1
|
Redeghieri P, Moray J, Kerff F, Gohy S, Leal T, Muyldermans S, Vanbever R, Morales‐Yánez FJ, Dumoulin M. Enzymatic, structural, and biophysical characterization of a single-domain antibody (VHH) selectively and tightly inhibiting neutrophil elastase and exhibiting favorable developability properties. Protein Sci 2024; 33:e5227. [PMID: 39604162 PMCID: PMC11602439 DOI: 10.1002/pro.5227] [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: 05/10/2024] [Revised: 10/28/2024] [Accepted: 10/28/2024] [Indexed: 11/29/2024]
Abstract
Human neutrophil elastase (hNE), a serine protease released by neutrophils during inflammation, plays a major role in the pathophysiology of several conditions especially in inflammatory lung diseases. Its inhibition constitutes, therefore, a promising therapeutic strategy to combat these diseases. In this work, we characterized the in vitro properties of a VHH (i.e., the antigen binding domain of camelid heavy chain-only antibodies), referred to as NbE201. This VHH is able to inhibit tightly, selectively and competitively both human and murine elastases with the inhibition constants (Ki) of 4.1 ± 0.9 nM and 36.8 ± 3.9 nM, respectively. The IC50 for the inhibition of the hydrolysis of elastin is in the same range to that of alpha-1 antitrypsin (i.e., the main endogenous inhibitor of hNE also used in the clinic) and 14 times better than that of Sivelestat (i.e., the 2nd clinically approved hNE inhibitor). The X-ray crystal structure of the NbE201-hNE complex reveals that the Complementarity Determining Regions CDR1 and CDR3 of the VHH bind into the substrate binding pocket of hNE and prevent the access to small or macromolecular substrates. They do not, however, bind deep enough into the pocket to be hydrolyzed. NbE201 is highly stable towards oxidation, deamidation, and chemical or thermal denaturation. NbE201 is therefore likely to tolerate manufacturing processes during drug development. These results highlight the high potential of NbE201 as a (pre)clinical tool to diagnose and treat diseases associated with excessive hNE activity, and for fundamental research to better understand the role of hNE in these conditions.
Collapse
Affiliation(s)
- Paola Redeghieri
- Nano‐Antibodies to Explore Protein Structure and Functions (NEPTUNS)Centre for Protein Engineering, InBios, Department of Life Sciences, University of LiègeLiègeBelgium
| | - Joël Moray
- Nano‐Antibodies to Explore Protein Structure and Functions (NEPTUNS)Centre for Protein Engineering, InBios, Department of Life Sciences, University of LiègeLiègeBelgium
| | - Frédéric Kerff
- Biological Macromolecule Crystallography, Centre for Protein Engineering, InBios, Department of Life SciencesUniversity of LiègeLiègeBelgium
| | - Sophie Gohy
- Department of Pneumology, ENT and Dermatology, Institute of Experimental and Clinical Research (IREC)Université Catholique de LouvainBrusselsBelgium
- Cystic Fibrosis Reference CentreCliniques Universitaires Saint‐LucBrusselsBelgium
| | - Teresinha Leal
- Louvain Center for Toxicology and Applied Pharmacology (LTAP), Institute of Experimental and Clinical Research (IREC)Université Catholique de LouvainBrusselsBelgium
| | - Serge Muyldermans
- Laboratory of Cellular and Molecular ImmunologyVrije Universiteit BrusselBrusselsBelgium
| | - Rita Vanbever
- Louvain Drug Research Institute (LDRI)Université Catholique de LouvainBrusselsBelgium
| | - Francisco Javier Morales‐Yánez
- Nano‐Antibodies to Explore Protein Structure and Functions (NEPTUNS)Centre for Protein Engineering, InBios, Department of Life Sciences, University of LiègeLiègeBelgium
| | - Mireille Dumoulin
- Nano‐Antibodies to Explore Protein Structure and Functions (NEPTUNS)Centre for Protein Engineering, InBios, Department of Life Sciences, University of LiègeLiègeBelgium
| |
Collapse
|
2
|
Lagoutte P, Bourhis JM, Mariano N, Gueguen-Chaignon V, Vandroux D, Moali C, Vadon-Le Goff S. Mono- and Bi-specific Nanobodies Targeting the CUB Domains of PCPE-1 Reduce the Proteolytic Processing of Fibrillar Procollagens. J Mol Biol 2024; 436:168667. [PMID: 38901640 DOI: 10.1016/j.jmb.2024.168667] [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: 04/03/2024] [Revised: 06/13/2024] [Accepted: 06/13/2024] [Indexed: 06/22/2024]
Abstract
The excessive deposition of fibrillar collagens is a hallmark of fibrosis. Collagen fibril formation requires proteolytic maturations by Procollagen N- and C-proteinases (PNPs and PCPs) to remove the N- and C-propeptides which maintain procollagens in the soluble form. Procollagen C-Proteinase Enhancer-1 (PCPE-1, a glycoprotein composed of two CUB domains and one NTR domain) is a regulatory protein that activates the C-terminal processing of procollagens by the main PCPs. It is often up-regulated in fibrotic diseases and represents a promising target for the development of novel anti-fibrotic strategies. Here, our objective was to develop the first antagonists of PCPE-1, based on the nanobody scaffold. Using both an in vivo selection through the immunization of a llama and an in vitro selection with a synthetic library, we generated 18 nanobodies directed against the CUB domains of PCPE1, which carry its enhancing activity. Among them, I5 from the immune library and H4 from the synthetic library have a high affinity for PCPE-1 and inhibit its interaction with procollagens. The crystal structure of the complex formed by PCPE-1, H4 and I5 showed that they have distinct epitopes and enabled the design of a biparatopic fusion, the diabody diab-D1. Diab-D1 has a sub-nanomolar affinity for PCPE-1 and is a potent antagonist of its activity, preventing the stimulation of procollagen cleavage in vitro. Moreover, Diab-D1 is also effective in reducing the proteolytic maturation of procollagen I in cultures of human dermal fibroblasts and hence holds great promise as a tool to modulate collagen deposition in fibrotic conditions.
Collapse
Affiliation(s)
- Priscillia Lagoutte
- Universite Claude Bernard Lyon 1, CNRS, Tissue Biology and Therapeutic Engineering Laboratory, LBTI, UMR5305, F-69367 Lyon, France
| | - Jean-Marie Bourhis
- Institut de Biologie Structurale, University Grenoble Alpes, CEA, CNRS, F-38000 Grenoble, France
| | - Natacha Mariano
- Universite Claude Bernard Lyon 1, CNRS, Tissue Biology and Therapeutic Engineering Laboratory, LBTI, UMR5305, F-69367 Lyon, France
| | - Virginie Gueguen-Chaignon
- Protein Science Facility, SFR BioSciences, Univ Lyon, CNRS UAR3444, Inserm US8, ENS de Lyon, F-69367 Lyon, France
| | | | - Catherine Moali
- Universite Claude Bernard Lyon 1, CNRS, Tissue Biology and Therapeutic Engineering Laboratory, LBTI, UMR5305, F-69367 Lyon, France
| | - Sandrine Vadon-Le Goff
- Universite Claude Bernard Lyon 1, CNRS, Tissue Biology and Therapeutic Engineering Laboratory, LBTI, UMR5305, F-69367 Lyon, France.
| |
Collapse
|
3
|
Bigi A, Napolitano L, Vadukul DM, Chiti F, Cecchi C, Aprile FA, Cascella R. A single-domain antibody detects and neutralises toxic Aβ 42 oligomers in the Alzheimer's disease CSF. Alzheimers Res Ther 2024; 16:13. [PMID: 38238842 PMCID: PMC10795411 DOI: 10.1186/s13195-023-01361-z] [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: 10/04/2023] [Accepted: 11/29/2023] [Indexed: 01/22/2024]
Abstract
BACKGROUND Amyloid-β42 (Aβ42) aggregation consists of a complex chain of nucleation events producing soluble oligomeric intermediates, which are considered the major neurotoxic agents in Alzheimer's disease (AD). Cerebral lesions in the brain of AD patients start to develop 20 years before symptom onset; however, no preventive strategies, effective treatments, or specific and sensitive diagnostic tests to identify people with early-stage AD are currently available. In addition, the isolation and characterisation of neurotoxic Aβ42 oligomers are particularly difficult because of their transient and heterogeneous nature. To overcome this challenge, a rationally designed method generated a single-domain antibody (sdAb), named DesAb-O, targeting Aβ42 oligomers. METHODS We investigated the ability of DesAb-O to selectively detect preformed Aβ42 oligomers both in vitro and in cultured neuronal cells, by using dot-blot, ELISA immunoassay and super-resolution STED microscopy, and to counteract the toxicity induced by the oligomers, monitoring their interaction with neuronal membrane and the resulting mitochondrial impairment. We then applied this approach to CSF samples (CSFs) from AD patients as compared to age-matched control subjects. RESULTS DesAb-O was found to selectively detect synthetic Aβ42 oligomers both in vitro and in cultured cells, and to neutralise their associated neuronal dysfunction. DesAb-O can also identify Aβ42 oligomers present in the CSFs of AD patients with respect to healthy individuals, and completely prevent cell dysfunction induced by the administration of CSFs to neuronal cells. CONCLUSIONS Taken together, our data indicate a promising method for the improvement of an early diagnosis of AD and for the generation of novel therapeutic approaches based on sdAbs for the treatment of AD and other devastating neurodegenerative conditions.
Collapse
Affiliation(s)
- Alessandra Bigi
- Department of Experimental and Clinical Biomedical Sciences, Section of Biochemistry, University of Florence, Florence, Italy
| | - Liliana Napolitano
- Department of Experimental and Clinical Biomedical Sciences, Section of Biochemistry, University of Florence, Florence, Italy
| | - Devkee M Vadukul
- Department of Chemistry, Molecular Sciences Research Hub, Imperial College London, London, UK
| | - Fabrizio Chiti
- Department of Experimental and Clinical Biomedical Sciences, Section of Biochemistry, University of Florence, Florence, Italy
| | - Cristina Cecchi
- Department of Experimental and Clinical Biomedical Sciences, Section of Biochemistry, University of Florence, Florence, Italy
| | - Francesco A Aprile
- Department of Chemistry, Molecular Sciences Research Hub, Imperial College London, London, UK
- Institute of Chemical Biology, Molecular Sciences Research Hub, Imperial College London, London, UK
| | - Roberta Cascella
- Department of Experimental and Clinical Biomedical Sciences, Section of Biochemistry, University of Florence, Florence, Italy.
| |
Collapse
|
4
|
Mortelecque J, Danis C, Landrieu I, Dupré E. Recombinant Production and Characterization of VHHs/Nanobodies Targeting Tau to Block Fibrillar Assembly. Methods Mol Biol 2024; 2754:131-146. [PMID: 38512665 DOI: 10.1007/978-1-0716-3629-9_8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/23/2024]
Abstract
Tau protein was extensively studied using nuclear magnetic resonance spectroscopy, providing a powerful way to determine interaction sites between Tau and partner proteins. Here we used this analytical tool to describe the epitopes of Tau-specific VHHs (variable domain of the heavy chain of the heavy chain-only antibodies, aka nanobodies) selected from a synthetic library. An in vitro Tau aggregation assay was subsequently used as a functional screen to check VHH efficacy as aggregation inhibitors. We have observed a correlation between the targeted epitope and the aggregation-inhibition capacity of a series of Tau-specific VHHs.
