1
|
Takeuchi LE, Kalia LV, Simmons CA. Vascular models of Alzheimer's disease: An overview of recent in vitro models of the blood-brain barrier. Neurobiol Dis 2025; 208:106864. [PMID: 40089165 DOI: 10.1016/j.nbd.2025.106864] [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: 11/03/2024] [Revised: 02/18/2025] [Accepted: 03/06/2025] [Indexed: 03/17/2025] Open
Abstract
Alzheimer's disease (AD) remains an overwhelming epidemiologic and economic burden on our healthcare systems, affecting an estimate of 11 % of individuals aged 65 years and older. Increasing evidence of the role of the blood-brain barrier (BBB) in AD pathology lends support to the vascular hypothesis of AD, which posits that damage to cerebral vasculature and impairments to cerebral blood flow are major contributors to neurodegeneration in AD. While the question remains whether the dysfunction of the BBB is the cause or consequence of the disease, understanding of the relationship between vascular pathology and AD is growing increasingly complex, warranting the need for better tools to study vasculature in AD. This review provides an overview of AD models in the context of studying vascular impairments and their relevance in pathology. Specifically, we summarize opportunities in in vitro models, cell sources, and phenotypic observations in sporadic and familial forms of AD. Further, we describe recent advances in generating models which recapitulate in vivo characteristics of the BBB in AD through the use of microfluidics, induced pluripotent stem cells (iPSC), and organoid technologies. Finally, we provide a searchable database of reported cell-based models of pathogenic AD gene variants.
Collapse
Affiliation(s)
- Lily E Takeuchi
- Institute of Biomedical Engineering, University of Toronto, Toronto, ON M5G 3G9, Canada; Translational Biology and Engineering Program, Ted Rogers Centre for Heart Research, Toronto, ON M5G 1M1, Canada.
| | - Lorraine V Kalia
- Division of Neurology, Department of Medicine, Toronto Western Hospital, University Health Network, University of Toronto, Toronto, ON M5T 2S8, Canada; Krembil Research Institute, University Health Network, Toronto, ON M5T 0S8, Canada; Tanz Centre for Research in Neurodegenerative Diseases, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Craig A Simmons
- Institute of Biomedical Engineering, University of Toronto, Toronto, ON M5G 3G9, Canada; Translational Biology and Engineering Program, Ted Rogers Centre for Heart Research, Toronto, ON M5G 1M1, Canada; Department of Mechanical and Industrial Engineering, University of Toronto, Toronto, ON M5S 3G8, Canada.
| |
Collapse
|
2
|
Kaur V, Sunkaria A. Unlocking the therapeutic promise of miRNAs in promoting amyloid-β clearance for Alzheimer's disease. Behav Brain Res 2025; 484:115505. [PMID: 40010509 DOI: 10.1016/j.bbr.2025.115505] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2024] [Revised: 01/06/2025] [Accepted: 02/21/2025] [Indexed: 02/28/2025]
Abstract
Alzheimer's disease (AD) is a neurological disorder that affects cognition and behavior, accounting for 60-70 % of dementia cases. Its mechanisms involve amyloid aggregates, hyperphosphorylated tau tangles, and loss of neural connections. Current treatments have limited efficacy due to a lack of specific targets. Recently, microRNAs (miRNAs) have emerged as key modulators in AD, regulating gene expression through interactions with mRNA. Dysregulation of specific miRNAs contributes to disease progression by disrupting clearance pathways. Antisense oligonucleotide (ASO)-based therapies show promise for AD treatment, particularly when combined with miRNA mimics or antagonists, targeting complex regulatory networks. However, miRNAs can interact with each other, complicating cellular processes and potentially leading to side effects. Our review emphasizes the role of miRNAs in regulating amyloid-beta (Aβ) clearance and highlights their potential as therapeutic targets and early biomarkers for AD, underscoring the need for further research to enhance their efficacy and safety.
Collapse
Affiliation(s)
- Vajinder Kaur
- Department of Biotechnology, Guru Nanak Dev University, Amritsar, Punjab 143005, India
| | - Aditya Sunkaria
- Department of Biotechnology, Guru Nanak Dev University, Amritsar, Punjab 143005, India.
| |
Collapse
|
3
|
Brumfield GL, Knoche SM, Doty KR, Larson AC, Poelaert BJ, Coulter DW, Solheim JC. Amyloid precursor-like protein 2 expression in macrophages: differentiation and M1/M2 macrophage dynamics. Front Oncol 2025; 15:1570955. [PMID: 40265027 PMCID: PMC12011594 DOI: 10.3389/fonc.2025.1570955] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2025] [Accepted: 03/13/2025] [Indexed: 04/24/2025] Open
Abstract
Amyloid precursor-like protein 2 (APLP2) has been previously associated with pro-tumor phenotypes in cancer cells, and in this current study we investigated the expression and functions of this protein in macrophages. Our findings showed that APLP2 expression was increased in monocyte-like U937 cells after cytokine-induced differentiation to macrophage-like cells. Evaluation of human mRNA data revealed that APLP2 is more highly expressed in human M2/anti-inflammatory (pro-tumor) macrophages than in M1 macrophages (which have a pro-inflammatory, anti-tumor phenotype). Consistent with the mRNA data, by immunoblotting we identified increased APLP2 protein expression in mouse M2/anti-inflammatory macrophages. Intratumoral infiltration of M2/anti-inflammatory macrophages has been reported in several cancers, including neuroblastoma (NB). We observed that treatment of macrophages with NB-conditioned media induced M2/anti-inflammatory and mixed phenotypes. Through comparison of macrophages from wild-type and APLP2-knockout mice, we correlated alterations in inflammation-associated markers with the presence of APLP2. This suggests that APLP2 influences macrophage polarization dynamics between M0/unpolarized and pro- and anti-inflammatory states, and populations altered by APLP2 KO resemble the macrophage profiles altered with NB-conditioned media treatment. In total, our work implicates APLP2 as a mediator of macrophage status, namely in the M0/unpolarized macrophage and the M1/pro-inflammatory and M2/anti-inflammatory axis.
Collapse
Affiliation(s)
- Gabrielle L. Brumfield
- Eppley Institute, University of Nebraska Medical Center, Omaha, NE, United States
- Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE, United States
| | - Shelby M. Knoche
- Eppley Institute, University of Nebraska Medical Center, Omaha, NE, United States
- Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE, United States
| | - Kenadie R. Doty
- Eppley Institute, University of Nebraska Medical Center, Omaha, NE, United States
- Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE, United States
| | - Alaina C. Larson
- Eppley Institute, University of Nebraska Medical Center, Omaha, NE, United States
- Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE, United States
| | - Brittany J. Poelaert
- Eppley Institute, University of Nebraska Medical Center, Omaha, NE, United States
- Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE, United States
| | - Don W. Coulter
- Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE, United States
- Department of Pediatrics, University of Nebraska Medical Center, Omaha, NE, United States
- Children's Nebraska, Omaha, NE, United States
| | - Joyce C. Solheim
- Eppley Institute, University of Nebraska Medical Center, Omaha, NE, United States
- Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE, United States
| |
Collapse
|
4
|
Marsland P, Vore AS, Lutzke A, Gano A, Fischer A, Trapp S, Savage LM, Deak T. Sex-specific effects of chronic alcohol consumption across the lifespan in the transgenic Alzheimer's Disease (TgF344-AD) rat model. Brain Behav Immun 2025; 128:192-207. [PMID: 40187669 DOI: 10.1016/j.bbi.2025.03.034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/16/2024] [Revised: 03/25/2025] [Accepted: 03/29/2025] [Indexed: 04/07/2025] Open
Abstract
Alcohol consumption across the lifespan contributes to mood fluctuations and cognitive dysfunction, two neurobehavioral features also associated with Alzheimer's Disease and Related Dementias (ADRD). Yet, few studies have used rodent models to determine how a history of ethanol consumption across the lifespan might contribute to neurobehavioral and neuropathological features of ADRD. We exposed Wild Type (WT) and transgenic Fischer 344 CE rats (TgF344-AD) that have been genetically modified to express the human Amyloid Precursor Protein (APP) and presenilin-1 genes with mutations, to ethanol using a chronic, intermittent ethanol consumption model. Beginning at P28, rats were given a single bottle 10 % ethanol solution for 2 consecutive days, followed by 2 days of tap water. This pattern (2 days on, days off) was repeated for a total of 12 cycles until rats reached the age of ∼ 3 months, and repeated at 6 (Exp 1 and Exp 2) and 9 months of age (Exp 2). In experiment 1, ethanol consumption decreased alternations in a spontaneous alternation task in females, only at the 3-month time point, whereas TgF344-AD females showed increased contextual fear conditioning in the test of retention and reinstatement tests at 6 months of age. In experiment 2, a battery of anxiety-like behaviors (Elevated Plus Maze, Marble Burying, and Novelty Induced Hypophagia) were assessed following a 2-week abstinence period at 3, 6, and 9 months of age in ethanol-consuming rats. Data from the EPM and marble burying tasks revealed evidence of heightened anxiety-like behavior in Tg-F344-AD rats that varied by sex and age, with no significant effects of ethanol. In the novelty-induced hypophagia task, males with a history of ethanol consumption had a lower latency to approach a familiar, salient reward at 3 months old, but effects of ethanol were overall minimal. Examination of dorsal hippocampal gene expression at 6 months of age under basal conditions also revealed predominantly genotype and sex-specific effects on inflammation- and AD-related genes (App, Il-6, Bace1, Rage, Lrp-1). When examined at 9 months old following LPS challenge, ethanol increased inflammatory genes in males (Il-1β, Il-6) in the hippocampus, whereas ethanol decreased several inflammatory and AD-related genes (Hmgb1, Rage, Bace1, Lrp-1) in TgF344-AD females. Overall, these data provide further evidence that females are especially vulnerable to AD, and that a history of ethanol consumption had selective, rather than global, effects on AD- and inflammation-related genes following an inflammatory stimulus.
Collapse
Affiliation(s)
- Paige Marsland
- Behavioral Neuroscience Program, Department of Psychology, Binghamton University, Binghamton, NY 13902-6000, United States
| | - Andrew S Vore
- Behavioral Neuroscience Program, Department of Psychology, Binghamton University, Binghamton, NY 13902-6000, United States
| | - Ashley Lutzke
- Behavioral Neuroscience Program, Department of Psychology, Binghamton University, Binghamton, NY 13902-6000, United States
| | - Anny Gano
- Behavioral Neuroscience Program, Department of Psychology, Binghamton University, Binghamton, NY 13902-6000, United States
| | - Abigail Fischer
- Behavioral Neuroscience Program, Department of Psychology, Binghamton University, Binghamton, NY 13902-6000, United States
| | - Sarah Trapp
- Behavioral Neuroscience Program, Department of Psychology, Binghamton University, Binghamton, NY 13902-6000, United States
| | - Lisa M Savage
- Behavioral Neuroscience Program, Department of Psychology, Binghamton University, Binghamton, NY 13902-6000, United States
| | - Terrence Deak
- Behavioral Neuroscience Program, Department of Psychology, Binghamton University, Binghamton, NY 13902-6000, United States.
| |
Collapse
|
5
|
Almeida ZL, Vaz DC, Brito RMM. Morphological and Molecular Profiling of Amyloid-β Species in Alzheimer's Pathogenesis. Mol Neurobiol 2025; 62:4391-4419. [PMID: 39446217 PMCID: PMC11880078 DOI: 10.1007/s12035-024-04543-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2024] [Accepted: 10/08/2024] [Indexed: 10/25/2024]
Abstract
Alzheimer's disease (AD) is the most common form of dementia around the world (~ 65%). Here, we portray the neuropathology of AD, biomarkers, and classification of amyloid plaques (diffuse, non-cored, dense core, compact). Tau pathology and its involvement with Aβ plaques and cell death are discussed. Amyloid cascade hypotheses, aggregation mechanisms, and molecular species formed in vitro and in vivo (on- and off-pathways) are described. Aβ42/Aβ40 monomers, dimers, trimers, Aβ-derived diffusible ligands, globulomers, dodecamers, amylospheroids, amorphous aggregates, protofibrils, fibrils, and plaques are characterized (structure, size, morphology, solubility, toxicity, mechanistic steps). An update on AD-approved drugs by regulatory agencies, along with new Aβ-based therapies, is presented. Beyond prescribing Aβ plaque disruptors, cholinergic agonists, or NMDA receptor antagonists, other therapeutic strategies (RNAi, glutaminyl cyclase inhibitors, monoclonal antibodies, secretase modulators, Aβ aggregation inhibitors, and anti-amyloid vaccines) are already under clinical trials. New drug discovery approaches based on "designed multiple ligands", "hybrid molecules", or "multitarget-directed ligands" are also being put forward and may contribute to tackling this highly debilitating and fatal form of human dementia.
Collapse
Affiliation(s)
- Zaida L Almeida
- Chemistry Department and Coimbra Chemistry Centre - Institute of Molecular Sciences (CQC-IMS), University of Coimbra, 3004-535, Coimbra, Portugal.
| | - Daniela C Vaz
- Chemistry Department and Coimbra Chemistry Centre - Institute of Molecular Sciences (CQC-IMS), University of Coimbra, 3004-535, Coimbra, Portugal.
- School of Health Sciences, Polytechnic Institute of Leiria, 2411-901, Leiria, Portugal.
- LSRE-LCM, Laboratory of Separation and Reaction Engineering and Laboratory of Catalysis and Materials, Leiria, 2411-901, Portugal.
- ALiCE - Associate Laboratory in Chemical Engineering, University of Porto, 4200-465, Porto, Portugal.
| | - Rui M M Brito
- Chemistry Department and Coimbra Chemistry Centre - Institute of Molecular Sciences (CQC-IMS), University of Coimbra, 3004-535, Coimbra, Portugal.
| |
Collapse
|
6
|
Zare H, Kasdorf MM, Bakhshian Nik A. Microfluidics in neural extracellular vesicles characterization for early Alzheimer's disease diagnosis. Mol Cell Neurosci 2025; 132:103982. [PMID: 39631514 DOI: 10.1016/j.mcn.2024.103982] [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: 08/13/2024] [Revised: 11/04/2024] [Accepted: 11/30/2024] [Indexed: 12/07/2024] Open
Abstract
Dementia is a general term for conditions impairing cognitive abilities including perception, reasoning, attention, judgment, memory, and daily brain function. Early diagnosis of Alzheimer's disease (AD), the most common form of dementia, using neural extracellular vesicles (nEVs) is the focus of the current study. These nEVs carry AD biomarkers including β-amyloid proteins and phosphorylated tau proteins. The novelty of this review lies in developing a microfluidic perspective by introducing the techniques using a microfluidic platform for early diagnosis of AD. A microfluidic device can detect small sample sizes with significantly low concentrations. These devices combine nEV isolation, enrichment, and detection, which makes them ideal candidates for early AD diagnosis.
Collapse
Affiliation(s)
- Hossein Zare
- Chemical and Biochemical Engineering Department, The University of Iowa, Iowa City, IA 52242, USA.
| | | | | |
Collapse
|
7
|
Muhammedkutty FNK, Zhou HX. Membrane-assisted Aβ40 aggregation pathways. CELL REPORTS. PHYSICAL SCIENCE 2025; 6:102436. [PMID: 40083905 PMCID: PMC11905421 DOI: 10.1016/j.xcrp.2025.102436] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 03/16/2025]
Abstract
Alzheimer's disease (AD) is caused by the assembly of amyloid-beta (Aβ) peptides into oligomers and fibrils. Endogenous Aβ aggregation may be assisted by cell membranes, which can accelerate the nucleation step enormously, but knowledge of membrane-assisted aggregation is still very limited. Here, we used extensive molecular dynamics (MD) simulations to structurally and energetically characterize key intermediates along the membrane-assisted aggregation pathways of Aβ40. Reinforcing experimental observations, the simulations reveal unique roles of GM1 ganglioside and cholesterol in stabilizing membrane-embedded β sheets and of Y10 and K28 in the ordered release of a small oligomeric seed into solution. The same seed leads to either an open-shaped or R-shaped fibril, with significant stabilization provided by inter- or intra-subunit interfaces between a straight β sheet (residues Q15-D23) and a bent β sheet (residues A30-V36). This work presents a comprehensive picture of membrane-assisted aggregation of Aβ40, with broad implications for developing AD therapies and rationalizing disease-specific polymorphisms of amyloidogenic proteins.
