1
|
Zhang S, Lu J, Jin Z, Xu H, Zhang D, Chen J, Wang J. Gut microbiota metabolites: potential therapeutic targets for Alzheimer's disease? Front Pharmacol 2024; 15:1459655. [PMID: 39355779 PMCID: PMC11442227 DOI: 10.3389/fphar.2024.1459655] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2024] [Accepted: 09/05/2024] [Indexed: 10/03/2024] Open
Abstract
Background Alzheimer's disease (AD) is a neurodegenerative disease characterized by progressive decline in cognitive function, which significantly increases pain and social burden. However, few therapeutic interventions are effective in preventing or mitigating the progression of AD. An increasing number of recent studies support the hypothesis that the gut microbiome and its metabolites may be associated with upstream regulators of AD pathology. Methods In this review, we comprehensively explore the potential mechanisms and currently available interventions targeting the microbiome for the improvement of AD. Our discussion is structured around modern research advancements in AD, the bidirectional communication between the gut and brain, the multi-target regulatory effects of microbial metabolites on AD, and therapeutic strategies aimed at modulating gut microbiota to manage AD. Results The gut microbiota plays a crucial role in the pathogenesis of AD through continuous bidirectional communication via the microbiota-gut-brain axis. Among these, microbial metabolites such as lipids, amino acids, bile acids and neurotransmitters, especially sphingolipids and phospholipids, may serve as central components of the gut-brain axis, regulating AD-related pathogenic mechanisms including β-amyloid metabolism, Tau protein phosphorylation, and neuroinflammation. Additionally, interventions such as probiotic administration, fecal microbiota transplantation, and antibiotic use have also provided evidence supporting the association between gut microbiota and AD. At the same time, we propose an innovative strategy for treating AD: a healthy lifestyle combined with targeted probiotics and other potential therapeutic interventions, aiming to restore intestinal ecology and microbiota balance. Conclusion Despite previous efforts, the molecular mechanisms by which gut microbes act on AD have yet to be fully described. However, intestinal microorganisms may become an essential target for connecting the gut-brain axis and improving the symptoms of AD. At the same time, it requires joint exploration by multiple centers and multiple disciplines.
Collapse
Affiliation(s)
- Shanshan Zhang
- The School to Changchun University of Chinese Medicine, Changchun, China
| | - Jing Lu
- Research Center of Traditional Chinese Medicine, The Affiliated Hospital to Changchun University of Chinese Medicine, Changchun, China
| | - Ziqi Jin
- The School to Changchun University of Chinese Medicine, Changchun, China
| | - Hanying Xu
- Department of Encephalopathy, The Affiliated Hospital to Changchun University of Chinese Medicine, Changchun, China
| | - Dongmei Zhang
- Research Center of Traditional Chinese Medicine, The Affiliated Hospital to Changchun University of Chinese Medicine, Changchun, China
| | - Jianan Chen
- The School to Changchun University of Chinese Medicine, Changchun, China
| | - Jian Wang
- Department of Encephalopathy, The Affiliated Hospital to Changchun University of Chinese Medicine, Changchun, China
| |
Collapse
|
2
|
Chen H, Li Y, Gao J, Cheng Q, Liu L, Cai R. Activation of Pgk1 Results in Reduced Protein Aggregation in Diverse Neurodegenerative Conditions. Mol Neurobiol 2023; 60:5090-5101. [PMID: 37249790 DOI: 10.1007/s12035-023-03389-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2022] [Accepted: 05/05/2023] [Indexed: 05/31/2023]
Abstract
The prevention of protein condensates has emerged as a new drug target to treat diverse neurodegenerative disorders. We previously reported that terazosin (TZ), a prescribed antagonist of the α1 adrenergic receptor, is an activator of phosphoglycerate kinase 1 (Pgk1) and Hsp90. In this study, we aimed to determine whether TZ prevents the formation of diverse pathological condensates in cell cultures and animal disease models. In primary neuron culture, TZ treatment reduced both the protein density and abundance of fused in sarcoma (FUS)-P525L-GFP, a disease-associated mutant form of FUS. Regarding the mechanism, we found that increased intracellular ATP levels were critical for the reduction in protein aggregate density. In addition, Hsp90 activation by TZ enhanced Hsp90 interaction with ULK1, a master regulator of autophagy. Through in vivo studies, we examined neuron-specific overexpression of tau in Drosophila, mouse models of APP/PS1 Alzheimer's disease (AD), and a rat model of multiple system atrophy (MSA) via the viral expression of α-synuclein in the striatum. TZ prevented and reversed the formation of pathological protein condensates. Together, our results suggest that activation of Pgk1 in cytosol may dissolve pathological protein aggregates via increased ATP levels and degrade these proteins via autophagy; the FUS-P525L degradation pathway in nucleus is unclear.
Collapse
Affiliation(s)
- Hao Chen
- Department of Biochemistry and Molecular Biology School of Basic Medicine, Capital Medical University, Youanmen, Beijing, 100069, China
| | - Yajie Li
- Department of Biochemistry and Molecular Biology School of Basic Medicine, Capital Medical University, Youanmen, Beijing, 100069, China
| | - Jingwen Gao
- Department of Biochemistry and Molecular Biology School of Basic Medicine, Capital Medical University, Youanmen, Beijing, 100069, China
| | - Qi Cheng
- Department of Biochemistry and Molecular Biology School of Basic Medicine, Capital Medical University, Youanmen, Beijing, 100069, China
| | - Lei Liu
- Department of Biochemistry and Molecular Biology School of Basic Medicine, Capital Medical University, Youanmen, Beijing, 100069, China.
| | - Rong Cai
- School of Engineering Medicine, Beihang University, Beijing, 100191, China.