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
- LabEx (Laboratory of Excellence) DISTALZ (Development of Innovative Strategies for a Transdisciplinary Approach to Alzheimer's Disease ANR-11-LABX-01), 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
- LabEx (Laboratory of Excellence) DISTALZ (Development of Innovative Strategies for a Transdisciplinary Approach to Alzheimer's Disease ANR-11-LABX-01), 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
- LabEx (Laboratory of Excellence) DISTALZ (Development of Innovative Strategies for a Transdisciplinary Approach to Alzheimer's Disease ANR-11-LABX-01), 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.
- LabEx (Laboratory of Excellence) DISTALZ (Development of Innovative Strategies for a Transdisciplinary Approach to Alzheimer's Disease ANR-11-LABX-01), Lille, France.
| |
Collapse
|
5
|
Lopes van den Broek S, García-Vázquez R, Andersen IV, Valenzuela-Nieto G, Shalgunov V, Battisti UM, Schwefel D, Modhiran N, Kramer V, Cheuquemilla Y, Jara R, Salinas-Varas C, Amarilla AA, Watterson D, Rojas-Fernandez A, Herth MM. Development and evaluation of an 18F-labeled nanobody to target SARS-CoV-2's spike protein. FRONTIERS IN NUCLEAR MEDICINE (LAUSANNE, SWITZERLAND) 2022; 2:1033697. [PMID: 39354971 PMCID: PMC11440877 DOI: 10.3389/fnume.2022.1033697] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Accepted: 10/21/2022] [Indexed: 10/03/2024]
Abstract
COVID-19, caused by the SARS-CoV-2 virus, has become a global pandemic that is still present after more than two years. COVID-19 is mainly known as a respiratory disease that can cause long-term consequences referred to as long COVID. Molecular imaging of SARS-CoV-2 in COVID-19 patients would be a powerful tool for studying the pathological mechanisms and viral load in different organs, providing insights into the disease and the origin of long-term consequences and assessing the effectiveness of potential COVID-19 treatments. Current diagnostic methods used in the clinic do not allow direct imaging of SARS-CoV-2. In this work, a nanobody (NB) - a small, engineered protein derived from alpacas - and an Fc-fused NB which selectively target the SARS-CoV-2 Spike protein were developed as imaging agents for positron emission tomography (PET). We used the tetrazine ligation to 18F-label the NB under mild conditions once the NBs were successfully modified with trans-cyclooctenes (TCOs). We confirmed binding to the Spike protein by SDS-PAGE. Dynamic PET scans in rats showed excretion through the liver for both constructs. Future work will evaluate in vivo binding to the Spike protein with our radioligands.
Collapse
Affiliation(s)
- Sara Lopes van den Broek
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Rocío García-Vázquez
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Ida Vang Andersen
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Guillermo Valenzuela-Nieto
- Institute of Medicine, Faculty of Medicine & Center for Interdisciplinary Studies on the Nervous System, CISNE, Universidad Austral de Chile, Valdivia, Chile
| | - Vladimir Shalgunov
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Umberto M. Battisti
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - David Schwefel
- Institute of Medical Physics and Biophysics, Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Naphak Modhiran
- School of Chemistry and Molecular Biosciences, University of Queensland, St Lucia, QLD, Australia
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, St Lucia, QLD, Australia
| | | | - Yorka Cheuquemilla
- Institute of Medicine, Faculty of Medicine & Center for Interdisciplinary Studies on the Nervous System, CISNE, Universidad Austral de Chile, Valdivia, Chile
| | - Ronald Jara
- Institute of Medicine, Faculty of Medicine & Center for Interdisciplinary Studies on the Nervous System, CISNE, Universidad Austral de Chile, Valdivia, Chile
| | - Constanza Salinas-Varas
- Institute of Medicine, Faculty of Medicine & Center for Interdisciplinary Studies on the Nervous System, CISNE, Universidad Austral de Chile, Valdivia, Chile
| | - Alberto A. Amarilla
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, St Lucia, QLD, Australia
| | - Daniel Watterson
- School of Chemistry and Molecular Biosciences, University of Queensland, St Lucia, QLD, Australia
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, St Lucia, QLD, Australia
| | - Alejandro Rojas-Fernandez
- Institute of Medicine, Faculty of Medicine & Center for Interdisciplinary Studies on the Nervous System, CISNE, Universidad Austral de Chile, Valdivia, Chile
- Berking Biotechnology, Valdivia, Chile
| | - Matthias M. Herth
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Department of Clinical Physiology, Nuclear Medicine & PET, Rigshospitalet Copenhagen University Hospital, Copenhagen, Denmark
| |
Collapse
|
6
|
Caers J, Duray E, Vrancken L, Marcion G, Bocuzzi V, De Veirman K, Krasniqi A, Lejeune M, Withofs N, Devoogdt N, Dumoulin M, Karlström AE, D’Huyvetter M. Radiotheranostic Agents in Hematological Malignancies. Front Immunol 2022; 13:911080. [PMID: 35865548 PMCID: PMC9294596 DOI: 10.3389/fimmu.2022.911080] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Accepted: 06/06/2022] [Indexed: 12/23/2022] Open
Abstract
Radioimmunotherapy (RIT) is a cancer treatment that combines radiation therapy with tumor-directed monoclonal antibodies (Abs). Although RIT had been introduced for the treatment of CD20 positive non-Hodgkin lymphoma decades ago, it never found a broad clinical application. In recent years, researchers have developed theranostic agents based on Ab fragments or small Ab mimetics such as peptides, affibodies or single-chain Abs with improved tumor-targeting capacities. Theranostics combine diagnostic and therapeutic capabilities into a single pharmaceutical agent; this dual application can be easily achieved after conjugation to radionuclides. The past decade has seen a trend to increased specificity, fastened pharmacokinetics, and personalized medicine. In this review, we discuss the different strategies introduced for the noninvasive detection and treatment of hematological malignancies by radiopharmaceuticals. We also discuss the future applications of these radiotheranostic agents.
Collapse
Affiliation(s)
- Jo Caers
- Laboratory of Hematology, GIGA I³, University of Liège, Liège, Belgium
- Department of Hematology, CHU de Liège, Liège, Belgium
- *Correspondence: Jo Caers,
| | - Elodie Duray
- Laboratory of Hematology, GIGA I³, University of Liège, Liège, Belgium
- Centre for Protein Engineering, Inbios, University of Liège, Liège, Belgium
| | - Louise Vrancken
- Laboratory of Hematology, GIGA I³, University of Liège, Liège, Belgium
- Department of Hematology, CHU de Liège, Liège, Belgium
| | - Guillaume Marcion
- Laboratory of Hematology, GIGA I³, University of Liège, Liège, Belgium
| | - Valentina Bocuzzi
- Laboratory of Hematology, GIGA I³, University of Liège, Liège, Belgium
| | - Kim De Veirman
- Department of Hematology and Immunology, Vrije Universiteit Brussel, Brussels, Belgium
| | - Ahmet Krasniqi
- Laboratory of In Vivo Cellular and Molecular Imaging Laboratory (ICMI), Vrije Universiteit Brussel, Brussels, Belgium
| | - Margaux Lejeune
- Laboratory of Hematology, GIGA I³, University of Liège, Liège, Belgium
| | - Nadia Withofs
- Department of Nuclear Medicine, CHU de Liège, Liège, Belgium
| | - Nick Devoogdt
- Laboratory of In Vivo Cellular and Molecular Imaging Laboratory (ICMI), Vrije Universiteit Brussel, Brussels, Belgium
| | - Mireille Dumoulin
- Centre for Protein Engineering, Inbios, University of Liège, Liège, Belgium
| | - Amelie Eriksson Karlström
- Department of Protein Science, School of Engineering Sciences in Chemistry, Biotechnology and Health, KTH Royal Institute of Technology, Stockholm, Sweden
| | - Matthias D’Huyvetter
- Laboratory of In Vivo Cellular and Molecular Imaging Laboratory (ICMI), Vrije Universiteit Brussel, Brussels, Belgium
| |
Collapse
|
7
|
Landrieu I, Dupré E, Sinnaeve D, El Hajjar L, Smet-Nocca C. Deciphering the Structure and Formation of Amyloids in Neurodegenerative Diseases With Chemical Biology Tools. Front Chem 2022; 10:886382. [PMID: 35646824 PMCID: PMC9133342 DOI: 10.3389/fchem.2022.886382] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Accepted: 04/20/2022] [Indexed: 11/24/2022] Open
Abstract
Protein aggregation into highly ordered, regularly repeated cross-β sheet structures called amyloid fibrils is closely associated to human disorders such as neurodegenerative diseases including Alzheimer's and Parkinson's diseases, or systemic diseases like type II diabetes. Yet, in some cases, such as the HET-s prion, amyloids have biological functions. High-resolution structures of amyloids fibrils from cryo-electron microscopy have very recently highlighted their ultrastructural organization and polymorphisms. However, the molecular mechanisms and the role of co-factors (posttranslational modifications, non-proteinaceous components and other proteins) acting on the fibril formation are still poorly understood. Whether amyloid fibrils play a toxic or protective role in the pathogenesis of neurodegenerative diseases remains to be elucidated. Furthermore, such aberrant protein-protein interactions challenge the search of small-molecule drugs or immunotherapy approaches targeting amyloid formation. In this review, we describe how chemical biology tools contribute to new insights on the mode of action of amyloidogenic proteins and peptides, defining their structural signature and aggregation pathways by capturing their molecular details and conformational heterogeneity. Challenging the imagination of scientists, this constantly expanding field provides crucial tools to unravel mechanistic detail of amyloid formation such as semisynthetic proteins and small-molecule sensors of conformational changes and/or aggregation. Protein engineering methods and bioorthogonal chemistry for the introduction of protein chemical modifications are additional fruitful strategies to tackle the challenge of understanding amyloid formation.
