Norde, WillemBiophysical Journal (2000), 78 (1), 394-404CODEN: BIOJAU; ISSN:0006-3495. (Biophysical Society)The denaturation of IgG was studied by different calorimetric methods and CD spectroscopy. The thermogram of the Ig showed two main transitions that are a superimposition of distinct denaturation steps. It was shown that the two transitions have different sensitivities to changes in temp. and pH. The two peaks represent the Fab and Fc fragments of the IgG mol. The Fab fragment is most sensitive to heat treatment, whereas the Fc fragment is most sensitive to decreasing pH. The transitions were independent, and the unfolding was immediately followed by an irreversible aggregation step. Below the unfolding temp., the unfolding is the rate-detg. step in the overall denaturation process. At higher temps. where a relatively high concn. of (partially) unfolded IgG mols. is present, the rate of aggregation is so fast that IgG mols. become locked in aggregates before they are completely denatured. Furthermore, the structure of the aggregates formed depends on the denaturation method. The CD spectrum of the IgG is also strongly affected by both heat treatment and low pH treatment. It was shown that a strong correlation exists between the denaturation transitions as obsd. by calorimetry and the changes in secondary structure derived from CD. After both heat- and low-pH-induced denaturation, a significant fraction of the secondary structure remains.
- 38Ries, J. SMAP: A Modular Super-Resolution Microscopy Analysis Platform for SMLM Data. Nat. Methods 2020, 17 (9), 870– 872, DOI: 10.1038/s41592-020-0938-1[Crossref], [PubMed], [CAS], Google Scholar38https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3cXhs1Cjtb%252FE&md5=55783f1f207c22674a59a0f60004011eSMAP: a modular super-resolution microscopy analysis platform for SMLM dataRies, JonasNature Methods (2020), 17 (9), 870-872CODEN: NMAEA3; ISSN:1548-7091. (Nature Research)There is no expanded citation for this reference.
Cited By
This article is cited by 17 publications.
- Wei-Hong Yeo, Yang Zhang, Amy E. Neely, Xiaomin Bao, Cheng Sun, Hao F. Zhang. Investigating Uncertainties in Single-Molecule Localization Microscopy Using Experimentally Informed Monte Carlo Simulation. Nano Letters 2023, 23 (16) , 7253-7259. https://doi.org/10.1021/acs.nanolett.3c00852
- Gang Wen, Volker Leen, Taoufik Rohand, Markus Sauer, Johan Hofkens. Current Progress in Expansion Microscopy: Chemical Strategies and Applications. Chemical Reviews 2023, 123 (6) , 3299-3323. https://doi.org/10.1021/acs.chemrev.2c00711
- Markus Mund, Aline Tschanz, Yu-Le Wu, Felix Frey, Johanna L. Mehl, Marko Kaksonen, Ori Avinoam, Ulrich S. Schwarz, Jonas Ries. Clathrin coats partially preassemble and subsequently bend during endocytosis. Journal of Cell Biology 2023, 222 (3) https://doi.org/10.1083/jcb.202206038
- Kai Kikuchi, Liam D. Adair, Jiarun Lin, Elizabeth J. New, Amandeep Kaur. Photochemical Mechanisms of Fluorophores Employed in Single‐Molecule Localization Microscopy. Angewandte Chemie 2023, 135 (1) https://doi.org/10.1002/ange.202204745
- Kai Kikuchi, Liam D. Adair, Jiarun Lin, Elizabeth J. New, Amandeep Kaur. Photochemical Mechanisms of Fluorophores Employed in Single‐Molecule Localization Microscopy. Angewandte Chemie International Edition 2023, 62 (1) https://doi.org/10.1002/anie.202204745
- Yu-Le Wu, Philipp Hoess, Aline Tschanz, Ulf Matti, Markus Mund, Jonas Ries. Maximum-likelihood model fitting for quantitative analysis of SMLM data. Nature Methods 2023, 20 (1) , 139-148. https://doi.org/10.1038/s41592-022-01676-z
- Abhinav Banerjee, Micky Anand, Mahipal Ganji. Labeling approaches for DNA-PAINT super-resolution imaging. Nanoscale 2023, 380 https://doi.org/10.1039/D2NR06541J
- Paola Cristofari, Mazarine Desplanque, Odile Poirel, Alison Hébert, Sylvie Dumas, Etienne Herzog, Lydia Danglot, David Geny, Jean-François Gilles, Audrey Geeverding, Susanne Bolte, Alexis Canette, Michaël Trichet, Véronique Fabre, Stéphanie Daumas, Nicolas Pietrancosta, Salah El Mestikawy, Véronique Bernard. Nanoscopic distribution of VAChT and VGLUT3 in striatal cholinergic varicosities suggests colocalization and segregation of the two transporters in synaptic vesicles. Frontiers in Molecular Neuroscience 2022, 15 https://doi.org/10.3389/fnmol.2022.991732
- Tiberiu S. Mihaila, Carina Bäte, Lynn M. Ostersehlt, Jasmin K. Pape, Jan Keller-Findeisen, Steffen J. Sahl, Stefan W. Hell. Enhanced incorporation of subnanometer tags into cellular proteins for fluorescence nanoscopy via optimized genetic code expansion. Proceedings of the National Academy of Sciences 2022, 119 (29) https://doi.org/10.1073/pnas.2201861119
- Paul Cordell, Glenn Carrington, Alistair Curd, Francine Parker, Darren Tomlinson, Michelle Peckham. Affimers and nanobodies as molecular probes and their applications in imaging. Journal of Cell Science 2022, 135 (14) https://doi.org/10.1242/jcs.259168
- Fabien Rideau, Audrey Villa, Pauline Belzanne, Emeline Verdier, Eric Hosy, Yonathan Arfi, , Makoto Miyata. Imaging Minimal Bacteria at the Nanoscale: a Reliable and Versatile Process to Perform Single-Molecule Localization Microscopy in Mycoplasmas. Microbiology Spectrum 2022, 10 (3) https://doi.org/10.1128/spectrum.00645-22
- Jan Christoph Thiele, Marvin Jungblut, Dominic A. Helmerich, Roman Tsukanov, Anna Chizhik, Alexey I. Chizhik, Martin J. Schnermann, Markus Sauer, Oleksii Nevskyi, Jörg Enderlein. Isotropic three-dimensional dual-color super-resolution microscopy with metal-induced energy transfer. Science Advances 2022, 8 (23) https://doi.org/10.1126/sciadv.abo2506
- Sheng Liu, Philipp Hoess, Jonas Ries. Super-Resolution Microscopy for Structural Cell Biology. Annual Review of Biophysics 2022, 51 (1) , 301-326. https://doi.org/10.1146/annurev-biophys-102521-112912
- Amandeep Kaur, Liam D. Adair, Sarah R. Ball, Elizabeth J. New, Margaret Sunde. A Fluorescent Sensor for Quantitative Super‐Resolution Imaging of Amyloid Fibril Assembly**. Angewandte Chemie 2022, 134 (10) https://doi.org/10.1002/ange.202112832
- Amandeep Kaur, Liam D. Adair, Sarah R. Ball, Elizabeth J. New, Margaret Sunde. A Fluorescent Sensor for Quantitative Super‐Resolution Imaging of Amyloid Fibril Assembly**. Angewandte Chemie International Edition 2022, 61 (10) https://doi.org/10.1002/anie.202112832
- Rachel E. Jackson, Benjamin Compans, Juan Burrone. Correlative Live-Cell and Super-Resolution Imaging to Link Presynaptic Molecular Organisation With Function. Frontiers in Synaptic Neuroscience 2022, 14 https://doi.org/10.3389/fnsyn.2022.830583
- Luciano A. Masullo, Alan M. Szalai, Lucía F. Lopez, Fernando D. Stefani. Fluorescence nanoscopy at the sub-10 nm scale. Biophysical Reviews 2021, 13 (6) , 1101-1112. https://doi.org/10.1007/s12551-021-00864-z
Abstract
Scheme 1
Scheme 1. Site-specific labeling of IgG antibodies. In a first reaction, N-linked glycans are removed by PNGaseF, and only the available glutamines (Q) at position −2 relative to the deglycosylated sites are modified with H2N-PEG3-N3 catalyzed by microbial transglutaminase (mTG). In a second reaction, fluorophores(-DIBO) or ssDNA(-DBCO) are attached to the azide-modified antibodies using strain-promoted azide–alkyne cycloaddition (SPAAC).Figure 1
Figure 1. Sequence alignment and evaluation of potential modification sites for different IgG subclasses from different host species. Sequences were sourced from the UniProt database and aligned with respect to the N-linked glycosylation sites (gray background). The glutamine at position −2 (black background) is preserved across the majority of IgG subtypes and species for which sequence information was available. *Potential conflict with additional glutamine marked in red; Fl = Florida.