Collapse
Affiliation(s)
| | - Huan-Xiang Zhou
- Department of Chemistry, University of Illinois Chicago, Chicago, IL, USA
- Department of Physics, University of Illinois Chicago, Chicago, IL, USA
- Lead contact
| |
Collapse
|
8
|
Kroll F, Donnelly J, Özcan GG, Mackay E, Rihel J. Behavioural pharmacology predicts disrupted signalling pathways and candidate therapeutics from zebrafish mutants of Alzheimer's disease risk genes. eLife 2025; 13:RP96839. [PMID: 39960847 PMCID: PMC11832171 DOI: 10.7554/elife.96839] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/20/2025] Open
Abstract
By exposing genes associated with disease, genomic studies provide hundreds of starting points that should lead to druggable processes. However, our ability to systematically translate these genomic findings into biological pathways remains limited. Here, we combine rapid loss-of-function mutagenesis of Alzheimer's risk genes and behavioural pharmacology in zebrafish to predict disrupted processes and candidate therapeutics. FramebyFrame, our expanded package for the analysis of larval behaviours, revealed that decreased night-time sleep was common to F0 knockouts of all four late-onset Alzheimer's risk genes tested. We developed an online tool, ZOLTAR, which compares any behavioural fingerprint to a library of fingerprints from larvae treated with 3677 compounds. ZOLTAR successfully predicted that sorl1 mutants have disrupted serotonin signalling and identified betamethasone as a drug which normalises the excessive day-time sleep of presenilin-2 knockout larvae with minimal side effects. Predictive behavioural pharmacology offers a general framework to rapidly link disease-associated genes to druggable pathways.
Collapse
Affiliation(s)
- François Kroll
- Department of Cell and Developmental Biology, University College LondonLondonUnited Kingdom
- Institut de la Vision, Sorbonne UniversitéParisFrance
| | - Joshua Donnelly
- Department of Cell and Developmental Biology, University College LondonLondonUnited Kingdom
| | - Güliz Gürel Özcan
- Department of Cell and Developmental Biology, University College LondonLondonUnited Kingdom
| | - Eirinn Mackay
- Department of Cell and Developmental Biology, University College LondonLondonUnited Kingdom
| | - Jason Rihel
- Department of Cell and Developmental Biology, University College LondonLondonUnited Kingdom
| |
Collapse
|
9
|
Aubrey LD, Radford SE. How is the Amyloid Fold Built? Polymorphism and the Microscopic Mechanisms of Fibril Assembly. J Mol Biol 2025:169008. [PMID: 39954780 DOI: 10.1016/j.jmb.2025.169008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2024] [Revised: 02/06/2025] [Accepted: 02/11/2025] [Indexed: 02/17/2025]
Abstract
For a given protein sequence, many, up to sometimes hundreds of different amyloid fibril folds, can be formed in vitro. Yet, fibrils extracted from, or found in, human tissue, usually at the end of a long disease process, are often structurally homogeneous. Through monitoring of amyloid assembly reactions in vitro, the scientific community has gained a detailed understanding of the kinetic mechanisms of fibril assembly and the rates at which the different processes involved occur. However, how this kinetic information relates to the structural changes as a protein transforms from its initial, native structure to the canonical cross-β structure of amyloid remain obscure. While cryoEM has yielded a plethora of high-resolution information that portray a vast variety of fibril structures, there remains little knowledge of how and why each particular structure resulted. Recent work has demonstrated that fibril structures can change over an assembly time course, despite the different fibril types having similar thermodynamic stability. This points to kinetic control of the fibrils formed, with structures that initiate or elongate faster becoming the dominant products of assembly. Annotating kinetic assembly mechanisms alongside structural analysis of the fibrils formed is required to truly understand the molecular mechanisms of amyloid formation. However, this is a complicated task. In this review, we discuss how embracing this challenge could open new frontiers in amyloid research and new opportunities for therapy.
Collapse
Affiliation(s)
- Liam D Aubrey
- Astbury Centre for Structural Molecular Biology, School of Molecular and Cellular Biology, Faculty of Biological Science, University of Leeds, Leeds LS2 9JT, United Kingdom
| | - Sheena E Radford
- Astbury Centre for Structural Molecular Biology, School of Molecular and Cellular Biology, Faculty of Biological Science, University of Leeds, Leeds LS2 9JT, United Kingdom.
| |
Collapse
|
10
|
Jáuregui GV, Parpura V. Neuron-Astrocyte Interactions in Aging and Alzheimer's Disease: Dysregulation of Amyloid Precursor Protein. AGEING & LONGEVITY 2025; 6:117-128. [PMID: 40098995 PMCID: PMC11911455 DOI: 10.47855/jal9020-2025-2-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/19/2025]
Abstract
Amyloid precursor protein (APP) is central to Alzheimer's disease (AD) by its role in Aβ build-up and in neuronal and astrocytic malfunction. The major risk factor for late-onset AD is aging, which increases APP processing in both neurons and astrocytes, and consequently increases Aβ production. This focused review covers the subjects of how aging and AD affect APP dynamics within the both cell types and how astrocytes dysfunction can enhance neuroinflammation and neuronal dysfunction and injury. We discuss the interplay between neurons and astrocytes in aging and AD brains, where bi-directional cellular interactions accelerate neurodegeneration.
Collapse
Affiliation(s)
- Gretsen Velezmoro Jáuregui
- International Translational Neuroscience Research Institute, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, P.R. China
| | - Vladimir Parpura
- International Translational Neuroscience Research Institute, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, P.R. China
| |
Collapse
|
11
|
Krishnamurthy HK, Jayaraman V, Krishna K, Wang T, Bei K, Changalath C, Rajasekaran JJ. An overview of the genes and biomarkers in Alzheimer's disease. Ageing Res Rev 2025; 104:102599. [PMID: 39612989 DOI: 10.1016/j.arr.2024.102599] [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: 09/03/2024] [Revised: 11/25/2024] [Accepted: 11/25/2024] [Indexed: 12/01/2024]
Abstract
Alzheimer's disease (AD) is the most common type of dementia and neurodegenerative disease characterized by neurofibrillary tangles (NFTs) and amyloid plaque. Familial AD is caused by mutations in the APP, PSEN1, and PSEN2 genes and these mutations result in the early onset of the disease. Sporadic AD usually affects older adults over the age of 65 years and is, therefore classified as late-onset AD (LOAD). Several risk factors associated with LOAD including the APOE gene have been identified. Moreover, GWAS studies have identified a wide array of genes and polymorphisms that are associated with LOAD risk. Currently, the diagnosis of AD involves the evaluation of memory and personality changes, cognitive impairment, and medical and family history to rule out other diseases. Laboratory tests to assess the biomarkers in the body fluids as well as MRI, CT, and PET scans to analyze the presence of plaques and NFTs are also included in the diagnosis of AD. It is important to diagnose AD before the onset of clinical symptoms, i.e. during the preclinical stage, to delay the progression and for better management of the disease. Research has been conducted to identify biomarkers of AD in the CSF, serum, saliva, and urine during the preclinical stage. Current research has identified several biomarkers and potential biomarkers in the body fluids that enhance diagnostic accuracy. Aside from genetics, other factors such as diet, physical activity, and lifestyle factors may influence the risk of developing AD. Clinical trials are underway to find potential biomarkers, diagnostic measures, and treatments for AD mainly in the preclinical stage. This review provides an overview of the genes and biomarkers of AD.
Collapse
Affiliation(s)
| | | | - Karthik Krishna
- Vibrant Sciences LLC., San Carlos, CA, United States of America.
| | - Tianhao Wang
- Vibrant Sciences LLC., San Carlos, CA, United States of America.
| | - Kang Bei
- Vibrant Sciences LLC., San Carlos, CA, United States of America.
| | | | | |
Collapse
|
12
|
Zheng Q, Wang X. Alzheimer's disease: insights into pathology, molecular mechanisms, and therapy. Protein Cell 2025; 16:83-120. [PMID: 38733347 PMCID: PMC11786724 DOI: 10.1093/procel/pwae026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2024] [Accepted: 05/06/2024] [Indexed: 05/13/2024] Open
Abstract
Alzheimer's disease (AD), the leading cause of dementia, is characterized by the accumulation of amyloid plaques and neurofibrillary tangles in the brain. This condition casts a significant shadow on global health due to its complex and multifactorial nature. In addition to genetic predispositions, the development of AD is influenced by a myriad of risk factors, including aging, systemic inflammation, chronic health conditions, lifestyle, and environmental exposures. Recent advancements in understanding the complex pathophysiology of AD are paving the way for enhanced diagnostic techniques, improved risk assessment, and potentially effective prevention strategies. These discoveries are crucial in the quest to unravel the complexities of AD, offering a beacon of hope for improved management and treatment options for the millions affected by this debilitating disease.
Collapse
Affiliation(s)
- Qiuyang Zheng
- Shenzhen Research Institute of Xiamen University, Shenzhen 518057, China
- State Key Laboratory of Cellular Stress Biology, Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, Institute of Neuroscience, Department of Neurology, the First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen 361005, China
| | - Xin Wang
- Shenzhen Research Institute of Xiamen University, Shenzhen 518057, China
- State Key Laboratory of Cellular Stress Biology, Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, Institute of Neuroscience, Department of Neurology, the First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen 361005, China
| |
Collapse
|
13
|
Martá-Ariza M, Leitner DF, Kanshin E, Suazo J, Giusti Pedrosa A, Thierry M, Lee EB, Devinsky O, Drummond E, Fortea J, Lleó A, Ueberheide B, Wisniewski T. Comparison of the amyloid plaque proteome in Down syndrome, early-onset Alzheimer's disease, and late-onset Alzheimer's disease. Acta Neuropathol 2025; 149:9. [PMID: 39825890 PMCID: PMC11742868 DOI: 10.1007/s00401-025-02844-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2024] [Revised: 01/02/2025] [Accepted: 01/04/2025] [Indexed: 01/20/2025]
Abstract
Down syndrome (DS) is strongly associated with Alzheimer's disease (AD) due to APP overexpression, exhibiting Amyloid-β (Aβ) and Tau pathology similar to early-onset (EOAD) and late-onset AD (LOAD). We evaluated the Aβ plaque proteome of DS, EOAD, and LOAD using unbiased localized proteomics on post-mortem paraffin-embedded tissues from four cohorts (n = 20/group): DS (59.8 ± 4.99 y/o), EOAD (63 ± 4.07 y/o), LOAD (82.1 ± 6.37 y/o), and controls (66.4 ± 13.04). We identified differentially abundant proteins when comparing Aβ plaques and neighboring non-plaque tissue (FDR < 5%, fold-change > 1.5) in DS (n = 132), EOAD (n = 192), and LOAD (n = 128), with 43 plaque-associated proteins shared across all groups. Positive correlations were observed between plaque-associated proteins in DS and EOAD (R2 = .77), DS and LOAD (R2 = .73), and EOAD and LOAD (R2 = .67). Top gene ontology biological processes (GOBP) included lysosomal transport (p = 1.29 × 10-5) for DS, immune system regulation (p = 4.33 × 10-5) for EOAD, and lysosome organization (p = 0.029) for LOAD. Protein networks revealed a plaque-associated protein signature involving APP metabolism, immune response, and lysosomal functions. In DS, EOAD, and LOAD non-plaque vs. control tissue, we identified 263, 269, and 301 differentially abundant proteins, with 65 altered proteins shared across all cohorts. Non-plaque proteins in DS showed modest correlations with EOAD (R2 = .59) and LOAD (R2 = .33) compared to the correlation between EOAD and LOAD (R2 = .79). Top GOBP term for all groups was chromatin remodeling (p < 0.001), with additional terms for DS including extracellular matrix, and protein-DNA complexes and gene expression regulation for EOAD and LOAD. Our study reveals key functional characteristics of the amyloid plaque proteome in DS, compared to EOAD and LOAD, highlighting shared pathways in endo/lysosomal functions and immune responses. The non-plaque proteome revealed distinct alterations in ECM and chromatin structure, underscoring unique differences between DS and AD subtypes. Our findings enhance our understanding of AD pathogenesis and identify potential biomarkers and therapeutic targets.
Collapse
Affiliation(s)
- Mitchell Martá-Ariza
- Department of Neurology, NYU Grossman School of Medicine, New York, NY, USA
- Center for Cognitive Neurology, NYU Grossman School of Medicine, New York, NY, USA
- Institut de Neurociències, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Dominique F Leitner
- Department of Neurology, NYU Grossman School of Medicine, New York, NY, USA
- Center for Cognitive Neurology, NYU Grossman School of Medicine, New York, NY, USA
- Comprehensive Epilepsy Center, Department of Neurology, NYU Langone Health and Grossman School of Medicine, New York, NY, USA
| | - Evgeny Kanshin
- Proteomics Laboratory, Division of Advanced Research Technologies, NYU Grossman School of Medicine, New York, NY, USA
- Department of Biochemistry and Molecular Pharmacology, NYU Grossman School of Medicine, New York, NY, USA
| | - Jianina Suazo
- Department of Neurology, NYU Grossman School of Medicine, New York, NY, USA
- Center for Cognitive Neurology, NYU Grossman School of Medicine, New York, NY, USA
| | | | - Manon Thierry
- Department of Neurology, NYU Grossman School of Medicine, New York, NY, USA
- Center for Cognitive Neurology, NYU Grossman School of Medicine, New York, NY, USA
| | - Edward B Lee
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - Orrin Devinsky
- Department of Neurology, NYU Grossman School of Medicine, New York, NY, USA
- Comprehensive Epilepsy Center, Department of Neurology, NYU Langone Health and Grossman School of Medicine, New York, NY, USA
| | - Eleanor Drummond
- Center for Cognitive Neurology, NYU Grossman School of Medicine, New York, NY, USA
- Brain and Mind Centre and School of Medical Sciences, University of Sydney, Camperdown, NSW, Australia
| | - Juan Fortea
- Memory Unit, Department of Neurology, Institut de Recerca Sant Pau, Hospital de Sant Pau, Universitat Autònoma de Barcelona, Barcelona, Spain
- Centro de Investigación Biomédica en Red en Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
- Barcelona Down Medical Center, Fundació Catalana de Síndrome de Down, Barcelona, Spain
| | - Alberto Lleó
- Memory Unit, Department of Neurology, Institut de Recerca Sant Pau, Hospital de Sant Pau, Universitat Autònoma de Barcelona, Barcelona, Spain
- Centro de Investigación Biomédica en Red en Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | - Beatrix Ueberheide
- Department of Neurology, NYU Grossman School of Medicine, New York, NY, USA
- Proteomics Laboratory, Division of Advanced Research Technologies, NYU Grossman School of Medicine, New York, NY, USA
- Department of Biochemistry and Molecular Pharmacology, NYU Grossman School of Medicine, New York, NY, USA
| | - Thomas Wisniewski
- Department of Neurology, NYU Grossman School of Medicine, New York, NY, USA.
- Center for Cognitive Neurology, NYU Grossman School of Medicine, New York, NY, USA.
- Department of Pathology, NYU Grossman School of Medicine, New York, NY, USA.
- Department of Psychiatry, NYU Grossman School of Medicine, New York, NY, USA.
| |
Collapse
|
14
|
Volloch V, Rits-Volloch S. Production of Amyloid-β in the Aβ-Protein-Precursor Proteolytic Pathway Is Discontinued or Severely Suppressed in Alzheimer's Disease-Affected Neurons: Contesting the 'Obvious'. Genes (Basel) 2025; 16:46. [PMID: 39858593 PMCID: PMC11764795 DOI: 10.3390/genes16010046] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2024] [Revised: 12/17/2024] [Accepted: 12/19/2024] [Indexed: 01/27/2025] Open
Abstract
A notion of the continuous production of amyloid-β (Aβ) via the proteolysis of Aβ-protein-precursor (AβPP) in Alzheimer's disease (AD)-affected neurons constitutes both a cornerstone and an article of faith in the Alzheimer's research field. The present Perspective challenges this assumption. It analyses the relevant empirical data and reaches an unexpected conclusion, namely that in AD-afflicted neurons, the production of AβPP-derived Aβ is either discontinued or severely suppressed, a concept that, if proven, would fundamentally change our understanding of the disease. This suppression, effectively self-suppression, occurs in the context of the global inhibition of the cellular cap-dependent protein synthesis as a consequence of the neuronal integrated stress response (ISR) elicited by AβPP-derived intraneuronal Aβ (iAβ; hence self-suppression) upon reaching certain levels. Concurrently with the suppression of the AβPP proteolytic pathway, the neuronal ISR activates in human neurons, but not in mouse neurons, the powerful AD-driving pathway generating the C99 fragment of AβPP independently of AβPP. The present study describes molecular mechanisms potentially involved in these phenomena, propounds novel approaches to generate transgenic animal models of AD, advocates for the utilization of human neuronal cells-based models of the disease, makes verifiable predictions, suggests experiments designed to validate the proposed concept, and considers its potential research and therapeutic implications. Remarkably, it opens up the possibility that the conventional production of AβPP, BACE enzymes, and γ-secretase components is also suppressed under the neuronal ISR conditions in AD-affected neurons, resulting in the dyshomeostasis of AβPP. It follows that whereas conventional AD is triggered by AβPP-derived iAβ accumulated to the ISR-eliciting levels, the disease, in its both conventional and unconventional (triggered by the neuronal ISR-eliciting stressors distinct from iAβ) forms, is driven not (or not only) by iAβ produced in the AβPP-independent pathway, as we proposed previously, but mainly, possibly exclusively, by the C99 fragment generated independently of AβPP and not cleaved at the γ-site due to the neuronal ISR-caused deficiency of γ-secretase (apparently, the AD-driving "substance X" predicted in our previous study), a paradigm consistent with a dictum by George Perry that Aβ is "central but not causative" in AD. The proposed therapeutic strategies would not only deplete the driver of the disease and abrogate the AβPP-independent production of C99 but also reverse the neuronal ISR and ameliorate the AβPP dyshomeostasis, a potentially significant contributor to AD pathology.