Collapse
Affiliation(s)
- Isabelle Landrieu
- University Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1167 - RID-AGE - Risk Factors and Molecular Determinants of Aging-Related Diseases, Lille, France
- CNRS EMR9002 Integrative Structural Biology, Lille, France
| | - Elian Dupré
- University Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1167 - RID-AGE - Risk Factors and Molecular Determinants of Aging-Related Diseases, Lille, France
- CNRS EMR9002 Integrative Structural Biology, Lille, France
| | - Davy Sinnaeve
- University Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1167 - RID-AGE - Risk Factors and Molecular Determinants of Aging-Related Diseases, Lille, France
- CNRS EMR9002 Integrative Structural Biology, Lille, France
| | - Léa El Hajjar
- University Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1167 - RID-AGE - Risk Factors and Molecular Determinants of Aging-Related Diseases, Lille, France
- CNRS EMR9002 Integrative Structural Biology, Lille, France
| | - Caroline Smet-Nocca
- University Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1167 - RID-AGE - Risk Factors and Molecular Determinants of Aging-Related Diseases, Lille, France
- CNRS EMR9002 Integrative Structural Biology, Lille, France
| |
Collapse
|
8
|
Danis C, Dupré E, Zejneli O, Caillierez R, Arrial A, Bégard S, Mortelecque J, Eddarkaoui S, Loyens A, Cantrelle FX, Hanoulle X, Rain JC, Colin M, Buée L, Landrieu I. Inhibition of Tau seeding by targeting Tau nucleation core within neurons with a single domain antibody fragment. Mol Ther 2022; 30:1484-1499. [PMID: 35007758 PMCID: PMC9077319 DOI: 10.1016/j.ymthe.2022.01.009] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2021] [Revised: 12/07/2021] [Accepted: 01/05/2022] [Indexed: 01/13/2023] Open
Abstract
Tau proteins aggregate into filaments in brain cells in Alzheimer's disease and related disorders referred to as tauopathies. Here, we used fragments of camelid heavy-chain-only antibodies (VHHs or single domain antibody fragments) targeting Tau as immuno-modulators of its pathologic seeding. A VHH issued from the screen against Tau of a synthetic phage-display library of humanized VHHs was selected for its capacity to bind Tau microtubule-binding domain, composing the core of Tau fibrils. This parent VHH was optimized to improve its biochemical properties and to act in the intra-cellular compartment, resulting in VHH Z70. VHH Z70 precisely binds the PHF6 sequence, known for its nucleation capacity, as shown by the crystal structure of the complex. VHH Z70 was more efficient than the parent VHH to inhibit in vitro Tau aggregation in heparin-induced assays. Expression of VHH Z70 in a cellular model of Tau seeding also decreased the aggregation-reporting fluorescence signal. Finally, intra-cellular expression of VHH Z70 in the brain of an established tauopathy mouse seeding model demonstrated its capacity to mitigate accumulation of pathological Tau. VHH Z70, by targeting Tau inside brain neurons, where most of the pathological Tau resides, provides an immunological tool to target the intra-cellular compartment in tauopathies.
Collapse
Affiliation(s)
- Clément Danis
- CNRS, EMR9002 BSI Integrative Structural Biology, 59000 Lille, France; Univ. Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1167 - RID-AGE - Risk Factors and Molecular Determinants of Aging-Related Diseases, 59000 Lille, France; Univ. Lille, Inserm, CHU Lille, U1172 - LilNCog - Lille Neuroscience & Cognition, F-59000 Lille, France
| | - Elian Dupré
- CNRS, EMR9002 BSI Integrative Structural Biology, 59000 Lille, France; Univ. Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1167 - RID-AGE - Risk Factors and Molecular Determinants of Aging-Related Diseases, 59000 Lille, France; Univ. Lille, Inserm, CHU Lille, U1172 - LilNCog - Lille Neuroscience & Cognition, F-59000 Lille, France
| | - Orgeta Zejneli
- CNRS, EMR9002 BSI Integrative Structural Biology, 59000 Lille, France; Univ. Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1167 - RID-AGE - Risk Factors and Molecular Determinants of Aging-Related Diseases, 59000 Lille, France; Univ. Lille, Inserm, CHU Lille, U1172 - LilNCog - Lille Neuroscience & Cognition, F-59000 Lille, France
| | - Raphaëlle Caillierez
- Univ. Lille, Inserm, CHU Lille, U1172 - LilNCog - Lille Neuroscience & Cognition, F-59000 Lille, France
| | - Alexis Arrial
- Hybrigenic Services, Evry-Courcouronnes 91000, France
| | - Séverine Bégard
- Univ. Lille, Inserm, CHU Lille, U1172 - LilNCog - Lille Neuroscience & Cognition, F-59000 Lille, France
| | - Justine Mortelecque
- CNRS, EMR9002 BSI Integrative Structural Biology, 59000 Lille, France; Univ. Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1167 - RID-AGE - Risk Factors and Molecular Determinants of Aging-Related Diseases, 59000 Lille, France
| | - Sabiha Eddarkaoui
- Univ. Lille, Inserm, CHU Lille, U1172 - LilNCog - Lille Neuroscience & Cognition, F-59000 Lille, France
| | - Anne Loyens
- Univ. Lille, Inserm, CHU Lille, U1172 - LilNCog - Lille Neuroscience & Cognition, F-59000 Lille, France
| | - François-Xavier Cantrelle
- CNRS, EMR9002 BSI Integrative Structural Biology, 59000 Lille, France; Univ. Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1167 - RID-AGE - Risk Factors and Molecular Determinants of Aging-Related Diseases, 59000 Lille, France
| | - Xavier Hanoulle
- CNRS, EMR9002 BSI Integrative Structural Biology, 59000 Lille, France; Univ. Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1167 - RID-AGE - Risk Factors and Molecular Determinants of Aging-Related Diseases, 59000 Lille, France
| | | | - Morvane Colin
- Univ. Lille, Inserm, CHU Lille, U1172 - LilNCog - Lille Neuroscience & Cognition, F-59000 Lille, France
| | - Luc Buée
- Univ. Lille, Inserm, CHU Lille, U1172 - LilNCog - Lille Neuroscience & Cognition, F-59000 Lille, France.
| | - Isabelle Landrieu
- CNRS, EMR9002 BSI Integrative Structural Biology, 59000 Lille, France; Univ. Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1167 - RID-AGE - Risk Factors and Molecular Determinants of Aging-Related Diseases, 59000 Lille, France.
| |
Collapse
|
9
|
Marino M, Zhou L, Rincon MY, Callaerts-Vegh Z, Verhaert J, Wahis J, Creemers E, Yshii L, Wierda K, Saito T, Marneffe C, Voytyuk I, Wouters Y, Dewilde M, Duqué SI, Vincke C, Levites Y, Golde TE, Saido TC, Muyldermans S, Liston A, De Strooper B, Holt MG. AAV-mediated delivery of an anti-BACE1 VHH alleviates pathology in an Alzheimer's disease model. EMBO Mol Med 2022; 14:e09824. [PMID: 35352880 PMCID: PMC8988209 DOI: 10.15252/emmm.201809824] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2019] [Revised: 02/11/2022] [Accepted: 02/14/2022] [Indexed: 01/18/2023] Open
Abstract
Single domain antibodies (VHHs) are potentially disruptive therapeutics, with important biological value for treatment of several diseases, including neurological disorders. However, VHHs have not been widely used in the central nervous system (CNS), largely because of their restricted blood-brain barrier (BBB) penetration. Here, we propose a gene transfer strategy based on BBB-crossing Adeno-associated virus (AAV)-based vectors to deliver VHH directly into the CNS. As a proof-of-concept, we explored the potential of AAV-delivered VHH to inhibit BACE1, a well-characterized target in Alzheimer's disease. First, we generated a panel of VHHs targeting BACE1, one of which, VHH-B9, shows high selectivity for BACE1 and efficacy in lowering BACE1 activity in vitro. We further demonstrate that a single systemic dose of AAV-VHH-B9 produces positive long-term (12 months plus) effects on amyloid load, neuroinflammation, synaptic function, and cognitive performance, in the AppNL-G-F Alzheimer's disease mouse model. These results constitute a novel therapeutic approach forneurodegenerative diseases, which is applicable to a range of CNS disease targets.
