1
|
Chelucci E, Scarfò G, Piccarducci R, Rizza A, Fusi J, Epifani F, Carpi S, Polini B, Betti L, Costa B, Taliani S, Cela V, Artini P, Daniele S, Martini C, Franzoni F. Sex Differences in Blood Accumulation of Neurodegenerative-Related Proteins and Antioxidant Responses to Regular Physical Exercise. J Mol Neurosci 2024; 74:105. [PMID: 39496982 DOI: 10.1007/s12031-024-02278-4] [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: 07/16/2024] [Accepted: 10/04/2024] [Indexed: 11/06/2024]
Abstract
Physical activity has been demonstrated to improve cognitive function, thereby preventing/slowing neurodegenerative diseases (NDs). Biological responses to physical activity and vulnerabilities to NDs are emerging to be gender-related. Herein, known ND-associated markers (β-amyloid, tau, α-synuclein), main sex steroid hormones, antioxidant responses, and key gene transcription modulators were evaluated in the blood of physically active and sedentary women and men. In our hands, females presented higher basal erythrocytes β-amyloid and α-synuclein amounts than males. Regular physical activity was able to significantly reduce the erythrocyte content of β-amyloid in females and the tau levels in males, suggesting that these differences may be mediated by organizational actions of sex steroid hormones during development. Furthermore, despite a comparable plasma antioxidant capability (AOC) between males and females, in the latter group, physical activity significantly enhances AOC versus peroxynitrite radicals only. Finally, regular physical activity modulated the levels of transcription factor Nrf2 in erythrocytes, as well as the plasma concentration of the microRNA miR-195 and miR-153, suggesting the promotion of antioxidant/autophagic processes associated with ND-related proteins. Overall, these results could shed light on how cerebral adaptations to physical activity differ between males and females, especially with regard to blood accumulation of ND proteins and mechanisms of antioxidant responses to regular exercise.
Collapse
Affiliation(s)
| | - Giorgia Scarfò
- Division of General Medicine, Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | | | - Antonio Rizza
- Interventional Cardiology Division, Gaetano Pasquinucci Heart Hospital, Fondazione Toscana Gabriele Monasterio, Massa, Italy
| | - Jonathan Fusi
- Division of General Medicine, Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Francesco Epifani
- Department of Juridical and Economic Sciences, Pegaso Telematic University, Fanfani Diagnostics and Health, Florence, Italy
| | - Sara Carpi
- Department of Health Sciences, University "Magna Graecia" of Catanzaro, Catanzaro, Italy
- National Enterprise for nanoScience and nanoTechnology (NEST), Istituto Nanoscienze-CNR and Scuola Normale Superiore, Pisa, Italy
| | | | - Laura Betti
- Department of Pharmacy, University of Pisa, Pisa, Italy
| | - Barbara Costa
- Department of Pharmacy, University of Pisa, Pisa, Italy
| | | | - Vito Cela
- Division of Gynecology and Obstetrics, Azienda Ospedaliero Universitaria Pisana, Pisa, Italy
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Paolo Artini
- Division of Gynecology and Obstetrics, Azienda Ospedaliero Universitaria Pisana, Pisa, Italy
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Simona Daniele
- Department of Pharmacy, University of Pisa, Pisa, Italy.
| | | | - Ferdinando Franzoni
- Division of General Medicine, Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| |
Collapse
|
2
|
Wu J, Wu J, Chen T, Cai J, Ren R. Protein aggregation and its affecting mechanisms in neurodegenerative diseases. Neurochem Int 2024; 180:105880. [PMID: 39396709 DOI: 10.1016/j.neuint.2024.105880] [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: 07/22/2024] [Revised: 10/09/2024] [Accepted: 10/11/2024] [Indexed: 10/15/2024]
Abstract
Protein aggregation serves as a critical pathological marker in a spectrum of neurodegenerative diseases (NDs), including the formation of amyloid β (Aβ) and Tau neurofibrillary tangles in Alzheimer's disease, as well as α-Synuclein (α-Syn) aggregates in Parkinson's disease, Parkinson's disease-related dementia (PDD), dementia with Lewy bodies (DLB), and multiple system atrophy (MSA). A significant proportion of patients with amyotrophic lateral sclerosis (ALS) exhibit TDP-43 aggregates. Moreover, a confluence of brain protein pathologies, such as Aβ, Tau, α-Syn, and TDP-43, has been identified in individual NDs cases, highlighting the intricate interplay among these proteins that is garnering heightened scrutiny. Importantly, protein aggregation is modulated by an array of factors, with burgeoning evidence suggesting that it frequently results from perturbations in protein homeostasis, influenced by the cellular membrane milieu, metal ion concentrations, post-translational modifications, and genetic mutations. This review delves into the pathological underpinnings of protein aggregation across various NDs and elucidates the intercommunication among disparate proteins within the same disease context. Additionally, we examine the pathogenic mechanisms by which diverse factors impinge upon protein aggregation, offering fresh perspectives for the future therapeutic intervention of NDs.
Collapse
Affiliation(s)
- Junyun Wu
- Neuroscience Care Unit, Second Affiliated Hospital of Zhejiang University School of Medicine, 88 Jiefang Road, Hangzhou, Zhejiang, 310009, China
| | - Jianan Wu
- Department of Neurosurgery, Second Affiliated Hospital of Zhejiang University School of Medicine, 88 Jiefang Road, Hangzhou, Zhejiang, 310009, China
| | - Tao Chen
- Neuroscience Care Unit, Second Affiliated Hospital of Zhejiang University School of Medicine, 88 Jiefang Road, Hangzhou, Zhejiang, 310009, China
| | - Jing Cai
- Neuroscience Care Unit, Second Affiliated Hospital of Zhejiang University School of Medicine, 88 Jiefang Road, Hangzhou, Zhejiang, 310009, China.
| | - Reng Ren
- Neuroscience Care Unit, Second Affiliated Hospital of Zhejiang University School of Medicine, 88 Jiefang Road, Hangzhou, Zhejiang, 310009, China.
| |
Collapse
|
3
|
Mayo P, Pascual J, Crisman E, Domínguez C, López MG, León R. Innovative pathological network-based multitarget approaches for Alzheimer's disease treatment. Med Res Rev 2024; 44:2367-2419. [PMID: 38678582 DOI: 10.1002/med.22045] [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: 11/02/2023] [Revised: 02/02/2024] [Accepted: 04/14/2024] [Indexed: 05/01/2024]
Abstract
Alzheimer's disease (AD) is the most prevalent neurodegenerative disease and is a major health threat globally. Its prevalence is forecasted to exponentially increase during the next 30 years due to the global aging population. Currently, approved drugs are merely symptomatic, being ineffective in delaying or blocking the relentless disease advance. Intensive AD research describes this disease as a highly complex multifactorial disease. Disclosure of novel pathological pathways and their interconnections has had a major impact on medicinal chemistry drug development for AD over the last two decades. The complex network of pathological events involved in the onset of the disease has prompted the development of multitarget drugs. These chemical entities combine pharmacological activities toward two or more drug targets of interest. These multitarget-directed ligands are proposed to modify different nodes in the pathological network aiming to delay or even stop disease progression. Here, we review the multitarget drug development strategy for AD during the last decade.
Collapse
Affiliation(s)
- Paloma Mayo
- Departamento de desarrollo preclínico, Fundación Teófilo Hernando, Las Rozas, Madrid, Spain
- Departamento de Farmacología y Terapéutica, Facultad de Medicina, Universidad Autónoma de Madrid, Madrid, Spain
- Instituto de Química Médica, Consejo Superior de Investigaciones Científicas (IQM-CSIC), Madrid, Spain
| | - Jorge Pascual
- Departamento de desarrollo preclínico, Fundación Teófilo Hernando, Las Rozas, Madrid, Spain
- Instituto de Química Médica, Consejo Superior de Investigaciones Científicas (IQM-CSIC), Madrid, Spain
| | - Enrique Crisman
- Instituto de Química Médica, Consejo Superior de Investigaciones Científicas (IQM-CSIC), Madrid, Spain
| | - Cristina Domínguez
- Instituto de Química Médica, Consejo Superior de Investigaciones Científicas (IQM-CSIC), Madrid, Spain
| | - Manuela G López
- Departamento de Farmacología y Terapéutica, Facultad de Medicina, Universidad Autónoma de Madrid, Madrid, Spain
| | - Rafael León
- Instituto de Química Médica, Consejo Superior de Investigaciones Científicas (IQM-CSIC), Madrid, Spain
| |
Collapse
|
4
|
Schäffler M, Wales DJ, Strodel B. The energy landscape of Aβ 42: a funnel to disorder for the monomer becomes a folding funnel for self-assembly. Chem Commun (Camb) 2024. [PMID: 39479923 DOI: 10.1039/d4cc02856b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2024]
Abstract
The aggregation of amyloid-β (Aβ) peptides, particularly Aβ1-42, plays a key role in Alzheimer's disease pathogenesis. In this study, we investigate how dimerisation transforms the free energy surface (FES) of the Aβ1-42 monomer when it interacts with another Aβ1-42 peptide. We find that the monomer FES is a structurally inverted funnel with a disordered state at the global minimum. However, in the presence of a second Aβ1-42 peptide, the landscape becomes a folding funnel, leading to a β-hairpin state. Using first passage time analysis, we analyse the pathway for the transition from disordered to the β-hairpin state, which highlights the initial formation of a D23-K28 salt bridge as the driving force, together with hydrophobic contacts.
Collapse
Affiliation(s)
- Moritz Schäffler
- Institute of Theoretical and Computational Chemistry, Heinrich Heine University Düsseldorf, 40225 Düsseldorf, Germany.
- Institute of Biological Information Processing, Structural Biochemistry (IBI-7), Forschungszentrum Jülich, 52428, Jülich, Germany
| | - David J Wales
- Yusuf Hamied Department of Chemistry, University of Cambridge, CB2 1EW Cambridge, UK
| | - Birgit Strodel
- Institute of Theoretical and Computational Chemistry, Heinrich Heine University Düsseldorf, 40225 Düsseldorf, Germany.
- Institute of Biological Information Processing, Structural Biochemistry (IBI-7), Forschungszentrum Jülich, 52428, Jülich, Germany
| |
Collapse
|
5
|
Caneus J, Autar K, Akanda N, Grillo M, Long CJ, Jackson M, Lindquist S, Guo X, Morgan D, Hickman JJ. Validation of a functional human AD model with four AD therapeutics utilizing patterned ipsc-derived cortical neurons integrated with microelectrode arrays. Sci Rep 2024; 14:24875. [PMID: 39438515 PMCID: PMC11496884 DOI: 10.1038/s41598-024-73869-9] [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: 04/23/2024] [Accepted: 09/23/2024] [Indexed: 10/25/2024] Open
Abstract
Preclinical methods are needed for screening potential Alzheimer's disease (AD) therapeutics that recapitulate phenotypes found in the Mild Cognitive Impairment (MCI) stage or even before this stage of the disease. This would require a phenotypic system that reproduces cognitive deficits without significant neuronal cell death to mimic the clinical manifestations of AD during these stages. Long-term potentiation (LTP), which is a correlate of learning and memory, was induced in mature human iPSC-derived cortical neurons cultured on microelectrode arrays utilizing circuit patterns connecting two adjacent electrodes. We demonstrated an LTP system that modeled the MCI and pre-MCI stages of Alzheimer's and validated this functional system utilizing four AD therapeutics, which was also verified utilizing patch-clamp electrophysiology. LTP was induced by tetanic electrical stimulation, and LTP maintenance was significantly reduced in the presence of Amyloid-Beta 42 (Aβ42) oligomers compared to the controls, however, co-treatment with AD therapeutics (Donepezil, Memantine, Rolipram and Saracatinib) corrected Aβ42-induced LTP impairment. The results illustrate the utility of the system as a validated platform to model MCI AD pathology, and potentially for the pre-MCI phase before significant neuronal death. This system also has the potential to become an ideal platform for high-content therapeutic screening for other neurodegenerative diseases.
Collapse
Affiliation(s)
- Julbert Caneus
- NanoScience Technology Center, University of Central Florida, 12424 Research Parkway, Suite 400, Orlando, FL, 32826, USA.
| | - Kaveena Autar
- NanoScience Technology Center, University of Central Florida, 12424 Research Parkway, Suite 400, Orlando, FL, 32826, USA
| | - Nesar Akanda
- NanoScience Technology Center, University of Central Florida, 12424 Research Parkway, Suite 400, Orlando, FL, 32826, USA
| | - Marcella Grillo
- NanoScience Technology Center, University of Central Florida, 12424 Research Parkway, Suite 400, Orlando, FL, 32826, USA
| | | | - Max Jackson
- Hesperos Inc., 12501 Research Pkwy #100, Orlando, FL, USA
| | | | - Xiufang Guo
- NanoScience Technology Center, University of Central Florida, 12424 Research Parkway, Suite 400, Orlando, FL, 32826, USA
| | - Dave Morgan
- Department of Translational Neuroscience, Michigan State University College of Human Medicine, Grand Rapids, MI, USA
| | - James J Hickman
- NanoScience Technology Center, University of Central Florida, 12424 Research Parkway, Suite 400, Orlando, FL, 32826, USA
- Hesperos Inc., 12501 Research Pkwy #100, Orlando, FL, USA
| |
Collapse
|
6
|
Zhang Z, Li R, Zhou Y, Huang S, Hou Y, Pei G. Dietary Flavonoid Chrysin Functions as a Dual Modulator to Attenuate Amyloid-β and Tau Pathology in the Models of Alzheimer's Disease. Mol Neurobiol 2024:10.1007/s12035-024-04557-y. [PMID: 39432184 DOI: 10.1007/s12035-024-04557-y] [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: 03/24/2024] [Accepted: 10/14/2024] [Indexed: 10/22/2024]
Abstract
Growing evidence indicates that healthy diets are associated with a slower progression of Alzheimer's disease (AD). Flavonoids are among the most abundant natural products in diets beneficial to AD, such as the Mediterranean diet. However, the effect and mechanism of these dietary flavonoids on AD remains incompletely understood. Here, we found that a representative dietary natural flavonoid, chrysin (Chr), significantly ameliorated cognitive impairment and AD pathology in APP/PS1 mice. Furthermore, mechanistic studies showed that Chr significantly reduced the levels of amyloid-β (Aβ) and phosphorylated tau (p-tau), along with dual inhibitory activity against β-site amyloid precursor protein cleaving enzyme 1 (BACE1) and glycogen synthase kinase 3β (GSK3β). Moreover, the effect of Chr was further confirmed by EW233, a structural analog of Chr that exhibited an improved pharmacokinetic profile. To further verify the role of Chr and EW233, we utilized our previously established chimeric human cerebral organoid (chCO) model for AD, in which astrogenesis was promoted to mimic the neuron-astrocyte ratio in human brain tissue, and similar dual inhibition of Aβ and p-tau was also observed. Altogether, our study not only reveals the molecular mechanisms through which dietary flavonoids, such as Chr, mitigate AD pathology, but also suggests that identifying a specific constituent that mimics some of the benefits of these healthy diets could serve as a promising approach to discover new treatments for AD.
Collapse
Affiliation(s)
- Zhen Zhang
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, 200031, China
| | - Rongyao Li
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, 200031, China
| | - Yue Zhou
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, 200031, China
| | - Shichao Huang
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, 200031, China
| | - Yujun Hou
- Institute for Regenerative Medicine, State Key Laboratory of Cardiology and Medical Innovation Center, Shanghai Key Laboratory of Signaling and Disease Research, Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Shanghai East Hospital, Tongji University, Shanghai, China
| | - Gang Pei
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, 200031, China.
- Shanghai Key Laboratory of Signaling and Disease Research, Laboratory of Receptor-Based Biomedicine, The Collaborative Innovation Center for Brain Science, School of Life Sciences and Technology, Tongji University, Shanghai, China.
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, China.
| |
Collapse
|
7
|
Arumugam D, Jamuna NA, Kamalakshan A, Mandal S. Modulation of AIE and Intramolecular Charge Transfer of a Pyrene-Based Probe for Discriminatory Detection and Imaging of Oligomers and Amyloid Fibrils. ACS APPLIED BIO MATERIALS 2024; 7:6343-6356. [PMID: 39291866 DOI: 10.1021/acsabm.4c00820] [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: 09/19/2024]
Abstract
Oligomers and amyloid fibrils formed at different stages of protein aggregation are important biomarkers for a variety of neurodegenerative diseases including Alzheimer's and Parkinson's diseases. The development of probes for the sensitive detection of oligomeric species is important for early stage diagnosis of amyloidogenic diseases. Many small molecular dyes have been developed to probe the dynamic growth of amyloid fibrils. However, there is a lack of discriminatory detection strategies to monitor the dynamics of both oligomers and amyloid fibrils based on the differential modulation of the photophysical properties of a single dye. Here we report a pyrene-based intramolecular charge transfer (ICT) dye with large Stokes shifted red-emitting aggregation induced emission (AIE) for monitoring the dynamic populations of both oligomers and fibrils during the aggregation of hen egg white lysozyme (HEWL) protein. At the early stage of protein aggregation, the accumulation of HEWL oligomers results in a rapid and substantial increase in the red AIE intensity at 660 nm. Later, as the oligomers transform into mature fibrils, the dye exhibits a distinct photophysical change. Binding of the dye to HEWL fibrils strongly suppresses the red AIE and enhances ICT emission. This is evidenced by a gradual decrease in the AIE intensity (∼660 nm) and an increase in LE (∼490 nm) and ICT (∼540 nm) emission intensities during the later stages of protein aggregation. Thus, the dye provides simultaneous measurements of the population dynamics of both HEWL oligomers and fibrils during protein aggregation based on the discriminatory modulation of AIE and ICT of the dye. The dye also enables imaging of both HEWL oligomers and fibrils simultaneously using different emission channels in super-resolution confocal fluorescence microscopy.
Collapse
Affiliation(s)
- Dharini Arumugam
- Department of Chemistry, National Institute of Technology, Tiruchirappalli, Tamil Nadu 620015, India
| | - Nidhi Anilkumar Jamuna
- Department of Chemistry, National Institute of Technology, Tiruchirappalli, Tamil Nadu 620015, India
| | - Adithya Kamalakshan
- Department of Chemistry, National Institute of Technology, Tiruchirappalli, Tamil Nadu 620015, India
| | - Sarthak Mandal
- Department of Chemistry, National Institute of Technology, Tiruchirappalli, Tamil Nadu 620015, India
| |
Collapse
|
8
|
Žvirblis M, Sakalauskas A, Ali Janvand SH, Dudutienė V, Žiaunys M, Sniečkutė R, Otzen DE, Smirnovas V, Matulis D. Structure-Activity Relationship of Fluorinated Benzenesulfonamides as Inhibitors of Amyloid-β Aggregation. Chemistry 2024; 30:e202402330. [PMID: 39109590 DOI: 10.1002/chem.202402330] [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] [Indexed: 09/25/2024]
Abstract
Amyloid-beta aggregation is considered one of the factors influencing the onset of the Alzheimer's disease. Early prevention of such aggregation should alleviate disease condition by applying small molecule compounds that shift the aggregation equilibrium toward the soluble form of the peptide or slow down the process. We have discovered that fluorinated benzenesulfonamides of particular structure slowed the amyloid-beta peptide aggregation process by more than three-fold. We synthesized a series of ortho-para and meta-para double-substituted fluorinated benzenesulfonamides that inhibited the aggregation process to a variable extent yielding a detailed picture of the structure-activity relationship. Analysis of compound chemical structure effect on aggregation in artificial cerebrospinal fluid showed the necessity to arrange the benzenesulfonamide, hydrophobic substituent, and benzoic acid in a particular way. The amyloid beta peptide aggregate fibril structures varied in cross-sectional height depending on the applied inhibitor indicating the formation of a complex with the compound. Application of selected inhibitors increased the survivability of cells affected by the amyloid beta peptide. Such compounds may be developed as drugs against Alzheimer's disease.
Collapse
Affiliation(s)
- Mantas Žvirblis
- Department of Biothermodynamics and Drug Design, Institute of Biotechnology, Life Sciences Center, Vilnius University, Saulėtekio av. 7, Vilnius, LT-10257, Lithuania
| | - Andrius Sakalauskas
- Sector of Amyloid Research, Institute of Biotechnology, Life Sciences Center, Vilnius University, Saulėtekio av. 7, Vilnius LT, 10257, Lithuania
| | - Saeid Hadi Ali Janvand
- Interdisciplinary Nanoscience Center (iNANO), Aarhus University, Gustav Wieds Vej 14, 8000, Aarhus C, Denmark
| | - Virginija Dudutienė
- Department of Biothermodynamics and Drug Design, Institute of Biotechnology, Life Sciences Center, Vilnius University, Saulėtekio av. 7, Vilnius, LT-10257, Lithuania
| | - Mantas Žiaunys
- Sector of Amyloid Research, Institute of Biotechnology, Life Sciences Center, Vilnius University, Saulėtekio av. 7, Vilnius LT, 10257, Lithuania
| | - Rūta Sniečkutė
- Sector of Amyloid Research, Institute of Biotechnology, Life Sciences Center, Vilnius University, Saulėtekio av. 7, Vilnius LT, 10257, Lithuania
| | - Daniel E Otzen
- Interdisciplinary Nanoscience Center (iNANO), Aarhus University, Gustav Wieds Vej 14, 8000, Aarhus C, Denmark
| | - Vytautas Smirnovas
- Sector of Amyloid Research, Institute of Biotechnology, Life Sciences Center, Vilnius University, Saulėtekio av. 7, Vilnius LT, 10257, Lithuania
| | - Daumantas Matulis
- Department of Biothermodynamics and Drug Design, Institute of Biotechnology, Life Sciences Center, Vilnius University, Saulėtekio av. 7, Vilnius, LT-10257, Lithuania
| |
Collapse
|
9
|
Madsen LS, Ismail R, Parbo P, Kjeldsen PL, Schaldemose JL, Hansen KV, Gottrup H, Aanerud J, Eskildsen SF, Brooks DJ. Microglial responses partially mediate the effect of Aβ on cognition in Alzheimer's disease. Alzheimers Dement 2024. [PMID: 39392185 DOI: 10.1002/alz.14298] [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: 05/07/2024] [Revised: 07/24/2024] [Accepted: 09/07/2024] [Indexed: 10/12/2024]
Abstract
INTRODUCTION Microglial responses are an integral part of Alzheimer's disease (AD) pathology and are associated with amyloid beta (Aβ) deposition. This study aimed to investigate the effects of Aβ and microglial responses on global cognitive impairment. METHODS In this longitudinal study, 28 patients with mild cognitive impairment and 11 healthy controls underwent 11C-PK11195 and 11C-Pittsburgh compound B positron emission tomography (PET), structural magnetic resonance imaging scans, and global cognitive ratings at baseline and 2-year follow-up. Correlations between PET uptake and global cognition were assessed. Additionally, the mediation effect of the microglial response on the association between Aβ load and global cognition was assessed. RESULTS Aβ load and the microglial response were both independently detrimental to global cognitive performance at baseline; however, at 2-year follow-up the association between Aβ load and global cognitive ratings was partially mediated by the microglial response. DISCUSSION As AD progresses, the associated microglial response partially mediates the detrimental effect of aggregated Aβ on cognition. HIGHLIGHTS This was a longitudinal study of amyloid beta (Aβ), microglial responses, and global cognitive performance. Aβ and microglial responses both affect cognition in early Alzheimer's disease. Microglial response partially mediates the effect of Aβ on cognition in later stages.
