1
|
Loreau V, Koolhaas WH, Chan EH, De Boissier P, Brouilly N, Avosani S, Sane A, Pitaval C, Reiter S, Luis NM, Mangeol P, von Philipsborn AC, Rupprecht JF, Görlich D, Habermann BH, Schnorrer F. Titin-dependent biomechanical feedback tailors sarcomeres to specialized muscle functions in insects. SCIENCE ADVANCES 2025; 11:eads8716. [PMID: 40344069 PMCID: PMC12063666 DOI: 10.1126/sciadv.ads8716] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/01/2024] [Accepted: 04/03/2025] [Indexed: 05/11/2025]
Abstract
Sarcomeres are the universal contractile units of muscles that enable animals to move. Insect muscles display a remarkable functional diversity: they operate at extremely different contraction frequencies (ranging from ~1 to 1000 hertz) and amplitudes during flying, walking, and crawling. This is puzzling because sarcomeres are built from essentially the same actin-myosin components. Here, we address how functionally different sarcomeres are made. We show that the giant protein titin and the regulation of developmental contractility are key for the sarcomere specializations. I-band titin spans and determines the length of the sarcomeric I-band in a muscle type-specific manner. Unexpectedly, I-band titin also rules the length of the force-generating myosin filament using a feedback mechanism that is modulated by myosin contractility. We propose a model of how sarcomere specializations in insects are tuned, provide evidence for this model, and discuss its validity beyond insects.
Collapse
Affiliation(s)
- Vincent Loreau
- Aix Marseille University, CNRS, IBDM, Turing Centre for Living Systems, Marseille, France
| | | | - Eunice HoYee Chan
- Aix Marseille University, CNRS, IBDM, Turing Centre for Living Systems, Marseille, France
| | - Paul De Boissier
- Aix Marseille University, CNRS, IBDM, Turing Centre for Living Systems, Marseille, France
| | - Nicolas Brouilly
- Aix Marseille University, CNRS, IBDM, Turing Centre for Living Systems, Marseille, France
| | - Sabina Avosani
- Department of Neuroscience and Movement Science, Medicine Section, University of Fribourg, Fribourg, Switzerland
| | - Aditya Sane
- Aix Marseille University, CNRS, IBDM, Turing Centre for Living Systems, Marseille, France
| | - Christophe Pitaval
- Aix Marseille University, CNRS, IBDM, Turing Centre for Living Systems, Marseille, France
| | - Stefanie Reiter
- Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
| | - Nuno Miguel Luis
- Aix Marseille University, CNRS, IBDM, Turing Centre for Living Systems, Marseille, France
| | - Pierre Mangeol
- Aix Marseille University, CNRS, IBDM, Turing Centre for Living Systems, Marseille, France
| | - Anne C. von Philipsborn
- Department of Neuroscience and Movement Science, Medicine Section, University of Fribourg, Fribourg, Switzerland
| | | | - Dirk Görlich
- Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
| | - Bianca H. Habermann
- Aix Marseille University, CNRS, IBDM, Turing Centre for Living Systems, Marseille, France
| | - Frank Schnorrer
- Aix Marseille University, CNRS, IBDM, Turing Centre for Living Systems, Marseille, France
- Max Planck Institute of Biochemistry, Martinsried, Germany
| |
Collapse
|
2
|
Maltseva M, Rossotti MA, Tanha J, Langlois MA. Characterization of Nanobody Binding to Distinct Regions of the SARS-CoV-2 Spike Protein by Flow Virometry. Viruses 2025; 17:571. [PMID: 40285013 PMCID: PMC12030927 DOI: 10.3390/v17040571] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2025] [Revised: 03/31/2025] [Accepted: 04/10/2025] [Indexed: 04/29/2025] Open
Abstract
Nanobodies, or single-domain antibodies (VHHs) from camelid heavy-chain-only antibodies, offer significant advantages in therapeutic and diagnostic applications due to their small size and ability to bind cryptic protein epitopes inaccessible to conventional antibodies. In this study, we examined nanobodies specific to regions of the SARS-CoV-2 spike glycoprotein, including the receptor-binding domain (RBD), N-terminal domain (NTD), and subunit 2 (S2). Using flow virometry, a high-throughput technique for viral quantification, we achieved the efficient detection of pseudotyped viruses expressing the spike glycoprotein. RBD-targeting nanobodies showed the most effective staining, followed by NTD-targeting ones, while S2-specific nanobodies exhibited limited resolution. The simple genetic structure of nanobodies enables the creation of multimeric formats, improving binding specificity and avidity. Bivalent VHH-Fc constructs (VHHs fused to the Fc region of human IgG) outperformed monovalent formats in resolving viral particles from background noise. However, S2-specific monovalent VHHs demonstrated improved staining efficiency, suggesting their smaller size better accesses restricted antigenic sites. Furthermore, direct staining of cell supernatants was possible without virus purification. This versatile nanobody platform, initially developed for antiviral therapy against SARS-CoV-2, can be readily adapted for flow virometry applications and other diagnostic assays.
Collapse
Affiliation(s)
- Mariam Maltseva
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, ON K1H 8M5, Canada; (M.M.); (J.T.)
| | - Martin A. Rossotti
- Human Health Therapeutics Research Centre, Life Sciences Division, National Research Council Canada, Ottawa, ON K1N 1J1, Canada;
| | - Jamshid Tanha
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, ON K1H 8M5, Canada; (M.M.); (J.T.)
- Human Health Therapeutics Research Centre, Life Sciences Division, National Research Council Canada, Ottawa, ON K1N 1J1, Canada;
- uOttawa Center for Infection, Immunity, and Inflammation (CI3), Ottawa, ON K1H 8L1, Canada
| | - Marc-André Langlois
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, ON K1H 8M5, Canada; (M.M.); (J.T.)
- uOttawa Center for Infection, Immunity, and Inflammation (CI3), Ottawa, ON K1H 8L1, Canada
| |
Collapse
|
3
|
de Zwaan K, Huo R, Hensgens MN, Wienecke HL, Tekpınar M, Geertsema H, Grußmayer K. High-Throughput Single-Molecule Microscopy with Adaptable Spatial Resolution Using Exchangeable Oligonucleotide Labels. ACS NANO 2025; 19:13149-13159. [PMID: 40145776 PMCID: PMC11984304 DOI: 10.1021/acsnano.4c18502] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/20/2024] [Revised: 03/13/2025] [Accepted: 03/14/2025] [Indexed: 03/28/2025]
Abstract
Super-resolution microscopy facilitates the visualization of cellular structures at a resolution approaching the molecular level. Especially, super-resolution techniques based on the localization of single molecules have relatively modest instrument requirements and are thus good candidates for adoption in bioimaging. However, their low-throughput nature hampers their applicability in biomolecular research and screening. Here, we propose a workflow for more efficient data collection, starting with the scanning of large areas using fast fluctuation-based imaging, followed by single-molecule localization microscopy of selected cells. To achieve this workflow, we exploit the versatility of DNA oligo hybridization kinetics with DNA-PAINT probes to tailor the fluorescent blinking toward high-throughput and high-resolution imaging. Additionally, we employ super-resolution optical fluctuation imaging (SOFI) to analyze statistical fluctuations in the DNA-PAINT binding kinetics, thereby tolerating much denser blinking and facilitating accelerated imaging speeds. Thus, we demonstrate 30-300-fold faster imaging of different cellular structures compared to conventional DNA-PAINT imaging, albeit at a lower resolution. Notably, by tuning the image medium and data processing though, we can flexibly switch between high-throughput SOFI (scanning an FOV of 0.65 mm × 0.52 mm within 4 min of total acquisition time) and super-resolution DNA-PAINT microscopy and thereby demonstrate that combining DNA-PAINT and SOFI enables one to adapt image resolution and acquisition time based on the imaging needs. We envision this approach to be especially powerful when combined with multiplexing and 3D imaging.
Collapse
Affiliation(s)
- Klarinda de Zwaan
- Department
of Bionanoscience, Kavli Institute of Nanoscience Delft, Delft University of Technology, 2629 HZ Delft, The Netherlands
| | - Ran Huo
- Department
of Bionanoscience, Kavli Institute of Nanoscience Delft, Delft University of Technology, 2629 HZ Delft, The Netherlands
| | - Myron N.F. Hensgens
- Department
of Imaging Physics, Delft University of
Technology, 2628 CJ Delft, The
Netherlands
| | - Hannah Lena Wienecke
- Department
of Bionanoscience, Kavli Institute of Nanoscience Delft, Delft University of Technology, 2629 HZ Delft, The Netherlands
| | - Miyase Tekpınar
- Department
of Bionanoscience, Kavli Institute of Nanoscience Delft, Delft University of Technology, 2629 HZ Delft, The Netherlands
| | - Hylkje Geertsema
- Department
of Imaging Physics, Delft University of
Technology, 2628 CJ Delft, The
Netherlands
| | - Kristin Grußmayer
- Department
of Bionanoscience, Kavli Institute of Nanoscience Delft, Delft University of Technology, 2629 HZ Delft, The Netherlands
| |
Collapse
|
4
|
Tröster V, Wong RP, Börgel A, Cakilkaya B, Renz C, Möckel MM, Eifler-Olivi K, Marinho J, Reinberg T, Furler S, Schaefer JV, Plückthun A, Wolf E, Ulrich HD. Custom affinity probes reveal DNA-damage-induced, ssDNA-independent chromatin SUMOylation in budding yeast. Cell Rep 2025; 44:115353. [PMID: 40019834 DOI: 10.1016/j.celrep.2025.115353] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2024] [Revised: 12/17/2024] [Accepted: 02/05/2025] [Indexed: 03/29/2025] Open
Abstract
The small ubiquitin-related modifier SUMO regulates cellular processes in eukaryotes either by modulating individual protein-protein interactions or with relaxed substrate selectivity by group modification. Here, we report the isolation and characterization of designed ankyrin repeat protein (DARPin)-based affinity probes directed against budding yeast SUMO (Smt3). We validate selected DARPins as compartment-specific inhibitors or neutral detection agents. Structural characterization reveals a recognition mode distinct from that of natural SUMO interactors. In vivo application pinpoints Smt3's essential function to the nucleus and demonstrates DARPin-mediated sensitization toward various stress conditions. A subset of selected clones is validated as SUMOylation reporters in cells. In this manner, we identify a DNA-damage-induced nuclear SUMOylation response that-in contrast to previously reported chromatin group SUMOylation-is independent of single-stranded DNA and the SUMO-E3 Siz2 but depends on Mms21 and likely reflects late intermediates of homologous recombination. Thus, Smt3-specific DARPins can provide insight into the dynamics of SUMOylation in defined subcellular structures.
Collapse
Affiliation(s)
- Vera Tröster
- Institute of Molecular Biology (IMB) gGmbH, 55128 Mainz, Germany
| | - Ronald P Wong
- Institute of Molecular Biology (IMB) gGmbH, 55128 Mainz, Germany
| | - Arne Börgel
- Institute of Molecular Physiology, Johannes Gutenberg University, 55128 Mainz, Germany
| | - Baris Cakilkaya
- Institute of Molecular Physiology, Johannes Gutenberg University, 55128 Mainz, Germany
| | - Christian Renz
- Institute of Molecular Biology (IMB) gGmbH, 55128 Mainz, Germany
| | - Martin M Möckel
- Institute of Molecular Biology (IMB) gGmbH, 55128 Mainz, Germany
| | | | - Joana Marinho
- Department of Biochemistry, University of Zurich, 8057 Zurich, Switzerland
| | - Thomas Reinberg
- Department of Biochemistry, University of Zurich, 8057 Zurich, Switzerland
| | - Sven Furler
- Department of Biochemistry, University of Zurich, 8057 Zurich, Switzerland
| | - Jonas V Schaefer
- Department of Biochemistry, University of Zurich, 8057 Zurich, Switzerland
| | - Andreas Plückthun
- Department of Biochemistry, University of Zurich, 8057 Zurich, Switzerland
| | - Eva Wolf
- Institute of Molecular Physiology, Johannes Gutenberg University, 55128 Mainz, Germany
| | - Helle D Ulrich
- Institute of Molecular Biology (IMB) gGmbH, 55128 Mainz, Germany.
| |
Collapse
|
5
|
Sescil J, Havens SM, Wang W. Principles and Design of Molecular Tools for Sensing and Perturbing Cell Surface Receptor Activity. Chem Rev 2025; 125:2665-2702. [PMID: 39999110 PMCID: PMC11934152 DOI: 10.1021/acs.chemrev.4c00582] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/27/2025]
Abstract
Cell-surface receptors are vital for controlling numerous cellular processes with their dysregulation being linked to disease states. Therefore, it is necessary to develop tools to study receptors and the signaling pathways they control. This Review broadly describes molecular approaches that enable 1) the visualization of receptors to determine their localization and distribution; 2) sensing receptor activation with permanent readouts as well as readouts in real time; and 3) perturbing receptor activity and mimicking receptor-controlled processes to learn more about these processes. Together, these tools have provided valuable insight into fundamental receptor biology and helped to characterize therapeutics that target receptors.
Collapse
Affiliation(s)
- Jennifer Sescil
- Department of Chemistry, University of Michigan, Ann Arbor,
MI, 48109
- Life Sciences Institute, University of Michigan, Ann Arbor,
MI, 48109
| | - Steven M. Havens
- Department of Chemistry, University of Michigan, Ann Arbor,
MI, 48109
- Life Sciences Institute, University of Michigan, Ann Arbor,
MI, 48109
| | - Wenjing Wang
- Department of Chemistry, University of Michigan, Ann Arbor,
MI, 48109
- Life Sciences Institute, University of Michigan, Ann Arbor,
MI, 48109
- Neuroscience Graduate Program, University of Michigan, Ann
Arbor, MI, 48109
- Program in Chemical Biology, University of Michigan, Ann
Arbor, MI, 48109
| |
Collapse
|
6
|
Martinez-Orengo N, Shah S, Lai J, Basuli F, Lyndaker A, Turner ML, Peiravi M, Sourabh S, Sampson K, Zhang P, Swenson RE, Lusso P, Maldarelli F, Nath A, Lau CY, Hammoud DA. PET imaging of HIV-1 envelope protein gp120 using 18F-labeled nanobodies. iScience 2025; 28:111795. [PMID: 39917021 PMCID: PMC11800091 DOI: 10.1016/j.isci.2025.111795] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2024] [Revised: 11/02/2024] [Accepted: 01/09/2025] [Indexed: 02/09/2025] Open
Abstract
Radiolabeled antibodies against the HIV-1 envelope protein, gp120, have been previously tested in animal models and in people with HIV (PWH). Nanobodies offer advantages over antibodies, including smaller size and faster clearance, which allow labeling with fluorine-18. In this study, three nanobodies (J3, 3E3, B9) chosen based on their binding properties to the conserved CD4-binding site of gp120 were labeled with fluorine-18 and used for PET imaging in mice bearing wild-type (WT) and/or gp120-expressing (Env+) tumors. [18F]J3 and [18F]3E3 selectively targeted Env+ tumors and not WT tumors, with minimal background signal. Switching from non-site-specific radiolabeling method to sortase A-mediated site-specific conjugation at the C-terminus improved binding to Env+ tumors for all nanobodies. Site-specifically 18F-labeled J3 nanobody is the most promising candidate with the highest level of binding. These results establish an Env+ imaging method that will enable next stage testing in an HIV-1 preclinical infection model and potentially in PWH.