- Key Laboratory of Biomechanics and Mechanobiology (Beihang University), Ministry of Education, Beijing, 100191, China.
| |
Collapse
|
3
|
Rollo J, Crawford J, Hardy J. A dynamical systems approach for multiscale synthesis of Alzheimer's pathogenesis. Neuron 2023; 111:2126-2139. [PMID: 37172582 DOI: 10.1016/j.neuron.2023.04.018] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2022] [Revised: 12/15/2022] [Accepted: 04/13/2023] [Indexed: 05/15/2023]
Abstract
Alzheimer's disease (AD) is a spatially dynamic pathology that implicates a growing volume of multiscale data spanning genetic, cellular, tissue, and organ levels of the organization. These data and bioinformatics analyses provide clear evidence for the interactions within and between these levels. The resulting heterarchy precludes a linear neuron-centric approach and necessitates that the numerous interactions are measured in a way that predicts their impact on the emergent dynamics of the disease. This level of complexity confounds intuition, and we propose a new methodology that uses non-linear dynamical systems modeling to augment intuition and that links with a community-wide participatory platform to co-create and test system-level hypotheses and interventions. In addition to enabling the integration of multiscale knowledge, key benefits include a more rapid innovation cycle and a rational process for prioritization of data campaigns. We argue that such an approach is essential to support the discovery of multilevel-coordinated polypharmaceutical interventions.
Collapse
Affiliation(s)
- Jennifer Rollo
- Department of Neurodegenerative Diseases, UCL Queen Square Institute of Neurology, Queen Square, London WC1N 3BG, UK.
| | - John Crawford
- Adam Smith Business School, University of Glasgow, Glasgow G12 8QQ, UK
| | - John Hardy
- Department of Neurodegenerative Diseases, UCL Queen Square Institute of Neurology, Queen Square, London WC1N 3BG, UK
| |
Collapse
|
4
|
Deaton CA, Johnson GVW. Presenilin 1 Regulates Membrane Homeostatic Pathways that are Dysregulated in Alzheimer's Disease. J Alzheimers Dis 2021; 77:961-977. [PMID: 32804090 DOI: 10.3233/jad-200598] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Mutations in the PSEN1 gene, encoding presenilin 1 (PS1), are the most common cause of familial Alzheimer's disease (fAD). Since the first mutations in the PSEN1 gene were discovered more than 25 years ago, many postulated functions of PS1 have been investigated. The majority of earlier studies focused on its role as the catalytic component of the γ-secretase complex, which in concert with β site amyloid precursor protein cleaving enzyme 1 (BACE1), mediates the formation of Aβ from amyloid-β protein precursor (AβPP). Though mutant PS1 was originally considered to cause AD by promoting Aβ pathology through its protease function, it is now becoming clear that PS1 is a multifunctional protein involved in regulating membrane dynamics and protein trafficking. Therefore, through loss of these abilities, mutant PS1 has the potential to impair numerous cellular functions such as calcium flux, organization of proteins in different compartments, and protein turnover via vacuolar metabolism. Impaired calcium signaling, vacuolar dysfunction, mitochondrial dysfunction, and increased ER stress, among other related membrane-dependent disturbances, have been considered critical to the development and progression of AD. Given that PS1 plays a key regulatory role in all these processes, this review will describe the role of PS1 in different cellular compartments and provide an integrated view of how PS1 dysregulation (due to mutations or other causes) could result in impairment of various cellular processes and result in a "multi-hit", integrated pathological outcome that could contribute to the etiology of AD.
Collapse
Affiliation(s)
- Carol A Deaton
- Cell Biology of Disease Program and the Department of Anesthesiology and Perioperative Medicine, University of Rochester Medical Center, Rochester, NY, USA
| | - Gail V W Johnson
- Cell Biology of Disease Program and the Department of Anesthesiology and Perioperative Medicine, University of Rochester Medical Center, Rochester, NY, USA
| |
Collapse
|
5
|
Mahan B, Antonelli MA, Burckel P, Turner S, Chung R, Habekost M, Jørgensen AL, Moynier F. Longitudinal biometal accumulation and Ca isotope composition of the Göttingen minipig brain. Metallomics 2020; 12:1585-1598. [PMID: 33084720 DOI: 10.1039/d0mt00134a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Biometals play a critical role in both the healthy and diseased brain's functioning. They accumulate in the normal aging brain, and are inherent to neurodegenerative disorders and their associated pathologies. A prominent example of this is the brain accumulation of metals such as Ca, Fe and Cu (and more ambiguously, Zn) associated with Alzheimer's disease (AD). The natural stable isotope compositions of such metals have also shown utility in constraining biological mechanisms, and in differentiating between healthy and diseased states, sometimes prior to conventional methods. Here we have detailed the distribution of the biologically relevant elements Mg, P, K, Ca, Fe, Cu and Zn in brain regions of Göttingen minipigs ranging in age from three months to nearly six years, including control animals and both a single- and double-transgenic model of AD (PS1, APP/PS1). Moreover, we have characterized the Ca isotope composition of the brain for the first time. Concentration data track rises in brain biometals with age, namely for Fe and Cu, as observed in the normal ageing brain and in AD, and biometal data point to increased soluble amyloid beta (Aβ) load prior to AD plaque identification via brain imaging. Calcium isotope results define the brain as the isotopically lightest permanent reservoir in the body, indicating that brain Ca dyshomeostasis may induce measurable isotopic disturbances in accessible downstream reservoirs such as biofluids.
