1
|
Cruz E, Nisbet RM, Padmanabhan P, van Waardenberg AJ, Graham ME, Nkajja G, Tapaswi S, Connor BJ, Robinson P, Götz J. Proteostasis as a fundamental principle of Tau immunotherapy. Brain 2025; 148:168-184. [PMID: 39074206 PMCID: PMC11706327 DOI: 10.1093/brain/awae254] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2023] [Revised: 05/01/2024] [Accepted: 07/08/2024] [Indexed: 07/31/2024] Open
Abstract
The microtubule-associated protein Tau is a driver of neuronal dysfunction in Alzheimer's disease and other tauopathies. In this process, Tau initially undergoes subtle changes to its abundance, subcellular localization and a vast array of post-translational modifications including phosphorylation that progressively result in the protein's somatodendritic accumulation and dysregulation of multiple Tau-dependent cellular processes. Given the various loss- and gain-of-functions of Tau in disease and the brain-wide changes in the proteome that characterize tauopathies, we asked whether targeting Tau would restore the alterations in proteostasis observed in disease. Therefore, by phage display, we generated a novel pan-Tau antibody, RNJ1, that preferentially binds human Tau and neutralizes proteopathic seeding activity in multiple cell lines and benchmarked it against a clinically tested pan-Tau antibody, HJ8.5 (murine version of tilavonemab). We then evaluated both antibodies, alone and in combination, in the K3 tauopathy mouse model, showing reduced Tau pathology and improvements in neuronal function following 14 weekly treatments, without obtaining synergy for the combination. These effects were more pronounced in female mice. To investigate the molecular mechanisms contributing to improvements in neuronal function, we employed quantitative proteomics, phosphoproteomics and kinase prediction analysis to first establish alterations in K3 mice relative to wild-type controls at the proteome level. In female K3 mice, we found 342 differentially abundant proteins, which are predominantly involved in metabolic and microtubule-associated processes, strengthening previously reported findings of defects in several functional domains in multiple tauopathy models. We next asked whether antibody-mediated Tau target engagement indirectly affects levels of deregulated proteins in the K3 model. Importantly, both immunotherapies, in particular RNJ1, induced abundance shifts towards a restoration to wild-type levels (proteostasis). A total of 257 of 342 (∼75%) proteins altered in K3 were closer in abundance to wild-type levels after RNJ1 treatment, and 73% after HJ8.5 treatment. However, the magnitude of these changes was less pronounced than that observed with RNJ1. Furthermore, analysis of the phosphoproteome showed an even stronger restoration effect with RNJ1, with ∼82% of altered phosphopeptides in K3 showing a shift to wild-type levels, and 75% with HJ8.5. Gene set over-representation analysis further confirmed that proteins undergoing restoration are involved in biological pathways affected in K3 mice. Together, our study suggests that a Tau immunotherapy-induced restoration of proteostasis links target engagement and treatment efficacy.
Collapse
Affiliation(s)
- Esteban Cruz
- Clem Jones Centre for Ageing Dementia Research (CJCADR), Queensland Brain Institute (QBI), The University of Queensland, St Lucia Campus (Brisbane), Brisbane, QLD 4072, Australia
| | - Rebecca M Nisbet
- Clem Jones Centre for Ageing Dementia Research (CJCADR), Queensland Brain Institute (QBI), The University of Queensland, St Lucia Campus (Brisbane), Brisbane, QLD 4072, Australia
- The Florey, The University of Melbourne, Parkville, Melbourne, VIC 3052, Australia
| | - Pranesh Padmanabhan
- Clem Jones Centre for Ageing Dementia Research (CJCADR), Queensland Brain Institute (QBI), The University of Queensland, St Lucia Campus (Brisbane), Brisbane, QLD 4072, Australia
| | | | - Mark E Graham
- Synapse Proteomics, Children’s Medical Research Institute, The University of Sydney, Westmead, NSW 2145, Australia
| | - Godfrey Nkajja
- Clem Jones Centre for Ageing Dementia Research (CJCADR), Queensland Brain Institute (QBI), The University of Queensland, St Lucia Campus (Brisbane), Brisbane, QLD 4072, Australia
| | - Swara Tapaswi
- Clem Jones Centre for Ageing Dementia Research (CJCADR), Queensland Brain Institute (QBI), The University of Queensland, St Lucia Campus (Brisbane), Brisbane, QLD 4072, Australia
| | - Bradley J Connor
- Clem Jones Centre for Ageing Dementia Research (CJCADR), Queensland Brain Institute (QBI), The University of Queensland, St Lucia Campus (Brisbane), Brisbane, QLD 4072, Australia
| | - Phil Robinson
- Cell Signalling Unit, Children's Medical Research Institute, The University of Sydney, Westmead, NSW 2145, Australia
| | - Jürgen Götz
- Clem Jones Centre for Ageing Dementia Research (CJCADR), Queensland Brain Institute (QBI), The University of Queensland, St Lucia Campus (Brisbane), Brisbane, QLD 4072, Australia
| |
Collapse
|
2
|
Zhang Y, Yu W, Zhang L, Li P. Application of engineered antibodies (scFvs and nanobodies) targeting pathological protein aggregates in Alzheimer's disease. Expert Opin Investig Drugs 2024; 33:1047-1062. [PMID: 39177331 DOI: 10.1080/13543784.2024.2396911] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2024] [Revised: 07/07/2024] [Accepted: 08/22/2024] [Indexed: 08/24/2024]
Abstract
INTRODUCTION The misfolding and aggregation of proteins are associated with various neurodegenerative diseases, such as Alzheimer's disease (AD). The small-molecule engineered antibodies, such as single-chain fragment variable (scFv) antibodies and nanobodies (Nbs), have gained attention in recent years due to their strong conformational specificity, ability to cross the blood-brain barrier (BBB), low immunogenicity, and enhanced proximity to active sites within aggregates. AREAS COVERED We have reviewed recent advances in therapies involving scFvs and Nbs that efficiently and specifically target pathological protein aggregates. Relevant publications were searched for in MEDLINE, GOOGLE SCHOLAR, Elsevier ScienceDirect and Wiley Online Library. EXPERT OPINION We reviewed the recent and specific targeting of pathological protein aggregates by scFvs and Nbs. These engineered antibodies can inhibit the aggregation or promote the disassembly of misfolded proteins by recognizing antigenic epitopes or through conformational specificity. Additionally, we discuss strategies for improving the effective application of engineered antibodies in treating AD. These technological strategies will lay the foundation for the clinical application of small-molecule antibody drugs in developing effective treatments for neurological diseases. Through rational application strategies, small-molecule engineered antibodies are expected to have significant potential in targeted therapy for neurological disorders.
Collapse
Affiliation(s)
- Yuan Zhang
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, China
| | - Wanpeng Yu
- Medical Collage, Qingdao University, Qingdao, China
| | - Lei Zhang
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, China
| | - Peifeng Li
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, China
| |
Collapse
|
3
|
Singh H, Das A, Khan MM, Pourmotabbed T. New insights into the therapeutic approaches for the treatment of tauopathies. Neural Regen Res 2024; 19:1020-1026. [PMID: 37862204 PMCID: PMC10749630 DOI: 10.4103/1673-5374.385288] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2023] [Revised: 07/05/2023] [Accepted: 08/10/2023] [Indexed: 10/22/2023] Open
Abstract
Tauopathies are a group of neurological disorders, including Alzheimer's disease and frontotemporal dementia, which involve progressive neurodegeneration, cognitive deficits, and aberrant tau protein accumulation. The development of tauopathies cannot currently be stopped or slowed down by treatment measures. Given the significant contribution of tau burden in primary tauopathies and the strong association between pathogenic tau accumulation and cognitive deficits, there has been a lot of interest in creating therapies that can alleviate tau pathology and render neuroprotective effects. Recently, small molecules, immunotherapies, and gene therapy have been used to reduce the pathological tau burden and prevent neurodegeneration in animal models of tauopathies. However, the major pitfall of the current therapeutic approach is the difficulty of drugs and gene-targeting modalities to cross the blood-brain barrier and their unintended side effects. In this review, the current therapeutic strategies used for tauopathies including the use of oligonucleotide-based gene therapy approaches that have shown a promising result for the treatment of tauopathies and Alzheimer's disease in preclinical animal models, have been discussed.
Collapse
Affiliation(s)
- Himanshi Singh
- Department of Microbiology, Immunology and Biochemistry, University of Tennessee Health Science Center, Memphis, TN, USA
- Department of Biotechnology, Delhi Technological University, Shahbad Daulatpur, Delhi, India
- Department of Neurology, College of Medicine, University of Tennessee Health Science Center, Memphis, TN, USA
| | - Asmita Das
- Department of Biotechnology, Delhi Technological University, Shahbad Daulatpur, Delhi, India
| | - Mohammad Moshahid Khan
- Department of Neurology, College of Medicine, University of Tennessee Health Science Center, Memphis, TN, USA
- Neuroscience Institute, University of Tennessee Health Science Center, Memphis, TN, USA
- Center for Muscle, Metabolism and Neuropathology, Division of Regenerative and Rehabilitation Sciences and Department of Physical Therapy, College of Health Professions, University of Tennessee Health Science Center, Memphis, TN, USA
| | - Tayebeh Pourmotabbed
- Department of Microbiology, Immunology and Biochemistry, University of Tennessee Health Science Center, Memphis, TN, USA
| |
Collapse
|
4
|
Wongsodirdjo P, Caruso AC, Yong AK, Lester MA, Vella LJ, Hung YH, Nisbet RM. Messenger RNA-encoded antibody approach for targeting extracellular and intracellular tau. Brain Commun 2024; 6:fcae100. [PMID: 38585667 PMCID: PMC10996922 DOI: 10.1093/braincomms/fcae100] [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: 10/25/2023] [Revised: 02/19/2024] [Accepted: 03/21/2024] [Indexed: 04/09/2024] Open
Abstract
Monoclonal antibodies have emerged as a leading therapeutic agent for the treatment of disease, including Alzheimer's disease. In the last year, two anti-amyloid monoclonal antibodies, lecanemab and aducanumab, have been approved in the USA for the treatment of Alzheimer's disease, whilst several tau-targeting monoclonal antibodies are currently in clinical trials. Such antibodies, however, are expensive and timely to produce and require frequent dosing regimens to ensure disease-modifying effects. Synthetic in vitro-transcribed messenger RNA encoding antibodies for endogenous protein expression holds the potential to overcome many of the limitations associated with protein antibody production. Here, we have generated synthetic in vitro-transcribed messenger RNA encoding a tau-specific antibody as a full-sized immunoglobulin and as a single-chain variable fragment. In vitro transfection of human neuroblastoma SH-SY5Y cells demonstrated the ability of the synthetic messenger RNA to be translated into a functional tau-specific antibody. Furthermore, we show that the translation of the tau-specific single-chain variable fragment as an intrabody results in the specific engagement of intracellular tau. This work highlights the utility of messenger RNA for the delivery of antibody therapeutics, including intrabodies, for the targeting of tau in Alzheimer's disease and other tauopathies.
Collapse
Affiliation(s)
- Patricia Wongsodirdjo
- The Florey Institute, Parkville, Victoria 3052, Australia
- Florey Department of Neuroscience and Mental Health, The University of Melbourne, Parkville, Victoria 3052, Australia
| | - Alayna C Caruso
- The Florey Institute, Parkville, Victoria 3052, Australia
- Florey Department of Neuroscience and Mental Health, The University of Melbourne, Parkville, Victoria 3052, Australia
| | - Alicia K Yong
- The Florey Institute, Parkville, Victoria 3052, Australia
| | - Madeleine A Lester
- The Florey Institute, Parkville, Victoria 3052, Australia
- Florey Department of Neuroscience and Mental Health, The University of Melbourne, Parkville, Victoria 3052, Australia
| | - Laura J Vella
- The Florey Institute, Parkville, Victoria 3052, Australia
- Department of Surgery, The Royal Melbourne Hospital, The University of Melbourne, Parkville, Victoria 3052, Australia
| | - Ya Hui Hung
- The Florey Institute, Parkville, Victoria 3052, Australia
- Florey Department of Neuroscience and Mental Health, The University of Melbourne, Parkville, Victoria 3052, Australia
| | - Rebecca M Nisbet
- The Florey Institute, Parkville, Victoria 3052, Australia
- Florey Department of Neuroscience and Mental Health, The University of Melbourne, Parkville, Victoria 3052, Australia
| |
Collapse
|
5
|
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: 84] [Impact Index Per Article: 42.0] [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
|
6
|
Düchs M, Blazevic D, Rechtsteiner P, Kenny C, Lamla T, Low S, Savistchenko J, Neumann M, Melki R, Schönberger T, Stierstorfer B, Wyatt D, Igney F, Ciossek T. AAV-mediated expression of a new conformational anti-aggregated α-synuclein antibody prolongs survival in a genetic model of α-synucleinopathies. NPJ Parkinsons Dis 2023; 9:91. [PMID: 37322068 DOI: 10.1038/s41531-023-00542-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2022] [Accepted: 06/05/2023] [Indexed: 06/17/2023] Open
Abstract
Prion-like transmission of pathology in α-synucleinopathies like Parkinson's disease or multiple system atrophy is increasingly recognized as one potential mechanism to address disease progression. Active and passive immunotherapies targeting insoluble, aggregated α-synuclein are already being actively explored in the clinic with mixed outcomes so far. Here, we report the identification of 306C7B3, a highly selective, aggregate-specific α-synuclein antibody with picomolar affinity devoid of binding to the monomeric, physiologic protein. 306C7B3 binding is Ser129-phosphorylation independent and shows high affinity to several different aggregated α-synuclein polymorphs, increasing the likelihood that it can also bind to the pathological seeds assumed to drive disease progression in patients. In support of this, highly selective binding to pathological aggregates in postmortem brains of MSA patients was demonstrated, with no staining in samples from other human neurodegenerative diseases. To achieve CNS exposure of 306C7B3, an adeno-associated virus (AAV) based approach driving expression of the secreted antibody within the brain of (Thy-1)-[A30P]-hα-synuclein mice was used. Widespread central transduction after intrastriatal inoculation was ensured by using the AAV2HBKO serotype, with transduction being spread to areas far away from the inoculation site. Treatment of (Thy-1)-[A30P]-hα-synuclein mice at the age of 12 months demonstrated significantly increased survival, with 306C7B3 concentration reaching 3.9 nM in the cerebrospinal fluid. These results suggest that AAV-mediated expression of 306C7B3, targeting extracellular, presumably disease-propagating aggregates of α-synuclein, has great potential as a disease-modifying therapy for α-synucleinopathies as it ensures CNS exposure of the antibody, thereby mitigating the selective permeability of the blood-brain barrier.