Collapse
Affiliation(s)
- Neysha Martinez-Orengo
- Center for Infectious Disease Imaging (CIDI), Radiology and Imaging Sciences, Clinical Center (CC), National Institutes of Health, Bethesda, MD, USA
| | - Swati Shah
- Center for Infectious Disease Imaging (CIDI), Radiology and Imaging Sciences, Clinical Center (CC), National Institutes of Health, Bethesda, MD, USA
| | - Jianhao Lai
- Center for Infectious Disease Imaging (CIDI), Radiology and Imaging Sciences, Clinical Center (CC), National Institutes of Health, Bethesda, MD, USA
| | - Falguni Basuli
- Chemistry and Synthesis Center, National Heart, Lung, and Blood Institute (NHLBI), National Institutes of Health, Rockville, MD, USA
| | - Anna Lyndaker
- Center for Infectious Disease Imaging (CIDI), Radiology and Imaging Sciences, Clinical Center (CC), National Institutes of Health, Bethesda, MD, USA
| | - Mitchell L. Turner
- Center for Infectious Disease Imaging (CIDI), Radiology and Imaging Sciences, Clinical Center (CC), National Institutes of Health, Bethesda, MD, USA
| | - Morteza Peiravi
- Center for Infectious Disease Imaging (CIDI), Radiology and Imaging Sciences, Clinical Center (CC), National Institutes of Health, Bethesda, MD, USA
| | - Suman Sourabh
- Center for Infectious Disease Imaging (CIDI), Radiology and Imaging Sciences, Clinical Center (CC), National Institutes of Health, Bethesda, MD, USA
| | - Kevon Sampson
- Division of Neuroimmunology and Neurovirology, National Institute of Neurological Disorders and Stroke (NINDS), National Institutes of Health, Bethesda, MD, USA
| | - Peng Zhang
- Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health, Bethesda, MD, USA
| | - Rolf E. Swenson
- Chemistry and Synthesis Center, National Heart, Lung, and Blood Institute (NHLBI), National Institutes of Health, Rockville, MD, USA
| | - Paolo Lusso
- Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health, Bethesda, MD, USA
| | - Frank Maldarelli
- HIV Dynamics and Replication Program, Center for Cancer Research, National Cancer Institute (NCI), National Institutes of Health, Bethesda, MD, USA
| | - Avindra Nath
- Division of Neuroimmunology and Neurovirology, National Institute of Neurological Disorders and Stroke (NINDS), National Institutes of Health, Bethesda, MD, USA
| | - Chuen-Yen Lau
- HIV Dynamics and Replication Program, Center for Cancer Research, National Cancer Institute (NCI), National Institutes of Health, Bethesda, MD, USA
| | - Dima A. Hammoud
- Center for Infectious Disease Imaging (CIDI), Radiology and Imaging Sciences, Clinical Center (CC), National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
7
|
Al-Seragi M, Chen Y, Duong van Hoa F. Advances in nanobody multimerization and multispecificity: from in vivo assembly to in vitro production. Biochem Soc Trans 2025; 53:BST20241419. [PMID: 39927832 DOI: 10.1042/bst20241419] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2024] [Revised: 01/08/2025] [Accepted: 01/10/2025] [Indexed: 02/11/2025]
Abstract
NANOBODIES® (Nbs) have emerged as valuable tools across therapeutic, diagnostic, and industrial applications owing to their small size and consequent ability to bind unique epitopes inaccessible to conventional antibodies. While Nbs retrieved from immune libraries normally possess sufficient affinity and specificity for their cognate antigens in the practical use case, their multimerization will often increase functional affinity via avidity effects. Therefore, to rescue binding affinity and broaden targeting specificities, recent efforts have focused on conjugating multiple Nb clones - of identical or unique antigen cognates - together. In vivo and in vitro approaches, including flexible linkers, antibody domains, self-assembling coiled coils, chemical conjugation, and self-clustering hydrophobic sequences, have been employed to produce multivalent and multispecific Nb constructs. Examples of successful Nb multimerization are diverse, ranging from immunoassaying reagents to virus-neutralizing moieties. This review aims to recapitulate the in vivo and in vitro modalities to produce multivalent and multispecific Nbs while highlighting the applications, advantages, and drawbacks tied to each method.
Collapse
Affiliation(s)
- Mohammed Al-Seragi
- Department of Biochemistry and Molecular Biology, Life Sciences Institute, University of British Columbia, Vancouver, British Columbia, Canada
| | - Yilun Chen
- Department of Biochemistry and Molecular Biology, Life Sciences Institute, University of British Columbia, Vancouver, British Columbia, Canada
| | - Franck Duong van Hoa
- Department of Biochemistry and Molecular Biology, Life Sciences Institute, University of British Columbia, Vancouver, British Columbia, Canada
| |
Collapse
|
8
|
Boonkaew S, Teodori L, Vendelbo MH, Kjems J, Ferapontova EE. Nanobodies' duo facilitates ultrasensitive serum HER-2/neu immunoassays via enhanced avidity interactions. Anal Chim Acta 2025; 1335:343472. [PMID: 39643321 DOI: 10.1016/j.aca.2024.343472] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2024] [Revised: 10/28/2024] [Accepted: 11/21/2024] [Indexed: 12/09/2024]
Abstract
BACKGROUND Existing liquid biopsy assays for protein biomarkers of cancer are mostly based on antibodies (Ab) contributing unfavorably to their high cost. Easy to express and modify in vitro, nanobodies may be a cost-effective alternative to Ab. RESULTS We show that serum HER-2/neu, a biomarker and target of aggressive HER-2/neu(+) cancers, can be accurately detected in a 1.2 h electrochemical cellulase-linked sandwich nanobody/aptamer assay on magnetic beads. Using a single nanobody receptor, 2Rs15d or 2Rb17c, reduces immunoassay's sensitivity by 35%-26 %. A combination of two nanobodies as a duo-receptor recovers the sensitivity of the enzyme-linked nanobody/aptamer-sorbent assay (ELNASA) to 11.9 ± 2.8 μC fM-1, due to the avidity effects making the nanobodies-duo binding properties comparable to those of Ab. Down to 0.1 fM HER-2/neu was detected by ELNASA in serum samples, with no interference from other blood-circulating proteins. In a 30 healthy-volunteers trial, ELNASA more accurately than optical ELISA assayed serum HER-2/neu. SIGNIFICANCE ELNASA performance rivals that of ELISA, yet estimated to be at least 200 times cheaper, due to the lower cost of nanobodies production, and may be better suited for routine clinical analysis of HER-2/neu, particularly, in low- and middle-income settings with limited resources. The ELNASA approach is generic and may be adapted for specific and ultrasensitive analysis of other blood-circulating proteins.
Collapse
Affiliation(s)
- Suchanat Boonkaew
- Interdisciplinary Nanoscience Center (iNANO), Faculty of Natural Sciences, Aarhus University, Gustav Wieds Vej 14, 8000, Aarhus C, Denmark
| | - Laura Teodori
- Interdisciplinary Nanoscience Center (iNANO), Faculty of Natural Sciences, Aarhus University, Gustav Wieds Vej 14, 8000, Aarhus C, Denmark
| | - Mikkel H Vendelbo
- Department of Nuclear Medicine & PET Centre, Aarhus University Hospital, Palle Juul-Jensens Boulevard 99, DK-8200, Aarhus N, Denmark; Department of Biomedicine, Aarhus University, Høegh-Guldbergs Gade 10, 8000, Aarhus C, Denmark
| | - Jørgen Kjems
- Interdisciplinary Nanoscience Center (iNANO), Faculty of Natural Sciences, Aarhus University, Gustav Wieds Vej 14, 8000, Aarhus C, Denmark; Department of Molecular Biology and Genetics, Aarhus University, Universitetsbyen 81, 8000 Aarhus C, Denmark
| | - Elena E Ferapontova
- Interdisciplinary Nanoscience Center (iNANO), Faculty of Natural Sciences, Aarhus University, Gustav Wieds Vej 14, 8000, Aarhus C, Denmark.
| |
Collapse
|
9
|
Wang J, Aceves AJ, Friesenhahn NJ, Mayo SL. CDRxAbs: antibody small-molecule conjugates with computationally designed target-binding synergy. Protein Eng Des Sel 2025; 38:gzaf004. [PMID: 40114302 DOI: 10.1093/protein/gzaf004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Revised: 02/05/2025] [Accepted: 02/17/2025] [Indexed: 03/22/2025] Open
Abstract
Bioconjugates as therapeutic modalities combine the advantages and offset the disadvantages of their constituent parts to achieve a refined spectrum of action. We combine the concept of bioconjugation with the full atomic simulation capability of computational protein design to define a new class of molecular recognition agents: CDR-extended antibodies, abbreviated as CDRxAbs. A CDRxAb incorporates a covalently attached small molecule into an antibody/target binding interface using computational protein design to create an antibody small-molecule conjugate that binds tighter to the target of the small molecule than the small molecule would alone. CDRxAbs are also expected to increase the target binding specificity of their associated small molecules. In a proof-of-concept study using monomeric streptavidin/biotin pairs at either a nanomolar or micromolar-level initial affinity, we designed nanobody-biotin conjugates that exhibited >20-fold affinity improvement against their protein targets with step-wise optimization of binding kinetics and overall protein stability. The workflow explored through this process promises a novel approach to optimize small-molecule based therapeutics and to explore new chemical and target space for molecular-recognition agents in general.
Collapse
Affiliation(s)
- Jingzhou Wang
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA 91125, United States
- Division of Chemistry and Chemical Engineering, California Institute of Technology, Pasadena, CA 91125, United States
- Merck Research Laboratories, 213 E Grand Ave, South San Francisco, CA 94080, USA
| | - Aiden J Aceves
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA 91125, United States
- Insight Partners, 1114 Avenue of the Americas, 36th Floor, New York, NY 10036, USA
| | - Nicholas J Friesenhahn
- Division of Chemistry and Chemical Engineering, California Institute of Technology, Pasadena, CA 91125, United States
| | - Stephen L Mayo
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA 91125, United States
- Division of Chemistry and Chemical Engineering, California Institute of Technology, Pasadena, CA 91125, United States
| |
Collapse
|
10
|
Ayrton JP, Ho C, Zhang H, Chudasama V, Frank S, Thomas MR. Multivalent nanobody engineering for enhanced physisorption and functional display on gold nanoparticles. NANOSCALE 2024; 16:19881-19896. [PMID: 39382227 DOI: 10.1039/d4nr02762k] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/10/2024]
Abstract
The ease of expression and engineering of single domain antibodies, known as nanobodies, make them attractive alternatives to conventional antibodies in point-of-care diagnostics such as lateral flow assays. In lateral flow assays, gold nanoparticle bioconjugates serve as labels which display affinity molecules on the gold surface. While examples of nanobody gold nanoparticle bioconjugates exist, few utilise the simple one-step approach of physisorption owing to undesirable nanoparticle aggregation and loss of functionality. Here we show that engineering nanobodies into multivalent structures can significantly enhance their functionality when physisorbed onto gold nanoparticles. This approach enables resulting bioconjugates to withstand multiple processing steps required for long-term nanoparticle storage within lateral flow assays. Specifically, we show that the trivalent version of VHHV nanobody (VHH3) against the S1 protein of SARS-CoV-2 can be immobilised onto gold nanoparticles through passive adsorption. Unlike its monovalent and bivalent nanobody counterparts, using VHHV3 preserves nanoparticle stability under salt stress, blocking, washing, and freeze-drying conditions while maintaining picomolar sensitivity to the S1 protein. We anticipate that this facile strategy is a significant advancement towards the integration of nanobodies in lateral flow assay development.
Collapse
Affiliation(s)
- John-Paul Ayrton
- London Centre for Nanotechnology, University College London, London, UK.
- Department of Biochemical Engineering, University College London, London, UK.
| | - Chapman Ho
- London Centre for Nanotechnology, University College London, London, UK.
- Department of Biochemical Engineering, University College London, London, UK.
| | - Haoran Zhang
- Department of Biochemical Engineering, University College London, London, UK.
| | - Vijay Chudasama
- Department of Chemistry, University College London, London, UK
| | - Stefanie Frank
- Department of Biochemical Engineering, University College London, London, UK.
| | - Michael R Thomas
- London Centre for Nanotechnology, University College London, London, UK.
- Department of Biochemical Engineering, University College London, London, UK.
| |
Collapse
|
11
|
Xu P, Zhang Y, Guo J, Li H, Konrath S, Zhou P, Cai L, Rao H, Chen H, Lin J, Cui Z, Ji B, Wang J, Li N, Liu DP, Renné T, Wang M. A single-domain antibody targeting factor XII inhibits both thrombosis and inflammation. Nat Commun 2024; 15:7898. [PMID: 39266545 PMCID: PMC11393108 DOI: 10.1038/s41467-024-51745-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2020] [Accepted: 08/16/2024] [Indexed: 09/14/2024] Open
Abstract
Factor XII (FXII) is the zymogen of the plasma protease FXIIa that activates the intrinsic coagulation pathway and the kallikrein kinin-system. The role of FXII in inflammation has been obscure. Here, we report a single-domain antibody (nanobody, Nb) fused to the Fc region of a human immunoglobulin (Nb-Fc) that recognizes FXII in a conformation-dependent manner and interferes with FXIIa formation. Nb-Fc treatment inhibited arterial thrombosis in male mice without affecting hemostasis. In a mouse model of extracorporeal membrane oxygenation (ECMO), FXII inhibition or knockout reduced thrombus deposition on oxygenator membranes and systemic microvascular thrombi. ECMO increased circulating levels of D-dimer, alkaline phosphatase, creatinine and TNF-α and triggered microvascular neutrophil adherence, platelet aggregation and their interaction, which were substantially attenuated by FXII blockade. Both Nb-Fc treatment and FXII knockout markedly ameliorated immune complex-induced local vasculitis and anti-neutrophil cytoplasmic antibody-induced systemic vasculitis, consistent with selectively suppressed neutrophil migration. In human blood microfluidic analysis, Nb-Fc treatment prevented collagen-induced fibrin deposition and neutrophil adhesion/activation. Thus, FXII is an important mediator of inflammatory responses in vasculitis and ECMO, and Nb-Fc provides a promising approach to alleviate thrombo-inflammatory disorders.
Collapse
Affiliation(s)
- Pengfei Xu
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yingjie Zhang
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Junyan Guo
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- College of Life Science, Zhejiang Normal University, Jinhua, Zhejiang, China
| | - Huihui Li
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Sandra Konrath
- Institute of Clinical Chemistry and Laboratory Medicine, University Medical Center Hamburg-Eppendorf (UKE), Hamburg, Germany
| | - Peng Zhou
- Synthetic and Functional Biomolecules Center, Beijing National Laboratory for Molecular Sciences, College of Chemistry and Molecular Engineering, Peking University, Beijing, China
| | - Liming Cai
- Department of Cardiopulmonary Bypass, State Key Laboratory of Cardiovascular Medicine, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Haojie Rao
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Hong Chen
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jian Lin
- Synthetic and Functional Biomolecules Center, Beijing National Laboratory for Molecular Sciences, College of Chemistry and Molecular Engineering, Peking University, Beijing, China
| | - Zhao Cui
- Renal Division, Peking University First Hospital, Beijing, China
| | - Bingyang Ji
- Department of Cardiopulmonary Bypass, State Key Laboratory of Cardiovascular Medicine, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jianwei Wang
- MOH Key Laboratory of Systems Biology of Pathogens, Institute of Pathogen Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Nailin Li
- Department of Medicine-Solna, Cardiovascular Medicine Unit, Karolinska Institute, Stockholm, Sweden
| | - De-Pei Liu
- Key Laboratory of Common Mechanism Research for Major Diseases, Department of Biochemistry and Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Thomas Renné
- Institute of Clinical Chemistry and Laboratory Medicine, University Medical Center Hamburg-Eppendorf (UKE), Hamburg, Germany
- Center for Thrombosis and Hemostasis (CTH), Johannes Gutenberg University Medical Center, Mainz, Germany
- Irish Centre for Vascular Biology, School of Pharmacy and Biomolecular Sciences, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Miao Wang
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
- Clinical Pharmacology Center, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
- National Health Commission Cardiovascular Disease Regenerative Medicine Research Key Laboratory, Central China Subcenter of National Center for Cardiovascular Diseases, Henan Cardiovascular Disease Center, Fuwai Central-China Cardiovascular Hospital, Central China Fuwai Hospital of Zhengzhou University, Zhengzhou, China.
| |
Collapse
|
12
|
Heidari MM, Shirazi EA, Cheraghi SF, Shahshahani R, Rahnama T, Khatami M. CDR grafting and site-directed mutagenesis approach for the generation and affinity maturation of Anti-CD20 nanobody. Mol Biol Rep 2024; 51:751. [PMID: 38874667 DOI: 10.1007/s11033-024-09684-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2024] [Accepted: 05/29/2024] [Indexed: 06/15/2024]
Abstract
BACKGROUND Recently, new and advanced techniques have been adopted to design and produce nanobodies, which are used in diagnostic and immunotherapy treatments. Traditionally, nanobodies are prepared from camelid immune libraries that require animal treatments. However, such approaches require large library sizes and complicated selection procedures. The current study has employed CDR grafting and site-directed mutagenesis techniques to create genetically engineered nanobodies against the tumor marker CD20 (anti-CD20 nanobodies) used in leukemia treatment. METHODS AND RESULTS In this study, we utilized the swapping method to graft CDRs from the VH Rituximab antibody to VHH CDRs. We aimed to enhance the binding affinity of the nanobodies by substituting the amino acids (Y101R-Y102R-Y107R) in the VHH-CDR3. To assess the binding capacity of the mutated nanobodies, we conducted an ELISA test. Moreover, through flow cytometry analysis, we compared the fluorescence intensity of the grafted CD20 and mutant nanobodies with that of the commercially available human anti-CD20 in Raji cells. The results showed a significant difference in the fluorescence intensity of the grafted nanobodies and mutant nanobodies when compared to the commercially available human anti-CD20. CONCLUSION The approach we followed in this study makes it possible to create multiple anti-CD20 nanobodies with varying affinities without the need for extensive selection efforts. Additionally, our research has demonstrated that computational tools are highly reliable in designing functional nanobodies.
Collapse
Affiliation(s)
| | | | | | | | - Tina Rahnama
- Department of Biology, Yazd University, Yazd, Iran
| | | |
Collapse
|
13
|
Rocha Tapia A, Abgottspon F, Nilvebrant J, Nygren PÅ, Duclos Ivetich S, Bello Hernandez AJ, Thanasi IA, Szijj PA, Sekkat G, Cuenot FM, Chudasama V, Aceto N, deMello AJ, Richards DA. Site-directed conjugation of single-stranded DNA to affinity proteins: quantifying the importance of conjugation strategy. Chem Sci 2024; 15:8982-8992. [PMID: 38873052 PMCID: PMC11168188 DOI: 10.1039/d4sc01838a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Accepted: 04/27/2024] [Indexed: 06/15/2024] Open
Abstract
Affinity protein-oligonucleotide conjugates are increasingly being explored as diagnostic and therapeutic tools. Despite growing interest, these probes are typically constructed using outdated, non-selective chemistries, and little has been done to investigate how conjugation to oligonucleotides influences the function of affinity proteins. Herein, we report a novel site-selective conjugation method for furnishing affinity protein-oligonucleotide conjugates in a 93% yield within fifteen minutes. Using SPR, we explore how the choice of affinity protein, conjugation strategy, and DNA length impact target binding and reveal the deleterious effects of non-specific conjugation methods. Furthermore, we show that these adverse effects can be minimised by employing our site-selective conjugation strategy, leading to improved performance in an immuno-PCR assay. Finally, we investigate the interactions between affinity protein-oligonucleotide conjugates and live cells, demonstrating the benefits of site-selective conjugation. This work provides critical insight into the importance of conjugation strategy when constructing affinity protein-oligonucleotide conjugates.