Collapse
Affiliation(s)
- Brandon Mahan
- Earth and Environmental Science, James Cook University, Townsville, Queensland 4811, Australia. and Thermo Fisher Isotope Development Hub, Department of Earth and Planetary Sciences, Macquarie University, Sydney, New South Wales 2109, Australia
| | - Michael A Antonelli
- Université de Paris, Institut de Physique du Globe de Paris, CNRS, 75238 Paris, France and Institute of Geochemistry and Petrology, Department of Earth Sciences, ETH Zürich, 8092 Zürich, Switzerland
| | - Pierre Burckel
- Université de Paris, Institut de Physique du Globe de Paris, CNRS, 75238 Paris, France
| | - Simon Turner
- Thermo Fisher Isotope Development Hub, Department of Earth and Planetary Sciences, Macquarie University, Sydney, New South Wales 2109, Australia
| | - Roger Chung
- Thermo Fisher Isotope Development Hub, Department of Earth and Planetary Sciences, Macquarie University, Sydney, New South Wales 2109, Australia
| | - Mette Habekost
- Department of Biomedicine, Aarhus University, 8000 Aarhus C, Denmark
| | | | - Frédéric Moynier
- Université de Paris, Institut de Physique du Globe de Paris, CNRS, 75238 Paris, France
| |
Collapse
|
6
|
Neddens J, Daurer M, Flunkert S, Beutl K, Loeffler T, Walker L, Attems J, Hutter-Paier B. Correlation of pyroglutamate amyloid β and ptau Ser202/Thr205 levels in Alzheimer's disease and related murine models. PLoS One 2020; 15:e0235543. [PMID: 32645028 PMCID: PMC7347153 DOI: 10.1371/journal.pone.0235543] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2020] [Accepted: 06/17/2020] [Indexed: 01/14/2023] Open
Abstract
Senile plaques frequently contain Aβ-pE(3), a N-terminally truncated Aβ species that is more closely linked to AD compared to other Aβ species. Tau protein is highly phosphorylated at several residues in AD, and specifically phosphorylation at Ser202/Thr205 is known to be increased in AD. Several studies suggest that formation of plaques and tau phosphorylation might be linked to each other. To evaluate if Aβ-pE(3) and ptau Ser202/Thr205 levels correlate in human and transgenic AD mouse models, we analyzed human cortical and hippocampal brain tissue of different Braak stages as well as murine brain tissue of two transgenic mouse models for levels of Aβ-pE(3) and ptau Ser202/Thr205 and correlated the data. Our results show that Aβ-pE(3) formation is increased at early Braak stages while ptau Ser202/Thr205 mostly increases at later stages. Further analyses revealed strongest correlations between the two pathologies in the temporal, frontal, cingulate, and occipital cortex, however correlation in the hippocampus was weaker. Evaluation of murine transgenic brain tissue demonstrated a slow but steady increase of Aβ-pE(3) from 6 to 12 months of age in the cortex and hippocampus of APPSL mice, and a very early and strong Aβ-pE(3) increase in 5xFAD mice. ptau Ser202/Thr205 levels increased at the age of 9 months in APPSL mice and at 6 months in 5xFAD mice. Our results show that Aβ-pE(3) and ptau Ser202/Thr205 levels strongly correlate in human as well as murine tissues, suggesting that tau phosphorylation might be amplified by Aβ-pE(3).
Collapse
Affiliation(s)
| | | | | | - Kerstin Beutl
- QPS Austria GmbH, Grambach, Austria
- FH Joanneum Graz, Graz, Austria
| | | | - Lauren Walker
- Translational and Clinical Research Institute and Newcastle University Institute for Ageing, Campus for Ageing and Vitality, Newcastle upon Tyne, United Kingdom
| | - Johannes Attems
- Translational and Clinical Research Institute and Newcastle University Institute for Ageing, Campus for Ageing and Vitality, Newcastle upon Tyne, United Kingdom
| | | |
Collapse
|
7
|
Wu HY, Kuo PC, Wang YT, Lin HT, Roe AD, Wang BY, Han CL, Hyman BT, Chen YJ, Tai HC. β-Amyloid Induces Pathology-Related Patterns of Tau Hyperphosphorylation at Synaptic Terminals. J Neuropathol Exp Neurol 2019; 77:814-826. [PMID: 30016458 DOI: 10.1093/jnen/nly059] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
A synergy between β-amyloid (Aβ) and tau appears to occur in Alzheimer disease (AD), but the mechanisms of interaction, and potential locations, are little understood. This study investigates the possibility of such interactions within the cortical synaptic compartments of APP/PS1 mice. We used label-free quantitative mass spectrometry to study the phosphoproteome of synaptosomes, covering 2400 phosphopeptides and providing an unbiased survey of phosphorylation changes associated with amyloid pathology. Hyperphosphorylation was detected on 36 synaptic proteins, many of which are associated with the cytoskeleton. Importantly, tau is one of the most hyperphosphorylated proteins at the synapse, upregulated at both proline-directed kinase (PDK) sites (S199/S202, S396/S404) and nonPDK sites (S400). These PDK sites correspond to well-known pathological tau epitopes in AD patients, recognized by AT8 and PHF-1 antibodies, respectively. Hyperphosphorylation at S199/S202, a rarely examined combination, was further validated in patient-derived human synaptosomes by immunoblotting. Global surveys of upregulated phosphosites revealed 2 potential kinase motifs, which resemble those of cyclin-dependent kinase 5 (CDK5, a PDK) and casein kinase II (CK2, a nonPDK). Our data demonstrate that, within synaptic compartments, amyloid pathology is associated with tau hyperphosphorylation at disease-relevant epitopes. This provides a plausible mechanism by which Aβ promotes the spreading of tauopathy.