Collapse
Affiliation(s)
- Matthias Düchs
- Boehringer Ingelheim Pharma GmbH & Co KG, Biberach an der Riss, Germany
| | - Dragica Blazevic
- Boehringer Ingelheim Pharma GmbH & Co KG, Biberach an der Riss, Germany
| | | | | | - Thorsten Lamla
- Boehringer Ingelheim Pharma GmbH & Co KG, Biberach an der Riss, Germany
| | - Sarah Low
- Boehringer Ingelheim USA, Ridgefield, CT, USA
| | | | - Manuela Neumann
- Molecular Neuropathology of Neurodegenerative Diseases, German Center for Neurodegenerative Diseases, Tübingen, Germany
- Department of Neuropathology, University Hospital of Tübingen, Tübingen, Germany
| | - Ronald Melki
- Institut Francois Jacob (MIRCen), CEA, CNRS, Fontenay-aux-Roses, France
| | - Tanja Schönberger
- Boehringer Ingelheim Pharma GmbH & Co KG, Biberach an der Riss, Germany
| | | | - David Wyatt
- Boehringer Ingelheim Pharma GmbH & Co KG, Biberach an der Riss, Germany
| | - Frederik Igney
- Boehringer Ingelheim Pharma GmbH & Co KG, Biberach an der Riss, Germany
| | - Thomas Ciossek
- Boehringer Ingelheim Pharma GmbH & Co KG, Biberach an der Riss, Germany.
| |
Collapse
|
7
|
Liang M, Gu L, Zhang H, Min J, Wang Z, Ma Z, Zhang C, Zeng S, Pan Y, Yan D, Shen Z, Huang W. Design, Synthesis, and Bioactivity of Novel Bifunctional Small Molecules for Alzheimer's disease. ACS OMEGA 2022; 7:26308-26315. [PMID: 35936449 PMCID: PMC9352321 DOI: 10.1021/acsomega.2c02130] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
The abnormal phosphorylation of the τ-protein is a typical early pathological feature of Alzheimer's disease (AD). The excessive phosphorylation of the τ-protein in the brain causes the formation of neurofibrillary tangles (NFTs) and increases the neurotoxicity of amyloid-β (Aβ). Thus, targeting the τ-protein is considered a promising strategy for treating AD. Herein, we designed and synthesized a series of molecules containing bifunctional groups to recognize the τ-protein and the E3 ligase. The molecules were examined in vitro, and their effects were tested on PC12 cells. In addition, we further studied the pharmacokinetics of compound I3 in healthy rats. Our data showed that compound I3 could effectively degrade τ-protein, reduce Aβ-induced cytotoxicity, and regulate the uneven distribution of mitochondria, which may open a new therapeutic strategy for the treatment of AD.
Collapse
Affiliation(s)
- Meihao Liang
- Affiliated
Yongkang First People’s Hospital and School of Pharmacy, Hangzhou Medical College, Hangzhou, Zhejiang 310013, China
- Key
Laboratory of Neuropsychiatric Drug Research of Zhejiang Province,
School of Pharmacy, Hangzhou Medical College, Hangzhou, Zhejiang 310013, P.R. China
| | - Lili Gu
- Key
Laboratory of Neuropsychiatric Drug Research of Zhejiang Province,
School of Pharmacy, Hangzhou Medical College, Hangzhou, Zhejiang 310013, P.R. China
| | - Hongjie Zhang
- Key
Laboratory of Neuropsychiatric Drug Research of Zhejiang Province,
School of Pharmacy, Hangzhou Medical College, Hangzhou, Zhejiang 310013, P.R. China
| | - Jingli Min
- Key
Laboratory of Neuropsychiatric Drug Research of Zhejiang Province,
School of Pharmacy, Hangzhou Medical College, Hangzhou, Zhejiang 310013, P.R. China
| | - Zunyuan Wang
- Key
Laboratory of Neuropsychiatric Drug Research of Zhejiang Province,
School of Pharmacy, Hangzhou Medical College, Hangzhou, Zhejiang 310013, P.R. China
| | - Zhen Ma
- Key
Laboratory of Neuropsychiatric Drug Research of Zhejiang Province,
School of Pharmacy, Hangzhou Medical College, Hangzhou, Zhejiang 310013, P.R. China
| | - Chixiao Zhang
- Key
Laboratory of Neuropsychiatric Drug Research of Zhejiang Province,
School of Pharmacy, Hangzhou Medical College, Hangzhou, Zhejiang 310013, P.R. China
| | - Shenxin Zeng
- Key
Laboratory of Neuropsychiatric Drug Research of Zhejiang Province,
School of Pharmacy, Hangzhou Medical College, Hangzhou, Zhejiang 310013, P.R. China
| | - Youlu Pan
- Key
Laboratory of Neuropsychiatric Drug Research of Zhejiang Province,
School of Pharmacy, Hangzhou Medical College, Hangzhou, Zhejiang 310013, P.R. China
| | - Dongmei Yan
- Key
Laboratory of Neuropsychiatric Drug Research of Zhejiang Province,
School of Pharmacy, Hangzhou Medical College, Hangzhou, Zhejiang 310013, P.R. China
| | - Zhengrong Shen
- Key
Laboratory of Neuropsychiatric Drug Research of Zhejiang Province,
School of Pharmacy, Hangzhou Medical College, Hangzhou, Zhejiang 310013, P.R. China
| | - Wenhai Huang
- Affiliated
Yongkang First People’s Hospital and School of Pharmacy, Hangzhou Medical College, Hangzhou, Zhejiang 310013, China
- Key
Laboratory of Neuropsychiatric Drug Research of Zhejiang Province,
School of Pharmacy, Hangzhou Medical College, Hangzhou, Zhejiang 310013, P.R. China
| |
Collapse
|
8
|
Marino M, Holt MG. AAV Vector-Mediated Antibody Delivery (A-MAD) in the Central Nervous System. Front Neurol 2022; 13:870799. [PMID: 35493843 PMCID: PMC9039256 DOI: 10.3389/fneur.2022.870799] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Accepted: 02/21/2022] [Indexed: 11/13/2022] Open
Abstract
In the last four decades, monoclonal antibodies and their derivatives have emerged as a powerful class of therapeutics, largely due to their exquisite targeting specificity. Several clinical areas, most notably oncology and autoimmune disorders, have seen the successful introduction of monoclonal-based therapeutics. However, their adoption for treatment of Central Nervous System diseases has been comparatively slow, largely due to issues of efficient delivery resulting from limited permeability of the Blood Brain Barrier. Nevertheless, CNS diseases are becoming increasingly prevalent as societies age, accounting for ~6.5 million fatalities worldwide per year. Therefore, harnessing the full therapeutic potential of monoclonal antibodies (and their derivatives) in this clinical area has become a priority. Adeno-associated virus-based vectors (AAVs) are a potential solution to this problem. Preclinical studies have shown that AAV vector-mediated antibody delivery provides protection against a broad range of peripheral diseases, such as the human immunodeficiency virus (HIV), influenza and malaria. The parallel identification and optimization of AAV vector platforms which cross the Blood Brain Barrier with high efficiency, widely transducing the Central Nervous System and allowing high levels of local transgene production, has now opened a number of interesting scenarios for the development of AAV vector-mediated antibody delivery strategies to target Central Nervous System proteinopathies.
Collapse
Affiliation(s)
- Marika Marino
- Laboratory of Glia Biology, VIB-KU Leuven, Center for Brain & Disease Research, Leuven, Belgium
- Department of Neurosciences, KU Leuven, Leuven, Belgium
| | - Matthew G. Holt
- Laboratory of Glia Biology, VIB-KU Leuven, Center for Brain & Disease Research, Leuven, Belgium
- Department of Neurosciences, KU Leuven, Leuven, Belgium
- Leuven Brain Institute, Leuven, Belgium
- Synapse Biology Group, Instituto de Investigação e Inovação em Saúde (i3S), University of Porto, Porto, Portugal
- *Correspondence: Matthew G. Holt
| |
Collapse
|
9
|
Mann CN, Devi SS, Kersting CT, Bleem AV, Karch CM, Holtzman DM, Gallardo G. Astrocytic α2-Na +/K + ATPase inhibition suppresses astrocyte reactivity and reduces neurodegeneration in a tauopathy mouse model. Sci Transl Med 2022; 14:eabm4107. [PMID: 35171651 DOI: 10.1126/scitranslmed.abm4107] [Citation(s) in RCA: 48] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Alzheimer's disease (AD) is the most dominant form of dementia characterized by the deposition of extracellular amyloid plaques and intracellular neurofibrillary tau tangles (NFTs). In addition to these pathologies, an emerging pathophysiological mechanism that influences AD is neuroinflammation. Astrocytes are a vital type of glial cell that contribute to neuroinflammation, and reactive astrocytes, or astrogliosis, are a well-known pathological feature of AD. However, the mechanisms by which astrocytes contribute to the neurodegenerative process in AD have not been fully elucidated. Here, we showed that astrocytic α2-Na+/K+ adenosine triphosphatase (α2-NKA) is elevated in postmortem human brain tissue from AD and progressive nuclear palsy, a primary tauopathy. The increased astrocytic α2-NKA was also recapitulated in a mouse model of tauopathy. Pharmacological inhibition of α2-NKA robustly suppressed neuroinflammation and reduced brain atrophy. In addition, α2-NKA knockdown in tauopathy mice halted the accumulation of tau pathology. We also demonstrated that α2-NKA promoted tauopathy, in part, by regulating the proinflammatory protein lipocalin-2 (Lcn2). Overexpression of Lcn2 in tauopathy mice increased tau pathology, and prolonged Lcn2 exposure to primary neurons promoted tau uptake in vitro. These studies collectively highlight the contribution of reactive astrocytes to tau pathogenesis in mice and define α2-NKA as a major regulator of astrocytic-dependent neuroinflammation.
Collapse
Affiliation(s)
- Carolyn N Mann
- Department of Neurology, Washington University School of Medicine, St. Louis, MO 63110, USA.,Hope Center for Neurological Disorders, Washington University, St. Louis, MO 63110, USA
| | - Shamulailatpam Shreedarshanee Devi
- Department of Neurology, Washington University School of Medicine, St. Louis, MO 63110, USA.,Hope Center for Neurological Disorders, Washington University, St. Louis, MO 63110, USA
| | - Corey T Kersting
- Department of Neurology, Washington University School of Medicine, St. Louis, MO 63110, USA.,Hope Center for Neurological Disorders, Washington University, St. Louis, MO 63110, USA
| | - Amber V Bleem
- Department of Neurology, Washington University School of Medicine, St. Louis, MO 63110, USA.,Hope Center for Neurological Disorders, Washington University, St. Louis, MO 63110, USA
| | - Celeste M Karch
- Hope Center for Neurological Disorders, Washington University, St. Louis, MO 63110, USA.,Department of Psychiatry, Washington University in St. Louis School of Medicine, St. Louis, MO 63110, USA.,Charles F. and Joanne Knight Alzheimer's Disease Research Center, Washington University, St. Louis, MO 63110, USA
| | - David M Holtzman
- Department of Neurology, Washington University School of Medicine, St. Louis, MO 63110, USA.,Hope Center for Neurological Disorders, Washington University, St. Louis, MO 63110, USA.,Charles F. and Joanne Knight Alzheimer's Disease Research Center, Washington University, St. Louis, MO 63110, USA
| | - Gilbert Gallardo
- Department of Neurology, Washington University School of Medicine, St. Louis, MO 63110, USA.,Hope Center for Neurological Disorders, Washington University, St. Louis, MO 63110, USA
| |
Collapse
|
10
|
Golde TE. Disease-Modifying Therapies for Alzheimer's Disease: More Questions than Answers. Neurotherapeutics 2022; 19:209-227. [PMID: 35229269 PMCID: PMC8885119 DOI: 10.1007/s13311-022-01201-2] [Citation(s) in RCA: 66] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/09/2022] [Indexed: 12/17/2022] Open
Abstract
Scientific advances over the last four decades have steadily infused the Alzheimer's disease (AD) field with great optimism that therapies targeting Aβ, amyloid, tau, and innate immune activation states in the brain would provide disease modification. Unfortunately, this optimistic scenario has not yet played out. Though a recent approval of the anti-Aβ aggregate binding antibody, Aduhelm (aducanumab), as a "disease-modifying therapy for AD" is viewed by some as a breakthrough, many remain unconvinced by the data underlying this approval. Collectively, we have not succeeded in changing AD from a largely untreatable, inevitable, and incurable disease to a treatable, preventable, and curable one. Here, I will review the major foci of the AD "disease-modifying" therapeutic pipeline and some of the "open questions" that remain in terms of these therapeutic approaches. I will conclude the review by discussing how we, as a field, might adjust our approach, learning from our past failures to ensure future success.
