1
|
Arastoo M, Penny LK, Lofthouse R, Abdallah A, Abrahamsson A, Marini P, Melis V, Riedel G, Harrington CR, Wischik CM, Porter A, Palliyil S. High-affinity antibodies specific to the core region of the tau protein exhibit diagnostic and therapeutic potential for Alzheimer's disease. Alzheimers Res Ther 2024; 16:209. [PMID: 39358820 PMCID: PMC11448309 DOI: 10.1186/s13195-024-01561-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Accepted: 08/15/2024] [Indexed: 10/04/2024]
Abstract
BACKGROUND Recent advances in blood-based biomarker discovery are paving the way for simpler, more accessible diagnostic tools that can detect early signs of Alzheimer's disease (AD). Recent successes in the development of amyloid-targeting immunotherapy approaches mark an important advancement in providing new options for the treatment of AD. We have developed a set of high-affinity monoclonal antibodies (mAbs) to tau protein that have the potential as tools for diagnosis and treatment of AD. METHODS Sheep were immunised with either full-length tau (1-441) or truncated paired helical filament (PHF)-core tau (297-391). A stringent bio-panning and epitope selection strategy, with a particular focus directed to epitopes within the disease-relevant PHF-core tau, was used to identify single-chain antibodies (scAbs). These scAbs were ranked by affinity for each epitope class, with leads converted to high-affinity mAbs. These antibodies and their potential utility were assessed by their performance in tau immunoassays, as well as their ability to prevent tau aggregation and propagation. Further characterisation of these antibodies was performed by immunohistochemical staining of brain sections and immuno-gold electronmicroscopy of isolated PHFs. RESULTS Our work resulted in a set of high-affinity antibodies reacting with multiple epitopes spanning the entire tau protein molecule. The tau antibodies directed against the core tau unit of the PHF inhibited pathological aggregation and seeding using several biochemical and cell assay systems. Through staining of brain sections and PHFs, the panel of antibodies revealed which tau epitopes were available, truncated, or occluded. In addition, highly sensitive immunoassays were developed with the ability to distinguish between and quantify various tau fragments. CONCLUSION This article introduces an alternative immunodiagnostic approach based on the concept of a "tauosome" - the diverse set of tau fragments present within biological fluids. The development of an antibody panel that can distinguish a range of different tau fragments provides the basis for a novel approach to potential diagnosis and monitoring of disease progression. Our results further support the notion that tau immunotherapy targeting the PHF-core needs to combine appropriate selection of both the target epitope and antibody affinity to optimise therapeutic potential.
Collapse
Affiliation(s)
- Mohammad Arastoo
- Institute of Medical Sciences, University of Aberdeen, Aberdeen, UK
- Scottish Biologics Facility, University of Aberdeen, Aberdeen, UK
| | - Lewis K Penny
- Institute of Medical Sciences, University of Aberdeen, Aberdeen, UK
- Scottish Biologics Facility, University of Aberdeen, Aberdeen, UK
- TauRx Therapeutics Ltd, Aberdeen, UK
| | - Richard Lofthouse
- Institute of Medical Sciences, University of Aberdeen, Aberdeen, UK
- Scottish Biologics Facility, University of Aberdeen, Aberdeen, UK
| | - Aya Abdallah
- Institute of Medical Sciences, University of Aberdeen, Aberdeen, UK
- Scottish Biologics Facility, University of Aberdeen, Aberdeen, UK
| | - Anna Abrahamsson
- Institute of Medical Sciences, University of Aberdeen, Aberdeen, UK
- Scottish Biologics Facility, University of Aberdeen, Aberdeen, UK
| | - Pietro Marini
- Institute of Medical Sciences, University of Aberdeen, Aberdeen, UK
| | - Valeria Melis
- Institute of Medical Sciences, University of Aberdeen, Aberdeen, UK
| | - Gernot Riedel
- Institute of Medical Sciences, University of Aberdeen, Aberdeen, UK
| | - Charles R Harrington
- Institute of Medical Sciences, University of Aberdeen, Aberdeen, UK
- GT Diagnostics (UK) Ltd, Aberdeen, UK
- TauRx Therapeutics Ltd, Aberdeen, UK
| | - Claude M Wischik
- Institute of Medical Sciences, University of Aberdeen, Aberdeen, UK
- GT Diagnostics (UK) Ltd, Aberdeen, UK
- TauRx Therapeutics Ltd, Aberdeen, UK
| | - Andrew Porter
- Institute of Medical Sciences, University of Aberdeen, Aberdeen, UK
- Scottish Biologics Facility, University of Aberdeen, Aberdeen, UK
| | - Soumya Palliyil
- Institute of Medical Sciences, University of Aberdeen, Aberdeen, UK.
- Scottish Biologics Facility, University of Aberdeen, Aberdeen, UK.
| |
Collapse
|
2
|
Islam MR, Rabbi MA, Hossain T, Sultana S, Uddin S. Mechanistic Approach to Immunity and Immunotherapy of Alzheimer's Disease: A Review. ACS Chem Neurosci 2024. [PMID: 39173186 DOI: 10.1021/acschemneuro.4c00360] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/24/2024] Open
Abstract
Alzheimer's disease (AD) is a debilitating neurodegenerative condition characterized by progressive cognitive decline and memory loss, affecting millions of people worldwide. Traditional treatments, such as cholinesterase inhibitors and NMDA receptor antagonists, offer limited symptomatic relief without addressing the underlying disease mechanisms. These limitations have driven the development of more potent and effective therapies. Recent advances in immunotherapy present promising avenues for AD treatment. Immunotherapy strategies, including both active and passive approaches, harness the immune system to target and mitigate AD-related pathology. Active immunotherapy stimulates the patient's immune response to produce antibodies against AD-specific antigens, while passive immunotherapy involves administering preformed antibodies or immune cells that specifically target amyloid-β (Aβ) or tau proteins. Monoclonal antibodies, such as aducanumab and lecanemab, have shown potential in reducing Aβ plaques and slowing cognitive decline in clinical trials, despite challenges related to adverse immune responses and the need for precise targeting. This comprehensive review explores the role of the immune system in AD, evaluates the current successes and limitations of immunotherapeutic approaches, and discusses future directions for enhancing the treatment efficacy.
Collapse
Affiliation(s)
- Md Rubiath Islam
- Department of Biochemistry and Molecular Biology, Shahjalal University of Science and Technology, Sylhet 3114, Bangladesh
| | - Md Afser Rabbi
- Department of Biochemistry and Molecular Biology, Shahjalal University of Science and Technology, Sylhet 3114, Bangladesh
| | - Tanbir Hossain
- Department of Biochemistry and Molecular Biology, Shahjalal University of Science and Technology, Sylhet 3114, Bangladesh
| | - Sadia Sultana
- Department of Biochemistry and Molecular Biology, Shahjalal University of Science and Technology, Sylhet 3114, Bangladesh
| | - Shihab Uddin
- Department of Bioengineering, King Fahad University of Petroleum & Minerals, Dhahran 31261, Saudi Arabia
- Interdisciplinary Research Center for Bio Systems and Machines, King Fahad University of Petroleum & Minerals, Dhahran-31261, Saudi Arabia
| |
Collapse
|
3
|
Shoemaker RL, Larsen RJ, Larsen PA. Single-domain antibodies and aptamers drive new opportunities for neurodegenerative disease research. Front Immunol 2024; 15:1426656. [PMID: 39238639 PMCID: PMC11374656 DOI: 10.3389/fimmu.2024.1426656] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2024] [Accepted: 07/22/2024] [Indexed: 09/07/2024] Open
Abstract
Neurodegenerative diseases (NDs) in mammals, such as Alzheimer's disease (AD), Parkinson's disease (PD), and transmissible spongiform encephalopathies (TSEs), are characterized by the accumulation of misfolded proteins in the central nervous system (CNS). Despite the presence of these pathogenic proteins, the immune response in affected individuals remains notably muted. Traditional immunological strategies, particularly those reliant on monoclonal antibodies (mAbs), face challenges related to tissue penetration, blood-brain barrier (BBB) crossing, and maintaining protein stability. This has led to a burgeoning interest in alternative immunotherapeutic avenues. Notably, single-domain antibodies (or nanobodies) and aptamers have emerged as promising candidates, as their reduced size facilitates high affinity antigen binding and they exhibit superior biophysical stability compared to mAbs. Aptamers, synthetic molecules generated from DNA or RNA ligands, present both rapid production times and cost-effective solutions. Both nanobodies and aptamers exhibit inherent qualities suitable for ND research and therapeutic development. Cross-seeding events must be considered in both traditional and small-molecule-based immunodiagnostic and therapeutic approaches, as well as subsequent neurotoxic impacts and complications beyond protein aggregates. This review delineates the challenges traditional immunological methods pose in ND research and underscores the potential of nanobodies and aptamers in advancing next-generation ND diagnostics and therapeutics.
Collapse
Affiliation(s)
- Rachel L Shoemaker
- Minnesota Center for Prion Research and Outreach (MNPRO), University of Minnesota, St. Paul, MN, United States
- Department of Biomedical and Veterinary Sciences, University of Minnesota College of Veterinary Medicine, St. Paul, MN, United States
| | - Roxanne J Larsen
- Department of Biomedical and Veterinary Sciences, University of Minnesota College of Veterinary Medicine, St. Paul, MN, United States
- Priogen Corp., St. Paul, MN, United States
| | - Peter A Larsen
- Minnesota Center for Prion Research and Outreach (MNPRO), University of Minnesota, St. Paul, MN, United States
- Department of Biomedical and Veterinary Sciences, University of Minnesota College of Veterinary Medicine, St. Paul, MN, United States
| |
Collapse
|
4
|
Miteva D, Vasilev GV, Velikova T. Role of Specific Autoantibodies in Neurodegenerative Diseases: Pathogenic Antibodies or Promising Biomarkers for Diagnosis. Antibodies (Basel) 2023; 12:81. [PMID: 38131803 PMCID: PMC10740538 DOI: 10.3390/antib12040081] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2023] [Revised: 12/04/2023] [Accepted: 12/05/2023] [Indexed: 12/23/2023] Open
Abstract
Neurodegenerative diseases (NDDs) affect millions of people worldwide. They develop due to the pathological accumulation and aggregation of various misfolded proteins, axonal and synaptic loss and dysfunction, inflammation, cytoskeletal abnormalities, defects in DNA and RNA, and neuronal death. This leads to the activation of immune responses and the release of the antibodies against them. Recently, it has become clear that autoantibodies (Aabs) can contribute to demyelination, axonal loss, and brain and cognitive dysfunction. This has significantly changed the understanding of the participation of humoral autoimmunity in neurodegenerative disorders. It is crucial to understand how neuroinflammation is involved in neurodegeneration, to aid in improving the diagnostic and therapeutic value of Aabs in the future. This review aims to provide data on the immune system's role in NDDs, the pathogenic role of some specific Aabs against molecules associated with the most common NDDs, and their potential role as biomarkers for monitoring and diagnosing NDDs. It is suggested that the autoimmune aspects of NDDs will facilitate early diagnosis and help to elucidate previously unknown aspects of the pathobiology of these diseases.
Collapse
Affiliation(s)
- Dimitrina Miteva
- Department of Genetics, Faculty of Biology, Sofia University “St. Kliment Ohridski”, 8 Dragan Tzankov Str., 1164 Sofia, Bulgaria
- Medical Faculty, Sofia University St. Kliment Ohridski, 1 Kozyak str, 1407 Sofia, Bulgaria; (G.V.V.); (T.V.)
| | - Georgi V. Vasilev
- Medical Faculty, Sofia University St. Kliment Ohridski, 1 Kozyak str, 1407 Sofia, Bulgaria; (G.V.V.); (T.V.)
- Clinic of Neurology, Department of Emergency Medicine UMHAT “Sv. Georgi”, 4000 Plovdiv, Bulgaria
| | - Tsvetelina Velikova
- Medical Faculty, Sofia University St. Kliment Ohridski, 1 Kozyak str, 1407 Sofia, Bulgaria; (G.V.V.); (T.V.)
| |
Collapse
|
5
|
Robbins M. Therapies for Tau-associated neurodegenerative disorders: targeting molecules, synapses, and cells. Neural Regen Res 2023; 18:2633-2637. [PMID: 37449601 PMCID: PMC10358644 DOI: 10.4103/1673-5374.373670] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2023] [Revised: 02/14/2023] [Accepted: 03/15/2023] [Indexed: 07/18/2023] Open
Abstract
Advances in experimental and computational technologies continue to grow rapidly to provide novel avenues for the treatment of neurodegenerative disorders. Despite this, there remain only a handful of drugs that have shown success in late-stage clinical trials for Tau-associated neurodegenerative disorders. The most commonly prescribed treatments are symptomatic treatments such as cholinesterase inhibitors and N-methyl-D-aspartate receptor blockers that were approved for use in Alzheimer's disease. As diagnostic screening can detect disorders at earlier time points, the field needs pre-symptomatic treatments that can prevent, or significantly delay the progression of these disorders (Koychev et al., 2019). These approaches may be different from late-stage treatments that may help to ameliorate symptoms and slow progression once symptoms have become more advanced should early diagnostic screening fail. This mini-review will highlight five key avenues of academic and industrial research for identifying therapeutic strategies to treat Tau-associated neurodegenerative disorders. These avenues include investigating (1) the broad class of chemicals termed "small molecules"; (2) adaptive immunity through both passive and active antibody treatments; (3) innate immunity with an emphasis on microglial modulation; (4) synaptic compartments with the view that Tau-associated neurodegenerative disorders are synaptopathies. Although this mini-review will focus on Alzheimer's disease due to its prevalence, it will also argue the need to target other tauopathies, as through understanding Alzheimer's disease as a Tau-associated neurodegenerative disorder, we may be able to generalize treatment options. For this reason, added detail linking back specifically to Tau protein as a direct therapeutic target will be added to each topic.
Collapse
Affiliation(s)
- Miranda Robbins
- MRC Laboratory of Molecular Biology, Cambridge Biomedical Campus, Francis Crick Ave, Trumpington, Cambridge, UK; University of Cambridge, Department of Zoology, Cambridge, UK
| |
Collapse
|
6
|
Role of Tau in Various Tauopathies, Treatment Approaches, and Emerging Role of Nanotechnology in Neurodegenerative Disorders. Mol Neurobiol 2023; 60:1690-1720. [PMID: 36562884 DOI: 10.1007/s12035-022-03164-z] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Accepted: 12/06/2022] [Indexed: 12/24/2022]
Abstract
A few protein kinases and phosphatases regulate tau protein phosphorylation and an imbalance in their enzyme activity results in tau hyper-phosphorylation. Aberrant tau phosphorylation causes tau to dissociate from the microtubules and clump together in the cytosol to form neurofibrillary tangles (NFTs), which lead to the progression of neurodegenerative disorders including Alzheimer's disease (AD) and other tauopathies. Hence, targeting hyperphosphorylated tau protein is a restorative approach for treating neurodegenerative tauopathies. The cyclin-dependent kinase (Cdk5) and the glycogen synthase kinase (GSK3β) have both been implicated in aberrant tau hyperphosphorylation. The limited transport of drugs through the blood-brain barrier (BBB) for reaching the central nervous system (CNS) thus represents a significant problem in the development of drugs. Drug delivery systems based on nanocarriers help solve this problem. In this review, we discuss the tau protein, regulation of tau phosphorylation and abnormal hyperphosphorylation, drugs in use or under clinical trials, and treatment strategies for tauopathies based on the critical role of tau hyperphosphorylation in the pathogenesis of the disease. Pathology of neurodegenerative disease due to hyperphosphorylation and various therapeutic approaches including nanotechnology for its treatment.