Collapse
Affiliation(s)
- Hsin-Yi Wu
- Instrumentation Center, National Taiwan University, Taipei, Taiwan
| | - Po-Cheng Kuo
- Department of Chemistry, National Taiwan University, Taipei, Taiwan
| | - Yi-Ting Wang
- Institute of Chemistry, Academia Sinica, Taipei, Taiwan
| | - Hao-Tai Lin
- Department of Chemistry, National Taiwan University, Taipei, Taiwan
| | - Allyson D Roe
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Bo Y Wang
- Department of Chemistry, National Taiwan University, Taipei, Taiwan
| | - Chia-Li Han
- Master Program for Clinical Pharmacogenomics and Pharmacoproteomics, College of Pharmacy, Taipei Medical University, Taipei, Taiwan
| | - Bradley T Hyman
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Yu-Ju Chen
- Department of Chemistry, National Taiwan University, Taipei, Taiwan.,Institute of Chemistry, Academia Sinica, Taipei, Taiwan
| | - Hwan-Ching Tai
- Department of Chemistry, National Taiwan University, Taipei, Taiwan
| |
Collapse
|
8
|
Koh JY, Kim HN, Hwang JJ, Kim YH, Park SE. Lysosomal dysfunction in proteinopathic neurodegenerative disorders: possible therapeutic roles of cAMP and zinc. Mol Brain 2019; 12:18. [PMID: 30866990 PMCID: PMC6417073 DOI: 10.1186/s13041-019-0439-2] [Citation(s) in RCA: 78] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2019] [Accepted: 03/04/2019] [Indexed: 12/22/2022] Open
Abstract
A number of neurodegenerative diseases, including Alzheimer's disease, Parkinson's disease, and amyotrophic lateral sclerosis, share intra- and/or extracellular deposition of protein aggregates as a common core pathology. While the species of accumulating proteins are distinct in each disease, an increasing body of evidence indicates that defects in the protein clearance system play a crucial role in the gradual accumulation of protein aggregates. Among protein degradation systems, the endosome-autophagosome-lysosome pathway (EALP) is the main degradation machinery, especially for large protein aggregates. Lysosomal dysfunction or defects in fusion with vesicles containing cargo are commonly observed abnormalities in proteinopathic neurodegenerative diseases. In this review, we discuss the available evidence for a mechanistic connection between components of the EALP-especially lysosomes-and neurodegenerative diseases. We also focus on lysosomal pH regulation and its significance in maintaining flux through the EALP. Finally, we suggest that raising cAMP and free zinc levels in brain cells may be beneficial in normalizing lysosomal pH and EALP flux.
Collapse
Affiliation(s)
- Jae-Young Koh
- Department of Neurology, University of Ulsan College of Medicine, Seoul, South Korea.
| | - Ha Na Kim
- Neural Injury Lab, Biomedical Research Center, Asan Institute for Life Sciences, Asan Medical Center, Seoul, South Korea
| | - Jung Jin Hwang
- Department of Convergence Medicine, University of Ulsan College of Medicine, Asan Medical Center, Seoul, South Korea
| | - Yang-Hee Kim
- Department of Integrative Bioscience and Biotechnology, Sejong University, Seoul, South Korea
| | - Sang Eun Park
- Asan Institute for Life Sciences, Asan Medical Center, Seoul, South Korea
| |
Collapse
|
9
|
Angulo SL, Orman R, Neymotin SA, Liu L, Buitrago L, Cepeda-Prado E, Stefanov D, Lytton WW, Stewart M, Small SA, Duff KE, Moreno H. Tau and amyloid-related pathologies in the entorhinal cortex have divergent effects in the hippocampal circuit. Neurobiol Dis 2017; 108:261-276. [PMID: 28860088 DOI: 10.1016/j.nbd.2017.08.015] [Citation(s) in RCA: 49] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2017] [Revised: 08/09/2017] [Accepted: 08/26/2017] [Indexed: 02/02/2023] Open
Abstract
The entorhinal cortex (EC) is affected early in Alzheimer's disease, an illness defined by a co-occurrence of tau and amyloid-related pathologies. How the co-occurrence of these pathologies in the EC affects the hippocampal circuit remains unknown. Here we address this question by performing electrophysiological analyses of the EC circuit in mice that express mutant human amyloid precursor protein (hAPP) or tau (hTau), or both in the EC. We show that the alterations in the hippocampal circuit are divergent, with hAPP increasing but hTau decreasing neuronal/circuit excitability. Most importantly, mice co-expressing hAPP and hTau show that hTau has a dominant effect, dampening the excitatory effects of hAPP. Additionally, compensatory synaptic downscaling, in response to increased excitability in EC was observed in subicular neurons of hAPP mice. Based on simulations, we propose that EC interneuron pruning can account for both EC hyperexcitability and subicular synaptic downscaling found in mice expressing hAPP.
Collapse
Affiliation(s)
- S L Angulo
- Department of Neurology, SUNY Downstate Medical Center, Brooklyn, NY 11203, United States; The Robert F. Furchgott Center for Neural and Behavioral Science, Department of Physiology/Pharmacology, SUNY Downstate Medical Center, Brooklyn, NY 11203, United States
| | - R Orman
- The Robert F. Furchgott Center for Neural and Behavioral Science, Department of Physiology/Pharmacology, SUNY Downstate Medical Center, Brooklyn, NY 11203, United States
| | - S A Neymotin
- Department of Neuroscience, Brown University, Providence, RI 02912, United States
| | - L Liu
- Department of Pathology, Cell Biology, Columbia University Medical Center, NY 10032, United States; Department of Psychiatry, Columbia University Medical Center, NY 10032, United States
| | - L Buitrago
- Department of Neurology, SUNY Downstate Medical Center, Brooklyn, NY 11203, United States; The Robert F. Furchgott Center for Neural and Behavioral Science, Department of Physiology/Pharmacology, SUNY Downstate Medical Center, Brooklyn, NY 11203, United States
| | - E Cepeda-Prado
- Department of Neurology, SUNY Downstate Medical Center, Brooklyn, NY 11203, United States; The Robert F. Furchgott Center for Neural and Behavioral Science, Department of Physiology/Pharmacology, SUNY Downstate Medical Center, Brooklyn, NY 11203, United States
| | - D Stefanov
- Scientific Computer Center, SUNY Downstate Medical Center, Brooklyn, NY 11203, United States
| | - W W Lytton
- Department of Neurology, SUNY Downstate Medical Center, Brooklyn, NY 11203, United States; The Robert F. Furchgott Center for Neural and Behavioral Science, Department of Physiology/Pharmacology, SUNY Downstate Medical Center, Brooklyn, NY 11203, United States
| | - M Stewart
- The Robert F. Furchgott Center for Neural and Behavioral Science, Department of Physiology/Pharmacology, SUNY Downstate Medical Center, Brooklyn, NY 11203, United States
| | - S A Small
- Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University, NY 10032, United States; Department of Neurology, Columbia University Medical Center, NY 10032, United States
| | - K E Duff
- Department of Pathology, Cell Biology, Columbia University Medical Center, NY 10032, United States; Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University, NY 10032, United States; Department of Psychiatry, Columbia University Medical Center, NY 10032, United States
| | - H Moreno
- Department of Neurology, SUNY Downstate Medical Center, Brooklyn, NY 11203, United States; The Robert F. Furchgott Center for Neural and Behavioral Science, Department of Physiology/Pharmacology, SUNY Downstate Medical Center, Brooklyn, NY 11203, United States.