Collapse
Affiliation(s)
- Vladimir Volloch
- Department of Developmental Biology, Harvard School of Dental Medicine, Boston, MA 02115, USA
| | - Sophia Rits-Volloch
- Division of Molecular Medicine, Children’s Hospital, Boston, MA 02115, USA
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA 02115, USA
| |
Collapse
|
15
|
Aisen P, Bateman RJ, Crowther D, Cummings J, Dwyer J, Iwatsubo T, Kosco‐Vilbois M, McDade E, Mohs R, Scheltens P, Sperling R, Selkoe D. The case for regulatory approval of amyloid-lowering immunotherapies in Alzheimer's disease based on clearcut biomarker evidence. Alzheimers Dement 2025; 21:e14342. [PMID: 39535341 PMCID: PMC11772734 DOI: 10.1002/alz.14342] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Revised: 09/23/2024] [Accepted: 09/27/2024] [Indexed: 11/16/2024]
Abstract
Decades of research have provided evidence that Alzheimer's disease (AD) is caused in part by cerebral accumulation of amyloid beta-protein (Aβ). In 2023, the US Food and Drug Administration gave full regulatory approval to a disease-modifying Aβ antibody for early AD. Secondary prevention trials with Aβ antibodies are underway. We summarize peer-reviewed evidence for targeting Aβ and argue that regulators should consider approving new agents working by similar mechanisms (Aβ antibodies and vaccines) based on robust amyloid lowering and reasonable safety. The urgent need to provide treatments to millions of mildly symptomatic patients suggests that AD should join other diseases for which standard approval is based on significant changes in mechanistically meaningful biomarkers coupled with safety. Robust amyloid lowering in secondary prevention trials of people who have amyloid plaques but are asymptomatic could also provide evidence of a change in the pathophysiological progression of AD as a basis for regulatory approval. HIGHLIGHTS: Thirteen key findings support amyloid beta as a cause of Alzheimer's disease (AD). Three immunotherapies lower amyloid and slow decline, allowing regulatory approval. New such agents could be considered for approval due to amyloid lowering and safety. Urgency suggests AD may join diseases with approval due to a key biomarker + safety.
Collapse
Affiliation(s)
- Paul Aisen
- USC Alzheimer's Therapeutic Research InstituteSan DiegoCaliforniaUSA
| | - Randall J. Bateman
- Department of NeurologyWashington University School of MedicineSt. LouisMissouriUSA
| | - Damian Crowther
- TRIMTECH Therapeutics Ltd and Medical and More LtdBostonMassachusettsUSA
| | - Jeff Cummings
- School of Integrated Health SciencesUniversity of Nevada Las VegasLas VegasNevadaUSA
| | - John Dwyer
- Global Alzheimer's Platform Foundation WashingtonWashingtonDistrict of ColumbiaUSA
| | | | | | - Eric McDade
- Department of NeurologyWashington University School of MedicineSt. LouisMissouriUSA
| | - Richard Mohs
- Global Alzheimer's Platform Foundation WashingtonWashingtonDistrict of ColumbiaUSA
| | - Philip Scheltens
- Medical Center and EQT Life SciencesAmsterdam UniversityAmsterdamThe Netherlands
| | - Reisa Sperling
- Dpartment of NeurologyBrigham and Women's Hospital and Harvard Medical SchoolBostonMassachusettsUSA
| | - Dennis Selkoe
- Dpartment of NeurologyBrigham and Women's Hospital and Harvard Medical SchoolBostonMassachusettsUSA
| |
Collapse
|
16
|
Ng LLH, Chow J, Lau KF. The AICD interactome: implications in neurodevelopment and neurodegeneration. Biochem Soc Trans 2024; 52:2539-2556. [PMID: 39670668 DOI: 10.1042/bst20241510] [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: 09/19/2024] [Revised: 11/25/2024] [Accepted: 11/26/2024] [Indexed: 12/14/2024]
Abstract
The pathophysiological mechanism involving the proteolytic processing of amyloid precursor protein (APP) and the generation of amyloid plaques is of significant interest in research on Alzheimer's disease (AD). The increasing significance of the downstream AD-related pathophysiological mechanisms has sparked research interest in other products of the APP processing cascades, including the APP intracellular domain (AICD). The potential importance of AICD in various cellular processes in the central nervous system has been established through the identification of its interactors. The interaction between AICD and its physiological binding partners is implicated in cellular events including regulation of transcriptional activity, cytoskeletal dynamics, neuronal growth, APP processing and cellular apoptosis. On the contrary, AICD is also implicated in neurodegeneration, which is a potential outcome of the functional fluctuation of AICD-mediated neuronal processes within the neuronal network. In this review, we summarize the neuronal functions and pathological manifestations of the dynamic AICD interaction network.
Collapse
Affiliation(s)
- Laura Lok-Haang Ng
- School of Life Sciences, Faculty of Science, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Jessica Chow
- School of Life Sciences, Faculty of Science, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Kwok-Fai Lau
- School of Life Sciences, Faculty of Science, The Chinese University of Hong Kong, Hong Kong SAR, China
| |
Collapse
|
17
|
Zimbone S, Giuffrida ML, Sciacca MFM, Carrotta R, Librizzi F, Milardi D, Grasso G. A VEGF Fragment Encompassing Residues 10-30 Inhibits Aβ1-42 Amyloid Aggregation and Exhibits Neuroprotective Properties Matching the Full-Length Protein. ACS Chem Neurosci 2024; 15:4580-4590. [PMID: 39587417 DOI: 10.1021/acschemneuro.4c00669] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2024] Open
Abstract
The intricate relationship between brain vascular diseases and neurodegeneration has garnered increased attention in the scientific community. With an aging population, the incidence of these two conditions is likely to increase, making it imperative to understand the underlying common molecular mechanisms and unveiling novel avenues for therapy. Prompted by the observation that Aβ peptide aggregation has been implicated in the development of cerebral amyloid angiopathy (CAA) and that elevated concentrations of vascular endothelial growth factor (VEGF) in the cerebrospinal fluid (CSF) have been correlated with less cognitive decline in Alzheimer's disease (AD), we demonstrate that a small peptide (Pep9) encompassing the 10-30 sequence of VEGF exhibits significant ability to inhibit the aggregation of the Aβ1-42 peptide, as well as the formation of toxic oligomers. AFM studies confirmed this inhibitory capacity, which is also paralleled by a significant reduction of the random coil to a beta-sheet conformational transition. Further studies have shown that Pep9 protects differentiated neuroblastoma SH-SY5Y cells from Aβ toxicity, being even more effective than full-length protein in preventing amyloid-induced neuronal death. The use of a control peptide wherein two histidines are substituted with glycines (H11G and H12G) suggests a close relationship between the peptide amino acid sequence and its antiaggregating/neuroprotective activity. Overall, this study provides insight into the role of VEGF in AD and suggests that specific VEGF fragments could be beneficial in the treatment of this condition.
Collapse
Affiliation(s)
- Stefania Zimbone
- Consiglio Nazionale delle Ricerche, Istituto di Cristallografia, Catania 95126, Italy
| | - M Laura Giuffrida
- Consiglio Nazionale delle Ricerche, Istituto di Cristallografia, Catania 95126, Italy
| | - Michele F M Sciacca
- Consiglio Nazionale delle Ricerche, Istituto di Cristallografia, Catania 95126, Italy
| | - Rita Carrotta
- Consiglio Nazionale delle Ricerche, Istituto di Biofisica, Palermo 90146, Italy
| | - Fabio Librizzi
- Consiglio Nazionale delle Ricerche, Istituto di Biofisica, Palermo 90146, Italy
| | - Danilo Milardi
- Consiglio Nazionale delle Ricerche, Istituto di Cristallografia, Catania 95126, Italy
| | - Giulia Grasso
- Consiglio Nazionale delle Ricerche, Istituto di Cristallografia, Catania 95126, Italy
| |
Collapse
|
18
|
Huang SH, Fang ST, Yang CH, Liou JW, Chen YC. Modulating Amyloid-β Toxicity: In Vitro Analysis of Aβ42(G37V) Variant Impact on Aβ42 Aggregation and Cytotoxicity. Int J Mol Sci 2024; 25:13219. [PMID: 39684928 DOI: 10.3390/ijms252313219] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2024] [Revised: 12/05/2024] [Accepted: 12/05/2024] [Indexed: 12/18/2024] Open
Abstract
Alzheimer's disease (AD) is primarily driven by the formation of toxic amyloid-β (Aβ) aggregates, with Aβ42 being a pivotal contributor to disease pathology. This study investigates a novel agent to mitigate Aβ42-induced toxicity by co-assembling Aβ42 with its G37V variant (Aβ42(G37V)), where Gly at position 37 is substituted with valine. Using a combination of Thioflavin-T (Th-T) fluorescence assays, Western blot analysis, atomic force microscopy (AFM)/transmission electron microscopy (TEM), and biochemical assays, we demonstrated that adding Aβ42(G37V) significantly accelerates Aβ42 aggregation rate and mass while altering the morphology of the resulting aggregates. Consequently, adding Aβ42(G37V) reduces the Aβ42 aggregates-induced cytotoxicity, as evidenced by improved cell viability assays. The possible mechanism for this effect is that adding Aβ42(G37V) reduces the production of reactive oxygen species (ROS) and lipid peroxidation, typically elevated in response to Aβ42, indicating its protective effects against oxidative stress. These findings suggest that Aβ42(G37V) could be a promising candidate for modulating Aβ42 aggregation dynamics and reducing its neurotoxic effects, providing a new avenue for potential therapeutic interventions in AD.
Collapse
Affiliation(s)
- Shu-Hsiang Huang
- Department of Medicine, MacKay Medical College, New Taipei City 252, Taiwan
| | - Shang-Ting Fang
- Department of Medicine, MacKay Medical College, New Taipei City 252, Taiwan
| | - Chin-Hao Yang
- Department of Biochemistry, School of Medicine, Tzu Chi University, Hualien City 970, Taiwan
| | - Je-Wen Liou
- Department of Biochemistry, School of Medicine, Tzu Chi University, Hualien City 970, Taiwan
| | - Yi-Cheng Chen
- Department of Medicine, MacKay Medical College, New Taipei City 252, Taiwan
| |
Collapse
|
19
|
Thai QM, Tung NT, Do Thi Mai D, Ngo ST. Dimerization of the Aβ 42 under the Influence of the Gold Nanoparticle: A REMD Study. J Phys Chem B 2024; 128:11705-11713. [PMID: 39508442 DOI: 10.1021/acs.jpcb.4c06224] [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: 11/15/2024]
Abstract
Advances in Alzheimer's disease (AD) are related to the oligomerization of Amyloid β (Aβ) peptides. Therefore, alteration of the process can prevent AD. We investigated the Aβ42 dimerization under the effects of gold nanoparticles using temperature replica-exchange molecular dynamics (REMD) simulations. The structural change of dimers in the presence and absence of the gold nanoparticle, Au55, was monitored over stable intervals. Physical insights into the oligomerization of Aβ were thus clarified. The computed metrics indicate that Au55 affects the progress of oligomerization. Specifically, the presence of the gold nanoparticle significantly modifies the structure of dimeric Aβ42. The β-content experienced a substantial decrease with the induction of Au55. The turn and coil-contents are also decreased under the effects of the gold nanoparticle. However, the α-content of the dimer exhibited a rigid increase. The influence of gold nanoparticles on the dimeric Aβ42 differs significantly from that of silver nanoparticles, which reduce β-content but increase coil-, turn-, and α-contents. The nature of inhibition will be discussed, in which the vdW interaction plays a driving force for the interaction between the Aβ42 dimer and the gold nanoparticle.
Collapse
Affiliation(s)
- Quynh Mai Thai
- Laboratory of Biophysics, Institute for Advanced Study in Technology, Ton Duc Thang University, Ho Chi Minh City 72915, Vietnam
- Faculty of Pharmacy, Ton Duc Thang University, Ho Chi Minh City 72915, Vietnam
| | - Nguyen Thanh Tung
- Institute of Materials Science, Vietnam Academy of Science and Technology, Hanoi 11307, Vietnam
- Graduate University of Science and Technology, Vietnam Academy of Science and Technology, Hanoi 11307, Vietnam
| | - Dung Do Thi Mai
- Faculty of Pharmaceutical Chemistry and Technology, Hanoi University of Pharmacy, 13-15 Le Thanh Tong, Hanoi 11021, Vietnam
| | - Son Tung Ngo
- Laboratory of Biophysics, Institute for Advanced Study in Technology, Ton Duc Thang University, Ho Chi Minh City 72915, Vietnam
- Faculty of Pharmacy, Ton Duc Thang University, Ho Chi Minh City 72915, Vietnam
| |
Collapse
|
20
|
Chen J, Li S, Zhang F, Chen J, Cai C, Guo Y, Lei Z, Zeng LH, Zi D, Shen Y, Tan J. The pathogenic APP N-terminal Val225Ala mutation alters tau protein liquid-liquid phase separation and exacerbates synaptic damage. Mol Psychiatry 2024:10.1038/s41380-024-02837-6. [PMID: 39558004 DOI: 10.1038/s41380-024-02837-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/09/2024] [Revised: 11/04/2024] [Accepted: 11/07/2024] [Indexed: 11/20/2024]
Abstract
Amyloid precursor protein (APP) is predominantly located in synapses of neurons and its mutations have been well recognized as the most important genetic causal factor for the familial Alzheimer's disease (AD). While most disease-causal mutations of APP occur within the Aβ-coding region or immediately proximal, the pathological impacts of mutations in the N-terminus of APP protein, which remote from the Aβ sequence, on neuron and synapse are still largely unknown. It was recently reported a pathogenic APP N-terminal Val225Ala mutation (APPV225A) with clinically featuring progressive dementia and typical AD pathologies in brain. In our present study, we further found that APPV225A mutation alters the N-terminal structure of APP, which enhances its binding affinity to tau protein and significantly increases APP-mediated endocytosis. Consequently, APPV225A promotes the uptake of extracellular tau into SH-SY5Y cells, further linking the structural change in APP to intracellular tau accumulation. In addition, APPV225A also notably alters the liquid-liquid phase separation (LLPS) of intracellular tau and intensified tau phosphorylation and aggregation in SH-SY5Y cells. Moreover, APPV225A promote AD-like tau pathology and synaptic damages in human induced pluripotent stem cells (hiPSCs)-derived neural progenitor cells and neurons, as well as in hiPSCs-derived human brain organoids and mouse brain, which can be ameliorated by tau knockdown. Proximity labeling identified several key APPV225A-interacting proteins, including HS3ST3A1, which was shown to directly regulate tau LLPS and phosphorylation. These findings nicely build on our previous work on roles for APP in tau-related pathological phenotypes and further highlight the involvement of N-terminal APP as the key region for both amyloidopathy and tauopathy, two aspects of AD pathogenesis and progression. Our study may also provide a theoretical breakthrough for AD therapy and highlight the important hub roles of APP and making previously neglected N-terminal APP as a potential target for the discovery of novel disease-modifying therapeutic agents against AD, holding significant scientific values and clinical promise.