Collapse
Affiliation(s)
- Lasse S Madsen
- Department of Clinical Medicine, Center of Functionally Integrative Neuroscience, Aarhus University, Aarhus, Denmark
| | - Rola Ismail
- Department of Nuclear Medicine, Sygehus Lillebaelt, Vejle, Denmark
| | - Peter Parbo
- Department of Nuclear Medicine, Odense University Hospital, Odense, Denmark
| | - Pernille L Kjeldsen
- Department of Nuclear Medicine and PET-Centre, Aarhus University Hospital, Aarhus, Denmark
- Department of Neurology, Aalborg University Hospital, Aalborg, Denmark
| | - Jeppe L Schaldemose
- Department of Nuclear Medicine and PET-Centre, Aarhus University Hospital, Aarhus, Denmark
| | - Kim V Hansen
- Department of Nuclear Medicine and PET-Centre, Aarhus University Hospital, Aarhus, Denmark
| | - Hanne Gottrup
- Department of Neurology, Aarhus University Hospital, Aarhus, Denmark
| | - Joel Aanerud
- Department of Nuclear Medicine and PET-Centre, Aarhus University Hospital, Aarhus, Denmark
| | - Simon F Eskildsen
- Department of Clinical Medicine, Center of Functionally Integrative Neuroscience, Aarhus University, Aarhus, Denmark
| | - David J Brooks
- Department of Nuclear Medicine and PET-Centre, Aarhus University Hospital, Aarhus, Denmark
- Institute of Translational and Clinical Research, University of Newcastle upon Tyne, Newcastle upon Tyne, UK
| |
Collapse
|
10
|
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 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
|
11
|
Pascoal TA, Aguzzoli CS, Lussier FZ, Crivelli L, Suemoto CK, Fortea J, Rosa-Neto P, Zimmer ER, Ferreira PCL, Bellaver B. Insights into the use of biomarkers in clinical trials in Alzheimer's disease. EBioMedicine 2024; 108:105322. [PMID: 39366844 DOI: 10.1016/j.ebiom.2024.105322] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2024] [Revised: 08/12/2024] [Accepted: 08/21/2024] [Indexed: 10/06/2024] Open
Abstract
Biomarkers have been instrumental in population selection and disease monitoring in clinical trials of recently FDA-approved drugs targeting amyloid-β to slow the progression of Alzheimer's disease (AD). As new therapeutic strategies and biomarker techniques emerge, the importance of biomarkers in drug development is growing exponentially. In this emerging landscape, biomarkers are expected to serve a wide range of contexts of use in clinical trials focusing on AD and related dementias. The joint FDA-NIH BEST (Biomarkers, EndpointS, and other Tools) framework provides standardised terminology to facilitate communication among stakeholders in this increasingly complex field. This review explores various applications of biomarkers relevant to AD clinical trials, using the BEST resource as a reference. For simplicity, we predominantly provide contextual characterizations of biomarkers use from the perspective of drugs targeting amyloid-β and tau proteins. However, general definitions and concepts can be extrapolated to other targets.
Collapse
Affiliation(s)
- Tharick A Pascoal
- Department of Psychiatry, School of Medicine, University of Pittsburgh, Pittsburgh, PA, 15213, USA; Department of Neurology, School of Medicine, University of Pittsburgh, Pittsburgh, PA, 15213, USA.
| | | | - Firoza Z Lussier
- Department of Psychiatry, School of Medicine, University of Pittsburgh, Pittsburgh, PA, 15213, USA
| | - Lucía Crivelli
- Department of Cognitive Neurology, Fleni, Buenos Aires, Argentina
| | - Claudia K Suemoto
- Division of Geriatrics, University of São Paulo Medical School, São Paulo, Brazil
| | - Juan Fortea
- Sant Pau Memory Unit, Department of Neurology, Hospital de la Santa Creu i Sant Pau, Biomedical Research Institute Sant Pau, Universitat Autònoma de Barcelona, Barcelona, Spain; Barcelona Down Medical Center, Fundació Catalana Síndrome de Down, Barcelona, Spain; Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas, CIBERNED, Madrid, Spain
| | - Pedro Rosa-Neto
- Translational Neuroimaging Laboratory, McGill University Research Centre for Studies in Aging, Alzheimer's Disease Research Unit, Douglas Research Institute McConnell Brain Imaging Centre, Montreal Neurological Institute, Montreal, Canada
| | - Eduardo R Zimmer
- Brain Institute of Rio Grande do Sul, PUCRS, Porto Alegre, Brazil; Department of Pharmacology, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, 90035-003, Brazil; Graduate Program in Biological Sciences, Biochemistry (PPGBioq), and Pharmacology and Therapeutics (PPGFT), Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, 90035-003, Brazil
| | - Pamela C L Ferreira
- Department of Psychiatry, School of Medicine, University of Pittsburgh, Pittsburgh, PA, 15213, USA
| | - Bruna Bellaver
- Department of Psychiatry, School of Medicine, University of Pittsburgh, Pittsburgh, PA, 15213, USA
| |
Collapse
|
12
|
Samuels JD, Lukens JR, Price RJ. Emerging roles for ITAM and ITIM receptor signaling in microglial biology and Alzheimer's disease-related amyloidosis. J Neurochem 2024; 168:3558-3573. [PMID: 37822118 DOI: 10.1111/jnc.15981] [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: 08/07/2023] [Revised: 09/11/2023] [Accepted: 09/21/2023] [Indexed: 10/13/2023]
Abstract
Microglia are critical responders to amyloid beta (Aβ) plaques in Alzheimer's disease (AD). Therefore, the therapeutic targeting of microglia in AD is of high clinical interest. While previous investigation has focused on the innate immune receptors governing microglial functions in response to Aβ plaques, how microglial innate immune responses are regulated is not well understood. Interestingly, many of these microglial innate immune receptors contain unique cytoplasmic motifs, termed immunoreceptor tyrosine-based activating and inhibitory motifs (ITAM/ITIM), that are commonly known to regulate immune activation and inhibition in the periphery. In this review, we summarize the diverse functions employed by microglia in response to Aβ plaques and also discuss the innate immune receptors and intracellular signaling players that guide these functions. Specifically, we focus on the role of ITAM and ITIM signaling cascades in regulating microglia innate immune responses. A better understanding of how microglial innate immune responses are regulated in AD may provide novel therapeutic avenues to tune the microglial innate immune response in AD pathology.
Collapse
Affiliation(s)
- Joshua D Samuels
- Department of Neuroscience, Center for Brain Immunology and Glia (BIG), University of Virginia (UVA), Charlottesville, Virginia, USA
- Neuroscience Graduate Program, University of Virginia, Charlottesville, Virginia, USA
- Department of Biomedical Engineering, University of Virginia, Charlottesville, Virginia, USA
| | - John R Lukens
- Department of Neuroscience, Center for Brain Immunology and Glia (BIG), University of Virginia (UVA), Charlottesville, Virginia, USA
- Neuroscience Graduate Program, University of Virginia, Charlottesville, Virginia, USA
| | - Richard J Price
- Neuroscience Graduate Program, University of Virginia, Charlottesville, Virginia, USA
- Department of Biomedical Engineering, University of Virginia, Charlottesville, Virginia, USA
| |
Collapse
|
13
|
Junyi L, Yueyang W, Bin L, Xiaohong D, Wenhui C, Ning Z, Hong Z. Gut Microbiota Mediates Neuroinflammation in Alzheimer's Disease: Unraveling Key Factors and Mechanistic Insights. Mol Neurobiol 2024:10.1007/s12035-024-04513-w. [PMID: 39317889 DOI: 10.1007/s12035-024-04513-w] [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: 03/24/2024] [Accepted: 09/18/2024] [Indexed: 09/26/2024]
Abstract
The gut microbiota, the complex community of microorganisms that inhabit the gastrointestinal tract, has emerged as a key player in the pathogenesis of neurodegenerative disorders, including Alzheimer's disease (AD). AD is characterized by progressive cognitive decline and neuronal loss, associated with the accumulation of amyloid-β plaques, neurofibrillary tangles, and neuroinflammation in the brain. Increasing evidence suggests that alterations in the composition and function of the gut microbiota, known as dysbiosis, may contribute to the development and progression of AD by modulating neuroinflammation, a chronic and maladaptive immune response in the central nervous system. This review aims to comprehensively analyze the current role of the gut microbiota in regulating neuroinflammation and glial cell function in AD. Its objective is to deepen our understanding of the pathogenesis of AD and to discuss the potential advantages and challenges of using gut microbiota modulation as a novel approach for the diagnosis, treatment, and prevention of AD.
Collapse
Affiliation(s)
- Liang Junyi
- Heilongjiang University of Traditional Chinese Medicine, Harbin, 150040, Heilongjiang Province, China
| | - Wang Yueyang
- Heilongjiang University of Traditional Chinese Medicine, Harbin, 150040, Heilongjiang Province, China
| | - Liu Bin
- Heilongjiang University of Traditional Chinese Medicine, Harbin, 150040, Heilongjiang Province, China.
| | - Dong Xiaohong
- Jiamusi College, Heilongjiang University of Traditional Chinese Medicine, Jiamusi, Heilongjiang Province, China
| | - Cai Wenhui
- Heilongjiang University of Traditional Chinese Medicine, Harbin, 150040, Heilongjiang Province, China
| | - Zhang Ning
- Heilongjiang University of Traditional Chinese Medicine, Harbin, 150040, Heilongjiang Province, China
| | - Zhang Hong
- Heilongjiang Jiamusi Central Hospital, Jiamusi, Heilongjiang Province, China
| |
Collapse
|
14
|
Ma L, Meng T, Wang Y, Xue Y, Zheng Y, Chen J, Xu D, Sun J, Yang F, Huang J, Yang X. Real-time analysis of the biomolecular interaction between gelsolin and Aβ 1-42 monomer and its implication for Alzheimer's disease. Talanta 2024; 282:126938. [PMID: 39357407 DOI: 10.1016/j.talanta.2024.126938] [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: 06/22/2024] [Revised: 07/30/2024] [Accepted: 09/23/2024] [Indexed: 10/04/2024]
Abstract
Biomolecular interaction acts a pivotal part in understanding the mechanisms underlying the development of Alzheimer's disease (AD). Herein, we built a biosensing platform to explore the interaction between gelsolin (GSN) and different β-amyloid protein 1-42 (Aβ1-42) species, including Aβ1-42 monomer (m-Aβ), Aβ1-42 oligomers with both low and high levels of aggregation (LLo-Aβ and HLo-Aβ) via dual polarization interferometry (DPI). Real-time molecular interaction process and kinetic analysis showed that m-Aβ had the strongest affinity and specificity with GSN compared with LLo-Aβ and HLo-Aβ. The impact of GSN on inhibiting aggregation of Aβ1-42 and solubilizing Aβ1-42 aggregates was evaluated by circular dichroism (CD) spectroscopy. The maintenance of random coil structure of m-Aβ and the reversal of β-sheet structure in HLo-Aβ were observed, demonstrating the beneficial effects of GSN on preventing Aβ from aggregation. In addition, the structure of m-Aβ/GSN complex was analyzed in detail by molecular dynamics (MD) simulation and molecular docking. The specific binding sites and crucial intermolecular forces were identified, which are believed to stabilize m-Aβ in its soluble state and to inhibit the fibrilization of Aβ1-42. Combined theoretical simulations and experiment results, we speculate that the success of GSN sequestration mechanism and the balance of GSN levels in cerebrospinal fluid and plasma of AD subjects may contribute to a delay in AD progression. This research not only unveils the molecular basis of the interaction between GSN and Aβ1-42, but also provides clues to understanding the crucial functions of GSN in AD and drives the development of AD drugs and therapeutic approaches.
Collapse
Affiliation(s)
- Limin Ma
- State Key Laboratory of Electroanalytical Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, China; School of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei, 230026, China
| | - Tian Meng
- State Key Laboratory of Electroanalytical Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, China; School of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei, 230026, China
| | - Yu Wang
- State Key Laboratory of Electroanalytical Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, China; School of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei, 230026, China
| | - Yu Xue
- State Key Laboratory of Electroanalytical Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, China; School of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei, 230026, China
| | - Yuxin Zheng
- State Key Laboratory of Electroanalytical Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, China; School of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei, 230026, China
| | - Jinghuang Chen
- State Key Laboratory of Electroanalytical Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, China; School of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei, 230026, China
| | - Dongming Xu
- State Key Laboratory of Electroanalytical Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, China; School of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei, 230026, China
| | - Jian Sun
- State Key Laboratory of Electroanalytical Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, China
| | - Fan Yang
- State Key Laboratory of Electroanalytical Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, China
| | - Jianshe Huang
- State Key Laboratory of Electroanalytical Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, China.
| | - Xiurong Yang
- State Key Laboratory of Electroanalytical Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, China; School of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei, 230026, China.
| |
Collapse
|
15
|
Zhaliazka K, Kurouski D. Nanoscale Structural Characterization of Amyloid β 1-42 Oligomers and Fibrils Grown in the Presence of Fatty Acids. ACS Chem Neurosci 2024; 15:3344-3353. [PMID: 39222387 DOI: 10.1021/acschemneuro.4c00275] [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: 09/04/2024] Open
Abstract
Mono- and polyunsaturated fatty acids (FAs) are broadly used as food supplements. However, their effect on the aggregation of amyloidogenic proteins remains unclear. In this study, we investigated the effect of a large number of mono- and polyunsaturated, as well as fully saturated FAs on the aggregation of amyloid β1-42 (Aβ1-42) peptide. A progressive aggregation of this peptide is the expected molecular cause of Alzheimer's disease (AD), one of the most common neurodegenerative pathologies in the world. We found that arachidonic and stearic acids delayed the aggregation of Aβ1-42. Using Nano-Infrared spectroscopy, we found that FAs caused very little if any changes in the secondary structure of Aβ1-42 oligomers and fibrils formed at different stages of protein aggregation. However, the analyzed mono- and polyunsaturated, as well as fully saturated FAs uniquely altered the toxicity of Aβ1-42 fibrils. We found a direct relationship between the degree of FAs unsaturation and toxicity of Aβ1-42 fibrils formed in their presence. Specifically, with an increase in the degree of unsaturation, the toxicity Aβ1-42/FA fibrils increased. These results indicate that fully saturated or monounsaturated FAs could be used to decrease the toxicity of amyloid aggregates and, consequently, decelerate the development of AD.
Collapse
Affiliation(s)
- Kiryl Zhaliazka
- Department of Biochemistry and Biophysics, Texas A&M University, College Station, Texas 77843, United States
| | - Dmitry Kurouski
- Department of Biochemistry and Biophysics, Texas A&M University, College Station, Texas 77843, United States
- Department of Biomedical Engineering, Texas A&M University, College Station, Texas 77843, United States
| |
Collapse
|
16
|
Kuhn MK, Proctor EA. Microglial Drivers of Alzheimer's Disease Pathology: An Evolution of Diverse Participating States. Proteins 2024. [PMID: 39219300 DOI: 10.1002/prot.26723] [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: 01/17/2024] [Revised: 06/05/2024] [Accepted: 06/12/2024] [Indexed: 09/04/2024]
Abstract
Microglia, the resident immune-competent cells of the brain, become dysfunctional in Alzheimer's disease (AD), and their aberrant immune responses contribute to the accumulation of pathological proteins and neuronal injury. Genetic studies implicate microglia in the development of AD, prompting interest in developing immunomodulatory therapies to prevent or ameliorate disease. However, microglia take on diverse functional states in disease, playing both protective and detrimental roles in AD, which largely overlap and may shift over the disease course, complicating the identification of effective therapeutic targets. Extensive evidence gathered using transgenic mouse models supports an active role of microglia in pathology progression, though results vary and can be contradictory between different types of models and the degree of pathology at the time of study. Here, we review microglial immune signaling and responses that contribute to the accumulation and spread of pathological proteins or directly affect neuronal health. We additionally explore the use of induced pluripotent stem cell (iPSC)-derived models to study living human microglia and how they have contributed to our knowledge of AD and may begin to fill in the gaps left by mouse models. Ultimately, mouse and iPSC-derived models have their own limitations, and a comprehensive understanding of microglial dysfunction in AD will only be established by an integrated view across models and an appreciation for their complementary viewpoints and limitations.
Collapse
Affiliation(s)
- Madison K Kuhn
- Department of Biomedical Engineering, The Pennsylvania State University, University Park, Pennsylvania, USA
- Department of Neurosurgery, Penn State College of Medicine, Hershey, Pennsylvania, USA
- Department of Pharmacology, Penn State College of Medicine, Hershey, Pennsylvania, USA
- Center for Neural Engineering, The Pennsylvania State University, University Park, Pennsylvania, USA
| | - Elizabeth A Proctor
- Department of Biomedical Engineering, The Pennsylvania State University, University Park, Pennsylvania, USA
- Department of Neurosurgery, Penn State College of Medicine, Hershey, Pennsylvania, USA
- Department of Pharmacology, Penn State College of Medicine, Hershey, Pennsylvania, USA
- Center for Neural Engineering, The Pennsylvania State University, University Park, Pennsylvania, USA
- Department of Engineering Science & Mechanics, The Pennsylvania State University, University Park, Pennsylvania, USA
- Penn State Neuroscience Institute, The Pennsylvania State University, University Park, Pennsylvania, USA
| |
Collapse
|
17
|
Marcatti M, Tumurbaatar B, Borghi M, Guptarak J, Zhang WR, Krishnan B, Kayed R, Fracassi A, Taglialatela G. Inhibition of Calcineurin with FK506 Reduces Tau Levels and Attenuates Synaptic Impairment Driven by Tau Oligomers in the Hippocampus of Male Mouse Models. Int J Mol Sci 2024; 25:9092. [PMID: 39201779 PMCID: PMC11354963 DOI: 10.3390/ijms25169092] [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: 07/11/2024] [Revised: 08/13/2024] [Accepted: 08/15/2024] [Indexed: 09/03/2024] Open
Abstract
Alzheimer's disease (AD) is the most common age-associated neurodegenerative disorder, characterized by progressive cognitive decline, memory impairment, and structural brain changes, primarily involving Aβ plaques and neurofibrillary tangles of hyperphosphorylated tau protein. Recent research highlights the significance of smaller Aβ and Tau oligomeric aggregates (AβO and TauO, respectively) in synaptic dysfunction and disease progression. Calcineurin (CaN), a key calcium/calmodulin-dependent player in regulating synaptic function in the central nervous system (CNS) is implicated in mediating detrimental effects of AβO on synapses and memory function in AD. This study aims to investigate the specific impact of CaN on both exogenous and endogenous TauO through the acute and chronic inhibition of CaN. We previously demonstrated the protective effect against AD of the immunosuppressant CaN inhibitor, FK506, but its influence on TauO remains unclear. In this study, we explored the short-term effects of acute CaN inhibition on TauO phosphorylation and TauO-induced memory deficits and synaptic dysfunction. Mice received FK506 post-TauO intracerebroventricular injection and TauO levels and phosphorylation were assessed, examining their impact on CaN and GSK-3β. The study investigated FK506 preventive/reversal effects on TauO-induced clustering of CaN and GSK-3β. Memory and synaptic function in TauO-injected mice were evaluated with/without FK506. Chronic FK506 treatment in 3xTgAD mice explored its influence on CaN, Aβ, and Tau levels. This study underscores the significant influence of CaN inhibition on TauO and associated AD pathology, suggesting therapeutic potential in targeting CaN for addressing various aspects of AD onset and progression. These findings provide valuable insights for potential interventions in AD, emphasizing the need for further exploration of CaN-targeted strategies.
Collapse
|
18
|
Zhang LY, Chu YH, You YF, Dong MH, Pang XW, Chen L, Zhu LF, Yang S, Zhou LQ, Shang K, Deng G, Xiao J, Wang W, Qin C, Tian DS. Systematic Druggable Genome-Wide Mendelian Randomization Identifies Therapeutic Targets for Functional Outcome After Ischemic Stroke. J Am Heart Assoc 2024; 13:e034749. [PMID: 39119979 DOI: 10.1161/jaha.124.034749] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/29/2024] [Accepted: 07/15/2024] [Indexed: 08/10/2024]
Abstract
BACKGROUND Stroke is a leading cause of death worldwide, with a lack of effective treatments for improving the prognosis. The aim of the present study was to identify novel therapeutic targets for functional outcome after ischemic stroke . METHODS AND RESULTS Cis-expression quantitative trait loci data for druggable genes were used as instrumental variables. The primary outcome was the modified Rankin Scale score at 3 months after ischemic stroke, evaluated as a dichotomous variable (3-6 versus 0-2) and also as an ordinal variable. Drug target Mendelian randomization, Steiger filtering analysis, and colocalization analysis were performed. Additionally, phenome-wide Mendelian randomization analysis was performed to identify the safety of the drug target genes at the genetic level. Among >2600 druggable genes, genetically predicted expression of 16 genes (ABCC2, ATRAID, BLK, CD93, CHST13, NR1H3, NRBP1, PI3, RIPK4, SEMG1, SLC22A4, SLC22A5, SLCO3A1, TEK, TLR4, and WNT10B) demonstrated the causal associations with ordinal modified Rankin Scale (P<1.892×10-5) or poor functional outcome (modified Rankin Scale 3-6 versus 0-2, P<1.893×10-5). Steiger filtering analysis suggested potential directional stability (P<0.05). Colocalization analysis provided further support for the associations between genetically predicted expression of ABCC2, NRBP1, PI3, and SEMG1 with functional outcome after ischemic stroke. Furthermore, phenome-wide Mendelian randomization revealed additional beneficial indications and few potential safety concerns of therapeutics targeting ABCC2, NRBP1, PI3, and SEMG1, but the robustness of these results was limited by low power. CONCLUSIONS The present study revealed 4 candidate therapeutic targets for improving functional outcome after ischemic stroke, while the underlying mechanisms need further investigation.
