1
|
Puangmalai N, Bhatt N, Bittar A, Jerez C, Shchankin N, Kayed R. Traumatic brain injury derived pathological tau polymorphs induce the distinct propagation pattern and neuroinflammatory response in wild type mice. Prog Neurobiol 2024; 232:102562. [PMID: 38135105 DOI: 10.1016/j.pneurobio.2023.102562] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2023] [Revised: 12/01/2023] [Accepted: 12/16/2023] [Indexed: 12/24/2023]
Abstract
The misfolding and aggregation of the tau protein into neurofibrillary tangles constitutes a central feature of tauopathies. Traumatic brain injury (TBI) has emerged as a potential risk factor, triggering the onset and progression of tauopathies. Our previous research revealed distinct polymorphisms in soluble tau oligomers originating from single versus repetitive mild TBIs. However, the mechanisms orchestrating the dissemination of TBI brain-derived tau polymorphs (TBI-BDTPs) remain elusive. In this study, we explored whether TBI-BDTPs could initiate pathological tau formation, leading to distinct pathogenic trajectories. Wild-type mice were exposed to TBI-BDTPs from sham, single-blast (SB), or repeated-blast (RB) conditions, and their memory function was assessed through behavioral assays at 2- and 8-month post-injection. Our findings revealed that RB-BDTPs induced cognitive and motor deficits, concurrently fostering the emergence of toxic tau aggregates within the injected hippocampus. Strikingly, this tau pathology propagated to cortical layers, intensifying over time. Importantly, RB-BDTP-exposed animals displayed heightened glial cell activation, NLRP3 inflammasome formation, and increased TBI biomarkers, particularly triggering the aggregation of S100B, which is indicative of a neuroinflammatory response. Collectively, our results shed light on the intricate mechanisms underlying TBI-BDTP-induced tau pathology and its association with neuroinflammatory processes. This investigation enhances our understanding of tauopathies and their interplay with neurodegenerative and inflammatory pathways following traumatic brain injury.
Collapse
Affiliation(s)
- Nicha Puangmalai
- Mitchell Center for Neurodegenerative Diseases, University of Texas Medical Branch, Galveston, TX 77555, USA; Departments of Neurology, Neuroscience and Cell Biology, University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Nemil Bhatt
- Mitchell Center for Neurodegenerative Diseases, University of Texas Medical Branch, Galveston, TX 77555, USA; Departments of Neurology, Neuroscience and Cell Biology, University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Alice Bittar
- Mitchell Center for Neurodegenerative Diseases, University of Texas Medical Branch, Galveston, TX 77555, USA; Departments of Neurology, Neuroscience and Cell Biology, University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Cynthia Jerez
- Mitchell Center for Neurodegenerative Diseases, University of Texas Medical Branch, Galveston, TX 77555, USA; Departments of Neurology, Neuroscience and Cell Biology, University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Nikita Shchankin
- Mitchell Center for Neurodegenerative Diseases, University of Texas Medical Branch, Galveston, TX 77555, USA; Departments of Neurology, Neuroscience and Cell Biology, University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Rakez Kayed
- Mitchell Center for Neurodegenerative Diseases, University of Texas Medical Branch, Galveston, TX 77555, USA; Departments of Neurology, Neuroscience and Cell Biology, University of Texas Medical Branch, Galveston, TX 77555, USA.
| |
Collapse
|
2
|
Canepa E, Parodi-Rullan R, Vazquez-Torres R, Gamallo-Lana B, Guzman-Hernandez R, Lemon NL, Angiulli F, Debure L, Ilies MA, Østergaard L, Wisniewski T, Gutiérrez-Jiménez E, Mar AC, Fossati S. FDA-approved carbonic anhydrase inhibitors reduce amyloid β pathology and improve cognition, by ameliorating cerebrovascular health and glial fitness. Alzheimers Dement 2023; 19:5048-5073. [PMID: 37186121 PMCID: PMC10600328 DOI: 10.1002/alz.13063] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Revised: 03/09/2023] [Accepted: 03/09/2023] [Indexed: 05/17/2023]
Abstract
INTRODUCTION Cerebrovascular pathology is an early and causal hallmark of Alzheimer's disease (AD), in need of effective therapies. METHODS Based on the success of our previous in vitro studies, we tested for the first time in a model of AD and cerebral amyloid angiopathy (CAA), the carbonic anhydrase inhibitors (CAIs) methazolamide and acetazolamide, Food and Drug Administration-approved against glaucoma and high-altitude sickness. RESULTS Both CAIs reduced cerebral, vascular, and glial amyloid beta (Aβ) accumulation and caspase activation, diminished gliosis, and ameliorated cognition in TgSwDI mice. The CAIs also improved microvascular fitness and induced protective glial pro-clearance pathways, resulting in the reduction of Aβ deposition. Notably, we unveiled that the mitochondrial carbonic anhydrase-VB (CA-VB) is upregulated in TgSwDI brains, CAA and AD+CAA human subjects, and in endothelial cells upon Aβ treatment. Strikingly, CA-VB silencing specifically reduces Aβ-mediated endothelial apoptosis. DISCUSSION This work substantiates the potential application of CAIs in clinical trials for AD and CAA.
Collapse
Affiliation(s)
- Elisa Canepa
- Alzheimer’s Center at Temple, Department of Neural Sciences, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, 19140, USA
| | - Rebecca Parodi-Rullan
- Alzheimer’s Center at Temple, Department of Neural Sciences, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, 19140, USA
| | - Rafael Vazquez-Torres
- Alzheimer’s Center at Temple, Department of Neural Sciences, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, 19140, USA
| | - Begona Gamallo-Lana
- Department of Neuroscience and Physiology, Neuroscience Institute, NYU Grossman School of Medicine, New York, NY, 10016, USA
| | - Roberto Guzman-Hernandez
- Alzheimer’s Center at Temple, Department of Neural Sciences, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, 19140, USA
| | - Nicole L. Lemon
- Alzheimer’s Center at Temple, Department of Neural Sciences, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, 19140, USA
| | - Federica Angiulli
- Alzheimer’s Center at Temple, Department of Neural Sciences, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, 19140, USA
| | - Ludovic Debure
- Department on Neurology, Center for Cognitive Neurology, NYU Grossman School of Medicine, New York, NY, 10016, USA
| | - Marc A. Ilies
- Department of Pharmaceutical Sciences and Moulder Center for Drug Discovery Research, Temple University School of Pharmacy, Temple University, Philadelphia, PA, 19140, USA
| | - Leif Østergaard
- Center of Functionally Integrative Neuroscience (CFIN), Department of Clinical Medicine, Aarhus University, 8200 Aarhus, Denmark
| | - Thomas Wisniewski
- Department on Neurology, Center for Cognitive Neurology, NYU Grossman School of Medicine, New York, NY, 10016, USA
| | - Eugenio Gutiérrez-Jiménez
- Center of Functionally Integrative Neuroscience (CFIN), Department of Clinical Medicine, Aarhus University, 8200 Aarhus, Denmark
| | - Adam C. Mar
- Department of Neuroscience and Physiology, Neuroscience Institute, NYU Grossman School of Medicine, New York, NY, 10016, USA
| | - Silvia Fossati
- Alzheimer’s Center at Temple, Department of Neural Sciences, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, 19140, USA
| |
Collapse
|
3
|
Zhao J, Wang X, He Y, Xu P, Lai L, Chung Y, Pan X. The Role of T Cells in Alzheimer's Disease Pathogenesis. Crit Rev Immunol 2023; 43:15-23. [PMID: 37943150 DOI: 10.1615/critrevimmunol.2023050145] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2023]
Abstract
Alzheimer's disease (AD) is a progressive neurodegenerative disorder associated with memory decline and cognitive impairment, which is related to hallmark protein aggregates, amyloid-β (Аβ) plaques and neurofibrillary tangles; the latter are accumulated with hyperphosphorylated Tau protein. Immune cells play an important role in AD pathogenesis. Although the role of T cells in AD remains controversial, studies have shown that T cell deficiency is associated with increased AD pathology. In contrast, transplantation of T cells reduces AD pathology. T cells can help B cells generate anti-Аβ antibody to neutralize the toxin of Аβ and hyperphosphorylated Tau. T cells also activate macrophages to phagocytose misfolded proteins including Аβ and Tau. Recent data have also shown that AD animals have a damaged thymic microenvironment, especially thymic epithelial cells (TECs), resulting in decreased T cell numbers, which contribute to AD pathology. Therefore, regulation of T cell regeneration, for example by rejuvenating the thymic microenvironment, has the potential to be used in the treatment of AD.
Collapse
Affiliation(s)
- Jin Zhao
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Southern Medical University, and Guangdong Provincial Key Laboratory of Single Cell Technology and Application, Guangzhou, Guangdong, 510515, China; ZhuHai Hengqin ImStem Biotechnology Co. Ltd., Hengqin New District Huandao Donglu 1889 Building 3, Zhuhai, Guangdong, 519000, China; Key Laboratory of Mental Health of the Ministry of Education, Guangdong Province Key Laboratory of Psychiatric Disorders, School of Basic Medical Sciences, Southern Medical University, Guangdong, 510515, China
| | - Xiaofang Wang
- ZhuHai Hengqin ImStem Biotechnology Co. Ltd., Hengqin New District Huandao Donglu 1889 Building 3, Zhuhai, Guangdong, 519000, China; ImStem Biotechnology, Inc., 400 Farmington Avenue R1808, Farmington, CT 06030, USA
| | - Yusheng He
- ZhuHai Hengqin ImStem Biotechnology Co. Ltd., Hengqin New District Huandao Donglu 1889 Building 3, Zhuhai, Guangdong, 519000, China
| | - Pingyi Xu
- Department of Neurology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, 510120, China
| | - Laijun Lai
- Department of Allied Health Sciences, University of Connecticut, Storrs, CT, USA; University of Connecticut Stem Cell Institute, University of Connecticut, Storrs, CT, USA
| | - Younggie Chung
- ZhuHai Hengqin ImStem Biotechnology Co. Ltd., Hengqin New District Huandao Donglu 1889 Building 3, Zhuhai, Guangdong, 519000, China; ImStem Biotechnology, Inc., 400 Farmington Avenue R1808, Farmington, CT 06030, USA
| | - Xinghua Pan
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Southern Medical University, and Guangdong Provincial Key Laboratory of Single Cell Technology and Application, Guangzhou, Guangdong, 510515, China; Key Laboratory of Mental Health of the Ministry of Education, Guangdong Province Key Laboratory of Psychiatric Disorders, School of Basic Medical Sciences, Southern Medical University, Guangdong, 510515, China
| |
Collapse
|
4
|
Assessment of sex-related neuropathology and cognitive deficits in the Tg-SwDI mouse model of Alzheimer’s disease. Behav Brain Res 2022; 428:113882. [DOI: 10.1016/j.bbr.2022.113882] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2021] [Revised: 03/17/2022] [Accepted: 04/03/2022] [Indexed: 11/21/2022]
|
5
|
Patel AG, Nehete PN, Krivoshik SR, Pei X, Cho EL, Nehete BP, Ramani MD, Shao Y, Williams LE, Wisniewski T, Scholtzova H. Innate immunity stimulation via CpG oligodeoxynucleotides ameliorates Alzheimer's disease pathology in aged squirrel monkeys. Brain 2021; 144:2146-2165. [PMID: 34128045 PMCID: PMC8502485 DOI: 10.1093/brain/awab129] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2020] [Revised: 12/09/2020] [Accepted: 12/17/2020] [Indexed: 11/15/2022] Open
Abstract
Alzheimer's disease is the most common cause of dementia and the only illness among the top 10 causes of death for which there is no disease-modifying therapy. The failure rate of clinical trials is very high, in part due to the premature translation of successful results in transgenic mouse models to patients. Extensive evidence suggests that dysregulation of innate immunity and microglia/macrophages plays a key role in Alzheimer's disease pathogenesis. Activated resident microglia and peripheral macrophages can display protective or detrimental phenotypes depending on the stimulus and environment. Toll-like receptors (TLRs) are a family of innate immune regulators known to play an important role in governing the phenotypic status of microglia. We have shown in multiple transgenic Alzheimer's disease mouse models that harnessing innate immunity via TLR9 agonist CpG oligodeoxynucleotides (ODNs) modulates age-related defects associated with immune cells and safely reduces amyloid plaques, oligomeric amyloid-β, tau pathology, and cerebral amyloid angiopathy (CAA) while promoting cognitive benefits. In the current study we have used a non-human primate model of sporadic Alzheimer's disease pathology that develops extensive CAA-elderly squirrel monkeys. The major complications in current immunotherapeutic trials for Alzheimer's disease are amyloid-related imaging abnormalities, which are linked to the presence and extent of CAA; hence, the prominence of CAA in elderly squirrel monkeys makes them a valuable model for studying the safety of the CpG ODN-based concept of immunomodulation. We demonstrate that long-term use of Class B CpG ODN 2006 induces a favourable degree of innate immunity stimulation without producing excessive or sustained inflammation, resulting in efficient amelioration of both CAA and tau Alzheimer's disease-related pathologies in association with behavioural improvements and in the absence of microhaemorrhages in aged elderly squirrel monkeys. CpG ODN 2006 has been well established in numerous human trials for a variety of diseases. The present evidence together with our earlier, extensive preclinical research, validates the beneficial therapeutic outcomes and safety of this innovative immunomodulatory approach, increasing the likelihood of CpG ODN therapeutic efficacy in future clinical trials.