Collapse
Affiliation(s)
- Todd E Golde
- Departments of Neuroscience and Neurology, Center for Translational Research in Neurodegenerative Disease, Evelyn F. and William L. McKnight Brain Institute, Norman Fixel Institute for Neurological Diseases, University of Florida, Gainesville, FL, USA.
| |
Collapse
|
11
|
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
|
12
|
Höglinger GU, Litvan I, Mendonca N, Wang D, Zheng H, Rendenbach-Mueller B, Lon HK, Jin Z, Fisseha N, Budur K, Gold M, Ryman D, Florian H, Ahmed A, Aiba I, Albanese A, Bertram K, Bordelon Y, Bower J, Brosch J, Claassen D, Colosimo C, Corvol JC, Cudia P, Daniele A, Defebvre L, Driver-Dunckley E, Duquette A, Eleopra R, Eusebio A, Fung V, Geldmacher D, Golbe L, Grandas F, Hall D, Hatano T, Höglinger GU, Honig L, Hui J, Kerwin D, Kikuchi A, Kimber T, Kimura T, Kumar R, Litvan I, Ljubenkov P, Lorenzl S, Ludolph A, Mari Z, McFarland N, Meissner W, Mir Rivera P, Mochizuki H, Morgan J, Munhoz R, Nishikawa N, O`Sullivan J, Oeda T, Oizumi H, Onodera O, Ory-Magne F, Peckham E, Postuma R, Quattrone A, Quinn J, Ruggieri S, Sarna J, Schulz PE, Slevin J, Tagliati M, Wile D, Wszolek Z, Xie T, Zesiewicz T. Safety and efficacy of tilavonemab in progressive supranuclear palsy: a phase 2, randomised, placebo-controlled trial. Lancet Neurol 2021; 20:182-192. [DOI: 10.1016/s1474-4422(20)30489-0] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Revised: 10/07/2020] [Accepted: 12/11/2020] [Indexed: 12/13/2022]
|
13
|
Li S, Yi Y, Cui K, Zhang Y, Chen Y, Han D, Sun L, Zhang X, Chen F, Zhang Y, Yang Y. A Single-Chain Variable Fragment Antibody Inhibits Aggregation of Phosphorylated Tau and Ameliorates Tau Toxicity in vitro and in vivo. J Alzheimers Dis 2021; 79:1613-1629. [PMID: 33459708 DOI: 10.3233/jad-191266] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
BACKGROUND Alzheimer's disease (AD) is a common cause of dementia among elderly people. Hyperphosphorylation and aggregation of tau correlates with the clinical progression of AD; therefore, therapies targeting the aggregation of tau may have potential applications for anti-AD drug development. Several inhibitors of tau aggregation, including small molecules and antibodies, have been found to decrease the aggregation of tau and the corresponding pathology. OBJECTIVE To screen one kind of single-chain variable fragment (scFv) antibody which could inhibit the aggregation of tau and ameliorate its cytotoxicity. METHODS/RESULTS Using phosphorylated tau (pTau) as an antigen, we obtained a scFv antibody via the screening of a high-capacity phage antibody library. Biochemical analysis revealed that this scFv antibody (scFv T1) had a strong ability to inhibit pTau aggregation both in dilute solutions and under conditions of macromolecular crowding. ScFv T1 could also depolymerize preformed pTau aggregates in vitro. Furthermore, scFv T1 was found to be able to inhibit the cytotoxicity of extracellular pTau aggregates and ameliorate tau-mediated toxicity when coexpressed with a hTauR406W mutant in the eye of transgenic Drosophila flies. CONCLUSION This scFv T1 antibody may be a potential new therapeutic agent against AD. Our methods can be used to develop novel strategies against protein aggregation for the treatment of neurodegenerative diseases.
Collapse
Affiliation(s)
- Sen Li
- Department of Biochemistry and Molecular Biology, College of Life Sciences, Beijing Normal University, Gene engineering and Biotechnology Beijing Key Laboratory, National Demonstration Center for Experimental Life Sciences & Biotechnology Education, Beijing, P. R. China
| | - Yushan Yi
- Institute of Life Sciences, Fuzhou University, Fuzhou, Fujian Province, P. R.China
| | - Ke Cui
- Department of Biochemistry and Molecular Biology, College of Life Sciences, Beijing Normal University, Gene engineering and Biotechnology Beijing Key Laboratory, National Demonstration Center for Experimental Life Sciences & Biotechnology Education, Beijing, P. R. China
| | - Yanqiu Zhang
- Department of Biochemistry and Molecular Biology, College of Life Sciences, Beijing Normal University, Gene engineering and Biotechnology Beijing Key Laboratory, National Demonstration Center for Experimental Life Sciences & Biotechnology Education, Beijing, P. R. China
| | - Yange Chen
- Department of Biochemistry and Molecular Biology, College of Life Sciences, Beijing Normal University, Gene engineering and Biotechnology Beijing Key Laboratory, National Demonstration Center for Experimental Life Sciences & Biotechnology Education, Beijing, P. R. China
| | - Dou Han
- Department of Biochemistry and Molecular Biology, College of Life Sciences, Beijing Normal University, Gene engineering and Biotechnology Beijing Key Laboratory, National Demonstration Center for Experimental Life Sciences & Biotechnology Education, Beijing, P. R. China
| | - Ling Sun
- Institute of Life Sciences, Fuzhou University, Fuzhou, Fujian Province, P. R.China
| | - Xiaohui Zhang
- Institute of Life Sciences, Fuzhou University, Fuzhou, Fujian Province, P. R.China
| | - Fei Chen
- Institute of Life Sciences, Fuzhou University, Fuzhou, Fujian Province, P. R.China
| | - Yixin Zhang
- B CUBE Center for Molecular Bioengineering, Technische Universität Dresden, Dresden, Germany
| | - Yufeng Yang
- Institute of Life Sciences, Fuzhou University, Fuzhou, Fujian Province, P. R.China
| |
Collapse
|
14
|
Goodwin MS, Sinyavskaya O, Burg F, O'Neal V, Ceballos-Diaz C, Cruz PE, Lewis J, Giasson BI, Davies P, Golde TE, Levites Y. Anti-tau scFvs Targeted to the Cytoplasm or Secretory Pathway Variably Modify Pathology and Neurodegenerative Phenotypes. Mol Ther 2021; 29:859-872. [PMID: 33128896 PMCID: PMC7854277 DOI: 10.1016/j.ymthe.2020.10.007] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2020] [Revised: 09/21/2020] [Accepted: 10/08/2020] [Indexed: 11/03/2022] Open
Abstract
Immunotherapies designed to treat neurodegenerative tauopathies that primarily engage extracellular tau may have limited efficacy as tau is primarily intracellular. We generated tau-targeting single-chain variable fragments (scFvs) and intrabodies (iBs) from the phosphorylated tau-specific antibodies CP13 and PHF1 and the pan-tau antibody Tau5. Recombinant adeno-associated virus (rAAV) was utilized to express these antibody fragments in homozygous JNPL3 P301L tau mice. Two iBs (CP13i, PHF1i) and one scFv (PHF1s) abrogated tau pathology and delayed time to severe hindlimb paralysis. In a second tauopathy model (rTg4510), CP13i and PHF1i reduced tau pathology, but cognate scFvs did not. These data demonstrate that (1) disease-modifying efficacy does not require antibody effector functions, (2) the intracellular targeting of tau with phosphorylated tau-specific iBs is more effective than extracellular targeting with the scFvs, and (3) robust effects on tau pathology before neurodegeneration only resulted in modest disease modification as assessed by delay of severe motor phenotype.
Collapse
Affiliation(s)
- Marshall S Goodwin
- Department of Neuroscience, College of Medicine, University of Florida, Gainesville, FL 32610, USA; Center for Translational Research in Neurodegenerative Disease, College of Medicine, University of Florida, Gainesville, FL 32610, USA; McKnight Brain Institute, College of Medicine, University of Florida, Gainesville, FL 32610, USA
| | - Olga Sinyavskaya
- Department of Neuroscience, College of Medicine, University of Florida, Gainesville, FL 32610, USA; Center for Translational Research in Neurodegenerative Disease, College of Medicine, University of Florida, Gainesville, FL 32610, USA
| | - Franklin Burg
- Department of Neuroscience, College of Medicine, University of Florida, Gainesville, FL 32610, USA; Center for Translational Research in Neurodegenerative Disease, College of Medicine, University of Florida, Gainesville, FL 32610, USA
| | - Veronica O'Neal
- Department of Neuroscience, College of Medicine, University of Florida, Gainesville, FL 32610, USA; Center for Translational Research in Neurodegenerative Disease, College of Medicine, University of Florida, Gainesville, FL 32610, USA
| | - Carolina Ceballos-Diaz
- Department of Neuroscience, College of Medicine, University of Florida, Gainesville, FL 32610, USA; Center for Translational Research in Neurodegenerative Disease, College of Medicine, University of Florida, Gainesville, FL 32610, USA
| | - Pedro E Cruz
- Department of Neuroscience, College of Medicine, University of Florida, Gainesville, FL 32610, USA; Center for Translational Research in Neurodegenerative Disease, College of Medicine, University of Florida, Gainesville, FL 32610, USA
| | - Jada Lewis
- Department of Neuroscience, College of Medicine, University of Florida, Gainesville, FL 32610, USA; Center for Translational Research in Neurodegenerative Disease, College of Medicine, University of Florida, Gainesville, FL 32610, USA; McKnight Brain Institute, College of Medicine, University of Florida, Gainesville, FL 32610, USA
| | - Benoit I Giasson
- Department of Neuroscience, College of Medicine, University of Florida, Gainesville, FL 32610, USA; Center for Translational Research in Neurodegenerative Disease, College of Medicine, University of Florida, Gainesville, FL 32610, USA; McKnight Brain Institute, College of Medicine, University of Florida, Gainesville, FL 32610, USA
| | - Peter Davies
- Litwin-Zucker Center for Research in Alzheimer's Disease, Feinstein Institute for Medical Research, North Shore/LIJ Health System, Manhasset, NY, USA
| | - Todd E Golde
- Department of Neuroscience, College of Medicine, University of Florida, Gainesville, FL 32610, USA; Center for Translational Research in Neurodegenerative Disease, College of Medicine, University of Florida, Gainesville, FL 32610, USA; McKnight Brain Institute, College of Medicine, University of Florida, Gainesville, FL 32610, USA.
| | - Yona Levites
- Department of Neuroscience, College of Medicine, University of Florida, Gainesville, FL 32610, USA; Center for Translational Research in Neurodegenerative Disease, College of Medicine, University of Florida, Gainesville, FL 32610, USA; McKnight Brain Institute, College of Medicine, University of Florida, Gainesville, FL 32610, USA.
| |
Collapse
|
15
|
Lennon MJ, Rigney G, Raymont V, Sachdev P. Genetic Therapies for Alzheimer's Disease: A Scoping Review. J Alzheimers Dis 2021; 84:491-504. [PMID: 34569966 DOI: 10.3233/jad-215145] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Effective, disease modifying therapies for Alzheimer's disease (AD) remain a quandary, following a panoply of expensive failures in human clinical trials. Given the stagnation in therapeutics, alternative approaches are needed. Recent successes of genetic therapies in other neurodegenerative diseases may highlight the way forward. This scoping review explores suggested targets of genetic therapy in AD, with a focus on vector-based approaches in pre-clinical and clinical trials. Putative targets of genetic therapies tested in pre-clinical trials include amyloid pathway intermediates and enzymes modulation, tau protein downregulation, APOE4 downregulation and APOE2 upregulation, neurotrophin expression (nerve growth factor (NGF) and brain-derived neurotrophic factor), and inflammatory cytokine alteration, among several other approaches. There have been three completed human clinical trials for genetic therapy in AD patients, all of which upregulated NGF in AD patients, showing some mixed evidence of benefit. Several impediments remain to be surpassed before genetic therapies can be successfully applied to AD, including the challenge of delivering monogenic genetic therapies for complex polygenic disorders, risks in the dominant delivery method (intracranial injection), stability of genetic therapies in vivo, poor translatability of pre-clinical AD models, and the expense of genetic therapy production. Genetic therapies represent an exciting opportunity within the world of AD therapeutics, but clinical applications likely remain a long term, rather than short term, possibility.
