1
|
McArthur N, Squire JD, Onyeachonam OJ, Bhatt NN, Jerez C, Holberton AL, Tessier PM, Wood LB, Kayed R, Kane RS. Generation of nanobodies with conformational specificity for tau oligomers that recognize tau aggregates from human Alzheimer's disease samples. Biomater Sci 2024. [PMID: 39434503 DOI: 10.1039/d4bm00707g] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2024]
Abstract
Tauopathies are neurodegenerative diseases that involve tau misfolding and aggregation in the brain. These diseases, including Alzheimer's disease (AD), are some of the least understood and most difficult to treat neurodegenerative disorders. Antibodies and antibody fragments that target tau oligomers, which are especially toxic forms of tau, are promising options for immunotherapies and diagnostic tools for tauopathies. In this study, we have developed conformational, tau oligomer-specific nanobodies, or single-domain antibodies. We demonstrate that these nanobodies, OT2.4 and OT2.6, are highly specific for tau oligomers relative to tau monomers and fibrils. We used epitope mapping to verify that these nanobodies bind to discontinuous epitopes on tau and to support the idea that they interact with a conformation present in the oligomeric, and not monomeric or fibrillar, forms of tau. We show that these nanobodies interact with tau oligomers in brain samples from AD patients and from healthy older adults with primary age-related tauopathy. Our results demonstrate the potential of these nanobodies as tau oligomer-specific binding reagents and future tauopathy therapeutics and diagnostics.
Collapse
Affiliation(s)
- Nikki McArthur
- School of Chemical & Biomolecular Engineering, Georgia Institute of Technology, Atlanta, Georgia 30332, USA.
| | - Jay D Squire
- School of Chemical & Biomolecular Engineering, Georgia Institute of Technology, Atlanta, Georgia 30332, USA.
| | - Ogechukwu J Onyeachonam
- School of Chemical & Biomolecular Engineering, Georgia Institute of Technology, Atlanta, Georgia 30332, USA.
| | - Nemil N Bhatt
- Mitchell Center for Neurodegenerative Disease, University of Texas Medical Branch, Galveston, Texas 77555, USA
- Department of Neurology, University of Texas Medical Branch, Galveston, Texas 77555, USA
| | - Cynthia Jerez
- Mitchell Center for Neurodegenerative Disease, University of Texas Medical Branch, Galveston, Texas 77555, USA
- Department of Neurology, University of Texas Medical Branch, Galveston, Texas 77555, USA
| | - Abigail L Holberton
- George W. Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, Georgia 30332, USA
| | - Peter M Tessier
- Department of Chemical Engineering, University of Michigan, North Campus Research Complex, 2800 Plymouth Road, Ann Arbor, MI 48109, USA
- Biointerfaces Institute, University of Michigan, Ann Arbor, MI 48109, USA
- Department of Pharmaceutical Sciences, University of Michigan, Ann Arbor, MI 48109, USA
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI 48109, USA
| | - Levi B Wood
- George W. Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, Georgia 30332, USA
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, Georgia 30332, USA
- Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, Georgia 30332, USA
| | - Rakez Kayed
- Mitchell Center for Neurodegenerative Disease, University of Texas Medical Branch, Galveston, Texas 77555, USA
- Department of Neurology, University of Texas Medical Branch, Galveston, Texas 77555, USA
| | - Ravi S Kane
- School of Chemical & Biomolecular Engineering, Georgia Institute of Technology, Atlanta, Georgia 30332, USA.
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, Georgia 30332, USA
- Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, Georgia 30332, USA
| |
Collapse
|
2
|
McArthur N, Kang B, Rivera Moctezuma FG, Shaikh AT, Loeffler K, Bhatt NN, Kidd M, Zupancic JM, Desai AA, Djeddar N, Bryksin A, Tessier PM, Kayed R, Wood LB, Kane RS. Development of a pan-tau multivalent nanobody that binds tau aggregation motifs and recognizes pathological tau aggregates. Biotechnol Prog 2024; 40:e3463. [PMID: 38568030 PMCID: PMC11447142 DOI: 10.1002/btpr.3463] [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: 11/14/2023] [Revised: 02/29/2024] [Accepted: 03/21/2024] [Indexed: 08/16/2024]
Abstract
Alzheimer's disease and other tauopathies are characterized by the misfolding and aggregation of the tau protein into oligomeric and fibrillar structures. Antibodies against tau play an increasingly important role in studying these neurodegenerative diseases and the generation of tools to diagnose and treat them. The development of antibodies that recognize tau protein aggregates, however, is hindered by complex immunization and antibody selection strategies and limitations to antigen presentation. Here, we have taken a facile approach to identify single-domain antibodies, or nanobodies, that bind to many forms of tau by screening a synthetic yeast surface display nanobody library against monomeric tau and creating multivalent versions of our lead nanobody, MT3.1, to increase its avidity for tau aggregates. We demonstrate that MT3.1 binds to tau monomer, oligomers, and fibrils, as well as pathogenic tau from a tauopathy mouse model, despite being identified through screens against monomeric tau. Through epitope mapping, we discovered binding epitopes of MT3.1 contain the key motif VQIXXK which drives tau aggregation. We show that our bivalent and tetravalent versions of MT3.1 have greatly improved binding ability to tau oligomers and fibrils compared to monovalent MT3.1. Our results demonstrate the utility of our nanobody screening and multivalent design approach in developing nanobodies that bind amyloidogenic protein aggregates. This approach can be extended to the generation of multivalent nanobodies that target other amyloid proteins and has the potential to advance the research and treatment of neurodegenerative diseases.
Collapse
Affiliation(s)
- Nikki McArthur
- School of Chemical & Biomolecular Engineering, Georgia Institute of Technology, Atlanta, Georgia 30332, USA
| | - Bokyung Kang
- School of Chemical & Biomolecular Engineering, Georgia Institute of Technology, Atlanta, Georgia 30332, USA
| | - Felix G. Rivera Moctezuma
- George W. Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, Georgia 30332, USA
| | - Akber T. Shaikh
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, Georgia 30332, USA
| | - Kathryn Loeffler
- School of Chemical & Biomolecular Engineering, Georgia Institute of Technology, Atlanta, Georgia 30332, USA
| | - Nemil N. Bhatt
- Mitchell Center for Neurodegenerative Disease, University of Texas Medical Branch, Galveston, Texas 77555, USA
- Department of Neurology, University of Texas Medical Branch, Galveston, Texas 77555, USA
| | - Madison Kidd
- Mitchell Center for Neurodegenerative Disease, University of Texas Medical Branch, Galveston, Texas 77555, USA
- Department of Neurology, University of Texas Medical Branch, Galveston, Texas 77555, USA
| | - Jennifer M. Zupancic
- Department of Chemical Engineering, University of Michigan, North Campus Research Complex, 2800 Plymouth Road, Ann Arbor, MI 48109, USA
- Biointerfaces Institute, University of Michigan, Ann Arbor, MI 48109, USA
| | - Alec A. Desai
- Department of Chemical Engineering, University of Michigan, North Campus Research Complex, 2800 Plymouth Road, Ann Arbor, MI 48109, USA
- Biointerfaces Institute, University of Michigan, Ann Arbor, MI 48109, USA
| | - Naima Djeddar
- Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, Georgia 30332, USA
| | - Anton Bryksin
- Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, Georgia 30332, USA
| | - Peter M. Tessier
- Department of Chemical Engineering, University of Michigan, North Campus Research Complex, 2800 Plymouth Road, Ann Arbor, MI 48109, USA
- Biointerfaces Institute, University of Michigan, Ann Arbor, MI 48109, USA
- Department of Pharmaceutical Sciences, University of Michigan, Ann Arbor, MI 48109, USA
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI 48109, USA
| | - Rakez Kayed
- Mitchell Center for Neurodegenerative Disease, University of Texas Medical Branch, Galveston, Texas 77555, USA
- Department of Neurology, University of Texas Medical Branch, Galveston, Texas 77555, USA
| | - Levi B. Wood
- George W. Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, Georgia 30332, USA
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, Georgia 30332, USA
- Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, Georgia 30332, USA
| | - Ravi S. Kane
- School of Chemical & Biomolecular Engineering, Georgia Institute of Technology, Atlanta, Georgia 30332, USA
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, Georgia 30332, USA
- Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, Georgia 30332, USA
| |
Collapse
|
3
|
Shoemaker RL, Larsen RJ, Larsen PA. Single-domain antibodies and aptamers drive new opportunities for neurodegenerative disease research. Front Immunol 2024; 15:1426656. [PMID: 39238639 PMCID: PMC11374656 DOI: 10.3389/fimmu.2024.1426656] [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: 05/01/2024] [Accepted: 07/22/2024] [Indexed: 09/07/2024] Open
Abstract
Neurodegenerative diseases (NDs) in mammals, such as Alzheimer's disease (AD), Parkinson's disease (PD), and transmissible spongiform encephalopathies (TSEs), are characterized by the accumulation of misfolded proteins in the central nervous system (CNS). Despite the presence of these pathogenic proteins, the immune response in affected individuals remains notably muted. Traditional immunological strategies, particularly those reliant on monoclonal antibodies (mAbs), face challenges related to tissue penetration, blood-brain barrier (BBB) crossing, and maintaining protein stability. This has led to a burgeoning interest in alternative immunotherapeutic avenues. Notably, single-domain antibodies (or nanobodies) and aptamers have emerged as promising candidates, as their reduced size facilitates high affinity antigen binding and they exhibit superior biophysical stability compared to mAbs. Aptamers, synthetic molecules generated from DNA or RNA ligands, present both rapid production times and cost-effective solutions. Both nanobodies and aptamers exhibit inherent qualities suitable for ND research and therapeutic development. Cross-seeding events must be considered in both traditional and small-molecule-based immunodiagnostic and therapeutic approaches, as well as subsequent neurotoxic impacts and complications beyond protein aggregates. This review delineates the challenges traditional immunological methods pose in ND research and underscores the potential of nanobodies and aptamers in advancing next-generation ND diagnostics and therapeutics.
Collapse
Affiliation(s)
- Rachel L Shoemaker
- Minnesota Center for Prion Research and Outreach (MNPRO), University of Minnesota, St. Paul, MN, United States
- Department of Biomedical and Veterinary Sciences, University of Minnesota College of Veterinary Medicine, St. Paul, MN, United States
| | - Roxanne J Larsen
- Department of Biomedical and Veterinary Sciences, University of Minnesota College of Veterinary Medicine, St. Paul, MN, United States
- Priogen Corp., St. Paul, MN, United States
| | - Peter A Larsen
- Minnesota Center for Prion Research and Outreach (MNPRO), University of Minnesota, St. Paul, MN, United States
- Department of Biomedical and Veterinary Sciences, University of Minnesota College of Veterinary Medicine, St. Paul, MN, United States
| |
Collapse
|
4
|
Nair S, Jiang Y, Marchal IS, Chernobelsky E, Huang HW, Suh S, Pan R, Kong XP, Ryoo HD, Sigurdsson EM. Anti-tau single domain antibodies clear pathological tau and attenuate its toxicity and related functional defects. Cell Death Dis 2024; 15:543. [PMID: 39079958 PMCID: PMC11289317 DOI: 10.1038/s41419-024-06927-9] [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: 01/29/2024] [Revised: 07/15/2024] [Accepted: 07/18/2024] [Indexed: 08/02/2024]
Abstract
Tauopathies are a group of neurodegenerative diseases characterized by the presence of tau inclusions. We have developed over fifty anti-tau single-domain antibodies (sdAbs) derived from phage display libraries of a llama immunized with recombinant and pathological tau immunogens. We examined the therapeutic potential of four of these sdAbs in a Drosophila tauopathy model following their transgenic expression either in all neurons or neuronal subtypes. Three of these sdAbs showed therapeutic potential in various assays, effectively clearing pathological tau and attenuating or preventing tau-induced phenotypes that typically manifest as defects in neuronal axonal transport, neurodegeneration, functional impairments, and shortened lifespan. Of these three, one sdAb was superior in every assay, which may at least in part be attributed to its tau-binding epitope. These findings support its development as a gene therapy for tauopathies.
Collapse
Affiliation(s)
- Sudershana Nair
- Department of Neuroscience and Physiology, Neuroscience Institute, New York University Grossman School of Medicine, New York, NY, USA
- Department of Cell Biology, New York University Grossman School of Medicine, New York, NY, USA
| | - Yixiang Jiang
- Department of Neuroscience and Physiology, Neuroscience Institute, New York University Grossman School of Medicine, New York, NY, USA
| | - Isabella S Marchal
- Department of Neuroscience and Physiology, Neuroscience Institute, New York University Grossman School of Medicine, New York, NY, USA
- Department of Cell Biology, New York University Grossman School of Medicine, New York, NY, USA
| | - Elizabeth Chernobelsky
- Department of Neuroscience and Physiology, Neuroscience Institute, New York University Grossman School of Medicine, New York, NY, USA
- Department of Cell Biology, New York University Grossman School of Medicine, New York, NY, USA
| | - Huai-Wei Huang
- Department of Neuroscience and Physiology, Neuroscience Institute, New York University Grossman School of Medicine, New York, NY, USA
- Department of Cell Biology, New York University Grossman School of Medicine, New York, NY, USA
| | - Sarah Suh
- Department of Cell Biology, New York University Grossman School of Medicine, New York, NY, USA
| | - Ruimin Pan
- Department of Biochemistry and Molecular Pharmacology, New York University Grossman School of Medicine, New York, NY, USA
| | - Xiang-Peng Kong
- Department of Biochemistry and Molecular Pharmacology, New York University Grossman School of Medicine, New York, NY, USA
| | - Hyung Don Ryoo
- Department of Cell Biology, New York University Grossman School of Medicine, New York, NY, USA.
| | - Einar M Sigurdsson
- Department of Neuroscience and Physiology, Neuroscience Institute, New York University Grossman School of Medicine, New York, NY, USA.
- Department of Psychiatry, New York University Grossman School of Medicine, New York, NY, USA.
| |
Collapse
|
5
|
Fernandez-Martinez D, Kong Y, Goussard S, Zavala A, Gastineau P, Rey M, Ayme G, Chamot-Rooke J, Lafaye P, Vos M, Mechaly A, Duménil G. Cryo-EM structures of type IV pili complexed with nanobodies reveal immune escape mechanisms. Nat Commun 2024; 15:2414. [PMID: 38499587 PMCID: PMC10948894 DOI: 10.1038/s41467-024-46677-y] [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: 10/03/2023] [Accepted: 03/01/2024] [Indexed: 03/20/2024] Open
Abstract
Type IV pili (T4P) are prevalent, polymeric surface structures in pathogenic bacteria, making them ideal targets for effective vaccines. However, bacteria have evolved efficient strategies to evade type IV pili-directed antibody responses. Neisseria meningitidis are prototypical type IV pili-expressing Gram-negative bacteria responsible for life threatening sepsis and meningitis. This species has evolved several genetic strategies to modify the surface of its type IV pili, changing pilin subunit amino acid sequence, nature of glycosylation and phosphoforms, but how these modifications affect antibody binding at the structural level is still unknown. Here, to explore this question, we determine cryo-electron microscopy (cryo-EM) structures of pili of different sequence types with sufficiently high resolution to visualize posttranslational modifications. We then generate nanobodies directed against type IV pili which alter pilus function in vitro and in vivo. Cyro-EM in combination with molecular dynamics simulation of the nanobody-pilus complexes reveals how the different types of pili surface modifications alter nanobody binding. Our findings shed light on the impressive complementarity between the different strategies used by bacteria to avoid antibody binding. Importantly, we also show that structural information can be used to make informed modifications in nanobodies as countermeasures to these immune evasion mechanisms.