Collapse
Affiliation(s)
- Lu-Yang Zhang
- Department of Neurology, Tongji Hospital, Tongji Medical College Huazhong University of Science and Technology Wuhan China
- Hubei Key Laboratory of Neural Injury and Functional Reconstruction Huazhong University of Science and Technology Wuhan China
- Key Laboratory of Vascular Aging, Ministry of Education, Tongji Hospital of Tongji Medical College Huazhong University of Science and Technology Wuhan China
| | - Yun-Hui Chu
- Department of Neurology, Tongji Hospital, Tongji Medical College Huazhong University of Science and Technology Wuhan China
- Hubei Key Laboratory of Neural Injury and Functional Reconstruction Huazhong University of Science and Technology Wuhan China
- Key Laboratory of Vascular Aging, Ministry of Education, Tongji Hospital of Tongji Medical College Huazhong University of Science and Technology Wuhan China
| | - Yun-Fan You
- Department of Neurology, Tongji Hospital, Tongji Medical College Huazhong University of Science and Technology Wuhan China
- Hubei Key Laboratory of Neural Injury and Functional Reconstruction Huazhong University of Science and Technology Wuhan China
- Key Laboratory of Vascular Aging, Ministry of Education, Tongji Hospital of Tongji Medical College Huazhong University of Science and Technology Wuhan China
| | - Ming-Hao Dong
- Department of Neurology, Tongji Hospital, Tongji Medical College Huazhong University of Science and Technology Wuhan China
- Hubei Key Laboratory of Neural Injury and Functional Reconstruction Huazhong University of Science and Technology Wuhan China
- Key Laboratory of Vascular Aging, Ministry of Education, Tongji Hospital of Tongji Medical College Huazhong University of Science and Technology Wuhan China
| | - Xiao-Wei Pang
- Department of Neurology, Tongji Hospital, Tongji Medical College Huazhong University of Science and Technology Wuhan China
- Hubei Key Laboratory of Neural Injury and Functional Reconstruction Huazhong University of Science and Technology Wuhan China
- Key Laboratory of Vascular Aging, Ministry of Education, Tongji Hospital of Tongji Medical College Huazhong University of Science and Technology Wuhan China
| | - Lian Chen
- Department of Neurology, Tongji Hospital, Tongji Medical College Huazhong University of Science and Technology Wuhan China
- Hubei Key Laboratory of Neural Injury and Functional Reconstruction Huazhong University of Science and Technology Wuhan China
- Key Laboratory of Vascular Aging, Ministry of Education, Tongji Hospital of Tongji Medical College Huazhong University of Science and Technology Wuhan China
| | - Li-Fang Zhu
- Department of Neurology, Tongji Hospital, Tongji Medical College Huazhong University of Science and Technology Wuhan China
- Hubei Key Laboratory of Neural Injury and Functional Reconstruction Huazhong University of Science and Technology Wuhan China
- Key Laboratory of Vascular Aging, Ministry of Education, Tongji Hospital of Tongji Medical College Huazhong University of Science and Technology Wuhan China
| | - Sheng Yang
- Department of Neurology, Tongji Hospital, Tongji Medical College Huazhong University of Science and Technology Wuhan China
- Hubei Key Laboratory of Neural Injury and Functional Reconstruction Huazhong University of Science and Technology Wuhan China
- Key Laboratory of Vascular Aging, Ministry of Education, Tongji Hospital of Tongji Medical College Huazhong University of Science and Technology Wuhan China
| | - Luo-Qi Zhou
- Department of Neurology, Tongji Hospital, Tongji Medical College Huazhong University of Science and Technology Wuhan China
- Hubei Key Laboratory of Neural Injury and Functional Reconstruction Huazhong University of Science and Technology Wuhan China
- Key Laboratory of Vascular Aging, Ministry of Education, Tongji Hospital of Tongji Medical College Huazhong University of Science and Technology Wuhan China
| | - Ke Shang
- Department of Neurology, Tongji Hospital, Tongji Medical College Huazhong University of Science and Technology Wuhan China
- Hubei Key Laboratory of Neural Injury and Functional Reconstruction Huazhong University of Science and Technology Wuhan China
- Key Laboratory of Vascular Aging, Ministry of Education, Tongji Hospital of Tongji Medical College Huazhong University of Science and Technology Wuhan China
| | - Gang Deng
- Department of Neurology, Tongji Hospital, Tongji Medical College Huazhong University of Science and Technology Wuhan China
- Hubei Key Laboratory of Neural Injury and Functional Reconstruction Huazhong University of Science and Technology Wuhan China
- Key Laboratory of Vascular Aging, Ministry of Education, Tongji Hospital of Tongji Medical College Huazhong University of Science and Technology Wuhan China
| | - Jun Xiao
- Department of Neurology, Tongji Hospital, Tongji Medical College Huazhong University of Science and Technology Wuhan China
- Hubei Key Laboratory of Neural Injury and Functional Reconstruction Huazhong University of Science and Technology Wuhan China
- Key Laboratory of Vascular Aging, Ministry of Education, Tongji Hospital of Tongji Medical College Huazhong University of Science and Technology Wuhan China
| | - Wei Wang
- Department of Neurology, Tongji Hospital, Tongji Medical College Huazhong University of Science and Technology Wuhan China
- Hubei Key Laboratory of Neural Injury and Functional Reconstruction Huazhong University of Science and Technology Wuhan China
- Key Laboratory of Vascular Aging, Ministry of Education, Tongji Hospital of Tongji Medical College Huazhong University of Science and Technology Wuhan China
| | - Chuan Qin
- Department of Neurology, Tongji Hospital, Tongji Medical College Huazhong University of Science and Technology Wuhan China
- Hubei Key Laboratory of Neural Injury and Functional Reconstruction Huazhong University of Science and Technology Wuhan China
- Key Laboratory of Vascular Aging, Ministry of Education, Tongji Hospital of Tongji Medical College Huazhong University of Science and Technology Wuhan China
| | - Dai-Shi Tian
- Department of Neurology, Tongji Hospital, Tongji Medical College Huazhong University of Science and Technology Wuhan China
- Hubei Key Laboratory of Neural Injury and Functional Reconstruction Huazhong University of Science and Technology Wuhan China
- Key Laboratory of Vascular Aging, Ministry of Education, Tongji Hospital of Tongji Medical College Huazhong University of Science and Technology Wuhan China
| |
Collapse
|
19
|
Liu N, Haziyihan A, Zhao W, Chen Y, Chao H. Trajectory of brain-derived amyloid beta in Alzheimer's disease: where is it coming from and where is it going? Transl Neurodegener 2024; 13:42. [PMID: 39160618 PMCID: PMC11331646 DOI: 10.1186/s40035-024-00434-9] [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: 01/30/2024] [Accepted: 07/25/2024] [Indexed: 08/21/2024] Open
Abstract
Alzheimer's disease (AD) is a progressive neurological disorder that primarily impacts cognitive function. Currently there are no disease-modifying treatments to stop or slow its progression. Recent studies have found that several peripheral and systemic abnormalities are associated with AD, and our understanding of how these alterations contribute to AD is becoming more apparent. In this review, we focuse on amyloid‑beta (Aβ), a major hallmark of AD, summarizing recent findings on the source of brain-derived Aβ and discussing where and how the brain-derived Aβ is cleared in vivo. Based on these findings, we propose future strategies for AD prevention and treatment, from a novel perspective on Aβ metabolism.
Collapse
Affiliation(s)
- Ni Liu
- Zhengzhou University, Zhengzhou, 450001, China
- Wuhan National Laboratory for Optoelectronics, Huazhong University of Science and Technology, Wuhan, 430074, China
| | | | - Wei Zhao
- Zhengzhou University, Zhengzhou, 450001, China
| | - Yu Chen
- Zhengzhou University, Zhengzhou, 450001, China
| | - Hongbo Chao
- Zhengzhou University, Zhengzhou, 450001, China.
- Huazhong University of Science and Technology, Wuhan, 430074, China.
| |
Collapse
|
20
|
Prasad AK, Samajdar R, Panwar AS, Martin LL. Origin of Secondary Structure Transitions and Peptide Self-Assembly Propensity in Trifluoroethanol-Water Mixtures. J Phys Chem B 2024; 128:7736-7749. [PMID: 39088441 DOI: 10.1021/acs.jpcb.4c02594] [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: 08/03/2024]
Abstract
Membrane-peptide interactions are key to the formation of helical intermediates in the early stages of amyloidogenesis. Aqueous solutions of 2,2,2-trifluoroethanol (TFE) provide a membrane-mimetic environment capable of promoting and stabilizing local peptide interactions. Uperin 3.5 (U3.5), a 17-residue and amidated antimicrobial peptide, is unstructured in water but self-assembles into fibrils in the presence of salt. Secondary structure transitions linked to U3.5 self-assembly were investigated in TFE/water mixtures, in both the absence and presence of salt, to assess the role of membrane-peptide interactions on peptide self-assembly and amyloid formation. A 5-to-7-fold increase in fibril yield of U3.5 was observed at low TFE concentrations (10% TFE/water v/v) compared with physiological buffer but only in the presence of salt. No aggregation was observed in salt-free TFE/water mixtures. Circular dichroism spectra showed that partial helical structures, initially stabilized by TFE, transitioned to β-sheet-rich aggregates in a saline buffer. Molecular dynamics simulations confirmed that TFE and salt act synergistically to enhance peptide-peptide interactions, resulting in β-sheet-rich U3.5 oligomers at low TFE concentrations. Specifically, TFE stabilized amphipathic, helical intermediates, leading to increased peptide-peptide attraction through hydrophobic interactions. The presence of salt further enhanced the peptide-peptide interactions by screening positively charged residues. Thus, the study revealed the role of a membrane mimic in stabilizing helical intermediates on the pathway to amyloid formation in the antimicrobial U3.5 peptide.
Collapse
Affiliation(s)
- Anup Kumar Prasad
- IITB-Monash Research Academy, Indian Institute of Technology Bombay, Mumbai 400076, India
- Department of Metallurgical Engineering & Materials Science, Indian Institute of Technology Bombay, Mumbai 400076, India
- School of Chemistry, Monash University, Clayton 3800, VIC, Australia
| | - Rajarshi Samajdar
- Department of Chemical Engineering, Institute of Chemical Technology, Mumbai 400019, India
| | - Ajay Singh Panwar
- Department of Metallurgical Engineering & Materials Science, Indian Institute of Technology Bombay, Mumbai 400076, India
| | - Lisandra L Martin
- School of Chemistry, Monash University, Clayton 3800, VIC, Australia
| |
Collapse
|
21
|
Senapati S, Tripathi K, Awad K, Rahimipour S. Multifunctional Liposomes Targeting Amyloid-β Oligomers for Early Diagnosis and Therapy of Alzheimer's Disease. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2311670. [PMID: 38461531 DOI: 10.1002/smll.202311670] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/14/2023] [Revised: 02/14/2024] [Indexed: 03/12/2024]
Abstract
Early detection and treatment are crucial for Alzheimer's disease (AD) management. Current diagnostic and therapeutic methods focus on late-stage amyloid fibrils and plaques, overlooking toxic soluble amyloid β oligomers (AβOs) accumulating early in AD. A multifunctional liposome-based platform is designed for early diagnosis and therapy of AD, leveraging a novel self-assembled cyclic d,l-α-peptide (CP-2) that selectively targets AβOs. Biocompatible CP-2 conjugated liposomes (CP-2-LPs) effectively disrupt Aβ aggregation and mitigate Aβ-mediated toxicity in human neuroblastoma cells. In transgenic Caenorhabditis elegans AD models, CP-2-LPs significantly outperformed free CP-2 by improving cognitive and behavioral functions, extending lifespan, and reducing toxic AβO levels. Intravenous injection of fluorescently labeled CP-2-LPs reveals effective blood-brain barrier penetration, with significantly higher brain fluorescence in transgenic mice than WT, enabling precise diagnosis. These findings underscore CP-2-LPs as a valuable tool for early detection and targeted therapy in AD.
Collapse
Affiliation(s)
- Sudipta Senapati
- Department of Chemistry, Bar-Ilan University, Ramat-Gan, 5290002, Israel
| | - Kuldeep Tripathi
- Department of Chemistry, Bar-Ilan University, Ramat-Gan, 5290002, Israel
| | - Khadeja Awad
- Department of Chemistry, Bar-Ilan University, Ramat-Gan, 5290002, Israel
| | - Shai Rahimipour
- Department of Chemistry, Bar-Ilan University, Ramat-Gan, 5290002, Israel
| |
Collapse
|
22
|
Chakrabarti KS, Bakhtiari D, Rezaei-Ghaleh N. Bifurcations in coupled amyloid-β aggregation-inflammation systems. NPJ Syst Biol Appl 2024; 10:80. [PMID: 39080352 PMCID: PMC11289389 DOI: 10.1038/s41540-024-00408-7] [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: 03/27/2024] [Accepted: 07/23/2024] [Indexed: 08/02/2024] Open
Abstract
A complex interplay between various processes underlies the neuropathology of Alzheimer's disease (AD) and its progressive course. Several lines of evidence point to the coupling between Aβ aggregation and neuroinflammation and its role in maintaining brain homeostasis during the long prodromal phase of AD. Little is however known about how this protective mechanism fails and as a result, an irreversible and progressive transition to clinical AD occurs. Here, we introduce a minimal model of a coupled system of Aβ aggregation and inflammation, numerically simulate its dynamical behavior, and analyze its bifurcation properties. The introduced model represents the following events: generation of Aβ monomers, aggregation of Aβ monomers into oligomers and fibrils, induction of inflammation by Aβ aggregates, and clearance of various Aβ species. Crucially, the rates of Aβ generation and clearance are modulated by inflammation level following a Hill-type response function. Despite its relative simplicity, the model exhibits enormously rich dynamics ranging from overdamped kinetics to sustained oscillations. We then specify the region of inflammation- and coupling-related parameters space where a transition to oscillatory dynamics occurs and demonstrate how changes in Aβ aggregation parameters could shift this oscillatory region in parameter space. Our results reveal the propensity of coupled Aβ aggregation-inflammation systems to oscillatory dynamics and propose prolonged sustained oscillations and their consequent immune system exhaustion as a potential mechanism underlying the transition to a more progressive phase of amyloid pathology in AD. The implications of our results in regard to early diagnosis of AD and anti-AD drug development are discussed.
Collapse
Affiliation(s)
- Kalyan S Chakrabarti
- Department of Biological Science and Chemistry, Krea University, Sri City, India
| | | | - Nasrollah Rezaei-Ghaleh
- Heinrich Heine University (HHU) Düsseldorf, Faculty of Mathematics and Natural Sciences, Institute of Physical Biology, Düsseldorf, Germany.
- Institute of Biological Information Processing, IBI-7: Structural Biochemistry, Forschungszentrum Jülich, Wilhelm-Johnen-Straße, Jülich, Germany.
| |
Collapse
|
23
|
Zott B, Nästle L, Grienberger C, Unger F, Knauer MM, Wolf C, Keskin-Dargin A, Feuerbach A, Busche MA, Skerra A, Konnerth A. β-amyloid monomer scavenging by an anticalin protein prevents neuronal hyperactivity in mouse models of Alzheimer's Disease. Nat Commun 2024; 15:5819. [PMID: 38987287 PMCID: PMC11237084 DOI: 10.1038/s41467-024-50153-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: 01/25/2023] [Accepted: 07/02/2024] [Indexed: 07/12/2024] Open
Abstract
Hyperactivity mediated by synaptotoxic β-amyloid (Aβ) oligomers is one of the earliest forms of neuronal dysfunction in Alzheimer's disease. In the search for a preventive treatment strategy, we tested the effect of scavenging Aβ peptides before Aβ plaque formation. Using in vivo two-photon calcium imaging and SF-iGluSnFR-based glutamate imaging in hippocampal slices, we demonstrate that an Aβ binding anticalin protein (Aβ-anticalin) can suppress early neuronal hyperactivity and synaptic glutamate accumulation in the APP23xPS45 mouse model of β-amyloidosis. Our results suggest that the sole targeting of Aβ monomers is sufficient for the hyperactivity-suppressing effect of the Aβ-anticalin at early disease stages. Biochemical and neurophysiological analyses indicate that the Aβ-anticalin-dependent depletion of naturally secreted Aβ monomers interrupts their aggregation to neurotoxic oligomers and, thereby, reverses early neuronal and synaptic dysfunctions. Thus, our results suggest that Aβ monomer scavenging plays a key role in the repair of neuronal function at early stages of AD.
Collapse
Affiliation(s)
- Benedikt Zott
- Institute of Neuroscience, Technical University of Munich, Munich, Germany.
- Department of Neuroradiology, MRI hospital of the Technical University of Munich, Munich, Germany.
- TUM Institute for Advanced Study, Garching, Germany.
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany.
| | - Lea Nästle
- Chair of Biological Chemistry, Technical University of Munich, Freising, Germany
| | - Christine Grienberger
- Institute of Neuroscience, Technical University of Munich, Munich, Germany
- Department of Biology and Volen National Center of Complex Systems, Brandeis University, Waltham, MA, USA
| | - Felix Unger
- Institute of Neuroscience, Technical University of Munich, Munich, Germany
- Department of Neuroradiology, MRI hospital of the Technical University of Munich, Munich, Germany
- TUM Institute for Advanced Study, Garching, Germany
| | - Manuel M Knauer
- Institute of Neuroscience, Technical University of Munich, Munich, Germany
| | - Christian Wolf
- Institute of Neuroscience, Technical University of Munich, Munich, Germany
- Department of Neuroradiology, MRI hospital of the Technical University of Munich, Munich, Germany
| | | | - Anna Feuerbach
- Chair of Biological Chemistry, Technical University of Munich, Freising, Germany
| | - Marc Aurel Busche
- Institute of Neuroscience, Technical University of Munich, Munich, Germany
- UK Dementia Research Institute at UCL, University College London, London, United Kingdom
| | - Arne Skerra
- Chair of Biological Chemistry, Technical University of Munich, Freising, Germany.
| | - Arthur Konnerth
- Institute of Neuroscience, Technical University of Munich, Munich, Germany.
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany.
| |
Collapse
|
24
|
Kobayashi R, Nabika H. Liquid-liquid phase separation induced by crowding condition affects amyloid-β aggregation mechanism. SOFT MATTER 2024; 20:5331-5342. [PMID: 38847095 DOI: 10.1039/d4sm00470a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/11/2024]
Abstract
Liquid-liquid phase separation (LLPS) is common in the aggregation of proteins associated with neurodegenerative diseases. Many efforts have been made to reproduce crowded conditions with artificial polymeric materials to understand the effect of LLPS in physiological conditions with significantly highly concentrated proteins, such as intrinsically disordered proteins. Although the possibility that LLPS is involved in intracellular amyloid-β (Aβ) aggregation, a protein related to the pathogenesis of Alzheimer's disease, has been investigated, the relationship between LLPS and the aggregation of Aβ is poorly characterized. Thus, in this study, we mimicked the intracellular crowding environment using polyethylene glycol and dextran, used commonly as model polymers, to examine the relationship of Aβ with LLPS and aggregation dynamics in vitro. We confirmed that Aβ undergoes LLPS under specific polymer coexistence conditions. Moreover, the addition of different electrolytes modulated LLPS and fibril formation. These results suggest that hydrophobic and electrostatic interactions are the driving forces for the LLPS of Aβ. Similar to the role of the liposome interface, the interface of droplets induced by LLPS functioned as the site for heterogeneous nucleation. These findings offer valuable insights into the complex mechanisms of Aβ aggregation in vivo and may be useful in establishing therapeutic methods for Alzheimer's disease.
Collapse
Affiliation(s)
- Ryuki Kobayashi
- Department of Science, Graduate School of Science and Engineering, Yamagata University, 1-4-12, Kojirakawa, Yamagata 990-8560, Japan
| | - Hideki Nabika
- Faculty of Science, Yamagata University, 1-4-12 Kojirakawa, Yamagata 990-8560, Japan.
| |
Collapse
|
25
|
Mastenbroek SE, Sala A, Vállez García D, Shekari M, Salvadó G, Lorenzini L, Pieperhoff L, Wink AM, Lopes Alves I, Wolz R, Ritchie C, Boada M, Visser PJ, Bucci M, Farrar G, Hansson O, Nordberg AK, Ossenkoppele R, Barkhof F, Gispert JD, Rodriguez-Vieitez E, Collij LE. Continuous β-Amyloid CSF/PET Imbalance Model to Capture Alzheimer Disease Heterogeneity. Neurology 2024; 103:e209419. [PMID: 38862136 PMCID: PMC11244744 DOI: 10.1212/wnl.0000000000209419] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2023] [Accepted: 03/29/2024] [Indexed: 06/13/2024] Open
Abstract
BACKGROUND AND OBJECTIVES Discordance between CSF and PET biomarkers of β-amyloid (Aβ) might reflect an imbalance between soluble and aggregated species, possibly reflecting disease heterogeneity. Previous studies generally used binary cutoffs to assess discrepancies in CSF/PET biomarkers, resulting in a loss of information on the extent of discordance. In this study, we (1) jointly modeled Aβ-CSF/PET data to derive a continuous measure of the imbalance between soluble and fibrillar pools of Aβ, (2) investigated factors contributing to this imbalance, and (3) examined associations with cognitive trajectories. METHODS Across 822 cognitively unimpaired (n = 261) and cognitively impaired (n = 561) Alzheimer's Disease Neuroimaging Initiative individuals (384 [46.7%] females, mean age 73.0 ± 7.4 years), we fitted baseline CSF-Aβ42 and global Aβ-PET to a hyperbolic regression model, deriving a participant-specific Aβ-aggregation score (standardized residuals); negative values represent more soluble relative to aggregated Aβ and positive values more aggregated relative to soluble Aβ. Using linear models, we investigated whether methodological factors, demographics, CSF biomarkers, and vascular burden contributed to Aβ-aggregation scores. With linear mixed models, we assessed whether Aβ-aggregation scores were predictive of cognitive functioning. Analyses were repeated in an early independent validation cohort of 383 Amyloid Imaging to Prevent Alzheimer's Disease Prognostic and Natural History Study individuals (224 [58.5%] females, mean age 65.2 ± 6.9 years). RESULTS The imbalance model could be fit (pseudo-R2 = 0.94) in both cohorts, across CSF kits and PET tracers. Although no associations were observed with the main methodological factors, lower Aβ-aggregation scores were associated with larger ventricular volume (β = 0.13, p < 0.001), male sex (β = -0.18, p = 0.019), and homozygous APOE-ε4 carriership (β = -0.56, p < 0.001), whereas higher scores were associated with increased uncorrected CSF p-tau (β = 0.17, p < 0.001) and t-tau (β = 0.16, p < 0.001), better baseline executive functioning (β = 0.12, p < 0.001), and slower global cognitive decline (β = 0.14, p = 0.006). In the validation cohort, we replicated the associations with APOE-ε4, CSF t-tau, and, although modestly, with cognition. DISCUSSION We propose a novel continuous model of Aβ CSF/PET biomarker imbalance, accurately describing heterogeneity in soluble vs aggregated Aβ pools in 2 independent cohorts across the full Aβ continuum. Aβ-aggregation scores were consistently associated with genetic and AD-associated CSF biomarkers, possibly reflecting disease heterogeneity beyond methodological influences.