Figure 2
Figure 2. 3D SMLM imaging of microtubule networks and determination of labeling shell dimensions for indirect and direct immunolabeling. (a–d) 3D STORM using Alexa Fluor 647 labeled antibodies in fixed U2OS cells. (a) Schematic (not to scale). (b) Indirect immunolabeling using a randomly labeled secondary donkey-anti-mouse (NHS 2°). (c) Indirect immunolabeling using a site-specifically-labeled secondary donkey-anti-mouse (2°). (d) Direct immunostaining using a site-specifically-labeled primary mouse-anti-α-tubulin (1°). (e–h) 3D DNA-PAINT using DNA-labeled antibodies in U2OS cells (f) and human platelets (g, h). (e) Schematic (not to scale). (f, g) Indirect immunolabeling using a site-specifically-labeled donkey-anti-mouse secondary (2°). (h) Direct labeling using a site-specifically-labeled primary mouse-anti-α-tubulin (1°). For all conditions, representative 3D SMLM images are shown (left). Labeling shell dimensions around microtubules were determined from averaged experimental yz cross-sections and fitted label distributions (right). The fitted label distribution was a Gaussian ring kernel (r: radius; w: full width at half-maximum) convolved with the localization precisions in y and z, respectively. (i) Comparison of center positions and widths of labeling shell dimensions for the different labeling strategies in b–d and f–h.
Figure 3
Figure 3. Monte Carlo simulations of antibody conformations at microtubules. (a) Location of the epitope of the anti-α-tubulin antibody in our geometric model (right) based on the cryo-EM structure of a microtubule (left; PDB 5SYF, ref (27); molecular surface rendered using Mol*, ref (33)). (b) Each IgG molecule was modeled by two segments corresponding to Fab and Fc fragments connected by a flexible hinge region. Right: Parametrization of Fab, Fc, and linker segments of the labeled primary antibody. Left: Dimensions were based on the crystallographic structure of IgG2a (PDB 1IGT, ref (30), visualization by Mol*). While the location of the modification is precisely known (Glu), the binding sites of polyclonal secondary antibodies are assumed to be evenly distributed over the Fc region. For a schematic of indirect immunolabeling, see Supplementary Figure S5c). Simulated reporter distribution for primary antibodies. Left: yz cross-section. Right: Radial distribution. (d) Simulated reporter distribution for primary plus secondary antibody complexes. Panels as in (c). (e) Comparison of reporter distributions for primary antibodies between simulations (left; convolved with the localization and experimental imprecision) and experiments (middle: pooled cross-sections as in Figure 2). Right: Normalized residuals of the difference between experiment and model. Top: STORM. Bottom: DNA-PAINT. (f) Comparison of reporter distributions for secondary antibodies between simulations and experiments. Panels as in (d).
References
ARTICLE SECTIONSThis article references 38 other publications.
- 1Huang, F.; Sirinakis, G.; Allgeyer, E. S.; Schroeder, L. K.; Duim, W. C.; Kromann, E. B.; Phan, T.; Rivera-Molina, F. E.; Myers, J. R.; Irnov, I.; Lessard, M.; Zhang, Y.; Handel, M. A.; Jacobs-Wagner, C.; Lusk, C. P.; Rothman, J. E.; Toomre, D.; Booth, M. J.; Bewersdorf, J. Ultra-High Resolution 3D Imaging of Whole Cells. Cell 2016, 166 (4), 1028– 1040, DOI: 10.1016/j.cell.2016.06.016[Crossref], [PubMed], [CAS], Google Scholar1https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC28XhtFCiu7rN&md5=45816ee5300c4b686789eba93ed8b28fUltra-High Resolution 3D Imaging of Whole CellsHuang, Fang; Sirinakis, George; Allgeyer, Edward S.; Schroeder, Lena K.; Duim, Whitney C.; Kromann, Emil B.; Phan, Thomy; Rivera-Molina, Felix E.; Myers, Jordan R.; Irnov, Irnov; Lessard, Mark; Zhang, Yongdeng; Handel, Mary Ann; Jacobs-Wagner, Christine; Lusk, C. Patrick; Rothman, James E.; Toomre, Derek; Booth, Martin J.; Bewersdorf, JoergCell (Cambridge, MA, United States) (2016), 166 (4), 1028-1040CODEN: CELLB5; ISSN:0092-8674. (Cell Press)Fluorescence nanoscopy, or super-resoln. microscopy, has become an important tool in cell biol. research. However, because of its usually inferior resoln. in the depth direction (50-80 nm) and rapidly deteriorating resoln. in thick samples, its practical biol. application has been effectively limited to two dimensions and thin samples. Here, the authors present the development of whole-cell 4Pi single-mol. switching nanoscopy (W-4PiSMSN), an optical nanoscope that allows imaging of three-dimensional (3D) structures at 10- to 20-nm resoln. throughout entire mammalian cells. The authors demonstrate the wide applicability of W-4PiSMSN across diverse research fields by imaging complex mol. architectures ranging from bacteriophages to nuclear pores, cilia, and synaptonemal complexes in large 3D cellular vols.
- 2Jungmann, R.; Avendaño, M. S.; Dai, M.; Woehrstein, J. B.; Agasti, S. S.; Feiger, Z.; Rodal, A.; Yin, P. Quantitative Super-Resolution Imaging with qPAINT. Nat. Methods 2016, 13 (5), 439– 442, DOI: 10.1038/nmeth.3804[Crossref], [PubMed], [CAS], Google Scholar2https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC28XkvV2jtLk%253D&md5=1ce6b7fdb5e94a4309c88942f751a172Quantitative super-resolution imaging with qPAINTJungmann, Ralf; Avendano, Maier S.; Dai, Mingjie; Woehrstein, Johannes B.; Agasti, Sarit S.; Feiger, Zachary; Rodal, Avital; Yin, PengNature Methods (2016), 13 (5), 439-442CODEN: NMAEA3; ISSN:1548-7091. (Nature Publishing Group)Counting mols. in complexes is challenging, even with super-resoln. microscopy. Here, we use the programmable and specific binding of dye-labeled DNA probes to count integer nos. of targets. This method, called quant. points accumulation in nanoscale topog. (qPAINT), works independently of dye photophysics for robust counting with high precision and accuracy over a wide dynamic range. qPAINT was benchmarked on DNA nanostructures and demonstrated for cellular applications by quantifying proteins in situ and the no. of single-mol. FISH probes bound to an mRNA target.
- 3Koch, B.; Nijmeijer, B.; Kueblbeck, M.; Cai, Y.; Walther, N.; Ellenberg, J. Generation and Validation of Homozygous Fluorescent Knock-In Cells Using CRISPR–Cas9 Genome Editing. Nat. Protoc. 2018, 13 (6), 1465– 1487, DOI: 10.1038/nprot.2018.042[Crossref], [PubMed], [CAS], Google Scholar3https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1cXhtVamurbP&md5=9a24e4b52eb37fe9f012717177b45f6bGeneration and validation of homozygous fluorescent knock-in cells using CRISPR-Cas9 genome editingKoch, Birgit; Nijmeijer, Bianca; Kueblbeck, Moritz; Cai, Yin; Walther, Nike; Ellenberg, JanNature Protocols (2018), 13 (6), 1465-1487CODEN: NPARDW; ISSN:1750-2799. (Nature Research)Gene tagging with fluorescent proteins is essential for investigations of the dynamic properties of cellular proteins. CRISPR-Cas9 technol. is a powerful tool for inserting fluorescent markers into all alleles of the gene of interest (GOI) and allows functionality and physiol. expression of the fusion protein. It is essential to evaluate such genome-edited cell lines carefully in order to preclude off-target effects caused by (i) incorrect insertion of the fluorescent protein, (ii) perturbation of the fusion protein by the fluorescent proteins or (iii) nonspecific genomic DNA damage by CRISPR-Cas9. In this protocol, we provide a step-by-step description of our systematic pipeline to generate and validate homozygous fluorescent knock-in cell lines. We have used the paired Cas9D10A nickase approach to efficiently insert tags into specific genomic loci via homol.-directed repair (HDR) with minimal off-target effects. It is time-consuming and costly to perform whole-genome sequencing of each cell clone to check for spontaneous genetic variations occurring in mammalian cell lines. Therefore, we have developed an efficient validation pipeline of the generated cell lines consisting of junction PCR, Southern blotting anal., Sanger sequencing, microscopy, western blotting anal. and live-cell imaging for cell-cycle dynamics. This protocol takes between 6 and 9 wk. With this protocol, up to 70% of the targeted genes can be tagged homozygously with fluorescent proteins, thus resulting in physiol. levels and phenotypically functional expression of the fusion proteins.
- 4Ghodke, H.; Caldas, V. E. A.; Punter, C. M.; van Oijen, A. M.; Robinson, A. Single-Molecule Specific Mislocalization of Red Fluorescent Proteins in Live Escherichia Coli. Biophys. J. 2016, 111 (1), 25– 27, DOI: 10.1016/j.bpj.2016.05.047[Crossref], [PubMed], [CAS], Google Scholar4https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC28XhtVWns7fK&md5=e4760ed7f31002c37e995eb59f14c1e8Single-Molecule Specific Mislocalization of Red Fluorescent Proteins in Live Escherichia coliGhodke, Harshad; Caldas, Victor E. A.; Punter, Christiaan M.; van Oijen, Antoine M.; Robinson, AndrewBiophysical Journal (2016), 111 (1), 25-27CODEN: BIOJAU; ISSN:0006-3495. (Cell Press)Tagging of individual proteins with genetically encoded fluorescent proteins (FPs) has been used extensively to study localization and interactions in live cells. Recent developments in single-mol. localization microscopy have enabled the dynamic visualization of individual tagged proteins inside living cells. However, tagging proteins with FPs is not without problems: formation of insol. aggregates and inhibition of native functions of the protein are well-known issues. Previously reported artifacts manifest themselves at all expression levels of the FP-tagged proteins, making the design of control expts. relatively straightforward. Here, we describe a previously uncharacterized mislocalization artifact of Entacmaea quadricolor red fluorescent protein variants that is detectable at the single-mol. level in live Escherichia coli cells.