Collapse
Affiliation(s)
- Andres Rocha Tapia
- Institute for Chemical and Bioengineering, ETH Zurich Vladimir-Prelog-Weg 1 8093 Zürich Switzerland
| | - Fabrice Abgottspon
- Institute for Chemical and Bioengineering, ETH Zurich Vladimir-Prelog-Weg 1 8093 Zürich Switzerland
| | - Johan Nilvebrant
- Department of Protein Science, KTH Royal Institute of Technology, AlbaNova University Center 106 91 Stockholm Sweden
| | - Per-Åke Nygren
- Department of Protein Science, KTH Royal Institute of Technology, AlbaNova University Center 106 91 Stockholm Sweden
| | - Sarah Duclos Ivetich
- Institute for Chemical and Bioengineering, ETH Zurich Vladimir-Prelog-Weg 1 8093 Zürich Switzerland
| | | | - Ioanna A Thanasi
- Department of Chemistry, University College London 20 Gordon Street WC1H 0AJ London UK
| | - Peter A Szijj
- Department of Chemistry, University College London 20 Gordon Street WC1H 0AJ London UK
| | - Ghali Sekkat
- Institute for Chemical and Bioengineering, ETH Zurich Vladimir-Prelog-Weg 1 8093 Zürich Switzerland
| | - François M Cuenot
- Department of Biology, Institute of Molecular Health Sciences, ETH Zurich Otto-Stern-Weg 7 8093 Zürich Switzerland
| | - Vijay Chudasama
- Department of Chemistry, University College London 20 Gordon Street WC1H 0AJ London UK
| | - Nicola Aceto
- Department of Biology, Institute of Molecular Health Sciences, ETH Zurich Otto-Stern-Weg 7 8093 Zürich Switzerland
| | - Andrew J deMello
- Institute for Chemical and Bioengineering, ETH Zurich Vladimir-Prelog-Weg 1 8093 Zürich Switzerland
| | - Daniel A Richards
- Institute for Chemical and Bioengineering, ETH Zurich Vladimir-Prelog-Weg 1 8093 Zürich Switzerland
| |
Collapse
|
14
|
Solà Colom M, Fu Z, Gunkel P, Güttler T, Trakhanov S, Srinivasan V, Gregor K, Pleiner T, Görlich D. A checkpoint function for Nup98 in nuclear pore formation suggested by novel inhibitory nanobodies. EMBO J 2024; 43:2198-2232. [PMID: 38649536 PMCID: PMC11148069 DOI: 10.1038/s44318-024-00081-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2023] [Revised: 03/11/2024] [Accepted: 03/13/2024] [Indexed: 04/25/2024] Open
Abstract
Nuclear pore complex (NPC) biogenesis is a still enigmatic example of protein self-assembly. We now introduce several cross-reacting anti-Nup nanobodies for imaging intact nuclear pore complexes from frog to human. We also report a simplified assay that directly tracks postmitotic NPC assembly with added fluorophore-labeled anti-Nup nanobodies. During interphase, NPCs are inserted into a pre-existing nuclear envelope. Monitoring this process is challenging because newly assembled NPCs are indistinguishable from pre-existing ones. We overcame this problem by inserting Xenopus-derived NPCs into human nuclear envelopes and using frog-specific anti-Nup nanobodies for detection. We further asked whether anti-Nup nanobodies could serve as NPC assembly inhibitors. Using a selection strategy against conserved epitopes, we obtained anti-Nup93, Nup98, and Nup155 nanobodies that block Nup-Nup interfaces and arrest NPC assembly. We solved structures of nanobody-target complexes and identified roles for the Nup93 α-solenoid domain in recruiting Nup358 and the Nup214·88·62 complex, as well as for Nup155 and the Nup98 autoproteolytic domain in NPC scaffold assembly. The latter suggests a checkpoint linking pore formation to the assembly of the Nup98-dominated permeability barrier.
Collapse
Affiliation(s)
- Mireia Solà Colom
- Department of Cellular Logistics, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
- AI Proteins, 20 Overland St., Boston, MA, USA
| | - Zhenglin Fu
- Department of Cellular Logistics, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
| | - Philip Gunkel
- Department of Cellular Logistics, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
| | - Thomas Güttler
- Department of Cellular Logistics, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
- Octapharma Biopharmaceuticals, Im Neuenheimer Feld 590, 69120, Heidelberg, Germany
| | - Sergei Trakhanov
- Department of Cellular Logistics, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
| | - Vasundara Srinivasan
- Department of Cellular Logistics, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
- Department of Chemistry, Institute of Biochemistry and Molecular Biology, Universität Hamburg, Hamburg, Germany
| | - Kathrin Gregor
- Department of Cellular Logistics, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
| | - Tino Pleiner
- Department of Cellular Logistics, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
- Department of Molecular and Cellular Physiology, Stanford University School of Medicine, Stanford, CA, USA
| | - Dirk Görlich
- Department of Cellular Logistics, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany.
| |
Collapse
|
15
|
Rizk SS, Moustafa DM, ElBanna SA, Nour El-Din HT, Attia AS. Nanobodies in the fight against infectious diseases: repurposing nature's tiny weapons. World J Microbiol Biotechnol 2024; 40:209. [PMID: 38771414 PMCID: PMC11108896 DOI: 10.1007/s11274-024-03990-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2024] [Accepted: 04/15/2024] [Indexed: 05/22/2024]
Abstract
Nanobodies are the smallest known antigen-binding molecules to date. Their small size, good tissue penetration, high stability and solubility, ease of expression, refolding ability, and negligible immunogenicity in the human body have granted them excellence over conventional antibodies. Those exceptional attributes of nanobodies make them promising candidates for various applications in biotechnology, medicine, protein engineering, structural biology, food, and agriculture. This review presents an overview of their structure, development methods, advantages, possible challenges, and applications with special emphasis on infectious diseases-related ones. A showcase of how nanobodies can be harnessed for applications including neutralization of viruses and combating antibiotic-resistant bacteria is detailed. Overall, the impact of nanobodies in vaccine design, rapid diagnostics, and targeted therapies, besides exploring their role in deciphering microbial structures and virulence mechanisms are highlighted. Indeed, nanobodies are reshaping the future of infectious disease prevention and treatment.
Collapse
Affiliation(s)
- Soha S Rizk
- Microbiology and Immunology Postgraduate Program, Faculty of Pharmacy, Cairo University, Cairo, 11562, Egypt
| | - Dina M Moustafa
- Department of Medical Sciences, Faculty of Dentistry, The British University in Egypt, El Sherouk City, Cairo, 11837, Egypt
| | - Shahira A ElBanna
- Department of Microbiology and Immunology, Faculty of Pharmacy, Cairo University, Cairo, 11562, Egypt
| | - Hanzada T Nour El-Din
- Department of Microbiology and Immunology, Faculty of Pharmacy, Cairo University, Cairo, 11562, Egypt
| | - Ahmed S Attia
- Department of Microbiology and Immunology, Faculty of Pharmacy, Cairo University, Cairo, 11562, Egypt.
| |
Collapse
|
16
|
Xiao Y, Dong H, Wu C, Zhang K, Jiang X, Chen J, Wang H, Xu S, Zhang F, Gu L. Nanobody in a Double "Y"-Shaped Assembly: A Promising Candidate for Lateral Flow Immunoassays. Anal Chem 2024; 96:7130-7137. [PMID: 38679866 DOI: 10.1021/acs.analchem.4c00509] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/01/2024]
Abstract
Derived from camelid heavy-chain antibodies, nanobodies (Nbs) are the smallest natural antibodies and are an ideal tool in biological studies because of their simple structure, high yield, and low cost. Nbs possess significant potential for developing highly specific and user-friendly diagnostic assays. Despite offering considerable advantages in detection applications, knowledge is limited regarding the exclusive use of Nbs in lateral flow immunoassay (LFIA) detection. Herein, we present a novel double "Y" architecture, achieved by using the SpyTag/SpyCatcher and Im7/CL7 systems. The double "Y" assemblies exhibited a significantly higher affinity for their epitopes, as particularly evident in the reduced dissociation rate. An LFIA employing double "Y" assemblies was effectively used to detect the severe acute respiratory syndrome coronavirus-2 N protein, with a detection limit of at least 500 pg/mL. This study helps broaden the array of tools available for the development of Nb-based diagnostic techniques.
Collapse
Affiliation(s)
- Yumeng Xiao
- State Key Laboratory of Microbial Technology, Shandong University, 72 Binhai Road, Qingdao 266237, P. R. China
| | - Hongjie Dong
- Shandong Institute of Parasitic Diseases, Shandong First Medical University & Shandong Academy of Medical Sciences, 11 Taibaizhong Road, Jining 272033, P. R. China
| | - Cancan Wu
- State Key Laboratory of Microbial Technology, Shandong University, 72 Binhai Road, Qingdao 266237, P. R. China
| | - Kundi Zhang
- State Key Laboratory of Microbial Technology, Shandong University, 72 Binhai Road, Qingdao 266237, P. R. China
| | - Xiaoqiong Jiang
- State Key Laboratory of Microbial Technology, Shandong University, 72 Binhai Road, Qingdao 266237, P. R. China
| | - Junyu Chen
- State Key Laboratory of Microbial Technology, Shandong University, 72 Binhai Road, Qingdao 266237, P. R. China
| | - Hongwei Wang
- State Key Laboratory of Microbial Technology, Shandong University, 72 Binhai Road, Qingdao 266237, P. R. China
| | - Sujuan Xu
- State Key Laboratory of Microbial Technology, Shandong University, 72 Binhai Road, Qingdao 266237, P. R. China
| | - Fengyu Zhang
- State Key Laboratory of Microbial Technology, Shandong University, 72 Binhai Road, Qingdao 266237, P. R. China
| | - Lichuan Gu
- State Key Laboratory of Microbial Technology, Shandong University, 72 Binhai Road, Qingdao 266237, P. R. China
| |
Collapse
|
17
|
Mortelecque J, Zejneli O, Bégard S, Simões MC, ElHajjar L, Nguyen M, Cantrelle FX, Hanoulle X, Rain JC, Colin M, Gomes CM, Buée L, Landrieu I, Danis C, Dupré E. A selection and optimization strategy for single-domain antibodies targeting the PHF6 linear peptide within the tau intrinsically disordered protein. J Biol Chem 2024; 300:107163. [PMID: 38484799 PMCID: PMC11007443 DOI: 10.1016/j.jbc.2024.107163] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2023] [Revised: 02/15/2024] [Accepted: 03/06/2024] [Indexed: 04/12/2024] Open
Abstract
The use of variable domain of the heavy-chain of the heavy-chain-only antibodies (VHHs) as disease-modifying biomolecules in neurodegenerative disorders holds promises, including targeting of aggregation-sensitive proteins. Exploitation of their clinical values depends however on the capacity to deliver VHHs with optimal physico-chemical properties for their specific context of use. We described previously a VHH with high therapeutic potential in a family of neurodegenerative diseases called tauopathies. The activity of this promising parent VHH named Z70 relies on its binding within the central region of the tau protein. Accordingly, we carried out random mutagenesis followed by yeast two-hybrid screening to obtain optimized variants. The VHHs selected from this initial screen targeted the same epitope as VHH Z70 as shown using NMR spectroscopy and had indeed improved binding affinities according to dissociation constant values obtained by surface plasmon resonance spectroscopy. The improved affinities can be partially rationalized based on three-dimensional structures and NMR data of three complexes consisting of an optimized VHH and a peptide containing the tau epitope. Interestingly, the ability of the VHH variants to inhibit tau aggregation and seeding could not be predicted from their affinity alone. We indeed showed that the in vitro and in cellulo VHH stabilities are other limiting key factors to their efficacy. Our results demonstrate that only a complete pipeline of experiments, here described, permits a rational selection of optimized VHH variants, resulting in the selection of VHH variants with higher affinities and/or acting against tau seeding in cell models.
Collapse
Affiliation(s)
- Justine Mortelecque
- CNRS EMR9002 - BSI - Integrative Structural Biology, Lille, France; Univ. Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1167 - RID-AGE - Risk Factors and Molecular Determinants of Aging-Related Diseases, Lille, France
| | - Orgeta Zejneli
- CNRS EMR9002 - BSI - Integrative Structural Biology, Lille, France; Univ. Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1167 - RID-AGE - Risk Factors and Molecular Determinants of Aging-Related Diseases, Lille, France; Univ. Lille, Inserm, CHU-Lille, U1172 - LilNCog - Lille Neuroscience & Cognition, Lille, France
| | - Séverine Bégard
- Univ. Lille, Inserm, CHU-Lille, U1172 - LilNCog - Lille Neuroscience & Cognition, Lille, France
| | - Margarida C Simões
- BioISI - Instituto de Biosistemas e Ciências Integrativas, Faculdade de Ciências, Universidade de Lisboa, Lisboa, Portugal; Departamento de Química e Bioquímica, Faculdade de Ciências, Universidade de Lisboa, Lisboa, Portugal
| | - Lea ElHajjar
- CNRS EMR9002 - BSI - Integrative Structural Biology, Lille, France; Univ. Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1167 - RID-AGE - Risk Factors and Molecular Determinants of Aging-Related Diseases, Lille, France
| | - Marine Nguyen
- CNRS EMR9002 - BSI - Integrative Structural Biology, Lille, France; Univ. Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1167 - RID-AGE - Risk Factors and Molecular Determinants of Aging-Related Diseases, Lille, France
| | - François-Xavier Cantrelle
- CNRS EMR9002 - BSI - Integrative Structural Biology, Lille, France; Univ. Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1167 - RID-AGE - Risk Factors and Molecular Determinants of Aging-Related Diseases, Lille, France
| | - Xavier Hanoulle
- CNRS EMR9002 - BSI - Integrative Structural Biology, Lille, France; Univ. Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1167 - RID-AGE - Risk Factors and Molecular Determinants of Aging-Related Diseases, Lille, France
| | | | - Morvane Colin
- Univ. Lille, Inserm, CHU-Lille, U1172 - LilNCog - Lille Neuroscience & Cognition, Lille, France
| | - Cláudio M Gomes
- BioISI - Instituto de Biosistemas e Ciências Integrativas, Faculdade de Ciências, Universidade de Lisboa, Lisboa, Portugal; Departamento de Química e Bioquímica, Faculdade de Ciências, Universidade de Lisboa, Lisboa, Portugal
| | - Luc Buée
- Univ. Lille, Inserm, CHU-Lille, U1172 - LilNCog - Lille Neuroscience & Cognition, Lille, France.
| | - Isabelle Landrieu
- CNRS EMR9002 - BSI - Integrative Structural Biology, Lille, France; Univ. Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1167 - RID-AGE - Risk Factors and Molecular Determinants of Aging-Related Diseases, Lille, France.
| | - Clément Danis
- CNRS EMR9002 - BSI - Integrative Structural Biology, Lille, France; Univ. Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1167 - RID-AGE - Risk Factors and Molecular Determinants of Aging-Related Diseases, Lille, France; Univ. Lille, Inserm, CHU-Lille, U1172 - LilNCog - Lille Neuroscience & Cognition, Lille, France
| | - Elian Dupré
- CNRS EMR9002 - BSI - Integrative Structural Biology, Lille, France; Univ. Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1167 - RID-AGE - Risk Factors and Molecular Determinants of Aging-Related Diseases, Lille, France.
| |
Collapse
|
18
|
Newstead S. Future opportunities in solute carrier structural biology. Nat Struct Mol Biol 2024; 31:587-590. [PMID: 38637662 DOI: 10.1038/s41594-024-01271-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2024] [Accepted: 03/07/2024] [Indexed: 04/20/2024]
Abstract
Solute carriers (SLCs) control the flow of small molecules and ions across biological membranes. Over the last 20 years, the pace of research in SLC biology has accelerated markedly, opening new opportunities to treat metabolic diseases, cancer and neurological disorders. Recently, new families of atypical SLCs, with roles in organelle biology, metabolite signaling and trafficking, have expanded their roles in the cell. This Perspective discusses work leading to current advances and the emerging opportunities to target and modulate SLCs to uncover new biology and treat human disease.
Collapse
Affiliation(s)
- Simon Newstead
- Department of Biochemistry, University of Oxford, Oxford, UK.