Collapse
Affiliation(s)
- Akash G Patel
- Center for Cognitive Neurology and Department of Neurology, New York University School of Medicine, New York, NY 10016, USA
| | - Pramod N Nehete
- Department of Comparative Medicine, the University of Texas MD Anderson Cancer Center, Bastrop, TX 78602, USA
- The University of Texas Graduate School of Biomedical Sciences, Houston, TX 77030, USA
| | - Sara R Krivoshik
- Center for Cognitive Neurology and Department of Neurology, New York University School of Medicine, New York, NY 10016, USA
| | - Xuewei Pei
- Center for Cognitive Neurology and Department of Neurology, New York University School of Medicine, New York, NY 10016, USA
| | - Elizabeth L Cho
- Center for Cognitive Neurology and Department of Neurology, New York University School of Medicine, New York, NY 10016, USA
| | - Bharti P Nehete
- Department of Comparative Medicine, the University of Texas MD Anderson Cancer Center, Bastrop, TX 78602, USA
| | - Margish D Ramani
- Center for Cognitive Neurology and Department of Neurology, New York University School of Medicine, New York, NY 10016, USA
| | - Yongzhao Shao
- Division of Biostatistics, Departments of Population Health and Environmental Medicine, New York University School of Medicine, New York, NY 10016, USA
| | - Lawrence E Williams
- Department of Comparative Medicine, the University of Texas MD Anderson Cancer Center, Bastrop, TX 78602, USA
| | - Thomas Wisniewski
- Center for Cognitive Neurology and Department of Neurology, New York University School of Medicine, New York, NY 10016, USA
- Department of Pathology, New York University School of Medicine, New York, NY 10016, USA
- Department of Psychiatry, New York University School of Medicine, New York, NY 10016, USA
| | - Henrieta Scholtzova
- Center for Cognitive Neurology and Department of Neurology, New York University School of Medicine, New York, NY 10016, USA
- Department of Comparative Medicine, the University of Texas MD Anderson Cancer Center, Bastrop, TX 78602, USA
| |
Collapse
|
6
|
Ashrafian H, Zadeh EH, Khan RH. Review on Alzheimer's disease: Inhibition of amyloid beta and tau tangle formation. Int J Biol Macromol 2020; 167:382-394. [PMID: 33278431 DOI: 10.1016/j.ijbiomac.2020.11.192] [Citation(s) in RCA: 192] [Impact Index Per Article: 38.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2020] [Revised: 11/18/2020] [Accepted: 11/28/2020] [Indexed: 12/19/2022]
Abstract
It is reported that approximately 40 million people are suffering from dementia, globally. Dementia is a group of symptoms that affect neurons and cause some mental disorders, such as losing memory. Alzheimer's disease (AD) which is known as the most common cause of dementia, is one of the top medical care concerns across the world. Although the exact sources of the disease are not understood, is it believed that aggregation of amyloid-beta (Aβ) outside of neuron cells and tau aggregation or neurofibrillary tangles (NFTs) formation inside the cell may play crucial roles. In this paper, we are going to review studies that targeted inhibition of amyloid plaque and tau protein tangle formation, to suppress or postpone AD.
Collapse
Affiliation(s)
- Hossein Ashrafian
- Department of Chemistry and Biochemistry, Ohio State University, Columbus, OH, USA; Department of Chemistry, Sharif University of Technology, Tehran, Iran.
| | | | | |
Collapse
|
7
|
Güell-Bosch J, Lope-Piedrafita S, Esquerda-Canals G, Montoliu-Gaya L, Villegas S. Progression of Alzheimer's disease and effect of scFv-h3D6 immunotherapy in the 3xTg-AD mouse model: An in vivo longitudinal study using Magnetic Resonance Imaging and Spectroscopy. NMR IN BIOMEDICINE 2020; 33:e4263. [PMID: 32067292 DOI: 10.1002/nbm.4263] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/31/2019] [Revised: 12/19/2019] [Accepted: 01/11/2020] [Indexed: 06/10/2023]
Abstract
Alzheimer's disease (AD) is an incurable disease that affects most of the 47 million people estimated as living with dementia worldwide. The main histopathological hallmarks of AD are extracellular β-amyloid (Aβ) plaques and intracellular neurofibrillary tangles (NFTs) composed of hyperphosphorylated tau protein. In recent years, Aβ-immunotherapy has been revealed as a potential tool in AD treatment. One strategy consists of using single-chain variable fragments (scFvs), which avoids the fragment crystallizable (Fc) effects that are supposed to trigger a microglial response, leading to microhemorrhages and vasogenic edemas, as evidenced in clinical trials with bapineuzumab. The scFv-h3D6 generated by our research group derives from this monoclonal antibody, which targets the N-terminal of the Aβ peptide and recognizes monomers, oligomers and fibrils. In this study, 3xTg-AD mice were intraperitoneally and monthly treated with 100 μg of scFv-h3D6 (a dose of ~3.3 mg/kg) or PBS, from 5 to 12 months of age (-mo), the age at which the mice were sacrificed and samples collected for histological and biochemical analyses. During treatments, four monitoring sessions using magnetic resonance imaging and spectroscopy (MRI/MRS) were performed at 5, 7, 9, and 12 months of age. MRI/MRS techniques are widely used in both human and mouse research, allowing to draw an in vivo picture of concrete aspects of the pathology in a non-invasive manner and allowing to monitor its development across time. Compared with the genetic background, 3xTg-AD mice presented a smaller volume in almost all cerebral regions and ages examined, an increase in both the intra and extracellular Aβ1-42 at 12-mo, and an inflammation process at this age, in both the hippocampus (IL-6 and mIns) and cortex (IL-6). In addition, treatment with scFv-h3D6 partially recovered the values in brain volume, and Aβ, IL-6, and mIns concentrations, among others, encouraging further studies with this antibody fragment.
Collapse
Affiliation(s)
- J Güell-Bosch
- Protein Design and Immunotherapy Group, Departament de Bioquímica i Biologia Molecular, Facultat de Biociències, Universitat Autònoma de Barcelona, Cerdanyola del Vallès, Barcelona, Spain
| | - S Lope-Piedrafita
- Servei de Ressonància Magnètica Nuclear, Universitat Autònoma de Barcelona, Cerdanyola del Vallès, Spain
- Centro de Investigación Biomédica en Red-Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Universitat Autònoma de Barcelona, Cerdanyola del Vallès, Spain
| | - G Esquerda-Canals
- Protein Design and Immunotherapy Group, Departament de Bioquímica i Biologia Molecular, Facultat de Biociències, Universitat Autònoma de Barcelona, Cerdanyola del Vallès, Barcelona, Spain
| | - L Montoliu-Gaya
- Protein Design and Immunotherapy Group, Departament de Bioquímica i Biologia Molecular, Facultat de Biociències, Universitat Autònoma de Barcelona, Cerdanyola del Vallès, Barcelona, Spain
| | - S Villegas
- Protein Design and Immunotherapy Group, Departament de Bioquímica i Biologia Molecular, Facultat de Biociències, Universitat Autònoma de Barcelona, Cerdanyola del Vallès, Barcelona, Spain
| |
Collapse
|
8
|
Abstract
This paper considers ethical issues related to early diagnosis and all forms of prevention of Alzheimer disease and related conditions. It offers a critical view of the current state of scientific, clinical, and social responses to the growing number of older people with cognitive challenges, and suggests how priorities going forward should be different from those receiving most attention today. We begin with a review of global policy efforts, consider the fundamental goals of prevention, examine issues surrounding early diagnosis, explore more deeply values associated with efforts to prevent age associated cognitive decline, and conclude by considering often unexplored ethical issues that contextualize the field and should influence our approaches to the future.
Collapse
Affiliation(s)
- Peter J Whitehouse
- Case Western Reserve University, Cleveland, Ohio, USA; Baycrest Health Center, Cleveland, Ohio, USA; Institute of Life Course and Aging, University of Toronto; Intergenerational Schools International, Toronto, Ontario, Canada
| |
Collapse
|
9
|
Yiannopoulou KG, Papageorgiou SG. Current and Future Treatments in Alzheimer Disease: An Update. J Cent Nerv Syst Dis 2020; 12:1179573520907397. [PMID: 32165850 PMCID: PMC7050025 DOI: 10.1177/1179573520907397] [Citation(s) in RCA: 461] [Impact Index Per Article: 92.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2019] [Accepted: 01/24/2020] [Indexed: 01/08/2023] Open
Abstract
Disease-modifying treatment strategies for Alzheimer disease (AD) are still under extensive research. Nowadays, only symptomatic treatments exist for this disease, all trying to counterbalance the neurotransmitter disturbance: 3 cholinesterase inhibitors and memantine. To block the progression of the disease, therapeutic agents are supposed to interfere with the pathogenic steps responsible for the clinical symptoms, classically including the deposition of extracellular amyloid β plaques and intracellular neurofibrillary tangle formation. Other underlying mechanisms are targeted by neuroprotective, anti-inflammatory, growth factor promotive, metabolic efficacious agents and stem cell therapies. Recent therapies have integrated multiple new features such as novel biomarkers, new neuropsychological outcomes, enrollment of earlier populations in the course of the disease, and innovative trial designs. In the near future different specific agents for every patient might be used in a “precision medicine” context, where aberrant biomarkers accompanied with a particular pattern of neuropsychological and neuroimaging findings could determine a specific treatment regimen within a customized therapeutic framework. In this review, we discuss potential disease-modifying therapies that are currently being studied and potential individualized therapeutic frameworks that can be proved beneficial for patients with AD.