Collapse
Affiliation(s)
- Sophie E Mastenbroek
- From the Department of Radiology and Nuclear Medicine (S.E.M., D.V.G., L.L., L.P., A.M.W., F.B., L.E.C.), Vrije Universiteit Amsterdam, Amsterdam University Medical Center, location VUmc; Amsterdam Neuroscience (S.E.M., D.V.G., L.L., L.P., A.M.W., F.B., L.E.C.), Brain Imaging, the Netherlands; Clinical Memory Research Unit (S.E.M., G.S., O.H., R.O., L.E.C.), Department of Clinical Sciences Malmö, Lund University; Division of Clinical Geriatrics (A.S., M. Bucci, A.K.N., E.R.-V.), Center for Alzheimer Research, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden; Coma Science Group (A.S.), GIGA-Consciousness, University of Liège; Centre du Cerveau2 (A.S.), University Hospital of Liège, Belgium; Barcelonaβeta Brain Research Center (BBRC) (M.S., G.S., J.D.G.), Pasqual Maragall Foundation; IMIM (Hospital del Mar Medical Research Institute) (M.S., J.D.G.), Barcelona; Centro de Investigación Biomédica en Red de Fragilidad y Envejecimiento Saludable (M.S., J.D.G.), Instituto de Salud Carlos III, Madrid; Universitat Pompeu Fabra (M.S.), Barcelona, Spain; Brain Research Center (I.L.A.), Amsterdam, the Netherlands; IXICO (R.W.), London; Centre for Clinical Brain Sciences (C.R.), University of Edinburgh, United Kingdom; Ace Alzheimer Center Barcelona (M. Boada), Universitat Internacional de Catalunya, Spain; Networking Research Center on Neurodegenerative Diseases (CIBERNED) (M. Boada), Instituto de Salud Carlos III, Madrid, Spain; Alzheimer Center Amsterdam (P.J.V., R.O.), Neurology, Vrije Universiteit Amsterdam, Amsterdam UMC location VUmc; Amsterdam Neuroscience (P.J.V.), Neurodegeneration; Alzheimer Center Limburg (P.J.V.), School for Mental Health and Neuroscience, Maastricht University, the Netherlands; Division of Neurogeriatrics (P.J.V.), Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden; Theme Inflammation and Aging (M. Bucci, A.K.N.), Karolinska University Hospital, Stockholm, Sweden; GE Healthcare (G.F.), Amersham, United Kingdom; Memory Clinic (O.H.), Skåne University Hospital, Malmö, Sweden; and Centre for Medical Image Computing (F.B.), and Queen Square Institute of Neurology, UCL, London, United Kingdom
| | - Arianna Sala
- From the Department of Radiology and Nuclear Medicine (S.E.M., D.V.G., L.L., L.P., A.M.W., F.B., L.E.C.), Vrije Universiteit Amsterdam, Amsterdam University Medical Center, location VUmc; Amsterdam Neuroscience (S.E.M., D.V.G., L.L., L.P., A.M.W., F.B., L.E.C.), Brain Imaging, the Netherlands; Clinical Memory Research Unit (S.E.M., G.S., O.H., R.O., L.E.C.), Department of Clinical Sciences Malmö, Lund University; Division of Clinical Geriatrics (A.S., M. Bucci, A.K.N., E.R.-V.), Center for Alzheimer Research, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden; Coma Science Group (A.S.), GIGA-Consciousness, University of Liège; Centre du Cerveau2 (A.S.), University Hospital of Liège, Belgium; Barcelonaβeta Brain Research Center (BBRC) (M.S., G.S., J.D.G.), Pasqual Maragall Foundation; IMIM (Hospital del Mar Medical Research Institute) (M.S., J.D.G.), Barcelona; Centro de Investigación Biomédica en Red de Fragilidad y Envejecimiento Saludable (M.S., J.D.G.), Instituto de Salud Carlos III, Madrid; Universitat Pompeu Fabra (M.S.), Barcelona, Spain; Brain Research Center (I.L.A.), Amsterdam, the Netherlands; IXICO (R.W.), London; Centre for Clinical Brain Sciences (C.R.), University of Edinburgh, United Kingdom; Ace Alzheimer Center Barcelona (M. Boada), Universitat Internacional de Catalunya, Spain; Networking Research Center on Neurodegenerative Diseases (CIBERNED) (M. Boada), Instituto de Salud Carlos III, Madrid, Spain; Alzheimer Center Amsterdam (P.J.V., R.O.), Neurology, Vrije Universiteit Amsterdam, Amsterdam UMC location VUmc; Amsterdam Neuroscience (P.J.V.), Neurodegeneration; Alzheimer Center Limburg (P.J.V.), School for Mental Health and Neuroscience, Maastricht University, the Netherlands; Division of Neurogeriatrics (P.J.V.), Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden; Theme Inflammation and Aging (M. Bucci, A.K.N.), Karolinska University Hospital, Stockholm, Sweden; GE Healthcare (G.F.), Amersham, United Kingdom; Memory Clinic (O.H.), Skåne University Hospital, Malmö, Sweden; and Centre for Medical Image Computing (F.B.), and Queen Square Institute of Neurology, UCL, London, United Kingdom
| | - David Vállez García
- From the Department of Radiology and Nuclear Medicine (S.E.M., D.V.G., L.L., L.P., A.M.W., F.B., L.E.C.), Vrije Universiteit Amsterdam, Amsterdam University Medical Center, location VUmc; Amsterdam Neuroscience (S.E.M., D.V.G., L.L., L.P., A.M.W., F.B., L.E.C.), Brain Imaging, the Netherlands; Clinical Memory Research Unit (S.E.M., G.S., O.H., R.O., L.E.C.), Department of Clinical Sciences Malmö, Lund University; Division of Clinical Geriatrics (A.S., M. Bucci, A.K.N., E.R.-V.), Center for Alzheimer Research, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden; Coma Science Group (A.S.), GIGA-Consciousness, University of Liège; Centre du Cerveau2 (A.S.), University Hospital of Liège, Belgium; Barcelonaβeta Brain Research Center (BBRC) (M.S., G.S., J.D.G.), Pasqual Maragall Foundation; IMIM (Hospital del Mar Medical Research Institute) (M.S., J.D.G.), Barcelona; Centro de Investigación Biomédica en Red de Fragilidad y Envejecimiento Saludable (M.S., J.D.G.), Instituto de Salud Carlos III, Madrid; Universitat Pompeu Fabra (M.S.), Barcelona, Spain; Brain Research Center (I.L.A.), Amsterdam, the Netherlands; IXICO (R.W.), London; Centre for Clinical Brain Sciences (C.R.), University of Edinburgh, United Kingdom; Ace Alzheimer Center Barcelona (M. Boada), Universitat Internacional de Catalunya, Spain; Networking Research Center on Neurodegenerative Diseases (CIBERNED) (M. Boada), Instituto de Salud Carlos III, Madrid, Spain; Alzheimer Center Amsterdam (P.J.V., R.O.), Neurology, Vrije Universiteit Amsterdam, Amsterdam UMC location VUmc; Amsterdam Neuroscience (P.J.V.), Neurodegeneration; Alzheimer Center Limburg (P.J.V.), School for Mental Health and Neuroscience, Maastricht University, the Netherlands; Division of Neurogeriatrics (P.J.V.), Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden; Theme Inflammation and Aging (M. Bucci, A.K.N.), Karolinska University Hospital, Stockholm, Sweden; GE Healthcare (G.F.), Amersham, United Kingdom; Memory Clinic (O.H.), Skåne University Hospital, Malmö, Sweden; and Centre for Medical Image Computing (F.B.), and Queen Square Institute of Neurology, UCL, London, United Kingdom
| | - Mahnaz Shekari
- From the Department of Radiology and Nuclear Medicine (S.E.M., D.V.G., L.L., L.P., A.M.W., F.B., L.E.C.), Vrije Universiteit Amsterdam, Amsterdam University Medical Center, location VUmc; Amsterdam Neuroscience (S.E.M., D.V.G., L.L., L.P., A.M.W., F.B., L.E.C.), Brain Imaging, the Netherlands; Clinical Memory Research Unit (S.E.M., G.S., O.H., R.O., L.E.C.), Department of Clinical Sciences Malmö, Lund University; Division of Clinical Geriatrics (A.S., M. Bucci, A.K.N., E.R.-V.), Center for Alzheimer Research, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden; Coma Science Group (A.S.), GIGA-Consciousness, University of Liège; Centre du Cerveau2 (A.S.), University Hospital of Liège, Belgium; Barcelonaβeta Brain Research Center (BBRC) (M.S., G.S., J.D.G.), Pasqual Maragall Foundation; IMIM (Hospital del Mar Medical Research Institute) (M.S., J.D.G.), Barcelona; Centro de Investigación Biomédica en Red de Fragilidad y Envejecimiento Saludable (M.S., J.D.G.), Instituto de Salud Carlos III, Madrid; Universitat Pompeu Fabra (M.S.), Barcelona, Spain; Brain Research Center (I.L.A.), Amsterdam, the Netherlands; IXICO (R.W.), London; Centre for Clinical Brain Sciences (C.R.), University of Edinburgh, United Kingdom; Ace Alzheimer Center Barcelona (M. Boada), Universitat Internacional de Catalunya, Spain; Networking Research Center on Neurodegenerative Diseases (CIBERNED) (M. Boada), Instituto de Salud Carlos III, Madrid, Spain; Alzheimer Center Amsterdam (P.J.V., R.O.), Neurology, Vrije Universiteit Amsterdam, Amsterdam UMC location VUmc; Amsterdam Neuroscience (P.J.V.), Neurodegeneration; Alzheimer Center Limburg (P.J.V.), School for Mental Health and Neuroscience, Maastricht University, the Netherlands; Division of Neurogeriatrics (P.J.V.), Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden; Theme Inflammation and Aging (M. Bucci, A.K.N.), Karolinska University Hospital, Stockholm, Sweden; GE Healthcare (G.F.), Amersham, United Kingdom; Memory Clinic (O.H.), Skåne University Hospital, Malmö, Sweden; and Centre for Medical Image Computing (F.B.), and Queen Square Institute of Neurology, UCL, London, United Kingdom
| | - Gemma Salvadó
- From the Department of Radiology and Nuclear Medicine (S.E.M., D.V.G., L.L., L.P., A.M.W., F.B., L.E.C.), Vrije Universiteit Amsterdam, Amsterdam University Medical Center, location VUmc; Amsterdam Neuroscience (S.E.M., D.V.G., L.L., L.P., A.M.W., F.B., L.E.C.), Brain Imaging, the Netherlands; Clinical Memory Research Unit (S.E.M., G.S., O.H., R.O., L.E.C.), Department of Clinical Sciences Malmö, Lund University; Division of Clinical Geriatrics (A.S., M. Bucci, A.K.N., E.R.-V.), Center for Alzheimer Research, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden; Coma Science Group (A.S.), GIGA-Consciousness, University of Liège; Centre du Cerveau2 (A.S.), University Hospital of Liège, Belgium; Barcelonaβeta Brain Research Center (BBRC) (M.S., G.S., J.D.G.), Pasqual Maragall Foundation; IMIM (Hospital del Mar Medical Research Institute) (M.S., J.D.G.), Barcelona; Centro de Investigación Biomédica en Red de Fragilidad y Envejecimiento Saludable (M.S., J.D.G.), Instituto de Salud Carlos III, Madrid; Universitat Pompeu Fabra (M.S.), Barcelona, Spain; Brain Research Center (I.L.A.), Amsterdam, the Netherlands; IXICO (R.W.), London; Centre for Clinical Brain Sciences (C.R.), University of Edinburgh, United Kingdom; Ace Alzheimer Center Barcelona (M. Boada), Universitat Internacional de Catalunya, Spain; Networking Research Center on Neurodegenerative Diseases (CIBERNED) (M. Boada), Instituto de Salud Carlos III, Madrid, Spain; Alzheimer Center Amsterdam (P.J.V., R.O.), Neurology, Vrije Universiteit Amsterdam, Amsterdam UMC location VUmc; Amsterdam Neuroscience (P.J.V.), Neurodegeneration; Alzheimer Center Limburg (P.J.V.), School for Mental Health and Neuroscience, Maastricht University, the Netherlands; Division of Neurogeriatrics (P.J.V.), Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden; Theme Inflammation and Aging (M. Bucci, A.K.N.), Karolinska University Hospital, Stockholm, Sweden; GE Healthcare (G.F.), Amersham, United Kingdom; Memory Clinic (O.H.), Skåne University Hospital, Malmö, Sweden; and Centre for Medical Image Computing (F.B.), and Queen Square Institute of Neurology, UCL, London, United Kingdom
| | - Luigi Lorenzini
- From the Department of Radiology and Nuclear Medicine (S.E.M., D.V.G., L.L., L.P., A.M.W., F.B., L.E.C.), Vrije Universiteit Amsterdam, Amsterdam University Medical Center, location VUmc; Amsterdam Neuroscience (S.E.M., D.V.G., L.L., L.P., A.M.W., F.B., L.E.C.), Brain Imaging, the Netherlands; Clinical Memory Research Unit (S.E.M., G.S., O.H., R.O., L.E.C.), Department of Clinical Sciences Malmö, Lund University; Division of Clinical Geriatrics (A.S., M. Bucci, A.K.N., E.R.-V.), Center for Alzheimer Research, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden; Coma Science Group (A.S.), GIGA-Consciousness, University of Liège; Centre du Cerveau2 (A.S.), University Hospital of Liège, Belgium; Barcelonaβeta Brain Research Center (BBRC) (M.S., G.S., J.D.G.), Pasqual Maragall Foundation; IMIM (Hospital del Mar Medical Research Institute) (M.S., J.D.G.), Barcelona; Centro de Investigación Biomédica en Red de Fragilidad y Envejecimiento Saludable (M.S., J.D.G.), Instituto de Salud Carlos III, Madrid; Universitat Pompeu Fabra (M.S.), Barcelona, Spain; Brain Research Center (I.L.A.), Amsterdam, the Netherlands; IXICO (R.W.), London; Centre for Clinical Brain Sciences (C.R.), University of Edinburgh, United Kingdom; Ace Alzheimer Center Barcelona (M. Boada), Universitat Internacional de Catalunya, Spain; Networking Research Center on Neurodegenerative Diseases (CIBERNED) (M. Boada), Instituto de Salud Carlos III, Madrid, Spain; Alzheimer Center Amsterdam (P.J.V., R.O.), Neurology, Vrije Universiteit Amsterdam, Amsterdam UMC location VUmc; Amsterdam Neuroscience (P.J.V.), Neurodegeneration; Alzheimer Center Limburg (P.J.V.), School for Mental Health and Neuroscience, Maastricht University, the Netherlands; Division of Neurogeriatrics (P.J.V.), Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden; Theme Inflammation and Aging (M. Bucci, A.K.N.), Karolinska University Hospital, Stockholm, Sweden; GE Healthcare (G.F.), Amersham, United Kingdom; Memory Clinic (O.H.), Skåne University Hospital, Malmö, Sweden; and Centre for Medical Image Computing (F.B.), and Queen Square Institute of Neurology, UCL, London, United Kingdom
| | - Leonard Pieperhoff
- From the Department of Radiology and Nuclear Medicine (S.E.M., D.V.G., L.L., L.P., A.M.W., F.B., L.E.C.), Vrije Universiteit Amsterdam, Amsterdam University Medical Center, location VUmc; Amsterdam Neuroscience (S.E.M., D.V.G., L.L., L.P., A.M.W., F.B., L.E.C.), Brain Imaging, the Netherlands; Clinical Memory Research Unit (S.E.M., G.S., O.H., R.O., L.E.C.), Department of Clinical Sciences Malmö, Lund University; Division of Clinical Geriatrics (A.S., M. Bucci, A.K.N., E.R.-V.), Center for Alzheimer Research, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden; Coma Science Group (A.S.), GIGA-Consciousness, University of Liège; Centre du Cerveau2 (A.S.), University Hospital of Liège, Belgium; Barcelonaβeta Brain Research Center (BBRC) (M.S., G.S., J.D.G.), Pasqual Maragall Foundation; IMIM (Hospital del Mar Medical Research Institute) (M.S., J.D.G.), Barcelona; Centro de Investigación Biomédica en Red de Fragilidad y Envejecimiento Saludable (M.S., J.D.G.), Instituto de Salud Carlos III, Madrid; Universitat Pompeu Fabra (M.S.), Barcelona, Spain; Brain Research Center (I.L.A.), Amsterdam, the Netherlands; IXICO (R.W.), London; Centre for Clinical Brain Sciences (C.R.), University of Edinburgh, United Kingdom; Ace Alzheimer Center Barcelona (M. Boada), Universitat Internacional de Catalunya, Spain; Networking Research Center on Neurodegenerative Diseases (CIBERNED) (M. Boada), Instituto de Salud Carlos III, Madrid, Spain; Alzheimer Center Amsterdam (P.J.V., R.O.), Neurology, Vrije Universiteit Amsterdam, Amsterdam UMC location VUmc; Amsterdam Neuroscience (P.J.V.), Neurodegeneration; Alzheimer Center Limburg (P.J.V.), School for Mental Health and Neuroscience, Maastricht University, the Netherlands; Division of Neurogeriatrics (P.J.V.), Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden; Theme Inflammation and Aging (M. Bucci, A.K.N.), Karolinska University Hospital, Stockholm, Sweden; GE Healthcare (G.F.), Amersham, United Kingdom; Memory Clinic (O.H.), Skåne University Hospital, Malmö, Sweden; and Centre for Medical Image Computing (F.B.), and Queen Square Institute of Neurology, UCL, London, United Kingdom
| | - Alle Meije Wink
- From the Department of Radiology and Nuclear Medicine (S.E.M., D.V.G., L.L., L.P., A.M.W., F.B., L.E.C.), Vrije Universiteit Amsterdam, Amsterdam University Medical Center, location VUmc; Amsterdam Neuroscience (S.E.M., D.V.G., L.L., L.P., A.M.W., F.B., L.E.C.), Brain Imaging, the Netherlands; Clinical Memory Research Unit (S.E.M., G.S., O.H., R.O., L.E.C.), Department of Clinical Sciences Malmö, Lund University; Division of Clinical Geriatrics (A.S., M. Bucci, A.K.N., E.R.-V.), Center for Alzheimer Research, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden; Coma Science Group (A.S.), GIGA-Consciousness, University of Liège; Centre du Cerveau2 (A.S.), University Hospital of Liège, Belgium; Barcelonaβeta Brain Research Center (BBRC) (M.S., G.S., J.D.G.), Pasqual Maragall Foundation; IMIM (Hospital del Mar Medical Research Institute) (M.S., J.D.G.), Barcelona; Centro de Investigación Biomédica en Red de Fragilidad y Envejecimiento Saludable (M.S., J.D.G.), Instituto de Salud Carlos III, Madrid; Universitat Pompeu Fabra (M.S.), Barcelona, Spain; Brain Research Center (I.L.A.), Amsterdam, the Netherlands; IXICO (R.W.), London; Centre for Clinical Brain Sciences (C.R.), University of Edinburgh, United Kingdom; Ace Alzheimer Center Barcelona (M. Boada), Universitat Internacional de Catalunya, Spain; Networking Research Center on Neurodegenerative Diseases (CIBERNED) (M. Boada), Instituto de Salud Carlos III, Madrid, Spain; Alzheimer Center Amsterdam (P.J.V., R.O.), Neurology, Vrije Universiteit Amsterdam, Amsterdam UMC location VUmc; Amsterdam Neuroscience (P.J.V.), Neurodegeneration; Alzheimer Center Limburg (P.J.V.), School for Mental Health and Neuroscience, Maastricht University, the Netherlands; Division of Neurogeriatrics (P.J.V.), Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden; Theme Inflammation and Aging (M. Bucci, A.K.N.), Karolinska University Hospital, Stockholm, Sweden; GE Healthcare (G.F.), Amersham, United Kingdom; Memory Clinic (O.H.), Skåne University Hospital, Malmö, Sweden; and Centre for Medical Image Computing (F.B.), and Queen Square Institute of Neurology, UCL, London, United Kingdom
| | - Isadora Lopes Alves
- From the Department of Radiology and Nuclear Medicine (S.E.M., D.V.G., L.L., L.P., A.M.W., F.B., L.E.C.), Vrije Universiteit Amsterdam, Amsterdam University Medical Center, location VUmc; Amsterdam Neuroscience (S.E.M., D.V.G., L.L., L.P., A.M.W., F.B., L.E.C.), Brain Imaging, the Netherlands; Clinical Memory Research Unit (S.E.M., G.S., O.H., R.O., L.E.C.), Department of Clinical Sciences Malmö, Lund University; Division of Clinical Geriatrics (A.S., M. Bucci, A.K.N., E.R.-V.), Center for Alzheimer Research, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden; Coma Science Group (A.S.), GIGA-Consciousness, University of Liège; Centre du Cerveau2 (A.S.), University Hospital of Liège, Belgium; Barcelonaβeta Brain Research Center (BBRC) (M.S., G.S., J.D.G.), Pasqual Maragall Foundation; IMIM (Hospital del Mar Medical Research Institute) (M.S., J.D.G.), Barcelona; Centro de Investigación Biomédica en Red de Fragilidad y Envejecimiento Saludable (M.S., J.D.G.), Instituto de Salud Carlos III, Madrid; Universitat Pompeu Fabra (M.S.), Barcelona, Spain; Brain Research Center (I.L.A.), Amsterdam, the Netherlands; IXICO (R.W.), London; Centre for Clinical Brain Sciences (C.R.), University of Edinburgh, United Kingdom; Ace Alzheimer Center Barcelona (M. Boada), Universitat Internacional de Catalunya, Spain; Networking Research Center on Neurodegenerative Diseases (CIBERNED) (M. Boada), Instituto de Salud Carlos III, Madrid, Spain; Alzheimer Center Amsterdam (P.J.V., R.O.), Neurology, Vrije Universiteit Amsterdam, Amsterdam UMC location VUmc; Amsterdam Neuroscience (P.J.V.), Neurodegeneration; Alzheimer Center Limburg (P.J.V.), School for Mental Health and Neuroscience, Maastricht University, the Netherlands; Division of Neurogeriatrics (P.J.V.), Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden; Theme Inflammation and Aging (M. Bucci, A.K.N.), Karolinska University Hospital, Stockholm, Sweden; GE Healthcare (G.F.), Amersham, United Kingdom; Memory Clinic (O.H.), Skåne University Hospital, Malmö, Sweden; and Centre for Medical Image Computing (F.B.), and Queen Square Institute of Neurology, UCL, London, United Kingdom
| | - Robin Wolz
- From the Department of Radiology and Nuclear Medicine (S.E.M., D.V.G., L.L., L.P., A.M.W., F.B., L.E.C.), Vrije Universiteit Amsterdam, Amsterdam University Medical Center, location VUmc; Amsterdam Neuroscience (S.E.M., D.V.G., L.L., L.P., A.M.W., F.B., L.E.C.), Brain Imaging, the Netherlands; Clinical Memory Research Unit (S.E.M., G.S., O.H., R.O., L.E.C.), Department of Clinical Sciences Malmö, Lund University; Division of Clinical Geriatrics (A.S., M. Bucci, A.K.N., E.R.-V.), Center for Alzheimer Research, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden; Coma Science Group (A.S.), GIGA-Consciousness, University of Liège; Centre du Cerveau2 (A.S.), University Hospital of Liège, Belgium; Barcelonaβeta Brain Research Center (BBRC) (M.S., G.S., J.D.G.), Pasqual Maragall Foundation; IMIM (Hospital del Mar Medical Research Institute) (M.S., J.D.G.), Barcelona; Centro de Investigación Biomédica en Red de Fragilidad y Envejecimiento Saludable (M.S., J.D.G.), Instituto de Salud Carlos III, Madrid; Universitat Pompeu Fabra (M.S.), Barcelona, Spain; Brain Research Center (I.L.A.), Amsterdam, the Netherlands; IXICO (R.W.), London; Centre for Clinical Brain Sciences (C.R.), University of Edinburgh, United Kingdom; Ace Alzheimer Center Barcelona (M. Boada), Universitat Internacional de Catalunya, Spain; Networking Research Center on Neurodegenerative Diseases (CIBERNED) (M. Boada), Instituto de Salud Carlos III, Madrid, Spain; Alzheimer Center Amsterdam (P.J.V., R.O.), Neurology, Vrije Universiteit Amsterdam, Amsterdam UMC location VUmc; Amsterdam Neuroscience (P.J.V.), Neurodegeneration; Alzheimer Center Limburg (P.J.V.), School for Mental Health and Neuroscience, Maastricht University, the Netherlands; Division of Neurogeriatrics (P.J.V.), Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden; Theme Inflammation and Aging (M. Bucci, A.K.N.), Karolinska University Hospital, Stockholm, Sweden; GE Healthcare (G.F.), Amersham, United Kingdom; Memory Clinic (O.H.), Skåne University Hospital, Malmö, Sweden; and Centre for Medical Image Computing (F.B.), and Queen Square Institute of Neurology, UCL, London, United Kingdom
| | - Craig Ritchie
- From the Department of Radiology and Nuclear Medicine (S.E.M., D.V.G., L.L., L.P., A.M.W., F.B., L.E.C.), Vrije Universiteit Amsterdam, Amsterdam University Medical Center, location VUmc; Amsterdam Neuroscience (S.E.M., D.V.G., L.L., L.P., A.M.W., F.B., L.E.C.), Brain Imaging, the Netherlands; Clinical Memory Research Unit (S.E.M., G.S., O.H., R.O., L.E.C.), Department of Clinical Sciences Malmö, Lund University; Division of Clinical Geriatrics (A.S., M. Bucci, A.K.N., E.R.-V.), Center for Alzheimer Research, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden; Coma Science Group (A.S.), GIGA-Consciousness, University of Liège; Centre du Cerveau2 (A.S.), University Hospital of Liège, Belgium; Barcelonaβeta Brain Research Center (BBRC) (M.S., G.S., J.D.G.), Pasqual Maragall Foundation; IMIM (Hospital del Mar Medical Research Institute) (M.S., J.D.G.), Barcelona; Centro de Investigación Biomédica en Red de Fragilidad y Envejecimiento Saludable (M.S., J.D.G.), Instituto de Salud Carlos III, Madrid; Universitat Pompeu Fabra (M.S.), Barcelona, Spain; Brain Research Center (I.L.A.), Amsterdam, the Netherlands; IXICO (R.W.), London; Centre for Clinical Brain Sciences (C.R.), University of Edinburgh, United Kingdom; Ace Alzheimer Center Barcelona (M. Boada), Universitat Internacional de Catalunya, Spain; Networking Research Center on Neurodegenerative Diseases (CIBERNED) (M. Boada), Instituto de Salud Carlos III, Madrid, Spain; Alzheimer Center Amsterdam (P.J.V., R.O.), Neurology, Vrije Universiteit Amsterdam, Amsterdam UMC location VUmc; Amsterdam Neuroscience (P.J.V.), Neurodegeneration; Alzheimer Center Limburg (P.J.V.), School for Mental Health and Neuroscience, Maastricht University, the Netherlands; Division of Neurogeriatrics (P.J.V.), Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden; Theme Inflammation and Aging (M. Bucci, A.K.N.), Karolinska University Hospital, Stockholm, Sweden; GE Healthcare (G.F.), Amersham, United Kingdom; Memory Clinic (O.H.), Skåne University Hospital, Malmö, Sweden; and Centre for Medical Image Computing (F.B.), and Queen Square Institute of Neurology, UCL, London, United Kingdom
| | - Mercè Boada