- The Kavli Institute for Nanoscience Discovery, University of Oxford, Oxford, UK.
| |
Collapse
|
19
|
Zhu XY, Li QX, Kong Y, Huang KK, Wang G, Wang YJ, Lu J, Hua GQ, Wu YL, Ying TL. A novel human single-domain antibody-drug conjugate targeting CEACAM5 exhibits potent in vitro and in vivo antitumor activity. Acta Pharmacol Sin 2024; 45:609-618. [PMID: 38030799 PMCID: PMC10834580 DOI: 10.1038/s41401-023-01200-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/09/2023] [Accepted: 11/13/2023] [Indexed: 12/01/2023]
Abstract
Leveraging the specificity of antibody to deliver cytotoxic agent into tumor, antibody-drug conjugates (ADCs) have become one of the hotspots in the development of anticancer therapies. Although significant progress has been achieved, there remain challenges to overcome, including limited penetration into solid tumors and potential immunogenicity. Fully human single-domain antibodies (UdAbs), with their small size and human nature, represent a promising approach for addressing these challenges. Carcinoembryonic antigen-related cell adhesion molecule 5 (CEACAM5) is a glycosylated cell surface protein that rarely expressed in normal adult tissues but overexpressed in diverse cancers, taking part in tumorigenesis, progression, and metastasis. In this study, we investigated the therapeutic potential of UdADC targeting CEACAM5. We performed biopanning in our library and obtained an antibody candidate B9, which bound potently and specifically to CEACAM5 protein (KD = 4.84 nM) and possessed excellent biophysical properties (low aggregation tendency, high homogeneity, and thermal stability). The conjugation of B9 with a potent cytotoxic agent, monomethyl auristatin E (MMAE), exhibited superior antitumor efficacy against CEACAM5-expressing human gastric cancer cell line MKN-45, human pancreatic carcinoma cell line BxPC-3 and human colorectal cancer cell line LS174T with IC50 values of 38.14, 25.60, and 101.4 nM, respectively. In BxPC-3 and MKN-45 xenograft mice, administration of UdADC B9-MMAE (5 mg/kg, i.v.) every 2 days for 4 times markedly inhibited the tumor growth without significant change in body weight. This study may have significant implications for the design of next-generation ADCs.
Collapse
Affiliation(s)
- Xiao-Yi Zhu
- MOE/NHC/CAMS Key Laboratory of Medical Molecular Virology, Shanghai Frontiers Science Center of Pathogenic Microorganisms and Infection, School of Basic Medical Sciences, Shanghai Medical College, Fudan University, Shanghai, 200032, China
- Shanghai Engineering Research Center for Synthetic Immunology, Shanghai, 200032, China
| | - Quan-Xiao Li
- MOE/NHC/CAMS Key Laboratory of Medical Molecular Virology, Shanghai Frontiers Science Center of Pathogenic Microorganisms and Infection, School of Basic Medical Sciences, Shanghai Medical College, Fudan University, Shanghai, 200032, China
- Shanghai Engineering Research Center for Synthetic Immunology, Shanghai, 200032, China
| | - Yu Kong
- MOE/NHC/CAMS Key Laboratory of Medical Molecular Virology, Shanghai Frontiers Science Center of Pathogenic Microorganisms and Infection, School of Basic Medical Sciences, Shanghai Medical College, Fudan University, Shanghai, 200032, China
- Shanghai Engineering Research Center for Synthetic Immunology, Shanghai, 200032, China
| | - Ke-Ke Huang
- MOE/NHC/CAMS Key Laboratory of Medical Molecular Virology, Shanghai Frontiers Science Center of Pathogenic Microorganisms and Infection, School of Basic Medical Sciences, Shanghai Medical College, Fudan University, Shanghai, 200032, China
- Shanghai Engineering Research Center for Synthetic Immunology, Shanghai, 200032, China
| | - Gang Wang
- MOE/NHC/CAMS Key Laboratory of Medical Molecular Virology, Shanghai Frontiers Science Center of Pathogenic Microorganisms and Infection, School of Basic Medical Sciences, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Yun-Ji Wang
- MOE/NHC/CAMS Key Laboratory of Medical Molecular Virology, Shanghai Frontiers Science Center of Pathogenic Microorganisms and Infection, School of Basic Medical Sciences, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Jun Lu
- Auckland Bioengineering Institute, University of Auckland, Auckland, 1010, New Zealand
| | - Guo-Qiang Hua
- Cancer Institute, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
| | - Yan-Ling Wu
- MOE/NHC/CAMS Key Laboratory of Medical Molecular Virology, Shanghai Frontiers Science Center of Pathogenic Microorganisms and Infection, School of Basic Medical Sciences, Shanghai Medical College, Fudan University, Shanghai, 200032, China.
- Shanghai Engineering Research Center for Synthetic Immunology, Shanghai, 200032, China.
| | - Tian-Lei Ying
- MOE/NHC/CAMS Key Laboratory of Medical Molecular Virology, Shanghai Frontiers Science Center of Pathogenic Microorganisms and Infection, School of Basic Medical Sciences, Shanghai Medical College, Fudan University, Shanghai, 200032, China.
- Shanghai Engineering Research Center for Synthetic Immunology, Shanghai, 200032, China.
| |
Collapse
|
20
|
Teodori L, Omer M, Kjems J. RNA nanostructures for targeted drug delivery and imaging. RNA Biol 2024; 21:1-19. [PMID: 38555519 PMCID: PMC10984137 DOI: 10.1080/15476286.2024.2328440] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Revised: 02/26/2024] [Accepted: 03/04/2024] [Indexed: 04/02/2024] Open
Abstract
The RNA molecule plays a pivotal role in many biological processes by relaying genetic information, regulating gene expression, and serving as molecular machines and catalyzers. This inherent versatility of RNA has fueled significant advancements in the field of RNA nanotechnology, driving the engineering of complex nanoscale architectures toward biomedical applications, including targeted drug delivery and bioimaging. RNA polymers, serving as building blocks, offer programmability and predictability of Watson-Crick base pairing, as well as non-canonical base pairing, for the construction of nanostructures with high precision and stoichiometry. Leveraging the ease of chemical modifications to protect the RNA from degradation, researchers have developed highly functional and biocompatible RNA architectures and integrated them into preclinical studies for the delivery of payloads and imaging agents. This review offers an educational introduction to the use of RNA as a biopolymer in the design of multifunctional nanostructures applied to targeted delivery in vivo, summarizing physical and biological barriers along with strategies to overcome them. Furthermore, we highlight the most recent progress in the development of both small and larger RNA nanostructures, with a particular focus on imaging reagents and targeted cancer therapeutics in pre-clinical models and provide insights into the prospects of this rapidly evolving field.
Collapse
Affiliation(s)
- Laura Teodori
- Interdisciplinary Nanoscience Center (iNANO), Aarhus University, Aarhus C, Denmark
- Center for Cellular Signal Patterns (CellPAT), Aarhus University, Aarhus, Denmark
- Center for RNA Therapeutics towards Metabolic Diseases (RNA-META), Aarhus University, Aarhus, Denmark
| | - Marjan Omer
- Interdisciplinary Nanoscience Center (iNANO), Aarhus University, Aarhus C, Denmark
- Center for Cellular Signal Patterns (CellPAT), Aarhus University, Aarhus, Denmark
| | - Jørgen Kjems
- Interdisciplinary Nanoscience Center (iNANO), Aarhus University, Aarhus C, Denmark
- Center for Cellular Signal Patterns (CellPAT), Aarhus University, Aarhus, Denmark
- Center for RNA Therapeutics towards Metabolic Diseases (RNA-META), Aarhus University, Aarhus, Denmark
- Department of Molecular Biology and Genetics, Aarhus University, Aarhus, Denmark
| |
Collapse
|
21
|
Stevens TA, Tomaleri GP, Hazu M, Wei S, Nguyen VN, DeKalb C, Voorhees RM, Pleiner T. A nanobody-based strategy for rapid and scalable purification of human protein complexes. Nat Protoc 2024; 19:127-158. [PMID: 37974029 DOI: 10.1038/s41596-023-00904-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2023] [Accepted: 08/18/2023] [Indexed: 11/19/2023]
Abstract
The isolation of proteins in high yield and purity is a major bottleneck for the analysis of their three-dimensional structure, function and interactome. Here, we present a streamlined workflow for the rapid production of proteins or protein complexes using lentiviral transduction of human suspension cells, combined with highly specific nanobody-mediated purification and proteolytic elution. Application of the method requires prior generation of a plasmid coding for a protein of interest (POI) fused to an N- or C-terminal GFP or ALFA peptide tag using a lentiviral plasmid toolkit we have designed. The plasmid is then used to generate human suspension cell lines stably expressing the tagged fusion protein by lentiviral transduction. By leveraging the picomolar affinity of the GFP and ALFA nanobodies for their respective tags, the POI can be specifically captured from the resulting cell lysate even when expressed at low levels and under a variety of conditions, including detergents and mild denaturants. Finally, rapid and specific elution of the POI (in its tagged or untagged form) under native conditions is achieved within minutes at 4 °C, using the engineered SUMO protease SENPEuB. We demonstrate the wide applicability of the method by purifying multiple challenging soluble and membrane protein complexes to high purity from human cells. Our strategy is also directly compatible with many widely used GFP-expression plasmids, cell lines and transgenic model organisms. Finally, our method is faster than alternative approaches, requiring only 8 d from plasmid to purified protein, and results in substantially improved yields and purity.
Collapse
Affiliation(s)
- Taylor Anthony Stevens
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, USA
| | - Giovani Pinton Tomaleri
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, USA
| | - Masami Hazu
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, USA
| | - Sophia Wei
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, USA
| | - Vy N Nguyen
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, USA
| | - Charlene DeKalb
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, USA
| | - Rebecca M Voorhees
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, USA.
| | - Tino Pleiner
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, USA.
- Department of Molecular and Cellular Physiology, Stanford University School of Medicine, Stanford, CA, USA.
| |
Collapse
|
22
|
Aksu M, Kumar P, Güttler T, Taxer W, Gregor K, Mußil B, Rymarenko O, Stegmann KM, Dickmanns A, Gerber S, Reineking W, Schulz C, Henneck T, Mohamed A, Pohlmann G, Ramazanoglu M, Mese K, Groß U, Ben-Yedidia T, Ovadia O, Fischer DW, Kamensky M, Reichman A, Baumgärtner W, von Köckritz-Blickwede M, Dobbelstein M, Görlich D. Nanobodies to multiple spike variants and inhalation of nanobody-containing aerosols neutralize SARS-CoV-2 in cell culture and hamsters. Antiviral Res 2024; 221:105778. [PMID: 38065245 DOI: 10.1016/j.antiviral.2023.105778] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2023] [Revised: 11/23/2023] [Accepted: 12/04/2023] [Indexed: 12/17/2023]
Abstract
The ongoing threat of COVID-19 has highlighted the need for effective prophylaxis and convenient therapies, especially for outpatient settings. We have previously developed highly potent single-domain (VHH) antibodies, also known as nanobodies, that target the Receptor Binding Domain (RBD) of the SARS-CoV-2 Spike protein and neutralize the Wuhan strain of the virus. In this study, we present a new generation of anti-RBD nanobodies with superior properties. The primary representative of this group, Re32D03, neutralizes Alpha to Delta as well as Omicron BA.2.75; other members neutralize, in addition, Omicron BA.1, BA.2, BA.4/5, and XBB.1. Crystal structures of RBD-nanobody complexes reveal how ACE2-binding is blocked and also explain the nanobodies' tolerance to immune escape mutations. Through the cryo-EM structure of the Ma16B06-BA.1 Spike complex, we demonstrated how a single nanobody molecule can neutralize a trimeric spike. We also describe a method for large-scale production of these nanobodies in Pichia pastoris, and for formulating them into aerosols. Exposing hamsters to these aerosols, before or even 24 h after infection with SARS-CoV-2, significantly reduced virus load, weight loss and pathogenicity. These results show the potential of aerosolized nanobodies for prophylaxis and therapy of coronavirus infections.
Collapse
Affiliation(s)
- Metin Aksu
- Max Planck Institute for Multidisciplinary Sciences, Dept. of Cellular Logistics, Am Fassberg 11, 37077 Göttingen, Germany
| | - Priya Kumar
- University Medical Center Göttingen, Dept. of Molecular Oncology, Justus von Liebig Weg 11, 37077 Göttingen, Germany
| | - Thomas Güttler
- Max Planck Institute for Multidisciplinary Sciences, Dept. of Cellular Logistics, Am Fassberg 11, 37077 Göttingen, Germany; Octapharma Biopharmaceuticals GmbH, Im Neuenheimer Feld 590, 69120 Heidelberg, Germany
| | - Waltraud Taxer
- Max Planck Institute for Multidisciplinary Sciences, Dept. of Cellular Logistics, Am Fassberg 11, 37077 Göttingen, Germany
| | - Kathrin Gregor
- Max Planck Institute for Multidisciplinary Sciences, Dept. of Cellular Logistics, Am Fassberg 11, 37077 Göttingen, Germany
| | - Bianka Mußil
- Max Planck Institute for Multidisciplinary Sciences, Dept. of Cellular Logistics, Am Fassberg 11, 37077 Göttingen, Germany
| | - Oleh Rymarenko
- Max Planck Institute for Multidisciplinary Sciences, Dept. of Cellular Logistics, Am Fassberg 11, 37077 Göttingen, Germany
| | - Kim M Stegmann
- University Medical Center Göttingen, Dept. of Molecular Oncology, Justus von Liebig Weg 11, 37077 Göttingen, Germany
| | - Antje Dickmanns
- University Medical Center Göttingen, Dept. of Molecular Oncology, Justus von Liebig Weg 11, 37077 Göttingen, Germany
| | - Sabrina Gerber
- University Medical Center Göttingen, Dept. of Molecular Oncology, Justus von Liebig Weg 11, 37077 Göttingen, Germany
| | - Wencke Reineking
- Department of Pathology, University of Veterinary Medicine Hannover, Bünteweg 17, 30559 Hannover, Germany
| | - Claudia Schulz
- Research Center for Emerging Infections and Zoonosis (RIZ), University of Veterinary Medicine Hannover, Bünteweg 17, 30559 Hannover, Germany
| | - Timo Henneck
- Research Center for Emerging Infections and Zoonosis (RIZ), University of Veterinary Medicine Hannover, Bünteweg 17, 30559 Hannover, Germany; Department of Biochemistry, University of Veterinary Medicine Hannover, Bünteweg 17, 30559 Hannover, Germany
| | - Ahmed Mohamed
- Research Center for Emerging Infections and Zoonosis (RIZ), University of Veterinary Medicine Hannover, Bünteweg 17, 30559 Hannover, Germany; Department of Biochemistry, University of Veterinary Medicine Hannover, Bünteweg 17, 30559 Hannover, Germany
| | - Gerhard Pohlmann
- Fraunhofer Institute for Toxicology and Experimental Medicine ITEM, Nikolai-Fuchs Str. 1, 30625 Hannover, Germany
| | - Mehmet Ramazanoglu
- Fraunhofer Institute for Toxicology and Experimental Medicine ITEM, Nikolai-Fuchs Str. 1, 30625 Hannover, Germany
| | - Kemal Mese
- University Medical Center Göttingen, Dept. of Medical Microbiology and Virology, Kreuzbergring 57, 37075 Göttingen, Germany
| | - Uwe Groß
- University Medical Center Göttingen, Dept. of Medical Microbiology and Virology, Kreuzbergring 57, 37075 Göttingen, Germany
| | - Tamar Ben-Yedidia
- Scinai Immunotherapeutics Ltd., Jerusalem BioPark, Hadassah Ein Kerem, Jerusalem, 9112001, Israel
| | - Oded Ovadia
- Scinai Immunotherapeutics Ltd., Jerusalem BioPark, Hadassah Ein Kerem, Jerusalem, 9112001, Israel
| | - Dalit Weinstein Fischer
- Scinai Immunotherapeutics Ltd., Jerusalem BioPark, Hadassah Ein Kerem, Jerusalem, 9112001, Israel
| | - Merav Kamensky
- Scinai Immunotherapeutics Ltd., Jerusalem BioPark, Hadassah Ein Kerem, Jerusalem, 9112001, Israel
| | - Amir Reichman
- Scinai Immunotherapeutics Ltd., Jerusalem BioPark, Hadassah Ein Kerem, Jerusalem, 9112001, Israel
| | - Wolfgang Baumgärtner
- Department of Pathology, University of Veterinary Medicine Hannover, Bünteweg 17, 30559 Hannover, Germany
| | - Maren von Köckritz-Blickwede
- Research Center for Emerging Infections and Zoonosis (RIZ), University of Veterinary Medicine Hannover, Bünteweg 17, 30559 Hannover, Germany; Department of Biochemistry, University of Veterinary Medicine Hannover, Bünteweg 17, 30559 Hannover, Germany
| | - Matthias Dobbelstein
- Max Planck Institute for Multidisciplinary Sciences, Dept. of Cellular Logistics, Am Fassberg 11, 37077 Göttingen, Germany; University Medical Center Göttingen, Dept. of Molecular Oncology, Justus von Liebig Weg 11, 37077 Göttingen, Germany.
| | - Dirk Görlich
- Max Planck Institute for Multidisciplinary Sciences, Dept. of Cellular Logistics, Am Fassberg 11, 37077 Göttingen, Germany.
| |
Collapse
|
23
|
Jeong S, Koh D, Gwak E, Srambickal CV, Seo D, Widengren J, Lee JC. Pushing the Resolution Limit of Stimulated Emission Depletion Optical Nanoscopy. Int J Mol Sci 2023; 25:26. [PMID: 38203197 PMCID: PMC10779414 DOI: 10.3390/ijms25010026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2023] [Revised: 12/08/2023] [Accepted: 12/13/2023] [Indexed: 01/12/2024] Open
Abstract
Optical nanoscopy, also known as super-resolution optical microscopy, has provided scientists with the means to surpass the diffraction limit of light microscopy and attain new insights into nanoscopic structures and processes that were previously inaccessible. In recent decades, numerous studies have endeavored to enhance super-resolution microscopy in terms of its spatial (lateral) resolution, axial resolution, and temporal resolution. In this review, we discuss recent efforts to push the resolution limit of stimulated emission depletion (STED) optical nanoscopy across multiple dimensions, including lateral resolution, axial resolution, temporal resolution, and labeling precision. We introduce promising techniques and methodologies building on the STED concept that have emerged in the field, such as MINSTED, isotropic STED, and event-triggered STED, and evaluate their respective strengths and limitations. Moreover, we discuss trade-off relationships that exist in far-field optical microscopy and how they come about in STED optical nanoscopy. By examining the latest developments addressing these aspects, we aim to provide an updated overview of the current state of STED nanoscopy and its potential for future research.