| |
Collapse
|
10
|
Abstract
Alzheimer's disease (AD) is a fatal neurodegenerative disorder that has no known cure, nor is there a clear mechanistic understanding of the disease process itself. Although amyloid plaques, neurofibrillary tangles, and cognitive decline are late-stage markers of the disease, it is unclear how they are initially generated, and if they represent a cause, effect, or end phase in the pathology process. Recent studies in AD models have identified marked dysregulations in calcium signaling and related downstream pathways, which occur long before the diagnostic histopathological or cognitive changes. Under normal conditions, intracellular calcium signals are coupled to effectors that maintain a healthy physiological state. Consequently, sustained up-regulation of calcium may have pathophysiological consequences. Indeed, upon reviewing the current body of literature, increased calcium levels are functionally linked to the major features and risk factors of AD: ApoE4 expression, presenilin and APP mutations, beta amyloid plaques, hyperphosphorylation of tau, apoptosis, and synaptic dysfunction. In turn, the histopathological features of AD, once formed, are capable of further increasing calcium levels, leading to a rapid feed-forward acceleration once the disease process has taken hold. The views proposed here consider that AD pathogenesis reflects long-term calcium dysregulations that ultimately serve an enabling role in the disease process. Therefore, “Calcinists” do not necessarily reject βAptist or Tauist doctrine, but rather believe that their genesis is associated with earlier calcium signaling dysregulations. NEUROSCIENTIST 13(5):546—559, 2007.
Collapse
Affiliation(s)
- Grace E Stutzmann
- Rosalind Franklin University of Medicine and Science, The Chicago Medical School, North Chicago, IL 60064, USA.
| |
Collapse
|
11
|
Javaid FZ, Brenton J, Guo L, Cordeiro MF. Visual and Ocular Manifestations of Alzheimer's Disease and Their Use as Biomarkers for Diagnosis and Progression. Front Neurol 2016; 7:55. [PMID: 27148157 PMCID: PMC4836138 DOI: 10.3389/fneur.2016.00055] [Citation(s) in RCA: 126] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2016] [Accepted: 03/29/2016] [Indexed: 12/21/2022] Open
Abstract
Alzheimer's disease (AD) is the most common form of dementia affecting the growing aging population today, with prevalence expected to rise over the next 35 years. Clinically, patients exhibit a progressive decline in cognition, memory, and social functioning due to deposition of amyloid β (Aβ) protein and intracellular hyperphosphorylated tau protein. These pathological hallmarks of AD are measured either through neuroimaging, cerebrospinal fluid analysis, or diagnosed post-mortem. Importantly, neuropathological progression occurs in the eye as well as the brain, and multiple visual changes have been noted in both human and animal models of AD. The eye offers itself as a transparent medium to cerebral pathology and has thus potentiated the development of ocular biomarkers for AD. The use of non-invasive screening, such as retinal imaging and visual testing, may enable earlier diagnosis in the clinical setting, minimizing invasive and expensive investigations. It also potentially improves disease management and quality of life for AD patients, as an earlier diagnosis allows initiation of medication and treatment. In this review, we explore the evidence surrounding ocular changes in AD and consider the biomarkers currently in development for early diagnosis.
Collapse
Affiliation(s)
- Fatimah Zara Javaid
- Glaucoma and Retinal Degeneration Research Group, Visual Neurosciences, UCL Institute of Ophthalmology, London, UK
| | - Jonathan Brenton
- Glaucoma and Retinal Degeneration Research Group, Visual Neurosciences, UCL Institute of Ophthalmology, London, UK
| | - Li Guo
- Glaucoma and Retinal Degeneration Research Group, Visual Neurosciences, UCL Institute of Ophthalmology, London, UK
| | - Maria F. Cordeiro
- Glaucoma and Retinal Degeneration Research Group, Visual Neurosciences, UCL Institute of Ophthalmology, London, UK
- Western Eye Hospital, Imperial College Healthcare NHS Trust, London, UK
| |
Collapse
|
12
|
Arun P, Oguntayo S, Albert SV, Gist I, Wang Y, Nambiar MP, Long JB. Acute decrease in alkaline phosphatase after brain injury: A potential mechanism for tauopathy. Neurosci Lett 2015; 609:152-8. [DOI: 10.1016/j.neulet.2015.10.036] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2015] [Revised: 10/01/2015] [Accepted: 10/12/2015] [Indexed: 12/29/2022]
|
13
|
Kroth H, Ansaloni A, Varisco Y, Jan A, Sreenivasachary N, Rezaei-Ghaleh N, Giriens V, Lohmann S, López-Deber MP, Adolfsson O, Pihlgren M, Paganetti P, Froestl W, Nagel-Steger L, Willbold D, Schrader T, Zweckstetter M, Pfeifer A, Lashuel HA, Muhs A. Discovery and structure activity relationship of small molecule inhibitors of toxic β-amyloid-42 fibril formation. J Biol Chem 2012; 287:34786-800. [PMID: 22891248 DOI: 10.1074/jbc.m112.357665] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Increasing evidence implicates Aβ peptides self-assembly and fibril formation as crucial events in the pathogenesis of Alzheimer disease. Thus, inhibiting Aβ aggregation, among others, has emerged as a potential therapeutic intervention for this disorder. Herein, we employed 3-aminopyrazole as a key fragment in our design of non-dye compounds capable of interacting with Aβ42 via a donor-acceptor-donor hydrogen bond pattern complementary to that of the β-sheet conformation of Aβ42. The initial design of the compounds was based on connecting two 3-aminopyrazole moieties via a linker to identify suitable scaffold molecules. Additional aryl substitutions on the two 3-aminopyrazole moieties were also explored to enhance π-π stacking/hydrophobic interactions with amino acids of Aβ42. The efficacy of these compounds on inhibiting Aβ fibril formation and toxicity in vitro was assessed using a combination of biophysical techniques and viability assays. Using structure activity relationship data from the in vitro assays, we identified compounds capable of preventing pathological self-assembly of Aβ42 leading to decreased cell toxicity.