Collapse
Affiliation(s)
- Matthew J Lennon
- Department of Physiology, Anatomy and Genetics, Sherrington Building, University of Oxford, Oxford, UK
- Centre for Healthy Brain Ageing (CHeBA), School of Psychiatry, Faculty of Medicine, University of New South Wales, Sydney, NSW, Australia
| | - Grant Rigney
- Department of Psychiatry, University of Oxford, Oxford, UK
| | | | - Perminder Sachdev
- Centre for Healthy Brain Ageing (CHeBA), School of Psychiatry, Faculty of Medicine, University of New South Wales, Sydney, NSW, Australia
- Neuropsychiatric Institute, Prince of Wales Hospital, Sydney, NSW, Australia
| |
Collapse
|
16
|
Antibody Fragments as Tools for Elucidating Structure-Toxicity Relationships and for Diagnostic/Therapeutic Targeting of Neurotoxic Amyloid Oligomers. Int J Mol Sci 2020; 21:ijms21238920. [PMID: 33255488 PMCID: PMC7727795 DOI: 10.3390/ijms21238920] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2020] [Revised: 10/01/2020] [Accepted: 10/01/2020] [Indexed: 02/06/2023] Open
Abstract
The accumulation of amyloid protein aggregates in tissues is the basis for the onset of diseases known as amyloidoses. Intriguingly, many amyloidoses impact the central nervous system (CNS) and usually are devastating diseases. It is increasingly apparent that neurotoxic soluble oligomers formed by amyloidogenic proteins are the primary molecular drivers of these diseases, making them lucrative diagnostic and therapeutic targets. One promising diagnostic/therapeutic strategy has been the development of antibody fragments against amyloid oligomers. Antibody fragments, such as fragment antigen-binding (Fab), scFv (single chain variable fragments), and VHH (heavy chain variable domain or single-domain antibodies) are an alternative to full-length IgGs as diagnostics and therapeutics for a variety of diseases, mainly because of their increased tissue penetration (lower MW compared to IgG), decreased inflammatory potential (lack of Fc domain), and facile production (low structural complexity). Furthermore, through the use of in vitro-based ligand selection, it has been possible to identify antibody fragments presenting marked conformational selectivity. In this review, we summarize significant reports on antibody fragments selective for oligomers associated with prevalent CNS amyloidoses. We discuss promising results obtained using antibody fragments as both diagnostic and therapeutic agents against these diseases. In addition, the use of antibody fragments, particularly scFv and VHH, in the isolation of unique oligomeric assemblies is discussed as a strategy to unravel conformational moieties responsible for neurotoxicity. We envision that advances in this field may lead to the development of novel oligomer-selective antibody fragments with superior selectivity and, hopefully, good clinical outcomes.
Collapse
|
17
|
Ng PY, Chang IS, Koh RY, Chye SM. Recent advances in tau-directed immunotherapy against Alzheimer's disease: an overview of pre-clinical and clinical development. Metab Brain Dis 2020; 35:1049-1066. [PMID: 32632666 DOI: 10.1007/s11011-020-00591-6] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/11/2019] [Accepted: 06/23/2020] [Indexed: 02/01/2023]
Abstract
Alzheimer's disease (AD) has been a worldwide concern for many years now. This is due to the fact that AD is an irreversible and progressive neurodegenerative disease that affects quality of life. Failure of some Phase II/III clinical trials in AD targeting accumulation of β-amyloid in the brain has led to an increase in interest in studying alternative treatments against tubulin-associated unit (Tau) pathology. These alternative treatments include active and passive immunisation. Based on numerous studies, Tau is reported as a potential immunotherapeutic target for tauopathy-related diseases including AD. Accumulation and aggregation of hyperphosphorylated Tau as neuropil threads and neurofibrillary tangles (NFT) are pathological hallmarks of AD. Both active and passive immunisation targeting Tau protein have shown the capabilities to decrease or prevent Tau pathology and improve either motor or cognitive impairment in various animal models. In this review, we summarise recent advances in active and passive immunisation targeting pathological Tau protein, and will discuss with data obtained from both animal and human trials. Together, we give a brief overview about problems being encountered in these immunotherapies.
Collapse
Affiliation(s)
- Pei Ying Ng
- School of Postgraduate, International Medical University, 57000, Kuala Lumpur, Malaysia
| | - I Shuen Chang
- School of Health Science, Division of Biomedical Science and Biotechnology, International Medical University, No. 126, Jalan Jalil Perkasa 19, Bukit Jalil, 57000, Kuala Lumpur, Malaysia
| | - Rhun Yian Koh
- School of Health Science, Division of Biomedical Science and Biotechnology, International Medical University, No. 126, Jalan Jalil Perkasa 19, Bukit Jalil, 57000, Kuala Lumpur, Malaysia
| | - Soi Moi Chye
- School of Health Science, Division of Biomedical Science and Biotechnology, International Medical University, No. 126, Jalan Jalil Perkasa 19, Bukit Jalil, 57000, Kuala Lumpur, Malaysia.
| |
Collapse
|
18
|
Sandusky-Beltran LA, Sigurdsson EM. Tau immunotherapies: Lessons learned, current status and future considerations. Neuropharmacology 2020; 175:108104. [PMID: 32360477 PMCID: PMC7492435 DOI: 10.1016/j.neuropharm.2020.108104] [Citation(s) in RCA: 49] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2019] [Revised: 03/10/2020] [Accepted: 04/06/2020] [Indexed: 12/15/2022]
Abstract
The majority of clinical trials targeting the tau protein in Alzheimer's disease and other tauopathies are tau immunotherapies. Because tau pathology correlates better with the degree of dementia than amyloid-β lesions, targeting tau is likely to be more effective in improving cognition than clearing amyloid-β in Alzheimer's disease. However, the development of tau therapies is in many ways more complex than for amyloid-β therapies as briefly outlined in this review. Most of the trials are on humanized antibodies, which may have very different properties than the original mouse antibodies. The impact of these differences are to a large extent unknown, can be difficult to decipher, and may not always be properly considered. Furthermore, the ideal antibody properties for efficacy are not well established and can depend on several factors. However, considering the varied approaches in clinical trials, there is a general optimism that at least some of these trials may provide functional benefits to patients suffering of various tauopathies. This article is part of the special issue entitled 'The Quest for Disease-Modifying Therapies for Neurodegenerative Disorders'.
Collapse
Affiliation(s)
- L A Sandusky-Beltran
- Department of Neuroscience and Physiology, New York University School of Medicine, New York, NY, 10016, USA; Department of Neuroscience Institute, New York University School of Medicine, New York, NY, 10016, USA
| | - E M Sigurdsson
- Department of Neuroscience and Physiology, New York University School of Medicine, New York, NY, 10016, USA; Department of Psychiatry, New York University School of Medicine, New York, NY, 10016, USA; Department of Neuroscience Institute, New York University School of Medicine, New York, NY, 10016, USA.
| |
Collapse
|
19
|
Long M, Lin F, Wang X, Chen X, Liu L, Zhang H, Dong K. Adenovirus-mediated anti-AEG-1 ScFv expression driven by stathmin promoter inhibits tumor growth in cervical cancer. Cancer Cell Int 2020; 20:79. [PMID: 32190003 PMCID: PMC7068931 DOI: 10.1186/s12935-020-1159-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2019] [Accepted: 02/29/2020] [Indexed: 12/14/2022] Open
Abstract
Background Astrocyte-elevated gene-1 (AEG-1) is over-expressed in many cancer cells and has multiple key functions in tumor initiation and progression. Currently, targeted-AEG-1 siRNA is one of the most common techniques to down-regulate AEG-1 expression, but the lack of tumor specificity and available delivery system make it difficult to enter clinical trials. Methods In this study, we creatively developed an adenovirus-mediated anti-AEG-1 single-chain antibody fragment (ScFv) expression system driven by a tumor specific promoter, and experimented with it in human cervical carcinoma cells to investigate the effect on tumor’s proliferation and apoptosis. Results The results showed that of HeLa and SiHa cells treated with this recombinant anti-AEG-1 ScFv adenovirus not only inhibited cell growth, but induced apoptosis both in vitro and in vivo. Furthermore, we also observed that the expressions of several apoptosis-related genes like Akt 1 and c-Myc decreased, while NF-κB (p65) and cleaved caspase 3 increased on protein levels in vivo. Conclusion We concluded that stathmin promoter-driving anti-AEG-1 ScFv adenoviral system may be a breakthrough for its dual-specificity, and serve as an adjuvant tumor specific therapy method in the treatment for human cervical cancers.
Collapse
Affiliation(s)
- Min Long
- Department of Medical Laboratory, Tangdu Hospital, Airforce Military Medical University, Xinsi Road, Xi'an, 710038 Shaanxi China
| | - Fang Lin
- Department of Medical Laboratory, Tangdu Hospital, Airforce Military Medical University, Xinsi Road, Xi'an, 710038 Shaanxi China
| | - Xi Wang
- Department of Medical Laboratory, Tangdu Hospital, Airforce Military Medical University, Xinsi Road, Xi'an, 710038 Shaanxi China
| | - Xi Chen
- Department of Medical Laboratory, Tangdu Hospital, Airforce Military Medical University, Xinsi Road, Xi'an, 710038 Shaanxi China
| | - Li Liu
- Department of Medical Laboratory, Tangdu Hospital, Airforce Military Medical University, Xinsi Road, Xi'an, 710038 Shaanxi China
| | - Huizhong Zhang
- Department of Medical Laboratory, Tangdu Hospital, Airforce Military Medical University, Xinsi Road, Xi'an, 710038 Shaanxi China
| | - Ke Dong
- Department of Medical Laboratory, Tangdu Hospital, Airforce Military Medical University, Xinsi Road, Xi'an, 710038 Shaanxi China
| |
Collapse
|
20
|
Krishnaswamy S, Huang HW, Marchal IS, Ryoo HD, Sigurdsson EM. Neuronally expressed anti-tau scFv prevents tauopathy-induced phenotypes in Drosophila models. Neurobiol Dis 2020; 137:104770. [PMID: 31982516 DOI: 10.1016/j.nbd.2020.104770] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2019] [Revised: 01/10/2020] [Accepted: 01/23/2020] [Indexed: 01/20/2023] Open
Abstract
We have derived single-chain variable fragments (scFv) from tau antibody hybridomas and previously shown their promise as imaging diagnostic agents. Here, we examined the therapeutic potential of anti-tau scFv in transgenic Drosophila models that express in neurons wild-type (WT) human tau (htau) or the human tauopathy mutation R406W. scFv expressing flies were crossed with the tauopathy flies and analyzed. Overall, the survival curves differed significantly (p < .0001). Control flies not expressing htau survived the longest, whereas R406W expressing flies had the shortest lifespan, which was greatly prolonged by co-expressing the anti-tau scFv (p < .0001). Likewise, htau WT expressing flies had a moderately short lifespan, which was prolonged by co-expressing the anti-tau scFv (p < .01). In addition, the htau expression impaired wing expansion after eclosion (p < .0001), and caused progressive abdomen expansion (p < .0001). These features were more severe in htau R406W flies than in htau WT flies. Importantly, both phenotypes were prevented by co-expression of the anti-tau scFv (p < .01-0.0001). Lastly, brain analyses revealed scFv-mediated tau clearance (p < .05-0.01), and its prevention of tau-mediated neurotoxicity (p < .05-0.001). In summary, these findings support the therapeutic potential of an anti-tau scFv, including as gene therapies, and the use of Drosophila models for such screening.
Collapse
Affiliation(s)
- Senthilkumar Krishnaswamy
- Department of Neuroscience and Physiology, New York University School of Medicine, New York, NY 10016, United States of America
| | - Huai-Wei Huang
- Department of Cell Biology, New York University School of Medicine, New York, NY 10016, United States of America
| | - Isabella S Marchal
- Department of Neuroscience and Physiology, New York University School of Medicine, New York, NY 10016, United States of America
| | - Hyung Don Ryoo
- Department of Cell Biology, New York University School of Medicine, New York, NY 10016, United States of America.
| | - Einar M Sigurdsson
- Department of Neuroscience and Physiology, New York University School of Medicine, New York, NY 10016, United States of America; Department of Psychiatry, New York University School of Medicine, New York, NY 10016, United States of America; Neuroscience Institute, New York University School of Medicine, New York, NY 10016, United States of America.
| |
Collapse
|
21
|
From the prion-like propagation hypothesis to therapeutic strategies of anti-tau immunotherapy. Acta Neuropathol 2020; 139:3-25. [PMID: 31686182 PMCID: PMC6942016 DOI: 10.1007/s00401-019-02087-9] [Citation(s) in RCA: 108] [Impact Index Per Article: 21.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2019] [Revised: 10/18/2019] [Accepted: 10/19/2019] [Indexed: 12/15/2022]
Abstract
The term “propagon” is used to define proteins that may transmit misfolding in vitro, in tissues or in organisms. Among propagons, misfolded tau is thought to be involved in the pathogenic mechanisms of various “tauopathies” that include Alzheimer's disease, progressive supranuclear palsy, and argyrophilic grain disease. Here, we review the available data in the literature and point out how the prion-like tau propagation has been extended from Alzheimer's disease to tauopathies. First, in Alzheimer’s disease, the progression of tau aggregation follows stereotypical anatomical stages which may be considered as spreading. The mechanisms of the propagation are now subject to intensive and controversial research. It has been shown that tau may be secreted in the interstitial fluid in an active manner as reflected by high and constant concentration of extracellular tau during Alzheimer’s pathology. Animal and cell models have been devised to mimic tau seeding and propagation, and despite their limitations, they have further supported to the prion-like propagation hypothesis. Finally, such new ways of thinking have led to different therapeutic strategies in anti-tau immunotherapy among tauopathies and have stimulated new clinical trials. However, it appears that the prion-like propagation hypothesis mainly relies on data obtained in Alzheimer’s disease. From this review, it appears that further studies are needed (1) to characterize extracellular tau species, (2) to find the right pathological tau species to target, (3) to follow in vivo tau pathology by brain imaging and biomarkers and (4) to interpret current clinical trial results aimed at reducing the progression of these pathologies. Such inputs will be essential to have a comprehensive view of these promising therapeutic strategies in tauopathies.