Collapse
Affiliation(s)
- Marika Marino
- VIB-KU Leuven Center for Brain and Disease Research, Leuven, Belgium.,Department of Neurosciences, KU Leuven, Leuven, Belgium
| | - Lujia Zhou
- VIB-KU Leuven Center for Brain and Disease Research, Leuven, Belgium.,Department of Neurosciences, KU Leuven, Leuven, Belgium
| | - Melvin Y Rincon
- VIB-KU Leuven Center for Brain and Disease Research, Leuven, Belgium.,Department of Neurosciences, KU Leuven, Leuven, Belgium
| | | | - Jens Verhaert
- VIB-KU Leuven Center for Brain and Disease Research, Leuven, Belgium.,Department of Neurosciences, KU Leuven, Leuven, Belgium
| | - Jérôme Wahis
- VIB-KU Leuven Center for Brain and Disease Research, Leuven, Belgium.,Department of Neurosciences, KU Leuven, Leuven, Belgium
| | - Eline Creemers
- VIB-KU Leuven Center for Brain and Disease Research, Leuven, Belgium.,Department of Neurosciences, KU Leuven, Leuven, Belgium.,Electrophysiology Expertise Unit, VIB-KU Leuven Center for Brain and Disease Research, Leuven, Belgium
| | - Lidia Yshii
- VIB-KU Leuven Center for Brain and Disease Research, Leuven, Belgium.,Department of Neurosciences, KU Leuven, Leuven, Belgium.,Department of Microbiology, Immunology and Transplantation, KU Leuven, Leuven, Belgium
| | - Keimpe Wierda
- VIB-KU Leuven Center for Brain and Disease Research, Leuven, Belgium.,Department of Neurosciences, KU Leuven, Leuven, Belgium.,Electrophysiology Expertise Unit, VIB-KU Leuven Center for Brain and Disease Research, Leuven, Belgium
| | - Takashi Saito
- Department of Neurocognitive Science, Institute of Brain Science, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | - Catherine Marneffe
- VIB-KU Leuven Center for Brain and Disease Research, Leuven, Belgium.,Department of Neurosciences, KU Leuven, Leuven, Belgium
| | - Iryna Voytyuk
- VIB-KU Leuven Center for Brain and Disease Research, Leuven, Belgium.,Department of Neurosciences, KU Leuven, Leuven, Belgium
| | - Yessica Wouters
- VIB-KU Leuven Center for Brain and Disease Research, Leuven, Belgium.,Department of Neurosciences, KU Leuven, Leuven, Belgium
| | - Maarten Dewilde
- VIB-KU Leuven Center for Brain and Disease Research, Leuven, Belgium.,Department of Neurosciences, KU Leuven, Leuven, Belgium
| | - Sandra I Duqué
- VIB-KU Leuven Center for Brain and Disease Research, Leuven, Belgium.,Department of Neurosciences, KU Leuven, Leuven, Belgium
| | - Cécile Vincke
- Laboratory of Cellular and Molecular Immunology, Vrije Universiteit Brussel, Brussels, Belgium
| | - Yona Levites
- Department of Neuroscience, Center for Translational Research in Neurodegenerative Disease, McKnight Brain Institute, College of Medicine, University of Florida, Gainesville, FL, USA
| | - Todd E Golde
- Department of Neuroscience, Center for Translational Research in Neurodegenerative Disease, McKnight Brain Institute, College of Medicine, University of Florida, Gainesville, FL, USA
| | - Takaomi C Saido
- Laboratory for Proteolytic Neuroscience, RIKEN Brain Science Institute, Wako-shi, Japan
| | - Serge Muyldermans
- Laboratory of Cellular and Molecular Immunology, Vrije Universiteit Brussel, Brussels, Belgium
| | - Adrian Liston
- VIB-KU Leuven Center for Brain and Disease Research, Leuven, Belgium.,Department of Microbiology, Immunology and Transplantation, KU Leuven, Leuven, Belgium.,Immunology Programme, The Babraham Institute, Cambridge, UK
| | - Bart De Strooper
- VIB-KU Leuven Center for Brain and Disease Research, Leuven, Belgium.,Department of Neurosciences, KU Leuven, Leuven, Belgium.,UK Dementia Research institute at UCL, London, UK.,Leuven Brain Institute, Leuven, Belgium
| | - Matthew G Holt
- VIB-KU Leuven Center for Brain and Disease Research, Leuven, Belgium.,Department of Neurosciences, KU Leuven, Leuven, Belgium.,Leuven Brain Institute, Leuven, Belgium.,Instituto de Investigação e Inovação em Saúde (i3S), University of Porto, Porto, Portugal
| |
Collapse
|
10
|
Wang J, Kang G, Yuan H, Cao X, Huang H, de Marco A. Research Progress and Applications of Multivalent, Multispecific and Modified Nanobodies for Disease Treatment. Front Immunol 2022; 12:838082. [PMID: 35116045 PMCID: PMC8804282 DOI: 10.3389/fimmu.2021.838082] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Accepted: 12/30/2021] [Indexed: 12/22/2022] Open
Abstract
Recombinant antibodies such as nanobodies are progressively demonstrating to be a valid alternative to conventional monoclonal antibodies also for clinical applications. Furthermore, they do not solely represent a substitute for monoclonal antibodies but their unique features allow expanding the applications of biotherapeutics and changes the pattern of disease treatment. Nanobodies possess the double advantage of being small and simple to engineer. This combination has promoted extremely diversified approaches to design nanobody-based constructs suitable for particular applications. Both the format geometry possibilities and the functionalization strategies have been widely explored to provide macromolecules with better efficacy with respect to single nanobodies or their combination. Nanobody multimers and nanobody-derived reagents were developed to image and contrast several cancer diseases and have shown their effectiveness in animal models. Their capacity to block more independent signaling pathways simultaneously is considered a critical advantage to avoid tumor resistance, whereas the mass of these multimeric compounds still remains significantly smaller than that of an IgG, enabling deeper penetration in solid tumors. When applied to CAR-T cell therapy, nanobodies can effectively improve the specificity by targeting multiple epitopes and consequently reduce the side effects. This represents a great potential in treating malignant lymphomas, acute myeloid leukemia, acute lymphoblastic leukemia, multiple myeloma and solid tumors. Apart from cancer treatment, multispecific drugs and imaging reagents built with nanobody blocks have demonstrated their value also for detecting and tackling neurodegenerative, autoimmune, metabolic, and infectious diseases and as antidotes for toxins. In particular, multi-paratopic nanobody-based constructs have been developed recently as drugs for passive immunization against SARS-CoV-2 with the goal of impairing variant survival due to resistance to antibodies targeting single epitopes. Given the enormous research activity in the field, it can be expected that more and more multimeric nanobody molecules will undergo late clinical trials in the next future. Systematic Review Registration.
Collapse
Affiliation(s)
- Jiewen Wang
- Department of Biochemical Engineering, School of Chemical Engineering and Technology, Tianjin University, Tianjin, China
- Frontiers Science Center for Synthetic Biology and Key Laboratory of Systems Bioengineering (Ministry of Education), Tianjin University, Tianjin, China
- Institute of Shaoxing, Tianjin University, Zhejiang, China
| | - Guangbo Kang
- Department of Biochemical Engineering, School of Chemical Engineering and Technology, Tianjin University, Tianjin, China
- Frontiers Science Center for Synthetic Biology and Key Laboratory of Systems Bioengineering (Ministry of Education), Tianjin University, Tianjin, China
- Institute of Shaoxing, Tianjin University, Zhejiang, China
| | - Haibin Yuan
- Department of Biochemical Engineering, School of Chemical Engineering and Technology, Tianjin University, Tianjin, China
- Frontiers Science Center for Synthetic Biology and Key Laboratory of Systems Bioengineering (Ministry of Education), Tianjin University, Tianjin, China
- Institute of Shaoxing, Tianjin University, Zhejiang, China
| | - Xiaocang Cao
- Department of Gastroenterology and Hepatology, Tianjin Medical University General Hospital, Tianjin Medical University, Tianjin, China
| | - He Huang
- Department of Biochemical Engineering, School of Chemical Engineering and Technology, Tianjin University, Tianjin, China
- Frontiers Science Center for Synthetic Biology and Key Laboratory of Systems Bioengineering (Ministry of Education), Tianjin University, Tianjin, China
- Institute of Shaoxing, Tianjin University, Zhejiang, China
| | - Ario de Marco
- Laboratory for Environmental and Life Sciences, University of Nova Gorica, Nova Gorica, Slovenia
| |
Collapse
|
11
|
Duray E, Lejeune M, Baron F, Beguin Y, Devoogdt N, Krasniqi A, Lauwers Y, Zhao YJ, D'Huyvetter M, Dumoulin M, Caers J. A non-internalised CD38-binding radiolabelled single-domain antibody fragment to monitor and treat multiple myeloma. J Hematol Oncol 2021; 14:183. [PMID: 34727950 PMCID: PMC8561907 DOI: 10.1186/s13045-021-01171-6] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Accepted: 09/22/2021] [Indexed: 12/21/2022] Open
Abstract
Background Antibody-based therapies targeting CD38 are currently used as single agents as well as in combination regimens for multiple myeloma, a malignant plasma cell disorder. In this study, we aimed to develop anti-CD38 single-domain antibodies (sdAbs) that can be used to trace CD38+ tumour cells and subsequently used for targeted radionuclide therapy. SdAbs are derived from Camelidae heavy-chain antibodies and have emerged as promising theranostic agents due to their favourable pharmacological properties. Methods Four different anti-CD38 sdAbs were produced, and their binding affinities and potential competition with the monoclonal antibody daratumumab were tested using biolayer interferometry. Their binding kinetics and potential cell internalisation were further studied after radiolabelling with the diagnostic radioisotope Indium-111. The resulting radiotracers were evaluated in vivo for their tumour-targeting potential and biodistribution through single-photon emission computed tomography (SPECT/CT) imaging and serial dissections. Finally, therapeutic efficacy of a lead anti-CD38 sdAb, radiolabelled with the therapeutic radioisotope Lutetium-177, was evaluated in a CD38+ MM xenograft model. Results We retained anti-CD38 sdAb #2F8 as lead based on its excellent affinity and superior stability, the absence of competition with daratumumab and the lack of receptor-mediated internalisation. When intravenously administered to tumour-xenografted mice, radiolabelled sdAb #2F8 revealed specific and sustained tumour retention with low accumulation in other tissues, except kidneys, resulting in high tumour-to-normal tissue ratios. In a therapeutic setting, myeloma-bearing mice received three consecutive intravenous administrations of a high (18.5 MBq) or a low radioactive dose (9.3 MBq) of 177Lu-DTPA-2F8 or an equal volume of vehicle solution. A dose-dependent tumour regression was observed, which translated into a prolonged median survival from 43 days for vehicle-treated mice, to 62 days (p = 0.027) in mice receiving the low and 65 days in mice receiving the high (p = 0.0007) radioactive dose regimen, respectively. Conclusions These results highlight the theranostic potential of radiolabelled anti-CD38 sdAbs for the monitoring and treatment of multiple myeloma. Supplementary Information The online version contains supplementary material available at 10.1186/s13045-021-01171-6.