Collapse
Affiliation(s)
- David Fernandez-Martinez
- Institut Pasteur, Université Paris Cité, INSERM UMR1225, Pathogenesis of Vascular Infections, 75015, Paris, France
| | - Youxin Kong
- Institut Pasteur, Université Paris Cité, INSERM UMR1225, Pathogenesis of Vascular Infections, 75015, Paris, France
- Sanofi R&D, Integrated Drug Discovery, CRVA, 94403, Vitry-sur-Seine, France
| | - Sylvie Goussard
- Institut Pasteur, Université Paris Cité, INSERM UMR1225, Pathogenesis of Vascular Infections, 75015, Paris, France
| | - Agustin Zavala
- Institut Pasteur, Université Paris Cité, INSERM UMR1225, Pathogenesis of Vascular Infections, 75015, Paris, France
| | - Pauline Gastineau
- Institut Pasteur, Université Paris Cité, INSERM UMR1225, Pathogenesis of Vascular Infections, 75015, Paris, France
| | - Martial Rey
- Institut Pasteur, Université Paris-Cité, CNRS, UAR 2024, Mass Spectrometry for Biology, 75015, Paris, France
| | - Gabriel Ayme
- Institut Pasteur, Université Paris-Cité, CNRS-UMR 3528, Antibody Engineering Platform, 75015, Paris, France
| | - Julia Chamot-Rooke
- Institut Pasteur, Université Paris-Cité, CNRS, UAR 2024, Mass Spectrometry for Biology, 75015, Paris, France
| | - Pierre Lafaye
- Institut Pasteur, Université Paris-Cité, CNRS-UMR 3528, Antibody Engineering Platform, 75015, Paris, France
| | - Matthijn Vos
- NanoImaging Core Facility, Center for Technological Resources and Research, Institut Pasteur, 75015, Paris, France
| | - Ariel Mechaly
- Institut Pasteur, Crystallography Platform-C2RT, CNRS-UMR 3528, Université Paris Cité, Paris, France
| | - Guillaume Duménil
- Institut Pasteur, Université Paris Cité, INSERM UMR1225, Pathogenesis of Vascular Infections, 75015, Paris, France.
| |
Collapse
|
6
|
De Leiris N, Perret P, Lombardi C, Gözel B, Chierici S, Millet P, Debiossat M, Bacot S, Tournier BB, Chames P, Lenormand JL, Ghezzi C, Fagret D, Moulin M. A single-domain antibody for the detection of pathological Tau protein in the early stages of oligomerization. J Transl Med 2024; 22:163. [PMID: 38365700 PMCID: PMC10870657 DOI: 10.1186/s12967-024-04987-1] [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: 10/23/2023] [Accepted: 02/12/2024] [Indexed: 02/18/2024] Open
Abstract
BACKGROUND Soluble oligomeric forms of Tau protein have emerged as crucial players in the propagation of Tau pathology in Alzheimer's disease (AD). Our objective is to introduce a single-domain antibody (sdAb) named 2C5 as a novel radiotracer for the efficient detection and longitudinal monitoring of oligomeric Tau species in the human brain. METHODS The development and production of 2C5 involved llama immunization with the largest human Tau isoform oligomers of different maturation states. Subsequently, 2C5 underwent comprehensive in vitro characterization for affinity and specificity via Enzyme-Linked Immunosorbent Assay and immunohistochemistry on human brain slices. Technetium-99m was employed to radiolabel 2C5, followed by its administration to healthy mice for biodistribution analysis. RESULTS 2C5 exhibited robust binding affinity towards Tau oligomers (Kd = 6.280 nM ± 0.557) and to Tau fibers (Kd = 5.024 nM ± 0.453), with relatively weaker binding observed for native Tau protein (Kd = 1791 nM ± 8.714) and amyloid peptide (Kd > 10,000 nM). Remarkably, this SdAb facilitated immuno-histological labeling of pathological forms of Tau in neurons and neuritic plaques, yielding a high-contrast outcome in AD patients, closely mirroring the performance of reference antibodies AT8 and T22. Furthermore, 2C5 SdAb was successfully radiolabeled with 99mTc, preserving stability for up to 6 h post-radiolabeling (radiochemical purity > 93%). However, following intravenous injection into healthy mice, the predominant uptake occurred in kidneys, amounting to 115.32 ± 3.67, 97.70 ± 43.14 and 168.20 ± 34.52% of injected dose per gram (% ID/g) at 5, 10 and 45 min respectively. Conversely, brain uptake remained minimal at all measured time points, registering at 0.17 ± 0.03, 0.12 ± 0.07 and 0.02 ± 0.01% ID/g at 5, 10 and 45 min post-injection respectively. CONCLUSION 2C5 demonstrates excellent affinity and specificity for pathological Tau oligomers, particularly in their early stages of oligomerization. However, the current limitation of insufficient blood-brain barrier penetration necessitates further modifications before considering its application in nuclear medicine imaging for humans.
Collapse
Affiliation(s)
- Nicolas De Leiris
- University Grenoble Alpes, Clinique Universitaire de Médecine Nucléaire, INSERM, Centre Hospitalier Universitaire Grenoble Alpes, LRB, CS 10217, 38043, Grenoble CEDEX 9, France.
| | - Pascale Perret
- University Grenoble Alpes, INSERM, LRB, 38000, Grenoble, France
| | | | - Bülent Gözel
- University Grenoble Alpes, INSERM, LRB, 38000, Grenoble, France
| | - Sabine Chierici
- University Grenoble Alpes, CNRS, DCM, 38000, Grenoble, France
| | - Philippe Millet
- Division of Adult Psychiatry, Department of Psychiatry, Geneva University Hospitals, Geneva, Switzerland
| | | | - Sandrine Bacot
- University Grenoble Alpes, INSERM, LRB, 38000, Grenoble, France
| | - Benjamin B Tournier
- Division of Adult Psychiatry, Department of Psychiatry, Geneva University Hospitals, Geneva, Switzerland
| | - Patrick Chames
- Aix Marseille University, CNRS, INSERM, Institut Paoli-Calmettes, CRCM, Marseille, France
| | | | | | - Daniel Fagret
- University Grenoble Alpes, INSERM, LRB, 38000, Grenoble, France
| | - Marcelle Moulin
- University Grenoble Alpes, INSERM, LRB, 38000, Grenoble, France
| |
Collapse
|
7
|
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
|
8
|
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
|
9
|
Congdon EE, Ji C, Tetlow AM, Jiang Y, Sigurdsson EM. Tau-targeting therapies for Alzheimer disease: current status and future directions. Nat Rev Neurol 2023; 19:715-736. [PMID: 37875627 PMCID: PMC10965012 DOI: 10.1038/s41582-023-00883-2] [Citation(s) in RCA: 36] [Impact Index Per Article: 36.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/20/2023] [Indexed: 10/26/2023]
Abstract
Alzheimer disease (AD) is the most common cause of dementia in older individuals. AD is characterized pathologically by amyloid-β (Aβ) plaques and tau neurofibrillary tangles in the brain, with associated loss of synapses and neurons, which eventually results in dementia. Many of the early attempts to develop treatments for AD focused on Aβ, but a lack of efficacy of these treatments in terms of slowing disease progression led to a change of strategy towards targeting of tau pathology. Given that tau shows a stronger correlation with symptom severity than does Aβ, targeting of tau is more likely to be efficacious once cognitive decline begins. Anti-tau therapies initially focused on post-translational modifications, inhibition of tau aggregation and stabilization of microtubules. However, trials of many potential drugs were discontinued because of toxicity and/or lack of efficacy. Currently, the majority of tau-targeting agents in clinical trials are immunotherapies. In this Review, we provide an update on the results from the initial immunotherapy trials and an overview of new therapeutic candidates that are in clinical development, as well as considering future directions for tau-targeting therapies.
Collapse
Affiliation(s)
- Erin E Congdon
- Department of Neuroscience and Physiology, New York University Grossman School of Medicine, New York, NY, USA
- Neuroscience Institute, New York University Grossman School of Medicine, New York, NY, USA
| | - Changyi Ji
- Department of Neuroscience and Physiology, New York University Grossman School of Medicine, New York, NY, USA
- Neuroscience Institute, New York University Grossman School of Medicine, New York, NY, USA
| | - Amber M Tetlow
- Department of Neuroscience and Physiology, New York University Grossman School of Medicine, New York, NY, USA
- Neuroscience Institute, New York University Grossman School of Medicine, New York, NY, USA
| | - Yixiang Jiang
- Department of Neuroscience and Physiology, New York University Grossman School of Medicine, New York, NY, USA
- Neuroscience Institute, New York University Grossman School of Medicine, New York, NY, USA
| | - Einar M Sigurdsson
- Department of Neuroscience and Physiology, New York University Grossman School of Medicine, New York, NY, USA.
- Neuroscience Institute, New York University Grossman School of Medicine, New York, NY, USA.
- Department of Psychiatry, New York University Grossman School of Medicine, New York, NY, USA.
| |
Collapse
|
10
|
Han X, Li PH, Wang S, Sanchez M, Aggarwal S, Blakely T, Schalek R, Meirovitch Y, Lin Z, Berger D, Wu Y, Aly F, Bay S, Delatour B, LaFaye P, Pfister H, Wei D, Jain V, Ploegh H, Lichtman J. A large-scale volumetric correlated light and electron microscopy study localizes Alzheimer's disease-related molecules in the hippocampus. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.10.24.563674. [PMID: 37961104 PMCID: PMC10634883 DOI: 10.1101/2023.10.24.563674] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/15/2023]
Abstract
Connectomics is a nascent neuroscience field to map and analyze neuronal networks. It provides a new way to investigate abnormalities in brain tissue, including in models of Alzheimer's disease (AD). This age-related disease is associated with alterations in amyloid-β (Aβ) and phosphorylated tau (pTau). These alterations correlate with AD's clinical manifestations, but causal links remain unclear. Therefore, studying these molecular alterations within the context of the local neuronal and glial milieu may provide insight into disease mechanisms. Volume electron microscopy (vEM) is an ideal tool for performing connectomics studies at the ultrastructural level, but localizing specific biomolecules within large-volume vEM data has been challenging. Here we report a volumetric correlated light and electron microscopy (vCLEM) approach using fluorescent nanobodies as immuno-probes to localize Alzheimer's disease-related molecules in a large vEM volume. Three molecules (pTau, Aβ, and a marker for activated microglia (CD11b)) were labeled without the need for detergents by three nanobody probes in a sample of the hippocampus of the 3xTg Alzheimer's disease model mouse. Confocal microscopy followed by vEM imaging of the same sample allowed for registration of the location of the molecules within the volume. This dataset revealed several ultrastructural abnormalities regarding the localizations of Aβ and pTau in novel locations. For example, two pTau-positive post-synaptic spine-like protrusions innervated by axon terminals were found projecting from the axon initial segment of a pyramidal cell. Three pyramidal neurons with intracellular Aβ or pTau were 3D reconstructed. Automatic synapse detection, which is necessary for connectomics analysis, revealed the changes in density and volume of synapses at different distances from an Aβ plaque. This vCLEM approach is useful to uncover molecular alterations within large-scale volume electron microscopy data, opening a new connectomics pathway to study Alzheimer's disease and other types of dementia.
Collapse
|
11
|
Jumapili NA, Zivalj M, Barthelmess RM, Raes G, De Groof TWM, Devoogdt N, Stijlemans B, Vincke C, Van Ginderachter JA. A few good reasons to use nanobodies for cancer treatment. Eur J Immunol 2023; 53:e2250024. [PMID: 37366246 DOI: 10.1002/eji.202250024] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2023] [Revised: 04/29/2023] [Accepted: 05/17/2023] [Indexed: 06/28/2023]
Abstract
mAbs have been instrumental for targeted cancer therapies. However, their relatively large size and physicochemical properties result in a heterogenous distribution in the tumor microenvironment, usually restricted to the first cell layers surrounding blood vessels, and a limited ability to penetrate the brain. Nanobodies are tenfold smaller, resulting in a deeper tumor penetration and the ability to reach cells in poorly perfused tumor areas. Nanobodies are rapidly cleared from the circulation, which generates a fast target-to-background contrast that is ideally suited for molecular imaging purposes but may be less optimal for therapy. To circumvent this problem, nanobodies have been formatted to noncovalently bind albumin, increasing their serum half-life without majorly increasing their size. Finally, nanobodies have shown superior qualities to infiltrate brain tumors as compared to mAbs. In this review, we discuss why these features make nanobodies prime candidates for targeted therapy of cancer.
Collapse
Affiliation(s)
- Neema Ahishakiye Jumapili
- Laboratory of Cellular and Molecular Immunology, Vrije Universiteit Brussel, Brussels, Belgium
- Myeloid Cell Immunology Laboratory, VIB Center for Inflammation Research, Brussels, Belgium
| | - Maida Zivalj
- Laboratory of Cellular and Molecular Immunology, Vrije Universiteit Brussel, Brussels, Belgium
- Myeloid Cell Immunology Laboratory, VIB Center for Inflammation Research, Brussels, Belgium
| | - Romina Mora Barthelmess
- Laboratory of Cellular and Molecular Immunology, Vrije Universiteit Brussel, Brussels, Belgium
- Myeloid Cell Immunology Laboratory, VIB Center for Inflammation Research, Brussels, Belgium
| | - Geert Raes
- Laboratory of Cellular and Molecular Immunology, Vrije Universiteit Brussel, Brussels, Belgium
- Myeloid Cell Immunology Laboratory, VIB Center for Inflammation Research, Brussels, Belgium
| | - Timo W M De Groof
- 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
| | - Benoît Stijlemans
- Laboratory of Cellular and Molecular Immunology, Vrije Universiteit Brussel, Brussels, Belgium
- Myeloid Cell Immunology Laboratory, VIB Center for Inflammation Research, Brussels, Belgium
| | - Cécile Vincke
- Laboratory of Cellular and Molecular Immunology, Vrije Universiteit Brussel, Brussels, Belgium
- Myeloid Cell Immunology Laboratory, VIB Center for Inflammation Research, Brussels, Belgium
| | - Jo A Van Ginderachter
- Laboratory of Cellular and Molecular Immunology, Vrije Universiteit Brussel, Brussels, Belgium
- Myeloid Cell Immunology Laboratory, VIB Center for Inflammation Research, Brussels, Belgium
| |
Collapse
|
12
|
Kunz S, Durandy M, Seguin L, Feral CC. NANOBODY ® Molecule, a Giga Medical Tool in Nanodimensions. Int J Mol Sci 2023; 24:13229. [PMID: 37686035 PMCID: PMC10487883 DOI: 10.3390/ijms241713229] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2023] [Revised: 08/22/2023] [Accepted: 08/23/2023] [Indexed: 09/10/2023] Open
Abstract
Although antibodies remain the most widely used tool for biomedical research, antibody technology is not flawless. Innovative alternatives, such as Nanobody® molecules, were developed to address the shortcomings of conventional antibodies. Nanobody® molecules are antigen-binding variable-domain fragments derived from the heavy-chain-only antibodies of camelids (VHH) and combine the advantageous properties of small molecules and monoclonal antibodies. Nanobody® molecules present a small size (~15 kDa, 4 nm long and 2.5 nm wide), high solubility, stability, specificity, and affinity, ease of cloning, and thermal and chemical resistance. Recombinant production in microorganisms is cost-effective, and VHH are also building blocks for multidomain constructs. These unique features led to numerous applications in fundamental research, diagnostics, and therapy. Nanobody® molecules are employed as biomarker probes and, when fused to radioisotopes or fluorophores, represent ideal non-invasive in vivo imaging agents. They can be used as neutralizing agents, receptor-ligand antagonists, or in targeted vehicle-based drug therapy. As early as 2018, the first Nanobody®, Cablivi (caplacizumab), a single-domain antibody (sdAb) drug developed by French pharmaceutical giant Sanofi for the treatment of adult patients with acquired thrombocytopenic purpura (aTTP), was launched. Nanobody® compounds are ideal tools for further development in clinics for diagnostic and therapeutic purposes.
Collapse
Affiliation(s)
- Sarah Kunz
- Université Côte d’Azur, CNRS UMR7284, INSERM U1081, IRCAN, 06107 Nice, France; (S.K.); (M.D.); (L.S.)