Collapse
Affiliation(s)
| | - Sokratis G Papageorgiou
- Cognitive Disorders/Dementia Unit, 2nd Neurological Department, National and Kapodistrian University of Athens, Attikon General University Hospital, Athens, Greece
| |
Collapse
|
10
|
Guillemaud O, Ceyzériat K, Saint-Georges T, Cambon K, Petit F, Ben Haim L, Carrillo-de Sauvage MA, Guillermier M, Bernier S, Hérard AS, Joséphine C, Bémelmans AP, Brouillet E, Hantraye P, Bonvento G, Escartin C. Complex roles for reactive astrocytes in the triple transgenic mouse model of Alzheimer disease. Neurobiol Aging 2020; 90:135-146. [PMID: 32171592 DOI: 10.1016/j.neurobiolaging.2020.02.010] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2019] [Revised: 02/06/2020] [Accepted: 02/12/2020] [Indexed: 12/29/2022]
Abstract
In Alzheimer disease (AD), astrocytes undergo complex changes and become reactive. The consequences of this reaction are still unclear. To evaluate the net impact of reactive astrocytes in AD, we developed viral vectors targeting astrocytes that either activate or inhibit the Janus kinase-signal transducer and activator of transcription 3 (JAK2-STAT3) pathway, a central cascade controlling astrocyte reaction. We aimed to evaluate whether reactive astrocytes contribute to tau as well as amyloid pathologies in the hippocampus of 3xTg-AD mice, an AD model that develops tau hyper-phosphorylation and amyloid deposition. JAK2-STAT3 pathway-mediated modulation of reactive astrocytes in 25% of the hippocampus of 3xTg-AD mice did not significantly influence tau phosphorylation or amyloid processing and deposition at early, advanced, and terminal disease stage. Interestingly, inhibition of the JAK2-STAT3 pathway in hippocampal astrocytes did not improve spatial memory in the Y maze but it did reduce anxiety in the elevated plus maze. Our unique approach to specifically manipulate reactive astrocytes in situ show they may impact behavioral outcomes without influencing tau or amyloid pathology.
Collapse
Affiliation(s)
- Océane Guillemaud
- Université Paris-Saclay, CEA, CNRS, MIRCen, Laboratoire des Maladies Neurodégénératives, Fontenay-aux-Roses, France
| | - Kelly Ceyzériat
- Université Paris-Saclay, CEA, CNRS, MIRCen, Laboratoire des Maladies Neurodégénératives, Fontenay-aux-Roses, France
| | - Thomas Saint-Georges
- Université Paris-Saclay, CEA, CNRS, MIRCen, Laboratoire des Maladies Neurodégénératives, Fontenay-aux-Roses, France
| | - Karine Cambon
- Université Paris-Saclay, CEA, CNRS, MIRCen, Laboratoire des Maladies Neurodégénératives, Fontenay-aux-Roses, France
| | - Fanny Petit
- Université Paris-Saclay, CEA, CNRS, MIRCen, Laboratoire des Maladies Neurodégénératives, Fontenay-aux-Roses, France
| | - Lucile Ben Haim
- Université Paris-Saclay, CEA, CNRS, MIRCen, Laboratoire des Maladies Neurodégénératives, Fontenay-aux-Roses, France
| | | | - Martine Guillermier
- Université Paris-Saclay, CEA, CNRS, MIRCen, Laboratoire des Maladies Neurodégénératives, Fontenay-aux-Roses, France
| | - Sueva Bernier
- Université Paris-Saclay, CEA, CNRS, MIRCen, Laboratoire des Maladies Neurodégénératives, Fontenay-aux-Roses, France
| | - Anne-Sophie Hérard
- Université Paris-Saclay, CEA, CNRS, MIRCen, Laboratoire des Maladies Neurodégénératives, Fontenay-aux-Roses, France
| | - Charlène Joséphine
- Université Paris-Saclay, CEA, CNRS, MIRCen, Laboratoire des Maladies Neurodégénératives, Fontenay-aux-Roses, France
| | - Alexis Pierre Bémelmans
- Université Paris-Saclay, CEA, CNRS, MIRCen, Laboratoire des Maladies Neurodégénératives, Fontenay-aux-Roses, France
| | - Emmanuel Brouillet
- Université Paris-Saclay, CEA, CNRS, MIRCen, Laboratoire des Maladies Neurodégénératives, Fontenay-aux-Roses, France
| | - Philippe Hantraye
- Université Paris-Saclay, CEA, CNRS, MIRCen, Laboratoire des Maladies Neurodégénératives, Fontenay-aux-Roses, France
| | - Gilles Bonvento
- Université Paris-Saclay, CEA, CNRS, MIRCen, Laboratoire des Maladies Neurodégénératives, Fontenay-aux-Roses, France
| | - Carole Escartin
- Université Paris-Saclay, CEA, CNRS, MIRCen, Laboratoire des Maladies Neurodégénératives, Fontenay-aux-Roses, France.
| |
Collapse
|
11
|
A MultiTEP platform-based epitope vaccine targeting the phosphatase activating domain (PAD) of tau: therapeutic efficacy in PS19 mice. Sci Rep 2019; 9:15455. [PMID: 31664089 PMCID: PMC6820729 DOI: 10.1038/s41598-019-51809-2] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2019] [Accepted: 10/08/2019] [Indexed: 11/30/2022] Open
Abstract
Pathological tau correlates well with cognitive impairments in Alzheimer’s disease (AD) patients and therefore represents a promising target for immunotherapy. Targeting an appropriate B cell epitope in pathological tau could in theory produce an effective reduction of pathology without disrupting the function of normal native tau. Recent data demonstrate that the N-terminal region of tau (aa 2-18), termed the “phosphatase activation domain (PAD)”, is hidden within native Tau in a ‘paperclip’-like conformation. Conversely, PAD is exposed in pathological tau and plays an essential role in the inhibition of fast axonal transport and tau polymerization. Thus, we hypothesized that anti-tau2-18 antibodies may safely and specifically reduce pathological tau and prevent further aggregation, which in turn would neutralize tau toxicity. Therefore, we evaluated the immunogenicity and therapeutic efficacy of our MultiTEP platform-based vaccine targeting tau2-18 formulated with AdvaxCpG adjuvant (AV-1980R/A) in PS19 tau transgenic mice. The AV-1980R/A induced extremely high antibody responses and the resulting sera recognized neurofibrillary tangles and plaque-associated dystrophic neurites in AD brain sections. In addition, under non-denaturing conditions AV-1980R/A sera preferentially recognized AD-associated tau. Importantly, vaccination also prevented age-related motor and cognitive deficits in PS19 mice and significantly reduced insoluble total and phosphorylated tau species. Taken together, these findings suggest that predominantly targeting misfolded tau with AV-1980R/A could represent an effective strategy for AD immunotherapy.
Collapse
|
12
|
Drummond E, Goñi F, Liu S, Prelli F, Scholtzova H, Wisniewski T. Potential Novel Approaches to Understand the Pathogenesis and Treat Alzheimer's Disease. J Alzheimers Dis 2019; 64:S299-S312. [PMID: 29562516 DOI: 10.3233/jad-179909] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
There is growing genetic and proteomic data highlighting the complexity of Alzheimer's disease (AD) pathogenesis. Greater use of unbiased "omics" approaches is being increasingly recognized as essential for the future development of effective AD research, that need to better reflect the multiple distinct pathway abnormalities that can drive AD pathology. The track record of success in AD clinical trials thus far has been very poor. In part, this high failure rate has been related to the premature translation of highly successful results in animal models that mirror only limited aspects of AD pathology to humans. We highlight our recent efforts to increase use of human tissue to gain a better understanding of the AD pathogenesis subtype variety and to develop several distinct therapeutic approaches tailored to address this diversity. These therapeutic approaches include the blocking of the Aβ/apoE interaction, stimulation of innate immunity, and the simultaneous blocking of Aβ/tau oligomer toxicity. We believe that future successful therapeutic approaches will need to be combined to better reflect the complexity of the abnormal pathways triggered in AD pathogenesis.
Collapse
Affiliation(s)
- Eleanor Drummond
- Department of Neurology, Center for Cognitive Neurology, NYU School of Medicine, New York, NY, USA
| | - Fernando Goñi
- Department of Neurology, Center for Cognitive Neurology, NYU School of Medicine, New York, NY, USA
| | - Shan Liu
- Department of Neurology, Center for Cognitive Neurology, NYU School of Medicine, New York, NY, USA
| | - Frances Prelli
- Department of Neurology, Center for Cognitive Neurology, NYU School of Medicine, New York, NY, USA
| | - Henrieta Scholtzova
- Department of Neurology, Center for Cognitive Neurology, NYU School of Medicine, New York, NY, USA
| | - Thomas Wisniewski
- Departments of Neurology, Pathology and Psychiatry, Center for Cognitive Neurology, NYU School of Medicine, New York, NY, USA
| |
Collapse
|
13
|
Boutajangout A, Lindberg H, Awwad A, Paul A, Baitalmal R, Almokyad I, Höidén-Guthenberg I, Gunneriusson E, Frejd FY, Härd T, Löfblom J, Ståhl S, Wisniewski T. Affibody-Mediated Sequestration of Amyloid β Demonstrates Preventive Efficacy in a Transgenic Alzheimer's Disease Mouse Model. Front Aging Neurosci 2019; 11:64. [PMID: 30967771 PMCID: PMC6440316 DOI: 10.3389/fnagi.2019.00064] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2018] [Accepted: 03/06/2019] [Indexed: 12/21/2022] Open
Abstract
Different strategies for treatment and prevention of Alzheimer’s disease (AD) are currently under investigation, including passive immunization with anti-amyloid β (anti-Aβ) monoclonal antibodies (mAbs). Here, we investigate the therapeutic potential of a novel type of Aβ-targeting agent based on an affibody molecule with fundamentally different properties to mAbs. We generated a therapeutic candidate, denoted ZSYM73-albumin-binding domain (ABD; 16.8 kDa), by genetic linkage of the dimeric ZSYM73 affibody for sequestering of monomeric Aβ-peptides and an ABD for extension of its in vivo half-life. Amyloid precursor protein (APP)/PS1 transgenic AD mice were administered with ZSYM73-ABD, followed by behavioral examination and immunohistochemistry. Results demonstrated rescued cognitive functions and significantly lower amyloid burden in the treated animals compared to controls. No toxicological symptoms or immunology-related side-effects were observed. To our knowledge, this is the first reported in vivo investigation of a systemically delivered scaffold protein against monomeric Aβ, demonstrating a therapeutic potential for prevention of AD.