Collapse
|
7
|
Luo J, Wu H, Li J, Xian W, Li W, Locascio JJ, Pei Z, Liu G. Joint Modeling Study Identifies Blood-Based Transcripts Link to Cognitive Decline in Parkinson's Disease. Mov Disord 2022; 37:2386-2395. [PMID: 36087011 DOI: 10.1002/mds.29213] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2022] [Revised: 08/05/2022] [Accepted: 08/17/2022] [Indexed: 01/13/2023] Open
Abstract
BACKGROUND Cognitive decline in Parkinson's disease (PD) is prevalent, insidious, and burdensome during the progression of the disease. OBJECTIVES We aimed to find transcriptome-wide biomarkers in blood to predict cognitive decline and identify patients at high risk with cognitive impairment in PD. METHODS We carried out joint modeling analysis to characterize transcriptome-wide longitudinal gene expression and its association with the progression of mild cognitive impairment (MCI) in PD patients. The average time-dependent area under the curves (AUCs) were used for evaluating the accuracy of the significant joint models. A cognitive survival score (CogSs) derived from joint model was leveraged to predict the occurrence of MCI. All predicting models were built in a discovery cohort with 272 patients and replicated in an independent cohort with 177 patients. RESULTS We identified five longitudinal varied expression of transcripts that were significantly associated with MCI progression in patients with PD. The most significant transcript IGLC1 joint model accurately predicted the progression of MCI in PD patients in the discovery and replication cohorts (average time-dependent AUCs >0.82). The CogSs derived from the optimal IGLC1 joint model had a high accuracy at early study stage in both cohorts (AUC ≥0.91). CONCLUSIONS Our transcriptome-wide joint modeling analysis uncovered five blood-based transcripts related to cognitive decline in PD. The joint models will serve as a useful resource for clinicians and researchers to screen PD patients with high risk of development of cognitive impairment and pave the path for Parkinson's personalized medicine. © 2022 International Parkinson and Movement Disorder Society.
Collapse
Affiliation(s)
- Junfeng Luo
- Neurobiology Research Center, School of Medicine, Shenzhen Campus of Sun Yat-sen University, Shenzhen, China
| | - Hao Wu
- Neurobiology Research Center, School of Medicine, Shenzhen Campus of Sun Yat-sen University, Shenzhen, China
| | - Jinxia Li
- Neurobiology Research Center, School of Medicine, Shenzhen Campus of Sun Yat-sen University, Shenzhen, China
| | - Wenbiao Xian
- Department of Neurology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Weimin Li
- Neurobiology Research Center, School of Medicine, Shenzhen Campus of Sun Yat-sen University, Shenzhen, China
| | - Joseph J Locascio
- Department of Neurology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Zhong Pei
- Department of Neurology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | | |
Collapse
|
8
|
Chen Y, Colonna M. Spontaneous and induced adaptive immune responses in Alzheimer's disease: new insights into old observations. Curr Opin Immunol 2022; 77:102233. [PMID: 35839620 DOI: 10.1016/j.coi.2022.102233] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Revised: 05/28/2022] [Accepted: 06/06/2022] [Indexed: 01/20/2023]
Abstract
Alzheimer's disease (AD) is the leading cause of dementia in the elderly. Although AD is primarily a neurological disorder distinguished by amyloid β plaques and intracellular neurofibrillary tangles, the immune system can impact the progression of the disease and may be targeted for therapeutic purposes. To date, most studies have focused on innate immune responses of microglia. However, emerging evidence implicates adaptive immune responses by T cells and B cells in the progression of AD. Moreover, the recent approval of an antibody that promotes amyloid β plaque clearance for AD therapy has pinpointed adaptive immunity as a fertile ground for the design of novel therapeutic approaches. Here, we highlight key studies delineating T cell and B cell responses in human AD and mouse models of AD, identify open questions on the specificity, development and impact of these responses and discuss outlooks for future studies and novel therapeutic avenues.
Collapse
Affiliation(s)
- Yun Chen
- Department of Pathology and Immunology and Department of Neurology, Washington University School of Medicine in St Louis, USA
| | - Marco Colonna
- Department of Pathology and Immunology and Department of Neurology, Washington University School of Medicine in St Louis, USA.
| |
Collapse
|
9
|
Stahr N, Galkina EV. Immune Response at the Crossroads of Atherosclerosis and Alzheimer's Disease. Front Cardiovasc Med 2022; 9:870144. [PMID: 35872901 PMCID: PMC9298512 DOI: 10.3389/fcvm.2022.870144] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2022] [Accepted: 06/02/2022] [Indexed: 11/30/2022] Open
Abstract
Alzheimer's disease (AD) and cardiovascular disease (CVD) are pathologies that are characterized by common signatures of vascular dysfunction and chronic inflammation that are accelerated with aging. Importantly, epidemiological studies report an independent interaction between AD and CVD and data suggest that chronic inflammation in CVD may accelerate AD development. Atherosclerosis affects most large to medium sized arteries including those supplying the cerebral circulation. Vascular dysfunction caused by atherosclerosis results in blood brain barrier breakdown, inflammation, an impaired clearance of amyloid-beta (Aβ), and finally ends with neurovascular dysfunction. Numerous data indicate that innate and adaptive immune responses shape atherogenesis and increasing evidence suggests an implication of the immune response in AD progression. Currently, mechanisms by which these two diseases are interconnected with each other are not well-defined. In this review, we discuss the recent advances in our understanding of the intertwined role of the immune response in atherosclerosis and AD and the implications of these findings for human health.
Collapse
|
10
|
Bozhanova NG, Flyak AI, Brown BP, Ruiz SE, Salas J, Rho S, Bombardi RG, Myers L, Soto C, Bailey JR, Crowe JE, Bjorkman PJ, Meiler J. Computational identification of HCV neutralizing antibodies with a common HCDR3 disulfide bond motif in the antibody repertoires of infected individuals. Nat Commun 2022; 13:3178. [PMID: 35676279 PMCID: PMC9177688 DOI: 10.1038/s41467-022-30865-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Accepted: 05/20/2022] [Indexed: 12/14/2022] Open
Abstract
Despite recent success in hepatitis C virus (HCV) treatment using antivirals, an HCV vaccine is still needed to prevent reinfections in treated patients, to avert the emergence of drug-resistant strains, and to provide protection for people with no access to the antiviral therapeutics. The early production of broadly neutralizing antibodies (bNAbs) associates with HCV clearance. Several potent bNAbs bind a conserved HCV glycoprotein E2 epitope using an unusual heavy chain complementarity determining region 3 (HCDR3) containing an intra-loop disulfide bond. Isolation of additional structurally-homologous bNAbs would facilitate the recognition of key determinants of such bNAbs and guide rational vaccine design. Here we report the identification of new antibodies containing an HCDR3 disulfide bond motif using computational screening with the Rosetta software. Using the newly-discovered and already-known members of this antibody family, we review the required HCDR3 amino acid composition and propose determinants for the bent versus straight HCDR3 loop conformation observed in these antibodies.
Collapse
Affiliation(s)
- Nina G Bozhanova
- Department of Chemistry, Vanderbilt University, Nashville, TN, 37235, USA
- Center for Structural Biology, Vanderbilt University, Nashville, TN, 37235, USA
| | - Andrew I Flyak
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, 91125, USA
| | - Benjamin P Brown
- Department of Chemistry, Vanderbilt University, Nashville, TN, 37235, USA
- Center for Structural Biology, Vanderbilt University, Nashville, TN, 37235, USA
| | - Stormy E Ruiz
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, 91125, USA
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
| | - Jordan Salas
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
| | - Semi Rho
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, 91125, USA
| | - Robin G Bombardi
- Vanderbilt Vaccine Center, Vanderbilt University Medical Center, Nashville, TN, 37232, USA
| | - Luke Myers
- Vanderbilt Vaccine Center, Vanderbilt University Medical Center, Nashville, TN, 37232, USA
| | - Cinque Soto
- Vanderbilt Vaccine Center, Vanderbilt University Medical Center, Nashville, TN, 37232, USA
- Department of Pediatrics, Vanderbilt University Medical Center, Nashville, TN, 37232, USA
| | - Justin R Bailey
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
| | - James E Crowe
- Vanderbilt Vaccine Center, Vanderbilt University Medical Center, Nashville, TN, 37232, USA
- Department of Pediatrics, Vanderbilt University Medical Center, Nashville, TN, 37232, USA
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, TN, 37232, USA
| | - Pamela J Bjorkman
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, 91125, USA
| | - Jens Meiler
- Department of Chemistry, Vanderbilt University, Nashville, TN, 37235, USA.
- Center for Structural Biology, Vanderbilt University, Nashville, TN, 37235, USA.
- Institute for Drug Discovery, Leipzig University Medical School, Leipzig, SAC, 04103, Germany.
| |
Collapse
|
11
|
Abstract
The realization that autoantibodies can contribute to dysfunction of the brain has brought about a paradigm shift in neurological diseases over the past decade, offering up important novel diagnostic and therapeutic opportunities. Detection of specific autoantibodies to neuronal or glial targets has resulted in a better understanding of central nervous system autoimmunity and in the reclassification of some diseases previously thought to result from infectious, 'idiopathic' or psychogenic causes. The most prominent examples, such as aquaporin 4 autoantibodies in neuromyelitis optica or NMDAR autoantibodies in encephalitis, have stimulated an entire field of clinical and experimental studies on disease mechanisms and immunological abnormalities. Also, these findings inspired the search for additional autoantibodies, which has been very successful to date and has not yet reached its peak. This Review summarizes this rapid development at a point in time where preclinical studies have started delivering fundamental new data for mechanistic understanding, where new technologies are being introduced into this field, and - most importantly - where the first specifically tailored immunotherapeutic approaches are emerging.
Collapse
Affiliation(s)
- Harald Prüss
- German Center for Neurodegenerative Diseases (DZNE) Berlin, Berlin, Germany.
- Department of Neurology and Experimental Neurology, Charité - Universitätsmedizin Berlin, Berlin, Germany.
| |
Collapse
|
12
|
Quint WH, Matečko-Burmann I, Schilcher I, Löffler T, Schöll M, Burmann BM, Vogels T. Bispecific Tau Antibodies with Additional Binding to C1q or Alpha-Synuclein. J Alzheimers Dis 2021; 80:813-829. [PMID: 33579845 DOI: 10.3233/jad-201334] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
BACKGROUND Alzheimer's disease (AD) and other tauopathies are neurodegenerative disorders characterized by cellular accumulation of aggregated tau protein. Tau pathology within these disorders is accompanied by chronic neuroinflammation, such as activation of the classical complement pathway by complement initiation factor C1q. Additionally, about half of the AD cases present with inclusions composed of aggregated alpha-synuclein called Lewy bodies. Lewy bodies in disorders such as Parkinson's disease and Lewy body dementia also frequently occur together with tau pathology. OBJECTIVE Immunotherapy is currently the most promising treatment strategy for tauopathies. However, the presence of multiple pathological processes within tauopathies makes it desirable to simultaneously target more than one disease pathway. METHODS Herein, we have developed three bispecific antibodies based on published antibody binding region sequences. One bispecific antibody binds to tau plus alpha-synuclein and two bispecific antibodies bind to tau plus C1q. RESULTS Affinity of the bispecific antibodies to their targets compared to their monospecific counterparts ranged from nearly identical to one order of magnitude lower. All bispecific antibodies retained binding to aggregated protein in patient-derived brain sections. The bispecific antibodies also retained their ability to inhibit aggregation of recombinant tau, regardless of whether the tau binding sites were in IgG or scFv format. Mono- and bispecific antibodies inhibited cellular seeding induced by AD-derived pathological tau with similar efficacy. Finally, both Tau-C1q bispecific antibodies completely inhibited the classical complement pathway. CONCLUSION Bispecific antibodies that bind to multiple pathological targets may therefore present a promising approach to treat tauopathies and other neurodegenerative disorders.
Collapse
Affiliation(s)
| | - Irena Matečko-Burmann
- Wallenberg Centre for Molecular and Translational Medicine, University of Gothenburg, Gothenburg, Sweden.,Department of Psychiatry and Neurochemistry, University of Gothenburg, Gothenburg, Sweden
| | | | - Tina Löffler
- QPS Austria GmbH, Neuropharmacology, Grambach, Austria
| | - Michael Schöll
- Wallenberg Centre for Molecular and Translational Medicine, University of Gothenburg, Gothenburg, Sweden.,Department of Psychiatry and Neurochemistry, University of Gothenburg, Gothenburg, Sweden.,Department of Neurodegenerative Disease, UCL Queen Square, Institute of Neurology, University College London, London, UK
| | - Björn Marcus Burmann
- Wallenberg Centre for Molecular and Translational Medicine, University of Gothenburg, Gothenburg, Sweden.,Department of Chemistry and Molecular Biology, University of Gothenburg, Gothenburg, Sweden
| | - Thomas Vogels
- Maptimmune BV, The Hague, The Netherlands.,Department of Psychiatry and Neurochemistry, University of Gothenburg, Gothenburg, Sweden.,Department of Neurodegenerative Disease, UCL Queen Square, Institute of Neurology, University College London, London, UK
| |
Collapse
|
13
|
Wu KM, Zhang YR, Huang YY, Dong Q, Tan L, Yu JT. The role of the immune system in Alzheimer's disease. Ageing Res Rev 2021; 70:101409. [PMID: 34273589 DOI: 10.1016/j.arr.2021.101409] [Citation(s) in RCA: 46] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2021] [Revised: 07/09/2021] [Accepted: 07/12/2021] [Indexed: 02/06/2023]
Abstract
Alzheimer's disease (AD) is the most common neurodegenerative disorder where the accumulation of amyloid plaques and the formation of tau tangles are the prominent pathological hallmarks. Increasing preclinical and clinical studies have revealed that different components of the immune system may act as important contributors to AD etiology and pathogenesis. The recognition of misfolded Aβ and tau by immune cells can trigger a series of complex immune responses in AD, and then lead to neuroinflammation and neurodegeneration. In parallel, genome-wide association studies have also identified several immune related loci associated with increased - risk of AD by interfering with the function of immune cells. Other immune related factors, such as impaired immunometabolism, defective meningeal lymphatic vessels and autoimmunity might also be involved in the pathogenesis of AD. Here, we review the data showing the alterations of immune cells in the AD trajectory and seek to demonstrate the crosstalk between the immune cell dysfunction and AD pathology. We then discuss the most relevant research findings in regards to the influences of gene susceptibility of immune cells for AD. We also consider impaired meningeal lymphatics, immunometabolism and autoimmune mechanisms in AD. In addition, immune related biomarkers and immunotherapies for AD are also mentioned in order to offer novel insights for future research.