Collapse
Affiliation(s)
- Jiang Chen
- Institute of Translational Medicine; Key Laboratory of Novel Targets and Drug Study for Neural Repair of Zhejiang Province, School of Medicine, Hangzhou City University, Hangzhou, 310015, China
- Department of Pharmacology, Zhejiang University School of Medicine, Hangzhou, 310058, China
- Key Laboratory of Endemic and Ethnic Diseases, Laboratory of Molecular Biology, Ministry of Education, Guizhou Medical University, Guiyang, 550025, China
| | - Song Li
- Center for Clinical Research on Neurological Diseases, the First Affiliated Hospital of Dalian Medical University, Dalian, 116021, China
| | - Fengning Zhang
- Key Laboratory of Endemic and Ethnic Diseases, Laboratory of Molecular Biology, Ministry of Education, Guizhou Medical University, Guiyang, 550025, China
| | - Junsheng Chen
- Key Laboratory of Endemic and Ethnic Diseases, Laboratory of Molecular Biology, Ministry of Education, Guizhou Medical University, Guiyang, 550025, China
| | - Chuanbin Cai
- Key Laboratory of Endemic and Ethnic Diseases, Laboratory of Molecular Biology, Ministry of Education, Guizhou Medical University, Guiyang, 550025, China
| | - Yi Guo
- Key Laboratory of Endemic and Ethnic Diseases, Laboratory of Molecular Biology, Ministry of Education, Guizhou Medical University, Guiyang, 550025, China
| | - Zhifeng Lei
- Key Laboratory of Endemic and Ethnic Diseases, Laboratory of Molecular Biology, Ministry of Education, Guizhou Medical University, Guiyang, 550025, China
| | - Ling-Hui Zeng
- Institute of Translational Medicine; Key Laboratory of Novel Targets and Drug Study for Neural Repair of Zhejiang Province, School of Medicine, Hangzhou City University, Hangzhou, 310015, China
| | - Dan Zi
- Department of Obstetrics and Gynecology, Guizhou Provincial People's Hospital, Guiyang, 550025, China
| | - Yong Shen
- Department of Neurology, Institute on Aging and Brain Disorders, The First Affiliated Hospital of USTC, Neurodegenerative Disorder Research Center, Division of Biological and Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China; CAS Key Laboratory of Brain Function and Disease, Anhui Provincial Key Laboratory of Biomedical Aging Research, University of Science and Technology of China, Hefei, 23006, China
| | - Jun Tan
- Institute of Translational Medicine; Key Laboratory of Novel Targets and Drug Study for Neural Repair of Zhejiang Province, School of Medicine, Hangzhou City University, Hangzhou, 310015, China.
- Key Laboratory of Endemic and Ethnic Diseases, Laboratory of Molecular Biology, Ministry of Education, Guizhou Medical University, Guiyang, 550025, China.
| |
Collapse
|
21
|
Kodosaki E, Bell R, Sogorb-Esteve A, Wiltshire K, Zetterberg H, Heslegrave A. More than microglia: myeloid cells and biomarkers in neurodegeneration. Front Neurosci 2024; 18:1499458. [PMID: 39544911 PMCID: PMC11560917 DOI: 10.3389/fnins.2024.1499458] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2024] [Accepted: 10/16/2024] [Indexed: 11/17/2024] Open
Abstract
The role of myeloid cells (granulocytes and monocytes) in neurodegeneration and neurodegenerative disorders (NDD) is indisputable. Here we discuss the roles of myeloid cells in neurodegenerative diseases, and the recent advances in biofluid and imaging myeloid biomarker research with a focus on methods that can be used in the clinic. For this review, evidence from three neurodegenerative diseases will be included, Alzheimer's disease (AD), Parkinson's disease (PD), and multiple sclerosis (MS). We discuss the potential for these biomarkers to be used in humans with suspected NDD as prognostic, diagnostic, or monitoring tools, identify knowledge gaps in literature, and propose potential approaches to further elucidate the role of myeloid cells in neurodegeneration and better utilize myeloid biomarkers in the understanding and treatment of NDD.
Collapse
Affiliation(s)
- Eleftheria Kodosaki
- Department of Neurodegenerative Disease, UCL Institute of Neurology, London, United Kingdom
- UK Dementia Research Institute at UCL, London, United Kingdom
| | - Rosie Bell
- Department of Neurodegenerative Disease, UCL Institute of Neurology, London, United Kingdom
- UK Dementia Research Institute at UCL, London, United Kingdom
| | - Aitana Sogorb-Esteve
- UK Dementia Research Institute at UCL, London, United Kingdom
- Dementia Research Centre, UCL Queen Square Institute of Neurology, University College London, London, United Kingdom
| | - Katharine Wiltshire
- Dementia Research Centre, UCL Queen Square Institute of Neurology, University College London, London, United Kingdom
| | - Henrik Zetterberg
- Department of Neurodegenerative Disease, UCL Institute of Neurology, London, United Kingdom
- UK Dementia Research Institute at UCL, London, United Kingdom
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
- Hong Kong Center for Neurodegenerative Diseases, Clear Water Bay, Hong Kong SAR, China
- Wisconsin Alzheimer’s Disease Research Center, University of Wisconsin School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, United States
| | - Amanda Heslegrave
- Department of Neurodegenerative Disease, UCL Institute of Neurology, London, United Kingdom
- UK Dementia Research Institute at UCL, London, United Kingdom
| |
Collapse
|
22
|
Martínez-Drudis L, Bérard M, Musiol D, Rivest S, Oueslati A. Pharmacological inhibition of PLK2 kinase activity mitigates cognitive decline but aggravates APP pathology in a sex-dependent manner in APP/PS1 mouse model of Alzheimer's disease. Heliyon 2024; 10:e39571. [PMID: 39498012 PMCID: PMC11532864 DOI: 10.1016/j.heliyon.2024.e39571] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2024] [Revised: 10/16/2024] [Accepted: 10/17/2024] [Indexed: 11/07/2024] Open
Abstract
Converging evidence from clinical and experimental studies suggest the potential significance of Polo-like kinase 2 (PLK2) in regulating the phosphorylation and toxicity of the Alzheimer's disease (AD)-related protein, amyloid precursor protein (APP). These findings have prompted various experimental trials aimed at inhibiting PLK2 kinase activity in different transgenic mouse models of AD. While positive impacts on cognitive decline were reported in these studies, the cellular effects remained controversial. In the present study, we sought to assess the cognitive and cellular consequences of chronic PLK2 inhibitor treatment in the APP/PS1 transgenic mouse model of AD. First, we confirmed that inhibiting PLK2 prevented cognitive decline in a sex-dependent manner, particularly by enhancing working memory in male APP/PS1 mice. Surprisingly, cellular analysis revealed that treatment with PLK2 inhibitor increased the load of amyloid plaques and elevated levels of soluble amyloid β (Aβ) 40 and Aβ42 in the cortex, as well as insoluble Aβ42 in the hippocampus of female mice, without affecting APP pathology in males. These results underscore the potential of PLK2 inhibition to mitigate cognitive symptoms in males. However, paradoxically, it intensifies amyloid pathology in females by enhancing APP amyloidogenic processing, creating a controversial aspect to its therapeutic impact. Overall, these data highlight the sex-dependent nature of the effects of PLK2 inhibition, which may also be influenced by the genetic background of the transgenic mouse model utilized.
Collapse
Affiliation(s)
- Laura Martínez-Drudis
- CHU de Québec-Université Laval Research Center, Neuroscience Axis, 2705 Boulevard Laurier, Quebec City, Canada
- Department of Molecular Medicine, Faculty of Medicine, Université Laval, Quebec City, Canada
| | - Morgan Bérard
- CHU de Québec-Université Laval Research Center, Neuroscience Axis, 2705 Boulevard Laurier, Quebec City, Canada
- Department of Molecular Medicine, Faculty of Medicine, Université Laval, Quebec City, Canada
| | - Dylan Musiol
- CHU de Québec-Université Laval Research Center, Neuroscience Axis, 2705 Boulevard Laurier, Quebec City, Canada
- Department of Molecular Medicine, Faculty of Medicine, Université Laval, Quebec City, Canada
| | - Serge Rivest
- CHU de Québec-Université Laval Research Center, Neuroscience Axis, 2705 Boulevard Laurier, Quebec City, Canada
- Department of Molecular Medicine, Faculty of Medicine, Université Laval, Quebec City, Canada
| | - Abid Oueslati
- CHU de Québec-Université Laval Research Center, Neuroscience Axis, 2705 Boulevard Laurier, Quebec City, Canada
- Department of Molecular Medicine, Faculty of Medicine, Université Laval, Quebec City, Canada
| |
Collapse
|
23
|
Wang M, Dinarvand D, Chan CTY, Bragin A, Li L. Photobiomodulation as a Potential Treatment for Alzheimer's Disease: A Review Paper. Brain Sci 2024; 14:1064. [PMID: 39595827 PMCID: PMC11591719 DOI: 10.3390/brainsci14111064] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2024] [Revised: 10/10/2024] [Accepted: 10/23/2024] [Indexed: 11/28/2024] Open
Abstract
BACKGROUND Alzheimer's disease (AD), the most prevalent form of dementia, is a leading neurodegenerative disorder currently affecting approximately 55 million individuals globally, a number projected to escalate to 139 million by 2050. Despite extensive research spanning several decades, the cure for AD remains at a developing stage. The only existing therapeutic options are limited to symptom management, and are often accompanied by adverse side effects. The pathological features of AD, including the accumulation of beta-amyloid plaques and tau protein tangles, result in progressive neuronal death, synaptic loss, and brain atrophy, leading to significant cognitive decline and a marked reduction in quality of life. OBJECTIVE In light of the shortcomings of existing pharmacological interventions, this review explores the potential of photobiomodulation (PBM) as a non-invasive therapeutic option for AD. PBM employs infrared light to facilitate cellular repair and regeneration, focusing on addressing the disease's underlying biomechanical mechanisms. METHOD This paper presents a comprehensive introduction to the mechanisms of PBM and an analysis of preclinical studies evaluating its impact on cellular health, cognitive function, and disease progression in AD.The review provides a comprehensive overview of the various wavelengths and application methods, evaluating their efficacy in mitigating AD-related symptoms. CONCLUSIONS The findings underscore the significant potential of PBM as a safe and effective alternative treatment for Alzheimer's disease, emphasizing the necessity for further research and clinical trials to establish its therapeutic efficacy conclusively.
Collapse
Affiliation(s)
- Miaomiao Wang
- Department of Biomedical Engineering, University of North Texas, Denton, TX 76207, USA; (M.W.); (D.D.); (C.T.Y.C.)
| | - Deeba Dinarvand
- Department of Biomedical Engineering, University of North Texas, Denton, TX 76207, USA; (M.W.); (D.D.); (C.T.Y.C.)
| | - Clement T. Y. Chan
- Department of Biomedical Engineering, University of North Texas, Denton, TX 76207, USA; (M.W.); (D.D.); (C.T.Y.C.)
| | - Anatol Bragin
- Department of Neurology, University of California Los Angeles, Los Angeles, CA 90095, USA;
- Brain Research Institute, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, USA
| | - Lin Li
- Department of Biomedical Engineering, University of North Texas, Denton, TX 76207, USA; (M.W.); (D.D.); (C.T.Y.C.)
- Department of Neurology, University of California Los Angeles, Los Angeles, CA 90095, USA;
| |
Collapse
|
24
|
Kaltschmidt B, Czaniera NJ, Schulten W, Kaltschmidt C. NF-κB in Alzheimer's Disease: Friend or Foe? Opposite Functions in Neurons and Glial Cells. Int J Mol Sci 2024; 25:11353. [PMID: 39518906 PMCID: PMC11545113 DOI: 10.3390/ijms252111353] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2024] [Revised: 10/11/2024] [Accepted: 10/18/2024] [Indexed: 11/16/2024] Open
Abstract
Alzheimer's disease (AD) is a devasting neurodegenerative disease afflicting mainly glutamatergic neurons together with a massive neuroinflammation mediated by the transcription factor NF-κB. A 65%-plus increase in Alzheimer's patients by 2050 might be a major threat to society. Hallmarks of AD are neurofibrillary tangles (NFTs) composed of hyperphosphorylated tau and amyloid beta (Aβ) plaques. Here, we review the potential involvement of transcription factor NF-κB by hereditary mutations of the tumor necrosis factor pathway in AD patients. One of the greatest genetic risk factors is APOE4. Recently, it was shown that the APOE4 allele functions as a null allele in human astrocytes not repressing NF-κB anymore. Moreover, NF-κB seems to be involved in the repair of DNA double-strand breaks during healthy learning and memory, a function blunted in AD. NF-κB could be a friend to healthy neurons by repressing apoptosis and necroptosis. But a loss of neuronal NF-κB and activation of glial NF-κB in AD makes it a foe of neuronal survival. Hopeful therapies include TNFR2 receptor bodies relieving the activation of glial NF-κB by TNFα.
Collapse
Affiliation(s)
- Barbara Kaltschmidt
- Molecular Neurobiology, University of Bielefeld, 33615 Bielefeld, Germany
- Department of Cell Biology, University of Bielefeld, 33615 Bielefeld, Germany; (N.J.C.); (W.S.); (C.K.)
- Forschungsverbund BioMedizin Bielefeld, Ostwestfalen-Lippe (OWL) (FBMB E.V.), 33615 Bielefeld, Germany
| | - Nele Johanne Czaniera
- Department of Cell Biology, University of Bielefeld, 33615 Bielefeld, Germany; (N.J.C.); (W.S.); (C.K.)
| | - Wiebke Schulten
- Department of Cell Biology, University of Bielefeld, 33615 Bielefeld, Germany; (N.J.C.); (W.S.); (C.K.)
| | - Christian Kaltschmidt
- Department of Cell Biology, University of Bielefeld, 33615 Bielefeld, Germany; (N.J.C.); (W.S.); (C.K.)
- Forschungsverbund BioMedizin Bielefeld, Ostwestfalen-Lippe (OWL) (FBMB E.V.), 33615 Bielefeld, Germany
| |
Collapse
|
25
|
Fanlo-Ucar H, Picón-Pagès P, Herrera-Fernández V, ILL-Raga G, Muñoz FJ. The Dual Role of Amyloid Beta-Peptide in Oxidative Stress and Inflammation: Unveiling Their Connections in Alzheimer's Disease Etiopathology. Antioxidants (Basel) 2024; 13:1208. [PMID: 39456461 PMCID: PMC11505517 DOI: 10.3390/antiox13101208] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2024] [Revised: 10/03/2024] [Accepted: 10/05/2024] [Indexed: 10/28/2024] Open
Abstract
Alzheimer's disease (AD) is a progressive neurodegenerative disease, and it is currently the seventh leading cause of death worldwide. It is characterized by the extracellular aggregation of the amyloid β-peptide (Aβ) into oligomers and fibrils that cause synaptotoxicity and neuronal death. Aβ exhibits a dual role in promoting oxidative stress and inflammation. This review aims to unravel the intricate connection between these processes and their contribution to AD progression. The review delves into oxidative stress in AD, focusing on the involvement of metals, mitochondrial dysfunction, and biomolecule oxidation. The distinct yet overlapping concept of nitro-oxidative stress is also discussed, detailing the roles of nitric oxide, mitochondrial perturbations, and their cumulative impact on Aβ production and neurotoxicity. Inflammation is examined through astroglia and microglia function, elucidating their response to Aβ and their contribution to oxidative stress within the AD brain. The blood-brain barrier and oligodendrocytes are also considered in the context of AD pathophysiology. We also review current diagnostic methodologies and emerging therapeutic strategies aimed at mitigating oxidative stress and inflammation, thereby offering potential treatments for halting or slowing AD progression. This comprehensive synthesis underscores the pivotal role of Aβ in bridging oxidative stress and inflammation, advancing our understanding of AD and informing future research and treatment paradigms.
Collapse
Affiliation(s)
- Hugo Fanlo-Ucar
- Laboratory of Molecular Physiology, Department of Medicine and Life Sciences, Faculty of Medicine and Life Sciences, Universitat Pompeu Fabra, 08003 Barcelona, Spain; (H.F.-U.); (P.P.-P.); (V.H.-F.); (G.I.-R.)
| | - Pol Picón-Pagès
- Laboratory of Molecular Physiology, Department of Medicine and Life Sciences, Faculty of Medicine and Life Sciences, Universitat Pompeu Fabra, 08003 Barcelona, Spain; (H.F.-U.); (P.P.-P.); (V.H.-F.); (G.I.-R.)