Collapse
Affiliation(s)
- Sejoo Jeong
- Department of New Biology, DGIST, Daegu 42988, Republic of Korea
| | - Dongbin Koh
- School of Undergraduate Studies, DGIST, Daegu 42988, Republic of Korea
| | - Eunha Gwak
- Department of New Biology, DGIST, Daegu 42988, Republic of Korea
| | - Chinmaya V. Srambickal
- Exp. Biomol. Physics, Dept. Applied Physics, KTH—Royal Institute of Technology, 106 91 Stockholm, Sweden
| | - Daeha Seo
- Department of Physics and Chemistry, DGIST, Daegu 42988, Republic of Korea
| | - Jerker Widengren
- Exp. Biomol. Physics, Dept. Applied Physics, KTH—Royal Institute of Technology, 106 91 Stockholm, Sweden
| | - Jong-Chan Lee
- Department of New Biology, DGIST, Daegu 42988, Republic of Korea
- New Biology Research Center, DGIST, Daegu 42988, Republic of Korea
| |
Collapse
|
24
|
Glück IM, Mathias GP, Strauss S, Rat V, Gialdini I, Ebert TS, Stafford C, Agam G, Manley S, Hornung V, Jungmann R, Sieben C, Lamb DC. Nanoscale organization of the endogenous ASC speck. iScience 2023; 26:108382. [PMID: 38047065 PMCID: PMC10690566 DOI: 10.1016/j.isci.2023.108382] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2022] [Revised: 06/15/2023] [Accepted: 10/31/2023] [Indexed: 12/05/2023] Open
Abstract
The NLRP3 inflammasome is a central component of the innate immune system. Its activation leads to formation of the ASC speck, a supramolecular assembly of the inflammasome adaptor protein ASC. Different models, based on ASC overexpression, have been proposed for the structure of the ASC speck. Using dual-color 3D super-resolution imaging (dSTORM and DNA-PAINT), we visualized the ASC speck structure following NLRP3 inflammasome activation using endogenous ASC expression. A complete structure was only obtainable by labeling with both anti-ASC antibodies and nanobodies. The complex varies in diameter between ∼800 and 1000 nm, and is composed of a dense core with emerging filaments. Dual-color confocal fluorescence microscopy indicated that the ASC speck does not colocalize with the microtubule-organizing center at late time points after Nigericin stimulation. From super-resolution images of whole cells, the ASC specks were sorted into a pseudo-time sequence indicating that they become denser but not larger during formation.
Collapse
Affiliation(s)
- Ivo M. Glück
- Department of Chemistry, Ludwig Maximilians-Universität München, Butenandtstraße 5-13, 81377 München, Germany
- Center for Nano Science (CENS), Ludwig Maximilians-Universität München, Butenandtstraße 5-13, 81377 München, Germany
| | - Grusha Primal Mathias
- Department of Chemistry, Ludwig Maximilians-Universität München, Butenandtstraße 5-13, 81377 München, Germany
| | - Sebastian Strauss
- Faculty of Physics and Center for Nanoscience, Ludwig Maximilian University, Munich, Germany
- Max Planck Institute of Biochemistry, Martinsried, Germany
| | - Virgile Rat
- Department of Chemistry, Ludwig Maximilians-Universität München, Butenandtstraße 5-13, 81377 München, Germany
- Center for Nano Science (CENS), Ludwig Maximilians-Universität München, Butenandtstraße 5-13, 81377 München, Germany
| | - Irene Gialdini
- Department of Chemistry, Ludwig Maximilians-Universität München, Butenandtstraße 5-13, 81377 München, Germany
- Center for Nano Science (CENS), Ludwig Maximilians-Universität München, Butenandtstraße 5-13, 81377 München, Germany
| | - Thomas Sebastian Ebert
- Gene Center and Department of Biochemistry, Ludwig-Maximilians-Universität, Munich, Germany
| | - Che Stafford
- Gene Center and Department of Biochemistry, Ludwig-Maximilians-Universität, Munich, Germany
| | - Ganesh Agam
- Department of Chemistry, Ludwig Maximilians-Universität München, Butenandtstraße 5-13, 81377 München, Germany
- Center for Nano Science (CENS), Ludwig Maximilians-Universität München, Butenandtstraße 5-13, 81377 München, Germany
| | - Suliana Manley
- Laboratory of Experimental Biophysics, École Polytechnique Fédérale de Lausanne, BSP 427 (Cubotron UNIL), Rte de la Sorge, CH-1015 Lausanne, Switzerland
| | - Veit Hornung
- Max Planck Institute of Biochemistry, Martinsried, Germany
- Gene Center and Department of Biochemistry, Ludwig-Maximilians-Universität, Munich, Germany
| | - Ralf Jungmann
- Faculty of Physics and Center for Nanoscience, Ludwig Maximilian University, Munich, Germany
- Max Planck Institute of Biochemistry, Martinsried, Germany
| | - Christian Sieben
- Laboratory of Experimental Biophysics, École Polytechnique Fédérale de Lausanne, BSP 427 (Cubotron UNIL), Rte de la Sorge, CH-1015 Lausanne, Switzerland
| | - Don C. Lamb
- Department of Chemistry, Ludwig Maximilians-Universität München, Butenandtstraße 5-13, 81377 München, Germany
- Center for Nano Science (CENS), Ludwig Maximilians-Universität München, Butenandtstraße 5-13, 81377 München, Germany
| |
Collapse
|
25
|
Frecot DI, Froehlich T, Rothbauer U. 30 years of nanobodies - an ongoing success story of small binders in biological research. J Cell Sci 2023; 136:jcs261395. [PMID: 37937477 DOI: 10.1242/jcs.261395] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2023] Open
Abstract
A milestone in the field of recombinant binding molecules was achieved 30 years ago with the discovery of single-domain antibodies from which antigen-binding variable domains, better known as nanobodies (Nbs), can be derived. Being only one tenth the size of conventional antibodies, Nbs feature high affinity and specificity, while being highly stable and soluble. In addition, they display accessibility to cryptic sites, low off-target accumulation and deep tissue penetration. Efficient selection methods, such as (semi-)synthetic/naïve or immunized cDNA libraries and display technologies, have facilitated the isolation of Nbs against diverse targets, and their single-gene format enables easy functionalization and high-yield production. This Review highlights recent advances in Nb applications in various areas of biological research, including structural biology, proteomics and high-resolution and in vivo imaging. In addition, we provide insights into intracellular applications of Nbs, such as live-cell imaging, biosensors and targeted protein degradation.
Collapse
Affiliation(s)
- Desiree I Frecot
- Pharmaceutical Biotechnology, Eberhard Karls University Tübingen, Auf der Morgenstelle 8, 72076 Tuebingen, Germany
- NMI Natural and Medical Sciences Institute at the University of Tübingen, Markwiesenstrasse 55, 72770 Reutlingen, Reutlingen, Germany
| | - Theresa Froehlich
- Pharmaceutical Biotechnology, Eberhard Karls University Tübingen, Auf der Morgenstelle 8, 72076 Tuebingen, Germany
| | - Ulrich Rothbauer
- Pharmaceutical Biotechnology, Eberhard Karls University Tübingen, Auf der Morgenstelle 8, 72076 Tuebingen, Germany
| |
Collapse
|
26
|
Chen KT. Novel Imaging Probes: From Design to Applications. Pharmaceuticals (Basel) 2023; 16:1506. [PMID: 37895977 PMCID: PMC10609778 DOI: 10.3390/ph16101506] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2023] [Revised: 10/23/2023] [Accepted: 10/23/2023] [Indexed: 10/29/2023] Open
Abstract
Molecular imaging has emerged as a powerful tool for clinical diagnosis [...].
Collapse
Affiliation(s)
- Kuo-Ting Chen
- Department of Chemistry, National Dong Hwa University, Hualien 974301, Taiwan
| |
Collapse
|
27
|
Monti G, Vincke C, Lunding M, Jensen AMG, Madsen P, Muyldermans S, Kjolby M, Andersen OM. Epitope mapping of nanobodies binding the Alzheimer's disease receptor SORLA. J Biotechnol 2023; 375:17-27. [PMID: 37634829 DOI: 10.1016/j.jbiotec.2023.08.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2023] [Revised: 08/20/2023] [Accepted: 08/24/2023] [Indexed: 08/29/2023]
Abstract
Reduced levels of the Sortilin-related receptor with A-type repeats (SORLA) in different brain regions as well as in the cerebrospinal fluid have been associated with Alzheimer's disease. Methods and reagents to develop reliable detection assays to quantify SORLA and its specific isoforms are therefore much needed. Nanobodies (Nbs) are unique biomolecules derived from the blood of camelids that display advantageous physicochemical and antigen affinity properties, making them attractive tools with great relevance to both diagnostic and therapeutic applications. Here, we purified and characterized eight Nbs that were isolated from the blood of an alpaca immunized with the recombinant extracellular domain of SORLA. The selected Nbs showed high affinity to SORLA in the low nanomolar range as observed by surface plasmon resonance. For mapping of the Nbs' epitopes within the antigen, we transiently transfected HEK293 cells with a panel of SORLA deletion constructs, and developed a protocol of immunostaining by applying fluorescent dye conjugated Nbs. With this method, we showed that the selected Nbs specifically recognize a part of SORLA containing Fibronectin-type III domains, representing promising tools not only for disease clarifying research, but also for translational medicine as candidates for clinical diagnostic purposes.
Collapse
Affiliation(s)
- Giulia Monti
- Department of Biomedicine, Aarhus University, Høegh‑Guldbergs Gade 10, 8000 Aarhus C, Denmark
| | - Cécile Vincke
- Laboratory of Cellular and Molecular Immunology, Vrije Universiteit Brussel, Brussels, Belgium; Myeloid Cell Immunology Lab, VIB Center for Inflammation Research, Brussels, Belgium
| | - Melanie Lunding
- Department of Biomedicine, Aarhus University, Høegh‑Guldbergs Gade 10, 8000 Aarhus C, Denmark
| | - Anne Mette G Jensen
- Department of Biomedicine, Aarhus University, Høegh‑Guldbergs Gade 10, 8000 Aarhus C, Denmark
| | - Peder Madsen
- Department of Biomedicine, Aarhus University, Høegh‑Guldbergs Gade 10, 8000 Aarhus C, Denmark
| | - Serge Muyldermans
- Laboratory of Cellular and Molecular Immunology, Vrije Universiteit Brussel, Brussels, Belgium
| | - Mads Kjolby
- Department of Biomedicine, Aarhus University, Høegh‑Guldbergs Gade 10, 8000 Aarhus C, Denmark; Department of Clinical Pharmacology and Steno Diabetes Center Aarhus, Aarhus University Hospital, Denmark
| | - Olav M Andersen
- Department of Biomedicine, Aarhus University, Høegh‑Guldbergs Gade 10, 8000 Aarhus C, Denmark.
| |
Collapse
|
28
|
Pleiner T, Hazu M, Pinton Tomaleri G, Nguyen VN, Januszyk K, Voorhees RM. A selectivity filter in the ER membrane protein complex limits protein misinsertion at the ER. J Cell Biol 2023; 222:e202212007. [PMID: 37199759 PMCID: PMC10200711 DOI: 10.1083/jcb.202212007] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2022] [Revised: 03/31/2023] [Accepted: 04/20/2023] [Indexed: 05/19/2023] Open
Abstract
Tail-anchored (TA) proteins play essential roles in mammalian cells, and their accurate localization is critical for proteostasis. Biophysical similarities lead to mistargeting of mitochondrial TA proteins to the ER, where they are delivered to the insertase, the ER membrane protein complex (EMC). Leveraging an improved structural model of the human EMC, we used mutagenesis and site-specific crosslinking to map the path of a TA protein from its cytosolic capture by methionine-rich loops to its membrane insertion through a hydrophilic vestibule. Positively charged residues at the entrance to the vestibule function as a selectivity filter that uses charge-repulsion to reject mitochondrial TA proteins. Similarly, this selectivity filter retains the positively charged soluble domains of multipass substrates in the cytosol, thereby ensuring they adopt the correct topology and enforcing the "positive-inside" rule. Substrate discrimination by the EMC provides a biochemical explanation for one role of charge in TA protein sorting and protects compartment integrity by limiting protein misinsertion.
Collapse
Affiliation(s)
- Tino Pleiner
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, USA
| | - Masami Hazu
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, USA
| | - Giovani Pinton Tomaleri
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, USA
| | - Vy N. Nguyen
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, USA
| | - Kurt Januszyk
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, USA
| | - Rebecca M. Voorhees
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, USA
| |
Collapse
|
29
|
Bocancia-Mateescu LA, Stan D, Mirica AC, Ghita MG, Stan D, Ruta LL. Nanobodies as Diagnostic and Therapeutic Tools for Cardiovascular Diseases (CVDs). Pharmaceuticals (Basel) 2023; 16:863. [PMID: 37375810 PMCID: PMC10301117 DOI: 10.3390/ph16060863] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2023] [Revised: 06/06/2023] [Accepted: 06/07/2023] [Indexed: 06/29/2023] Open
Abstract
The aim of this review is to summarize some of the most recent work in the field of cardiovascular disease (CVD) diagnosis and therapy, focusing mainly on the role of nanobodies in the development of non-invasive imaging methods, diagnostic devices, and advanced biotechnological therapy tools. In the context of the increased number of people suffering from CVDs due to a variety of factors such as sedentariness, poor nutrition, stress, and smoking, there is an urgent need for new and improved diagnostic and therapeutic methods. Nanobodies can be easily produced in prokaryotes, lower eukaryotes, and plant and mammalian cells, and offer great advantages. In the diagnosis domain, they are mainly used as labeled probes that bind to certain surface receptors or other target molecules and give important information on the severity and extent of atherosclerotic lesions, using imaging methods such as contrast-enhanced ultrasound molecular imaging (CEUMI), positron emission tomography (PET), single-photon emission computed tomography coupled with computed tomography (SPECT/CT), and PET/CT. As therapy tools, nanobodies have been used either for transporting drug-loaded vesicles to specific targets or as inhibitors for certain enzymes and receptors, demonstrated to be involved in various CVDs.
Collapse
Affiliation(s)
| | - Dana Stan
- DDS Diagnostic, 7 Vulcan Judetu, 031427 Bucharest, Romania; (L.-A.B.-M.); (D.S.); (A.-C.M.); (M.G.G.); (D.S.)
- Medicine Doctoral School, Titu Maiorescu University, 031593 Bucharest, Romania
| | - Andreea-Cristina Mirica
- DDS Diagnostic, 7 Vulcan Judetu, 031427 Bucharest, Romania; (L.-A.B.-M.); (D.S.); (A.-C.M.); (M.G.G.); (D.S.)
- Advanced Polymer Materials Group, University Politehnica of Bucharest, 1–7 Gh. Polizu Street, 011061 Bucharest, Romania
| | - Miruna Gabriela Ghita
- DDS Diagnostic, 7 Vulcan Judetu, 031427 Bucharest, Romania; (L.-A.B.-M.); (D.S.); (A.-C.M.); (M.G.G.); (D.S.)
| | - Diana Stan
- DDS Diagnostic, 7 Vulcan Judetu, 031427 Bucharest, Romania; (L.-A.B.-M.); (D.S.); (A.-C.M.); (M.G.G.); (D.S.)
| | - Lavinia Liliana Ruta
- Faculty of Chemistry, University of Bucharest, 90-92 Panduri Street, 050663 Bucharest, Romania
| |
Collapse
|
30
|
Liang H, Ma Z, Wang Z, Zhong P, Li R, Jiang H, Zong X, Zhong C, Liu X, Liu P, Liu J, Zhu H, Liu R, Ding Y. Structural Insights into the Binding of Red Fluorescent Protein mCherry-Specific Nanobodies. Int J Mol Sci 2023; 24:ijms24086952. [PMID: 37108116 PMCID: PMC10138814 DOI: 10.3390/ijms24086952] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2023] [Revised: 03/31/2023] [Accepted: 04/06/2023] [Indexed: 04/29/2023] Open
Abstract
Red fluorescent proteins (RFPs) have broad applications in life science research, and the manipulation of RFPs using nanobodies can expand their potential uses. However, the structural information available for nanobodies that bind with RFPs is still insufficient. In this study, we cloned, expressed, purified, and crystallized complexes formed by mCherry with LaM1, LaM3, and LaM8. Then, we analyzed the biochemical properties of the complexes using mass spectrometry (MS), fluorescence-detected size exclusion chromatography (FSEC), isothermal titration calorimetry (ITC), and bio-layer interferometry (BLI) technology. We determined the crystal structure of mCherry-LaM1, mCherry-LaM3, and mCherry-LaM8, with resolutions of 2.05 Å, 3.29 Å, and 1.31 Å, respectively. In this study, we systematically compared various parameters of several LaM series nanobodies, including LaM1, LaM3, and LaM8, with previously reported data on LaM2, LaM4, and LaM6, specifically examining their structural information. After designing multivalent tandem LaM1-LaM8 and LaM8-LaM4 nanobodies based on structural information, we characterized their properties, revealing their higher affinity and specificity to mCherry. Our research provides novel structural insights that could aid in understanding nanobodies targeting a specific target protein. This could provide a starting point for developing enhanced mCherry manipulation tools.