Collapse
|
14
|
Therapeutic Ultrasound as a Treatment Modality for Physiological and Pathological Ageing Including Alzheimer's Disease. Pharmaceutics 2021; 13:pharmaceutics13071002. [PMID: 34371696 PMCID: PMC8309087 DOI: 10.3390/pharmaceutics13071002] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Revised: 06/29/2021] [Accepted: 06/29/2021] [Indexed: 01/09/2023] Open
Abstract
Physiological and pathological ageing (as exemplified by Alzheimer's disease, AD) are characterized by a progressive decline that also includes cognition. How this decline can be slowed or even reversed is a critical question. Here, we discuss therapeutic ultrasound as a novel modality to achieve this goal. In our studies, we explored three fundamental strategies, (i) scanning ultrasound on its own (SUSonly), (ii) therapeutic ultrasound in concert with intravenously injected microbubbles (which transiently opens the blood-brain barrier, SUS+MB), and (iii) SUS+MB in combination with therapeutic antibodies (SUS+MB+mAb). These studies show SUS+MB effectively clears amyloid and restores memory in amyloid-depositing mice and partially clears Tau and ameliorates memory impairments in Tau transgenic mice, with additional improvements found in combination trials (SUS+MB+mAb). Interestingly, both SUSonly and SUS+MB restored the induction of long-term potentiation (LTP, electrophysiological correlate of memory) in senescent wild-type mice. Both lead to increased neurogenesis, and SUSonly, in particular, resulted in improved spatial memory. We discuss these findings side-by-side with our findings obtained in AD mouse models. We conclude that therapeutic ultrasound is a non-invasive, pleiotropic modality that may present a treatment option not only for AD but also for enhancing cognition in physiological ageing.
Collapse
|
15
|
Bao C, Bajrami B, Marcotte DJ, Chodaparambil JV, Kerns HM, Henderson J, Wei R, Gao B, Dillon GM. Mechanisms of Regulation and Diverse Activities of Tau-Tubulin Kinase (TTBK) Isoforms. Cell Mol Neurobiol 2021; 41:669-685. [PMID: 32424773 DOI: 10.1007/s10571-020-00875-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2020] [Accepted: 05/11/2020] [Indexed: 10/24/2022]
Abstract
Tau-tubulin kinase 1 (TTBK1) is a CNS-specific, kinase that has been implicated in the pathological phosphorylation of tau in Alzheimer's Disease (AD) and Frontotemporal Dementia (FTD). TTBK1 is a challenging therapeutic target because it shares a highly conserved catalytic domain with its homolog, TTBK2, a ubiquitously expressed kinase genetically linked to the disease spinocerebellar ataxia type 11. The present study attempts to elucidate the functional distinctions between the TTBK isoforms and increase our understanding of them as distinct targets for the treatment of neurodegenerative disease. We demonstrate that in cortical neurons, TTBK1, not TTBK2, is the isoform responsible for tau phosphorylation at epitopes enriched in tauopathies such as Serine 422. In addition, although our elucidation of the crystal structure of the TTBK2 kinase domain indicates almost identical structural similarity with TTBK1, biochemical and cellular assays demonstrate that the enzymatic activity of these two proteins is regulated by a combination of unique extra-catalytic sequences and autophosphorylation events. Finally, we have identified an unbiased list of neuronal interactors and phosphorylation substrates for TTBK1 and TTBK2 that highlight the unique cellular pathways and functional networks that each isoform is involved in. This data address an important gap in knowledge regarding the implications of targeting TTBK kinases and may prove valuable in the development of potential therapies for disease.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Ru Wei
- Biogen, Cambridge, MA, 02134, USA
| | | | | |
Collapse
|
16
|
Arbaciauskaite M, Lei Y, Cho YK. High-specificity antibodies and detection methods for quantifying phosphorylated tau from clinical samples. Antib Ther 2021; 4:34-44. [PMID: 33928234 PMCID: PMC7944500 DOI: 10.1093/abt/tbab004] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2020] [Revised: 01/14/2021] [Accepted: 02/05/2021] [Indexed: 12/14/2022] Open
Abstract
The ability to measure total and phosphorylated tau levels in clinical samples is transforming the detection of Alzheimer’s disease (AD) and other neurodegenerative diseases. In particular, recent reports indicate that accurate detection of low levels of phosphorylated tau (p-tau) in plasma provides a reliable biomarker of AD long before sensing memory loss. Therefore, the diagnosis and monitoring of neurodegenerative diseases progression using blood samples is becoming a reality. These major advances were achieved by using antibodies specific to p-tau as well as sophisticated high-sensitivity immunoassay platforms. This review focuses on these enabling advances in high-specificity antibody development, engineering, and novel signal detection methods. We will draw insights from structural studies on p-tau antibodies, engineering efforts to improve their binding properties, and efforts to validate their specificity. A comprehensive survey of high-sensitivity p-tau immunoassay platforms along with sensitivity limits will be provided. We conclude that although robust approaches for detecting certain p-tau species have been established, systematic efforts to validate antibodies for assay development is still needed for the recognition of biomarkers for AD and other neurodegenerative diseases.
Collapse
Affiliation(s)
- Monika Arbaciauskaite
- Department of Chemical and Biomolecular Engineering, University of Connecticut, Storrs, CT 06269, USA
| | - Yu Lei
- Department of Chemical and Biomolecular Engineering, University of Connecticut, Storrs, CT 06269, USA
| | - Yong Ku Cho
- Department of Chemical and Biomolecular Engineering, University of Connecticut, Storrs, CT 06269, USA
| |
Collapse
|
17
|
Savastano A, Jaipuria G, Andreas L, Mandelkow E, Zweckstetter M. Solid-state NMR investigation of the involvement of the P2 region in tau amyloid fibrils. Sci Rep 2020; 10:21210. [PMID: 33273615 PMCID: PMC7712923 DOI: 10.1038/s41598-020-78161-0] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Accepted: 11/20/2020] [Indexed: 11/08/2022] Open
Abstract
The aggregation of hyperphosphorylated tau into amyloid fibrils is closely linked to the progression of Alzheimer's disease. To gain insight into the link between amyloid structure and disease, the three-dimensional structure of tau fibrils has been studied using solid-state NMR (ssNMR) and cryogenic electron microscopy (cryo-EM). The proline-rich region of tau remains poorly defined in the context of tau amyloid structures, despite the clustering of several phosphorylation sites, which have been associated with Alzheimer's disease. In order to gain insight into the contribution of the proline-rich region P2 of tau to amyloid fibrils, we studied in vitro aggregated amyloid fibrils of tau constructs, which contain both the proline-rich region P2 and the pseudo-repeats. Using ssNMR we show that the sequence [Formula: see text], the most hydrophobic patch within the P2 region, loses its flexibility upon formation of amyloid fibrils. The data suggest a contribution of the P2 region to tau amyloid fibril formation, which might account for some of the unassigned electron density in cryo-EM studies of tau fibrils and could be modulated by tau phosphorylation at the disease-associated AT180 epitope T231/S235.
Collapse
Affiliation(s)
- Adriana Savastano
- German Center for Neurodegenerative Diseases (DZNE), Von-Siebold-Str. 3a, 37075, Göttingen, Germany
| | - Garima Jaipuria
- German Center for Neurodegenerative Diseases (DZNE), Von-Siebold-Str. 3a, 37075, Göttingen, Germany
| | - Loren Andreas
- Max Planck Institute for Biophysical Chemistry, Am Faßberg 11, 37077, Göttingen, Germany
| | - Eckhard Mandelkow
- German Center for Neurodegenerative Diseases (DZNE), Venusberg-Campus 1, 53127, Bonn, Germany
- CAESAR Research Center, Ludwig-Erhard-Allee 2, 53175, Bonn, Germany
| | - Markus Zweckstetter
- German Center for Neurodegenerative Diseases (DZNE), Von-Siebold-Str. 3a, 37075, Göttingen, Germany.
- Max Planck Institute for Biophysical Chemistry, Am Faßberg 11, 37077, Göttingen, Germany.
| |
Collapse
|
18
|
Silva MC, Haggarty SJ. Tauopathies: Deciphering Disease Mechanisms to Develop Effective Therapies. Int J Mol Sci 2020; 21:ijms21238948. [PMID: 33255694 PMCID: PMC7728099 DOI: 10.3390/ijms21238948] [Citation(s) in RCA: 50] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Revised: 11/20/2020] [Accepted: 11/22/2020] [Indexed: 12/13/2022] Open
Abstract
Tauopathies are neurodegenerative diseases characterized by the pathological accumulation of microtubule-associated protein tau (MAPT) in the form of neurofibrillary tangles and paired helical filaments in neurons and glia, leading to brain cell death. These diseases include frontotemporal dementia (FTD) and Alzheimer's disease (AD) and can be sporadic or inherited when caused by mutations in the MAPT gene. Despite an incredibly high socio-economic burden worldwide, there are still no effective disease-modifying therapies, and few tau-focused experimental drugs have reached clinical trials. One major hindrance for therapeutic development is the knowledge gap in molecular mechanisms of tau-mediated neuronal toxicity and death. For the promise of precision medicine for brain disorders to be fulfilled, it is necessary to integrate known genetic causes of disease, i.e., MAPT mutations, with an understanding of the dysregulated molecular pathways that constitute potential therapeutic targets. Here, the growing understanding of known and proposed mechanisms of disease etiology will be reviewed, together with promising experimental tau-directed therapeutics, such as recently developed tau degraders. Current challenges faced by the fields of tau research and drug discovery will also be addressed.
Collapse
|
19
|
Avdeeva NV. Novel mGluR4 agonist Rapitalam ameliorates motor dysfunction in mice with tau-associated neurodegeneration. RESEARCH RESULTS IN PHARMACOLOGY 2020. [DOI: 10.3897/rrpharmacology.6.52098] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Introduction: Tau protein is classically involved in the pathogenesis of a neurodegenerative processes, such as Parkinson’s disease. This study was aimed at testing the novel mGluR4 selective agonist using it in transgenic mice with tau-associated neurodegeneration.
Materials and methods: Mice with Human P301S Tau hyperexpression were divided into 3 groups: Rapitalam 6 mg/kg and 20 mg/kg by gavage 3 times a week; and Control (Sham). The motor functions of animals were evaluated at 12th, 14th, 16th, 18th, and 20th weeks of life using the grip-test, rotarod and hanging wire test. In addition, the time of symptoms onset and death was recorded.
Research results: The use of Rapitalam at a dose of 6 mg/kg and 20 mg/kg significantly restored the holding impulse on a hanging wire, increasing it from 5.06±1.25 kg×sec to 6.42±0.97 kg×sec and 8.84±1.17 kg×sec, respectively. A similar trend was observed in the grip test: Rapitalam recovered grip strength from 28.43±5.04 N in the control group to 44.27±5.54 N (6 mg/kg) and 59.53±7.95 (20 mg/kg). Finally, the two-month use of Rapitalam neither delayed the manifestation of symptoms, nor increased the survival of mice.
Discussion: The cause of the loss of nerve cells in the mouse-tau line is autophagy. Apparently, Rapitalam is not able to simulate this process by reducing excitotoxicity, but against the background of the neurodegenerative process, it increases the activity of the nerve cells.
Conclusion: Rapitalam improves motor dysfunction in mice with tauopathy, with no effect on the survival of animals.
Collapse
|
20
|
Dillon GM, Henderson JL, Bao C, Joyce JA, Calhoun M, Amaral B, King KW, Bajrami B, Rabah D. Acute inhibition of the CNS-specific kinase TTBK1 significantly lowers tau phosphorylation at several disease relevant sites. PLoS One 2020; 15:e0228771. [PMID: 32255788 PMCID: PMC7138307 DOI: 10.1371/journal.pone.0228771] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2019] [Accepted: 01/07/2020] [Indexed: 11/24/2022] Open
Abstract
Hyperphosphorylated tau protein is a pathological hallmark of numerous neurodegenerative diseases and the level of tau pathology is correlated with the degree of cognitive impairment. Tau hyper-phosphorylation is thought to be an early initiating event in the cascade leading to tau toxicity and neuronal death. Inhibition of tau phosphorylation therefore represents an attractive therapeutic strategy. However, the widespread expression of most kinases and promiscuity of their substrates, along with poor selectivity of most kinase inhibitors, have resulted in systemic toxicities that have limited the advancement of tau kinase inhibitors into the clinic. We therefore focused on the CNS-specific tau kinase, TTBK1, and investigated whether selective inhibition of this kinase could represent a viable approach to targeting tau phosphorylation in disease. In the current study, we demonstrate that TTBK1 regulates tau phosphorylation using overexpression or knockdown of this kinase in heterologous cells and primary neurons. Importantly, we find that TTBK1-specific phosphorylation of tau leads to a loss of normal protein function including a decrease in tau-tubulin binding and deficits in tubulin polymerization. We then describe the use of a novel, selective small molecule antagonist, BIIB-TTBK1i, to study the acute effects of TTBK1 inhibition on tau phosphorylation in vivo. We demonstrate substantial lowering of tau phosphorylation at multiple sites implicated in disease, suggesting that TTBK1 inhibitors may represent an exciting new approach in the search for neurodegenerative disease therapies.
Collapse
Affiliation(s)
| | | | - Channa Bao
- Biogen, Cambridge, MA, United States of America
| | | | | | | | | | | | - Dania Rabah
- Biogen, Cambridge, MA, United States of America
| |
Collapse
|
21
|
Frontzek K, Carta M, Losa M, Epskamp M, Meisl G, Anane A, Brandel JP, Camenisch U, Castilla J, Haïk S, Knowles T, Lindner E, Lutterotti A, Minikel EV, Roiter I, Safar JG, Sanchez-Valle R, Žáková D, Hornemann S, Aguzzi A. Autoantibodies against the prion protein in individuals with PRNP mutations. Neurology 2020; 95:e2028-e2037. [PMID: 32098855 DOI: 10.1212/wnl.0000000000009183] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2019] [Accepted: 12/04/2019] [Indexed: 12/15/2022] Open
Abstract
OBJECTIVE To determine whether naturally occurring autoantibodies against the prion protein are present in individuals with genetic prion disease mutations and controls, and if so, whether they are protective against prion disease. METHODS In this case-control study, we collected 124 blood samples from individuals with a variety of pathogenic PRNP mutations and 78 control individuals with a positive family history of genetic prion disease but lacking disease-associated PRNP mutations. Antibody reactivity was measured using an indirect ELISA for the detection of human immunoglobulin G1-4 antibodies against wild-type human prion protein. Multivariate linear regression models were constructed to analyze differences in autoantibody reactivity between (1) PRNP mutation carriers vs controls and (2) asymptomatic vs symptomatic PRNP mutation carriers. Robustness of results was examined in matched cohorts. RESULTS We found that antibody reactivity was present in a subset of both PRNP mutation carriers and controls. Autoantibody levels were not influenced by PRNP mutation status or clinical manifestation of prion disease. Post hoc analyses showed anti-PrPC autoantibody titers to be independent of personal history of autoimmune disease and other immunologic disorders, as well as PRNP codon 129 polymorphism. CONCLUSIONS Pathogenic PRNP variants do not notably stimulate antibody-mediated anti-PrPC immunity. Anti-PrPC immunoglobulin G autoantibodies are not associated with the onset of prion disease. The presence of anti-PrPC autoantibodies in the general population without any disease-specific association suggests that relatively high titers of naturally occurring antibodies are well-tolerated. CLINICALTRIALSGOV IDENTIFIER NCT02837705.