- Laboratory of Molecular and Cellular Neurobiotechnology, Institute of Bioengineering of Catalonia (IBEC), 08028 Barcelona, Spain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), 08028 Barcelona, Spain
| | - Víctor Herrera-Fernández
- Laboratory of Molecular Physiology, Department of Medicine and Life Sciences, Faculty of Medicine and Life Sciences, Universitat Pompeu Fabra, 08003 Barcelona, Spain; (H.F.-U.); (P.P.-P.); (V.H.-F.); (G.I.-R.)
| | - Gerard ILL-Raga
- Laboratory of Molecular Physiology, Department of Medicine and Life Sciences, Faculty of Medicine and Life Sciences, Universitat Pompeu Fabra, 08003 Barcelona, Spain; (H.F.-U.); (P.P.-P.); (V.H.-F.); (G.I.-R.)
| | - Francisco J. Muñoz
- Laboratory of Molecular Physiology, Department of Medicine and Life Sciences, Faculty of Medicine and Life Sciences, Universitat Pompeu Fabra, 08003 Barcelona, Spain; (H.F.-U.); (P.P.-P.); (V.H.-F.); (G.I.-R.)
| |
Collapse
|
26
|
Wang J, Fourriere L, Gleeson PA. Advances in the cell biology of the trafficking and processing of amyloid precursor protein: impact of familial Alzheimer's disease mutations. Biochem J 2024; 481:1297-1325. [PMID: 39302110 PMCID: PMC11555708 DOI: 10.1042/bcj20240056] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2024] [Revised: 09/02/2024] [Accepted: 09/05/2024] [Indexed: 09/22/2024]
Abstract
The production of neurotoxic amyloid-β peptides (Aβ) is central to the initiation and progression of Alzheimer's disease (AD) and involves sequential cleavage of the amyloid precursor protein (APP) by β- and γ-secretases. APP and the secretases are transmembrane proteins and their co-localisation in the same membrane-bound sub-compartment is necessary for APP cleavage. The intracellular trafficking of APP and the β-secretase, BACE1, is critical in regulating APP processing and Aβ production and has been studied in several cellular systems. Here, we summarise the intracellular distribution and transport of APP and its secretases, and the intracellular location for APP cleavage in non-polarised cells and neuronal models. In addition, we review recent advances on the potential impact of familial AD mutations on APP trafficking and processing. This is critical information in understanding the molecular mechanisms of AD progression and in supporting the development of novel strategies for clinical treatment.
Collapse
Affiliation(s)
- Jingqi Wang
- Department of Biochemistry and Pharmacology, Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, Melbourne, Victoria 3010, Australia
| | - Lou Fourriere
- Department of Biochemistry and Pharmacology, Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, Melbourne, Victoria 3010, Australia
| | - Paul A. Gleeson
- Department of Biochemistry and Pharmacology, Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, Melbourne, Victoria 3010, Australia
| |
Collapse
|
27
|
Kurakin S, Ivankov O, Dushanov E, Murugova T, Ermakova E, Efimov S, Mukhametzyanov T, Smerdova S, Klochkov V, Kuklin A, Kučerka N. Calcium ions do not influence the Aβ(25-35) triggered morphological changes of lipid membranes. Biophys Chem 2024; 313:107292. [PMID: 39018778 DOI: 10.1016/j.bpc.2024.107292] [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: 03/06/2024] [Revised: 07/04/2024] [Accepted: 07/09/2024] [Indexed: 07/19/2024]
Abstract
We have studied the effect of calcium ions (Ca2+) at various concentrations on the structure of lipid vesicles in the presence of amyloid-beta peptide Aβ(25-35). In particular, we have investigated the influence of calcium ions on the formation of recently documented bicelle-like structures (BLSs) emerged as a result of Aβ(25-35) triggered membrane disintegration. First, we have shown by using small-angle X-ray and neutron scattering that peptide molecules rigidify the lipid bilayer of gel phase DPPC unilamellar vesicles (ULVs), while addition of the calcium ions to the system hinders this effect of Aβ(25-35). Secondly, the Aβ(25-35) demonstrates a critical peptide concentration at which the BLSs reorganize from ULVs due to heating and cooling the samples through the lipid main phase transition temperature (Tm). However, addition of calcium ions does not affect noticeably the Aβ-induced formation of BLSs and their structural parameters, though the changes in peptide's secondary structure, e.g. the increased α-helix fraction, has been registered by circular dichroism spectroscopy. Finally, according to 31P nuclear magnetic resonance (NMR) measurements, calcium ions do not affect the lipid-peptide arrangement in BLSs and their ability to align in the magnetic field of NMR spectrometer. The influences of various concentrations of calcium ions on the lipid-peptide interactions may prove biologically important because their local concentrations vary widely in in-vivo conditions. In the present work, calcium ions were investigated as a possible tool aimed at regulating the lipid-peptide interactions that demonstrated the disruptive effect of Aβ(25-35) on lipid membranes.
Collapse
Affiliation(s)
- Sergei Kurakin
- Frank Laboratory of Neutron Physics, Joint Institute for Nuclear Research, Joliot-Curie 6, Dubna, Moscow Region 141980, Russia; Institute of Physics, Kazan Federal University, Kremlevskaya 18, Kazan 420008, Russia.
| | - Oleksandr Ivankov
- Frank Laboratory of Neutron Physics, Joint Institute for Nuclear Research, Joliot-Curie 6, Dubna, Moscow Region 141980, Russia
| | - Ermuhammad Dushanov
- Laboratory of Radiation Biology, Joint Institute for Nuclear Research, Joliot-Curie 6, Dubna, Moscow Region 141980, Russia; Department of Biophysics, Dubna State University, Universitetskaya 19, Dubna, Moscow Region 141982, Russia
| | - Tatiana Murugova
- Frank Laboratory of Neutron Physics, Joint Institute for Nuclear Research, Joliot-Curie 6, Dubna, Moscow Region 141980, Russia
| | - Elena Ermakova
- Frank Laboratory of Neutron Physics, Joint Institute for Nuclear Research, Joliot-Curie 6, Dubna, Moscow Region 141980, Russia
| | - Sergey Efimov
- Institute of Physics, Kazan Federal University, Kremlevskaya 18, Kazan 420008, Russia
| | - Timur Mukhametzyanov
- Butlerov Chemistry Institute, Kazan Federal University, Kremlevskaya 18, Kazan 420008, Russia
| | - Svetlana Smerdova
- Kazan National Research Technological University, Karl Marx 68, Kazan 420015, Russia
| | - Vladimir Klochkov
- Institute of Physics, Kazan Federal University, Kremlevskaya 18, Kazan 420008, Russia
| | - Alexander Kuklin
- Frank Laboratory of Neutron Physics, Joint Institute for Nuclear Research, Joliot-Curie 6, Dubna, Moscow Region 141980, Russia; Moscow Institute of Physics and Technology, Instytutskiy Pereulok 9, Dolgoprudny, Moscow Region 141701, Russia
| | - Norbert Kučerka
- Frank Laboratory of Neutron Physics, Joint Institute for Nuclear Research, Joliot-Curie 6, Dubna, Moscow Region 141980, Russia; Department of Physical Chemistry of Drugs, Faculty of Pharmacy, Comenius University Bratislava, Odbojárov 10, Bratislava 832 32, Slovakia.
| |
Collapse
|
28
|
Türküner MS, Yazıcı A, Özcan F. SIK2 Controls the Homeostatic Character of the POMC Secretome Acutely in Response to Pharmacological ER Stress Induction. Cells 2024; 13:1565. [PMID: 39329749 PMCID: PMC11430698 DOI: 10.3390/cells13181565] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2024] [Revised: 09/04/2024] [Accepted: 09/10/2024] [Indexed: 09/28/2024] Open
Abstract
The neuronal etiology of obesity is centered around a diet-induced inflammatory state in the arcuate nucleus of the hypothalamus, which impairs the functionality of pro-opiomelanocortine neurons (POMCs) responsible for whole-body energy homeostasis and feeding behavior. Intriguingly, systemic salt inducible kinase 2 (SIK2) knockout mice demonstrated reduced food intake and energy expenditure along with modestly dysregulated metabolic parameters, suggesting a causal link between the absence of SIK2 activity in POMCs and the observed phenotype. To test this hypothesis, we conducted a comparative secretomics study from POMC neurons following pharmacologically induced endoplasmic reticulum (ER) stress induction, a hallmark of metabolic inflammation and POMC dysregulation in diet-induced obese (DIO) mice. Our data provide significant in vitro evidence for the POMC-specific SIK2 activity in controlling energy metabolism and feeding in DIO mice by regulating the nature of the related POMC secretome. Our data also suggest that under physiological stress conditions, SIK2 may act as a gatekeeper for the secreted inflammatory factors and signaling molecules critical for cellular survival and energy homeostasis. On the other hand, in the absence of SIK2, the gate opens, leading to a surge of inflammatory cytokines and apoptotic cues concomitant with the dysregulation of POMC neurons.
Collapse
Affiliation(s)
- Mehmet Soner Türküner
- Department of Molecular Biology and Genetics, Graduate School of Natural and Applied Sciences, Gebze Technical University (GTU), Gebze, Kocaeli 41400, Turkey; (M.S.T.); (A.Y.)
- Cellular Proteomics Laboratory, Gebze Technical University—Central Research Laboratory, Application and Research Center Laboratory (GTU-MAR), Gebze, Kocaeli 41400, Turkey
| | - Ayşe Yazıcı
- Department of Molecular Biology and Genetics, Graduate School of Natural and Applied Sciences, Gebze Technical University (GTU), Gebze, Kocaeli 41400, Turkey; (M.S.T.); (A.Y.)
- Cellular Proteomics Laboratory, Gebze Technical University—Central Research Laboratory, Application and Research Center Laboratory (GTU-MAR), Gebze, Kocaeli 41400, Turkey
| | - Ferruh Özcan
- Department of Molecular Biology and Genetics, Graduate School of Natural and Applied Sciences, Gebze Technical University (GTU), Gebze, Kocaeli 41400, Turkey; (M.S.T.); (A.Y.)
- Cellular Proteomics Laboratory, Gebze Technical University—Central Research Laboratory, Application and Research Center Laboratory (GTU-MAR), Gebze, Kocaeli 41400, Turkey
| |
Collapse
|
29
|
Volloch V, Rits-Volloch S. Quintessential Synergy: Concurrent Transient Administration of Integrated Stress Response Inhibitors and BACE1 and/or BACE2 Activators as the Optimal Therapeutic Strategy for Alzheimer's Disease. Int J Mol Sci 2024; 25:9913. [PMID: 39337400 PMCID: PMC11432332 DOI: 10.3390/ijms25189913] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2024] [Revised: 09/01/2024] [Accepted: 09/03/2024] [Indexed: 09/30/2024] Open
Abstract
The present study analyzes two potential therapeutic approaches for Alzheimer's disease (AD). One is the suppression of the neuronal integrated stress response (ISR). Another is the targeted degradation of intraneuronal amyloid-beta (iAβ) via the activation of BACE1 (Beta-site Aβ-protein-precursor Cleaving Enzyme) and/or BACE2. Both approaches are rational. Both are promising. Both have substantial intrinsic limitations. However, when combined in a carefully orchestrated manner into a composite therapy they display a prototypical synergy and constitute the apparently optimal, potentially most effective therapeutic strategy for AD.
Collapse
Affiliation(s)
- Vladimir Volloch
- Department of Developmental Biology, Harvard School of Dental Medicine, Boston, MA 02115, USA
| | - Sophia Rits-Volloch
- Division of Molecular Medicine, Children’s Hospital, Boston, MA 02115, USA
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA 02115, USA
| |
Collapse
|
30
|
Muhammedkutty FNK, Zhou HX. Membrane-assisted Aβ40 aggregation pathways. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.05.611426. [PMID: 39282376 PMCID: PMC11398458 DOI: 10.1101/2024.09.05.611426] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 10/21/2024]
Abstract
Alzheimer's disease (AD) is caused by the assembly of amyloid-beta (Aβ) peptides into oligomers and fibrils. Endogenous Aβ aggregation may be assisted by cell membranes, which can accelerate the nucleation step enormously, but knowledge of membrane-assisted aggregation is still very limited. Here we used extensive MD simulations to structurally and energetically characterize key intermediates along the membrane-assisted aggregation pathways of Aβ40. Reinforcing experimental observations, the simulations reveal unique roles of GM1 ganglioside and cholesterol in stabilizing membrane-embedded β-sheets and of Y10 and K28 in the ordered release of a small oligomeric seed into solution. The same seed leads to either an open-shaped or R-shaped fibril, with significant stabilization provided by inter- or intra-subunit interfaces between a straight β-sheet (residues Q15-D23) and a bent β-sheet (residues A30-V36). This work presents the first comprehensive picture of membrane-assisted aggregation of Aβ40, with broad implications for developing AD therapies and rationalizing disease-specific polymorphisms of amyloidogenic proteins.
Collapse
Affiliation(s)
| | - Huan-Xiang Zhou
- Department of Chemistry, University of Illinois Chicago, Chicago, IL, USA
- Department of Physics, University of Illinois Chicago, Chicago, IL, USA
| |
Collapse
|
31
|
Behl C. In 2024, the amyloid-cascade-hypothesis still remains a working hypothesis, no less but certainly no more. Front Aging Neurosci 2024; 16:1459224. [PMID: 39295642 PMCID: PMC11408168 DOI: 10.3389/fnagi.2024.1459224] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2024] [Accepted: 08/27/2024] [Indexed: 09/21/2024] Open
Abstract
The amyloid-cascade-hypothesis of the pathogenesis of Alzheimer's disease (AD) was introduced 32 years ago, in 1992. From early on, this clear and straight forward hypothesis received a lot of attention, but also a lot of substantial criticism. Foremost, there have always been massive doubts that a complex age-associated disorder of the most intricate organ of the human body, the brain, can be explained by a linear, one-dimensional cause-and-effect model. The amyloid-cascade defines the generation, aggregation, and deposition of the amyloid beta peptide as the central pathogenic mechanism in AD, as the ultimate trigger of the disease, and, consequently, as the key pharmacological target. Certainly, the original 1992 version of this hypothesis has been refined by various means, and the 'formulating fathers' followed up with a few reappraisals and partly very open reflections in 2002, 2006, 2009, and 2016. However, up until today, for the supporters of this hypothesis, the central and initial steps of the cascade are believed to be driven by amyloid beta-even if now displayed somewhat more elaborate. In light of the recently published clinical results achieved with anti-amyloid antibodies, the controversy in the field about (1) the clinical meaningfulness of this approach, (2) the significance of clearance of the amyloid beta peptide, and last but not least (3) the relevance of the amyloid-cascade-hypothesis is gaining momentum. This review addresses the interesting manifestation of the amyloid-cascade-hypothesis as well as its ups and downs over the decades.
Collapse
Affiliation(s)
- Christian Behl
- The-Autophagy-Lab, Institute of Pathobiochemistry, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| |
Collapse
|
32
|
Baek Y, Lee M. Exploring the complexity of amyloid-beta fibrils: structural polymorphisms and molecular interactions. Biochem Soc Trans 2024; 52:1631-1646. [PMID: 39034652 DOI: 10.1042/bst20230854] [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: 04/29/2024] [Revised: 07/09/2024] [Accepted: 07/12/2024] [Indexed: 07/23/2024]
Abstract
The aggregation of amyloid-beta (Aβ) peptides into cross-β structures forms a variety of distinct fibril conformations, potentially correlating with variations in neurodegenerative disease progression. Recent advances in techniques such as X-ray crystallography, solid-state NMR, and cryo-electron microscopy have enabled the development of high-resolution molecular structures of these polymorphic amyloid fibrils, which are either grown in vitro or isolated from human and transgenic mouse brain tissues. This article reviews our current understanding of the structural polymorphisms in amyloid fibrils formed by Aβ40 and Aβ42, as well as disease-associated mutants of Aβ peptides. The aim is to enhance our understanding of various molecular interactions, including hydrophobic and ionic interactions, within and among cross-β structures.
Collapse
Affiliation(s)
- Yoongyeong Baek
- Department of Chemistry, Drexel University, Philadelphia, PA 19104, U.S.A
| | - Myungwoon Lee
- Department of Chemistry, Drexel University, Philadelphia, PA 19104, U.S.A
| |
Collapse
|
33
|
Sampani K, Ness S, Tuz-Zahra F, Aytan N, Spurlock EE, Alluri S, Chen X, Siegel NH, Alosco ML, Xia W, Tripodis Y, Stein TD, Subramanian ML. Neurodegenerative biomarkers in different chambers of the eye relative to plasma: an agreement validation study. Alzheimers Res Ther 2024; 16:192. [PMID: 39187891 PMCID: PMC11346268 DOI: 10.1186/s13195-024-01556-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2024] [Accepted: 08/11/2024] [Indexed: 08/28/2024]
Abstract
BACKGROUND Protein biomarkers have been broadly investigated in cerebrospinal fluid and blood for the detection of neurodegenerative diseases, yet a clinically useful diagnostic test to detect early, pre-symptomatic Alzheimer's disease (AD) remains elusive. We conducted this study to quantify Aβ40, Aβ42, total Tau (t-Tau), hyperphosphorylated Tau (ptau181), glial fibrillary acidic protein (GFAP) and neurofilament light chain (NfL) in eye fluids relative to blood. METHODS In this cross-sectional study we collected vitreous humor, aqueous humor, tear fluid and plasma in patients undergoing surgery for eye disease. All six biomarkers were quantitatively measured by digital immunoassay. Spearman and Bland-Altman correlation analyses were performed to assess the agreement of levels between ocular fluids and plasma. RESULTS Seventy-nine adults underwent pars-plana vitrectomy in at least one eye. Of the 79, there were 77 vitreous, 67 blood, 56 tear fluid, and 51 aqueous samples. All six biomarkers were quantified in each bio-sample, except GFAP and NfL in tear fluid due to low sample volume. All six biomarkers were elevated in vitreous humor compared to plasma samples. T-Tau, ptau181, GFAP and NfL were higher in aqueous than in plasma, and t-Tau and ptau181 concentrations were higher in tear fluid than in plasma. Significant correlations were found between Aβ40 in plasma and tears (r = 0.5; p = 0.019), t-Tau in plasma and vitreous (r = 0.4; p = 0.004), NfL in plasma and vitreous (r = 0.3; p = 0.006) and plasma and aqueous (r = 0.5; p = 0.004). No significant associations were found for Aβ42, ptau181 and GFAP among ocular fluids relative to plasma. Bland-Altman analysis showed aqueous humor had the closest agreement to plasma across all biomarkers. Biomarker levels in ocular fluids revealed statistically significant associations between vitreous and aqueous for t-Tau (r = 0.5; p = 0.001), GFAP (r = 0.6; p < 0.001) and NfL (r = 0.7; p < 0.001). CONCLUSION AD biomarkers are detectable in greater quantities in eye fluids than in plasma and show correlations with levels in plasma. Future studies are needed to assess the utility of ocular fluid biomarkers as diagnostic and prognostic markers for AD, especially in those at risk with eye disease.