Collapse
Affiliation(s)
- Allal Boutajangout
- Center for Cognitive Neurology, New York University Langone Health, New York, NY, United States.,Department of Neurology, New York University Langone Health, New York, NY, United States.,Department of Psychiatry, New York University Langone Health, New York, NY, United States.,Department of Physiology & Neuroscience, New York University Langone Medical Center, New York, NY, United States
| | - Hanna Lindberg
- Department of Protein Science, School of Engineering Sciences in Chemistry, Biotechnology and Health, KTH Royal Institute of Technology, Stockholm, Sweden
| | - Abdulaziz Awwad
- School of Medicine, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Arun Paul
- Center for Cognitive Neurology, New York University Langone Health, New York, NY, United States.,Department of Neurology, New York University Langone Health, New York, NY, United States
| | - Rabaa Baitalmal
- Center for Cognitive Neurology, New York University Langone Health, New York, NY, United States.,Department of Neurology, New York University Langone Health, New York, NY, United States
| | - Ismail Almokyad
- Center for Cognitive Neurology, New York University Langone Health, New York, NY, United States.,Department of Neurology, New York University Langone Health, New York, NY, United States
| | | | | | | | - Torleif Härd
- Department of Chemistry and Biotechnology, Swedish University of Agricultural Sciences (SLU), Uppsala, Sweden
| | - John Löfblom
- Department of Protein Science, School of Engineering Sciences in Chemistry, Biotechnology and Health, KTH Royal Institute of Technology, Stockholm, Sweden
| | - Stefan Ståhl
- Department of Protein Science, School of Engineering Sciences in Chemistry, Biotechnology and Health, KTH Royal Institute of Technology, Stockholm, Sweden
| | - Thomas Wisniewski
- Center for Cognitive Neurology, New York University Langone Health, New York, NY, United States.,Department of Neurology, New York University Langone Health, New York, NY, United States.,Department of Psychiatry, New York University Langone Health, New York, NY, United States.,Department of Pathology, New York University School of Medicine, New York, NY, United States
| |
Collapse
|
14
|
Wisniewski T, Drummond E. Future horizons in Alzheimer's disease research. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2019; 168:223-241. [PMID: 31699317 DOI: 10.1016/bs.pmbts.2019.08.001] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
There are growing genetic, transcriptomic and proteomic data pointing to the complexity of Alzheimer's disease (AD) pathogenesis. Unbiased "omics" approaches are essential for the future development of effective AD research, which will need to be combined and personalized, given that multiple distinct pathways can drive AD pathology. It is essential to gain a better understanding of the AD pathogenesis subtype variety and to develop several distinct therapeutic approaches tailored to address this diversity, as well as the common presence of mixed pathologies. These nonmutually exclusive therapeutic approaches include the targeting of multiple toxic oligomeric species concurrently, targeting the apolipoprotein E/amyloid β interaction and the modulation of innate immunity, as well as more "out of the box" ideas such as targeting infectious agents that may play a role in AD.
Collapse
Affiliation(s)
- Thomas Wisniewski
- Departments of Neurology, Pathology and Psychiatry, Center for Cognitive Neurology, NYU School of Medicine, New York, NY, United States.
| | - Eleanor Drummond
- Central Clinical School, Faculty of Medicine and Health, University of Sydney, Sydney, NSW, Australia
| |
Collapse
|
15
|
Herline K, Drummond E, Wisniewski T. Recent advancements toward therapeutic vaccines against Alzheimer's disease. Expert Rev Vaccines 2018; 17:707-721. [PMID: 30005578 DOI: 10.1080/14760584.2018.1500905] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
INTRODUCTION Alzheimer's disease (AD) is a devastating neurodegenerative disease characterized by protein aggregates of amyloid β (Aβ) and tau. These proteins have normal physiological functions, but in AD, they undergo a conformational change and aggregate as toxic oligomeric and fibrillar species with a high β-sheet content. AREAS COVERED Active and passive immunotherapeutic approaches are among the most attractive methods for targeting misfolded Aβ and tau. Promising preclinical testing of various immunotherapeutic approaches has yet to translate to cognitive benefits in human clinical trials. Knowledge gained from these past failures has led to the development of second-generation Aβ-active immunotherapies, anti-Aβ monoclonal antibodies targeting a wide array of Aβ conformations, and to a number of immunotherapies targeting pathological tau. This review covers the more recent advances in vaccine development for AD from 2016 to present. EXPERT COMMENTARY Due to the complex pathophysiology of AD, greatest clinical efficacy will most likely be achieved by concurrently targeting the most toxic forms of both Aβ and tau.
Collapse
Affiliation(s)
- Krystal Herline
- a Center for Cognitive Neurology , New York University School of Medicine , New York , NY , USA.,b Departments of Neurology , New York University School of Medicine , New York , NY , USA
| | - Eleanor Drummond
- a Center for Cognitive Neurology , New York University School of Medicine , New York , NY , USA.,b Departments of Neurology , New York University School of Medicine , New York , NY , USA
| | - Thomas Wisniewski
- a Center for Cognitive Neurology , New York University School of Medicine , New York , NY , USA.,b Departments of Neurology , New York University School of Medicine , New York , NY , USA.,c Pathology , New York University School of Medicine , New York , NY , USA.,d Psychiatry , New York University School of Medicine , New York , NY , USA
| |
Collapse
|
16
|
Herline K, Prelli F, Mehta P, MacMurray C, Goñi F, Wisniewski T. Immunotherapy to improve cognition and reduce pathological species in an Alzheimer's disease mouse model. ALZHEIMERS RESEARCH & THERAPY 2018; 10:54. [PMID: 29914551 PMCID: PMC6006698 DOI: 10.1186/s13195-018-0384-9] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/21/2018] [Accepted: 05/10/2018] [Indexed: 12/11/2022]
Abstract
Background Alzheimer’s disease (AD) is characterized by physiologically endogenous proteins amyloid beta (Aβ) and tau undergoing a conformational change and accumulating as soluble oligomers and insoluble aggregates. Tau and Aβ soluble oligomers, which contain extensive β-sheet secondary structure, are thought to be the most toxic forms. The objective of this study was to determine the ability of TWF9, an anti-β-sheet conformation antibody (aβComAb), to selectively recognize pathological Aβ and phosphorylated tau in AD human tissue compared with cognitively normal age-matched controls and to improve the performance of old 3xTg-AD mice with advanced pathology in behavioral testing after acute treatment with TWF9. Methods In this study, we used immunohistochemistry, immunoprecipitation, and enzyme-linked immunosorbent assay (ELISA) to characterize TWF9 specificity. We further assessed cognitive performance in old (18–22 months) 3xTg-AD mice using both a Barnes maze and novel object recognition after intraperitoneal administration of TWF9 (4 mg/kg) biweekly for 2 weeks before the start of behavioral testing. Injections continued for the duration of the behavioral testing, which lasted 2 weeks. Results Histological analysis of TWF9 in formalin-fixed paraffin-embedded human control and AD (ABC score: A3B3C3) brain tissue revealed preferential cytoplasmic immunoreactivity in neurons in the AD tissue compared with controls (p < 0.05). Furthermore, ELISA using oligomeric and monomeric Aβ showed a preferential affinity for oligomeric Aβ. Immunoprecipitation studies showed that TWF9 extracted both phosphorylated tau (p < 0.01) and Aβ (p < 0.01) from fresh frozen brain tissues. Results show that treated old 3xTg-AD mice have an enhanced novel object recognition memory (p < 0.01) and Barnes maze performance (p = 0.05) compared with control animals. Overall plaque burden, neurofibrillary tangles, microgliosis, and astrocytosis remained unchanged. Soluble phosphorylated tau was significantly reduced in TWF9-treated mice (p < 0.05), and there was a trend for a reduction in soluble Aβ levels in the brain homogenates of female 3xTg-AD mice (p = 0.06). Conclusions This study shows that acute treatment with an aβComAb can effectively improve performance in behavioral testing without reduction of amyloid plaque burden, and that peripherally administered IgG can affect levels of pathological species in the brain.
Collapse
Affiliation(s)
- Krystal Herline
- Center for Cognitive Neurology and Department of Neurology, New York University School of Medicine, Alexandria, ERSP Rm 802, 450 East 29th Street, New York, NY, USA
| | - Frances Prelli
- Center for Cognitive Neurology and Department of Neurology, New York University School of Medicine, Alexandria, ERSP Rm 802, 450 East 29th Street, New York, NY, USA
| | - Pankaj Mehta
- Department of Immunology, New York State Institute for Basic Research in Developmental Disabilities, Staten Island, USA
| | | | - Fernando Goñi
- Center for Cognitive Neurology and Department of Neurology, New York University School of Medicine, Alexandria, ERSP Rm 802, 450 East 29th Street, New York, NY, USA
| | - Thomas Wisniewski
- Center for Cognitive Neurology and Department of Neurology, New York University School of Medicine, Alexandria, ERSP Rm 802, 450 East 29th Street, New York, NY, USA. .,Departments of Pathology and Psychiatry, New York University School of Medicine, New York, NY, USA.
| |
Collapse
|
17
|
Goñi F, Martá-Ariza M, Herline K, Peyser D, Boutajangout A, Mehta P, Drummond E, Prelli F, Wisniewski T. Anti-β-sheet conformation monoclonal antibody reduces tau and Aβ oligomer pathology in an Alzheimer's disease model. ALZHEIMERS RESEARCH & THERAPY 2018; 10:10. [PMID: 29378642 PMCID: PMC5789573 DOI: 10.1186/s13195-018-0337-3] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/28/2017] [Accepted: 01/04/2018] [Indexed: 02/08/2023]
Abstract
Background Oligomeric forms of amyloid-β (Aβ) and tau are increasing being recognized as key toxins in the pathogenesis of Alzheimer’s disease (AD). Methods We developed a novel monoclonal antibody (mAb), GW-23B7, that recognizes β-sheet secondary structure on pathological oligomers of neurodegenerative diseases. Results The pentameric immunoglobulin M kappa chain (IgMκp) we developed specifically distinguishes intra- and extracellular pathology in human AD brains. Purified GW-23B7 showed a dissociation constant in the nanomolar range for oligomeric Aβ and did not bind monomeric Aβ. In enzyme-linked immunosorbent assays, it recognized oligomeric forms of both Aβ and hyperphosphorylated tau. Aged triple-transgenic AD mice with both Aβ and tau pathology infused intraperitoneally for 2 months showed IgMκp in the soluble brain homogenate, peaking at 24 h postinoculation. Treated mice exhibited significant cognitive rescue on radial arm maze testing compared with vehicle control-infused mice. Immunohistochemically, treatment resulted in a significant decrease of extracellular pathology. Biochemically, treatment resulted in significant reductions of oligomeric forms of Aβ and tau. Conclusions These results suggest that GW-23B7, an anti-β-sheet conformational mAb humanized for clinical trials, may be an effective therapeutic agent for human AD. Electronic supplementary material The online version of this article (10.1186/s13195-018-0337-3) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Fernando Goñi
- Center for Cognitive Neurology and Department of Neurology, New York University School of Medicine, Alexandria, ERSP Rm 802, 450 East 29th Street, New York, NY, USA.