Collapse
Affiliation(s)
- Elodie Duray
- Laboratory of Haematology, GIGA-I3, University of Liège, Liège, Belgium.,NEPTUNS, Nanobodies To Explore Protein Structure and Functions, Centre for Protein Engineering (CIP), University of Liège, Liège, Belgium
| | - Margaux Lejeune
- Laboratory of Haematology, GIGA-I3, University of Liège, Liège, Belgium
| | - Frederic Baron
- Laboratory of Haematology, GIGA-I3, University of Liège, Liège, Belgium.,Division of Haematology, Department of Medicine, University and CHU of Liège, Liège, Belgium
| | - Yves Beguin
- Laboratory of Haematology, GIGA-I3, University of Liège, Liège, Belgium.,Division of Haematology, Department of Medicine, University and CHU of Liège, Liège, Belgium
| | - Nick Devoogdt
- Department of Medical Imaging, Laboratory for In Vivo Cellular and Molecular Imaging, Vrije Universiteit Brussel, Brussels, Belgium
| | - Ahmet Krasniqi
- Department of Medical Imaging, Laboratory for In Vivo Cellular and Molecular Imaging, Vrije Universiteit Brussel, Brussels, Belgium
| | - Yoline Lauwers
- Department of Medical Imaging, Laboratory for In Vivo Cellular and Molecular Imaging, Vrije Universiteit Brussel, Brussels, Belgium
| | - Yong Juan Zhao
- School of Chemical Biology and Biotechnology, University Shenzhen Graduate School, Peking, China
| | - Matthias D'Huyvetter
- Department of Medical Imaging, Laboratory for In Vivo Cellular and Molecular Imaging, Vrije Universiteit Brussel, Brussels, Belgium
| | - Mireille Dumoulin
- NEPTUNS, Nanobodies To Explore Protein Structure and Functions, Centre for Protein Engineering (CIP), University of Liège, Liège, Belgium
| | - Jo Caers
- Laboratory of Haematology, GIGA-I3, University of Liège, Liège, Belgium. .,Division of Haematology, Department of Medicine, University and CHU of Liège, Liège, Belgium.
| |
Collapse
|
12
|
Vettore N, Moray J, Brans A, Herman R, Charlier P, Kumita JR, Kerff F, Dobson CM, Dumoulin M. Characterisation of the structural, dynamic and aggregation properties of the W64R amyloidogenic variant of human lysozyme. Biophys Chem 2021; 271:106563. [PMID: 33640796 DOI: 10.1016/j.bpc.2021.106563] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2020] [Revised: 02/05/2021] [Accepted: 02/06/2021] [Indexed: 10/22/2022]
Abstract
The accumulation in vital organs of amyloid fibrils made of mutational variants of lysozyme (HuL) is associated with a human systemic amyloid disease. The detailed comparison of the in vitro properties of the I56T and D67H amyloidogenic variants to those of the T70N non-amyloidogenic variant and the wild-type (WT) protein suggested that the deposition of large amounts of aggregated disease-related lysozyme variants is initiated by the formation of transient intermediate species. The ability to populate such intermediates is essentially due to the destabilisation of the protein and the loss of the global structural cooperativity under physiologically relevant conditions. Here, we report the characterisation of a third naturally occurring amyloidogenic lysozyme variant, W64R, in comparison with the I56T and WT proteins. The X-ray crystal structure of the W64R variant at 1.15 Å resolution is very similar to that of the WT protein; a few interactions within the β-domain and at the interface between the α- and β-domains differ, however, from those in the WT protein. Consequently, the W64R mutation destabilizes the protein to an extent that is similar to that observed for the I56T and D67H mutations. The ΔG°NU(H2O) is reduced by 24 kJ·mol-1 and the Tm is about 12 °C lower than that of the WT protein. Under native conditions, the W64R and I56T proteins are readily digested by proteinase K, while the WT protein remains intact. These results suggest that the two variant proteins transiently populate similar partially unfolded states in which proteinase K cleavage sites are accessible to the protease. Moreover, the in vitro aggregation properties of the W64R protein are similar to those of the I56T variant. Altogether, these results indicate that the properties of the W64R protein are astonishingly similar to those of the I56T variant. They further corroborate the idea that HuL variants associated with the disease are those whose stability and global structural cooperativity are sufficiently reduced to allow the formation of aggregation prone partially folded intermediates under physiological conditions.
Collapse
Affiliation(s)
- Nicola Vettore
- Centre for Protein Engineering, InBioS, Department of Life Sciences, University of Liège, (Sart-Tilman) 4000 Liège, Belgium
| | - Joël Moray
- Centre for Protein Engineering, InBioS, Department of Life Sciences, University of Liège, (Sart-Tilman) 4000 Liège, Belgium
| | - Alain Brans
- Centre for Protein Engineering, InBioS, Department of Life Sciences, University of Liège, (Sart-Tilman) 4000 Liège, Belgium
| | - Raphaël Herman
- Centre for Protein Engineering, InBioS, Department of Life Sciences, University of Liège, (Sart-Tilman) 4000 Liège, Belgium
| | - Paulette Charlier
- Centre for Protein Engineering, InBioS, Department of Life Sciences, University of Liège, (Sart-Tilman) 4000 Liège, Belgium
| | - Janet R Kumita
- Department of Chemistry, University of Cambridge, Lensfield Road, Cambridge CB2 1EW, United Kingdom
| | - Frédéric Kerff
- Centre for Protein Engineering, InBioS, Department of Life Sciences, University of Liège, (Sart-Tilman) 4000 Liège, Belgium
| | - Christopher M Dobson
- Department of Chemistry, University of Cambridge, Lensfield Road, Cambridge CB2 1EW, United Kingdom
| | - Mireille Dumoulin
- Centre for Protein Engineering, InBioS, Department of Life Sciences, University of Liège, (Sart-Tilman) 4000 Liège, Belgium.
| |
Collapse
|
13
|
Antibody Fragments as Tools for Elucidating Structure-Toxicity Relationships and for Diagnostic/Therapeutic Targeting of Neurotoxic Amyloid Oligomers. Int J Mol Sci 2020; 21:ijms21238920. [PMID: 33255488 PMCID: PMC7727795 DOI: 10.3390/ijms21238920] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2020] [Revised: 10/01/2020] [Accepted: 10/01/2020] [Indexed: 02/06/2023] Open
Abstract
The accumulation of amyloid protein aggregates in tissues is the basis for the onset of diseases known as amyloidoses. Intriguingly, many amyloidoses impact the central nervous system (CNS) and usually are devastating diseases. It is increasingly apparent that neurotoxic soluble oligomers formed by amyloidogenic proteins are the primary molecular drivers of these diseases, making them lucrative diagnostic and therapeutic targets. One promising diagnostic/therapeutic strategy has been the development of antibody fragments against amyloid oligomers. Antibody fragments, such as fragment antigen-binding (Fab), scFv (single chain variable fragments), and VHH (heavy chain variable domain or single-domain antibodies) are an alternative to full-length IgGs as diagnostics and therapeutics for a variety of diseases, mainly because of their increased tissue penetration (lower MW compared to IgG), decreased inflammatory potential (lack of Fc domain), and facile production (low structural complexity). Furthermore, through the use of in vitro-based ligand selection, it has been possible to identify antibody fragments presenting marked conformational selectivity. In this review, we summarize significant reports on antibody fragments selective for oligomers associated with prevalent CNS amyloidoses. We discuss promising results obtained using antibody fragments as both diagnostic and therapeutic agents against these diseases. In addition, the use of antibody fragments, particularly scFv and VHH, in the isolation of unique oligomeric assemblies is discussed as a strategy to unravel conformational moieties responsible for neurotoxicity. We envision that advances in this field may lead to the development of novel oligomer-selective antibody fragments with superior selectivity and, hopefully, good clinical outcomes.
Collapse
|
14
|
Mabbott NA, Bradford BM, Pal R, Young R, Donaldson DS. The Effects of Immune System Modulation on Prion Disease Susceptibility and Pathogenesis. Int J Mol Sci 2020; 21:E7299. [PMID: 33023255 PMCID: PMC7582561 DOI: 10.3390/ijms21197299] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2020] [Revised: 09/25/2020] [Accepted: 09/29/2020] [Indexed: 12/17/2022] Open
Abstract
Prion diseases are a unique group of infectious chronic neurodegenerative disorders to which there are no cures. Although prion infections do not stimulate adaptive immune responses in infected individuals, the actions of certain immune cell populations can have a significant impact on disease pathogenesis. After infection, the targeting of peripherally-acquired prions to specific immune cells in the secondary lymphoid organs (SLO), such as the lymph nodes and spleen, is essential for the efficient transmission of disease to the brain. Once the prions reach the brain, interactions with other immune cell populations can provide either host protection or accelerate the neurodegeneration. In this review, we provide a detailed account of how factors such as inflammation, ageing and pathogen co-infection can affect prion disease pathogenesis and susceptibility. For example, we discuss how changes to the abundance, function and activation status of specific immune cell populations can affect the transmission of prion diseases by peripheral routes. We also describe how the effects of systemic inflammation on certain glial cell subsets in the brains of infected individuals can accelerate the neurodegeneration. A detailed understanding of the factors that affect prion disease transmission and pathogenesis is essential for the development of novel intervention strategies.
Collapse
Affiliation(s)
- Neil A. Mabbott
- The Roslin Institute & Royal (Dick) School of Veterinary Studies, University of Edinburgh, Easter Bush, Midlothian EH25 9RG, UK; (B.M.B.); (R.P.); (R.Y.); (D.S.D.)
| | | | | | | | | |
Collapse
|
15
|
Sandusky-Beltran LA, Sigurdsson EM. Tau immunotherapies: Lessons learned, current status and future considerations. Neuropharmacology 2020; 175:108104. [PMID: 32360477 PMCID: PMC7492435 DOI: 10.1016/j.neuropharm.2020.108104] [Citation(s) in RCA: 49] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2019] [Revised: 03/10/2020] [Accepted: 04/06/2020] [Indexed: 12/15/2022]
Abstract
The majority of clinical trials targeting the tau protein in Alzheimer's disease and other tauopathies are tau immunotherapies. Because tau pathology correlates better with the degree of dementia than amyloid-β lesions, targeting tau is likely to be more effective in improving cognition than clearing amyloid-β in Alzheimer's disease. However, the development of tau therapies is in many ways more complex than for amyloid-β therapies as briefly outlined in this review. Most of the trials are on humanized antibodies, which may have very different properties than the original mouse antibodies. The impact of these differences are to a large extent unknown, can be difficult to decipher, and may not always be properly considered. Furthermore, the ideal antibody properties for efficacy are not well established and can depend on several factors. However, considering the varied approaches in clinical trials, there is a general optimism that at least some of these trials may provide functional benefits to patients suffering of various tauopathies. This article is part of the special issue entitled 'The Quest for Disease-Modifying Therapies for Neurodegenerative Disorders'.