- 5Sahl, S. J.; Hell, S. W.; Jakobs, S. Fluorescence Nanoscopy in Cell Biology. Nat. Rev. Mol. Cell Biol. 2017, 18 (11), 685– 701, DOI: 10.1038/nrm.2017.71[Crossref], [PubMed], [CAS], Google Scholar5https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2sXhsVChs7rO&md5=c72d990fae52915e9a9623e2eb065cceFluorescence nanoscopy in cell biologySahl, Steffen J.; Hell, Stefan W.; Jakobs, StefanNature Reviews Molecular Cell Biology (2017), 18 (11), 685-701CODEN: NRMCBP; ISSN:1471-0072. (Nature Research)A review. Fluorescence nanoscopy uniquely combines minimally invasive optical access to the internal nanoscale structure and dynamics of cells and tissues with mol. detection specificity. While the basic phys. principles of 'super-resoln.' imaging were discovered in the 1990s, with initial exptl. demonstrations following in 2000, the broad application of super-resoln. imaging to address cell-biol. questions has only more recently emerged. Nanoscopy approaches have begun to facilitate discoveries in cell biol. and to add new knowledge. One current direction for method improvement is the ambition to quant. account for each mol. under investigation and assess true mol. colocalization patterns via multi-color analyses. In pursuing this goal, the labeling of individual mols. to enable their visualization has emerged as a central challenge. Extending nanoscale imaging into (sliced) tissue and whole-animal contexts is a further goal. In this Review we describe the successes to date and discuss current obstacles and possibilities for further development.
- 6Schlichthaerle, T.; Eklund, A. S.; Schueder, F.; Strauss, M. T.; Tiede, C.; Curd, A.; Ries, J.; Peckham, M.; Tomlinson, D. C.; Jungmann, R. Site-Specific Labeling of Affimers for DNA-PAINT Microscopy. Angew. Chem., Int. Ed. 2018, 57 (34), 11060– 11063, DOI: 10.1002/anie.201804020[Crossref], [CAS], Google Scholar6https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1cXhtlWqtL3L&md5=85559fed38b1e5ff592fb2f67975029fSite-Specific Labeling of Affimers for DNA-PAINT MicroscopySchlichthaerle, Thomas; Eklund, Alexandra S.; Schueder, Florian; Strauss, Maximilian T.; Tiede, Christian; Curd, Alistair; Ries, Jonas; Peckham, Michelle; Tomlinson, Darren C.; Jungmann, RalfAngewandte Chemie, International Edition (2018), 57 (34), 11060-11063CODEN: ACIEF5; ISSN:1433-7851. (Wiley-VCH Verlag GmbH & Co. KGaA)Optical super-resoln. techniques allow fluorescence imaging below the classical diffraction limit of light. From a technol. standpoint, recent methods are approaching mol.-scale spatial resoln. However, this remarkable achievement is not easily translated to imaging of cellular components, since current labeling approaches are limited by either large label sizes (antibodies) or the sparse availability of small and efficient binders (nanobodies, aptamers, genetically-encoded tags). In this work, we combined recently developed Affimer reagents with site-specific DNA modification for high-efficiency labeling and imaging using DNA-PAINT. We assayed our approach using an actin Affimer. The small DNA-conjugated affinity binders could provide a soln. for efficient multitarget super-resoln. imaging in the future.
- 7Shaw, A.; Hoffecker, I. T.; Smyrlaki, I.; Rosa, J.; Grevys, A.; Bratlie, D.; Sandlie, I.; Michaelsen, T. E.; Andersen, J. T.; Högberg, B. Binding to Nanopatterned Antigens Is Dominated by the Spatial Tolerance of Antibodies. Nat. Nanotechnol. 2019, 14, 1, DOI: 10.1038/s41565-018-0336-3
- 8Mikhaylova, M.; Cloin, B. M. C.; Finan, K.; van den Berg, R.; Teeuw, J.; Kijanka, M. M.; Sokolowski, M.; Katrukha, E. a; Maidorn, M.; Opazo, F.; Moutel, S.; Vantard, M.; Perez, F.; van Bergen En Henegouwen, P. M. P.; Hoogenraad, C. C.; Ewers, H.; Kapitein, L. C. Resolving Bundled Microtubules Using Anti-Tubulin Nanobodies. Nat. Commun. 2015, 6 (May), 7933, DOI: 10.1038/ncomms8933[Crossref], [PubMed], [CAS], Google Scholar8https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2MXhsVKhtbjJ&md5=ccb0d03ce3408f23623cfa0049540572Resolving bundled microtubules using anti-tubulin nanobodiesMikhaylova, Marina; Cloin, Bas M. C.; Finan, Kieran; van den Berg, Robert; Teeuw, Jalmar; Kijanka, Marta M.; Sokolowski, Mikolaj; Katrukha, Eugene A.; Maidorn, Manuel; Opazo, Felipe; Moutel, Sandrine; Vantard, Marylin; Perez, Frank; van Bergen en Henegouwen, Paul M. P.; Hoogenraad, Casper C.; Ewers, Helge; Kapitein, Lukas C.Nature Communications (2015), 6 (), 7933CODEN: NCAOBW; ISSN:2041-1723. (Nature Publishing Group)Microtubules are hollow biopolymers of 25-nm diam. and are key constituents of the cytoskeleton. In neurons, microtubules are organized differently between axons and dendrites, but their precise organization in different compartments is not completely understood. Super-resoln. microscopy techniques can detect specific structures at an increased resoln., but the narrow spacing between neuronal microtubules poses challenges because most existing labeling strategies increase the effective microtubule diam. by 20-40 nm and will thereby blend neighboring microtubules into one structure. Here we develop single-chain antibody fragments (nanobodies) against tubulin to achieve super-resoln. imaging of microtubules with a decreased apparent diam. To test the resolving power of these novel probes, we generate microtubule bundles with a known spacing of 50-70 nm and successfully resolve individual microtubules. Individual bundled microtubules can also be resolved in different mammalian cells, including hippocampal neurons, allowing novel insights into fundamental mechanisms of microtubule organization in cell- and neurobiol.
- 9Ries, J.; Kaplan, C.; Platonova, E.; Eghlidi, H.; Ewers, H. A Simple, Versatile Method for GFP-Based Super-Resolution Microscopy via Nanobodies. Nat. Methods 2012, 9 (6), 582– 584, DOI: 10.1038/nmeth.1991[Crossref], [PubMed], [CAS], Google Scholar9https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC38Xmt1Gksbs%253D&md5=678f32bb3f032e786fb8ca42d893030aA simple, versatile method for GFP-based super-resolution microscopy via nanobodiesRies, Jonas; Kaplan, Charlotte; Platonova, Evgenia; Eghlidi, Hadi; Ewers, HelgeNature Methods (2012), 9 (6), 582-584CODEN: NMAEA3; ISSN:1548-7091. (Nature Publishing Group)We developed a method to use any GFP-tagged construct in single-mol. super-resoln. microscopy. By targeting GFP with small, high-affinity antibodies coupled to org. dyes, we achieved nanometer spatial resoln. and minimal linkage error when analyzing microtubules, living neurons and yeast cells. We show that in combination with libraries encoding GFP-tagged proteins, virtually any known protein can immediately be used in super-resoln. microscopy and that simplified labeling schemes allow high-throughput super-resoln. imaging.
- 10Opazo, F.; Levy, M.; Byrom, M.; Schäfer, C.; Geisler, C.; Groemer, T. W.; Ellington, A. D.; Rizzoli, S. O. Aptamers as Potential Tools for Super-Resolution Microscopy. Nat. Methods 2012, 9 (10), 938– 939, DOI: 10.1038/nmeth.2179[Crossref], [PubMed], [CAS], Google Scholar10https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC38XhsVWntb%252FE&md5=854e68813111f273803bc0b79db829d4Aptamers as potential tools for super-resolution microscopyOpazo, Felipe; Levy, Matthew; Byrom, Michelle; Schaefer, Christina; Geisler, Claudia; Groemer, Teja W.; Ellington, Andrew D.; Rizzoli, Silvio O.Nature Methods (2012), 9 (10), 938-939CODEN: NMAEA3; ISSN:1548-7091. (Nature Publishing Group)This article shows that aptamers, which are single-stranded DNA or RNA oligonucleotides, are potential probes that would complement nanobodies for super-resoln. imaging. Three aptamers were tested for mols. involved in endosomal trafficking: the transferrin receptor (TfnR; aptamer size ∼15 kDa), the prostate-specific membrane antigen (PSMA; ∼15 kDa) and the epidermal growth factor receptor (EGFR; ∼36 kDa). Epitope recognition with aptamers was higher than with conventional antibodies, and this allowed the aptamers to provide superior super-resoln. images, just as the nanobodies did, when investigated using stimulated emission depletion (STED) microscopy. The high epitope recognition also permitted rapid live super-resoln. imaging.