Collapse
Affiliation(s)
- Konstantina Sampani
- Beetham Eye Institute, Joslin Diabetes Center, Boston, MA, 02115, USA
- Department of Medicine, Harvard Medical School, Boston, MA, 02115, USA
| | - Steven Ness
- Department of Ophthalmology, Boston Medical Center, Boston, MA, 02118, USA
- Department of Ophthalmology, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, USA
| | - Fatima Tuz-Zahra
- Department of Biostatistics, Boston University School of Public Health, Boston, MA, USA
| | - Nurgul Aytan
- Boston University Alzheimer's Disease Research Center and CTE Center, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
| | - Elizabeth E Spurlock
- Boston University Alzheimer's Disease Research Center and CTE Center, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
| | - Sreevardhan Alluri
- Department of Ophthalmology, Boston Medical Center, Boston, MA, 02118, USA
| | - Xuejing Chen
- Department of Ophthalmology, Boston Medical Center, Boston, MA, 02118, USA
- Department of Ophthalmology, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, USA
| | - Nicole H Siegel
- Department of Ophthalmology, Boston Medical Center, Boston, MA, 02118, USA
- Department of Ophthalmology, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, USA
| | - Michael L Alosco
- Boston University Alzheimer's Disease Research Center and CTE Center, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
- Department of Psychology, University of Arizona, Tucson, AZ, USA
| | - Weiming Xia
- Department of Pharmacology and Experimental Therapeutics, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
- Geriatric Research Education and Clinical Center, Bedford Veterans Affairs Medical Center, Bedford, MA, USA
| | - Yorghos Tripodis
- Department of Biostatistics, Boston University School of Public Health, Boston, MA, USA
- Boston University Alzheimer's Disease Research Center and CTE Center, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
| | - Thor D Stein
- Boston University Alzheimer's Disease Research Center and CTE Center, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA.
- Department of Pathology and Laboratory Medicine, Boston Medical Center, Boston University School of Medicine, Boston, MA, USA.
- Department of Veterans Affairs Medical Center, VA Boston Healthcare System, Boston, MA, USA.
- Department of Veterans Affairs Medical Center, VA Bedford Healthcare System, Bedford, MA, USA.
| | - Manju L Subramanian
- Department of Ophthalmology, Boston Medical Center, Boston, MA, 02118, USA.
- Department of Ophthalmology, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, USA.
| |
Collapse
|
34
|
Paterno G, Moore BD, Bell BM, Gorion KMM, Ran Y, Prokop S, Golde TE, Giasson BI. Novel Monoclonal Antibody Specific toward Amyloid-β Binds to a Unique Epitope within the N-Terminal Region. Antibodies (Basel) 2024; 13:68. [PMID: 39189239 PMCID: PMC11348109 DOI: 10.3390/antib13030068] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2024] [Revised: 08/04/2024] [Accepted: 08/06/2024] [Indexed: 08/28/2024] Open
Abstract
Amyloid-β (Aβ) deposition throughout the neuroaxis is a classical hallmark of several neurodegenerative diseases, most notably Alzheimer's disease (AD). Aβ peptides of varied length and diverse structural conformations are deposited within the parenchyma and vasculature in the brains of individuals with AD. Neuropathologically, Aβ pathology can be assessed using antibodies to label and characterize their features, which in turn leads to a more extensive understanding of the pathological process. In the present study, we generated a novel monoclonal antibody, which we found to be specific for the N-terminal region of Aβ. This antibody reacted to amyloid precursor protein expressed in cultured cells and labels Aβ plaques and cerebral amyloid angiopathy in brain tissue from a mouse model of amyloidosis as well as post-mortem brain tissue from patients diagnosed with AD. This highly specific novel antibody will serve as a unique tool for future studies investigating Aβ deposition in novel mouse models and cross-sectional studies using post-mortem human tissue.
Collapse
Affiliation(s)
- Giavanna Paterno
- Department of Neuroscience, College of Medicine, University of Florida, Gainesville, FL 32610, USA; (G.P.); (B.M.B.); (K.-M.M.G.)
- Center for Translational Research in Neurodegenerative Disease, College of Medicine, University of Florida, Gainesville, FL 32610, USA;
| | - Brenda D. Moore
- Department of Pharmacology and Chemical Biology, Emory University School of Medicine, Atlanta, GA 30322, USA; (B.D.M.); (Y.R.); (T.E.G.)
- Center for Neurodegenerative Disease, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Brach M. Bell
- Department of Neuroscience, College of Medicine, University of Florida, Gainesville, FL 32610, USA; (G.P.); (B.M.B.); (K.-M.M.G.)
- Center for Translational Research in Neurodegenerative Disease, College of Medicine, University of Florida, Gainesville, FL 32610, USA;
| | - Kimberly-Marie M. Gorion
- Department of Neuroscience, College of Medicine, University of Florida, Gainesville, FL 32610, USA; (G.P.); (B.M.B.); (K.-M.M.G.)
- Center for Translational Research in Neurodegenerative Disease, College of Medicine, University of Florida, Gainesville, FL 32610, USA;
| | - Yong Ran
- Department of Pharmacology and Chemical Biology, Emory University School of Medicine, Atlanta, GA 30322, USA; (B.D.M.); (Y.R.); (T.E.G.)
- Center for Neurodegenerative Disease, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Stefan Prokop
- Center for Translational Research in Neurodegenerative Disease, College of Medicine, University of Florida, Gainesville, FL 32610, USA;
- McKnight Brain Institute, College of Medicine, University of Florida, Gainesville, FL 32610, USA
- Department of Pathology, College of Medicine, University of Florida, Gainesville, FL 32610, USA
| | - Todd E. Golde
- Department of Pharmacology and Chemical Biology, Emory University School of Medicine, Atlanta, GA 30322, USA; (B.D.M.); (Y.R.); (T.E.G.)
- Center for Neurodegenerative Disease, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Benoit I. Giasson
- Department of Neuroscience, College of Medicine, University of Florida, Gainesville, FL 32610, USA; (G.P.); (B.M.B.); (K.-M.M.G.)
- Center for Translational Research in Neurodegenerative Disease, College of Medicine, University of Florida, Gainesville, FL 32610, USA;
- McKnight Brain Institute, College of Medicine, University of Florida, Gainesville, FL 32610, USA
| |
Collapse
|
35
|
Kramer M, Hoang TH, Yang H, Shchyglo O, Böge J, Neubacher U, Colitti-Klausnitzer J, Manahan-Vaughan D. Intracerebral inoculation of healthy non-transgenic rats with a single aliquot of oligomeric amyloid-β (1-42) profoundly and progressively alters brain function throughout life. Front Aging Neurosci 2024; 16:1397901. [PMID: 39156737 PMCID: PMC11327071 DOI: 10.3389/fnagi.2024.1397901] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2024] [Accepted: 07/09/2024] [Indexed: 08/20/2024] Open
Abstract
One of the puzzling aspects of sporadic Alzheimer's disease (AD) is how it commences. Changes in one key brain peptide, amyloid-beta (Aβ), accompany disease progression, but whether this comprises a trigger or a consequence of AD is still a topic of debate. It is clear however that the cerebral presence of oligomeric Aβ (1-42) is a key factor in early AD-pathogenesis. Furthermore, treatment of rodent brains with oligomeric Aβ (1-42) either in vitro or in vivo, acutely impairs hippocampal synaptic plasticity, creating a link between Aβ-pathology and learning impairments. Here, we show that a once-off inoculation of the brains of healthy adult rats with oligomeric Aβ (1-42) exerts debilitating effects on the long-term viability of the hippocampus, one of the primary targets of AD. Changes are progressive: months after treatment, synaptic plasticity, neuronal firing and spatial learning are impaired and expression of plasticity-related proteins are changed, in the absence of amyloid plaques. Early changes relate to activation of microglia, whereas later changes are associated with a reconstruction of astroglial morphology. These data suggest that a disruption of Aβ homeostasis may suffice to trigger an irreversible cascade, underlying progressive loss of hippocampal function, that parallels the early stages of AD.
Collapse
|
36
|
Lippi A, Krisko A. Protein aggregation: A detrimental symptom or an adaptation mechanism? J Neurochem 2024; 168:1426-1441. [PMID: 37694504 DOI: 10.1111/jnc.15955] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2023] [Revised: 08/21/2023] [Accepted: 08/22/2023] [Indexed: 09/12/2023]
Abstract
Protein quality control mechanisms oversee numerous aspects of protein lifetime. From the point of protein synthesis, protein homeostasis machineries take part in folding, solubilization, and/or degradation of impaired proteins. Some proteins follow an alternative path upon loss of their solubility, thus are secluded from the cytosol and form protein aggregates. Protein aggregates differ in their function and composition, rendering protein aggregation a complex phenomenon that continues to receive plenty of attention in the scientific and medical communities. Traditionally, protein aggregates have been associated with aging and a large spectrum of protein folding diseases, such as neurodegenerative diseases, type 2 diabetes, or cataract. However, a body of evidence suggests that they may act as an adaptive mechanism to overcome transient stressful conditions, serving as a sink for the removal of misfolded proteins from the cytosol or storage compartments for machineries required upon stress release. In this review, we present examples and evidence elaborating different possible roles of protein aggregation and discuss their potential roles in stress survival, aging, and disease, as well as possible anti-aggregation interventions.
Collapse
Affiliation(s)
- Alice Lippi
- Department of Experimental Neurodegeneration, University Medical Center Göttingen, Göttingen, Germany
| | - Anita Krisko
- Department of Experimental Neurodegeneration, University Medical Center Göttingen, Göttingen, Germany
| |
Collapse
|
37
|
Petralla S, Panayotova M, Franchina E, Fricker G, Puris E. Low-Density Lipoprotein Receptor-Related Protein 1 as a Potential Therapeutic Target in Alzheimer's Disease. Pharmaceutics 2024; 16:948. [PMID: 39065645 PMCID: PMC11279518 DOI: 10.3390/pharmaceutics16070948] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2024] [Revised: 07/15/2024] [Accepted: 07/16/2024] [Indexed: 07/28/2024] Open
Abstract
Alzheimer's disease (AD) is a progressive neurodegenerative disease impacting the lives of millions of people worldwide. The formation of amyloid β (Aβ) plagues in the brain is the main pathological hallmark of AD. The Aβ deposits are formed due to the imbalance between the production and Aβ clearance in the brain and across the blood-brain barrier (BBB). In this respect, low-density lipoprotein receptor-related protein 1 (LRP1) plays a significant role by mediating both brain Aβ production and clearance. Due to its important role in AD pathogenesis, LRP1 is considered an attractive drug target for AD therapies. In the present review, we summarize the current knowledge about the role of LRP1 in AD pathogenesis as well as recent findings on changes in LRP1 expression and function in AD. Finally, we discuss the advances in utilizing LRP1 as a drug target for AD treatments as well as future perspectives on LRP1 research.
Collapse
Affiliation(s)
| | | | | | | | - Elena Puris
- Institute of Pharmacy and Molecular Biotechnology, Ruprecht-Karls-University, Im Neuenheimer Feld 329, 69120 Heidelberg, Germany; (S.P.); (M.P.); (E.F.); (G.F.)
| |
Collapse
|
38
|
Martá-Ariza M, Leitner DF, Kanshin E, Suazo J, Pedrosa AG, Thierry M, Lee EB, Devinsky O, Drummond E, Fortea J, Lleó A, Ueberheide B, Wisniewski T. Comparison of the Amyloid Plaque Proteome in Down Syndrome, Early-Onset Alzheimer's Disease and Late-Onset Alzheimer's Disease. RESEARCH SQUARE 2024:rs.3.rs-4469045. [PMID: 39070643 PMCID: PMC11275979 DOI: 10.21203/rs.3.rs-4469045/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/30/2024]
Abstract
Background Down syndrome (DS) is strongly associated with Alzheimer's disease (AD), attributable to APP overexpression. DS exhibits Amyloid-β (Aβ) and Tau pathology similar to early-onset AD (EOAD) and late-onset AD (LOAD). The study aimed to evaluate the Aβ plaque proteome of DS, EOAD and LOAD. Methods Using unbiased localized proteomics, we analyzed amyloid plaques and adjacent plaque-devoid tissue ('non-plaque') from post-mortem paraffin-embedded tissues in four cohorts (n = 20/group): DS (59.8 ± 4.99 y/o), EOAD (63 ± 4.07 y/o), LOAD (82.1 ± 6.37 y/o) and controls (66.4 ± 13.04). We assessed functional associations using Gene Ontology (GO) enrichment and protein interaction networks. Results We identified differentially abundant Aβ plaque proteins vs. non-plaques (FDR < 5%, fold-change > 1.5) in DS (n = 132), EOAD (n = 192) and in LOAD (n = 128); there were 43 plaque-associated proteins shared between all groups. Positive correlations (p < 0.0001) were observed between plaque-associated proteins in DS and EOAD (R2 = 0.77), DS and LOAD (R2 = 0.73), and EOAD vs. LOAD (R2 = 0.67). Top Biological process (BP) GO terms (p < 0.0001) included lysosomal transport for DS, immune system regulation for EOAD, and lysosome organization for LOAD. Protein networks revealed a plaque enriched signature across all cohorts involving APP metabolism, immune response, and lysosomal functions. In DS, EOAD and LOAD non-plaque vs. control tissue, we identified 263, 269, and 301 differentially abundant proteins, including 65 altered non-plaque proteins across all cohorts. Differentially abundant non-plaque proteins in DS showed a significant (p < 0.0001) but weaker positive correlation with EOAD (R2 = 0.59) and LOAD (R2 = 0.33) compared to the stronger correlation between EOAD and LOAD (R2 = 0.79). The top BP GO term for all groups was chromatin remodeling (DS p = 0.0013, EOAD p = 5.79×10- 9, and LOAD p = 1.69×10- 10). Additional GO terms for DS included extracellular matrix (p = 0.0068), while EOAD and LOAD were associated with protein-DNA complexes and gene expression regulation (p < 0.0001). Conclusions We found strong similarities among the Aβ plaque proteomes in individuals with DS, EOAD and LOAD, and a robust association between the plaque proteomes and lysosomal and immune-related pathways. Further, non-plaque proteomes highlighted altered pathways related to chromatin structure and extracellular matrix (ECM), the latter particularly associated with DS. We identified novel Aβ plaque proteins, which may serve as biomarkers or therapeutic targets.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Edward B Lee
- University of Pennsylvania Perelman School of Medicine
| | | | | | - Juan Fortea
- Universitat Autònoma de Barcelona: Universitat Autonoma de Barcelona
| | - Alberto Lleó
- Universitat Autònoma de Barcelona: Universitat Autonoma de Barcelona
| | | | | |
Collapse
|
39
|
Żukowska J, Moss SJ, Subramanian V, Acharya KR. Molecular basis of selective amyloid-β degrading enzymes in Alzheimer's disease. FEBS J 2024; 291:2999-3029. [PMID: 37622248 DOI: 10.1111/febs.16939] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2023] [Revised: 07/31/2023] [Accepted: 08/22/2023] [Indexed: 08/26/2023]
Abstract
The accumulation of the small 42-residue long peptide amyloid-β (Aβ) has been proposed as a major trigger for the development of Alzheimer's disease (AD). Within the brain, the concentration of Aβ peptide is tightly controlled through production and clearance mechanisms. Substantial experimental evidence now shows that reduced levels of Aβ clearance are present in individuals living with AD. This accumulation of Aβ can lead to the formation of large aggregated amyloid plaques-one of two detectable hallmarks of the disease. Aβ-degrading enzymes (ADEs) are major players in the clearance of Aβ. Stimulating ADE activity or expression, in order to compensate for the decreased clearance in the AD phenotype, provides a promising therapeutic target. It has been reported in mice that upregulation of ADEs can reduce the levels of Aβ peptide and amyloid plaques-in some cases, this led to improved cognitive function. Among several known ADEs, neprilysin (NEP), endothelin-converting enzyme-1 (ECE-1), insulin degrading enzyme (IDE) and angiotensin-1 converting enzyme (ACE) from the zinc metalloprotease family have been identified as important. These ADEs have the capacity to digest soluble Aβ which, in turn, cannot form the toxic oligomeric species. While they are known for their amyloid degradation, they exhibit complexity through promiscuous nature and a broad range of substrates that they can degrade. This review highlights current structural and functional understanding of these key ADEs, giving some insight into the molecular interactions that leads to the hydrolysis of peptide substrates, the crucial tasks performed by them and the potential for therapeutic use in the future.