- From the Department of Radiology and Nuclear Medicine (S.E.M., D.V.G., L.L., L.P., A.M.W., F.B., L.E.C.), Vrije Universiteit Amsterdam, Amsterdam University Medical Center, location VUmc; Amsterdam Neuroscience (S.E.M., D.V.G., L.L., L.P., A.M.W., F.B., L.E.C.), Brain Imaging, the Netherlands; Clinical Memory Research Unit (S.E.M., G.S., O.H., R.O., L.E.C.), Department of Clinical Sciences Malmö, Lund University; Division of Clinical Geriatrics (A.S., M. Bucci, A.K.N., E.R.-V.), Center for Alzheimer Research, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden; Coma Science Group (A.S.), GIGA-Consciousness, University of Liège; Centre du Cerveau2 (A.S.), University Hospital of Liège, Belgium; Barcelonaβeta Brain Research Center (BBRC) (M.S., G.S., J.D.G.), Pasqual Maragall Foundation; IMIM (Hospital del Mar Medical Research Institute) (M.S., J.D.G.), Barcelona; Centro de Investigación Biomédica en Red de Fragilidad y Envejecimiento Saludable (M.S., J.D.G.), Instituto de Salud Carlos III, Madrid; Universitat Pompeu Fabra (M.S.), Barcelona, Spain; Brain Research Center (I.L.A.), Amsterdam, the Netherlands; IXICO (R.W.), London; Centre for Clinical Brain Sciences (C.R.), University of Edinburgh, United Kingdom; Ace Alzheimer Center Barcelona (M. Boada), Universitat Internacional de Catalunya, Spain; Networking Research Center on Neurodegenerative Diseases (CIBERNED) (M. Boada), Instituto de Salud Carlos III, Madrid, Spain; Alzheimer Center Amsterdam (P.J.V., R.O.), Neurology, Vrije Universiteit Amsterdam, Amsterdam UMC location VUmc; Amsterdam Neuroscience (P.J.V.), Neurodegeneration; Alzheimer Center Limburg (P.J.V.), School for Mental Health and Neuroscience, Maastricht University, the Netherlands; Division of Neurogeriatrics (P.J.V.), Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden; Theme Inflammation and Aging (M. Bucci, A.K.N.), Karolinska University Hospital, Stockholm, Sweden; GE Healthcare (G.F.), Amersham, United Kingdom; Memory Clinic (O.H.), Skåne University Hospital, Malmö, Sweden; and Centre for Medical Image Computing (F.B.), and Queen Square Institute of Neurology, UCL, London, United Kingdom
| | - Pieter Jelle Visser
- From the Department of Radiology and Nuclear Medicine (S.E.M., D.V.G., L.L., L.P., A.M.W., F.B., L.E.C.), Vrije Universiteit Amsterdam, Amsterdam University Medical Center, location VUmc; Amsterdam Neuroscience (S.E.M., D.V.G., L.L., L.P., A.M.W., F.B., L.E.C.), Brain Imaging, the Netherlands; Clinical Memory Research Unit (S.E.M., G.S., O.H., R.O., L.E.C.), Department of Clinical Sciences Malmö, Lund University; Division of Clinical Geriatrics (A.S., M. Bucci, A.K.N., E.R.-V.), Center for Alzheimer Research, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden; Coma Science Group (A.S.), GIGA-Consciousness, University of Liège; Centre du Cerveau2 (A.S.), University Hospital of Liège, Belgium; Barcelonaβeta Brain Research Center (BBRC) (M.S., G.S., J.D.G.), Pasqual Maragall Foundation; IMIM (Hospital del Mar Medical Research Institute) (M.S., J.D.G.), Barcelona; Centro de Investigación Biomédica en Red de Fragilidad y Envejecimiento Saludable (M.S., J.D.G.), Instituto de Salud Carlos III, Madrid; Universitat Pompeu Fabra (M.S.), Barcelona, Spain; Brain Research Center (I.L.A.), Amsterdam, the Netherlands; IXICO (R.W.), London; Centre for Clinical Brain Sciences (C.R.), University of Edinburgh, United Kingdom; Ace Alzheimer Center Barcelona (M. Boada), Universitat Internacional de Catalunya, Spain; Networking Research Center on Neurodegenerative Diseases (CIBERNED) (M. Boada), Instituto de Salud Carlos III, Madrid, Spain; Alzheimer Center Amsterdam (P.J.V., R.O.), Neurology, Vrije Universiteit Amsterdam, Amsterdam UMC location VUmc; Amsterdam Neuroscience (P.J.V.), Neurodegeneration; Alzheimer Center Limburg (P.J.V.), School for Mental Health and Neuroscience, Maastricht University, the Netherlands; Division of Neurogeriatrics (P.J.V.), Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden; Theme Inflammation and Aging (M. Bucci, A.K.N.), Karolinska University Hospital, Stockholm, Sweden; GE Healthcare (G.F.), Amersham, United Kingdom; Memory Clinic (O.H.), Skåne University Hospital, Malmö, Sweden; and Centre for Medical Image Computing (F.B.), and Queen Square Institute of Neurology, UCL, London, United Kingdom
| | - Marco Bucci
- From the Department of Radiology and Nuclear Medicine (S.E.M., D.V.G., L.L., L.P., A.M.W., F.B., L.E.C.), Vrije Universiteit Amsterdam, Amsterdam University Medical Center, location VUmc; Amsterdam Neuroscience (S.E.M., D.V.G., L.L., L.P., A.M.W., F.B., L.E.C.), Brain Imaging, the Netherlands; Clinical Memory Research Unit (S.E.M., G.S., O.H., R.O., L.E.C.), Department of Clinical Sciences Malmö, Lund University; Division of Clinical Geriatrics (A.S., M. Bucci, A.K.N., E.R.-V.), Center for Alzheimer Research, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden; Coma Science Group (A.S.), GIGA-Consciousness, University of Liège; Centre du Cerveau2 (A.S.), University Hospital of Liège, Belgium; Barcelonaβeta Brain Research Center (BBRC) (M.S., G.S., J.D.G.), Pasqual Maragall Foundation; IMIM (Hospital del Mar Medical Research Institute) (M.S., J.D.G.), Barcelona; Centro de Investigación Biomédica en Red de Fragilidad y Envejecimiento Saludable (M.S., J.D.G.), Instituto de Salud Carlos III, Madrid; Universitat Pompeu Fabra (M.S.), Barcelona, Spain; Brain Research Center (I.L.A.), Amsterdam, the Netherlands; IXICO (R.W.), London; Centre for Clinical Brain Sciences (C.R.), University of Edinburgh, United Kingdom; Ace Alzheimer Center Barcelona (M. Boada), Universitat Internacional de Catalunya, Spain; Networking Research Center on Neurodegenerative Diseases (CIBERNED) (M. Boada), Instituto de Salud Carlos III, Madrid, Spain; Alzheimer Center Amsterdam (P.J.V., R.O.), Neurology, Vrije Universiteit Amsterdam, Amsterdam UMC location VUmc; Amsterdam Neuroscience (P.J.V.), Neurodegeneration; Alzheimer Center Limburg (P.J.V.), School for Mental Health and Neuroscience, Maastricht University, the Netherlands; Division of Neurogeriatrics (P.J.V.), Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden; Theme Inflammation and Aging (M. Bucci, A.K.N.), Karolinska University Hospital, Stockholm, Sweden; GE Healthcare (G.F.), Amersham, United Kingdom; Memory Clinic (O.H.), Skåne University Hospital, Malmö, Sweden; and Centre for Medical Image Computing (F.B.), and Queen Square Institute of Neurology, UCL, London, United Kingdom
| | - Gill Farrar
- From the Department of Radiology and Nuclear Medicine (S.E.M., D.V.G., L.L., L.P., A.M.W., F.B., L.E.C.), Vrije Universiteit Amsterdam, Amsterdam University Medical Center, location VUmc; Amsterdam Neuroscience (S.E.M., D.V.G., L.L., L.P., A.M.W., F.B., L.E.C.), Brain Imaging, the Netherlands; Clinical Memory Research Unit (S.E.M., G.S., O.H., R.O., L.E.C.), Department of Clinical Sciences Malmö, Lund University; Division of Clinical Geriatrics (A.S., M. Bucci, A.K.N., E.R.-V.), Center for Alzheimer Research, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden; Coma Science Group (A.S.), GIGA-Consciousness, University of Liège; Centre du Cerveau2 (A.S.), University Hospital of Liège, Belgium; Barcelonaβeta Brain Research Center (BBRC) (M.S., G.S., J.D.G.), Pasqual Maragall Foundation; IMIM (Hospital del Mar Medical Research Institute) (M.S., J.D.G.), Barcelona; Centro de Investigación Biomédica en Red de Fragilidad y Envejecimiento Saludable (M.S., J.D.G.), Instituto de Salud Carlos III, Madrid; Universitat Pompeu Fabra (M.S.), Barcelona, Spain; Brain Research Center (I.L.A.), Amsterdam, the Netherlands; IXICO (R.W.), London; Centre for Clinical Brain Sciences (C.R.), University of Edinburgh, United Kingdom; Ace Alzheimer Center Barcelona (M. Boada), Universitat Internacional de Catalunya, Spain; Networking Research Center on Neurodegenerative Diseases (CIBERNED) (M. Boada), Instituto de Salud Carlos III, Madrid, Spain; Alzheimer Center Amsterdam (P.J.V., R.O.), Neurology, Vrije Universiteit Amsterdam, Amsterdam UMC location VUmc; Amsterdam Neuroscience (P.J.V.), Neurodegeneration; Alzheimer Center Limburg (P.J.V.), School for Mental Health and Neuroscience, Maastricht University, the Netherlands; Division of Neurogeriatrics (P.J.V.), Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden; Theme Inflammation and Aging (M. Bucci, A.K.N.), Karolinska University Hospital, Stockholm, Sweden; GE Healthcare (G.F.), Amersham, United Kingdom; Memory Clinic (O.H.), Skåne University Hospital, Malmö, Sweden; and Centre for Medical Image Computing (F.B.), and Queen Square Institute of Neurology, UCL, London, United Kingdom
| | - Oskar Hansson
- From the Department of Radiology and Nuclear Medicine (S.E.M., D.V.G., L.L., L.P., A.M.W., F.B., L.E.C.), Vrije Universiteit Amsterdam, Amsterdam University Medical Center, location VUmc; Amsterdam Neuroscience (S.E.M., D.V.G., L.L., L.P., A.M.W., F.B., L.E.C.), Brain Imaging, the Netherlands; Clinical Memory Research Unit (S.E.M., G.S., O.H., R.O., L.E.C.), Department of Clinical Sciences Malmö, Lund University; Division of Clinical Geriatrics (A.S., M. Bucci, A.K.N., E.R.-V.), Center for Alzheimer Research, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden; Coma Science Group (A.S.), GIGA-Consciousness, University of Liège; Centre du Cerveau2 (A.S.), University Hospital of Liège, Belgium; Barcelonaβeta Brain Research Center (BBRC) (M.S., G.S., J.D.G.), Pasqual Maragall Foundation; IMIM (Hospital del Mar Medical Research Institute) (M.S., J.D.G.), Barcelona; Centro de Investigación Biomédica en Red de Fragilidad y Envejecimiento Saludable (M.S., J.D.G.), Instituto de Salud Carlos III, Madrid; Universitat Pompeu Fabra (M.S.), Barcelona, Spain; Brain Research Center (I.L.A.), Amsterdam, the Netherlands; IXICO (R.W.), London; Centre for Clinical Brain Sciences (C.R.), University of Edinburgh, United Kingdom; Ace Alzheimer Center Barcelona (M. Boada), Universitat Internacional de Catalunya, Spain; Networking Research Center on Neurodegenerative Diseases (CIBERNED) (M. Boada), Instituto de Salud Carlos III, Madrid, Spain; Alzheimer Center Amsterdam (P.J.V., R.O.), Neurology, Vrije Universiteit Amsterdam, Amsterdam UMC location VUmc; Amsterdam Neuroscience (P.J.V.), Neurodegeneration; Alzheimer Center Limburg (P.J.V.), School for Mental Health and Neuroscience, Maastricht University, the Netherlands; Division of Neurogeriatrics (P.J.V.), Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden; Theme Inflammation and Aging (M. Bucci, A.K.N.), Karolinska University Hospital, Stockholm, Sweden; GE Healthcare (G.F.), Amersham, United Kingdom; Memory Clinic (O.H.), Skåne University Hospital, Malmö, Sweden; and Centre for Medical Image Computing (F.B.), and Queen Square Institute of Neurology, UCL, London, United Kingdom
| | - Agneta K Nordberg
- From the Department of Radiology and Nuclear Medicine (S.E.M., D.V.G., L.L., L.P., A.M.W., F.B., L.E.C.), Vrije Universiteit Amsterdam, Amsterdam University Medical Center, location VUmc; Amsterdam Neuroscience (S.E.M., D.V.G., L.L., L.P., A.M.W., F.B., L.E.C.), Brain Imaging, the Netherlands; Clinical Memory Research Unit (S.E.M., G.S., O.H., R.O., L.E.C.), Department of Clinical Sciences Malmö, Lund University; Division of Clinical Geriatrics (A.S., M. Bucci, A.K.N., E.R.-V.), Center for Alzheimer Research, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden; Coma Science Group (A.S.), GIGA-Consciousness, University of Liège; Centre du Cerveau2 (A.S.), University Hospital of Liège, Belgium; Barcelonaβeta Brain Research Center (BBRC) (M.S., G.S., J.D.G.), Pasqual Maragall Foundation; IMIM (Hospital del Mar Medical Research Institute) (M.S., J.D.G.), Barcelona; Centro de Investigación Biomédica en Red de Fragilidad y Envejecimiento Saludable (M.S., J.D.G.), Instituto de Salud Carlos III, Madrid; Universitat Pompeu Fabra (M.S.), Barcelona, Spain; Brain Research Center (I.L.A.), Amsterdam, the Netherlands; IXICO (R.W.), London; Centre for Clinical Brain Sciences (C.R.), University of Edinburgh, United Kingdom; Ace Alzheimer Center Barcelona (M. Boada), Universitat Internacional de Catalunya, Spain; Networking Research Center on Neurodegenerative Diseases (CIBERNED) (M. Boada), Instituto de Salud Carlos III, Madrid, Spain; Alzheimer Center Amsterdam (P.J.V., R.O.), Neurology, Vrije Universiteit Amsterdam, Amsterdam UMC location VUmc; Amsterdam Neuroscience (P.J.V.), Neurodegeneration; Alzheimer Center Limburg (P.J.V.), School for Mental Health and Neuroscience, Maastricht University, the Netherlands; Division of Neurogeriatrics (P.J.V.), Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden; Theme Inflammation and Aging (M. Bucci, A.K.N.), Karolinska University Hospital, Stockholm, Sweden; GE Healthcare (G.F.), Amersham, United Kingdom; Memory Clinic (O.H.), Skåne University Hospital, Malmö, Sweden; and Centre for Medical Image Computing (F.B.), and Queen Square Institute of Neurology, UCL, London, United Kingdom
| | - Rik Ossenkoppele
- From the Department of Radiology and Nuclear Medicine (S.E.M., D.V.G., L.L., L.P., A.M.W., F.B., L.E.C.), Vrije Universiteit Amsterdam, Amsterdam University Medical Center, location VUmc; Amsterdam Neuroscience (S.E.M., D.V.G., L.L., L.P., A.M.W., F.B., L.E.C.), Brain Imaging, the Netherlands; Clinical Memory Research Unit (S.E.M., G.S., O.H., R.O., L.E.C.), Department of Clinical Sciences Malmö, Lund University; Division of Clinical Geriatrics (A.S., M. Bucci, A.K.N., E.R.-V.), Center for Alzheimer Research, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden; Coma Science Group (A.S.), GIGA-Consciousness, University of Liège; Centre du Cerveau2 (A.S.), University Hospital of Liège, Belgium; Barcelonaβeta Brain Research Center (BBRC) (M.S., G.S., J.D.G.), Pasqual Maragall Foundation; IMIM (Hospital del Mar Medical Research Institute) (M.S., J.D.G.), Barcelona; Centro de Investigación Biomédica en Red de Fragilidad y Envejecimiento Saludable (M.S., J.D.G.), Instituto de Salud Carlos III, Madrid; Universitat Pompeu Fabra (M.S.), Barcelona, Spain; Brain Research Center (I.L.A.), Amsterdam, the Netherlands; IXICO (R.W.), London; Centre for Clinical Brain Sciences (C.R.), University of Edinburgh, United Kingdom; Ace Alzheimer Center Barcelona (M. Boada), Universitat Internacional de Catalunya, Spain; Networking Research Center on Neurodegenerative Diseases (CIBERNED) (M. Boada), Instituto de Salud Carlos III, Madrid, Spain; Alzheimer Center Amsterdam (P.J.V., R.O.), Neurology, Vrije Universiteit Amsterdam, Amsterdam UMC location VUmc; Amsterdam Neuroscience (P.J.V.), Neurodegeneration; Alzheimer Center Limburg (P.J.V.), School for Mental Health and Neuroscience, Maastricht University, the Netherlands; Division of Neurogeriatrics (P.J.V.), Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden; Theme Inflammation and Aging (M. Bucci, A.K.N.), Karolinska University Hospital, Stockholm, Sweden; GE Healthcare (G.F.), Amersham, United Kingdom; Memory Clinic (O.H.), Skåne University Hospital, Malmö, Sweden; and Centre for Medical Image Computing (F.B.), and Queen Square Institute of Neurology, UCL, London, United Kingdom
| | - Frederik Barkhof
- From the Department of Radiology and Nuclear Medicine (S.E.M., D.V.G., L.L., L.P., A.M.W., F.B., L.E.C.), Vrije Universiteit Amsterdam, Amsterdam University Medical Center, location VUmc; Amsterdam Neuroscience (S.E.M., D.V.G., L.L., L.P., A.M.W., F.B., L.E.C.), Brain Imaging, the Netherlands; Clinical Memory Research Unit (S.E.M., G.S., O.H., R.O., L.E.C.), Department of Clinical Sciences Malmö, Lund University; Division of Clinical Geriatrics (A.S., M. Bucci, A.K.N., E.R.-V.), Center for Alzheimer Research, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden; Coma Science Group (A.S.), GIGA-Consciousness, University of Liège; Centre du Cerveau2 (A.S.), University Hospital of Liège, Belgium; Barcelonaβeta Brain Research Center (BBRC) (M.S., G.S., J.D.G.), Pasqual Maragall Foundation; IMIM (Hospital del Mar Medical Research Institute) (M.S., J.D.G.), Barcelona; Centro de Investigación Biomédica en Red de Fragilidad y Envejecimiento Saludable (M.S., J.D.G.), Instituto de Salud Carlos III, Madrid; Universitat Pompeu Fabra (M.S.), Barcelona, Spain; Brain Research Center (I.L.A.), Amsterdam, the Netherlands; IXICO (R.W.), London; Centre for Clinical Brain Sciences (C.R.), University of Edinburgh, United Kingdom; Ace Alzheimer Center Barcelona (M. Boada), Universitat Internacional de Catalunya, Spain; Networking Research Center on Neurodegenerative Diseases (CIBERNED) (M. Boada), Instituto de Salud Carlos III, Madrid, Spain; Alzheimer Center Amsterdam (P.J.V., R.O.), Neurology, Vrije Universiteit Amsterdam, Amsterdam UMC location VUmc; Amsterdam Neuroscience (P.J.V.), Neurodegeneration; Alzheimer Center Limburg (P.J.V.), School for Mental Health and Neuroscience, Maastricht University, the Netherlands; Division of Neurogeriatrics (P.J.V.), Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden; Theme Inflammation and Aging (M. Bucci, A.K.N.), Karolinska University Hospital, Stockholm, Sweden; GE Healthcare (G.F.), Amersham, United Kingdom; Memory Clinic (O.H.), Skåne University Hospital, Malmö, Sweden; and Centre for Medical Image Computing (F.B.), and Queen Square Institute of Neurology, UCL, London, United Kingdom
| | - Juan Domingo Gispert
- From the Department of Radiology and Nuclear Medicine (S.E.M., D.V.G., L.L., L.P., A.M.W., F.B., L.E.C.), Vrije Universiteit Amsterdam, Amsterdam University Medical Center, location VUmc; Amsterdam Neuroscience (S.E.M., D.V.G., L.L., L.P., A.M.W., F.B., L.E.C.), Brain Imaging, the Netherlands; Clinical Memory Research Unit (S.E.M., G.S., O.H., R.O., L.E.C.), Department of Clinical Sciences Malmö, Lund University; Division of Clinical Geriatrics (A.S., M. Bucci, A.K.N., E.R.-V.), Center for Alzheimer Research, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden; Coma Science Group (A.S.), GIGA-Consciousness, University of Liège; Centre du Cerveau2 (A.S.), University Hospital of Liège, Belgium; Barcelonaβeta Brain Research Center (BBRC) (M.S., G.S., J.D.G.), Pasqual Maragall Foundation; IMIM (Hospital del Mar Medical Research Institute) (M.S., J.D.G.), Barcelona; Centro de Investigación Biomédica en Red de Fragilidad y Envejecimiento Saludable (M.S., J.D.G.), Instituto de Salud Carlos III, Madrid; Universitat Pompeu Fabra (M.S.), Barcelona, Spain; Brain Research Center (I.L.A.), Amsterdam, the Netherlands; IXICO (R.W.), London; Centre for Clinical Brain Sciences (C.R.), University of Edinburgh, United Kingdom; Ace Alzheimer Center Barcelona (M. Boada), Universitat Internacional de Catalunya, Spain; Networking Research Center on Neurodegenerative Diseases (CIBERNED) (M. Boada), Instituto de Salud Carlos III, Madrid, Spain; Alzheimer Center Amsterdam (P.J.V., R.O.), Neurology, Vrije Universiteit Amsterdam, Amsterdam UMC location VUmc; Amsterdam Neuroscience (P.J.V.), Neurodegeneration; Alzheimer Center Limburg (P.J.V.), School for Mental Health and Neuroscience, Maastricht University, the Netherlands; Division of Neurogeriatrics (P.J.V.), Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden; Theme Inflammation and Aging (M. Bucci, A.K.N.), Karolinska University Hospital, Stockholm, Sweden; GE Healthcare (G.F.), Amersham, United Kingdom; Memory Clinic (O.H.), Skåne University Hospital, Malmö, Sweden; and Centre for Medical Image Computing (F.B.), and Queen Square Institute of Neurology, UCL, London, United Kingdom
| | - Elena Rodriguez-Vieitez
- From the Department of Radiology and Nuclear Medicine (S.E.M., D.V.G., L.L., L.P., A.M.W., F.B., L.E.C.), Vrije Universiteit Amsterdam, Amsterdam University Medical Center, location VUmc; Amsterdam Neuroscience (S.E.M., D.V.G., L.L., L.P., A.M.W., F.B., L.E.C.), Brain Imaging, the Netherlands; Clinical Memory Research Unit (S.E.M., G.S., O.H., R.O., L.E.C.), Department of Clinical Sciences Malmö, Lund University; Division of Clinical Geriatrics (A.S., M. Bucci, A.K.N., E.R.-V.), Center for Alzheimer Research, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden; Coma Science Group (A.S.), GIGA-Consciousness, University of Liège; Centre du Cerveau2 (A.S.), University Hospital of Liège, Belgium; Barcelonaβeta Brain Research Center (BBRC) (M.S., G.S., J.D.G.), Pasqual Maragall Foundation; IMIM (Hospital del Mar Medical Research Institute) (M.S., J.D.G.), Barcelona; Centro de Investigación Biomédica en Red de Fragilidad y Envejecimiento Saludable (M.S., J.D.G.), Instituto de Salud Carlos III, Madrid; Universitat Pompeu Fabra (M.S.), Barcelona, Spain; Brain Research Center (I.L.A.), Amsterdam, the Netherlands; IXICO (R.W.), London; Centre for Clinical Brain Sciences (C.R.), University of Edinburgh, United Kingdom; Ace Alzheimer Center Barcelona (M. Boada), Universitat Internacional de Catalunya, Spain; Networking Research Center on Neurodegenerative Diseases (CIBERNED) (M. Boada), Instituto de Salud Carlos III, Madrid, Spain; Alzheimer Center Amsterdam (P.J.V., R.O.), Neurology, Vrije Universiteit Amsterdam, Amsterdam UMC location VUmc; Amsterdam Neuroscience (P.J.V.), Neurodegeneration; Alzheimer Center Limburg (P.J.V.), School for Mental Health and Neuroscience, Maastricht University, the Netherlands; Division of Neurogeriatrics (P.J.V.), Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden; Theme Inflammation and Aging (M. Bucci, A.K.N.), Karolinska University Hospital, Stockholm, Sweden; GE Healthcare (G.F.), Amersham, United Kingdom; Memory Clinic (O.H.), Skåne University Hospital, Malmö, Sweden; and Centre for Medical Image Computing (F.B.), and Queen Square Institute of Neurology, UCL, London, United Kingdom
| | - Lyduine E Collij
- From the Department of Radiology and Nuclear Medicine (S.E.M., D.V.G., L.L., L.P., A.M.W., F.B., L.E.C.), Vrije Universiteit Amsterdam, Amsterdam University Medical Center, location VUmc; Amsterdam Neuroscience (S.E.M., D.V.G., L.L., L.P., A.M.W., F.B., L.E.C.), Brain Imaging, the Netherlands; Clinical Memory Research Unit (S.E.M., G.S., O.H., R.O., L.E.C.), Department of Clinical Sciences Malmö, Lund University; Division of Clinical Geriatrics (A.S., M. Bucci, A.K.N., E.R.-V.), Center for Alzheimer Research, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden; Coma Science Group (A.S.), GIGA-Consciousness, University of Liège; Centre du Cerveau2 (A.S.), University Hospital of Liège, Belgium; Barcelonaβeta Brain Research Center (BBRC) (M.S., G.S., J.D.G.), Pasqual Maragall Foundation; IMIM (Hospital del Mar Medical Research Institute) (M.S., J.D.G.), Barcelona; Centro de Investigación Biomédica en Red de Fragilidad y Envejecimiento Saludable (M.S., J.D.G.), Instituto de Salud Carlos III, Madrid; Universitat Pompeu Fabra (M.S.), Barcelona, Spain; Brain Research Center (I.L.A.), Amsterdam, the Netherlands; IXICO (R.W.), London; Centre for Clinical Brain Sciences (C.R.), University of Edinburgh, United Kingdom; Ace Alzheimer Center Barcelona (M. Boada), Universitat Internacional de Catalunya, Spain; Networking Research Center on Neurodegenerative Diseases (CIBERNED) (M. Boada), Instituto de Salud Carlos III, Madrid, Spain; Alzheimer Center Amsterdam (P.J.V., R.O.), Neurology, Vrije Universiteit Amsterdam, Amsterdam UMC location VUmc; Amsterdam Neuroscience (P.J.V.), Neurodegeneration; Alzheimer Center Limburg (P.J.V.), School for Mental Health and Neuroscience, Maastricht University, the Netherlands; Division of Neurogeriatrics (P.J.V.), Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden; Theme Inflammation and Aging (M. Bucci, A.K.N.), Karolinska University Hospital, Stockholm, Sweden; GE Healthcare (G.F.), Amersham, United Kingdom; Memory Clinic (O.H.), Skåne University Hospital, Malmö, Sweden; and Centre for Medical Image Computing (F.B.), and Queen Square Institute of Neurology, UCL, London, United Kingdom
| |
Collapse
|
26
|
Satarker S, Gurram PC, Nassar A, Manandhar S, Vibhavari R, Yarlagadda DL, Mudgal J, Lewis S, Arora D, Nampoothiri M. Evaluating the Role of N-Acetyl-L-Tryptophan in the Aβ 1-42-Induced Neuroinflammation and Cognitive Decline in Alzheimer's Disease. Mol Neurobiol 2024; 61:4421-4440. [PMID: 38091207 PMCID: PMC11236887 DOI: 10.1007/s12035-023-03844-4] [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: 09/27/2023] [Accepted: 11/29/2023] [Indexed: 07/11/2024]
Abstract
Alzheimer's disease (AD), a neurodegenerative condition previously known to affect the older population, is also now seen in younger individuals. AD is often associated with cognitive decline and neuroinflammation elevation primarily due to amyloid β (Aβ) accumulation. Multiple pathological complications in AD call for therapies with a wide range of neuroprotection. Our study aims to evaluate the effect of N-acetyl-L-tryptophan (NAT) in ameliorating the cognitive decline and neuroinflammation induced by Aβ 1-42 oligomers and to determine the therapeutic concentration of NAT in the brain. We administered Aβ 1-42 oligomers in rats via intracerebroventricular (i.c.v.) injection to induce AD-like conditions. The NAT-treated animals lowered the cognitive decline in the Morris water maze characterized by shorter escape latency and increased path efficiency and platform entries. Interestingly, the hippocampus and frontal cortex showed downregulation of tumor necrosis factor, interleukin-6, and substance P levels. NAT treatment also reduced acetylcholinesterase activity and total and phosphorylated nuclear factor kappa B and Tau levels. Lastly, we observed upregulation of cAMP response element-binding protein 1 (CREB1) signaling. Surprisingly, our HPLC method was not sensitive enough to detect the therapeutic levels of NAT in the brain, possibly due to NAT concentrations being below the lowest limit of quantification of our validated method. To summarize, the administration of NAT significantly lowered cognitive decline, neuroinflammatory pathways, and Tau protein and triggered the upregulation of CREB1 signaling, suggesting its neuroprotective role in AD-like conditions.