Collapse
Affiliation(s)
- Hui Liang
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Fudan University, Shanghai 200438, China
| | - Zhiqiang Ma
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Fudan University, Shanghai 200438, China
| | - Ziying Wang
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Fudan University, Shanghai 200438, China
| | - Peiyu Zhong
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Fudan University, Shanghai 200438, China
| | - Ran Li
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Fudan University, Shanghai 200438, China
| | - He Jiang
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Fudan University, Shanghai 200438, China
| | - Xin Zong
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Fudan University, Shanghai 200438, China
| | - Chao Zhong
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Fudan University, Shanghai 200438, China
| | - Xihuan Liu
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Fudan University, Shanghai 200438, China
| | - Peng Liu
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Fudan University, Shanghai 200438, China
| | - Jiayuan Liu
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Fudan University, Shanghai 200438, China
| | - Haoran Zhu
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Fudan University, Shanghai 200438, China
| | - Rui Liu
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Fudan University, Shanghai 200438, China
| | - Yu Ding
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Fudan University, Shanghai 200438, China
| |
Collapse
|
31
|
Yong J, Mellick AS, Whitelock J, Wang J, Liang K. A Biomolecular Toolbox for Precision Nanomotors. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2023; 35:e2205746. [PMID: 36055646 DOI: 10.1002/adma.202205746] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/23/2022] [Revised: 08/31/2022] [Indexed: 06/15/2023]
Abstract
The application of nanomotors for cancer diagnosis and therapy is a new and exciting area of research, which when combined with precision nanomedicine, promises to solve many of the issues encountered by previous development of passive nanoparticles. The goal of this article is to introduce nanomotor and nanomedicine researchers to the deep pool of knowledge available regarding cancer cell biology and biochemistry, as well as provide a greater appreciation of the complexity of cell membrane compositions, extracellular surfaces, and their functional consequences. A short description of the nanomotor state-of-art for cancer therapy and diagnosis is first provided, as well as recommendations for future directions of the field. Then, a biomolecular targeting toolbox has been collated for researchers looking to apply their nanomaterial of choice to a biological setting, as well as providing a glimpse into currently available clinical therapies and technologies. This toolbox contains an overview of different classes of targeting molecules available for high affinity and specific targeting and cell surface targets to aid researchers in the selection of a clinical disease model and targeting methodology. It is hoped that this review will provide biological context, inspiration, and direction to future nanomotor and nanomedicine research.
Collapse
Affiliation(s)
- Joel Yong
- School of Chemical Engineering and Australian Centre for NanoMedicine, The University of New South Wales, Kensington, New South Wales, 2052, Australia
| | - Albert S Mellick
- Graduate School of Biomedical Engineering, The University of New South Wales, Kensington, New South Wales, 2052, Australia
- Ingham Institute for Applied Medical Research, Liverpool, New South Wales, 2170, Australia
| | - John Whitelock
- Graduate School of Biomedical Engineering, The University of New South Wales, Kensington, New South Wales, 2052, Australia
| | - Joseph Wang
- Department of Nanoengineering, University of California San Diego, La Jolla, CA, 92093, USA
| | - Kang Liang
- School of Chemical Engineering and Australian Centre for NanoMedicine, The University of New South Wales, Kensington, New South Wales, 2052, Australia
- Graduate School of Biomedical Engineering, The University of New South Wales, Kensington, New South Wales, 2052, Australia
| |
Collapse
|
32
|
van den Bor J, Bergkamp ND, Anbuhl SM, Dekker F, Comez D, Perez Almeria CV, Bosma R, White CW, Kilpatrick LE, Hill SJ, Siderius M, Smit MJ, Heukers R. NanoB 2 to monitor interactions of ligands with membrane proteins by combining nanobodies and NanoBRET. CELL REPORTS METHODS 2023; 3:100422. [PMID: 37056381 PMCID: PMC10088090 DOI: 10.1016/j.crmeth.2023.100422] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Revised: 01/31/2023] [Accepted: 02/17/2023] [Indexed: 03/14/2023]
Abstract
The therapeutic potential of ligands targeting disease-associated membrane proteins is predicted by ligand-receptor binding constants, which can be determined using NanoLuciferase (NanoLuc)-based bioluminescence resonance energy transfer (NanoBRET) methods. However, the broad applicability of these methods is hampered by the restricted availability of fluorescent probes. We describe the use of antibody fragments, like nanobodies, as universal building blocks for fluorescent probes for use in NanoBRET. Our nanobody-NanoBRET (NanoB2) workflow starts with the generation of NanoLuc-tagged receptors and fluorescent nanobodies, enabling homogeneous, real-time monitoring of nanobody-receptor binding. Moreover, NanoB2 facilitates the assessment of receptor binding of unlabeled ligands in competition binding experiments. The broad significance is illustrated by the successful application of NanoB2 to different drug targets (e.g., multiple G protein-coupled receptors [GPCRs] and a receptor tyrosine kinase [RTK]) at distinct therapeutically relevant binding sites (i.e., extracellular and intracellular).
Collapse
Affiliation(s)
- Jelle van den Bor
- Receptor Biochemistry and Signaling group, Division of Medicinal Chemistry, Amsterdam Institute for Molecular and Life Science (AIMMS), Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
| | - Nick D. Bergkamp
- Receptor Biochemistry and Signaling group, Division of Medicinal Chemistry, Amsterdam Institute for Molecular and Life Science (AIMMS), Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
| | - Stephanie M. Anbuhl
- Receptor Biochemistry and Signaling group, Division of Medicinal Chemistry, Amsterdam Institute for Molecular and Life Science (AIMMS), Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
- QVQ Holding B.V., Utrecht, the Netherlands
| | - Françoise Dekker
- Receptor Biochemistry and Signaling group, Division of Medicinal Chemistry, Amsterdam Institute for Molecular and Life Science (AIMMS), Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
| | - Dehan Comez
- Cell Signalling Research Group, Division of Physiology, Pharmacology and Neuroscience, School of Life Sciences, University of Nottingham, Nottingham, UK
- Centre of Membrane Proteins and Receptors (COMPARE), University of Birmingham and University of Nottingham, the Midlands, UK
| | - Claudia V. Perez Almeria
- Receptor Biochemistry and Signaling group, Division of Medicinal Chemistry, Amsterdam Institute for Molecular and Life Science (AIMMS), Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
| | - Reggie Bosma
- Receptor Biochemistry and Signaling group, Division of Medicinal Chemistry, Amsterdam Institute for Molecular and Life Science (AIMMS), Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
| | - Carl W. White
- Cell Signalling Research Group, Division of Physiology, Pharmacology and Neuroscience, School of Life Sciences, University of Nottingham, Nottingham, UK
- Centre of Membrane Proteins and Receptors (COMPARE), University of Birmingham and University of Nottingham, the Midlands, UK
| | - Laura E. Kilpatrick
- Centre of Membrane Proteins and Receptors (COMPARE), University of Birmingham and University of Nottingham, the Midlands, UK
- Division of Bimolecular Science and Medicinal Chemistry, School of Pharmacy, Biodiscovery Institute, University of Nottingham, Nottingham, UK
| | - Stephen J. Hill
- Cell Signalling Research Group, Division of Physiology, Pharmacology and Neuroscience, School of Life Sciences, University of Nottingham, Nottingham, UK
- Centre of Membrane Proteins and Receptors (COMPARE), University of Birmingham and University of Nottingham, the Midlands, UK
| | - Marco Siderius
- Receptor Biochemistry and Signaling group, Division of Medicinal Chemistry, Amsterdam Institute for Molecular and Life Science (AIMMS), Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
| | - Martine J. Smit
- Receptor Biochemistry and Signaling group, Division of Medicinal Chemistry, Amsterdam Institute for Molecular and Life Science (AIMMS), Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
| | - Raimond Heukers
- Receptor Biochemistry and Signaling group, Division of Medicinal Chemistry, Amsterdam Institute for Molecular and Life Science (AIMMS), Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
- QVQ Holding B.V., Utrecht, the Netherlands
| |
Collapse
|
33
|
Remmel M, Scheiderer L, Butkevich AN, Bossi ML, Hell SW. Accelerated MINFLUX Nanoscopy, through Spontaneously Fast-Blinking Fluorophores. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2023; 19:e2206026. [PMID: 36642798 DOI: 10.1002/smll.202206026] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Revised: 11/24/2022] [Indexed: 06/17/2023]
Abstract
The introduction of MINFLUX nanoscopy allows single molecules to be localized with one nanometer precision in as little as one millisecond. However, current applications have so far focused on increasing this precision by optimizing photon collection, rather than minimizing the localization time. Concurrently, commonly used fluorescent switches are specifically designed for stochastic methods (e.g., STORM), optimized for a high photon yield and rather long on-times (tens of milliseconds). Here, accelerated MINFLUX nanoscopy with up to a 30-fold gain in localization speed is presented. The improvement is attained by designing spontaneously blinking fluorescent markers with remarkably fast on-times, down to 1-3 ms, matching the iterative localization process used in a MINFLUX microscope. This design utilizes a silicon rhodamine amide core, shifting the spirocyclization equilibrium toward an uncharged closed form at physiological conditions and imparting intact live cell permeability, modified with a fused (benzo)thiophene spirolactam fragment. The best candidate for MINFLUX microscopy (also suitable for STORM imaging) is selected through detailed characterization of the blinking behavior of single fluorophores, bound to different protein tags. Finally, optimization of the localization routines, customized to the fast blinking times, renders a significant speed improvement on a commercial MINFLUX microscope.
Collapse
Affiliation(s)
- Michael Remmel
- Department of Optical Nanoscopy, Max Planck Institute for Medical Research, Jahnstrasse 29, 69120, Heidelberg, Germany
| | - Lukas Scheiderer
- Department of Optical Nanoscopy, Max Planck Institute for Medical Research, Jahnstrasse 29, 69120, Heidelberg, Germany
| | - Alexey N Butkevich
- Department of NanoBiophotonics, Max Planck Institute for Multidisciplinary Sciences, Am Fassberg 11, 37077, Göttingen, Germany
| | - Mariano L Bossi
- Department of NanoBiophotonics, Max Planck Institute for Multidisciplinary Sciences, Am Fassberg 11, 37077, Göttingen, Germany
| | - Stefan W Hell
- Department of Optical Nanoscopy, Max Planck Institute for Medical Research, Jahnstrasse 29, 69120, Heidelberg, Germany
- Department of NanoBiophotonics, Max Planck Institute for Multidisciplinary Sciences, Am Fassberg 11, 37077, Göttingen, Germany
| |
Collapse
|
34
|
Loreau V, Rees R, Chan EH, Taxer W, Gregor K, Mußil B, Pitaval C, Luis NM, Mangeol P, Schnorrer F, Görlich D. A nanobody toolbox to investigate localisation and dynamics of Drosophila titins and other key sarcomeric proteins. eLife 2023; 12:79343. [PMID: 36645120 PMCID: PMC9886281 DOI: 10.7554/elife.79343] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2022] [Accepted: 12/16/2022] [Indexed: 01/17/2023] Open
Abstract
Measuring the positions and dynamics of proteins in intact tissues or whole animals is key to understanding protein function. However, to date, this is challenging, as the accessibility of large antibodies to dense tissues is often limited, and fluorescent proteins inserted close to a domain of interest may affect protein function. These complications apply in particular to muscle sarcomeres, arguably one of the most protein-dense assemblies in nature, which complicates studying sarcomere morphogenesis at molecular resolution. Here, we introduce a toolbox of nanobodies recognising various domains of the two Drosophila titin homologs, Sallimus and Projectin, as well as the key sarcomeric proteins Obscurin, α-Actinin, and Zasp52. We verified the superior labelling qualities of our nanobodies in muscle tissue as compared to antibodies. By applying our toolbox to larval muscles, we found a gigantic Sallimus isoform stretching more than 2 µm to bridge the sarcomeric I-band, while Projectin covers almost the entire myosin filaments in a polar orientation. Transgenic expression of tagged nanobodies confirmed their high affinity-binding without affecting target protein function. Finally, adding a degradation signal to anti-Sallimus nanobodies suggested that it is difficult to fully degrade Sallimus in mature sarcomeres; however, expression of these nanobodies caused developmental lethality. These results may inspire the generation of similar toolboxes for other large protein complexes in Drosophila or mammals.
Collapse
Affiliation(s)
- Vincent Loreau
- Turing Centre for Living Systems, Aix-Marseille University, CNRS, IDBMMarseilleFrance
| | - Renate Rees
- Department of Cellular Logistics, Max Planck Institute for Multidisciplinary SciencesGöttingenGermany
| | - Eunice HoYee Chan
- Turing Centre for Living Systems, Aix-Marseille University, CNRS, IDBMMarseilleFrance
| | - Waltraud Taxer
- Department of Cellular Logistics, Max Planck Institute for Multidisciplinary SciencesGöttingenGermany
| | - Kathrin Gregor
- Department of Cellular Logistics, Max Planck Institute for Multidisciplinary SciencesGöttingenGermany
| | - Bianka Mußil
- Department of Cellular Logistics, Max Planck Institute for Multidisciplinary SciencesGöttingenGermany
| | - Christophe Pitaval
- Turing Centre for Living Systems, Aix-Marseille University, CNRS, IDBMMarseilleFrance
| | - Nuno Miguel Luis
- Turing Centre for Living Systems, Aix-Marseille University, CNRS, IDBMMarseilleFrance
| | - Pierre Mangeol
- Turing Centre for Living Systems, Aix-Marseille University, CNRS, IDBMMarseilleFrance
| | - Frank Schnorrer
- Turing Centre for Living Systems, Aix-Marseille University, CNRS, IDBMMarseilleFrance
| | - Dirk Görlich
- Department of Cellular Logistics, Max Planck Institute for Multidisciplinary SciencesGöttingenGermany
| |
Collapse
|
35
|
Schueder F, Mangeol P, Chan EH, Rees R, Schünemann J, Jungmann R, Görlich D, Schnorrer F. Nanobodies combined with DNA-PAINT super-resolution reveal a staggered titin nanoarchitecture in flight muscles. eLife 2023; 12:e79344. [PMID: 36645127 PMCID: PMC9886278 DOI: 10.7554/elife.79344] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2022] [Accepted: 11/22/2022] [Indexed: 01/17/2023] Open
Abstract
Sarcomeres are the force-producing units of all striated muscles. Their nanoarchitecture critically depends on the large titin protein, which in vertebrates spans from the sarcomeric Z-disc to the M-band and hence links actin and myosin filaments stably together. This ensures sarcomeric integrity and determines the length of vertebrate sarcomeres. However, the instructive role of titins for sarcomeric architecture outside of vertebrates is not as well understood. Here, we used a series of nanobodies, the Drosophila titin nanobody toolbox, recognising specific domains of the two Drosophila titin homologs Sallimus and Projectin to determine their precise location in intact flight muscles. By combining nanobodies with DNA-PAINT super-resolution microscopy, we found that, similar to vertebrate titin, Sallimus bridges across the flight muscle I-band, whereas Projectin is located at the beginning of the A-band. Interestingly, the ends of both proteins overlap at the I-band/A-band border, revealing a staggered organisation of the two Drosophila titin homologs. This architecture may help to stably anchor Sallimus at the myosin filament and hence ensure efficient force transduction during flight.
Collapse
Affiliation(s)
- Florian Schueder
- Faculty of Physics and Center for Nanoscience, Ludwig Maximilian UniversityMunichGermany
- Max Planck Institute of BiochemistryMartinsriedGermany
| | - Pierre Mangeol
- Aix Marseille University, CNRS, IBDM, Turing Centre for Living SystemsMarseilleFrance
| | - Eunice HoYee Chan
- Aix Marseille University, CNRS, IBDM, Turing Centre for Living SystemsMarseilleFrance
| | - Renate Rees
- Max Planck Institute for Multidisciplinary SciencesGöttingenGermany
| | | | - Ralf Jungmann
- Faculty of Physics and Center for Nanoscience, Ludwig Maximilian UniversityMunichGermany
- Max Planck Institute of BiochemistryMartinsriedGermany
| | - Dirk Görlich
- Max Planck Institute for Multidisciplinary SciencesGöttingenGermany
| | - Frank Schnorrer
- Aix Marseille University, CNRS, IBDM, Turing Centre for Living SystemsMarseilleFrance
| |
Collapse
|
36
|
Kumar MS, Fowler-Magaw ME, Kulick D, Boopathy S, Gadd DH, Rotunno M, Douthwright C, Golebiowski D, Yusuf I, Xu Z, Brown RH, Sena-Esteves M, O’Neil AL, Bosco DA. Anti-SOD1 Nanobodies That Stabilize Misfolded SOD1 Proteins Also Promote Neurite Outgrowth in Mutant SOD1 Human Neurons. Int J Mol Sci 2022; 23:ijms232416013. [PMID: 36555655 PMCID: PMC9784173 DOI: 10.3390/ijms232416013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2022] [Revised: 12/09/2022] [Accepted: 12/11/2022] [Indexed: 12/23/2022] Open
Abstract
ALS-linked mutations induce aberrant conformations within the SOD1 protein that are thought to underlie the pathogenic mechanism of SOD1-mediated ALS. Although clinical trials are underway for gene silencing of SOD1, these approaches reduce both wild-type and mutated forms of SOD1. Here, we sought to develop anti-SOD1 nanobodies with selectivity for mutant and misfolded forms of human SOD1 over wild-type SOD1. Characterization of two anti-SOD1 nanobodies revealed that these biologics stabilize mutant SOD1 in vitro. Further, SOD1 expression levels were enhanced and the physiological subcellular localization of mutant SOD1 was restored upon co-expression of anti-SOD1 nanobodies in immortalized cells. In human motor neurons harboring the SOD1 A4V mutation, anti-SOD1 nanobody expression promoted neurite outgrowth, demonstrating a protective effect of anti-SOD1 nanobodies in otherwise unhealthy cells. In vitro assays revealed that an anti-SOD1 nanobody exhibited selectivity for human mutant SOD1 over endogenous murine SOD1, thus supporting the preclinical utility of anti-SOD1 nanobodies for testing in animal models of ALS. In sum, the anti-SOD1 nanobodies developed and presented herein represent viable biologics for further preclinical testing in human and mouse models of ALS.