Collapse
|
22
|
Ising C, Venegas C, Zhang S, Scheiblich H, Schmidt SV, Vieira-Saecker A, Schwartz S, Albasset S, McManus RM, Tejera D, Griep A, Santarelli F, Brosseron F, Opitz S, Stunden J, Merten M, Kayed R, Golenbock DT, Blum D, Latz E, Buée L, Heneka MT. NLRP3 inflammasome activation drives tau pathology. Nature 2019; 575:669-673. [PMID: 31748742 DOI: 10.1038/s41586-019-1769-z] [Citation(s) in RCA: 895] [Impact Index Per Article: 149.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2019] [Accepted: 10/02/2019] [Indexed: 12/21/2022]
Abstract
Alzheimer's disease is characterized by the accumulation of amyloid-beta in plaques, aggregation of hyperphosphorylated tau in neurofibrillary tangles and neuroinflammation, together resulting in neurodegeneration and cognitive decline1. The NLRP3 inflammasome assembles inside of microglia on activation, leading to increased cleavage and activity of caspase-1 and downstream interleukin-1β release2. Although the NLRP3 inflammasome has been shown to be essential for the development and progression of amyloid-beta pathology in mice3, the precise effect on tau pathology remains unknown. Here we show that loss of NLRP3 inflammasome function reduced tau hyperphosphorylation and aggregation by regulating tau kinases and phosphatases. Tau activated the NLRP3 inflammasome and intracerebral injection of fibrillar amyloid-beta-containing brain homogenates induced tau pathology in an NLRP3-dependent manner. These data identify an important role of microglia and NLRP3 inflammasome activation in the pathogenesis of tauopathies and support the amyloid-cascade hypothesis in Alzheimer's disease, demonstrating that neurofibrillary tangles develop downstream of amyloid-beta-induced microglial activation.
Collapse
Affiliation(s)
- Christina Ising
- Department of Neurodegenerative Diseases and Geriatric Psychiatry, University Hospital of Bonn, Bonn, Germany.,German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
| | - Carmen Venegas
- Department of Neurodegenerative Diseases and Geriatric Psychiatry, University Hospital of Bonn, Bonn, Germany
| | - Shuangshuang Zhang
- Department of Neurodegenerative Diseases and Geriatric Psychiatry, University Hospital of Bonn, Bonn, Germany.,German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
| | - Hannah Scheiblich
- Department of Neurodegenerative Diseases and Geriatric Psychiatry, University Hospital of Bonn, Bonn, Germany.,German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
| | - Susanne V Schmidt
- Institute of Innate Immunity, University Hospital Bonn, Bonn, Germany
| | - Ana Vieira-Saecker
- Department of Neurodegenerative Diseases and Geriatric Psychiatry, University Hospital of Bonn, Bonn, Germany.,German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
| | - Stephanie Schwartz
- Department of Neurodegenerative Diseases and Geriatric Psychiatry, University Hospital of Bonn, Bonn, Germany.,German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
| | - Shadi Albasset
- Department of Neurodegenerative Diseases and Geriatric Psychiatry, University Hospital of Bonn, Bonn, Germany.,German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
| | - Róisín M McManus
- Department of Neurodegenerative Diseases and Geriatric Psychiatry, University Hospital of Bonn, Bonn, Germany.,German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
| | - Dario Tejera
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
| | - Angelika Griep
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
| | | | | | - Sabine Opitz
- Department of Neurodegenerative Diseases and Geriatric Psychiatry, University Hospital of Bonn, Bonn, Germany.,German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
| | | | - Maximilian Merten
- Department of Neurodegenerative Diseases and Geriatric Psychiatry, University Hospital of Bonn, Bonn, Germany
| | - Rakez Kayed
- Mitchell Center for Neurodegenerative Diseases and Departments of Neurology, Neuroscience and Cell Biology, University of Texas Medical Branch, Galveston, TX, USA
| | - Douglas T Golenbock
- Divison of Infectious Diseases and Immunology, University of Massachusetts Medical School, Worcester, MA, USA
| | - David Blum
- University of Lille, Inserm, CHU-Lille, UMR-S 1172, "Alzheimer & Tauopathies", Labex DISTALZ, Lille, France
| | - Eicke Latz
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany.,Institute of Innate Immunity, University Hospital Bonn, Bonn, Germany.,Divison of Infectious Diseases and Immunology, University of Massachusetts Medical School, Worcester, MA, USA
| | - Luc Buée
- University of Lille, Inserm, CHU-Lille, UMR-S 1172, "Alzheimer & Tauopathies", Labex DISTALZ, Lille, France
| | - Michael T Heneka
- Department of Neurodegenerative Diseases and Geriatric Psychiatry, University Hospital of Bonn, Bonn, Germany. .,German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany. .,Divison of Infectious Diseases and Immunology, University of Massachusetts Medical School, Worcester, MA, USA.
| |
Collapse
|
23
|
Gallardo G, Wong CH, Ricardez SM, Mann CN, Lin KH, Leyns CEG, Jiang H, Holtzman DM. Targeting tauopathy with engineered tau-degrading intrabodies. Mol Neurodegener 2019; 14:38. [PMID: 31640765 PMCID: PMC6805661 DOI: 10.1186/s13024-019-0340-6] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2019] [Accepted: 10/09/2019] [Indexed: 12/21/2022] Open
Abstract
Background The accumulation of pathological tau is the main component of neurofibrillary tangles and other tau aggregates in several neurodegenerative diseases, referred to as tauopathies. Recently, immunotherapeutic approaches targeting tau have been demonstrated to be beneficial in decreasing tauopathy in animal models. We previously found that passive immunotherapy with anti-tau antibody to human tau or expression of an anti-tau secreted single-chain variable fragment (scFv) in the central nervous system of a mouse model of tauopathy decreased but did not remove all tau-associated pathology. Although these and other studies demonstrate that conventional immunotherapeutic approaches targeting tau can influence tau pathogenesis, the majority of pathological tau remains in the cytosol of cells, not typically accessible to an extracellular antibody. Therefore, we reasoned targeting intracellular tau might be more efficacious in preventing or decreasing tauopathy. Methods By utilizing our anti-tau scFv, we generated anti-tau intrabodies for the expression in the cytosol of neurons. To enhance the degradation capacity of conventional intrabodies, we engineered chimeric anti-tau intrabodies fused to ubiquitin harboring distinct mutations that shuttle intracellular tau for either the proteasome or lysosomal mediated degradation. To evaluate the efficacy in delaying or eliminating tauopathy, we expressed our tau degrading intrabodies or controls in human tau transgenic mice by adeno-associated virus prior to overt tau pathology and after tau deposition. Results Our results demonstrate, the expression of chimeric anti-tau intrabodies significantly reduce tau protein levels in primary neuronal cultures expression human tau relative to a non-modified anti-tau intrabody. We found the expression of engineered tau-degrading intrabodies destined for proteasomal-mediated degradation are more effective in delaying or eliminating tauopathy than a conventional intrabody in aged human tau transgenic mice. Conclusion This study, harnesses the strength of intrabodies that are amendable for targeting specific domains or modifications with the cell-intrinsic mechanisms that regulate protein degradation providing a new immunotherapeutic approach with potentially improved efficacy.
Collapse
Affiliation(s)
- Gilbert Gallardo
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, 63110, USA. .,Hope Center for Neurological Disorders, Washington University, Campus Box 8111, 660 S. Euclid Avenue, St. Louis, MO, 63110, USA.
| | - Connie H Wong
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, 63110, USA.,Hope Center for Neurological Disorders, Washington University, Campus Box 8111, 660 S. Euclid Avenue, St. Louis, MO, 63110, USA.,Department of Molecular Genetics and Center for Translational Neurodegeneration Research, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Sara M Ricardez
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, 63110, USA.,Hope Center for Neurological Disorders, Washington University, Campus Box 8111, 660 S. Euclid Avenue, St. Louis, MO, 63110, USA
| | - Carolyn N Mann
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, 63110, USA.,Hope Center for Neurological Disorders, Washington University, Campus Box 8111, 660 S. Euclid Avenue, St. Louis, MO, 63110, USA
| | - Kent H Lin
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, 63110, USA.,Hope Center for Neurological Disorders, Washington University, Campus Box 8111, 660 S. Euclid Avenue, St. Louis, MO, 63110, USA
| | - Cheryl E G Leyns
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, 63110, USA.,Hope Center for Neurological Disorders, Washington University, Campus Box 8111, 660 S. Euclid Avenue, St. Louis, MO, 63110, USA.,Charles F. and Joanne Knight Alzheimer's Disease Research Center, Washington University, St. Louis, MO, USA.,Neuroscience Discovery, Merck Research Laboratories, Boston, MA, 02115, USA
| | - Hong Jiang
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, 63110, USA.,Hope Center for Neurological Disorders, Washington University, Campus Box 8111, 660 S. Euclid Avenue, St. Louis, MO, 63110, USA.,Charles F. and Joanne Knight Alzheimer's Disease Research Center, Washington University, St. Louis, MO, USA
| | - David M Holtzman
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, 63110, USA. .,Hope Center for Neurological Disorders, Washington University, Campus Box 8111, 660 S. Euclid Avenue, St. Louis, MO, 63110, USA. .,Charles F. and Joanne Knight Alzheimer's Disease Research Center, Washington University, St. Louis, MO, USA.
| |
Collapse
|
24
|
Dupré E, Danis C, Arrial A, Hanoulle X, Homa M, Cantrelle FX, Merzougui H, Colin M, Rain JC, Buée L, Landrieu I. Single Domain Antibody Fragments as New Tools for the Detection of Neuronal Tau Protein in Cells and in Mice Studies. ACS Chem Neurosci 2019; 10:3997-4006. [PMID: 31380615 DOI: 10.1021/acschemneuro.9b00217] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Tau is a neuronal protein linked to pathologies called tauopathies, including Alzheimer's disease. In Alzheimer's disease, tau aggregates into filaments, leading to the observation of intraneuronal fibrillary tangles. Molecular mechanisms resulting in tau aggregation and in tau pathology spreading through the brain regions are still not fully understood. New tools are thus needed to decipher tau pathways involved in the diseases. In this context, a family of novel single domain antibody fragments, or VHHs, directed against tau were generated and characterized. Among the selected VHHs obtained from screening of a synthetic library, a family of six VHHs shared the same CDR3 recognition loop and recognized the same epitope, located in the C-terminal domain of tau. Affinity parameters characterizing the tau/VHHs interaction were next evaluated using surface plasmon resonance spectroscopy. The equilibrium constants KD were in the micromolar range, but despite conservation of the CDR3 loop sequence, a range of affinities was observed for this VHH family. One of these VHHs, named F8-2, was additionally shown to bind tau upon expression in a neuronal cell line model. Optimization of VHH F8-2 by yeast two-hybrid allowed the generation of an optimized VHH family characterized by lower KD than that of the F8-2 wild-type counterpart, and recognizing the same epitope. The optimized VHHs can also be used as antibodies for detecting tau in transgenic mice brain tissues. These results validate the use of these VHHs for in vitro studies, but also their potential for in-cell expression and assays in mouse models, to explore the mechanisms underlying tau physiopathology.
Collapse
Affiliation(s)
- Elian Dupré
- Univ. Lille, CNRS, UMR 8576 - UGSF - Unité de Glycobiologie Structurale et Fonctionnelle, F-59000 Lille, France
| | - Clément Danis
- Univ. Lille, CNRS, UMR 8576 - UGSF - Unité de Glycobiologie Structurale et Fonctionnelle, F-59000 Lille, France
- Univ. Lille, Inserm, CHU Lille, UMR-S 1172 - JPArc - Centre de Recherche Jean-Pierre AUBERT Neurosciences et Cancer, F-59000 Lille, France
| | | | - Xavier Hanoulle
- Univ. Lille, CNRS, UMR 8576 - UGSF - Unité de Glycobiologie Structurale et Fonctionnelle, F-59000 Lille, France
| | - Mégane Homa
- Univ. Lille, Inserm, CHU Lille, UMR-S 1172 - JPArc - Centre de Recherche Jean-Pierre AUBERT Neurosciences et Cancer, F-59000 Lille, France
| | - François-Xavier Cantrelle
- Univ. Lille, CNRS, UMR 8576 - UGSF - Unité de Glycobiologie Structurale et Fonctionnelle, F-59000 Lille, France
| | - Hamida Merzougui
- Univ. Lille, CNRS, UMR 8576 - UGSF - Unité de Glycobiologie Structurale et Fonctionnelle, F-59000 Lille, France
| | - Morvane Colin
- Univ. Lille, Inserm, CHU Lille, UMR-S 1172 - JPArc - Centre de Recherche Jean-Pierre AUBERT Neurosciences et Cancer, F-59000 Lille, France
| | | | - Luc Buée
- Univ. Lille, Inserm, CHU Lille, UMR-S 1172 - JPArc - Centre de Recherche Jean-Pierre AUBERT Neurosciences et Cancer, F-59000 Lille, France
| | - Isabelle Landrieu
- Univ. Lille, CNRS, UMR 8576 - UGSF - Unité de Glycobiologie Structurale et Fonctionnelle, F-59000 Lille, France
| |
Collapse
|
25
|
Vogels T, Murgoci AN, Hromádka T. Intersection of pathological tau and microglia at the synapse. Acta Neuropathol Commun 2019; 7:109. [PMID: 31277708 PMCID: PMC6612163 DOI: 10.1186/s40478-019-0754-y] [Citation(s) in RCA: 121] [Impact Index Per Article: 20.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2019] [Accepted: 06/19/2019] [Indexed: 02/07/2023] Open
Abstract
Tauopathies are a heterogenous class of diseases characterized by cellular accumulation of aggregated tau and include diseases such as Alzheimer’s disease (AD), progressive supranuclear palsy and chronic traumatic encephalopathy. Tau pathology is strongly linked to neurodegeneration and clinical symptoms in tauopathy patients. Furthermore, synapse loss is an early pathological event in tauopathies and is the strongest correlate of cognitive decline. Tau pathology is additionally associated with chronic neuroinflammatory processes, such as reactive microglia, astrocytes, and increased levels of pro-inflammatory molecules (e.g. complement proteins, cytokines). Recent studies show that as the principal immune cells of the brain, microglia play a particularly important role in the initiation and progression of tau pathology and associated neurodegeneration. Furthermore, AD risk genes such as Triggering receptor expressed on myeloid cells 2 (TREM2) and Apolipoprotein E (APOE) are enriched in the innate immune system and modulate the neuroinflammatory response of microglia to tau pathology. Microglia can play an active role in synaptic dysfunction by abnormally phagocytosing synaptic compartments of neurons with tau pathology. Furthermore, microglia are involved in synaptic spreading of tau – a process which is thought to underlie the progressive nature of tau pathology propagation through the brain. Spreading of pathological tau is also the predominant target for tau-based immunotherapy. Active tau vaccines, therapeutic tau antibodies and other approaches targeting the immune system are actively explored as treatment options for AD and other tauopathies. This review describes the role of microglia in the pathobiology of tauopathies and the mechanism of action of potential therapeutics targeting the immune system in tauopathies.