Collapse
Affiliation(s)
- Sairaj Satarker
- Department of Pharmacology, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, Karnataka, 576104, India
| | - Prasada Chowdari Gurram
- Department of Pharmacology, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, Karnataka, 576104, India
| | - Ajmal Nassar
- Department of Pharmacology, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, Karnataka, 576104, India
| | - Suman Manandhar
- Department of Pharmacology, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, Karnataka, 576104, India
| | - Rja Vibhavari
- Department of Pharmacology, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, Karnataka, 576104, India
| | - Dani Lakshman Yarlagadda
- Department of Pharmaceutical Quality Assurance, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, Karnataka, 576104, India
| | - Jayesh Mudgal
- Department of Pharmacology, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, Karnataka, 576104, India
| | - Shaila Lewis
- Department of Pharmaceutics, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, Karnataka, 576104, India
| | - Devinder Arora
- Department of Pharmacology, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, Karnataka, 576104, India
- School of Pharmacy and Medical Sciences, Griffith University, QLD, Gold Coast, 4222, Australia
| | - Madhavan Nampoothiri
- Department of Pharmacology, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, Karnataka, 576104, India.
| |
Collapse
|
27
|
Smardz P, Anila MM, Rogowski P, Li MS, Różycki B, Krupa P. A Practical Guide to All-Atom and Coarse-Grained Molecular Dynamics Simulations Using Amber and Gromacs: A Case Study of Disulfide-Bond Impact on the Intrinsically Disordered Amyloid Beta. Int J Mol Sci 2024; 25:6698. [PMID: 38928405 PMCID: PMC11204378 DOI: 10.3390/ijms25126698] [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: 05/09/2024] [Revised: 06/12/2024] [Accepted: 06/13/2024] [Indexed: 06/28/2024] Open
Abstract
Intrinsically disordered proteins (IDPs) pose challenges to conventional experimental techniques due to their large-scale conformational fluctuations and transient structural elements. This work presents computational methods for studying IDPs at various resolutions using the Amber and Gromacs packages with both all-atom (Amber ff19SB with the OPC water model) and coarse-grained (Martini 3 and SIRAH) approaches. The effectiveness of these methodologies is demonstrated by examining the monomeric form of amyloid-β (Aβ42), an IDP, with and without disulfide bonds at different resolutions. Our results clearly show that the addition of a disulfide bond decreases the β-content of Aβ42; however, it increases the tendency of the monomeric Aβ42 to form fibril-like conformations, explaining the various aggregation rates observed in experiments. Moreover, analysis of the monomeric Aβ42 compactness, secondary structure content, and comparison between calculated and experimental chemical shifts demonstrates that all three methods provide a reasonable choice to study IDPs; however, coarse-grained approaches may lack some atomistic details, such as secondary structure recognition, due to the simplifications used. In general, this study not only explains the role of disulfide bonds in Aβ42 but also provides a step-by-step protocol for setting up, conducting, and analyzing molecular dynamics (MD) simulations, which is adaptable for studying other biomacromolecules, including folded and disordered proteins and peptides.
Collapse
Affiliation(s)
| | | | | | | | | | - Pawel Krupa
- Institute of Physics Polish Academy of Sciences, Al. Lotników 32/46, 02-668 Warsaw, Poland; (P.S.); (M.M.A.); (P.R.); (M.S.L.); (B.R.)
| |
Collapse
|
28
|
Yin X, Zhou H, Cao T, Yang X, Meng F, Dai X, Wang Y, Li S, Zhai W, Yang Z, Chen N, Zhou R. Rational Design of Dual-Functionalized Gd@C 82 Nanoparticles to Relieve Neuronal Cytotoxicity in Alzheimer's Disease via Inhibition of Aβ Aggregation. ACS NANO 2024; 18:15416-15431. [PMID: 38840269 DOI: 10.1021/acsnano.3c08823] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/07/2024]
Abstract
The accumulation of amyloid-β (Aβ) peptides is a major hallmark of Alzheimer's disease (AD) and plays a crucial role in its pathogenesis. Particularly, the structured oligomeric species rich in β-sheet formations were implicated in neuronal organelle damage. Addressing this formidable challenge requires identifying candidates capable of inhibiting peptide aggregation or disaggregating preformed oligomers for effective antiaggregation-based AD therapy. Here, we present a dual-functional nanoinhibitor meticulously designed to target the aggregation driving force and amyloid fibril spatial structure. Leveraging the exceptional structural stability and facile tailoring capability of endohedral metallofullerene Gd@C82, we introduce desired hydrogen-binding sites and charged groups, which are abundant on its surface for specific designs. Impressively, these designs endow the resultant functionalized-Gd@C82 nanoparticles (f-Gd@C82 NPs) with high capability of redirecting peptide self-assembly toward disordered, off-pathway species, obstructing the early growth of protofibrils, and disaggregating the preformed well-ordered protofibrils or even mature Aβ fibrils. This results in considerable alleviation of Aβ peptide-induced neuronal cytotoxicity, rescuing neuronal death and synaptic loss in primary neuron models. Notably, these modifications significantly improved the dispersibility of f-Gd@C82 NPs, thus substantially enhancing its bioavailability. Moreover, f-Gd@C82 NPs demonstrate excellent cytocompatibility with various cell lines and possess the ability to penetrate the blood-brain barrier in mice. Large-scale molecular dynamics simulations illuminate the inhibition and disaggregation mechanisms. Our design successfully overcomes the limitations of other nanocandidates, which often overly rely on hydrophobic interactions or photothermal conversion properties, and offers a viable direction for developing anti-AD agents through the inhibition and even reversal of Aβ aggregation.
Collapse
Affiliation(s)
- Xiuhua Yin
- Institute of Quantitative Biology, Shanghai Institute for Advanced Study, College of Life Sciences, Zhejiang University, Hangzhou 310027, China
- State Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X), Soochow University, Suzhou 215123, China
| | - Hong Zhou
- Institute of Quantitative Biology, Shanghai Institute for Advanced Study, College of Life Sciences, Zhejiang University, Hangzhou 310027, China
| | - Tiantian Cao
- College of Chemistry, Chemical Engineering and Materials Science, Soochow University, Suzhou 215123, China
- Suzhou Institute of Trade and Commerce, Suzhou 215009, China
| | - Xiner Yang
- State Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X), Soochow University, Suzhou 215123, China
| | - Fei Meng
- State Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X), Soochow University, Suzhou 215123, China
| | - Xing Dai
- State Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X), Soochow University, Suzhou 215123, China
| | - Yifan Wang
- State Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X), Soochow University, Suzhou 215123, China
| | - Sijie Li
- State Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X), Soochow University, Suzhou 215123, China
| | - Wangsong Zhai
- State Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X), Soochow University, Suzhou 215123, China
| | - Zaixing Yang
- Institute of Quantitative Biology, Shanghai Institute for Advanced Study, College of Life Sciences, Zhejiang University, Hangzhou 310027, China
- State Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X), Soochow University, Suzhou 215123, China
| | - Ning Chen
- College of Chemistry, Chemical Engineering and Materials Science, Soochow University, Suzhou 215123, China
| | - Ruhong Zhou
- Institute of Quantitative Biology, Shanghai Institute for Advanced Study, College of Life Sciences, Zhejiang University, Hangzhou 310027, China
- Department of Chemistry, Columbia University, New York, New York 10027, United States
| |
Collapse
|
29
|
Pan M, Wu Y, Sun C, Ma H, Ye X, Li X. Polygonati Rhizoma: A review on the extraction, purification, structural characterization, biosynthesis of the main secondary metabolites and anti-aging effects. JOURNAL OF ETHNOPHARMACOLOGY 2024; 327:118002. [PMID: 38437890 DOI: 10.1016/j.jep.2024.118002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Revised: 02/13/2024] [Accepted: 02/29/2024] [Indexed: 03/06/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Polygonati Rhizome (PR) is a plant that is extensively widespread in the temperate zones of the Northern Hemisphere. It is a member of the Polygonatum family of Asparagaceae. PR exhibits diverse pharmacological effects and finds applications in ethnopharmacology, serving as a potent tonic for more than two millennia. PR's compounds endow it with various pharmacological properties, including anti-aging, antioxidant, anti-fatigue, anti-inflammatory, and sleep-enhancing effects, as well as therapeutic potential for osteoporosis and age-related diseases. AIM OF THE STUDY This review seeks to offer a thorough overview of the processing, purification, extraction, structural characterization, and biosynthesis pathways of PR. Furthermore, it delves into the anti-aging mechanism of PR, using organ protection as an entry point. MATERIALS AND METHODS Information on PR was obtained from scientific databases (Google Scholar, Web of Science, ScienceDirect, SciFinder, PubMed, CNKI) and books, doctoral theses, and master's dissertations. RESULTS In this investigation, 49 polysaccharides were extracted from PR, and the impact of various processing, extraction, and purification techniques on the structure and activity of these polysaccharides was evaluated. Additionally, 163 saponins and 46 flavonoids were identified, and three key biosynthesis pathways of secondary metabolites were outlined. Notably, PR and Polygonat Rhizomai polysaccharides (PRP) exhibit remarkable protective effects against age-induced injuries to the brain, liver, kidney, intestine, heart, and vessels, thereby promoting longevity and ameliorating the aging process. CONCLUSIONS PR, a culinary and therapeutic herb, is rich in active components and pharmacological activities. Based on this review, PR plays a meaningful role in lifespan extension and anti-aging, which can be attributed to PRP. Future research should delve deeper into the structural aspects of PRP that underlie its anti-aging effects and explore potential synergistic interactions with other compounds. Moreover, exploring the potential applications of PR in functional foods and pharmaceutical formulations is recommended to advance the development of industries and resources focused on healthy aging.
Collapse
Affiliation(s)
- Miao Pan
- Engineering Research Center of Coptis Development and Utilization (Ministry of Education), College of Pharmaceutical Sciences, Southwest University, Chongqing, China.
| | - Yajing Wu
- Engineering Research Center of Coptis Development and Utilization (Ministry of Education), College of Pharmaceutical Sciences, Southwest University, Chongqing, China.
| | - Chunyong Sun
- Engineering Research Center of Coptis Development and Utilization (Ministry of Education), College of Pharmaceutical Sciences, Southwest University, Chongqing, China.
| | - Hang Ma
- Engineering Research Center of Coptis Development and Utilization (Ministry of Education), College of Pharmaceutical Sciences, Southwest University, Chongqing, China.
| | - Xiaoli Ye
- School of Life Sciences, Southwest University, Chongqing, 400715, China.
| | - Xuegang Li
- Engineering Research Center of Coptis Development and Utilization (Ministry of Education), College of Pharmaceutical Sciences, Southwest University, Chongqing, China.
| |
Collapse
|
30
|
Wilson DL, Carreon A, Chinnam S, Sharifan H, Ahlawat J, Narayan M. Screening Carbon Nano Materials for Preventing Amyloid Protein Aggregation by Adopting a Facile Method. Cell Biochem Biophys 2024; 82:1389-1395. [PMID: 38802601 DOI: 10.1007/s12013-024-01293-x] [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] [Accepted: 04/24/2024] [Indexed: 05/29/2024]
Abstract
The soluble-to-toxic transformation of intrinsically disordered amyloidogenic proteins such as amyloid beta (Aβ), α-synuclein, mutant Huntingtin Protein (mHTT) and islet amyloid polypeptide (IAPP) among others are associated with disorders such as Alzheimer's disease (AD), Parkinson's disease (PD), Huntington's disease (HD) and Type 2 Diabetes (T2D), respectively. The dissolution of mature fibrils and toxic amyloidogenic intermediates, including oligomers, continues to be the pinnacle in the treatment of neurodegenerative disorders. Yet, methods to effectively and quantitatively report on the interconversion between amyloid monomers, oligomers and mature fibrils fall short. Here we describe a simplified method that implements the use of gel electrophoresis to address the transformation between soluble monomeric amyloid proteins and mature amyloid fibrils. The technique implements an optimized but well-known, simple, inexpensive, and quantitative assessment previously used to assess the oligomerization of amyloid monomers and subsequent amyloid fibrils. This method facilitates the screening of small molecules that disintegrate oligomers and fibrils into monomers, dimers, and trimers and/or retain amyloid proteins in their monomeric forms. Most importantly, our optimized method diminishes existing barriers associated with existing (alternative) techniques to evaluate fibril formation and intervention.
Collapse
Affiliation(s)
- Daisy L Wilson
- The Environmental Science & Engineering Program, The University of Texas at El Paso, El Paso, TX, 79968, USA
| | - Ana Carreon
- Department of Chemistry and Biochemistry, the University of Texas at El Paso (UTEP), El Paso, TX, 79968, USA
| | - Sampath Chinnam
- Department of Chemistry, M.S. Ramaiah Institute of Technology (Autonoumous Institution, Affiliated to Visvesvaraya Technological University, Belgaum), Bengaluru, Karnataka, 560054, India
| | - Hamidreza Sharifan
- Department of Chemistry and Biochemistry, the University of Texas at El Paso (UTEP), El Paso, TX, 79968, USA
| | - Jyoti Ahlawat
- Department of Chemistry and Biochemistry, the University of Texas at El Paso (UTEP), El Paso, TX, 79968, USA.
| | - Mahesh Narayan
- Department of Chemistry and Biochemistry, the University of Texas at El Paso (UTEP), El Paso, TX, 79968, USA.
| |
Collapse
|
31
|
Nguyen DLB, Okolicsanyi RK, Haupt LM. Heparan sulfate proteoglycans: Mediators of cellular and molecular Alzheimer's disease pathogenic factors via tunnelling nanotubes? Mol Cell Neurosci 2024; 129:103936. [PMID: 38750678 DOI: 10.1016/j.mcn.2024.103936] [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: 03/07/2024] [Revised: 04/14/2024] [Accepted: 05/01/2024] [Indexed: 05/19/2024] Open
Abstract
Neurological disorders impact around one billion individuals globally (15 % approx.), with significant implications for disability and mortality with their impact in Australia currently amounts to 6.8 million deaths annually. Heparan sulfate proteoglycans (HSPGs) are complex extracellular molecules implicated in promoting Tau fibril formation resulting in Tau tangles, a hallmark of Alzheimer's disease (AD). HSPG-Tau protein interactions contribute to various AD stages via aggregation, toxicity, and clearance, largely via interactions with the glypican 1 and syndecan 3 core proteins. The tunnelling nanotubes (TNTs) pathway is emerging as a facilitator of intercellular molecule transport, including Tau and Amyloid β proteins, across extensive distances. While current TNT-associated evidence primarily stems from cancer models, their role in Tau propagation and its effects on recipient cells remain unclear. This review explores the interplay of TNTs, HSPGs, and AD-related factors and proposes that HSPGs influence TNT formation in neurodegenerative conditions such as AD.
Collapse
Affiliation(s)
- Duy L B Nguyen
- Stem Cell and Neurogenesis Group, Genomics Research Centre, Centre for Genomics and Personalised Health, School of Biomedical Sciences, Queensland University of Technology (QUT), 60 Musk Ave., Kelvin Grove, Queensland 4059, Australia
| | - Rachel K Okolicsanyi
- Stem Cell and Neurogenesis Group, Genomics Research Centre, Centre for Genomics and Personalised Health, School of Biomedical Sciences, Queensland University of Technology (QUT), 60 Musk Ave., Kelvin Grove, Queensland 4059, Australia; ARC Training Centre for Cell and Tissue Engineering Technologies, Queensland University of Technology (QUT), Australia
| | - Larisa M Haupt
- Stem Cell and Neurogenesis Group, Genomics Research Centre, Centre for Genomics and Personalised Health, School of Biomedical Sciences, Queensland University of Technology (QUT), 60 Musk Ave., Kelvin Grove, Queensland 4059, Australia; Centre for Biomedical Technologies, Queensland University of Technology (QUT), 60 Musk Ave., Kelvin Grove, QLD 4059, Australia; ARC Training Centre for Cell and Tissue Engineering Technologies, Queensland University of Technology (QUT), Australia; Max Planck Queensland Centre for the Materials Sciences of Extracellular Matrices, Queensland University of Technology (QUT), Australia.
| |
Collapse
|
32
|
Jarero-Basulto JJ, Gasca-Martínez Y, Rivera-Cervantes MC, Gasca-Martínez D, Carrillo-González NJ, Beas-Zárate C, Gudiño-Cabrera G. Cytotoxic Effect of Amyloid-β1-42 Oligomers on Endoplasmic Reticulum and Golgi Apparatus Arrangement in SH-SY5Y Neuroblastoma Cells. NEUROSCI 2024; 5:141-157. [PMID: 39483494 PMCID: PMC11469764 DOI: 10.3390/neurosci5020010] [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: 03/19/2024] [Revised: 04/28/2024] [Accepted: 04/30/2024] [Indexed: 11/03/2024] Open
Abstract
Amyloid-β oligomers are a cytotoxic structure that is key for the establishment of the beginning stages of Alzheimer's disease (AD). These structures promote subcellular alterations that cause synaptic dysfunction, loss of cell communication, and even cell death, generating cognitive deficits. The aim of this study was to investigate the cytotoxic effects of amyloid-β1-42 oligomers (AβOs) on the membranous organelles involved in protein processing: the endoplasmic reticulum (ER) and Golgi apparatus (GA). The results obtained with 10 μM AβOs in SH-SY5Y neuroblastoma cells showed that oligomeric structures are more toxic than monomers because they cause cell viability to decrease as exposure time increases. Survivor cells were analyzed to further understand the toxic effects of AβOs on intracellular organelles. Survivor cells showed morphological alterations associated with abnormal cytoskeleton modification 72-96 h after exposure to AβOs. Moreover, the ER and GA presented rearrangement throughout the cytoplasmic space, which could be attributed to a lack of constitutive protein processing or to previous abnormal cytoskeleton modification. Interestingly, the disorganization of both ER and GA organelles exposed to AβOs is likely an early pathological alteration that could be related to aberrant protein processing and accumulation in AD.