Collapse
|
40
|
Mallesh R, Khan J, Gharai PK, Arshi MU, Garg S, Gupta S, Ghosh S. Hydrophobic C-Terminal Peptide Analog Aβ 31-41 Protects the Neurons from Aβ-Induced Toxicity. ACS Chem Neurosci 2024; 15:2372-2385. [PMID: 38822790 DOI: 10.1021/acschemneuro.4c00032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/03/2024] Open
Abstract
Spontaneous aggregation of amyloid beta (Aβ) leads to the formation of neurotoxic senile plaque considered as the most crucial event in Alzheimer's disease (AD) progression. Inhibition or disruption of this deadly aggregate formation is one of the most efficient strategies for the development of potential therapeutics, and extensive research is in progress by various research groups. In this direction, the development of a peptide analogous to that of the native Aβ peptide is an attractive strategy. Based on this rationale, β-sheet breakers were developed from the Aβ central hydrophobic core. These peptide derivatives will bind to the full length of the parent Aβ and interfere in self-recognition, thereby preventing the folding of the Aβ peptide into cross β-sheet neurotoxic aggregates. However, this approach is effective in the inhibition of fibrillar aggregation, but this strategy is ineffective in the Aβ neurotoxic oligomer formation. Therefore, an alternative and efficient approach is to use the Aβ peptide analogous to the C-terminal region, which arbitrates fibrillation and oligomerization. Herein, we developed the Aβ C-terminal fragment (ACT-1 to ACT-7) for inhibition of oligomerization as well as fibrillar aggregation. Screening of these seven peptides resulted in an efficient anti-Aβ peptide aggregative agent (ACT-7), which was evaluated by the ThT assay peptide. The ThT assay reveals complete inhibition and showed significant neuroprotection of PC-12-derived neurons from Aβ-induced toxicity and reduced cell apoptosis. Further, analysis using CD and FTIR spectroscopy reveals that the ACT-7 peptide efficiently inhibits the formation of the β-sheet secondary structure content. HR-TEM microscopic analysis confirmed the inhibition of formation. Therefore, the inhibition of β-sheet Aβ fibrillary aggregation by the protease-stable ACT-7 peptide may provide a beneficial effect on AD treatment to control the Aβ aggregates. Finally, we anticipate that our newly designed ACT peptides may also assist as a template molecular scaffold for designing potential anti-AD therapeutics.
Collapse
Affiliation(s)
- Rathnam Mallesh
- Department of Bioscience & Bioengineering, Indian Institute of Technology, Jodhpur, NH 65, Surpura Bypass Road, Karwar, Jodhpur, Rajasthan 342037, India
- Organic and Medicinal Chemistry and Structural Biology and Bioinformatics Division, CSIR-Indian Institute of Chemical Biology, 4, Raja S. C. Mullick Road, Jadavpur, Kolkata, WB 700 032, India
- National Institute of Pharmaceutical Education and Research, Kolkata, Chunilal Bhawan 168, Maniktala Main Road, Kolkata 700054, India
| | - Juhee Khan
- Department of Bioscience & Bioengineering, Indian Institute of Technology, Jodhpur, NH 65, Surpura Bypass Road, Karwar, Jodhpur, Rajasthan 342037, India
- Organic and Medicinal Chemistry and Structural Biology and Bioinformatics Division, CSIR-Indian Institute of Chemical Biology, 4, Raja S. C. Mullick Road, Jadavpur, Kolkata, WB 700 032, India
| | - Prabir Kumar Gharai
- Department of Bioscience & Bioengineering, Indian Institute of Technology, Jodhpur, NH 65, Surpura Bypass Road, Karwar, Jodhpur, Rajasthan 342037, India
- Organic and Medicinal Chemistry and Structural Biology and Bioinformatics Division, CSIR-Indian Institute of Chemical Biology, 4, Raja S. C. Mullick Road, Jadavpur, Kolkata, WB 700 032, India
| | - Mohammad Umar Arshi
- Department of Bioscience & Bioengineering, Indian Institute of Technology, Jodhpur, NH 65, Surpura Bypass Road, Karwar, Jodhpur, Rajasthan 342037, India
| | - Shubham Garg
- Department of Bioscience & Bioengineering, Indian Institute of Technology, Jodhpur, NH 65, Surpura Bypass Road, Karwar, Jodhpur, Rajasthan 342037, India
| | - Sanju Gupta
- Department of Bioscience & Bioengineering, Indian Institute of Technology, Jodhpur, NH 65, Surpura Bypass Road, Karwar, Jodhpur, Rajasthan 342037, India
| | - Surajit Ghosh
- Department of Bioscience & Bioengineering, Indian Institute of Technology, Jodhpur, NH 65, Surpura Bypass Road, Karwar, Jodhpur, Rajasthan 342037, India
- Organic and Medicinal Chemistry and Structural Biology and Bioinformatics Division, CSIR-Indian Institute of Chemical Biology, 4, Raja S. C. Mullick Road, Jadavpur, Kolkata, WB 700 032, India
- National Institute of Pharmaceutical Education and Research, Kolkata, Chunilal Bhawan 168, Maniktala Main Road, Kolkata 700054, India
| |
Collapse
|
41
|
Brandão-Teles C, Antunes ASLM, de Moraes Vrechi TA, Martins-de-Souza D. The Roles of hnRNP Family in the Brain and Brain-Related Disorders. Mol Neurobiol 2024; 61:3578-3595. [PMID: 37999871 DOI: 10.1007/s12035-023-03747-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Accepted: 10/25/2023] [Indexed: 11/25/2023]
Abstract
Heterogeneous nuclear ribonucleoproteins (hnRNPs) belong to a complex family of RNA-binding proteins that are essential to control alternative splicing, mRNA trafficking, synaptic plasticity, stress granule formation, cell cycle regulation, and axonal transport. Over the past decade, hnRNPs have been associated with different brain disorders such as Alzheimer's disease, multiple sclerosis, and schizophrenia. Given their essential role in maintaining cell function and integrity, it is not surprising that dysregulated hnRNP levels lead to neurological implications. This review aims to explore the primary functions of hnRNPs in neurons, oligodendrocytes, microglia, and astrocytes, and their roles in brain disorders. We also discuss proteomics and other technologies and their potential for studying and evaluating hnRNPs in brain disorders, including the discovery of new therapeutic targets and possible pharmacological interventions.
Collapse
Affiliation(s)
- Caroline Brandão-Teles
- Laboratory of Neuroproteomics, Department of Biochemistry and Tissue Biology, Institute of Biology, University of Campinas, Campinas, Brazil.
| | - André S L M Antunes
- Brazilian Biosciences National Laboratory (LNBio), Brazilian Center for Research in Energy and Materials (CNPEM), Campinas, Brazil
| | - Talita Aparecida de Moraes Vrechi
- Laboratory of Neuroproteomics, Department of Biochemistry and Tissue Biology, Institute of Biology, University of Campinas, Campinas, Brazil
| | - Daniel Martins-de-Souza
- Laboratory of Neuroproteomics, Department of Biochemistry and Tissue Biology, Institute of Biology, University of Campinas, Campinas, Brazil.
- D'Or Institute for Research and Education (IDOR), São Paulo, Brazil.
- Experimental Medicine Research Cluster (EMRC), University of Campinas, Campinas, SP, 13083-862, Brazil.
- INCT in Modelling Human Complex Diseases with 3D Platforms (Model3D), São Paulo, Brazil.
- Conselho Nacional de Desenvolvimento Científico e Tecnológico, Instituto Nacional de Biomarcadores em Neuropsiquiatria, São Paulo, Brazil.
| |
Collapse
|
42
|
Volloch V, Rits-Volloch S. ACH2.0/E, the Consolidated Theory of Conventional and Unconventional Alzheimer's Disease: Origins, Progression, and Therapeutic Strategies. Int J Mol Sci 2024; 25:6036. [PMID: 38892224 PMCID: PMC11172602 DOI: 10.3390/ijms25116036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2024] [Revised: 05/19/2024] [Accepted: 05/21/2024] [Indexed: 06/21/2024] Open
Abstract
The centrality of amyloid-beta (Aβ) is an indisputable tenet of Alzheimer's disease (AD). It was initially indicated by the detection (1991) of a mutation within Aβ protein precursor (AβPP) segregating with the disease, which served as a basis for the long-standing Amyloid Cascade Hypothesis (ACH) theory of AD. In the intervening three decades, this notion was affirmed and substantiated by the discovery of numerous AD-causing and AD-protective mutations with all, without an exception, affecting the structure, production, and intraneuronal degradation of Aβ. The ACH postulated that the disease is caused and driven by extracellular Aβ. When it became clear that this is not the case, and the ACH was largely discredited, a new theory of AD, dubbed ACH2.0 to re-emphasize the centrality of Aβ, was formulated. In the ACH2.0, AD is caused by physiologically accumulated intraneuronal Aβ (iAβ) derived from AβPP. Upon reaching the critical threshold, it triggers activation of the autonomous AβPP-independent iAβ generation pathway; its output is retained intraneuronally and drives the AD pathology. The bridge between iAβ derived from AβPP and that generated independently of AβPP is the neuronal integrated stress response (ISR) elicited by the former. The ISR severely suppresses cellular protein synthesis; concurrently, it activates the production of a small subset of proteins, which apparently includes components necessary for operation of the AβPP-independent iAβ generation pathway that are absent under regular circumstances. The above sequence of events defines "conventional" AD, which is both caused and driven by differentially derived iAβ. Since the ISR can be elicited by a multitude of stressors, the logic of the ACH2.0 mandates that another class of AD, referred to as "unconventional", has to occur. Unconventional AD is defined as a disease where a stressor distinct from AβPP-derived iAβ elicits the neuronal ISR. Thus, the essence of both, conventional and unconventional, forms of AD is one and the same, namely autonomous, self-sustainable, AβPP-independent production of iAβ. What distinguishes them is the manner of activation of this pathway, i.e., the mode of causation of the disease. In unconventional AD, processes occurring at locations as distant from and seemingly as unrelated to the brain as, say, the knee can potentially trigger the disease. The present study asserts that these processes include traumatic brain injury (TBI), chronic traumatic encephalopathy, viral and bacterial infections, and a wide array of inflammatory conditions. It considers the pathways which are common to all these occurrences and culminate in the elicitation of the neuronal ISR, analyzes the dynamics of conventional versus unconventional AD, shows how the former can morph into the latter, explains how a single TBI can hasten the occurrence of AD and why it takes multiple TBIs to trigger the disease, and proposes the appropriate therapeutic strategies. It posits that yet another class of unconventional AD may occur where the autonomous AβPP-independent iAβ production pathway is initiated by an ISR-unrelated activator, and consolidates the above notions in a theory of AD, designated ACH2.0/E (for expanded ACH2.0), which incorporates the ACH2.0 as its special case and retains the centrality of iAβ produced independently of AβPP as the driving agent of the disease.
Collapse
Affiliation(s)
- Vladimir Volloch
- Department of Developmental Biology, Harvard School of Dental Medicine, Boston, MA 02115, USA
| | - Sophia Rits-Volloch
- Division of Molecular Medicine, Children’s Hospital, Boston, MA 02115, USA
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA 02115, USA
| |
Collapse
|
43
|
Yu Y, Yu S, Battaglia G, Tian X. Amyloid-β in Alzheimer's disease: Structure, toxicity, distribution, treatment, and prospects. IBRAIN 2024; 10:266-289. [PMID: 39346788 PMCID: PMC11427815 DOI: 10.1002/ibra.12155] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/21/2024] [Revised: 05/06/2024] [Accepted: 05/07/2024] [Indexed: 10/01/2024]
Abstract
Amyloid-β (Aβ) is a pivotal biomarker in Alzheimer's disease (AD), attracting considerable attention from numerous researchers. There is uncertainty regarding whether clearing Aβ is beneficial or harmful to cognitive function. This question has been a central topic of research, especially given the lack of success in developing Aβ-targeted drugs for AD. However, with the Food and Drug Administration's approval of Lecanemab as the first anti-Aβ medication in July 2023, there is a significant shift in perspective on the potential of Aβ as a therapeutic target for AD. In light of this advancement, this review aims to illustrate and consolidate the molecular structural attributes and pathological ramifications of Aβ. Furthermore, it elucidates the determinants influencing its expression levels while delineating the gamut of extant Aβ-targeted pharmacotherapies that have been subjected to clinical or preclinical evaluation. Subsequently, a comprehensive analysis is presented, dissecting the research landscape of Aβ across the domains above, culminating in the presentation of informed perspectives. Concluding reflections contemplate the supplementary advantages conferred by nanoparticle constructs, conceptualized within the framework of multivalent theory, within the milieu of AD diagnosis and therapeutic intervention, supplementing conventional modalities.
Collapse
Affiliation(s)
- Yifan Yu
- Institute for Bioengineering of Catalunya (IBEC)The Barcelona Institute of Science and Technology (BIST), Barcelona (Spain), Carrer Baldiri I ReixacBarcelonaSpain
- Catalan Institution for Research and Advanced Studies (ICREA)BarcelonaSpain
- Department of Radiology and Huaxi MR Research Center (HMRRC), Functional and Molecular Imaging Key Laboratory of Sichuan Province, West China HospitalSichuan UniversityChengduChina
| | - Shilong Yu
- Institute for Bioengineering of Catalunya (IBEC)The Barcelona Institute of Science and Technology (BIST), Barcelona (Spain), Carrer Baldiri I ReixacBarcelonaSpain
- Catalan Institution for Research and Advanced Studies (ICREA)BarcelonaSpain
- Department of Radiology and Huaxi MR Research Center (HMRRC), Functional and Molecular Imaging Key Laboratory of Sichuan Province, West China HospitalSichuan UniversityChengduChina
| | - Giuseppe Battaglia
- Institute for Bioengineering of Catalunya (IBEC)The Barcelona Institute of Science and Technology (BIST), Barcelona (Spain), Carrer Baldiri I ReixacBarcelonaSpain
- Catalan Institution for Research and Advanced Studies (ICREA)BarcelonaSpain
- Department of Radiology and Huaxi MR Research Center (HMRRC), Functional and Molecular Imaging Key Laboratory of Sichuan Province, West China HospitalSichuan UniversityChengduChina
| | - Xiaohe Tian
- Institute for Bioengineering of Catalunya (IBEC)The Barcelona Institute of Science and Technology (BIST), Barcelona (Spain), Carrer Baldiri I ReixacBarcelonaSpain
- Catalan Institution for Research and Advanced Studies (ICREA)BarcelonaSpain
- Department of Radiology and Huaxi MR Research Center (HMRRC), Functional and Molecular Imaging Key Laboratory of Sichuan Province, West China HospitalSichuan UniversityChengduChina
| |
Collapse
|
44
|
Abukuri DN. Novel Biomarkers for Alzheimer's Disease: Plasma Neurofilament Light and Cerebrospinal Fluid. Int J Alzheimers Dis 2024; 2024:6668159. [PMID: 38779175 PMCID: PMC11111307 DOI: 10.1155/2024/6668159] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2023] [Revised: 03/18/2024] [Accepted: 04/24/2024] [Indexed: 05/25/2024] Open
Abstract
Neurodegenerative disorders such as Alzheimer's disease (AD) represent an increasingly significant public health concern. As clinical diagnosis faces challenges, biomarkers are becoming increasingly important in research, trials, and patient assessments. While biomarkers like amyloid-β peptide, tau proteins, CSF levels (Aβ, tau, and p-tau), and neuroimaging techniques are commonly used in AD diagnosis, they are often limited and invasive in monitoring and diagnosis. For this reason, blood-based biomarkers are the optimal choice for detecting neurodegeneration in brain diseases due to their noninvasiveness, affordability, reliability, and consistency. This literature review focuses on plasma neurofilament light (NfL) and CSF NfL as blood-based biomarkers used in recent AD diagnosis. The findings revealed that the core CSF biomarkers of neurodegeneration (T-tau, P-tau, and Aβ42), CSF NFL, and plasma T-tau were strongly associated with Alzheimer's disease, and the core biomarkers were strongly associated with mild cognitive impairment due to Alzheimer's disease. Elevated levels of plasma and cerebrospinal fluid NfL were linked to decreased [18F]FDG uptake in corresponding brain areas. In participants with Aβ positivity (Aβ+), NfL correlated with reduced metabolism in regions susceptible to Alzheimer's disease. In addition, CSF NfL levels correlate with brain atrophy and predict cognitive changes, while plasma total tau does not. Plasma P-tau, especially in combination with Aβ42/Aβ40, is promising for symptomatic AD stages. Though not AD-exclusive, blood NfL holds promise for neurodegeneration detection and assessing treatment efficacy. Given the consistent levels of T-tau, P-tau, Aβ42, and NFL in CSF, their incorporation into both clinical practice and research is highly recommended.