Collapse
|
26
|
Nisbet RM, Götz J. Amyloid-β and Tau in Alzheimer's Disease: Novel Pathomechanisms and Non-Pharmacological Treatment Strategies. J Alzheimers Dis 2019; 64:S517-S527. [PMID: 29562514 DOI: 10.3233/jad-179907] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Accumulation of the peptide amyloid-β (Aβ) and the protein tau in Alzheimer's disease (AD) brains is a gradual process that involves the post-translational modification and assembly of monomeric forms into larger structures that eventually form fibrillar inclusions. This process is thought to both drive and initiate AD. However, why the axonally enriched tau in the course of AD accumulates in the somatodendritic domain is not fully understood. We discuss new data that provide a possible explanation that involves de novo protein synthesis, induced by Aβ and mediated through the kinase Fyn. We further discuss how in a pathological state, tau, being a scaffolding protein, impairs nuclear and mitochondrial functions and reduces action potential generation at the axon initial segment. Pathological tau can further be packaged into exosomes, released by one neuron and taken up by another, contributing to its pathogenicity. We also present our new work that suggests ultrasound as a new treatment modality to clear pathological Aβ and tau. We put this work into perspective, discussing current vaccination strategies and improved brain delivery methods involving antibody engineering and viral approaches. We propose that rather than reducing post-translational modifications of tau, its levels and de novo synthesis need to be reduced. We anticipate a surge in combinatorial strategies, simultaneously targeting multiple pathologies, and an improved drug delivery to the brain facilitated by emerging technologies such as ultrasound.
Collapse
Affiliation(s)
- Rebecca M Nisbet
- Clem Jones Centre for Ageing Dementia Research, Queensland Brain Institute, The University of Queensland, Brisbane (St Lucia Campus), QLD, Australia
| | - Jürgen Götz
- Clem Jones Centre for Ageing Dementia Research, Queensland Brain Institute, The University of Queensland, Brisbane (St Lucia Campus), QLD, Australia
| |
Collapse
|
27
|
Ittner LM, Klugmann M, Ke YD. Adeno-associated virus-based Alzheimer's disease mouse models and potential new therapeutic avenues. Br J Pharmacol 2019; 176:3649-3665. [PMID: 30817847 DOI: 10.1111/bph.14637] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2018] [Revised: 12/23/2018] [Accepted: 02/15/2019] [Indexed: 12/22/2022] Open
Abstract
Alzheimer's disease (AD) is a highly prevalent neurodegenerative condition that presents with cognitive decline. The current understanding of underlying disease mechanisms remains incomplete. Genetically modified mouse models have been instrumental in deciphering pathomechanisms in AD. While these models were typically generated by classical transgenesis and genome editing, the use of adeno-associated viruses (AAVs) to model and investigate AD in mice, as well as to develop novel gene-therapy approaches, is emerging. Here, we reviewed literature that used AAVs to study and model AD and discuss potential gene therapy strategies. LINKED ARTICLES: This article is part of a themed section on Therapeutics for Dementia and Alzheimer's Disease: New Directions for Precision Medicine. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v176.18/issuetoc.
Collapse
Affiliation(s)
- Lars M Ittner
- Dementia Research Centre and Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Macquarie University, Sydney, NSW, Australia.,School of Medical Sciences, University of New South Wales, Sydney, NSW, Australia
| | - Matthias Klugmann
- School of Medical Sciences, University of New South Wales, Sydney, NSW, Australia
| | - Yazi D Ke
- Dementia Research Centre and Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Macquarie University, Sydney, NSW, Australia.,School of Medical Sciences, University of New South Wales, Sydney, NSW, Australia
| |
Collapse
|
28
|
Regional vulnerability and spreading of hyperphosphorylated tau in seeded mouse brain. Neurobiol Dis 2019; 127:398-409. [PMID: 30878534 DOI: 10.1016/j.nbd.2019.03.010] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2018] [Revised: 03/01/2019] [Accepted: 03/12/2019] [Indexed: 11/20/2022] Open
Abstract
We have exploited whole brain microscopy to map the progressive deposition of hyperphosphorylated tau in intact, cleared mouse brain. We found that the three-dimensional spreading pattern of hyperphosphorylated tau in the brain of an aging Tau.P301L mouse model did not resemble that observed in AD patients. Injection of synthetic or patient-derived tau fibrils in the CA1 region resulted in a more faithful spreading pattern. Atlas-guided volumetric analysis showed a connectome-dependent spreading from the injection site and also revealed hyperphosphorylated tau deposits beyond the direct anatomical connections. In fibril-injected brains, we also detected a persistent subpopulation of rod-like and swollen microglia. Furthermore, we showed that the hyperphosphorylated tau load could be reduced by intracranial co-administration of, and to a lesser extent, by repeated systemic dosing with an antibody targeting the microtubule-binding domain of tau. Thus, the combination of targeted seeding and in toto staging of tau pathology allowed assessing regional vulnerability in a comprehensive manner, and holds potential as a preclinical drug validation tool.
Collapse
|
29
|
Li H, Liu CC, Zheng H, Huang TY. Amyloid, tau, pathogen infection and antimicrobial protection in Alzheimer's disease -conformist, nonconformist, and realistic prospects for AD pathogenesis. Transl Neurodegener 2018; 7:34. [PMID: 30603085 PMCID: PMC6306008 DOI: 10.1186/s40035-018-0139-3] [Citation(s) in RCA: 77] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2018] [Accepted: 12/02/2018] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND Alzheimer's disease (AD) is a fatal disease that threatens the quality of life of an aging population at a global scale. Various hypotheses on the etiology of AD have been developed over the years to guide efforts in search of therapeutic strategies. MAIN BODY In this review, we focus on four AD hypotheses currently relevant to AD onset: the prevailing amyloid cascade hypothesis, the well-recognized tau hypothesis, the increasingly popular pathogen (viral infection) hypothesis, and the infection-related antimicrobial protection hypothesis. In briefly reviewing the main evidence supporting each hypothesis and discussing the questions that need to be addressed, we hope to gain a better understanding of the complicated multi-layered interactions in potential causal and/or risk factors in AD pathogenesis. As a defining feature of AD, the existence of amyloid deposits is likely fundamental to AD onset but is insufficient to wholly reproduce many complexities of the disorder. A similar belief is currently also applied to hyperphosphorylated tau aggregates within neurons, where tau has been postulated to drive neurodegeneration in the presence of pre-existing Aβ plaques in the brain. Although infection of the central nerve system by pathogens such as viruses may increase AD risk, it is yet to be determined whether this phenomenon is applicable to all cases of sporadic AD and whether it is a primary trigger for AD onset. Lastly, the antimicrobial protection hypothesis provides insight into a potential physiological role for Aβ peptides, but how Aβ/microbial interactions affect AD pathogenesis during aging awaits further validation. Nevertheless, this hypothesis cautions potential adverse effects in Aβ-targeting therapies by hindering potential roles for Aβ in anti-viral protection. CONCLUSION AD is a multi-factor complex disorder, which likely requires a combinatorial therapeutic approach to successfully slow or reduce symptomatic memory decline. A better understanding of how various causal and/or risk factors affecting disease onset and progression will enhance the likelihood of conceiving effective treatment paradigms, which may involve personalized treatment strategies for individual patients at varying stages of disease progression.
Collapse
Affiliation(s)
- Hongmei Li
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL USA
| | - Chia-Chen Liu
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL USA
| | - Hui Zheng
- Huffington Center on Aging, Baylor College of Medicine, Houston, TX USA
| | - Timothy Y. Huang
- Neuroscience Initiative, Sanford Burnham Prebys Medical Discovery Institute, San Diego, CA USA
| |
Collapse
|
30
|
Kumar NN, Pizzo ME, Nehra G, Wilken-Resman B, Boroumand S, Thorne RG. Passive Immunotherapies for Central Nervous System Disorders: Current Delivery Challenges and New Approaches. Bioconjug Chem 2018; 29:3937-3966. [PMID: 30265523 PMCID: PMC7234797 DOI: 10.1021/acs.bioconjchem.8b00548] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Passive immunotherapy, i.e., the administration of exogenous antibodies that recognize a specific target antigen, has gained significant momentum as a potential treatment strategy for several central nervous system (CNS) disorders, including Alzheimer's disease, Parkinson's disease, Huntington's disease, and brain cancer, among others. Advances in antibody engineering to create therapeutic antibody fragments or antibody conjugates have introduced new strategies that may also be applied to treat CNS disorders. However, drug delivery to the CNS for antibodies and other macromolecules has thus far proven challenging, due in large part to the blood-brain barrier and blood-cerebrospinal fluid barriers that greatly restrict transport of peripherally administered molecules from the systemic circulation into the CNS. Here, we summarize the various passive immunotherapy approaches under study for the treatment of CNS disorders, with a primary focus on disease-specific and target site-specific challenges to drug delivery and new, cutting edge methods.
Collapse
Affiliation(s)
- Niyanta N. Kumar
- Pharmaceutical Sciences Division, University of
Wisconsin-Madison School of Pharmacy
| | - Michelle E. Pizzo
- Pharmaceutical Sciences Division, University of
Wisconsin-Madison School of Pharmacy
- Clinical Neuroengineering Training Program, University of
Wisconsin-Madison, Madison, Wisconsin 53705, United States
| | - Geetika Nehra
- Pharmaceutical Sciences Division, University of
Wisconsin-Madison School of Pharmacy
| | - Brynna Wilken-Resman
- Pharmaceutical Sciences Division, University of
Wisconsin-Madison School of Pharmacy
| | - Sam Boroumand
- Pharmaceutical Sciences Division, University of
Wisconsin-Madison School of Pharmacy
| | - Robert G. Thorne
- Pharmaceutical Sciences Division, University of
Wisconsin-Madison School of Pharmacy
- Clinical Neuroengineering Training Program, University of
Wisconsin-Madison, Madison, Wisconsin 53705, United States
- Neuroscience Training Program & Center for
Neuroscience, University of Wisconsin-Madison, Madison, Wisconsin 53705, United
States
- Cellular and Molecular Pathology Graduate Training Program,
University of Wisconsin-Madison, Madison, Wisconsin 53705, United States
| |
Collapse
|
31
|
Vitale F, Giliberto L, Ruiz S, Steslow K, Marambaud P, d'Abramo C. Anti-tau conformational scFv MC1 antibody efficiently reduces pathological tau species in adult JNPL3 mice. Acta Neuropathol Commun 2018; 6:82. [PMID: 30134961 PMCID: PMC6103963 DOI: 10.1186/s40478-018-0585-2] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2018] [Accepted: 08/13/2018] [Indexed: 01/07/2023] Open
Abstract
Tau, the main component of the neurofibrillary tangles (NFTs), is an attractive target for immunotherapy in Alzheimer's disease (AD) and other tauopathies. MC1/Alz50 are currently the only antibodies targeting a disease-specific conformational modification of tau. Passive immunization experiments using intra-peritoneal injections have previously shown that MC1 is effective at reducing tau pathology in the forebrain of tau transgenic JNPL3 mice. In order to reach a long-term and sustained brain delivery, and avoid multiple injection protocols, we tested the efficacy of the single-chain variable fragment of MC1 (scFv-MC1) to reduce tau pathology in the same animal model, with focus on brain regional differences. ScFv-MC1 was cloned into an AAV delivery system and was directly injected into the hippocampus of adult JNPL3 mice. Specific promoters were employed to selectively target neurons or astrocytes for scFv-MC1 expression. ScFv-MC1 was able to decrease soluble, oligomeric and insoluble tau species, in our model. The effect was evident in the cortex, hippocampus and hindbrain. The astrocytic machinery appeared more efficient than the neuronal, with significant reduction of pathology in areas distant from the site of injection. To our knowledge, this is the first evidence that an anti-tau conformational scFv antibody, delivered directly into the mouse adult brain, is able to reduce pathological tau, providing further insight into the nature of immunotherapy strategies.