| | - Mitchell Martá-Ariza
- Center for Cognitive Neurology and Department of Neurology, New York University School of Medicine, Alexandria, ERSP Rm 802, 450 East 29th Street, New York, NY, USA
| | - Krystal Herline
- Center for Cognitive Neurology and Department of Neurology, New York University School of Medicine, Alexandria, ERSP Rm 802, 450 East 29th Street, New York, NY, USA
| | - Daniel Peyser
- Center for Cognitive Neurology and Department of Neurology, New York University School of Medicine, Alexandria, ERSP Rm 802, 450 East 29th Street, New York, NY, USA
| | - Allal Boutajangout
- Center for Cognitive Neurology and Department of Neurology, New York University School of Medicine, Alexandria, ERSP Rm 802, 450 East 29th Street, New York, NY, USA.,Department of Psychiatry, New York University School of Medicine, New York, NY, USA
| | - Pankaj Mehta
- Department of Immunology, New York State Institute for Basic Research in Developmental Disabilities, Staten Island, NY, USA
| | - Eleanor Drummond
- Center for Cognitive Neurology and Department of Neurology, New York University School of Medicine, Alexandria, ERSP Rm 802, 450 East 29th Street, New York, NY, USA
| | - Frances Prelli
- Center for Cognitive Neurology and Department of Neurology, New York University School of Medicine, Alexandria, ERSP Rm 802, 450 East 29th Street, New York, NY, USA
| | - Thomas Wisniewski
- Center for Cognitive Neurology and Department of Neurology, New York University School of Medicine, Alexandria, ERSP Rm 802, 450 East 29th Street, New York, NY, USA. .,Department of Pathology, New York University School of Medicine, New York, NY, USA. .,Department of Psychiatry, New York University School of Medicine, New York, NY, USA.
| |
Collapse
|
18
|
Abstract
Currently all prion diseases are without effective treatment and are universally fatal. It is increasingly being recognized that the pathogenesis of many neurodegenerative diseases, such as Alzheimer disease (AD), includes "prion-like" properties. Hence, any effective therapeutic intervention for prion disease could have significant implications for other neurodegenerative diseases. Conversely, therapies that are effective in AD might also be therapeutically beneficial for prion disease. AD-like prion disease has no effective therapy. However, various vaccine and immunomodulatory approaches have shown great success in animal models of AD, with numerous ongoing clinical trials of these potential immunotherapies. More limited evidence suggests that immunotherapies may be effective in prion models and in naturally occurring prion disease. In particular, experimental data suggest that mucosal vaccination against prions can be effective for protection against orally acquired prion infection. Many prion diseases, including natural sheep scrapie, bovine spongiform encephalopathy, chronic wasting disease, and variant Creutzfeldt-Jakob disease, are thought to be acquired peripherally, mainly by oral exposure. Mucosal vaccination would be most applicable to this form of transmission. In this chapter we review various immunologically based strategies which are under development for prion infection.
Collapse
Affiliation(s)
- Thomas Wisniewski
- Center for Cognitive Neurology, New York University School of Medicine, New York, NY, United States; Department of Neurology, New York University School of Medicine, New York, NY, United States; Department of Pathology, New York University School of Medicine, New York, NY, United States; Department of Psychiatry, New York University School of Medicine, New York, NY, United States.
| | - Fernando Goñi
- Center for Cognitive Neurology, New York University School of Medicine, New York, NY, United States; Department of Neurology, New York University School of Medicine, New York, NY, United States
| |
Collapse
|
19
|
Verbeeck C, Carrano A, Chakrabarty P, Jankowsky JL, Das P. Combination of Aβ Suppression and Innate Immune Activation in the Brain Significantly Attenuates Amyloid Plaque Deposition. THE AMERICAN JOURNAL OF PATHOLOGY 2017; 187:2886-2894. [PMID: 28919107 PMCID: PMC5718090 DOI: 10.1016/j.ajpath.2017.08.010] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/04/2017] [Revised: 08/01/2017] [Accepted: 08/07/2017] [Indexed: 01/22/2023]
Abstract
Anti-Aβ clinical trials are currently under way to determine whether preventing amyloid deposition will be beneficial in arresting progression of Alzheimer disease. Both clinical and preclinical studies suggest that antiamyloid strategies are only effective if started at early stages of the disease process in a primary prevention strategy. Because this approach will be difficult to deploy, strategies for secondary prevention aimed at later stages of disease are also needed. In this study, we asked whether combining innate immune activation in the brain with concurrent Aβ suppression could enhance plaque clearance and could improve pathologic outcomes in mice with moderate amyloid pathologic disorder. Starting at 5 months of age, tet-off amyloid precursor protein transgenic mice were treated with doxycycline (dox) to suppress further amyloid precursor protein/Aβ production, and at the same time mice were intracranially injected with adeno-associated virus 1 expressing murine IL-6 (AAV1-mIL-6). Three months later, mice treated with the combination of Aβ suppression and AAV1-mIL-6 showed significantly less plaque pathologic disorder than dox or AAV1-mIL-6 only groups. The combination of AAV1-mIL-6 + dox treatment lowered total plaque burden by >60% versus untreated controls. Treatment with either dox or AAV1-mIL-6 alone was less effective than the combination. Our results suggest a synergistic mechanism by which the up-regulation of mIL-6 was able to improve plaque clearance in the setting of Aβ suppression.
Collapse
Affiliation(s)
- Christophe Verbeeck
- Department of Neuroscience, Mayo Clinic College of Medicine, Jacksonville, Florida
| | - Anna Carrano
- Department of Neuroscience, Mayo Clinic College of Medicine, Jacksonville, Florida
| | - Paramita Chakrabarty
- Department of Neuroscience, Center for Translational Research in Neurodegenerative Disease, McKnight Brain Institute, University of Florida, Gainesville, Florida
| | | | - Pritam Das
- Department of Neuroscience, Mayo Clinic College of Medicine, Jacksonville, Florida.
| |
Collapse
|
20
|
Production of Monoclonal Antibodies to Pathologic β-sheet Oligomeric Conformers in Neurodegenerative Diseases. Sci Rep 2017; 7:9881. [PMID: 28852189 PMCID: PMC5575137 DOI: 10.1038/s41598-017-10393-z] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2017] [Accepted: 08/07/2017] [Indexed: 01/12/2023] Open
Abstract
We describe a novel approach to produce conformational monoclonal antibodies selected to specifically react with the β-sheet secondary structure of pathological oligomeric conformers, characteristic of many neurodegenerative diseases. Contrary to past and current efforts, we utilize a mammalian non-self-antigen as an immunogen. The small, non-self peptide selected was covalently polymerized with glutaraldehyde until it reached a high β-sheet secondary structure content, and species between 10–100kDa that are immunogenic, stable and soluble (p13Bri). Inoculation of p13Bri in mice elicited antibodies to the peptide and the β-sheet secondary structure conformation. Hybridomas were produced and clones selected for their reactivity with at least two different oligomeric conformers from Alzheimer’s, Parkinson and/or Prion diseases. The resulting conformational monoclonals are able to detect pathological oligomeric forms in different human neurodegenerative diseases by ELISA, immunohistochemistry and immunoblots. This technological approach may be useful to develop tools for detection, monitoring and treatment of multiple misfolding disorders.
Collapse
|
21
|
Liu S, Park S, Allington G, Prelli F, Sun Y, Martá-Ariza M, Scholtzova H, Biswas G, Brown B, Verghese PB, Mehta PD, Kwon YU, Wisniewski T. Targeting Apolipoprotein E/Amyloid β Binding by Peptoid CPO_Aβ17-21 P Ameliorates Alzheimer's Disease Related Pathology and Cognitive Decline. Sci Rep 2017; 7:8009. [PMID: 28808293 PMCID: PMC5556019 DOI: 10.1038/s41598-017-08604-8] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2017] [Accepted: 07/12/2017] [Indexed: 02/01/2023] Open
Abstract
Inheritance of the apolipoprotein E4 (apoE4) genotype has been identified as the major genetic risk factor for late onset Alzheimer's disease (AD). Studies have shown that apoE, apoE4 in particular, binds to amyloid-β (Aβ) peptides at residues 12-28 of Aβ and this binding modulates Aβ accumulation and disease progression. We have previously shown in several AD transgenic mice lines that blocking the apoE/Aβ interaction with Aβ12-28 P reduced Aβ and tau-related pathology, leading to cognitive improvements in treated AD mice. Recently, we have designed a small peptoid library derived from the Aβ12-28 P sequence to screen for new apoE/Aβ binding inhibitors with higher efficacy and safety. Peptoids are better drug candidates than peptides due to their inherently more favorable pharmacokinetic properties. One of the lead peptoid compounds, CPO_Aβ17-21 P, diminished the apoE/Aβ interaction and attenuated the apoE4 pro-fibrillogenic effects on Aβ aggregation in vitro as well as apoE4 potentiation of Aβ cytotoxicity. CPO_Aβ17-21 P reduced Aβ-related pathology coupled with cognitive improvements in an AD APP/PS1 transgenic mouse model. Our study suggests the non-toxic, non-fibrillogenic peptoid CPO_Aβ17-21 P has significant promise as a new AD therapeutic agent which targets the Aβ related apoE pathway, with improved efficacy and pharmacokinetic properties.
Collapse
Affiliation(s)
- Shan Liu
- Center for Cognitive Neurology, Department of Neurology, New York University School of Medicine, New York, USA
| | - Shinae Park
- Department of Chemistry and Nanoscience, Ewha Womans University, Seoul, Korea
| | | | - Frances Prelli
- Center for Cognitive Neurology, Department of Neurology, New York University School of Medicine, New York, USA
| | - Yanjie Sun
- Center for Cognitive Neurology, Department of Neurology, New York University School of Medicine, New York, USA
| | - Mitchell Martá-Ariza
- Center for Cognitive Neurology, Department of Neurology, New York University School of Medicine, New York, USA
| | - Henrieta Scholtzova
- Center for Cognitive Neurology, Department of Neurology, New York University School of Medicine, New York, USA
| | - Goutam Biswas
- Department of Chemistry and Nanoscience, Ewha Womans University, Seoul, Korea
| | - Bernard Brown
- Center for Cognitive Neurology, Department of Neurology, New York University School of Medicine, New York, USA
| | - Philip B Verghese
- C2N Diagnostics, Center for Emerging Technologies, 4041 Forest Park Avenue, St. Louis, MO, 63108, USA
| | - Pankaj D Mehta
- Department of Immunology, New York State Institute for Basic Research in Developmental Disabilities, New York, USA
| | - Yong-Uk Kwon
- Department of Chemistry and Nanoscience, Ewha Womans University, Seoul, Korea.
| | - Thomas Wisniewski
- Center for Cognitive Neurology, Departments of Neurology, Psychiatry and Pathology, Neuroscience Institute, New York University School of Medicine, New York, USA.