Collapse
Affiliation(s)
- Heiko Kroth
- AC Immune SA, PSE Building B, Swiss Federal Institute of Technology Lausanne (EPFL), CH-1015 Lausanne, Switzerland
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
14
|
Bugiani O. Alzheimer’s disease: ageing-related or age-related? New hypotheses from an old debate. Neurol Sci 2011; 32:1241-7. [DOI: 10.1007/s10072-011-0614-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2011] [Accepted: 04/28/2011] [Indexed: 11/28/2022]
|
15
|
The γ-Secretase Modulator CHF5074 Reduces the Accumulation of Native Hyperphosphorylated Tau in a Transgenic Mouse Model of Alzheimer’s Disease. J Mol Neurosci 2010; 45:22-31. [DOI: 10.1007/s12031-010-9482-2] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2010] [Accepted: 12/02/2010] [Indexed: 01/29/2023]
|
16
|
Lin J, Li X, Yuan F, Lin L, Cook CL, Rao CV, Lei Z. Genetic ablation of luteinizing hormone receptor improves the amyloid pathology in a mouse model of Alzheimer disease. J Neuropathol Exp Neurol 2010; 69:253-61. [PMID: 20142765 PMCID: PMC2869088 DOI: 10.1097/nen.0b013e3181d072cf] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
Amyloid-beta peptide (Abeta) plays an essential pathophysiologic role in Alzheimer disease, and elevation of luteinizing hormone (LH) levels during aging has been implicated in its pathogenesis. To assess the effect of LH receptor deficiency on Abeta accumulation, we generated a bigenic mouse model, APPsw(+)/Lhr(-/-), which expresses human amyloid precursor protein (APPsw) in the background of LH receptor (Lhr) knockout. Genetic ablation of Lhr resulted in a significant decrease in the number of Abeta plaques and protein content in the hippocampus and cerebral cortex in both male and female mice. Accordingly, several Abeta deposition-related neuropathologic features and functionally relevant molecules were markedly improved, including decreased astrogliosis, reductions of elevated phosphorylated tau, c-fos, alpha7-nicotinic acetylcholine receptor, and restoration of the altered neuropeptide Y receptors Y1 and Y2. Diminution of Abeta accumulation in the absence of LH receptor supports the contention that dysregulation of LH may impact the pathogenesis of Alzheimer disease. The APPsw(+)/Lhr(-/-) mouse may be a useful tool for advancing understanding of the role of LH-mediated events in Alzheimer disease and a model in which to test therapeutic interventions.
Collapse
MESH Headings
- Alzheimer Disease/genetics
- Alzheimer Disease/metabolism
- Alzheimer Disease/physiopathology
- Amyloid beta-Peptides/metabolism
- Amyloid beta-Protein Precursor/genetics
- Amyloid beta-Protein Precursor/metabolism
- Animals
- Brain/metabolism
- Brain/pathology
- Brain/physiopathology
- Cerebral Cortex/metabolism
- Cerebral Cortex/pathology
- Cerebral Cortex/physiopathology
- Disease Models, Animal
- Female
- Gliosis/genetics
- Gliosis/metabolism
- Gliosis/physiopathology
- Hippocampus/metabolism
- Hippocampus/pathology
- Hippocampus/physiopathology
- Humans
- Luteinizing Hormone/metabolism
- Male
- Mice
- Mice, Knockout
- Mice, Transgenic
- Plaque, Amyloid/genetics
- Plaque, Amyloid/metabolism
- Plaque, Amyloid/pathology
- Proto-Oncogene Proteins c-fos/metabolism
- Receptors, LH/genetics
- Receptors, Neuropeptide Y/metabolism
- Receptors, Nicotinic/genetics
- Receptors, Nicotinic/metabolism
- alpha7 Nicotinic Acetylcholine Receptor
- tau Proteins/metabolism
Collapse
Affiliation(s)
- Jing Lin
- Department of Obstetrics, Gynecology and Women’s Health, University of Louisville School of Medicine, Louisville, Kentucky
| | - Xian Li
- Department of Obstetrics, Gynecology and Women’s Health, University of Louisville School of Medicine, Louisville, Kentucky
| | - Fangping Yuan
- Department of Obstetrics, Gynecology and Women’s Health, University of Louisville School of Medicine, Louisville, Kentucky
| | - Ling Lin
- Department of Obstetrics, Gynecology and Women’s Health, University of Louisville School of Medicine, Louisville, Kentucky
| | - Christine L. Cook
- Department of Obstetrics, Gynecology and Women’s Health, University of Louisville School of Medicine, Louisville, Kentucky
| | - Ch V. Rao
- Department of Obstetrics, Gynecology and Women’s Health, University of Louisville School of Medicine, Louisville, Kentucky
| | - Zhenmin Lei
- Department of Obstetrics, Gynecology and Women’s Health, University of Louisville School of Medicine, Louisville, Kentucky
| |
Collapse
|
17
|
Wilcock DM, Colton CA. Immunotherapy, vascular pathology, and microhemorrhages in transgenic mice. CNS & NEUROLOGICAL DISORDERS DRUG TARGETS 2009; 8:50-64. [PMID: 19275636 PMCID: PMC2659468 DOI: 10.2174/187152709787601858] [Citation(s) in RCA: 72] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Alzheimer's disease (AD) is a progressive, neurodegenerative disorder that results in severe cognitive decline. Amyloid plaques are a principal pathology found in AD and are composed of aggregated amyloid-beta (Abeta) peptides. According to the amyloid hypothesis, Abeta peptides initiate the other pathologies characteristic for AD including cognitive deficits. Immunotherapy against Abeta is a potential therapeutic for the treatment of humans with AD. While anti-Abeta immunotherapy has been shown to reduce amyloid burden in both mouse models and in humans, immunotherapy also exacerbates vascular pathologies. Cerebral amyloid angiopathy (CAA), that is, the accumulation of amyloid in the cerebrovasculature, is increased with immunotherapy in humans with AD and in mouse models of amyloid deposition. CAA persists in the brains of clinical trial patients that show removal of parenchymal amyloid. Mouse model studies also show that immunotherapy results in multiple small bleeds in the brain, termed microhemorrhages. The neurovascular unit is a term used to describe the cerebrovasculature and its associated cells-astrocytes, neurons, pericytes and microglia. CAA affects brain perfusion and there is now evidence that the neurovascular unit is affected in AD when CAA is present. Understanding the type of damage to the neurovascular unit caused by CAA in AD and the underlying cause of microhemorrhage after immunotherapy is essential to the success of therapeutic vaccines as a treatment for AD.