Collapse
Affiliation(s)
- Karl Frontzek
- From the Institute of Neuropathology (K.F., M.C., M.L., M.E., S. Hornemann, A.A.), Institute of Surgical Pathology (U.C.), and Department of Neurology, Neuroimmunology and MS Research (NIMS) (A.L.), University of Zurich, Switzerland; Department of Chemistry (G.M., T.K.), University of Cambridge, UK; CJD Foundation Israel (A.A.), Pardes Hanna; ICM (J.-P.B.), Salpêtrière Hospital, Sorbonne University, Paris, France; CIC bioGUNE and IKERBASQUE (J.C.), Basque Foundation for Science, Bizkaia, Spain; Sorbonne University (S. Haïk), ICM, Salpêtrière Hospital, Paris, France; Ophthalmology Division (E.L.), University of Graz, Austria; Broad Institute (E.V.M.), Cambridge, MA; Treviso Hospital (I.R.), Italy; Department of Pathology, Neurology, and National Prion Disease Pathology Surveillance Center (J.G.S.), Case Western Reserve University, Cleveland, OH; Alzheimer's Disease and Other Cognitive Disorders Unit (R.S.-V.), Hospital Clinic, IDIBAPS, University of Barcelona, Spain; and Department of Prion Diseases (D.Ž.), Slovak Medical University, Bratislava, Slovakia.
| | - Manfredi Carta
- From the Institute of Neuropathology (K.F., M.C., M.L., M.E., S. Hornemann, A.A.), Institute of Surgical Pathology (U.C.), and Department of Neurology, Neuroimmunology and MS Research (NIMS) (A.L.), University of Zurich, Switzerland; Department of Chemistry (G.M., T.K.), University of Cambridge, UK; CJD Foundation Israel (A.A.), Pardes Hanna; ICM (J.-P.B.), Salpêtrière Hospital, Sorbonne University, Paris, France; CIC bioGUNE and IKERBASQUE (J.C.), Basque Foundation for Science, Bizkaia, Spain; Sorbonne University (S. Haïk), ICM, Salpêtrière Hospital, Paris, France; Ophthalmology Division (E.L.), University of Graz, Austria; Broad Institute (E.V.M.), Cambridge, MA; Treviso Hospital (I.R.), Italy; Department of Pathology, Neurology, and National Prion Disease Pathology Surveillance Center (J.G.S.), Case Western Reserve University, Cleveland, OH; Alzheimer's Disease and Other Cognitive Disorders Unit (R.S.-V.), Hospital Clinic, IDIBAPS, University of Barcelona, Spain; and Department of Prion Diseases (D.Ž.), Slovak Medical University, Bratislava, Slovakia
| | - Marco Losa
- From the Institute of Neuropathology (K.F., M.C., M.L., M.E., S. Hornemann, A.A.), Institute of Surgical Pathology (U.C.), and Department of Neurology, Neuroimmunology and MS Research (NIMS) (A.L.), University of Zurich, Switzerland; Department of Chemistry (G.M., T.K.), University of Cambridge, UK; CJD Foundation Israel (A.A.), Pardes Hanna; ICM (J.-P.B.), Salpêtrière Hospital, Sorbonne University, Paris, France; CIC bioGUNE and IKERBASQUE (J.C.), Basque Foundation for Science, Bizkaia, Spain; Sorbonne University (S. Haïk), ICM, Salpêtrière Hospital, Paris, France; Ophthalmology Division (E.L.), University of Graz, Austria; Broad Institute (E.V.M.), Cambridge, MA; Treviso Hospital (I.R.), Italy; Department of Pathology, Neurology, and National Prion Disease Pathology Surveillance Center (J.G.S.), Case Western Reserve University, Cleveland, OH; Alzheimer's Disease and Other Cognitive Disorders Unit (R.S.-V.), Hospital Clinic, IDIBAPS, University of Barcelona, Spain; and Department of Prion Diseases (D.Ž.), Slovak Medical University, Bratislava, Slovakia
| | - Mirka Epskamp
- From the Institute of Neuropathology (K.F., M.C., M.L., M.E., S. Hornemann, A.A.), Institute of Surgical Pathology (U.C.), and Department of Neurology, Neuroimmunology and MS Research (NIMS) (A.L.), University of Zurich, Switzerland; Department of Chemistry (G.M., T.K.), University of Cambridge, UK; CJD Foundation Israel (A.A.), Pardes Hanna; ICM (J.-P.B.), Salpêtrière Hospital, Sorbonne University, Paris, France; CIC bioGUNE and IKERBASQUE (J.C.), Basque Foundation for Science, Bizkaia, Spain; Sorbonne University (S. Haïk), ICM, Salpêtrière Hospital, Paris, France; Ophthalmology Division (E.L.), University of Graz, Austria; Broad Institute (E.V.M.), Cambridge, MA; Treviso Hospital (I.R.), Italy; Department of Pathology, Neurology, and National Prion Disease Pathology Surveillance Center (J.G.S.), Case Western Reserve University, Cleveland, OH; Alzheimer's Disease and Other Cognitive Disorders Unit (R.S.-V.), Hospital Clinic, IDIBAPS, University of Barcelona, Spain; and Department of Prion Diseases (D.Ž.), Slovak Medical University, Bratislava, Slovakia
| | - Georg Meisl
- From the Institute of Neuropathology (K.F., M.C., M.L., M.E., S. Hornemann, A.A.), Institute of Surgical Pathology (U.C.), and Department of Neurology, Neuroimmunology and MS Research (NIMS) (A.L.), University of Zurich, Switzerland; Department of Chemistry (G.M., T.K.), University of Cambridge, UK; CJD Foundation Israel (A.A.), Pardes Hanna; ICM (J.-P.B.), Salpêtrière Hospital, Sorbonne University, Paris, France; CIC bioGUNE and IKERBASQUE (J.C.), Basque Foundation for Science, Bizkaia, Spain; Sorbonne University (S. Haïk), ICM, Salpêtrière Hospital, Paris, France; Ophthalmology Division (E.L.), University of Graz, Austria; Broad Institute (E.V.M.), Cambridge, MA; Treviso Hospital (I.R.), Italy; Department of Pathology, Neurology, and National Prion Disease Pathology Surveillance Center (J.G.S.), Case Western Reserve University, Cleveland, OH; Alzheimer's Disease and Other Cognitive Disorders Unit (R.S.-V.), Hospital Clinic, IDIBAPS, University of Barcelona, Spain; and Department of Prion Diseases (D.Ž.), Slovak Medical University, Bratislava, Slovakia
| | - Alice Anane
- From the Institute of Neuropathology (K.F., M.C., M.L., M.E., S. Hornemann, A.A.), Institute of Surgical Pathology (U.C.), and Department of Neurology, Neuroimmunology and MS Research (NIMS) (A.L.), University of Zurich, Switzerland; Department of Chemistry (G.M., T.K.), University of Cambridge, UK; CJD Foundation Israel (A.A.), Pardes Hanna; ICM (J.-P.B.), Salpêtrière Hospital, Sorbonne University, Paris, France; CIC bioGUNE and IKERBASQUE (J.C.), Basque Foundation for Science, Bizkaia, Spain; Sorbonne University (S. Haïk), ICM, Salpêtrière Hospital, Paris, France; Ophthalmology Division (E.L.), University of Graz, Austria; Broad Institute (E.V.M.), Cambridge, MA; Treviso Hospital (I.R.), Italy; Department of Pathology, Neurology, and National Prion Disease Pathology Surveillance Center (J.G.S.), Case Western Reserve University, Cleveland, OH; Alzheimer's Disease and Other Cognitive Disorders Unit (R.S.-V.), Hospital Clinic, IDIBAPS, University of Barcelona, Spain; and Department of Prion Diseases (D.Ž.), Slovak Medical University, Bratislava, Slovakia
| | - Jean-Philippe Brandel
- From the Institute of Neuropathology (K.F., M.C., M.L., M.E., S. Hornemann, A.A.), Institute of Surgical Pathology (U.C.), and Department of Neurology, Neuroimmunology and MS Research (NIMS) (A.L.), University of Zurich, Switzerland; Department of Chemistry (G.M., T.K.), University of Cambridge, UK; CJD Foundation Israel (A.A.), Pardes Hanna; ICM (J.-P.B.), Salpêtrière Hospital, Sorbonne University, Paris, France; CIC bioGUNE and IKERBASQUE (J.C.), Basque Foundation for Science, Bizkaia, Spain; Sorbonne University (S. Haïk), ICM, Salpêtrière Hospital, Paris, France; Ophthalmology Division (E.L.), University of Graz, Austria; Broad Institute (E.V.M.), Cambridge, MA; Treviso Hospital (I.R.), Italy; Department of Pathology, Neurology, and National Prion Disease Pathology Surveillance Center (J.G.S.), Case Western Reserve University, Cleveland, OH; Alzheimer's Disease and Other Cognitive Disorders Unit (R.S.-V.), Hospital Clinic, IDIBAPS, University of Barcelona, Spain; and Department of Prion Diseases (D.Ž.), Slovak Medical University, Bratislava, Slovakia
| | - Ulrike Camenisch
- From the Institute of Neuropathology (K.F., M.C., M.L., M.E., S. Hornemann, A.A.), Institute of Surgical Pathology (U.C.), and Department of Neurology, Neuroimmunology and MS Research (NIMS) (A.L.), University of Zurich, Switzerland; Department of Chemistry (G.M., T.K.), University of Cambridge, UK; CJD Foundation Israel (A.A.), Pardes Hanna; ICM (J.-P.B.), Salpêtrière Hospital, Sorbonne University, Paris, France; CIC bioGUNE and IKERBASQUE (J.C.), Basque Foundation for Science, Bizkaia, Spain; Sorbonne University (S. Haïk), ICM, Salpêtrière Hospital, Paris, France; Ophthalmology Division (E.L.), University of Graz, Austria; Broad Institute (E.V.M.), Cambridge, MA; Treviso Hospital (I.R.), Italy; Department of Pathology, Neurology, and National Prion Disease Pathology Surveillance Center (J.G.S.), Case Western Reserve University, Cleveland, OH; Alzheimer's Disease and Other Cognitive Disorders Unit (R.S.-V.), Hospital Clinic, IDIBAPS, University of Barcelona, Spain; and Department of Prion Diseases (D.Ž.), Slovak Medical University, Bratislava, Slovakia
| | - Joaquín Castilla
- From the Institute of Neuropathology (K.F., M.C., M.L., M.E., S. Hornemann, A.A.), Institute of Surgical Pathology (U.C.), and Department of Neurology, Neuroimmunology and MS Research (NIMS) (A.L.), University of Zurich, Switzerland; Department of Chemistry (G.M., T.K.), University of Cambridge, UK; CJD Foundation Israel (A.A.), Pardes Hanna; ICM (J.-P.B.), Salpêtrière Hospital, Sorbonne University, Paris, France; CIC bioGUNE and IKERBASQUE (J.C.), Basque Foundation for Science, Bizkaia, Spain; Sorbonne University (S. Haïk), ICM, Salpêtrière Hospital, Paris, France; Ophthalmology Division (E.L.), University of Graz, Austria; Broad Institute (E.V.M.), Cambridge, MA; Treviso Hospital (I.R.), Italy; Department of Pathology, Neurology, and National Prion Disease Pathology Surveillance Center (J.G.S.), Case Western Reserve University, Cleveland, OH; Alzheimer's Disease and Other Cognitive Disorders Unit (R.S.-V.), Hospital Clinic, IDIBAPS, University of Barcelona, Spain; and Department of Prion Diseases (D.Ž.), Slovak Medical University, Bratislava, Slovakia
| | - Stéphane Haïk
- From the Institute of Neuropathology (K.F., M.C., M.L., M.E., S. Hornemann, A.A.), Institute of Surgical Pathology (U.C.), and Department of Neurology, Neuroimmunology and MS Research (NIMS) (A.L.), University of Zurich, Switzerland; Department of Chemistry (G.M., T.K.), University of Cambridge, UK; CJD Foundation Israel (A.A.), Pardes Hanna; ICM (J.-P.B.), Salpêtrière Hospital, Sorbonne University, Paris, France; CIC bioGUNE and IKERBASQUE (J.C.), Basque Foundation for Science, Bizkaia, Spain; Sorbonne University (S. Haïk), ICM, Salpêtrière Hospital, Paris, France; Ophthalmology Division (E.L.), University of Graz, Austria; Broad Institute (E.V.M.), Cambridge, MA; Treviso Hospital (I.R.), Italy; Department of Pathology, Neurology, and National Prion Disease Pathology Surveillance Center (J.G.S.), Case Western Reserve University, Cleveland, OH; Alzheimer's Disease and Other Cognitive Disorders Unit (R.S.-V.), Hospital Clinic, IDIBAPS, University of Barcelona, Spain; and Department of Prion Diseases (D.Ž.), Slovak Medical University, Bratislava, Slovakia
| | - Tuomas Knowles
- From the Institute of Neuropathology (K.F., M.C., M.L., M.E., S. Hornemann, A.A.), Institute of Surgical Pathology (U.C.), and Department of Neurology, Neuroimmunology and MS Research (NIMS) (A.L.), University of Zurich, Switzerland; Department of Chemistry (G.M., T.K.), University of Cambridge, UK; CJD Foundation Israel (A.A.), Pardes Hanna; ICM (J.-P.B.), Salpêtrière Hospital, Sorbonne University, Paris, France; CIC bioGUNE and IKERBASQUE (J.C.), Basque Foundation for Science, Bizkaia, Spain; Sorbonne University (S. Haïk), ICM, Salpêtrière Hospital, Paris, France; Ophthalmology Division (E.L.), University of Graz, Austria; Broad Institute (E.V.M.), Cambridge, MA; Treviso Hospital (I.R.), Italy; Department of Pathology, Neurology, and National Prion Disease Pathology Surveillance Center (J.G.S.), Case Western Reserve University, Cleveland, OH; Alzheimer's Disease and Other Cognitive Disorders Unit (R.S.-V.), Hospital Clinic, IDIBAPS, University of Barcelona, Spain; and Department of Prion Diseases (D.Ž.), Slovak Medical University, Bratislava, Slovakia
| | - Ewald Lindner
- From the Institute of Neuropathology (K.F., M.C., M.L., M.E., S. Hornemann, A.A.), Institute of Surgical Pathology (U.C.), and Department of Neurology, Neuroimmunology and MS Research (NIMS) (A.L.), University of Zurich, Switzerland; Department of Chemistry (G.M., T.K.), University of Cambridge, UK; CJD Foundation Israel (A.A.), Pardes Hanna; ICM (J.-P.B.), Salpêtrière Hospital, Sorbonne University, Paris, France; CIC bioGUNE and IKERBASQUE (J.C.), Basque Foundation for Science, Bizkaia, Spain; Sorbonne University (S. Haïk), ICM, Salpêtrière Hospital, Paris, France; Ophthalmology Division (E.L.), University of Graz, Austria; Broad Institute (E.V.M.), Cambridge, MA; Treviso Hospital (I.R.), Italy; Department of Pathology, Neurology, and National Prion Disease Pathology Surveillance Center (J.G.S.), Case Western Reserve University, Cleveland, OH; Alzheimer's Disease and Other Cognitive Disorders Unit (R.S.-V.), Hospital Clinic, IDIBAPS, University of Barcelona, Spain; and Department of Prion Diseases (D.Ž.), Slovak Medical University, Bratislava, Slovakia
| | - Andreas Lutterotti