| |
Collapse
|
22
|
Kuo YC, Rajesh R. A critical overview of therapeutic strategy and advancement for Alzheimer's disease treatment. J Taiwan Inst Chem Eng 2017. [DOI: 10.1016/j.jtice.2017.05.011] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
|
23
|
Innate Immunity Stimulation via Toll-Like Receptor 9 Ameliorates Vascular Amyloid Pathology in Tg-SwDI Mice with Associated Cognitive Benefits. J Neurosci 2017; 37:936-959. [PMID: 28123027 DOI: 10.1523/jneurosci.1967-16.2016] [Citation(s) in RCA: 61] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2016] [Revised: 11/29/2016] [Accepted: 12/06/2016] [Indexed: 11/21/2022] Open
Abstract
Alzheimer's disease (AD) is characterized by the presence of parenchymal amyloid-β (Aβ) plaques, cerebral amyloid angiopathy (CAA) and neurofibrillary tangles. Currently there are no effective treatments for AD. Immunotherapeutic approaches under development are hampered by complications related to ineffectual clearance of CAA. Genome-wide association studies have demonstrated the importance of microglia in AD pathogenesis. Microglia are the primary innate immune cells of the brain. Depending on their activation state and environment, microglia can be beneficial or detrimental. In our prior work, we showed that stimulation of innate immunity with Toll-like receptor 9 agonist, class B CpG (cytosine-phosphate-guanine) oligodeoxynucleotides (ODNs), can reduce amyloid and tau pathologies without causing toxicity in Tg2576 and 3xTg-AD mouse models. However, these transgenic mice have relatively little CAA. In the current study, we evaluated the therapeutic profile of CpG ODN in a triple transgenic mouse model, Tg-SwDI, with abundant vascular amyloid, in association with low levels of parenchymal amyloid deposits. Peripheral administration of CpG ODN, both before and after the development of CAA, negated short-term memory deficits, as assessed by object-recognition tests, and was effective at improving spatial and working memory evaluated using a radial arm maze. These findings were associated with significant reductions of CAA pathology lacking adverse effects. Together, our extensive evidence suggests that this innovative immunomodulation may be a safe approach to ameliorate all hallmarks of AD pathology, supporting the potential clinical applicability of CpG ODN. SIGNIFICANCE STATEMENT Recent genetic studies have underscored the emerging role of microglia in Alzheimer's disease (AD) pathogenesis. Microglia lose their amyloid-β-clearing capabilities with age and as AD progresses. Therefore, the ability to modulate microglia profiles offers a promising therapeutic avenue for reducing AD pathology. Current immunotherapeutic approaches have been limited by poor clearance of a core AD lesion, cerebral amyloid angiopathy (CAA). The present study used Tg-SwDI mice, which have extensive CAA. We found that stimulation of the innate immune system and microglia/macrophage activation via Toll-like receptor 9 using CpG (cytosine-phosphate-guanine) oligodeoxynucleotides (ODNs) leads to cognitive improvements and CAA reduction, without associated toxicity. Our data indicate that this novel concept of immunomodulation represents a safer method to reduce all aspects of AD pathology and provide essential information for potential clinical use of CpG ODN.
Collapse
|
24
|
MultiTEP platform-based DNA epitope vaccine targeting N-terminus of tau induces strong immune responses and reduces tau pathology in THY-Tau22 mice. Vaccine 2017; 35:2015-2024. [PMID: 28320590 DOI: 10.1016/j.vaccine.2017.03.020] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2016] [Revised: 01/31/2017] [Accepted: 03/08/2017] [Indexed: 12/22/2022]
Abstract
BACKGROUND By the time clinical symptoms of Alzheimer's disease (AD) manifest in patients there is already substantial tau pathology in the brain. Recent evidence also suggests that tau pathology can become self-propagating, further accelerating disease progression. Over the last decade several groups have tested the efficacy of protein-based anti-tau immunotherapeutics in various animal models of tauopathy. Here we report on the immunological and therapeutic potency of the first anti-tau DNA vaccine based on the MultiTEP platform, AV-1980D, in THY-Tau22 transgenic mice. METHODS Starting at 3months of age, mice were immunized intramuscularly with AV-1980D vaccine targeting a tau B cell epitope spanning aa2-18 followed by electroporation (EP). Humoral and cellular immune responses in vaccinated animals were analyzed by ELISA and ELISpot, respectively. Neuropathological changes in the brains of experimental and control mice were then analyzed by biochemical (WB and ELISA) and immunohistochemical (IHC) methods at 9months of age. RESULTS EP-mediated AV-1980D vaccinations of THY-Tau22 mice induced activation of Th cells specific to the MultiTEP vaccine platform and triggered robust humoral immunity response specific to tau. Importantly, no activation of potentially harmful autoreactive Th cell responses specific to endogenous tau species was detected. The maximum titers of anti-tau antibodies were reached after two immunizations and remained slightly lower, but steady during five subsequent monthly immunizations. Vaccinations with AV-1980D followed by EP significantly reduced total tau and pS199 and AT180 phosphorylated tau levels in the brains extracts of vaccinated mice, but produced on subtle non-significant effects on other phosphorylated tau species. CONCLUSIONS These data demonstrate that MultiTEP-based DNA epitope vaccination targeting the N-terminus of tau is highly immunogenic and therapeutically potent in the THY-Tau22 mouse model of tauopathy and indicate that EP-mediated DNA immunization is an attractive alternative to protein-based adjuvanted vaccines for inducing high concentrations of anti-tau antibodies.
Collapse
|
25
|
Wang J, Li N, Ma J, Gu Z, Yu L, Fu X, Liu X, Wang J. Effects of an amyloid-beta 1-42 oligomers antibody screened from a phage display library in APP/PS1 transgenic mice. Brain Res 2016; 1635:169-79. [PMID: 26820640 PMCID: PMC4801032 DOI: 10.1016/j.brainres.2016.01.028] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2015] [Revised: 01/01/2016] [Accepted: 01/17/2016] [Indexed: 01/05/2023]
Abstract
We screened anti-Aβ1-42 antibodies from a human Alzheimer's disease (AD) specific single chain variable fragment (scFv) phage display library and assessed their effects in APP/PS1 transgenic mice. Reverse transcription-PCR was used to construct the scFv phage display library, and screening identified 11A5 as an anti-Aβ1-42 antibody. We mixed 11A5 and the monoclonal antibody 6E10 with Aβ1-42 and administered the mixture to Sprague-Dawley rats via intracerebroventricular injection. After 30 days, rats injected with the antibody/Aβ1-42 mixture and those injected with Aβ1-42 alone were tested on the Morris water maze. We also injected 11A5 and 6E10 into APP/PS1 transgenic mice and assessed the concentrations of Aβ in brain and peripheral blood by ELISA at 1-month intervals for 3 months. Finally we evaluated behavior changes in the Morris water maze. Rats injected with Aβ1-42 and mixed antibodies showed better performance in the Morris water maze than did rats injected with Aβ1-42 alone. In APP/PS1 transgenic mice, Aβ concentration was lower in the brains of the antibody-treated group than in the control group, but higher in the peripheral blood. The antibody-treated mice also exhibited improved behavioral performance in the Morris water maze. In conclusion, anti-Aβ1-42 antibodies (11A5) screened from the human scFv antibody phage display library promoted the efflux or clearance of Aβ1-42 and effectively decreased the cerebral Aβ burden in an AD mouse model.
Collapse
Affiliation(s)
- Jianping Wang
- Department of Neurology, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China.
| | - Nan Li
- Department of Neurology, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China; Department of Neurology, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450014, China
| | - Jun Ma
- Department of Gastroenterology, The second Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450014, China
| | - Zhiqiang Gu
- Department of Radiology, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China
| | - Lie Yu
- Department of Neurology, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China
| | - Xiaojie Fu
- Department of Neurology, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China
| | - Xi Liu
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China
| | - Jian Wang
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University, School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
26
|
Ye M, Chung HS, Lee C, Yoon MS, Yu AR, Kim JS, Hwang DS, Shim I, Bae H. Neuroprotective effects of bee venom phospholipase A2 in the 3xTg AD mouse model of Alzheimer's disease. J Neuroinflammation 2016; 13:10. [PMID: 26772975 PMCID: PMC4715334 DOI: 10.1186/s12974-016-0476-z] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2015] [Accepted: 01/05/2016] [Indexed: 11/10/2022] Open
Abstract
Background Alzheimer’s disease (AD) is a severe neuroinflammatory disease. CD4+Foxp3+ regulatory T cells (Tregs) modulate various inflammatory diseases via suppressing Th cell activation. There are increasing evidences that Tregs have beneficial roles in neurodegenerative diseases. Previously, we found the population of Treg cells was significantly increased by bee venom phospholipase A2 (bvPLA2) treatment in vivo and in vitro. Methods To examine the effects of bvPLA2 on AD, bvPLA2 was administered to 3xTg-AD mice, mouse model of Alzheimer’s disease. The levels of amyloid beta (Aβ) deposits in the hippocampus, glucose metabolism in the brain, microglia activation, and CD4+ T cell infiltration were analyzed to evaluate the neuroprotective effect of bvPLA2. Results bvPLA2 treatment significantly enhanced the cognitive function of the 3xTg-AD mice and increased glucose metabolism, as assessed with 18F-2 fluoro-2-deoxy-D-glucose ([F-18] FDG) positron emission tomography (PET). The levels of Aβ deposits in the hippocampus were dramatically decreased by bvPLA2 treatment. This neuroprotective effect of bvPLA2 was associated with microglial deactivation and reduction in CD4+ T cell infiltration. Interestingly, the neuroprotective effects of bvPLA2 were abolished in Treg-depleted mice. Conclusions The present studies strongly suggest that the increase of Treg population by bvPLA2 treatment might inhibit progression of AD in the 3xTg AD mice.
Collapse
Affiliation(s)
- Minsook Ye
- Department of Physiology, College of Korean Medicine, Kyung Hee University, #1 Hoegi-dong, Dongdaemoon-ku, Seoul, 130-701, Republic of Korea.
| | - Hwan-Suck Chung
- Korean Medicine (KM)-Application Center, Korea Institute of Oriental Medicine (KIOM), 70, Cheomdan-ro, Dong-gu, Daegu, 41062, Republic of Korea.
| | - Chanju Lee
- Department of Physiology, College of Korean Medicine, Kyung Hee University, #1 Hoegi-dong, Dongdaemoon-ku, Seoul, 130-701, Republic of Korea.
| | - Moon Sik Yoon
- Department of Physiology, College of Korean Medicine, Kyung Hee University, #1 Hoegi-dong, Dongdaemoon-ku, Seoul, 130-701, Republic of Korea.
| | - A Ram Yu
- Molecular Imaging Research Center, Korea Institute of Radiological and Medical Sciences, University of Science and Technology, #215-4 Gongneug-dong, Nowon-ku, Seoul, 139-241, Republic of Korea.
| | - Jin Su Kim
- Molecular Imaging Research Center, Korea Institute of Radiological and Medical Sciences, University of Science and Technology, #215-4 Gongneug-dong, Nowon-ku, Seoul, 139-241, Republic of Korea.
| | - Deok-Sang Hwang
- Department of Obstetrics and Gynecology, College of Korean Medicine, Kyung Hee University, #1 Hoegi-dong, Dongdaemoon-ku, Seoul, 130-701, Republic of Korea.
| | - Insop Shim
- Acupuncture and Meridian Science Research Center, College of Korean Medical Science Graduate School, Kyung Hee University, #1 Hoegi-dong, Dongdaemoon-ku, Seoul, 130-701, Republic of Korea.
| | - Hyunsu Bae
- Department of Physiology, College of Korean Medicine, Kyung Hee University, #1 Hoegi-dong, Dongdaemoon-ku, Seoul, 130-701, Republic of Korea.
| |
Collapse
|
27
|
Wisniewski T, Drummond E. Developing therapeutic vaccines against Alzheimer's disease. Expert Rev Vaccines 2015; 15:401-15. [PMID: 26577574 PMCID: PMC4940858 DOI: 10.1586/14760584.2016.1121815] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2015] [Accepted: 11/16/2015] [Indexed: 01/08/2023]
Abstract
Alzheimer's disease (AD) is the most common form of dementia worldwide. It is characterized by an imbalance between the production and clearance of amyloid β (Aβ) and tau proteins. In AD these normal proteins accumulate, leading to aggregation and a conformational change forming oligomeric and fibrillary species with a high β-sheet content. Active and passive immunotherapeutic approaches result in dramatic reduction of Aβ pathology in AD animal models. However, there is much more limited evidence in human studies of significant clinical benefits from these strategies and it is becoming apparent that they may only be effective very early in AD. Vaccination targeting only tau pathology has shown benefits in some mouse studies but human studies are limited. Greater therapeutic efficacy for the next generation of vaccine approaches will likely benefit from specifically targeting the most toxic species of Aβ and tau, ideally simultaneously.