- 11Agarwal, P.; Bertozzi, C. R. Site-Specific Antibody-Drug Conjugates: The Nexus of Bioorthogonal Chemistry, Protein Engineering, and Drug Development. Bioconjugate Chem. 2015, 26 (2), 176– 192, DOI: 10.1021/bc5004982[ACS Full Text
], [CAS], Google Scholar
11https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2cXitFCrsrfK&md5=c3d822b0e53e5cdb074e5b63b8060ccfSite-Specific Antibody-Drug Conjugates: The Nexus of Bioorthogonal Chemistry, Protein Engineering, and Drug DevelopmentAgarwal, Paresh; Bertozzi, Carolyn R.Bioconjugate Chemistry (2015), 26 (2), 176-192CODEN: BCCHES; ISSN:1043-1802. (American Chemical Society)A review. Antibody-drug conjugates (ADCs) combine the specificity of antibodies with the potency of small mols. to create targeted drugs. Despite the simplicity of this concept, generation of clin. successful ADCs has been very difficult. Over the past several decades, scientists have learned a great deal about the constraints on antibodies, linkers, and drugs as they relate to successful construction of ADCs. Once these components are in hand, most ADCs are prepd. by nonspecific modification of antibody lysine or cysteine residues with drug-linker reagents, which results in heterogeneous product mixts. that cannot be further purified. With advances in the fields of bioorthogonal chem. and protein engineering, there is growing interest in producing ADCs by site-specific conjugation to the antibody, yielding more homogeneous products that have demonstrated benefits over their heterogeneous counterparts in vivo. Here, we chronicle the development of a multitude of site-specific conjugation strategies for assembly of ADCs and provide a comprehensive account of key advances and their roots in the fields of bioorthogonal chem. and protein engineering. - 12Beck, A.; Goetsch, L.; Dumontet, C.; Corvaïa, N. Strategies and Challenges for the Next Generation of Antibody–Drug Conjugates. Nat. Rev. Drug Discovery 2017, 16 (5), 315– 337, DOI: 10.1038/nrd.2016.268[Crossref], [PubMed], [CAS], Google Scholar12https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2sXkt1Onsbc%253D&md5=7358b6080dd1aed1d21e98c078cd9ef1Strategies and challenges for the next generation of antibody-drug conjugatesBeck, Alain; Goetsch, Liliane; Dumontet, Charles; Corvaia, NathalieNature Reviews Drug Discovery (2017), 16 (5), 315-337CODEN: NRDDAG; ISSN:1474-1776. (Nature Publishing Group)A review. Antibody-drug conjugates (ADCs) are one of the fastest growing classes of oncol. therapeutics. After half a century of research, the approvals of brentuximab vedotin (in 2011) and trastuzumab emtansine (in 2013) have paved the way for ongoing clin. trials that are evaluating more than 60 further ADC candidates. The limited success of first-generation ADCs (developed in the early 2000s) informed strategies to bring second-generation ADCs to the market, which have higher levels of cytotoxic drug conjugation, lower levels of naked antibodies and more-stable linkers between the drug and the antibody. Furthermore, lessons learned during the past decade are now being used in the development of third-generation ADCs. In this Review, we discuss strategies to select the best target antigens as well as suitable cytotoxic drugs; the design of optimized linkers; the discovery of bioorthogonal conjugation chemistries; and toxicity issues. The selection and engineering of antibodies for site-specific drug conjugation, which will result in higher homogeneity and increased stability, as well as the quest for new conjugation chemistries and mechanisms of action, are priorities in ADC research.
- 13Li, X.; Fang, T.; Boons, G. J. Preparation of Well-Defined Antibody-Drug Conjugates through Glycan Remodeling and Strain-Promoted Azide-Alkyne Cycloadditions. Angew. Chem., Int. Ed. 2014, 53 (28), 7179– 7182, DOI: 10.1002/anie.201402606[Crossref], [CAS], Google Scholar13https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2cXos1aks7w%253D&md5=d59a17169fb0891a63e88960285c4f55Preparation of well-defined antibody-drug conjugates through glycan remodeling and strain-promoted azide-alkyne cycloadditionsLi, Xiuru; Fang, Tao; Boons, Geert-JanAngewandte Chemie, International Edition (2014), 53 (28), 7179-7182CODEN: ACIEF5; ISSN:1433-7851. (Wiley-VCH Verlag GmbH & Co. KGaA)Antibody-drug conjugates hold considerable promise as anticancer agents, however, producing them remains a challenge and there is a need for mild, broadly applicable, site-specific conjugation methods that yield homogenous products. It was envisaged that enzymic remodeling of the oligosaccharides of an antibody would enable the introduction of reactive groups that can be exploited for the site-specific attachment of cytotoxic drugs. This is based on the observation that glycosyltransferases often tolerate chem. modifications in their sugar nucleotide substrates, thus allowing the installation of reactive functionalities. An azide was incorporated because this functional group is virtually absent in biol. systems and can be reacted by strain-promoted alkyne-azide cycloaddn. This method, which does not require genetic engineering, was used to produce an anti-CD22 antibody modified with doxorubicin to selectively target and kill lymphoma cells.
- 14Van Geel, R.; Wijdeven, M. A.; Heesbeen, R.; Verkade, J. M. M.; Wasiel, A. A.; Van Berkel, S. S.; Van Delft, F. L. Chemoenzymatic Conjugation of Toxic Payloads to the Globally Conserved N-Glycan of Native mAbs Provides Homogeneous and Highly Efficacious Antibody-Drug Conjugates. Bioconjugate Chem. 2015, 26 (11), 2233– 2242, DOI: 10.1021/acs.bioconjchem.5b00224[ACS Full Text
], [CAS], Google Scholar
14https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2MXhtVShtLjL&md5=3473e419ded9df111414a8788bcdce43Chemoenzymatic conjugation of toxic payloads to the globally conserved N-glycan of native mAbs provides homogeneous and highly efficacious antibody-drug conjugatesvan Geel, Remon; Wijdeven, Marloes A.; Heesbeen, Ryan; Verkade, Jorge M. M.; Wasiel, Anna A.; van Berkel, Sander S.; van Delft, Floris L.Bioconjugate Chemistry (2015), 26 (11), 2233-2242CODEN: BCCHES; ISSN:1043-1802. (American Chemical Society)A robust, generally applicable, nongenetic technol. is presented to convert monoclonal antibodies into stable and homogeneous ADCs. Starting from a native (nonengineered) mAb, a chemoenzymic protocol allows for the highly controlled attachment of any given payload to the N-glycan residing at asparagine-297, based on a two-stage process: first, enzymic remodeling (trimming and tagging with azide), followed by ligation of the payload based on copper-free click chem. The technol., termed GlycoConnect, is applicable to any IgG isotype irresp. of glycosylation profile. Application to trastuzumab and maytansine, both components of the marketed ADC Kadcyla, demonstrate a favorable in vitro and in vivo efficacy for GlycoConnect ADC. Moreover, the superiority of the native glycan as attachment site was demonstrated by in vivo comparison to a range of trastuzumab-based glycosylation mutants. A side-by-side comparison of the copper-free click probes bicyclononyne (BCN) and a dibenzoannulated cyclooctyne (DBCO) showed a surprising difference in conjugation efficiency in favor of BCN, which could be even further enhanced by introduction of electron-withdrawing fluoride substitutions onto the azide. The resulting mAb-conjugates were in all cases found to be highly stable, which in combination with the demonstrated efficacy warrants ADCs with a superior therapeutic index. - 15Rosen, C. B.; Kodal, A. L. B.; Nielsen, J. S.; Schaffert, D. H.; Scavenius, C.; Okholm, A. H.; Voigt, N. V.; Enghild, J. J.; Kjems, J.; Tørring, T.; Gothelf, K. V. Template-Directed Covalent Conjugation of DNA to Native Antibodies, Transferrin and Other Metal-Binding Proteins. Nat. Chem. 2014, 6 (9), 804– 809, DOI: 10.1038/nchem.2003[Crossref], [PubMed], [CAS], Google Scholar15https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2cXhtFygs7bL&md5=8a402d92b83e7d605e841cba7fb72b27Template-directed covalent conjugation of DNA to native antibodies, transferrin and other metal-binding proteinsRosen, Christian B.; Kodal, Anne L. B.; Nielsen, Jesper S.; Schaffert, David H.; Scavenius, Carsten; Okholm, Anders H.; Voigt, Niels V.; Enghild, Jan J.; Kjems, Joergen; Toerring, Thomas; Gothelf, Kurt V.Nature Chemistry (2014), 6 (9), 804-809CODEN: NCAHBB; ISSN:1755-4330. (Nature Publishing Group)DNA-protein conjugates are important in bioanal. chem., mol. diagnostics and bionanotechnol., as the DNA provides a unique handle to identify, functionalize or otherwise manipulate proteins. To maintain protein activity, conjugation of a single DNA handle to a specific location on the protein is often needed. However, prepg. such high-quality site-specific conjugates often requires genetically engineered proteins, which is a laborious and tech. challenging approach. Here we demonstrate a simpler method to create site-selective DNA-protein conjugates. Using a guiding DNA strand modified with a metal-binding functionality, we directed a second DNA strand to the vicinity of a metal-binding site of His6-tagged or wild-type metal-binding proteins, such as serotransferrin, where it subsequently reacted with lysine residues at that site. This method, DNA-templated protein conjugation, facilitates the prodn. of site-selective protein conjugates, and also conjugation to IgG1 antibodies via a histidine cluster in the const. domain.