Collapse
Affiliation(s)
- Meenakshi Sundaram Kumar
- Department of Neurology, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA
- Biochemistry and Molecular Biotechnology Program, Morningside Graduate School of Biomedical Sciences, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA
| | - Megan E. Fowler-Magaw
- Department of Neurology, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA
- Neuroscience Program, Morningside Graduate School of Biomedical Sciences, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA
| | - Daniel Kulick
- Department of Biology, Neuroscience and Behavior Program, Wesleyan University, Middletown, CT 06459, USA
| | - Sivakumar Boopathy
- Department of Neurology, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA
- Biochemistry and Molecular Biotechnology Program, Morningside Graduate School of Biomedical Sciences, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA
| | - Del Hayden Gadd
- Department of Neurology, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA
| | - Melissa Rotunno
- Department of Neurology, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA
- Biochemistry and Molecular Biotechnology Program, Morningside Graduate School of Biomedical Sciences, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA
| | - Catherine Douthwright
- Department of Neurology, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA
- Biochemistry and Molecular Biotechnology Program, Morningside Graduate School of Biomedical Sciences, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA
| | - Diane Golebiowski
- Department of Neurology, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA
- Horae Gene Therapy Center, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA
| | - Issa Yusuf
- Department of Biochemistry and Molecular Biotechnology, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA
| | - Zuoshang Xu
- Department of Biochemistry and Molecular Biotechnology, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA
| | - Robert H. Brown
- Department of Neurology, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA
| | - Miguel Sena-Esteves
- Department of Neurology, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA
- Horae Gene Therapy Center, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA
| | - Alison L. O’Neil
- Department of Chemistry, Neuroscience and Behavior Program, Wesleyan University, Middletown, CT 06459, USA
| | - Daryl A. Bosco
- Department of Neurology, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA
- Department of Biochemistry and Molecular Biotechnology, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA
- Correspondence: ; Tel.: +1-(774)-445-3745; Fax: +1-(508)-856-6750
| |
Collapse
|
37
|
Modular Site-Specific Conjugation of Nanobodies Using Two Co-Associating Tags. Int J Mol Sci 2022; 23:ijms232214405. [PMID: 36430882 PMCID: PMC9696751 DOI: 10.3390/ijms232214405] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2022] [Revised: 11/12/2022] [Accepted: 11/17/2022] [Indexed: 11/22/2022] Open
Abstract
The homogeneous labeling of antibodies and their fragments is a critical step for the generation of robust probes used in immuno-detection applications. To date, numerous chemical, genetic and peptide-based site-specific coupling methods have been developed. Among these methods, co-assembling peptide-tags is one of the most straightforward and versatile solutions. Here, we describe site-specific labeling of nanobodies through the use of two co-associating peptides tags, E3 and K3, originating from the tetramerization domain of p53. These E3 and K3-tags provide a simple and robust method for associating stoichiometric amount of VHH and fluorescent probes, either fluorescent proteins or fluorochromes, at specific positions. As a proof of concept, a nanobody targeting the human epidermal growth factor receptor 2 (HER2), the nano-HER2 was genetically fused to the E3 and associated with different fluorescent K3-derivates. Entities were produced separately in Escherichia coli in soluble forms at high yields and co-assembled in vitro. These molecular probes present high binding specificity on HER2-overexpressing cells in flow-cytometry with relative binding constants in the low nanomolar range and are stable enough to stain HER2-receptor on living cells followed detection using fluorescent confocal microscopy. Altogether, our results demonstrate that the non-covalent conjugation method using these two co-associating peptides can be easily implemented for the modular engineering of molecular probes for cell immuno-staining.
Collapse
|
38
|
Zhang Q, Miyamoto A, Watanabe S, Arimori T, Sakai M, Tomisaki M, Kiuchi T, Takagi J, Watanabe N. Engineered fast-dissociating antibody fragments for multiplexed super-resolution microscopy. CELL REPORTS METHODS 2022; 2:100301. [PMID: 36313806 PMCID: PMC9606137 DOI: 10.1016/j.crmeth.2022.100301] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/11/2022] [Revised: 07/07/2022] [Accepted: 08/31/2022] [Indexed: 05/22/2023]
Abstract
Image reconstruction by integrating exchangeable single-molecule localization (IRIS) achieves multiplexed super-resolution imaging by high-density labeling with fast exchangeable fluorescent probes. However, previous methods to develop probes for individual targets required a great amount of time and effort. Here, we introduce a method for generating recombinant IRIS probes with a new mutagenesis strategy that can be widely applied to existing antibody sequences. Several conserved tyrosine residues at the base of complementarity-determining regions were identified as candidate sites for site-directed mutagenesis. With a high probability, mutations at candidate sites accelerated the off rate of recombinant antibody-based probes without compromising specific binding. We were able to develop IRIS probes from five monoclonal antibodies and three single-domain antibodies. We demonstrate multiplexed localization of endogenous proteins in primary neurons that visualizes small synaptic connections with high binding density. It is now practically feasible to generate fast-dissociating fluorescent probes for multitarget super-resolution imaging.
Collapse
Affiliation(s)
- Qianli Zhang
- Laboratory of Single-Molecule Cell Biology, Kyoto University Graduate School of Biostudies, Kyoto 606-8501, Japan
| | - Akitoshi Miyamoto
- Laboratory of Single-Molecule Cell Biology, Kyoto University Graduate School of Biostudies, Kyoto 606-8501, Japan
| | - Shin Watanabe
- Laboratory of Single-Molecule Cell Biology, Kyoto University Graduate School of Biostudies, Kyoto 606-8501, Japan
| | - Takao Arimori
- Institute for Protein Research, Osaka University, Suita, Osaka 565-0871, Japan
| | - Masanori Sakai
- Kyoto University Faculty of Engineering, Kyoto 606-8317, Japan
| | - Madoka Tomisaki
- Laboratory of Single-Molecule Cell Biology, Kyoto University Graduate School of Biostudies, Kyoto 606-8501, Japan
| | - Tai Kiuchi
- Department of Pharmacology, Kyoto University Graduate School of Medicine, Kyoto 606-8501, Japan
| | - Junichi Takagi
- Institute for Protein Research, Osaka University, Suita, Osaka 565-0871, Japan
| | - Naoki Watanabe
- Laboratory of Single-Molecule Cell Biology, Kyoto University Graduate School of Biostudies, Kyoto 606-8501, Japan
- Department of Pharmacology, Kyoto University Graduate School of Medicine, Kyoto 606-8501, Japan
| |
Collapse
|
39
|
Guna A, Stevens TA, Inglis AJ, Replogle JM, Esantsi TK, Muthukumar G, Shaffer KCL, Wang ML, Pogson AN, Jones JJ, Lomenick B, Chou TF, Weissman JS, Voorhees RM. MTCH2 is a mitochondrial outer membrane protein insertase. Science 2022; 378:317-322. [PMID: 36264797 PMCID: PMC9674023 DOI: 10.1126/science.add1856] [Citation(s) in RCA: 78] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
In the mitochondrial outer membrane, α-helical transmembrane proteins play critical roles in cytoplasmic-mitochondrial communication. Using genome-wide CRISPR screens, we identified MTCH2, and its paralog MTCH1, and showed that it is required for insertion of biophysically diverse tail-anchored (TA), signal-anchored, and multipass proteins, but not outer membrane β-barrel proteins. Purified MTCH2 was sufficient to mediate insertion into reconstituted proteoliposomes. Functional and mutational studies suggested that MTCH2 has evolved from a solute carrier transporter. MTCH2 uses membrane-embedded hydrophilic residues to function as a gatekeeper for the outer membrane, controlling mislocalization of TAs into the endoplasmic reticulum and modulating the sensitivity of leukemia cells to apoptosis. Our identification of MTCH2 as an insertase provided a mechanistic explanation for the diverse phenotypes and disease states associated with MTCH2 dysfunction. We showed that MTCH2 was both necessary and sufficient for insertion of diverse α-helical proteins into the mitochondrial outer membrane, and was the defining member of a family of insertases that have co-opted the SLC25 transporter fold.
Collapse
Affiliation(s)
- Alina Guna
- Whitehead Institute for Biomedical Research, Massachusetts Institute of Technology, Cambridge, MA 02142, USA
| | - Taylor A Stevens
- Division of Biology and Biological Engineering, California Institute of Technology, 1200 East California Avenue, Pasadena, CA 91125, USA
| | - Alison J Inglis
- Division of Biology and Biological Engineering, California Institute of Technology, 1200 East California Avenue, Pasadena, CA 91125, USA
| | - Joseph M Replogle
- Whitehead Institute for Biomedical Research, Massachusetts Institute of Technology, Cambridge, MA 02142, USA.,Medical Scientist Training Program, University of California, San Francisco, San Francisco, CA 94158, USA.,Tetrad Graduate Program, University of California, San Francisco, San Francisco, CA 94158, USA.,Howard Hughes Medical Institute, Massachusetts Institute of Technology, Cambridge, MA 02142, USA
| | - Theodore K Esantsi
- Whitehead Institute for Biomedical Research, Massachusetts Institute of Technology, Cambridge, MA 02142, USA.,Howard Hughes Medical Institute, Massachusetts Institute of Technology, Cambridge, MA 02142, USA
| | - Gayathri Muthukumar
- Whitehead Institute for Biomedical Research, Massachusetts Institute of Technology, Cambridge, MA 02142, USA.,Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02142, USA
| | - Kelly C L Shaffer
- Division of Biology and Biological Engineering, California Institute of Technology, 1200 East California Avenue, Pasadena, CA 91125, USA
| | - Maxine L Wang
- Whitehead Institute for Biomedical Research, Massachusetts Institute of Technology, Cambridge, MA 02142, USA.,Division of Biology and Biological Engineering, California Institute of Technology, 1200 East California Avenue, Pasadena, CA 91125, USA.,Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02142, USA
| | - Angela N Pogson
- Whitehead Institute for Biomedical Research, Massachusetts Institute of Technology, Cambridge, MA 02142, USA.,Howard Hughes Medical Institute, Massachusetts Institute of Technology, Cambridge, MA 02142, USA
| | - Jeff J Jones
- Division of Biology and Biological Engineering, California Institute of Technology, 1200 East California Avenue, Pasadena, CA 91125, USA
| | - Brett Lomenick
- Division of Biology and Biological Engineering, California Institute of Technology, 1200 East California Avenue, Pasadena, CA 91125, USA
| | - Tsui-Fen Chou
- Division of Biology and Biological Engineering, California Institute of Technology, 1200 East California Avenue, Pasadena, CA 91125, USA
| | - Jonathan S Weissman
- Whitehead Institute for Biomedical Research, Massachusetts Institute of Technology, Cambridge, MA 02142, USA.,Howard Hughes Medical Institute, Massachusetts Institute of Technology, Cambridge, MA 02142, USA.,Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02142, USA.,David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02142, USA
| | - Rebecca M Voorhees
- Division of Biology and Biological Engineering, California Institute of Technology, 1200 East California Avenue, Pasadena, CA 91125, USA
| |
Collapse
|
40
|
A multifaceted strategy to improve recombinant expression and structural characterisation of a Trypanosoma invariant surface protein. Sci Rep 2022; 12:12706. [PMID: 35882923 PMCID: PMC9325691 DOI: 10.1038/s41598-022-16958-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2022] [Accepted: 07/19/2022] [Indexed: 11/16/2022] Open
Abstract
Identification of a protein minimal fragment amenable to crystallisation can be time- and labour intensive especially if large amounts are required and the protein has a complex fold and functionally important post-translational modifications. In addition, a lack of homologues and structural information can further complicate the design of a minimal expression construct. Recombinant expression in E. coli promises high yields, low costs and fast turnover times, but falls short for many extracellular, eukaryotic proteins. Eukaryotic expression systems provide an alternative but are costly, slow and require special handling and equipment. Using a member of a structurally uncharacterized, eukaryotic receptor family as an example we employ hydrogen–deuterium exchange mass spectrometry (HDX-MS) guided construct design in conjunction with truncation scanning and targeted expression host switching to identify a minimal expression construct that can be produced with high yields and moderate costs.
Collapse
|
41
|
Cordell P, Carrington G, Curd A, Parker F, Tomlinson D, Peckham M. Affimers and nanobodies as molecular probes and their applications in imaging. J Cell Sci 2022; 135:276020. [PMID: 35848463 PMCID: PMC9450889 DOI: 10.1242/jcs.259168] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Antibodies are the most widely used, traditional tool for labelling molecules in cells. In the past five to ten years, many new labelling tools have been developed with significant advantages over the traditional antibody. Here, we focus on nanobodies and the non-antibody binding scaffold proteins called Affimers. We explain how they are generated, selected and produced, and we describe how their small size, high binding affinity and specificity provides them with many advantages compared to antibodies. Of particular importance, their small size enables them to better penetrate dense cytoskeletal regions within cells, as well as tissues, providing them with specific advantage for super-resolution imaging, as they place the fluorophore with a few nanometres of the target protein being imaged. We expect these novel tools to be of broad interest to many cell biologists and anticipate them becoming the tools of choice for super-resolution imaging.
Collapse
|
42
|
Li B, Qin X, Mi LZ. Nanobodies: from structure to applications in non-injectable and bispecific biotherapeutic development. NANOSCALE 2022; 14:7110-7122. [PMID: 35535618 DOI: 10.1039/d2nr00306f] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
The increasing demand for convenient, miniaturized and multifunctional antibodies necessitates the development of novel antigen-recognition molecules for biological and medical studies. Nanobodies, the functional variable regions of camelid heavy-chain-only antibodies, as a new tool, complement the conventional antibodies and are in the stage of rapid development. The outstanding advantages of nanobodies include a stable structure, easy production, excellent water solubility, high affinity toward antigens and low immunogenicity. With promising application potential, nanobodies are now increasingly applied to various studies, including protein structure analysis, microscopic imaging, medical diagnosis, and drug development. The approval of the first nanobody drug Caplacizumab by the FDA disclosed the therapeutic potential of nanobodies. The outbreak of COVID-19 accelerated the development of nanobody drugs in non-injectable and bispecific biotherapeutic applications. Herein, we reviewed recent studies on the nanobody structure, screening and their applications in protein structure analysis and nanobody drugs, especially on non-injectable nanobody and bispecific nanobody development.
Collapse
Affiliation(s)
- Bingxuan Li
- School of Life Sciences, Tianjin University, No. 92 Weijin Road, Nankai District, Tianjin 300072, China.
| | - Xiaohong Qin
- School of Life Sciences, Tianjin University, No. 92 Weijin Road, Nankai District, Tianjin 300072, China.
| | - Li-Zhi Mi
- School of Life Sciences, Tianjin University, No. 92 Weijin Road, Nankai District, Tianjin 300072, China.
| |
Collapse
|
43
|
Awad RM, Meeus F, Ceuppens H, Ertveldt T, Hanssens H, Lecocq Q, Mateusiak L, Zeven K, Valenta H, De Groof TWM, De Vlaeminck Y, Krasniqi A, De Veirman K, Goyvaerts C, D'Huyvetter M, Hernot S, Devoogdt N, Breckpot K. Emerging applications of nanobodies in cancer therapy. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2022; 369:143-199. [PMID: 35777863 DOI: 10.1016/bs.ircmb.2022.03.010] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Cancer is a heterogeneous disease, requiring treatment tailored to the unique phenotype of the patient's tumor. Monoclonal antibodies (mAbs) and variants thereof have enabled targeted therapies to selectively target cancer cells. Cancer cell-specific mAbs have been used for image-guided surgery and targeted delivery of radionuclides or toxic agents, improving classical treatment strategies. Cancer cell-specific mAbs can further inhibit tumor cell growth or can stimulate immune-mediated destruction of cancer cells, a feature that has also been achieved through mAb-mediated manipulation of immune cells and pathways. Drawbacks of mAbs and their variants, together with the discovery of camelid heavy chain-only antibodies and the many advantageous features of their variable domains, referred to as VHHs, single domain antibodies or nanobodies (Nbs), resulted in the exploration of Nbs as an alternative targeting moiety. We therefore review the state-of-the-art as well as novel exploitation strategies of Nbs for targeted cancer therapy.