- From the Institute of Neuropathology (K.F., M.C., M.L., M.E., S. Hornemann, A.A.), Institute of Surgical Pathology (U.C.), and Department of Neurology, Neuroimmunology and MS Research (NIMS) (A.L.), University of Zurich, Switzerland; Department of Chemistry (G.M., T.K.), University of Cambridge, UK; CJD Foundation Israel (A.A.), Pardes Hanna; ICM (J.-P.B.), Salpêtrière Hospital, Sorbonne University, Paris, France; CIC bioGUNE and IKERBASQUE (J.C.), Basque Foundation for Science, Bizkaia, Spain; Sorbonne University (S. Haïk), ICM, Salpêtrière Hospital, Paris, France; Ophthalmology Division (E.L.), University of Graz, Austria; Broad Institute (E.V.M.), Cambridge, MA; Treviso Hospital (I.R.), Italy; Department of Pathology, Neurology, and National Prion Disease Pathology Surveillance Center (J.G.S.), Case Western Reserve University, Cleveland, OH; Alzheimer's Disease and Other Cognitive Disorders Unit (R.S.-V.), Hospital Clinic, IDIBAPS, University of Barcelona, Spain; and Department of Prion Diseases (D.Ž.), Slovak Medical University, Bratislava, Slovakia
| | - Eric Vallabh Minikel
- From the Institute of Neuropathology (K.F., M.C., M.L., M.E., S. Hornemann, A.A.), Institute of Surgical Pathology (U.C.), and Department of Neurology, Neuroimmunology and MS Research (NIMS) (A.L.), University of Zurich, Switzerland; Department of Chemistry (G.M., T.K.), University of Cambridge, UK; CJD Foundation Israel (A.A.), Pardes Hanna; ICM (J.-P.B.), Salpêtrière Hospital, Sorbonne University, Paris, France; CIC bioGUNE and IKERBASQUE (J.C.), Basque Foundation for Science, Bizkaia, Spain; Sorbonne University (S. Haïk), ICM, Salpêtrière Hospital, Paris, France; Ophthalmology Division (E.L.), University of Graz, Austria; Broad Institute (E.V.M.), Cambridge, MA; Treviso Hospital (I.R.), Italy; Department of Pathology, Neurology, and National Prion Disease Pathology Surveillance Center (J.G.S.), Case Western Reserve University, Cleveland, OH; Alzheimer's Disease and Other Cognitive Disorders Unit (R.S.-V.), Hospital Clinic, IDIBAPS, University of Barcelona, Spain; and Department of Prion Diseases (D.Ž.), Slovak Medical University, Bratislava, Slovakia
| | - Ignazio Roiter
- From the Institute of Neuropathology (K.F., M.C., M.L., M.E., S. Hornemann, A.A.), Institute of Surgical Pathology (U.C.), and Department of Neurology, Neuroimmunology and MS Research (NIMS) (A.L.), University of Zurich, Switzerland; Department of Chemistry (G.M., T.K.), University of Cambridge, UK; CJD Foundation Israel (A.A.), Pardes Hanna; ICM (J.-P.B.), Salpêtrière Hospital, Sorbonne University, Paris, France; CIC bioGUNE and IKERBASQUE (J.C.), Basque Foundation for Science, Bizkaia, Spain; Sorbonne University (S. Haïk), ICM, Salpêtrière Hospital, Paris, France; Ophthalmology Division (E.L.), University of Graz, Austria; Broad Institute (E.V.M.), Cambridge, MA; Treviso Hospital (I.R.), Italy; Department of Pathology, Neurology, and National Prion Disease Pathology Surveillance Center (J.G.S.), Case Western Reserve University, Cleveland, OH; Alzheimer's Disease and Other Cognitive Disorders Unit (R.S.-V.), Hospital Clinic, IDIBAPS, University of Barcelona, Spain; and Department of Prion Diseases (D.Ž.), Slovak Medical University, Bratislava, Slovakia
| | - Jiri G Safar
- From the Institute of Neuropathology (K.F., M.C., M.L., M.E., S. Hornemann, A.A.), Institute of Surgical Pathology (U.C.), and Department of Neurology, Neuroimmunology and MS Research (NIMS) (A.L.), University of Zurich, Switzerland; Department of Chemistry (G.M., T.K.), University of Cambridge, UK; CJD Foundation Israel (A.A.), Pardes Hanna; ICM (J.-P.B.), Salpêtrière Hospital, Sorbonne University, Paris, France; CIC bioGUNE and IKERBASQUE (J.C.), Basque Foundation for Science, Bizkaia, Spain; Sorbonne University (S. Haïk), ICM, Salpêtrière Hospital, Paris, France; Ophthalmology Division (E.L.), University of Graz, Austria; Broad Institute (E.V.M.), Cambridge, MA; Treviso Hospital (I.R.), Italy; Department of Pathology, Neurology, and National Prion Disease Pathology Surveillance Center (J.G.S.), Case Western Reserve University, Cleveland, OH; Alzheimer's Disease and Other Cognitive Disorders Unit (R.S.-V.), Hospital Clinic, IDIBAPS, University of Barcelona, Spain; and Department of Prion Diseases (D.Ž.), Slovak Medical University, Bratislava, Slovakia
| | - Raquel Sanchez-Valle
- From the Institute of Neuropathology (K.F., M.C., M.L., M.E., S. Hornemann, A.A.), Institute of Surgical Pathology (U.C.), and Department of Neurology, Neuroimmunology and MS Research (NIMS) (A.L.), University of Zurich, Switzerland; Department of Chemistry (G.M., T.K.), University of Cambridge, UK; CJD Foundation Israel (A.A.), Pardes Hanna; ICM (J.-P.B.), Salpêtrière Hospital, Sorbonne University, Paris, France; CIC bioGUNE and IKERBASQUE (J.C.), Basque Foundation for Science, Bizkaia, Spain; Sorbonne University (S. Haïk), ICM, Salpêtrière Hospital, Paris, France; Ophthalmology Division (E.L.), University of Graz, Austria; Broad Institute (E.V.M.), Cambridge, MA; Treviso Hospital (I.R.), Italy; Department of Pathology, Neurology, and National Prion Disease Pathology Surveillance Center (J.G.S.), Case Western Reserve University, Cleveland, OH; Alzheimer's Disease and Other Cognitive Disorders Unit (R.S.-V.), Hospital Clinic, IDIBAPS, University of Barcelona, Spain; and Department of Prion Diseases (D.Ž.), Slovak Medical University, Bratislava, Slovakia
| | - Dana Žáková
- From the Institute of Neuropathology (K.F., M.C., M.L., M.E., S. Hornemann, A.A.), Institute of Surgical Pathology (U.C.), and Department of Neurology, Neuroimmunology and MS Research (NIMS) (A.L.), University of Zurich, Switzerland; Department of Chemistry (G.M., T.K.), University of Cambridge, UK; CJD Foundation Israel (A.A.), Pardes Hanna; ICM (J.-P.B.), Salpêtrière Hospital, Sorbonne University, Paris, France; CIC bioGUNE and IKERBASQUE (J.C.), Basque Foundation for Science, Bizkaia, Spain; Sorbonne University (S. Haïk), ICM, Salpêtrière Hospital, Paris, France; Ophthalmology Division (E.L.), University of Graz, Austria; Broad Institute (E.V.M.), Cambridge, MA; Treviso Hospital (I.R.), Italy; Department of Pathology, Neurology, and National Prion Disease Pathology Surveillance Center (J.G.S.), Case Western Reserve University, Cleveland, OH; Alzheimer's Disease and Other Cognitive Disorders Unit (R.S.-V.), Hospital Clinic, IDIBAPS, University of Barcelona, Spain; and Department of Prion Diseases (D.Ž.), Slovak Medical University, Bratislava, Slovakia
| | - Simone Hornemann
- From the Institute of Neuropathology (K.F., M.C., M.L., M.E., S. Hornemann, A.A.), Institute of Surgical Pathology (U.C.), and Department of Neurology, Neuroimmunology and MS Research (NIMS) (A.L.), University of Zurich, Switzerland; Department of Chemistry (G.M., T.K.), University of Cambridge, UK; CJD Foundation Israel (A.A.), Pardes Hanna; ICM (J.-P.B.), Salpêtrière Hospital, Sorbonne University, Paris, France; CIC bioGUNE and IKERBASQUE (J.C.), Basque Foundation for Science, Bizkaia, Spain; Sorbonne University (S. Haïk), ICM, Salpêtrière Hospital, Paris, France; Ophthalmology Division (E.L.), University of Graz, Austria; Broad Institute (E.V.M.), Cambridge, MA; Treviso Hospital (I.R.), Italy; Department of Pathology, Neurology, and National Prion Disease Pathology Surveillance Center (J.G.S.), Case Western Reserve University, Cleveland, OH; Alzheimer's Disease and Other Cognitive Disorders Unit (R.S.-V.), Hospital Clinic, IDIBAPS, University of Barcelona, Spain; and Department of Prion Diseases (D.Ž.), Slovak Medical University, Bratislava, Slovakia
| | - Adriano Aguzzi
- From the Institute of Neuropathology (K.F., M.C., M.L., M.E., S. Hornemann, A.A.), Institute of Surgical Pathology (U.C.), and Department of Neurology, Neuroimmunology and MS Research (NIMS) (A.L.), University of Zurich, Switzerland; Department of Chemistry (G.M., T.K.), University of Cambridge, UK; CJD Foundation Israel (A.A.), Pardes Hanna; ICM (J.-P.B.), Salpêtrière Hospital, Sorbonne University, Paris, France; CIC bioGUNE and IKERBASQUE (J.C.), Basque Foundation for Science, Bizkaia, Spain; Sorbonne University (S. Haïk), ICM, Salpêtrière Hospital, Paris, France; Ophthalmology Division (E.L.), University of Graz, Austria; Broad Institute (E.V.M.), Cambridge, MA; Treviso Hospital (I.R.), Italy; Department of Pathology, Neurology, and National Prion Disease Pathology Surveillance Center (J.G.S.), Case Western Reserve University, Cleveland, OH; Alzheimer's Disease and Other Cognitive Disorders Unit (R.S.-V.), Hospital Clinic, IDIBAPS, University of Barcelona, Spain; and Department of Prion Diseases (D.Ž.), Slovak Medical University, Bratislava, Slovakia.
| | | |
Collapse
|
22
|
B cells in autoimmune and neurodegenerative central nervous system diseases. Nat Rev Neurosci 2019; 20:728-745. [DOI: 10.1038/s41583-019-0233-2] [Citation(s) in RCA: 128] [Impact Index Per Article: 25.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/30/2019] [Indexed: 12/16/2022]
|
23
|
Albus A, Jördens M, Möller M, Dodel R. Encoding the Sequence of Specific Autoantibodies Against beta-Amyloid and alpha-Synuclein in Neurodegenerative Diseases. Front Immunol 2019; 10:2033. [PMID: 31507618 PMCID: PMC6718452 DOI: 10.3389/fimmu.2019.02033] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2019] [Accepted: 08/12/2019] [Indexed: 01/18/2023] Open
Abstract
There is no effective disease-modifying therapy for Alzheimer's or Parkinson's disease. As pathological hallmarks, the specific peptide amyloid-β and the specific protein α-Synuclein aggregate and deposit in and destabilize neurons, which lead to their degeneration. Within the context of a potential immunization strategy for these diseases, naturally occurring autoantibodies could play a crucial role in treatment due to their ability to inhibit peptide/protein aggregation and mediate their phagocytosis. We developed a procedure to extract the genetic information of such amyloid-β- and α-Synuclein- specific naturally occurring autoantibodies for future passive immunization strategies. We performed FACS-based single-cell sorting on whole blood donated from healthy individuals and performed single-cell RT-PCR analysis to amplify the coding sequences of antigen-binding regions of each antibody-secreting B1 cell. Sequences were further analyzed to determine CDR sequences and germline expression. Therefore, only low percentages of B1 cells obtained were amyloid-β+/α-Synuclein+. After cell sorting, the variable regions of full IgGs were sequenced, demonstrating preferred usage of IGVH3 and IGKV1. The study we present herein describes an approaching for extracting and amplifying the sequence information of autoantibodies based on single-cell analysis of donated blood and producing a recombinant antibody pool for potential passive immunization against neurodegenerative diseases. We sorted a small pool of CD20+ CD27+ CD43+ CD69− IgG+ and Aβ+/α-Syn+ B cells.
Collapse
Affiliation(s)
- Alexandra Albus
- Chair of Geriatric Medicine, University Hospital Essen, University Duisburg-Essen, Essen, Germany.,Department of Neurology, Philipps-University, Marburg, Germany
| | - Marit Jördens
- Department of Neurology, Philipps-University, Marburg, Germany
| | - Moritz Möller
- Department of Neurology, Philipps-University, Marburg, Germany
| | - Richard Dodel
- Chair of Geriatric Medicine, University Hospital Essen, University Duisburg-Essen, Essen, Germany.,Department of Neurology, Philipps-University, Marburg, Germany
| |
Collapse
|
24
|
Lindestam Arlehamn CS, Pham J, Alcalay RN, Frazier A, Shorr E, Carpenter C, Sidney J, Dhanwani R, Agin-Liebes J, Garretti F, Amara AW, Standaert DG, Phillips EJ, Mallal SA, Peters B, Sulzer D, Sette A. Widespread Tau-Specific CD4 T Cell Reactivity in the General Population. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2019; 203:84-92. [PMID: 31085590 PMCID: PMC6581570 DOI: 10.4049/jimmunol.1801506] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/13/2018] [Accepted: 04/20/2019] [Indexed: 12/11/2022]
Abstract
Tau protein is found to be aggregated and hyperphosphorylated (p-tau) in many neurologic disorders, including Parkinson disease (PD) and related parkinsonisms, Alzheimer disease, traumatic brain injury, and even in normal aging. Although not known to produce autoimmune responses, we hypothesized that the appearance of aggregated tau and p-tau with disease could activate the immune system. We thus compared T cell responses to tau and p-tau-derived peptides between PD patients, age-matched healthy controls, and young healthy controls (<35 y old; who are less likely to have high levels of tau aggregates). All groups exhibited CD4+ T cell responses to tau-derived peptides, which were associated with secretion of IFN-γ, IL-5, and/or IL-4. The PD and control participants exhibited a similar magnitude and breadth of responses. Some tau-derived epitopes, consisting of both unmodified and p-tau residues, were more highly represented in PD participants. These results were verified in an independent set of PD and control donors (either age-matched or young controls). Thus, T cells recognizing tau epitopes escape central and peripheral tolerance in relatively high numbers, and the magnitude and nature of these responses are not modulated by age or PD disease.