Collapse
Affiliation(s)
- Thomas Wisniewski
- Center for Cognitive Neurology, New York University School of Medicine, Alexandria ERSP, 450 East 29 Street, New York, NY 10016
- Department of Neurology, New York University School of Medicine, Alexandria ERSP, 450 East 29 Street, New York, NY 10016
- Department of Pathology, New York University School of Medicine, Alexandria ERSP, 450 East 29 Street, New York, NY 10016
- Department of Psychiatry, New York University School of Medicine, Alexandria ERSP, 450 East 29 Street, New York, NY 10016
| | - Eleanor Drummond
- Center for Cognitive Neurology, New York University School of Medicine, Alexandria ERSP, 450 East 29 Street, New York, NY 10016
- Department of Neurology, New York University School of Medicine, Alexandria ERSP, 450 East 29 Street, New York, NY 10016
| |
Collapse
|
28
|
Teoh CL, Su D, Sahu S, Yun SW, Drummond E, Prelli F, Lim S, Cho S, Ham S, Wisniewski T, Chang YT. Chemical Fluorescent Probe for Detection of Aβ Oligomers. J Am Chem Soc 2015; 137:13503-9. [PMID: 26218347 PMCID: PMC4756585 DOI: 10.1021/jacs.5b06190] [Citation(s) in RCA: 141] [Impact Index Per Article: 14.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Aggregation of amyloid β-peptide (Aβ) is implicated in the pathology of Alzheimer's disease (AD), with the soluble, Aβ oligomeric species thought to be the critical pathological species. Identification and characterization of intermediate species formed during the aggregation process is crucial to the understanding of the mechanisms by which oligomeric species mediate neuronal toxicity and following disease progression. Probing these species proved to be extremely challenging, as evident by the lack of reliable sensors, due to their heterogeneous and transient nature. We describe here an oligomer-specific fluorescent chemical probe, BoDipy-Oligomer (BD-Oligo), developed through the use of the diversity-oriented fluorescent library approach (DOFLA) and high-content, imaging-based screening. This probe enables dynamic oligomer monitoring during fibrillogenesis in vitro and shows in vivo Aβ oligomers staining possibility in the AD mice model.
Collapse
Affiliation(s)
- Chai Lean Teoh
- Laboratory of Bioimaging Probe Development, Singapore Bioimaging Consortium (SBIC), Agency for Science, Technology and Research (A*STAR), 11 Biopolis Way, 02-02 Helios, Biopolis, Singapore 138667, Singapore
| | - Dongdong Su
- Laboratory of Bioimaging Probe Development, Singapore Bioimaging Consortium (SBIC), Agency for Science, Technology and Research (A*STAR), 11 Biopolis Way, 02-02 Helios, Biopolis, Singapore 138667, Singapore
| | - Srikanta Sahu
- Laboratory of Bioimaging Probe Development, Singapore Bioimaging Consortium (SBIC), Agency for Science, Technology and Research (A*STAR), 11 Biopolis Way, 02-02 Helios, Biopolis, Singapore 138667, Singapore
| | - Seong-Wook Yun
- Laboratory of Bioimaging Probe Development, Singapore Bioimaging Consortium (SBIC), Agency for Science, Technology and Research (A*STAR), 11 Biopolis Way, 02-02 Helios, Biopolis, Singapore 138667, Singapore
| | - Eleanor Drummond
- Department of Neurology and the Center for Cognitive Neurology, New York University School of Medicine, Alexandria ERSP, Room 802, 450 East 29th Street, New York, New York 10016, United States
| | - Frances Prelli
- Department of Neurology and the Center for Cognitive Neurology, New York University School of Medicine, Alexandria ERSP, Room 802, 450 East 29th Street, New York, New York 10016, United States
| | - Sulgi Lim
- Department of Chemistry and the Center for Fluctuating Thermodynamics, Sookmyung Women’s University, Cheongpa-ro 47-gil 100, Yongsan-Ku, Seoul 140-742, Korea
| | - Sunhee Cho
- Department of Chemistry and the Center for Fluctuating Thermodynamics, Sookmyung Women’s University, Cheongpa-ro 47-gil 100, Yongsan-Ku, Seoul 140-742, Korea
| | - Sihyun Ham
- Department of Chemistry and the Center for Fluctuating Thermodynamics, Sookmyung Women’s University, Cheongpa-ro 47-gil 100, Yongsan-Ku, Seoul 140-742, Korea
| | - Thomas Wisniewski
- Departments of Neurology, Pathology and Psychiatry and the Center for Cognitive Neurology, New York University School of Medicine, Alexandria ERSP, Room 802, 450 East 29th Street, New York, New York 10016, United States
| | - Young-Tae Chang
- Laboratory of Bioimaging Probe Development, Singapore Bioimaging Consortium (SBIC), Agency for Science, Technology and Research (A*STAR), 11 Biopolis Way, 02-02 Helios, Biopolis, Singapore 138667, Singapore
- Department of Chemistry and MedChem Program, Life Sciences Institute, National University of Singapore, 3 Science Drive 3, Singapore 117543, Singapore
| |
Collapse
|
29
|
Abstract
Alzheimer's disease (AD) is the most prevalent form of dementia worldwide and is an emerging global epidemic. It is characterized by an imbalance between production and clearance of amyloid β (Aβ) and tau proteins. Oligomeric forms of Aβ and tau are believed to be the most toxic. Dramatic results from AD animal models showed great promise for active and passive immune therapies targeting Aβ. However, there is very limited evidence in human studies of the clinical benefits from these approaches. Immunotherapies targeting only tau pathology have had some success but are limited so far to mouse models. The majority of current methods is based on immunological targeting of a self-protein; hence, benefits need to be balanced against risks of stimulating excessive autoimmune toxic inflammation. For greater efficacy the next generation of vaccines needs to focus more on concurrently targeting all the intermediate toxic conformers of oligomeric Aβ and tau species.
Collapse
Affiliation(s)
- Thomas Wisniewski
- Department of Neurology, Center for Cognitive Neurology, New York University School of Medicine, Alexandria ERSP, 450 East 29(th) Street, New York, NY 10016, USA; Department of Pathology, New York University School of Medicine, Alexandria ERSP, 450 East 29(th) Street, New York, NY 10016, USA; Department of Psychiatry, New York University School of Medicine, Alexandria ERSP, 450 East 29(th) Street, New York, NY 10016, USA.
| | - Fernando Goñi
- Department of Neurology, Center for Cognitive Neurology, New York University School of Medicine, Alexandria ERSP, 450 East 29(th) Street, New York, NY 10016, USA
| |
Collapse
|
30
|
Kohyama K, Matsumoto Y. Alzheimer's disease and immunotherapy: what is wrong with clinical trials? Immunotargets Ther 2015; 4:27-34. [PMID: 27471709 PMCID: PMC4918245 DOI: 10.2147/itt.s49923] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Alzheimer’s disease (AD) is characterized by progressive neurodegeneration and is the most common cause of dementia. Immunotherapy has recently been regarded as a potential treatment for AD. This stems from the fact that the clinical and pathological findings from the active AD vaccine trial suggests that such vaccine therapy may be effective for AD. However, this trial was halted because of the occurrence of meningoencephalitis in some patients. Avoiding excessive immune reaction is necessary for the success of vaccine therapy. For this purpose, adjuvant-free vaccine therapies (eg, passive immunization or DNA vaccines) are currently under investigation. However, the results of clinical trials employing both active and passive anti-amyloid-beta immunotherapy have been unsatisfactory. In this article, we will analyze the reasons for the limited efficacy of currently available immunotherapies and discuss the effectiveness of new vaccine therapies. Finally, we will speculate on the possibility of its clinical application.
Collapse
Affiliation(s)
- Kuniko Kohyama
- Department of Brain Development and Neural Regeneration, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan
| | - Yoh Matsumoto
- Department of Sensory and Motor Systems, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan; Immunotherapy Development Inc., Saitama, Japan; Geriatric Health Services Facility "Asahigaoka", Saitama, Japan
| |
Collapse
|
31
|
Goñi F, Mathiason CK, Yim L, Wong K, Hayes-Klug J, Nalls A, Peyser D, Estevez V, Denkers N, Xu J, Osborn DA, Miller KV, Warren RJ, Brown DR, Chabalgoity JA, Hoover EA, Wisniewski T. Mucosal immunization with an attenuated Salmonella vaccine partially protects white-tailed deer from chronic wasting disease. Vaccine 2014; 33:726-33. [PMID: 25539804 DOI: 10.1016/j.vaccine.2014.11.035] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2014] [Revised: 11/13/2014] [Accepted: 11/19/2014] [Indexed: 10/24/2022]
Abstract
Prion disease is a unique category of illness, affecting both animals and humans, in which the underlying pathogenesis is related to a conformational change of a normal, self-protein called PrP(C) (C for cellular) to a pathological and infectious conformer known as PrP(Sc) (Sc for scrapie). Bovine spongiform encephalopathy (BSE), a prion disease believed to have arisen from feeding cattle with prion contaminated meat and bone meal products, crossed the species barrier to infect humans. Chronic wasting disease (CWD) infects large numbers of deer and elk, with the potential to infect humans. Currently no prionosis has an effective treatment. Previously, we have demonstrated we could prevent transmission of prions in a proportion of susceptible mice with a mucosal vaccine. In the current study, white-tailed deer were orally inoculated with attenuated Salmonella expressing PrP, while control deer were orally inoculated with vehicle attenuated Salmonella. Once a mucosal response was established, the vaccinated animals were boosted orally and locally by application of polymerized recombinant PrP onto the tonsils and rectal mucosa. The vaccinated and control animals were then challenged orally with CWD-infected brain homogenate. Three years post CWD oral challenge all control deer developed clinical CWD (median survival 602 days), while among the vaccinated there was a significant prolongation of the incubation period (median survival 909 days; p=0.012 by Weibull regression analysis) and one deer has remained CWD free both clinically and by RAMALT and tonsil biopsies. This negative vaccinate has the highest titers of IgA in saliva and systemic IgG against PrP. Western blots showed that immunoglobulins from this vaccinate react to PrP(CWD). We document the first partially successful vaccination for a prion disease in a species naturally at risk.