- 16Kline, T.; Steiner, A. R.; Penta, K.; Sato, A. K.; Hallam, T. J.; Yin, G. Methods to Make Homogenous Antibody Drug Conjugates. Pharm. Res. 2015, 32 (11), 3480– 3493, DOI: 10.1007/s11095-014-1596-8[Crossref], [PubMed], [CAS], Google Scholar16https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2cXitFCru7vO&md5=3e933915d0c26eac721d100dbcf28f08Methods to Make Homogenous Antibody Drug ConjugatesKline, Toni; Steiner, Alexander R.; Penta, Kalyani; Sato, Aaron K.; Hallam, Trevor J.; Yin, GangPharmaceutical Research (2015), 32 (11), 3480-3493CODEN: PHREEB; ISSN:0724-8741. (Springer)Antibody drug conjugates (ADCs) have progressed from hypothesis to approved therapeutics in less than 30 years, and the technologies available to modify both the antibodies and the cytotoxic drugs are expanding rapidly. For reasons well reviewed previously, the field is trending strongly toward homogeneous, defined antibody conjugation. In this review we present the antibody and small mol. chemistries that are currently used and being explored to develop specific, homogeneous ADCs.
- 17Jeger, S.; Zimmermann, K.; Blanc, A.; Grünberg, J.; Honer, M.; Hunziker, P.; Struthers, H.; Schibli, R. Site-Specific and Stoichiometric Modification of Antibodies by Bacterial Transglutaminase. Angew. Chem., Int. Ed. 2010, 49 (51), 9995– 9997, DOI: 10.1002/anie.201004243[Crossref], [CAS], Google Scholar17https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3cXhsFGhsLnI&md5=6c2bad043af729dcfa3a7b27a122b98fSite-Specific and Stoichiometric Modification of Antibodies by Bacterial TransglutaminaseJeger, Simone; Zimmermann, Kurt; Blanc, Alain; Gruenberg, Juergen; Honer, Michael; Hunziker, Peter; Struthers, Harriet; Schibli, RogerAngewandte Chemie, International Edition (2010), 49 (51), 9995-9997, S9995/1-S9995/46CODEN: ACIEF5; ISSN:1433-7851. (Wiley-VCH Verlag GmbH & Co. KGaA)We have shown that enzymic modification of mAbs using bacterial transglutaminase (BTG) is site-specific and versatile (with the potential to be readily scaled up), and leads to homogeneous immunoconjugates with defined stoichiometries. The in vivo characteristics of such immunoconjugates are superior to those prepd. using chem. coupling methods. Since position 295 is located in the const. Fc region, the enzymic conjugation approach is applicable not only to other human IgG1s, but also to mAbs belonging to subtypes IgG2, IgG3, and IgG4, all of which conserve the Q295 residue. Thus, the method is broadly applicable and permits the systematic assessment and improvement of immunoconjugates.
- 18Dennler, P.; Chiotellis, A.; Fischer, E.; Brégeon, D.; Belmant, C.; Gauthier, L.; Lhospice, F.; Romagne, F.; Schibli, R. Transglutaminase-Based Chemo-Enzymatic Conjugation Approach Yields Homogeneous Antibody–Drug Conjugates. Bioconjugate Chem. 2014, 25 (3), 569– 578, DOI: 10.1021/bc400574z[ACS Full Text
], [CAS], Google Scholar
18https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2cXhs1GntLs%253D&md5=7d70123f2e5cea5bbda003eb0c0d55e0Transglutaminase-Based Chemo-Enzymatic Conjugation Approach Yields Homogeneous Antibody-Drug ConjugatesDennler, Patrick; Chiotellis, Aristeidis; Fischer, Eliane; Bregeon, Delphine; Belmant, Christian; Gauthier, Laurent; Lhospice, Florence; Romagne, Francois; Schibli, RogerBioconjugate Chemistry (2014), 25 (3), 569-578CODEN: BCCHES; ISSN:1043-1802. (American Chemical Society)Most chem. techniques used to produce antibody-drug conjugates (ADCs) result in a heterogeneous mixt. of species with variable drug-to-antibody ratios (DAR) which will potentially display different pharmacokinetics, stability, and safety profiles. Here we investigated two strategies to obtain homogeneous ADCs based on site-specific modification of deglycosylated antibodies by microbial transglutaminase (MTGase), which forms isopeptidic bonds between Gln and Lys residues. We have previously shown that MTGase solely recognizes Gln295 within the heavy chain of IgGs as a substrate and can therefore be exploited to generate ADCs with an exact DAR of 2. The first strategy included the direct, one-step attachment of the antimitotic toxin monomethyl auristatin E (MMAE) to the antibody via different spacer entities with a primary amine functionality that is recognized as a substrate by MTGase. The second strategy was a chemo-enzymic, two-step approach whereby a reactive spacer entity comprising a bio-orthogonal thiol or azide function was attached to the antibody by MTGase and subsequently reacted with a suitable MMAE-deriv. To this aim, we investigated two different chem. approaches, namely, thiol-maleimide and strain-promoted azide-alkyne cycloaddn. (SPAAC). Direct enzymic attachment of MMAE-spacer derivs. at an 80 M excess of drug yielded heterogeneous ADCs with a DAR of between 1.0 to 1.6. In contrast to this, the chemo-enzymic approach only required a 2.5 M excess of toxin to yield homogeneous ADCs with a DAR of 2.0 in the case of SPAAC and 1.8 for the thiol-maleimide approach. As a proof-of-concept, trastuzumab (Herceptin) was armed with the MMAE via the chemo-enzymic approach using SPAAC and tested in vitro. Trastuzumab-MMAE efficiently killed BT-474 and SK-BR-3 cells with an IC50 of 89.0 pM and 21.7 pM, resp. Thus, the chemo-enzymic approach using MTGase is an elegant strategy to form ADCs with a defined DAR of 2. Furthermore, the approach is directly applicable to a broad variety of antibodies as it does not require prior genetic modifications of the antibody sequence. - 19Agard, N. J.; Prescher, J. A.; Bertozzi, C. R. A Strain-Promoted [3 + 2] Azide-Alkyne Cycloaddition for Covalent Modification of Biomolecules in Living Systems. J. Am. Chem. Soc. 2004, 126 (46), 15046– 15047, DOI: 10.1021/ja044996f[ACS Full Text
], [CAS], Google Scholar
19https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD2cXpt1Sks7s%253D&md5=37af3dbaa89ae4cffaba2dee30e50ec0A strain-promoted [3+2] azide-alkyne cycloaddition for covalent modification of biomolecules in living systemsAgard, Nicholas J.; Prescher, Jennifer A.; Bertozzi, Carolyn R.Journal of the American Chemical Society (2004), 126 (46), 15046-15047CODEN: JACSAT; ISSN:0002-7863. (American Chemical Society)Selective chem. reactions that are orthogonal to the diverse functionality of biol. systems have become important tools in the field of chem. biol. Two notable examples are the Staudinger ligation of azides and phosphines and the Cu(I)-catalyzed [3+2] cycloaddn. of azides and alkynes ("click chem."). The Staudinger ligation has sufficient biocompatibility for performance in living animals but suffers from phosphine oxidn. and synthetic challenges. Click chem. obviates the requirement of phosphines, but the Cu(I) catalyst is toxic to cells, thereby precluding in vivo applications. Here we present a strain-promoted [3+2] cycloaddn. between cyclooctynes and azides that proceeds under physiol. conditions without the need for a catalyst. The utility of the reaction was demonstrated by selective modification of biomols. in vitro and on living cells, with no apparent toxicity. - 20Helmerich, D. A.; Beliu, G.; Sauer, M. Multiple-Labeled Antibodies Behave Like Single Emitters in Photoswitching Buffer. ACS Nano 2020, 14 (10), 12629– 12641, DOI: 10.1021/acsnano.0c06099[ACS Full Text
], [CAS], Google Scholar
20https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BB38fmtl2ltw%253D%253D&md5=8510150e6bd05409d8c6b812609171f7Multiple-Labeled Antibodies Behave Like Single Emitters in Photoswitching BufferHelmerich Dominic A; Beliu Gerti; Sauer MarkusACS nano (2020), 14 (10), 12629-12641 ISSN:.The degree of labeling (DOL) of antibodies has so far been optimized for high brightness and specific and efficient binding. The influence of the DOL on the blinking performance of antibodies used in direct stochastic optical reconstruction microscopy (dSTORM) has so far attained limited attention. Here, we investigated the spectroscopic characteristics of IgG antibodies labeled at DOLs of 1.1-8.3 with Alexa Fluor 647 (Al647) at the ensemble and single-molecule level. Multiple-Al647-labeled antibodies showed weak and strong quenching interactions in aqueous buffer but could all be used for dSTORM imaging with spatial resolutions of ∼20 nm independent of the DOL. Single-molecule fluorescence trajectories and photon antibunching experiments revealed that individual multiple-Al647-labeled antibodies show complex photophysics in aqueous buffer but behave as single emitters in photoswitching buffer independent of the DOL. We developed a model that explains the observed blinking of multiple-labeled antibodies and can be used for the development of improved fluorescent probes for dSTORM experiments. - 21Pleiner, T.; Bates, M.; Görlich, D. A Toolbox of Anti-Mouse and Anti-Rabbit IgG Secondary Nanobodies. J. Cell Biol. 2018, 217 (3), 1143– 1154, DOI: 10.1083/jcb.201709115[Crossref], [PubMed], [CAS], Google Scholar21https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1cXhslWmsbvF&md5=47efb96d0aeb73ae07eaafad355a0277A toolbox of anti-mouse and anti-rabbit IgG secondary nanobodiesPleiner, Tino; Bates, Mark; Goerlich, DirkJournal of Cell Biology (2018), 217 (3), 1143-1159CODEN: JCLBA3; ISSN:1540-8140. (Rockefeller University Press)Polyclonal anti-IgG (anti-IgG) secondary antibodies are essential tools for many mol. biol. techniques and diagnostic tests. Their animal-based prodn. is, however, a major ethical problem. Here, we introduce a sustainable alternative, namely nanobodies against all mouse IgG subclasses and rabbit IgG. They can be produced at large scale in Escherichia coli and could thus make secondary antibody prodn. in animals obsolete. Their recombinant nature allows fusion with affinity tags or reporter enzymes as well as efficient maleimide chem. for fluorophore coupling. We demonstrate their superior performance in Western blotting, in both peroxidase- and fluorophore-linked form. Their site-specific labeling with multiple fluorophores creates bright imaging reagents for confocal and superresoln. microscopy with much smaller label displacement than traditional secondary antibodies. They also enable simpler and faster immunostaining protocols, and allow multitarget localization with primary IgGs from the same species and of the same class.