Collapse
Affiliation(s)
| | - John Pham
- Division of Vaccine Discovery, La Jolla Institute for Immunology, La Jolla, CA 92037
| | - Roy N Alcalay
- Department of Neurology, Columbia University Irving Medical Center, New York State Psychiatric Institute, New York, NY 10032
| | - April Frazier
- Division of Vaccine Discovery, La Jolla Institute for Immunology, La Jolla, CA 92037
| | - Evan Shorr
- Department of Psychiatry, Columbia University Irving Medical Center, New York State Psychiatric Institute, New York, NY 10032
- Department of Pharmacology, Columbia University Irving Medical Center, New York State Psychiatric Institute, New York, NY 10032
| | - Chelsea Carpenter
- Division of Vaccine Discovery, La Jolla Institute for Immunology, La Jolla, CA 92037
| | - John Sidney
- Division of Vaccine Discovery, La Jolla Institute for Immunology, La Jolla, CA 92037
| | - Rekha Dhanwani
- Division of Vaccine Discovery, La Jolla Institute for Immunology, La Jolla, CA 92037
| | - Julian Agin-Liebes
- Department of Psychiatry, Columbia University Irving Medical Center, New York State Psychiatric Institute, New York, NY 10032
- Department of Pharmacology, Columbia University Irving Medical Center, New York State Psychiatric Institute, New York, NY 10032
| | - Francesca Garretti
- Department of Psychiatry, Columbia University Irving Medical Center, New York State Psychiatric Institute, New York, NY 10032
- Department of Pharmacology, Columbia University Irving Medical Center, New York State Psychiatric Institute, New York, NY 10032
| | - Amy W Amara
- Department of Neurology, University of Alabama at Birmingham, Birmingham, AL 35233
| | - David G Standaert
- Department of Neurology, University of Alabama at Birmingham, Birmingham, AL 35233
| | - Elizabeth J Phillips
- Vanderbilt University School of Medicine, Nashville, TN 37235
- Institute for Immunology and Infectious Diseases, Murdoch University, Perth, Western Australia 6150, Australia; and
| | - Simon A Mallal
- Vanderbilt University School of Medicine, Nashville, TN 37235
- Institute for Immunology and Infectious Diseases, Murdoch University, Perth, Western Australia 6150, Australia; and
| | - Bjoern Peters
- Division of Vaccine Discovery, La Jolla Institute for Immunology, La Jolla, CA 92037
- Department of Medicine, University of California, San Diego, La Jolla, CA 92093
| | - David Sulzer
- Department of Neurology, Columbia University Irving Medical Center, New York State Psychiatric Institute, New York, NY 10032
- Department of Psychiatry, Columbia University Irving Medical Center, New York State Psychiatric Institute, New York, NY 10032
- Department of Pharmacology, Columbia University Irving Medical Center, New York State Psychiatric Institute, New York, NY 10032
| | - Alessandro Sette
- Division of Vaccine Discovery, La Jolla Institute for Immunology, La Jolla, CA 92037;
- Department of Medicine, University of California, San Diego, La Jolla, CA 92093
| |
Collapse
|
25
|
Li X, Koudstaal W, Fletcher L, Costa M, van Winsen M, Siregar B, Inganäs H, Kim J, Keogh E, Macedo J, Holland T, Perry S, Bard F, Hoozemans JJ, Goudsmit J, Apetri A, Pascual G. Naturally occurring antibodies isolated from PD patients inhibit synuclein seeding in vitro and recognize Lewy pathology. Acta Neuropathol 2019; 137:825-836. [PMID: 30805666 PMCID: PMC6482120 DOI: 10.1007/s00401-019-01974-5] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2019] [Revised: 02/06/2019] [Accepted: 02/08/2019] [Indexed: 12/24/2022]
Abstract
Deposition of α-synuclein into Lewy bodies and Lewy neurites is the hallmark of Parkinson’s disease (PD). It is hypothesized that α-synuclein pathology spreads by a “prion-like” mechanism (i.e., by seeded aggregation or templated misfolding). Therefore, various extracellular α-synuclein conformers and/or posttranslational modifications may serve as biomarkers of disease or potential targets for novel interventions. To explore whether the antibody repertoires of PD patients contain anti-α-synuclein antibodies that can potentially be used as markers or immunotherapy, we interrogated peripheral IgG+ memory B cells from PD patients for reactivity to α-synuclein. In total, ten somatically mutated antibodies were recovered, suggesting the presence of an ongoing antigen-driven immune response. The three antibodies that had the highest affinity to recombinant full-length α-synuclein, aSyn-323.1, aSyn-336.1 and aSyn-338.1, were characterized further and shown to recognize epitopes in the C terminus of α-synuclein with binding affinities between 0.3 and 2.8 μM. Furthermore, all three antibodies were able to neutralize the “seeding” of intracellular synuclein aggregates in an in vitro α-synuclein seeding assay. Finally, differential reactivities were observed for all three human anti-α-synuclein antibodies across tissue treatment conditions by immunohistochemistry. Our results suggest that the memory B-cell repertoire of PD patients might represent a potential source of biomarkers and therapies.
Collapse
Affiliation(s)
- Xinyi Li
- Janssen Prevention Center, Janssen Pharmaceutical Companies of Johnson & Johnson, 3210 Merryfield Row, San Diego, CA 92121 USA
| | - Wouter Koudstaal
- Janssen Prevention Center, Janssen Pharmaceutical Companies of Johnson & Johnson, Archimedesweg 6, 2333 CN Leiden, The Netherlands
- Present Address: Lucidity Biomedical Consulting, Calle Emir 11, 18006 Granada, Spain
| | - Lauren Fletcher
- Janssen Prevention Center, Janssen Pharmaceutical Companies of Johnson & Johnson, 3210 Merryfield Row, San Diego, CA 92121 USA
| | - Martha Costa
- Janssen Prevention Center, Janssen Pharmaceutical Companies of Johnson & Johnson, 3210 Merryfield Row, San Diego, CA 92121 USA
| | - Margot van Winsen
- Janssen Prevention Center, Janssen Pharmaceutical Companies of Johnson & Johnson, Archimedesweg 6, 2333 CN Leiden, The Netherlands
| | - Berdien Siregar
- Janssen Prevention Center, Janssen Pharmaceutical Companies of Johnson & Johnson, Archimedesweg 6, 2333 CN Leiden, The Netherlands
| | - Hanna Inganäs
- Janssen Prevention Center, Janssen Pharmaceutical Companies of Johnson & Johnson, Archimedesweg 6, 2333 CN Leiden, The Netherlands
| | - Julie Kim
- Janssen Prevention Center, Janssen Pharmaceutical Companies of Johnson & Johnson, 3210 Merryfield Row, San Diego, CA 92121 USA
| | - Elissa Keogh
- Janssen Prevention Center, Janssen Pharmaceutical Companies of Johnson & Johnson, 3210 Merryfield Row, San Diego, CA 92121 USA
| | - Jeremy Macedo
- Janssen Prevention Center, Janssen Pharmaceutical Companies of Johnson & Johnson, 3210 Merryfield Row, San Diego, CA 92121 USA
| | - Trevin Holland
- Janssen Prevention Center, Janssen Pharmaceutical Companies of Johnson & Johnson, 3210 Merryfield Row, San Diego, CA 92121 USA
| | - Stuart Perry
- Janssen Prevention Center, Janssen Pharmaceutical Companies of Johnson & Johnson, 3210 Merryfield Row, San Diego, CA 92121 USA
| | - Frederique Bard
- Janssen Prevention Center, Janssen Pharmaceutical Companies of Johnson & Johnson, 3210 Merryfield Row, San Diego, CA 92121 USA
| | - Jeroen J. Hoozemans
- Department of Pathology, Amsterdam Neuroscience, VU University Medical Center, De Boelelaan 1117, 1081 HV Amsterdam, The Netherlands
| | - Jaap Goudsmit
- Janssen Prevention Center, Janssen Pharmaceutical Companies of Johnson & Johnson, Archimedesweg 6, 2333 CN Leiden, The Netherlands
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, 677 Huntington Avenue, Boston, MA 02115 USA
- Department of Neurology, Amsterdam Neuroscience, Academic Medical Center, Meidreefberg 9, 1105 AZ Amsterdam, The Netherlands
| | - Adrian Apetri
- Janssen Prevention Center, Janssen Pharmaceutical Companies of Johnson & Johnson, Archimedesweg 6, 2333 CN Leiden, The Netherlands
| | - Gabriel Pascual
- Janssen Prevention Center, Janssen Pharmaceutical Companies of Johnson & Johnson, 3210 Merryfield Row, San Diego, CA 92121 USA
| |
Collapse
|
26
|
Castro-Sánchez S, García-Yagüe ÁJ, Kügler S, Lastres-Becker I. CX3CR1-deficient microglia shows impaired signalling of the transcription factor NRF2: Implications in tauopathies. Redox Biol 2019; 22:101118. [PMID: 30769286 PMCID: PMC6375000 DOI: 10.1016/j.redox.2019.101118] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2018] [Revised: 01/16/2019] [Accepted: 01/20/2019] [Indexed: 12/11/2022] Open
Abstract
TAU protein aggregation is the main characteristic of neurodegenerative diseases known as tauopathies. Low-grade chronic inflammation is also another hallmark that indicates crosstalk between damaged neurons and glial cells. Previously, we have demonstrated that neurons overexpressing TAUP301L release CX3CL1, which activates the transcription factor NRF2 signalling to limit over-activation in microglial cells in vitro and in vivo. However, the connection between CX3CL1/CX3CR1 and NRF2 system and its functional implications in microglia are poorly described. We evaluated CX3CR1/NRF2 axis in the context of tauopathies and its implication in neuroinflammation. Regarding the molecular mechanisms that connect CX3CL1/CX3CR1 and NRF2 systems, we observed that in primary microglia from Cx3cr1-/- mice the mRNA levels of Nrf2 and its related genes were significantly decreased, establishing a direct linking between both systems. To determine functional relevance of CX3CR1, migration and phagocytosis assays were evaluated. CX3CR1-deficient microglia showed impaired cell migration and deficiency of phagocytosis, as previously described for NRF2-deficient microglia, reinforcing the idea of the relevance of the CX3CL1/CX3CR1 axis in these events. The importance of these findings was evident in a tauopathy mouse model where the effects of sulforaphane (SFN), an NRF2 inducer, were examined on neuroinflammation in Cx3cr1+/+ and Cx3cr1-/- mice. Interestingly, the treatment with SFN was able to modulate astrogliosis but failed to reduce microgliosis in Cx3cr1-/- mice. These findings suggest an essential role of the CX3CR1/NRF2 axis in microglial function and in tauopathies. Therefore, polymorphisms with loss of function in CX3CR1 or NRF2 have to be taken into account for the development of therapeutic strategies. CX3CR1-deficient primary microglial cells present impaired expression of the transcription factor NRF2 signature. TAM receptors expression is decreased in CX3CR1-deficient microglia. AXL receptor is a NRF2-dependent gene. Loss of CX3CR1 expression led to impaired phagocytosis and migration of microglia. Sulforaphane treatment did not reverse rAAV-TAUP301L induced microgliosis in CX3CR1-deficient mice.
Collapse
Affiliation(s)
- Sara Castro-Sánchez
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Instituto de Investigación Sanitaria La Paz (IdiPaz), Instituto de Investigaciones Biomédicas "Alberto Sols" UAM-CSIC, Madrid, Spain; Department of Biochemistry, School of Medicine, Universidad Autónoma de Madrid, Spain.
| | - Ángel J García-Yagüe
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Instituto de Investigación Sanitaria La Paz (IdiPaz), Instituto de Investigaciones Biomédicas "Alberto Sols" UAM-CSIC, Madrid, Spain; Department of Biochemistry, School of Medicine, Universidad Autónoma de Madrid, Spain.
| | - Sebastian Kügler
- Department of Neurology, Center Nanoscale Microscopy and Molecular Physiology of the Brain (CNMPB), University of Medicine Göttingen, Göttingen, Germany.
| | - Isabel Lastres-Becker
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Instituto de Investigación Sanitaria La Paz (IdiPaz), Instituto de Investigaciones Biomédicas "Alberto Sols" UAM-CSIC, Madrid, Spain; Department of Biochemistry, School of Medicine, Universidad Autónoma de Madrid, Spain.
| |
Collapse
|
27
|
Zhang H, Zhu X, Pascual G, Wadia JS, Keogh E, Hoozemans JJ, Siregar B, Inganäs H, Stoop EJM, Goudsmit J, Apetri A, Koudstaal W, Wilson IA. Structural Basis for Recognition of a Unique Epitope by a Human Anti-tau Antibody. Structure 2018; 26:1626-1634.e4. [PMID: 30318466 DOI: 10.1016/j.str.2018.08.012] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2018] [Revised: 07/23/2018] [Accepted: 08/27/2018] [Indexed: 01/08/2023]
Abstract
Aggregation of the hyperphosphorylated protein tau into neurofibrillary tangles and neuropil threads is a hallmark of Alzheimer disease (AD). Identification and characterization of the epitopes recognized by anti-tau antibodies might shed light on the molecular mechanisms of AD pathogenesis. Here we report on the biochemical and structural characterization of a tau-specific monoclonal antibody CBTAU-24.1, which was isolated from the human memory B cell repertoire. Immunohistochemical staining with CBTAU-24.1 specifically detects pathological tau structures in AD brain samples. The crystal structure of CBTAU-24.1 Fab with a phosphorylated tau peptide revealed recognition of a unique epitope (Ser235-Leu243) in the tau proline-rich domain. Interestingly, the antibody can bind tau regardless of phosphorylation state of its epitope region and also recognizes both monomeric and paired helical filament tau irrespective of phosphorylation status. This human anti-tau antibody and its unique epitope may aid in development of diagnostics and/or therapeutic AD strategies.