- Department of Oncology, Sanofi Research Center, 94400 Vitry-sur-Seine, France
| | - Manon Durandy
- Université Côte d’Azur, CNRS UMR7284, INSERM U1081, IRCAN, 06107 Nice, France; (S.K.); (M.D.); (L.S.)
| | - Laetitia Seguin
- Université Côte d’Azur, CNRS UMR7284, INSERM U1081, IRCAN, 06107 Nice, France; (S.K.); (M.D.); (L.S.)
| | - Chloe C. Feral
- Université Côte d’Azur, CNRS UMR7284, INSERM U1081, IRCAN, 06107 Nice, France; (S.K.); (M.D.); (L.S.)
| |
Collapse
|
13
|
Zupancic JM, Smith MD, Trzeciakiewicz H, Skinner ME, Ferris SP, Makowski EK, Lucas MJ, McArthur N, Kane RS, Paulson HL, Tessier PM. Quantitative flow cytometric selection of tau conformational nanobodies specific for pathological aggregates. Front Immunol 2023; 14:1164080. [PMID: 37622125 PMCID: PMC10445546 DOI: 10.3389/fimmu.2023.1164080] [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: 02/12/2023] [Accepted: 05/15/2023] [Indexed: 08/26/2023] Open
Abstract
Single-domain antibodies, also known as nanobodies, are broadly important for studying the structure and conformational states of several classes of proteins, including membrane proteins, enzymes, and amyloidogenic proteins. Conformational nanobodies specific for aggregated conformations of amyloidogenic proteins are particularly needed to better target and study aggregates associated with a growing class of associated diseases, especially neurodegenerative disorders such as Alzheimer's and Parkinson's diseases. However, there are few reported nanobodies with both conformational and sequence specificity for amyloid aggregates, especially for large and complex proteins such as the tau protein associated with Alzheimer's disease, due to difficulties in selecting nanobodies that bind to complex aggregated proteins. Here, we report the selection of conformational nanobodies that selectively recognize aggregated (fibrillar) tau relative to soluble (monomeric) tau. Notably, we demonstrate that these nanobodies can be directly isolated from immune libraries using quantitative flow cytometric sorting of yeast-displayed libraries against tau aggregates conjugated to quantum dots, and this process eliminates the need for secondary nanobody screening. The isolated nanobodies demonstrate conformational specificity for tau aggregates in brain samples from both a transgenic mouse model and human tauopathies. We expect that our facile approach will be broadly useful for isolating conformational nanobodies against diverse amyloid aggregates and other complex antigens.
Collapse
Affiliation(s)
- Jennifer M. Zupancic
- Department of Chemical Engineering, University of Michigan, Ann Arbor, MI, United States
- Biointerfaces Institute, University of Michigan, Ann Arbor, MI, United States
| | - Matthew D. Smith
- Department of Chemical Engineering, University of Michigan, Ann Arbor, MI, United States
- Biointerfaces Institute, University of Michigan, Ann Arbor, MI, United States
| | - Hanna Trzeciakiewicz
- Department of Translational Neuroscience, Michigan State University, Grand Rapids, MI, United States
| | - Mary E. Skinner
- Department of Neurology, University of Michigan, Ann Arbor, MI, United States
| | - Sean P. Ferris
- Department of Pathology, University of Michigan, Ann Arbor, MI, United States
| | - Emily K. Makowski
- Biointerfaces Institute, University of Michigan, Ann Arbor, MI, United States
- Department of Pharmaceutical Sciences, University of Michigan, Ann Arbor, MI, United States
| | - Michael J. Lucas
- Department of Chemical Engineering, University of Michigan, Ann Arbor, MI, United States
- Biointerfaces Institute, University of Michigan, Ann Arbor, MI, United States
- Department of Pharmaceutical Sciences, University of Michigan, Ann Arbor, MI, United States
| | - Nikki McArthur
- School of Chemical and Biomolecular Engineering, Georgia Institute of Technology, Atlanta, GA, United States
| | - Ravi S. Kane
- School of Chemical and Biomolecular Engineering, Georgia Institute of Technology, Atlanta, GA, United States
| | - Henry L. Paulson
- Department of Neurology, University of Michigan, Ann Arbor, MI, United States
- Protein Folding Disease Initiative, University of Michigan, Ann Arbor, MI, United States
- Michigan Alzheimer’s Disease Center, University of Michigan, Ann Arbor, MI, United States
| | - Peter M. Tessier
- Department of Chemical Engineering, University of Michigan, Ann Arbor, MI, United States
- Biointerfaces Institute, University of Michigan, Ann Arbor, MI, United States
- Department of Pharmaceutical Sciences, University of Michigan, Ann Arbor, MI, United States
- Protein Folding Disease Initiative, University of Michigan, Ann Arbor, MI, United States
- Michigan Alzheimer’s Disease Center, University of Michigan, Ann Arbor, MI, United States
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, United States
| |
Collapse
|
14
|
Cuche C, Mastrogiovanni M, Juzans M, Laude H, Ungeheuer MN, Krentzel D, Gariboldi MI, Scott-Algara D, Madec M, Goyard S, Floch C, Chauveau-Le Friec G, Lafaye P, Renaudat C, Le Bidan M, Micallef C, Schmutz S, Mella S, Novault S, Hasan M, Duffy D, Di Bartolo V, Alcover A. T cell migration and effector function differences in familial adenomatous polyposis patients with APC gene mutations. Front Immunol 2023; 14:1163466. [PMID: 37533857 PMCID: PMC10393261 DOI: 10.3389/fimmu.2023.1163466] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2023] [Accepted: 05/11/2023] [Indexed: 08/04/2023] Open
Abstract
Familial adenomatous polyposis (FAP) is an inherited disease characterized by the development of large number of colorectal adenomas with high risk of evolving into colorectal tumors. Mutations of the Adenomatous polyposis coli (APC) gene is often at the origin of this disease, as well as of a high percentage of spontaneous colorectal tumors. APC is therefore considered a tumor suppressor gene. While the role of APC in intestinal epithelium homeostasis is well characterized, its importance in immune responses remains ill defined. Our recent work indicates that the APC protein is involved in various phases of both CD4 and CD8 T cells responses. This prompted us to investigate an array of immune cell features in FAP subjects carrying APC mutations. A group of 12 FAP subjects and age and sex-matched healthy controls were studied. We characterized the immune cell repertoire in peripheral blood and the capacity of immune cells to respond ex vivo to different stimuli either in whole blood or in purified T cells. A variety of experimental approaches were used, including, pultiparamater flow cytometry, NanosString gene expression profiling, Multiplex and regular ELISA, confocal microscopy and computer-based image analyis methods. We found that the percentage of several T and natural killer (NK) cell populations, the expression of several genes induced upon innate or adaptive immune stimulation and the production of several cytokines and chemokines was different. Moreover, the capacity of T cells to migrate in response to chemokine was consistently altered. Finally, immunological synapses between FAP cytotoxic T cells and tumor target cells were more poorly structured. Our findings of this pilot study suggest that mild but multiple immune cell dysfunctions, together with intestinal epithelial dysplasia in FAP subjects, may facilitate the long-term polyposis and colorectal tumor development. Although at an initial discovery phase due to the limited sample size of this rare disease cohort, our findings open new perspectives to consider immune cell abnormalities into polyposis pathology.
Collapse
Affiliation(s)
- Céline Cuche
- Institut Pasteur, Université Paris Cité, INSERM-U1224, Unité Biologie Cellulaire des Lymphocytes, Ligue Nationale Contre le Cancer-Équipe Labellisée Ligue 2018, Paris, France
| | - Marta Mastrogiovanni
- Institut Pasteur, Université Paris Cité, INSERM-U1224, Unité Biologie Cellulaire des Lymphocytes, Ligue Nationale Contre le Cancer-Équipe Labellisée Ligue 2018, Paris, France
- Sorbonne Université, Collège Doctoral, Paris, France
| | - Marie Juzans
- Institut Pasteur, Université Paris Cité, INSERM-U1224, Unité Biologie Cellulaire des Lymphocytes, Ligue Nationale Contre le Cancer-Équipe Labellisée Ligue 2018, Paris, France
- Sorbonne Université, Collège Doctoral, Paris, France
| | - Hélène Laude
- Institut Pasteur, Université Paris Cité, ICAReB-Clin, Paris, France
| | | | - Daniel Krentzel
- Institut Pasteur, Université Paris Cité, CNRS-UMR3691, Unité Imagerie et Modélisation, Paris, France
| | - Maria Isabella Gariboldi
- Institut Pasteur, Université Paris Cité, CNRS-UMR3691, Unité Imagerie et Modélisation, Paris, France
| | - Daniel Scott-Algara
- Institut Pasteur, Université Paris Cité, INSERM-U1224, Unité Biologie Cellulaire des Lymphocytes, Ligue Nationale Contre le Cancer-Équipe Labellisée Ligue 2018, Paris, France
| | - Marianne Madec
- Institut Pasteur, Université Paris Cité, INSERM-U1224, Unité Biologie Cellulaire des Lymphocytes, Ligue Nationale Contre le Cancer-Équipe Labellisée Ligue 2018, Paris, France
| | - Sophie Goyard
- Institut Pasteur, Université Paris Cité, INSERM-U1224, Unité Biologie Cellulaire des Lymphocytes, Ligue Nationale Contre le Cancer-Équipe Labellisée Ligue 2018, Paris, France
- Institut Pasteur, Université Paris Cité, Plateforme d’Innovation et de Développement de Tests Diagnostiques, Paris, France
| | - Camille Floch
- Institut Pasteur, Université Paris Cité, INSERM-U1224, Unité Biologie Cellulaire des Lymphocytes, Ligue Nationale Contre le Cancer-Équipe Labellisée Ligue 2018, Paris, France
- Institut Pasteur, Université Paris Cité, Plateforme d’Innovation et de Développement de Tests Diagnostiques, Paris, France
| | - Gaëlle Chauveau-Le Friec
- Institut Pasteur, Université Paris Cité, CNRS-UMR3528, Plateforme d’Ingénierie des Anticorps, Paris, France
| | - Pierre Lafaye
- Institut Pasteur, Université Paris Cité, CNRS-UMR3528, Plateforme d’Ingénierie des Anticorps, Paris, France
| | | | | | | | - Sandrine Schmutz
- Institut Pasteur, Université Paris Cité, Unité de Technologie et Service Cytométrie et Biomarqueurs, Paris, France
| | - Sébastien Mella
- Institut Pasteur, Université Paris Cité, Unité de Technologie et Service Cytométrie et Biomarqueurs, Paris, France
- Institut Pasteur, Université Paris Cité, Hub Bioinformatique et Biostatistique, Paris, France
| | - Sophie Novault
- Institut Pasteur, Université Paris Cité, Unité de Technologie et Service Cytométrie et Biomarqueurs, Paris, France
| | - Milena Hasan
- Institut Pasteur, Université Paris Cité, Unité de Technologie et Service Cytométrie et Biomarqueurs, Paris, France
| | - Darragh Duffy
- Institut Pasteur, Université Paris Cité, Unité Immunologie Translationnelle, Paris, France
| | - Vincenzo Di Bartolo
- Institut Pasteur, Université Paris Cité, INSERM-U1224, Unité Biologie Cellulaire des Lymphocytes, Ligue Nationale Contre le Cancer-Équipe Labellisée Ligue 2018, Paris, France
| | - Andrés Alcover
- Institut Pasteur, Université Paris Cité, INSERM-U1224, Unité Biologie Cellulaire des Lymphocytes, Ligue Nationale Contre le Cancer-Équipe Labellisée Ligue 2018, Paris, France
| |
Collapse
|
15
|
Li Q, Nemecz Á, Aymé G, Dejean de la Bâtie G, Prevost MS, Pons S, Barilone N, Baachaoui R, Maskos U, Lafaye P, Corringer PJ. Generation of nanobodies acting as silent and positive allosteric modulators of the α7 nicotinic acetylcholine receptor. Cell Mol Life Sci 2023; 80:164. [PMID: 37231269 DOI: 10.1007/s00018-023-04779-8] [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: 12/15/2022] [Revised: 03/28/2023] [Accepted: 04/07/2023] [Indexed: 05/27/2023]
Abstract
The α7 nicotinic acetylcholine receptor (nAChR), a potential drug target for treating cognitive disorders, mediates communication between neuronal and non-neuronal cells. Although many competitive antagonists, agonists, and partial-agonists have been found and synthesized, they have not led to effective therapeutic treatments. In this context, small molecules acting as positive allosteric modulators binding outside the orthosteric, acetylcholine, site have attracted considerable interest. Two single-domain antibody fragments, C4 and E3, against the extracellular domain of the human α7-nAChR were generated through alpaca immunization with cells expressing a human α7-nAChR/mouse 5-HT3A chimera, and are herein described. They bind to the α7-nAChR but not to the other major nAChR subtypes, α4β2 and α3β4. E3 acts as a slowly associating positive allosteric modulator, strongly potentiating the acetylcholine-elicited currents, while not precluding the desensitization of the receptor. An E3-E3 bivalent construct shows similar potentiating properties but displays very slow dissociation kinetics conferring quasi-irreversible properties. Whereas, C4 does not alter the receptor function, but fully inhibits the E3-evoked potentiation, showing it is a silent allosteric modulator competing with E3 binding. Both nanobodies do not compete with α-bungarotoxin, localizing at an allosteric extracellular binding site away from the orthosteric site. The functional differences of each nanobody, as well as the alteration of functional properties through nanobody modifications indicate the importance of this extracellular site. The nanobodies will be useful for pharmacological and structural investigations; moreover, they, along with the extracellular site, have a direct potential for clinical applications.
Collapse
Affiliation(s)
- Qimeng Li
- Institut Pasteur, Université Paris Cité, CNRS UMR 3571, Channel-Receptors Unit, Paris, France
- Institut Pasteur, Université Paris Cité, CNRS UMR 3528, Antibody Engineering Platform, Paris, France
- Lanzhou Institute of Biological Product Co., Lanzhou, China
- Sorbonne Université, Collège Doctoral, Paris, France
| | - Ákos Nemecz
- Institut Pasteur, Université Paris Cité, CNRS UMR 3571, Channel-Receptors Unit, Paris, France.
| | - Gabriel Aymé
- Institut Pasteur, Université Paris Cité, CNRS UMR 3528, Antibody Engineering Platform, Paris, France.
| | | | - Marie S Prevost
- Institut Pasteur, Université Paris Cité, CNRS UMR 3571, Channel-Receptors Unit, Paris, France
| | - Stéphanie Pons
- Institut Pasteur, Université Paris Cité, CNRS UMR 3571, Integrative Neurobiology of Cholinergic Systems Unit, Paris, France
| | - Nathalie Barilone
- Institut Pasteur, Université Paris Cité, CNRS UMR 3571, Channel-Receptors Unit, Paris, France
| | - Rayen Baachaoui
- Institut Pasteur, Université Paris Cité, CNRS UMR 3571, Channel-Receptors Unit, Paris, France
- Institut Pasteur, Université Paris Cité, CNRS UMR 3528, Antibody Engineering Platform, Paris, France
| | - Uwe Maskos
- Institut Pasteur, Université Paris Cité, CNRS UMR 3571, Integrative Neurobiology of Cholinergic Systems Unit, Paris, France
| | - Pierre Lafaye
- Institut Pasteur, Université Paris Cité, CNRS UMR 3528, Antibody Engineering Platform, Paris, France
| | - Pierre-Jean Corringer
- Institut Pasteur, Université Paris Cité, CNRS UMR 3571, Channel-Receptors Unit, Paris, France.
| |
Collapse
|
16
|
Ramirez DM, Whitesell JD, Bhagwat N, Thomas TL, Ajay AD, Nawaby A, Delatour B, Bay S, LaFaye P, Knox JE, Harris JA, Meeks JP, Diamond MI. Endogenous pathology in tauopathy mice progresses via brain networks. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.05.23.541792. [PMID: 37293074 PMCID: PMC10245958 DOI: 10.1101/2023.05.23.541792] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Neurodegenerative tauopathies are hypothesized to propagate via brain networks. This is uncertain because we have lacked precise network resolution of pathology. We therefore developed whole-brain staining methods with anti-p-tau nanobodies and imaged in 3D PS19 tauopathy mice, which have pan-neuronal expression of full-length human tau containing the P301S mutation. We analyzed patterns of p-tau deposition across established brain networks at multiple ages, testing the relationship between structural connectivity and patterns of progressive pathology. We identified core regions with early tau deposition, and used network propagation modeling to determine the link between tau pathology and connectivity strength. We discovered a bias towards retrograde network-based propagation of tau. This novel approach establishes a fundamental role for brain networks in tau propagation, with implications for human disease.