- 22Vaughan, J. C.; Dempsey, G. T.; Sun, E.; Zhuang, X. Phosphine-Quenching of Cyanine Dyes as a Versatile Tool for Fluorescence Microscopy. J. Am. Chem. Soc. 2013, 135, 1197– 1200, DOI: 10.1021/ja3105279[ACS Full Text
], [CAS], Google Scholar
22https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3sXmvF2rug%253D%253D&md5=163fc5e390c184b71edd202f2d80ea7ePhosphine Quenching of Cyanine Dyes as a Versatile Tool for Fluorescence MicroscopyVaughan, Joshua C.; Dempsey, Graham T.; Sun, Eileen; Zhuang, XiaoweiJournal of the American Chemical Society (2013), 135 (4), 1197-1200CODEN: JACSAT; ISSN:0002-7863. (American Chemical Society)The authors report that the cyanine dye Cy5 and several of its structural relatives are reversibly quenched by the phosphine tris(2-carboxyethyl)phosphine (TCEP). Using Cy5 as a model, the quenching reaction occurs by 1,4-addn. of the phosphine to the polymethine bridge of Cy5 to form a covalent adduct. Illumination with UV light dissocs. the adduct and returns the dye to the fluorescent state. TCEP quenching can be used for super-resoln. imaging as well as for other applications, such as differentiating between mols. inside and outside the cell. - 23Olivier, N.; Keller, D.; Gönczy, P.; Manley, S. Resolution Doubling in 3D-STORM Imaging through Improved Buffers. PLoS One 2013, 8 (7), 1– 9, DOI: 10.1371/journal.pone.0069004
- 24Endesfelder, U.; Malkusch, S.; Fricke, F.; Heilemann, M. A Simple Method to Estimate the Average Localization Precision of a Single-Molecule Localization Microscopy Experiment. Histochem. Cell Biol. 2014, 141 (6), 629– 638, DOI: 10.1007/s00418-014-1192-3[Crossref], [PubMed], [CAS], Google Scholar24https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2cXisFWhs7Y%253D&md5=a804a9d46ee82438cbe478fa25034140A simple method to estimate the average localization precision of a single-molecule localization microscopy experimentEndesfelder, Ulrike; Malkusch, Sebastian; Fricke, Franziska; Heilemann, MikeHistochemistry and Cell Biology (2014), 141 (6), 629-638CODEN: HCBIFP; ISSN:0948-6143. (Springer)The localization precision is a crucial and important parameter for single-mol. localization microscopy (SMLM) and directly influences the achievable spatial resoln. It primarily depends on exptl. imaging conditions and the registration potency of the algorithm used. We propose a new and simple routine to est. the av. exptl. localization precision in SMLM, based on the nearest neighbor anal. By exploring different exptl. and simulated targets, we show that this approach can be generally used for any 2D or 3D SMLM data and that reliable values for the localization precision σSMLM are obtained. Knowing σSMLM is a prerequisite for consistent visualization or any quant. structural anal., e.g., cluster anal. or colocalization studies.
- 25Li, Y.; Mund, M.; Hoess, P.; Deschamps, J.; Matti, U.; Nijmeijer, B.; Sabinina, V. J.; Ellenberg, J.; Schoen, I.; Ries, J. Real-Time 3D Single-Molecule Localization Using Experimental Point Spread Functions. Nat. Methods 2018, 15 (5), 367– 369, DOI: 10.1038/nmeth.4661[Crossref], [PubMed], [CAS], Google Scholar25https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1cXnt1Sns7c%253D&md5=c58c4361da24656c77566dd8ee4ea120Real-time 3D single-molecule localization using experimental point spread functionsLi, Yiming; Mund, Markus; Hoess, Philipp; Deschamps, Joran; Matti, Ulf; Nijmeijer, Bianca; Sabinina, Vilma Jimenez; Ellenberg, Jan; Schoen, Ingmar; Ries, JonasNature Methods (2018), 15 (5), 367-369CODEN: NMAEA3; ISSN:1548-7091. (Nature Research)We present a real-time fitter for 3D single-mol. localization microscopy using exptl. point spread functions (PSFs) that achieves minimal uncertainty in 3D on any microscope and is compatible with any PSF engineering approach. We used this method to image cellular structures and attained unprecedented image quality for astigmatic PSFs. The fitter compensates for most optical aberrations and makes accurate 3D super-resoln. microscopy broadly accessible, even on std. microscopes without dedicated 3D optics.
- 26Auer, A.; Schlichthaerle, T.; Woehrstein, J. B.; Schueder, F.; Strauss, M. T.; Grabmayr, H.; Jungmann, R. Nanometer-Scale Multiplexed Super-Resolution Imaging with an Economic 3D-DNA-PAINT Microscope. ChemPhysChem 2018, 19 (22), 3024– 3034, DOI: 10.1002/cphc.201800630[Crossref], [PubMed], [CAS], Google Scholar26https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1cXhvVGltLzM&md5=de5d6cc8edf56158bd04e01fee92015bNanometer-scale Multiplexed Super-Resolution Imaging with an Economic 3D-DNA-PAINT MicroscopeAuer, Alexander; Schlichthaerle, Thomas; Woehrstein, Johannes B.; Schueder, Florian; Strauss, Maximilian T.; Grabmayr, Heinrich; Jungmann, RalfChemPhysChem (2018), 19 (22), 3024-3034CODEN: CPCHFT; ISSN:1439-4235. (Wiley-VCH Verlag GmbH & Co. KGaA)Optical super-resoln. microscopy is rapidly changing the way imaging studies in the biol. and biomedical sciences are conducted. Due to the unique capability of achieving mol. contrast using fluorescent labels and sub-diffraction resoln. down to a few tens of nanometers, super-resoln. is developing as an attractive imaging modality. While the increased spatial resoln. has already enabled structural studies at unprecedented mol. detail, the wide-spread use of super-resoln. approaches as a std. characterization technique in biol. labs. has thus far been prevented by mainly two issues: (1) Intricate sample prepn. and image acquisition and (2) costly and complex instrumentation. We here introduce a combination of the recently developed super-resoln. technique DNA-PAINT (DNA points accumulation for imaging in nanoscale topog.) with an easy-to-replicate, custom-built 3D single-mol. microscope (termed liteTIRF) that is an order of magnitude more economic in cost compared to most com. systems. We assay the performance of our system using synthetic two- and three-dimensional DNA origami structures and show the applicability to single- and multiplexed cellular imaging.
- 27Kellogg, E. H.; Hejab, N. M. A.; Howes, S.; Northcote, P.; Miller, J. H.; Díaz, J. F.; Downing, K. H.; Nogales, E. Insights into the Distinct Mechanisms of Action of Taxane and Non-Taxane Microtubule Stabilizers from Cryo-EM Structures. J. Mol. Biol. 2017, 429 (5), 633– 646, DOI: 10.1016/j.jmb.2017.01.001[Crossref], [PubMed], [CAS], Google Scholar27https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2sXhtFOgurk%253D&md5=2fc94e61ef199ff77990dda297020180Insights into the Distinct Mechanisms of Action of Taxane and Non-Taxane Microtubule Stabilizers from Cryo-EM StructuresKellogg, Elizabeth H.; Hejab, Nisreen M. A.; Howes, Stuart; Northcote, Peter; Miller, John H.; Diaz, J. Fernando; Downing, Kenneth H.; Nogales, EvaJournal of Molecular Biology (2017), 429 (5), 633-646CODEN: JMOBAK; ISSN:0022-2836. (Elsevier Ltd.)A no. of microtubule (MT)-stabilizing agents (MSAs) have demonstrated or predicted potential as anticancer agents, but a detailed structural basis for their mechanism of action is still lacking. We have obtained high-resoln. (3.9-4.2 Å) cryo-electron microscopy (cryo-EM) reconstructions of MTs stabilized by the taxane-site binders Taxol and zampanolide, and by peloruside, which targets a distinct, non-taxoid pocket on β-tubulin. We find that each mol. has unique distinct structural effects on the MT lattice structure. Peloruside acts primarily at lateral contacts and has an effect on the "seam" of heterologous interactions, enforcing a conformation more similar to that of homologous (i.e., non-seam) contacts by which it regularizes the MT lattice. In contrast, binding of either Taxol or zampanolide induces MT heterogeneity. In doubly bound MTs, peloruside overrides the heterogeneity induced by Taxol binding. Our structural anal. illustrates distinct mechanisms of these drugs for stabilizing the MT lattice and is of relevance to the possible use of combinations of MSAs to regulate MT activity and improve therapeutic potential.