Collapse
Affiliation(s)
- L A Sandusky-Beltran
- Department of Neuroscience and Physiology, New York University School of Medicine, New York, NY, 10016, USA; Department of Neuroscience Institute, New York University School of Medicine, New York, NY, 10016, USA
| | - E M Sigurdsson
- Department of Neuroscience and Physiology, New York University School of Medicine, New York, NY, 10016, USA; Department of Psychiatry, New York University School of Medicine, New York, NY, 10016, USA; Department of Neuroscience Institute, New York University School of Medicine, New York, NY, 10016, USA.
| |
Collapse
|
16
|
Sroga P, Safronetz D, Stein DR. Nanobodies: a new approach for the diagnosis and treatment of viral infectious diseases. Future Virol 2020. [DOI: 10.2217/fvl-2019-0167] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
With the rise of viral infections and antibiotic resistance, there is a constant need for the development of more sensitive and effective treatment and diagnostic tools. Since their discovery in the early 1990s, Camelidae antibodies have been investigated as potential tools due to their unique structure and favorable characteristics. Members of this family produce conventional IgG antibodies as well as heavy-chain only IgG antibodies that do not possess light chains. The variable domain (VHH), or nanobody, demonstrates unique antigen-binding capabilities, enhanced stability, and its small size allows for delivery into the body using a nebulizer, thereby eliminating the unfavorable use of injections. In addition, the cost-effective and easy in vitro production of these antibodies are an attractive quality in terms of mass production. This review covers the past and current nanobody treatment and diagnostic developments aimed at viral infectious diseases, including a brief overview of protozoal, bacterial, and veterinary viral approaches.
Collapse
Affiliation(s)
- Patrycja Sroga
- Department of Medical Microbiology, University of Manitoba, Winnipeg, MB, Canada
| | - David Safronetz
- Department of Medical Microbiology, University of Manitoba, Winnipeg, MB, Canada
- Zoonotic Diseases & Special Pathogens, National Microbiology Laboratory, Public Health Agency of Canada, Winnipeg, MB, Canada
| | | |
Collapse
|
17
|
Verhelle A, Van Overbeke W, Peleman C, De Smet R, Zwaenepoel O, Lahoutte T, Van Dorpe J, Devoogdt N, Gettemans J. Non-Invasive Imaging of Amyloid Deposits in a Mouse Model of AGel Using 99mTc-Modified Nanobodies and SPECT/CT. Mol Imaging Biol 2017; 18:887-897. [PMID: 27130233 DOI: 10.1007/s11307-016-0960-y] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
PURPOSE Gelsolin amyloidosis (AGel), also known as familial amyloidosis, Finnish type (FAF), is an autosomal, dominant, incurable disease caused by a point mutation (G654A/T) in the gelsolin (GSN) gene. The mutation results in loss of a Ca2+-binding site in the second gelsolin domain. Subsequent incorrect folding exposes a cryptic furin cleavage site, leading to the formation of a 68-kDa C-terminal cleavage product (C68) in the trans-Golgi network. This C68 fragment is cleaved by membrane type 1-matrix metalloproteinase (MT1-MMP) during secretion into the extracellular environment, releasing 8- and 5-kDa amyloidogenic peptides. These peptides aggregate and cause disease-associated symptoms. We set out to investigate whether AGel-specific nanobodies could be used to monitor amyloidogenic gelsolin buildup. PROCEDURES Three nanobodies (FAF Nb1-3) raised against the 8-kDa fragment were screened as AGel amyloid imaging agents in WT and AGel mice using 99mTc-based single-photon emission computed tomography (SPECT)/X-ray tomography (CT), biodistribution analysis, and immunofluorescence (IF). The quantitative characteristics were analyzed in a follow-up study with a Nb11-expressing mouse model. RESULTS All three nanobodies possess the characteristics desired for a 99mTc-based SPECT/CT imaging agent, high specificity and a low background signal. FAF Nb1 was identified as the most potent, based on its superior signal-to-noise ratio and signal specificity. As a proof of concept, we implemented 99mTc-FAF Nb1 in a follow-up study of the Nb11-expressing AGel mouse model. Using biodistribution analysis and immunofluorescence, we demonstrated the validity of the data acquired via 99mTc-FAF Nb1 SPECT/CT. CONCLUSION These findings demonstrate the potential of this nanobody as a non-invasive tool to image amyloidogenic gelsolin deposition and assess the therapeutic capacity of AGel therapeutics currently under development. We propose that this approach can be extended to other amyloid diseases, thereby contributing to the development of specific therapies.
Collapse
Affiliation(s)
- Adriaan Verhelle
- Department of Biochemistry, Faculty of Medicine and Health Sciences, Ghent University, Albert Baertsoenkaai 3, B-9000, Ghent, Belgium
| | - Wouter Van Overbeke
- Department of Biochemistry, Faculty of Medicine and Health Sciences, Ghent University, Albert Baertsoenkaai 3, B-9000, Ghent, Belgium
| | - Cindy Peleman
- In Vivo Cellular and Molecular Imaging Laboratory, Vrije Universiteit Brussel, Brussels, Belgium
| | - Rebecca De Smet
- Department of Medical and Forensic Pathology, Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium
| | - Olivier Zwaenepoel
- Department of Biochemistry, Faculty of Medicine and Health Sciences, Ghent University, Albert Baertsoenkaai 3, B-9000, Ghent, Belgium
| | - Tony Lahoutte
- In Vivo Cellular and Molecular Imaging Laboratory, Vrije Universiteit Brussel, Brussels, Belgium
| | - Jo Van Dorpe
- Department of Medical and Forensic Pathology, Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium
| | - Nick Devoogdt
- In Vivo Cellular and Molecular Imaging Laboratory, Vrije Universiteit Brussel, Brussels, Belgium
| | - Jan Gettemans
- Department of Biochemistry, Faculty of Medicine and Health Sciences, Ghent University, Albert Baertsoenkaai 3, B-9000, Ghent, Belgium.
| |
Collapse
|
18
|
Swasthi HM, Mukhopadhyay S. Electrostatic lipid-protein interactions sequester the curli amyloid fold on the lipopolysaccharide membrane surface. J Biol Chem 2017; 292:19861-19872. [PMID: 29021250 DOI: 10.1074/jbc.m117.815522] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2017] [Revised: 10/04/2017] [Indexed: 12/14/2022] Open
Abstract
Curli is a functional amyloid protein in the extracellular matrix of enteric Gram-negative bacteria. Curli is assembled at the cell surface and consists of CsgA, the major subunit of curli, and a membrane-associated nucleator protein, CsgB. Oligomeric intermediates that accumulate during the lag phase of amyloidogenesis are generally toxic, but the underlying mechanism by which bacterial cells overcome this toxicity during curli assembly at the surface remains elusive. Here, we elucidated the mechanism of curli amyloidogenesis and provide molecular insights into the strategy by which bacteria can potentially bypass the detrimental consequences of toxic amyloid intermediates. Using a diverse range of biochemical and biophysical tools involving circular dichroism, fluorescence, Raman spectroscopy, and atomic force microscopy imaging, we characterized the molecular basis of the interaction of CsgB with a membrane-mimetic anionic surfactant as well as with lipopolysaccharide (LPS) constituting the outer leaflet of Gram-negative bacteria. Aggregation studies revealed that the electrostatic interaction of the positively charged C-terminal region of the protein with a negatively charged head group of surfactant/LPS promotes a protein-protein interaction that results in facile amyloid formation without a detectable lag phase. We also show that CsgB, in the presence of surfactant/LPS, accelerates the fibrillation rate of CsgA by circumventing the lag phase during nucleation. Our findings suggest that the electrostatic interactions between lipid and protein molecules play a pivotal role in efficiently sequestering the amyloid fold of curli on the membrane surface without significant accumulation of toxic oligomeric intermediates.
Collapse
Affiliation(s)
- Hema M Swasthi
- From the Centre for Protein Science, Design and Engineering.,Chemical Sciences, Indian Institute of Science Education and Research (IISER), Mohali, Sector 81, Knowledge City, Mohali, Punjab, India
| | - Samrat Mukhopadhyay
- From the Centre for Protein Science, Design and Engineering, .,Chemical Sciences, Indian Institute of Science Education and Research (IISER), Mohali, Sector 81, Knowledge City, Mohali, Punjab, India.,Departments of Biological Sciences and
| |
Collapse
|
19
|
Iljina M, Hong L, Horrocks MH, Ludtmann MH, Choi ML, Hughes CD, Ruggeri FS, Guilliams T, Buell AK, Lee JE, Gandhi S, Lee SF, Bryant CE, Vendruscolo M, Knowles TPJ, Dobson CM, De Genst E, Klenerman D. Nanobodies raised against monomeric ɑ-synuclein inhibit fibril formation and destabilize toxic oligomeric species. BMC Biol 2017; 15:57. [PMID: 28673288 PMCID: PMC5496350 DOI: 10.1186/s12915-017-0390-6] [Citation(s) in RCA: 56] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2017] [Accepted: 06/06/2017] [Indexed: 11/16/2022] Open
Abstract
Background The aggregation of the protein ɑ-synuclein (ɑS) underlies a range of increasingly common neurodegenerative disorders including Parkinson’s disease. One widely explored therapeutic strategy for these conditions is the use of antibodies to target aggregated ɑS, although a detailed molecular-level mechanism of the action of such species remains elusive. Here, we characterize ɑS aggregation in vitro in the presence of two ɑS-specific single-domain antibodies (nanobodies), NbSyn2 and NbSyn87, which bind to the highly accessible C-terminal region of ɑS. Results We show that both nanobodies inhibit the formation of ɑS fibrils. Furthermore, using single-molecule fluorescence techniques, we demonstrate that nanobody binding promotes a rapid conformational conversion from more stable oligomers to less stable oligomers of ɑS, leading to a dramatic reduction in oligomer-induced cellular toxicity. Conclusions The results indicate a novel mechanism by which diseases associated with protein aggregation can be inhibited, and suggest that NbSyn2 and NbSyn87 could have significant therapeutic potential. Electronic supplementary material The online version of this article (doi:10.1186/s12915-017-0390-6) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Marija Iljina
- Department of Chemistry, University of Cambridge, Lensfield Road, Cambridge, CB2 1EW, UK
| | - Liu Hong
- Department of Chemistry, University of Cambridge, Lensfield Road, Cambridge, CB2 1EW, UK.,Zhou Pei-Yuan Center for Applied Mathematics, Tsinghua University, Beijing, 100084, China
| | - Mathew H Horrocks
- Department of Chemistry, University of Cambridge, Lensfield Road, Cambridge, CB2 1EW, UK.,Wellcome Trust-Medical Research Council Cambridge Stem Cell Institute, University of Cambridge, Cambridge, CB2 1QR, UK
| | - Marthe H Ludtmann
- Department of Molecular Neuroscience, University College London, Institute of Neurology, Queen Square, London, WC1N 3BG, UK
| | - Minee L Choi
- Department of Molecular Neuroscience, University College London, Institute of Neurology, Queen Square, London, WC1N 3BG, UK
| | - Craig D Hughes
- Department of Veterinary Medicine, University of Cambridge, Madingley Road, Cambridge, CB3 0ES, UK
| | - Francesco S Ruggeri
- Department of Chemistry, University of Cambridge, Lensfield Road, Cambridge, CB2 1EW, UK
| | - Tim Guilliams
- Department of Chemistry, University of Cambridge, Lensfield Road, Cambridge, CB2 1EW, UK.,Present address: Healx Ltd., St John's Innovation Centre, Cowley Road, Cambridge, CB4 0WS, UK
| | - Alexander K Buell
- Department of Chemistry, University of Cambridge, Lensfield Road, Cambridge, CB2 1EW, UK.,Present address: Institute of Physical Biology, University of Düsseldorf, Universitätsstr. 1, 40225, Düsseldorf, Germany
| | - Ji-Eun Lee
- Department of Chemistry, University of Cambridge, Lensfield Road, Cambridge, CB2 1EW, UK
| | - Sonia Gandhi
- Department of Molecular Neuroscience, University College London, Institute of Neurology, Queen Square, London, WC1N 3BG, UK
| | - Steven F Lee
- Department of Chemistry, University of Cambridge, Lensfield Road, Cambridge, CB2 1EW, UK
| | - Clare E Bryant
- Department of Veterinary Medicine, University of Cambridge, Madingley Road, Cambridge, CB3 0ES, UK
| | - Michele Vendruscolo
- Department of Chemistry, University of Cambridge, Lensfield Road, Cambridge, CB2 1EW, UK
| | - Tuomas P J Knowles
- Department of Chemistry, University of Cambridge, Lensfield Road, Cambridge, CB2 1EW, UK
| | - Christopher M Dobson
- Department of Chemistry, University of Cambridge, Lensfield Road, Cambridge, CB2 1EW, UK.