Collapse
Affiliation(s)
- Denise M.O. Ramirez
- Department of Neurology, Peter O’Donnell Jr. Brain Institute, University of Texas Southwestern Medical Center; Dallas, TX, USA
| | - Jennifer D. Whitesell
- Allen Institute for Brain Science; Seattle, WA, USA
- Cajal Neuroscience; Seattle, WA, USA
| | - Nikhil Bhagwat
- Allen Institute for Brain Science; Seattle, WA, USA
- McConnell Brain Imaging Centre, The Neuro (Montreal Neurological Institute-Hospital), McGill University; Montreal, Quebec, Canada
| | - Talitha L. Thomas
- Center for Alzheimer’s and Neurodegenerative Diseases, Peter O’Donnell Jr. Brain Institute, University of Texas Southwestern Medical Center; Dallas, TX, USA
| | - Apoorva D. Ajay
- Department of Neurology, Peter O’Donnell Jr. Brain Institute, University of Texas Southwestern Medical Center; Dallas, TX, USA
| | - Ariana Nawaby
- Department of Neurology, Peter O’Donnell Jr. Brain Institute, University of Texas Southwestern Medical Center; Dallas, TX, USA
| | - Benoît Delatour
- Paris Brain Institute (ICM), CNRS UMR 7225, INSERM U1127, Sorbonne Université, Hôpital de la Pitié-Salpêtrière; Paris, France
| | - Sylvie Bay
- Unité de Chimie des Biomolécules, Institut Pasteur, Université Paris Cité, CNRS UMR 3523; Paris, France
| | - Pierre LaFaye
- Antibody Engineering Platform, Institut Pasteur, Université Paris Cité, CNRS UMR 3528; Paris, France
| | | | | | - Julian P. Meeks
- Department of Neuroscience, University of Rochester Medical School; Rochester, NY, USA
| | - Marc I. Diamond
- Center for Alzheimer’s and Neurodegenerative Diseases, Peter O’Donnell Jr. Brain Institute, University of Texas Southwestern Medical Center; Dallas, TX, USA
| |
Collapse
|
17
|
Jiang Y, Lin Y, Krishnaswamy S, Pan R, Wu Q, Sandusky-Beltran LA, Liu M, Kuo MH, Kong XP, Congdon EE, Sigurdsson EM. Single-domain antibody-based noninvasive in vivo imaging of α-synuclein or tau pathology. SCIENCE ADVANCES 2023; 9:eadf3775. [PMID: 37163602 PMCID: PMC10171817 DOI: 10.1126/sciadv.adf3775] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/19/2022] [Accepted: 04/11/2023] [Indexed: 05/12/2023]
Abstract
Intracellular deposition of α-synuclein and tau are hallmarks of synucleinopathies and tauopathies, respectively. Recently, several dye-based imaging probes with selectivity for tau aggregates have been developed, but suitable imaging biomarkers for synucleinopathies are still unavailable. Detection of both of these aggregates early in the disease process may allow for prophylactic therapies before functional impairments have manifested, highlighting the importance of developing specific imaging probes for these lesions. In contrast to the β sheet dyes, single-domain antibodies, found in camelids and a few other species, are highly specific, and their small size allows better brain entry and distribution than whole antibodies. Here, we have developed such imaging ligands via phage display libraries derived from llamas immunized with α-synuclein and tau preparations, respectively. These probes allow noninvasive and specific in vivo imaging of α-synuclein versus tau pathology in mice, with the brain signal correlating strongly with lesion burden. These small antibody derivatives have great potential for in vivo diagnosis of these diseases.
Collapse
Affiliation(s)
- Yixiang Jiang
- Department of Neuroscience and Physiology, Neuroscience Institute, New York University Grossman School of Medicine, 435 East 30th Street, New York, NY 10016, USA
| | - Yan Lin
- Department of Neuroscience and Physiology, Neuroscience Institute, New York University Grossman School of Medicine, 435 East 30th Street, New York, NY 10016, USA
| | - Senthilkumar Krishnaswamy
- Department of Neuroscience and Physiology, Neuroscience Institute, New York University Grossman School of Medicine, 435 East 30th Street, New York, NY 10016, USA
| | - Ruimin Pan
- Department of Biochemistry and Molecular Pharmacology, New York University Grossman School of Medicine, 550 First Avenue, New York, NY 10016, USA
| | - Qian Wu
- Department of Neuroscience and Physiology, Neuroscience Institute, New York University Grossman School of Medicine, 435 East 30th Street, New York, NY 10016, USA
| | - Leslie A. Sandusky-Beltran
- Department of Neuroscience and Physiology, Neuroscience Institute, New York University Grossman School of Medicine, 435 East 30th Street, New York, NY 10016, USA
| | - Mengyu Liu
- Department of Biochemistry and Molecular Biology, Michigan State University, 603 Wilson Road, East Lansing, MI 48824, USA
| | - Min-Hao Kuo
- Department of Biochemistry and Molecular Biology, Michigan State University, 603 Wilson Road, East Lansing, MI 48824, USA
| | - Xiang-Peng Kong
- Department of Biochemistry and Molecular Pharmacology, New York University Grossman School of Medicine, 550 First Avenue, New York, NY 10016, USA
| | - Erin E. Congdon
- Department of Neuroscience and Physiology, Neuroscience Institute, New York University Grossman School of Medicine, 435 East 30th Street, New York, NY 10016, USA
| | - Einar M. Sigurdsson
- Department of Neuroscience and Physiology, Neuroscience Institute, New York University Grossman School of Medicine, 435 East 30th Street, New York, NY 10016, USA
- Department of Psychiatry, New York University Grossman School of Medicine, 435 East 30th Street, New York, NY 10016, USA
| |
Collapse
|
18
|
Lerouge F, Ong E, Rositi H, Mpambani F, Berner LP, Bolbos R, Olivier C, Peyrin F, Apputukan VK, Monnereau C, Andraud C, Chaput F, Berthezène Y, Braun B, Jucker M, Åslund AK, Nyström S, Hammarström P, R Nilsson KP, Lindgren M, Wiart M, Chauveau F, Parola S. In vivo targeting and multimodal imaging of cerebral amyloid-β aggregates using hybrid GdF 3 nanoparticles. Nanomedicine (Lond) 2023; 17:2173-2187. [PMID: 36927004 DOI: 10.2217/nnm-2022-0252] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/18/2023] Open
Abstract
Aim: To propose a new multimodal imaging agent targeting amyloid-β (Aβ) plaques in Alzheimer's disease. Materials & methods: A new generation of hybrid contrast agents, based on gadolinium fluoride nanoparticles grafted with a pentameric luminescent-conjugated polythiophene, was designed, extensively characterized and evaluated in animal models of Alzheimer's disease through MRI, two-photon microscopy and synchrotron x-ray phase-contrast imaging. Results & conclusion: Two different grafting densities of luminescent-conjugated polythiophene were achieved while preserving colloidal stability and fluorescent properties, and without affecting biodistribution. In vivo brain uptake was dependent on the blood-brain barrier status. Nevertheless, multimodal imaging showed successful Aβ targeting in both transgenic mice and Aβ fibril-injected rats.
Collapse
Affiliation(s)
- Frédéric Lerouge
- University of Lyon, École Normale Supérieure de Lyon, Laboratoire de Chimie, University of Lyon 1, CNRS UMR, 5182, Lyon, France
| | - Elodie Ong
- University of Lyon, Lyon Neuroscience Research Center, CNRS UMR, 5292, INSERM U1028, University of Lyon 1, Lyon, France
| | - Hugo Rositi
- University of Clermont Auvergne, Clermont Auvergne INP, Institut Pascal, CNRS UMR, 6602, Clermont-Ferrand, France
| | - Francis Mpambani
- University of Lyon, École Normale Supérieure de Lyon, Laboratoire de Chimie, University of Lyon 1, CNRS UMR, 5182, Lyon, France
| | - Lise-Prune Berner
- University of Lyon, CREATIS, INSA-Lyon, University of Lyon 1, CNRS UMR, 5220, INSERM U1206, Villeurbanne, France
| | | | - Cécile Olivier
- University of Lyon, CREATIS, INSA-Lyon, University of Lyon 1, CNRS UMR, 5220, INSERM U1206, Villeurbanne, France
| | - Françoise Peyrin
- University of Lyon, CREATIS, INSA-Lyon, University of Lyon 1, CNRS UMR, 5220, INSERM U1206, Villeurbanne, France
| | - Vinu K Apputukan
- University of Lyon, École Normale Supérieure de Lyon, Laboratoire de Chimie, University of Lyon 1, CNRS UMR, 5182, Lyon, France
| | - Cyrille Monnereau
- University of Lyon, École Normale Supérieure de Lyon, Laboratoire de Chimie, University of Lyon 1, CNRS UMR, 5182, Lyon, France
| | - Chantal Andraud
- University of Lyon, École Normale Supérieure de Lyon, Laboratoire de Chimie, University of Lyon 1, CNRS UMR, 5182, Lyon, France
| | - Frederic Chaput
- University of Lyon, École Normale Supérieure de Lyon, Laboratoire de Chimie, University of Lyon 1, CNRS UMR, 5182, Lyon, France
| | - Yves Berthezène
- University of Lyon, CREATIS, INSA-Lyon, University of Lyon 1, CNRS UMR, 5220, INSERM U1206, Villeurbanne, France
| | - Bettina Braun
- Department of Cellular Neurology, Hertie Institute for Clinical Brain Research, University Tübingen, Tübingen, Germany
| | - Mathias Jucker
- Department of Cellular Neurology, Hertie Institute for Clinical Brain Research, University Tübingen, Tübingen, Germany
| | - Andreas Ko Åslund
- Department of Physics, Chemistry, & Biology, Linköping University, Linköping, Sweden
| | - Sofie Nyström
- Department of Physics, Chemistry, & Biology, Linköping University, Linköping, Sweden
| | - Per Hammarström
- Department of Physics, Chemistry, & Biology, Linköping University, Linköping, Sweden
| | - K Peter R Nilsson
- Department of Physics, Chemistry, & Biology, Linköping University, Linköping, Sweden
| | - Mikael Lindgren
- Department of Physics, Norwegian University of Science & Technology, Trondheim, Norway
| | - Marlène Wiart
- University of Lyon, CarMeN laboratory, INSERM U1060, INRA, U1397, University of Lyon 1, INSA-Lyon, Oullins, France.,CNRS, Villeurbanne, France
| | - Fabien Chauveau
- University of Lyon, Lyon Neuroscience Research Center, CNRS UMR, 5292, INSERM U1028, University of Lyon 1, Lyon, France
| | - Stephane Parola
- University of Lyon, École Normale Supérieure de Lyon, Laboratoire de Chimie, University of Lyon 1, CNRS UMR, 5182, Lyon, France
| |
Collapse
|
19
|
Tsitokana ME, Lafon PA, Prézeau L, Pin JP, Rondard P. Targeting the Brain with Single-Domain Antibodies: Greater Potential Than Stated So Far? Int J Mol Sci 2023; 24:ijms24032632. [PMID: 36768953 PMCID: PMC9916958 DOI: 10.3390/ijms24032632] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Revised: 01/12/2023] [Accepted: 01/25/2023] [Indexed: 02/03/2023] Open
Abstract
Treatments for central nervous system diseases with therapeutic antibodies have been increasingly investigated over the last decades, leading to some approved monoclonal antibodies for brain disease therapies. The detection of biomarkers for diagnosis purposes with non-invasive antibody-based imaging approaches has also been explored in brain cancers. However, antibodies generally display a low capability of reaching the brain, as they do not efficiently cross the blood-brain barrier. As an alternative, recent studies have focused on single-domain antibodies (sdAbs) that correspond to the antigen-binding fragment. While some reports indicate that the brain uptake of these small antibodies is still low, the number of studies reporting brain-penetrating sdAbs is increasing. In this review, we provide an overview of methods used to assess or evaluate brain penetration of sdAbs and discuss the pros and cons that could affect the identification of brain-penetrating sdAbs of therapeutic or diagnostic interest.
Collapse
|
20
|
Maali A, Gholizadeh M, Feghhi-Najafabadi S, Noei A, Seyed-Motahari SS, Mansoori S, Sharifzadeh Z. Nanobodies in cell-mediated immunotherapy: On the road to fight cancer. Front Immunol 2023; 14:1012841. [PMID: 36761751 PMCID: PMC9905824 DOI: 10.3389/fimmu.2023.1012841] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2022] [Accepted: 01/09/2023] [Indexed: 01/27/2023] Open
Abstract
The immune system is essential in recognizing and eliminating tumor cells. The unique characteristics of the tumor microenvironment (TME), such as heterogeneity, reduced blood flow, hypoxia, and acidity, can reduce the efficacy of cell-mediated immunity. The primary goal of cancer immunotherapy is to modify the immune cells or the TME to enable the immune system to eliminate malignancies successfully. Nanobodies, known as single-domain antibodies, are light chain-free antibody fragments produced from Camelidae antibodies. The unique properties of nanobodies, including high stability, reduced immunogenicity, enhanced infiltration into the TME of solid tumors and facile genetic engineering have led to their promising application in cell-mediated immunotherapy. They can promote the cancer therapy either directly by bridging between tumor cells and immune cells and by targeting cancer cells using immune cell-bound nanobodies or indirectly by blocking the inhibitory ligands/receptors. The T-cell activation can be engaged through anti-CD3 and anti-4-1BB nanobodies in the bispecific (bispecific T-cell engagers (BiTEs)) and trispecific (trispecific T-cell engager (TriTEs)) manners. Also, nanobodies can be used as natural killer (NK) cell engagers (BiKEs, TriKEs, and TetraKEs) to create an immune synapse between the tumor and NK cells. Nanobodies can redirect immune cells to attack tumor cells through a chimeric antigen receptor (CAR) incorporating a nanobody against the target antigen. Various cancer antigens have been targeted by nanobody-based CAR-T and CAR-NK cells for treating both hematological and solid malignancies. They can also cause the continuation of immune surveillance against tumor cells by stopping inappropriate inhibition of immune checkpoints. Other roles of nanobodies in cell-mediated cancer immunotherapy include reprogramming macrophages to reduce metastasis and angiogenesis, as well as preventing the severe side effects occurring in cell-mediated immunotherapy. Here, we highlight the critical functions of various immune cells, including T cells, NK cells, and macrophages in the TME, and discuss newly developed immunotherapy methods based on the targeted manipulation of immune cells and TME with nanobodies.
Collapse
Affiliation(s)
- Amirhosein Maali
- Department of Immunology, Pasteur Institute of Iran, Tehran, Iran,Department of Medical Biotechnology, Faculty of Allied Medicine, Qazvin University of Medical Sciences, Qazvin, Iran
| | - Monireh Gholizadeh
- Department of Immunology, Pasteur Institute of Iran, Tehran, Iran,Department of Medical Biotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | - Ahmad Noei
- Department of Immunology, Pasteur Institute of Iran, Tehran, Iran
| | - Seyedeh Sheila Seyed-Motahari
- Department of Immunology, Pasteur Institute of Iran, Tehran, Iran,Department of Biology, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | | | - Zahra Sharifzadeh
- Department of Immunology, Pasteur Institute of Iran, Tehran, Iran,*Correspondence: Zahra Sharifzadeh,
| |
Collapse
|
21
|
ImmunoPET Directed to the Brain: A New Tool for Preclinical and Clinical Neuroscience. Biomolecules 2023; 13:biom13010164. [PMID: 36671549 PMCID: PMC9855881 DOI: 10.3390/biom13010164] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Revised: 01/08/2023] [Accepted: 01/10/2023] [Indexed: 01/14/2023] Open
Abstract
Immuno-positron emission tomography (immunoPET) is a non-invasive in vivo imaging method based on tracking and quantifying radiolabeled monoclonal antibodies (mAbs) and other related molecules, such as antibody fragments, nanobodies, or affibodies. However, the success of immunoPET in neuroimaging is limited because intact antibodies cannot penetrate the blood-brain barrier (BBB). In neuro-oncology, immunoPET has been successfully applied to brain tumors because of the compromised BBB. Different strategies, such as changes in antibody properties, use of physiological mechanisms in the BBB, or induced changes to BBB permeability, have been developed to deliver antibodies to the brain. These approaches have recently started to be applied in preclinical central nervous system PET studies. Therefore, immunoPET could be a new approach for developing more specific PET probes directed to different brain targets.