- 28Brandt, J. P.; Patapoff, T. W.; Aragon, S. R. Construction, MD Simulation, and Hydrodynamic Validation of an All-Atom Model of a Monoclonal IgG Antibody. Biophys. J. 2010, 99 (3), 905– 913, DOI: 10.1016/j.bpj.2010.05.003[Crossref], [PubMed], [CAS], Google Scholar28https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3cXpvFWms7s%253D&md5=de25c2118b7897401ff71b6c0516cce3Construction, MD Simulation, and Hydrodynamic Validation of an All-Atom Model of a Monoclonal IgG AntibodyBrandt, J. Paul; Patapoff, Thomas W.; Aragon, Sergio R.Biophysical Journal (2010), 99 (3), 905-913CODEN: BIOJAU; ISSN:0006-3495. (Cell Press)At 150 kDa, antibodies of the IgG class are too large for their structure to be detd. with current NMR methodologies. Because of hinge-region flexibility, it is difficult to obtain at.-level structural information from the crystal, and questions regarding antibody structure and dynamics in soln. remain unaddressed. Here we describe the construction of a model of a human IgG1 monoclonal antibody (trastuzumab) from the crystal structures of fragments. We use a combination of mol.-dynamics (MD) simulation, continuum hydrodynamics modeling, and exptl. diffusion measurements to explore antibody behavior in aq. soln. Hydrodynamic modeling provides a link between the at.-level details of MD simulation and the size- and shape-dependent data provided by hydrodynamic measurements. Eight independent 40 ns MD trajectories were obtained with the AMBER program suite. The ensemble av. of the computed transport properties over all of the MD trajectories agrees remarkably well with the value of the translational diffusion coeff. obtained with dynamic light scattering at 20°C and 27°C, and the intrinsic viscosity measured at 20°C. Therefore, our MD results likely represent a realistic sampling of the conformational space that an antibody explores in aq. soln.
- 29De Michele, C.; De Los Rios, P.; Foffi, G.; Piazza, F. Simulation and Theory of Antibody Binding to Crowded Antigen-Covered Surfaces. PLoS Comput. Biol. 2016, 12 (3), 1– 17, DOI: 10.1371/journal.pcbi.1004752
- 30Harris, L. J.; Larson, S. B.; Hasel, K. W.; McPherson, A. Refined Structure of an Intact IgG2a Monoclonal Antibody. Biochemistry 1997, 36 (7), 1581– 1597, DOI: 10.1021/bi962514+[ACS Full Text
], [CAS], Google Scholar
30https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADyaK2sXht1Ohs78%253D&md5=762b87a6094c1cf0eb698314f64ebfafRefined Structure of an Intact IgG2a Monoclonal AntibodyHarris, Lisa J.; Larson, Steven B.; Hasel, Karl W.; McPherson, AlexanderBiochemistry (1997), 36 (7), 1581-1597CODEN: BICHAW; ISSN:0006-2960. (American Chemical Society)The structure of an intact, anti-canine lymphoma monoclonal antibody (Mab231) was detd. by mol. replacement and refined in a triclinic cell to an R-value of 20.9%, using synchrotron diffraction data from 2.8 to 20 Å resoln. All segments of the antibody, including the hinge region and carbohydrate component, are visible in electron d. maps. There is no overall symmetry to the antibody, as the Fc is disposed in an entirely oblique manner with respect to the Fabs. The CH2 and CH3 domains do, however, possess a nearly exact, local 2-fold relation. The Fab segments are related by a second, independent, local dyad axis, exact only with respect to const. domains. Variable domains exhibit no symmetry relation as a consequence of the 16° difference in Fab elbow angles. Variable domain pair assocns. VL:VH for the Fabs are virtually the same, and corresponding CDRs of the 2 Fabs also are nearly identical in structure. CDR-H3 displays the greatest difference. Hypervariable loops of both Fabs are involved in contacts with symmetry-related Fc segments at the CH2-CH3 switch junction, suggesting a "complex" structure. The hinge segment connecting Fabs with the Fc is quite extended and exhibits thermal factors indicative of a high degree of mobility. It consists of a well-defined upper hinge that partially maintains dyad symmetry and a fairly rigid core bounded above and below by fluid polypeptides that provide segmental flexibility. This structure represents the first visualization by x-ray anal. of a murine Fc segment, and its CH2 domains exhibit substantial rigid body conformational changes with respect to the human Fc used as an initial mol. replacement model. The oligosaccharides were found by difference Fourier syntheses to be very similar to those of the free human Fc fragment, although differences are present in the terminal residues. The detailed structure of the IgG presented here, and the distribution of effector binding sites, appears consistent with effector activation mechanisms involving translocation and/or aggregation of the Fc following antigen binding by the Fabs. - 31Bongini, L.; Fanelli, D.; Piazza, F.; De Los Rios, P.; Sandin, S.; Skoglund, U. Freezing Immunoglobulins to See Them Move. Proc. Natl. Acad. Sci. U. S. A. 2004, 101 (17), 6466– 6471, DOI: 10.1073/pnas.0400119101[Crossref], [PubMed], [CAS], Google Scholar31https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD2cXjvVyitLg%253D&md5=78a91c90a7605ec67627cd5ae249a50cFreezing immunoglobulins to see them moveBongini, L.; Fanelli, D.; Piazza, F.; De Los Rios, P.; Sandin, S.; Skoglund, U.Proceedings of the National Academy of Sciences of the United States of America (2004), 101 (17), 6466-6471CODEN: PNASA6; ISSN:0027-8424. (National Academy of Sciences)The issue of protein dynamics and its implications in the biol. function of proteins are arousing greater and greater interest in different fields of mol. biol. In cryo-electron tomog. expts. one may take several snapshots of a given biol. macromol. In principle, a large enough collection of snapshots of the mol. may then be used to calc. its equil. configuration in terms of the exptl. accessible degrees of freedom and, hence, to est. its potential energy. This information would be crucial in order to analyze the biol. functions of biomols. by directly accessing the relevant dynamical indicators. In this article, we analyze the results of cryo-electron tomog. expts. performed on monoclonal murine IgG2a antibodies. We measure the equil. distribution of the mol. in terms of the relevant angular coordinates and build a mech. model of the antibody dynamics. This approach enables us to derive an explicit expression of the IgG potential energy. Furthermore, we discuss the configuration space at equil. in relation to results from other techniques, and we set our discussion in the context of the current debate regarding conformation and flexibility of antibodies.
- 32Dani, A.; Huang, B.; Bergan, J.; Dulac, C.; Zhuang, X. Superresolution Imaging of Chemical Synapses in the Brain. Neuron 2010, 68 (5), 843– 856, DOI: 10.1016/j.neuron.2010.11.021[Crossref], [PubMed], [CAS], Google Scholar32https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3cXhsFGhu73I&md5=07c554f9890e13d6767bc4205c3001f6Superresolution Imaging of Chemical Synapses in the BrainDani, Adish; Huang, Bo; Bergan, Joseph; Dulac, Catherine; Zhuang, Xiao-WeiNeuron (2010), 68 (5), 843-856CODEN: NERNET; ISSN:0896-6273. (Cell Press)Detn. of the mol. architecture of synapses requires nanoscopic image resoln. and specific mol. recognition, a task that has so far defied many conventional imaging approaches. Here, we present a superresoln. fluorescence imaging method to visualize the mol. architecture of synapses in the brain. Using multicolor, three-dimensional stochastic optical reconstruction microscopy, the distributions of synaptic proteins can be measured with nanometer precision. Furthermore, the wide-field, volumetric imaging method enables high-throughput, quant. anal. of a large no. of synapses from different brain regions. To demonstrate the capabilities of this approach, we have detd. the organization of ten protein components of the presynaptic active zone and the postsynaptic d. Variations in synapse morphol., neurotransmitter receptor compn., and receptor distribution were obsd. both among synapses and across different brain regions. Combination with optogenetics further allowed mol. events assocd. with synaptic plasticity to be resolved at the single-synapse level.
- 33Sehnal, D.; Rose, A. S.; Koča, J.; Burley, S. K.; Velankar, S. Mol*: Towards a Common Library and Tools for Web Molecular Graphics. In Workshop on Molecular Graphics and Visual Analysis of Molecular Data; Byska, J., Krone, M., Sommer, B., Eds.; The Eurographics Association, 2018; pp 29– 33 DOI: 10.2312/molva.20181103 .