Collapse
Affiliation(s)
- Robin Maximilian Awad
- Laboratory for Molecular and Cellular Therapy, Department of Biomedical Sciences, Vrije Universiteit Brussel, Brussels, Belgium
| | - Fien Meeus
- Laboratory for Molecular and Cellular Therapy, Department of Biomedical Sciences, Vrije Universiteit Brussel, Brussels, Belgium
| | - Hannelore Ceuppens
- Laboratory for Molecular and Cellular Therapy, Department of Biomedical Sciences, Vrije Universiteit Brussel, Brussels, Belgium
| | - Thomas Ertveldt
- Laboratory for Molecular and Cellular Therapy, Department of Biomedical Sciences, Vrije Universiteit Brussel, Brussels, Belgium
| | - Heleen Hanssens
- In Vivo Cellular and Molecular Imaging Laboratory, Department of Medical Imaging, Vrije Universiteit Brussel, Brussels, Belgium
| | - Quentin Lecocq
- Laboratory for Molecular and Cellular Therapy, Department of Biomedical Sciences, Vrije Universiteit Brussel, Brussels, Belgium
| | - Lukasz Mateusiak
- In Vivo Cellular and Molecular Imaging Laboratory, Department of Medical Imaging, Vrije Universiteit Brussel, Brussels, Belgium
| | - Katty Zeven
- In Vivo Cellular and Molecular Imaging Laboratory, Department of Medical Imaging, Vrije Universiteit Brussel, Brussels, Belgium
| | - Hana Valenta
- Lab for Nanobiology, Department of Chemistry, KU Leuven, Leuven, Belgium
| | - Timo W M De Groof
- In Vivo Cellular and Molecular Imaging Laboratory, Department of Medical Imaging, Vrije Universiteit Brussel, Brussels, Belgium
| | - Yannick De Vlaeminck
- Laboratory for Molecular and Cellular Therapy, Department of Biomedical Sciences, Vrije Universiteit Brussel, Brussels, Belgium
| | - Ahmet Krasniqi
- In Vivo Cellular and Molecular Imaging Laboratory, Department of Medical Imaging, Vrije Universiteit Brussel, Brussels, Belgium
| | - Kim De Veirman
- Laboratory for Hematology and Immunology, Department of Biomedical Sciences, Vrije Universiteit Brussel, Brussels, Belgium
| | - Cleo Goyvaerts
- Laboratory for Molecular and Cellular Therapy, Department of Biomedical Sciences, Vrije Universiteit Brussel, Brussels, Belgium
| | - Matthias D'Huyvetter
- In Vivo Cellular and Molecular Imaging Laboratory, Department of Medical Imaging, Vrije Universiteit Brussel, Brussels, Belgium
| | - Sophie Hernot
- In Vivo Cellular and Molecular Imaging Laboratory, Department of Medical Imaging, Vrije Universiteit Brussel, Brussels, Belgium
| | - Nick Devoogdt
- In Vivo Cellular and Molecular Imaging Laboratory, Department of Medical Imaging, Vrije Universiteit Brussel, Brussels, Belgium
| | - Karine Breckpot
- Laboratory for Molecular and Cellular Therapy, Department of Biomedical Sciences, Vrije Universiteit Brussel, Brussels, Belgium.
| |
Collapse
|
44
|
Haueis L, Stech M, Kubick S. A Cell-free Expression Pipeline for the Generation and Functional Characterization of Nanobodies. Front Bioeng Biotechnol 2022; 10:896763. [PMID: 35573250 PMCID: PMC9096027 DOI: 10.3389/fbioe.2022.896763] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Accepted: 04/11/2022] [Indexed: 11/13/2022] Open
Abstract
Cell-free systems are well-established platforms for the rapid synthesis, screening, engineering and modification of all kinds of recombinant proteins ranging from membrane proteins to soluble proteins, enzymes and even toxins. Also within the antibody field the cell-free technology has gained considerable attention with respect to the clinical research pipeline including antibody discovery and production. Besides the classical full-length monoclonal antibodies (mAbs), so-called "nanobodies" (Nbs) have come into focus. A Nb is the smallest naturally-derived functional antibody fragment known and represents the variable domain (VHH, ∼15 kDa) of a camelid heavy-chain-only antibody (HCAb). Based on their nanoscale and their special structure, Nbs display striking advantages concerning their production, but also their characteristics as binders, such as high stability, diversity, improved tissue penetration and reaching of cavity-like epitopes. The classical way to produce Nbs depends on the use of living cells as production host. Though cell-based production is well-established, it is still time-consuming, laborious and hardly amenable for high-throughput applications. Here, we present for the first time to our knowledge the synthesis of functional Nbs in a standardized mammalian cell-free system based on Chinese hamster ovary (CHO) cell lysates. Cell-free reactions were shown to be time-efficient and easy-to-handle allowing for the "on demand" synthesis of Nbs. Taken together, we complement available methods and demonstrate a promising new system for Nb selection and validation.
Collapse
Affiliation(s)
- Lisa Haueis
- Fraunhofer Institute for Cell Therapy and Immunology (IZI), Branch Bioanalytics and Bioprocesses (IZI-BB), Potsdam, Germany.,Institute of Biochemistry and Biology, University of Potsdam, Potsdam, Germany
| | - Marlitt Stech
- Fraunhofer Institute for Cell Therapy and Immunology (IZI), Branch Bioanalytics and Bioprocesses (IZI-BB), Potsdam, Germany
| | - Stefan Kubick
- Fraunhofer Institute for Cell Therapy and Immunology (IZI), Branch Bioanalytics and Bioprocesses (IZI-BB), Potsdam, Germany.,Institute of Chemistry and Biochemistry, Freie Universität Berlin, Berlin, Germany.,Faculty of Health Sciences, Joint Faculty of the Brandenburg University of Technology Cottbus-Senftenberg, The Brandenburg Medical School Theodor Fontane and the University of Potsdam, Potsdam, Germany
| |
Collapse
|
45
|
Bas AL, Mikolajek H, Huo J, Dormon J, Naismith JH, Owens RJ, Owens R. Production and Crystallization of Nanobodies in Complex with the Receptor Binding Domain of the SARS-CoV-2 Spike Protein. Bio Protoc 2022; 12:e4406. [PMID: 35800465 PMCID: PMC9090526 DOI: 10.21769/bioprotoc.4406] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2022] [Accepted: 03/17/2022] [Indexed: 12/29/2022] Open
Abstract
The receptor binding domain (RBD) of the spike protein of SARS-CoV-2 binds angiotensin converting enzyme-2 (ACE-2) on the surface of epithelial cells, leading to fusion, and entry of the virus into the cell. This interaction can be blocked by the binding of llama-derived nanobodies (VHHs) to the RBD, leading to virus neutralisation. Structural analysis of VHH-RBD complexes by X-ray crystallography enables VHH epitopes to be precisely mapped, and the effect of variant mutations to be interpreted and predicted. Key to this is a protocol for the reproducible production and crystallization of the VHH-RBD complexes. Based on our experience, we describe a workflow for expressing and purifying the proteins, and the screening conditions for generating diffraction quality crystals of VHH-RBD complexes. Production and crystallization of protein complexes takes approximately twelve days, from construction of vectors to harvesting and freezing crystals for data collection.
Collapse
Affiliation(s)
- Audrey Le Bas
- Structural Biology, The Rosalind Franklin Institute, Harwell Science Campus, UK
| | | | - Jiandong Huo
- Structural Biology, The Rosalind Franklin Institute, Harwell Science Campus, UK
,Division of Structural Biology, The Wellcome Centre for Human Genetics, University of Oxford
| | - Joshua Dormon
- Structural Biology, The Rosalind Franklin Institute, Harwell Science Campus, UK
| | - James H. Naismith
- Structural Biology, The Rosalind Franklin Institute, Harwell Science Campus, UK
,Division of Structural Biology, The Wellcome Centre for Human Genetics, University of Oxford
,
*For correspondence: ;
| | - Raymond J. Owens
- Structural Biology, The Rosalind Franklin Institute, Harwell Science Campus, UK
,Division of Structural Biology, The Wellcome Centre for Human Genetics, University of Oxford
,
*For correspondence: ;
| | | |
Collapse
|
46
|
Tagliatti E, Cortese K. Imaging Endocytosis Dynamics in Health and Disease. MEMBRANES 2022; 12:membranes12040393. [PMID: 35448364 PMCID: PMC9028293 DOI: 10.3390/membranes12040393] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Revised: 03/16/2022] [Accepted: 03/29/2022] [Indexed: 02/06/2023]
Abstract
Endocytosis is a critical process for cell growth and viability. It mediates nutrient uptake, guarantees plasma membrane homeostasis, and generates intracellular signaling cascades. Moreover, it plays an important role in dead cell clearance and defense against external microbes. Finally, endocytosis is an important cellular route for the delivery of nanomedicines for therapeutic treatments. Thus, it is not surprising that both environmental and genetic perturbation of endocytosis have been associated with several human conditions such as cancer, neurological disorders, and virus infections, among others. Over the last decades, a lot of research has been focused on developing advanced imaging methods to monitor endocytosis events with high resolution in living cells and tissues. These include fluorescence imaging, electron microscopy, and correlative and super-resolution microscopy. In this review, we outline the major endocytic pathways and briefly discuss how defects in the molecular machinery of these pathways lead to disease. We then discuss the current imaging methodologies used to study endocytosis in different contexts, highlighting strengths and weaknesses.
Collapse
Affiliation(s)
- Erica Tagliatti
- Laboratory of Pharmacology and Brain Pathology, Humanitas Clinical and Research Center, Via Manzoni 56, 20089 Milano, Italy
- Department of Clinical and Experimental Epilepsy, UCL Queen Square Institute of Neurology, University College London, London WC1E 6BT, UK
- Correspondence: (E.T.); (K.C.)
| | - Katia Cortese
- Cellular Electron Microscopy Laboratory, Department of Experimental Medicine (DIMES), Human Anatomy, Università di Genova, Via Antonio de Toni 14, 16132 Genova, Italy
- Correspondence: (E.T.); (K.C.)
| |
Collapse
|
47
|
Wu Y, Li Q, Kong Y, Wang Z, Lei C, Li J, Ding L, Wang C, Cheng Y, Wei Y, Song Y, Yang Z, Tu C, Ding Y, Ying T. A highly stable human single-domain antibody-drug conjugate exhibits superior penetration and treatment of solid tumors. Mol Ther 2022; 30:2785-2799. [PMID: 35462042 PMCID: PMC9372316 DOI: 10.1016/j.ymthe.2022.04.013] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Revised: 03/04/2022] [Accepted: 04/18/2022] [Indexed: 10/18/2022] Open
Abstract
The inefficient tumor penetration of therapeutic antibodies has hampered their effective use in treating solid tumors. Here, we report the identification of a fully human single-domain antibody (UdAb), designated as n501, targeting the oncofetal antigen 5T4. The high-resolution crystal structure indicates that n501 adopts a compact structure very similar to that of camelid nanobodies, and binds tightly to all eight leucine-rich repeats of 5T4. Furthermore, the UdAb n501 exhibits exceptionally high stability, with no apparent activity changes over 4 weeks of storage at various temperatures. Importantly, the UdAb-based antibody-drug conjugate (n501-SN38) showed much deeper tumor penetration, significantly higher tumor uptake, and faster accumulation at tumor sites than conventional IgG1-based antibody-drug conjugate (m603-SN38), resulting in improved tumor inhibition. These results highlight the potential of UdAb-based antibody-drug conjugates as a potential class of antitumor therapeutics with characteristics of high stability and strong tumor penetration for the effective treatment of solid tumors.
Collapse
|
48
|
Yang E, Liu Q, Huang G, Liu J, Wei W. Engineering nanobodies for next-generation molecular imaging. Drug Discov Today 2022; 27:1622-1638. [PMID: 35331925 DOI: 10.1016/j.drudis.2022.03.013] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2021] [Revised: 02/04/2022] [Accepted: 03/17/2022] [Indexed: 12/12/2022]
Abstract
In recent years, nanobodies have emerged as ideal imaging agents for molecular imaging. Molecular nanobody imaging combines the specificity of nanobodies with the sensitivity of state-of-the-art molecular imaging modalities, such as positron emission tomography (PET). Given that modifications of nanobodies alter their pharmacokinetics (PK), the engineering strategies that combine nanobodies with radionuclides determine the effectiveness, reliability, and safety of the molecular imaging probes. In this review, we introduce conjugation strategies that have been applied to nanobodies, including random conjugation, 99mTc tricarbonyl chemistry, sortase A-mediated site-specific conjugation, maleimide-cysteine chemistry, and click chemistries. We also summarize the latest advances in nanobody tracers, emphasizing their preclinical and clinical use. In addition, we elaborate on nanobody-based near-infrared fluorescence (NIRF) imaging and image-guided surgery.
Collapse
Affiliation(s)
- Erpeng Yang
- Department of Nuclear Medicine, Institute of Clinical Nuclear Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200217, China
| | - Qiufang Liu
- Department of Nuclear Medicine, Fudan University Shanghai Cancer Center, Fudan University, Shanghai 200032, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Gang Huang
- Department of Nuclear Medicine, Institute of Clinical Nuclear Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200217, China
| | - Jianjun Liu
- Department of Nuclear Medicine, Institute of Clinical Nuclear Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200217, China.
| | - Weijun Wei
- Department of Nuclear Medicine, Institute of Clinical Nuclear Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200217, China.
| |
Collapse
|
49
|
Ji F, Ren J, Vincke C, Jia L, Muyldermans S. Nanobodies: From Serendipitous Discovery of Heavy Chain-Only Antibodies in Camelids to a Wide Range of Useful Applications. METHODS IN MOLECULAR BIOLOGY (CLIFTON, N.J.) 2022; 2446:3-17. [PMID: 35157266 DOI: 10.1007/978-1-0716-2075-5_1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
The presence of unique heavy chain-only antibodies (HCAbs) in camelids was discovered at Vrije Universiteit Brussel (VUB, Brussels, Belgium) at a time when many researchers were exploring the cloning and expression of smaller antigen-binding fragments (Fv and Fab) from hybridoma-derived antibodies. The potential importance of this discovery was anticipated, and efforts were immediately undertaken to understand the emergence and ontogeny of these HCAbs as well as to investigate the applications of the single-domain antigen-binding variable domains of HCAbs (nanobodies). Nanobodies were demonstrated to possess multiple biochemical and biophysical advantages over other antigen-binding antibody fragments and alternative scaffolds. Today, nanobodies have a significant and growing impact on research, biotechnology, and medicine.
Collapse
Affiliation(s)
- Fangling Ji
- Liaoning Key Laboratory of Molecular Recognition and Imaging, School of Bioengineering, Dalian University of Technology, Dalian, Liaoning, China
| | - Jun Ren
- Liaoning Key Laboratory of Molecular Recognition and Imaging, School of Bioengineering, Dalian University of Technology, Dalian, Liaoning, China
| | - Cécile Vincke
- Cellular and Molecular Immunology Laboratory, Vrije Universiteit Brussel, Brussels, Belgium.,Myeloid Cell Immunology Laboratory, VIB Center for Inflammation Research, Brussels, Belgium
| | - Lingyun Jia
- Liaoning Key Laboratory of Molecular Recognition and Imaging, School of Bioengineering, Dalian University of Technology, Dalian, Liaoning, China
| | - Serge Muyldermans
- Liaoning Key Laboratory of Molecular Recognition and Imaging, School of Bioengineering, Dalian University of Technology, Dalian, Liaoning, China. .,Cellular and Molecular Immunology Laboratory, Vrije Universiteit Brussel, Brussels, Belgium.
| |
Collapse
|
50
|
Unterauer EM, Jungmann R. Quantitative Imaging With DNA-PAINT for Applications in Synaptic Neuroscience. Front Synaptic Neurosci 2022; 13:798267. [PMID: 35197837 PMCID: PMC8860300 DOI: 10.3389/fnsyn.2021.798267] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2021] [Accepted: 12/21/2021] [Indexed: 12/02/2022] Open
Abstract
Super-resolution (SR) microscopy techniques have been advancing the understanding of neuronal protein networks and interactions. Unraveling the arrangement of proteins with molecular resolution provided novel insights into neuron cytoskeleton structure and actin polymerization dynamics in synaptic spines. Recent improvements in quantitative SR imaging have been applied to synaptic protein clusters and with improved multiplexing technology, the interplay of multiple protein partners in synaptic active zones has been elucidated. While all SR techniques come with benefits and drawbacks, true molecular quantification is a major challenge with the most complex requirements for labeling reagents and careful experimental design. In this perspective, we provide an overview of quantitative SR multiplexing and discuss in greater detail the quantification and multiplexing capabilities of the SR technique DNA-PAINT. Using predictable binding kinetics of short oligonucleotides, DNA-PAINT provides two unique approaches to address multiplexed molecular quantification: qPAINT and Exchange-PAINT. With precise and accurate quantification and spectrally unlimited multiplexing, DNA-PAINT offers an attractive route to unravel complex protein interaction networks in neurons. Finally, while the SR community has been pushing technological advances from an imaging technique perspective, the development of universally available, small, efficient, and quantitative labels remains a major challenge in the field.
Collapse
Affiliation(s)
- Eduard M. Unterauer
- Max Planck Institute of Biochemistry, Martinsried, Germany
- Faculty of Physics and Center for Nanoscience, Ludwig Maximilian University, Munich, Germany
| | - Ralf Jungmann
- Max Planck Institute of Biochemistry, Martinsried, Germany
- Faculty of Physics and Center for Nanoscience, Ludwig Maximilian University, Munich, Germany
| |
Collapse
|