Collapse
|
22
|
Zheng F, Pang Y, Li L, Pang Y, Zhang J, Wang X, Raes G. Applications of nanobodies in brain diseases. Front Immunol 2022; 13:978513. [PMID: 36426363 PMCID: PMC9679430 DOI: 10.3389/fimmu.2022.978513] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2022] [Accepted: 09/30/2022] [Indexed: 03/31/2024] Open
Abstract
Nanobodies are antibody fragments derived from camelids, naturally endowed with properties like low molecular weight, high affinity and low immunogenicity, which contribute to their effective use as research tools, but also as diagnostic and therapeutic agents in a wide range of diseases, including brain diseases. Also, with the success of Caplacizumab, the first approved nanobody drug which was established as a first-in-class medication to treat acquired thrombotic thrombocytopenic purpura, nanobody-based therapy has received increasing attention. In the current review, we first briefly introduce the characterization and manufacturing of nanobodies. Then, we discuss the issue of crossing of the brain-blood-barrier (BBB) by nanobodies, making use of natural methods of BBB penetration, including passive diffusion, active efflux carriers (ATP-binding cassette transporters), carrier-mediated influx via solute carriers and transcytosis (including receptor-mediated transport, and adsorptive mediated transport) as well as various physical and chemical methods or even more complicated methods such as genetic methods via viral vectors to deliver nanobodies to the brain. Next, we give an extensive overview of research, diagnostic and therapeutic applications of nanobodies in brain-related diseases, with emphasis on Alzheimer's disease, Parkinson's disease, and brain tumors. Thanks to the advance of nanobody engineering and modification technologies, nanobodies can be linked to toxins or conjugated with radionuclides, photosensitizers and nanoparticles, according to different requirements. Finally, we provide several perspectives that may facilitate future studies and whereby the versatile nanobodies offer promising perspectives for advancing our knowledge about brain disorders, as well as hopefully yielding diagnostic and therapeutic solutions.
Collapse
Affiliation(s)
- Fang Zheng
- The Key Laboratory of Environment and Genes Related to Disease of Ministry of Education, Health Science Center, Xi’an Jiaotong University, Xi’an, China
| | - Yucheng Pang
- The Key Laboratory of Environment and Genes Related to Disease of Ministry of Education, Health Science Center, Xi’an Jiaotong University, Xi’an, China
| | - Luyao Li
- The Key Laboratory of Environment and Genes Related to Disease of Ministry of Education, Health Science Center, Xi’an Jiaotong University, Xi’an, China
| | - Yuxing Pang
- School of Electronic Science and Engineering, University of Electronic Science and Technology of China, Chengdu, China
| | - Jiaxin Zhang
- The Key Laboratory of Environment and Genes Related to Disease of Ministry of Education, Health Science Center, Xi’an Jiaotong University, Xi’an, China
| | - Xinyi Wang
- The Key Laboratory of Environment and Genes Related to Disease of Ministry of Education, Health Science Center, Xi’an Jiaotong University, Xi’an, China
| | - Geert Raes
- Research Group of Cellular and Molecular Immunology, Vrije Universiteit Brussel, Brussels, Belgium
- Myeloid Cell Immunology Lab, Vlaams Instituut voor Biotechnologie (VIB) Center for Inflammation Research, Brussels, Belgium
| |
Collapse
|
23
|
Zhang X, Liu C, Xie Y, Hu Q, Chen Y, Li J. Identification and characterization of blocking nanobodies against human CD70. Acta Biochim Biophys Sin (Shanghai) 2022; 54:1518-1527. [PMID: 36239354 PMCID: PMC9827822 DOI: 10.3724/abbs.2022141] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
CD70 is overexpressed in a variety of solid and hematological tumors and plays a role in tumor proliferation and evasion of immune surveillance. Targeting and blocking its binding to the receptor CD27 have the potential to treat CD70-dependent tumors. To generate novel CD70 blocking agents, we screen a human CD70-immunized camel VHH phage display library and isolate two blocking nanobodies against human CD70 targeting different epitopes. Upon enrichment by three rounds of biopanning, two strategies are employed to identify CD70 blockers. One named affinity selection is used for detecting clones with CD70 binding by conventional PE-ELISA. However, no clone with a blocking effect is obtained from 188 enriched clones by this method. The alternative strategy named competitive selection is based on the inhibiting capacity of CD70-CD27 binding by enriched VHHs. By this method, two clones, Nb-2B3 and Nb-3B6, with strong blocking capacity are obtained from 20 enriched VHHs, suggesting the efficiency of this strategy. Furthermore, Nb-2B3 and Nb-3B6 specifically bind to CD70-positive SKOV3 and Raji cells at low concentrations. Meanwhile, Nb-2B3 has no competitive effect on the binding of Nb-3B6 to CD70, and vice versa, indicating that they target two different epitopes on CD70. Our data show that nanobodies Nb-2B3 and Nb-3B6 are potential attractive theranostic agents for CD70-expressing cancers.
Collapse
Affiliation(s)
- Xin Zhang
- Xinjiang Key Laboratory of Biological Resources and Genetic EngineeringCollege of Life Science and TechnologyXinjiang UniversityUrumqi830046China
| | - Chang Liu
- Xinjiang Key Laboratory of Biological Resources and Genetic EngineeringCollege of Life Science and TechnologyXinjiang UniversityUrumqi830046China
| | - Yuan Xie
- Xinjiang Key Laboratory of Biological Resources and Genetic EngineeringCollege of Life Science and TechnologyXinjiang UniversityUrumqi830046China
| | - Qianqian Hu
- Xinjiang Key Laboratory of Biological Resources and Genetic EngineeringCollege of Life Science and TechnologyXinjiang UniversityUrumqi830046China
| | | | - Jiangwei Li
- Xinjiang Key Laboratory of Biological Resources and Genetic EngineeringCollege of Life Science and TechnologyXinjiang UniversityUrumqi830046China,Xinjiang Unique Mab BioTech Co.LtdUrumqi830002China,Correspondence: Tel: +86-18099167036;
| |
Collapse
|
24
|
Pinto-Espinoza C, Guillou C, Rissiek B, Wilmes M, Javidi E, Schwarz N, Junge M, Haag F, Liaukouskaya N, Wanner N, Nicke A, Stortelers C, Tan YV, Adriouch S, Magnus T, Koch-Nolte F. Effective targeting of microglial P2X7 following intracerebroventricular delivery of nanobodies and nanobody-encoding AAVs. Front Pharmacol 2022; 13:1029236. [PMID: 36299894 PMCID: PMC9589454 DOI: 10.3389/fphar.2022.1029236] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2022] [Accepted: 09/23/2022] [Indexed: 11/19/2022] Open
Abstract
The P2X7 ion channel is a key sensor for extracellular ATP and a key trigger of sterile inflammation. Intravenous injection of nanobodies that block P2X7 has shown to be beneficial in mouse models of systemic inflammation. P2X7 has also emerged as an attractive therapeutic target for inflammatory brain diseases. However, little is known about the ability of nanobodies to cross the BBB. Here we evaluated the ability of P2X7-specific nanobodies to reach and to block P2X7 on microglia following intravenous or intracerebral administration. For this study, we reformatted and sequence-optimized P2X7 nanobodies for higher stability and elevated isoelectric point. Following injection of nanobodies or nanobody-encoding adeno-associated viral vectors (AAV), we monitored the occupancy and blockade of microglial P2X7 in vivo using ex vivo flow cytometry. Our results show that P2X7 on microglia was within minutes completely occupied and blocked by intracerebroventricularly injected nanobodies, even at low doses. In contrast, very high doses were required to achieve similar effects when injected intravenously. The endogenous production of P2X7-antagonistic nanobodies following intracerebral or intramuscular injection of nanobody-encoding AAVs resulted in a long-term occupancy and blockade of P2X7 on microglia. Our results provide new insights into the conditions for the delivery of nanobodies to microglial P2X7 and point to AAV-mediated delivery of P2X7 nanobodies as a promising strategy for the treatment of sterile brain inflammation.
Collapse
Affiliation(s)
- Carolina Pinto-Espinoza
- Institute of Immunology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Department of Neurology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Charlotte Guillou
- Normandie Univ, UNIROUEN, INSERM U1234, Pathophysiology, Autoimmunity and Immunotherapy (PanTHER), Rouen, France
| | - Björn Rissiek
- Department of Neurology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Maximilian Wilmes
- Department of Neurology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Ehsan Javidi
- Department of Neurology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Nicole Schwarz
- Institute of Immunology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- MSH- Medical School Hamburg- Dep. Anatomy, Hamburg, Germany
| | - Marten Junge
- Institute of Immunology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Friedrich Haag
- Institute of Immunology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | | | - Nicola Wanner
- Department of Nephrology, University Medical Centre Hamburg-Eppendorf, Hamburg, Germany
| | - Annette Nicke
- Walther Straub Institute of Pharmacology and Toxicology, Faculty of Medicine, LMU Munich, Munich, Germany
| | | | - Yossan-Var Tan
- Normandie Univ, UNIROUEN, INSERM U1234, Pathophysiology, Autoimmunity and Immunotherapy (PanTHER), Rouen, France
| | - Sahil Adriouch
- Normandie Univ, UNIROUEN, INSERM U1234, Pathophysiology, Autoimmunity and Immunotherapy (PanTHER), Rouen, France
| | - Tim Magnus
- Department of Neurology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Friedrich Koch-Nolte
- Institute of Immunology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- *Correspondence: Friedrich Koch-Nolte,
| |
Collapse
|
25
|
Mitusova K, Peltek OO, Karpov TE, Muslimov AR, Zyuzin MV, Timin AS. Overcoming the blood-brain barrier for the therapy of malignant brain tumor: current status and prospects of drug delivery approaches. J Nanobiotechnology 2022; 20:412. [PMID: 36109754 PMCID: PMC9479308 DOI: 10.1186/s12951-022-01610-7] [Citation(s) in RCA: 58] [Impact Index Per Article: 29.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2022] [Accepted: 08/18/2022] [Indexed: 01/06/2023] Open
Abstract
Besides the broad development of nanotechnological approaches for cancer diagnosis and therapy, currently, there is no significant progress in the treatment of different types of brain tumors. Therapeutic molecules crossing the blood-brain barrier (BBB) and reaching an appropriate targeting ability remain the key challenges. Many invasive and non-invasive methods, and various types of nanocarriers and their hybrids have been widely explored for brain tumor treatment. However, unfortunately, no crucial clinical translations were observed to date. In particular, chemotherapy and surgery remain the main methods for the therapy of brain tumors. Exploring the mechanisms of the BBB penetration in detail and investigating advanced drug delivery platforms are the key factors that could bring us closer to understanding the development of effective therapy against brain tumors. In this review, we discuss the most relevant aspects of the BBB penetration mechanisms, observing both invasive and non-invasive methods of drug delivery. We also review the recent progress in the development of functional drug delivery platforms, from viruses to cell-based vehicles, for brain tumor therapy. The destructive potential of chemotherapeutic drugs delivered to the brain tumor is also considered. This review then summarizes the existing challenges and future prospects in the use of drug delivery platforms for the treatment of brain tumors.
Collapse
Affiliation(s)
- Ksenia Mitusova
- Peter The Great St. Petersburg Polytechnic University, Polytechnicheskaya 29, St. Petersburg, 195251, Russian Federation
| | - Oleksii O Peltek
- School of Physics and Engineering, ITMO University, Lomonosova 9, St. Petersburg, 191002, Russian Federation
| | - Timofey E Karpov
- Peter The Great St. Petersburg Polytechnic University, Polytechnicheskaya 29, St. Petersburg, 195251, Russian Federation
| | - Albert R Muslimov
- Peter The Great St. Petersburg Polytechnic University, Polytechnicheskaya 29, St. Petersburg, 195251, Russian Federation
- Sirius University of Science and Technology, Olympic Ave 1, Sirius, 354340, Russian Federation
| | - Mikhail V Zyuzin
- School of Physics and Engineering, ITMO University, Lomonosova 9, St. Petersburg, 191002, Russian Federation
| | - Alexander S Timin
- Peter The Great St. Petersburg Polytechnic University, Polytechnicheskaya 29, St. Petersburg, 195251, Russian Federation.
- School of Physics and Engineering, ITMO University, Lomonosova 9, St. Petersburg, 191002, Russian Federation.
| |
Collapse
|
26
|
Ishizuka Y, Mergiya TF, Baldinotti R, Xu J, Hallin EI, Markússon S, Kursula P, Bramham CR. Development and Validation of Arc Nanobodies: New Tools for Probing Arc Dynamics and Function. Neurochem Res 2022; 47:2656-2666. [PMID: 35307777 PMCID: PMC9463278 DOI: 10.1007/s11064-022-03573-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2022] [Revised: 03/04/2022] [Accepted: 03/07/2022] [Indexed: 11/06/2022]
Abstract
Activity-regulated cytoskeleton-associated (Arc) protein plays key roles in long-term synaptic plasticity, memory, and cognitive flexibility. However, an integral understanding of Arc mechanisms is lacking. Arc is proposed to function as an interaction hub in neuronal dendrites and the nucleus, yet Arc can also form retrovirus-like capsids with proposed roles in intercellular communication. Here, we sought to develop anti-Arc nanobodies (ArcNbs) as new tools for probing Arc dynamics and function. Six ArcNbs representing different clonal lines were selected from immunized alpaca. Immunoblotting with recombinant ArcNbs fused to a small ALFA-epitope tag demonstrated binding to recombinant Arc as well as endogenous Arc from rat cortical tissue. ALFA-tagged ArcNb also provided efficient immunoprecipitation of stimulus-induced Arc after carbachol-treatment of SH-SY5Y neuroblastoma cells and induction of long-term potentiation in the rat dentate gyrus in vivo. Epitope mapping showed that all Nbs recognize the Arc C-terminal region containing the retroviral Gag capsid homology domain, comprised of tandem N- and C-lobes. ArcNbs E5 and H11 selectively bound the N-lobe, which harbors a peptide ligand binding pocket specific to mammals. Four additional ArcNbs bound the region containing the C-lobe and C-terminal tail. For use as genetically encoded fluorescent intrabodies, we show that ArcNbs fused to mScarlet-I are uniformly expressed, without aggregation, in the cytoplasm and nucleus of HEK293FT cells. Finally, mScarlet-I-ArcNb H11 expressed as intrabody selectively bound the N-lobe and enabled co-immunoprecipitation of full-length intracellular Arc. ArcNbs are versatile tools for live-cell labeling and purification of Arc, and interrogation of Arc capsid domain specific functions.
Collapse
Affiliation(s)
- Yuta Ishizuka
- Department of Biomedicine, University of Bergen, Jonas Lies 91, 5009, Bergen, Norway.
- Department of Pathophysiology and Metabolism, Kawasaki Medical School, 577 Matsushima, Kurashiki, Okayama, 701-0192, Japan.
| | - Tadiwos F Mergiya
- Department of Biomedicine, University of Bergen, Jonas Lies 91, 5009, Bergen, Norway
- Mohn Center for Research on the Brain, University of Bergen, Bergen, Norway
| | - Rodolfo Baldinotti
- Department of Biomedicine, University of Bergen, Jonas Lies 91, 5009, Bergen, Norway
- Mohn Center for Research on the Brain, University of Bergen, Bergen, Norway
| | - Ju Xu
- Department of Biomedicine, University of Bergen, Jonas Lies 91, 5009, Bergen, Norway
| | - Erik I Hallin
- Department of Biomedicine, University of Bergen, Jonas Lies 91, 5009, Bergen, Norway
| | - Sigurbjörn Markússon
- Department of Biomedicine, University of Bergen, Jonas Lies 91, 5009, Bergen, Norway
| | - Petri Kursula
- Department of Biomedicine, University of Bergen, Jonas Lies 91, 5009, Bergen, Norway
- Faculty of Biochemistry and Molecular Medicine, University of Oulu, Oulu, Finland
- Biocenter Oulu, University of Oulu, Oulu, Finland
| | - Clive R Bramham
- Department of Biomedicine, University of Bergen, Jonas Lies 91, 5009, Bergen, Norway.