Collapse
Affiliation(s)
- José J Jarero-Basulto
- Cellular Neurobiology Laboratory, Cell and Molecular Biology Department, University Center of Biological and Agricultural Sciences (CUCBA), University of Guadalajara, Zapopan 45220, Mexico; (J.J.J.-B.); (M.C.R.-C.)
| | - Yadira Gasca-Martínez
- Development and Neural Regeneration Laboratory, Cell and Molecular Biology Department, University Center of Biological and Agricultural Sciences (CUCBA), University of Guadalajara, Zapopan 45220, Mexico; (Y.G.-M.); (N.J.C.-G.)
| | - Martha C Rivera-Cervantes
- Cellular Neurobiology Laboratory, Cell and Molecular Biology Department, University Center of Biological and Agricultural Sciences (CUCBA), University of Guadalajara, Zapopan 45220, Mexico; (J.J.J.-B.); (M.C.R.-C.)
| | - Deisy Gasca-Martínez
- Behavioral Analysis Unit, Neurobiology Institute, Campus UNAM, Juriquilla 76230, Mexico;
| | - Nidia Jannette Carrillo-González
- Development and Neural Regeneration Laboratory, Cell and Molecular Biology Department, University Center of Biological and Agricultural Sciences (CUCBA), University of Guadalajara, Zapopan 45220, Mexico; (Y.G.-M.); (N.J.C.-G.)
| | - Carlos Beas-Zárate
- Neurobiotechnology Laboratory, Cell and Molecular Biology Department, University Center of Biological and Agricultural Sciences (CUCBA), University of Guadalajara, Zapopan 45220, Mexico;
| | - Graciela Gudiño-Cabrera
- Development and Neural Regeneration Laboratory, Cell and Molecular Biology Department, University Center of Biological and Agricultural Sciences (CUCBA), University of Guadalajara, Zapopan 45220, Mexico; (Y.G.-M.); (N.J.C.-G.)
| |
Collapse
|
33
|
Deng J, Liu B, Tao Q, Luo Y, Zhu Y, Huang X, Yue F. The Co-oligomers of Aβ42 and Human Islet Amyloid Polypeptide Exacerbate Neurotoxicity and Alzheimer-like Pathology at Cellular Level. Neuroscience 2024; 547:37-55. [PMID: 38604526 DOI: 10.1016/j.neuroscience.2024.04.002] [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: 01/26/2024] [Revised: 03/28/2024] [Accepted: 04/04/2024] [Indexed: 04/13/2024]
Abstract
The Aβ hypothesis has long been central to Alzheimer's disease (AD) theory, with a recent surge in attention following drug approvals targeting Aβ plaque clearance. Aβ42 oligomers (AβO) are key neurotoxins. While β-amyloid (Aβ) buildup is a hallmark of AD, postmortem brain analyses have unveiled human islet amyloid polypeptide (hIAPP) deposition in AD patients, suggesting a potential role in Alzheimer's pathology. This study investigates the neurotoxic effects of co-aggregates of Aβ42 and hIAPP, specifically focusing on their impact on cell survival, apoptosis, and AD-like pathology. We analyzed and compared the impact of AβO and Aβ42-hIAPP on cell survival in SH-SY5Y cells, apoptosis and inducing AD-like pathology in glutamatergic neurons. Aβ42-hIAPP co-oligomers exhibited significantly greater toxicity, causing 2.3-3.5 times higher cell death compared to AβO alone. Furthermore, apoptosis rates were significantly exacerbated in glutamatergic neurons when exposed to Aβ42-hIAPP co-oligomers. The study also revealed that Aβ42-hIAPP co-oligomers induced typical AD-like pathology in glutamatergic neurons, including the presence of Aβ deposits (detected by 6E10 and 4G8 immunofluorescence) and alterations in tau protein (changes in total tau HT7, phosphorylated tau AT8, AT180). Notably, Aβ42-hIAPP co-oligomers induced a more severe AD pathology compared to AβO alone. These findings provide compelling evidence for the heightened toxicity of Aβ42-hIAPP co-oligomers on neurons and their role in exacerbating AD pathology. The study contributes novel insights into the pathogenesis of Alzheimer's disease, highlighting the potential involvement of hIAPP in AD pathology. Together, these findings offer novel insights into AD pathogenesis and routes for constructing animal models.
Collapse
Affiliation(s)
- Jiajun Deng
- Key Laboratory of Biomedical Engineering of Hainan Province, School of Biomedical Engineering, Hainan University, Sanya 572025, China; Collaborative Innovation Center of One Health Institute, Hainan University. Haikou 570228, China
| | - Bin Liu
- Key Laboratory of Biomedical Engineering of Hainan Province, School of Biomedical Engineering, Hainan University, Sanya 572025, China; Collaborative Innovation Center of One Health Institute, Hainan University. Haikou 570228, China
| | - Qian Tao
- State Key Laboratory of Primate Biomedical Research, Kunming University of Science and Technology, Kunming 650000, China
| | - Yanyu Luo
- Key Laboratory of Biomedical Engineering of Hainan Province, School of Biomedical Engineering, Hainan University, Sanya 572025, China; Collaborative Innovation Center of One Health Institute, Hainan University. Haikou 570228, China
| | - Yi Zhu
- Key Laboratory of Biomedical Engineering of Hainan Province, School of Biomedical Engineering, Hainan University, Sanya 572025, China; Collaborative Innovation Center of One Health Institute, Hainan University. Haikou 570228, China
| | - Xinxin Huang
- Key Laboratory of Biomedical Engineering of Hainan Province, School of Biomedical Engineering, Hainan University, Sanya 572025, China; Collaborative Innovation Center of One Health Institute, Hainan University. Haikou 570228, China
| | - Feng Yue
- Key Laboratory of Biomedical Engineering of Hainan Province, School of Biomedical Engineering, Hainan University, Sanya 572025, China; Collaborative Innovation Center of One Health Institute, Hainan University. Haikou 570228, China.
| |
Collapse
|
34
|
Karunarathne K, Kee TR, Jeon H, Cazzaro S, Gamage YI, Pan J, Woo JAA, Kang DE, Muschol M. Crystal Violet Selectively Detects Aβ Oligomers but Not Fibrils In Vitro and in Alzheimer's Disease Brain Tissue. Biomolecules 2024; 14:615. [PMID: 38927020 PMCID: PMC11201545 DOI: 10.3390/biom14060615] [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: 03/28/2024] [Revised: 05/02/2024] [Accepted: 05/05/2024] [Indexed: 06/28/2024] Open
Abstract
Deposition of extracellular Amyloid Beta (Aβ) and intracellular tau fibrils in post-mortem brains remains the only way to conclusively confirm cases of Alzheimer's Disease (AD). Substantial evidence, though, implicates small globular oligomers instead of fibrils as relevant biomarkers of, and critical contributors to, the clinical symptoms of AD. Efforts to verify and utilize amyloid oligomers as AD biomarkers in vivo have been limited by the near-exclusive dependence on conformation-selective antibodies for oligomer detection. While antibodies have yielded critical evidence for the role of both Aβ and tau oligomers in AD, they are not suitable for imaging amyloid oligomers in vivo. Therefore, it would be desirable to identify a set of oligomer-selective small molecules for subsequent development into Positron Emission Tomography (PET) probes. Using a kinetics-based screening assay, we confirm that the triarylmethane dye Crystal Violet (CV) is oligomer-selective for Aβ42 oligomers (AβOs) grown under near-physiological solution conditions in vitro. In postmortem brains of an AD mouse model and human AD patients, we demonstrate that A11 antibody-positive oligomers but not Thioflavin S (ThioS)-positive fibrils colocalize with CV staining, confirming in vitro results. Therefore, our kinetic screen represents a robust approach for identifying new classes of small molecules as candidates for oligomer-selective dyes (OSDs). Such OSDs, in turn, provide promising starting points for the development of PET probes for pre-mortem imaging of oligomer deposits in humans.
Collapse
Affiliation(s)
| | - Teresa R. Kee
- Department of Molecular Medicine, USF Health, Morsani College of Medicine, Tampa, FL 33620, USA
- Department of Pathology, School of Medicine, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Hanna Jeon
- Department of Pathology, School of Medicine, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Sara Cazzaro
- Department of Molecular Medicine, USF Health, Morsani College of Medicine, Tampa, FL 33620, USA
| | - Yasith I. Gamage
- Department of Physics, University of South Florida, Tampa, FL 33620, USA
| | - Jianjun Pan
- Department of Physics, University of South Florida, Tampa, FL 33620, USA
| | - Jung-A. A. Woo
- Department of Pathology, School of Medicine, Case Western Reserve University, Cleveland, OH 44106, USA
| | - David E. Kang
- Department of Pathology, School of Medicine, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Martin Muschol
- Department of Physics, University of South Florida, Tampa, FL 33620, USA
| |
Collapse
|
35
|
Wei YC, Kung YC, Lin CP, Chen CK, Lin C, Tseng RY, Chen YL, Huang WY, Chen PY, Chong ST, Shyu YC, Chang WC, Yeh CH. White matter alterations and their associations with biomarkers and behavior in subjective cognitive decline individuals: a fixel-based analysis. BEHAVIORAL AND BRAIN FUNCTIONS : BBF 2024; 20:12. [PMID: 38778325 PMCID: PMC11110460 DOI: 10.1186/s12993-024-00238-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/06/2023] [Accepted: 05/04/2024] [Indexed: 05/25/2024]
Abstract
BACKGROUND Subjective cognitive decline (SCD) is an early stage of dementia linked to Alzheimer's disease pathology. White matter changes were found in SCD using diffusion tensor imaging, but there are known limitations in voxel-wise tensor-based methods. Fixel-based analysis (FBA) can help understand changes in white matter fibers and how they relate to neurodegenerative proteins and multidomain behavior data in individuals with SCD. METHODS Healthy adults with normal cognition were recruited in the Northeastern Taiwan Community Medicine Research Cohort in 2018-2022 and divided into SCD and normal control (NC). Participants underwent evaluations to assess cognitive abilities, mental states, physical activity levels, and susceptibility to fatigue. Neurodegenerative proteins were measured using an immunomagnetic reduction technique. Multi-shell diffusion MRI data were collected and analyzed using whole-brain FBA, comparing results between groups and correlating them with multidomain assessments. RESULTS The final enrollment included 33 SCD and 46 NC participants, with no significant differences in age, sex, or education between the groups. SCD had a greater fiber-bundle cross-section than NC (pFWE < 0.05) at bilateral frontal superior longitudinal fasciculus II (SLFII). These white matter changes correlate negatively with plasma Aβ42 level (r = -0.38, p = 0.01) and positively with the AD8 score for subjective cognitive complaints (r = 0.42, p = 0.004) and the Hamilton Anxiety Rating Scale score for the degree of anxiety (Ham-A, r = 0.35, p = 0.019). The dimensional analysis of FBA metrics and blood biomarkers found positive correlations of plasma neurofilament light chain with fiber density at the splenium of corpus callosum (pFWE < 0.05) and with fiber-bundle cross-section at the right thalamus (pFWE < 0.05). Further examination of how SCD grouping interacts between the correlations of FBA metrics and multidomain assessments showed interactions between the fiber density at the corpus callosum with letter-number sequencing cognitive score (pFWE < 0.01) and with fatigue to leisure activities (pFWE < 0.05). CONCLUSION Based on FBA, our investigation suggests white matter structural alterations in SCD. The enlargement of SLFII's fiber cross-section is linked to plasma Aβ42 and neuropsychiatric symptoms, which suggests potential early axonal dystrophy associated with Alzheimer's pathology in SCD. The splenium of the corpus callosum is also a critical region of axonal degeneration and cognitive alteration for SCD.
Collapse
Affiliation(s)
- Yi-Chia Wei
- Department of Neurology, Chang Gung Memorial Hospital, Keelung, 204, Taiwan
- Community Medicine Research Center, Chang Gung Memorial Hospital, Keelung, 204, Taiwan
- College of Medicine, Chang Gung University, Taoyuan, 333, Taiwan
- Institute of Neuroscience, National Yang Ming Chiao Tung University, Taipei, 112, Taiwan
| | - Yi-Chia Kung
- Department of Radiology, Tri-Service General Hospital, National Defense Medical Center, Taipei, 114, Taiwan
| | - Ching-Po Lin
- Institute of Neuroscience, National Yang Ming Chiao Tung University, Taipei, 112, Taiwan
- Department of Education and Research, Taipei City Hospital, Taipei, Taiwan
| | - Chih-Ken Chen
- Community Medicine Research Center, Chang Gung Memorial Hospital, Keelung, 204, Taiwan
- College of Medicine, Chang Gung University, Taoyuan, 333, Taiwan
- Department of Psychiatry, Chang Gung Memorial Hospital, Keelung, 204, Taiwan
| | - Chemin Lin
- Community Medicine Research Center, Chang Gung Memorial Hospital, Keelung, 204, Taiwan
- College of Medicine, Chang Gung University, Taoyuan, 333, Taiwan
- Department of Psychiatry, Chang Gung Memorial Hospital, Keelung, 204, Taiwan
| | - Rung-Yu Tseng
- Department of Medical Imaging and Radiological Sciences, Chang Gung University, Taoyuan, 333, Taiwan
| | - Yao-Liang Chen
- Department of Medical Imaging and Radiological Sciences, Chang Gung University, Taoyuan, 333, Taiwan
- Department of Radiology, Chang Gung Memorial Hospital, Keelung, 204, Taiwan
| | - Wen-Yi Huang
- Department of Neurology, Chang Gung Memorial Hospital, Keelung, 204, Taiwan
- College of Medicine, Chang Gung University, Taoyuan, 333, Taiwan
| | - Pin-Yuan Chen
- Community Medicine Research Center, Chang Gung Memorial Hospital, Keelung, 204, Taiwan
- College of Medicine, Chang Gung University, Taoyuan, 333, Taiwan
- Department of Neurosurgery, Chang Gung Memorial Hospital, Keelung, 204, Taiwan
| | - Shin-Tai Chong
- Institute of Neuroscience, National Yang Ming Chiao Tung University, Taipei, 112, Taiwan
| | - Yu-Chiau Shyu
- Community Medicine Research Center, Chang Gung Memorial Hospital, Keelung, 204, Taiwan
- Department of Nursing, Chang Gung University of Science and Technology, Taoyuan, 333, Taiwan
| | - Wei-Chou Chang
- Department of Radiology, Tri-Service General Hospital, National Defense Medical Center, Taipei, 114, Taiwan
| | - Chun-Hung Yeh
- Department of Medical Imaging and Radiological Sciences, Chang Gung University, Taoyuan, 333, Taiwan.
- Department of Psychiatry, Chang Gung Memorial Hospital at Linkou, Taoyuan, 333, Taiwan.
| |
Collapse
|
36
|
Caneus J, Autar K, Akanda N, Grillo M, Long C, Jackson M, Lindquist S, Guo X, Morgan D, Hickman JJ. Validation of a functional human AD model with four AD therapeutics utilizing patterned iPSC-derived cortical neurons integrated with microelectrode arrays. RESEARCH SQUARE 2024:rs.3.rs-4313679. [PMID: 38826367 PMCID: PMC11142300 DOI: 10.21203/rs.3.rs-4313679/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: 06/04/2024]
Abstract
Preclinical methods are needed for screening potential Alzheimer's disease (AD) therapeutics that recapitulate phenotypes found in the Mild Cognitive Impairment (MCI) stage or even before this stage of the disease. This would require a phenotypic system that reproduces cognitive deficits without significant neuronal cell death to mimic the clinical manifestations of AD during these stages. A potential functional parameter to be monitored is long-term potentiation (LTP), which is a correlate of learning and memory, that would be one of the first functions effected by AD onset. Mature human iPSC-derived cortical neurons and primary astrocytes were co-cultured on microelectrode arrays (MEA) where surface chemistry was utilized to create circuit patterns connecting two adjacent electrodes to model LTP function. LTP maintenance was significantly reduced in the presence of Amyloid-Beta 42 (Aβ42) oligomers compared to the controls, however, co-treatment with AD therapeutics (Donepezil, Memantine, Rolipram and Saracatinib) corrected Aβ42 induced LTP impairment. The results presented here illustrate the significance of the system as a validated platform that can be utilized to model and study MCI AD pathology, and potentially for the pre-MCI phase before the occurrence of significant cell death. It also has the potential to become an ideal platform for high content therapeutic screening for other neurodegenerative diseases.
Collapse
|
37
|
Ren W, Yan XS, Fan JC, Huo DS, Wang XX, Jia JX, Yang ZJ. Effect of total flavonoids of Dracocephalum moldavica L. On neuroinflammation in Alzheimer's disease model amyloid-β (Aβ1-42)-peptide-induced astrocyte activation. JOURNAL OF TOXICOLOGY AND ENVIRONMENTAL HEALTH. PART A 2024; 87:436-447. [PMID: 38557424 DOI: 10.1080/15287394.2024.2336570] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/04/2024]
Abstract
One of the main pathological features noted in Alzheimer's disease (AD) is the presence of plagues of aggregated β-amyloid (Aβ1-42)-peptides. Excess deposition of amyloid-β oligomers (AβO) are known to promote neuroinflammation. Sequentially, following neuroinflammation astrocytes become activated with cellular characteristics to initiate activated astrocytes. The purpose of this study was to determine whether total flavonoids derived from Dracocephalum moldavica L. (TFDM) inhibited Aβ1-42-induced damage attributed to activated C8-D1A astrocytes. Western blotting and ELISA were used to determine the expression of glial fibrillary acidic protein (GFAP), and complement C3 to establish the activation status of astrocytes following induction from exposure to Aβ1-42. Data demonstrated that stimulation of C8-D1A astrocytes by treatment with 40 μM Aβ1-42 for 24 hr produced significant elevation in protein expression and protein levels of acidic protein (GFAP) and complement C3 accompanied by increased expression and levels of inflammatory cytokines. Treatment with TFDM or the clinically employed drug donepezil in AD therapy reduced production of inflammatory cytokines, and toxicity initiated following activation of C8-D1A astrocytes following exposure to Aβ1-42. Therefore, TFDM similar to donepezil inhibited inflammatory secretion in reactive astrocytes, suggesting that TFDM may be considered as a potential compound to be utilized in AD therapy.
Collapse
Affiliation(s)
- Wei Ren
- Department of Human Anatomy, Baotou Medical College, Inner Mongolia, China
- Key Laboratory of Human Anatomy, Education Department of Inner Mongolia Autonomous Region
| | - Xu-Sheng Yan
- Department of Human Anatomy, Baotou Medical College, Inner Mongolia, China
- Key Laboratory of Human Anatomy, Education Department of Inner Mongolia Autonomous Region
| | - Jia-Cheng Fan
- Department of Human Anatomy, Baotou Medical College, Inner Mongolia, China
- Key Laboratory of Human Anatomy, Education Department of Inner Mongolia Autonomous Region
| | - Dong-Sheng Huo
- Department of Human Anatomy, Baotou Medical College, Inner Mongolia, China
- Key Laboratory of Human Anatomy, Education Department of Inner Mongolia Autonomous Region
| | - Xin-Xin Wang
- Key Laboratory of Human Anatomy, Education Department of Inner Mongolia Autonomous Region
- Department of pathology, Baotou Medical College, Inner Mongolia, China
| | - Jian-Xin Jia
- Department of Human Anatomy, Baotou Medical College, Inner Mongolia, China
- Key Laboratory of Human Anatomy, Education Department of Inner Mongolia Autonomous Region
| | - Zhan-Jun Yang
- Key Laboratory of Human Anatomy, Education Department of Inner Mongolia Autonomous Region
- Department of Human Anatomy, Chifeng University, Inner Mongolia, China
| |
Collapse
|
38
|
Mukherjee A, Biswas S, Roy I. Immunotherapy: An emerging treatment option for neurodegenerative diseases. Drug Discov Today 2024; 29:103974. [PMID: 38555032 DOI: 10.1016/j.drudis.2024.103974] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2023] [Revised: 03/18/2024] [Accepted: 03/26/2024] [Indexed: 04/02/2024]
Abstract
Accumulation of misfolded proteins and protein aggregates leading to degeneration of neurons is a hallmark of several neurodegenerative diseases. Therapy mostly relies on symptomatic relief. Immunotherapy offers a promising approach for the development of disease-modifying routes. Such strategies have shown remarkable results in oncology, and this promise is increasingly being realized for neurodegenerative diseases in advanced preclinical and clinical studies. This review highlights cases of passive and active immunotherapies in Parkinson's and Alzheimer's diseases. The reasons for success and failure, wherever available, and strategies to cross the blood-brain barrier, are discussed. The need for conditional modulation of the immune response is also reflected on.
Collapse
Affiliation(s)
- Abhiyanta Mukherjee
- Department of Biotechnology, National Institute of Pharmaceutical Education and Research, Sector 67, S.A.S. Nagar, Punjab 160062, India
| | - Soumojit Biswas
- Department of Biotechnology, National Institute of Pharmaceutical Education and Research, Sector 67, S.A.S. Nagar, Punjab 160062, India
| | - Ipsita Roy
- Department of Biotechnology, National Institute of Pharmaceutical Education and Research, Sector 67, S.A.S. Nagar, Punjab 160062, India.
| |
Collapse
|
39
|
Ciuperca I, Pujo-Menjouet L, Matar-Tine L, Torres N, Volpert V. A qualitative analysis of an A β-monomer model with inflammation processes for Alzheimer's disease. ROYAL SOCIETY OPEN SCIENCE 2024; 11:231536. [PMID: 39076807 PMCID: PMC11285901 DOI: 10.1098/rsos.231536] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/13/2023] [Revised: 02/13/2024] [Accepted: 02/13/2024] [Indexed: 07/31/2024]
Abstract
We introduce and study a new model for the progression of Alzheimer's disease (AD) incorporating the interactions of A β -monomers, oligomers, microglial cells and interleukins with neurons through different mechanisms such as protein polymerization, inflammation processes and neural stress reactions. To understand the complete interactions between these elements, we study a spatially homogeneous simplified model that allows us to determine the effect of key parameters such as degradation rates in the asymptotic behaviour of the system and the stability of equilibrium. We observe that inflammation appears to be a crucial factor in the initiation and progression of AD through a phenomenon of hysteresis with respect to the oligomer degradation rate d . This means that depending on the advanced state of the disease (given by the value of the A β -monomer degradation rate d : large value for an early stage and low value for an advanced stage) there exists a critical threshold of initial concentration of interleukins that determines if the disease persists or not in the long term. These results give perspectives on possible anti-inflammatory treatments that could be applied to mitigate the progression of AD. We also present numerical simulations that allow us to observe the effect of initial inflammation and monomer concentration in our model.
Collapse
Affiliation(s)
- Ionel Ciuperca
- CNRS, Ecole Centrale de Lyon, Université Jean Monnet, Universite Claude Bernard Lyon 1, ICJ UMR5208, INSA Lyon, Lyon, Villeurbanne69622, France
| | - Laurent Pujo-Menjouet
- CNRS, Ecole Centrale de Lyon, Université Jean Monnet, Universite Claude Bernard Lyon 1, ICJ UMR5208, INSA Lyon, Lyon, Villeurbanne69622, France
| | - Leon Matar-Tine
- CNRS, Ecole Centrale de Lyon, Université Jean Monnet, Universite Claude Bernard Lyon 1, ICJ UMR5208, INSA Lyon, Lyon, Villeurbanne69622, France
| | - Nicolas Torres
- Departamento de Matemática Aplicada, Universidad de Granada, Granada, Andalusia, Spain
| | - Vitaly Volpert
- CNRS, Ecole Centrale de Lyon, Université Jean Monnet, Universite Claude Bernard Lyon 1, ICJ UMR5208, INSA Lyon, Lyon, Villeurbanne69622, France
- Peoples’ Friendship University of Russia, Moscow117198, Russia
| |
Collapse
|
40
|
Ruttenberg SM, Nowick JS. A turn for the worse: Aβ β-hairpins in Alzheimer's disease. Bioorg Med Chem 2024; 105:117715. [PMID: 38615460 DOI: 10.1016/j.bmc.2024.117715] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2024] [Revised: 04/04/2024] [Accepted: 04/05/2024] [Indexed: 04/16/2024]
Abstract
Amyloid-β (Aβ) oligomers are a cause of neurodegeneration in Alzheimer's disease (AD). These soluble aggregates of the Aβ peptide have proven difficult to study due to their inherent metastability and heterogeneity. Strategies to isolate and stabilize homogenous Aβ oligomer populations have emerged such as mutations, covalent cross-linking, and protein fusions. These strategies along with molecular dynamics simulations have provided a variety of proposed structures of Aβ oligomers, many of which consist of molecules of Aβ in β-hairpin conformations. β-Hairpins are intramolecular antiparallel β-sheets composed of two β-strands connected by a loop or turn. Three decades of research suggests that Aβ peptides form several different β-hairpin conformations, some of which are building blocks of toxic Aβ oligomers. The insights from these studies are currently being used to design anti-Aβ antibodies and vaccines to treat AD. Research suggests that antibody therapies designed to target oligomeric Aβ may be more successful at treating AD than antibodies designed to target linear epitopes of Aβ or fibrillar Aβ. Aβ β-hairpins are good epitopes to use in antibody development to selectively target oligomeric Aβ. This review summarizes the research on β-hairpins in Aβ peptides and discusses the relevance of this conformation in AD pathogenesis and drug development.