Collapse
Affiliation(s)
- Donna M Wilcock
- Duke University Medical Center, Division of Neurology, Research Dr, Durham, NC 27710, USA.
| | | |
Collapse
|
18
|
Nishitsuji K, Tomiyama T, Ishibashi K, Kametani F, Ozawa K, Okada R, Maat-Schieman ML, Roos RAC, Iwai K, Mori H. Cerebral vascular accumulation of Dutch-type Abeta42, but not wild-type Abeta42, in hereditary cerebral hemorrhage with amyloidosis, Dutch type. J Neurosci Res 2008; 85:2917-23. [PMID: 17628026 DOI: 10.1002/jnr.21413] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Hereditary cerebral hemorrhage with amyloidosis, Dutch type (HCHWA-D), is an autosomal dominant disorder caused by the Dutch mutation (E693Q) in the beta-amyloid precursor protein. This mutation produces an aberrant amyloid beta (Abeta) species (AbetaE22Q) and causes severe meningocortical vascular Abeta deposition. We analyzed the Abeta composition of the vascular amyloid in the brains of HCHWA-D patients. Immunohistochemistry demonstrated that the vascular amyloid contained both Abeta40 and Abeta42, with a high Abeta40/Abeta42 ratio. In Western blotting of cerebral microvessel fractions isolated from the brains, both wild-type and Dutch-type Abeta40 were observed as major species. Reverse-phase HPLC-mass spectrometric analysis of the fractions revealed both wild-type and Dutch-type Abeta38 as the other main components of the vascular amyloid. Moreover, we detected peaks corresponding to Dutch-type Abeta42 but not to wild-type Abeta42. These results suggest a pathogenic role for the mutant Abeta42 in addition to the mutant Abeta40 in the cerebral amyloid angiopathy of HCHWA-D.
Collapse
Affiliation(s)
- Kazuchika Nishitsuji
- Department of Neuroscience, Osaka City University Graduate School of Medicine, Osaka, Japan
| | | | | | | | | | | | | | | | | | | |
Collapse
|
19
|
Bellucci A, Rosi MC, Grossi C, Fiorentini A, Luccarini I, Casamenti F. Abnormal processing of tau in the brain of aged TgCRND8 mice. Neurobiol Dis 2007; 27:328-38. [PMID: 17656099 DOI: 10.1016/j.nbd.2007.06.008] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2007] [Revised: 05/02/2007] [Accepted: 06/04/2007] [Indexed: 12/21/2022] Open
Abstract
Amyloid plaques and neurofibrillary tangles are the main histopathological hallmarks of Alzheimer's disease (AD). In the neocortex and hippocampus of aged TgCRND8 mice, tau is hyperphosphorylated at different sites recognized by PHF-1, AT100, AT8 and CP13 antibodies. Phospho-SAPK/JNK levels were increased in the tg mouse brain, where activated SAPK/JNK co-localizes with PHF-1-positive cells. Phosphorylated tau-positive cells showed Bielschowsky- and Thioflavine S-positive intraneuronal deposits. PHF-1 and nitrotyrosine immunoreactivity merged within neurons surrounding amyloid deposits in cortical and hippocampal areas and immunoprecipitation studies confirmed that tau is nitrosylated. Our findings, demonstrating the presence of hyperphosphorylated and nitrosylated tau protein as well as of insoluble aggregates after the onset of amyloid deposition in the TgCRND8 mouse brain, indicate that the abnormal processing of tau may occur subsequently to cerebral amyloidosis and that activation of SAPK/JNK and induction of nitrosative stress are the more likely connecting factors between amyloidosis and tauopathy in AD.
Collapse
Affiliation(s)
- Arianna Bellucci
- Biomedical Sciences and Biotechnology, Division of Pharmacology, University of Brescia, Viale Europa, 11, 25123, Italy
| | | | | | | | | | | |
Collapse
|
20
|
Kim J, Onstead L, Randle S, Price R, Smithson L, Zwizinski C, Dickson DW, Golde T, McGowan E. Abeta40 inhibits amyloid deposition in vivo. J Neurosci 2007; 27:627-33. [PMID: 17234594 PMCID: PMC6672801 DOI: 10.1523/jneurosci.4849-06.2007] [Citation(s) in RCA: 291] [Impact Index Per Article: 16.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Numerous studies have established a pivotal role for Abeta42 in Alzheimer's disease (AD) pathogenesis. In contrast, although Abeta40 is the predominant form of amyloid beta (Abeta) produced and accumulates to a variable degree in the human AD brain, its role in AD pathogenesis has not been established. It has generally been assumed that an increase in Abeta40 would accelerate amyloid plaque formation in vivo. We have crossed BRI-Abeta40 mice that selectively express high levels of Abeta40 with both Tg2576 (APPswe, K670N+M671L) mice and BRI-Abeta42A mice expressing Abeta42 selectively and analyzed parenchymal and cerebrovascular Abeta deposition in the bitransgenic mice compared with their singly transgenic littermates. In the bitransgenic mice, the increased steady-state levels of Abeta40 decreased Abeta deposition by 60-90%. These results demonstrate that Abeta42 and Abeta40 have opposing effects on amyloid deposition: Abeta42 promotes amyloid deposition but Abeta40 inhibits it. In addition, increasing Abeta40 levels protected BRI-Abeta40/Tg2576 mice from the premature-death phenotype observed in Tg2576 mice. The protective properties of Abeta40 with respect to amyloid deposition suggest that strategies that preferentially target Abeta40 may actually worsen the disease course and that selective increases in Abeta40 levels may actually reduce the risk for development of AD.