| | - Erwin De Genst
- Department of Chemistry, University of Cambridge, Lensfield Road, Cambridge, CB2 1EW, UK. .,Present address: Astra Zeneca, Innovative Medicines Discovery Sciences Unit 310, Darwin Building, Cambridge Science Park, Milton Road, Cambridge, CB4 0WG, UK.
| | - David Klenerman
- Department of Chemistry, University of Cambridge, Lensfield Road, Cambridge, CB2 1EW, UK.
| |
Collapse
|
20
|
Vandesquille M, Li T, Po C, Ganneau C, Lenormand P, Dudeffant C, Czech C, Grueninger F, Duyckaerts C, Delatour B, Dhenain M, Lafaye P, Bay S. Chemically-defined camelid antibody bioconjugate for the magnetic resonance imaging of Alzheimer's disease. MAbs 2017; 9:1016-1027. [PMID: 28657418 DOI: 10.1080/19420862.2017.1342914] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Today, molecular imaging of neurodegenerative diseases is mainly based on small molecule probes. Alternatively, antibodies are versatile tools that may be developed as new imaging agents. Indeed, they can be readily obtained to specifically target any antigen of interest and their scaffold can be functionalized. One of the critical issues involved in translating antibody-based probes to the clinic is the design and synthesis of perfectly-defined conjugates. Camelid single-domain antibody-fragments (VHHs) are very small and stable antibodies that are able to diffuse in tissues and potentially cross the blood brain barrier (BBB). Here, we selected a VHH (R3VQ) specifically targeting one of the main lesions of Alzheimer's disease (AD), namely the amyloid-beta (Aß) deposits. It was used as a scaffold for the design of imaging probes for magnetic resonance imaging (MRI) and labeled with the contrastophore gadolinium using either a random or site-specific approach. In contrast to the random strategy, the site-specific conjugation to a single reduced cysteine in the C-terminal part of the R3VQ generates a well-defined bioconjugate in a high yield process. This new imaging probe is able to cross the BBB and label Aß deposits after intravenous injection. Also, it displays improved r1 and r2 relaxivities, up to 30 times higher than a widely used clinical contrast agent, and it allows MRI detection of amyloid deposits in post mortem brain tissue of a mouse model of AD. The ability to produce chemically-defined VHH conjugates that cross the BBB opens the way for future development of tailored imaging probes targeting intracerebral antigens.
Collapse
Affiliation(s)
- Matthias Vandesquille
- a Institut Pasteur, Unité de Chimie des Biomolécules, Département Biologie Structurale et Chimie , Paris , France.,b CNRS UMR 3523, 75724 , France.,d French Alternative Energies and Atomic Energy Commission , Institute of Biomedical Imaging, Molecular Imaging Research Center , Fontenay-aux-Roses , France
| | - Tengfei Li
- c Institut Pasteur, CITECH, Plateforme d'Ingénierie des Anticorps , 75724 , Paris , France.,e Sorbonne Universités, UPMC Univ. Paris 06 UMR S 1127, and Inserm, U 1127, and CNRS UMR 7225, and ICM, 75013, Paris , France.,f Université Paris Diderot-Paris 7, Paris , France
| | - Chrystelle Po
- a Institut Pasteur, Unité de Chimie des Biomolécules, Département Biologie Structurale et Chimie , Paris , France.,b CNRS UMR 3523, 75724 , France.,d French Alternative Energies and Atomic Energy Commission , Institute of Biomedical Imaging, Molecular Imaging Research Center , Fontenay-aux-Roses , France
| | - Christelle Ganneau
- a Institut Pasteur, Unité de Chimie des Biomolécules, Département Biologie Structurale et Chimie , Paris , France.,b CNRS UMR 3523, 75724 , France
| | - Pascal Lenormand
- c Institut Pasteur, CITECH, Plateforme d'Ingénierie des Anticorps , 75724 , Paris , France
| | - Clémence Dudeffant
- e Sorbonne Universités, UPMC Univ. Paris 06 UMR S 1127, and Inserm, U 1127, and CNRS UMR 7225, and ICM, 75013, Paris , France
| | - Christian Czech
- g F. Hoffmann-La Roche AG, Pharmaceutical Research and Early Development, NORD DTA, Roche Innovation Center Basel , Basel , Switzerland
| | - Fiona Grueninger
- g F. Hoffmann-La Roche AG, Pharmaceutical Research and Early Development, NORD DTA, Roche Innovation Center Basel , Basel , Switzerland
| | - Charles Duyckaerts
- e Sorbonne Universités, UPMC Univ. Paris 06 UMR S 1127, and Inserm, U 1127, and CNRS UMR 7225, and ICM, 75013, Paris , France
| | - Benoît Delatour
- e Sorbonne Universités, UPMC Univ. Paris 06 UMR S 1127, and Inserm, U 1127, and CNRS UMR 7225, and ICM, 75013, Paris , France
| | - Marc Dhenain
- d French Alternative Energies and Atomic Energy Commission , Institute of Biomedical Imaging, Molecular Imaging Research Center , Fontenay-aux-Roses , France
| | - Pierre Lafaye
- c Institut Pasteur, CITECH, Plateforme d'Ingénierie des Anticorps , 75724 , Paris , France
| | - Sylvie Bay
- a Institut Pasteur, Unité de Chimie des Biomolécules, Département Biologie Structurale et Chimie , Paris , France.,b CNRS UMR 3523, 75724 , France
| |
Collapse
|
21
|
Salahuddin P, Siddiqi MK, Khan S, Abdelhameed AS, Khan RH. Mechanisms of protein misfolding: Novel therapeutic approaches to protein-misfolding diseases. J Mol Struct 2016. [DOI: 10.1016/j.molstruc.2016.06.046] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
22
|
Bruce VJ, Ta AN, McNaughton BR. Minimalist Antibodies and Mimetics: An Update and Recent Applications. Chembiochem 2016; 17:1892-1899. [DOI: 10.1002/cbic.201600303] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2016] [Indexed: 12/14/2022]
Affiliation(s)
- Virginia J. Bruce
- Department of Chemistry; Colorado State University; Fort Collins CO 80523 USA
| | - Angeline N. Ta
- Department of Chemistry; Colorado State University; Fort Collins CO 80523 USA
| | - Brian R. McNaughton
- Department of Chemistry; Colorado State University; Fort Collins CO 80523 USA
- Department of Biochemistry and Molecular Biology; Colorado State University; Fort Collins CO 80523 USA
| |
Collapse
|
23
|
Abstract
Bispecific antibodies that engage immune cells to kill cancer cells have been actively studied in cancer immunotherapy. In this study, we present a novel bispecific format, S-Fab, fabricated by linking a single-domain anti-carcinoembryonic antigen VHH to a conventional anti-CD3 Fab. In contrast to most bispecific antibodies, the S-Fab bispecific antibody can be efficiently expressed and purified from bacteria. The purified S-Fab is stable in serum and is able to recruit T cells to drive potent cancer cell killing. In xenograft models, the S-Fab antibody suppresses tumor growth in the presence of human immune cells. Our study suggested that the bispecific S-Fab format can be applied to a wide range of immunotherapies.
Collapse
|
24
|
Steeland S, Vandenbroucke RE, Libert C. Nanobodies as therapeutics: big opportunities for small antibodies. Drug Discov Today 2016; 21:1076-113. [DOI: 10.1016/j.drudis.2016.04.003] [Citation(s) in RCA: 196] [Impact Index Per Article: 21.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2015] [Revised: 02/26/2016] [Accepted: 04/04/2016] [Indexed: 12/28/2022]
|
25
|
Abstract
Cancer immunotherapy has recently generated much excitement after the continuing success of the immunomodulating anti-CTLA-4 and anti-PD-1 antibodies against various types of cancers. Aside from these immunomodulating antibodies, bispecific antibodies, chimeric antigen receptor T cells, and other technologies are being actively studied. Among the various approaches to cancer immunotherapy, 2 bispecific antibodies are currently approved for patient care. Many more bispecific antibodies are now in various phases of clinical development and will become the next generation of antibody-based therapies. Further understanding of immunology and advances in protein engineering will help to generate a greater variety of bispecific antibodies to fight cancer. Here, we focus on bispecific antibodies that recruit immune cells to engage and kill tumor cells.