Collapse
Affiliation(s)
- Sarah M Ruttenberg
- Department of Chemistry, University of California, Irvine, Irvine, CA 92697-2025, United States
| | - James S Nowick
- Department of Chemistry, University of California, Irvine, Irvine, CA 92697-2025, United States.
| |
Collapse
|
41
|
Maity B, Kameyama S, Tian J, Pham TT, Abe S, Chatani E, Murata K, Ueno T. Fusion of amyloid beta with ferritin yields an isolated oligomeric beta-sheet-rich aggregate inside the ferritin cage. Biomater Sci 2024; 12:2408-2417. [PMID: 38511491 DOI: 10.1039/d4bm00173g] [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: 03/22/2024]
Abstract
Alzheimer's disease is a severe brain condition caused by the formation of amyloid plaques composed of amyloid beta (Aβ) peptides. These peptides form oligomers, protofibrils, and fibrils before deposition into amyloid plaques. Among these intermediates, Aβ oligomers (AβOs) were found to be the most toxic and therefore an appealing target for drug development and understanding their role in the disease. However, precise isolation and characterization of AβOs have proven challenging because AβOs tend to aggregate and form heterogeneous mixtures in solution. As a solution, we genetically fused the Aβ peptide with a ferritin monomer. Such fusion allowed the encapsulation of precisely 24 Aβ peptides inside the 24-mer ferritin cage. Using high-speed atomic force microscopy (HS-AFM), we disassembled ferritin and directly visualized the Aβ core enclosed within the cage. The thioflavin-T assay (ThT) and attenuated total reflection infrared spectroscopy (ATR-IR) revealed the presence of a β-sheet structure in the encapsulated oligomeric aggregate. Gallic acid, an amyloid inhibitor, can inhibit the fluorescence of ThT bound AβOs. Our approach represents a significant advancement in the isolation and characterization of β-sheet rich AβOs and is expected to be useful for future studies of other disordered peptides such as α-synuclein and tau.
Collapse
Affiliation(s)
- Basudev Maity
- School of Life Science and Technology, Tokyo Institute of Technology, Nagatsuta-cho, 4259, Midori-ku, Yokohama 226 8501, Japan.
| | - Shiori Kameyama
- School of Life Science and Technology, Tokyo Institute of Technology, Nagatsuta-cho, 4259, Midori-ku, Yokohama 226 8501, Japan.
| | - Jiaxin Tian
- School of Life Science and Technology, Tokyo Institute of Technology, Nagatsuta-cho, 4259, Midori-ku, Yokohama 226 8501, Japan.
| | - Thuc Toan Pham
- School of Life Science and Technology, Tokyo Institute of Technology, Nagatsuta-cho, 4259, Midori-ku, Yokohama 226 8501, Japan.
| | - Satoshi Abe
- School of Life Science and Technology, Tokyo Institute of Technology, Nagatsuta-cho, 4259, Midori-ku, Yokohama 226 8501, Japan.
| | - Eri Chatani
- Department of Chemistry, Graduate School of Science, Kobe University, Kobe, Hyogo 657-8501, Japan
| | - Kazuyoshi Murata
- Exploratory Research Center on Life and Living Systems (ExCELLS), National Institute for Natural Sciences, Okazaki, Aichi, 444-8585, Japan
- National Institute for Physiological Sciences (NIPS), National Institute for Natural Sciences, Okazaki, Aichi, 444-8585, Japan
| | - Takafumi Ueno
- School of Life Science and Technology, Tokyo Institute of Technology, Nagatsuta-cho, 4259, Midori-ku, Yokohama 226 8501, Japan.
- Living Systems Materialogy (LiSM) Research Group, International Research Frontiers Initiative (IRFI), Tokyo Institute of Technology, Nagatsuta-cho 4259, Midori-ku, Yokohama 226-8501, Japan
| |
Collapse
|
42
|
Chen Y, Lai M, Tao M. Evaluating the efficacy and safety of Alzheimer's disease drugs: A meta-analysis and systematic review. Medicine (Baltimore) 2024; 103:e37799. [PMID: 38640313 PMCID: PMC11029996 DOI: 10.1097/md.0000000000037799] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/17/2023] [Revised: 03/07/2024] [Accepted: 03/14/2024] [Indexed: 04/21/2024] Open
Abstract
BACKGROUND Alzheimer's disease (AD) is a progressive neurodegenerative disorder. Dementia severity was assessed mainly through cognitive function, psychobehavioral symptoms, and daily living ability. Currently, there are not many drugs that can be selected to treat mild to moderate AD, and the value of drugs remains controversial. OBJECTIVE The aim of this study is to quantitatively evaluate the efficacy and safety of cholinesterase inhibitors (ChEIs), memantine, and sodium oligomannate (GV-971) in the treatment of patients with AD. Additionally, molecular docking analysis will be used to investigate the binding affinities of donepezil, galantamine, rivastigmine, and memantine with key receptor proteins associated with AD, including beta-amyloid (Abeta), microtubule-associated protein (MAP), apolipoprotein E4 (APOE4), and Mitofusin-2 (MFN2), to further validate the results of the meta-analysis. METHODS We obtained clinical trials characterized by randomization, placebo control, and double-blinded methodologies concerning ChEIs, memantine, and GV-971. Statistical analysis was performed using Review Manager Version 5.4 software. Molecular docking was also conducted to evaluate the results. RESULTS All drugs improved the cognitive function, with the effect value ranging from -1.23 (95% CI -2.17 to -0.30) for 20 mg memantine to -3.29 (95% CI -4.14 to -2.45) for 32 mg galantamine. Although 32 mg galanthamine and GV-971 did not improve the clinicians' Global Impression of Change scale, other drugs showed significant results compared with placebo. On NPI, only 10 mg of donepezil and 24 mg of galantamine had improvement effects. On ADCS/ADL, only 20 mg memantine and 900 mg GV-971 had no significant difference from the placebo. Donepezil 5 mg and GV-971 900 mg did not increase the drug withdrawal rates due to various reasons or adverse reactions when compared to the placebo. Donepezil demonstrated superior binding to the protein and exhibited greater efficacy compared to other drugs. CONCLUSION ChEIs, memantine, and GV-971 all can slow the progression of AD but have different effects on respective assessments. Donepezil and GV-971 were relatively well tolerated.
Collapse
Affiliation(s)
- Yan Chen
- The Second Clinical Medical College of Zhejiang Chinese Medical University, Hangzhou, China
| | - Min Lai
- The Second Clinical Medical College of Zhejiang Chinese Medical University, Hangzhou, China
| | - Ming Tao
- The Second Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, China
| |
Collapse
|
43
|
Wilson DL, Carreon A, Chinnam S, Sharifan H, Ahlawat J, Narayan M. Screening Carbon Nano Materials for preventing amyloid protein aggregation by adopting a facile method. RESEARCH SQUARE 2024:rs.3.rs-4164618. [PMID: 38585783 PMCID: PMC10996794 DOI: 10.21203/rs.3.rs-4164618/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: 04/09/2024]
Abstract
The soluble-to-toxic transformation of intrinsically disordered amyloidogenic proteins such as amyloid beta (Aβ), α-synuclein, mutant Huntingtin Protein (mHTT) and islet amyloid polypeptide (IAPP) among others is associated with disorders such as Alzheimer's disease (AD), Parkinson's disease (PD), Huntington's disease (HD) and Type 2 Diabetes (T2D), respectively. The dissolution of mature fibrils and toxic amyloidogenic intermediates including oligomers continues to be the pinnacle in the treatment of neurodegenerative disorders. Yet, methods to effectively, and quantitatively, report on the interconversion between amyloid monomers, oligomers and mature fibrils fall short. Here we describe a simplified method that implements the use of gel electrophoresis to address the transformation between soluble monomeric amyloid proteins and mature amyloid fibrils. The technique implements an optimized but well-known, simple, inexpensive and quantitative assessment previously used to assess the oligomerization of amyloid monomers and subsequent amyloid fibrils. This method facilitates the screening of small molecules that disintegrate oligomers and fibrils into monomers, dimers, and trimers and/or retain amyloid proteins in their monomeric forms. Most importantly, our optimized method diminishes existing barriers associated with existing (alternative) techniques to evaluate fibril formation and intervention.
Collapse
Affiliation(s)
| | | | - Sampath Chinnam
- M.S. Ramaiah Institute of Technology (Autonoumous Institution, Affiliated to Visvesvaraya Technological University
| | | | | | | |
Collapse
|
44
|
Yu Z, Luo F. The Role of Reactive Oxygen Species in Alzheimer's Disease: From Mechanism to Biomaterials Therapy. Adv Healthc Mater 2024:e2304373. [PMID: 38508583 DOI: 10.1002/adhm.202304373] [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: 12/09/2023] [Revised: 03/13/2024] [Indexed: 03/22/2024]
Abstract
Alzheimer's disease (AD) is a chronic, insidious, and progressive neurodegenerative disease that remains a clinical challenge for society. The fully approved drug lecanemab exhibits the prospect of therapy against the pathological processes, while debatable adverse events conflict with the drug concentration required for the anticipated therapeutic effects. Reactive oxygen species (ROS) are involved in the pathological progression of AD, as has been demonstrated in much research regarding oxidative stress (OS). The contradiction between anticipated dosage and adverse event may be resolved through targeted transport by biomaterials and get therapeutic effects through pathological progression via regulation of ROS. Besides, biomaterials fix delivery issues by promoting the penetration of drugs across the blood-brain barrier (BBB), protecting the drug from peripheral degradation, and elevating bioavailability. The goal is to comprehensively understand the mechanisms of ROS in the progression of AD disease and the potential of ROS-related biomaterials in the treatment of AD. This review focuses on OS and its connection with AD and novel biomaterials in recent years against AD via OS to inspire novel biomaterial development. Revisiting these biomaterials and mechanisms associated with OS in AD via thorough investigations presents a considerable potential and bright future for improving effective interventions for AD.
Collapse
Affiliation(s)
- Zhuohang Yu
- State Key Laboratory of Oral Diseases and National Center for Stomatology and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Feng Luo
- State Key Laboratory of Oral Diseases and National Center for Stomatology and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, 610041, China
| |
Collapse
|
45
|
Chen S, Guo D, Zhu Y, Xiao S, Xie J, Zhang Z, Hu Y, Huang J, Ma X, Ning Z, Cao L, Cheng J, Tang Y. Amyloid β oligomer induces cerebral vasculopathy via pericyte-mediated endothelial dysfunction. Alzheimers Res Ther 2024; 16:56. [PMID: 38475929 PMCID: PMC10935813 DOI: 10.1186/s13195-024-01423-w] [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: 08/26/2023] [Accepted: 02/28/2024] [Indexed: 03/14/2024]
Abstract
BACKGROUND Although abnormal accumulation of amyloid beta (Aβ) protein is thought to be the main cause of Alzheimer's disease (AD), emerging evidence suggests a pivotal vascular contribution to AD. Aberrant amyloid β induces neurovascular dysfunction, leading to changes in the morphology and function of the microvasculature. However, little is known about the underlying mechanisms between Aβ deposition and vascular injuries. Recent studies have revealed that pericytes play a substantial role in the vasculopathy of AD. Additional research is imperative to attain a more comprehensive understanding. METHODS Two-photon microscopy and laser speckle imaging were used to examine cerebrovascular dysfunction. Aβ oligomer stereotactic injection model was established to explain the relationship between Aβ and vasculopathy. Immunofluorescence staining, western blot, and real-time PCR were applied to detect the morphological and molecular alternations of pericytes. Primary cultured pericytes and bEnd.3 cells were employed to explore the underlying mechanisms. RESULTS Vasculopathy including BBB damage, hypoperfusion, and low vessel density were found in the cortex of 8 to 10-month-old 5xFAD mice. A similar phenomenon accompanied by pericyte degeneration appeared in an Aβ-injected model, suggesting a direct relationship between Aβ and vascular dysfunction. Pericytes showed impaired features including low PDGFRβ expression and increased pro-inflammatory chemokines secretion under the administration of Aβ in vitro, of which supernatant cultured with bEND.3 cells led to significant endothelial dysfunction characterized by TJ protein deficiency. CONCLUSIONS Our results provide new insights into the pathogenic mechanism underlying Aβ-induced vasculopathy. Targeting pericyte therapies are promising to ameliorate vascular dysfunction in AD.
Collapse
Affiliation(s)
- Siqi Chen
- Department of Anesthesiology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, China
| | - Daji Guo
- Department of Neurology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, China
- Nanhai Translational Innovation Center of Precision Immunology, Sun Yat-sen Memorial Hospital, Foshan, 528200, China
| | - Yuanyuan Zhu
- Department of Neurology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, China
| | - Songhua Xiao
- Department of Neurology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, China
| | - Jiatian Xie
- Department of Neurology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, China
| | - Zhan Zhang
- Department of Neurology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, China
| | - Yu Hu
- Department of Neurology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, China
| | - Jialin Huang
- Department of Neurology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, China
| | - Xueying Ma
- Department of Neurology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, China
| | - Zhiyuan Ning
- Department of Neurology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, China
| | - Lin Cao
- Department of Anesthesiology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, China.
- Brain Research Center, Sun Yat-sen Memorial Hospital, Sun Yat‑sen University, Guangzhou, 510120, China.
| | - Jinping Cheng
- Department of Neurology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, China.
- Brain Research Center, Sun Yat-sen Memorial Hospital, Sun Yat‑sen University, Guangzhou, 510120, China.
- Nanhai Translational Innovation Center of Precision Immunology, Sun Yat-sen Memorial Hospital, Foshan, 528200, China.
| | - Yamei Tang
- Department of Neurology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, China.
- Brain Research Center, Sun Yat-sen Memorial Hospital, Sun Yat‑sen University, Guangzhou, 510120, China.
- Nanhai Translational Innovation Center of Precision Immunology, Sun Yat-sen Memorial Hospital, Foshan, 528200, China.
| |
Collapse
|
46
|
Gales L. Detection and clearance in Alzheimer's disease: leading with illusive chemical, structural and morphological features of the targets. Neural Regen Res 2024; 19:497-498. [PMID: 37721271 PMCID: PMC10581563 DOI: 10.4103/1673-5374.380897] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2023] [Revised: 05/20/2023] [Accepted: 05/31/2023] [Indexed: 09/19/2023] Open
Affiliation(s)
- Luís Gales
- i3S - Instituto de Investigaçã o e Inovaçã o em Saúde, Rua Alfredo Allen, Porto, Portugal
- IBMC − Instituto de Biologia Molecular e Celular, Universidade do Porto, Rua Alfredo Allen, Porto, Portugal
- ICBAS - Instituto de Ciências Biomédicas Abel Salazar, Rua de Jorge Viterbo Ferreira, Porto, Portugal
| |
Collapse
|
47
|
Xue H, Chen J, Zeng L, Fan W. Causal relationship between circulating immune cells and the risk of Alzheimer's disease: A Mendelian randomization study. Exp Gerontol 2024; 187:112371. [PMID: 38301877 DOI: 10.1016/j.exger.2024.112371] [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: 12/10/2023] [Revised: 01/23/2024] [Accepted: 01/26/2024] [Indexed: 02/03/2024]
Abstract
BACKGROUND Increasing evidence has shown a link between immune cells and Alzheimer's disease (AD). Comprehensive two-sample Mendelian randomization (MR) analysis was performed to determine the causal association between 731 immune cell signatures and AD in this study. METHODS We extracted genetic variants of 731 immune cell traits and AD from the publicly available GWAS dataset. The immune features included median fluorescence intensity (MFI), relative cellular (RC), absolute cellular (AC) and morphological parameters (MP). The inverse variance weighted (IVW) method was the main MR analysis method, and sensitivity analyses were used to validate the robustness, heterogeneity and horizontal pleiotropy of the results. RESULTS After FDR adjustment, seven immune phenotypes were found to be associated significantly with AD risk: HLA DR on CD33-HLA DR+ (OR = 0.938, PFDR = 0.001), Secreting Treg %CD4 (OR = 0.972, PFDR = 0.021), HLA DR+T cell AC (OR = 0.928, PFDR = 0.041), Activated & resting Treg % CD4 Treg (OR = 1.031, PFDR = 0.002), CD33 on CD33dim HLA DR+CD11b+ (OR = 1.025, PFDR = 0.025), CD33 on CD14+monocyte (OR = 1.026, PFDR = 0.027) and CD33 on CD66b++myeloid cell (OR = 1.027, PFDR = 0.036). CONCLUSIONS These findings demonstrated seven immune phenotypes were significantly associated with AD risk. This may provide researchers with a new perspective in exploring the biological mechanisms of AD and may lead to the exploration of earlier treatment.
Collapse
Affiliation(s)
- Hua Xue
- Department of Neurology, Sichuan Taikang Hospital, Chengdu, Sichuan, China.
| | - Jiajia Chen
- College of Chemical Engineering, Sichuan University of Science & Engineering, Zigong, China
| | - Li Zeng
- Department of Respiratory, Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, Guangxi, China
| | - Wenhui Fan
- Department of Neurology, Sichuan Taikang Hospital, Chengdu, Sichuan, China.
| |
Collapse
|
48
|
Parekh P, Badachhape AA, Tanifum EA, Annapragada AV, Ghaghada KB. Advances in nanoprobes for molecular MRI of Alzheimer's disease. WILEY INTERDISCIPLINARY REVIEWS. NANOMEDICINE AND NANOBIOTECHNOLOGY 2024; 16:e1946. [PMID: 38426638 PMCID: PMC10983770 DOI: 10.1002/wnan.1946] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/21/2023] [Revised: 01/11/2024] [Accepted: 01/30/2024] [Indexed: 03/02/2024]
Abstract
Alzheimer's disease is the most common cause of dementia and a leading cause of mortality in the elderly population. Diagnosis of Alzheimer's disease has traditionally relied on evaluation of clinical symptoms for cognitive impairment with a definitive diagnosis requiring post-mortem demonstration of neuropathology. However, advances in disease pathogenesis have revealed that patients exhibit Alzheimer's disease pathology several decades before the manifestation of clinical symptoms. Magnetic resonance imaging (MRI) plays an important role in the management of patients with Alzheimer's disease. The clinical availability of molecular MRI (mMRI) contrast agents can revolutionize the diagnosis of Alzheimer's disease. In this article, we review advances in nanoparticle contrast agents, also referred to as nanoprobes, for mMRI of Alzheimer's disease. This article is categorized under: Diagnostic Tools > In Vivo Nanodiagnostics and Imaging Therapeutic Approaches and Drug Discovery > Nanomedicine for Neurological Disease.
Collapse
Affiliation(s)
- Parag Parekh
- Department of Radiology, Baylor College of Medicine, Houston, Texas 77030
- Department of Radiology, Texas Children's Hospital, Houston, Texas 77030
| | - Andrew A. Badachhape
- Department of Radiology, Baylor College of Medicine, Houston, Texas 77030
- Department of Radiology, Texas Children's Hospital, Houston, Texas 77030
| | - Eric A. Tanifum
- Department of Radiology, Baylor College of Medicine, Houston, Texas 77030
- Department of Radiology, Texas Children's Hospital, Houston, Texas 77030
| | - Ananth V. Annapragada
- Department of Radiology, Baylor College of Medicine, Houston, Texas 77030
- Department of Radiology, Texas Children's Hospital, Houston, Texas 77030
| | - Ketan B. Ghaghada
- Department of Radiology, Baylor College of Medicine, Houston, Texas 77030
- Department of Radiology, Texas Children's Hospital, Houston, Texas 77030
| |
Collapse
|
49
|
Tolar M, Hey JA, Power A, Abushakra S. The Single Toxin Origin of Alzheimer's Disease and Other Neurodegenerative Disorders Enables Targeted Approach to Treatment and Prevention. Int J Mol Sci 2024; 25:2727. [PMID: 38473975 PMCID: PMC10932387 DOI: 10.3390/ijms25052727] [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: 01/15/2024] [Revised: 02/20/2024] [Accepted: 02/23/2024] [Indexed: 03/14/2024] Open
Abstract
New data suggest that the aggregation of misfolded native proteins initiates and drives the pathogenic cascade that leads to Alzheimer's disease (AD) and other age-related neurodegenerative disorders. We propose a unifying single toxin theory of brain neurodegeneration that identifies new targets and approaches to the development of disease-modifying treatments. An extensive body of genetic evidence suggests soluble aggregates of beta-amyloid (Aβ) as the primary neurotoxin in the pathogenesis of AD. New insights from fluid biomarkers, imaging, and clinical studies provide further evidence for the decisive impact of toxic Aβ species in the initiation and progression of AD. Understanding the distinct roles of soluble and insoluble amyloid aggregates on AD pathogenesis has been the key missing piece of the Alzheimer's puzzle. Data from clinical trials with anti-amyloid agents and recent advances in the diagnosis of AD demonstrate that the driving insult in biologically defined AD is the neurotoxicity of soluble Aβ aggregates, called oligomers and protofibrils, rather than the relatively inert insoluble mature fibrils and amyloid plaques. Amyloid oligomers appear to be the primary factor causing the synaptic impairment, neuronal stress, spreading of tau pathology, and eventual cell death that lead to the clinical syndrome of AD dementia. All other biochemical effects and neurodegenerative changes in the brain that are observed in AD are a response to or a downstream effect of this initial toxic insult by oligomers. Other neurodegenerative disorders follow a similar pattern of pathogenesis, in which normal brain proteins with important biological functions become trapped in the aging brain due to impaired clearance and then misfold and aggregate into neurotoxic species that exhibit prion-like behavior. These aggregates then spread through the brain and cause disease-specific neurodegeneration. Targeting the inhibition of this initial step in neurodegeneration by blocking the misfolding and aggregation of healthy proteins has the potential to slow or arrest disease progression, and if treatment is administered early in the course of AD and other neurodegenerative disorders, it may delay or prevent the onset of clinical symptoms.
Collapse
|
50
|
Tanaka M, Hirayoshi Y, Minatani S, Hasegawa I, Itoh Y. Diffusion Mediates Molecular Transport through the Perivascular Space in the Brain. Int J Mol Sci 2024; 25:2480. [PMID: 38473727 DOI: 10.3390/ijms25052480] [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: 01/24/2024] [Revised: 02/13/2024] [Accepted: 02/19/2024] [Indexed: 03/14/2024] Open
Abstract
The perivascular space has been proposed as a clearance pathway for degradation products in the brain, including amyloid β, the accumulation of which may induce Alzheimer's disease. Live images were acquired using a two-photon microscope through a closed cranial window in mice. In topical application experiments, the dynamics of FITC-dextran were evaluated from 30 to 150 min after the application and closure of the window. In continuous injection experiments, image acquisition began before the continuous injection of FITC-dextran. The transport of dextran molecules of different sizes was evaluated. In topical application experiments, circumferential accumulation around the penetrating arteries, veins, and capillaries was observed, even at the beginning of the observation period. No further increases were detected. In continuous injection experiments, a time-dependent increase in the fluorescence intensity was observed around the penetrating arteries and veins. Lower-molecular-weight dextran was transported more rapidly than higher-molecular-weight dextran, especially around the arteries. The largest dextran molecules were not transported significantly during the observation period. The size-dependent transport of dextran observed in the present study strongly suggests that diffusion is the main mechanism mediating substance transport in the perivascular space.
Collapse
Affiliation(s)
- Marie Tanaka
- Department of Neurology, Osaka City University Graduate School of Medicine, Osaka 545-8585, Japan
| | - Yoko Hirayoshi
- Department of Neurology, Osaka City University Graduate School of Medicine, Osaka 545-8585, Japan
| | - Shinobu Minatani
- Department of Neurology, Osaka City University Graduate School of Medicine, Osaka 545-8585, Japan
| | - Itsuki Hasegawa
- Department of Neurology, Osaka City University Graduate School of Medicine, Osaka 545-8585, Japan
| | - Yoshiaki Itoh
- Department of Neurology, Osaka City University Graduate School of Medicine, Osaka 545-8585, Japan
| |
Collapse
|