Collapse
|
45
|
Thai QM, Tran PT, Phung HTT, Pham MQ, Ngo ST. Silver nanoparticles alter the dimerization of Aβ 42 studied by REMD simulations. RSC Adv 2024; 14:15112-15119. [PMID: 38720971 PMCID: PMC11078207 DOI: 10.1039/d4ra02197e] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2024] [Accepted: 05/03/2024] [Indexed: 01/06/2025] Open
Abstract
The aggregation of amyloid beta (Aβ) peptides is associated with the development of Alzheimer's disease (AD). However, there has been a growing belief that the oligomerization of Aβ species in different environments has a neurotoxic effect on the patient's brain, causing damage. It is necessary to comprehend the compositions of Aβ oligomers in order to develop medications that may effectively inhibit these neurotoxic forms that affect the nervous system of AD patients. Thus, dissociation or inhibition of Aβ aggregation may be able to prevent AD. To date, the search for traditional agents and biomolecules has largely been unsuccessful. In this context, nanoparticles have emerged as potential candidates to directly inhibit the formation of Aβ oligomers. The oligomerization of the dimeric Aβ peptides with or without the influence of a silver nanoparticle was thus investigated using temperature replica-exchange molecular dynamics (REMD) simulations. The physical insights into the dimeric Aβ oligomerization were clarified by analyzing intermolecular contact maps, the free energy landscape of the dimeric oligomer, secondary structure terms, etc. The difference in obtained metrics between Aβ with or without a silver nanoparticle provides a picture of the influence of silver nanoparticles on the oligomerization process. The underlying mechanisms that are involved in altering Aβ oligomerization will be discussed. The obtained results may play an important role in searching for Aβ inhibitor pathways.
Collapse
Affiliation(s)
- Quynh Mai Thai
- Laboratory of Biophysics, Institute of Advanced Study in Technology, Ton Duc Thang University Ho Chi Minh City Vietnam
- Faculty of Pharmacy, Ton Duc Thang University Ho Chi Minh City Vietnam
| | | | - Huong T T Phung
- NTT Hi-Tech Institute, Nguyen Tat Thanh University Ho Chi Minh City Vietnam
| | - Minh Quan Pham
- Institute of Natural Products Chemistry, Vietnam Academy of Science and Technology Hanoi Vietnam
- Graduate University of Science and Technology, Vietnam Academy of Science and Technology Hanoi Vietnam
| | - Son Tung Ngo
- Laboratory of Biophysics, Institute of Advanced Study in Technology, Ton Duc Thang University Ho Chi Minh City Vietnam
- Faculty of Pharmacy, Ton Duc Thang University Ho Chi Minh City Vietnam
| |
Collapse
|
46
|
Dehghani H, Holzapfel GA, Mittelbronn M, Zilian A. Cell adhesion affects the properties of interstitial fluid flow: A study using multiscale poroelastic composite modeling. J Mech Behav Biomed Mater 2024; 153:106486. [PMID: 38428205 DOI: 10.1016/j.jmbbm.2024.106486] [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: 07/06/2023] [Revised: 02/20/2024] [Accepted: 02/26/2024] [Indexed: 03/03/2024]
Abstract
In this study, we conduct a multiscale, multiphysics modeling of the brain gray matter as a poroelastic composite. We develop a customized representative volume element based on cytoarchitectural features that encompass important microscopic components of the tissue, namely the extracellular space, the capillaries, the pericapillary space, the interstitial fluid, cell-cell and cell-capillary junctions, and neuronal and glial cell bodies. Using asymptotic homogenization and direct numerical simulation, the effective properties at the tissue level are identified based on microscopic properties. To analyze the influence of various microscopic elements on the effective/macroscopic properties and tissue response, we perform sensitivity analyses on cell junction (cluster) stiffness, cell junction diameter (dimensions), and pericapillary space width. The results of this study suggest that changes in cell adhesion can greatly affect both mechanical and hydraulic (interstitial fluid flow and porosity) features of brain tissue, consistent with the effects of neurodegenerative diseases.
Collapse
Affiliation(s)
- Hamidreza Dehghani
- Institute of Computational Engineering and Sciences, Department of Engineering, Faculty of Science, Technology and Medicine, University of Luxembourg, Esch-sur-Alzette, Luxembourg.
| | - Gerhard A Holzapfel
- Institute of Biomechanics, Graz University of Technology, 8010 Graz, Austria; Department of Structural Engineering, Norwegian University of Science and Technology (NTNU), 7491 Trondheim, Norway
| | - Michel Mittelbronn
- National Center of Pathology (NCP), Laboratoire National de Santé (LNS), Dudelange, Luxembourg; Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, Belval, Luxembourg; Luxembourg Center of Neuropathology (LCNP), Dudelange, Luxembourg; Department of Oncology (DONC), Luxembourg Institute of Health (LIH), Luxembourg; Department of Life Sciences and Medicine (DLSM), University of Luxembourg, Esch-sur-Alzette, Luxembourg; Faculty of Science, Technology and Medicine (FSTM), University of Luxembourg, Esch-sur-Alzette, Luxembourg
| | - Andreas Zilian
- Institute of Computational Engineering and Sciences, Department of Engineering, Faculty of Science, Technology and Medicine, University of Luxembourg, Esch-sur-Alzette, Luxembourg
| |
Collapse
|
47
|
Ayoubi R, Fotouhi M, Worrall D, Southern K, Laflamme C. A guide to selecting high-performing antibodies for amyloid-beta precursor protein for use in Western Blot, immunoprecipitation and immunofluorescence. F1000Res 2024; 12:956. [PMID: 39359612 PMCID: PMC11445640 DOI: 10.12688/f1000research.139867.2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 04/04/2024] [Indexed: 10/04/2024] Open
Abstract
The amyloid-beta precursor protein is a transmembrane protein expressed in many tissues and highly concentrated in the brain. The protein is of significant interest due to its involvement in the generation of amyloidogenic β-amyloid peptides, prone to plaque formation that is characteristic of Alzheimer's Disease. The scientific community would benefit from the availability of high-quality anti-amyloid-beta precursor protein antibodies to enhance reproducible research on this target. In this study, we characterized eleven amyloid-beta precursor protein commercial antibodies for Western blot, immunoprecipitation, and immunofluorescence using a standardized experimental protocol based on comparing read-outs in knockout cell lines and isogenic parental controls. These studies are part of a larger, collaborative initiative seeking to address antibody reproducibility issues by characterizing commercially available antibodies for human proteins and publishing the results openly as a resource for the scientific community. While use of antibodies and protocols vary between laboratories, we encourage readers to use this report as a guide to select the most appropriate antibodies for their specific needs.
Collapse
Affiliation(s)
- Riham Ayoubi
- Department of Neurology and Neurosurgery, Structural Genomics Consortium, The Montreal Neurological Institute, McGill University, Montreal, Québec, H3A 2B4, Canada
| | - Maryam Fotouhi
- Department of Neurology and Neurosurgery, Structural Genomics Consortium, The Montreal Neurological Institute, McGill University, Montreal, Québec, H3A 2B4, Canada
| | - Donovan Worrall
- Department of Neurology and Neurosurgery, Structural Genomics Consortium, The Montreal Neurological Institute, McGill University, Montreal, Québec, H3A 2B4, Canada
| | - Kathleen Southern
- Department of Neurology and Neurosurgery, Structural Genomics Consortium, The Montreal Neurological Institute, McGill University, Montreal, Québec, H3A 2B4, Canada
| | - Carl Laflamme
- Department of Neurology and Neurosurgery, Structural Genomics Consortium, The Montreal Neurological Institute, McGill University, Montreal, Québec, H3A 2B4, Canada
| | | | - ABIF consortium
- Department of Neurology and Neurosurgery, Structural Genomics Consortium, The Montreal Neurological Institute, McGill University, Montreal, Québec, H3A 2B4, Canada
| |
Collapse
|
48
|
Selkoe DJ. The advent of Alzheimer treatments will change the trajectory of human aging. NATURE AGING 2024; 4:453-463. [PMID: 38641654 DOI: 10.1038/s43587-024-00611-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/21/2023] [Accepted: 03/08/2024] [Indexed: 04/21/2024]
Abstract
Slowing neurodegenerative disorders of late life has lagged behind progress on other chronic diseases. But advances in two areas, biochemical pathology and human genetics, have now identified early pathogenic events, enabling molecular hypotheses and disease-modifying treatments. A salient example is the discovery that antibodies to amyloid ß-protein, long debated as a causative factor in Alzheimer's disease (AD), clear amyloid plaques, decrease levels of abnormal tau proteins and slow cognitive decline. Approval of amyloid antibodies as the first disease-modifying treatments means a gradually rising fraction of the world's estimated 60 million people with symptomatic disease may decline less or even stabilize. Society is entering an era in which the unchecked devastation of AD is no longer inevitable. This Perspective considers the impact of slowing AD and other neurodegenerative disorders on the trajectory of aging, allowing people to survive into late life with less functional decline. The implications of this moment for medicine and society are profound.
Collapse
Affiliation(s)
- Dennis J Selkoe
- Ann Romney Center for Neurologic Diseases Brigham and Women's Hospital Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
49
|
Wicks SL, Roberts JA, Hurtt MJ, Hernandez BP, Jones JJ, Taylor AL, Logan JK, Schreiber WJ, Murray MG, Crenshaw BL, Stevens CB, Lammi RK, Hanna JM. Synthesis of symmetrical and unsymmetrical tetrahydroxybiphenyls and their evaluation as amyloid-β aggregation inhibitors. LETT ORG CHEM 2024; 21:964-972. [PMID: 40191153 PMCID: PMC11970624 DOI: 10.2174/0115701786286700240322065602] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Revised: 03/07/2024] [Accepted: 03/08/2024] [Indexed: 04/09/2025]
Abstract
Our group recently reported that the polyhydroxy aromatic compound 3,3',4,4'-biphenyltetrol (2a) is a successful inhibitor of amyloid-β peptide (Aβ) aggregation, decreasing Aβ aggregation by 50 % when present in equimolar concentrations. In the present study, several additional biphenyltetrols were prepared and examined for their in vitro activity against aggregation of Aβ, to investigate the effect of the relative positions of hydrogen-bond donors on the aggregation process. Congo red spectral shift assays have shown that, of the eight (8) additional biphenyltetrol compounds prepared, three (3) successfully inhibit association of Aβ monomers - two symmetrical isomers, 2,2',5,5'-biphenyltetrol (2c), and 2,2',3,3'-biphenyltetrol (2d), along with one unsymmetrical isomer, 2,3',4',5-biphenyltetrol (2g). These results, along with previously reported results of 2a, strongly suggest that hydroxyl group position affects the ability of the inhibitor to bind to Aβ assemblies, thus impacting inhibitory efficacy.
Collapse
Affiliation(s)
- Sarah L. Wicks
- Department of Chemistry, Physics, Geology, and the Environment, Winthrop University, Rock Hill, SC
| | - Jake A. Roberts
- Department of Chemistry, Physics, Geology, and the Environment, Winthrop University, Rock Hill, SC
| | - Matthew J. Hurtt
- Department of Chemistry, Physics, Geology, and the Environment, Winthrop University, Rock Hill, SC
| | - Benjamin P. Hernandez
- Department of Chemistry, Physics, Geology, and the Environment, Winthrop University, Rock Hill, SC
| | - Jason J. Jones
- Department of Chemistry, Physics, Geology, and the Environment, Winthrop University, Rock Hill, SC
| | - Andrea L. Taylor
- Department of Chemistry, Physics, Geology, and the Environment, Winthrop University, Rock Hill, SC
| | - Jessica K. Logan
- Department of Chemistry, Physics, Geology, and the Environment, Winthrop University, Rock Hill, SC
| | - William J. Schreiber
- Department of Chemistry, Physics, Geology, and the Environment, Winthrop University, Rock Hill, SC
| | - Mouskudah G. Murray
- Department of Chemistry, Physics, Geology, and the Environment, Winthrop University, Rock Hill, SC
| | - Brandy L. Crenshaw
- Department of Chemistry, Physics, Geology, and the Environment, Winthrop University, Rock Hill, SC
| | - Craig B. Stevens
- Department of Chemistry, Physics, Geology, and the Environment, Winthrop University, Rock Hill, SC
| | - Robin K. Lammi
- Department of Chemistry, Physics, Geology, and the Environment, Winthrop University, Rock Hill, SC
| | - James M. Hanna
- Department of Chemistry, Physics, Geology, and the Environment, Winthrop University, Rock Hill, SC
| |
Collapse
|
50
|
Amartumur S, Nguyen H, Huynh T, Kim TS, Woo RS, Oh E, Kim KK, Lee LP, Heo C. Neuropathogenesis-on-chips for neurodegenerative diseases. Nat Commun 2024; 15:2219. [PMID: 38472255 PMCID: PMC10933492 DOI: 10.1038/s41467-024-46554-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2023] [Accepted: 02/28/2024] [Indexed: 03/14/2024] Open
Abstract
Developing diagnostics and treatments for neurodegenerative diseases (NDs) is challenging due to multifactorial pathogenesis that progresses gradually. Advanced in vitro systems that recapitulate patient-like pathophysiology are emerging as alternatives to conventional animal-based models. In this review, we explore the interconnected pathogenic features of different types of ND, discuss the general strategy to modelling NDs using a microfluidic chip, and introduce the organoid-on-a-chip as the next advanced relevant model. Lastly, we overview how these models are being applied in academic and industrial drug development. The integration of microfluidic chips, stem cells, and biotechnological devices promises to provide valuable insights for biomedical research and developing diagnostic and therapeutic solutions for NDs.
Collapse
Affiliation(s)
- Sarnai Amartumur
- Department of Biophysics, Institute of Quantum Biophysics, Sungkyunkwan University, Suwon, 16419, Korea
| | - Huong Nguyen
- Department of Biophysics, Institute of Quantum Biophysics, Sungkyunkwan University, Suwon, 16419, Korea
| | - Thuy Huynh
- Department of Biophysics, Institute of Quantum Biophysics, Sungkyunkwan University, Suwon, 16419, Korea
| | - Testaverde S Kim
- Center for Integrated Nanostructure Physics (CINAP), Institute for Basic Science (IBS), Suwon, 16419, Korea
| | - Ran-Sook Woo
- Department of Anatomy and Neuroscience, College of Medicine, Eulji University, Daejeon, 34824, Korea
| | - Eungseok Oh
- Department of Neurology, Chungnam National University Hospital, Daejeon, 35015, Korea
| | - Kyeong Kyu Kim
- Department of Precision Medicine, Graduate School of Basic Medical Science (GSBMS), Institute for Anti-microbial Resistance Research and Therapeutics, Sungkyunkwan University School of Medicine, Suwon, 16419, Korea
| | - Luke P Lee
- Department of Biophysics, Institute of Quantum Biophysics, Sungkyunkwan University, Suwon, 16419, Korea.
- Harvard Medical School, Division of Engineering in Medicine and Renal Division, Department of Medicine, Brigham and Women's Hospital, Boston, MA, 02115, USA.
- Department of Bioengineering, Department of Electrical Engineering and Computer Science, University of California, Berkeley, CA, 94720, USA.
| | - Chaejeong Heo
- Department of Biophysics, Institute of Quantum Biophysics, Sungkyunkwan University, Suwon, 16419, Korea.
- Center for Integrated Nanostructure Physics (CINAP), Institute for Basic Science (IBS), Suwon, 16419, Korea.
| |
Collapse
|