- 34Zheng, K.; Bantog, C.; Bayer, R. The Impact of Glycosylation on Monoclonal Antibody Conformation and Stability. mAbs 2011, 3 (6), 568– 576, DOI: 10.4161/mabs.3.6.17922[Crossref], [PubMed], [CAS], Google Scholar34https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BC383htFGquw%253D%253D&md5=7a337ca1fb8a8e12a30b3d1a679f9849The impact of glycosylation on monoclonal antibody conformation and stabilityZheng Kai; Bantog Christopher; Bayer RobertmAbs (2011), 3 (6), 568-76 ISSN:.Antibody glycosylation is a common post-translational modification and has a critical role in antibody effector function. The use of glycoengineering to produce antibodies with specific glycoforms may be required to achieve the desired therapeutic efficacy. However, the modified molecule could have unusual behavior during development due to the alteration of its intrinsic properties and stability. In this study, we focused on the differences between glycosylated and deglycosylated antibodies, as aglycosyl antibodies are often chosen when effector function is not desired or unimportant. We selected three human IgG1 antibodies and used PNGase F to remove their oligosaccharide chains. Although there were no detected secondary or tertiary structural changes after deglycosylation, other intrinsic properties of the antibody were altered with the removal of oligosaccharide chains in the Fc region. The apparent molecular hydrodynamic radius increased after deglycosylation based on size-exclusion chromatography analysis. Deglycosylated antibodies exhibited less thermal stability for the CH2 domain and less resistance to GdnHCl induced unfolding. Susceptibility to proteolytic cleavage demonstrated that the deglycosylated version was more susceptible to papain. An accelerated stability study revealed that deglycosylated antibodies had higher aggregation rates. These changes may impact the development of aglycosyl antibody biotherapeutics.
- 35Mimura, Y.; Church, S.; Ghirlando, R.; Ashton, P. R.; Dong, S.; Goodall, M.; Lund, J.; Jefferis, R. The Influence of Glycosylation on the Thermal Stability and Effector Function Expression of Human IgG1-Fc: Properties of a Series of Truncated Glycoforms. Mol. Immunol. 2000, 37 (12–13), 697– 706, DOI: 10.1016/S0161-5890(00)00105-X[Crossref], [PubMed], [CAS], Google Scholar35https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BD3M7os1Srsw%253D%253D&md5=07eed4544a1af8d14f58c52d6a625a88The influence of glycosylation on the thermal stability and effector function expression of human IgG1-Fc: properties of a series of truncated glycoformsMimura Y; Church S; Ghirlando R; Ashton P R; Dong S; Goodall M; Lund J; Jefferis RMolecular immunology (2000), 37 (12-13), 697-706 ISSN:0161-5890.Antibodies are multifunctional molecules that following the formation of antibody antigen complexes, may activate mechanisms to effect the clearance and destruction of the antigen (pathogen). The IgG molecule is comprised of three globular protein moieties (2Fab+Fc) linked through a flexible hinge region. While the Fabs bind antigens, the Fc triggers effector mechanisms through interactions with specific ligands, e.g. cellular receptors (FcgammaR), and the C1 component of complement. Glycosylation of IgG-Fc has been shown to be essential for efficient activation of FcgammaR and C1. We report the generation of a series of truncated glycoforms of IgG-Fc, and the analysis of the contribution of the residual oligosaccharide to IgG-Fc function and thermal stability. Differential scanning microcalorimetry has been used to compare the stabilities of the homogeneous glycoforms of IgG1-Fc. The results show that all truncated oligosaccharides confer a degree of functional activity, and thermodynamic stability to the IgG1-Fc, in comparison with deglycosylated IgG1-Fc. The same truncated glycoforms of an intact IgG1 anti-MHC Class II antibody are shown to exhibit differential functional activity for FcgammaRI and C1 ligands, relative to deglycosylated IgG1. The minimal glycoform investigated had a trisaccharide attached to each heavy chain and can be expected to influence protein structure primarily in the proximity of the N-terminal region of the C(H)2 domain, implicated as a binding site for multiple effector ligands. These data provide a thermodynamic rationale for the modulation of antibody effector functions by different glycoforms.
- 36Szenczi, Á.; Kardos, J.; Medgyesi, G. A.; Závodszky, P. The Effect of Solvent Environment on the Conformation and Stability of Human Polyclonal IgG in Solution. Biologicals 2006, 34, 5– 14, DOI: 10.1016/j.biologicals.2005.06.007[Crossref], [PubMed], [CAS], Google Scholar36https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD28Xhslylsrs%253D&md5=f17aff81c43bc676ceb4fedc3e1ed7e1The effect of solvent environment on the conformation and stability of human polyclonal IgG in solutionSzenczi, Arpad; Kardos, Jozsef; Medgyesi, Gyoergy A.; Zavodszky, PeterBiologicals (2006), 34 (1), 5-14CODEN: BILSEC; ISSN:1045-1056. (Elsevier B.V.)Stability of therapeutic IgG prepns. is an important issue as adequate efficacy and safety has to be ensured throughout a long shelf life. To this end, denaturation and aggregation have to be avoided. In many cases sugars are applied for stabilizing IgG in relatively high concn. (5-10%). However, certain sugars (sucrose, maltose) are responsible for adverse effects including renal failure. In this work we reassessed the effect of pH and stabilizers to optimize the solvent environment and minimize the amt. of additives without endangering quality and stability. Since both biol. function and aggregation depend on the conformational properties of individual IgG mols., two sensitive and rapid phys. methods were introduced to assess conformational changes and structural stability as a function of pH and addn. of std. stabilizers. It was obsd. that the conformational stability decreases with decreasing pH, while the resistance against aggregation improves. The optimum pH range for storage is 5.0-6.0, as a compromise between conformational stability and the tendency for oligomerization. Intriguingly, additives in physiol. acceptable concn. have no effect on the thermal stability of IgG. On the other hand, glucose or sorbitol, even at a concn. as low as 1%, have significant effect on the tertiary structure as revealed by near-UV-CD spectroscopy, reflecting changes in the environment of arom. side-chains. Although, 0.3% leucine does not increase conformational stability, it decreases the aggregation tendency even more efficiently than 1% glucose or sorbitol. Both pH and storage temp. are decisive factors for the long-term stability of IgG solns. An increase in the dimer content was obsd. upon storage at 5°C which was partly reverted upon incubation at 37°C. Storage at temps. higher than 5°C may help to maintain an optimal proportion of dimers. Regarding the known side effects, and their limited stabilizing capacity at low concn., it is advisable to omit sugars at i.v. Ig (IVIG) formulation. Hydrophobic amino acids give promising alternatives.
- 37Vermeer, A. W. P.; Norde, W. The Thermal Stability of Immunoglobulin: Unfolding and Aggregation of a Multi-Domain Protein. Biophys. J. 2000, 78 (1), 394– 404, DOI: 10.1016/S0006-3495(00)76602-1[Crossref], [PubMed], [CAS], Google Scholar37https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD3cXktFeqsQ%253D%253D&md5=3664753139bb8ca404f4c73051cc2adeThe thermal stability of immunoglobulin: unfolding and aggregation of a multi-domain proteinVermeer, Arnoldus W. P.; Norde, WillemBiophysical Journal (2000), 78 (1), 394-404CODEN: BIOJAU; ISSN:0006-3495. (Biophysical Society)The denaturation of IgG was studied by different calorimetric methods and CD spectroscopy. The thermogram of the Ig showed two main transitions that are a superimposition of distinct denaturation steps. It was shown that the two transitions have different sensitivities to changes in temp. and pH. The two peaks represent the Fab and Fc fragments of the IgG mol. The Fab fragment is most sensitive to heat treatment, whereas the Fc fragment is most sensitive to decreasing pH. The transitions were independent, and the unfolding was immediately followed by an irreversible aggregation step. Below the unfolding temp., the unfolding is the rate-detg. step in the overall denaturation process. At higher temps. where a relatively high concn. of (partially) unfolded IgG mols. is present, the rate of aggregation is so fast that IgG mols. become locked in aggregates before they are completely denatured. Furthermore, the structure of the aggregates formed depends on the denaturation method. The CD spectrum of the IgG is also strongly affected by both heat treatment and low pH treatment. It was shown that a strong correlation exists between the denaturation transitions as obsd. by calorimetry and the changes in secondary structure derived from CD. After both heat- and low-pH-induced denaturation, a significant fraction of the secondary structure remains.
- 38Ries, J. SMAP: A Modular Super-Resolution Microscopy Analysis Platform for SMLM Data. Nat. Methods 2020, 17 (9), 870– 872, DOI: 10.1038/s41592-020-0938-1[Crossref], [PubMed], [CAS], Google Scholar38https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3cXhs1Cjtb%252FE&md5=55783f1f207c22674a59a0f60004011eSMAP: a modular super-resolution microscopy analysis platform for SMLM dataRies, JonasNature Methods (2020), 17 (9), 870-872CODEN: NMAEA3; ISSN:1548-7091. (Nature Research)There is no expanded citation for this reference.
Supporting Information
Supporting Information
ARTICLE SECTIONSThe Supporting Information is available free of charge at https://pubs.acs.org/doi/10.1021/acsnano.1c03677.
Experimental methods on mass spectrometry, SMLM setups, SMLM imaging, the determination of residual experimental errors, and circular dichroism spectroscopy measurements; Supplementary Figures S1 to S8; Supplementary Tables S1 to S6 (PDF)
Terms & Conditions
Most electronic Supporting Information files are available without a subscription to ACS Web Editions. Such files may be downloaded by article for research use (if there is a public use license linked to the relevant article, that license may permit other uses). Permission may be obtained from ACS for other uses through requests via the RightsLink permission system: http://pubs.acs.org/page/copyright/permissions.html.