- Mohn Center for Research on the Brain, University of Bergen, Bergen, Norway.
| |
Collapse
|
27
|
Hmila I, Vaikath NN, Majbour NK, Erskine D, Sudhakaran IP, Gupta V, Ghanem SS, Islam Z, Emara MM, Abdesselem HB, Kolatkar PR, Achappa DK, Vinardell T, El‐Agnaf OMA. Novel engineered nanobodies specific for N‐terminal region of alpha‐synuclein recognize Lewy‐body pathology and inhibit
in‐vitro
seeded aggregation and toxicity. FEBS J 2022; 289:4657-4673. [PMID: 35090199 PMCID: PMC9545584 DOI: 10.1111/febs.16376] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2021] [Revised: 09/28/2021] [Accepted: 01/26/2022] [Indexed: 12/22/2022]
Abstract
Nanobodies (Nbs), the single‐domain antigen‐binding fragments of dromedary heavy‐chain antibodies (HCAb), are excellent candidates as therapeutic and diagnostic tools in synucleinopathies because of their small size, solubility and stability. Here, we constructed an immune nanobody library specific to the monomeric form of alpha‐synuclein (α‐syn). Phage display screening of the library allowed the identification of a nanobody, Nbα‐syn01, specific for α‐syn. Unlike previously developed nanobodies, Nbα‐syn01 recognized the N‐terminal region which is critical for in vitro and in vivo aggregation and contains many point mutations involved in early PD cases. The affinity of the monovalent Nbα‐syn01 and the engineered bivalent format BivNbα‐syn01 measured by isothermal titration calorimetry revealed unexpected results where Nbα‐syn01 and its bivalent format recognized preferentially α‐syn fibrils compared to the monomeric form. Nbα‐syn01 and BivNbα‐syn01 were also able to inhibit α‐syn‐seeded aggregation in vitro and reduced α‐syn‐seeded aggregation and toxicity in cells showing their potential to reduce α‐syn pathology. Moreover, both nanobody formats were able to recognize Lewy‐body pathology in human post‐mortem brain tissue from PD and DLB cases. Additionally, we present evidence through structural docking that Nbα‐syn01 binds the N‐terminal region of the α‐syn aggregated form. Overall, these results highlight the potential of Nbα‐syn01 and BivNbα‐syn01 in developing into a diagnostic or a therapeutic tool for PD and related disorders.
Collapse
Affiliation(s)
- Issam Hmila
- Neurological Disorder Research Center Qatar Biomedical Research Institute (QBRI) Hamad Bin Khalifa University (HBKU) Qatar Foundation Doha Qatar
| | - Nishant N. Vaikath
- Neurological Disorder Research Center Qatar Biomedical Research Institute (QBRI) Hamad Bin Khalifa University (HBKU) Qatar Foundation Doha Qatar
| | - Nour K. Majbour
- Neurological Disorder Research Center Qatar Biomedical Research Institute (QBRI) Hamad Bin Khalifa University (HBKU) Qatar Foundation Doha Qatar
| | - Daniel Erskine
- Translational and Clinical Research Institute Newcastle University UK
| | - Indulekha P. Sudhakaran
- Neurological Disorder Research Center Qatar Biomedical Research Institute (QBRI) Hamad Bin Khalifa University (HBKU) Qatar Foundation Doha Qatar
| | - Vijay Gupta
- Neurological Disorder Research Center Qatar Biomedical Research Institute (QBRI) Hamad Bin Khalifa University (HBKU) Qatar Foundation Doha Qatar
| | - Simona S. Ghanem
- Neurological Disorder Research Center Qatar Biomedical Research Institute (QBRI) Hamad Bin Khalifa University (HBKU) Qatar Foundation Doha Qatar
| | - Zeyaul Islam
- Diabetes Center Qatar Biomedical Research Institute (QBRI) Hamad Bin Khalifa University (HBKU) Qatar Foundation Doha Qatar
| | - Mohamed M. Emara
- Basic Medical Sciences Department College of Medicine QU Health Qatar University Doha Qatar
- Biomedical and Pharmaceutical Research Unit QU Health Qatar University Doha Qatar
| | - Houari B. Abdesselem
- Neurological Disorder Research Center Qatar Biomedical Research Institute (QBRI) Hamad Bin Khalifa University (HBKU) Qatar Foundation Doha Qatar
| | - Prasanna R. Kolatkar
- Diabetes Center Qatar Biomedical Research Institute (QBRI) Hamad Bin Khalifa University (HBKU) Qatar Foundation Doha Qatar
| | | | - Tatiana Vinardell
- Equine Veterinary Medical Center Qatar Foundation Doha Qatar
- College of Health & Life Science Hamad Bin Khalifa University Qatar Foundation Doha Qatar
| | - Omar M. A. El‐Agnaf
- Neurological Disorder Research Center Qatar Biomedical Research Institute (QBRI) Hamad Bin Khalifa University (HBKU) Qatar Foundation Doha Qatar
| |
Collapse
|
28
|
Shamshiripour P, Hajiahmadi F, Lotfi S, Esmaeili NR, Zare A, Akbarpour M, Ahmadvand D. Next-Generation Anti-Angiogenic Therapies as a Future Prospect for Glioma Immunotherapy; From Bench to Bedside. Front Immunol 2022; 13:859633. [PMID: 35757736 PMCID: PMC9231436 DOI: 10.3389/fimmu.2022.859633] [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: 01/21/2022] [Accepted: 03/24/2022] [Indexed: 11/13/2022] Open
Abstract
Glioblastoma (grade IV glioma) is the most aggressive histopathological subtype of glial tumors with inordinate microvascular proliferation as one of its key pathological features. Extensive angiogenesis in the tumor microenvironment supplies oxygen and nutrients to tumoral cells; retains their survival under hypoxic conditions; and induces an immunosuppressive microenvironment. Anti-angiogenesis therapy for high-grade gliomas has long been studied as an adjuvant immunotherapy strategy to overcome tumor growth. In the current review, we discussed the underlying molecular mechanisms contributing to glioblastoma aberrant angiogenesis. Further, we discussed clinical applications of monoclonal antibodies, tyrosine kinase inhibitors, and aptamers as three major subgroups of anti-angiogenic immunotherapeutics and their limitations. Moreover, we reviewed clinical and preclinical applications of small interfering RNAs (siRNAs) as the next-generation anti-angiogenic therapeutics and summarized their potential advantages and limitations. siRNAs may serve as next-generation anti-angiogenic therapeutics for glioma. Additionally, application of nanoparticles as a delivery vehicle could increase their selectivity and lower their off-target effects.
Collapse
Affiliation(s)
- Parisa Shamshiripour
- Faculty of Medicine, Iran University of Medical Sciences, Tehran, Iran.,Department of Molecular Imaging, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Fahimeh Hajiahmadi
- Department of Molecular Imaging, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Shahla Lotfi
- Department of Molecular Imaging, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Niloofar Robab Esmaeili
- Department of Biomedical Engineering, Amirkabir University of Technology (Tehran Polytechnic), Tehran, Iran
| | - Amir Zare
- Faculty of Medicine, Iran University of Medical Sciences, Tehran, Iran.,Department of Surgery, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Mahzad Akbarpour
- Advanced Cellular Therapeutics Facility, David and Etta Jonas Center for Cellular Therapy, Hematopoietic Cellular Therapy Program, The University of Chicago Medical Center, Chicago, IL, United States.,Immunology Board for Transplantation and Cell-Based Therapeutics (Immuno-TACT), Universal Science and Education Research Network (USERN), Tehran, Iran
| | - Davoud Ahmadvand
- Department of Molecular Imaging, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran, Iran.,Neuroscience Research Center, Iran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
29
|
Antman-Passig M, Wong E, Frost GR, Cupo C, Shah J, Agustinus A, Chen Z, Mancinelli C, Kamel M, Li T, Jonas LA, Li YM, Heller DA. Optical Nanosensor for Intracellular and Intracranial Detection of Amyloid-Beta. ACS NANO 2022; 16:7269-7283. [PMID: 35420796 PMCID: PMC9710299 DOI: 10.1021/acsnano.2c00054] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Amyloid-beta (Aβ) deposition occurs in the early stages of Alzheimer's disease (AD), but the early detection of Aβ is a persistent challenge. Herein, we engineered a near-infrared optical nanosensor capable of detecting Aβ intracellularly in live cells and intracranially in vivo. The sensor is composed of single-walled carbon nanotubes functionalized with Aβ wherein Aβ-Aβ interactions drive the response. We found that the Aβ nanosensors selectively responded to Aβ via solvatochromic modulation of the near-infrared emission of the nanotube. The sensor tracked Aβ accumulation in live cells and, upon intracranial administration in a genetic model of AD, signaled distinct responses in aged mice. This technology enables the interrogation of molecular mechanisms underlying Aβ neurotoxicity in the development of AD in living systems.
Collapse
Affiliation(s)
- Merav Antman-Passig
- Molecular Pharmacology Program, Memorial Sloan Kettering Cancer Center, New York, New York 10065, United States
| | - Eitan Wong
- Chemical Biology Program, Memorial Sloan Kettering Cancer Center, New York, New York 10065, United States
| | - Georgia R Frost
- Chemical Biology Program, Memorial Sloan Kettering Cancer Center, New York, New York 10065, United States
| | - Christian Cupo
- Department of Mechanical Engineering, Columbia University, New York, New York 10027, United States
| | - Janki Shah
- Molecular Pharmacology Program, Memorial Sloan Kettering Cancer Center, New York, New York 10065, United States
| | - Albert Agustinus
- Program of Pharmacology, Weill Graduate School of Medical Sciences of Cornell University, New York, New York 10065, United States
| | - Ziyu Chen
- Program of Physiology, Biophysics, & Systems Biology, Weill Graduate School of Medical Sciences of Cornell University, New York, New York 10065, United States
| | - Chiara Mancinelli
- Program of Pharmacology, Weill Graduate School of Medical Sciences of Cornell University, New York, New York 10065, United States
| | - Maikel Kamel
- Sophie Davis School of Biomedical Education, CUNY School of Medicine, New York, New York 10031, United States
| | - Thomas Li
- Chemical Biology Program, Memorial Sloan Kettering Cancer Center, New York, New York 10065, United States
- Program of Neurosciences, Weill Graduate School of Medical Sciences of Cornell University, New York, New York 10065, United States
| | - Lauren A Jonas
- Chemical Biology Program, Memorial Sloan Kettering Cancer Center, New York, New York 10065, United States
- Program of Pharmacology, Weill Graduate School of Medical Sciences of Cornell University, New York, New York 10065, United States
| | - Yue-Ming Li
- Chemical Biology Program, Memorial Sloan Kettering Cancer Center, New York, New York 10065, United States
- Program of Pharmacology, Weill Graduate School of Medical Sciences of Cornell University, New York, New York 10065, United States
- Program of Neurosciences, Weill Graduate School of Medical Sciences of Cornell University, New York, New York 10065, United States
| | - Daniel A Heller
- Molecular Pharmacology Program, Memorial Sloan Kettering Cancer Center, New York, New York 10065, United States
- Program of Pharmacology, Weill Graduate School of Medical Sciences of Cornell University, New York, New York 10065, United States
- Program of Physiology, Biophysics, & Systems Biology, Weill Graduate School of Medical Sciences of Cornell University, New York, New York 10065, United States
| |
Collapse
|
30
|
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: 11] [Impact Index Per Article: 5.5] [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
|
31
|
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: 0] [Impact Index Per Article: 0] [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
|
32
|
Menon S, Armstrong S, Hamzeh A, Visanji NP, Sardi SP, Tandon A. Alpha-Synuclein Targeting Therapeutics for Parkinson's Disease and Related Synucleinopathies. Front Neurol 2022; 13:852003. [PMID: 35614915 PMCID: PMC9124903 DOI: 10.3389/fneur.2022.852003] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2022] [Accepted: 04/01/2022] [Indexed: 12/14/2022] Open
Abstract
α-Synuclein (asyn) is a key pathogenetic factor in a group of neurodegenerative diseases generically known as synucleinopathies, including Parkinson's disease (PD), dementia with Lewy bodies (DLB) and multiple system atrophy (MSA). Although the initial triggers of pathology and progression are unclear, multiple lines of evidence support therapeutic targeting of asyn in order to limit its prion-like misfolding. Here, we review recent pre-clinical and clinical work that offers promising treatment strategies to sequester, degrade, or silence asyn expression as a means to reduce the levels of seed or substrate. These diverse approaches include removal of aggregated asyn with passive or active immunization or by expression of vectorized antibodies, modulating kinetics of misfolding with small molecule anti-aggregants, lowering asyn gene expression by antisense oligonucleotides or inhibitory RNA, and pharmacological activation of asyn degradation pathways. We also discuss recent technological advances in combining low intensity focused ultrasound with intravenous microbubbles to transiently increase blood-brain barrier permeability for improved brain delivery and target engagement of these large molecule anti-asyn biologics.
Collapse
Affiliation(s)
- Sindhu Menon
- Tanz Centre for Research in Neurodegenerative Diseases, Toronto, ON, Canada
| | - Sabrina Armstrong
- Tanz Centre for Research in Neurodegenerative Diseases, Toronto, ON, Canada
| | - Amir Hamzeh
- Tanz Centre for Research in Neurodegenerative Diseases, Toronto, ON, Canada
| | - Naomi P. Visanji
- Tanz Centre for Research in Neurodegenerative Diseases, Toronto, ON, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada
- Krembil Research Institute, Toronto, ON, Canada
| | | | - Anurag Tandon
- Tanz Centre for Research in Neurodegenerative Diseases, Toronto, ON, Canada
- Department of Medicine, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
33
|
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: 31] [Impact Index Per Article: 15.5] [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
|
34
|
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: 13] [Impact Index Per Article: 6.5] [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
|
35
|
Eshghi D, Kafi M, Sharifiyazdi H, Azari M, Ahmadi N, Ghasrodashti AR, Sadeghi M. Intrauterine infusion of blood serum of dromedary camel improves the uterine health and fertility in high producing dairy cows with endometritis. Anim Reprod Sci 2022; 240:106973. [DOI: 10.1016/j.anireprosci.2022.106973] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2022] [Revised: 03/20/2022] [Accepted: 03/25/2022] [Indexed: 12/30/2022]
|
36
|
Diagnosis of Glioblastoma by Immuno-Positron Emission Tomography. Cancers (Basel) 2021; 14:cancers14010074. [PMID: 35008238 PMCID: PMC8750680 DOI: 10.3390/cancers14010074] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2021] [Revised: 12/16/2021] [Accepted: 12/21/2021] [Indexed: 02/06/2023] Open
Abstract
Simple Summary Neuroimaging has transformed the way brain tumors are diagnosed and treated. Although different non-invasive modalities provide very helpful information, in some situations, they present a limited value. By merging the specificity of antibodies with the resolution, sensitivity, and quantitative capabilities of positron emission tomography (PET), “Immuno-PET” allows us to conduct the non-invasive diagnosis and monitoring of patients over time using antibody-based probes as an in vivo, integrated, quantifiable, 3D, full-body “immunohistochemistry”, like a “virtual biopsy”. This review provides and focuses on immuno-PET applications and future perspectives of this promising imaging approach for glioblastoma. Abstract Neuroimaging has transformed neuro-oncology and the way that glioblastoma is diagnosed and treated. Magnetic Resonance Imaging (MRI) is the most widely used non-invasive technique in the primary diagnosis of glioblastoma. Although MRI provides very powerful anatomical information, it has proven to be of limited value for diagnosing glioblastomas in some situations. The final diagnosis requires a brain biopsy that may not depict the high intratumoral heterogeneity present in this tumor type. The revolution in “cancer-omics” is transforming the molecular classification of gliomas. However, many of the clinically relevant alterations revealed by these studies have not yet been integrated into the clinical management of patients, in part due to the lack of non-invasive biomarker-based imaging tools. An innovative option for biomarker identification in vivo is termed “immunotargeted imaging”. By merging the high target specificity of antibodies with the high spatial resolution, sensitivity, and quantitative capabilities of positron emission tomography (PET), “Immuno-PET” allows us to conduct the non-invasive diagnosis and monitoring of patients over time using antibody-based probes as an in vivo, integrated, quantifiable, 3D, full-body “immunohistochemistry” in patients. This review provides the state of the art of immuno-PET applications and future perspectives on this imaging approach for glioblastoma.