Collapse
Affiliation(s)
- Heng Zhang
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA 92037, USA; Beijing Synchrotron Radiation Facility, Institute of High Energy Physics, Chinese Academy of Sciences, Beijing 100049, People's Republic of China
| | - Xueyong Zhu
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Gabriel Pascual
- Janssen Prevention Center, Janssen Pharmaceutical Companies of Johnson & Johnson, 3210 Merryfield Row, San Diego, CA 92121, USA
| | - Jehangir S Wadia
- Janssen Prevention Center, Janssen Pharmaceutical Companies of Johnson & Johnson, 3210 Merryfield Row, San Diego, CA 92121, USA
| | - Elissa Keogh
- Janssen Prevention Center, Janssen Pharmaceutical Companies of Johnson & Johnson, 3210 Merryfield Row, San Diego, CA 92121, USA
| | - Jeroen J Hoozemans
- Department of Pathology, Amsterdam Neuroscience, VU University Medical Center, de Boelelaan 1117, 1081 HV Amsterdam, the Netherlands
| | - Berdien Siregar
- Janssen Prevention Center, Janssen Pharmaceutical Companies of Johnson & Johnson, Archimedesweg 6, 2333 CN Leiden, the Netherlands
| | - Hanna Inganäs
- Janssen Prevention Center, Janssen Pharmaceutical Companies of Johnson & Johnson, Archimedesweg 6, 2333 CN Leiden, the Netherlands
| | - Esther J M Stoop
- Janssen Prevention Center, Janssen Pharmaceutical Companies of Johnson & Johnson, Archimedesweg 6, 2333 CN Leiden, the Netherlands
| | - Jaap Goudsmit
- Janssen Prevention Center, Janssen Pharmaceutical Companies of Johnson & Johnson, Archimedesweg 6, 2333 CN Leiden, the Netherlands; Department of Neurology, Amsterdam Neuroscience, Academic Medical Center, Meidreefberg 9, 1105 AZ Amsterdam, the Netherlands; Department of Epidemiology, Harvard T.H. Chan School of Public Health, 677 Huntington Avenue, Boston, MA 02115, USA
| | - Adrian Apetri
- Janssen Prevention Center, Janssen Pharmaceutical Companies of Johnson & Johnson, Archimedesweg 6, 2333 CN Leiden, the Netherlands
| | - Wouter Koudstaal
- Janssen Prevention Center, Janssen Pharmaceutical Companies of Johnson & Johnson, Archimedesweg 6, 2333 CN Leiden, the Netherlands
| | - Ian A Wilson
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA 92037, USA; Skaggs Institute for Chemical Biology, The Scripps Research Institute, La Jolla, CA 92037, USA.
| |
Collapse
|
28
|
Wang J, Jin WS, Bu XL, Zeng F, Huang ZL, Li WW, Shen LL, Zhuang ZQ, Fang Y, Sun BL, Zhu J, Yao XQ, Zeng GH, Dong ZF, Yu JT, Hu Z, Song W, Zhou HD, Jiang JX, Liu YH, Wang YJ. Physiological clearance of tau in the periphery and its therapeutic potential for tauopathies. Acta Neuropathol 2018; 136:525-536. [PMID: 30074071 DOI: 10.1007/s00401-018-1891-2] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2018] [Revised: 07/18/2018] [Accepted: 07/18/2018] [Indexed: 02/07/2023]
Abstract
Accumulation of pathological tau is the hallmark of Alzheimer's disease and other tauopathies and is closely correlated with cognitive decline. Clearance of pathological tau from the brain is a major therapeutic strategy for tauopathies. The physiological capacity of the periphery to clear brain-derived tau and its therapeutic potential remain largely unknown. Here, we found that cisterna magna injected 131I-labelled synthetic tau dynamically effluxed from the brain and was mainly cleared from the kidney, blood, and liver in mice; we also found that plasma tau levels in inferior vena cava were lower than those in femoral artery in humans. These findings suggest that tau proteins can efflux out of the brain and be cleared in the periphery under physiological conditions. Next, we showed that lowering blood tau levels via peritoneal dialysis could reduce interstitial fluid (ISF) tau levels in the brain, and tau levels in the blood and ISF were dynamically correlated; furthermore, tau efflux from the brain was accelerated after the addition of another set of peripheral system in a parabiosis model. Finally, we established parabiosis mouse models using tau transgenic mice and their wild-type littermates and found that brain tau levels and related pathologies in parabiotic transgenic mice were significantly reduced after parabiosis, suggesting that chronic enhancement of peripheral tau clearance alleviates pathological tau accumulation and neurodegeneration in the brain. Our study provides the first evidence of physiological clearance of brain-derived pathological tau in the periphery, suggesting that enhancing peripheral tau clearance is a potential therapeutic strategy for tauopathies.
Collapse
Affiliation(s)
- Jun Wang
- Department of Neurology, Daping Hospital, Third Military Medical University, Chongqing, China
| | - Wang-Sheng Jin
- Department of Neurology, Daping Hospital, Third Military Medical University, Chongqing, China
| | - Xian-Le Bu
- Department of Neurology, Daping Hospital, Third Military Medical University, Chongqing, China
| | - Fan Zeng
- Department of Neurology, Daping Hospital, Third Military Medical University, Chongqing, China
| | - Zhi-Lin Huang
- Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Translational Medical Research in Cognitive Development and Learning and Memory Disorders, Children's Hospital of Chongqing Medical University, Chongqing, China
| | - Wei-Wei Li
- Department of Neurology, Daping Hospital, Third Military Medical University, Chongqing, China
| | - Lin-Lin Shen
- Department of Neurology, Daping Hospital, Third Military Medical University, Chongqing, China
| | - Zhen-Qian Zhuang
- Department of Neurology, Daping Hospital, Third Military Medical University, Chongqing, China
| | - Yuqiang Fang
- Department of Cardiology, Daping Hospital, Third Military Medical University, Chongqing, China
| | - Bin-Lu Sun
- Department of Neurology, Daping Hospital, Third Military Medical University, Chongqing, China
| | - Jie Zhu
- Department of Neurology, Daping Hospital, Third Military Medical University, Chongqing, China
| | - Xiu-Qing Yao
- Department of Neurology, Daping Hospital, Third Military Medical University, Chongqing, China
| | - Gui-Hua Zeng
- Department of Neurology, Daping Hospital, Third Military Medical University, Chongqing, China
| | - Zhi-Fang Dong
- Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Translational Medical Research in Cognitive Development and Learning and Memory Disorders, Children's Hospital of Chongqing Medical University, Chongqing, China
| | - Jin-Tai Yu
- Department of Neurology, Qingdao Municipal Hospital, School of Medicine, Qingdao University, Qingdao, China
| | - Zhian Hu
- Department of Physiology, Collaborative Innovation Center for Brain Science, Third Military Medical University, Chongqing, China
| | - Weihong Song
- Townsend Family Laboratories, Department of Psychiatry, Brain Research Center, The University of British Columbia, Vancouver, BC, V6T 1Z3, Canada
| | - Hua-Dong Zhou
- Department of Neurology, Daping Hospital, Third Military Medical University, Chongqing, China
| | - Jian-Xin Jiang
- State Key Laboratory of Trauma, Burn and Combined Injury, Institute of Surgery Research, Daping Hospital, Third Military Medical University, Chongqing, China
| | - Yu-Hui Liu
- Department of Neurology, Daping Hospital, Third Military Medical University, Chongqing, China.
| | - Yan-Jiang Wang
- Department of Neurology, Daping Hospital, Third Military Medical University, Chongqing, China.
- State Key Laboratory of Trauma, Burn and Combined Injury, Institute of Surgery Research, Daping Hospital, Third Military Medical University, Chongqing, China.
- Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China.
| |
Collapse
|
29
|
A Closer Look into the Role of Protein Tau in the Identification of Promising Therapeutic Targets for Alzheimer's Disease. Brain Sci 2018; 8:brainsci8090162. [PMID: 30149687 PMCID: PMC6162660 DOI: 10.3390/brainsci8090162] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2018] [Revised: 08/24/2018] [Accepted: 08/24/2018] [Indexed: 01/09/2023] Open
Abstract
One of the most commonly known chronic neurodegenerative disorders, Alzheimer's disease (AD), manifests the common type of dementia in 60⁻80% of cases. From a clinical standpoint, a patent cognitive decline and a severe change in personality, as caused by a loss of neurons, is usually evident in AD with about 50 million people affected in 2016. The disease progression in patients is distinguished by a gradual plummet in cognitive functions, eliciting symptoms such as memory loss, and eventually requiring full-time medical care. From a histopathological standpoint, the defining characteristics are intracellular aggregations of hyper-phosphorylated tau protein, known as neurofibrillary tangles (NFT), and depositions of amyloid β-peptides (Aβ) in the brain. The abnormal phosphorylation of tau protein is attributed to a wide gamut of neurological disorders known as tauopathies. In addition to the hyperphosphorylated tau lesions, neuroinflammatory processes could occur in a sustained manner through astro-glial activation, resulting in the disease progression. Recent findings have suggested a strong interplay between the mechanism of Tau phosphorylation, disruption of microtubules, and synaptic loss and pathology of AD. The mechanisms underlying these interactions along with their respective consequences in Tau pathology are still ill-defined. Thus, in this review: (1) we highlight the interplays existing between Tau pathology and AD; and (2) take a closer look into its role while identifying some promising therapeutic advances including state of the art imaging techniques.
Collapse
|
30
|
Ganz AB, Beker N, Hulsman M, Sikkes S, Netherlands Brain Bank, Scheltens P, Smit AB, Rozemuller AJM, Hoozemans JJM, Holstege H. Neuropathology and cognitive performance in self-reported cognitively healthy centenarians. Acta Neuropathol Commun 2018; 6:64. [PMID: 30037350 PMCID: PMC6055341 DOI: 10.1186/s40478-018-0558-5] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2018] [Accepted: 06/21/2018] [Indexed: 11/23/2022] Open
Abstract
With aging, the incidence of neuropathological hallmarks of neurodegenerative diseases increases in the brains of cognitively healthy individuals. It is currently unclear to what extent these hallmarks associate with symptoms of disease at extreme ages. Forty centenarians from the 100-plus Study cohort donated their brain. Centenarians self-reported to be cognitively healthy at baseline, which was confirmed by a proxy. Objective ante-mortem measurements of cognitive performance were associated with the prevalence, distribution and quantity of age- and AD-related neuropathological hallmarks. Despite self-reported cognitive health, objective neuropsychological testing suggested varying levels of ante-mortem cognitive functioning. Post-mortem, we found that neuropathological hallmarks related to age and neurodegenerative diseases, such as Aβ and Tau pathology, as well as atherosclerosis, were abundantly present in most or all centenarians, whereas Lewy body and pTDP-43 pathology were scarce. We observed that increased pathology loads correlated across pathology subtypes, and an overall trend of higher pathology loads to associate with a lower cognitive test performance. This trend was carried especially by the presence of neurofibrillary tangles (NFTs) and granulovacuolar degeneration (GVD) and to a lesser extent by Aβ-associated pathologies. Cerebral Amyloid Angiopathy (CAA) specifically associated with lower executive functioning in the centenarians. In conclusion, we find that while the centenarians in this cohort escaped or delayed cognitive impairment until extreme ages, their brains reveal varying levels of disease-associated neuropathological hallmarks, some of which associate with cognitive performance.
Collapse
Affiliation(s)
- Andrea B Ganz
- Department of Molecular and Cellular Neuroscience, Center for Neurogenomics and Cognitive Research, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, De Boelelaan 1085, 1081 HV, Amsterdam, The Netherlands
- Department of Pathology, Amsterdam Neuroscience, VU University Medical Center, PO Box 7057, 1007 MB, Amsterdam, The Netherlands
- Alzheimer Centre, Department of Neurology, Amsterdam Neuroscience, VU University Medical Center, Postbus 7057, 1007 MB, Amsterdam, The Netherlands
| | - Nina Beker
- Alzheimer Centre, Department of Neurology, Amsterdam Neuroscience, VU University Medical Center, Postbus 7057, 1007 MB, Amsterdam, The Netherlands
| | - Marc Hulsman
- Department of Clinical Genetics, Amsterdam Neuroscience, VU University Medical Center, de Boelelaan, 1118 1081 HV, Amsterdam, The Netherlands
| | - Sietske Sikkes
- Alzheimer Centre, Department of Neurology, Amsterdam Neuroscience, VU University Medical Center, Postbus 7057, 1007 MB, Amsterdam, The Netherlands
| | - Netherlands Brain Bank
- Netherlands Institute for Neuroscience, Meibergdreef 47, 1105 BA, Amsterdam, The Netherlands
| | - Philip Scheltens
- Alzheimer Centre, Department of Neurology, Amsterdam Neuroscience, VU University Medical Center, Postbus 7057, 1007 MB, Amsterdam, The Netherlands
| | - August B Smit
- Department of Molecular and Cellular Neuroscience, Center for Neurogenomics and Cognitive Research, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, De Boelelaan 1085, 1081 HV, Amsterdam, The Netherlands
| | - Annemieke J M Rozemuller
- Department of Pathology, Amsterdam Neuroscience, VU University Medical Center, PO Box 7057, 1007 MB, Amsterdam, The Netherlands
| | - Jeroen J M Hoozemans
- Department of Pathology, Amsterdam Neuroscience, VU University Medical Center, PO Box 7057, 1007 MB, Amsterdam, The Netherlands
| | - Henne Holstege
- Alzheimer Centre, Department of Neurology, Amsterdam Neuroscience, VU University Medical Center, Postbus 7057, 1007 MB, Amsterdam, The Netherlands.
- Department of Clinical Genetics, Amsterdam Neuroscience, VU University Medical Center, de Boelelaan, 1118 1081 HV, Amsterdam, The Netherlands.
| |
Collapse
|
31
|
van Ameijde J, Crespo R, Janson R, Juraszek J, Siregar B, Verveen H, Sprengers I, Nahar T, Hoozemans JJ, Steinbacher S, Willems R, Delbroek L, Borgers M, Dockx K, Van Kolen K, Mercken M, Pascual G, Koudstaal W, Apetri A. Enhancement of therapeutic potential of a naturally occurring human antibody targeting a phosphorylated Ser 422 containing epitope on pathological tau. Acta Neuropathol Commun 2018; 6:59. [PMID: 30001207 PMCID: PMC6042391 DOI: 10.1186/s40478-018-0562-9] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2018] [Accepted: 06/28/2018] [Indexed: 11/24/2022] Open
Abstract
Aggregation of tau protein and spreading of tau aggregates are pivotal pathological processes in a range of neurological disorders. Accumulating evidence suggests that immunotherapy targeting tau may be a viable therapeutic strategy. We have previously described the isolation of antibody CBTAU-22.1 from the memory B-cell repertoire of healthy human donors. CBTAU-22.1 was shown to specifically bind a disease-associated phosphorylated epitope in the C-terminus of tau (Ser422) and to be able to inhibit the spreading of pathological tau aggregates from P301S spinal cord lysates in vitro, albeit with limited potency. Using a combination of rational design and random mutagenesis we have derived a variant antibody with improved affinity while maintaining the specificity of the parental antibody. This affinity improved antibody showed greatly enhanced potency in a cell-based immunodepletion assay using paired helical filaments (PHFs) derived from human Alzheimer’s disease (AD) brain tissue. Moreover, the affinity improved antibody limits the in vitro aggregation propensity of full length tau species specifically phosphorylated at position 422 produced by employing a native chemical ligation approach. Together, these results indicate that in addition to being able to inhibit the spreading of pathological tau aggregates, the matured antibody can potentially also interfere with the nucleation of tau which is believed to be the first step of the pathogenic process. Finally, the functionality in a P301L transgenic mice co-injection model highlights the therapeutic potential of human antibody dmCBTAU-22.1.