Collapse
Affiliation(s)
- Francesca Vitale
- Litwin-Zucker Center for Research in Alzheimer's Disease, The Feinstein Institute for Medical Research, Northwell Health, 350 Community Drive, Manhasset, NY, 11030, USA
| | - Luca Giliberto
- Litwin-Zucker Center for Research in Alzheimer's Disease, The Feinstein Institute for Medical Research, Northwell Health, 350 Community Drive, Manhasset, NY, 11030, USA
| | - Santiago Ruiz
- Litwin-Zucker Center for Research in Alzheimer's Disease, The Feinstein Institute for Medical Research, Northwell Health, 350 Community Drive, Manhasset, NY, 11030, USA
| | - Kristen Steslow
- Litwin-Zucker Center for Research in Alzheimer's Disease, The Feinstein Institute for Medical Research, Northwell Health, 350 Community Drive, Manhasset, NY, 11030, USA
| | - Philippe Marambaud
- Litwin-Zucker Center for Research in Alzheimer's Disease, The Feinstein Institute for Medical Research, Northwell Health, 350 Community Drive, Manhasset, NY, 11030, USA
| | - Cristina d'Abramo
- Litwin-Zucker Center for Research in Alzheimer's Disease, The Feinstein Institute for Medical Research, Northwell Health, 350 Community Drive, Manhasset, NY, 11030, USA.
| |
Collapse
|
32
|
Deverman BE, Ravina BM, Bankiewicz KS, Paul SM, Sah DWY. Gene therapy for neurological disorders: progress and prospects. Nat Rev Drug Discov 2018; 17:641-659. [DOI: 10.1038/nrd.2018.110] [Citation(s) in RCA: 175] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
|
33
|
Li D, Wang L, Maziuk BF, Yao X, Wolozin B, Cho YK. Directed evolution of a picomolar-affinity, high-specificity antibody targeting phosphorylated tau. J Biol Chem 2018; 293:12081-12094. [PMID: 29899114 PMCID: PMC6078456 DOI: 10.1074/jbc.ra118.003557] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2018] [Revised: 06/12/2018] [Indexed: 01/03/2023] Open
Abstract
Antibodies are essential biochemical reagents for detecting protein post-translational modifications (PTMs) in complex samples. However, recent efforts in developing PTM-targeting antibodies have reported frequent nonspecific binding and limited affinity of such antibodies. To address these challenges, we investigated whether directed evolution could be applied to improve the affinity of a high-specificity antibody targeting phosphothreonine 231 (pThr-231) of the human microtubule-associated protein tau. On the basis of existing structural information, we hypothesized that improving antibody affinity may come at the cost of loss in specificity. To test this hypothesis, we developed a novel approach using yeast surface display to quantify the specificity of PTM-targeting antibodies. When we affinity-matured the single-chain variable antibody fragment through directed evolution, we found that its affinity can be improved >20-fold over that of the WT antibody, reaching a picomolar range. We also discovered that most of the high-affinity variants exhibit cross-reactivity toward the nonphosphorylated target site but not to the phosphorylation site with a scrambled sequence. However, systematic quantification of the specificity revealed that such a tradeoff between the affinity and specificity did not apply to all variants and led to the identification of a picomolar-affinity variant that has a matching high specificity of the original phosphotau antibody. In cell- and tissue-imaging experiments, the high-affinity variant gave significantly improved signal intensity while having no detectable nonspecific binding. These results demonstrate that directed evolution is a viable approach for obtaining high-affinity PTM-specific antibodies and highlight the importance of assessing the specificity in the antibody engineering process.
Collapse
Affiliation(s)
- Dan Li
- Department of Biomedical Engineering, University of Connecticut, Storrs, Connecticut 06269
| | - Lei Wang
- Department of Chemistry, University of Connecticut, Storrs, Connecticut 06269
| | - Brandon F Maziuk
- Department of Pharmacology and Experimental Therapeutics, Boston University School of Medicine, Boston, Massachusetts 02118
| | - Xudong Yao
- Department of Chemistry, University of Connecticut, Storrs, Connecticut 06269; Department of Institute for Systems Genomics, University of Connecticut, Storrs, Connecticut 06269
| | - Benjamin Wolozin
- Department of Pharmacology and Experimental Therapeutics, Boston University School of Medicine, Boston, Massachusetts 02118
| | - Yong Ku Cho
- Department of Biomedical Engineering, University of Connecticut, Storrs, Connecticut 06269; Department of Institute for Systems Genomics, University of Connecticut, Storrs, Connecticut 06269; Department of Chemical and Biomolecular Engineering, University of Connecticut, Storrs, Connecticut 06269.
| |
Collapse
|
34
|
Loera-Valencia R, Piras A, Ismail MAM, Manchanda S, Eyjolfsdottir H, Saido TC, Johansson J, Eriksdotter M, Winblad B, Nilsson P. Targeting Alzheimer's disease with gene and cell therapies. J Intern Med 2018; 284:2-36. [PMID: 29582495 DOI: 10.1111/joim.12759] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Alzheimer's disease (AD) causes dementia in both young and old people affecting more than 40 million people worldwide. The two neuropathological hallmarks of the disease, amyloid beta (Aβ) plaques and neurofibrillary tangles consisting of protein tau are considered the major contributors to the disease. However, a more complete picture reveals significant neurodegeneration and decreased cell survival, neuroinflammation, changes in protein and energy homeostasis and alterations in lipid and cholesterol metabolism. In addition, gene and cell therapies for severe neurodegenerative disorders have recently improved technically in terms of safety and efficiency and have translated to the clinic showing encouraging results. Here, we review broadly current data within the field for potential targets that could modify AD through gene and cell therapy strategies. We envision that not only Aβ will be targeted in a disease-modifying treatment strategy but rather that a combination of treatments, possibly at different intervention times may prove beneficial in curing this devastating disease. These include decreased tau pathology, neuronal growth factors to support neurons and modulation of neuroinflammation for an appropriate immune response. Furthermore, cell based therapies may represent potential strategies in the future.
Collapse
Affiliation(s)
- R Loera-Valencia
- Division of Neurogeriatrics, Department of Neurobiology, Care Sciences and Society, Center for Alzheimer Research, Karolinska Institutet, Solna, Sweden
| | - A Piras
- Division of Neurogeriatrics, Department of Neurobiology, Care Sciences and Society, Center for Alzheimer Research, Karolinska Institutet, Solna, Sweden
| | - M A M Ismail
- Division of Neurogeriatrics, Department of Neurobiology, Care Sciences and Society, Center for Alzheimer Research, Karolinska Institutet, Solna, Sweden.,Theme Neuro, Diseases of the Nervous System Patient Flow, Karolinska University Hospital, Huddinge, Sweden
| | - S Manchanda
- Division of Neurogeriatrics, Department of Neurobiology, Care Sciences and Society, Center for Alzheimer Research, Karolinska Institutet, Solna, Sweden
| | - H Eyjolfsdottir
- Division of Clinical Geriatrics, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Huddinge, Sweden.,Theme Aging, Karolinska University Hospital, Huddinge, Sweden
| | - T C Saido
- RIKEN Brain Science Institute, Wako, Saitama, Japan
| | - J Johansson
- Division of Neurogeriatrics, Department of Neurobiology, Care Sciences and Society, Center for Alzheimer Research, Karolinska Institutet, Solna, Sweden
| | - M Eriksdotter
- Division of Clinical Geriatrics, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Huddinge, Sweden.,Theme Aging, Karolinska University Hospital, Huddinge, Sweden
| | - B Winblad
- Division of Neurogeriatrics, Department of Neurobiology, Care Sciences and Society, Center for Alzheimer Research, Karolinska Institutet, Solna, Sweden.,Theme Aging, Karolinska University Hospital, Huddinge, Sweden
| | - P Nilsson
- Division of Neurogeriatrics, Department of Neurobiology, Care Sciences and Society, Center for Alzheimer Research, Karolinska Institutet, Solna, Sweden
| |
Collapse
|
35
|
Spencer B, Brüschweiler S, Sealey-Cardona M, Rockenstein E, Adame A, Florio J, Mante M, Trinh I, Rissman RA, Konrat R, Masliah E. Selective targeting of 3 repeat Tau with brain penetrating single chain antibodies for the treatment of neurodegenerative disorders. Acta Neuropathol 2018; 136:69-87. [PMID: 29934874 PMCID: PMC6112111 DOI: 10.1007/s00401-018-1869-0] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2018] [Revised: 05/22/2018] [Accepted: 05/22/2018] [Indexed: 01/20/2023]
Abstract
Alzheimer's disease (AD) is the most common form of dementia in the elderly affecting more than 5 million people in the U.S. AD is characterized by the accumulation of β-amyloid (Aβ) and Tau in the brain, and is manifested by severe impairments in memory and cognition. Therefore, removing tau pathology has become one of the main therapeutic goals for the treatment of AD. Tau (tubulin-associated unit) is a major neuronal cytoskeletal protein found in the CNS encoded by the gene MAPT. Alternative splicing generates two major isoforms of tau containing either 3 or 4 repeat (R) segments. These 3R or 4RTau species are differentially expressed in neurodegenerative diseases. Previous studies have been focused on reducing Tau accumulation with antibodies against total Tau, 4RTau or phosphorylated isoforms. Here, we developed a brain penetrating, single chain antibody that specifically recognizes a pathogenic 3RTau. This single chain antibody was modified by the addition of a fragment of the apoB protein to facilitate trafficking into the brain, once in the CNS these antibody fragments reduced the accumulation of 3RTau and related deficits in a transgenic mouse model of tauopathy. NMR studies showed that the single chain antibody recognized an epitope at aa 40-62 of 3RTau. This single chain antibody reduced 3RTau transmission and facilitated the clearance of Tau via the endosomal-lysosomal pathway. Together, these results suggest that targeting 3RTau with highly specific, brain penetrating, single chain antibodies might be of potential value for the treatment of tauopathies such as Pick's Disease.
Collapse
Affiliation(s)
- Brian Spencer
- Department of Neurosciences, University of California, La Jolla, San Diego, CA, USA
| | - Sven Brüschweiler
- Department of Computational and Structural Biology, University of Vienna, Vienna, Austria
| | - Marco Sealey-Cardona
- Department of Computational and Structural Biology, University of Vienna, Vienna, Austria
| | - Edward Rockenstein
- Department of Neurosciences, University of California, La Jolla, San Diego, CA, USA
| | - Anthony Adame
- Department of Neurosciences, University of California, La Jolla, San Diego, CA, USA
| | - Jazmin Florio
- Department of Neurosciences, University of California, La Jolla, San Diego, CA, USA
| | - Michael Mante
- Department of Neurosciences, University of California, La Jolla, San Diego, CA, USA
| | - Ivy Trinh
- Department of Neurosciences, University of California, La Jolla, San Diego, CA, USA
| | - Robert A Rissman
- Department of Neurosciences, University of California, La Jolla, San Diego, CA, USA
- Veterans Affairs San Diego Healthcare System, San Diego, USA
| | - Robert Konrat
- Department of Computational and Structural Biology, University of Vienna, Vienna, Austria
| | - Eliezer Masliah
- Department of Neurosciences, University of California, La Jolla, San Diego, CA, USA.
- Department of Pathology, University of California, La Jolla, San Diego, CA, USA.
- Molecular Neuropathology Section, Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, 7201 Wisconsin Ave, Bethesda, MD, 20892, USA.
| |
Collapse
|
36
|
Abstract
Alzheimer disease (AD) is the most common form of dementia. Pathologically, AD is characterized by amyloid plaques and neurofibrillary tangles in the brain, with associated loss of synapses and neurons, resulting in cognitive deficits and eventually dementia. Amyloid-β (Aβ) peptide and tau protein are the primary components of the plaques and tangles, respectively. In the decades since Aβ and tau were identified, development of therapies for AD has primarily focused on Aβ, but tau has received more attention in recent years, in part because of the failure of various Aβ-targeting treatments in clinical trials. In this article, we review the current status of tau-targeting therapies for AD. Initially, potential anti-tau therapies were based mainly on inhibition of kinases or tau aggregation, or on stabilization of microtubules, but most of these approaches have been discontinued because of toxicity and/or lack of efficacy. Currently, the majority of tau-targeting therapies in clinical trials are immunotherapies, which have shown promise in numerous preclinical studies. Given that tau pathology correlates better with cognitive impairments than do Aβ lesions, targeting of tau is expected to be more effective than Aβ clearance once the clinical symptoms are evident. With future improvements in diagnostics, these two hallmarks of the disease might be targeted prophylactically.
Collapse
Affiliation(s)
- Erin E Congdon
- Department of Neuroscience and Physiology, New York University School of Medicine, New York, NY, USA
| | - Einar M Sigurdsson
- Department of Neuroscience and Physiology, New York University School of Medicine, New York, NY, USA.