Collapse
Affiliation(s)
- Siqi Chen
- a School of Pharmaceutical Sciences, Sun Yat-Sen University , Guangzhou , China.,b Center for Cellular & Structural Biology, Sun Yat-Sen University , Guangzhou , China
| | - Jing Li
- a School of Pharmaceutical Sciences, Sun Yat-Sen University , Guangzhou , China.,b Center for Cellular & Structural Biology, Sun Yat-Sen University , Guangzhou , China
| | - Qing Li
- a School of Pharmaceutical Sciences, Sun Yat-Sen University , Guangzhou , China.,b Center for Cellular & Structural Biology, Sun Yat-Sen University , Guangzhou , China
| | - Zhong Wang
- a School of Pharmaceutical Sciences, Sun Yat-Sen University , Guangzhou , China.,b Center for Cellular & Structural Biology, Sun Yat-Sen University , Guangzhou , China
| |
Collapse
|
26
|
Burmistrova DA, Tillib SV, Shcheblyakov DV, Dolzhikova IV, Shcherbinin DN, Zubkova OV, Ivanova TI, Tukhvatulin AI, Shmarov MM, Logunov DY, Naroditsky BS, Gintsburg AL. Genetic Passive Immunization with Adenoviral Vector Expressing Chimeric Nanobody-Fc Molecules as Therapy for Genital Infection Caused by Mycoplasma hominis. PLoS One 2016; 11:e0150958. [PMID: 26962869 PMCID: PMC4786110 DOI: 10.1371/journal.pone.0150958] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2015] [Accepted: 02/22/2016] [Indexed: 12/18/2022] Open
Abstract
Developing pathogen-specific recombinant antibody fragments (especially nanobodies) is a very promising strategy for the treatment of infectious disease. Nanobodies have great potential for gene therapy application due to their single-gene nature. Historically, Mycoplasma hominis has not been considered pathogenic bacteria due to the lack of acute infection and partially due to multiple studies demonstrating high frequency of isolation of M. hominis samples from asymptomatic patients. However, recent studies on the role of latent M. hominis infection in oncologic transformation, especially prostate cancer, and reports that M. hominis infects Trichomonas and confers antibiotic resistance to Trichomonas, have generated new interest in this field. In the present study we have generated specific nanobody against M. hominis (aMh), for which the identified target is the ABC-transporter substrate-binding protein. aMh exhibits specific antibacterial action against M. hominis. In an attempt to improve the therapeutic properties, we have developed the adenoviral vector-based gene therapy approach for passive immunization with nanobodies against M. hominis. For better penetration into the mucous layer of the genital tract, we fused aMh with the Fc-fragment of IgG. Application of this comprehensive approach with a single systemic administration of recombinant adenovirus expressing aMh-Fc demonstrated both prophylactic and therapeutic effects in a mouse model of genital M. hominis infection.
Collapse
Affiliation(s)
- Daria A. Burmistrova
- Department of Immunobiotechnology, Gamaleya Research Center of Epidemiology and Microbiology, Moscow, Russia
- * E-mail: (D. Shcheblyakov); (DB)
| | - Sergey V. Tillib
- Department of Molecular Biotechnology, Institute of Gene Biology, Moscow, Russia
| | - Dmitry V. Shcheblyakov
- Department of Immunobiotechnology, Gamaleya Research Center of Epidemiology and Microbiology, Moscow, Russia
- * E-mail: (D. Shcheblyakov); (DB)
| | - Inna V. Dolzhikova
- Department of Cellular Microbiology, Gamaleya Research Center of Epidemiology and Microbiology, Moscow, Russia
| | - Dmitry N. Shcherbinin
- Department of Molecular Biotechnology, Gamaleya Research Center of Epidemiology and Microbiology, Moscow, Russia
| | - Olga V. Zubkova
- Department of Molecular Biotechnology, Gamaleya Research Center of Epidemiology and Microbiology, Moscow, Russia
| | - Tatiana I. Ivanova
- Department of Molecular Biotechnology, Institute of Gene Biology, Moscow, Russia
| | - Amir I. Tukhvatulin
- Department of Cellular Microbiology, Gamaleya Research Center of Epidemiology and Microbiology, Moscow, Russia
| | - Maxim M. Shmarov
- Department of Molecular Biotechnology, Gamaleya Research Center of Epidemiology and Microbiology, Moscow, Russia
| | - Denis Y. Logunov
- Department of Cellular Microbiology, Gamaleya Research Center of Epidemiology and Microbiology, Moscow, Russia
| | - Boris S. Naroditsky
- Department of Immunobiotechnology, Gamaleya Research Center of Epidemiology and Microbiology, Moscow, Russia
| | | |
Collapse
|
27
|
Dong B, Zhou C, He P, Li J, Chen S, Miao J, Li Q, Wang Z. A novel bispecific antibody, BiSS, with potent anti-cancer activities. Cancer Biol Ther 2016; 17:364-70. [PMID: 26828900 DOI: 10.1080/15384047.2016.1139266] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
One of the most active fields in cancer immunotherapy is the study of bispecific antibodies, which engage immune cells to kill cancer cells. However, a variety of issues are associated with most of current bispecific antibody formats. In this study, we present a novel bispecific antibody, BiSS (Bispecific antibody with Single domain, Single domain antibodies), which was constructed by linking 2 single domain antibodies, anti-CEA and anti-CD16, in tandem. Unlike most other bispecific antibodies, the BiSS antibody can be expressed and purified from E.coli in large quantities. By recruiting natural killer cells (NK cells) to CEA-positive cancer cells, BiSS led to cancer cell death in vitro. In xenograft models, the BiSS protein blocked cancer progression. The data suggested that the single domain-based bispecific antibody BiSS was functional and can be potentially applied to a broad range of immunotherapies.
Collapse
Affiliation(s)
- Bin Dong
- a School of Pharmaceutical Sciences , Sun Yat-Sen University , Guangzhou , China.,b Centre for Cellular & Structural Biology , Sun Yat-Sen University , Guangzhou , China
| | - Changhua Zhou
- a School of Pharmaceutical Sciences , Sun Yat-Sen University , Guangzhou , China.,b Centre for Cellular & Structural Biology , Sun Yat-Sen University , Guangzhou , China
| | - Ping He
- a School of Pharmaceutical Sciences , Sun Yat-Sen University , Guangzhou , China.,b Centre for Cellular & Structural Biology , Sun Yat-Sen University , Guangzhou , China
| | - Jing Li
- a School of Pharmaceutical Sciences , Sun Yat-Sen University , Guangzhou , China.,b Centre for Cellular & Structural Biology , Sun Yat-Sen University , Guangzhou , China
| | - Siqi Chen
- a School of Pharmaceutical Sciences , Sun Yat-Sen University , Guangzhou , China.,b Centre for Cellular & Structural Biology , Sun Yat-Sen University , Guangzhou , China
| | - Ji Miao
- a School of Pharmaceutical Sciences , Sun Yat-Sen University , Guangzhou , China.,b Centre for Cellular & Structural Biology , Sun Yat-Sen University , Guangzhou , China
| | - Qing Li
- a School of Pharmaceutical Sciences , Sun Yat-Sen University , Guangzhou , China.,b Centre for Cellular & Structural Biology , Sun Yat-Sen University , Guangzhou , China
| | - Zhong Wang
- a School of Pharmaceutical Sciences , Sun Yat-Sen University , Guangzhou , China.,b Centre for Cellular & Structural Biology , Sun Yat-Sen University , Guangzhou , China
| |
Collapse
|
28
|
Ji K, Zhao Y, Yu T, Wang Z, Gong H, Yang X, Liu Y, Huang K. Inhibition effects of tanshinone on the aggregation of α-synuclein. Food Funct 2016; 7:409-16. [DOI: 10.1039/c5fo00664c] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Bioactive tanshinone compounds inhibit the aggregation of α-synuclein and extend the life span of aC. elegansmodel of Parkinson's disease.
Collapse
Affiliation(s)
- Kaige Ji
- Tongji School of Pharmacy
- Huazhong University of Science and Technology
- Wuhan
- China
| | - Yudan Zhao
- Tongji School of Pharmacy
- Huazhong University of Science and Technology
- Wuhan
- China
| | - Tianhong Yu
- Tongji School of Pharmacy
- Huazhong University of Science and Technology
- Wuhan
- China
| | - Zhuoyi Wang
- Tongji School of Pharmacy
- Huazhong University of Science and Technology
- Wuhan
- China
| | - Hao Gong
- Tongji School of Pharmacy
- Huazhong University of Science and Technology
- Wuhan
- China
| | - Xin Yang
- Tongji School of Pharmacy
- Huazhong University of Science and Technology
- Wuhan
- China
| | - Yang Liu
- Synergy Innovation Center of Biological Peptide Antidiabetics of Hubei Province
- School of Life Science
- Wuchang University of Technology
- Wuhan
- China
| | - Kun Huang
- Tongji School of Pharmacy
- Huazhong University of Science and Technology
- Wuhan
- China
- Center for Biomedicine Research
| |
Collapse
|
29
|
Moreno-Del Álamo M, de la Espina SMD, Fernández-Tresguerres ME, Giraldo R. Pre-amyloid oligomers of the proteotoxic RepA-WH1 prionoid assemble at the bacterial nucleoid. Sci Rep 2015; 5:14669. [PMID: 26423724 PMCID: PMC4589793 DOI: 10.1038/srep14669] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2015] [Accepted: 09/03/2015] [Indexed: 12/13/2022] Open
Abstract
Upon binding to short specific dsDNA sequences in vitro, the N-terminal WH1 domain of the plasmid DNA replication initiator RepA assembles as amyloid fibres. These are bundles of single or double twisted tubular filaments in which distorted RepA-WH1 monomers are the building blocks. When expressed in Escherichia coli, RepA-WH1 triggers the first synthetic amyloid proteinopathy in bacteria, recapitulating some of the features of mammalian prion diseases: it is vertically transmissible, albeit non-infectious, showing up in at least two phenotypically distinct and interconvertible strains. Here we report B3h7, a monoclonal antibody specific for oligomers of RepA-WH1, but which does not recognize the mature amyloid fibres. Unlike a control polyclonal antibody generated against the soluble protein, B3h7 interferes in vitro with DNA-promoted or amyloid-seeded assembly of RepA-WH1 fibres, thus the targeted oligomers are on-pathway amyloidogenic intermediates. Immuno-electron microscopy with B3h7 on thin sections of E. coli cells expressing RepA-WH1 consistently labels the bacterial nucleoid, but not the large cytoplasmic aggregates of the protein. This observation points to the nucleoid as the place where oligomeric amyloid precursors of RepA-WH1 are generated, and suggests that, once nucleated by DNA, further growth must continue in the cytoplasm due to entropic exclusion.
Collapse
Affiliation(s)
- María Moreno-Del Álamo
- Department of Cellular and Molecular Biology, Centro de Investigaciones Biológicas - CSIC, Madrid E28040, Spain
| | | | | | - Rafael Giraldo
- Department of Cellular and Molecular Biology, Centro de Investigaciones Biológicas - CSIC, Madrid E28040, Spain
| |
Collapse
|