Collapse
|
37
|
Soleimanizadeh A, Dinter H, Schindowski K. Central Nervous System Delivery of Antibodies and Their Single-Domain Antibodies and Variable Fragment Derivatives with Focus on Intranasal Nose to Brain Administration. Antibodies (Basel) 2021; 10:antib10040047. [PMID: 34939999 PMCID: PMC8699001 DOI: 10.3390/antib10040047] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2021] [Revised: 11/10/2021] [Accepted: 11/25/2021] [Indexed: 02/06/2023] Open
Abstract
IgG antibodies are some of the most important biopharmaceutical molecules with a high market volume. In spite of the fact that clinical therapies with antibodies are broadly utilized in oncology, immunology and hematology, their delivery strategies and biodistribution need improvement, their limitations being due to their size and poor ability to penetrate into tissues. In view of their small size, there is a rising interest in derivatives, such as single-domain antibodies and single-chain variable fragments, for clinical diagnostic but also therapeutic applications. Smaller antibody formats combine several benefits for clinical applications and can be manufactured at reduced production costs compared with full-length IgGs. Moreover, such formats have a relevant potential for targeted drug delivery that directs drug cargo to a specific tissue or across the blood–brain barrier. In this review, we give an overview of the challenges for antibody drug delivery in general and focus on intranasal delivery to the central nervous system with antibody formats of different sizes.
Collapse
Affiliation(s)
- Arghavan Soleimanizadeh
- Institute of Applied Biotechnology, Biberach University of Applied Science, 88400 Biberach, Germany; (A.S.); (H.D.)
- Faculty of Medicine, University of Ulm, 89081 Ulm, Germany
| | - Heiko Dinter
- Institute of Applied Biotechnology, Biberach University of Applied Science, 88400 Biberach, Germany; (A.S.); (H.D.)
- Department of Pharmacy and Biochemistry, University of Tübingen, 72076 Tübingen, Germany
| | - Katharina Schindowski
- Institute of Applied Biotechnology, Biberach University of Applied Science, 88400 Biberach, Germany; (A.S.); (H.D.)
- Correspondence:
| |
Collapse
|
38
|
A Targeted Catalytic Nanobody (T-CAN) with Asparaginolytic Activity. Cancers (Basel) 2021; 13:cancers13225637. [PMID: 34830793 PMCID: PMC8616244 DOI: 10.3390/cancers13225637] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Revised: 11/08/2021] [Accepted: 11/09/2021] [Indexed: 11/25/2022] Open
Abstract
Simple Summary The therapy of Acute Lymphoblastic Leukemia (ALL) is based on Escherichia coli (E. coli) L-asparaginase, which is a very effective drug in most cases. However, its side effects sometimes prevent its usage or impose its interruption. The main issues derive from its bacterial origin, which elicits a strong immune response in the patient, and from its generalized action on all body compartments. In this work, we describe how we generated a fully active and miniaturized form of L-asparaginase starting from a camel single domain antibody, a class of antibodies known to have a very limited immunogenicity in humans. We also targeted it onto tumor cells by attaching it to an antibody fragment directed onto the CD19 B-cell surface receptor, expressed on ALL cells. We named this new molecule “Targeted Catalytic Nanobody” (T-CAN). The T-CAN is active and successfully binds to CD19 expressing cells in vitro. Thanks to its reduced immunogenic potential, it represents a new tool which deserves further development. Abstract E. coli L-asparaginase is an amidohydrolase (EC 3.5.1.1) which has been successfully used for the treatment of Acute Lymphoblastic Leukemia for over 50 years. Despite its efficacy, its side effects, and especially its intrinsic immunogenicity, hamper its usage in a significant subset of cases, thus limiting therapeutic options. Innovative solutions to improve on these drawbacks have been attempted, but none of them have been truly successful so far. In this work, we fully replaced the enzyme scaffold, generating an active, miniaturized form of L-asparaginase by protein engineering of a camel single domain antibody, a class of antibodies known to have a limited immunogenicity in humans. We then targeted it onto tumor cells by an antibody scFv fragment directed onto the CD19 B-cell surface receptor expressed on ALL cells. We named this new type of nanobody-based antibody-drug conjugate “Targeted Catalytic Nanobody” (T-CAN). The new molecule retains the catalytic activity and the binding capability of the original modules and successfully targets CD19 expressing cells in vitro. Thanks to its theoretically reduced immunogenic potential compared to the original molecule, the T-CAN can represent a novel approach to tackle current limitations in L-asparaginase usage.
Collapse
|
39
|
Gransagne M, Aymé G, Brier S, Chauveau-Le Friec G, Meriaux V, Nowakowski M, Dejardin F, Levallois S, Dias de Melo G, Donati F, Prot M, Brûlé S, Raynal B, Bellalou J, Goncalves P, Montagutelli X, Di Santo JP, Lazarini F, England P, Petres S, Escriou N, Lafaye P. Development of a highly specific and sensitive VHH-based sandwich immunoassay for the detection of the SARS-CoV-2 nucleoprotein. J Biol Chem 2021; 298:101290. [PMID: 34678315 PMCID: PMC8526496 DOI: 10.1016/j.jbc.2021.101290] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2021] [Revised: 09/15/2021] [Accepted: 10/06/2021] [Indexed: 12/20/2022] Open
Abstract
The current COVID-19 pandemic illustrates the importance of obtaining reliable methods for the rapid detection of SARS-CoV-2. A highly specific and sensitive diagnostic test able to differentiate the SARS-CoV-2 virus from common human coronaviruses is therefore needed. Coronavirus nucleoprotein (N) localizes to the cytoplasm and the nucleolus and is required for viral RNA synthesis. N is the most abundant coronavirus protein, so it is of utmost importance to develop specific antibodies for its detection. In this study, we developed a sandwich immunoassay to recognize the SARS-CoV-2 N protein. We immunized one alpaca with recombinant SARS-CoV-2 N and constructed a large single variable domain on heavy chain (VHH) antibody library. After phage display selection, seven VHHs recognizing the full N protein were identified by ELISA. These VHHs did not recognize the nucleoproteins of the four common human coronaviruses. Hydrogen Deuterium eXchange–Mass Spectrometry (HDX-MS) analysis also showed that these VHHs mainly targeted conformational epitopes in either the C-terminal or the N-terminal domains. All VHHs were able to recognize SARS-CoV-2 in infected cells or on infected hamster tissues. Moreover, the VHHs could detect the SARS variants B.1.17/alpha, B.1.351/beta, and P1/gamma. We propose that this sandwich immunoassay could be applied to specifically detect the SARS-CoV-2 N in human nasal swabs.
Collapse
Affiliation(s)
| | - Gabriel Aymé
- Plateforme d'Ingénierie des Anticorps, C2RT, Institut Pasteur, CNRS UMR 3528, Paris, France
| | - Sébastien Brier
- Plateforme technologique de RMN biologique, C2RT, Institut Pasteur, CNRS UMR 3528, Paris, France
| | | | - Véronique Meriaux
- Plateforme d'Ingénierie des Anticorps, C2RT, Institut Pasteur, CNRS UMR 3528, Paris, France
| | - Mireille Nowakowski
- Plateforme Technologique Production et Purification de Protéines Recombinantes, C2RT, Institut Pasteur, CNRS UMR 3528, Paris, France
| | - François Dejardin
- Plateforme Technologique Production et Purification de Protéines Recombinantes, C2RT, Institut Pasteur, CNRS UMR 3528, Paris, France
| | - Sylvain Levallois
- Biology of Infection Unit, Institut Pasteur, Inserm U1117, Paris, France
| | | | - Flora Donati
- Molecular Genetics of RNA viruses, UMR 3569 CNRS, University of Paris, Institut Pasteur, Paris, France; National Reference Center for Respiratory Viruses, Institut Pasteur, Paris, France
| | - Matthieu Prot
- Evolutionary Genomics of RNA viruses, Institut Pasteur, Paris, France
| | - Sébastien Brûlé
- Plateforme de Biophysique Moléculaire, C2RT, Institut Pasteur, CNRS UMR 3528, Paris, France
| | - Bertrand Raynal
- Plateforme de Biophysique Moléculaire, C2RT, Institut Pasteur, CNRS UMR 3528, Paris, France
| | - Jacques Bellalou
- Plateforme Technologique Production et Purification de Protéines Recombinantes, C2RT, Institut Pasteur, CNRS UMR 3528, Paris, France
| | - Pedro Goncalves
- Unité d'Immunité Innée, Institut Pasteur, Paris, France; INSERM U1223, Paris, France
| | | | - James P Di Santo
- Unité d'Immunité Innée, Institut Pasteur, Paris, France; INSERM U1223, Paris, France
| | - Françoise Lazarini
- Perception and Memory Unit, Institut Pasteur, CNRS UMR 3571, Paris, France
| | - Patrick England
- Plateforme de Biophysique Moléculaire, C2RT, Institut Pasteur, CNRS UMR 3528, Paris, France
| | - Stéphane Petres
- Plateforme Technologique Production et Purification de Protéines Recombinantes, C2RT, Institut Pasteur, CNRS UMR 3528, Paris, France
| | - Nicolas Escriou
- Département de Santé Globale, Institut Pasteur, Paris, France
| | - Pierre Lafaye
- Plateforme d'Ingénierie des Anticorps, C2RT, Institut Pasteur, CNRS UMR 3528, Paris, France.
| |
Collapse
|
40
|
Pharmacokinetics of Single Domain Antibodies and Conjugated Nanoparticles Using a Hybrid near Infrared Method. Int J Mol Sci 2021; 22:ijms22168695. [PMID: 34445399 PMCID: PMC8395466 DOI: 10.3390/ijms22168695] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Revised: 08/05/2021] [Accepted: 08/09/2021] [Indexed: 11/17/2022] Open
Abstract
Iron oxide nanoparticles and single domain antibodies from camelids (VHHs) have been increasingly recognized for their potential uses for medical diagnosis and treatment. However, there have been relatively few detailed characterizations of their pharmacokinetics (PK). The aim of this study was to develop imaging methods and pharmacokinetic models to aid the future development of a novel family of brain MRI molecular contrast agents. An efficient near-infrared (NIR) imaging method was established to monitor VHH and VHH conjugated nanoparticle kinetics in mice using a hybrid approach: kinetics in blood were assessed by direct sampling, and kinetics in kidney, liver, and brain were assessed by serial in vivo NIR imaging. These studies were performed under "basal" circumstances in which the VHH constructs and VHH-conjugated nanoparticles do not substantially interact with targets nor cross the blood brain barrier. Using this approach, we constructed a five-compartment PK model that fits the data well for single VHHs, engineered VHH trimers, and iron oxide nanoparticles conjugated to VHH trimers. The establishment of the feasibility of these methods lays a foundation for future PK studies of candidate brain MRI molecular contrast agents.
Collapse
|
41
|
Ruiz-López E, Schuhmacher AJ. Transportation of Single-Domain Antibodies through the Blood-Brain Barrier. Biomolecules 2021; 11:biom11081131. [PMID: 34439797 PMCID: PMC8394617 DOI: 10.3390/biom11081131] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2021] [Revised: 07/26/2021] [Accepted: 07/28/2021] [Indexed: 02/06/2023] Open
Abstract
Single-domain antibodies derive from the heavy-chain-only antibodies of Camelidae (camel, dromedary, llama, alpaca, vicuñas, and guananos; i.e., nanobodies) and cartilaginous fishes (i.e., VNARs). Their small size, antigen specificity, plasticity, and potential to recognize unique conformational epitopes represent a diagnostic and therapeutic opportunity for many central nervous system (CNS) pathologies. However, the blood–brain barrier (BBB) poses a challenge for their delivery into the brain parenchyma. Nevertheless, numerous neurological diseases and brain pathologies, including cancer, result in BBB leakiness favoring single-domain antibodies uptake into the CNS. Some single-domain antibodies have been reported to naturally cross the BBB. In addition, different strategies and methods to deliver both nanobodies and VNARs into the brain parenchyma can be exploited when the BBB is intact. These include device-based and physicochemical disruption of the BBB, receptor and adsorptive-mediated transcytosis, somatic gene transfer, and the use of carriers/shuttles such as cell-penetrating peptides, liposomes, extracellular vesicles, and nanoparticles. Approaches based on single-domain antibodies are reaching the clinic for other diseases. Several tailoring methods can be followed to favor the transport of nanobodies and VNARs to the CNS, avoiding the limitations imposed by the BBB to fulfill their therapeutic, diagnostic, and theragnostic promises for the benefit of patients suffering from CNS pathologies.
Collapse
Affiliation(s)
- Eduardo Ruiz-López
- Molecular Oncology Group, Instituto de Investigación Sanitaria Aragón (IIS Aragón), 50009 Zaragoza, Spain;
| | - Alberto J. Schuhmacher
- Molecular Oncology Group, Instituto de Investigación Sanitaria Aragón (IIS Aragón), 50009 Zaragoza, Spain;
- Fundación Aragonesa para la Investigación y el Desarrollo (ARAID), 500018 Zaragoza, Spain
- Correspondence:
| |
Collapse
|
42
|
A Novel Nanobody Precisely Visualizes Phosphorylated Histone H2AX in Living Cancer Cells under Drug-Induced Replication Stress. Cancers (Basel) 2021; 13:cancers13133317. [PMID: 34282773 PMCID: PMC8267817 DOI: 10.3390/cancers13133317] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2021] [Revised: 06/24/2021] [Accepted: 06/25/2021] [Indexed: 12/21/2022] Open
Abstract
Simple Summary γ-H2AX, a phosphorylated variant of histone H2A, is a widely used biomarker of DNA replication stress. To develop an immunological probe able to detect and track γ-H2AX in live cancer cells, we have isolated single domain antibodies (called nanobodies) that are easily expressed as functional recombinant proteins and here we report the extensive characterization of a novel nanobody that specifically recognizes γ-H2AX. The interaction of this nanobody with the C-terminal end of γ-H2AX was determined by X-ray crystallography. Moreover, the generation of a bivalent nanobody allowed us to precisely detect γ-H2AX foci in drug-treated cells as efficiently as with commercially available conventional antibodies. Furthermore, we tracked γ-H2AX foci in live cells upon intracellular delivery of the bivalent nanobody fused to the red fluorescent protein dTomato, making, consequently, this new cost-effective reagent useful for studying drug-induced replication stress in both fixed and living cancer cells. Abstract Histone H2AX phosphorylated at serine 139 (γ-H2AX) is a hallmark of DNA damage, signaling the presence of DNA double-strand breaks and global replication stress in mammalian cells. While γ-H2AX can be visualized with antibodies in fixed cells, its detection in living cells was so far not possible. Here, we used immune libraries and phage display to isolate nanobodies that specifically bind to γ-H2AX. We solved the crystal structure of the most soluble nanobody in complex with the phosphopeptide corresponding to the C-terminus of γ-H2AX and show the atomic constituents behind its specificity. We engineered a bivalent version of this nanobody and show that bivalency is essential to quantitatively visualize γ-H2AX in fixed drug-treated cells. After labelling with a chemical fluorophore, we were able to detect γ-H2AX in a single-step assay with the same sensitivity as with validated antibodies. Moreover, we produced fluorescent nanobody-dTomato fusion proteins and applied a transduction strategy to visualize with precision γ-H2AX foci present in intact living cells following drug treatment. Together, this novel tool allows performing fast screenings of genotoxic drugs and enables to study the dynamics of this particular chromatin modification in individual cancer cells under a variety of conditions.
Collapse
|
43
|
Brilhante-da-Silva N, de Oliveira Sousa RM, Arruda A, Dos Santos EL, Marinho ACM, Stabeli RG, Fernandes CFC, Pereira SDS. Camelid Single-Domain Antibodies for the Development of Potent Diagnosis Platforms. Mol Diagn Ther 2021; 25:439-456. [PMID: 34146333 DOI: 10.1007/s40291-021-00533-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/29/2021] [Indexed: 11/26/2022]
Abstract
The distinct biophysical and pharmaceutical properties of camelid single-domain antibodies, referred to as VHHs or nanobodies, are associated with their nanometric dimensions, elevated stability, and antigen recognition capacity. These biomolecules can circumvent a number of diagnostic system limitations, especially those related to the size and stability of conventional immunoglobulins currently used in enzyme-linked immunosorbent assays and point-of-care, electrochemical, and imaging assays. In these formats, VHHs are directionally conjugated to different molecules, such as metallic nanoparticles, small peptides, and radioisotopes, which demonstrates their comprehensive versatility. Thus, the application of VHHs in diagnostic systems range from the identification of cancer cells to the detection of degenerative disease biomarkers, viral antigens, bacterial toxins, and insecticides. The improvements of sensitivity and specificity are among the central benefits resulting from the use of VHHs, which are indispensable parameters for high-quality diagnostics. Therefore, this review highlights the main biotechnological advances related to camelid single-domain antibodies and their use in in vitro and in vivo diagnostic approaches for human health.