Collapse
Affiliation(s)
- Fernando Goñi
- Department of Neurology, New York University School of Medicine, 550 First Avenue, New York, NY 10016, United States
| | - Candace K Mathiason
- College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, CO, United States
| | - Lucia Yim
- Laboratory for Vaccine Research, Department of Biotechnology, Instituto de Higiene, Facultad de Medicina, Universidad de la República, Montevideo, Uruguay
| | - Kinlung Wong
- Department of Neurology, New York University School of Medicine, 550 First Avenue, New York, NY 10016, United States
| | - Jeanette Hayes-Klug
- College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, CO, United States
| | - Amy Nalls
- College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, CO, United States
| | - Daniel Peyser
- Department of Neurology, New York University School of Medicine, 550 First Avenue, New York, NY 10016, United States
| | - Veronica Estevez
- Laboratory for Vaccine Research, Department of Biotechnology, Instituto de Higiene, Facultad de Medicina, Universidad de la República, Montevideo, Uruguay
| | - Nathaniel Denkers
- College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, CO, United States
| | - Jinfeng Xu
- Department of Population Health, New York University School of Medicine, 550 First Avenue, New York, NY 10016, United States
| | - David A Osborn
- Warnell School of Forestry and Natural Resources, University of Georgia, United States
| | - Karl V Miller
- Warnell School of Forestry and Natural Resources, University of Georgia, United States
| | - Robert J Warren
- Warnell School of Forestry and Natural Resources, University of Georgia, United States
| | - David R Brown
- Department of Biology and Biochemistry, University of Bath, UK
| | - Jose A Chabalgoity
- Laboratory for Vaccine Research, Department of Biotechnology, Instituto de Higiene, Facultad de Medicina, Universidad de la República, Montevideo, Uruguay
| | - Edward A Hoover
- College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, CO, United States
| | - Thomas Wisniewski
- Department of Neurology, New York University School of Medicine, 550 First Avenue, New York, NY 10016, United States; Department of Pathology, New York University School of Medicine, 550 First Avenue, New York, NY 10016, United States; Department of Psychiatry, New York University School of Medicine, 550 First Avenue, New York, NY 10016, United States.
| |
Collapse
|
32
|
Scholtzova H, Chianchiano P, Pan J, Sun Y, Goñi F, Mehta PD, Wisniewski T. Amyloid β and Tau Alzheimer's disease related pathology is reduced by Toll-like receptor 9 stimulation. Acta Neuropathol Commun 2014; 2:101. [PMID: 25178404 PMCID: PMC4171548 DOI: 10.1186/s40478-014-0101-2] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2014] [Accepted: 08/11/2014] [Indexed: 12/20/2022] Open
Abstract
Alzheimer’s disease (AD) is the most common cause of dementia, and currently, there is no effective treatment. The major neuropathological lesions in AD are accumulation of amyloid β (Aβ) as amyloid plaques and congophilic amyloid angiopathy, as well as aggregated tau in the form of neurofibrillary tangles (NFTs). In addition, inflammation and microglia/macrophage function play an important role in AD pathogenesis. We have hypothesized that stimulation of the innate immune system via Toll-like receptor 9 (TLR9) agonists, such as type B CpG oligodeoxynucleotides (ODNs), might be an effective way to ameliorate AD related pathology. We have previously shown in the Tg2576 AD model that CpG ODN can reduce amyloid deposition and prevent cognitive deficits. In the present study, we used the 3xTg-AD mice with both Aβ and tau related pathology. The mice were divided into 2 groups treated from 7 to 20 months of age, prior to onset of pathology and from 11 to 18 months of age, when pathology is already present. We demonstrated that immunomodulatory treatment with CpG ODN reduces both Aβ and tau pathologies, as well as levels of toxic oligomers, in the absence of any apparent inflammatory toxicity, in both animal groups. This pathology reduction is associated with a cognitive rescue in the 3xTg-AD mice. Our data indicate that modulation of microglial function via TLR9 stimulation is effective at ameliorating all the cardinal AD related pathologies in an AD mouse model mice suggesting such an approach would have a greater chance of achieving clinical efficacy.
Collapse
|
33
|
Wisniewski T, Goñi F. Immunotherapy for Alzheimer's disease. Biochem Pharmacol 2014; 88:499-507. [PMID: 24412277 PMCID: PMC3972315 DOI: 10.1016/j.bcp.2013.12.020] [Citation(s) in RCA: 71] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2013] [Revised: 12/24/2013] [Accepted: 12/24/2013] [Indexed: 02/08/2023]
Abstract
Alzheimer's disease (AD) is the most common cause of dementia worldwide. In AD the normal soluble amyloid β (sAβ) peptide is converted into oligomeric/fibrillar Aβ. The oligomeric forms of Aβ are thought to be the most toxic, while fibrillar Aβ becomes deposited as amyloid plaques and congophilic angiopathy, which serve as neuropathological markers of the disease. In addition the accumulation of abnormally phosphorylated tau as soluble toxic oligomers and as neurofibrillary tangles is a critical part of the pathology. Numerous therapeutic interventions are under investigation to prevent and treat AD. Among the more exciting and advanced of these approaches is vaccination. Active and passive Immunotherapy targeting only Aβ has been successful in many AD model animal trials; however, the more limited human data has shown much less benefit so far, with encephalitis occurring in a minority of patients treated with active immunization and vasogenic edema or amyloid-related imaging abnormalities (ARIA) being a complication in some passive immunization trials. Therapeutic intervention targeting only tau has been tested only in mouse models; and no approaches targeting both pathologies concurrently has been attempted, until very recently. The immune approaches tried so far were targeting a self-protein, albeit in an abnormal conformation; however, effective enhanced clearance of the disease associated conformer has to be balanced with the potential risk of stimulating excessive toxic inflammation. The design of future more effective immunomodulatory approaches will need to target all aspects of AD pathology, as well as specifically targeting pathological oligomeric conformers, without the use of any self-antigen.
Collapse
Affiliation(s)
- Thomas Wisniewski
- Departments of Neurology, New York University School of Medicine, Alexandria ERSP, 450 East 29th Street, New York, NY 10016, United States; Departments of Pathology, New York University School of Medicine, Alexandria ERSP, 450 East 29th Street, New York, NY 10016, United States; Departments of Psychiatry, New York University School of Medicine, Alexandria ERSP, 450 East 29th Street, New York, NY 10016, United States.
| | - Fernando Goñi
- Departments of Neurology, New York University School of Medicine, Alexandria ERSP, 450 East 29th Street, New York, NY 10016, United States
| |
Collapse
|
34
|
Boutajangout A, Wisniewski T. Tau-based therapeutic approaches for Alzheimer's disease - a mini-review. Gerontology 2014; 60:381-5. [PMID: 24732638 PMCID: PMC4149810 DOI: 10.1159/000358875] [Citation(s) in RCA: 67] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2013] [Accepted: 01/21/2014] [Indexed: 11/19/2022] Open
Abstract
The accumulation of aggregated, hyperphosphorylated tau as neurofibrillary tangles and neuropil threads are cardinal features of Alzheimer's disease (AD). The other lesions found in AD include amyloid plaques and congophilic amyloid angiopathy, both associated with the extracellular accumulation of the amyloid-beta (Aβ) peptide. AD is the most common cause of dementia globally. Currently, there are no effective means to treat AD or even to slow it down. The dominant theory for the causation of AD is the amyloid cascade hypothesis, which suggests that the aggregation of Aβ as oligomers and amyloid plaques is central to the pathogenesis of AD. Numerous therapies have been developed directed to Aβ-related pathology, in particular various immunotherapeutic approaches. So far all of these have failed in clinical trials. Recently, there has been more focus on therapy directed to tau-related pathology, which correlates better with the cognitive status of patients, compared to the amyloid burden. Immunotherapeutic targeting of tau pathology has shown great potential in treating tau pathologies in mouse models of AD. A number of studies have shown the efficacy of both passive and active immunization. This review summarizes recent advances in therapy targeting pathological tau protein, in particular focusing on immunotherapeutic approaches which are showing great promise.
Collapse
Affiliation(s)
- Allal Boutajangout
- Department of Neurology, New York University School of Medicine, 450 East 29 Street, New York. NY. 10016
- Department of Psychiatry, New York University School of Medicine, 450 East 29 Street, New York. NY. 10016
- Department of Physiology & Neuroscience, New York University School of Medicine, 450 East 29 Street, New York. NY. 10016
| | - Thomas Wisniewski
- Department of Neurology, New York University School of Medicine, 450 East 29 Street, New York. NY. 10016
- Department of Pathology, New York University School of Medicine, 450 East 29 Street, New York. NY. 10016
- Department of Psychiatry, New York University School of Medicine, 450 East 29 Street, New York. NY. 10016
| |
Collapse
|
35
|
Sabharwal P, Wisniewski T. Novel immunological approaches for the treatment of Alzheimer's disease. ZHONGGUO XIAN DAI SHEN JING JI BING ZA ZHI 2014; 14:139-151. [PMID: 25429302 PMCID: PMC4241771 DOI: 10.3969/j.issn.1672-6731.2014.03.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Alzheimer's disease (AD), the most prevalent form of dementia worldwide, can be deemed as the next global health epidemic. The biochemistry underlying deposition of amyloid beta (A β) and hyperphosphorylated tau aggregates in AD has been extensively studied. The oligomeric forms of A β that are derived from the normal soluble A β peptides are believed to be the most toxic. However, it is the fibrillar Aβ form that aggregates as amyloid plaques and cerebral amyloid angiopathy, which serve as pathological hallmarks of AD. Moreover, deposits of abnormally phosphorylated tau that form soluble toxic oligomers and then accumulate as neurofibrillary tangles are an essential part of AD pathology. Currently, many strategies are being tested that either inhibit, eradicate or prevent the development of plaques in AD. An exciting new approach on the horizon is the immunization approach. Dramatic results from AD animal models have shown promise for active and passive immune therapies targeting A β. However, there is very limited data in humans that suggests a clear benefit. Some hurdles faced with these studies arise from complications noted with therapy. Encephalitis has been reported in trials of active immunization and vasogenic edema or amyloid - related imaging abnormalities (ARIA) has been reported with passive immunization in a minority of patients. As yet, therapies targeting only tau are still limited to mouse models with few studies targeting both pathologies. As the majority of approaches tried so far are based on targeting a self - protein, though in an abnormal conformation, benefits of therapy need to be balanced against the possible risks of stimulating excessive toxic inflammation. For better efficacy, future strategies will need to focus on the toxic oligomers and targeting all aspects of AD pathology.
Collapse
Affiliation(s)
- Priyanka Sabharwal
- Department of Neurology, New York University School of Medicine, New York, USA
| | - Thomas Wisniewski
- Department of Neurology, New York University School of Medicine, New York, USA
- Department of Pathology, New York University School of Medicine, New York, USA
- Department of Psychiatry, New York University School of Medicine, New York, USA
| |
Collapse
|