Collapse
|
32
|
Yang C, Li X, Gao W, Wang Q, Zhang L, Li Y, Li L, Zhang L. Cornel Iridoid Glycoside Inhibits Tau Hyperphosphorylation via Regulating Cross-Talk Between GSK-3β and PP2A Signaling. Front Pharmacol 2018; 9:682. [PMID: 29997510 PMCID: PMC6028923 DOI: 10.3389/fphar.2018.00682] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2018] [Accepted: 06/06/2018] [Indexed: 12/15/2022] Open
Abstract
Neurofibrillary pathology contributes to neuronal dysfunction and correlates with the clinical progression of Alzheimer's disease (AD). Tau phosphorylation is mainly regulated by a balance of glycogen synthase kinase-3β (GSK-3β) and protein phosphatase 2A (PP2A) activities. Cornel iridoid glycoside (CIG) is a main component extracted from Cornus officinalis. The purpose of this study was to investigate the effects of CIG on GSK-3β and PP2A, thus to explore the mechanisms of CIG to inhibit tau hyperphosphorylation. The rat model of tau hyperphosphorylation was established by intraventricular injection of wortmannin and GF-109203X (GFX) to activate GSK-3β. The results showed that intragastrical administration of CIG inhibited tau hyperphosphorylation in the brain of rats induced by wortmannin/GFX. The results in vivo and in vitro exhibited that CIG inhibited tau hyperphosphorylation and GSK-3β over-activation. In the mechanism of action, CIG's attenuating GSK-3β activity was found to be dependent on PI3K/AKT signaling pathway. PP2A catalytic C subunit (PP2Ac) siRNA abrogated the effect of CIG on PI3K/AKT/GSK-3β. Additionally and crucially, we also found that CIG inhibited the demethylation of PP2Ac at Leu309 in vivo and in vitro. It enhanced PP2A activity, decreased tau hyperphosphorylation, and protected cell morphology in okadaic acid (OA)-induced cell model in vitro. PP2Ac siRNA abated the inhibitory effect of CIG on tau hyperphosphorylation. Moreover, CIG inhibited protein phosphatase methylesterase-1 (PME-1) and demethylation of PP2Ac, enhanced PP2A activity, and decreased tau hyperphosphorylation in PME-1-transfectd cells. Taken together, CIG inhibited GSK-3β activity via promoting P13K/AKT and PP2A signaling pathways. In addition, CIG also elevated PP2A activity via inhibiting PME-1-induced PP2Ac demethylation to inhibit GSK-3β activity, thus regulated the cross-talk between GSK-3β and PP2A signaling and consequently inhibited tau hyperphosphorylation. These results suggest that CIG may be a promising agent for AD therapy.
Collapse
Affiliation(s)
- Cuicui Yang
- Department of Pharmacy, Xuanwu Hospital of Capital Medical University, Beijing, China.,Beijing Institute for Brain Disorders, Beijing, China.,Beijing Engineering Research Center for Nerve System Drugs, Beijing, China.,Key Laboratory for Neurodegenerative Diseases of Ministry of Education, Beijing, China
| | - Xuelian Li
- Institute of Clinical Pharmacology, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Wenbin Gao
- Institute of Clinical Pharmacology, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Qi Wang
- Institute of Clinical Pharmacology, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Li Zhang
- Department of Pharmacy, Xuanwu Hospital of Capital Medical University, Beijing, China.,Beijing Institute for Brain Disorders, Beijing, China.,Beijing Engineering Research Center for Nerve System Drugs, Beijing, China.,Key Laboratory for Neurodegenerative Diseases of Ministry of Education, Beijing, China
| | - Yali Li
- Department of Pharmacy, Xuanwu Hospital of Capital Medical University, Beijing, China.,Beijing Institute for Brain Disorders, Beijing, China.,Beijing Engineering Research Center for Nerve System Drugs, Beijing, China.,Key Laboratory for Neurodegenerative Diseases of Ministry of Education, Beijing, China
| | - Lin Li
- Department of Pharmacy, Xuanwu Hospital of Capital Medical University, Beijing, China.,Beijing Institute for Brain Disorders, Beijing, China.,Beijing Engineering Research Center for Nerve System Drugs, Beijing, China.,Key Laboratory for Neurodegenerative Diseases of Ministry of Education, Beijing, China
| | - Lan Zhang
- Department of Pharmacy, Xuanwu Hospital of Capital Medical University, Beijing, China.,Beijing Institute for Brain Disorders, Beijing, China.,Beijing Engineering Research Center for Nerve System Drugs, Beijing, China.,Key Laboratory for Neurodegenerative Diseases of Ministry of Education, Beijing, China
| |
Collapse
|
33
|
Apetri A, Crespo R, Juraszek J, Pascual G, Janson R, Zhu X, Zhang H, Keogh E, Holland T, Wadia J, Verveen H, Siregar B, Mrosek M, Taggenbrock R, Ameijde J, Inganäs H, van Winsen M, Koldijk MH, Zuijdgeest D, Borgers M, Dockx K, Stoop EJM, Yu W, Brinkman-van der Linden EC, Ummenthum K, van Kolen K, Mercken M, Steinbacher S, de Marco D, Hoozemans JJ, Wilson IA, Koudstaal W, Goudsmit J. A common antigenic motif recognized by naturally occurring human V H5-51/V L4-1 anti-tau antibodies with distinct functionalities. Acta Neuropathol Commun 2018; 6:43. [PMID: 29855358 PMCID: PMC5984341 DOI: 10.1186/s40478-018-0543-z] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2018] [Accepted: 05/07/2018] [Indexed: 01/16/2023] Open
Abstract
Misfolding and aggregation of tau protein are closely associated with the onset and progression of Alzheimer’s Disease (AD). By interrogating IgG+ memory B cells from asymptomatic donors with tau peptides, we have identified two somatically mutated VH5–51/VL4–1 antibodies. One of these, CBTAU-27.1, binds to the aggregation motif in the R3 repeat domain and blocks the aggregation of tau into paired helical filaments (PHFs) by sequestering monomeric tau. The other, CBTAU-28.1, binds to the N-terminal insert region and inhibits the spreading of tau seeds and mediates the uptake of tau aggregates into microglia by binding PHFs. Crystal structures revealed that the combination of VH5–51 and VL4–1 recognizes a common Pro-Xn-Lys motif driven by germline-encoded hotspot interactions while the specificity and thereby functionality of the antibodies are defined by the CDR3 regions. Affinity improvement led to improvement in functionality, identifying their epitopes as new targets for therapy and prevention of AD.
Collapse
|
34
|
Tau and neuroinflammation: What impact for Alzheimer's Disease and Tauopathies? Biomed J 2018; 41:21-33. [PMID: 29673549 PMCID: PMC6138617 DOI: 10.1016/j.bj.2018.01.003] [Citation(s) in RCA: 211] [Impact Index Per Article: 35.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2017] [Revised: 01/09/2018] [Accepted: 01/11/2018] [Indexed: 01/03/2023] Open
Abstract
Alzheimer's Disease (AD) is a chronic neurodegenerative disorder and the most common type of dementia (60–80% of cases). In 2016, nearly 44 million people were affected by AD or related dementia. AD is characterized by progressive neuronal damages leading to subtle and latter obvious decline in cognitive functions including symptoms such as memory loss or confusion, which ultimately require full-time medical care. Its neuropathology is defined by the extracellular accumulation of amyloid-β (Aβ) peptide into amyloid plaques, and intraneuronal neurofibrillary tangles (NFT) consisting of aggregated hyper- and abnormal phosphorylation of tau protein. The latter, identified also as Tau pathology, is observed in a broad spectrum of neurological diseases commonly referred to as “Tauopathies”. Besides these lesions, sustained neuroinflammatory processes occur, involving notably micro- and astro-glial activation, which contribute to disease progression. Recent findings from genome wide association studies further support an instrumental role of neuroinflammation. While the interconnections existing between this innate immune response and the amyloid pathogenesis are widely characterized and described as complex, elaborated and evolving, only few studies focused on Tau pathology. An adaptive immune response takes place conjointly during the disease course, as indicated by the presence of vascular and parenchymal T-cell in AD patients' brain. The underlying mechanisms of this infiltration and its consequences with regards to Tau pathology remain understudied so far. In the present review, we highlight the interplays existing between Tau pathology and the innate/adaptive immune responses.
Collapse
|
35
|
Klaver AC, Coffey MP, Bennett DA, Loeffler DA. Specific serum antibody binding to phosphorylated and non-phosphorylated tau in non-cognitively impaired, mildly cognitively impaired, and Alzheimer's disease subjects: an exploratory study. Transl Neurodegener 2017; 6:32. [PMID: 29204273 PMCID: PMC5701435 DOI: 10.1186/s40035-017-0100-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2017] [Accepted: 10/30/2017] [Indexed: 11/28/2022] Open
Abstract
Background Tau vaccination and administration of anti-tau antibodies can prevent pathology and cognitive impairment in transgenic mouse models of tauopathy, suggesting that therapies which increase anti-tau antibodies might slow the development and/or progression of Alzheimer’s disease (AD). The extent to which individuals with no cognitive impairment (NCI) possess serum anti-tau antibodies, and whether their concentrations of these antibodies differ from anti-tau antibody levels in persons with mild cognitive impairment (MCI) or AD, are unclear. Previous studies measuring these antibodies did not account for antibody polyvalent binding, which can be extensive, nor that antibody binding to phosphorylated tau peptides could be due to binding to non-phosphorylated epitopes on those peptides. Methods An ELISA controlling for these factors was used to measure the specific binding of serum IgG and IgM to phosphorylated (“pTau;” phosphorylated at Serine-199 and Serine-202) and non-phosphorylated (“non-pTau”) tau 196-207 in subjects with NCI, MCI, or AD (n = 10/group). Between-group differences in these antibody levels were evaluated for statistical significance, and correlations were examined in pooled data from all subjects between these antibody levels and subject age, global cognitive functioning, and NFT counts. Results Specific IgG binding to pTau and non-pTau was detected in all subjects except for one NCI control. Specific IgM binding was detected to pTau in all subjects except for two AD patients, and to non-pTau in all subjects. Mean pTau IgG was increased in MCI subjects by 53% and 70% vs. AD and NCI subjects respectively (both p < 0.05), while no significant differences were found between groups for non-pTau IgG (p = 0.052), pTau IgM, or non-pTau IgM. Non-pTau IgG was negatively associated with global cognition (Spearman rho = −0.50). Conclusions Specific binding of serum IgG and IgM to phosphorylated and non-phosphorylated tau may be present in older persons regardless of their cognitive status. Serum IgG to phosphorylated tau may be increased in individuals with MCI, but this unexpected finding requires confirmation. The approach used in this study to measure specific serum antibodies to phosphorylated tau should be useful for measuring antibodies to other post-translationally-modified proteins that are of relevance to neurodegenerative disorders.
Collapse
Affiliation(s)
- Andrea C Klaver
- Department of Neurology, Beaumont Research Institute, Beaumont Health, Royal Oak, MI USA
| | - Mary P Coffey
- Department of Biostatistics, Beaumont Research Institute, Beaumont Health, Royal Oak, MI USA
| | | | - David A Loeffler
- Department of Neurology, Beaumont Research Institute, Beaumont Health, Royal Oak, MI USA
| |
Collapse
|
36
|
Rojas-Gutierrez E, Muñoz-Arenas G, Treviño S, Espinosa B, Chavez R, Rojas K, Flores G, Díaz A, Guevara J. Alzheimer's disease and metabolic syndrome: A link from oxidative stress and inflammation to neurodegeneration. Synapse 2017. [PMID: 28650104 DOI: 10.1002/syn.21990] [Citation(s) in RCA: 119] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Alzheimer's disease (AD) is the most common cause of dementia and one of the most important causes of morbidity and mortality among the aging population. AD diagnosis is made post-mortem, and the two pathologic hallmarks, particularly evident in the end stages of the illness, are amyloid plaques and neurofibrillary tangles. Currently, there is no curative treatment for AD. Additionally, there is a strong relation between oxidative stress, metabolic syndrome, and AD. The high levels of circulating lipids and glucose imbalances amplify lipid peroxidation that gradually diminishes the antioxidant systems, causing high levels of oxidative metabolism that affects cell structure, leading to neuronal damage. Accumulating evidence suggests that AD is closely related to a dysfunction of both insulin signaling and glucose metabolism in the brain, leading to an insulin-resistant brain state. Four drugs are currently used for this pathology: Three FDA-approved cholinesterase inhibitors and one NMDA receptor antagonist. However, wide varieties of antioxidants are promissory to delay or prevent the symptoms of AD and may help in treating the disease. Therefore, therapeutic efforts to achieve attenuation of oxidative stress could be beneficial in AD treatment, attenuating Aβ-induced neurotoxicity and improve neurological outcomes in AD. The term inflammaging characterizes a widely accepted paradigm that aging is accompanied by a low-grade chronic up-regulation of certain pro-inflammatory responses in the absence of overt infection, and is a highly significant risk factor for both morbidity and mortality in the elderly.
Collapse
Affiliation(s)
- Eduardo Rojas-Gutierrez
- Departamento de Bioquímica, Facultad de Medicina, Universidad Nacional Autónoma de México, Ciudad de México, Mexico
| | - Guadalupe Muñoz-Arenas
- Facultad de Ciencias Químicas, Benemérita Universidad Autónoma de Puebla, Puebla, Pue, Mexico
| | - Samuel Treviño
- Facultad de Ciencias Químicas, Benemérita Universidad Autónoma de Puebla, Puebla, Pue, Mexico
| | - Blanca Espinosa
- Departamento de Bioquímica, Instituto Nacional de Enfermedades Respiratorias-INER, Ciudad de México, Mexico
| | - Raúl Chavez
- Departamento de Bioquímica, Facultad de Medicina, Universidad Nacional Autónoma de México, Ciudad de México, Mexico
| | - Karla Rojas
- Departamento de Bioquímica, Facultad de Medicina, Universidad Nacional Autónoma de México, Ciudad de México, Mexico
| | - Gonzalo Flores
- Instituto de Fisiología, Benemérita Universidad Autónoma de Puebla, Puebla, Pue, Mexico
| | - Alfonso Díaz
- Facultad de Ciencias Químicas, Benemérita Universidad Autónoma de Puebla, Puebla, Pue, Mexico
| | - Jorge Guevara
- Departamento de Bioquímica, Facultad de Medicina, Universidad Nacional Autónoma de México, Ciudad de México, Mexico
| |
Collapse
|
37
|
Yin W, M. Stover C. The potential of circulating autoantibodies in the early diagnosis of Alzheimer’s disease. AIMS ALLERGY AND IMMUNOLOGY 2017. [DOI: 10.3934/allergy.2017.2.62] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
|