- Department of Psychiatry, New York University School of Medicine, New York, NY, USA.
| |
Collapse
|
37
|
Zhuang J, Wang S, Shan Q, Zhang ZF, Li MQ, Zheng GH, Fan SH, Wu DM, Hu B, Lu J, Zheng YL. Adeno-associated virus vector-mediated expression of DJ-1 attenuates learning and memory deficits in 2, 2´, 4, 4´-tetrabromodiphenyl ether (BDE-47)-treated mice. JOURNAL OF HAZARDOUS MATERIALS 2018; 347:390-402. [PMID: 29335220 DOI: 10.1016/j.jhazmat.2018.01.008] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/18/2017] [Revised: 12/18/2017] [Accepted: 01/03/2018] [Indexed: 06/07/2023]
Abstract
Evidence indicates that oxidative stress is the central pathological feature of 2, 2´, 4, 4´-tetrabromodiphenyl ether (BDE-47)-induced neurotoxicity. Protein kinase C delta (PKCδ), an oxidative stress-sensitive kinase, can be proteolytically cleaved to yield a catalytically active fragment (PKCδ-CF) that is involved in various neurodegenerative disorders. Here, we showed that BDE-47 treatment increased ROS, malondialdehyde, and protein carbonyl levels in the mouse hippocampus. In turn, excessive ROS induced caspase-3-dependent PKCδ activation and stimulated NF-κB p65 nuclear translocation, resulting in inflammation in the mouse hippocampus. These changes caused learning and memory deficits in BDE-47-treated mice. Treatment with Z-DEVD-fmk, a caspase-3 inhibitor, or N-acetyl-L-cysteine, an antioxidant, blocked PKCδ activation and subsequently inhibited inflammation, thereby improving learning and memory deficits in BDE-47-treated mice. Our data further showed that activation of ROS-PKCδ signaling was associated with DJ-1 downregulation, which exerted neuroprotective effects against oxidative stress induced by different neurotoxic agents. Adeno-associated viral vector-mediated DJ-1 overexpression in the hippocampus effectively inhibited excessive ROS production, suppressed caspase-3-dependent PKCδ cleavage, blunted inflammation and ultimately reversed learning and memory deficits in BDE-47-treated mice. Taken together, our results demonstrate that DJ-1 plays a pivotal role in BDE-47-induced neurotoxic effects and learning and memory deficits.
Collapse
Affiliation(s)
- Juan Zhuang
- School of Environment Science and Spatial Informatics, China University of Mining and Technology, Xuzhou 221116, Jiangsu Province, China; Key Laboratory for Biotechnology on Medicinal Plants of Jiangsu Province, School of Life Science, Jiangsu Normal University, 101 Shanghai Road, Xuzhou 221116, Jiangsu Province, China; Jiangsu Collaborative Innovation Center of Regional Modern Agriculture & Environmental Protection, Huaiyin Normal University, Huaian 223300, China; Jiangsu Key Laboratory for Eco-Agricultural Biotechnology around Hongze Lake, Huaiyin Normal University, Huaian 223300, China; School of Life Sciences, Huaiyin Normal University, Huaian 223300, China
| | - Shan Wang
- Key Laboratory for Biotechnology on Medicinal Plants of Jiangsu Province, School of Life Science, Jiangsu Normal University, 101 Shanghai Road, Xuzhou 221116, Jiangsu Province, China; College of Health Sciences, Jiangsu Normal University, 101 Shanghai Road, Xuzhou 221116, Jiangsu Province, China
| | - Qun Shan
- School of Environment Science and Spatial Informatics, China University of Mining and Technology, Xuzhou 221116, Jiangsu Province, China; Key Laboratory for Biotechnology on Medicinal Plants of Jiangsu Province, School of Life Science, Jiangsu Normal University, 101 Shanghai Road, Xuzhou 221116, Jiangsu Province, China; College of Health Sciences, Jiangsu Normal University, 101 Shanghai Road, Xuzhou 221116, Jiangsu Province, China
| | - Zi-Feng Zhang
- Key Laboratory for Biotechnology on Medicinal Plants of Jiangsu Province, School of Life Science, Jiangsu Normal University, 101 Shanghai Road, Xuzhou 221116, Jiangsu Province, China; College of Health Sciences, Jiangsu Normal University, 101 Shanghai Road, Xuzhou 221116, Jiangsu Province, China
| | - Meng-Qiu Li
- Key Laboratory for Biotechnology on Medicinal Plants of Jiangsu Province, School of Life Science, Jiangsu Normal University, 101 Shanghai Road, Xuzhou 221116, Jiangsu Province, China; College of Health Sciences, Jiangsu Normal University, 101 Shanghai Road, Xuzhou 221116, Jiangsu Province, China
| | - Gui-Hong Zheng
- Key Laboratory for Biotechnology on Medicinal Plants of Jiangsu Province, School of Life Science, Jiangsu Normal University, 101 Shanghai Road, Xuzhou 221116, Jiangsu Province, China; College of Health Sciences, Jiangsu Normal University, 101 Shanghai Road, Xuzhou 221116, Jiangsu Province, China
| | - Shao-Hua Fan
- Key Laboratory for Biotechnology on Medicinal Plants of Jiangsu Province, School of Life Science, Jiangsu Normal University, 101 Shanghai Road, Xuzhou 221116, Jiangsu Province, China; College of Health Sciences, Jiangsu Normal University, 101 Shanghai Road, Xuzhou 221116, Jiangsu Province, China
| | - Dong-Mei Wu
- Key Laboratory for Biotechnology on Medicinal Plants of Jiangsu Province, School of Life Science, Jiangsu Normal University, 101 Shanghai Road, Xuzhou 221116, Jiangsu Province, China; College of Health Sciences, Jiangsu Normal University, 101 Shanghai Road, Xuzhou 221116, Jiangsu Province, China.
| | - Bin Hu
- Key Laboratory for Biotechnology on Medicinal Plants of Jiangsu Province, School of Life Science, Jiangsu Normal University, 101 Shanghai Road, Xuzhou 221116, Jiangsu Province, China; College of Health Sciences, Jiangsu Normal University, 101 Shanghai Road, Xuzhou 221116, Jiangsu Province, China
| | - Jun Lu
- Key Laboratory for Biotechnology on Medicinal Plants of Jiangsu Province, School of Life Science, Jiangsu Normal University, 101 Shanghai Road, Xuzhou 221116, Jiangsu Province, China; College of Health Sciences, Jiangsu Normal University, 101 Shanghai Road, Xuzhou 221116, Jiangsu Province, China.
| | - Yuan-Lin Zheng
- Key Laboratory for Biotechnology on Medicinal Plants of Jiangsu Province, School of Life Science, Jiangsu Normal University, 101 Shanghai Road, Xuzhou 221116, Jiangsu Province, China; College of Health Sciences, Jiangsu Normal University, 101 Shanghai Road, Xuzhou 221116, Jiangsu Province, China.
| |
Collapse
|
38
|
Alzheimer’s Disease and Frontotemporal Lobar Degeneration: Mouse Models. NEURODEGENER DIS 2018. [DOI: 10.1007/978-3-319-72938-1_10] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/17/2022] Open
|
39
|
Sigurdsson EM. Tau Immunotherapies for Alzheimer's Disease and Related Tauopathies: Progress and Potential Pitfalls. J Alzheimers Dis 2018; 64:S555-S565. [PMID: 29865056 PMCID: PMC6171771 DOI: 10.3233/jad-179937] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Tau immunotherapies have now advanced from proof-of-concept studies to Phase II clinical trials. This review briefly outlines developments in the field and discusses how these therapies may work, which involves multiple variables that are connected in complex ways. These various factors are likely to define therapeutic success in humans and have not been thoroughly investigated, at least based on published reports.
Collapse
Affiliation(s)
- Einar M. Sigurdsson
- New York University School of Medicine, Departments of Neuroscience and Physiology, and Psychiatry, Neuroscience Institute, Science Building, 435 East 30 Street, Room SB1115, New York, NY 10016,
| |
Collapse
|
40
|
Raikwar SP, Thangavel R, Dubova I, Ahmed ME, Selvakumar PG, Kempuraj D, Zaheer S, Iyer S, Zaheer A. Neuro-Immuno-Gene- and Genome-Editing-Therapy for Alzheimer's Disease: Are We There Yet? J Alzheimers Dis 2018; 65:321-344. [PMID: 30040732 PMCID: PMC6130335 DOI: 10.3233/jad-180422] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/26/2018] [Indexed: 12/16/2022]
Abstract
Alzheimer's disease (AD) is a highly complex neurodegenerative disorder and the current treatment strategies are largely ineffective thereby leading to irreversible and progressive cognitive decline in AD patients. AD continues to defy successful treatment despite significant advancements in the field of molecular medicine. Repeatedly, early promising preclinical and clinical results have catapulted into devastating setbacks leading to multi-billion dollar losses not only to the top pharmaceutical companies but also to the AD patients and their families. Thus, it is very timely to review the progress in the emerging fields of gene therapy and stem cell-based precision medicine. Here, we have made sincere efforts to feature the ongoing progress especially in the field of AD gene therapy and stem cell-based regenerative medicine. Further, we also provide highlights in elucidating the molecular mechanisms underlying AD pathogenesis and describe novel AD therapeutic targets and strategies for the new drug discovery. We hope that the quantum leap in the scientific advancements and improved funding will bolster novel concepts that will propel the momentum toward a trajectory leading to a robust AD patient-specific next generation precision medicine with improved cognitive function and excellent life quality.
Collapse
Affiliation(s)
- Sudhanshu P. Raikwar
- Department of Neurology, Center for Translational Neuroscience, School of Medicine, University of Missouri, Columbia, MO, USA
- U.S. Department of Veterans Affairs, Harry S. Truman Memorial Veteran’s Hospital, Columbia, MO, USA
| | - Ramasamy Thangavel
- Department of Neurology, Center for Translational Neuroscience, School of Medicine, University of Missouri, Columbia, MO, USA
- U.S. Department of Veterans Affairs, Harry S. Truman Memorial Veteran’s Hospital, Columbia, MO, USA
| | - Iuliia Dubova
- Department of Neurology, Center for Translational Neuroscience, School of Medicine, University of Missouri, Columbia, MO, USA
| | - Mohammad Ejaz Ahmed
- Department of Neurology, Center for Translational Neuroscience, School of Medicine, University of Missouri, Columbia, MO, USA
- U.S. Department of Veterans Affairs, Harry S. Truman Memorial Veteran’s Hospital, Columbia, MO, USA
| | - Pushpavathi Govindhasamy Selvakumar
- Department of Neurology, Center for Translational Neuroscience, School of Medicine, University of Missouri, Columbia, MO, USA
- U.S. Department of Veterans Affairs, Harry S. Truman Memorial Veteran’s Hospital, Columbia, MO, USA
| | - Duraisamy Kempuraj
- Department of Neurology, Center for Translational Neuroscience, School of Medicine, University of Missouri, Columbia, MO, USA
- U.S. Department of Veterans Affairs, Harry S. Truman Memorial Veteran’s Hospital, Columbia, MO, USA
| | - Smita Zaheer
- Department of Neurology, Center for Translational Neuroscience, School of Medicine, University of Missouri, Columbia, MO, USA
| | - Shankar Iyer
- Department of Neurology, Center for Translational Neuroscience, School of Medicine, University of Missouri, Columbia, MO, USA
- U.S. Department of Veterans Affairs, Harry S. Truman Memorial Veteran’s Hospital, Columbia, MO, USA
| | - Asgar Zaheer
- Department of Neurology, Center for Translational Neuroscience, School of Medicine, University of Missouri, Columbia, MO, USA
- U.S. Department of Veterans Affairs, Harry S. Truman Memorial Veteran’s Hospital, Columbia, MO, USA
| |
Collapse
|
41
|
TREM2 deficiency attenuates neuroinflammation and protects against neurodegeneration in a mouse model of tauopathy. Proc Natl Acad Sci U S A 2017; 114:11524-11529. [PMID: 29073081 PMCID: PMC5663386 DOI: 10.1073/pnas.1710311114] [Citation(s) in RCA: 336] [Impact Index Per Article: 42.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Alzheimer’s disease (AD) is the most common cause of dementia and is a major public health problem for which there is currently no disease-modifying treatment. There is an urgent need for greater understanding of the molecular mechanisms underlying neurodegeneration in patients to create better therapeutic options. Recently, genetic studies uncovered novel AD risk variants in the microglial receptor, triggering receptor expressed on myeloid cells 2 (TREM2). Previous studies suggested that loss of TREM2 function worsens amyloid-β (Aβ) plaque-related toxicity. In contrast, we observe TREM2 deficiency mitigates neuroinflammation and protects against brain atrophy in the context of tau pathology. These findings indicate dual roles for TREM2 and microglia in the context of amyloid versus tau pathology, which are important to consider for potential treatments targeting TREM2. Variants in the gene encoding the triggering receptor expressed on myeloid cells 2 (TREM2) were recently found to increase the risk for developing Alzheimer’s disease (AD). In the brain, TREM2 is predominately expressed on microglia, and its association with AD adds to increasing evidence implicating a role for the innate immune system in AD initiation and progression. Thus far, studies have found TREM2 is protective in the response to amyloid pathology while variants leading to a loss of TREM2 function impair microglial signaling and are deleterious. However, the potential role of TREM2 in the context of tau pathology has not yet been characterized. In this study, we crossed Trem2+/+ (T2+/+) and Trem2−/− (T2−/−) mice to the PS19 human tau transgenic line (PS) to investigate whether loss of TREM2 function affected tau pathology, the microglial response to tau pathology, or neurodegeneration. Strikingly, by 9 mo of age, T2−/−PS mice exhibited significantly less brain atrophy as quantified by ventricular enlargement and preserved cortical volume in the entorhinal and piriform regions compared with T2+/+PS mice. However, no TREM2-dependent differences were observed for phosphorylated tau staining or insoluble tau levels. Rather, T2−/−PS mice exhibited significantly reduced microgliosis in the hippocampus and piriform cortex compared with T2+/+PS mice. Gene expression analyses and immunostaining revealed microglial activation was significantly attenuated in T2−/−PS mice, and there were lower levels of inflammatory cytokines and astrogliosis. These unexpected findings suggest that impairing microglial TREM2 signaling reduces neuroinflammation and is protective against neurodegeneration in the setting of pure tauopathy.
Collapse
|
42
|
Tau-based therapies in neurodegeneration: opportunities and challenges. Nat Rev Drug Discov 2017; 16:863-883. [DOI: 10.1038/nrd.2017.155] [Citation(s) in RCA: 155] [Impact Index Per Article: 19.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|