Collapse
Affiliation(s)
- Nairo Brilhante-da-Silva
- Laboratório de Engenharia de Anticorpos, Fundação Oswaldo Cruz, Fiocruz, Unidade Rondônia, Porto Velho, RO, 76812-245, Brazil
- Programa de Pós-Graduação em Biologia Celular e Molecular, Instituto Oswaldo Cruz, IOC, Rio de Janeiro, Brazil
| | - Rosa Maria de Oliveira Sousa
- Laboratório de Engenharia de Anticorpos, Fundação Oswaldo Cruz, Fiocruz, Unidade Rondônia, Porto Velho, RO, 76812-245, Brazil
| | - Andrelisse Arruda
- Laboratório de Engenharia de Anticorpos, Fundação Oswaldo Cruz, Fiocruz, Unidade Rondônia, Porto Velho, RO, 76812-245, Brazil
| | - Eliza Lima Dos Santos
- Laboratório de Engenharia de Anticorpos, Fundação Oswaldo Cruz, Fiocruz, Unidade Rondônia, Porto Velho, RO, 76812-245, Brazil
| | - Anna Carolina Machado Marinho
- Plataforma de Desenvolvimento de Anticorpos e Nanocorpos, Fundação Oswaldo Cruz, Fiocruz Ceará, Eusebio, Brazil
- Programa de Pós-Graduação em Ciências Farmacêuticas, Universidade Federal do Ceará, Fortaleza, Brazil
| | - Rodrigo Guerino Stabeli
- Plataforma Bi-institucional de Medicina Translacional.Fundação Oswaldo Cruz-USP, Ribeirão Preto, São Paulo, Brazil
| | - Carla Freire Celedonio Fernandes
- Programa de Pós-Graduação em Biologia Celular e Molecular, Instituto Oswaldo Cruz, IOC, Rio de Janeiro, Brazil
- Plataforma de Desenvolvimento de Anticorpos e Nanocorpos, Fundação Oswaldo Cruz, Fiocruz Ceará, Eusebio, Brazil
- Programa de Pós-Graduação em Ciências Farmacêuticas, Universidade Federal do Ceará, Fortaleza, Brazil
| | - Soraya Dos Santos Pereira
- Laboratório de Engenharia de Anticorpos, Fundação Oswaldo Cruz, Fiocruz, Unidade Rondônia, Porto Velho, RO, 76812-245, Brazil.
- Programa de Pós-Graduação em Biologia Celular e Molecular, Instituto Oswaldo Cruz, IOC, Rio de Janeiro, Brazil.
- Programa de Pós-graduação em Biologia Experimental, Universidade Federal de Rondônia, Porto Velho, Brazil.
| |
Collapse
|
44
|
Ji C, Sigurdsson EM. Current Status of Clinical Trials on Tau Immunotherapies. Drugs 2021; 81:1135-1152. [PMID: 34101156 DOI: 10.1007/s40265-021-01546-6] [Citation(s) in RCA: 56] [Impact Index Per Article: 18.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/22/2021] [Indexed: 12/12/2022]
Abstract
Tau immunotherapies have advanced from proof-of-concept studies to over a dozen clinical trials for Alzheimer's disease (AD) and other tauopathies. Mechanistic studies in animal and culture models have provided valuable insight into how these therapies may work but multiple pathways are likely involved. Different groups have emphasized the importance of intracellular vs extracellular antibody-mediated clearance of the tau protein and there is no consensus on which pool of tau should ideally be targeted. Likewise, various normal and disease-selective epitopes are being targeted, and the antibody isotypes either favor phagocytosis of the tau-antibody complex or are neutral in that aspect. Most of the clinical trials are in early stages, thus their efficacy is not yet known, but all have been without any major adverse effects and some have reported target engagement. A few have been discontinued. One in phase I, presumably because of a poor pharmacokinetic profile, and three in phase II for a lack of efficacy although this trial stage is not well powered for efficacy measures. In these phase II studies, trials with two antibodies in patients with progressive supranuclear palsy or other primary tauopathies were halted but are continuing in patients with AD, and one antibody trial was stopped in early-stage AD but is continuing in moderate AD. These three antibodies have been reported to only work extracellularly and tau is not increased in the cerebrospinal fluid of primary tauopathies, which may explain the failures of two of them. In the discontinued AD trial, there are some concerns about how much of extracellular tau contains the N-terminal epitope that is being targeted. In addition, extracellular tau is only a small part of total tau, compared to intracellular tau. Targeting only the former may not be sufficient for functional benefits. Given these outcomes, decision makers within the pharmaceutical companies who green light these trials should attempt to target tau not only extracellularly but also intracellularly to increase their chances of success. Hopefully, some of the ongoing trials will provide some functional benefits to the large number of patients with tauopathies.
Collapse
Affiliation(s)
- Changyi Ji
- Department of Neuroscience and Physiology, Neuroscience Institute, New York University Grossman School of Medicine, Science Building, 11th floor, 435 East 30th Street, New York, NY, 10016, USA
| | - Einar M Sigurdsson
- Department of Neuroscience and Physiology, Neuroscience Institute, New York University Grossman School of Medicine, Science Building, 11th floor, 435 East 30th Street, New York, NY, 10016, USA. .,Department of Psychiatry, Neuroscience Institute, New York University Grossman School of Medicine, New York, NY, USA.
| |
Collapse
|
45
|
Habiba U, Descallar J, Kreilaus F, Adhikari UK, Kumar S, Morley JW, Bui BV, Hamaoui MK, Tayebi M. Detection of retinal and blood Aβ oligomers with nanobodies. ALZHEIMER'S & DEMENTIA: DIAGNOSIS, ASSESSMENT & DISEASE MONITORING 2021; 13:e12193. [PMID: 33977118 PMCID: PMC8101010 DOI: 10.1002/dad2.12193] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/06/2021] [Revised: 04/13/2021] [Accepted: 04/13/2021] [Indexed: 12/11/2022]
Abstract
Introduction Abnormal retinal changes are increasingly recognized as an early pathological change in Alzheimer's disease (AD). Although amyloid beta oligomers (Aβo) have been shown to accumulate in the blood and retina of AD patients and animals, it is not known whether the early Aβo deposition precedes their accumulation in brain. Methods and results Using nanobodies targeting Aβ1‐40 and Aβ1‐42 oligomers we were able to detect Aβ oligomers in the retina and blood but not in the brain of 3‐month‐old APP/PS1 mice. Furthermore, Aβ plaques were detected in the brain but not the retina of 3‐month‐old APP/PS1 mice. Conclusion These results suggest that retinal accumulation of Aβo originates from peripheral blood and precedes cognitive decline and Aβo deposition in the brain. This provides a very strong basis to develop and implement an “eye test” for early detection of AD using nanobodies targeting retinal Aβ.
Collapse
Affiliation(s)
- Umma Habiba
- School of Medicine Western Sydney University Campbelltown New South Wales Australia
| | - Joseph Descallar
- South Western Sydney Clinical School, Faculty of Medicine UNSW, Liverpool Hospital Liverpool New South Wales Australia.,Ingham Institute of Applied Medical Research Liverpool New South Wales Australia
| | - Fabian Kreilaus
- School of Medicine Western Sydney University Campbelltown New South Wales Australia
| | - Utpal K Adhikari
- School of Medicine Western Sydney University Campbelltown New South Wales Australia
| | - Sachin Kumar
- School of Medicine Western Sydney University Campbelltown New South Wales Australia
| | - John W Morley
- School of Medicine Western Sydney University Campbelltown New South Wales Australia
| | - Bang V Bui
- Department of Optometry and Vision Sciences University of Melbourne Melbourne Victoria Australia
| | - Maya K Hamaoui
- Department of Neurosurgery Maxine Dunitz Neurosurgical Research Institute and Department of Biomedical Sciences Cedars-Sinai Medical Center Los Angeles California USA
| | - Mourad Tayebi
- School of Medicine Western Sydney University Campbelltown New South Wales Australia
| |
Collapse
|
46
|
Sun ZT, Ma C, Li GJ, Zheng XY, Hao YT, Yang Y, Wang X. Application of Antibody Fragments Against Aβ With Emphasis on Combined Application With Nanoparticles in Alzheimer's Disease. Front Pharmacol 2021; 12:654611. [PMID: 33967797 PMCID: PMC8100690 DOI: 10.3389/fphar.2021.654611] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2021] [Accepted: 04/09/2021] [Indexed: 11/13/2022] Open
Abstract
Alzheimer’s disease (AD) is one of the most common neurodegenerative diseases and accumulating evidences suggest a key role of amyloid-β (Aβ) peptide in the pathogenesis of AD. According to the amyloid cascade hypothesis, the imbalance of producing and clearing Aβ is the beginning of neurodegeneration and dementia. Consequently, immunotherapy becomes popular through using antibodies against Aβ. However, many studies of monoclonal antibodies were stopped because adverse effects appeared or there were no evident benefits observed. Some antibody fragments have many advantages over monoclonal antibodies, such as small sizes, lack of the crystallizable fraction (Fc) and so on. There are three main antibody fragments, including single chain variable fragments (scFvs), Fab fragments and single-domain antibody fragments. Nanoparticles can facilitate the entry of drug molecules across the blood-brain barrier, making them become excellent carriers. Various kinds of nanoparticles have been applied in the treatment of AD. The combination of nanoparticles and antibody fragments against amyloid-β can be used in the diagnosis and treatment of Alzheimer’s disease. In this review, we summarize the progress of antibody fragments against amyloid-β in AD, focusing on the combined application with nanoparticles in the diagnosis and treatment of AD.
Collapse
Affiliation(s)
- Zhi-Ting Sun
- Department of Neurology and Neuroscience Center, The First Hospital of Jilin University, Changchun, China
| | - Chi Ma
- Department of Neurosurgery, The First Hospital of Jilin University, Changchun, China
| | - Guang-Jian Li
- Department of Neurology and Neuroscience Center, The First Hospital of Jilin University, Changchun, China
| | - Xiang-Yu Zheng
- Department of Neurology and Neuroscience Center, The First Hospital of Jilin University, Changchun, China
| | - Yi-Tong Hao
- Department of Neurology and Neuroscience Center, The First Hospital of Jilin University, Changchun, China
| | - Yu Yang
- Department of Neurology and Neuroscience Center, The First Hospital of Jilin University, Changchun, China
| | - Xu Wang
- Department of Neurology and Neuroscience Center, The First Hospital of Jilin University, Changchun, China
| |
Collapse
|
47
|
Forlani G, Di Ventura B. A light way for nuclear cell biologists. J Biochem 2021; 169:273-286. [PMID: 33245128 PMCID: PMC8053400 DOI: 10.1093/jb/mvaa139] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Accepted: 11/12/2020] [Indexed: 12/11/2022] Open
Abstract
The nucleus is a very complex organelle present in eukaryotic cells. Having the crucial task to safeguard, organize and manage the genetic information, it must tightly control its molecular constituents, its shape and its internal architecture at any given time. Despite our vast knowledge of nuclear cell biology, much is yet to be unravelled. For instance, only recently we came to appreciate the existence of a dynamic nuclear cytoskeleton made of actin filaments that regulates processes such as gene expression, DNA repair and nuclear expansion. This suggests further exciting discoveries ahead of us. Modern cell biologists embrace a new methodology relying on precise perturbations of cellular processes that require a reversible, highly spatially confinable, rapid, inexpensive and tunEable external stimulus: light. In this review, we discuss how optogenetics, the state-of-the-art technology that uses genetically encoded light-sensitive proteins to steer biological processes, can be adopted to specifically investigate nuclear cell biology.
Collapse
Affiliation(s)
- Giada Forlani
- Spemann Graduate School of Biology and Medicine (SGBM)
- Centers for Biological Signalling Studies BIOSS and CIBSS
- Faculty of Biology, Institute of Biology II, Albert Ludwigs University of Freiburg, Freiburg, Germany
| | - Barbara Di Ventura
- Centers for Biological Signalling Studies BIOSS and CIBSS
- Faculty of Biology, Institute of Biology II, Albert Ludwigs University of Freiburg, Freiburg, Germany
| |
Collapse
|
48
|
Nanobodies as Versatile Tool for Multiscale Imaging Modalities. Biomolecules 2020; 10:biom10121695. [PMID: 33353213 PMCID: PMC7767244 DOI: 10.3390/biom10121695] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2020] [Revised: 12/11/2020] [Accepted: 12/14/2020] [Indexed: 02/07/2023] Open
Abstract
Molecular imaging is constantly growing in different areas of preclinical biomedical research. Several imaging methods have been developed and are continuously updated for both in vivo and in vitro applications, in order to increase the information about the structure, localization and function of molecules involved in physiology and disease. Along with these progresses, there is a continuous need for improving labeling strategies. In the last decades, the single domain antigen-binding fragments nanobodies (Nbs) emerged as important molecular imaging probes. Indeed, their small size (~15 kDa), high stability, affinity and modularity represent desirable features for imaging applications, providing higher tissue penetration, rapid targeting, increased spatial resolution and fast clearance. Accordingly, several Nb-based probes have been generated and applied to a variety of imaging modalities, ranging from in vivo and in vitro preclinical imaging to super-resolution microscopy. In this review, we will provide an overview of the state-of-the-art regarding the use of Nbs in several imaging modalities, underlining their extreme versatility and their enormous potential in targeting molecules and cells of interest in both preclinical and clinical studies.
Collapse
|
49
|
Abstract
Unique, functional, homodimeric heavy chain-only antibodies, devoid of light chains, are circulating in the blood of Camelidae. These antibodies recognize their cognate antigen via one single domain, known as VHH or Nanobody. This serendipitous discovery made three decades ago has stimulated a growing number of researchers to generate highly specific Nanobodies against a myriad of targets. The small size, strict monomeric state, robustness, and easy tailoring of these Nanobodies have inspired many groups to design innovative Nanobody-based multi-domain constructs to explore novel applications. As such, Nanobodies have been employed as an exquisite research tool in structural, cell, and developmental biology. Furthermore, Nanobodies have demonstrated their benefit for more sensitive diagnostic tests. Finally, several Nanobody-based constructs have been designed to develop new therapeutic products.
Collapse
Affiliation(s)
- Serge Muyldermans
- Cellular and Molecular Immunology, Vrije Universiteit Brussel, 1050 Brussels, Belgium; .,Liaoning Key Laboratory of Molecular Recognition and Imaging, School of Bioengineering, Dalian University of Technology, Dalian 116023, Liaoning, People's Republic of China
| |
Collapse
|
50
|
Gao Y, Zhu J, Lu H. Single domain antibody-based vectors in the delivery of biologics across the blood-brain barrier: a review. Drug Deliv Transl Res 2020; 11:1818-1828. [PMID: 33155179 DOI: 10.1007/s13346-020-00873-7] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/19/2020] [Indexed: 10/23/2022]
Abstract
Biologics are a promising and effective method for the treatment of central nervous system (CNS) diseases. The blood-brain barrier (BBB) is a natural barrier for the delivery of biologics into the brain, which decreases the effective concentration of drugs in the CNS. A range of strategies has been explored to transport biologics across the BBB endothelium, typically via receptor-mediated transcytosis (RMT), which involving molecules for endogenous BBB receptors to be fused with biologics. This review emphasized a category of novel alternative RMT-targeting vectors: single domain antibodies (sdAb). SdAbs are a unique category of antibodies derived from naturally occurring heavy-chain-only antibodies. Herein, we describe their properties, mechanisms, modifications, and translational perspectives for their ability to transmigrate across the BBB in vitro and in vivo in detail.
Collapse
Affiliation(s)
- Yang Gao
- Engineering Research Center of Cell and Therapeutic Antibody, Ministry of Education, School of Pharmacy, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai, 200240, China
| | - Jianwei Zhu
- Engineering Research Center of Cell and Therapeutic Antibody, Ministry of Education, School of Pharmacy, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai, 200240, China
| | - Huili Lu
- Engineering Research Center of Cell and Therapeutic Antibody, Ministry of Education, School of Pharmacy, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai, 200240, China.
| |
Collapse
|