Collapse
Affiliation(s)
- Jungsu Kim
- Department of Neuroscience, Mayo Clinic College of Medicine, Jacksonville, Florida 32224
| | - Luisa Onstead
- Department of Neuroscience, Mayo Clinic College of Medicine, Jacksonville, Florida 32224
| | - Suzanne Randle
- Department of Neuroscience, Mayo Clinic College of Medicine, Jacksonville, Florida 32224
| | - Robert Price
- Department of Neuroscience, Mayo Clinic College of Medicine, Jacksonville, Florida 32224
| | - Lisa Smithson
- Department of Neuroscience, Mayo Clinic College of Medicine, Jacksonville, Florida 32224
| | - Craig Zwizinski
- Department of Neuroscience, Mayo Clinic College of Medicine, Jacksonville, Florida 32224
| | - Dennis W. Dickson
- Department of Neuroscience, Mayo Clinic College of Medicine, Jacksonville, Florida 32224
| | - Todd Golde
- Department of Neuroscience, Mayo Clinic College of Medicine, Jacksonville, Florida 32224
| | - Eileen McGowan
- Department of Neuroscience, Mayo Clinic College of Medicine, Jacksonville, Florida 32224
| |
Collapse
|
21
|
Abstract
AbstractWe appear to be on the brink of a new epoch of treatment for Alzheimer's disease. Compelling evidence suggests that Aβ42 secretion is the triggering event in the pathogenesis of Alzheimer's disease, and that tau aggregation may be an important secondary event linked to neurodegeneration. Prophylactic administration of anti-amyloid agents designed to prevent Aβ accumulation in persons with subclinical disease is likely to be more effective than therapeutic interventions in established Alzheimer's disease. Drug development programs in Alzheimer's disease focus primarily on agents with anti-amyloid disease-modifying properties, and many different pharmacologic approaches to reducing amyloid pathology and tauopathy are being studied. Classes of therapeutic modalities currently in advanced-stage clinical trial testing include forms of immunotherapy (active β-amyloid immunoconjugate and human intravenous immunoglobulin), a γ-secretase inhibitor, the selective Aβ42-lowering agent R-flurbiprofen, and the anti-aggregation agent tramiprosate. Non-traditional dementia therapies such as the HMG-CoA reductase inhibitors (statins), valproate, and lithium are now being assessed for clinical benefit as anti-amyloid disease-modifying treatments. Positive findings of efficacy and safety from clinical studies are necessary but not sufficient to demonstrate that a drug has disease-modifying properties. Definitive proof of disease-modification requires evidence from validated animal models of Alzheimer's disease; rigorously controlled clinical trials showing a significantly improved, stabilized, or slowed rate of decline in cognitive and global function compared to placebo; and prospectively obtained evidence from surrogate biomarkers that the treatment resulted in measurable biological changes associated with the underlying disease process.
Collapse
|
22
|
Kurata T, Kawarabayashi T, Murakami T, Miyazaki K, Morimoto N, Ohta Y, Takehisa Y, Nagai M, Ikeda M, Matsubara E, Westaway D, Hyslop PSG, Harigaya Y, Kamiya T, Shoji M, Abe K. Enhanced accumulation of phosphorylated α-synuclein in double transgenic mice expressing mutant β-amyloid precursor protein and presenilin-1. J Neurosci Res 2007; 85:2246-52. [PMID: 17526016 DOI: 10.1002/jnr.21352] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
A recent report showed that the accumulation of alpha-synuclein (alpha-syn) was detected in the brains of one-third of Alzheimer's disease and Down syndrome patients. However, the relationship between amyloid-beta protein (Abeta) and alpha-syn remains unclear. We analyzed the relation between the mutation of presenilin-1 (PS-1) and the pathological features of beta-amyloidosis and alpha-synucleinopathy. We generated doubly transgenic mice overexpressing mutant beta-amyloid precursor protein (betaAPP; Tg2576) and mutant PS-1 (PS1L286Vtg; line 198) and analyzed 19 double Tg betaAPP(+)/PS(+) mice at 5-23 months (young to old), 23 age-matched single Tg betaAPP(+)/PS(-) mice, and 11 non-Tg littermates. Immunohistochemical comparison was performed in these three groups by counting the area and the number of alpha-syn- or phosphorylated alpha-syn (palpha-syn)-positive dystrophic neurites per plaque (ASPDN, pASPDN). The acceleration of Abeta pathology was found with earlier onset and exaggerated numbers in double Tg betaAPP(+)/PS(+) compared with single Tg betaAPP(+)/PS(-) mouse brains. The accumulation of ASPDN and pASPDN was also accelerated in double Tg betaAPP(+)/PS(+) compared with single Tg betaAPP(+)/PS(-) mouse brains, especially in pASPDN. The number and area of alpha-syn and palpha-syn, and the ratio of palpha-syn positive neurites were significantly higher in double Tg betaAPP(+)/PS(+) than in single Tg betaAPP(+)/PS(-) mouse brains in middle-aged and old groups. Additional overexpression of mutant PS-1 accelerated Abeta-induced alpha-synucleinopathy and further facilitated the phosphorylation of alpha-syn, suggesting a direct association between mutant PS-1 and phosphorylation of alpha-syn.
Collapse
Affiliation(s)
- Tomoko Kurata
- Department of Neurology, Okayama University Graduate School of Medicine and Dentistry, Okayama, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|